Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Nat Chem Biol ; 20(3): 365-372, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37828400

ABSTRACT

Stimulator of interferon genes (STING) is a dimeric transmembrane adapter protein that plays a key role in the human innate immune response to infection and has been therapeutically exploited for its antitumor activity. The activation of STING requires its high-order oligomerization, which could be induced by binding of the endogenous ligand, cGAMP, to the cytosolic ligand-binding domain. Here we report the discovery through functional screens of a class of compounds, named NVS-STGs, that activate human STING. Our cryo-EM structures show that NVS-STG2 induces the high-order oligomerization of human STING by binding to a pocket between the transmembrane domains of the neighboring STING dimers, effectively acting as a molecular glue. Our functional assays showed that NVS-STG2 could elicit potent STING-mediated immune responses in cells and antitumor activities in animal models.


Subject(s)
Adaptor Proteins, Signal Transducing , Membrane Proteins , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Biological Assay , Cytosol , Immunity, Innate , Ligands , Membrane Proteins/metabolism
3.
Cells ; 12(3)2023 01 25.
Article in English | MEDLINE | ID: mdl-36766748

ABSTRACT

Hepatitis C Virus NS3/NS4A, a serine protease complex, has been found to interact with many host proteins and cause various adverse effects on cellular function and immune response. For example, the cleavage of important immune factors by NS3/NS4A has been suggested as a mechanism for the hepatitis C virus to evade innate immunity. The spectrum of susceptible substrates for NS3/NS4A cleavage certainly includes important immune modulator kinases such as IKKα, IKKß, IKKε, and TBK1, as demonstrated in this paper. We show that the kinase activities of these four host kinases were transformed in unexpected ways by NS3/NS4A. Treatment with NS3/NS4A caused a significant reduction in the kinase activities of both IKKα and IKKß, suggesting that HCV might use its NS3/NS4A protease activity to deactivate the NF-κB-associated innate immune responses. In contrast, the kinase activities of both IKKε and TBK1 were enhanced after NS3/NS4A treatment, and more strikingly, the enhancement was more than 10-fold within 20 min of treatment. Our mass spectroscopic results suggested that the cleavage after Cys89 in the kinase domain of IKKε by NS3/NS4A led to their higher kinase activities, and three potential mechanisms were discussed. The observed kinase activity enhancement might facilitate the activation of both IKKε- and TBK1-dependent cellular antiviral pathways, likely contributing to spontaneous clearance of the virus and observed acute HCV infection. After longer than 20 min cleavage, both IKKε- and TBK1 gradually lost their kinase activities and the relevant antiviral pathways were expected to be inactivated, facilitating the establishment of chronic HCV infection.


Subject(s)
Hepatitis C , I-kappa B Kinase , Humans , I-kappa B Kinase/metabolism , Hepacivirus/metabolism , Viral Nonstructural Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Serine Proteases/metabolism , Antiviral Agents/metabolism
4.
Clin Cancer Res ; 29(1): 110-121, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36282874

ABSTRACT

PURPOSE: The stimulator of IFN genes (STING) is a transmembrane protein that plays a role in the immune response to tumors. Single-agent STING agonist MIW815 (ADU-S100) has demonstrated immune activation but limited antitumor activity. This phase Ib, multicenter, dose-escalation study assessed the safety and tolerability of MIW815 plus spartalizumab (PDR001), a humanized IgG4 antibody against PD-1, in 106 patients with advanced solid tumors or lymphomas. PATIENTS AND METHODS: Patients were treated with weekly intratumoral injections of MIW815 (50-3,200 µg) on a 3-weeks-on/1-week-off schedule or once every 4 weeks, plus a fixed dose of spartalizumab (400 mg) intravenously every 4 weeks. RESULTS: Common adverse events were pyrexia (n = 23; 22%), injection site pain (n = 21; 20%), and diarrhea (n = 12; 11%). Overall response rate was 10.4%. The MTD was not reached. Pharmacodynamic biomarker analysis demonstrated on-target activity. CONCLUSIONS: The combination of MIW815 and spartalizumab was well tolerated in patients with advanced/metastatic cancers, including in patients with anti-PD-1 refractory disease. Minimal antitumor responses were seen.


Subject(s)
Lymphoma , Neoplasms, Second Primary , Neoplasms , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/pathology , Lymphoma/drug therapy , Neoplasms, Second Primary/drug therapy
5.
Proc Natl Acad Sci U S A ; 119(22): e2200568119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35588144

ABSTRACT

Cyclic dinucleotides (CDN) and Toll-like receptor (TLR) ligands mobilize antitumor responses by natural killer (NK) cells and T cells, potentially serving as complementary therapies to immune checkpoint therapy. In the clinic thus far, however, CDN therapy targeting stimulator of interferon genes (STING) protein has yielded mixed results, perhaps because it initiates responses potently but does not provide signals to sustain activation and proliferation of activated cytotoxic lymphocytes. To improve efficacy, we combined CDN with a half life-extended interleukin-2 (IL-2) superkine, H9-MSA (mouse serum albumin). CDN/H9-MSA therapy induced dramatic long-term remissions of the most difficult to treat major histocompatibility complex class I (MHC I)­deficient and MHC I+ tumor transplant models. H9-MSA combined with CpG oligonucleotide also induced potent responses. Mechanistically, tumor elimination required CD8 T cells and not NK cells in the case of MHC I+ tumors and NK cells but not CD8 T cells in the case of MHC-deficient tumors. Furthermore, combination therapy resulted in more prolonged and more intense NK cell activation, cytotoxicity, and expression of cytotoxic effector molecules in comparison with monotherapy. Remarkably, in a primary autochthonous sarcoma model that is refractory to PD-1 checkpoint therapy, the combination of CDN/H9-MSA with checkpoint therapy yielded long-term remissions in the majority of the animals, mediated by T cells and NK cells. This combination therapy has the potential to activate responses in tumors resistant to current therapies and prevent MHC I loss accompanying acquired resistance of tumors to checkpoint therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Histocompatibility Antigens Class I , Immunotherapy , Interleukin-2 , Membrane Proteins , Neoplasms , Nucleotides, Cyclic , Oligodeoxyribonucleotides , Serum Albumin , Animals , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/genetics , Humans , Immunotherapy/methods , Interleukin-2/immunology , Killer Cells, Natural/immunology , Membrane Proteins/agonists , Mice , Neoplasms/genetics , Neoplasms/therapy , Nucleotides, Cyclic/therapeutic use , Oligodeoxyribonucleotides/therapeutic use , Serum Albumin/therapeutic use
6.
J Immunol ; 208(2): 407-419, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34965963

ABSTRACT

Tuberculosis consistently causes more deaths worldwide annually than any other single pathogen, making new effective vaccines an urgent priority for global public health. Among potential adjuvants, STING-activating cyclic dinucleotides (CDNs) uniquely stimulate a cytosolic sensing pathway activated only by pathogens. Recently, we demonstrated that a CDN-adjuvanted protein subunit vaccine robustly protects against tuberculosis infection in mice. In this study, we delineate the mechanistic basis underlying the efficacy of CDN vaccines for tuberculosis. CDN vaccines elicit CD4 T cells that home to lung parenchyma and penetrate into macrophage lesions in the lung. Although CDNs, like other mucosal vaccines, generate B cell-containing lymphoid structures in the lungs, protection is independent of B cells. Mucosal vaccination with a CDN vaccine induces Th1, Th17, and Th1-Th17 cells, and protection is dependent upon both IL-17 and IFN-γ. Single-cell RNA sequencing experiments reveal that vaccination enhances a metabolic state in Th17 cells reflective of activated effector function and implicate expression of Tnfsf8 (CD153) in vaccine-induced protection. Finally, we demonstrate that simply eliciting Th17 cells via mucosal vaccination with any adjuvant is not sufficient for protection. A vaccine adjuvanted with deacylated monophosphoryl lipid A (MPLA) failed to protect against tuberculosis infection when delivered mucosally, despite eliciting Th17 cells, highlighting the unique promise of CDNs as adjuvants for tuberculosis vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interleukin-17/immunology , Mycobacterium tuberculosis/immunology , Th17 Cells/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , CD30 Ligand/metabolism , Interferon-gamma/immunology , Lung/cytology , Lung/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Tuberculosis, Pulmonary/immunology , Vaccination
7.
Clin Cancer Res ; 28(4): 677-688, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34716197

ABSTRACT

PURPOSE: This phase I study assessed the safety, pharmacokinetics (PKs), and efficacy of MIW815 (ADU-S100), a novel synthetic cyclic dinucleotide that activates the stimulator of IFN genes (STING) pathway, in patients with advanced/metastatic cancers. PATIENTS AND METHODS: Patients (n = 47) received weekly i.t. injections of MIW815, 50 to 6,400 µg, on a 3-weeks-on/1-week-off schedule. RESULTS: A maximum tolerated dose was not reached. Most common treatment-related adverse events were pyrexia (17%), chills, and injection-site pain (each 15%). MIW815 was rapidly absorbed from the injection site with dose-proportional PK, a rapid terminal plasma half-life (approximately 24 minutes), and high interindividual variability. One patient had a partial response (PR; Merkel cell carcinoma); two patients had unconfirmed PR (parotid cancer, myxofibrosarcoma). Lesion size was stable or decreased in 94% of evaluable, injected lesions. RNA expression and immune infiltration assessments in paired tumor biopsies did not reveal significant on-treatment changes. However, increases in inflammatory cytokines and peripheral blood T-cell clonal expansion suggested systemic immune activation. CONCLUSIONS: MIW815 was well tolerated in patients with advanced/metastatic cancers. Clinical activity of single-agent MIW815 was limited in this first-in-human study; however, evidence of systemic immune activation was seen.


Subject(s)
Lymphoma , Neoplasms, Second Primary , Neoplasms , Adult , Humans , Immunotherapy , Maximum Tolerated Dose , Neoplasms/drug therapy , Neoplasms/pathology
8.
Immunity ; 53(1): 78-97, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32668230

ABSTRACT

Innate immune sensors that detect nucleic acids are attractive targets for therapeutic intervention because of their diverse roles in many disease processes. In detecting RNA and DNA from either self or non-self, nucleic acid sensors mediate the pathogenesis of many autoimmune and inflammatory conditions. Despite promising pre-clinical data and investigational use in the clinic, relatively few drugs targeting nucleic acid sensors are approved for therapeutic use. Nevertheless, there is growing appreciation for the untapped potential of nucleic acid sensors as therapeutic targets, driven by the need for better therapies for cancer, infectious diseases, and autoimmune disorders. This review highlights the diverse mechanisms by which nucleic acid sensors are activated and exert their biological effects in the context of various disease settings. We discuss current therapeutic strategies utilizing agonists and antagonists targeting nucleic acid sensors to treat infectious disease, cancer, and autoimmune and inflammatory disorders.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , DNA/immunology , Immunity, Innate/immunology , RNA/immunology , Animals , Humans , Immunologic Factors/pharmacology , Neoplasms/immunology , Neoplasms/therapy , Receptors, Pattern Recognition/immunology , Signal Transduction/immunology
9.
Nature ; 579(7800): E12, 2020 03.
Article in English | MEDLINE | ID: mdl-32144410

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Sci Immunol ; 5(45)2020 03 20.
Article in English | MEDLINE | ID: mdl-32198222

ABSTRACT

Several immunotherapy approaches that mobilize CD8+ T cell responses stimulate tumor rejection, and some, such as checkpoint blockade, have been approved for several cancer indications and show impressive increases in patient survival. However, tumors may evade CD8+ T cell recognition via loss of MHC molecules or because they contain few or no neoantigens. Therefore, approaches are needed to combat CD8+ T cell-resistant cancers. STING-activating cyclic dinucleotides (CDNs) are a new class of immune-stimulating agents that elicit impressive CD8+ T cell-mediated tumor rejection in preclinical tumor models and are now being tested in clinical trials. Here, we demonstrate powerful CDN-induced, natural killer (NK) cell-mediated tumor rejection in numerous tumor models, independent of CD8+ T cells. CDNs enhanced NK cell activation, cytotoxicity, and antitumor effects in part by inducing type I interferon (IFN). IFN acted in part directly on NK cells in vivo and in part indirectly via the induction of IL-15 and IL-15 receptors, which were important for CDN-induced NK activation and tumor control. After in vivo administration of CDNs, dendritic cells (DCs) up-regulated IL-15Rα in an IFN-dependent manner. Mice lacking the type I IFN receptor specifically on DCs had reduced NK cell activation and tumor control. Therapeutics that activate NK cells, such as CDNs, checkpoint inhibitors, NK cell engagers, and cytokines, may represent next-generation approaches to cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Membrane Proteins/agonists , Neoplasms/drug therapy , Neoplasms/immunology , Animals , CRISPR-Cas Systems/drug effects , CRISPR-Cas Systems/immunology , Interferon Type I/pharmacology , Killer Cells, Natural/drug effects , Membrane Proteins/immunology , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/pathology , Tumor Cells, Cultured
12.
Nature ; 573(7774): 434-438, 2019 09.
Article in English | MEDLINE | ID: mdl-31511694

ABSTRACT

The accumulation of DNA in the cytosol serves as a key immunostimulatory signal associated with infections, cancer and genomic damage1,2. Cytosolic DNA triggers immune responses by activating the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway3. The binding of DNA to cGAS activates its enzymatic activity, leading to the synthesis of a second messenger, cyclic guanosine monophosphate-adenosine monophosphate (2'3'-cGAMP)4-7. This cyclic dinucleotide (CDN) activates STING8, which in turn activates the transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), promoting the transcription of genes encoding type I interferons and other cytokines and mediators that stimulate a broader immune response. Exogenous 2'3'-cGAMP produced by malignant cells9 and other CDNs, including those produced by bacteria10-12 and synthetic CDNs used in cancer immunotherapy13,14, must traverse the cell membrane to activate STING in target cells. How these charged CDNs pass through the lipid bilayer is unknown. Here we used a genome-wide CRISPR-interference screen to identify the reduced folate carrier SLC19A1, a folate-organic phosphate antiporter, as the major transporter of CDNs. Depleting SLC19A1 in human cells inhibits CDN uptake and functional responses, and overexpressing SLC19A1 increases both uptake and functional responses. In human cell lines and primary cells ex vivo, CDN uptake is inhibited by folates as well as two medications approved for treatment of inflammatory diseases, sulfasalazine and the antifolate methotrexate. The identification of SLC19A1 as the major transporter of CDNs into cells has implications for the immunotherapeutic treatment of cancer13, host responsiveness to CDN-producing pathogenic microorganisms11 and-potentially-for some inflammatory diseases.


Subject(s)
DNA/metabolism , Nucleotides, Cyclic/metabolism , Reduced Folate Carrier Protein/metabolism , Animals , Cytosol , DNA/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferon Regulatory Factor-3/metabolism , Nucleotides, Cyclic/immunology , Nucleotidyltransferases/metabolism , Reduced Folate Carrier Protein/immunology
13.
Cell Rep ; 25(11): 3074-3085.e5, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540940

ABSTRACT

Intratumoral (IT) STING activation results in tumor regression in preclinical models, yet factors dictating the balance between innate and adaptive anti-tumor immunity are unclear. Here, clinical candidate STING agonist ADU-S100 (S100) is used in an IT dosing regimen optimized for adaptive immunity to uncover requirements for a T cell-driven response compatible with checkpoint inhibitors (CPIs). In contrast to high-dose tumor ablative regimens that result in systemic S100 distribution, low-dose immunogenic regimens induce local activation of tumor-specific CD8+ effector T cells that are responsible for durable anti-tumor immunity and can be enhanced with CPIs. Both hematopoietic cell STING expression and signaling through IFNAR are required for tumor-specific T cell activation, and in the context of optimized T cell responses, TNFα is dispensable for tumor control. In a poorly immunogenic model, S100 combined with CPIs generates a survival benefit and durable protection. These results provide fundamental mechanistic insights into STING-induced anti-tumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity , Membrane Proteins/metabolism , Neoplasms/immunology , Animals , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Cytokines/metabolism , Dose-Response Relationship, Immunologic , Drug Resistance, Neoplasm , Hematopoiesis , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/pathology , Programmed Cell Death 1 Receptor/metabolism , S100 Proteins/administration & dosage , S100 Proteins/immunology
14.
JCI Insight ; 3(20)2018 10 18.
Article in English | MEDLINE | ID: mdl-30333318

ABSTRACT

Tumor neoantigens arising from somatic mutations in the cancer genome are less likely to be subject to central immune tolerance and are therefore attractive targets for vaccine immunotherapy. We utilized whole-exome sequencing, RNA sequencing (RNASeq), and an in silico immunogenicity prediction algorithm, NetMHC, to generate a neoantigen-targeted vaccine, PancVAX, which was administered together with the STING adjuvant ADU-V16 to mice bearing pancreatic adenocarcinoma (Panc02) cells. PancVAX activated a neoepitope-specific T cell repertoire within the tumor and caused transient tumor regression. When given in combination with two checkpoint modulators, namely anti-PD-1 and agonist OX40 antibodies, PancVAX resulted in enhanced and more durable tumor regression and a survival benefit. The addition of OX40 to vaccine reduced the coexpression of T cell exhaustion markers, Lag3 and PD-1, and resulted in rejection of tumors upon contralateral rechallenge, suggesting the induction of T cell memory. Together, these data provide the framework for testing personalized neoantigen-based combinatorial vaccine strategies in patients with pancreatic and other nonimmunogenic cancers.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/administration & dosage , Immunotherapy/methods , Pancreatic Neoplasms/therapy , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Combined Modality Therapy/methods , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Immunogenicity, Vaccine , Membrane Proteins/immunology , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, OX40/agonists , Receptors, OX40/immunology , Treatment Outcome , Tumor Escape/drug effects , Tumor Escape/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
15.
Cell Rep ; 23(5): 1435-1447, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29719256

ABSTRACT

There are a limited number of adjuvants that elicit effective cell-based immunity required for protection against intracellular bacterial pathogens. Here, we report that STING-activating cyclic dinucleotides (CDNs) formulated in a protein subunit vaccine elicit long-lasting protective immunity to Mycobacterium tuberculosis in the mouse model. Subcutaneous administration of this vaccine provides equivalent protection to that of the live attenuated vaccine strain Bacille Calmette-Guérin (BCG). Protection is STING dependent but type I IFN independent and correlates with an increased frequency of a recently described subset of CXCR3-expressing T cells that localize to the lung parenchyma. Intranasal delivery results in superior protection compared with BCG, significantly boosts BCG-based immunity, and elicits both Th1 and Th17 immune responses, the latter of which correlates with enhanced protection. Thus, a CDN-adjuvanted protein subunit vaccine has the capability of eliciting a multi-faceted immune response that results in protection from infection by an intracellular pathogen.


Subject(s)
Adjuvants, Immunologic/pharmacology , BCG Vaccine/pharmacology , Membrane Proteins/immunology , Mycobacterium tuberculosis/immunology , Th17 Cells/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , BCG Vaccine/immunology , Disease Models, Animal , Immunity, Cellular/drug effects , Mice , Mice, Knockout , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/pathology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
16.
Cancer Immunol Res ; 6(4): 422-433, 2018 04.
Article in English | MEDLINE | ID: mdl-29472271

ABSTRACT

The cGAS-STING cytosolic DNA sensing pathway may play an integral role in the initiation of antitumor immune responses. Studies evaluating the immunogenicity of various cyclic dinucleotide (CDN) STING agonists administered by intratumoral (i.t.) injection showed potent induction of inflammation, tumor necrosis, and, in some cases, durable tumor-specific adaptive immunity. However, the specific immune mechanisms underlying these responses remain incompletely defined. The majority of these studies have focused on the effect of CDNs on immune cells but have not conclusively interrogated the role of stromal cells in the acute rejection of the CDN-injected tumor. Here, we revealed a mechanism of STING agonist-mediated tumor response that relied on both stromal and immune cells to achieve tumor regression and clearance. Using knockout and bone marrow chimeric mice, we showed that although bone marrow-derived TNFα was necessary for CDN-induced necrosis, STING signaling in radioresistant stromal cells was also essential for CDN-mediated tumor rejection. These results provide evidence for crosstalk between stromal and hematopoietic cells during CDN-mediated tumor collapse after i.t. administration. These mechanistic insights may prove critical in the clinical development of STING agonists. Cancer Immunol Res; 6(4); 422-33. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Membrane Proteins/agonists , Neoplasms/etiology , Neoplasms/metabolism , Nucleotides, Cyclic/pharmacology , Radiation Tolerance , Stromal Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Bone Marrow/metabolism , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Immunity, Innate , Interferon-beta/metabolism , Melanoma, Experimental , Mice , Mice, Knockout , Necrosis/metabolism , Necrosis/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Signal Transduction/drug effects , Stromal Cells/pathology , Stromal Cells/radiation effects , Tumor Burden/drug effects , Tumor Microenvironment/immunology
18.
J Clin Invest ; 126(7): 2404-11, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27367184

ABSTRACT

A major subset of human cancers shows evidence for spontaneous adaptive immunity, which is reflected by the presence of infiltrating CD8+ T cells specific for tumor antigens within the tumor microenvironment. This observation has raised the question of which innate immune sensing pathway might detect the presence of cancer and lead to a natural adaptive antitumor immune response in the absence of exogenous infectious pathogens. Evidence for a critical functional role for type I IFNs led to interrogation of candidate innate immune sensing pathways that might be triggered by tumor presence and induce type I IFN production. Such analyses have revealed a major role for the stimulator of IFN genes pathway (STING pathway), which senses cytosolic tumor-derived DNA within the cytosol of tumor-infiltrating DCs. Activation of this pathway is correlated with IFN-ß production and induction of antitumor T cells. Based on the biology of this natural immune response, pharmacologic agonists of the STING pathway are being developed to augment and optimize STING activation as a cancer therapy. Intratumoral administration of STING agonists results in remarkable therapeutic activity in mouse models, and STING agonists are being carried forward into phase I clinical testing.


Subject(s)
Interferons/immunology , Neoplasms/immunology , Neoplasms/metabolism , Adaptive Immunity , Animals , Antigen-Presenting Cells/immunology , Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/cytology , Cytosol/metabolism , DNA, Neoplasm/analysis , Dendritic Cells/immunology , Humans , Immunity, Innate , Interferon-beta/immunology , Mice , Tumor Microenvironment/immunology
19.
J Immunol ; 196(7): 3191-8, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26927800

ABSTRACT

Recent evidence has indicated that innate immune sensing of cytosolic DNA in dendritic cells via the host STING pathway is a major mechanism leading to spontaneous T cell responses against tumors. However, the impact of the other major pathway triggered by intracellular DNA, the absent in melanoma 2 (AIM2) inflammasome, on the functional output from the stimulator of IFN genes (STING) pathway is poorly understood. We found that dendritic cells and macrophages deficient in AIM2, apoptosis-associated specklike protein, or caspase-1 produced markedly higher IFN-ß in response to DNA. Biochemical analyses showed enhanced generation of cyclic GMP-AMP, STING aggregation, and TANK-binding kinase 1 and IFN regulatory factor 3 phosphorylation in inflammasome-deficient cells. Induction of pyroptosis by the AIM2 inflammasome was a major component of this effect, and inhibition of caspase-1 reduced cell death, augmenting phosphorylation of TANK-binding kinase 1/IFN regulatory factor 3 and production of IFN-ß. Our data suggest that in vitro activation of the AIM2 inflammasome in murine macrophages and dendritic cells leads to reduced activation of the STING pathway, in part through promoting caspase-1-dependent cell death.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/immunology , DNA/metabolism , Inflammasomes , Membrane Proteins/metabolism , Signal Transduction , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Caspase 1/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Interferon-gamma/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Nucleotidyltransferases/metabolism , Pyroptosis/genetics , Pyroptosis/immunology
20.
Cell Rep ; 11(7): 1018-30, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25959818

ABSTRACT

Spontaneous tumor-initiated T cell priming is dependent on IFN-ß production by tumor-resident dendritic cells. On the basis of recent observations indicating that IFN-ß expression was dependent upon activation of the host STING pathway, we hypothesized that direct engagement of STING through intratumoral (IT) administration of specific agonists would result in effective anti-tumor therapy. After proof-of-principle studies using the mouse STING agonist DMXAA showed a potent therapeutic effect, we generated synthetic cyclic dinucleotide (CDN) derivatives that activated all human STING alleles as well as murine STING. IT injection of STING agonists induced profound regression of established tumors in mice and generated substantial systemic immune responses capable of rejecting distant metastases and providing long-lived immunologic memory. Synthetic CDNs have high translational potential as a cancer therapeutic.


Subject(s)
Antineoplastic Agents/pharmacology , Membrane Proteins/antagonists & inhibitors , Neoplasms, Experimental/immunology , Nucleotides, Cyclic/pharmacology , Tumor Microenvironment/immunology , Animals , Antineoplastic Agents/chemical synthesis , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Gene Knockout Techniques , Humans , Macrophages , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Nucleotides, Cyclic/chemical synthesis , Polymerase Chain Reaction , Transfection , Xanthones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...