Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 342
Filter
1.
Nat Commun ; 15(1): 5842, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992037

ABSTRACT

Activating interferon responses with STING agonists (STINGa) is a current cancer immunotherapy strategy, and therapeutic modalities that enable tumor-targeted delivery via systemic administration could be beneficial. Here we demonstrate that tumor cell-directed STING agonist antibody-drug-conjugates (STINGa ADCs) activate STING in tumor cells and myeloid cells and induce anti-tumor innate immune responses in in vitro, in vivo (in female mice), and ex vivo tumor models. We show that the tumor cell-directed STINGa ADCs are internalized into myeloid cells by Fcγ-receptor-I in a tumor antigen-dependent manner. Systemic administration of STINGa ADCs in mice leads to STING activation in tumors, with increased anti-tumor activity and reduced serum cytokine elevations compared to a free STING agonist. Furthermore, STINGa ADCs induce type III interferons, which contribute to the anti-tumor activity by upregulating type I interferon and other key chemokines/cytokines. These findings reveal an important role for type III interferons in the anti-tumor activity elicited by STING agonism and provide rationale for the clinical development of tumor cell-directed STINGa ADCs.


Subject(s)
Immunity, Innate , Immunoconjugates , Interferons , Membrane Proteins , Animals , Membrane Proteins/agonists , Membrane Proteins/immunology , Immunity, Innate/drug effects , Female , Humans , Mice , Cell Line, Tumor , Immunoconjugates/pharmacology , Immunoconjugates/administration & dosage , Interferons/metabolism , Interferon Lambda , Neoplasms/immunology , Neoplasms/drug therapy , Interferon Type I/immunology , Cytokines/metabolism , Myeloid Cells/immunology , Myeloid Cells/drug effects , Immunotherapy/methods , Mice, Inbred C57BL , Receptors, IgG/agonists , Receptors, IgG/metabolism , Receptors, IgG/immunology
2.
ACS Appl Bio Mater ; 7(6): 3877-3889, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38832760

ABSTRACT

Adjuvants and immunomodulators that effectively drive a Th17-skewed immune response are not part of the standard vaccine toolkit. Vaccine adjuvants and delivery technologies that can induce Th17 or Th1/17 immunity and protection against bacterial pathogens, such as tuberculosis (TB), are urgently needed. Th17-polarized immune response can be induced using agonists that bind and activate C-type lectin receptors (CLRs) such as macrophage inducible C-type lectin (Mincle). A simple but effective strategy was developed for codelivering Mincle agonists with the recombinant Mycobacterium tuberculosis fusion antigen, M72, using tunable silica nanoparticles (SNP). Anionic bare SNP, hydrophobic phenyl-functionalized SNP (P-SNP), and cationic amine-functionalized SNP (A-SNP) of different sizes were coated with three synthetic Mincle agonists, UM-1024, UM-1052, and UM-1098, and evaluated for adjuvant activity in vitro and in vivo. The antigen and adjuvant were coadsorbed onto SNP via electrostatic and hydrophobic interactions, facilitating multivalent display and delivery to antigen presenting cells. The cationic A-SNP showed the highest coloading efficiency for the antigen and adjuvant. In addition, the UM-1098-adsorbed A-SNP formulation demonstrated slow-release kinetics in vitro, excellent stability over 12 months of storage, and strong IL-6 induction from human peripheral blood mononuclear cells. Co-adsorption of UM-1098 and M72 on A-SNP significantly improved antigen-specific humoral and Th17-polarized immune responses in vivo in BALB/c mice relative to the controls. Taken together, A-SNP is a promising platform for codelivery and proper presentation of adjuvants and antigens and provides the basis for their further development as a vaccine delivery platform for immunization against TB or other diseases for which Th17 immunity contributes to protection.


Subject(s)
Antigens, Bacterial , Lectins, C-Type , Nanoparticles , Silicon Dioxide , Th17 Cells , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/agonists , Nanoparticles/chemistry , Th17 Cells/immunology , Animals , Silicon Dioxide/chemistry , Mice , Antigens, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/chemistry , Mycobacterium tuberculosis/immunology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Particle Size , Materials Testing , Humans , Female , Membrane Proteins/immunology , Membrane Proteins/agonists
3.
Viruses ; 16(6)2024 May 27.
Article in English | MEDLINE | ID: mdl-38932148

ABSTRACT

The devastating effects of COVID-19 have highlighted the importance of prophylactic and therapeutic strategies to combat respiratory diseases. Stimulator of interferon gene (STING) is an essential component of the host defense mechanisms against respiratory viral infections. Although the role of the cGAS/STING signaling axis in the innate immune response to DNA viruses has been thoroughly characterized, mounting evidence shows that it also plays a key role in the prevention of RNA virus infections. In this study, we investigated the role of STING activation during Influenza virus (IFV) infection. In both mouse bone marrow-derived macrophages and monocytic cell line THP-1 differentiated with PMA, we found that dimeric amidobenzimidazole (diABZI), a STING agonist, had substantial anti-IFV activity against multiple strains of IFV, including A/H1N1, A/H3N2, B/Yamagata, and B/Victoria. On the other hand, a pharmacological antagonist of STING (H-151) or the loss of STING in human macrophages leads to enhanced viral replication but suppressed IFN expression. Furthermore, diABZI was antiviral against IFV in primary air-liquid interface cultures of nasal epithelial cells. Our data suggest that STING agonists may serve as promising therapeutic antiviral agents to combat IFV.


Subject(s)
Antiviral Agents , Immunity, Innate , Macrophages , Membrane Proteins , Animals , Humans , Immunity, Innate/drug effects , Mice , Antiviral Agents/pharmacology , Macrophages/immunology , Macrophages/drug effects , Macrophages/virology , Membrane Proteins/agonists , Membrane Proteins/metabolism , Membrane Proteins/genetics , THP-1 Cells , Virus Replication/drug effects , Influenza, Human/immunology , Influenza, Human/virology , Influenza, Human/drug therapy , Dogs , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Orthomyxoviridae/drug effects , Orthomyxoviridae/immunology , Orthomyxoviridae/physiology , Benzimidazoles/pharmacology , Signal Transduction/drug effects
4.
ACS Nano ; 18(24): 15499-15516, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38832815

ABSTRACT

T cell exhaustion has emerged as a major hurdle that impedes the clinical translation of stimulator of interferon genes (STING) agonists. It is crucial to explore innovative strategies to rejuvenate exhausted T cells and potentiate the antitumor efficacy. Here, we propose an approach utilizing MSA-2 as a STING agonist, along with nanoparticle-mediated delivery of mRNA encoding interleukin-12 (IL-12) to restore the function of T cells. We developed a lipid nanoparticle (DMT7-IL12 LNP) that encapsulated IL12 mRNA. Our findings convincingly demonstrated that the combination of MSA-2 and DMT7-IL12 LNP can effectively reverse the exhausted T cell phenotype, as evidenced by the enhanced secretion of cytokines, such as tumor necrosis factor alpha, interferon gamma, and Granzyme B, coupled with reduced levels of inhibitory molecules such as T cell immunoglobulin and mucin domain-3 and programmed cell death protein-1 on CD8+ T cells. Furthermore, this approach led to improved survival and tumor regression without causing any systemic toxicity in melanoma and lung metastasis models. These findings suggest that mRNA encoding IL-12 in conjunction with STING agonists has the potential to confer superior clinical outcomes, representing a promising advancement in cancer immunotherapy.


Subject(s)
Interleukin-12 , Mice, Inbred C57BL , RNA, Messenger , Interleukin-12/genetics , Animals , RNA, Messenger/genetics , RNA, Messenger/metabolism , Mice , Nanoparticles/chemistry , Membrane Proteins/agonists , Membrane Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Humans , Female , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Cell Line, Tumor , T-Cell Exhaustion
5.
Microbiologyopen ; 13(3): e23, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867416

ABSTRACT

The G protein-coupled estrogen receptor, also known as GPER1 or originally GPR30, is found in various tissues, indicating its diverse functions. It is typically present in immune cells, suggesting its role in regulating immune responses to infectious diseases. Our previous studies have shown that G-1, a selective GPER agonist, can limit the pathogenesis mediated by Staphylococcus aureus alpha-hemolysin (Hla). It aids in clearing bacteria in a mouse skin infection model and restricts the surface display of the Hla receptor, ADAM10 (a disintegrin and metalloprotease 10) in HaCaT keratinocytes. In this report, we delve into the modulation of GPER in human immune cells in relation to the NLRP3 inflammasome. We used macrophage-like differentiated THP-1 cells for our study. We found that treating these cells with G-1 reduces ATP release, decreases the activity of the caspase-1 enzyme, and lessens cell death following Hla intoxication. This is likely due to the reduced levels of ADAM10 and NLRP3 proteins, as well as the decreased display of the ADAM10 receptor in the G-1-treated THP-1 cells. Our studies, along with our previous work, suggest the potential therapeutic use of G-1 in reducing Hla susceptibility in humans. This highlights the importance of GPER in immune regulation and its potential as a therapeutic target.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Bacterial Toxins , Hemolysin Proteins , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Estrogen , Receptors, G-Protein-Coupled , Staphylococcus aureus , ADAM10 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Humans , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Hemolysin Proteins/metabolism , Inflammasomes/metabolism , Bacterial Toxins/metabolism , THP-1 Cells , Receptors, Estrogen/metabolism , Amyloid Precursor Protein Secretases/metabolism , Staphylococcus aureus/drug effects , Membrane Proteins/metabolism , Membrane Proteins/agonists , Caspase 1/metabolism , Adenosine Triphosphate/metabolism , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Dipeptides , Hydroxamic Acids
6.
Biomed Pharmacother ; 176: 116877, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38850654

ABSTRACT

Multiple myeloma (MM) progression is closely dependent on cells in the bone marrow (BM) microenvironment, including fibroblasts (FBs) and immune cells. In their BM niche, MM cells adhere to FBs sustaining immune evasion, drug resistance and the undetectable endurance of tumor cells known as minimal residual disease (MRD). Here, we describe the novel bi-specific designed ankyrin repeat protein (DARPin) α-FAPx4-1BB (MP0310) with FAP-dependent 4-1BB agonistic activity. The α-FAPx4-1BB DARPin simultaneously binds to FAP and 4-1BB overexpressed by activated FBs and immune cells, respectively. Although flow cytometry analysis showed that T and NK cells from MM patients were not activated and did not express 4-1BB, stimulation with daratumumab or elotuzumab, monoclonal antibodies (mAbs) currently used for the treatment of MM, significantly upregulated 4-1BB both in vitro and in MM patients following mAb-based therapy. The mAb-induced 4-1BB overexpression allowed the engagement of α-FAPx4-1BB that acted as a bridge between FAP+FBs and 4-1BB+NK cells. Therefore, α-FAPx4-1BB enhanced both the adhesion of daratumumab-treated NK cells on FBs as well as their activation by improving release of CD107a and perforin, hence MM cell killing via antibody-mediated cell cytotoxicity (ADCC). Interestingly, α-FAPx4-1BB significantly potentiated daratumumab-mediated ADCC in the presence of FBs, suggesting that it may overcome the BM FBs' immunosuppressive effect. Overall, we speculate that treatment with α-FAPx4-1BB may represent a valuable strategy to improve mAb-induced NK cell activity fostering MRD negativity in MM patients through the eradication of latent MRD cells.


Subject(s)
Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal , Killer Cells, Natural , Multiple Myeloma , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Humans , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Membrane Proteins/metabolism , Membrane Proteins/agonists , Endopeptidases
7.
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
8.
J Clin Invest ; 134(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38941297

ABSTRACT

STING agonists can reprogram the tumor microenvironment to induce immunological clearance within the central nervous system. Using multiplexed sequential immunofluorescence (SeqIF) and the Ivy Glioblastoma Atlas, STING expression was found in myeloid populations and in the perivascular space. The STING agonist 8803 increased median survival in multiple preclinical models of glioblastoma, including QPP8, an immune checkpoint blockade-resistant model, where 100% of mice were cured. Ex vivo flow cytometry profiling during the therapeutic window demonstrated increases in myeloid tumor trafficking and activation, alongside enhancement of CD8+ T cell and NK effector responses. Treatment with 8803 reprogrammed microglia to express costimulatory CD80/CD86 and iNOS, while decreasing immunosuppressive CD206 and arginase. In humanized mice, where tumor cell STING is epigenetically silenced, 8803 therapeutic activity was maintained, further attesting to myeloid dependency and reprogramming. Although the combination with a STAT3 inhibitor did not further enhance STING agonist activity, the addition of anti-PD-1 antibodies to 8803 treatment enhanced survival in an immune checkpoint blockade-responsive glioma model. In summary, 8803 as a monotherapy demonstrates marked in vivo therapeutic activity, meriting consideration for clinical translation.


Subject(s)
Glioblastoma , Membrane Proteins , Tumor Microenvironment , Animals , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , Tumor Microenvironment/immunology , Mice , Membrane Proteins/immunology , Membrane Proteins/genetics , Membrane Proteins/agonists , Humans , Cell Line, Tumor , Brain Neoplasms/immunology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/genetics
9.
Oncoimmunology ; 13(1): 2364958, 2024.
Article in English | MEDLINE | ID: mdl-38872753

ABSTRACT

We have recently demonstrated that inhibiting VPS34 enhances T-cell-recruiting chemokines through the activation of the cGAS/STING pathway using the STING agonist ADU-S100. Combining VPS34 inhibitors with ADU-S100 increased cytokine release and improved tumor control in mouse models, suggesting a potential synergy between VPS34 inhibition and therapies based on STING agonists.


Subject(s)
Autophagy , Class III Phosphatidylinositol 3-Kinases , Membrane Proteins , Neoplasms , Animals , Membrane Proteins/agonists , Membrane Proteins/metabolism , Humans , Mice , Autophagy/drug effects , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Class III Phosphatidylinositol 3-Kinases/metabolism , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors
10.
Expert Opin Ther Pat ; 34(5): 297-313, 2024 May.
Article in English | MEDLINE | ID: mdl-38849323

ABSTRACT

INTRODUCTION: Stimulator of Interferon Genes (STING) is an innate immune sensor. Activation of STING triggers a downstream response that results in the expression of proinflammatory cytokines (TNF-α, IL-1ß) via nuclear factor kappa-B (NF-κB) or the expression of type I interferons (IFNs) via an interferon regulatory factor 3 (IRF3). IFNs can eventually result in promotion of the adaptive immune response including activation of tumor-specific CD8+ T cells to abolish the tumor. Consequently, activation of STING has been considered as a potential strategy for cancer treatment. AREAS COVERED: This article provides an overview on structures and pharmacological data of CDN-like and non-nucleotide STING agonists acting as anticancer agents (January 2021 to October 2023) from a medicinal chemistry perspective. The data in this review come from EPO, WIPO, RCSB PDB, CDDI. EXPERT OPINION: In recent years, several structurally diverse STING agonists have been identified. As an immune enhancer, they are used in the treatment of tumors, which has received extensive attention from scientific community and pharmaceutical companies. Despite the multiple challenges that have appeared, STING agonists may offer opportunities for immunotherapy.


Subject(s)
Antineoplastic Agents , Membrane Proteins , Neoplasms , Patents as Topic , Humans , Animals , Neoplasms/drug therapy , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Membrane Proteins/agonists , Membrane Proteins/metabolism , Membrane Proteins/genetics , Immunity, Innate/drug effects , Immunotherapy/methods
11.
J Control Release ; 371: 273-287, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38789087

ABSTRACT

The stimulator of interferon genes (STING) connects the innate and adaptive immune system and plays a significant role in antitumor immunity. Over the past decades, endogenous and CDN-derived STING agonists have been a hot topic in the research of cancer immunotherapies. However, these STING agonists are either in infancy with limited biological effects or have failed in clinical trials. In 2020, a non-nucleotide STING agonist MSA-2 was identified, which exhibited satisfactory antitumor effects in animal studies and is amenable to oral administration. Due to its distinctive binding mode and enhanced bioavailability, there have been accumulating interests and an array of studies on MSA-2 and its derivatives, spanning its structure-activity relationship, delivery systems, applications in combination therapies, etc. Here, we provide a comprehensive review of MSA-2 and interventional strategies based on this family of STING agonists to help more researchers extend the investigation on MSA-2 in the future.


Subject(s)
Membrane Proteins , Humans , Membrane Proteins/agonists , Animals , Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use
12.
Cancer Lett ; 594: 216992, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38797231

ABSTRACT

Intrahepatic cholangiocarcinoma (iCCA) is an aggressive cancer with an extremely poor prognosis, and new treatment options are needed. Recently, immunotherapy has emerged as an efficient treatment against malignant tumors, but less effective in iCCA. Activation of stimulator of interferon genes (STING) signaling could reignite immunologically inert tumors, but the expression and role of STING in iCCA remains to be determined. Here, we show STING is expressed in iCCA, and patients with high expression of STING in early-stage iCCA have a longer overall survival than those have low expression. Increased immune cell infiltration in early-stage iCCA corresponds to elevated STING expression. In mice iCCA models, treatment with the STING agonist MSA-2 show stage-specific inhibitory effects on tumors, with beneficial effects in early-stage tumors but not with advanced-stage cancer. This discrepancy was associated with greater programmed cell death ligand 1 (PD-L1) expression in advanced-stage tumors. Combination therapy targeting PD-L1 and MSA-2 strikingly reduced tumor burden in such tumors compared to either monotherapy. Cumulatively, these data demonstrate that STING agonism monotherapy improves the immune landscape of the tumor microenvironment in early-stage iCCA, while combination therapy ameliorates advanced-stage iCCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Membrane Proteins , Cholangiocarcinoma/immunology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Cholangiocarcinoma/drug therapy , Animals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/agonists , Humans , Mice , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Neoplasm Staging , Cell Line, Tumor , Tumor Microenvironment/immunology , Male , Female , Signal Transduction
13.
Chembiochem ; 25(13): e202400321, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38720428

ABSTRACT

Cyclic dinucleotides (CDNs) have garnered popularity over the last decade as immunotherapeutic agents, which activate the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway to trigger an immune response. Many analogs of 2'3'-cGAMP, c-di-GMP, and c-di-AMP have been developed and shown as effective cancer vaccines and immunomodulators for the induction of both the adaptive and innate immune systems. Unfortunately, the effectiveness of these CDNs is limited by their chemical and enzymatic instability. We recently introduced 5'-endo-phosphorothoiate 2'3'-cGAMP analogs as potent STING agonist with improved resistance to cleavage by clinically relevant phosphodiesterases. We herein report the synthesis of locked nucleic acid-functionalized (LNA) endo-S-CDNs and evaluate their ability to activate STING in THP1 monocytes. Interestingly, some of our synthesized LNA 3'3'-endo-S-CDNs can moderately activate hSTING REF haplotype (R232H), which exhibit diminished response to both 2'3'-cGAMP and ADU-S100. Also, we show that one of our most potent endo-S-CDNs has remarkable chemical (oxidants I2 and H2O2) and phosphodiesterase stability.


Subject(s)
Membrane Proteins , Oligonucleotides , Membrane Proteins/metabolism , Membrane Proteins/agonists , Humans , Oligonucleotides/chemistry , Oligonucleotides/pharmacology , Oligonucleotides/chemical synthesis , Nucleotides, Cyclic/pharmacology , Nucleotides, Cyclic/chemistry , Nucleotides, Cyclic/metabolism , THP-1 Cells
14.
Nat Commun ; 15(1): 3933, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730243

ABSTRACT

As a strategy to improve the therapeutic success of chimeric antigen receptor T cells (CART) directed against solid tumors, we here test the combinatorial use of CART and IMSA101, a newly developed stimulator of interferon genes (STING) agonist. In two syngeneic tumor models, improved overall survival is observed when mice are treated with intratumorally administered IMSA101 in addition to intravenous CART infusion. Transcriptomic analyses of CART isolated from tumors show elevated T cell activation, as well as upregulated cytokine pathway signatures, in particular IL-18, in the combination treatment group. Also, higher levels of IL-18 in serum and tumor are detected with IMSA101 treatment. Consistent with this, the use of IL-18 receptor negative CART impair anti-tumor responses in mice receiving combination treatment. In summary, we find that IMSA101 enhances CART function which is facilitated through STING agonist-induced IL-18 secretion.


Subject(s)
Interleukin-18 , Membrane Proteins , Receptors, Chimeric Antigen , Animals , Interleukin-18/metabolism , Membrane Proteins/agonists , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Humans , Cell Line, Tumor , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Lymphocyte Activation/drug effects , Immunotherapy, Adoptive/methods , Female , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy
15.
Curr Opin Biotechnol ; 87: 103105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38461748

ABSTRACT

Agonists of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway, a critical mediator of innate immune response to foreign invaders with DNA, have gained significant interest in cancer immunotherapy. STING agonists are envisioned as a way of complementing the antitumor activity of the patient's immune system and immune checkpoint blockade therapy. However, their clinical development has been challenging due to the poor pharmacokinetic and physicochemical properties. This review discusses drug delivery efforts to circumvent the challenges, their accomplishment, and unmet needs based on the last five years of literature.


Subject(s)
Immunotherapy , Membrane Proteins , Neoplasms , Humans , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/immunology , Immunotherapy/methods , Membrane Proteins/agonists , Animals , Drug Delivery Systems/methods
16.
Phytomedicine ; 128: 155404, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38507852

ABSTRACT

BACKGROUND: The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon (IFN) genes (STING) pathway is critical in the innate immune system and can be mobilized by cytosolic DNA. The various inflammatory and autoimmune diseases progression is highly correlated with aberrant cGAS-STING pathway activation. While some cGAS-STING pathway inhibitor were identified, there are no drugs that can be applied to the clinic. Compound Danshen Dripping Pill (CDDP) has been successfully used in clinic around the world, but the most common application is limited to cardiovascular disease. Therefore, the purpose of the present investigation was to examine whether CDDP inhibits the cGAS-STING pathway and could be used as a therapeutic agent for multiple cGAS-STING-triggered diseases. METHODS: BMDMs, THP1 cells or Trex1-/- BMDMs were stimulated with various cGAS-STING-agonists after pretreatment with CDDP to detect the function of CDDP on IFN-ß and ISGs productionn. Next, we detect the influence on IRF3 and P65 nuclear translocation, STING oligomerization and STING-TBK1-IRF3 complex formation of CDDP. Additionally, the DMXAA-mediated activation mice model of cGAS-STING pathway was used to study the effects of CDDP. Trex1-/- mice model and HFD-mediated obesity model were established to clarify the efficacy of CDDP on inflammatory and autoimmune diseases. RESULTS: CDDP efficacy suppressed the IRF3 phosphorylation or the generation of IFN-ß, ISGs, IL-6 and TNF-α. Mechanistically, CDDP did not influence the STING oligomerization and IRF3-TBK1 and STING-IRF3 interaction, but remarkably eliminated the STING-TBK1 interaction, ultimately blocking the downstream responses. In addition, we also clarified that CDDP could suppress cGAS-STING pathway activation triggered by DMXAA, in vivo. Consistently, CDDP could alleviate multi-organ inflammatory responses in Trex1-/- mice model and attenuate the inflammatory disorders, incleding obesity-induced insulin resistance. CONCLUSION: CDDP is a specifically cGAS-STING pathway inhibitor. Furthermore, we provide novel mechanism for CDDP and discovered a clinical agent for the therapy of cGAS-STING-triggered inflammatory and autoimmune diseases.


Subject(s)
Autoimmune Diseases , Camphanes , Drugs, Chinese Herbal , Inflammation , Panax notoginseng , Salvia miltiorrhiza , Mice , Mice, Inbred C57BL , Salvia miltiorrhiza/chemistry , Panax notoginseng/chemistry , Immunity, Innate , Membrane Proteins/agonists , Membrane Proteins/metabolism , Signal Transduction , THP-1 Cells , Exodeoxyribonucleases/genetics , Phosphoproteins/genetics , Macrophages , Interferon-beta/metabolism , Interferon Regulatory Factor-3/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus , Autoimmune Diseases/drug therapy , Inflammation/drug therapy , Obesity/drug therapy , Diet, High-Fat , Protein Serine-Threonine Kinases/metabolism , Humans
17.
Med Res Rev ; 44(4): 1768-1799, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38323921

ABSTRACT

Adjuvants are of critical value in vaccine development as they act on enhancing immunogenicity of antigen and inducing long-lasting immunity. However, there are only a few adjuvants that have been approved for clinical use, which highlights the need for exploring and developing new adjuvants to meet the growing demand for vaccination. Recently, emerging evidence demonstrates that the cGAS-STING pathway orchestrates innate and adaptive immunity by generating type I interferon responses. Many cGAS-STING pathway agonists have been developed and tested in preclinical research for the treatment of cancer or infectious diseases with promising results. As adjuvants, cGAS-STING agonists have demonstrated their potential to activate robust defense immunity in various diseases, including COVID-19 infection. This review summarized the current developments in the field of cGAS-STING agonists with a special focus on the latest applications of cGAS-STING agonists as adjuvants in vaccination. Potential challenges were also discussed in the hope of sparking future research interests to further the development of cGAS-STING as vaccine adjuvants.


Subject(s)
Membrane Proteins , Nucleotidyltransferases , Humans , Nucleotidyltransferases/metabolism , Membrane Proteins/agonists , Membrane Proteins/immunology , Membrane Proteins/metabolism , Animals , Adjuvants, Vaccine/pharmacology , Adjuvants, Vaccine/chemistry , Signal Transduction/drug effects , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , SARS-CoV-2/drug effects , Immunity, Innate/drug effects , Adjuvants, Immunologic/pharmacology , COVID-19 Vaccines/immunology
18.
Org Lett ; 26(4): 819-823, 2024 02 02.
Article in English | MEDLINE | ID: mdl-38236576

ABSTRACT

The stimulator of interferon genes (STING) protein plays a crucial role in the activation of the innate immune response. Activation of STING is initiated by cyclic dinucleotides (CDNs) which prompted the community to synthesize structural analogues to enhance their biological properties. We present here the synthesis and biological evaluation of four novel CDN analogues composed of an N-acylsulfonamide linkage. These CDNs were obtained in high overall yields via the sulfo-click reaction as a key step.


Subject(s)
Nucleotides, Cyclic , Nucleotides, Cyclic/chemistry , Nucleotides, Cyclic/metabolism , Membrane Proteins/agonists , Membrane Proteins/chemistry , Click Chemistry/methods
19.
Org Biomol Chem ; 22(2): 302-308, 2024 01 03.
Article in English | MEDLINE | ID: mdl-38054844

ABSTRACT

The STING pathway is critical to innate immunity and is being investigated as a potential therapeutic target. Existing agents targeting STING suffer from several undesirable effects, particularly the possibility of systematic activation, which increases the risk of autoimmune disorders. In this proof-of-concept study, we report the development of a light-activated STING agonist, based on the potent compound SR-717. We first screened the activity of the non-caged agonist toward 5 human STING variants to identify the most viable target. A photocaged agonist was designed and synthesized in order to block an essential interaction between the carboxy acid group of the ligand with the R238 residue of the STING protein. We then investigated the selective activation of STING with the photocaged agonist, demonstrating an irradiation-dependent response. The development and characterization of this selective agonist expands the growing toolbox of conditionally controlled STING agonists to avoid systematic immune activation.


Subject(s)
Immunity, Innate , Membrane Proteins , Humans , Membrane Proteins/agonists
20.
Adv Mater ; 35(51): e2305255, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37541432

ABSTRACT

Stimuli-responsive nanomaterials have the potential to improve the performance and overcome existing barriers of conventional nanotherapeutics. Molecular cooperativity design in stimuli-responsive nanomedicine can amplify physiological signals, enabling a cooperative response for improved diagnostic and therapeutic precision. Previously, this work reported an ultra-pH-sensitive polymer, PEG-b-PC7A, that possesses innate immune activating properties by binding to the stimulator of interferon genes (STING) through polyvalent phase condensation. This interaction enhances STING activation and synergizes with the endogenous STING ligand for robust cancer immunotherapy. Despite its successes in innate immune activation, the fundamental physicochemical and pH-responsive properties of PC7A require further investigation. Here, this study elucidates the protonation cooperativity driven by the phase transition of PC7A copolymer. The highly cooperative system displays an "all-or-nothing" proton distribution between highly charged unimer (all) and neutral micelle (nothing) states without gradually protonated intermediates. The binary protonation behavior is further illustrated in pH-precision-controlled release of a representative anticancer drug, ß-lapachone, by PC7A micelles over a noncooperative PE5A polymer. Furthermore, the bimodal distribution of protons is represented by a high Hill coefficient (nH  > 9), featuring strong positive cooperativity. This study highlights the nanoscale pH cooperativity of an immune activating polymer, providing insights into the physicochemical characterization and design parameters for future nanotherapeutics development.


Subject(s)
Antineoplastic Agents , Membrane Proteins , Nanostructures , Hydrogen-Ion Concentration , Micelles , Phase Transition , Polymers/chemistry , Membrane Proteins/agonists , Membrane Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL