Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Nat Immunol ; 22(4): 460-470, 2021 04.
Article in English | MEDLINE | ID: mdl-33767425

ABSTRACT

Targeting the p53-MDM2 pathway to reactivate tumor p53 is a chemotherapeutic approach. However, the involvement of this pathway in CD8+ T cell-mediated antitumor immunity is unknown. Here, we report that mice with MDM2 deficiency in T cells exhibit accelerated tumor progression and a decrease in tumor-infiltrating CD8+ T cell survival and function. Mechanistically, MDM2 competes with c-Cbl for STAT5 binding, reduces c-Cbl-mediated STAT5 degradation and enhances STAT5 stability in tumor-infiltrating CD8+ T cells. Targeting the p53-MDM2 interaction with a pharmacological agent, APG-115, augmented MDM2 in T cells, thereby stabilizing STAT5, boosting T cell immunity and synergizing with cancer immunotherapy. Unexpectedly, these effects of APG-115 were dependent on p53 and MDM2 in T cells. Clinically, MDM2 abundance correlated with T cell function and interferon-γ signature in patients with cancer. Thus, the p53-MDM2 pathway controls T cell immunity, and targeting this pathway may treat patients with cancer regardless of tumor p53 status.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , Lymphocytes, Tumor-Infiltrating/enzymology , Neoplasms/enzymology , Proto-Oncogene Proteins c-mdm2/metabolism , STAT5 Transcription Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Combined Modality Therapy , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-mdm2/genetics , STAT5 Transcription Factor/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Nat Immunol ; 22(2): 193-204, 2021 02.
Article in English | MEDLINE | ID: mdl-33398181

ABSTRACT

Metabolic reprograming toward aerobic glycolysis is a pivotal mechanism shaping immune responses. Here we show that deficiency in NF-κB-inducing kinase (NIK) impairs glycolysis induction, rendering CD8+ effector T cells hypofunctional in the tumor microenvironment. Conversely, ectopic expression of NIK promotes CD8+ T cell metabolism and effector function, thereby profoundly enhancing antitumor immunity and improving the efficacy of T cell adoptive therapy. NIK regulates T cell metabolism via a NF-κB-independent mechanism that involves stabilization of hexokinase 2 (HK2), a rate-limiting enzyme of the glycolytic pathway. NIK prevents autophagic degradation of HK2 through controlling cellular reactive oxygen species levels, which in turn involves modulation of glucose-6-phosphate dehydrogenase (G6PD), an enzyme that mediates production of the antioxidant NADPH. We show that the G6PD-NADPH redox system is important for HK2 stability and metabolism in activated T cells. These findings establish NIK as a pivotal regulator of T cell metabolism and highlight a post-translational mechanism of metabolic regulation.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , Colonic Neoplasms/enzymology , Energy Metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/enzymology , Melanoma, Experimental/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Cytotoxicity, Immunologic , Enzyme Stability , Female , Glucosephosphate Dehydrogenase/metabolism , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Knockout , NADP/metabolism , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Microenvironment , NF-kappaB-Inducing Kinase
3.
Gastroenterology ; 161(6): 1813-1829, 2021 12.
Article in English | MEDLINE | ID: mdl-34606846

ABSTRACT

Chronic inflammation is a known risk factor for gastrointestinal cancer. The evidence that nonsteroidal anti-inflammatory drugs suppress the incidence, growth, and metastasis of gastrointestinal cancer supports the concept that a nonsteroidal anti-inflammatory drug target, cyclooxygenase, and its downstream bioactive lipid products may provide one of the links between inflammation and cancer. Preclinical studies have demonstrated that the cyclooxygenase-2-prostaglandin E2 pathway can promote gastrointestinal cancer development. Although the role of this pathway in cancer has been investigated extensively for 2 decades, only recent studies have described its effects on host defenses against transformed epithelial cells. Overcoming tumor-immune evasion remains one of the major challenges in cancer immunotherapy. This review summarizes the impacts of the cyclooxygenase-2-prostaglandin E2 pathway on gastrointestinal cancer development. Our focus was to highlight recent advances in our understanding of how this pathway induces tumor immune evasion.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Gastrointestinal Neoplasms/enzymology , Inflammation Mediators/metabolism , Tumor Escape , Tumor Microenvironment/immunology , Animals , Antineoplastic Agents/therapeutic use , Cancer-Associated Fibroblasts/enzymology , Cancer-Associated Fibroblasts/immunology , Cyclooxygenase 2 Inhibitors/therapeutic use , Epithelial Cells/enzymology , Epithelial Cells/immunology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/pathology , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Signal Transduction , Tumor Escape/drug effects , Tumor-Associated Macrophages/enzymology , Tumor-Associated Macrophages/immunology
4.
Cancer Metastasis Rev ; 38(3): 417-430, 2019 09.
Article in English | MEDLINE | ID: mdl-31482487

ABSTRACT

In the past decade, immune-based therapies such as monoclonal antibodies against tumor epitopes or immune checkpoint inhibitors have become an integral part of contemporary cancer treatment in many entities. However, a fundamental prerequisite for the success of such therapies is a sufficient trafficking of tumor-infiltrating lymphocytes into the tumor microenvironment. This infiltration is facilitated by chemokines, a group of about 50 small proteins capable of chemotactically guiding leukocytes. Proteolytic inactivation of chemokines leading to an impaired infiltration of immune effector cells appears to be an efficient immune escape mechanism of solid cancers.The CXCR3 and CX3CR1 chemokine receptor ligands CXCL9-11 and CX3CL1, respectively, are mainly responsible for the tumor-suppressive lymphocytic infiltration into the tumor micromilieu. Their structure explains the biochemical basis of their proteolytic cleavage, while in vivo data from mouse models and patient samples shed light on the corresponding processes in cancer. The emerging roles of proteases, e.g., matrix metalloproteinases, cathepsins, and dipeptidyl peptidase 4, in chemokine inactivation define new resistance mechanisms against immunotherapies and identify attractive new targets to enhance immune intervention in cancer.


Subject(s)
Chemokines/immunology , Chemokines/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Amino Acid Sequence , Animals , CX3C Chemokine Receptor 1/immunology , CX3C Chemokine Receptor 1/metabolism , Chemokine CXCL10/chemistry , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Chemokine CXCL11/chemistry , Chemokine CXCL11/immunology , Chemokine CXCL11/metabolism , Chemokine CXCL9/chemistry , Chemokine CXCL9/immunology , Chemokine CXCL9/metabolism , Chemokines/chemistry , Humans , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/pathology , Models, Molecular , Neoplasms/enzymology , Neoplasms/pathology , Peptide Hydrolases/metabolism , Proteolysis , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism
5.
Oncology ; 98(4): 252-258, 2020.
Article in English | MEDLINE | ID: mdl-32053815

ABSTRACT

BACKGROUND: Very little is known about receptor tyrosine kinase (RTK) expression on peripheral blood mononuclear cells (PBMC) in humans including renal cell carcinoma (RCC) patients. OBJECTIVES: The primary objective of this study was to evaluate expression levels of major RTKs on PBMC and tumor-infiltrating lymphocytes (TIL) isolated from RCC patients. The secondary aim was to compare levels of RTK expression in RCC patients before surgery and on the 180th day after surgery (lymphocyte lifetime) and to compare them with the expression in healthy donors. In addition, we compared RTK and PD-L1 expression in TIL. METHODS: Tumor and blood samples were obtained from 20 patients with primary RCC immediately after surgical resection. Blood samples were collected from 20 healthy donors. Tumors were harvested into RPMI 1640 medium (Gibco) and processed within 4 h. TIL isolation was performed using a modified protocol [Baldan et al. Br J Cancer. 2015;112:1510-18]. Expression of RTKs was evaluated with NovoExpress Software. Twenty tumors from the same patients were stained with PD-L1 IHC assay (clone SP142; Ventana). RESULTS: PBMC and TIL express RTKs in humans. The RTK expression level was significantly lower on peripheral blood cells in patients with RCC (mean 41%, range 27.1-62.6%) as compared with healthy donor PBMC (mean 77.1%, range 72.1-80.1%, all p < 0.05). Furthermore, RTK expression was significantly downregulated on intratumoral cells (mean 40%, range 23.2-52.3%) in comparison with healthy donor PBMC. There was no significant recovery of RTK expression on the 180th day except for VEGFR2. Five of 20 (25%) patients were PD-L1 positive. PD-L1 expression on TIL was strongly associated with downregulated expression of PDGFRα (p = 0.017) and PDGFRß (p = 0.024). CONCLUSIONS: PBMC and TIL had similar low RTK expression levels in RCC patients. PBMC of healthy humans had a significantly higher expression of RTK. PD-L1 and PDGFRα-ß expression could correlate. Comprehensive basic and clinical studies will be needed to define a biological role of RTKs on different lymphocyte subsets and correlations between clinical outcomes and expression levels.


Subject(s)
Blood Donors , Carcinoma, Renal Cell/enzymology , Kidney Neoplasms/enzymology , Leukocytes, Mononuclear/enzymology , Lymphocytes, Tumor-Infiltrating/enzymology , Receptor Protein-Tyrosine Kinases/blood , Adult , Aged , B7-H1 Antigen/blood , Female , Humans , Male , Middle Aged , Pilot Projects , Receptors, Platelet-Derived Growth Factor/blood
6.
Immunology ; 157(3): 210-218, 2019 07.
Article in English | MEDLINE | ID: mdl-31107985

ABSTRACT

Tumour infiltration by regulatory T (Treg) cells contributes to suppression of the anti-tumour immune response, which limits the efficacy of immune-mediated cancer therapies. The phosphoinositide 3-kinase (PI3K) pathway has key roles in mediating the function of many immune cell subsets, including Treg cells. Treg function is context-dependent and depends on input from different cell surface receptors, many of which can activate the PI3K pathway. In this review, we explore how PI3Kδ contributes to signalling through several major immune cell receptors, including the T-cell receptor and co-stimulatory receptors such as CD28 and ICOS, but is antagonized by the immune checkpoint receptors CTLA-4 and PD-1. Understanding how PI3Kδ inhibition affects Treg signalling events will help to inform how best to use PI3Kδ inhibitors in clinical cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Animals , Antineoplastic Agents/adverse effects , Class Ib Phosphatidylinositol 3-Kinase/immunology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Humans , Immunotherapy/adverse effects , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/immunology , Neoplasms/pathology , Phenotype , Protein Kinase Inhibitors/adverse effects , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Tumor Escape , Tumor Microenvironment
7.
Immunology ; 147(2): 251-64, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26595239

ABSTRACT

Decreased expression of CD3-ζ chain, an adaptor protein associated with T-cell signalling, is well documented in patients with oral cancer, but the mechanistic justifications are fragmentary. Previous studies in patients with oral cancer have shown that decreased expression of CD3-ζ chain was associated with decreased responsiveness of T cells. Tumours are known to induce localized as well as systemic immune suppression. This study provides evidence that oral tumour-derived factors promote immune suppression by down-regulating CD3-ζ chain expression. 2'5'-Oligoadenylate synthetase 2 (OAS2) was identified by the proteomic approach and our results established a causative link between CD3-ζ chain down-regulation and OAS2 stimulation. The surrogate situation was established by over-expressing OAS2 in a HEK293 cell line and cell-free supernatant was collected. These supernatants when incubated with T cells resulted in down-regulation of CD3-ζ chain, which shows that the secreted OAS2 is capable of regulating CD3-ζ chain expression. Incubation of T cells with cell-free supernatants of oral tumours or recombinant human OAS2 (rh-OAS2) induced caspase-3 activation, which resulted in CD3-ζ chain down-regulation. Caspase-3 inhibition/down-regulation using pharmacological inhibitor or small interfering RNA restored down-regulated CD3-ζ chain expression in T cells induced by cell-free tumour supernatant or rh-OAS2. Collectively these results show that OAS2 leads to impairment in CD3-ζ chain expression, so offering an explanation that might be applicable to the CD3-ζ chain deficiency observed in cancer and diverse disease conditions.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , CD3 Complex/metabolism , Caspase 3/metabolism , Lymphocytes, Tumor-Infiltrating/enzymology , Mouth Neoplasms/enzymology , T-Lymphocytes/enzymology , 2',5'-Oligoadenylate Synthetase/genetics , CD3 Complex/immunology , Case-Control Studies , Caspase 3/genetics , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mouth Neoplasms/genetics , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Paracrine Communication , Proteomics/methods , RNA Interference , Recombinant Proteins/metabolism , Signal Transduction , T-Lymphocytes/immunology , Time Factors , Transfection , Tumor Cells, Cultured
8.
Tissue Antigens ; 83(6): 409-13, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24673566

ABSTRACT

The purpose of this work was the assessment of cytotoxic reaction mediators - granzymes A and B in the serum of women with ovarian tumors. The study included 120 women with proven ovarian tumors. The control group consisted of 60 healthy women in whom no pathological changes within the reproductive system were detected. Concentrations of granzymes A and B were measured by enzyme-linked immunosorbent (ELISA) assay. The highest concentrations of the studied parameters were observed in serum of women with ovarian cancer. Moreover, the concentrations of granzymes A and B in patients with ovarian cancer were substantially increased in comparison to concentrations in patients with ovarian cystadenomas (P < 0.0001) or ovarian teratomas (P < 0.0001).


Subject(s)
Cystadenocarcinoma, Serous/blood , Cystadenoma, Serous/blood , Granzymes/blood , Neoplasm Proteins/blood , Ovarian Neoplasms/blood , Teratoma/blood , Adult , Apoptosis , CA-125 Antigen/blood , Case-Control Studies , Cystadenocarcinoma, Serous/immunology , Cystadenoma, Serous/immunology , Cytotoxicity, Immunologic , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Menopause/blood , Menstrual Cycle/blood , Middle Aged , Ovarian Neoplasms/immunology , Teratoma/immunology , Young Adult
10.
Dig Dis Sci ; 58(12): 3494-502, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23979437

ABSTRACT

BACKGROUNDS: Although many researchers have concentrated on the mechanism of T cell anergy resulting from over-expression of Indoleamine 2,3-dioxygenase (IDO), it remains unclear what alterations will developed in memory T cells (Tm) under over-expression of IDO. METHODS: Immunohistochemical staining for IDO expression in gastric cancer tissues (n=50) was carried out. Tumor-infiltrating memory Tm cells were counted by flow cytometry. The association between IDO expression and the subsets of tumor infiltrating Tm are discussed. RESULTS: The higher IDO expressions were significantly associated with deeper invasion (P=0.016) and higher frequencies (P=0.038) of lymph node metastasis. The lower tumor-infiltrating CD4+Tm were significantly associated with the advanced clinical stage (P=0.026) and lymph node metastasis (P=0.016). The lower percentages of CD8+Tm were significantly related to undifferentiated histological type (P=0.042) and lymph node metastasis (P=0.037). However, the lower percentage of CD8+Tem was significantly correlated to differentiated histological type (P=0.017), lower frequencies of lymph node metastasis (P=0.014), and earlier clinical stage (P=0.008). The higher IDO expression patients had significantly lower percentages of CD4+Tm (P=0.012) and CD8+Tm (P=0.033). Nevertheless, it was confirmed that the higher level of IDO expression correlated with higher percentages of CD8+Tm cells in univariate and multivariate analysis (P=0.011). CONCLUSION: IDO over-expression and Tm in tumor microenvironments were correlated with the disease stage and histological type of gastric cancer. Higher IDO expression was related to the lower percentage of CD4+Tm and CD8+Tm, whereas the higher level of IDO expression related with a higher percentage of CD8+Tem.


Subject(s)
Adenocarcinoma/pathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Lymphocytes, Tumor-Infiltrating/pathology , Stomach Neoplasms/pathology , Adenocarcinoma/enzymology , Adenocarcinoma/immunology , Adult , Aged , Female , Humans , Logistic Models , Lymphatic Metastasis/pathology , Lymphocytes, Tumor-Infiltrating/enzymology , Male , Middle Aged , Neoplasm Invasiveness , Stomach/immunology , Stomach/pathology , Stomach Neoplasms/enzymology , Stomach Neoplasms/immunology
11.
J Immunol ; 183(12): 7752-60, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19933867

ABSTRACT

Previous cancer vaccination approaches have shown some efficiency in generating measurable immune responses, but they have rarely led to tumor regression. It is therefore possible that tumors emerge with the capacity to down-regulate immune counterparts, through the local production of immunosuppressive molecules, such as IDO. Although it is known that IDO exerts suppressive effects on T cell functions, the mechanisms of IDO regulation in tumor cells remain to be characterized. Here, we demonstrate that activated T cells can induce functional IDO expression in breast and kidney tumor cell lines, and that this is partly attributable to IFN-gamma. Moreover, we found that IL-13, a Th2 cytokine, has a negative modulatory effect on IDO expression. Furthermore, we report IDO expression in the majority of breast and kidney carcinoma samples, with infiltration of activated Th1-polarized T cells in human tumors. These findings demonstrate complex control of immune activity within tumors. Future immune therapeutic interventions should thus include strategies to counteract these negative mechanisms.


Subject(s)
Breast Neoplasms/immunology , Carcinoma, Renal Cell/immunology , Gene Expression Regulation, Enzymologic/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Lymphocyte Activation/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/immunology , Coculture Techniques , Humans , Immunophenotyping , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Interferon-gamma/metabolism , Interferon-gamma/physiology , Lymphocyte Activation/genetics , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Th1 Cells/enzymology , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/enzymology , Th2 Cells/pathology , Tumor Cells, Cultured
12.
Front Immunol ; 12: 667850, 2021.
Article in English | MEDLINE | ID: mdl-33995401

ABSTRACT

Proprotein convertases (PC) are a family of 9 serine proteases involved in the processing of cellular pro-proteins. They trigger the activation, inactivation or functional changes of many hormones, neuropeptides, growth factors and receptors. Therefore, these enzymes are essential for cellular homeostasis in health and disease. Nine PC subtilisin/kexin genes (PCSK1 to PCSK9) encoding for PC1/3, PC2, furin, PC4, PC5/6, PACE4, PC7, SKI-1/S1P and PCSK9 are known. The expression of PC1/3, PC2, PC5/6, Furin and PC7 in lymphoid organs such as lymph nodes, thymus and spleen has suggested a role for these enzymes in immunity. In fact, knock-out of Furin in T cells was associated with high secretion of pro-inflammatory cytokines and autoantibody production in mice. This suggested a key role for this enzyme in immune tolerance. Moreover, Furin through its proteolytic activity, regulates the suppressive functions of Treg and thus prevents chronic inflammation and autoimmune diseases. In macrophages, Furin is also involved in the regulation of their inflammatory phenotype. Similarly, PC1/3 inhibition combined with TLR4 stimulation triggers the activation of the NF-κB signaling pathway with an increased secretion of pro-inflammatory cytokines. Factors secreted by PC1/3 KD macrophages stimulated with LPS exert a chemoattractive effect on naive auxiliary T lymphocytes (Th0) and anti-tumoral activities. The link between TLR and PCs is thus very important in inflammatory response regulation. Furin regulates TL7 and TLR8 processing and trafficking whereas PC1/3 controls TLR4 and TLR9 trafficking. Since PC1/3 and Furin are key regulators of both the innate and adaptive immune responses their inhibition may play a major role in oncoimmune therapy. The role of PCs in the oncoimmune response and therapeutic strategies based on PCs inhibition are proposed in the present review.


Subject(s)
Adaptive Immunity , Immunity, Innate , Lymphocytes, Tumor-Infiltrating/enzymology , Neoplasms/enzymology , Proprotein Convertases/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/enzymology , Animals , Histocompatibility Antigens Class I/metabolism , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Neoplasms/therapy , Signal Transduction , Toll-Like Receptors/metabolism , Tumor-Associated Macrophages/immunology
13.
Biosci Rep ; 41(6)2021 06 25.
Article in English | MEDLINE | ID: mdl-34060602

ABSTRACT

Immune checkpoint inhibitor (ICI) immunotherapies have vastly improved therapeutic outcomes for patients with certain cancer types, but these responses only manifest in a small percentage of all cancer patients. The goal of the present study was to improve checkpoint therapy efficacy by utilizing an engineered vaccinia virus to improve the trafficking of lymphocytes to the tumor, given that such lymphocyte trafficking is positively correlated with patient checkpoint inhibitor response rates. We developed an oncolytic vaccinia virus (OVV) platform expressing manganese superoxide dismutase (MnSOD) for use as both a monotherapy and together with anti-PD-L1. Intratumoral OVV-MnSOD injection in immunocompetent mice resulted in inflammation within poorly immunogenic tumors, thereby facilitating marked tumor regression. OVV-MnSOD administration together with anti-PD-L1 further improved antitumor therapy outcomes in models in which these monotherapy approaches were ineffective. Overall, our results emphasize the value of further studying these therapeutic approaches in patients with minimally or non-inflammatory tumors.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/virology , Lymphoma/therapy , Oncolytic Virotherapy , Superoxide Dismutase/metabolism , Vaccinia virus/enzymology , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Drug Resistance, Neoplasm , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma/enzymology , Lymphoma/immunology , Lymphoma/virology , Mice, Inbred C57BL , Superoxide Dismutase/genetics , Tumor Burden , Tumor Microenvironment/immunology , Vaccinia virus/genetics , Vaccinia virus/pathogenicity
14.
Virchows Arch ; 479(6): 1153-1165, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34448895

ABSTRACT

In recent years, breakthroughs in the field of tumor immunotherapy with immune checkpoint inhibitors (ICIs) have made a therapeutic revolution, which has been shown to improve the prognosis of patients with hepatocellular carcinoma (HCC). Immune infiltrates represent a major component of tumor microenvironment (TME), and play an essential role in both tumor progression and therapeutic response. The major unmet challenge in tumor immunotherapy is exploring the intrinsic and extrinsic mechanisms of TME promoting the management of HCC. Lysyl oxidase like 3 (LOXL3) participates in the remodeling of extracellular matrix (ECM) and the cross-linking of collagen and elastic fibers. It has been reported that LOXL3 is associated with the development and tumorigenesis of multiple types of cancer. RNA sequencing data and corresponding clinical information were extracted from The Cancer Genome Atlas (TCGA) databases, then subjected to gene expression, tumor microenvironment, survival, enrichment analyses utilizing R packages. In this study, we first found that LOXL3 gene was upregulated in tumor tissues compared with the normal tissues. Furthermore, LOXL3 expression is positively correlated with the infiltration of multiple immune cells and the expression of immune checkpoint genes in HCC. Meanwhile, high LOXL3 expression predicted poor outcomes of the patients with HCC. Functional enrichment analysis suggested that LOXL3 was mainly linked to extracellular structure and matrix organization, cell-cell adhesion, and T cell activation. This is the first comprehensive study to indicate that LOXL3 is correlated with immune infiltrates and may serve as a novel biomarker predicting prognosis and immunotherapy in HCC.


Subject(s)
Amino Acid Oxidoreductases/analysis , Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Lymphocytes, Tumor-Infiltrating/enzymology , Tumor Microenvironment , Amino Acid Oxidoreductases/genetics , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , DNA Damage , DNA Methylation , DNA Repair , Databases, Genetic , Humans , Immune Checkpoint Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Prognosis , Up-Regulation
15.
J Immunother Cancer ; 9(4)2021 04.
Article in English | MEDLINE | ID: mdl-33883257

ABSTRACT

BACKGROUND: In the tumor microenvironment, tumor cells are able to suppress antitumor immunity by competing for essential nutrients, including amino acids. However, whether amino acid depletion modulates the activity of CD8+ tumor-infiltrating lymphocytes (TILs) is unclear. METHOD: In this study, we evaluated the roles of amino acids and the Rag complex in regulating mammalian target of rapamycin complex 1 (mTORC1) signaling in CD8+ TILs. RESULTS: We discovered that the Rag complex, particularly RagD, was crucial for CD8+ T-cell antitumor immunity. RagD expression was positively correlated with the antitumor response of CD8+ TILs in both murine syngeneic tumor xenografts and clinical human colon cancer samples. On RagD deficiency, CD8+ T cells were rendered more dysfunctional, as demonstrated by attenuation of mTORC1 signaling and reductions in proliferation and cytokine secretion. Amino acids maintained RagD-mediated mTORC1 translocation to the lysosome, thereby achieving maximal mTORC1 activity in CD8+ T cells. Moreover, the limited T-cell access to leucine (LEU), overshadowed by tumor cell amino acid consumption, led to impaired RagD-dependent mTORC1 activity. Finally, combined with antiprogrammed cell death protein 1 antibody, LEU supplementation improved T-cell immunity in MC38 tumor-bearing mice in vivo. CONCLUSION: Our results revealed that robust signaling of amino acids by RagD and downstream mTORC1 signaling were crucial for T-cell receptor-initiated antitumor immunity. The characterization the role of RagD and LEU in nutrient mTORC1 signaling in TILs might suggest potential therapeutic strategies based on the manipulation of RagD and its upstream pathway.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , Leucine/metabolism , Lymphocytes, Tumor-Infiltrating/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Melanoma, Experimental/enzymology , Monomeric GTP-Binding Proteins/metabolism , Skin Neoplasms/enzymology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Enzyme Activation , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Escape , Tumor Microenvironment
16.
Front Immunol ; 12: 624284, 2021.
Article in English | MEDLINE | ID: mdl-33717133

ABSTRACT

Natural killer (NK) cells are effector cells of the innate immune system involved in defense against virus-infected and transformed cells. The effector function of NK cells is linked to their ability to migrate to sites of inflammation or damage. Therefore, understanding the factors regulating NK cell migration is of substantial interest. Here, we show that in the absence of aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor, NK cells have reduced capacity to migrate and infiltrate tumors in vivo. Analysis of differentially expressed genes revealed that ankyrin repeat and SOCS Box containing 2 (Asb2) expression was dramatically decreased in Ahr-/- NK cells and that AhR ligands modulated its expression. Further, AhR directly regulated the promoter region of the Asb2 gene. Similar to what was observed with murine Ahr-/- NK cells, ASB2 knockdown inhibited the migration of human NK cells. Activation of AHR by its agonist FICZ induced ASB2-dependent filamin A degradation in NK cells; conversely, knockdown of endogenous ASB2 inhibited filamin A degradation. Reduction of filamin A increased the migration of primary NK cells and restored the invasion capacity of AHR-deficient NK cells. Our study introduces AHR as a new regulator of NK cell migration, through an AHR-ASB2-filamin A axis and provides insight into a potential therapeutic target for NK cell-based immunotherapies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement , Filamins/metabolism , Killer Cells, Natural/enzymology , Receptors, Aryl Hydrocarbon/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Humans , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred C57BL , Mice, Knockout , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Suppressor of Cytokine Signaling Proteins/genetics , Tumor Microenvironment , Ubiquitination
17.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33753566

ABSTRACT

BACKGROUND: Regorafenib and other multikinase inhibitors may enhance antitumor efficacy of anti-program cell death-1 (anti-PD1) therapy in hepatocellular carcinoma (HCC). Its immunomodulatory effects, besides anti-angiogenesis, were not clearly defined. METHODS: In vivo antitumor efficacy was tested in multiple syngeneic liver cancer models. Murine bone marrow-derived macrophages (BMDMs) were tested in vitro for modulation of polarization by regorafenib and activation of cocultured T cells. Markers of M1/M2 polarization were measured by quantitative reverse transcription PCR (RT-PCR), arginase activity, flow cytometry, and ELISA. Knockdown of p38 kinase and downstream Creb1/Klf4 signaling on macrophage polarization were confirmed by using knockdown of the upstream MAPK14 kinase, chemical p38 kinase inhibitor, and chromatin immunoprecipitation. RESULTS: Regorafenib (5 mg/kg/day, corresponding to about half of human clinical dosage) inhibited tumor growth and angiogenesis in vivo similarly to DC-101 (anti-VEGFR2 antibody) but produced higher T cell activation and M1 macrophage polarization, increased the ratio of M1/M2 polarized BMDMs and proliferation/activation of cocultured T cells in vitro, indicating angiogenesis-independent immunomodulatory effects. Suppression of p38 kinase phosphorylation and downstream Creb1/Klf4 activity in BMDMs by regorafenib reversed M2 polarization. Regorafenib enhanced antitumor efficacy of adoptively transferred antigen-specific T cells. Synergistic antitumor efficacy between regorafenib and anti-PD1 was associated with multiple immune-related pathways in the tumor microenvironment. CONCLUSION: Regorafenib may enhance antitumor immunity through modulation of macrophage polarization, independent of its anti-angiogenic effects. Optimization of regorafenib dosage for rational design of combination therapy regimen may improve the therapeutic index in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cyclic AMP Response Element-Binding Protein/metabolism , Liver Neoplasms/drug therapy , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Tumor-Associated Macrophages/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/immunology , Cell Line, Tumor , Coculture Techniques , Kruppel-Like Factor 4/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/immunology , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Signal Transduction , Tumor Microenvironment , Tumor-Associated Macrophages/enzymology , Tumor-Associated Macrophages/immunology
18.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33658304

ABSTRACT

INTRODUCTION: The use of immune-checkpoint inhibitors has drastically improved the management of patients with non-small cell lung cancer (NSCLC), but innate and acquired resistances are hurdles needed to be solved. Immunomodulatory drugs that can reinvigorate the immune cytotoxic activity, in combination with antiprogrammed cell death 1 (PD-1) antibody, are a great promise to overcome resistance. We evaluated the impact of the SRC family kinases (SFKs) on NSCLC prognosis, and the immunomodulatory effect of the SFK inhibitor dasatinib, in combination with anti-PD-1, in clinically relevant mouse models of NSCLC. METHODS: A cohort of patients from University Clinic of Navarra (n=116) was used to study immune infiltrates by multiplex immunofluorescence (mIF) and YES1 protein expression in tumor samples. Publicly available resources (TCGA, Km Plotter, and CIBERSORT) were used to study patient's survival based on expression of SFKs and tumor infiltrates. Syngeneic NSCLC mouse models 393P and UNSCC680AJ were used for in vivo drug testing. RESULTS: Among the SFK members, YES1 expression showed the highest association with poor prognosis. Patients with high YES1 tumor levels also showed high infiltration of CD4+/FOXP3+ cells (regulatory T cells (Tregs)), suggesting an immunosuppressive phenotype. After testing for YES1 expression in a panel of murine cell lines, 393P and UNSCC680AJ were selected for in vivo studies. In the 393P model, dasatinib+anti-PD-1 treatment resulted in synergistic activity, with 87% tumor regressions and development of immunological memory that impeded tumor growth when mice were rechallenged. In vivo depletion experiments further showed that CD8+ and CD4+ cells are necessary for the therapeutic effect of the combination. The antitumor activity was accompanied by a very significant decrease in the number of Tregs, which was validated by mIF in tumor sections. In the UNSCC680AJ model, the antitumor effects of dasatinib+anti-PD-1 were milder but similar to the 393P model. In in vitro assays, we demonstrated that dasatinib blocks proliferation and transforming growth factor beta-driven conversion of effector CD4+ cells into Tregs through targeting of phospholymphocyte-specific protein tyrosine kinase and downstream effectors pSTAT5 and pSMAD3. CONCLUSIONS: YES1 protein expression is associated with increased numbers of Tregs in patients with NSCLC. Dasatinib synergizes with anti-PD-1 to impair tumor growth in NSCLC experimental models. This study provides the preclinical rationale for the combined use of dasatinib and PD-1/programmed death-ligand 1 blockade to improve outcomes of patients with NSCLC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation/drug effects , Dasatinib/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-yes/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, 129 Strain , Phenotype , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment
19.
J Cancer Res Clin Oncol ; 146(10): 2607-2620, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32388585

ABSTRACT

PURPOSE: Programmed death ligand 1 (PD-L1) and indoleamine 2,3-dioxygenase 1 (IDO1) are immunosuppressive proteins known to be associated with poor prognosis in various cancers. However, their expression and clinical relevance in osteosarcoma remain unknown. In this study, the relationships of PD-L1 and IDO1 expression with clinicopathological features and prognosis were explored. METHODS: The expression of PD-L1, IDO1, CD3, CD4, and CD8 in 112 formalin-fixed, paraffin-embedded tumor tissues collected by biopsy or surgical resection from 56 osteosarcoma patients was evaluated immunohistochemically. Moreover, four osteosarcoma cell lines were evaluated for the effects of IFNγ on PD-L1 and IDO1 mRNA expression by real-time reverse-transcription polymerase chain reaction. RESULTS: In pre-neoadjuvant chemotherapy (NAC) primary specimens, 10 cases (17%) showed PD-L1 expression and 12 (21%) showed IDO1 expression. Six of ten cases (60%) with PD-L1 positivity co-expressed IDO1. In post-NAC metastatic lesions, the frequency of immunoexpression of PD-L1 and IDO1 was increased compared with that in pre-NAC specimens. PD-L1 and/or IDO1 expression was not associated with poor prognosis. PD-L1 immunoexpression was significantly associated with the infiltration of CD3+ T cells, CD4+ T cells, and CD8+ T cells; while, IDO1 immunoexpression was significantly associated with the infiltration of CD3+ T cells and CD4+ T cells. In all osteosarcoma cell lines, PD-L1 and IDO1 expression was upregulated by stimulation with IFNγ. CONCLUSION: Our results suggest that the PD-L1 and IDO1 immune checkpoint inhibitors may provide clinical benefit in osteosarcoma patients with metastatic lesions after conventional chemotherapy.


Subject(s)
B7-H1 Antigen/biosynthesis , Bone Neoplasms/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Osteosarcoma/immunology , Adolescent , Adult , B7-H1 Antigen/immunology , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Child , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Lymphocytes, Tumor-Infiltrating/enzymology , Male , Middle Aged , Neoplasm Staging , Osteosarcoma/enzymology , Osteosarcoma/pathology , Osteosarcoma/secondary , Prognosis , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/immunology , Young Adult
20.
Oxid Med Cell Longev ; 2020: 7095902, 2020.
Article in English | MEDLINE | ID: mdl-33312338

ABSTRACT

The formation of reactive oxygen species (ROS) by the myeloid cell NADPH oxidase NOX2 is critical for the destruction of engulfed microorganisms. However, recent studies imply that ROS, formed by NOX2+ myeloid cells in the malignant microenvironment, exert multiple actions of relevance to the growth and spread of neoplastic cells. By generating ROS, tumor-infiltrating myeloid cells and NOX2+ leukemic myeloid cells may thus (i) compromise the function and viability of adjacent cytotoxic lymphocytes, including natural killer (NK) cells and T cells, (ii) oxidize DNA to trigger cancer-promoting somatic mutations, and (iii) affect the redox balance in cancer cells to control their proliferation and survival. Here, we discuss the impact of NOX2-derived ROS for tumorigenesis, tumor progression, regulation of antitumor immunity, and metastasis. We propose that NOX2 may be a targetable immune checkpoint in cancer.


Subject(s)
Carcinogenesis , Leukemia , Mutation , NADPH Oxidase 2 , Neoplasm Proteins , Reactive Oxygen Species , Tumor Microenvironment , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/metabolism , Humans , Killer Cells, Natural/enzymology , Killer Cells, Natural/immunology , Leukemia/enzymology , Leukemia/genetics , Leukemia/immunology , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid Cells/enzymology , Myeloid Cells/immunology , NADPH Oxidase 2/genetics , NADPH Oxidase 2/immunology , NADPH Oxidase 2/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL