Abstract
Tumor antigen-specific T cells rapidly lose energy and effector function in tumors. The cellular mechanisms by which energy loss and inhibition of effector function occur in tumor infiltrating lymphocytes (TILs) are ill-defined, and methods to identify tumor-antigen-specific TILs that experience such stress are unknown. Processes upstream of the mitochondria guide cell-intrinsic energy depletion. We hypothesized that a mechanism of T cell-intrinsic energy consumption was the process of oxidative protein folding and disulfide-bond formation that takes place in the endoplasmic reticulum (ER) guided by protein kinase R-like endoplasmic reticulum kinase (PERK) and downstream PERK axis target ER oxidoreductase 1 (ERO1α. To test this hypothesis, we created TCR transgenic mice with a T cell-specific PERK gene deletion (OT1+Lckcre+PERKf/f, PERK KO). We found that PERK KO and T cells that were pharmacologically inhibited by PERK or ERO1α maintained reserve energy and exhibited a protein profile consistent with reduced oxidative stress. These T cell groups displayed superior tumor control compared to T effectors. We identified a biomarker of ER-induced mitochondrial exhaustion in T cells as mitochondrial reactive oxygen species (mtROS), and found that PD-1+ tumor antigen-specific CD8+ TILs express mtROS. In vivo treatment with a PERK inhibitor abrogated mtROS in PD-1+ CD8+ TILs and bolstered CD8+ TIL viability. Combination therapy enabled 100% survival and 71% tumor clearance in a sarcoma mouse model. Our data identify the ER as a regulator of T cell energetics and indicate that ER elements are effective targets to improve cancer immunotherapy.
Keywords: T cell, PERK, ER Stress, Metabolism, PD-1
INTRODUCTION
In response to antigen, T cells undergo rapid expansion, engaging in up to 15-20 cell divisions, and proliferation is characterized by differentiation to an effector phenotype from a naïve state (1). To support the bioenergetic demands of growth and proliferation, naïve T cells that primarily engage oxidative phosphorylation (OXPHOS) shift to generate adenosine triphosphate (ATP) via increased OXPHOS and aerobic glycolysis as they become functional effector cells (2,3). Highly differentiated effector T cells rely primarily on glycolysis and are characterized by a loss of mitochondrial integrity (4) that likely accounts for their inability to effectively continue OXPHOS. The cell-intrinsic mechanisms that consume metabolic energy and impart mitochondrial exhaustion in T cells are unknown.
Programmed cell death protein 1 (PD-1) is expressed on highly differentiated effector T cells that have become exhausted due to chronic antigen exposure (5). In cancers, programmed death-ligand 1 (PD-L1) is expressed by tumor cells to further impair anti-tumor effector function of PD-1+ T cells (6). Checkpoint blockade therapy is a means to reinvigorate effector function of exhausted T cells by inhibition of the PD-1-PD-L1 interaction (7). Unfortunately, anti-PD-1 therapy remains ineffective for the majority of cancer patients due to initial lack of response or loss of durable responses (8-10). In early-stage exhaustion, PD-1+ T cells show diminished OXPHOS and glycolysis, and long-term chronically exhausted PD-1high T cells exhibit a dependence on glycolysis due to dysfunctional mitochondria (11). T cell-intrinsic factors that drive PD-1+ CD8+ tumor infiltrating lymphocyte (TIL) metabolic exhaustion in response to tumor antigen are unknown, and simple methods to characterize the metabolic state of PD-1+ tumor infiltrating lymphocytes (TILs) in patient tumors are ill-defined.
T cell expansion initiated by antigen recognition requires T effector cells to greatly increase new protein synthesis and initiate post-translational modifications (12). Imbalances of unfolded and misfolded proteins are detected by endoplasmic reticulum (ER) stress sensors inositol-requiring enzyme-1 (IRE1α), protein kinase R-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6) (13). The acute arm of the stress response aims to alleviate the burden of unfolded or misfolded proteins in the ER to restore proteostasis. In the face of insurmountable stress, the unfolded protein response (UPR) initiates cell death. PERK mediates the terminal UPR through regulation of transcription factors activating transcription factor 4 (ATF4) and C/EBPα homologous protein (CHOP) which induce downstream target ER oxidoreductase 1 (ERO1α) (14-17). As a catalyst of disulfide bond formation in the ER, ERO1α facilitates oxidation-reduction reactions (Redox). Excess ERO1α produced in response to chronic protein folding induces accumulation of reactive oxygen species (ROS) and impairs survival (15,16). The PERK-specific cell stress response has not been measured in T cells or applied to the rubric of anti-tumor immunity.
Here we report that the PERK axis contributes to ERO1α activation in T effector cells. We find that this axis consumes metabolic energy in T cells and drives a protein profile synonymous with oxidative stress. We show that accumulation of mitochondrial ROS (mtROS) is a hallmark of ER-induced mitochondrial exhaustion, connecting ER biology to metabolic function in T cells. Memory T cells show reduced expression of mtROS and ERO1α compared to T effectors. PERK KO, and PERK, or ERO1α inhibitor-treated T cells exhibited superior tumor control compared to T effectors. Tumor antigen-specific PD-1+ CD8+ TILs acquire mtROS in tumors. In sarcoma patients and a sarcoma mouse model, we find that high proportions of PD-1+ CD8+ TILs express mtROS. In vivo treatment with a PERK inhibitor reduced mtROS in PD-1+ CD8+ TILs, increased tumor clearance, and extended animal survival in combination with anti-PD-1 therapy.
MATERIALS & METHODS
Mice
T cell-specific deletion of PERK on a C57BL/6J background was accomplished by cross of PERKloxP (Eif2ak3tm1.2Drc/J) mice with OT-1 (C57BL/6-Tg(TcraTcrb)1100Mjb/J) or Lck-Cre (B6.Cg-Tg(Lck-icre)3779Nik/J) mice to obtain OT-1xPERKf/f and LckCrexPERKf/w mice, respectively. These lines were crossed to obtain OT1+Lckcre+PERKf/f mice and OT1+Lckcre−PERKf/f littermate controls. Lck-Cre, PERKloxP, OT-1, C57BL/6J, Ly5.2 (B6.SJL-Ptprca Pepcb/BoyJ), and P-mel (B6.CgThy1a/CyTg(TcraTcrb)8Rest/J) mice were obtained from the Jackson Laboratory. All animal experiments were approved by the Medical University of South Carolina (MUSC) Institutional Animal Care and Use Committee and the Division of Laboratory Animal Resources at MUSC maintained all mice.
Human Samples
This work was determined by MUSC Institutional Review Board to be exempt under protocols Pro00055941 & Pro00055960. Normal donor patients undergoing routine non–cancer-associated surgery (Pro00055941) or patients undergoing surgical removal of high grade deep pleomorphic undifferentiated sarcomas (Pro00055960) granted consent under MUSC Biorepository surgical consent forms. Tissue samples were de-identified. Studies were conducted in accordance with the Declaration of Helsinki, International Ethical Guidelines for Biomedical Research Involving Human Subjects (CIOMS), Belmont Report, or U.S. Common Rule. Blood (8 mL) was collected in EDTA coated tubes and PBMC were isolated via Histopaque-1077 centrifugation (Sigma). Sarcoma tissue was collected on ice and immediately cut into 2mm3 pieces and dissociated to a single cell suspension using Human Tumor Dissociation Kit and gentleMACS dissociator (Miltenyi Biotec) according to manufacturer’s protocol.
RT-PCR and Immunoblot analysis
RNA was isolated with RNeasy Mini Kit (QIAGEN) and single-strand cDNA was made with High Capacity RNA-to-cDNA Kit (Applied Biosystems, Thermo Fisher Scientific). Taqman gene expression assays (Applied Biosystems, Thermo Fisher) were used to perform real-time PCR using the StepOnePlus Real-Time PCR system (Applied Biosystems, Thermo Fisher). Gene expression for PERK (Eif2ak3), ATF4 (Atf4), CHOP (Ddit3), and ERO1α (Ero1) were normalized to Gapdh. For immunoblots cell lysates were prepared in RIPA buffer (Sigma-Aldrich) and probed with PERK (Cell Signaling Technology), ERO1α (Santa Cruz Biotechnology) antibodies or β-actin (Cell Signaling Technology) loading control.
Cell Sort, FACS Staining & Analysis
Fluorochrome-conjugated monoclonal antibodies and respective isotype controls listed in Supplementary Table S1 were purchased from ThermoFisher (Ebioscience). Extracellular stains were performed in PBS supplemented with 2% FBS. IFN-γ was probed after 4-6 hours of cell restimulation with Cell Stimulation Cocktail (eBioscience) and Golgi Plug Protein Transport Inhibitor (eBioscience). Foxp3/Transcription Factor Intracellular Staining Buffer Set was used according to manufacturer’s protocol (eBioscience). Mitochondrial reactive oxygen species (mtROS) were measured with MitoSOX Red Mitochondrial Superoxide Indicator (3μM) loaded at 37°C for 30 minutes in RT PBS. Extracellular stains were added post dye incubation. For Annexin staining, MitoSOX dye loaded cells were washed and stained for using Annexin V-FITC Apoptosis Detection Kit (eBioscience) according to manufacturers’ protocol. Samples were run directly on a BD Accuri C6 flow cytometer. For FACS sorting of mtROS+ and mtROS− cell fractions, samples were stained with MitoSOX Red Mitochondrial Superoxide Indicator and CD8 and sorted on a FACS Aria IIu (BD Biosciences). Further analysis was performed post-sort for confirmation of population purity. For RNA ex vivo gene analysis, lymphocytes were isolated via Histopaque gradient (Sigma) and CD8+ T cell Isolation Kit (Miltenyi) was used to obtain >90% purity from spleens and tumors.
Cell lines
MCA-205-OVA (Gift of William Redmond) and MC38 (Kerafast Inc.) cell lines were maintained in DMEM 10% FBS. B16F1-OVA and B16F10 (Gifts of Mark Rubinstein) were maintained in RPMI complete T cell media. Cell lines were last determined to be mycoplasma free in March 2017 (MCA-205-OVA), January 2018 (MC38), 2012 (B16F1-OVA, B16F10). Cell lines were not authenticated in the past year. All growth media were supplemented with Plasmocin mycoplasma prophylactic (Invivogen). All cell lines were passaged three times prior to in vivo tumor inoculation, and a fresh vial of cells was prepared for individual experiments.
T cell activation and treatment
Spleens from OT-1 mice were dissociated and RBC lysis was performed using ACK Lysing (Thermo-Fisher Scientific) to achieve a single cell suspension. T cells were activated and expanded for indicated time points from total splenocytes incubated with 1μg/mL OVA 257-264 (Invivogen) or hgp100 25-33 (GenScript) peptide in complete T cell media (200U rhIL-2, NCI). Cells were washed and media was changed after 3 days of initial activation and expansion. For inhibitor treatments, PERK (100nM, GSK2606414, Tocris) or ERO1α inhibitors (10μM, EN460, Cayman Chemical) were incubated with splenocytes for 10 min prior to introduction of peptide and re-introduced at the d3 cell split. For central memory T cell development, IL-2 was replaced in T cell media with rhIL-15 (50ng/mL, Shenandoah) at the day 3 cell split.
Metabolic & Proteomic Analysis
Oxygen consumption rate (OCR) was measured in non-buffered RS media supplemented with HEPES under basal conditions and in response to 1μM oligomycin, 1.5 μM FCCP, and 2μM rotenone + 1μM Antimycin A using the XFe96 Extracellular Flux Analyzer (Seahorse Bioscience). Cell-Tak (Corning) was used for T-cell adherence.
Liquid Chromatography-Tandem Mass Spectrometry
Proteomics were performed by Bioproximity, LLC. Samples were prepared for digestion using the suspension-trapping (S-trap, Protifi) method. Digested peptides were collected by centrifugation. Peptides were eluted with 80% acetonitrile, 5% ammonium hydroxide and lyophilized in a SpeedVac (Thermo Savant) to remove volatile components. Digestion mixtures were analyzed by UHPLC-MS/MS. LC was performed on an Easy-nLC 1000 UHPLC system (Thermo) interfaced to a quadrupole-Orbitrap mass spectrometer (Q-Exactive HF-X, Thermo Fisher) via nano-electrospray ionization using a source with an integrated column heater (Thermo Easy Spray source).
Data Processing and Library Searching
Tandem mass spectra were searched using X!Tandem and Open Mass Spectrometry Search Algorithm (OMSSA), requiring expectation value scores of 0.01 or better to be considered a match. Protein intensity values were calculated using OpenMS to measure the area under the curve of identified peptides. Searches were performed on Amazon Web Services-based cluster compute instances using the Proteome Cluster interface which builds species- and genus-specific protein sequence libraries monthly from current UniProtKB distributions. Gene annotations were obtained from Ensembl Release 93 database, Mouse genes (v93GRCm38.p6) data set through the BioMart website. Official Gene Ontology categories to annotate mouse proteins based on corresponding gene symbols were used.
Tumor mouse models and In vivo treatments
For transfer of 7-day expanded OT-1+ or PERK KO T cells, C57BL/6 mice were implanted subcutaneously (s.c.) with 2.5×105 B16F1-OVA tumor cells and 5×105 T cells were infused via tail vein to 5Gy irradiated mice after 7 days of tumor growth. For transfer of 7-day expanded Pmel or Pmel inhibitor-treated T cells, C57BL/6 mice were implanted subcutaneously (s.c.) with 2.5×105 B16F10 tumor cells and 2×106 T cells were infused via tail vein to 5Gy irradiated mice after 7 days of tumor growth. Tumor growth was measured every other day for 3 weeks. For tracking acquisition of mtROS/PD-1+ status in tumor antigen-specific CD8+ TILs, 2.5×105 MCA-205-OVA were implanted s.c. to Ly5.2 mice. Naïve OT-1+ T cells were obtained via Mouse CD8+ T Cell Isolation Kit (Miltenyi Biotec), and 1×106 CD8+ T cells were transferred via tail vein on day 7 of tumor growth. After 7 days of in vivo expansion, tumor draining lymph nodes (TDLNs) and tumors were harvested. Tumors were processed to single cell suspension using Mouse Tumor Dissociation Kit (Miltenyi Biotec) according to manufacturer’s protocol. For combination therapy experiments, 2.5×105 MCA-205-OVA were injected s.c. to C57BL/6 mice and tumors were established for 7 days. 50mg/kg PERK Inhibitor (GSK2606414, GlaxoSmithKline) or vehicle was administered twice daily via oral gavage as a suspension of 0.5% hydroxypropylmethyl cellulose + 0.1% Tween-80 in water. 200μg Anti-PD-1 (RMP1-14) or RatIgG2a isotype control (2A3) (Bio X Cell) was administered every 4 days after 12 days of initial tumor growth. For CD8α cell depletion, 100μg Anti-CD8α (53-6.7, Bio X Cell) was administered three times per week after 5 days of tumor growth.
RESULTS
PERK contributes to activation of chronic ER stress in T effector cells
Previously we found that ER activation contributed to mitochondrial stimulation in CD4+ T cells through inositol 1,4,5-triphosphate receptor (IP(3)R)-mediated Ca2+ signaling. Inhibition of IP(3)R shifted T cell fate and promoted cell persistence in tumors (18). Continuous disruptions in Ca2+ homeostasis impair proper protein folding and a high burden of misfolded proteins induces chronic stress on the ER. In the face of irresolvable stress, PERK integrates signals from Ca2+ binding protein BiP/grp78 to initiate the terminal UPR through activation of transcription factors ATF4 and CHOP. A key downstream target of ATF4/CHOP is ERO1α. As a facilitator of Redox reactions, excessive ERO1α activity can induce oxidative stress and impair cell survival as cells lose capability to scavenge ROS through anti-oxidant systems (17)(19,20). We asked how the ER stress sensor PERK and the terminal UPR were regulated in differentiating T effector cells. We found that gene expression of PERK (Eif2ak3), ATF4 (Atf4), CHOP (Ddit3), and ERO1α (Eor1l) were significantly increased over the course of T cell activation and differentiation (Fig. 1A-D). PERK protein content increased in T cells and peaked after 5 days of expansion. ERO1α protein increased abundance as T cells differentiated (Fig. 1E). PERK gene expression peaked 7 days post activation, but protein expression was reduced after the 5-day time point. Our data point out a discrepancy between gene and protein regulation of PERK in T cells and indicate that further study of how PERK is post-transcriptionally regulated is needed.
To measure the role of PERK in induction of the terminal UPR in T cells, we created T cell receptor (TCR) transgenic T cell-specific conditional knock out mice (OT1+Lckcre+PERKf/f, PERK KO). We measured genes associated with the chronic PERK response; Atf4, Ddit3, and Ero1l in naïve and 7-day expanded WT and PERK KO T cells. As expected, Eif2ak3, Atf4, Ddit3, and Ero1l were significantly reduced in PERK KO T cells (Figs. 1F-I). In line with these data, PERK and ERO1α proteins were decreased in PERK KO T cells (Fig. 1J). A longer exposure for PERK was performed to convey absence of PERK protein. These data reveal a role for chronic ER stress as a component of differentiating T effector cells.
PERK axis consumes energy in CD8+ T effector cells
Increased spare respiratory capacity (SRC) is a property associated with superior anti-tumor function of T cells (21). The molecular processes that consume T cell metabolic energy are not well understood. Redox reactions for protein folding and disulfide bond formation in the ER are energy consumptive processes that integrate ER activation with mitochondrial bioenergetics (17,19,22). We measured oxygen consumption rates (OCR) in WT and PERK KO T cells and found that PERK KO T cells maintained superior SRC compared to WT cells. To assess a potential contribution of ERO1α to this phenomenon, we treated T cells with a target-specific PERK (PERK I) (23) or ERO1α inhibitor (ERO1 I) (24) across the course of differentiation and expansion. We found that PERK I, or ERO1 I-treated T cells preserved metabolic energy compared to WT controls (Fig. 2A-C). Loss of SRC is associated with a reduction in T cell effector function (25). We measured capacity to secrete IFN-γ in the abovementioned T cell groups, and found that PERK KO, PERK I, or ERO1 I T cells had increased IFN-γ secretion compared to WT controls (Fig. 2D). Inhibitor treatments did not significantly reduce cell numbers and experimental T cell groups exhibited increased viability compared to respective WT controls (Supplementary Fig. S1A-B).
Activation of chronic PERK UPR mediated by ATF4/CHOP-ERO1α signaling has previously been implicated to drive protein synthesis, consume cellular energy, and contribute to death through oxidative cell stress (16,17,19). To gain insight into how PERK and downstream target ERO1α shape T effector cells, we assayed the proteomes of T effector, PERK KO, and ERO1α I-treated T cells. Shotgun liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to assess the top 100 proteins with greatest intensity expression in T effectors compared to PERK KO or ERO1α I-treated T cells (Supplementary Table S2). User-defined categories were used to qualify proteins into biologically functional groups. Proteomics revealed protein groups associated with energy and metabolism, redox reactions and oxidative stress/ DNA damage with increased expression in T effectors compared to PERK KO and ERO1α I-treated T cell groups (Fig. 2E-F). Data are represented as fold increased expression values in T effectors compared to PERK KO or ERO1α I-treated T cell groups. Of user-defined categories the greatest overlap in PERK KO and ERO1α-treated T cells was in the “ER transport/ cell stress” group (n=6 proteins). 3/7 of redox-associated proteins overlapped as highly expressed in T effectors compared to both PERK KO and ERO1α I-treated T cells, highlighting lactate dehydrogenases, Thioredoxins, and Glutathione-S-transferases as protein families of potential relevance. Further analysis to validate these protein pathways to elucidate the role of Redox and oxidative stress in T cells is warranted.
Mitochondrial ROS are a hallmark of mitochondrial exhaustion in T cells
Persistent ERO1α activation is bioenergetically costly to cells and OXPHOS may be stimulated in efforts to replenish depleted ATP stores (19). We wished to establish a biomarker of energy loss and aging in T cells that may be associated with chronic stress on the ER consistent with pathogenic ERO1α activation. We reasoned that persistent mitochondrial activation through continued OXPHOS would result in accumulation of mitochondrial reactive oxygen species (mtROS) in T cells. The live cell dye MitoSOX Red detects the superoxide anion, a precursor to the majority of cellular ROS, in mitochondrial membranes and is detectable by flow cytometry (26). Under normal physiological conditions, detoxification of superoxide occurs through superoxide dismutase-mediated conversion to hydrogen peroxide (27). However, excess ROS is associated with disease and aging (28). We measured mtROS in OT-1+ T cells over the course of T effector cell differentiation. As naïve T cells lost CD62L expression, a measure of stemness, mtROS accumulated in T cell mitochondria (Fig. 3A). Human CD8+ T cells underwent the same pattern over the course of 3 weeks of in vitro expansion (Fig. 3B).
To determine whether mtROS accumulation was a hallmark of energy loss in T cells, we FACS sorted day 3 and day 5 mtROS− and mtROS+ CD8+ T cell subsets. The lowest 25% mtROS− and highest 25% mtROS+ cells were obtained (Fig. 3C) and SRC among groups was measured. Accumulation of mtROS indicated energy loss in T cells (Fig. 3D). We next measured the capacity for IFN-γ secretion among day 5 and 7 expanded mtROS− and mtROS+ T cell groups. We found that IFN-γ secretion underwent a similar pattern to SRC among sorted T cell populations (Fig. 3E). In day 7 cells, we asked whether mtROS+ may indicate early stage apoptosis. We found that ~8% of CD8+ mtROShigh T cells harvested on day 7 were in early stage apoptosis (Fig. 3F). This was only detectable in day 7 samples, not in cells harvested at earlier time points. Our data show that mtROS can be used as a hallmark of metabolic energy loss and aging in T cells.
PERK axis contributes to mitochondrial exhaustion of T effector cells
Through ERO1α activation, oxidative protein folding in the stressed ER has been linked to generation of cellular ROS and inhibition of cell survival (19). However, the specific effects of the PERK axis on mitochondrial activation have not been assessed. Given the maintenance of SRC and diminished oxidative stress-associated proteomic signature in PERK KO and ERO1α I T cells (Fig. 2), we reasoned that these T cell groups may show reduced mitochondrial activation and low mtROS accumulation. We found that mtROS was significantly diminished in PERK I, ERO1 I, and PERK KO T cells compared to T effector controls (Fig. 4A). Indeed, in the abovementioned T cell groups, maintained expression of CD62L was a mark of reduced cell aging (Fig. 4B).
Reacquisition (29) or continued expression of CD62L has been associated with memory T cell formation (25,30). We developed memory T cells in vitro with IL-15 cytokine conditioning and found that memory T cells expressed reduced mtROS compared to T effector controls (Fig. 4C). These data are consistent with maintenance SRC of memory cells (25). We asked if memory T cells experience reduced activation of the chronic PERK axis. We found that Atf4, Ddit3, and ERO1l genes were reduced in memory T cells compared to T effectors (Fig. 4D). As expected, ERO1α protein expression was increased in T effectors, and ERO1α protein was not highly expressed in memory T cells. Gene expression of Eif2ak3 was not significantly different between T effector and T memory cells, but protein expression of PERK was reduced (Fig. 4D-E). These data, similar to data obtained in Fig. 1, may suggest post-transcriptional regulation of PERK in T cells. Together, the data indicate that ER programs may impact T cell lineage fate.
PERK and ERO1α hinder T cell-mediated tumor control
Memory T cells promote durable anti-tumor responses (30). We used adoptive T cell therapy mouse models to measure how PERK and ERO1α impact T cell-specific tumor control. Mice bearing B16F1-OVA-expressing tumors were left untreated or treated with OT-1 (T eff) or PERK KO T cells and tumor control was measured every other day for 3 weeks. OT-1-PERK KO T cells exhibited significantly greater tumor control compared to T effectors (Fig. 5A). Survival was significantly extended in tumor-bearing mice treated with PERK KO T cells (Fig. 5B). Although PERK can be acutely protective, the PERK axis can also impair T cell-mediated anti-tumor immunity. To further test how the PERK axis contributes to T cell-specific tumor control, P-mel T cells-conditioned with PERK I or ERO1α I were transferred to mice bearing pathogenic B16F10 melanomas and tumor growth was monitored for 3 weeks. Both inhibitor treatments of T cells yielded significant increases in tumor control compared to T effectors (Fig. 5C-D).
Tumor antigen-specific PD-1+ CD8+ TILs exhibit mitochondrial exhaustion
We next asked how the terminal PERK UPR is expressed among endogenous CD8+ TILs. CD8+ cells were sorted from spleens and tumors of mice bearing immunogenic MCA-OVA sarcomas and gene expression was measured between splenocytes and TILs. The chronic PERK signaling axis was significantly upregulated in CD8+ TILs compared to splenic-matched controls (Fig. 6A). We next asked whether mtROS accumulated in CD8+ TILs cells in mice bearing immunogenic tumors. In multiple tumor models, CD8+ TILs accrued mtROS compared to T cells in tumor draining lymph nodes (TDLNs). To expand this finding, we asked whether mtROS was a property of exhausted T cells in tumors. We measured co-expression of mtROS and PD-1 in CD8+ T cells from TDLNs and tumors of mice bearing MCA-205-OVA sarcomas or MC-38 colon carcinomas. In both mouse models mtROS+ cells were localized to the PD-1+ compartment and mtROS expression was significantly increased in TILs compared to TDLNs (Fig. 6B-C). In agreement with gene expression data, splenocytes from MCA-205-OVA tumor-bearing mice also expressed reduced mtROS/PD-1+ CD8+ T cells compared to TILs (Supplementary Fig. S2A).
PD-1+ TILs comprise a pool of T cells enriched for tumor antigen specificity (31) and tumor-specific T cells home to and proliferate in tumors within 7 days of adoptive transfer (32). We reasoned that metabolically exhausted TILs would be in the tumor antigen-specific T cell compartment. To address this hypothesis, we transferred naïve OT-1+ T cells to mice bearing 7 day-established MCA-205-OVA sarcomas and measured mtROS/PD-1+ co-expression among endogenous (CD45.1) and transferred (CD45.2) T cells recovered from tumors after 7 days of expansion (Fig. 6D). ~70% of transferred CD45.2 CD8+ TILs co-expressed mtROS/PD-1 compared to ~20-30% in the endogenous polyclonal pool of TILs, and this expression could be visualized in TDLNs (Fig. 6E). Our data agree with reports that PD-1+ T cells experience mitochondrial depolarization (33), and shed light on the molecular events within T cells that contribute to mitochondrial exhaustion.
PERK inhibition improves anti-PD-1 therapy
We wished to determine whether mtROS+ CD8+ TILs could be identified in patients. Bone and soft tissue sarcomas are immunogenic human tumor types, but have shown poor responsiveness to anti-PD-1 and combination checkpoint blockade therapies (34). We obtained tumor tissue from patients undergoing routine surgical removal of high grade deep pleomorphic undifferentiated (HGD PU) sarcomas. These patients had not received radiation and had not undergone recent chemotherapy. Dissociation and analysis of CD8+ TILs from fresh HGD PU sarcomas showed a CD8+ T cell infiltrate in all tumors and FACS analysis of PBMC versus autologous TILs showed a significant increase in mtROS/PD-1+ CD8+ T cells in tumors (Fig. 7A). Our data show that PERK inhibition overcomes mitochondrial exhaustion in T cells in vitro (Fig. 4). MCA-205-OVA tumor-bearing mice were treated with PERK I for 7 days during the time period when T cells become functionally exhausted. Day 14 harvest of TILs showed that PERK I-treated PD-1+ CD8+ TILs expressed significantly reduced mtROS compared to vehicle treated controls (Fig. 7B). In line with these data, PERK I treated mice exhibited increased absolute numbers of CD8+ T cells in tumors (Fig. 7C). mtROS+ TILs comprise the pool of highly activated TILs measured by CD44 (Supplementary Fig. S2B). A reduction in mtROS+ TILs was concordant with reduced CD44+ expression among PERK I-treated TILs in both MCA-205-OVA and MC-38 tumor models (Supplementary Fig. S2C). These data suggest that PERK I effects on CD8+ TILs were not due to increased activation associated with reduced formation of immunosuppressive TIL populations or reduced tumor growth. In vivo PERK I treatment augmented anti-PD-1 therapy as 5/7 combination therapy treated mice achieved complete response compared to 2/7 mice treated with anti-PD-1 therapy alone. Depletions of CD8+ cells over the course of therapy demonstrated that CD8+ cells were necessary for combination therapy effect (Fig. 7D-E). Combination therapy-treated mice exhibited 100% survival compared to ~28% survival in the anti-PD-1 therapy condition (Fig. 7F). Together, our data indicate that targeting ER stress in combination with traditional immunotherapy may improve responses in patients.
DISCUSSION
Works by several groups have brought the ER to the forefront of programming pro- and anti-tumor immunity (35). ROS in the tumor microenvironment drive MDSC cell death through an ER stress-mediated mechanism. The shortened lifespan of MDSCs promotes their continued expansion in bone marrow (36). In multiple human tumor types, MDSCs in peripheral blood directly correlate with low overall survival rates in patients (37). Similarly, the tumor microenvironment appears to impair tumor-associated dendritic cell (tDC) antigen-presentation, and antigen presentation is enhanced by impairing the IRE1α-stress pathway in tDCs (38). Lastly, the role of ER stress, and in particular PERK, in augmenting tumor cell development is well defined (39). Together, these data indicate that targeting ER stress pathways in cancer patients may be a formidable strategy to promote anti-tumor immune function and impair immune suppression and tumor cell growth.
Few reports have focused on the role of ER stress in control of T cell anti-tumor immunity, and little attention has been paid to the role of the ER in defining T cell biology. Our previous work found that inhibition of ER activation improved T cell mitochondrial function and aided anti-tumor immunity (18). Our proteomic screen yielded increased expression of multiple proteins associated with ER-mitochondrial crosstalk enriched in T effectors compared to PERK KO T cells. A second report found that inhibition of IRE1α stress signaling led to memory T cell development in response to acute infection (40). These data agree with our data presented here that uncover a role for ER stress signaling a response that affects T cell differentiation and effector cell maturation.
Modulation of ER stress elements alone was able to impact mitochondrial bioenergetics and enhance T cell-specific tumor control. Mechanistically, acute PERK-specific ER stress is protective to cells through momentary attenuation of protein synthesis mediated by phosphorylation of eukaryotic translation initiation factor 2A (EIF2α) (13). The chronic PERK axis drives protein synthesis, energy consumption, oxidative cell stress and death through activation of transcription factors ATF4 and CHOP (16,17). Though PERK gene and protein expression were reduced in our PERK KO T cells, we do not discount the possibility of remnant PERK expression that may have conferred the protective effect of the acute PERK response combined with reduced lethal effects from the terminal UPR. Based on our findings, T cells present a paradigm in which to study the effect of biologically relevant chronic ER stress on cell development and death. Our data show that activation of ERO1α in rapidly dividing T effectors/TILs governs a cell-intrinsic mechanism that drives energy loss and oxidative stress. How the PERK-mediated transcription factors control expression of ERO1α in T cells remains unknown. This question needs to be addressed due to potential implications for cancer immunotherapy (41,42).
Based on the durability of anti-tumor immunity associated with memory T cells in vivo (4,21,25,43), we note that modulation of the PERK pathway could promote in vivo T cell durability and contribute to long-term efficacy of anti-PD-1 therapy. Multiple potential benefits of targeting chronic ER stress have been identified in modulation of pro and anti-tumor immunity (35). Development of novel ER stress axis inhibitors that may circumvent previous toxicities associated with inhibitors is on the horizon (44-48). Future research that defines the role of ER stress in T cell biology is needed. Work that measures how tumor microenvironments drive cell stress signaling that impacts T cell anti-tumor function is essential.
Supplementary Material
Acknowledgements
We are thankful to Gyda Beeson and Dr. Craig C. Beeson for support with metabolic assays and to Dr. Jeffrey Axten for insight and thoughtful commentary on the manuscript. We thank Dr. B. Arman Aksoy for qualification of protein groups. We are grateful to Drs. Zihai Li, Chrystal Paulos, Michael Lilly, and Gustavo Leone for mentorship within the Hollings Cancer Center. Funding sources for this work were from NCI K12 CA157688, ACS IRG-97-219-14, and ACS IRG-16-185-17 to J.E. Thaxton.
Financial Support: K12 CA157688 was granted via HHS ∣ NIH ∣ National Cancer Institute (NCI) to L. Michael and ACS IRG-97-219-14 and ACS IRG-16-185-17 to J. E. Thaxton.
Footnotes
Conflicts of interest: The authors declare no potential conflicts of interest
REFERENCES
- 1.Williams MA, Bevan MJ. Effector and memory CTL differentiation. Annual review of immunology 2007;25:171–92 doi 10.1146/annurev.immunol.25.022106.141548. [DOI] [PubMed] [Google Scholar]
- 2.Macintyre AN, Rathmell JC. Activated lymphocytes as a metabolic model for carcinogenesis. Cancer & metabolism 2013;1(1):5 doi 10.1186/2049-3002-1-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Buck MD, O’Sullivan D, Pearce EL. T cell metabolism drives immunity. The Journal of experimental medicine 2015;212(9):1345–60 doi 10.1084/jem.20151159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Buck MD, O’Sullivan D, Klein Geltink RI, Curtis JD, Chang CH, Sanin DE, et al. Mitochondrial Dynamics Controls T Cell Fate through Metabolic Programming. Cell 2016;166(1):63–76 doi 10.1016/j.cell.2016.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wherry EJ. T cell exhaustion. Nature immunology 2011;12(6):492–9. [DOI] [PubMed] [Google Scholar]
- 6.Chamoto K, Chowdhury PS, Kumar A, Sonomura K, Matsuda F, Fagarasan S, et al. Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proceedings of the National Academy of Sciences of the United States of America 2017;114(5):E761–e70 doi 10.1073/pnas.1620433114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer cell 2015;27(4):450–61 doi 10.1016/j.ccell.2015.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Munhoz RR, Postow MA. Clinical Development of PD-1 in Advanced Melanoma. Cancer journal (Sudbury, Mass) 2018;24(1):7–14 doi 10.1097/ppo.0000000000000299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Polk A, Svane IM, Andersson M, Nielsen D. Checkpoint inhibitors in breast cancer - Current status. Cancer treatment reviews 2018;63:122–34 doi 10.1016/j.ctrv.2017.12.008. [DOI] [PubMed] [Google Scholar]
- 10.Zolkind P, Uppaluri R. Checkpoint immunotherapy in head and neck cancers. Cancer metastasis reviews 2017;36(3):475–89 doi 10.1007/s10555-017-9694-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Schurich A, Pallett LJ, Jajbhay D, Wijngaarden J, Otano I, Gill US, et al. Distinct Metabolic Requirements of Exhausted and Functional Virus-Specific CD8 T Cells in the Same Host. Cell reports 2016;16(5):1243–52 doi 10.1016/j.celrep.2016.06.078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Araki K, Morita M, Bederman AG, Konieczny BT, Kissick HT, Sonenberg N, et al. Translation is actively regulated during the differentiation of CD8(+) effector T cells. Nature immunology 2017;18(9):1046–57 doi 10.1038/ni.3795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Rutkowski DT, Kaufman RJ. A trip to the ER: coping with stress. Trends in cell biology 2004;14(1):20–8. [DOI] [PubMed] [Google Scholar]
- 14.Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, Schapira M, et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Molecular cell 2000;6(5):1099–108. [DOI] [PubMed] [Google Scholar]
- 15.Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Molecular cell 2003;11(3):619–33. [DOI] [PubMed] [Google Scholar]
- 16.Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, et al. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes & development 2004;18(24):3066–77 doi 10.1101/gad.1250704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Han J, Back SH, Hur J, Lin YH, Gildersleeve R, Shan J, et al. ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nature cell biology 2013;15(5):481–90 doi 10.1038/ncb2738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Thaxton JE, Wallace C, Riesenberg B, Zhang Y, Paulos CM, Beeson CC, et al. Modulation of Endoplasmic Reticulum Stress Controls CD4(+) T-cell Activation and Antitumor Function. Cancer immunology research 2017;5(8):666–75 doi 10.1158/2326-6066.Cir-17-0081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kaufman RJ, Malhotra JD. Calcium trafficking integrates endoplasmic reticulum function with mitochondrial bioenergetics. Biochimica et biophysica acta 2014;1843(10):2233–9 doi 10.1016/j.bbamcr.2014.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.van Vliet AR, Garg AD, Agostinis P. Coordination of stress, Ca2+, and immunogenic signaling pathways by PERK at the endoplasmic reticulum. Biological chemistry 2016;397(7):649–56 doi 10.1515/hsz-2016-0108. [DOI] [PubMed] [Google Scholar]
- 21.van der Windt GJ, Pearce EL. Metabolic switching and fuel choice during T-cell differentiation and memory development. Immunological reviews 2012;249(1):27–42 doi 10.1111/j.1600-065X.2012.01150.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Braakman I, Helenius J, Helenius A. Role of ATP and disulphide bonds during protein folding in the endoplasmic reticulum. Nature 1992;356(6366):260–2 doi 10.1038/356260a0. [DOI] [PubMed] [Google Scholar]
- 23.Axten JM, Medina JR, Feng Y, Shu A, Romeril SP, Grant SW, et al. Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}−2,3-dihydro-1H-indol-5-yl)-7H-p yrrolo[2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK). Journal of medicinal chemistry 2012;55(16):7193–207 doi 10.1021/jm300713s. [DOI] [PubMed] [Google Scholar]
- 24.Blais JD, Chin KT, Zito E, Zhang Y, Heldman N, Harding HP, et al. A small molecule inhibitor of endoplasmic reticulum oxidation 1 (ERO1) with selectively reversible thiol reactivity. The Journal of biological chemistry 2010;285(27):20993–1003 doi 10.1074/jbc.M110.126599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.van der Windt GJ, O’Sullivan D, Everts B, Huang SC, Buck MD, Curtis JD, et al. CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability. Proceedings of the National Academy of Sciences of the United States of America 2013;110(35):14336–41 doi 10.1073/pnas.1221740110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zhao H, Kalivendi S, Zhang H, Joseph J, Nithipatikom K, Vasquez-Vivar J, et al. Superoxide reacts with hydroethidine but forms a fluorescent product that is distinctly different from ethidium: potential implications in intracellular fluorescence detection of superoxide. Free radical biology & medicine 2003;34(11):1359–68. [DOI] [PubMed] [Google Scholar]
- 27.Venditti P, Di Stefano L, Di Meo S. Mitochondrial metabolism of reactive oxygen species. Mitochondrion 2013;13(2):71–82 doi 10.1016/j.mito.2013.01.008. [DOI] [PubMed] [Google Scholar]
- 28.Navarro A, Boveris A. The mitochondrial energy transduction system and the aging process. American journal of physiology Cell physiology 2007;292(2):C670–86 doi 10.1152/ajpcell.00213.2006. [DOI] [PubMed] [Google Scholar]
- 29.Youngblood B, Hale JS, Kissick HT, Ahn E, Xu X, Wieland A, et al. Effector CD8 T cells dedifferentiate into long-lived memory cells. Nature 2017;552(7685):404–9 doi 10.1038/nature25144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Gattinoni L, Restifo NP. Moving T memory stem cells to the clinic. Blood 2013;121(4):567–8 doi 10.1182/blood-2012-11-468660. [DOI] [PubMed] [Google Scholar]
- 31.Fernandez-Poma SM, Salas-Benito D, Lozano T, Casares N, Riezu-Boj JI, Mancheno U, et al. Expansion of Tumor-Infiltrating CD8(+) T cells Expressing PD-1 Improves the Efficacy of Adoptive T-cell Therapy. Cancer research 2017;77(13):3672–84 doi 10.1158/0008-5472.Can-17-0236. [DOI] [PubMed] [Google Scholar]
- 32.Thompson ED, Enriquez HL, Fu YX, Engelhard VH. Tumor masses support naive T cell infiltration, activation, and differentiation into effectors. The Journal of experimental medicine 2010;207(8):1791–804 doi 10.1084/jem.20092454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bengsch B, Johnson AL, Kurachi M, Odorizzi PM, Pauken KE, Attanasio J, et al. Bioenergetic Insufficiencies Due to Metabolic Alterations Regulated by the Inhibitory Receptor PD-1 Are an Early Driver of CD8(+) T Cell Exhaustion. Immunity 2016;45(2):358–73 doi 10.1016/j.immuni.2016.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Nathenson MJ, Conley AP, Sausville E. Immunotherapy: A New (and Old) Approach to Treatment of Soft Tissue and Bone Sarcomas. The oncologist 2018;23(1):71–83 doi 10.1634/theoncologist.2016-0025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Cubillos-Ruiz JR, Bettigole SE, Glimcher LH. Tumorigenic and Immunosuppressive Effects of Endoplasmic Reticulum Stress in Cancer. Cell 2017;168(4):692–706 doi 10.1016/j.cell.2016.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Condamine T, Kumar V, Ramachandran IR, Youn JI, Celis E, Finnberg N, et al. ER stress regulates myeloid-derived suppressor cell fate through TRAIL-R-mediated apoptosis. The Journal of clinical investigation 2014;124(6):2626–39 doi 10.1172/jci74056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tobin RP, Davis D, Jordan KR, McCarter MD. The clinical evidence for targeting human myeloid-derived suppressor cells in cancer patients. Journal of leukocyte biology 2017;102(2):381–91 doi 10.1189/jlb.5VMR1016-449R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, Chopra S, Perales-Puchalt A, Song M, et al. ER Stress Sensor XBP1 Controls Anti-tumor Immunity by Disrupting Dendritic Cell Homeostasis. Cell 2015;161(7):1527–38 doi 10.1016/j.cell.2015.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Urra H, Dufey E, Avril T, Chevet E, Hetz C. Endoplasmic Reticulum Stress and the Hallmarks of Cancer. Trends in cancer 2016;2(5):252–62 doi 10.1016/j.trecan.2016.03.007. [DOI] [PubMed] [Google Scholar]
- 40.Kamimura D, Bevan MJ. Endoplasmic reticulum stress regulator XBP-1 contributes to effector CD8+ T cell differentiation during acute infection. Journal of immunology (Baltimore, Md : 1950) 2008;181(8):5433–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Franchina DG, He F, Brenner D. Survival of the fittest: Cancer challenges T cell metabolism. Cancer letters 2018;412:216–23 doi 10.1016/j.canlet.2017.10.014. [DOI] [PubMed] [Google Scholar]
- 42.Zhang L, Romero P. Metabolic Control of CD8(+) T Cell Fate Decisions and Antitumor Immunity. Trends in molecular medicine 2018;24(1):30–48 doi 10.1016/j.molmed.2017.11.005. [DOI] [PubMed] [Google Scholar]
- 43.O’Sullivan D, van der Windt GJ, Huang SC, Curtis JD, Chang CH, Buck MD, et al. Memory CD8(+) T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 2014;41(1):75–88 doi 10.1016/j.immuni.2014.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Atkins C, Liu Q, Minthorn E, Zhang SY, Figueroa DJ, Moss K, et al. Characterization of a novel PERK kinase inhibitor with antitumor and antiangiogenic activity. Cancer research 2013;73(6):1993–2002 doi 10.1158/0008-5472.Can-12-3109. [DOI] [PubMed] [Google Scholar]
- 45.Cavener DR, Gupta S, McGrath BC. PERK in beta cell biology and insulin biogenesis. Trends in endocrinology and metabolism: TEM 2010;21(12):714–21 doi 10.1016/j.tem.2010.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gupta S, McGrath B, Cavener DR. PERK regulates the proliferation and development of insulin-secreting beta-cell tumors in the endocrine pancreas of mice. PloS one 2009;4(11):e8008 doi 10.1371/journal.pone.0008008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang R, Munoz EE, Zhu S, McGrath BC, Cavener DR. Perk gene dosage regulates glucose homeostasis by modulating pancreatic beta-cell functions. PloS one 2014;9(6):e99684 doi 10.1371/journal.pone.0099684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Halliday M, Radford H, Sekine Y, Moreno J, Verity N, le Quesne J, et al. Partial restoration of protein synthesis rates by the small molecule ISRIB prevents neurodegeneration without pancreatic toxicity. Cell death & disease 2015;6:e1672 doi 10.1038/cddis.2015.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.