Stromal cells of mesenchymal origin reside below the epithelial compartment and provide structural support in the intestine. These intestinal stromal cells interact with both the epithelial cell compartments, as well as infiltrating hematopoietic immune cells. The importance of these cells in regulating immune homeostasis during inflammation is well recognized. However, little is known about their function and phenotype in the inflammatory tumor microenvironment. Using a syngeneic, immunogenic model of colorectal cancer, we showed that TNFα-initiated inflammatory signaling in CT26 colorectal cancer cells selectively induced PD-L1 expression in stromal cells. Using CD274 shRNA and antibody-mediated approaches, we showed that stromal cell PD-L1 potentiated enhanced immunosuppression, characterized by inhibition of activated CD8+ granzyme B-secreting T cells in vitro, and the inhibition of CD8+ effector cells was associated with enhanced tumor progression. Stromal cell immunosuppressive and tumor-promoting effects could be reversed with administration of anti–PD-1 in vivo. We validated our findings of stromal cell CD274 expression in two cohorts of clinical samples and also observed PD-L1 induction on human stromal cells in response to exposure to the inflammatory secretome from human colon cancer cells, irrespective of microsatellite instability. Collectively, our data showed that tumor-associated stromal cells support T-cell suppression by PD-L1 induction, which is dependent on colon cancer inflammatory signaling. Our findings reveal a key role of mesenchymal stromal cells PD-L1 in suppression of CD8+ antitumor immune responses and potentiation of colorectal cancer progression. Cancer Immunol Res; 6(11); 1426–41. ©2018 AACR.

Colon cancer development and metastasis is a multistep process, during which accumulation of genetic mutations drives tumor development through acquisition of hallmarks, including the ability to evade immune surveillance (1). Colon tumors are composed of numerous cellular components that can facilitate an immunosuppressive microenvironment, including immune, stromal, and endothelial cells (2–5). In colon cancer, the type, density, and location of immune cells within tumors can predict clinical outcome (6), and colon cancer patients without tumor recurrence are shown to have higher immune cell densities (CD3, CD8, granzyme B) than patients whose tumors recurred after treatment (6). These observations highlight the need to better understand the cellular and molecular mechanisms that dictate an immunosuppressive microenvironment in colon cancer.

In the colon cancer microenvironment, activated T cells traffic from the blood or lymph nodes into the tumor. The stromal cells in the colon are a heterogeneous population (7, 8), with a large proportion being of mesenchymal origin. They are positioned between the epithelial cells and the underlying vasculature and can passively or actively impair immune cell trafficking and activation. Stromal cell influence in the tumor microenvironment has been highlighted in several studies that identify a stromal cell signature from colon tumors that is associated with tumor progression and a poorer prognosis in colon cancer patients (9). However, it remains poorly understood if the prognosis associated with these signatures is due to the inherent tumor-promoting or immunomodulatory potential of mesenchymal cells, which constitute a significant proportion of stromal cells in the intestinal microenvironment. These findings highlight the need to better understand the immunomodulatory role of the tumor stromal compartment to improve the development of effective stroma-targeting strategies.

Inflammation can promote tumorigenesis and is recognized as one of the hallmarks of cancer (1, 10). Inflammatory bowel disease is causally associated with colon cancer promotion (11). The transcription factor NF-κB, a key regulator of both inflammation and cancer, regulates the expression of many proinflammatory chemokines and cytokines and is associated with cancer progression and metastasis (8, 12). Many NF-κB–dependent soluble factors, including TNFα, can directly activate stromal cells to inhibit adaptive and innate immune effector mechanisms (13). Mesenchymal stromal cells (MSC) can suppress effector T lymphocytes, but little is known about how these stromal cell properties are altered in the inflammatory colon tumor microenvironment (14).

One important mechanism by which tumors avoid clearance by the immune system is by upregulating the expression of immunomodulatory ligands. In colorectal cancer, programmed death-ligand 1 (PD-L1) expression on tumor cells and tumor-infiltrating lymphocytes is of particular importance to this immune escape phenotype (15, 16). The receptor for PD-L1, PD-1, is expressed on activated T cells and, when bound by the ligand, results in downregulated proliferation and inhibition of effector function (15). The PD-1/PD-L1 pathway represents an adaptive immune resistance mechanism that is exerted by tumor cells in response to endogenous antitumor activity. An antibody targeting PD-1 has been granted FDA approval for metastatic colon cancer (17). Although the PD-1/PD-L1 signaling axis is important in colorectal cancer, anti–PD-1 therapy is not always effective in this setting, with some patients experiencing disease progression following treatment (18). Efforts to better stratify patients for anti–PD-1 immunotherapy have relied on the measurement of tumor or immune cell PD-L1 expression, but reports show that some patients deemed tumor PD-L1–negative respond positively to anti–PD-1 therapy (19).

The stromal compartment of the tumor microenvironment has received little attention to date in terms of stratifying patients for immunotherapy despite the known immunomodulatory potential of these cells. We, therefore, hypothesized that stromal cells in the inflammatory colon tumor microenvironment can directly modulate T-cell–mediated antitumor immunity through cell contact–dependent mechanisms. We showed that stromal cell PD-L1 expression induced by TNFα signaling in colon cancer cells led to decreased CD8+ T-cell proliferation, activation, and promotion of colon cancer growth in vivo.

Animals

Eight- to 14-week-old female Balb/c mice were purchased from Charles River. Experimental animals were housed in a specific pathogen-free facility and fed a standard chow diet. All procedures performed were conducted in a fully accredited animal housing facility at NUI Galway under a license granted by the Health Products Regulatory Authority, Ireland, and were approved by the NUI Galway Animal Care Research Ethics Committee.

Cell culture, MSC isolation, and conditioning

Mouse colon adenocarcinoma cells CT26, derived from Balb/c mice, were purchased from the European Collection of Cell Cultures (ECACC) and cultured in DMEM (Biosciences-Gibco) supplemented with 10% fetal bovine serum (FBS; Sigma) and 1% penicillin/streptomycin (Sigma). To assess the effects of CT26 NF--κB signaling, stable CT26 clones expressing a mutant form of the human IкB-α with serine 32 and 36 mutated into alanine (IкB-α SR) were generated as previously described (4). Human colon cancer cell lines HCT116 and HT29 were purchased from the ATCC and cultured in McCoys 5A medium (Sigma) supplemented with 10% FBS (Fisher, Hyclone), 1% l-glutamine, and 1% penicillin/streptomycin (Sigma). Mouse and human cell master stocks were authenticated by ECACC/ATCC, confirmed mycoplasma negative (MycoAlert, Lonza), expanded, frozen, and used within 15 passages of testing for all subsequent experiments.

For murine MSC isolation, Balb/c mice were euthanized by CO2 inhalation, and the femur and tibia were removed, cleaned, and placed in MSC culture medium; MEM-α (Biosciences-Gibco) supplemented with 10% FBS (Fisher, Hyclone), 10% Equine serum (Fisher, Hyclone), and 1% penicillin/streptomycin (Sigma). MSC were flushed from the bones, filtered, and plated at a density of 9 × 105 per cm2 in a T175 flask. Cells were incubated at 37°C, 5% CO2, and nonadherent cells were removed 24 hours later. This process was repeated 3 times per week until the cells reached confluency. MSC were characterized according to the criteria published by the ISCT cell-surface characterization (Supplementary Fig. S1A), and trilineage differentiation was carried out at passage 4 (Supplementary Fig. S1B–S1D). All subsequent experiments were carried out with mouse MSC between passages 5 and 14.

Human MSC (hMSC) were isolated from the bone marrow of three healthy volunteers at Galway University Hospital under an ethically approved protocol (NUIG Research Ethics Committee, Ref: 08/May/14) according to a standardized procedure. Written consent was obtained from the volunteers. Briefly, bone marrow cell suspensions were layered onto a Ficoll density gradient, and the nucleated cell fraction was collected, washed, and resuspended in MSC culture medium. After 24 hours of cultivation, nonadherent cells were removed, fresh medium was added, and individual colonies of fibroblast-like cells were allowed to expand and approach confluence prior to passage. hMSC were grown in alpha MEM (Gibco, Invitrogen) supplemented with 10% FBS (Sigma-Aldrich), 1% penicillin–streptomycin, and fibroblast growth factor 2 (FGF2, 1 ng/mL; Sigma-Aldrich).

For MSC tumor conditioning (MSCTCM), CT26, HCT116, and/or HT29 cells were seeded in a T175 flask (Nunc, Fisher) at a density of 1 × 106 cells per flask in 25 mL of the corresponding tumor cell culture medium. Cells were left to grow at 37°C at 5% CO2 (normoxia) for a total of 72 hours, at which point conditioned medium was collected, spun at 1,000 × g to pellet any cellular debris, and stored at −80°C. For TNFα conditioning (MSCTNF-TCM), TNFα (100 ng/mL; PeproTech) was added to tumor cell cultures 24 hours prior to collection of tumor cell medium. For conditioning mouse MSC, cells were seeded at a density of 0.03 × 106 cells per well of a 6-well plate in 2 mL MSC culture medium. Human MSC were seeded at a density of 0.1 × 106 cells per well of a 6-well plate. Twenty-four hours after seeding, the culture medium was removed and replaced with 40% fresh MSC medium and 60% tumor-conditioned medium. Fresh DMEM (MSCControl) and DMEM with TNFα (100 ng/mL; MSCTNF) were included as controls. MSC were analyzed at 24 and 72 hours after addition of conditioned medium. In vitro immunosuppression assays and in vivo experiments, described below, were all carried out using MSC that had been conditioned for 72 hours.

NF-κB reporter assay

The NF-κB luciferase reporters driven by 5 × wild-type (5 × NF-κB-Luc; pNF-κB-Luc plasmid; Stratagene) were used in this study. CT26/EV or CT26/IкB-α SR cells were generated and validated as previously described (4), plated in a 96-well plate, and cotransfected with 500 ng of pNF-κB-Luc or 500 ng of pLuc control vector (Stratagene) and 50 ng of RSV-pRL reporter (Promega Corp.). Twenty-four hours after transfection, cells were treated with TNFα at doses of 10 to 100 ng/mL for 24 hours. NF-κB activity was determined by analysis of luciferase activity with the Dual-Glo Luciferase Reporter Assay System (Promega) to validate reduced NF-κB activity in CT26/IкB-α SR cells (Supplementary Fig. S2A).

Lentivirus production

Four PD-L1 (CD274) mouse-specific shRNA expression vectors in pGFP-C-shLenti plasmids (CD274 shRNA Lenti-plasmid) and one scrambled negative control noneffective shRNA cassette in pGFP-C-shLenti plasmid (scramble shRNA Lenti-plasmid) were purchased from OriGene (OriGene Technologies; Cat. No. TL503436). Lentiviruses were generated by cotransfection of 293 T cells with either CD274 shRNA or scramble shRNA Lenti-plasmids, as well as with packaging (psPAX2.2), envelope (pMD2.G), and additional (pRSV-rev) plasmids using a standard protocol (Invitrogen). Supernatants were harvested 48 and 72 hours after transfection, combined, filtered through 0.45-μm pore size filters, and frozen at −80°C until required.

Lentiviral transduction of MSC

Balb/c MSC were removed from liquid nitrogen storage and passaged twice before transduction. Because pGFP-C-shLenti plasmids encode a puromycin resistance gene, MSC were treated with 7 μg/mL of puromycin (Gibco-Biosciences) for 96 hours, followed by 72 hours recovery in fresh MSC media. Following this, cells were imaged by fluorescent microscopy to identify GFP+ cells (Supplementary Fig. S4A). GFP expression was observed in both the CD274 shRNA– and scramble shRNA–transduced MSC, demonstrating successful transduction (Supplementary Fig. S4A).

RNA isolation and qRT-PCR

To determine if successful knockdown of PD-L1 was achieved, CD274 shRNA– and scramble shRNA–transduced MSC were centrifuged at 800 × g, supernatants were removed, and RNA was isolated from cell pellets using the Isolate II RNA MiniKit (Bioline) according to the manufacturer's instructions. RNA was resuspended in 40 μL of nuclease-free water and quantified by NanoDrop. cDNA was synthesized using RevertAid H Minus Reverse Transcriptase (Fermentas) with random primers. Two-step qRT-PCR was performed to determine the mRNA expression of CD274 (IDT Primetime qPCR assay: Mm.PT.58.11921659, Integrated DNA Technologies) by comparing Ct values with that of the housekeeping gene GAPDH (Primetime qPCR assay: Mm.PT.39a.1, Integrated DNA Technologies). Quantitative real-time PCR was performed according to the standard program on the ABI Step-one machine (Applied Biosystems). CD274 mRNA expression in CD274 shRNA– and scramble shRNA–transduced MSC are illustrated in Supplementary Fig. S4B.

Validation of tumor-conditioned shRNA PD-L1 MSC

MSC were seeded at a concentration of 1 × 105 cells per 6-well in 2 mL of mouse MSC media and allowed to adhere overnight. Lentivirus-containing supernatants from the different preparations were mixed with mouse MSC media at a ratio of 1:5 (mouse MSC media:lentiviral supernatant) to a final volume of 2 mL per well. Plates were incubated at 37°C, 5% CO2 for 24 hours. After 24 hours, the virus-containing media were removed and replaced with fresh mouse MSC media mixed with conditioned medium from CT26 cells preactivated with TNFα (MSCTNF-TCM; 40% MSC media and 60% TNFα TCM) and incubated for 24 hours. To test the transduction efficiency, 48 hours after transduction GFP+ cells were analyzed by flow cytometry (Supplementary Fig. S4D). In the same experiment, successful inhibition of PD-L1 expression on PD-L1 shRNA MSC was confirmed (Supplementary Fig. S4E and see Supplementary Fig. S4C for gating strategy).

T-cell immunosuppression assay

To obtain primary Balb/c lymphocytes, inguinal, mesenteric, and cervical lymph nodes, as well as spleens, were harvested from healthy animals following CO2 asphyxiation. Single-cell suspensions were obtained by mechanical disruption (using a 1-mL syringe plunger) of the tissue in culture medium: RPMI 1640 (Fisher, Lonza) supplemented with 10% FBS (Sigma), 1% sodium pyruvate, 1% nonessential amino acids, l-glutamine, 1% penicillin/streptomycin (all Sigma), and 0.01% β-mercaptoethanol. Cells were washed, resuspended in PBS, and erythrocytes lysed using ammonium–chloride–potassium lysing buffer (Sigma-Aldrich) for 5 minutes on ice. Cells were resuspended in culture medium, and a combination of 90% lymphocyte/10% splenocyte was used for immunosuppression assays.

To assess proliferation, cells were stained with carboxyfluorescein diacetate succinimidyl ester (CFSE) using the CellTrace cell proliferation kit (Invitrogen). For assessment of effects of shRNA MSC, lymphocytes were stained with cell trace violet (CTV) using a CellTrace Violet Cell Proliferation Kit (Thermo Fisher Scientific) in order to exclude MSC GFP expression from analysis. Cells were counted and resuspended in 1 mL PBS at a concentration of 1 × 107 cells/mL. CFSE and CTV were reconstituted in dimethyl sulfoxide, and 2 μL of reconstituted dye was added for each 1 mL of BSA/PBS. Cells were protected from light and incubated at 37°C for 6 minutes. Ice-cold culture medium was added to neutralize the CFSE, and cells were washed twice more. Cells were then activated using CD3/CD28 Mouse T-Activator Dynabeads (Life Technologies) for the duration of the 72-hour coculture.

CFSE-labeled and activated cells (0.1 × 106) from whole-cell isolates (90% lymphocyte/10% splenocyte) were cocultured with naïve or tumor-conditioned Balb/c MSC or TNFα tumor-conditioned PD-L1 shRNA– and scramble shRNA–transduced MSC at a ratio of 1 MSC:50 or 10 lymphocytes in a 96-well round bottom cell culture plate (Sarstedt). After 72 hours, medium was removed from the cocultures, aliquoted, and stored at −80°C for further analysis by ELISA. Cells were resuspended in FACS buffer (PBS supplemented with 2% FBS and 0.05% sodium azide) and were incubated with CD8a (APC, Clone 53-6.7; BioLegend) or CD4 (PerCP-Clone GK1.5; BioLegend) and/or CD25 (PE-clone PC61; BioLegend) for 15 minutes at 4°C. Cell viability was assessed following resuspension in 100 μL of FACS buffer by addition of the viability dye SYTOX blue (Thermo Fisher, S34857; Supplementary Fig. S2B) according to the product instructions. Cells were washed twice, and proliferation was measured using a FACSCanto cytometer (Becton Dickinson). Data were analyzed using FlowJo software (Version X, TreeStar Inc.). T-cell suppression was calculated according to the formula: 100 − ((% positive control divided/% sample divided) × 100) for all population doublings greater than the second generation (Supplementary Fig. S2C and S2D).

IFNγ, TNFα, and granzyme B enzyme-linked immunosorbent assays (ELISA)

Supernatants from MSC and T-cell cocultures were analyzed using Ready-SET-Go! ELISA kits (Affymetrix, eBioscience) for secretion of IFNγ, TNFα, and granzyme B. Flat-bottom 96-well ELISA plates were coated overnight at 4°C with capture antibody according to the manufacturer's protocol. The following morning, plates were washed with wash buffer (1× PBS with 0.05% Tween-20), and plates were blocked using the supplied assay diluent for 1 hour and then washed. Standards, as supplied with the assays and 100 μL of samples (1:2 dilution), were added to the plates and incubated for 24 hours at 4°C. Following 3 washes, detection antibody (100 μL) was added and incubated at room temperature for 1 hour. Following washing, 100 μL avidin horseradish peroxidase (HRP) was added for 30 minutes and washed a further 3 times. Tetramethylbenzidine substrate solution (100 μL) was then added and left at room temperature for 15 minutes before 50 μL stop solution (2N sulfuric acid) was added, and the plates were read at 450 and 570 nm on a Wallac 1420 plate reader (PerkinElmer).

Subcutaneous tumor model

Tumors were induced in 8- to 14-week-old female Balb/c mice by subcutaneous injection into the left flank of 2 × 105 CT26 cells ± 0.5 × 105 MSC (−/+ in vitro tumor preconditioning MSCControl/MSCTNF-TCM) in a total volume of 100 μL PBS. Animals receiving PD-1 antibody therapy received an intraperitoneal injection of 200 μg of anti–PD-1 (clone RMP1-14; Bio X Cell, 2B Scientific) in 100 μL PBS on days 7 and 14 after tumor induction. Tumor growth was monitored daily until sacrifice on day 21. At day 21, animals were euthanized, and tumors were harvested from left flank and, where tumor invasion had occurred as determined by visual tumor deposits, the peritoneal cavity. Tumor measurements were taken using digital calipers, and tumor volume was calculated according to the rational ellipse formula: (M1⁁2 × M2 × π/6). Images of tumors were taken of tumor tissue dissected from animals at day 21.

Cell-surface characterization of MSC or CT26 cells by flow cytometry

MSC or CT26 cells were trypsinized, counted, and washed in FACS buffer (PBS supplemented with 2% FBS and 0.05% sodium azide) prior to staining. Staining was carried out by incubating the cells with the antibody of interest in FACS buffer at 4°C for 10 minutes. Following this incubation, cells were washed twice in FACS buffer, stained with the viability dye SYTOX blue (Thermo Fisher, S34857), and analyzed straight after or prepared for intracellular staining (protocol below). Antibodies used were as follows: MHC-I (APC, clone SF1-1.1), MHC-II (FITC, clone 39-10-8), CD80 (PE, clone 16-10A1), CD86 (PE, clone PO3), PD-L1 (APC, clone 10F.9G2), PD-L2 (PE, clone TY25), and FasL (biotin with streptavidin–PE). All antibodies were purchased from BioLegend. Streptavidin was purchased from eBioscience. For characterization, 100,000 CT26 or MSC were stained per marker.

Analysis of tumors by flow cytometry

To analyze tumor immune cell infiltrates, tumors were digested in 1 mL HBSS (Gibco-Biosciences) containing 150 U/mL collagenase IV (Biosciences) and 200 U/mL DNase (Sigma). Samples were left at 37°C for 2 hours and then filtered through 40-μm cell strainers and washed with PBS. Single-cell suspensions were counted and stained with markers of interest: CD3 (FITC, clone 17A2), CD8 (APC, clone 53-6.7), CD25 (PE, clone PC61), CD4 (APC, clone GK1.5), all BioLegend. Cell-surface staining was carried out as before using 1 × 106 cells per sample and appropriate FMOs were used to ensure staining accuracy.

To stain for intracellular granzyme B (PE, clone REA226; Miltenyi), cells were washed twice after surface staining had been completed and fixed [2% paraformaldehyde (Sigma) for 10 minutes at 4°C] for intracellular staining. Cells were then incubated with granzyme B antibody diluted in permeabilization buffer (1% BSA/PBS with 0.5% saponin) according to the manufacturer's instructions. Samples were again incubated at 4°C for 10 minutes, washed twice in FACS buffer, and analyzed on a FACSCanto II.

Transcriptional data sets

Gene-expression profiles from two independent colorectal cancer data sets were downloaded from the NCBI Gene-Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) under accession numbers GSE35602 and GSE70468. GSE35602 contains microarray profiles separately profiled from laser-capture microdissected stroma or epithelium regions from 13 colorectal cancer primary tumors (20). GSE70468 contains microarray profiles from primary fibroblasts derived using colon primary tumor and morphologically normal colonic mucosa tissue isolated from fresh colorectal cancer resection material. Both studies indicate that they were performed after approval by an institutional review board (IRB), and informed written consent was obtained from the subjects (20, 21).

Bioinformatics analysis

Partek Genomics Suite software (version 6.6; Partek Inc.) was used for analysis of the independent data sets. For the purpose of clustering, data matrices were standardized to the median value of probe set expression. Standardization of the data allows for comparison of expression for different probe sets. Following standardization, two-dimensional hierarchical clustering was performed (samples × probe sets/genes). Euclidean distance was used to calculate the distance matrix, a multidimensional matrix representing the distance from each data point (probe set-sample pair) to all the other data points. Ward's linkage method was subsequently applied to join samples and genes together, with the minimum variance, to find compact clusters based on the calculated distance matrix.

Statistical analysis

Statistical analysis was carried out using GraphPad Version X (GraphPad Software). Data were assessed for normal distribution using D’Agostino–Pearson omnibus normality test. Data sets with two groups were analyzed using an unpaired t test. Data sets with more than two groups were analyzed by ordinary one-way ANOVA followed by the Tukey multiple comparisons test. For analysis of correlation, Pearson correlation coefficient was calculated. To test for a trend in tumor invasion data, a χ2 test was carried out, and to test for the effect of antibody treatment on individual treatment groups, Fisher exact test was used. Results were considered statistically significant at P < 0.05.

Inflammatory tumor-conditioned stromal cells inhibit T-cell proliferation and activation

A large proportion of the stromal component of the healthy colon and colon tumor microenvironment are of mesenchymal origin, which is an important barrier to overcome for infiltrating immune cells (22). However, the role of these stromal cells in modulating the immune response in the tumor microenvironment, and the influence of the tumor secretome on this capability, is unknown. To test the functional consequences of tumor conditioning on the ability of stromal cells to inhibit lymphocyte proliferation and effector function, we used an ex vivo syngeneic culture system, whereby primary Balb/c lymphocytes were cocultured with stromal cells conditioned by the tumor secretome in the presence (MSCTNF-TCM) or absence (MSCTCM) of inflammation [Fig. 1A(i–iv)]. A dose-dependent induction of NF-κB in CT26 in response to TNFα treatment was confirmed by a luciferase reporter gene assay (Supplementary Fig. S2A). Primary bone marrow stromal cells represent a robust model cell type for investigation of the effects of tumor conditioning on stromal cell components of the tumor microenvironment, considering they share numerous characteristics with intestinal stromal cells in terms of origin, surface marker expression, gene signatures, and immunomodulatory capacity (23–25). As previously documented, control MSC had limited ability to reduce CD4+ and CD8+ T-cell proliferation relative to positive controls [stimulated T cells and no stromal cells; Fig. 1B(i)]. However, this suppressive capacity was significantly increased by stromal cell exposure to the inflammatory tumor secretome [Fig. 1B(ii–iii)]. To identify the mechanism of stromal cell–mediated suppression, we assessed T-cell viability after coculture. No significant differences were observed in either CD4+ or CD8+ T cells cocultured with MSCControl, MSCTCM, or MSCTNF-TCM, suggesting induction of T-cell anergy or exhaustion rather than activation-induced cell death (Fig. 1C). In addition to enhanced T-cell suppression, inflammatory tumor-conditioned stromal cells, compared with control or TNFα-primed stromal cells, also significantly reduced lymphocyte activation, measured by TNFα and IFNγ release [Fig. 1D(i) and (ii); ref. 26]. A reduction in the cytolytic capacity of lymphocytes was seen when cultured with inflammatory tumor-conditioned stromal cells, measured by a reduction in granzyme B [Fig. 1D(iii)]. The inhibitory effect exerted by tumor-conditioned stromal cells on T-cell effector phenotype was most potent following stromal cell exposure to the secretome from TNFα pretreated CT26 cells, indicating a requirement for tumor cell inflammatory signaling in dictating stromal cell immunomodulatory function. These data demonstrate an enhanced ability of stromal cells to prevent the proliferation and effector function of T cells following exposure to the inflammatory tumor secretome. Considering the close contact between the tumor cells and the stromal niche in the colon, these data may imply stromal cells utilize the induction of PD-L1 to protect tumor cells from destruction by CD8+ cytotoxic T lymphocytes by inhibiting cytotoxicity and inducing T-cell anergy and exhaustion (27).

Figure 1.

Inflammatory tumor conditioning of stromal cells results in a significantly enhanced capacity to inhibit T-cell proliferation and activation. A, Experimental outline. MSC were seeded in 6-well plates, left to adhere for 24 hours, and subsequently treated with 60% tumor-conditioned medium or controls for 72 hours. Treatment groups were as follows: (i) untreated (MSCControl), (ii) TNFα-treated (MSCTNF-α), (iii) CT26-conditioned medium (MSCTCM), and (iv) conditioned medium from CT26 preactivated with TNFα (MSCTNF-TCM). B, (i) Representative histograms displaying CD4+ and CD8+ T-cell suppression following 72-hour coculture with tumor-conditioned MSC. Bar charts for (ii) CD4+ and (iii) CD8+ suppression greater than two generations. C, Cell viability assessed in CD4+ and CD8+ T cells by flow cytometry after 72-hour coculture with MSCControl, MSCTCM, MSCTNF-α, or MSCTNF-TCM. SYTOX blue negativity was used as an indicator of live cells. D, Culture supernatants were analyzed for the presence of (i) TNFα (ii) IFNγ, and (iii) granzyme B by ELISA following 72-hour coculture. Error bars, mean ± SEM; *, P < 0.05; **, P < 0.01; ***, P < 0.001 by one-way ANOVA and Tukey post hoc test; ****, P < 0.0001; n = 3 samples/group, 3–5 independent experiments.

Figure 1.

Inflammatory tumor conditioning of stromal cells results in a significantly enhanced capacity to inhibit T-cell proliferation and activation. A, Experimental outline. MSC were seeded in 6-well plates, left to adhere for 24 hours, and subsequently treated with 60% tumor-conditioned medium or controls for 72 hours. Treatment groups were as follows: (i) untreated (MSCControl), (ii) TNFα-treated (MSCTNF-α), (iii) CT26-conditioned medium (MSCTCM), and (iv) conditioned medium from CT26 preactivated with TNFα (MSCTNF-TCM). B, (i) Representative histograms displaying CD4+ and CD8+ T-cell suppression following 72-hour coculture with tumor-conditioned MSC. Bar charts for (ii) CD4+ and (iii) CD8+ suppression greater than two generations. C, Cell viability assessed in CD4+ and CD8+ T cells by flow cytometry after 72-hour coculture with MSCControl, MSCTCM, MSCTNF-α, or MSCTNF-TCM. SYTOX blue negativity was used as an indicator of live cells. D, Culture supernatants were analyzed for the presence of (i) TNFα (ii) IFNγ, and (iii) granzyme B by ELISA following 72-hour coculture. Error bars, mean ± SEM; *, P < 0.05; **, P < 0.01; ***, P < 0.001 by one-way ANOVA and Tukey post hoc test; ****, P < 0.0001; n = 3 samples/group, 3–5 independent experiments.

Close modal

The inflammatory tumor secretome induces PD-L1 expression on stromal cells

Stromal cells have been shown to inhibit T-cell responses by both cell contact–dependent mechanisms, via cell-surface protein expression, and cell contact–independent mechanisms, via release of soluble mediators (28, 29). Our initial finding of an enhanced immunosuppressive stromal cell potential following exposure to the inflammatory tumor secretome was observed following contact-dependent interactions between the stromal cells and lymphocytes, and this prompted a comprehensive analysis of an array of ligands on the stromal cell surface known to have immunomodulatory capacity. A significant increase in MHC-I expression following stromal cell exposure to the tumor secretome was seen, and this effect was significantly enhanced by inflammatory tumor activation [Fig. 2A(i)]. This observation may have important consequences, considering tumor antigens can be presented on MHC-I complexes potentially resulting in dysfunctional activation of antigen-specific CD8+ T cells (30). Compared with control stromal cells, tumor conditioning, in the presence or absence of inflammation, had no effect on the surface expression of MHC-II [Fig. 2A(ii)]. CD80 expression decreased, irrespective of inflammation, but with expression remaining high [Fig. 2A(iii)], whereas CD86 expression was unchanged [Fig. 2A(iv)].

Figure 2.

Exposure to the inflammatory tumor secretome induces markers of enhanced immunosuppressive ability on the stromal cell surface. A, Representative histograms and bar charts displaying MFI of MSC, cultured under the indicated conditions, for (i) MHC-I, (ii) MHC-II, (iii) CD80, and (iv) CD86. B, Histograms and bar charts displaying MFI of MSC, cultured under the indicated conditions, for (i) PD-L1, (ii) PD-L2, and (iii) FasL. C, Representative histograms and bar chart of PD-L1 expression on stromal cells compared with CT26 tumor cells. D, Bar chart showing PD-L1 induction following addition of TNFα directly to MSCTCM. Error bars, mean ± SEM; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by one-way ANOVA and Tukey post hoc test; n = 3 samples/group, 3–-5 independent experiments.

Figure 2.

Exposure to the inflammatory tumor secretome induces markers of enhanced immunosuppressive ability on the stromal cell surface. A, Representative histograms and bar charts displaying MFI of MSC, cultured under the indicated conditions, for (i) MHC-I, (ii) MHC-II, (iii) CD80, and (iv) CD86. B, Histograms and bar charts displaying MFI of MSC, cultured under the indicated conditions, for (i) PD-L1, (ii) PD-L2, and (iii) FasL. C, Representative histograms and bar chart of PD-L1 expression on stromal cells compared with CT26 tumor cells. D, Bar chart showing PD-L1 induction following addition of TNFα directly to MSCTCM. Error bars, mean ± SEM; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by one-way ANOVA and Tukey post hoc test; n = 3 samples/group, 3–-5 independent experiments.

Close modal

PD-L1 was found to be specifically increased following tumor conditioning, which was further increased in the presence of inflammation [Fig. 2B(i); refs. 31, 32]. Expression of PD-L2 and Fas Ligand (FasL), two negative regulators of T-cell function, was found to be low or almost absent, respectively, and remained unchanged following exposure to the tumor secretome [Fig. 2B(ii) and (iii); refs. 33–35]. These findings confirmed our previous observations that T-cell viability remained unchanged following coculture. Compared with controls or TNFα-treated stromal cells, where PD-L1 expression was low, inflammatory tumor-conditioned stromal cells expressed PD-L1 to a higher extent than CT26 tumor cells (Fig. 2C). This finding points to an important role for the stromal niche in dictating the fate of tumor-infiltrating, PD-1 receptor–expressing immune cells. We confirmed that TNFα, when added to MSCTCM, did not induce stromal cell PD-L1 expression in contrast to MSCTNF-TCM. This suggests that TNFα-induced NF-κB signaling in CT26 cells is responsible for the PD-L1 induction (Fig. 2D). This finding was further confirmed using conditioned media from TNFα-treated NF-κB–deficient CT26 tumor cells. PD-L1 expression was significantly reduced on MSCTNF-TCM conditioned with CT26/IκB-α SR cells when compared with CT26/EV control cells [Supplementary Fig. S3(i) and (ii)].

Tumor stromal cell suppression of cytolytic CD8+ T cells is reversed by blocking PD-1

To confirm a definitive role for induced stromal cell PD-L1 in the suppression of T-cell proliferation and activation, we targeted the PD-1/PD-L1 signaling axis using a monoclonal blocking antibody to the PD-1 receptor. We observed high PD-1 expression on CD4+ and CD8+ T cells (Fig. 3A) ex vivo. Treatment with anti–PD-1 significantly reduced the ability of inflammatory tumor-conditioned stromal cells to suppress the proliferation of CD8+ T cells [Fig. 3B(i–ii)]. Following anti–PD-1 treatment, CD8+ T-cell suppression elicited by stromal cells exposed to the inflammatory tumor secretome was comparable with that induced by stromal cells exposed to the tumor secretome alone or TNFα-treated MSC [Fig. 3B(ii)]. These findings indicated that the enhanced CD8+ T-cell suppressive capacity of these cells is dependent on stromal PD-L1 expression induced by inflammatory signaling in the tumor microenvironment. No significant differences were seen in the capacity of inflammatory tumor-conditioned stromal cells to suppress CD4+ T cells, upon treatment with anti–PD-1 [Fig. 3B(iii)]. These findings may indicate a separate unknown mechanism by which stromal cells in the tumor microenvironment modulate CD4+ T cells. We further confirmed these findings using shRNA CD274 knockdown MSC. shRNA CD274 and scramble shRNA MSC were extensively characterized for shRNA transduction and knockdown efficiency by microscopy (Supplementary Fig. S4A), mRNA expression (Supplementary Fig. S4B), and flow cytometry (Supplementary Fig. S4C). Quantification of the percentage of GFP+ shRNA CD274 and scrambled MSC (Supplementary Fig. S4D) and of the percentage of PD-L1+ MSC from the GFP+ population (Supplementary Fig. S4E) were assessed prior to coculture with T cells in vitro. Compared with MSC controls, shRNA PD-L1 MSCTNF-TCM significantly reversed the ability of MSCTNF-TCM to suppress CD8+ T-cell proliferation [Fig. 3C(i)]. Similar to our findings using anti–PD-1, this effect was specific to CD8+ T cells, as the observation was not evident in the CD4+ T-cell subset [Fig. 3C(ii)]. We also observed a restoration of CD25 expression on CD8+ T cells following coculture with shRNA CD274 MSCTNF-TCM compared with MSCTNF-TCM [Fig. 3D(i)]. The CD25 restoration was not evident on the CD4+ T cells [Fig. 3D(ii)], validating that the PD-L1–mediated effects were predominantly on the CD8+ T cells. PD-1 blockade was sufficient to restore lymphocyte activation and cytolytic potential, measured by IFNγ, TNFα, and granzyme B secretion (Fig. 3E). This demonstrates the critical role of the PD-1/PD-L1 signaling axis in the ability of tumor-conditioned stromal cells to inhibit CD8+ T-cell–mediated antitumor immune effector functions.

Figure 3.

Blocking PD-1 signaling reverses the increased CD8+ T-cell suppression induced by inflammatory tumor-conditioned stromal cells. A, Representative histograms of PD-1 expression on murine CD4+ and CD8+ T cells isolated from lymph nodes and spleen (n = 3). B, (i) Representative histograms of CD4+ and CD8+ T-cell proliferation with (dark gray/check bars) and without (light gray histogram/black bars) PD-1 blocking antibody in the presence of MSCTNF-TCM. Bar charts showing (ii) % CD8+ and (iii) % CD4+ T-cell suppression with and without treatment with PD-1 blocking antibody. C, Bar charts and representative histograms of (i) CD8+ and (ii) CD4+ T-cell proliferation following coculture with CD274 shRNA or scramble shRNA MSC. D, Bar charts showing CD25 expression (MFI) on (i) CD8+ and (ii) CD4+ T cells following coculture with CD274 shRNA or scramble shRNA MSC. E, Measurement of TNFα, IFNγ, and granzyme B by ELISA in culture supernatants without (black bars) and with anti–PD-1 (striped bars). Error bars, mean ± SEM; *, P < 0.05; **, P < 0.01; ***, P < 0.00; and ****, P < 0.0001; by one-way ANOVA or unpaired t test and Tukey post hoc test; n = 3 samples/group, 3–5 independent experiments.

Figure 3.

Blocking PD-1 signaling reverses the increased CD8+ T-cell suppression induced by inflammatory tumor-conditioned stromal cells. A, Representative histograms of PD-1 expression on murine CD4+ and CD8+ T cells isolated from lymph nodes and spleen (n = 3). B, (i) Representative histograms of CD4+ and CD8+ T-cell proliferation with (dark gray/check bars) and without (light gray histogram/black bars) PD-1 blocking antibody in the presence of MSCTNF-TCM. Bar charts showing (ii) % CD8+ and (iii) % CD4+ T-cell suppression with and without treatment with PD-1 blocking antibody. C, Bar charts and representative histograms of (i) CD8+ and (ii) CD4+ T-cell proliferation following coculture with CD274 shRNA or scramble shRNA MSC. D, Bar charts showing CD25 expression (MFI) on (i) CD8+ and (ii) CD4+ T cells following coculture with CD274 shRNA or scramble shRNA MSC. E, Measurement of TNFα, IFNγ, and granzyme B by ELISA in culture supernatants without (black bars) and with anti–PD-1 (striped bars). Error bars, mean ± SEM; *, P < 0.05; **, P < 0.01; ***, P < 0.00; and ****, P < 0.0001; by one-way ANOVA or unpaired t test and Tukey post hoc test; n = 3 samples/group, 3–5 independent experiments.

Close modal

In the context of immunotherapy, biologics targeting PD-L1 and PD-1 are showing great promise, but only in a subset of patients. A number of clinical trials have relied on measurements of PD-L1 expression on tumor cells or tumor-infiltrating immune cells in an effort to predict those patients who will respond best, but this method of stratification has had only limited success, with evidence now showing that some patients deemed PD-L1 negative will respond to this therapy (19, 36). Our findings may highlight an important role for stromal cell PD-L1 expression in modulating antitumor T-cell responses and in selecting optimal immunotherapeutic strategies for patients.

Inflammatory tumor-conditioned stromal cells reduce CD8+ T-cell cytotoxicity

The effect of stromal cells on the progression of colon cancer has been tested in vivo, but much of this work has relied on the use of immunocompromised mice, limiting the ability to assess the antitumor immune response (37, 38). In order to test the functional consequences of stromal PD-L1 expression in vivo, we used a fully immunocompetent, syngeneic Balb/c subcutaneous tumor model, whereby the immunogenic CT26 cell line was administered with or without control or inflammatory tumor-conditioned stromal cells (39). Because our in vitro analysis showed that increased PD-L1 expression was unique to stromal cells exposed to the inflammatory tumor secretome, we assessed this group compared with control MSC in vivo (Fig. 4A). Untreated MSC were used as controls as PD-L1 expression on these cells was not significantly different to MSCTNF-α or MSCTCM (Fig. 2). The preliminary experiments we carried out as part of this study showed that control stromal cells induced increased tumor growth compared with that of CT26 tumor cells alone (Supplementary Fig. S5A), and tumor burden was not different from that induced by MSCTNF-α or MSCTCM (Supplementary Fig. S5B). The use of untreated MSC, thereby, allowed us to minimize the number of variables tested in our in vivo system.

Figure 4.

Inflammatory tumor-conditioned stromal cells reduce the cytolytic capacity of tumor-infiltrating CD8+ T cells resulting in enhanced tumor growth and invasiveness. A, Schematic of experimental design. Balb/c mice were injected subcutaneously with CT26 cells alone or in combination with control or inflammatory tumor-conditioned MSC at day 0. Tumors were harvested for analysis at day 21. B, Tumors were processed to single-cell suspensions and analyzed by flow cytometry for the presence of infiltrating (i) CD3+, (ii) CD4+, (iii) CD8+, and (iv) Granzyme B+ CD8+ lymphocytes. (v) MFI was used to compare granzyme B expression on CD8+ T cells between groups. *, P < 0.05 by one-way ANOVA and Tukey post hoc test; n = 3–9 mice/group. C, Percentage of animals with an invasive tumor phenotype, defined as any tumor that penetrated skin tissue or grew across the peritoneal membrane with tumor deposits found attached to the liver and colon. Error bars, mean ± SEM; ****, P < 0.0001 by χ2 test for trend (n = 3–9 mice/group). D, Tumor volume measured by the rational ellipse formula (M1⁁2 × M2 × π/6). Error bars, mean ± SEM; *, P < 0.05; **, P < 0.01 one-way ANOVA, Tukey post hoc test. n = 3–9 mice/group, two independent experiments.

Figure 4.

Inflammatory tumor-conditioned stromal cells reduce the cytolytic capacity of tumor-infiltrating CD8+ T cells resulting in enhanced tumor growth and invasiveness. A, Schematic of experimental design. Balb/c mice were injected subcutaneously with CT26 cells alone or in combination with control or inflammatory tumor-conditioned MSC at day 0. Tumors were harvested for analysis at day 21. B, Tumors were processed to single-cell suspensions and analyzed by flow cytometry for the presence of infiltrating (i) CD3+, (ii) CD4+, (iii) CD8+, and (iv) Granzyme B+ CD8+ lymphocytes. (v) MFI was used to compare granzyme B expression on CD8+ T cells between groups. *, P < 0.05 by one-way ANOVA and Tukey post hoc test; n = 3–9 mice/group. C, Percentage of animals with an invasive tumor phenotype, defined as any tumor that penetrated skin tissue or grew across the peritoneal membrane with tumor deposits found attached to the liver and colon. Error bars, mean ± SEM; ****, P < 0.0001 by χ2 test for trend (n = 3–9 mice/group). D, Tumor volume measured by the rational ellipse formula (M1⁁2 × M2 × π/6). Error bars, mean ± SEM; *, P < 0.05; **, P < 0.01 one-way ANOVA, Tukey post hoc test. n = 3–9 mice/group, two independent experiments.

Close modal

Evidence exists to suggest that tumor-infiltrating lymphocytes contribute significantly to the prognosis, responsiveness to treatment, and likelihood of disease relapse in cancer patients (6). Our analysis of tumor immune cell infiltrates showed no change in the frequency of CD3+, CD3+CD4+, or CD3+CD8+ cells [Fig. 4B(i–iii); Supplementary Fig. S6]. There was a trend toward reduced frequency of granzyme B+ CD8+ T cells [Fig. 4B(iv)], but this did not reach statistical significance. Considering the previously published observation of increased granzyme B expression in colon tumors correlating with favorable patient outcomes, we measured granzyme B expression on CD8+ T cells by median fluorescent intensity (MFI) and found a significant reduction in the expression in tumors formed by the coadministration of CT26 with inflammatory tumor-conditioned stromal cells compared with CT26 cells alone. This result was dependent on the exposure of the stromal population to the inflammatory tumor secretome and, thus, may indicate a role for stromal PD-L1 in this effect. Cytotoxic CD8+ T lymphocytes are important in antitumor immune responses, and a reduction in granzyme B expression could result in an inhibition of their tumor cell clearing capacity (40).

Inflammatory tumor-conditioned stromal cells significantly enhance tumorigenesis

In addition to altering the tumor immune cell infiltrate, stromal cells have been shown to promote the growth and invasion of tumors (41, 42). In our model, injection of CT26 alone induced subcutaneous tumor formation with no evidence of tumor cell invasion and metastasis. In 50% of animals receiving control stromal cells, tumors were observed to have invaded tissue surrounding the primary tumor site and metastasized (Fig. 4C). This effect was significantly potentiated by the coadministration of stromal cells exposed to the inflammatory tumor secretome (Fig. 4C). In the clinical setting, an invasive or metastatic tumor is associated with a much less favorable outcome for patient survival. This holds true in the setting of colorectal cancer, where the emergence of metastasis is particularly grave (43). A significant increase in tumor volume was observed upon the coinjection of stromal cells, with inflammatory tumor-conditioned stromal cells inducing greater increases in volume than CT26 alone (Fig. 4D). These results highlight a role for inflammatory tumor signaling in altering the functional characteristics of the tumor stroma. The observations of smaller, though significant, increases in invasiveness and volume upon administration of control stromal cells may be indicative of in situ induction of PD-L1 expression on control stromal cells when in contact with tumor cells. The enhanced increases observed following inflammatory tumor-conditioned stromal cells point to an important role for early polarization of the stromal cell phenotype prior to or at the beginning of tumor formation.

To confirm the immunomodulatory role of inflammatory tumor-conditioned stromal cells and to test the effect of increasing stromal cell number, a second group of animals was coadministered CT26 with double the number of stromal cells used previously (0.5 × 105 vs. 1 × 105). This was investigated using both control stromal cells and those exposed to the inflammatory tumor secretome. No differences were found in tumor invasiveness or tumor volumes between animals that had received the lower or higher stromal cell number (Supplementary Fig. S7). Hence, all further experiments and analysis were carried out using the lower number of MSC, in consideration of physiologic levels of MSC in the TME. These data confirmed that the presence of an immunosuppressive stromal niche in a tumor is sufficient to significantly affect ability of immune cells to access and eliminate that tumor. We have shown that the presence of this stromal population caused biologically relevant and potent effects on tumor promotion, even when present in low numbers.

Anti–PD-1 restores CD8+ T-cell activation and reverses tumorigenesis

To confirm a role for PD-L1 in the enhanced immunosuppressive and tumor-promoting ability of coadministered stromal cells observed in vivo, three additional groups of animals were treated with anti–PD-1 at 7 and 14 days after tumor induction (Fig. 5A). In line with previous data demonstrating only mild responses to PD-1 monotherapy, in CT26 derived tumors, PD-1 antibody treatment had no significant effect on the immune cell infiltrate of tumors, and anti–PD-1 treatment also did not alter the frequency of CD3+, CD3+CD4+, or CD3+CD8+ T cells infiltrating coinjected CT26 and stromal cell tumors [Fig. 5B(i–iii)]. However, a trend toward restoration of the cytolytic capacity of intratumoral CD8+ T cells was seen, measured by granzyme B expression, in tumors formed by coadministration of CT26 with inflammatory tumor-conditioned stromal cells [Fig. 5B(iv); refs. 44, 45]. The immune cell checkpoint inhibition induced by anti–PD-1 was sufficient to reverse the increased tumor invasiveness that had been observed with the coadministration of inflammatory tumor-conditioned stromal cells (Fig. 5C). This suggests that stromal cell PD-L1 expression may have a central role in enabling the tumor to inhibit CD8+ T-cell–mediated inhibition of metastatic tumor spread. A similar significant reduction in tumor volume was observed upon treatment with anti–PD-1 (Fig. 5D). Images of tumors excised from treated animals indicated the increased number of invasive nodules on tumors in the CT26 coinjected with MSCTNF-TCM group compared with either CT26 alone or CT26 coinjected with MSCControl. This observation of enhanced invasiveness of tumors was reversed after treatment with anti–PD-1 therapy (Fig. 5E). Tumor volume in animals administered inflammatory tumor-conditioned stromal cells treated with PD-1 monoclonal antibody was similar to that of animals administered CT26 alone. These data point to a central role for stromal cell PD-L1 expression in obstructing the activity of antitumor CD8+ immune responder cells, allowing tumors to grow through immune evasion.

Figure 5.

Treatment with anti–PD-1 restored antitumor immune responses resulting in decreased invasiveness and tumor growth. A, Schematic of experimental design. Balb/c mice were administered CT26 cells alone or in combination with control or inflammatory tumor-conditioned MSC at day 0. Anti–PD-1 was administered i.p. on days 7 and 14. Tumors were harvested for analysis at day 21. B, Tumors were processed to single-cell suspension and analyzed by flow cytometry for the presence of infiltrating (i) CD3+, (ii) CD4+, and (iii) CD8+ T cells. For CT26 controls, MSCControl and MSCTNF-TCM from Fig. 4B (i–iii) were used. (iv) Median fluorescent intensity (MFI) was used to compare granzyme B expression on CD8+ T cells between groups. One-way ANOVA and Tukey post hoc test. C, Percentage of animals with an invasive tumor phenotype (defined in 4C). Error bars, mean ± SEM; *, P < 0.05; ****, P < 0.0001 by Fisher exact test. D, Tumor volume measured by the rational ellipse formula (M1⁁2 × M2 × π/6). For CT26 controls, MSCControl and MSCTNF-TCM from Fig. 4B (v) were used. Error bars, mean ± SEM; *, P < 0.05 by one-way ANOVA and Tukey post hoc test; E, Representative images of tumor tissue dissected from animals at day 21 with (right) or without (left) anti–PD-1. n = 3–9 per group. n = 2 independent experiments.

Figure 5.

Treatment with anti–PD-1 restored antitumor immune responses resulting in decreased invasiveness and tumor growth. A, Schematic of experimental design. Balb/c mice were administered CT26 cells alone or in combination with control or inflammatory tumor-conditioned MSC at day 0. Anti–PD-1 was administered i.p. on days 7 and 14. Tumors were harvested for analysis at day 21. B, Tumors were processed to single-cell suspension and analyzed by flow cytometry for the presence of infiltrating (i) CD3+, (ii) CD4+, and (iii) CD8+ T cells. For CT26 controls, MSCControl and MSCTNF-TCM from Fig. 4B (i–iii) were used. (iv) Median fluorescent intensity (MFI) was used to compare granzyme B expression on CD8+ T cells between groups. One-way ANOVA and Tukey post hoc test. C, Percentage of animals with an invasive tumor phenotype (defined in 4C). Error bars, mean ± SEM; *, P < 0.05; ****, P < 0.0001 by Fisher exact test. D, Tumor volume measured by the rational ellipse formula (M1⁁2 × M2 × π/6). For CT26 controls, MSCControl and MSCTNF-TCM from Fig. 4B (v) were used. Error bars, mean ± SEM; *, P < 0.05 by one-way ANOVA and Tukey post hoc test; E, Representative images of tumor tissue dissected from animals at day 21 with (right) or without (left) anti–PD-1. n = 3–9 per group. n = 2 independent experiments.

Close modal

PD-L1 is differentially expressed on cancerous stroma in clinical samples

Because our experiments to date had been carried out using murine tissue, we next wanted to confirm the phenomenon of stromal cell PD-L1 induction on stromal tissue in the human tumor microenvironment. We examined transcriptional profiles of colorectal cancer resection samples (data set GSE35602), which had been laser-capture microdissected (LCM) to isolate the stromal and epithelial fractions prior to microarray profiling (20). Assessment of PD-L1 gene expression (CD274) indicated significantly higher expression of PD-L1 in the stromal compartment of CRCs compared with the epithelial cells [Fig. 6A(i)]. Assessment of EpCam (EPCAM), E-caderhin (CDH1), as markers of epithelial lineage, and α-SMA (ACTA2) and vimentin (VIM), as markers of mesenchymal lineages, further confirmed the purity of the samples following LCM [Fig. 6A(ii)]. Assessment of PD-L1, PDGFR-α (PDGFRA), CD90 (THY1), and CD105 (ENG) indicated that gene expression of these markers was associated with the stromal compartment in colorectal cancer tissue samples. Because of this observation, we next investigated if exposure to the tumor secretome would induce human stromal cell PD-L1 expression in a manner similar to that seen for our murine cells. Using primary human bone marrow stromal cells from healthy donors, which we showed were CD90, CD105, and PD-L1–positive [Fig. 6B(i–iii)], and the secretome from microsatellite (HCT116) or chromosomal (HT29) unstable cells, we repeated the conditioning protocol outlined in Fig. 1A (46). We observed a significant increase in PD-L1 expression upon exposure to the inflammatory tumor secretome, and this induction occurred irrespective of tumor cell microsatellite instability (HCT116) or chromosomal instability [HT29; Fig. 6B(iii–v); ref. 46].

Figure 6.

PD-L1 (CD274) expression is differentially expressed on cancerous stroma in clinical samples and is also induced on human stromal cells exposed to the inflammatory tumor secretome. A, (i) Relative CD274 (PD-L1) gene-expression profile on epithelial and stromal cells from colorectal cancer patients (n = 13; data set GSE35602). (ii) Clustering for gene-expression profiles of PDGFR-α (PDGFRA), PD-L1 (CD274), CD90 (THY1), CD105 (ENG), Vimentin (VIM), α-SMA (ACTA2), E-cadherin (CDH1), and EPCAM (EPCAM). B, Healthy donor human stromal cells expression of (i) CD90, (ii) CD105, and (iii) PD-L1. Primary human MSCs (n = 1 donor) were treated with conditioned medium from control or TNFα-activated (iv) HCT116 or (v) HT29 human colon tumor cells and PD-L1 expression (MFI) was measured by flow cytometry (n = 3 independent experiments). Error bars: mean ± SEM, P < 0.05 by one-way ANOVA and Tukey post hoc test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by unpaired t test. C, Relative PD-L1 gene expression in normal and CAFs from colorectal cancer patients (n = 7; data set GSE70468). Error bars, min–max; UNPAIRED t test.

Figure 6.

PD-L1 (CD274) expression is differentially expressed on cancerous stroma in clinical samples and is also induced on human stromal cells exposed to the inflammatory tumor secretome. A, (i) Relative CD274 (PD-L1) gene-expression profile on epithelial and stromal cells from colorectal cancer patients (n = 13; data set GSE35602). (ii) Clustering for gene-expression profiles of PDGFR-α (PDGFRA), PD-L1 (CD274), CD90 (THY1), CD105 (ENG), Vimentin (VIM), α-SMA (ACTA2), E-cadherin (CDH1), and EPCAM (EPCAM). B, Healthy donor human stromal cells expression of (i) CD90, (ii) CD105, and (iii) PD-L1. Primary human MSCs (n = 1 donor) were treated with conditioned medium from control or TNFα-activated (iv) HCT116 or (v) HT29 human colon tumor cells and PD-L1 expression (MFI) was measured by flow cytometry (n = 3 independent experiments). Error bars: mean ± SEM, P < 0.05 by one-way ANOVA and Tukey post hoc test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 by unpaired t test. C, Relative PD-L1 gene expression in normal and CAFs from colorectal cancer patients (n = 7; data set GSE70468). Error bars, min–max; UNPAIRED t test.

Close modal

Our data in both mouse and human models have indicated that PD-L1 expression is elevated in MSC following exposure to conditioned media from inflammatory stimulated colon cancer cells. Cancer-associated fibroblasts (CAF) represent a major component of colorectal cancer stroma and provide a suitable model to test our findings in colorectal cancer clinical samples. Therefore, we utilized gene-expression profiles (data set GSE70468) derived from patient-matched primary fibroblasts (n = 14 samples from 7 patients) isolated from within colorectal cancer tissue (CAFs), to represent MSC following cancer cell exposure, or from adjacent, distant, normal mucosal tissue (normal), to represent unconditioned MSC (20). Analysis of overall gene-expression profiles indicated a nonsignificant trend (P 0.10; range, 1.15–1.08-fold change) toward increased PD-L1 gene expression in CAFs compared with normal fibroblasts (Fig. 6C). A pairwise analysis of patient-matched samples further confirmed this finding, with 6 of the 7 matched CAFs displaying an increase in PD-L1 gene expression compared with their patient-matched normal fibroblast (Supplementary Fig. S8). This analysis in two sets of independent colorectal cancer clinical tissue cohorts supports our observations in the mouse model and adds further weight to our suggestion of an important role for stromal cell PD-L1 induction in colorectal cancer.

In this article, we described a central role for stromal cell PD-L1 expression in the tumor microenvironment in aiding the tumor in avoiding immune cell–mediated clearance. This dampening of the antitumor immune responses by the stromal compartment resulted in enhanced tumor burden and invasiveness (Fig. 7). The stromal compartment is located adjacent to both the cancerous epithelial cells and the colonic vasculature and lymphatic network, representing a physical barrier to entry for immune cells in response to inflammation or epithelial transformation (47). In addition to this structural role, these cells have been shown to produce soluble mediators and express proteins that can influence immune and epithelial cells in diseased tissue (48). We have now shown that PD-L1, induced in response to activation of inflammatory signaling in the tumor, is a critical mediator of stromal cell immunomodulatory capacity. More specifically, we identified tumor cell NF-κB as an important signaling pathway in the induction of a tumor-promoting stromal cell phenotype, demonstrated by a lack of induced stromal cell PD-L1 expression in the absence of TNFα-induced NF-κB activation in CT26 cells. Given the inflammatory nature of the colon tumor microenvironment and the important role for NF-κB signaling in the pathogenesis of colon cancer, these results may highlight a mechanism by which NF-κB signaling in colon tumor cells enables communication with the adjacent stromal compartment, thereby dictating their immunomodulatory function (49, 50). Interestingly, a study by Lakins and colleagues has shown that CAFs have similar properties as those described here, in terms of their ability to suppress antitumor responses. However, the effects highlighted in their study were shown to be dependent on T-cell death mediated by FasL and PD-L2. In contrast, we highlighted a significant role for tumor-induced stromal cell PD-L1 in cell death–independent suppression of CD8+ T-cell proliferation and activation, indicating induction of anergy. These results may highlight the importance of the nature of the tumor microenvironment on the induction of stromal cell–mediated immunosuppression (i.e., the presence of inflammation or hypoxia). Increasing our understanding of the impact of stromal cell functional status, as well as other tumor-associated immunosuppressive cell populations in different tumor microenvironments, will be essential to improving the efficacy of immunotherapies in the future.

Figure 7.

TNFα-induced colon cancer signaling induces stromal cell PD-L1, which inhibits cytotoxic CD8+ T-cell antitumor immune responses and promotes colon cancer. Using CD274 shRNA and antibody-mediated approaches, we showed that stromal cell PD-L1 potentiated enhanced immunosuppression, characterized by the inhibition of activated CD8+ granzyme B-secreting T cells in vitro, and the inhibition of CD8+ effector cells was associated with enhanced tumor progression. Targeting the PD-L1/PD-1 signaling axis reversed the MSC PD-L1–induced immunosuppression and tumor-promoting effects.

Figure 7.

TNFα-induced colon cancer signaling induces stromal cell PD-L1, which inhibits cytotoxic CD8+ T-cell antitumor immune responses and promotes colon cancer. Using CD274 shRNA and antibody-mediated approaches, we showed that stromal cell PD-L1 potentiated enhanced immunosuppression, characterized by the inhibition of activated CD8+ granzyme B-secreting T cells in vitro, and the inhibition of CD8+ effector cells was associated with enhanced tumor progression. Targeting the PD-L1/PD-1 signaling axis reversed the MSC PD-L1–induced immunosuppression and tumor-promoting effects.

Close modal

By utilizing an immunocompetent, syngeneic murine model, we could overcome the limitations in assessing immune cell infiltrate associated with the use of xenografts in immunocompromised mice. This allowed us to identify CD8+ T-cell granzyme B as a potential effector molecule inhibited by the stromal cell barrier in the tumor microenvironment, which was restored by anti–PD-1 therapy. With specific regard to colon cancer, higher intratumoral expression of granzyme B has been shown to correlate with improved survival in patients (51). Collectively, these data demonstrate the importance of the stromal compartment in modulating the CD8+ T-cell–mediated antitumor immune responses in a PD-L1–dependent manner.

Expression of PD-L1 has been primarily detected on the surface of epithelial neoplastic cells in a number of cancers. However, in colorectal carcinoma, IHC-based studies of small cohorts have detected high PD-L1 expression in the stromal and immune compartments (18, 52, 53). In light of studies showing positive responses to PD-1 immunotherapy in patients whose tumors have been deemed PD-L1–negative, we provide a rationale for the assessment of stromal cell PD-L1 expression in order to better stratify patients for immunotherapy (19).

We also showed the phenomenon of inflammatory tumor-induced PD-L1 expression on human stromal cells in response to the secretome from microsatellite- or chromosomal instable human colon tumor cells. This is particularly important in light of data showing a better response to PD-1 immunotherapy in MSI-high colon cancer patients. We suggest that this is, as was discussed by Huang and Wu, a result of a lower mutational load attracting much fewer tumor-infiltrating CD8+ T cells (54). In this setting, PD-1 inhibition is of limited use because a paucity of T cells, whose effector function can be disinhibited by such therapy, exists. However, as the authors suggest, treatment with a second immune-stimulatory agent, such as MEKi, could lead to CD8+ T-cell infiltration and, thus, synergistic and favorable responses in these patients (54, 55). We validated our in vitro and in vivo findings in two independent colorectal cancer clinical resection cohorts. Using transcriptional profiles specific for the stromal or epithelial fractions of overall colorectal cancer tissue (20), we identified significantly higher gene expression for PD-L1 in the stroma, and we also identified increased PD-L1 gene expression in CAFs compared with patient-matched normal colon fibroblasts.

The precise molecular mechanisms underpinning this enhanced PD-L1 expression on stromal cells remain to be elucidated and represent an important avenue of pursuit for the future in our laboratory. Identification of the factors released from tumor cells under conditions of inflammation, and the signaling pathways activated in MSC, may facilitate the development of novel adjuvants for immunotherapy to enhance clinical therapeutic effects. In summary, targeting stromal cell PD-L1 may be key to breaking the cycle of immune evasion and immunosuppression established by the stromal compartment of the tumor microenvironment, leading to more favorable and durable outcomes for patients.

A.E. Ryan reports receiving commercial research support from Janssen Pharmaceuticals and Bristol-Myers Squibb and has ownership interest in U.S. Patent app 15/798,670. No potential conflicts of interest were disclosed by the other authors.

Conception and design: T. Ritter, L.J. Egan, A.E. Ryan

Development of methodology: G. O'Malley, K. Lynch, S.D. Naicker, A.E. Ryan

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G. O'Malley, O. Treacy, K. Lynch, S.D. Naicker, N.A. Leonard, P. Lohan, A.E. Ryan

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G. O'Malley, K. Lynch, S.D. Naicker, P.D. Dunne, T. Ritter, L.J. Egan, A.E. Ryan

Writing, review, and/or revision of the manuscript: G. O'Malley, K. Lynch, T. Ritter, L.J. Egan, A.E. Ryan

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G. O'Malley, O. Treacy, K. Lynch, S.D. Naicker, N.A. Leonard, P.D. Dunne, A.E. Ryan

Study supervision: A.E. Ryan

The authors would like to acknowledge financial support from the Irish Cancer Society (CRF12RYA) and Science Foundation Ireland (15/SIRG/3456) and Galway University Foundation to Dr. A.E. Ryan, a postgraduate scholarship from the Irish Research Council (GOIPG/2013/998) to G O’Malley, and a grant from Science Foundation Ireland (12/IA/1624) to T. Ritter.

The authors thank Professor Matthew Griffin, Professor Rhodri Ceredig, and Dr. Declan McKernan for helpful discussions; Ms. Georgina Shaw, Dr. Joana Cabral, and Ms. Athina Rigalou for technical assistance; and Dr. Shirley Hanley in the NUIG Flow Cytometry core facility for technical expertise.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
2.
Hanahan
D
,
Coussens
LM
. 
Accessories to the crime: functions of cells recruited to the tumor microenvironment
.
Cancer Cell
2012
;
21
:
309
22
.
3.
O'Malley
G
,
Heijltjes
M
,
Houston
AM
,
Rani
S
,
Ritter
T
,
Egan
LJ
, et al
Mesenchymal stromal cells (MSCs) and colorectal cancer - a troublesome twosome for the anti-tumour immune response?
Oncotarget
2016
;
7
:
60752
74
.
4.
Ryan
AE
,
Colleran
A
,
O'Gorman
A
,
O'Flynn
L
,
Pindjacova
J
,
Lohan
P
, et al
Targeting colon cancer cell NF-kappaB promotes an anti-tumour M1-like macrophage phenotype and inhibits peritoneal metastasis
.
Oncogene
2015
;
34
:
1563
74
.
5.
Ryan
AE
,
Shanahan
F
,
O'Connell
J
,
Houston
AM
. 
Fas ligand promotes tumor immune evasion of colon cancer in vivo
.
Cell Cycle
2006
;
5
:
246
9
.
6.
Galon
J
,
Costes
A
,
Sanchez-Cabo
F
,
Kirilovsky
A
,
Mlecnik
B
,
Lagorce-Pages
C
, et al
Type, density, and location of immune cells within human colorectal tumors predict clinical outcome
.
Science
2006
;
313
:
1960
4
.
7.
Koliaraki
V
,
Pasparakis
M
,
Kollias
G
. 
IKKbeta in intestinal mesenchymal cells promotes initiation of colitis-associated cancer
.
J Exp Med
2015
;
212
:
2235
51
.
8.
Greten
FR
,
Eckmann
L
,
Greten
TF
,
Park
JM
,
Li
ZW
,
Egan
LJ
, et al
IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer
.
Cell
2004
;
118
:
285
96
.
9.
Calon
A
,
Lonardo
E
,
Berenguer-Llergo
A
,
Espinet
E
,
Hernando-Momblona
X
,
Iglesias
M
, et al
Stromal gene expression defines poor-prognosis subtypes in colorectal cancer
.
Nat Genet
2015
;
47
:
320
9
.
10.
Dvorak
HF
. 
Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing
.
N Engl J Med
1986
;
315
:
1650
9
.
11.
Terzic
J
,
Grivennikov
S
,
Karin
E
,
Karin
M
. 
Inflammation and colon cancer
.
Gastroenterology
2010
;
138
:
2101
14
e5
.
12.
Luo
JL
,
Maeda
S
,
Hsu
LC
,
Yagita
H
,
Karin
M
. 
Inhibition of NF-kappaB in cancer cells converts inflammation- induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression
.
Cancer Cell
2004
;
6
:
297
305
.
13.
Ren
G
,
Zhao
X
,
Wang
Y
,
Zhang
X
,
Chen
X
,
Xu
C
, et al
CCR2-dependent recruitment of macrophages by tumor-educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFalpha
.
Cell Stem Cell
2012
;
11
:
812
24
.
14.
Shi
Y
,
Du
L
,
Lin
L
,
Wang
Y
. 
Tumour-associated mesenchymal stem/stromal cells: emerging therapeutic targets
.
Nat Rev Drug Discov
2017
;
16
:
35
52
.
15.
Li
Y
,
Liang
L
,
Dai
W
,
Cai
G
,
Xu
Y
,
Li
X
, et al
Prognostic impact of programed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor infiltrating lymphocytes in colorectal cancer
.
Mol Cancer
2016
;
15
:
55
.
16.
Rosenbaum
MW
,
Bledsoe
JR
,
Morales-Oyarvide
V
,
Huynh
TG
,
Mino-Kenudson
M
. 
PD-L1 expression in colorectal cancer is associated with microsatellite instability, BRAF mutation, medullary morphology and cytotoxic tumor-infiltrating lymphocytes
.
Mod Pathol
2016
;
29
:
1104
12
.
17.
FDA
. 
FDA grants nivolumab accelerated approval for MSI-H or dMMR colorectal cancer
. 
2017
[Available from
: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm569366.htm.
18.
Dunne
PD
,
McArt
DG
,
O'Reilly
PG
,
Coleman
HG
,
Allen
WL
,
Loughrey
M
, et al
Immune-derived PD-L1 gene expression defines a subgroup of stage II/III colorectal cancer patients with favorable prognosis who may be harmed by adjuvant chemotherapy
.
Cancer Immunol Res
2016
;
4
:
582
91
.
19.
Ma
W
,
Gilligan
BM
,
Yuan
J
,
Li
T
. 
Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy
.
J Hematol Oncol
2016
;
9
:
47
.
20.
Nishida
N
,
Nagahara
M
,
Sato
T
,
Mimori
K
,
Sudo
T
,
Tanaka
F
, et al
Microarray analysis of colorectal cancer stromal tissue reveals upregulation of two oncogenic miRNA clusters
.
Clin Cancer Res
2012
;
18
:
3054
70
.
21.
Ferrer-Mayorga
G
,
Gomez-Lopez
G
,
Barbachano
A
,
Fernandez-Barral
A
,
Pena
C
,
Pisano
DG
, et al
Vitamin D receptor expression and associated gene signature in tumour stromal fibroblasts predict clinical outcome in colorectal cancer
.
Gut
2017
;
66
:
1449
62
.
22.
Owens
BM
. 
Inflammation, innate immunity, and the intestinal stromal cell niche: opportunities and challenges
.
Front Immunol
2015
;
6
:
319
.
23.
Mishra
PJ
,
Mishra
PJ
,
Humeniuk
R
,
Medina
DJ
,
Alexe
G
,
Mesirov
JP
, et al
Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells
.
Cancer Res
2008
;
68
:
4331
9
.
24.
Spaeth
EL
,
Dembinski
JL
,
Sasser
AK
,
Watson
K
,
Klopp
A
,
Hall
B
, et al
Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression
.
PLoS One
2009
;
4
:
e4992
.
25.
Spaeth
EL
,
Labaff
AM
,
Toole
BP
,
Klopp
A
,
Andreeff
M
,
Marini
FC
. 
Mesenchymal CD44 expression contributes to the acquisition of an activated fibroblast phenotype via TWIST activation in the tumor microenvironment
.
Cancer Res
2013
;
73
:
5347
59
.
26.
Hodge
G
,
Barnawi
J
,
Jurisevic
C
,
Moffat
D
,
Holmes
M
,
Reynolds
PN
, et al
Lung cancer is associated with decreased expression of perforin, granzyme B and interferon (IFN)-gamma by infiltrating lung tissue T cells, natural killer (NK) T-like and NK cells
.
Clin Exp Immunol
2014
;
178
:
79
85
.
27.
Lakins
MA
,
Ghorani
E
,
Munir
H
,
Martins
CP
,
Shields
JD
. 
Cancer-associated fibroblasts induce antigen-specific deletion of CD8 (+) T cells to protect tumour cells
.
Nat Commun
2018
;
9
:
948
.
28.
Chinnadurai
R
,
Copland
IB
,
Patel
SR
,
Galipeau
J
. 
IDO-independent suppression of T cell effector function by IFN-gamma-licensed human mesenchymal stromal cells
.
J Immunol
2014
;
192
:
1491
501
.
29.
Di Nicola
M
,
Carlo-Stella
C
,
Magni
M
,
Milanesi
M
,
Longoni
PD
,
Matteucci
P
, et al
Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli
.
Blood
2002
;
99
:
3838
43
.
30.
Hirosue
S
,
Dubrot
J
. 
Modes of antigen presentation by lymph node stromal cells and their immunological implications
.
Front Immunol
2015
;
6
:
446
.
31.
He
J
,
Hu
Y
,
Hu
M
,
Li
B
. 
Development of PD-1/PD-L1 pathway in tumor immune microenvironment and treatment for non-small cell lung cancer
.
Sci Rep
2015
;
5
:
13110
.
32.
Juneja
VR
,
McGuire
KA
,
Manguso
RT
,
LaFleur
MW
,
Collins
N
,
Haining
WN
, et al
PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity
.
J Exp Med
2017
;
214
:
895
904
.
33.
Igney
FH
,
Krammer
PH
. 
Tumor counterattack: fact or fiction?
Cancer Immunol Immunother
2005
;
54
:
1127
36
.
34.
Latchman
Y
,
Wood
CR
,
Chernova
T
,
Chaudhary
D
,
Borde
M
,
Chernova
I
, et al
PD-L2 is a second ligand for PD-1 and inhibits T cell activation
.
Nat Immunol
2001
;
2
:
261
8
.
35.
Ryan
AE
,
Shanahan
F
,
O'Connell
J
,
Houston
AM
. 
Addressing the "Fas counterattack" controversy: blocking fas ligand expression suppresses tumor immune evasion of colon cancer in vivo
.
Cancer Res
2005
;
65
:
9817
23
.
36.
Ribas
A
,
Hu-Lieskovan
S
. 
What does PD-L1 positive or negative mean?
J Exp Med
2016
;
213
:
2835
40
.
37.
Huang
WH
,
Chang
MC
,
Tsai
KS
,
Hung
MC
,
Chen
HL
,
Hung
SC
. 
Mesenchymal stem cells promote growth and angiogenesis of tumors in mice
.
Oncogene
2013
;
32
:
4343
54
.
38.
Lin
JT
,
Wang
JY
,
Chen
MK
,
Chen
HC
,
Chang
TH
,
Su
BW
, et al
Colon cancer mesenchymal stem cells modulate the tumorigenicity of colon cancer through interleukin 6
.
Exp Cell Res
2013
;
319
:
2216
29
.
39.
Castle
JC
,
Loewer
M
,
Boegel
S
,
de Graaf
J
,
Bender
C
,
Tadmor
AD
, et al
Immunomic, genomic and transcriptomic characterization of CT26 colorectal carcinoma
.
BMC Genomics
2014
;
15
:
190
.
40.
Giordano
M
,
Henin
C
,
Maurizio
J
,
Imbratta
C
,
Bourdely
P
,
Buferne
M
, et al
Molecular profiling of CD8 T cells in autochthonous melanoma identifies Maf as driver of exhaustion
.
EMBO J
2015
;
34
:
2042
58
.
41.
Shinagawa
K
,
Kitadai
Y
,
Tanaka
M
,
Sumida
T
,
Kodama
M
,
Higashi
Y
, et al
Mesenchymal stem cells enhance growth and metastasis of colon cancer
.
Int J Cancer
2010
;
127
:
2323
33
.
42.
De Boeck
A
,
Pauwels
P
,
Hensen
K
,
Rummens
JL
,
Westbroek
W
,
Hendrix
A
, et al
Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression through paracrine neuregulin 1/HER3 signalling
.
Gut
2013
;
62
:
550
60
.
43.
Riihimaki
M
,
Hemminki
A
,
Sundquist
J
,
Hemminki
K
. 
Patterns of metastasis in colon and rectal cancer
.
Sci Rep
2016
;
6
:
29765
.
44.
Selby
MJ
,
Engelhardt
JJ
,
Johnston
RJ
,
Lu
LS
,
Han
M
,
Thudium
K
, et al
Preclinical development of ipilimumab and nivolumab combination immunotherapy: mouse tumor models, in vitro functional studies, and cynomolgus macaque toxicology
.
PLoS One
2016
;
11
:
e0161779
.
45.
Duraiswamy
J
,
Kaluza
KM
,
Freeman
GJ
,
Coukos
G
. 
Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors
.
Cancer Res
2013
;
73
:
3591
603
.
46.
Dunican
DS
,
McWilliam
P
,
Tighe
O
,
Parle-McDermott
A
,
Croke
DT
. 
Gene expression differences between the microsatellite instability (MIN) and chromosomal instability (CIN) phenotypes in colorectal cancer revealed by high-density cDNA array hybridization
.
Oncogene
2002
;
21
:
3253
7
.
47.
Turley
SJ
,
Cremasco
V
,
Astarita
JL
. 
Immunological hallmarks of stromal cells in the tumour microenvironment
.
Nat Rev Immunol
2015
;
15
:
669
82
.
48.
Han
Z
,
Tian
Z
,
Lv
G
,
Zhang
L
,
Jiang
G
,
Sun
K
, et al
Immunosuppressive effect of bone marrow-derived mesenchymal stem cells in inflammatory microenvironment favours the growth of B16 melanoma cells
.
J Cell Mol Med
2011
;
15
:
2343
52
.
49.
Ben-Neriah
Y
,
Karin
M
. 
Inflammation meets cancer, with NF-kappaB as the matchmaker
.
Nat Immunol
2011
;
12
:
715
23
.
50.
Karin
M
. 
NF-kappaB as a critical link between inflammation and cancer
.
Cold Spring Harb Perspect Biol
2009
;
1
:
a000141
.
51.
Prizment
AE
,
Vierkant
RA
,
Smyrk
TC
,
Tillmans
LS
,
Nelson
HH
,
Lynch
CF
, et al
Cytotoxic T-cells and granzyme B associated with improved colorectal cancer survival in a prospective cohort of older women
.
Cancer Epidemiol Biomarkers Prev
2017
;
26
:
622
31
.
52.
Llosa
NJ
,
Cruise
M
,
Tam
A
,
Wicks
EC
,
Hechenbleikner
EM
,
Taube
JM
, et al
The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints
.
Cancer Discov
2015
;
5
:
43
51
.
53.
Taube
JM
,
Klein
A
,
Brahmer
JR
,
Xu
H
,
Pan
X
,
Kim
JH
, et al
Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy
.
Clin Cancer Res
2014
;
20
:
5064
74
.
54.
Huang
K
,
Wu
J
. 
Targeting the PD-1 pathway in MSI-stable metastatic colorectal cancer
.
Chemo Open Access
2017
;
6
. e132.
55.
Ebert
PJ
,
Cheung
J
,
Yang
Y
,
McNamara
E
,
Hong
R
,
Moskalenko
M
, et al
MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade
.
Immunity
2016
;
44
:
609
21
.