Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Cell Rep ; 39(1): 110609, 2022 04 05.
Article En | MEDLINE | ID: mdl-35385733

Tumor-associated macrophages (TAMs) are a major cellular component in the tumor microenvironment (TME). However, the relationship between the phenotype and metabolic pattern of TAMs remains poorly understood. We performed single-cell transcriptome profiling on hepatic TAMs from mice bearing liver metastatic tumors. We find that TAMs manifest high heterogeneity at the levels of transcription, development, metabolism, and function. Integrative analyses and validation experiments indicate that increased purine metabolism is a feature of TAMs with pro-tumor and terminal differentiation phenotypes. Like mouse TAMs, human TAMs are highly heterogeneous. Human TAMs with increased purine metabolism exhibit a pro-tumor phenotype and correlate with poor therapeutic efficacy to immune checkpoint blockade. Altogether, our work demonstrates that TAMs are developmentally, metabolically, and functionally heterogeneous and purine metabolism may be a key metabolic feature of a pro-tumor macrophage population.


Liver Neoplasms , Tumor Microenvironment , Animals , Gene Expression Profiling , Liver Neoplasms/pathology , Macrophages/metabolism , Mice , Tumor-Associated Macrophages
2.
Nat Med ; 27(1): 152-164, 2021 01.
Article En | MEDLINE | ID: mdl-33398162

Metastasis is the primary cause of cancer mortality, and cancer frequently metastasizes to the liver. It is not clear whether liver immune tolerance mechanisms contribute to cancer outcomes. We report that liver metastases diminish immunotherapy efficacy systemically in patients and preclinical models. Patients with liver metastases derive limited benefit from immunotherapy independent of other established biomarkers of response. In multiple mouse models, we show that liver metastases siphon activated CD8+ T cells from systemic circulation. Within the liver, activated antigen-specific Fas+CD8+ T cells undergo apoptosis following their interaction with FasL+CD11b+F4/80+ monocyte-derived macrophages. Consequently, liver metastases create a systemic immune desert in preclinical models. Similarly, patients with liver metastases have reduced peripheral T cell numbers and diminished tumoral T cell diversity and function. In preclinical models, liver-directed radiotherapy eliminates immunosuppressive hepatic macrophages, increases hepatic T cell survival and reduces hepatic siphoning of T cells. Thus, liver metastases co-opt host peripheral tolerance mechanisms to cause acquired immunotherapy resistance through CD8+ T cell deletion, and the combination of liver-directed radiotherapy and immunotherapy could promote systemic antitumor immunity.


Immunotherapy , Liver Neoplasms, Experimental/secondary , Liver Neoplasms, Experimental/therapy , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Macrophages/immunology , T-Lymphocytes/immunology , Animals , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cohort Studies , Combined Modality Therapy , Female , Humans , Liver Neoplasms/immunology , Liver Neoplasms, Experimental/immunology , Lymphocyte Activation , Male , Melanoma/immunology , Melanoma/secondary , Melanoma/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Radiotherapy, Adjuvant , T-Lymphocytes/classification , T-Lymphocytes/pathology , Treatment Failure , Treatment Outcome , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
3.
Cancer Cell ; 39(4): 480-493.e6, 2021 04 12.
Article En | MEDLINE | ID: mdl-33513345

Immunotherapy induces durable clinical responses in a fraction of patients with cancer. However, therapeutic resistance poses a major challenge to current immunotherapies. Here, we identify that expression of tumor stanniocalcin 1 (STC1) correlates with immunotherapy efficacy and is negatively associated with patient survival across diverse cancer types. Gain- and loss-of-function experiments demonstrate that tumor STC1 supports tumor progression and enables tumor resistance to checkpoint blockade in murine tumor models. Mechanistically, tumor STC1 interacts with calreticulin (CRT), an "eat-me" signal, and minimizes CRT membrane exposure, thereby abrogating membrane CRT-directed phagocytosis by antigen-presenting cells (APCs), including macrophages and dendritic cells. Consequently, this impairs APC capacity of antigen presentation and T cell activation. Thus, tumor STC1 inhibits APC phagocytosis and contributes to tumor immune evasion and immunotherapy resistance. We suggest that STC1 is a previously unappreciated phagocytosis checkpoint and targeting STC1 and its interaction with CRT may sensitize to cancer immunotherapy.


Glycoproteins/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Phagocytosis/immunology , Tumor Escape/immunology , Animals , Antigen Presentation/immunology , Immunotherapy/methods , Macrophages/metabolism , Mice , Phagocytosis/drug effects , Receptors, Immunologic/immunology
4.
Nature ; 585(7824): 277-282, 2020 09.
Article En | MEDLINE | ID: mdl-32879489

Abnormal epigenetic patterns correlate with effector T cell malfunction in tumours1-4, but the cause of this link is unknown. Here we show that tumour cells disrupt methionine metabolism in CD8+ T cells, thereby lowering intracellular levels of methionine and the methyl donor S-adenosylmethionine (SAM) and resulting in loss of dimethylation at lysine 79 of histone H3 (H3K79me2). Loss of H3K79me2 led to low expression of STAT5 and impaired T cell immunity. Mechanistically, tumour cells avidly consumed methionine and outcompeted T cells for methionine by expressing high levels of the methionine transporter SLC43A2. Genetic and biochemical inhibition of tumour SLC43A2 restored H3K79me2 in T cells, thereby boosting spontaneous and checkpoint-induced tumour immunity. Moreover, methionine supplementation improved the expression of H3K79me2 and STAT5 in T cells, and this was accompanied by increased T cell immunity in tumour-bearing mice and patients with colon cancer. Clinically, tumour SLC43A2 correlated negatively with T cell histone methylation and functional gene signatures. Our results identify a mechanistic connection between methionine metabolism, histone patterns, and T cell immunity in the tumour microenvironment. Thus, cancer methionine consumption is an immune evasion mechanism, and targeting cancer methionine signalling may provide an immunotherapeutic approach.


Amino Acid Transport System L/metabolism , CD8-Positive T-Lymphocytes/metabolism , Histones/metabolism , Methionine/metabolism , Methylation , Neoplasms/metabolism , Amino Acid Transport System L/deficiency , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Epigenesis, Genetic , Female , Histones/chemistry , Humans , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, T-Cell/metabolism , STAT5 Transcription Factor/metabolism
5.
Cancer Discov ; 9(12): 1673-1685, 2019 12.
Article En | MEDLINE | ID: mdl-31554642

A challenge in oncology is to rationally and effectively integrate immunotherapy with traditional modalities, including radiotherapy. Here, we demonstrate that radiotherapy induces tumor-cell ferroptosis. Ferroptosis agonists augment and ferroptosis antagonists limit radiotherapy efficacy in tumor models. Immunotherapy sensitizes tumors to radiotherapy by promoting tumor-cell ferroptosis. Mechanistically, IFNγ derived from immunotherapy-activated CD8+ T cells and radiotherapy-activated ATM independently, yet synergistically, suppresses SLC7A11, a unit of the glutamate-cystine antiporter xc-, resulting in reduced cystine uptake, enhanced tumor lipid oxidation and ferroptosis, and improved tumor control. Thus, ferroptosis is an unappreciated mechanism and focus for the development of effective combinatorial cancer therapy. SIGNIFICANCE: This article describes ferroptosis as a previously unappreciated mechanism of action for radiotherapy. Further, it shows that ferroptosis is a novel point of synergy between immunotherapy and radiotherapy. Finally, it nominates SLC7A11, a critical regulator of ferroptosis, as a mechanistic determinant of synergy between radiotherapy and immunotherapy.This article is highlighted in the In This Issue feature, p. 1631.


Amino Acid Transport System y+/genetics , Antineoplastic Agents, Immunological/administration & dosage , Melanoma, Experimental/therapy , Sulfasalazine/administration & dosage , Animals , Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Down-Regulation , Ferroptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy/methods , Interferon-gamma , Lipid Metabolism/drug effects , Lipid Metabolism/radiation effects , Melanoma, Experimental/genetics , Mice , Oxidation-Reduction , Sulfasalazine/pharmacology , Xenograft Model Antitumor Assays
6.
Nature ; 569(7755): 270-274, 2019 05.
Article En | MEDLINE | ID: mdl-31043744

Cancer immunotherapy restores or enhances the effector function of CD8+ T cells in the tumour microenvironment1,2. CD8+ T cells activated by cancer immunotherapy clear tumours mainly by inducing cell death through perforin-granzyme and Fas-Fas ligand pathways3,4. Ferroptosis is a form of cell death that differs from apoptosis and results from iron-dependent accumulation of lipid peroxide5,6. Although it has been investigated in vitro7,8, there is emerging evidence that ferroptosis might be implicated in a variety of pathological scenarios9,10. It is unclear whether, and how, ferroptosis is involved in T cell immunity and cancer immunotherapy. Here we show that immunotherapy-activated CD8+ T cells enhance ferroptosis-specific lipid peroxidation in tumour cells, and that increased ferroptosis contributes to the anti-tumour efficacy of immunotherapy. Mechanistically, interferon gamma (IFNγ) released from CD8+ T cells downregulates the expression of SLC3A2 and SLC7A11, two subunits of the glutamate-cystine antiporter system xc-, impairs the uptake of cystine by tumour cells, and as a consequence, promotes tumour cell lipid peroxidation and ferroptosis. In mouse models, depletion of cystine or cysteine by cyst(e)inase (an engineered enzyme that degrades both cystine and cysteine) in combination with checkpoint blockade synergistically enhanced T cell-mediated anti-tumour immunity and induced ferroptosis in tumour cells. Expression of system xc- was negatively associated, in cancer patients, with CD8+ T cell signature, IFNγ expression, and patient outcome. Analyses of human transcriptomes before and during nivolumab therapy revealed that clinical benefits correlate with reduced expression of SLC3A2 and increased IFNγ and CD8. Thus, T cell-promoted tumour ferroptosis is an anti-tumour mechanism, and targeting this pathway in combination with checkpoint blockade is a potential therapeutic approach.


CD8-Positive T-Lymphocytes/immunology , Ferroptosis , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Amino Acid Transport System y+/metabolism , Animals , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Cysteine/metabolism , Female , Ferroptosis/drug effects , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Humans , Interferon-gamma/immunology , Lipid Peroxidation , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Melanoma/therapy , Mice , Neoplasms/metabolism , Nivolumab/therapeutic use , Reactive Oxygen Species/metabolism , Treatment Outcome
8.
J Clin Invest ; 128(2): 805-815, 2018 02 01.
Article En | MEDLINE | ID: mdl-29337305

Programmed death-1 receptor (PD-L1, B7-H1) and programmed cell death protein 1 (PD-1) pathway blockade is a promising therapy for treating cancer. However, the mechanistic contribution of host and tumor PD-L1 and PD-1 signaling to the therapeutic efficacy of PD-L1 and PD-1 blockade remains elusive. Here, we evaluated 3 tumor-bearing mouse models that differ in their sensitivity to PD-L1 blockade and demonstrated a loss of therapeutic efficacy of PD-L1 blockade in immunodeficient mice and in PD-L1- and PD-1-deficient mice. In contrast, neither knockout nor overexpression of PD-L1 in tumor cells had an effect on PD-L1 blockade efficacy. Human and murine studies showed high levels of functional PD-L1 expression in dendritic cells and macrophages in the tumor microenvironments and draining lymph nodes. Additionally, expression of PD-L1 on dendritic cells and macrophages in ovarian cancer and melanoma patients correlated with the efficacy of treatment with either anti-PD-1 alone or in combination with anti-CTLA-4. Thus, PD-L1-expressing dendritic cells and macrophages may mechanistically shape and therapeutically predict clinical efficacy of PD-L1/PD-1 blockade.


B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cytokines/metabolism , Dendrites/metabolism , Dendritic Cells/metabolism , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Lymph Nodes/pathology , Macrophages/metabolism , Male , Melanoma/drug therapy , Melanoma/pathology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, SCID , Ovarian Neoplasms/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Time Factors , Tumor Microenvironment
9.
Nat Immunol ; 18(12): 1332-1341, 2017 Dec.
Article En | MEDLINE | ID: mdl-29083399

Live regulatory T cells (Treg cells) suppress antitumor immunity, but how Treg cells behave in the metabolically abnormal tumor microenvironment remains unknown. Here we show that tumor Treg cells undergo apoptosis, and such apoptotic Treg cells abolish spontaneous and PD-L1-blockade-mediated antitumor T cell immunity. Biochemical and functional analyses show that adenosine, but not typical suppressive factors such as PD-L1, CTLA-4, TGF-ß, IL-35, and IL-10, contributes to apoptotic Treg-cell-mediated immunosuppression. Mechanistically, apoptotic Treg cells release and convert a large amount of ATP to adenosine via CD39 and CD73, and mediate immunosuppression via the adenosine and A2A pathways. Apoptosis in Treg cells is attributed to their weak NRF2-associated antioxidant system and high vulnerability to free oxygen species in the tumor microenvironment. Thus, the data support a model wherein tumor Treg cells sustain and amplify their suppressor capacity through inadvertent death via oxidative stress. This work highlights the oxidative pathway as a metabolic checkpoint that controls Treg cell behavior and affects the efficacy of therapeutics targeting cancer checkpoints.


Apoptosis/immunology , B7-H1 Antigen/metabolism , Immune Tolerance/immunology , Ovarian Neoplasms/immunology , Oxidative Stress/physiology , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , CTLA-4 Antigen/metabolism , Female , GPI-Linked Proteins/genetics , Humans , Interleukin-10/metabolism , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxygen/metabolism , Receptor, Adenosine A2A/metabolism , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/immunology
10.
Oncoimmunology ; 5(8): e1105430, 2016 Aug.
Article En | MEDLINE | ID: mdl-27622054

Foxp3(+)CD4(+) regulatory T (Treg) cells are thought to express negligible levels of effector cytokines, and inhibit immune responses and inflammation. Here, we have identified a population of IL-8(+)Foxp3(+)CD4(+) T cells in human peripheral blood, which is selectively increased in the microenvironments of ulcerative colitis and colon carcinoma. Phenotypically, this population is minimally overlapping with IL-17(+)Foxp3(+)CD4(+) T cells, and is different from IL-8(-)Foxp3(+)CD4(+) T cells in the same microenvironment. 40-60% of IL-8(+)Foxp3(+)CD4(+) T cells exhibit naive phenotype and express CD127, whereas IL-8(-)Foxp3(+)CD4(+) cells are basically memory T cells and express minimal CD127. The levels of CXCR5 expression are higher in IL-8(+)Foxp3(+) cells than in IL-8(-)Foxp3(+) cells. IL-2 and TGFß induce IL-8(+)Foxp3(+) T cells. Exogenous Foxp3 expression promotes IL-8(+)Foxp3(+) T cells and inhibits effector cytokine IFNγ and IL-2 expression. Furthermore, Foxp3 binds to IL-8 proximal promoter and increases its activity. Functionally, IL-8(+)Foxp3(+) T cells inhibit T cell proliferation and effector cytokine production, but stimulate inflammatory cytokine production in the colon tissues, and promote neutrophil trafficking through IL-8. Thus, IL-8(+)Foxp3(+) cells may be an "inflammatory" Treg subset, and possess inflammatory and immunosuppressive dual biological activities. Given their dual roles and localization, these cells may be in a unique position to support tumor initiation and development in human chronic inflammatory environment.

11.
Cell ; 165(5): 1092-1105, 2016 May 19.
Article En | MEDLINE | ID: mdl-27133165

Effector T cells and fibroblasts are major components in the tumor microenvironment. The means through which these cellular interactions affect chemoresistance is unclear. Here, we show that fibroblasts diminish nuclear accumulation of platinum in ovarian cancer cells, resulting in resistance to platinum-based chemotherapy. We demonstrate that glutathione and cysteine released by fibroblasts contribute to this resistance. CD8(+) T cells abolish the resistance by altering glutathione and cystine metabolism in fibroblasts. CD8(+) T-cell-derived interferon (IFN)γ controls fibroblast glutathione and cysteine through upregulation of gamma-glutamyltransferases and transcriptional repression of system xc(-) cystine and glutamate antiporter via the JAK/STAT1 pathway. The presence of stromal fibroblasts and CD8(+) T cells is negatively and positively associated with ovarian cancer patient survival, respectively. Thus, our work uncovers a mode of action for effector T cells: they abrogate stromal-mediated chemoresistance. Capitalizing upon the interplay between chemotherapy and immunotherapy holds high potential for cancer treatment.


CD8-Positive T-Lymphocytes/metabolism , Drug Resistance, Neoplasm , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Culture Techniques , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Fibroblasts/metabolism , Glutathione/metabolism , Humans , Interferon-gamma/metabolism , Mice , Mice, Inbred NOD , Mice, Nude
12.
Cancer Res ; 76(11): 3156-65, 2016 06 01.
Article En | MEDLINE | ID: mdl-27197152

Myeloid-derived suppressor cells (MDSC) contribute to immune suppression in cancer, but the mechanisms through which they drive metastatic progression are not fully understood. In this study, we show how MDSC convey stem-like qualities to breast cancer cells that coordinately help enable immune suppression and escape. We found that MDSC promoted tumor formation by enhancing breast cancer cell stem-like properties as well as by suppressing T-cell activation. Mechanistic investigations indicated that these effects relied upon cross-talk between the STAT3 and NOTCH pathways in cancer cells, with MDSC inducing IL6-dependent phosphorylation of STAT3 and activating NOTCH through nitric oxide leading to prolonged STAT3 activation. In clinical specimens of breast cancer, the presence of MDSC correlated with the presence of cancer stem-like cells (CSC) and independently predicted poor survival outcomes. Collectively, our work revealed an immune-associated mechanism that extrinsically confers cancer cell stemness properties and affects patient outcome. We suggest that targeting STAT3-NOTCH cross-talk between MDSC and CSC could offer a unique locus to improve cancer treatment, by coordinately targeting a coupled mechanism that enables cancer stemness and immune escape. Cancer Res; 76(11); 3156-65. ©2016 AACR.


Breast Neoplasms/pathology , Interleukin-6/metabolism , Myeloid-Derived Suppressor Cells/pathology , Neoplastic Stem Cells/pathology , Nitric Oxide/metabolism , Receptors, Notch/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Cohort Studies , Female , Flow Cytometry , Follow-Up Studies , Humans , Immunoenzyme Techniques , Interleukin-6/genetics , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Staging , Neoplastic Stem Cells/metabolism , Phosphorylation , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor Cross-Talk , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Nat Immunol ; 17(1): 95-103, 2016 Jan.
Article En | MEDLINE | ID: mdl-26523864

Aerobic glycolysis regulates T cell function. However, whether and how primary cancer alters T cell glycolytic metabolism and affects tumor immunity in cancer patients remains a question. Here we found that ovarian cancers imposed glucose restriction on T cells and dampened their function via maintaining high expression of microRNAs miR-101 and miR-26a, which constrained expression of the methyltransferase EZH2. EZH2 activated the Notch pathway by suppressing Notch repressors Numb and Fbxw7 via trimethylation of histone H3 at Lys27 and, consequently, stimulated T cell polyfunctional cytokine expression and promoted their survival via Bcl-2 signaling. Moreover, small hairpin RNA-mediated knockdown of human EZH2 in T cells elicited poor antitumor immunity. EZH2(+)CD8(+) T cells were associated with improved survival in patients. Together, these data unveil a metabolic target and mechanism of cancer immune evasion.


Gene Expression Regulation, Neoplastic/immunology , MicroRNAs , Neoplasms/immunology , Polycomb Repressive Complex 2/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Animals , Cell Separation , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Female , Flow Cytometry , Fluorescent Antibody Technique , Glycolysis , Humans , Immunoblotting , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Transfection
14.
Cancer Res ; 76(2): 275-82, 2016 Jan 15.
Article En | MEDLINE | ID: mdl-26567139

Infiltration of tumors with effector T cells is positively associated with therapeutic efficacy and patient survival. However, the mechanisms underlying effector T-cell trafficking to the tumor microenvironment remain poorly understood in patients with colon cancer. The polycomb repressive complex 2 (PRC2) is involved in cancer progression, but the regulation of tumor immunity by epigenetic mechanisms has yet to be investigated. In this study, we examined the relationship between the repressive PRC2 machinery and effector T-cell trafficking. We found that PRC2 components and demethylase JMJD3-mediated histone H3 lysine 27 trimethylation (H3K27me3) repress the expression and subsequent production of Th1-type chemokines CXCL9 and CXCL10, mediators of effector T-cell trafficking. Moreover, the expression levels of PRC2 components, including EZH2, SUZ12, and EED, were inversely associated with those of CD4, CD8, and Th1-type chemokines in human colon cancer tissue, and this expression pattern was significantly associated with patient survival. Collectively, our findings reveal that PRC2-mediated epigenetic silencing is not only a crucial oncogenic mechanism, but also a key circuit controlling tumor immunosuppression. Therefore, targeting epigenetic programs may have significant implications for improving the efficacy of current cancer immunotherapies relying on effective T-cell-mediated immunity at the tumor site.


Chemokines/immunology , Colonic Neoplasms/genetics , Polycomb Repressive Complex 2/genetics , T-Lymphocytes/immunology , Th1 Cells/immunology , Cell Movement/genetics , Cell Movement/immunology , Cell Proliferation/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Epigenesis, Genetic , Humans , Polycomb Repressive Complex 2/immunology , T-Lymphocytes/metabolism , Th1 Cells/metabolism , Transfection
15.
Nature ; 527(7577): 249-53, 2015 Nov 12.
Article En | MEDLINE | ID: mdl-26503055

Epigenetic silencing including histone modifications and DNA methylation is an important tumorigenic mechanism. However, its role in cancer immunopathology and immunotherapy is poorly understood. Using human ovarian cancers as our model, here we show that enhancer of zeste homologue 2 (EZH2)-mediated histone H3 lysine 27 trimethylation (H3K27me3) and DNA methyltransferase 1 (DNMT1)-mediated DNA methylation repress the tumour production of T helper 1 (TH1)-type chemokines CXCL9 and CXCL10, and subsequently determine effector T-cell trafficking to the tumour microenvironment. Treatment with epigenetic modulators removes the repression and increases effector T-cell tumour infiltration, slows down tumour progression, and improves the therapeutic efficacy of programmed death-ligand 1 (PD-L1; also known as B7-H1) checkpoint blockade and adoptive T-cell transfusion in tumour-bearing mice. Moreover, tumour EZH2 and DNMT1 are negatively associated with tumour-infiltrating CD8(+) T cells and patient outcome. Thus, epigenetic silencing of TH1-type chemokines is a novel immune-evasion mechanism of tumours. Selective epigenetic reprogramming alters the T-cell landscape in cancer and may enhance the clinical efficacy of cancer therapy.


Chemokines/genetics , Epigenesis, Genetic , Gene Silencing , Immunotherapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Th1 Cells/metabolism , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL9/biosynthesis , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Chemokines/biosynthesis , Chemokines/immunology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic/drug effects , Female , Histones/chemistry , Histones/metabolism , Humans , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Lysine/metabolism , Mice , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/metabolism , Prognosis , Th1 Cells/immunology , Tumor Cells, Cultured , Tumor Escape/immunology , Xenograft Model Antitumor Assays
16.
Immunity ; 40(5): 772-784, 2014 May 15.
Article En | MEDLINE | ID: mdl-24816405

Little is known about how the immune system impacts human colorectal cancer invasiveness and stemness. Here we detected interleukin-22 (IL-22) in patient colorectal cancer tissues that was produced predominantly by CD4(+) T cells. In a mouse model, migration of these cells into the colon cancer microenvironment required the chemokine receptor CCR6 and its ligand CCL20. IL-22 acted on cancer cells to promote activation of the transcription factor STAT3 and expression of the histone 3 lysine 79 (H3K79) methytransferase DOT1L. The DOT1L complex induced the core stem cell genes NANOG, SOX2, and Pou5F1, resulting in increased cancer stemness and tumorigenic potential. Furthermore, high DOT1L expression and H3K79me2 in colorectal cancer tissues was a predictor of poor patient survival. Thus, IL-22(+) cells promote colon cancer stemness via regulation of stemness genes that negatively affects patient outcome. Efforts to target this network might be a strategy in treating colorectal cancer patients.


CD4-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Interleukins/immunology , Methyltransferases/immunology , Neoplastic Stem Cells/immunology , STAT3 Transcription Factor/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Chemokine CCL20/immunology , Chemokine CCL20/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Enzyme Activation/immunology , HT29 Cells , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Humans , Methyltransferases/metabolism , Mice , Nanog Homeobox Protein , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/immunology , Octamer Transcription Factor-3/metabolism , Receptors, CCR6/immunology , Receptors, CCR6/metabolism , SOXB1 Transcription Factors/immunology , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Interleukin-22
17.
Immunity ; 39(3): 611-21, 2013 Sep 19.
Article En | MEDLINE | ID: mdl-24012420

Myeloid-derived suppressor cells (MDSCs) and cancer stem cells (CSCs) are important cellular components in the cancer microenvironment and may affect cancer phenotype and patient outcome. The nature of MDSCs and their interaction with CSCs in ovarian carcinoma are unclear. We examined the interaction between MDSCs and CSCs in patients with ovarian carcinoma and showed that MDSCs inhibited T cell activation and enhanced CSC gene expression, sphere formation, and cancer metastasis. MDSCs triggered miRNA101 expression in cancer cells. miRNA101 subsequently repressesed the corepressor gene C-terminal binding protein-2 (CtBP2), and CtBP2 directly targeted stem cell core genes resulting in increased cancer cell stemness and increasing metastatic and tumorigenic potential. Increased MDSC density and tumor microRNA101 expression predict poor survival, as does decreased tumor CtBP2 expression, independent of each other. Collectively, our work identifies an immune-associated cellular, molecular, and clinical network involving MDSCs-microRNA101-CtBP2-stem cell core genes, which extrinsically controls cancer stemness and impacts patient outcome.


Alcohol Oxidoreductases/metabolism , MicroRNAs/metabolism , Myeloid Cells/metabolism , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Ovarian Neoplasms/immunology , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/genetics , Cell Communication , Co-Repressor Proteins , Female , Humans , Lymphocyte Activation , MicroRNAs/genetics , Myeloid Cells/cytology , Myeloid Cells/immunology , Neoplasm Metastasis , Neoplastic Stem Cells/immunology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Ovarian Neoplasms/metabolism , RNA Interference , RNA, Small Interfering , T-Lymphocytes/immunology
18.
Int J Cancer ; 130(1): 29-39, 2012 Jan 01.
Article En | MEDLINE | ID: mdl-21480217

Identification of cancer stem cells is crucial for advancing cancer biology and therapy. Several markers including CD24, CD44, CD117, CD133, the G subfamily of ATP-binding cassette transporters (ABCG), epithelial specific antigen (ESA) and aldehyde dehydrogenase (ALDH) are used to identify and investigate human epithelial cancer stem cells in the literature. We have now systemically analyzed and compared the expression of these markers in fresh ovarian epithelial carcinomas. Although the expression levels of these markers were unexpectedly variable and partially overlapping in fresh ovarian cancer cells from different donors, we reliably detected important levels of CD133 and ALDH in the majority of fresh ovarian cancer. Furthermore, most of these stem cell markers including CD133 and ALDH were gradually lost following in vitro passage of primary tumor cells. However, the expression of ALDH and CD133, but not CD24, CD44 and CD117, could be partially rescued by the in vitro serum-free and sphere cultures and by the in vivo passage in the immune-deficient xenografts. ALDH+ and CD133+ cells formed three-dimensional spheres more efficiently than their negative counterparts. These sphere-forming cells expressed high levels of stem cell core gene transcripts and could be expanded and form additional spheres in long-term culture. ALDH+ , CD133+ and ALDH+ CD133+ cells from fresh tumors developed larger tumors more rapidly than their negative counterparts. This property was preserved in the xenografted tumors. Altogether, the data suggest that ALDH+ and CD133+ cells are enriched with ovarian cancer-initiating (stem) cells and that ALDH and CD133 may be widely used as reliable markers to investigate ovarian cancer stem cell biology.


ATP-Binding Cassette Transporters/metabolism , Antigens, CD/metabolism , Glycoproteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peptides/metabolism , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Antigens, CD/genetics , Carcinoma, Ovarian Epithelial , Cell Proliferation , Female , Flow Cytometry , Fluorescent Antibody Technique , Glycoproteins/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Peptides/genetics , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
19.
Sci Transl Med ; 3(104): 104ra100, 2011 Oct 12.
Article En | MEDLINE | ID: mdl-21998407

T helper 17 (TH17) cells have been shown to contribute to multiple disease systems. However, the functional phenotype and survival pattern of TH17 cells as well as the underlying mechanisms that control TH17 cells have been poorly investigated in humans, significantly hampering the clinical targeting of these cells. Here, we studied human TH17 cells in the pathological microenvironments of graft-versus-host disease, ulcerative colitis, and cancer; TH17 cell numbers were increased in the chronic phase of these diseases. Human TH17 cells phenotypically resembled terminally differentiated memory T cells but were distinct from central memory, exhausted, and senescent T cells. Despite their phenotypic markers of terminal differentiation, TH17 cells mediated and promoted long-term antitumor immunity in in vivo adoptive transfer experiments. Furthermore, TH17 cells had a high capacity for proliferative self-renewal, potent persistence, and apoptotic resistance in vivo, as well as plasticity-converting into other types of TH cells. These cells expressed a relatively specific gene signature including abundant antiapoptotic genes. We found that hypoxia-inducible factor-1α and Notch collaboratively controlled key antiapoptosis Bcl-2 family gene expression and function in TH17 cells. Together, these data indicate that human TH17 cells may be a long-lived proliferating effector memory T cell population with unique genetic and functional characteristics. Targeting TH17-associated signaling pathway would be therapeutically meaningful for treating patients with autoimmune disease and advanced tumor.


Immunologic Memory , Th17 Cells/cytology , Animals , Apoptosis , Autoimmune Diseases/immunology , Cell Differentiation , Cell Line, Tumor , Cellular Senescence , Gene Expression Profiling , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Phenotype , Receptors, Notch/metabolism
20.
J Immunol ; 186(7): 4388-95, 2011 Apr 01.
Article En | MEDLINE | ID: mdl-21357259

Foxp3(+)CD4(+) regulatory T (Treg) cells inhibit immune responses and temper inflammation. IL-17(+)CD4(+) T (Th17) cells mediate inflammation of autoimmune diseases. A small population of IL-17(+)Foxp3(+)CD4(+) T cells has been observed in peripheral blood in healthy human beings. However, the biology of IL-17(+)Foxp3(+)CD4(+) T cells remains poorly understood in humans. We investigated their phenotype, cytokine profile, generation, and pathological relevance in patients with ulcerative colitis. We observed that high levels of IL-17(+)Foxp3(+)CD4(+) T cells were selectively accumulated in the colitic microenvironment and associated colon carcinoma. The phenotype and cytokine profile of IL-17(+)Foxp3(+)CD4(+) T cells was overlapping with Th17 and Treg cells. Myeloid APCs, IL-2, and TGF-ß are essential for their induction from memory CCR6(+) T cells or Treg cells. IL-17(+)Foxp3(+)CD4(+) T cells functionally suppressed T cell activation and stimulated inflammatory cytokine production in the colitic tissues. Our data indicate that IL-17(+)Foxp3(+) cells may be "inflammatory" Treg cells in the pathological microenvironments. These cells may contribute to the pathogenesis of ulcerative colitis through inducing inflammatory cytokines and inhibiting local T cell immunity, and in turn may mechanistically link human chronic inflammation to tumor development. Our data therefore challenge commonly held beliefs of the anti-inflammatory role of Treg cells and suggest a more complex Treg cell biology, at least in the context of human chronic inflammation and associated carcinoma.


Inflammation Mediators/physiology , Interleukin-17/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cells, Cultured , Chronic Disease , Coculture Techniques , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytokines/biosynthesis , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/physiology , Growth Inhibitors/biosynthesis , Growth Inhibitors/physiology , Humans , Immune Tolerance/immunology , Inflammation Mediators/metabolism , Interleukin-17/biosynthesis , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , T-Lymphocytes, Regulatory/metabolism
...