Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cancer Immunol Immunother ; 73(8): 148, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38832958

ABSTRACT

Immunotherapy is one of the most promising anti-cancer treatment. It involves activating the host's own immune system to eliminate cancer cells. Activation of cGAS-STING pathway is promising therapeutic approach for cancer immunotherapy. However, in human clinical trials, targeting cGAS-STING pathway results in insufficient or unsustainable anti-tumor response. To enhance its effectiveness, combination with other anti-cancer therapies seems essential to achieve synergistic systemic anti-tumor response.The aim of this study was to evaluate whether the combination of STING agonist-cGAMP with anti-vascular RGD-(KLAKLAK)2 peptide results in a better anti-tumor response in poorly immunogenic tumors with various STING protein and αvß3 integrin status.Combination therapy inhibited growth of murine breast carcinoma more effectively than melanoma. In melanoma, the administration of STING agonist alone was sufficient to obtain a satisfactory therapeutic effect. In both tumor models we have noted stimulation of innate immune response following cGAMP administration alone or in combination. The largest population of immune cells infiltrating the TME after therapy were activated NK cells. Increased infiltration of cytotoxic CD8+ T lymphocytes within the TME was only observed in melanoma tumors. However, they also expressed the "exhaustion" PD-1 receptor. In contrast, in breast carcinoma tumors each therapy caused the drop in the number of infiltrating CD8+ T cells.The obtained results indicate an additional therapeutic benefit from combining STING agonist with an anti-vascular agent. However, this effect depends on the type of tumor, the status of its microenvironment and the expression of specific proteins such as STING and αvß3 family integrin.


Subject(s)
Membrane Proteins , Animals , Mice , Membrane Proteins/agonists , Female , Humans , Oligopeptides/pharmacology , Nucleotides, Cyclic/pharmacology , Nucleotides, Cyclic/administration & dosage , Immunotherapy/methods , Mice, Inbred C57BL , Cell Line, Tumor , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
2.
Cell Rep ; 37(13): 110150, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965418

ABSTRACT

Enteric pathogens overcome barrier immunity within the intestinal environment that includes the endogenous flora. The microbiota produces diverse ligands, and the full spectrum of microbial products that are sensed by the epithelium and prime protective immunity is unknown. Using Drosophila, we find that the gut presents a high barrier to infection, which is partially due to signals from the microbiota, as loss of the microbiota enhances oral viral infection. We report cyclic dinucleotide (CDN) feeding is sufficient to protect microbiota-deficient flies from enhanced oral infection, suggesting that bacterial-derived CDNs induce immunity. Mechanistically, we find CDN protection is dSTING- and dTBK1-dependent, leading to NF-kB-dependent gene expression. Furthermore, we identify the apical nucleoside transporter, CNT2, as required for oral CDN protection. Altogether, our studies define a role for bacterial products in priming immune defenses in the gut.


Subject(s)
Alphavirus Infections/immunology , Antiviral Agents/pharmacology , Drosophila melanogaster/immunology , Enterocytes/immunology , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Nucleotides, Cyclic/administration & dosage , Alphavirus Infections/drug therapy , Alphavirus Infections/virology , Animals , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enterocytes/drug effects , Enterocytes/virology , Female , Immunity, Innate , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sindbis Virus/immunology
3.
J Immunol ; 206(9): 2015-2028, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33820855

ABSTRACT

The cGAS-cyclic GMP-AMP (cGAMP)-stimulator of IFN genes (STING) pathway induces a powerful type I IFN (IFN-I) response and is a prime candidate for augmenting immunity in cancer immunotherapy and vaccines. IFN-I also has immune-regulatory functions manifested in several autoimmune diseases and is a first-line therapy for relapsing-remitting multiple sclerosis. However, it is only moderately effective and can induce adverse effects and neutralizing Abs in recipients. Targeting cGAMP in autoimmunity is unexplored and represents a challenge because of the intracellular location of its receptor, STING. We used microparticle (MP)-encapsulated cGAMP to increase cellular delivery, achieve dose sparing, and reduce potential toxicity. In the C57BL/6 experimental allergic encephalomyelitis (EAE) model, cGAMP encapsulated in MPs (cGAMP MPs) administered therapeutically protected mice from EAE in a STING-dependent fashion, whereas soluble cGAMP was ineffective. Protection was also observed in a relapsing-remitting model. Importantly, cGAMP MPs protected against EAE at the peak of disease and were more effective than rIFN-ß. Mechanistically, cGAMP MPs showed both IFN-I-dependent and -independent immunosuppressive effects. Furthermore, it induced the immunosuppressive cytokine IL-27 without requiring IFN-I. This augmented IL-10 expression through activated ERK and CREB. IL-27 and subsequent IL-10 were the most important cytokines to mitigate autoreactivity. Critically, cGAMP MPs promoted IFN-I as well as the immunoregulatory cytokines IL-27 and IL-10 in PBMCs from relapsing-remitting multiple sclerosis patients. Collectively, this study reveals a previously unappreciated immune-regulatory effect of cGAMP that can be harnessed to restrain T cell autoreactivity.


Subject(s)
Cell-Derived Microparticles/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon Type I/immunology , Membrane Proteins/immunology , Nucleotides, Cyclic/immunology , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell-Derived Microparticles/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Humans , Interferon Type I/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Membrane Proteins/agonists , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Nucleotides, Cyclic/administration & dosage , Nucleotides, Cyclic/metabolism , Signal Transduction/drug effects
4.
Mol Ther ; 29(5): 1758-1771, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33571681

ABSTRACT

DNA methylation abnormality is closely related to tumor occurrence and development. Chemical inhibitors targeting DNA methyltransferase (DNMTis) have been used in treating cancer. However, the impact of DNMTis on antitumor immunity has not been well elucidated. In this study, we show that zebularine (a demethylating agent) treatment of cancer cells led to increased levels of interferon response in a cyclic guanosine monophosphate-AMP (cGAMP) synthase (cGAS)- and stimulator of interferon genes (STING)-dependent manner. This treatment also specifically sensitized the cGAS-STING pathway in response to DNA stimulation. Incorporation of zebularine into genomic DNA caused demethylation and elevated expression of a group of genes, including STING. Without causing DNA damage, zebularine led to accumulation of DNA species in the cytoplasm of treated cells. In syngeneic tumor models, administration of zebularine alone reduced tumor burden and extended mice survival. This effect synergized with cGAMP and immune checkpoint blockade therapy. The efficacy of zebularine was abolished in nude mice and in cGAS-/- or STING-/- mice, indicating its dependency on host immunity. Analysis of tumor cells indicates upregulation of interferon-stimulated genes (ISGs) following zebularine administration. Zebularine promoted infiltration of CD8 T cells and natural killer (NK) cells into tumor and therefore suppressed tumor growth. This study unveils the role of zebularine in sensitizing the cGAS-STING pathway to promote anti-tumor immunity and provides the foundation for further therapeutic development.


Subject(s)
Cytidine/analogs & derivatives , Melanoma, Experimental/drug therapy , Membrane Proteins/genetics , Nucleotides, Cyclic/administration & dosage , Nucleotidyltransferases/genetics , Administration, Oral , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytidine/administration & dosage , Cytidine/pharmacology , Drug Synergism , Humans , Killer Cells, Natural/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Mice, Nude , Nucleotides, Cyclic/pharmacology , Promoter Regions, Genetic , THP-1 Cells , Tumor Microenvironment , Xenograft Model Antitumor Assays
5.
Nat Biomed Eng ; 5(5): 455-466, 2021 05.
Article in English | MEDLINE | ID: mdl-33558734

ABSTRACT

The stimulator of interferon genes (STING) is an endoplasmic reticulum transmembrane protein that is a target of therapeutics for infectious diseases and cancer. However, early-phase clinical trials of small-molecule STING agonists have shown limited antitumour efficacy and dose-limiting toxicity. Here, we show that a polyvalent STING agonist-a pH-sensitive polymer bearing a seven-membered ring with a tertiary amine (PC7A)-activates innate-immunity pathways through the polymer-induced formation of STING-PC7A condensates. In contrast to the natural STING ligand 2',3'-cyclic-GMP-AMP (cGAMP), PC7A stimulates the prolonged production of pro-inflammatory cytokines by binding to a non-competitive STING surface site that is distinct from the cGAMP binding pocket. PC7A induces antitumour responses that are dependent on STING expression and CD8+ T-cell activity, and the combination of PC7A and cGAMP led to synergistic therapeutic outcomes (including the activation of cGAMP-resistant STING variants) in mice bearing subcutaneous tumours and in resected human tumours and lymph nodes. The activation of the STING pathway through polymer-induced STING condensation may offer new therapeutic opportunities.


Subject(s)
Immunity, Innate , Membrane Proteins/agonists , Neoplasms/therapy , Nucleotides, Cyclic/administration & dosage , Polymers/administration & dosage , Animals , CD8-Positive T-Lymphocytes/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Neoplasms/immunology , Nucleotides, Cyclic/pharmacology , Polymers/pharmacology , THP-1 Cells
6.
Chem Commun (Camb) ; 57(4): 504-507, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33331360

ABSTRACT

A novel STING agonist, CDGSF, ipsilaterally modified with phosphorothioate and fluorine, was synthesized. The phosphorothioate in CDGSF might be a site for covalent conjugation. Injection of CDGSF generated an immunogenic ("hot") tumor microenvironment to suppress melanoma, more efficiently than dithio CDG. In particular, immunization with SARS-CoV-2 spike protein using CDGSF as an adjuvant elicited an exceptionally high antibody titer and a robust T cell response, overcoming the drawbacks of aluminum hydroxide. These results highlighted the therapeutic potential of CDGSF for cancer immunotherapy and the adjuvant potential of the STING agonist in the SARS-CoV-2 vaccine for the first time.


Subject(s)
Adjuvants, Immunologic/administration & dosage , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , Melanoma, Experimental/drug therapy , Membrane Proteins/agonists , Nucleotides, Cyclic/administration & dosage , Skin Neoplasms/drug therapy , Adjuvants, Immunologic/chemical synthesis , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/chemistry , Animals , Antibodies, Viral/biosynthesis , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/virology , COVID-19/immunology , COVID-19/virology , COVID-19 Vaccines/chemistry , Enzyme-Linked Immunospot Assay , Humans , Immunotherapy/methods , Interferon-gamma/biosynthesis , Melanoma, Experimental/immunology , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Nucleotides, Cyclic/chemical synthesis , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Vaccination/methods
7.
Nat Commun ; 11(1): 4909, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32999291

ABSTRACT

Effectively activating macrophages against cancer is promising but challenging. In particular, cancer cells express CD47, a 'don't eat me' signal that interacts with signal regulatory protein alpha (SIRPα) on macrophages to prevent phagocytosis. Also, cancer cells secrete stimulating factors, which polarize tumor-associated macrophages from an antitumor M1 phenotype to a tumorigenic M2 phenotype. Here, we report that hybrid cell membrane nanovesicles (known as hNVs) displaying SIRPα variants with significantly increased affinity to CD47 and containing M2-to-M1 repolarization signals can disable both mechanisms. The hNVs block CD47-SIRPα signaling axis while promoting M2-to-M1 repolarization within tumor microenvironment, significantly preventing both local recurrence and distant metastasis in malignant melanoma models. Furthermore, by loading a stimulator of interferon genes (STING) agonist, hNVs lead to potent tumor inhibition in a poorly immunogenic triple negative breast cancer model. hNVs are safe, stable, drug loadable, and suitable for genetic editing. These properties, combined with the capabilities inherited from source cells, make hNVs an attractive immunotherapy.


Subject(s)
Cell-Derived Microparticles/immunology , Immunotherapy/methods , Macrophages/immunology , Melanoma/therapy , Neoplasm Recurrence, Local/prevention & control , Triple Negative Breast Neoplasms/therapy , Animals , CD47 Antigen/metabolism , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , HEK293 Cells , Humans , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Melanoma/immunology , Melanoma/secondary , Membrane Proteins/agonists , Membrane Proteins/immunology , Mice , Nanoparticles/administration & dosage , Neoplasm Recurrence, Local/immunology , Nucleotides, Cyclic/administration & dosage , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Escape/drug effects , Tumor Escape/immunology , Tumor Microenvironment/immunology
8.
Science ; 367(6480)2020 02 21.
Article in English | MEDLINE | ID: mdl-32079747

ABSTRACT

Current influenza vaccines only confer protection against homologous viruses. We synthesized pulmonary surfactant (PS)-biomimetic liposomes encapsulating 2',3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), an agonist of the interferon gene inducer STING (stimulator of interferon genes). The adjuvant (PS-GAMP) vigorously augmented influenza vaccine-induced humoral and CD8+ T cell immune responses in mice by simulating the early phase of viral infection without concomitant excess inflammation. Two days after intranasal immunization with PS-GAMP-adjuvanted H1N1 vaccine, strong cross-protection was elicited against distant H1N1 and heterosubtypic H3N2, H5N1, and H7N9 viruses for at least 6 months while maintaining lung-resident memory CD8+ T cells. Adjuvanticity was then validated in ferrets. When alveolar epithelial cells (AECs) lacked Sting or gap junctions were blocked, PS-GAMP-mediated adjuvanticity was substantially abrogated in vivo. Thus, AECs play a pivotal role in configuring heterosubtypic immunity.


Subject(s)
Biomimetic Materials , Influenza Vaccines/immunology , Nanoparticles , Nucleotides, Cyclic/administration & dosage , Orthomyxoviridae Infections/prevention & control , Pulmonary Surfactants/immunology , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Biomimetic Materials/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Ferrets , Immunologic Memory , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/administration & dosage , Liposomes , Membrane Proteins/agonists , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Nanoparticles/administration & dosage , Nucleotides, Cyclic/pharmacology , Pulmonary Surfactants/administration & dosage
9.
Front Immunol ; 11: 583251, 2020.
Article in English | MEDLINE | ID: mdl-33603732

ABSTRACT

Current strategies for improving protective response to influenza vaccines during immunosenescence do not adequately protect individuals over 65 years of age. Here, we used an aged mouse model to investigate the potential of co-delivery of influenza vaccine with the recently identified combination of a saponin adjuvant Quil-A and an activator of the STING pathway, 2'3 cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) via dissolving microneedle patches (MNPs) applied to skin. We demonstrate that synergy between the two adjuvant components is observed after their incorporation with H1N1 vaccine into MNPs as revealed by analysis of the immune responses in adult mice. Aged 21-month-old mice were found to be completely protected against live influenza challenge after vaccination with the MNPs adjuvanted with the Quil-A/cGAMP combination (5 µg each) and demonstrated significantly reduced morbidity compared to the observed responses in these mice vaccinated with unadjuvanted MNPs. Analysis of the lung lysates of the surviving aged mice post challenge revealed the lowest level of residual inflammation in the adjuvanted groups. We conclude that combining influenza vaccine with a STING pathway activator and saponin-based adjuvant in MNPs is a novel option for skin vaccination of the immunosenescent population, which is at high risk for influenza.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Drug Delivery Systems/instrumentation , Influenza Vaccines/administration & dosage , Nucleotides, Cyclic/administration & dosage , Saponins/administration & dosage , Aging , Animals , Disease Models, Animal , Female , Humans , Immunosenescence/drug effects , Immunosenescence/immunology , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Transdermal Patch
10.
Nat Commun ; 10(1): 5108, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31704921

ABSTRACT

Mounting evidence suggests that the tumor microenvironment is profoundly immunosuppressive. Thus, mitigating tumor immunosuppression is crucial for inducing sustained antitumor immunity. Whereas previous studies involved intratumoral injection, we report here an inhalable nanoparticle-immunotherapy system targeting pulmonary antigen presenting cells (APCs) to enhance anticancer immunity against lung metastases. Inhalation of phosphatidylserine coated liposome loaded with STING agonist cyclic guanosine monophosphate-adenosine monophosphate (NP-cGAMP) in mouse models of lung metastases enables rapid distribution of NP-cGAMP to both lungs and subsequent uptake by APCs without causing immunopathology. NP-cGAMP designed for enhanced cytosolic release of cGAMP stimulates STING signaling and type I interferons production in APCs, resulting in the pro-inflammatory tumor microenvironment in multifocal lung metastases. Furthermore, fractionated radiation delivered to one tumor-bearing lung synergizes with inhaled NP-cGAMP, eliciting systemic anticancer immunity, controlling metastases in both lungs, and conferring long-term survival in mice with lung metastases and with repeated tumor challenge.


Subject(s)
Antigen-Presenting Cells/drug effects , Immunotherapy , Lung Neoplasms/secondary , Lung/drug effects , Melanoma, Experimental/secondary , Membrane Proteins/agonists , Nanoparticles , Nucleotides, Cyclic/pharmacology , Radiotherapy , Administration, Inhalation , Animals , Antigen-Presenting Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Interferon Type I/drug effects , Interferon Type I/immunology , Liposomes , Lung/immunology , Lung/radiation effects , Macrophages/drug effects , Macrophages/immunology , Mice , Nucleotides, Cyclic/administration & dosage , Phosphatidylserines
11.
J Immunother Cancer ; 7(1): 199, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31362778

ABSTRACT

BACKGROUND: Despite major advancements in immunotherapy among a number of solid tumors, response rates among ovarian cancer patients remain modest. Standard treatment for ovarian cancer is still surgery followed by taxane- and platinum-based chemotherapy. Thus, there is an urgent need to develop novel treatment options for clinical translation. METHODS: Our approach was to analyze the effects of standard chemotherapy in the tumor microenvironment of mice harboring orthotopic, syngeneic ID8-Vegf-Defb29 ovarian tumors in order to mechanistically determine a complementary immunotherapy combination. Specifically, we interrogated the molecular and cellular consequences of chemotherapy by analyzing gene expression and flow cytometry data. RESULTS: These data show that there is an immunosuppressive shift in the myeloid compartment, with increased expression of IL-10 and ARG1, but no activation of CD3+ T cells shortly after chemotherapy treatment. We therefore selected immunotherapies that target both the innate and adaptive arms of the immune system. Survival studies revealed that standard chemotherapy was complemented most effectively by a combination of anti-IL-10, 2'3'-cGAMP, and anti-PD-L1. Immunotherapy dramatically decreased the immunosuppressive myeloid population while chemotherapy effectively activated dendritic cells. Together, combination treatment increased the number of activated T and dendritic cells as well as expression of cytotoxic factors. It was also determined that the immunotherapy had to be administered concurrently with the chemotherapy to reverse the acute immunosuppression caused by chemotherapy. Mechanistic studies revealed that antitumor immunity in this context was driven by CD4+ T cells, which acquired a highly activated phenotype. Our data suggest that these CD4+ T cells can kill cancer cells directly via granzyme B-mediated cytotoxicity. Finally, we showed that this combination therapy is also effective at delaying tumor growth substantially in an aggressive model of lung cancer, which is also treated clinically with taxane- and platinum-based chemotherapy. CONCLUSIONS: This work highlights the importance of CD4+ T cells in tumor immunology. Furthermore, the data support the initiation of clinical trials in ovarian cancer that target both innate and adaptive immunity, with a focus on optimizing dosing schedules.


Subject(s)
Adaptive Immunity/drug effects , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Gene Expression Profiling/methods , Immunity, Innate/drug effects , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Carboplatin/administration & dosage , Carboplatin/pharmacology , Combined Modality Therapy , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-10/antagonists & inhibitors , Mice , Molecular Targeted Therapy , Nucleotides, Cyclic/administration & dosage , Nucleotides, Cyclic/pharmacology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Survival Analysis , Treatment Outcome , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
12.
Virology ; 531: 233-239, 2019 05.
Article in English | MEDLINE | ID: mdl-30928701

ABSTRACT

A persistent hepatitis B virus (HBV) infection is characterized by a lack of or a weak immune response to HBV. Efficient induction of the HBV-specific immune response leads to the clearance of HBV. Stimulator of interferon (IFN) genes (STING) is a cytoplasmic sensor of intracellular DNA from microbes and host cells. In the present study, we examined the efficacy of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) that is a ligand of the STING pathway as an HBV vaccine adjuvant. Wild-type (WT) mice and HBV-transgenic (HBV-Tg) mice were immunized with hepatitis B surface antigen (HBsAg) and cGAMP. The vaccination with HBsAg and cGAMP significantly enhanced the humoral and cellular immune response to HBsAg in WT and HBV-Tg mice. Cytokine production related to Th1 and Th2 responses and the activation of antigen-presenting cells in lymphoid tissues were induced by cGAMP. Vaccination using cGAMP may overcome tolerance in patients with chronic HBV infection.


Subject(s)
Hepatitis B Antibodies/immunology , Hepatitis B Vaccines/administration & dosage , Hepatitis B virus/physiology , Hepatitis B/immunology , Nucleotides, Cyclic/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Cytokines/immunology , Female , Hepatitis B/genetics , Hepatitis B/virology , Hepatitis B Surface Antigens/administration & dosage , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/immunology , Hepatitis B virus/genetics , Humans , Immunity, Cellular , Immunity, Humoral , Ligands , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Nucleotides, Cyclic/administration & dosage , Th1 Cells/immunology , Th2 Cells/immunology , Up-Regulation , Vaccination
13.
JCI Insight ; 4(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30944257

ABSTRACT

Pathogen recognition receptor (PRR) agonists are currently being developed and tested as adjuvants in various formulations to optimize the immunogenicity and efficacy of vaccines. Using an original in vitro approach to prime naive precursors from unfractionated human peripheral blood mononuclear cells, we assessed the influence of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), a ligand for the stimulator of interferon genes (STING), on the induction of antigen-specific CD8+ T cells. We found that 2'3'-cGAMP and 3'3'-cGAMP were especially potent adjuvants in this system, driving the expansion and maturation of functionally replete antigen-specific CD8+ T cells via the induction of type I IFNs. The biological relevance of these findings was confirmed in vivo using two mouse models, in which 2'3'-cGAMP-adjuvanted vaccination elicited protective antitumor or antiviral CD8+ T cell responses. These results identify particular isoforms of cGAMP as effective adjuvants that may find utility in the development of novel immunotherapies and vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Membrane Proteins/agonists , Nucleotides, Cyclic/immunology , Vaccination/methods , Animals , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Cells, Cultured , Disease Models, Animal , Female , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/immunology , Humans , Immunogenicity, Vaccine , Interferon Type I/immunology , Interferon Type I/metabolism , Ligands , Mice , Nucleotides, Cyclic/administration & dosage , Primary Cell Culture , Thymoma/immunology , Thymoma/pathology , Thymoma/prevention & control , Thymus Neoplasms/immunology , Thymus Neoplasms/pathology , Thymus Neoplasms/prevention & control , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
14.
JCI Insight ; 3(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30429378

ABSTRACT

Triple-negative breast cancer (TNBC) has few therapeutic options, and alternative approaches are urgently needed. Stimulator of IFN genes (STING) is becoming an exciting target for therapeutic adjuvants. However, STING resides inside the cell, and the intracellular delivery of CDNs, such as cGAMP, is required for the optimal activation of STING. We show that liposomal nanoparticle-delivered cGAMP (cGAMP-NP) activates STING more effectively than soluble cGAMP. These particles induce innate and adaptive host immune responses to preexisting tumors in both orthotopic and genetically engineered models of basal-like TNBC. cGAMP-NPs also reduce melanoma tumor load, with limited responsivity to anti-PD-L1. Within the tumor microenvironment, cGAMP-NPs direct both mouse and human macrophages (M), reprograming from protumorigenic M2-like phenotype toward M1-like phenotype; enhance MHC and costimulatory molecule expression; reduce M2 biomarkers; increase IFN-γ-producing T cells; augment tumor apoptosis; and increase CD4+ and CD8+ T cell infiltration. Activated T cells are required for tumor suppression, as their depletion reduces antitumor activity. Importantly, cGAMP-NPs prevent the formation of secondary tumors, and a single dose is sufficient to inhibit TNBC. These data suggest that a minimal system comprised of cGAMP-NP alone is sufficient to modulate the tumor microenvironment to effectively control PD-L1-insensitive TNBC.


Subject(s)
B7-H1 Antigen/immunology , Membrane Proteins/genetics , Nanoparticles/therapeutic use , Nucleotides, Cyclic/pharmacology , Triple Negative Breast Neoplasms/immunology , Animals , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunity, Innate/drug effects , Immunotherapy , Interferon Type I/genetics , Liposomes , Macrophages/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nucleotides, Cyclic/administration & dosage , T-Lymphocytes/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/therapy
15.
Mol Pharm ; 15(11): 4933-4946, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30281314

ABSTRACT

Vaccines are the most effective tool for preventing infectious diseases; however, subunit vaccines, considered the safest type, suffer from poor immunogenicity and require adjuvants to create a strong and sustained immune response. As adjuvants, pathogen-associated molecular patterns (PAMPs) offer potent immunostimulatory properties and defined mechanisms of action through their cognate pattern recognition receptors (PRRs). Their activity can be further enhanced through combining two or more PAMPs, particularly those that activate multiple immune signaling pathways. However, the cytosolic localization of many PRRs requires intracellular delivery of PAMPs for optimal biological activity, which is particularly true of the stimulator of interferon genes (STING) PRR. Using acetalated dextran (Ace-DEX) microparticles (MPs) encapsulating STING agonist 3'3'-cyclic GMP-AMP (cGAMP) combined with soluble PAMPS, we screened the effect of codelivery of adjuvants using primary mouse bone marrow derived dendritic cells (BMDCs). We identified that codelivery of cGAMP MPs and soluble Toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) elicited the broadest cytokine response. cGAMP and R848 were then coencapsulated within Ace-DEX MPs via electrospray. Using the model antigen ovalbumin, we observed that Ace-DEX MPs coencapsulating cGAMP and R848 (cGAMP/R848 Ace-DEX MPs) induced antigen-specific cellular immunity, and a balanced Th1/Th2 humoral response that was greater than cGAMP Ace-DEX MPs alone and PAMPs delivered in separate MPs. These data indicate that polymeric Ace-DEX MPs loaded with STING and TLR7/8 agonists represent a potent cellular and humoral vaccine adjuvant.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Drug Carriers/chemistry , Drug Compounding/methods , Pathogen-Associated Molecular Pattern Molecules/administration & dosage , Acetylation , Animals , Cells, Cultured , Dendritic Cells , Dextrans/chemistry , Female , Imidazoles/administration & dosage , Immunity, Cellular/drug effects , Immunogenicity, Vaccine , Male , Mice , Mice, Inbred C57BL , Models, Animal , Nucleotides, Cyclic/administration & dosage , Pathogen-Associated Molecular Pattern Molecules/immunology , Primary Cell Culture , Receptors, Pattern Recognition/antagonists & inhibitors , Receptors, Pattern Recognition/immunology , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/antagonists & inhibitors , Toll-Like Receptor 8/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
16.
Biomaterials ; 163: 67-75, 2018 05.
Article in English | MEDLINE | ID: mdl-29454236

ABSTRACT

Recent advancements in the field of immunotherapy have yielded encouraging results for the treatment of advanced cancers. Cyclic dinucleotides (CDNs) are a powerful new class of immunotherapy drugs known as STING (Stimulator of Interferon Genes) agonists, currently in clinical trials. However, previous studies of CDNs in murine cancer models have required multiple injections, and improve survival only in relatively nonaggressive tumor models. Therefore, we sought to improve the efficacy of CDN immunotherapy by developing a novel biomaterial we call "STINGel." STINGel is an injectable peptide hydrogel that localizes and provides controlled release of CDN delivery, showing an 8-fold slower release rate compared to a standard collagen hydrogel. The carrier hydrogel is a positively charged, MultiDomain Peptide (MDP) which self-assembles to form a nanofibrous matrix and is easily delivered by syringe. The highly localized delivery of CDN from this nanostructured biomaterial affects the local histological response in a subcutaneous model, and dramatically improves overall survival in a challenging murine model of head and neck cancer compared to CDN alone or CDN delivered from a collagen hydrogel. This study demonstrates the feasibility of biomaterial-based immunotherapy platforms like STINGel as strategies for increasing the efficacy of CDN immunotherapies.


Subject(s)
Antineoplastic Agents/chemistry , Neoplasms/therapy , Nucleotides, Cyclic/chemistry , Peptides/chemistry , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival , Collagen/chemistry , Delayed-Action Preparations , Dimerization , Drug Carriers/chemistry , Drug Liberation , Female , Humans , Hydrogels , Immunotherapy/methods , Injections, Intralesional , Mice, Inbred C57BL , Nanofibers/chemistry , Neoplasms/immunology , Nucleotides, Cyclic/administration & dosage , Particle Size , Static Electricity
17.
J Control Release ; 270: 1-13, 2018 01 28.
Article in English | MEDLINE | ID: mdl-29170142

ABSTRACT

Most FDA-approved adjuvants for infectious agents boost humoral but not cellular immunity, and have poorly-understood mechanisms. Stimulator of interferon genes (STING, also known as MITA, MPYS, or ERIS) is an exciting adjuvant target due to its role in cyclic dinucleotide (CDN)-driven anti-viral immunity; however, a major hindrance is STING's cytosolic localization which requires intracellular delivery of its agonists. As a result, STING agonists administered in a soluble form have elicited suboptimal immune responses. Delivery of STING agonists via particle platforms has proven a more successful strategy, but the opportunity for improved formulations and bioactivity remains. In this study we evaluated the adjuvant activity of the potent STING agonist, CDN 3'3'-cGAMP (cGAMP), encapsulated in acid-sensitive acetalated dextran (Ace-DEX) polymeric microparticles (MPs) which passively target antigen-presenting cells for intracellular release. This formulation was superior to all particle delivery systems evaluated and maintained its bioactivity following a sterilizing dose of gamma irradiation. Compared to soluble cGAMP, the Ace-DEX cGAMP MPs enhanced type-I interferon responses nearly 1000-fold in vitro and 50-fold in vivo, caused up to a 104-fold boost in antibody titers, increased Th1-associated responses, and expanded germinal center B cells and memory T cells. Furthermore, the encapsulated cGAMP elicited no observable toxicity in animals and achieved protective immunity against a lethal influenza challenge seven months post-immunization when using CDN adjuvant doses up to 100-fold lower than previous reports. For these reasons, Ace-DEX MP-encapsulated cGAMP represents a potent vaccine adjuvant of humoral and cellular immunity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Drug Carriers/administration & dosage , Membrane Proteins/immunology , Nucleotides, Cyclic/administration & dosage , Animals , Cells, Cultured , Dextrans/administration & dosage , Female , Immunity, Cellular , Immunity, Humoral , Male , Mice , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Vaccination
18.
Nanomedicine ; 14(2): 237-246, 2018 02.
Article in English | MEDLINE | ID: mdl-29127039

ABSTRACT

Therapeutic cancer vaccines require adjuvants leading to robust type I interferon and proinflammatory cytokine responses in the tumor microenvironment to induce an anti-tumor response. Cyclic dinucleotides (CDNs), a potent Stimulator of Interferon Receptor (STING) agonist, are currently in phase I trials. However, their efficacy may be limited to micromolar concentrations due to the cytosolic residence of STING in the ER membrane. Here we utilized biodegradable, poly(beta-amino ester) (PBAE) nanoparticles to deliver CDNs to the cytosol leading to robust immune response at >100-fold lower extracellular CDN concentrations in vitro. The leading CDN PBAE nanoparticle formulation induced a log-fold improvement in potency in treating established B16 melanoma tumors in vivo when combined with PD-1 blocking antibody in comparison to free CDN without nanoparticles. This nanoparticle-mediated cytosolic delivery method for STING agonists synergizes with checkpoint inhibitors and has strong potential for enhanced cancer immunotherapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunotherapy , Melanoma, Experimental/therapy , Membrane Proteins/agonists , Nanoparticles/administration & dosage , Nucleotides, Cyclic/administration & dosage , Animals , Antineoplastic Agents/chemistry , Female , Interferon Regulatory Factor-3/metabolism , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Nucleotides, Cyclic/chemistry , Polymers/chemistry , Tumor Cells, Cultured
19.
Sci Rep ; 7(1): 6355, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743914

ABSTRACT

Endogenous cyclic GMP-AMP (cGAMP) binds and activates STING to induce type I interferons. However, whether cGAMP plays any roles in regulating metabolic homeostasis remains unknown. Here we show that exogenous cGAMP ameliorates obesity-associated metabolic dysregulation and uniquely alters proinflammatory responses. In obese mice, treatment with cGAMP significantly decreases diet-induced proinflammatory responses in liver and adipose tissues and ameliorates metabolic dysregulation. Strikingly, cGAMP exerts cell-type-specific anti-inflammatory effects on macrophages, hepatocytes, and adipocytes, which is distinct from the effect of STING activation by DMXAA on enhancing proinflammatory responses. While enhancing insulin-stimulated Akt phosphorylation in hepatocytes and adipocytes, cGAMP weakens the effects of glucagon on stimulating hepatocyte gluconeogenic enzyme expression and glucose output and blunts palmitate-induced hepatocyte fat deposition in an Akt-dependent manner. Taken together, these results suggest an essential role for cGAMP in linking innate immunity and metabolic homeostasis, indicating potential applications of cGAMP in treating obesity-associated inflammatory and metabolic diseases.


Subject(s)
Adipocytes/immunology , Diet, High-Fat/adverse effects , Hepatocytes/immunology , Nucleotides, Cyclic/administration & dosage , Obesity/metabolism , Protein Serine-Threonine Kinases/metabolism , Adipocytes/drug effects , Animals , Hepatocytes/drug effects , Humans , Immunity, Innate , Interferon Type I/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Membrane Proteins/metabolism , Mice , Nucleotides, Cyclic/pharmacology , Obesity/chemically induced , Obesity/immunology , Phosphorylation , Xanthones/administration & dosage , Xanthones/pharmacology
20.
Cancer Immunol Immunother ; 66(6): 705-716, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28243692

ABSTRACT

Stimulator of IFN genes (STING) spontaneously contributes to anti-tumor immunity by inducing type I interferons (IFNs) following sensing of tumor-derived genomic DNAs in the tumor-bearing host. Although direct injection of STING ligands such as cyclic diguanylate monophosphate (c-di-GMP) and cyclic [G(2',5')pA(3',5')p] (cGAMP) into the tumor microenvironment exerts anti-tumor effects through strong induction of type I IFNs and activation of innate and adaptive immunity, the precise events caused by STING in the tumor microenvironment remain to be elucidated. We describe here our finding that a CD45+ CD11bmid Ly6C+ cell subset transiently accumulated in mouse tumor microenvironment of 4T1 breast cancer, squamous cell carcinomas, CT26 colon cancer, or B16F10 melanoma tissue after intratumoral injection of cGAMP. The accumulated cells displayed a macrophage (M ) phenotype since the cells were positive for F4/80 and MHC class II and negative for Ly6G. Intratumoral cGAMP treatment did not induce Mφ accumulation in STING-deficient mice. Depletion of CD8+ T cell using anti-CD8 mAb impaired the anti-tumor effects of cGAMP treatment. Depletion of the Mφ using clodronate liposomes impaired the anti-tumor effects of cGAMP treatment. Functional analysis indicated that the STING-triggered tumor-migrating Mφ exhibited phagocytic activity, production of tumor necrosis factor alpha TNFα), and high expression levels of T cell-recruiting chemokines, Cxcl10 and Cxcl11, IFN-induced molecules, MX dynamin-like GTPase 1 (Mx1) and 2'-5' oligoadenylate synthetase-like 1 (Oasl1), nitric oxide synthase 2 (Nos2), and interferon beta 1 (Ifnb1). These results indicate that the STING-triggered tumor-migrating Mφ participate in the anti-tumor effects of STING-activating compounds.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/immunology , Carcinoma, Squamous Cell/immunology , Colonic Neoplasms/immunology , Macrophages/immunology , Melanoma, Experimental/immunology , Nucleotides, Cyclic/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/prevention & control , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Female , Immunotherapy , Injections, Intralesional , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/drug effects , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nucleotides, Cyclic/pharmacology , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL
...