ArticlePDF Available

TIGIT and PD-1 Immune Checkpoint Pathways Are Associated With Patient Outcome and Anti-Tumor Immunity in Glioblastoma

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

Glioblastoma (GBM) remains an aggressive brain tumor with a high rate of mortality. Immune checkpoint (IC) molecules are expressed on tumor infiltrating lymphocytes (TILs) and promote T cell exhaustion upon binding to IC ligands expressed by the tumor cells. Interfering with IC pathways with immunotherapy has promoted reactivation of anti-tumor immunity and led to success in several malignancies. However, IC inhibitors have achieved limited success in GBM patients, suggesting that other checkpoint molecules may be involved with suppressing TIL responses. Numerous IC pathways have been described, with current testing of inhibitors underway in multiple clinical trials. Identification of the most promising checkpoint pathways may be useful to guide the future trials for GBM. Here, we analyzed the The Cancer Genome Atlas (TCGA) transcriptomic database and identified PD1 and TIGIT as top putative targets for GBM immunotherapy. Additionally, dual blockade of PD1 and TIGIT improved survival and augmented CD8⁺ TIL accumulation and functions in a murine GBM model compared with either single agent alone. Furthermore, we demonstrated that this combination immunotherapy affected granulocytic/polymorphonuclear (PMN) myeloid derived suppressor cells (MDSCs) but not monocytic (Mo) MDSCs in in our murine gliomas. Importantly, we showed that suppressive myeloid cells express PD1, PD-L1, and TIGIT-ligands in human GBM tissue, and demonstrated that antigen specific T cell proliferation that is inhibited by immunosuppressive myeloid cells can be restored by TIGIT/PD1 blockade. Our data provide new insights into mechanisms of GBM αPD1/αTIGIT immunotherapy.
This content is subject to copyright.
TIGIT and PD-1 Immune Checkpoint
Pathways Are Associated With
Patient Outcome and Anti-Tumor
Immunity in Glioblastoma
Itay Raphael
1
, Rajeev Kumar
1
, Lauren H. McCarl
1
, Karsen Shoger
1
, Lin Wang
2
,
Poorva Sandlesh
1
, Chaim T. Sneiderman
1
, Jordan Allen
1
, Shuyan Zhai
3
,
Marissa Lynn Campagna
1
, Alexandra Foster
1
, Tullia C. Bruno
4
, Sameer Agnihotri
1
,
Baoli Hu
1
, Brandyn A. Castro
5
, Frank S. Lieberman
6
, Alberto Broniscer
7
, Aaron A. Diaz
2
,
Nduka M. Amankulor
1
, Dhivyaa Rajasundaram
7
, Ian F. Pollack
1
and Gary Kohanbash
1,4
*
1
Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States,
2
Departments of Neurological
Surgery, University of California, San Francisco, CA, United States,
3
University of Pittsburgh Medical Center (UPMC) Hillman
Cancer Center Biostatistics Facility, University of Pittsburgh, Pittsburgh, PA, United States,
4
Department of Pediatrics,
University of Pittsburgh, Pittsburgh, PA, United States,
5
Departments of Neurology, University of Chicago, Chicago, IL,
United States,
6
Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States,
7
Department of Pediatrics,
Division of Health Informatics, Childrens Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh,
PA, United States
Glioblastoma (GBM) remains an aggressive brain tumor with a high rate of mortality.
Immune checkpoint (IC) molecules are expressed on tumor inltrating lymphocytes (TILs)
and promote T cell exhaustion upon binding to IC ligands expressed by the tumor cells.
Interfering with IC pathways with immunotherapy has promoted reactivation of anti-tumor
immunity and led to success in several malignancies. However, IC inhibitors have
achieved limited success in GBM patients, suggesting that other checkpoint molecules
may be involved with suppressing TIL responses. Numerous IC pathways have been
described, with current testing of inhibitors underway in multiple clinical trials. Identication
of the most promising checkpoint pathways may be useful to guide the future trials for
GBM. Here, we analyzed the The Cancer Genome Atlas (TCGA) transcriptomic database
and identied PD1 and TIGIT as top putative targets for GBM immunotherapy.
Additionally, dual blockade of PD1 and TIGIT improved survival and augmented CD8
+
TIL accumulation and functions in a murine GBM model compared with either single agent
alone. Furthermore, we demonstrated that this combination immunotherapy affected
granulocytic/polymorphonuclear (PMN) myeloid derived suppressor cells (MDSCs) but
not monocytic (Mo) MDSCs in in our murine gliomas. Importantly, we showed that
suppressive myeloid cells express PD1, PD-L1, and TIGIT-ligands in human GBM tissue,
and demonstrated that antigen specic T cell proliferation that is inhibited by
immunosuppressive myeloid cells can be restored by TIGIT/PD1 blockade. Our data
provide new insights into mechanisms of GBM aPD1/aTIGIT immunotherapy.
Keywords: glioblastoma, immunotherapy, PD1, TIGIT, MDSCs, myeloid suppressor cell, gene network analyses
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371461
Edited by:
Sarah E. Church,
NanoString Technologies,
United States
Reviewed by:
Shane Curran,
EMD Millipore, United States
Patrick Wen,
DanaFarber Cancer Institute,
United States
*Correspondence:
Gary Kohanbash
gary.kohanbash2@chp.edu
These authors have contributed
equally to the work
Specialty section:
This article was submitted to
Cancer Immunity
and Immunotherapy,
a section of the journal
Frontiers in Immunology
Received: 02 December 2020
Accepted: 12 April 2021
Published: 07 May 2021
Citation:
Raphael I, Kumar R, McCarl LH,
Shoger K, Wang L, Sandlesh P,
Sneiderman CT, Allen J, Zhai S,
Campagna ML, Foster A, Bruno TC,
Agnihotri S, Hu B, Castro BA,
Lieberman FS, Broniscer A, Diaz AA,
Amankulor NM, Rajasundaram D,
Pollack IF and Kohanbash G (2021)
TIGIT and PD-1 Immune Checkpoint
Pathways Are Associated With Patient
Outcome and Anti-Tumor
Immunity in Glioblastoma.
Front. Immunol. 12:637146.
doi: 10.3389/fimmu.2021.637146
ORIGINAL RESEARCH
published: 07 May 2021
doi: 10.3389/fimmu.2021.637146
INTRODUCTION
Malignant gliomas are the most common primary malignant
central nervous system (CNS) tumor in adults (1). Glioblastoma
(GBM) are highly aggressive brain cancers and the most
common type of high-grade glioma (HGG) (2). The current
standard of care for GBM patients include a combination of
surgery, radiation therapy, and chemotherapy. However, even
with standard of care, the median overall survival times remain
less than two years (3,4). Therefore, identication of novel GBM
treatment strategies is warranted.
Theimmunesystemcanmountspecic and durable
responses against tumors (5,6). However, cancer cells, tumor-
myeloid cells, and tumor inltrating regulatory T cells (Tregs)
can express negative regulators of the immune system including
immune checkpoint (IC) molecules, thereby limiting effective
anti-tumor immunity (7,8). In recent years, the development of
immunoregulatory drugs that block IC pathways, such as PD1/
PD-L1 inhibitors, have emerged as a promising treatment
strategy against a variety of malignancies, including melanoma,
lung cancers, and head and neck cancers (9,10). Although anti-
PD1/PD-L1 immunotherapy shows durable response in other
types of malignancies, its efcacy is limited to approximately 10%
of GBM patients (1113), thus highlighting the need for more
effective and novel approaches, including the combination of
additional IC inhibitors (ICIs) to target several IC
pathways simultaneously.
T cell immunoreceptor with Ig and ITIM domain (TIGIT) is
an IC receptor expressed on activated T cells, NK cells, and Tregs
(14). Elevated TIGIT expression on TILs correlates with reduced
TIL cytokine production and poor overall survival (14). TIGIT
binds with high-afnity to CD155 (PVR) and with low-afnity to
CD112 (PVRL2; nectin-2) which are expressed in the tumor
microenvironment (TME) by antigen presenting cells (APCs)
and tumor cells (15). The binding of TIGIT to CD155 suppresses
the activation of TILs. CD155 can also bind CD226, which is
expressed on T cells and provides a stimulatory signal which
promotes T cell activation, thus competing with TIGIT binding
to CD155. However, TIGIT has a signicantly greater afnity to
CD155 than CD226 (15,16). While blocking the interaction
between TIGIT and CD155 has been identied as a potential
therapeutic target in treatment of malignancies, its effects in
GBM are poorly understood (17).
MDSCs are myeloid-lineage regulatory cell that act as negative
immune regulators in the TME (18). MDSCs consist of two major
subtypes based on phenotype:PMN-MDSCsmatchedwith
granulocytes, and Mo-MDSC resembling inhibitory monocytes
(18). In mice, PMN-MDSC are dened a CD11b
+
Ly6C
low
Ly6G
high
,
and Mo-MDSC as CD11b
+
Ly6C
high
Ly6G
low
whereas in human,
PMN-MDSC are dened as CD14
-
CD11b
+
CD33
+
CD15
+
and Mo-
MDSC as CD14
+
CD11b
+
HLA-DR
low
. Some studies have shown that
increased presence of MDSCs within the TME is related to poor
clinical outcome in patients treated with ICI (19). Consequently,
reduced inltration of MDSCs in TME has shown enhanced anti-
tumor efcacy of ICI in pre-clinical tumor models (20,21).
In order to identify putative IC targets in GBM, we rst
analyzed of The Cancer Genome Atlas (TCGA) dataset and
identied IC molecules whose expression is associated with poor
survival in GBM patients. We found that upregulated expression
of PD1 and TIGIT, but not other ICs or their ligands, are
associated with reduced patient survival. We demonstrate that
dual treatment with aPD1/aTIGIT prolonged survival in a
murine GBM model, at least in part by targeting MDSCs.
Together, our data provide new insights into mechanisms of
immunotherapy in GBM.
MATERIALS AND METHODS
TCGA Data Analysis
The Cancer Genome Atlas (TCGA) database was used to assess
survival of patients with GBM in accordance with gene
expression levels of immune checkpoint molecules. Survival
analysis was performed through the cBioPortal platform using
a z-score of 1.0 for all checkpoint receptors and their respective
ligands. The correlation of checkpoint gene expression with z
score >2.0 was considered upregulated expression. Kaplan-Meier
(KM) survival curves were generated to determine overall
survival (OS) and disease-free survival (DFS).
RNA Sequencing (RNA-seq) and
Pathway Analysis
The study uses RNA-seq datasets of GBM tissue from The Cancer
Genome Atlas (TCGA), and the raw expression les were
downloaded from TCGA Genomic Data Commons (GDC) Data
Portal. Reads were quantied and mapped to human genome
(Ensembl GRCh38 Homo sapiens) Salmon version 0.8.2 (22).
Transcript-per-kilobase-million (TPM) were used for gene-
correlation and pathway analyses. Pearsons rank correlation
analysis was performed for TIGIT and PDCD1. Genes with
statistically signicant correlation (p value < 0.05 and false
discovery rate (FDR) p < 0.05) were used to determine pathway
enrichment using Gene Ontology (GO) (23)forReactome
(version 65 Released 2020-11-17) (24)andPANTHER
Overrepresentation Test (Released 20200728) (25)curated
pathways. Pathway enrichment cutoff was set for p<0.05 using
Fishers Exact test and FDR p<0.05 and enrichment scores greater
than 1. Immunological network analysis was performed using
ClueGo v2.5.7 (26) and Cytoscape 3.8 (27) with the current
parameters: GO ImmuneSystemProcess EBI-UniPort, GO term
fusion, network specicity was set to medium-detailed, pathways
p value <0.05 with Benjamini-Hochberg correction. Positively and
negatively correlated genes were used to determine positively and
negatively associated networks, respectively.
Single Cell RNA-seq (scRNA-seq) Analysis
scRNA-seq data were obtained from Wang et al. (28)and
processed as described previously (28). Briey, the neoplastic
cells and non-neoplastic cells were separated via copy number
variation (CNV). The presence/absence of CNVs was assessed
with CONICSmat (29), and the primary cell types of non-
neoplastic cells (i.e. monocytes/myeloid) were identied by
using ELSA (30). CD11b
+
monocyte/myeloid cell population
was sampled for further analysis using Seurat package on
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371462
Bioconductor (R) (31). Following Elbow Plot analysis, the
number of principal components analysis (PCA) was set up to
3 with 0.2 resolution for UMAP clustering.
Cell Lines
GL261 cells were cultured in Dulbeccos Modied Eagle Medium
(DMEM, Gibco) supplemented with 10% fetal bovine serum
(FBS, Hyclone), 1x antimycotic-antimycotic solution (Gibco),
1% L-glutamine, ß-mercaptoethanol, 200 µg sodium pyruvate,
and 1x NEAA. Cell lines were kept in a 37°C humidied
incubator with 5% CO2. Cell number and viability were
measured using the trypan exclusion method (0.4% trypan
Blue, Gibco).
Mice
C57BL/6J mice (Stock No. 000664) and B6.Cg-Thy1 a/Cy Tg
(TcraTcrb)8Rest/J (PMEL; Stock No. 005023 (32)) were purchased
from the Jackson Laboratory and housed in animal facility of the
UPMC Childrens Hospital of Pittsburgh. Animals were kept in the
facility for at least one week prior to performing any procedures to
minimize stress-related symptoms. 56-week-old female were used
in the experiments. All experiments were conducted following
protocols approved by the University of Pittsburgh Institutional
Animal Care and Use Committee (IACUC).
Intracranial Tumor Model and
Antibodies Treatment
Mice were anesthetized by mask inhalation of 1.5% vaporized
isourane throughout the surgical procedure. GL261 cells
(100,000 cells in 2 mL DPBS) were stereotactically implanted
into the caudate nucleus using the following coordinates
relative to bregma: x = +2.5 mm (lateral), y = +1.5 mm
(anterior), and z = 2-3.0 mm (inferior) (33). MRI was
performed 7 days post tumor cell implantation to conrm
tumor presence, and again at day 40 to measure tumor size
growth in control-treated animals and aTIGIT & aPD1 dual
blockade-treated animals. All mice were randomized prior to
their separation into treatment groups. IgG1 (clone MOPC-21),
IgG2a (clone RTK2758), aPD1 (clone RMP1-14) and aTIGIT
(clone 1G9) antibodies were obtained from Bio-X-Cell
Antibodies were dosed at 200 mg per animal and administered
via intraperitoneal (i.p.) injection, as described previously (34)
for both the survival and immunophenotyping studies. Anti-
TIGIT and anti-PD1 treatments were given on the same day
twice per week starting on day 8, for a total of 7 doses. Mice were
euthanized after receiving seven doses of immune-checkpoint
inhibitor therapeutic antibodies (aTIGIT/aPD1) to investigate
biological endpoint and immune cell phenotype. Mice were
monitored for weight loss and morbidity symptoms for
survival study. All survival experiments were repeated in
triplicate with at 4-6 animals per group.
Mouse Immune Cell Isolation
For the biological endpoint study, mice were euthanized on day
22 (CO2 asphyxiation followed by cervical dislocation) post-
tumor inoculation. Brains were dissected and processed for ow
cytometry analysis. Brains were homogenized in Collagenase IV
Cocktail (3.2 mg/mL collagenase type IV, 1.0 mg/mL
deoxyribonuclease I, 2 mg/mL Soybean Trypsin Inhibitor).
Samples were centrifuged for 5 minutes at 1500 rpm, followed
by red-blood cell (RBC) lysis using ACK lysing buffer (Lonza).
Cell viability was measured using the trypan blue exclusion
method. Cells were resuspended in FACS Buffer (DPBS with
1% BSA) and centrifuged for 5 minutes at 300 g, after which the
pellet was resuspended in FACS buffer. The cells were then
stained with appropriate antibodies and acquired on a BD LSR
Fortessa ow cytometer.
Isolation of TILs From GBM Patients
Patient-derived GBM tissue was dissociated, using Accutase
(1:10), to form a single cell suspension (SCS). SCS was
centrifuged at 1500rpm for 5 mins. The pellet was resuspended
in 5mL of 70% Percoll solution. A Percoll gradient of 5mL of 37
and 5mL of 30% Percoll sequentially, was then overlaid onto the
tumor-containing 70% Percoll solution. The tumor gradient
solution was centrifuged at 2400 rpm for 20 minutes. Immune
cells at the interphase were collected and washed once with PBS.
The cells were then stained with appropriate antibodies and
acquired on a BD LSR Fortessa ow cytometer.
Isolation of PBMCs
Peripheral blood samples were collected in preservative-free
heparin tubes (10 U/mL) and layered into an equal volume of
Ficoll-Hypaque density gradient solution (Amersham Pharmacia
Biotech Ltd., Little Chalfont, UK). Samples were then centrifuged
at 2250 rpm for 20 minutes. After removal of the top layer
(plasma), the mononuclear cells (PBMCs) were collected and
washed twice with PBS (Hyclone,GEHealthcare).Cell
viability was determined by trypan blue exclusion and
exceeded 95%. The cells were then stained with appropriate
antibodies and acquired on a BD LSR Fortessa ow cytometer.
Generation of Immunosuppressive Myeloid
Cells From Bone Marrow (BM)
Immunosuppressive myeloid cells were generated as described
previously (35). Briey, tibia and femur-derived BM cell from
C57BL/6j mice were cultured in complete DMEM media
supplemented with 10 ng/ml each of GM-CSF and IL-4. On
day 3, oating cells were removed, and medium was replaced
with 1:1 complete DMEM media to GL261 tumor-derived
conditioned media (TCM), supplemented with GM-CSF and
IL-4 for 3 additional days prior to use.
Suppression of T Cell Proliferation Assay
T cell suppression assay was performed as described previously
(21,36). In brief, hGP100-restricted (B6.Cg-Thy1a/Cy TCR-
transgenic) CD8
+
T cells were isolated from PMEL-mice (32)
using magnetic bead separation (Miltenyi Biotec) and labeled
with Cell-Trace proliferation dye (Invitrogen. Cat. No C34557)
according to the manufacture guidelines. Feeder cells (antigen
presenting cells) were generated from non-CD8
+
cell fraction
and were treated with 10 mg/ml of mitomycin at 37°C for 1 hour
to cease proliferation (37). T cells and feeder cells were co-
cultured with BM-derived immunosuppressive myeloid cells in
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371463
the presence of 100 U/mL hIL-2 (PeproTech), 100 µg/mL
hGP10025-33 peptide (antigen), and 10 µg/mL of either IgG2a
(RTK2758) as control, or aPD1 (RPMI14) and aTIGIT (1G9).
Cells were collected and analyzed on day 4 by ow cytometry.
Flow Cytometry
Prior to cell surface staining, samples were stained with cell
viability dyes (GhostDye or 7AAD) in PBS for 20 minutes in 4°C
and then washed with FACS buffer. For mouse immune cell
staining, the cell suspensions were blocked with 1% anti-mouse
Fc-receptor (CD16/CD32) in FACS buffer for 20 minutes, then
washed and stained with uorescently labeled anti-mouse
antibodies for 45 minutes in FACS buffer at 4°C. TILs (n=5)
and PBMCs from 2 matched, 3 unmatched and 3 healthy donor
(HD) patient samples (n=8) were washed with PBS and stained
with cell-surface antibodies for 30 minutes at 4°C per the
manufacture guidelines. After staining, cells were washed with
FACS buffer and xed with xation buffer (BD Cytox/
Cytoperm buffer). The cells were washed with FACS buffer,
resuspended in FACS buffer and analyzed by ow cytometry.
The antibody clones were purchased from BioLegend or
eBioscience and used for ow cytometry as follows: For mouse
cell staining: CD4 (GK1.5), CD8 (53-6.7), CD11b (M1/70), CD45
(30-F11), Gr-1 (RB6-8C5), CD3 (17A2, and 145-2C11),
Granzyme B (QA16A02). For human cell staining: CD45
(2D1), CD11b (ICRF44), CD3 (C3e/1308), CD8 (OKT-8), PD-
1 (EH12.2H7), PD-L1 (MIH2), CD33 (WM53), CD226 (11A8),
TIGIT (A15153G), CD155/PVR (SKII.4). GhostDyes
(TONOBO) UV450 and Red-780, and 7AAD were used to
stain for cell viability (live/dead) according to the manufacture
guidelines. Gating was performed on live CD45
+
cells to
designate all immune cells. All samples were analyzed on a BD
LSRFortessa (BD Biosciences). Data were analyzed using BD
FACSDiva (BD Biosciences) and FlowJo V10 data analysis
software (FlowJo LLC).
Statistical Analysis and Software
Kaplan-Meier survival curves were generated to determine
survival and then compared using the log-rank Mantel Cox
test. One-way analysis of variance (ANOVA) with Kruskal
Wallis multiple comparisons test was used to compare assays
containing more than two groups. Statistical signicance was
considered as p <0.05. Normal distribution was assumed unless
specied overwise in the text or gure legend. The analyses were
performed using GraphPad Prism 8 or Bioconductor (R
programing) on RStudio.
RESULTS
High Expression Level of Immune
Checkpoint Molecules Associated With
Overall Survival (OS) and Disease-Free
Survival (DFS) in GBM
To identify putative immunotherapy targets for GBM, we
evaluated the expression of IC genes and their ligands in RNA
sequencing (RNA-seq) data of 153 GBM tumor samples in the
TCGA database (38). We rst assessed the correlation of 15
established IC gene expression levels with overall survival (OS)
and disease-free survival (DFS) (39,40). Upregulated expression
was dened as expression z score greater than 2. Our data
demonstrate that upregulated expression (red lines) of TIGIT
and PDCD1 (gene encoding PD1) were associated with poor
patient outcome and increased mortality as compared with
patients who had no change in TIGIT and PDCD1 RNA
expression (green lines) (Figures 1A, B). Upregulated ICOS
expression was also associated with reduced OS and DFS,
although the data did not reach signicance (Figures 1A, B).
However, upregulated expression of other IC receptor genes,
including CTLA4,LAG3, TIM3 (HVAC1), BTLA4, and CD224
were not associated with changes in OS and DFS. Interestingly,
expression of genes for CD155 (PVR), PD-L1, and ICOS-L, the
ligands for TIGIT, PD1, and ICOS, respectively, was also
signicantly associated with decreased OS and DFS, whereas
upregulated expression of other IC gene ligands did not affect
these parameters in GBM patients (Figures 2A, B). Although our
survival analysis assessed patients with elevated expressed based
on Z score (i.e. compared to mean expression of that gene), we
further examined the absolute expression of each gene to
determine the extent of therapeutic utility among all patients.
Our data show that a large portion of patients showed to have
physiologically relevant expression levels (TPM>1) of the genes
encoding to the checkpoint receptors PD-L1 (94%) and CD155
(PVR; 100%) (Supplemental Figure 1). Additionally, PD1 and
TIGIT were reported to be expressed by large frequencies of
GBM CD4
+
and CD8
+
TILs (34,41). Taken together these data
suggest that PD1/TIGIT-targeted therapy may be relevant for
many patients with GBM.
TIGIT and PD1 Are Co-Expressed, Share
Common Gene Networks, but Are Also
Associated With Distinct Pathways in GBM
Our data revealed that TIGIT/CD155 and PD1/PD-L1
checkpoint genes were signicantly associated with GBM
clinical outcome, thus we next analyzed RNA-seq data from
these patients to identify genes and pathways which may be
involved with TIGIT and PD1 expression in GBM. Notably, the
expression of TIGIT and PDCD1 were signicantly correlated
with each other (Figure 3A), suggesting a rationale for dual
blockade of these checkpoint molecules in GBM patients. Despite
their signicant correlation, TIGIT and PDCD1 may be
associated with unique gene networks and pathways (42).
Therefore, we next interrogated the gene networks associated
with the expression of TIGIT and/or PDCD1 in GBM. We
identied a total of 6347 genes which correlated with TIGIT
and PDCD1 expression with high statistical signicance (p<0.05
and FDR<0.05) (Figure 3B). While many genes correlated with
both TIGIT and PD1 expression, we also identied a large
number of genes and pathways uniquely correlated with either
TIGIT or PD1 (Figures 3B, C).
TIGIT/PD1 (shared)-associated pathways included immune
related pathways, such as Toll-like receptor (TLR) signaling,
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371464
interleukins signaling (such as IL-10 and IL-2), TCR signaling
and T cell activation, and innate immune system pathways
(Figure 3D). Interestingly, TIGIT-associated pathways included
Treg development, MHC class I presentation, caspases and
death-receptors signaling, control of cell cycle transition,
regulation of TLR and Nf-kB signaling, and p53 regulation
(Figure 3D). PD1-associated pathways included cell motility,
oxidation and phagocytosis, IL-12 mediated Jak-STAT signaling,
MHC class II and antigen presentation, and EGF receptor
signaling (Figure 3D). Immunological network analysis showed
that many immune responses were strongly associated with the
expression of TIGIT and PD1, mostly T cell activating and
regulation of immunity, but also innate immune functions such
as leukocytes degranulation, and functions of macrophages and
dendritic cells (Figure 3E).
Together, these data suggest that upregulated expression of
TIGIT and PD1 may confer immunosuppression and tumor
aggression in GBM patients through both shared and distinct
pathways, and therefore targeting both these pathways may be
benecial for improving clinical outcome of GBM patients.
Combination of aTIGIT and aPD1
Immunotherapy Increases Numbers of TIL
Cytolytic CD8
+
T Cells and Prolongs Long-
Term Survival of GBM-Bearing Mice
Our data suggest a benecial outcome for IC blockade of TIGIT
and/or PD1 in GBM. To investigate this hypothesis, C57BL/6
mice were intracranially injected with syngeneic GL261 cells,
followed by 7 doses of immunotherapy with (1) isotype control
antibodies, (2) aPD1, (3) aTIGIT, or (4) a combination of
aTIGIT/aPD1 therapeutic antibodies, administered twice per-
week starting on day 8 post-tumor injection (Figure 4A).
Analysis of immune cells was performed uniformly across
groups on day 22 post-tumor implantation (biological
endpoint) followed by MRI analysis for tumor size on day 40
for control and dual aTIGIT/aPD1-treatment groups (Figure
4A). Control mice (Isotype; black line) displayed median survival
of 33 days (range: 29-51 days) with severe morbidity signs and
did not reach long-term survival endpoint (Figure 4B). While
aTIGIT monotherapy (green line) moderately improved
survival, treatment with aPD1 (blue line) or a combination
FIGURE 1 | Immune-checkpoint receptor genes associated with GBM patient outcome. TCGA patient survival data obtained from cBioPortal, and patients were
grouped based on gene expression z-scores to upregulated expression (z 2; red line) or no change expression (z <2; green line). The (A) overall survival rate and
(B) disease free survival rate, were plotted using Kaplan-Meier survival curves. P values reect one-way ANOVA with Kruskal Wallis comparison test. n=153.
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371465
treatment of aTIGIT/aPD1 (red line) signicantly prolonged
animal survival as compared with isotype treated animals
(Figure 4B). The median survival of aTIGIT treatment was 34
days (range: 32-43 days) while aPD1 monotherapy (green line)
was 37 days (range 32-74 days). Notably, aTIGIT/aPD1 dual
treatment most signicantly prolonged mice survival with
median survival of 48 days (range 39-74 days) (Figure 4B).
MRI analysis showed that in aTIGIT/aPD1 treated animals the
tumor size was signicantly smaller than tumors in isotype-
treated animals (Figure 4C). These data conrm previous results
in which immunotherapy combination of aPD1 with aTIGIT
reduced tumor burden and improved survival of mice with
glioma (34).
To examine a mechanism by which the combination therapy
improved anti-tumor immunity, we explored the effect of
treatment on tumor-inltrating lymphocytes (TILs) and their
cytolytic phenotype on day 22 post tumor implantation.
Although aPD1 monotherapy did not signicantly affected the
percentages of CD4
+
TILs, treatment with either aTIGIT or
aTIGIT/aPD1 resulted in a signicant increase of CD4
+
TILs as
compared with control animals (Figure 4D). Additionally, we
noted a signicant increase in percentages of CD8
+
TILs in
tumors in animals treated with aPD1 or aTIGIT/aPD1, but not
in aTIGIT-monotherapy treated animals (Figure 4E). Analysis
showed that treatment with either aPD1 or aTIGIT
monotherapy resulted in a mild increase in the percentages of
CD8
+
granzyme-B
+
TILs, while this effect was signicantly
increased in aTIGIT/aPD1 combination treatment
(Figure 4F). These data are complementary to previous results
showing that aPD1 or aTIGIT immunotherapy enhances the
expression of TNFaand IFNgin TILs from GBM (34) as well as
other cancers (43), and suggest that the therapeutic effect of
checkpoint blockade with aTIGIT/aPD1 may work through
distinct mechanisms to affect CD4
+
and CD8
+
TILs and
promote anti-glioma immunity.
Dual Blockade of TIGIT and PD1
Regulates MDSCs in GBM Murine Model
Previous reports have shown that MDSCs stimulate suppressive
mechanism to develop a pre-metastatic niche, promote tumor
growth, inhibit anti-tumor function of TILs, and negate
immunotherapy which results as resistance to IC blockade (44,
FIGURE 2 | Immune-checkpoint ligand genes associated with GBM patient outcome. TCGA patient survival data obtained from cBioPortal, and patients were
grouped based on gene expression z-scores to upregulated expression (z 2; red line) or no change expression (z <2; green line). The (A) overall survival rate, and
(B) disease free survival rates were plotted using Kaplan-Meier survival curves. P values reect one-way ANOVA with Kruskal Wallis comparison test. n=153.
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371466
FIGURE 3 |TIGIT and PDCD1 (PD1) exhibit shared immunological networks but have unique regulatory pathways in GBM. GBM patientsRNA-seq data was
obtained from TCGA, transcript per million (TPM) normalized reads were calculated per each patient and Pearsons correlation analysis was performed. n=153.
Genes with a statistically signicant (p<0.05 and FDR<0.05) positive correlation and negative correlation to TIGIT and PDCD1 expression were identied.
(A) Pearsons correlation analysis of TIGIT and PDCD1 expression. (B) Venn diagrams showing number of statistically signicant correlated genes unique and
overlapping within TIGIT and PD1 gene groups. (C) Number of statistically signicant (p<0.05 and FDR<0.05) pathway enriched in each corresponding gene group.
(D) Representative pathways which are positively and negatively enriched in the shared-gene group, TIGIT-associated group, and PD1-associated group.
(E) Network analysis for Gene Ontology (GO) Immunological Processes associated with TIGIT and PDCD1 positively correlated gene network. Statistically signicant
gene correlation and pathway enrichments were corrected for false discovery rate (FDR) using Benjamini-Hochberg test.
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371467
45). Furthermore, MDSCs have been shown to contribute to
immunosuppressive microenvironment in gliomas, including
GBM (4547). MDSCs were shown to express PD-L1 (48).
Additionally, inhibition of TIGIT was reported to abrogate
MDSC immunosuppressive capacity in vitro (49). Together,
these data suggest that targeting PD1 and TIGIT pathways may
affect MDSCs in GBM. However, the effects of these checkpoint on
MDSC inltration in gliomas are ill dened (45). We, therefore,
investigated if MDSCs were affected by immunotherapy in our
model on day 22 (biological endpoint; end of immunotherapy).
Shown in Figure 5A,gliomainltrating MDSC subsets were
characterized by the expression of Gr1 and CD11b as follows:
PMN MDSCs were dened as CD11b
+
Gr1
high
cells, whereas Mo
MDSCs were dened as CD11b
+
Gr1
intermediate (int)
cells (18). We
evaluated the levels of MDSCs and their subsets following
immunotherapy (Figure 5A; lower panel). Our data show, that
compared with isotype treatment, both aTIGIT monotherapy and
dual blockade of TIGIT & PD1 signicantly reduced the
frequencies of GL261 glioma inltrating MDSCs
(CD45
+
CD11b
+
Gr1
+
cells), most strikingly for aTIGIT/aPD1
combination therapy (Figure 5B). Treatment with aPD1
showed a trend of decreasing MDSC percentages, though the
results did not achieve statistical signicance (Figure 5B). Analysis
of MDSC subsets revealed that aTIGIT/aPD1 dual treatment
signicantly decreased the frequencies of PMN MDSCs (Figure 5C),
while Mo MDSCs levels remained mostly unaltered (Figure 5D).
Furthermore, we observed a statistically signicant increase in ratios
of CD8
+
T cells over total MDSCs in tumors when mice were treated
with aTIGIT monotherapy or aTIGIT/aPD1 combination therapy
(Figure 5E). Blockade of PD1 alone did not signicantly increase the
CD8
+
T cells/MDSCs ratios (Figure 5E). Together, our data reveal a
mechanism of TIGIT/PD1 blockade in glioma and suggest distinct
roles of these ICs on MDSC subsets and in regulating
tumor immunity.
Myeloid Cells Upregulate PD-L1 and
TIGIT-Ligands in GBM Which Inhibit
T Cell Functions
We next evaluated the potential of aTIGIT/PD1-immunotheraphy
to impact MDSC-like cell in GBM patients. For that, we rst
analyzed single-cell (sc)RNA-seq data of CD11b
+
myeloid cells
from GBM patients (28) for the expression of PD1, TIGIT, and their
ligands. Myeloid cells were conrmed based on the expression of
CD45 (PTPRC), CD14,andCSF1R genes (Figure 6A)(28,50).
Of note, we identied 4 unique clusters of tumor-associated
myeloid/macrophage cells (TAMs) in GBM, which had distinct
expression proles (Figure 6A and Supplemental Figure 2A). The
expression of CD33, an hematopoietic progenitor cell marker which
commonly used to identify pan-MDSCs (51), was distributed
throughout the TAM clusters. Nonetheless, the expression of
inhibitory and suppressive markers, including genes for IL-4R,
IL-10, IL-6, VEGFA, CCL2 and IL-1b(Figure 6A and
FIGURE 4 | Anti-TIGIT and anti-PD1 combination improves survival of GL261 glioma bearing mice. GL261 glioma cells were injected stereotactically in the caudate
putamen of C56BL/6J mice followed by immunotherapy treatment starting on day 8 post tumor injection. Mice were evaluated for T cell responses on day 22
(biological endpoint) and for tumor size by day MRI on day 40. (A) Schematic showing induction of GL261 glioma in mice following treatment regimen using anti-PD1
and anti-TIGIT immunotherapies. (B) Survival curves with Log-rank (Mantel-Cox) curve comparison test. Pooled data from 3 independent experiments. (C) Pooled
data and representative MRI images of tumor growth in murine GL261 glioma model in anti-PD1/anti-TIGIT treated group and isotype (control) treated animals.
Unpaired ttest with Welchs correction. n=5 per group. (DF) Percentages (%) of CD45
+
glioma-inltrating CD4
+
T cells (D), CD8
+
T cells (E), and CD8
+
granzyme
B
+
T cells (F), on day 22 following anti-TIGIT/anti-PD1 immunotherapy. Representative data of 3 experiments. n=5 per group. One-way ANOVA with multiple
comparisons test corrected for false discovery rate (FDR) using Benjamini-Hochberg test. Pvalues are as followed: *0.05, **0.01, ***0.001. NS, not signicant.
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371468
Supplemental Figure 2A), were mostly expressed by cluster 0,
suggesting that these cells had a tumor-promoting and immune-
suppressing functions, which resemble MDSC-like cells (52).
Interestingly, PDCD1 (PD1), CD274 (PD-L1), and CD226,were
also predominantly expressed by cluster 0 (Figure 6B and
Supplemental Figure 2B). CD155 (PVR) was also associated and
expressed by cluster 0, although less frequent than CD226, PD1, and
PD-L1 (Figure 6B and Supplemental Figure 2B). PVRL2, another
inhibitory receptor that bind to TIGIT (53), was also expressed by
TAMs, with highest expression levels in cluster 0 (Figure 6B and
Supplemental Figure 2B). We did not detect TIGIT expression in
TAMs by scRNA-seq (Figure 6B). Additionally, we noted high
expression and associated of ICOS-L with cluster 0 (Supplemental
Figure 2B), which interestingly was also associated with GBM
patientOSandDFS(Figures 2A, B). These data suggest that
immunosuppressive TAMs, such as MDSCs, express genes for PD1,
PD-L1 and TIGIT-ligands. Therefore, evaluated the protein
expression of these markers on CD45
+
immune cells in patient
derived GBM tissue and PBMCs, and healthy donor (HD) PBMCs.
The frequencies of CD11b
+
CD33
+
cells in GBM TILs were on
average higher compared to GBM PBMCs, and were signicantly
higher than of HD PBMCs (Figure 6C and Supplemental Figure
2C), suggesting that MDSCs are present at high levels in GBM and
could contribute to the TME immunosuppression (46). Consistent
with our scRNA-seq data, CD11b
+
CD33
+
cells had higher
expression levels of PD1, PD-L1, PVR, and CD226 in TILs, as
compared with CD11b
+
CD33
+
cells from PBMCs of GBM patients
and HDs, most notably for PD-L1 and CD226 expression (Figure
6D). Moreover, as compared to PBMCs samples we noted an
increased expression of PD1, PD-L1 and TIGIT on CD8
+
TILs,
while CD4
+
TILs had mostly upregulated expression of TIGIT
(Figure 6E). Together, these data suggest that CD11b
+
CD33
+
TAMs may promote immunosuppressive functions at-least in part
through expression of PD1/TIGIT-checkpoint ligands. To test this
hypothesis, hGP100-restricted naïve T cells isolated from pmel mice
(32) were activated in vitro with hGP100
2533
peptide and feeder cell
FIGURE 5 | Anti-PD1/TIGIT immunotherapy is associated with altered myeloid-derived suppressor cells (MDSCs) in GL261 murine model. GL261 glioma cells were
injected stereotactically in the caudate putamen of C56BL/6J mice followed by immunotherapy treatment starting on day 8 post tumor injection, and the frequencies
of MDSCs were determined on day 22. (A) Representative ow cytometry plots showing gating strategy of PMN MDCSs and Mo MDCSs based on the expression
of Gr1 and CD11b. (BD) Percentages (%) of CD45
+
glioma-inltrating total MDSCs (CD11b
+
Gr1
+
)(B), PMN MDSCs (CD11b
+
Gr1
high
)(C), and Mo MDSCs
(CD11b
+
Gr1
low
)(D), on day 22 following treatment. (E) Ratios of tumor inltrating CD8+ T cell to total MDSCs. Representative data of 3 experiments. n=5 per
group. One-way ANOVA with multiple comparisons test corrected for false discovery rate (FDR) using Benjamini-Hochberg test. Pvalues are as followed: *0.05,
**0.01, NS, not signicant.
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 6371469
FIGURE 6 | PD1, PD-L1 and TIGIT-ligands are expressed on myeloid suppressor cells in GBM and contribute to T cell dysfunction. Single cell (sc) RNA-seq analysis
was performed on myeloid cells from GBM patients. (A) UMAP clustering and expression (z-scores) of suppressive myeloid cell markers. (B) Expression z-scores of
PD1/TIGIT-associated checkpoint molecules in the scRNA-seq clusters. (CE) Healthy donor (HD) PBMCs and GBM patient PBMCs and TILs analyzed ow
cytometry for myeloid cells, T cells, and IC markers. n = 4 HD; n = 5 GBM patients. (C) Representative ow cytometry plots and percentages (%) of CD11b
+
CD33
+
myeloid cells. (D) Representative histograms and mean uorescence intensity (MFI) of PD1, PD-L1, PVR, and CD226 on CD11b
+
CD33
+
cells. (E) MFI of PD1, PD-
L1, PVR, and CD226 on CD8
+
T cells and CD4
+
T cells. (F) T cell proliferation assay of murine hGP100-reactive CD8
+
T cells cultured with immunosuppressive
myeloid cells with aTIGIT and aPD1. Representative histogram plots and percentages (%) of proliferated CD8
+
T cells at different culture conditions as indicated in
the table lagend. n=4 per group. One-way ANOVA with Tukey multiple comparisons correction. ns, not signicant. p = *<0.05, **<0.01, ***<0.001, ****<0.0001.
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 63714610
(antigen presenting cells; APCs) in the presence of bone-marrow
derived myeloid cells (putatively MDSC-like cells) cultured in
GL261 cell-derived tumor-conditioned media and treated with
aTIGIT and aPD1. Our data indicated that glioma-conditioned
immunosuppressive myeloid cells signicantly inhibited CD8
+
T
cell proliferation, which was restored by the addition of aTIGIT or
aPD1 (Figure 6F). In summary, these new data suggest that
immunosuppressive myeloid cells, and presumably MDSCs,
suppress anti-tumor immunity by inhibiting antigen-specicT
cell function in GBM, at least in part via TIGIT and PD1
pathways, which may have major implication to patient
treatments by immunotherapy.
DISCUSSION
In cancer, the dysregulationof immune checkpoints, such as TIGIT
and PD1, is directly associated with tumor progression and
enhanced immune evasion (5457). In the past decade, an
increasing number of IC-targeted immunotherapies have proven
to have substantially benecial outcomes for a wide variety of
malignancies and provide durable tumor immunity and long-
term patient survival (58). Nonetheless, evidence supporting the
efcacy of IC immunotherapy in glioma remains insufcient (11).
In this study we interrogated RNA-seq data of 153 GBM
patients in the TCGA database to identify IC genes whose
upregulated expression is associated with poor outcome. We
found that upregulated expression of TIGIT and PDCD1, as well
as their ligands CD155 (PVR) and PD-L1 (respectively), was
signicantly correlated with poor DFS and OS. Other checkpoint
pathways with inhibitors currently in development, including
LAG3 and TIM3, were not associated with either patient OS or
DFS. We posit that interrogation of TIGIT and PD1 -associated
regulatory gene networks in responding and non-responding GBM
patients would be of great interest to identify biomarker of ICIs.
PD1isanimmunecheckpointexpressedonactivated
immune cells, including CD4
+
and CD8
+
TILs. The binding of
PD1 to its ligand, PD-L1 on tumor and stromal cells, delivers a
signal that inhibits effector functions such as cytokine production
and cytolytic activity in the tumor microenvironment (TME)
(59). PD-L1, like many other IC ligands, is hijacked by tumor
cells in order to evade anti-tumor immunity. Accordingly,
blockade of PD1/PD-L1 pathway with antibodies have been
shown to improve T cell function and reduce tumor burden in
several types of tumors (60,61). Previous studies demonstrated
elevated levels of TIGIT expression in human gliomas (34);
however, the therapeutic effects of targeting this pathway in
glioma patients remain poorly understood. TIGIT has recently
emerged as an important checkpoint that is also expressed by
activated CD4
+
and CD8
+
TILs. TIGIT has a higher binding
afnity to CD155 than CD226; thus, once TIGIT is upregulated,
the inhibitory signal becomes more dominant (6264). Similarly,
interfering with TIGIT/CD155 interaction has been identied as
a potential therapeutic target for malignancies (65). Interestingly,
blocking PD1/PD-L1 signaling was shown to increase the
expression of TIGIT on Tregs in head and neck squamous cell
carcinoma (HNSCC) patients (49), suggesting a resistant
mechanism for aPD1 immunotherapy mediated by TIGIT.
Accordingly, our data support prior studies that combination
immunotherapy treatment targeting the PD1 and TIGIT
pathways leads to prolonged survival in GBM murine models
(34,66). Furthermore, we showed that aTIGIT/aPD1 dual
treatment increased the numbers of CD8
+
TILs and enhanced
their lytic function in GBM, supporting previous ndings that
this treatment can enhances IFN-gexpression in glioma-
inltrating T cells (34). Importantly, our data indicate that
combined immunotherapy with aTIGIT/aPD1 affects MDSCs
in the glioma TME.
MDSCs are a heterogenous population of immature myeloid
cells that contribute to tumor growth, accumulation of
additional immunosuppressive cells, and immunotherapy
resistance (66,67). Furthermore, MDSCs express large amounts
of immunosuppressive factors, multiple anti-inammatory
cytokines and chemokines that directly stimulate tumor
progression (68). Notably, a long-term survival study in
melanoma patients showed that elevated numbers of MDSCs
were highly associated with ICI resistance and negative
therapeutic outcomes (69). Additionally, elevated numbers of
tumor inltrating MDSCs are correlated with CD8
+
TIL
dysfunction and induced tumor cell expression of IC ligands;
thus, MDSCs may promote and sustain an immunosuppressive
glioma TME (7072). Here, we showed that TIGIT blockade
stimulated anti-tumor cytotoxic T cell (CTL) responses and
reduced the immunosuppressive MDSCs in a murine model of
GBM. Moreover, we found that PMN MDSC, but not Mo MDSC
accumulation was reduced by dual blockade of TIGIT and PD1,
compared with controls. Thus, our data suggest that PMN and Mo
MDSCs might have different mechanisms to confer resistance
against ICI immunotherapy, but may also be a target of ICI in
glioma.We positedthat future studies shouldfocus on unveiling the
crosstalk and mechanisms by which ICIs affect MDSCs in glioma.
Along these lines, we showed that suppressive myeloid cells express
PD1, PD-L1, and TIGIT-ligands in human GBM tissue. Moreover,
we demonstrated that antigen specic T cell proliferation is
inhibited by immunosuppressive myeloid cells can be restored by
TIGIT/PD1 blockade. This suggests that CTL exhaustion might be
regulatedat least in part bythe expression of IC ligands on MDSCs
in GBM.
Treg cells are major components of the immune suppressive TME
which express many ICs (73). The expression of TIGIT and PD1 by
Treg cells was shown to enhances their immunosuppressive
functions and contribute to tumor progression both in glioma
murine models and GBM patients (74). Importantly, Treg cells are
major source of IL-10 in GBM (74,75), and IL-10 can induce MDSC
development and enhance their suppressive functions (76,77), as well
as increasing the expression of PD1 myeloid cells (78). Additionally,
TIGIT is important for IL-10 expression by Treg cells (55). Therefore,
it is possible that aTIGIT might also regulate MDSC cell numbers
and functions by suppressing Treg expression of IL-10. Future studies
should focus on the mechanisms and crosstalk between Treg cells and
MDSCs via checkpoint molecules in the GBM TME and their
contribution to ICI resistant.
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 63714611
In summary, our data support the concept of treating GBM
patients with dual blockade of PD1 and TIGIT and provides new
insights into mechanisms of GBM immunotherapy to facilitates
the development of novel treatments.
DATA AVAILABILITY STATEMENT
The datasets presented in this study can be found in https://www.
ncbi.nlm.nih.gov/gap/ online repository, under accession
number phs000178.
ETHICS STATEMENT
The animal study was reviewed and approved by University of
Pittsburgh Institutional Animal Care and Use Committee.
AUTHOR CONTRIBUTIONS
Performed experiments and collected data: IR, RK, LM, KS, PS,
CS, JN, NA, MLC, AF and TB. Formal data analysis and gures:
IR and RK. Statistical analysis: IR, SZ, and DR. RNA-seq and
single cell RNA-seq analysis: IR, LW, and DR. Resources,
concepts and/or manuscript revisions: IR, BH, SA, AB, FSL, IP,
NA, BC, AD and GK. IR wrote the manuscript. IR revised the
manuscript with assistance from GK. IR and GK designed the
experiments. GK supervised and nanced the study. All authors
contributed to the article and approved the submitted version.
FUNDING
This research was supported by National Institute of Health
(NIH)/National Cancer Institute (NCI) grants R01CA244520
and NIH R01CA222804 (to GK), NIH/National Institute of
Biomedical Imaging and Bioengineering (NIBIB) R21EB029650
(to GK), The Walter L.Copeland Fund of The Pittsburgh
Foundation (to IR, GK) and The Brain Tumor Funders
Collaborative (GK). IR was supported by a fellowship from
UPMC Childrens Hospital of Pittsburgh. This work utilized
the Hillman Cancer Center Flow Cytometry Core, a shared
resource at the University of Pittsburgh supported by the
CCSG P30 CA047904.
ACKNOWLEDGMENTS
The results shown here are in whole or part based upon data
generated by the TCGA Research Network: https://www.cancer.
gov/tcga. The authors would like to thank Dr. Yijen Wu, director
of Rangos Research Center Animal Imaging Core for assistance
with MRI imaging and analysis.
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found online
at: https://www.frontiersin.org/articles/10.3389/mmu.2021.
637146/full#supplementary-material
REFERENCES
1. Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS Statistical Report:
Primary Brain and Central Nervous System Tumors Diagnosed in the United
States in 2005-2009. Neuro Oncol (2012) 14(Suppl 5):v149. doi: 10.1093/
neuonc/nos218
2. Lah TT, Novak M, Breznik B. Brain Malignancies: Glioblastoma and Brain
Metastases. Semin Cancer Biol (2020) 60:26273. doi: 10.1016/
j.semcancer.2019.10.010
3. Braunstein S, Raleigh D, Bindra R, Mueller S, Haas-Kogan D. Pediatric High-
Grade Glioma: Current Molecular Landscape and Therapeutic Approaches.
J Neurooncol (2017) 134(3):5419. doi: 10.1007/s11060-017-2393-0
4. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB,
et al. Radiotherapy Plus Concomitant and Adjuvant Temozolomide for
Glioblastoma. NEnglJMed(2005) 352(10):98796. doi: 10.1056/
NEJMoa043330
5. Buerki RA, Chheda ZS, Okada H. Immunotherapy of Primary Brain Tumors:
Facts and Hopes. Clin Cancer Res (2018) 24(21):5198205. doi: 10.1158/1078-
0432.CCR-17-2769
6. Yang Y. Cancer Immunotherapy: Harnessing the Immune System to Battle
Cancer. J Clin Invest (2015) 125(9):33357. doi: 10.1172/JCI83871
7. Valkenburg KC, de Groot AE, Pienta KJ. Targeting the Tumour Stroma to
Improve Cancer Therapy. Nat Rev Clin Oncol (2018) 15:36681. doi: 10.1038/
s41571-018-0007-1
8. Liu T, Han C, Wang S, Fang P, Ma Z, Xu L, et al. Cancer-Associated
Fibroblasts: An Emerging Target of Anti-Cancer Immunotherapy. J Hematol
Oncol (2019) 12(1):86. doi: 10.1186/s13045-019-0770-1
9. Iwai Y, Hamanishi J, Chamot o K, Honjo T. Cancer Immunotherapies
Targeting the PD-1 Signaling Pathway. J BioMed Sci (2017) 24(1):26. doi:
10.1186/s12929-017-0329-9
10. Tang J, Yu JX, Hubbard-Lucey VM, Neftelinov ST, Hodge JP, Lin Y. Trial
Watch: The Clinical Trial Landscape for PD1/PDL1 Immune Checkpoint
Inhibitors. Nat Rev Drug Discov (2018) 17(12):8545. doi: 10.1038/
nrd.2018.210
11. Yarchoan M, Hopkins A, Jaffee EM. Tumor Mutational Burden and Response
Rate to PD-1 Inhibition. N Engl J Med (2017) 377(25):25001. doi: 10.1056/
NEJMc1713444
12. Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick A, et al.
Effect of Nivolumab vs Bevacizumab in Patients With Recurrent
Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial.
JAMA Oncol (2020) 6(7):100310. doi: 10.1001/jamaoncol.2020.1024
13. Upadhaya S, Hubbard-Lucey VM, Yu JX. Immuno-Oncology Drug
Development Forges on Despite COVID-19. Nat Rev Drug Discov (2020)
19(11):7512. doi: 10.1038/d41573-020-00166-1
14. Chauvin JM, Zarour HM. TIGIT in Cancer Immunotherapy. J Immunother
Cancer (2020) 8(2):e000957. doi: 10.1136/jitc-2020-000957
15. Gorvel L, Olive D. Targeting the PVR-TIGIT AxisWith Immune
Checkpoint Therapies. F1000Res (2020) 9:35. doi: 10.12688/
f1000research.22877.1
16. Blake SJ, Dougall WC, Miles JJ, Teng MW, Smyth MJ. Molecular Pathways:
Targeting CD96 and TIGIT for Cancer Immunotherapy. Clin Cancer Res
(2016) 22(21):51838. doi: 10.1158/1078-0432.CCR-16-0933
17. Park J, Kwon M, Shin EC. Immune Checkpoint Inhibitors for Cancer
Treatment. Arch Pharm Res (2016) 39(11):157787. doi: 10.1007/s12272-
016-0850-5
18. Awad RM, De Vlaeminck Y, Maebe J, Goyvaerts C, Breckpot K. Turn Back the
TIMe: Targeting Tumor Inltrating Myeloid Cells to Revert Cancer
Progression. Front Immunol (2018) 9:1977. doi: 10.3389/mmu.2018.01977
19. Meyer C, Cagnon L, Costa-Nunes CM, Baumgaertner P, Montandon N,
Leyvraz L, et al. Frequencies of Circulating MDSC Correlate With Clinical
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 63714612
Outcome of Melanoma Patients Treated With Ipilimumab. Cancer Immunol
Immunother (2014) 63(3):24757. doi: 10.1007/s00262-013-1508-5
20. Fujita M, Kohanbash G, Fellows-Mayle W, Hamilton RL, Komohara Y,
Decker SA, et al. COX-2 Blockade Suppresses Gliomagenesis by Inhibiting
Myeloid-Derived Suppressor Cells. Cancer Res (2011) 71(7):266474. doi:
10.1158/0008-5472.CAN-10-3055
21. Kohanbash G, McKaveney K, Sakaki M, Ueda R, Mintz AH, Amankulor N,
et al. GM-CSF Promotes the Immunosuppressive Activity of Glioma-
Inltrating Myeloid Cells Through Interleukin-4 Receptor-Alpha. Cancer
Res (2013) 73(21):641323. doi: 10.1158/0008-5472.CAN-12-4124
22. Patro R, Duggal G, Love MI, Irizarry RA, Kingsford C. Salmon Provides Fast
and Bias-Aware Quantication of Transcript Expression. Nat Methods (2017)
14(4):4179. doi: 10.1038/nmeth.4197
23. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene
Ontology: Tool for the Unication of Biology. Gene Ontology Consortium.
Nat Genet (2000) 25(1):259. doi: 10.1038/75556
24. Jassal B, Matthews L, Viteri G, Gong C, Lorente P, Fabregat A, et al. The
Reactome Pathway Knowledgebase. Nucleic Acids Res (2020) 48(D1):D498
503. doi: 10.1093/nar/gkz1031
25. Mi H, Muruganujan A, Ebert D, Huang X, Thomas PD. PANTHER Version
14: More Genomes, a New PANTHER GO-Slim and Improvements in
Enrichment Analysis Tools. Nucleic Acids Res (2019) 47(D1):D419D26.
doi: 10.1093/nar/gky1038
26. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, et al.
ClueGO: A Cytoscape Plug-in to Decipher Functionally Grouped Gene
Ontology and Pathway Annotation Networks. Bioinformatics (2009) 25
(8):10913. doi: 10.1093/bioinformatics/btp101
27. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al.
Cytoscape: A Software Environment for Integrated Models of Biomolecular
Interaction Networks. Genome Res (2003) 13(11):2498504. doi: 10.1101/
gr.1239303
28. Wang L, Babikir H, Muller S, Yagnik G, Shamardani K, Catalan F, et al. The
Phenotypes of Proliferating Glioblastoma Cells Reside on a Single Axis of
Variation. Cancer Discov (2019) 9(12):170819. doi: 10.1158/2159-8290.CD-
19-0329
29. Muller S, Cho A, Liu SJ, Lim DA, Diaz A. CONICS Integrates scRNA-seq
With DNA Sequencing to Map Gene Expression to Tumor Sub-Clones.
Bioinformatics (2018) 34(18):32179. doi: 10.1093/bioinformatics/bty316
30. Wang L, Catalan F, Shamardani K, Babikir H, Diaz A. Ensemble Learning for
Classifying Single-Cell Data and Projection Across Reference Atlases.
Bioinformatics (2020) 36(11):35857. doi: 10.1093/bioinformatics/btaa137
31. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM,3, et al.
Comprehensive Integration of Single-Cell Data. Cell (2019) 177(7):1888902
e21. doi: 10.1016/j.cell.2019.05.031
32. Overwijk WW, Theoret MR, Finkelstein SE, Surman DR, de Jong LA, Vyth-
Dreese FA, et al. Tumor Regression and Autoimmunity After Reversal of a
Functionally Tolerant State of Self-Reactive CD8+ T Cells. J Exp Med (2003)
198(4):56980. doi: 10.1084/jem.20030590
33. Nigam S, McCarl L, Kumar R, Edinger RS, Kurland BF, Anderson CJ, et al.
Preclinical ImmunoPET Imaging of Glioblastoma-Inltrating Myeloid Cells
Using Zirconium-89 Labeled Anti-CD11b Antibody. Mol Imaging Biol (2019)
22:68594. doi: 10.1101/614511
34. Hung AL, Maxwell R, Theodros D, Belcaid Z, Mathios D, Luksik AS, et al.
TIGIT and PD-1 Dual Checkpoint Blockade Enhances Antitumor Immunity
and Survival in GBM. Oncoimmunology (2018) 7(8):e1466769. doi: 10.1080/
2162402X.2018.1466769
35. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of Myeloid-Derived
Suppressor Cells in Tumor-Bearing Mice. J Immunol (2008) 181(8):5791802.
doi: 10.4049/jimmunol.181.8.5791
36. Bruger AM, Vanhaver C, Bruderek K, Amodio G, Tavukcuoglu E,
Esendagli G, et al. Protocol to Assess the Suppression of T-Cell
Proliferation by Human MDSC. Methods Enzymol (2020) 632:15592. doi:
10.1016/bs.mie.2019.05.046
37. Rezazadeh H, Astaneh M, Tehrani M, Hossein-Nataj H, Zaboli E, Shekarriz R,
et al. Blockade of PD-1 and TIM-3 Immune Checkpoints Fails to Restore the
Function of Exhausted CD8(+) T Cells in Early Clinical Stages of Chronic
Lymphocytic Leukemia. Immunol Res (2020) 68(5):26979. doi: 10.1007/
s12026-020-09146-4
38. Cancer Genome Atlas Research N. Comprehensive Genomic Characterization
Denes Human Glioblastoma Genes and Core Pathways. Nature (2008) 455
(7216):10618. doi: 10.1038/nature07385
39. Zarour HM. Reversing T-cell Dysfunction and Exhaustion in Cancer. Clin
Cancer Res (2016) 22(8):185664. doi: 10.1158/1078-0432.CCR-15-1849
40. ChristoT, Baritaki S, Falzone L, Libra M, Zaravinos A. Current Perspectives
in Cancer Immunotherapy. Cancers (Basel) (2019) 11(10):1472. doi: 10.3390/
cancers11101472
41. Fu W, Wang W, Li H, Jiao Y, Huo R, Yan Z, et al. Single-Cell Atlas Reveals
Complexity of the Immunosuppressive Microenvironment of Initial and
Recurrent Glioblastoma. Front Immunol (2020) 11:835. doi: 10.3389/
mmu.2020.00835
42. Chase Huizar C, Raphael I, Forsthuber TG. Genomic, Proteomic, and Systems
Biology Approaches in Biomarker Discovery for Multiple Sclerosis. Cell
Immunol (2020) 358:104219. doi: 10.1016/j.cellimm.2020.104219
43. Chauvin JM, Pagliano O, Fourcade J, Sun Z, Wang H, Sander C, et al. TIGIT
and PD-1 Impair Tumor Antigen-Specic CD8(+) T Cells in Melanoma
Patients. J Clin Invest (2015) 125(5):204658. doi: 10.1172/JCI80445
44. Weber R, Fleming V, Hu X, Nagibin V, Groth C, Altevogt P, et al. Myeloid-
Derived Suppressor Cells Hinder the Anti-Cancer Activity of Immune
Checkpoint Inhibitors. Front Immunol (2018) 9:1310. doi: 10.3389/
mmu.2018.01310
45. Mi Y, Guo N, Luan J, Cheng J, Hu Z, Jiang P, et al. The Emerging Role of
Myeloid-Derived Suppressor Cells in the Glioma Immune Suppressive
Microenvironment. Front Immunol (2020) 11:737. doi: 10.3389/
mmu.2020.00737
46. Kohanbash G, Okada H. Myeloid-Derived Suppressor Cells (MDSCs) in
Gliomas and Glioma-Development. Immunol Invest (2012) 41(6-7):65879.
doi: 10.3109/08820139.2012.689591
47. Raychaudhuri B, Rayman P, Ireland J, Ko J, Rini B, Borden EC, et al. Myeloid-
Derived Suppressor Cell Accumulation and Function in Patients With Newly
Diagnosed Glioblastoma. Neuro Oncol (2011) 13(6):5919. doi: 10.1093/
neuonc/nor042
48. Ballbach M, Dannert A, Singh A, Siegmund DM, Handgretinger R, Piali L,
et al. Expression of Checkpoint Molecules on Myeloid-Derived Suppressor
Cells. Immunol Lett (2017) 192:16. doi: 10.1016/j.imlet.2017.10.001
49. Wu L, Mao L, Liu JF, Chen L, Yu GT, Yang LL, et al. Blockade of TIGIT/
CD155 Signaling Reverses T-Cell Exhaustion and Enhances Antitumor
Capability in Head and Neck Squamous Cell Carcinoma. Cancer Immunol
Res (2019) 7(10):170013. doi: 10.1158/2326-6066.CIR-18-0725
50. Muller S, Kohanbash G, Liu SJ, Alvarado B, Carrera D, Bhaduri A, et al.
Single-Cell Proling of Human Gliomas Reveals Macrophage Ontogeny as a
Basis for Regional Differences in Macrophage Activation in the Tumor
Microenvironment. Genome Biol (2017) 18(1):234. doi: 10.1186/s13059-
017-1362-4
51. Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-Induced Myeloid
Deviation: When Myeloid-Derived Suppressor Cells Meet Tumor-Associated
Macrophages. J Clin Invest (2015) 125(9):336576. doi: 10.1172/JCI80006
52. Sieow JL, Gun SY, Wong SC. The Sweet Surrender: How Myeloid Cell
Metabolic Plasticity Shapes the Tumor Microenvironment. Front Cell Dev
Biol (2018) 6:168. doi: 10.3389/fcell.2018.00168
53. Stanietsky N, Simic H, Arapovic J, Toporik A, Levy O, Novik A, et al. The
Interaction of TIGIT With PVR and PVRL2 Inhibits Human NK Cell
Cytotoxicity. Proc Natl Acad Sci USA (2009) 106(42):1785863. doi:
10.1073/pnas.0903474106
54. Johnston RJ, Comps-Agrar L, Hackney J, Yu X, Huseni M, Yang Y, et al. The
Immunoreceptor TIGIT Regulates Antitumor and Antiviral CD8(+) T Cell
Effector Function. Cancer Cell (2014) 26(6):92337. doi: 10.1016/
j.ccell.2014.10.018
55. Kurtulus S, Sakuishi K, Ngiow SF, Joller N, Tan DJ, Teng MW, et al. TIGIT
Predominantly Regulates the Immune Response Via Regulatory T Cells. J Clin
Invest (2015) 125(11):405362. doi: 10.1172/JCI81187
56. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF,
et al. Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in
Cancer. N Engl J Med (2012) 366(26):244354. doi: 10.1056/NEJMoa1200690
57. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al.
Mismatch Repair Deciency Predicts Response of Solid Tumors to PD-1
Blockade. Science (2017) 357(6349):40913. doi: 10.1126/science.aan6733
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 63714613
58. Darvin P, Toor SM, Nair VS, Elkord E. Immune Checkpoint Inhibitors:
Recent Progress and Potential Biomarkers. Exp Mol Med (2018) 50:11. doi:
10.1038/s12276-018-0191-1
59. Lim TS, Chew V, Sieow JL, Goh S,Yeong JPS, Soon AL, et al. PD-1 Expression
on Dendritic Cells Suppresses CD8(+) T Cell Function and Antitumor
Immunity. Oncoimmunology (2016) 5(3):8. doi: 10.1080/2162402X.2015.
1085146
60. Dong YN, Sun Q, Zhang XW. PD-1 and its Ligands are Important Immune
Checkpoints in Cancer. Oncotarget (2017) 8(2):217186. doi: 10.18632/
oncotarget.13895
61. Pardoll DM. The Blockade of Immune Checkpoints in Cancer
Immunotherapy. Nat Rev Cancer (2012) 12(4):25264. doi: 10.1038/nrc3239
62. Yu X, Harden K, Gonzalez LC, Francesco M, Chiang E, Irving B, et al. The
Surface Protein TIGIT Suppresses T Cell Activation by Promoting the
Generation of Mature Immunoregulatory Dendritic Cells. Nat Immunol
(2009) 10(1):4857. doi: 10.1038/ni.1674
63. Pauken KE, Wherry EJ. TIGIT and CD226: Tipping the Balance Between
Costimulatory and Coinhibitory Molecules to Augment the Cancer
Immunotherapy Toolkit. Cancer Cell (2014) 26(6):7857. doi: 10.1016/
j.ccell.2014.11.016
64. Lozano E, Dominguez-Villar M, Kuchroo V, Haer DA. The TIGIT/CD226
Axis Regulates Human T Cell Function. J Immunol (2012) 188(8):386975.
doi: 10.4049/jimmunol.1103627
65. Blake SJ, Stannard K, Liu J, Allen S, Yong MCR, Mittal D, et al. Suppression of
Metastases Using a New Lymphocyte Checkpoint Target for Cancer
Immunotherapy. Cancer Discov (2016) 6(4):44659. doi: 10.1158/2159-
8290.CD-15-0944
66. Kohanbash G, Okada H. Myeloid-Derived Suppressor Cells (MDSCs) in
Gliomas and Glioma-Development. Immunol Invest (2012) 41(6-7):65879.
doi: 10.3109/08820139.2012.689591
67. Kamran N, Kadiyala P, Saxena M, CandolM, Li YP, Moreno-Ayala MA,
et al. Immunosuppressive Myeloid CellsBlockade in the Glioma
Microenvironment Enhances the Efcacy of Immune-Stimulatory Gene
Therapy. Mol Ther (2017) 25(1):23248. doi: 10.1016/j.ymthe.2016.10.003
68. Veglia F, PeregoM, Gabrilovich D. Myeloid-Derived Suppressor Cells Coming of
Age. Nat Immunol (2018) 19(2):10819. doi: 10.1038/s41590-017-0022-x
69. Fujimura T, Kambayashi Y, Aiba S. Crosstalk Between Regulatory T Cells
(Tregs) and Myeloid Derived Suppressor Cells (MDSCs) During Melanoma
Growth. Oncoimmunology (2012) 1(8):14334. doi: 10.4161/onci.21176
70. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of Myeloid-Derived
Suppressor Cells in Tumor-Bearing Mice. J Immunol (2008) 181(8):5791802.
doi: 10.4049/jimmunol.181.8.5791
71. de Coana YP, Poschke I, Gentilcore G, Mao YM, Nystrom M, Hansson J, et al.
Ipilimumab Treatment Results in an Early Decrease in the Frequency of
Circulating Granulocytic Myeloid-Derived Suppressor Cells as Well as Their
Arginase1 Production. Cancer Immunol Res (2013) 1(3):15862. doi: 10.1158/
2326-6066.CIR-13-0016
72. Pitt JM, Vetizou M, Daillere R, Roberti MP, Yamazaki T, Routy B, et al.
Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-
Intrinsic and -Extrinsic Factors. Immunity (2016) 44(6):125569. doi:
10.1016/j.immuni.2016.06.001
73. Nishikawa H, Sakaguchi S. Regulatory T Cells in Tumor Immunity. Int J
Cancer (2010) 127(4):75967. doi: 10.1002/ijc.25429
74. Ooi YC, Tran P, Ung N, Thill K, Trang A, Fong BM, et al. The Role of
Regulatory T-Cells in Glioma Immunology. Clin Neurol Neurosurg (2014)
119:12532. doi: 10.1016/j.clineuro.2013.12.004
75. Raphael I, Nalawade S, Eagar TN, Forsthuber TG. T Cell Subsets and Their
Signature Cytokines in Autoimmune and Inammatory Diseases. Cytokine
(2015) 74(1):517. doi: 10.1016/j.cyto.2014.09.011
76. Bah I, Kumbhare A, Nguyen L, McCall CE, El Gazzar M. IL-10 Induces an
Immune Repressor Pathway in Sepsis by Promoting S100A9 Nuclear
Localization and MDSC Development. Cell Immunol (2018) 332:328. doi:
10.1016/j.cellimm.2018.07.003
77. Yaseen MM, Abuharfeil NM, Darmani H, Daoud A. Mechanisms of Immune
Suppression by Myeloid-Derived Suppressor Cells: The Role of Interleukin-10
as a Key Immunoregulatory Cytokine. Open Biol (2020) 10(9):200111. doi:
10.1098/rsob.200111
78. Lamichhane P, Karyampudi L, Shreeder B, Krempski J, Bahr D, Daum J, et al.
IL10 Release Upon PD-1 Blockade Sustains Immunosuppression in Ovarian
Cancer. Cancer Res (2017) 77(23):666778. doi: 10.1158/0008-5472.CAN-17-
0740
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Copyright © 2021 Raphael, Kumar, McCarl, Shoger, Wang, Sandlesh, Sneiderman,
Allen, Zhai, Campagna, Foster, Bruno, Agnihotri, Hu, Castro, Lieberman, Broniscer,
Diaz, Amankulor, Rajasundaram, Pollack and Kohanbash. This is an open-access
article distributed under the terms of the Creative Commons Attribution License
(CC BY). The use,distribution or reproduction in other forums is permitted, provided
the original author(s) and the copyright owner(s) are credited and that the original
publicationin this journalis cited, in accordance with accepted academic practice. No
use, distribution or reproduction is permitted which does not comply with these terms.
Raphael et al. aTIGIT/aPD1 Immunotherapy in GBM
Frontiers in Immunology | www.frontiersin.org May 2021 | Volume 12 | Article 63714614
... However, when disease states occur, there is a hindrance of normal differentiation contributing to the presence of MDSCs (61-63). MDSCs are a heterogenous population of immature myeloid cells associated with many cancers which can inhibit T-cell proliferation (64). There are ample studies that show these cells are present in adult gliomas and several studies have also shown their presence in some pHGG types, with limited knowledge in pGBM (65)(66)(67)(68)(69)(70). ...
... In mice, MDSCs are often characterized by the expression of the CD11b and Gr1 (ly6C/Ly6G) receptors depending on the phenotype of the MDSC. Ly6C and Ly6G is also often used to distinguish between myeloid type (M) and polymorphonuclear or granulocytic (PMN/G)-MDSC, respectively (64). In patients, MDSCs that are in the monocytic phenotype commonly are described by their expression as CD11b + CD14 + HLA-DR −/lo CD15 − , while polymorphonuclear (PMN) MDSCs are associated with CD11b + CD14 − CD15 + or CD11b + CD14 − CD66b + (63). ...
Article
Full-text available
Pediatric high-grade glioma (pHGG) including pediatric glioblastoma (pGBM) are highly aggressive pediatric central nervous system (CNS) malignancies. pGBM comprises approximately 3% of all pediatric CNS malignancies and has a 5-year survival rate of approximately 20%. Surgical resection and chemoradiation are often the standard of care for pGBM and pHGG, however, even with these interventions, survival for children diagnosed with pGBM and pHGG remains poor. Due to shortcomings associated with the standard of care, many efforts have been made to create novel immunotherapeutic approaches targeted to these malignancies. These efforts include the use of vaccines, cell-based therapies, and immune-checkpoint inhibitors. However, it is believed that in many pediatric glioma patients an immunosuppressive tumor microenvironment (TME) possess barriers that limit the efficacy of immune-based therapies. One of these barriers includes the presence of immunosuppressive myeloid cells. In this review we will discuss the various types of myeloid cells present in the glioma TME, including macrophages and microglia, myeloid-derived suppressor cells, and dendritic cells, as well as the specific mechanisms these cells can employ to enable immunosuppression. Finally, we will highlight therapeutic strategies targeted to these cells that are aimed at impeding myeloid-cell derived immunosuppression.
... Spleen and tumor tissues were collected from GL261-bearing mice, and enzymatically processed into singlecell suspensions as previously described 9,13 . For staining of GL261 cells, non-specific Fc binding was blocked using TruStain FcX™ (human or anti-mouse CD16/32, clone 93), for ex vivo spleen and tumor cells, staining was performed using anti-CD3 (clone 17A2), anti-CD11B (M1/70) and anti-TIGIT (clone 1G9); for in vitro GL261 staining, IgG (anti-mouse IgG1, clone MOPC-21), anti-TIGIT (anti-mouse TIGIT, clone 1G9), and anti-PD-L1 (anti-mouse CD274, clone 10F.9G2) were used. ...
... We previously examined TIGIT expression in 28 primary human gliomas by scRNAseq and found that CD11b + TAMs, including myeloid-derived suppressor cells, expressed genes for PD1, PD-L1, and TIGIT ligands 9 . The overall findings in that study supported the idea that TIGIT and PD-1 pathways contribute to immunosuppression and tumor aggression in glioma 13 . ...
Article
Full-text available
Glioblastoma (GBM) is the most common primary malignant brain tumor. Currently, there are few effective treatment options for GBM beyond surgery and chemo-radiation, and even with these interventions, median patient survival remains poor. While immune checkpoint inhibitors (ICIs) have demonstrated therapeutic efficacy against non-central nervous system cancers, ICI trials for GBM have typically had poor outcomes. TIGIT is an immune checkpoint receptor that is expressed on activated T-cells and has a role in the suppression of T-cell and Natural Killer (NK) cell function. As TIGIT expression is reported as both prognostic and a biomarker for anti-TIGIT therapy, we constructed a molecular imaging agent, [ 89 Zr]Zr-DFO-anti-TIGIT (89 Zr-αTIGIT), to visualize TIGIT in preclinical GBM by immunoPET imaging. PET imaging and biodistribution analysis of 89 Zr-αTIGIT demonstrated uptake in the tumor microenvironment of GBM-bearing mice. Blocking antibody and irrelevant antibody tracer studies demonstrated specificity of 89 Zr-αTIGIT with significance at a late time point post-tracer injection. However, the magnitude of 89 Zr-αTIGIT uptake in tumor, relative to the IgG tracer was minimal. These findings highlight the features and limitations of using 89 Zr-αTIGIT to visualize TIGIT in the GBM microenvironment.
... PD-1/TIGIT-axis emerged as putative immunotherapy targets in GBM in the analysis of TCGA datasets from 153 GBM patients. GBM patients with upregulated PD-1 and TIGIT genes and their cognate ligand genes displayed reduced OS and PFS compared to patients who did not upregulate the same genes [78]. In a GBM mouse model, treatment with anti-TIGIT/anti-PD-1 resulted in increased survival of the mice, reduced tumor volume, and increased immune infiltration in tumor lesions compared to single ICBs or isotype treatments [78]. ...
... GBM patients with upregulated PD-1 and TIGIT genes and their cognate ligand genes displayed reduced OS and PFS compared to patients who did not upregulate the same genes [78]. In a GBM mouse model, treatment with anti-TIGIT/anti-PD-1 resulted in increased survival of the mice, reduced tumor volume, and increased immune infiltration in tumor lesions compared to single ICBs or isotype treatments [78]. A phase I clinical trial has been set up to study an anti-TIGIT antibody (AB154) in combination with anti-PD-1 therapy in rGBM (NCT04656535). ...
Article
Full-text available
Simple Summary Glioblastoma is a lethal primary brain tumor, and so far, immunotherapeutic strategies have not significantly improved patients’ prognosis, both in newly diagnosed and recurrent settings. Understanding the features of the immune environment of the central nervous system is crucial to designing new treatment strategies able to counteract site-specific immunosuppressive and pro-tumorigenic factors. Here, we review next-generation immunomodulating therapeutic strategies such as immune check-point blockade, vaccinations, and adoptive cell therapies, aiming to re-shape the tumor microenvironment and restore active anti-tumor immunity. The door for immunotherapeutic strategies in glioblastoma treatment is not completely closed; researchers should test combinatorial treatments and design trials with solid translational analyses to gain more close and deep insight into on-treatment modifications of the tumor microenvironment. Abstract Glioblastoma (GBM) is the most aggressive and lethal primary brain tumor, bearing a survival estimate below 10% at five years, despite standard chemoradiation treatment. At recurrence, systemic treatment options are limited and the standard of care is not well defined, with inclusion in clinical trials being highly encouraged. So far, the use of immunotherapeutic strategies in GBM has not proved to significantly improve patients’ prognosis in the treatment of newly diagnosed GBM, nor in the recurrent setting. Probably this has to do with the unique immune environment of the central nervous system, which harbors several immunosuppressive/pro-tumorigenic factors, both soluble (e.g., TGF-β, IL-10, STAT3, prostaglandin E2, and VEGF) and cellular (e.g., Tregs, M2 phenotype TAMs, and MDSC). Here we review the immune composition of the GBMs microenvironment, specifically focusing on the phenotype and function of the T cell compartment. Moreover, we give hints on the therapeutic strategies, such as immune checkpoint blockade, vaccinations, and adoptive cell therapy, that, interacting with tumor-infiltrating lymphocytes, might both target in different ways the tumor microenvironment and potentiate the activity of standard therapies. The path to be followed in advancing clinical research on immunotherapy for GBM treatment relies on a twofold strategy: testing combinatorial treatments, aiming to restore active immune anti-tumor responses, tackling immunosuppression, and additionally, designing more phase 0 and window opportunity trials with solid translational analyses to gain deeper insight into the on-treatment shaping of the GBM microenvironment.
... Another study analysed the TCGA transcriptome database and identified PD1 and TIGIT as preferred targets for GBM immunotherapy. Furthermore, they also reported that the dual blockade of PD1 and TIGIT improved survival and increased CD8+ TIL accumulation and function compared with either drug alone in a murine GBM model (116). Therefore, TIGIT represents a promising target for immunotherapy in patients with GBM, and ongoing clinical trials are evaluating anti-TIGIT combined with anti-PD-1 antibody treatment for recurrent GBM (NCT04656535). ...
Article
Full-text available
High-grade gliomas (HGG) pose significant challenges in modern tumour therapy due to the distinct biological properties and limitations of the blood-brain barrier. This review discusses recent advancements in HGG treatment, particularly in the context of immunotherapy and cellular therapy. Initially, treatment strategies focus on targeting tumour cells guided by the molecular characteristics of various gliomas, encompassing chemotherapy, radiotherapy and targeted therapy for enhanced precision. Additionally, technological enhancements are augmenting traditional treatment modalities. Furthermore, immunotherapy, emphasising comprehensive tumour management, has gained widespread attention. Immune checkpoint inhibitors, vaccines and CAR-T cells exhibit promising efficacy against recurrent HGG. Moreover, emerging therapies such as tumour treating fields (TTFields) offer additional treatment avenues for patients with HGG. The combination of diverse treatments holds promise for improving the prognosis of HGG, particularly in cases of recurrence.
... Yet, TIGIT monotherapy has so far resulted in divergent NK cell responses. Because co-expression of TIGIT correlates to other inhibitory receptors on immune cells 11,12 , its apparent role as a co-receptor has most often benefited from dual blockade approaches. On NK cells, this has translated into the requirement of co-activation via IL-15 stimulation 13,14 or agonism of 4-1BB. ...
Article
Full-text available
Severe heterogeneity within glioblastoma has spurred the notion that disrupting the interplay between multiple elements on immunosuppression is at the core of meaningful anti-tumor responses. T cell immunoreceptor with Ig and ITIM domains (TIGIT) and its glioblastoma-associated antigen, CD155, form a highly immunosuppressive axis in glioblastoma and other solid tumors, yet targeting of TIGIT, a functionally heterogeneous receptor on tumor-infiltrating immune cells, has largely been ineffective as monotherapy, suggesting that disruption of its inhibitory network might be necessary for measurable responses. It is within this context that we show that the usurpation of the TIGIT − CD155 axis via engineered synNotch-mediated activation of induced pluripotent stem cell-derived natural killer (NK) cells promotes transcription factor-mediated activation of a downstream signaling cascade that results in the controlled, localized blockade of CD73 to disrupt purinergic activity otherwise resulting in the production and accumulation of immunosuppressive extracellular adenosine. Such “decoy” receptor engages CD155 binding to TIGIT, but tilts inhibitory TIGIT/CD155 interactions toward activation via downstream synNotch signaling. Usurping activities of TIGIT and CD73 promotes the function of adoptively transferred NK cells into intracranial patient-derived models of glioblastoma and enhances their natural cytolytic functions against this tumor to result in complete tumor eradication. In addition, targeting both receptors, in turn, reprograms the glioblastoma microenvironment via the recruitment of T cells and the downregulation of M2 macrophages. This study demonstrates that TIGIT/CD155 and CD73 are targetable receptor partners in glioblastoma. Our data show that synNotch-engineered pluripotent stem cell-derived NK cells are not only effective mediators of anti-glioblastoma responses within the setting of CD73 and TIGIT/CD155 co-targeting, but represent a powerful allogeneic treatment option for this tumor.
... T-cell immune receptor with immunoglobulin and ITIM structural domains (TIGIT) is a promising new target for cancer immunotherapy [16]. Preclinical studies have shown that TIGIT blockade can prevent the progression of various solid and hematologic cancers [17][18][19][20][21][22][23][24]. TIGIT interacts with the CD155 expressed on antigen-presenting or tumor cells to impair T-cell and natural killer (NK) cell function [16,17,[25][26][27][28][29][30]. ...
Preprint
Full-text available
Heat shock protein 47 (HSP47) plays an essential role in correcting protein folding, and abnormal protein folding is closely related to tumorigenesis. However, the relationship between HSP47 and cancer immune response is poorly studied. Herein, HSP47 was found to be frequently overexpressed in human osteosarcomas. In animal models, HSP47 inhibition resulted in enhanced immune cell infiltration and function. Transcriptome data revealed that HSP47 negatively regulated CD155, a ligand of TIGIT. Immune checkpoint blockade therapy targeting the novel immune checkpoint molecule TIGIT is effective in limited patients. Further investigations are urgently needed to harness a robust response of this treatment. TIGIT antibody and HSP47-targeted therapy significantly inhibited the progression of osteosarcoma in mice and consequently prolonged survival. Mechanistically, inhibition of HSP47 attenuated TRAF2 protein ubiquitination and subsequently facilitated NF-κB-mediated CD155 transcription in HSP47-overexpressed osteosarcomas. Similarly, CD155 expression was significantly weakened in TRAF2-inhibited osteosarcoma cells. Collectively, our data revealed that targeting HSP47 could reinforce the expression of CD155 and therefore enhance the efficacy of anti-TIGIT treatment, providing a promising strategy for cancer immunotherapy.
... Surprisingly, an exploratory phase I clinical trial combining nivolumab and ipilimumab, 42 a CTLA-4 inhibitor, showed a lack of benefit over bevacizumab in contrast to preclinical experiments that showed an anti-PD-1, anti-CTLA-4 combination therapy capable of curing 75% of GBM immunocompetent mice, 43 somewhat emphasizing the gap between murine models and clinical translation. In addition, we know that there are multiple other immune checkpoint signals [44][45][46][47][48] and epigenetic signals 49,50 that maintain GBM immunoevasion. 3 A third explanation is that the timing of immune checkpoint inhibitor administration may matter. In fact, a small phase 2 trial comparing adjuvant versus neoadjuvant pembrolizumab in subjects with rGBM showed significantly improved survival in the latter group. ...
Article
Glioblastoma (GBM)’s median overall survival is almost 21 months. Six phase 3 immunotherapy clinical trials have recently been published, yet 5/6 did not meet approval by regulatory bodies. For the sixth, approval is uncertain. Trial failures result from multiple factors, ranging from intrinsic tumor biology to clinical trial design. Understanding the clinical and basic science of these 6 trials is compelled by other immunotherapies reaching the point of advanced phase 3 clinical trial testing. We need to understand more of the science in human GBMs in early trials: the “window of opportunity” design may not be best to understand complex changes brought about by immunotherapeutic perturbations of the GBM microenvironment. The convergence of increased safety of image-guided biopsies with “multi-omics” of small cell numbers now permits longitudinal sampling of tumor and biofluids to dissect the complex temporal changes in the GBM microenvironment as a function of the immunotherapy.
Article
Full-text available
Glioblastoma (GBM), the predominant and primary malignant intracranial tumor, poses a formidable challenge due to its immunosuppressive microenvironment, thereby confounding conventional therapeutic interventions. Despite the established treatment regimen comprising surgical intervention, radiotherapy, temozolomide administration, and the exploration of emerging modalities such as immunotherapy and integration of medicine and engineering technology therapy, the efficacy of these approaches remains constrained, resulting in suboptimal prognostic outcomes. In recent years, intensive scrutiny of the inhibitory and immunosuppressive milieu within GBM has underscored the significance of cellular constituents of the GBM microenvironment and their interactions with malignant cells and neurons. Novel immune and targeted therapy strategies have emerged, offering promising avenues for advancing GBM treatment. One pivotal mechanism orchestrating immunosuppression in GBM involves the aggregation of myeloid-derived suppressor cells (MDSCs), glioma-associated macrophage/microglia (GAM), and regulatory T cells (Tregs). Among these, MDSCs, though constituting a minority (4–8%) of CD45⁺ cells in GBM, play a central component in fostering immune evasion and propelling tumor progression, angiogenesis, invasion, and metastasis. MDSCs deploy intricate immunosuppressive mechanisms that adapt to the dynamic tumor microenvironment (TME). Understanding the interplay between GBM and MDSCs provides a compelling basis for therapeutic interventions. This review seeks to elucidate the immune regulatory mechanisms inherent in the GBM microenvironment, explore existing therapeutic targets, and consolidate recent insights into MDSC induction and their contribution to GBM immunosuppression. Additionally, the review comprehensively surveys ongoing clinical trials and potential treatment strategies, envisioning a future where targeting MDSCs could reshape the immune landscape of GBM. Through the synergistic integration of immunotherapy with other therapeutic modalities, this approach can establish a multidisciplinary, multi-target paradigm, ultimately improving the prognosis and quality of life in patients with GBM.
Article
The immune checkpoint TIGIT/PVR blockade exhibits significant antitumor effects through activation of NK and CD8+ T cell-mediated cytotoxicity. Immune checkpoint blockade (ICB) could induce tumor ferroptosis through IFN-γ released by immune cells, indicating the synergetic effects of ICB with ferroptosis in inhibiting tumor growth. However, the development of TIGIT/PVR inhibitors with ferroptosis-inducing effects has not been explored yet. In this study, the small molecule Hemin that could bind with TIGIT to block TIGIT/PVR interaction was screened by virtual molecular docking and cell-based blocking assay. Hemin could effectively restore the IL-2 secretion from Jurkat-hTIGIT cells. Hemin reinvigorated the function of CD8+ T cells to secrete IFN-γ and the elevated IFN-γ could synergize with Hemin to induce ferroptosis in tumor cells. Hemin inhibited tumor growth by boosting CD8+ T cell immune response and inducing ferroptosis in CT26 tumor model. More importantly, Hemin in combination with PD-1/PD-L1 blockade exhibited more effective antitumor efficacy in anti-PD-1 resistant B16 tumor model. In summary, our finding indicated that Hemin blocked TIGIT/PVR interaction and induced tumor cell ferroptosis, which provided a new therapeutic strategy to combine immunotherapy and ferroptosis for cancer treatment.
Article
Full-text available
Tumors evade immune-mediated recognition through multiple mechanisms of immune escape. On chronic tumor antigen exposure, T cells become dysfunctional/exhausted and upregulate various checkpoint inhibitory receptors (IRs) that limit T cells' survival and function. During the last decade, immunotherapies targeting IRs such as programmed cell death receptor 1 (PD-1) and anticytotoxic T lymphocyte-associated antigen 4 (CTLA-4) have provided ample evidence of clinical benefits in many solid tumors. Beyond CTLA-4 and PD-1, multiple other IRs are also targeted with immune checkpoint blockade in the clinic. Specifically, T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is a promising new target for cancer immunotherapy. TIGIT is upregulated by immune cells, including activated T cells, natural killer cells, and regulatory T cells. TIGIT binds to two ligands, CD155 (PVR) and CD112 (PVRL2, nectin-2), that are expressed by tumor cells and antigen-presenting cells in the tumor microenvironment. There is now ample evidence that the TIGIT pathway regulates T cell-mediated and natural killer cell-mediated tumor recognition in vivo and in vitro. Dual PD-1/TIGIT blockade potently increases tumor antigen-specific CD8+ T cell expansion and function in vitro and promotes tumor rejection in mouse tumor models. These findings support development of ongoing clinical trials with dual PD-1/TIGIT blockade in patients with cancer.
Article
Full-text available
Chronic immune activation and inflammation are unwanted consequences of many pathological conditions, since they could lead to tissue damage and immune exhaustion, both of which can worsen the pathological condition status. In fact, the immune system is naturally equipped with immunoregulatory cells that can limit immune activation and inflammation. However, chronic activation of downregulatory immune responses is also associated with unwanted consequences that, in turn, could lead to disease progression as seen in the case of cancer and chronic infections. Myeloid-derived suppressor cells (MDSC) are now considered to play a pivotal role in the pathogenesis of different inflammatory pathological conditions, including different types of cancer and chronic infections. As a potent immunosuppressor cell population, MDSC can inhibit specific and non-specific immune responses via different mechanisms that, in turn, lead to disease persistence. One such mechanism by which MDSC can activate their immunosuppressive effects is accomplished by secreting copious amounts of immunosuppressant molecules such as interleukin-10 (IL-10). In this article, we will focus on the pathological role of MDSC expansion in chronic inflammatory conditions, particularly in cancer, as well as some of the mechanisms by which MDSCs/IL-10 contribute to the disease progression in such conditions.
Article
Full-text available
Blocking antibodies targeting immune checkpoint molecules achieved invaluable success in tumor therapy and amazing clinical responses in a variety of cancers. Although common treatment protocols have improved overall survival in patients with chronic lymphocytic leukemia (CLL), they continue to relapse and progress. In the present in vitro study, the application of anti-PD-1 and anti-TIM-3 blocking antibodies was studied to restore the function of exhausted CD8+ T cells in CLL. CD8+ T cells were isolated from peripheral blood of 20 patients with CLL, treated with blocking antibodies, and cocultured with mitomycin-frozen non-CD8+ T cell fraction as target cells. Cultures were stimulated with anti-CD3/CD28 antibodies to assess the proliferation of CD8+ T cells by MTT and stimulated with PMA/ionomycin to measure the levels of CD107a expression and cytokine production by flow cytometry and ELISA, respectively. Our results showed that the blockade of PD-1 and TIM-3 does not improve the proliferation of CD8+ T cells in CLL patients. No significant difference was found between control and blocked groups in terms of degranulation properties and production of IFN-γ, TNF-α, IL-2, and IL-10 by CD8+ T cells. We observed that pre-treatment of CD8+ T cells with blocking antibodies in CLL patients at early clinical stages had no effects on restoring their functional properties. Further in vitro and in vivo complementary studies are required to more explore the utility of checkpoint inhibitors for CLL patients.
Article
Full-text available
Importance Clinical outcomes for glioblastoma remain poor. Treatment with immune checkpoint blockade has shown benefits in many cancer types. To our knowledge, data from a randomized phase 3 clinical trial evaluating a programmed death-1 (PD-1) inhibitor therapy for glioblastoma have not been reported. Objective To determine whether single-agent PD-1 blockade with nivolumab improves survival in patients with recurrent glioblastoma compared with bevacizumab. Design, Setting, and Participants In this open-label, randomized, phase 3 clinical trial, 439 patients with glioblastoma at first recurrence following standard radiation and temozolomide therapy were enrolled, and 369 were randomized. Patients were enrolled between September 2014 and May 2015. The median follow-up was 9.5 months at data cutoff of January 20, 2017. The study included 57 multicenter, multinational clinical sites. Interventions Patients were randomized 1:1 to nivolumab 3 mg/kg or bevacizumab 10 mg/kg every 2 weeks until confirmed disease progression, unacceptable toxic effects, or death. Main Outcomes and Measures The primary end point was overall survival (OS). Results A total of 369 patients were randomized to nivolumab (n = 184) or bevacizumab (n = 185). The MGMT promoter was methylated in 23.4% (43/184; nivolumab) and 22.7% (42/185; bevacizumab), unmethylated in 32.1% (59/184; nivolumab) and 36.2% (67/185; bevacizumab), and not reported in remaining patients. At median follow-up of 9.5 months, median OS (mOS) was comparable between groups: nivolumab, 9.8 months (95% CI, 8.2-11.8); bevacizumab, 10.0 months (95% CI, 9.0-11.8); HR, 1.04 (95% CI, 0.83-1.30); P = .76. The 12-month OS was 42% in both groups. The objective response rate was higher with bevacizumab (23.1%; 95% CI, 16.7%-30.5%) vs nivolumab (7.8%; 95% CI, 4.1%-13.3%). Grade 3/4 treatment-related adverse events (TRAEs) were similar between groups (nivolumab, 33/182 [18.1%]; bevacizumab, 25/165 [15.2%]), with no unexpected neurological TRAEs or deaths due to TRAEs. Conclusions and Relevance Although the primary end point was not met in this randomized clinical trial, mOS was comparable between nivolumab and bevacizumab in the overall patient population with recurrent glioblastoma. The safety profile of nivolumab in patients with glioblastoma was consistent with that in other tumor types. Trial Registration ClinicalTrials.gov Identifier: NCT02017717
Article
Full-text available
Checkpoint inhibitors have become an efficient way to treat cancers. Indeed, anti-CTLA-4, anti-PD1, and anti-PDL-1 antibodies are now used as therapies for cancers. However, while these therapies are very efficient in certain tumors, they remain poorly efficient in others. This might be explained by the immune infiltrate, the expression of target molecules, and the influence of the tumor microenvironment. It is therefore critical to identify checkpoint antigens that represent alternative targets for immunotherapies. PVR-like molecules play regulatory roles in immune cell functions. These proteins are expressed by different cell types and have been shown to be upregulated in various malignancies. PVR and Nectin-2 are expressed by tumor cells as well as myeloid cells, while TIGIT, CD96, and DNAM-1 are expressed on effector lymphoid cells. PVR is able to bind DNAM-1, CD96, and TIGIT, which results in two distinct profiles of effector cell activation. Indeed, while binding to DNAM-1 induces the release of cytokines and cytotoxicity of cytotoxic effector cells, binding TIGIT induces an immunosuppressive and non-cytotoxic profile. PVR is also able to bind CD96, which induces an immunosuppressive response in murine models. Unfortunately, in humans, results remain contradictory, and this interaction might induce the activation or the suppression of the immune response. Similarly, Nectin-2 was shown to bind TIGIT and to induce regulatory profiles in effectors cells such as NK and T cells. Therefore, these data highlight the potential of each of the molecules of the “PVR–TIGIT axis” as a potential target for immune checkpoint therapy. However, many questions remain to be answered to fully understand the mechanisms of this synapse, in particular for human CD96 and Nectin-2, which are still understudied. Here, we review the recent advances in “PVR–TIGIT axis” research and discuss the potential of targeting this axis by checkpoint immunotherapies.
Article
Full-text available
The Glioblastoma (GBM) immune microenvironment plays a critical role in tumor development, progression, and prognosis. A comprehensive understanding of the intricate milieu and its interactions remains unclear, and single-cell analysis is crucially needed. Leveraging mass cytometry (CyTOF), we analyzed immunocytes from 13 initial and three recurrent GBM samples and their matched peripheral blood mononuclear cells (pPBMCs). Using a panel of 30 markers, we provide a high-dimensional view of the complex GBM immune microenvironment. Hematoxylin and eosin staining and polychromatic immunofluorescence were used for verification of the key findings. In the initial and recurrent GBMs, glioma-associated microglia/macrophages (GAMs) constituted 59.05 and 27.87% of the immunocytes, respectively; programmed cell death-ligand 1 (PD-L1), T cell immunoglobulin domain and mucin domain-3 (TIM-3), lymphocyte activation gene-3 (LAG-3), interleukin-10 (IL-10) and transforming growth factor-β (TGFβ) demonstrated different expression levels in the GAMs among the patients. GAMs could be subdivided into different subgroups with different phenotypes. Both the exhausted T cell and regulatory T (Treg) cell percentages were significantly higher in tumors than in pPBMCs. The natural killer (NK) cells that infiltrated into the tumor lesions expressed higher levels of CXC chemokine receptor 3 (CXCR3), as these cells expressed lower levels of interferon-γ (IFNγ). The immune microenvironment in the initial and recurrent GBMs displayed similar suppressive changes. Our study confirmed that GAMs, as the dominant infiltrating immunocytes, present great inter- and intra-tumoral heterogeneity and that GAMs, increased exhausted T cells, infiltrating Tregs, and nonfunctional NK cells contribute to local immune suppressive characteristics. Recurrent GBMs share similar immune signatures with the initial GBMs except the proportion of GAMs decreases.
Article
Full-text available
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid progenitor and precursor cells at different stages of differentiation, which play an important role in tumor immunosuppression. Glioma is the most common and deadliest primary malignant tumor of the brain, and ample evidence supports key contributions of MDSCs to the immunosuppressive tumor microenvironment, which is a key factor stimulating glioma progression. In this review, we summarize the source and characterization of MDSCs, discuss their immunosuppressive functions, and current approaches that target MDSCs for tumor control. Overall, the review provides insights into the roles of MDSC immunosuppression in the glioma microenvironment and suggests that MDSC control is a powerful cellular therapeutic target for currently incurable glioma tumors.
Article
Multiple sclerosis (MS) is a neuroinflammatory disorder characterized by autoimmune-mediated inflammatory lesions in CNS leading to myelin damage and axonal loss. MS is a heterogenous disease with variable and unpredictable disease course. Due to its complex nature, MS is difficult to diagnose and responses to specific treatments may vary between individuals. Therefore, there is an indisputable need for biomarkers for early diagnosis, prediction of disease exacerbations, monitoring the progression of disease, and for measuring responses to therapy. Genomic and proteomic studies have sought to understand the molecular basis of MS and find biomarker candidates. Advances in next-generation sequencing and mass-spectrometry techniques have yielded an unprecedented amount of genomic and proteomic data; yet, translation of the results into the clinic has been underwhelming. This has prompted the development of novel data science techniques for exploring these large datasets to identify biologically relevant relationships and ultimately point towards useful bio-markers. Herein we discuss optimization of omics study designs, advances in the generation of omics data, and systems biology approaches aimed at improving biomarker discovery and translation to the clinic for MS.
Article
Single-cell transcriptomics has transformed our ability to characterize cell states, but deep biological understanding requires more than a taxonomic listing of clusters. As new methods arise to measure distinct cellular modalities, a key analytical challenge is to integrate these datasets to better understand cellular identity and function. Here, we develop a strategy to "anchor" diverse datasets together, enabling us to integrate single-cell measurements not only across scRNA-seq technologies, but also across different modalities. After demonstrating improvement over existing methods for integrating scRNA-seq data, we anchor scRNA-seq experiments with scATAC-seq to explore chromatin differences in closely related interneuron subsets and project protein expression measurements onto a bone marrow atlas to characterize lymphocyte populations. Lastly, we harmonize in situ gene expression and scRNA-seq datasets, allowing transcriptome-wide imputation of spatial gene expression patterns. Our work presents a strategy for the assembly of harmonized references and transfer of information across datasets.