Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 98(9): e0035424, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39171925

RESUMEN

Development of next-generation influenza virus vaccines is crucial to improve protection against circulating and emerging viruses. Current vaccine formulations have to be updated annually due to mutations in seasonal strains and do not offer protection against strains with pandemic potential. Computationally optimized broadly reactive antigen (COBRA) methodology has been utilized by our group to generate broadly reactive immunogens for individual influenza subtypes, which elicit protective immune responses against a broad range of strains over numerous seasons. Octavalent mixtures of COBRA hemagglutinin (HA) (H1, H2, H3, H5, H7, and influenza B virus) plus neuraminidase (NA) (N1 and N2) recombinant proteins mixed with c-di-AMP adjuvant were administered intranasally to naive or pre-immune ferrets in prime-boost fashion. Four weeks after final vaccination, collected sera were analyzed for breadth of antibody response, and the animals were challenged with seasonal or pre-pandemic strains. The octavalent COBRA vaccine elicited antibodies that recognized a broad panel of strains representing different subtypes, and these vaccinated animals were protected against influenza virus challenges. Overall, this study demonstrated that the mixture of eight COBRA HA/NA proteins mixed with an intranasal adjuvant is a promising candidate for a universal influenza vaccine. IMPORTANCE: Influenza is a respiratory virus which infects around a billion people globally every year, with millions experiencing severe illness. Commercial vaccine efficacy varies year to year and can be low due to mismatch of circulating virus strains. Thus, the formulation of current vaccines has to be adapted accordingly every year. The development of a broadly reactive influenza vaccine would lessen the global economic and public health burden caused by the different types of influenza viruses. The significance of our research is producing a promising universal vaccine candidate which provides protection against a wider range of virus strains over a wider range of time.


Asunto(s)
Administración Intranasal , Anticuerpos Antivirales , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Animales , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Neuraminidasa/inmunología , Neuraminidasa/genética , Estaciones del Año , Adyuvantes Inmunológicos/administración & dosificación , Vacunación/métodos , Gripe Humana/prevención & control , Gripe Humana/inmunología , Gripe Humana/virología , Humanos , Femenino , Protección Cruzada/inmunología , Pandemias/prevención & control
2.
Proc Natl Acad Sci U S A ; 119(22): e2200568119, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35588144

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Antígenos de Histocompatibilidad Clase I , Inmunoterapia , Interleucina-2 , Proteínas de la Membrana , Neoplasias , Nucleótidos Cíclicos , Oligodesoxirribonucleótidos , Albúmina Sérica , Animales , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Inmunoterapia/métodos , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/agonistas , Ratones , Neoplasias/genética , Neoplasias/terapia , Nucleótidos Cíclicos/uso terapéutico , Oligodesoxirribonucleótidos/uso terapéutico , Albúmina Sérica/uso terapéutico
3.
Med Res Rev ; 44(4): 1768-1799, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38323921

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana , Nucleotidiltransferasas , Humanos , Nucleotidiltransferasas/metabolismo , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Animales , Adyuvantes de Vacunas/farmacología , Adyuvantes de Vacunas/química , Transducción de Señal/efectos de los fármacos , COVID-19/inmunología , COVID-19/prevención & control , SARS-CoV-2/inmunología , SARS-CoV-2/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Vacunas contra la COVID-19/inmunología
4.
Chembiochem ; 25(13): e202400321, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38720428

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana , Oligonucleótidos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/agonistas , Humanos , Oligonucleótidos/química , Oligonucleótidos/farmacología , Oligonucleótidos/síntesis química , Nucleótidos Cíclicos/farmacología , Nucleótidos Cíclicos/química , Nucleótidos Cíclicos/metabolismo , Células THP-1
5.
Mol Cell Biochem ; 479(7): 1697-1705, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38592428

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most prevalent and lethal subtype of kidney cancer, patients with ccRCC usually have very poor prognosis and short survival. Therefore, it is urgent to develop more effective therapeutics or medications to suppress ccRCC progression. Here, we demonstrated that STING agonist, MSA-2 significantly inhibits tumor progress and prolongs the survival of ccRCC mice by promoting cytokines secretion. Moreover, MSA-2 triggered the trafficking and infiltration of CD8+ T cells, supported by the generation of a chemokine milieu that promoted recruitment and modulation of the immunosuppressive TME in ccRCC. These findings suggest that MSA-2 potentially serves an effective and preferable adjuvant immunotherapy of ccRCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Proteínas de la Membrana , Microambiente Tumoral , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Animales , Neoplasias Renales/patología , Neoplasias Renales/metabolismo , Neoplasias Renales/tratamiento farmacológico , Ratones , Proteínas de la Membrana/metabolismo , Humanos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral
6.
Handb Exp Pharmacol ; 284: 113-132, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37059911

RESUMEN

Adjuvants have been extensively and essentially formulated in subunits and certain inactivated vaccines for enhancing and prolonging protective immunity against infections and diseases. According to the types of infectious diseases and the required immunity, adjuvants with various acting mechanisms have been designed and applied in human vaccines. In this chapter, we introduce the advances in vaccine adjuvants based on nanomaterials and small molecules. By reviewing the immune mechanisms induced by adjuvants with different characteristics, we aim to establish structure-activity relationships between the physicochemical properties of adjuvants and their immunostimulating capability for the development of adjuvants for more effective preventative and therapeutic vaccines.


Asunto(s)
Nanoestructuras , Vacunas , Humanos , Adyuvantes de Vacunas , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química
7.
Turk J Med Sci ; 54(3): 607-614, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39049995

RESUMEN

Background/aim: Glioblastoma is one of the most aggressive tumours, resistant to all applied therapy regiments and prone to relapse. Median survival rates are therefore only expressed as months. STING agonists are immunomodulatory molecules that activate type I interferon expression, making them potentially useful in regulating the tumour microenvironment. Since PTEN serves as a critical phosphatase in activating interferon-regulating transcription factors and is frequently mutated in glioblastoma cells, this study aimed to investigate STING activation in glioblastoma cell lines, examining whether they harbour the PTEN protein or not.°. Materials and methods: T98G and U118MG glioblastoma cell lines were treated with the 2'3'-c-di-AM(PS)2(Rp,Rp) STING agonist together with or without the chemotherapeutic agent temozolomide. cGAS/STING pathway components were subsequently analysed using qRT-PCR, western blot, and ELISA methods. Results: Our results showed that PTEN-harbouring T98G cells responded well to STING activation, leading to increased temozolomide efficacy. In contrast, STING activation in U118MG cells did not affect the response to temozolomide. mRNA expression levels of STING, IRF3, NF-KB, and RELA genes were significantly increased at the combined treatment groups in T98G cell line. Conversely, combined treatment with STING agonist and temozolomide did not affect mRNA expression levels of cGAS/STING pathway genes in U118MG cells. Conclusion: Our data offers new evidence suggesting that STING agonists can effectively be used to increase temozolomide response in the presence of PTEN protein. Therefore, increased GBM therapy success rates can be achieved by employing the PTEN expression status as a predictive biomarker before treating patients with a chemotherapeutic agent in combination with STING agonist.


Asunto(s)
Glioblastoma , Proteínas de la Membrana , Fosfohidrolasa PTEN , Temozolomida , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Temozolomida/farmacología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Línea Celular Tumoral , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Factor 3 Regulador del Interferón/metabolismo
8.
Angew Chem Int Ed Engl ; : e202417027, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39375150

RESUMEN

The activation of the stimulator of interferon genes (STING) protein by cyclic dinucleotide metabolites plays a critical role in antitumor immunity. However, synthetic STING agonists like 4-(5,6-dimethoxybenzo[b]thiophen-2-yl)-4-oxobutanoic acid (MSA-2) exhibit suboptimal pharmacokinetics and fail to sustain STING activation in tumors for effective antitumor responses. Here, we report the design of MOF/MSA-2, a bifunctional MSA-2 conjugated nanoscale metal-organic framework (MOF) based on Hf6 secondary building units (SBUs) and hexakis(4'-carboxy[1,1'-biphenyl]-4-yl)benzene bridging ligands, for potent cancer radio-immunotherapy. By leveraging the high-Z properties of the Hf6 SBUs, the MOF enhances the therapeutic effect of X-ray radiation and elicits potent immune stimulation in the tumor microenvironment. MOF/MSA-2 further enhances radiotherapeutic effects of X-rays by enabling sustained STING activation and promoting the infiltration and activation of immune cells in the tumors. MOF/MSA-2 plus low-dose X-ray irradiation elicits strong STING activation and potent tumor regression, and when combined with an immune checkpoint inhibitor, effectively suppresses both primary and distant tumors through systemic immune activation.

9.
Cell Struct Funct ; 48(1): 59-70, 2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36575042

RESUMEN

Stimulator of interferon genes (STING) is an ER-localized transmembrane protein and the receptor for 2',3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), which is a second messenger produced by cGAMP synthase (cGAS), a cytosolic double-stranded DNA sensor. The cGAS-STING pathway plays a critical role in the innate immune response to infection of a variety of DNA pathogens through the induction of the type I interferons. Pharmacological activation of STING is a promising therapeutic strategy for cancer, thus the development of potent and selective STING agonists has been pursued. Here we report that mouse STING can be activated by phenylarsine oxide (PAO), a membrane permeable trivalent arsenic compound that preferentially reacts with thiol group of cysteine residue (Cys). The activation of STING with PAO does not require cGAS or cGAMP. Mass spectrometric analysis of the peptides generated by trypsin and chymotrypsin digestion of STING identifies several PAO adducts, suggesting that PAO covalently binds to STING. Screening of STING variants with single Cys to serine residues (Ser) reveals that Cys88 and Cys291 are critical to the response to PAO. STING activation with PAO, as with cGAMP, requires the ER-to-Golgi traffic and palmitoylation of STING. Our results identify a non-nucleotide STING agonist that does not target the cGAMP-binding pocket, and demonstrate that Cys of STING can be a novel target for the development of STING agonist.Key words: STING agonist, cysteine modification, innate immunity, phenylarsine oxide.


Asunto(s)
Cisteína , Transducción de Señal , Ratones , Animales , Proteínas de la Membrana/metabolismo , Inmunidad Innata , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , ADN
10.
Int J Cancer ; 152(8): 1685-1697, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36495276

RESUMEN

Recent studies have shown that activation of the cGAS-STING pathway is a key process in antitumor immune responses and various kinds of STING agonists have been developed for cancer immunotherapy. Despite promising preclinical studies, preliminary clinical results have shown only a modest effect of STING agonists. There is therefore a need to develop more effective treatment strategies. Based on previous observations that COX-2 is frequently overexpressed not only in a variety of cancers but also in tumor myeloid cells and that it suppresses antitumor immunity and promotes tumor survival by producing PGE2, we investigated the antitumor effects of combination therapy with a STING agonist cGAMP and the selective COX-2 inhibitor celecoxib in mouse models. Combination treatment with cGAMP and celecoxib inhibited tumor growth compared with either monotherapy, and the combination therapy induced both local and systemic antitumor immunity. cGAMP treatment decreased PD-1 expression on tumor-infiltrating T-cells and enhanced T-cell activation in tumor-draining lymph nodes regardless of the presence of celecoxib. Meanwhile, although celecoxib treatment did not alter the frequency of CD4+ CD25+ Foxp3+ regulatory T-cells, it enhanced the expression of costimulatory molecules and glycolysis-associated genes in tumor-infiltrating CD11b+ Ly6G+ cells. Moreover, we also found that celecoxib decreased lactate efflux and increased the frequency of IFN-γ- and TNF-α-producing CD8+ T-cells in the tumor microenvironment. Taken together, our findings suggest that combined treatment with celecoxib may be an effective strategy to improve the antitumor efficacy of STING agonists.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Ratones , Animales , Celecoxib/farmacología , Neoplasias/patología , Inhibidores de la Ciclooxigenasa 2/farmacología , Inmunoterapia , Glucosa , Microambiente Tumoral
11.
Cancer Immunol Immunother ; 72(11): 3593-3608, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37526659

RESUMEN

Reovirus, a naturally occurring oncolytic virus, initiates the lysis of tumor cells while simultaneously releasing tumor antigens or proapoptotic cytokines in the tumor microenvironment to augment anticancer immunity. However, reovirus has developed a strategy to evade antiviral immunity via its inhibitory effect on interferon production, which negatively affects the induction of antitumor immune responses. The mammalian adaptor protein Stimulator of Interferon Genes (STING) was identified as a key regulator that orchestrates immune responses by sensing cytosolic DNA derived from pathogens or tumors, resulting in the production of type I interferon. Recent studies reported the role of STING in innate immune responses to RNA viruses leading to the restriction of RNA virus replication. In the current study, we found that reovirus had a reciprocal reaction with a STING agonist regarding type I interferon responses in vitro; however, we found that the combination of reovirus and STING agonist enhanced anti-tumor immunity by enhancing cytotoxic T cell trafficking into tumors, leading to significant tumor regression and survival benefit in a syngeneic colorectal cancer model. Our data indicate the combination of reovirus and a STING agonist to enhance inflammation in the tumor microenvironment might be a strategy to improve oncolytic reovirus immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Interferón Tipo I , Reoviridae , Animales , Ratones , Reoviridae/metabolismo , Inmunidad Innata , Citocinas , Interferón Tipo I/metabolismo , Neoplasias Colorrectales/terapia , Mamíferos/metabolismo , Microambiente Tumoral
12.
Cancer Immunol Immunother ; 72(11): 3491-3505, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37550427

RESUMEN

STING is a pivotal mediator of effective innate and adaptive anti-tumor immunity; however, intratumoral administration of STING agonists have shown limited therapeutic benefit in clinical trials. The systemic effect of the intravenous delivery of STING agonists in cancer is not well-defined. Here, we demonstrated that systemic administration of STING agonist inhibited melanoma growth, improved inflammatory effector cell infiltration, and induced bone marrow mobilization and extramedullary hematopoiesis, causing widespread changes in immune components in the peripheral blood. The systemically administered STING agonist promoted HSC expansion and influenced lineage fate commitment, which was manifested as the differentiation of HSPCs was skewed toward myeloid cells at the expense of B-cell lymphopoiesis and erythropoiesis. Transcriptome analysis revealed upregulation of myeloid lineage differentiation-related and type I interferon-related genes. This myeloid-biased differentiation promoted the production and maturation of myeloid cells toward an activated phenotype. Furthermore, depletion of Gr-1+ myeloid cells attenuated the anti-tumor immunity of STING agonist. Our findings reveal the anti-tumor mechanism of systemic administration of STING agonist that involves modulating HSPC differentiation and promoting myeloid cells maturation. Our study may help explain the limited clinical activity of STING agonists administered intratumorally.


Asunto(s)
Médula Ósea , Neoplasias , Humanos , Diferenciación Celular , Médula Ósea/metabolismo , Células Madre Hematopoyéticas , Células Mieloides , Inmunidad Adaptativa
13.
J Med Virol ; 95(2): e28475, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36606607

RESUMEN

Global coronavirus disease 2019 (COVID-19) pandemics highlight the need of developing vaccines with universal and durable protection against emerging SARS-CoV-2 variants. Here we developed an extended-release vaccine delivery system (GP-diABZI-RBD), consisting the original SARS-CoV-2 WA1 strain receptor-binding domain (RBD) as the antigen and diABZI stimulator of interferon genes (STING) agonist in conjunction with yeast ß-glucan particles (GP-diABZI) as the platform. GP-diABZI-RBD could activate STING pathway and inhibit SARS-CoV-2 replication. Compared to diABZI-RBD, intraperitoneal injection of GP-diABZI-RBD elicited robust cellular and humoral immune responses in mice. Using SARS-CoV-2 GFP/ΔN transcription and replication-competent virus-like particle system (trVLP), we demonstrated that GP-diABZI-RBD-prototype vaccine exhibited the strongest and durable humoral immune responses and antiviral protection; whereas GP-diABZI-RBD-Omicron displayed minimum neutralization responses against trVLP. By using pseudotype virus (PsVs) neutralization assay, we found that GP-diABZI-RBD-Prototype, GP-diABZI-RBD-Delta, and GP-diABZI-RBD-Gamma immunized mice sera could efficiently neutralize Delta and Gamma PsVs, but had weak protection against Omicron PsVs. In contrast, GP-diABZI-RBD-Omicron immunized mice sera displayed the strongest neutralization response to Omicron PsVs. Taken together, the results suggest that GP-diABZI can serve as a promising vaccine delivery system for enhancing durable humoral and cellular immunity against broad SARS-CoV-2 variants. Our study provides important scientific basis for developing SARS-CoV-2 VOC-specific vaccines.


Asunto(s)
COVID-19 , Vacunas , Animales , Humanos , Ratones , SARS-CoV-2 , Vacunas contra la COVID-19 , Inmunidad Celular , Anticuerpos Neutralizantes , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Antivirales
14.
Curr Oncol Rep ; 25(3): 189-199, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36705879

RESUMEN

PURPOSE OF REVIEW: New therapies are needed to potentiate the effects of current immunotherapies and overcome resistance. The stimulator of interferon genes genes (STING) pathway is an innate immune activating cascade that may enhance current cancer immunotherapies. RECENT FINDINGS: Preclinical data has shown that the addition of a STING agonist enhances the effect of current treatments such as immune checkpoint inhibitor antibodies and radiation therapy. Early phase trials have demonstrated modest efficacy of STING agonists and revealed new mechanistic and technical challenges. STING agonists are a new class of agents that activate the immune response to improve tumor control. A wide range of preclinical experiments, translational data, and ongoing clinical trials support the therapeutic use of STING agonists in patients. Trials to determine optimal drug combinations and novel delivery mechanisms are continuing in development.


Asunto(s)
Proteínas de la Membrana , Neoplasias , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Inmunidad Innata
15.
Nano Lett ; 22(5): 2048-2058, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35133159

RESUMEN

Herein, we integrate the Hepa1-6 liver cancer-specific neoantigen, toll-like receptor 9 agonist and stimulator of interferon genes agonist by silk-hydrogel package, and combine with TIM-3 blockade to elicit robust antitumor immunity for effectively suppressing orthotopic hepatocellular carcinoma (HCC) progression. Unlike intradermal injection of simple mixed components with short-term immune protection, the neoantigen immunotherapeutic-gels evoke long-term immune protection to achieve significant prophylactic and therapeutic activity against HCC through only one-shot administration without any side effects. Notably, the synergized immunotherapy by further combining NGC-gels with TIM-3 antibody significantly reduces regulatory T-cells and increases the IFN-γ and IL-12p70 levels in tumor tissues for promoting the infiltration of IFN-γ+CD8+T-cells and 41BB+CD8+T-cells to achieve complete remission (4/7) and prevent pulmonary metastasis in orthotopic HCC, and establish long-term memory against tumor rechallenge with remarkably longer survival time (180 days). Overall, this study provides an attractive and promising synergistic strategy for HCC immunotherapy with possible clinical translation prospects.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Linfocitos T CD8-positivos , Carcinoma Hepatocelular/tratamiento farmacológico , Geles , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Inmunoterapia , Neoplasias Hepáticas/tratamiento farmacológico
16.
Int J Mol Sci ; 24(22)2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38003463

RESUMEN

Stimulator of interferon genes (STING) agonists have shown potent anti-tumor efficacy in various mouse tumor models and have the potential to overcome resistance to immune checkpoint inhibitors (ICI) by linking the innate and acquired immune systems. First-generation STING agonists are administered intratumorally; however, a systemic delivery route would greatly expand the clinical use of STING agonists. Biochemical and cell-based experiments, as well as syngeneic mouse efficacy models, were used to demonstrate the anti-tumoral activity of ALG-031048, a novel STING agonist. In vitro, ALG-031048 is highly stable in plasma and liver microsomes and is resistant to degradation via phosphodiesterases. The high stability in biological matrices translated to good cellular potency in a HEK 293 STING R232 reporter assay, efficient activation and maturation of primary human dendritic cells and monocytes, as well as long-lasting, antigen-specific anti-tumor activity in up to 90% of animals in the CT26 mouse colon carcinoma model. Significant reductions in tumor growth were observed in two syngeneic mouse tumor models following subcutaneous administration. Combinations of ALG-031048 and ICIs further enhanced the in vivo anti-tumor activity. This initial demonstration of anti-tumor activity after systemic administration of ALG-031048 warrants further investigation, while the combination of systemically administered ALG-031048 with ICIs offers an attractive approach to overcome key limitations of ICIs in the clinic.


Asunto(s)
Neoplasias del Colon , Neoplasias , Ratones , Animales , Humanos , Células HEK293 , Neoplasias/patología , Neoplasias del Colon/tratamiento farmacológico , Modelos Animales de Enfermedad , Inmunoterapia , Microambiente Tumoral
17.
Int J Mol Sci ; 25(1)2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38203256

RESUMEN

To improve the chemical regulation on the activity of cyclic dinucleotides (CDNs), we here designed a reduction-responsive dithioethanol (DTE)-based dCDN prodrug 9 (DTE-dCDN). Prodrug 9 improved the cell permeability with the intracellular levels peaking in 2 h in THP-1 cells. Under the reductive substance such as GSH or DTT, prodrug 9 could be quickly decomposed in 30 min to release the parent dCDN. In THP1-Lucia cells, prodrug 9 also retained a high bioactivity with the EC50 of 0.96 µM, which was 51-, 43-, and 3-fold more than the 2',3'-cGAMP (EC50 = 48.6 µM), the parent compound 3',3'-c-di-dAMP (EC50 = 41.3 µM), and ADU-S100 (EC50 = 2.9 µM). The high bioactivity of prodrug 9 was validated to be highly correlated with the activation of the STING signaling pathway. Furthermore, prodrug 9 could also improve the transcriptional expression levels of IFN-ß, CXCL10, IL-6, and TNF-α in THP-1 cells. These results will be helpful to the development of chemically controllable CDN prodrugs with a high cellular permeability and potency.


Asunto(s)
Desoxirribosa , Profármacos , Alarminas , Fosfatos de Dinucleósidos , Permeabilidad , Profármacos/farmacología
18.
Molecules ; 28(7)2023 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-37049669

RESUMEN

The stimulator-of-interferon-gene (STING) protein is involved in innate immunity. The drug DMXAA (5,6-dimethylxanthenone-4-acetic acid) proved to be a potent murine-STING (mSTING) agonist but had little effect on human-STING (hSTING). In this paper, we draw upon the comparison of different crystal structures and protein-ligand interaction relationships analysis to venture the hypothesis that the drug design of DMXAA variants has the potential to convert STING agonists to inhibitors. Based on our previous discovery of two DMXAA analogs, 3 and 4 (both could bind to STING), we structurally optimized them and synthesized new derivatives, respectively. In binding assays, we found compounds 11 and 27 to represent STING binders that were superior to the original structures and discussed the structure-activity relationships. All target compounds were inactive in cellular assays for the screening of STING agonistic activity. Gratifyingly, we identified 11 and 27 as STING inhibitors with micromolar activity in both hSTING and mSTING pathways. In addition, 11 and 27 inhibited the induction of interferon and inflammatory cytokines activated by 2'3'-cGAMP without apparent cytotoxicity. These findings break the rigid thinking that DMXAA provides the structural basis specifically for STING agonists and open up more possibilities for developing novel STING agonists or inhibitors.


Asunto(s)
Proteínas de la Membrana , Xantonas , Ratones , Humanos , Animales , Proteínas de la Membrana/metabolismo , Xantonas/química , Inmunidad Innata , Interferones
19.
Mol Pharm ; 19(11): 3877-3883, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36018674

RESUMEN

The innovation of cancer immunotherapy is improving the prognosis of colorectal cancer (CRC) in clinics. Nevertheless, due to tumor heterogeneity and complex underlying inhibitory mechanisms, the therapeutic response greatly varies among different patients. To optimize the clinical management of CRC patients, it is critical to develop novel approaches for response monitoring and prediction. In the current study, we developed a novel near-infrared fluorescence (NIRF) imaging probe (Cy5.5-ICOS mAb) targeting the inducible T-cell costimulatory receptor (ICOS or CD278) and assessed its capacity for the detection of ICOS+-activated T cells in vivo. ICOS expression was evaluated by flow cytometry and immunofluorescence staining in subcutaneous MC38 models treated with the stimulator of interferon genes (STING) agonist (STINGa). NIRF imaging study was performed 1 day after the last treatment, and tumor volume was monitored every other day with a caliper. A significantly higher optical signal could be detected at tumor regions in STINGa group, compared with that in the PBS group at all time points imaged, and this was in line with ex vivo imaging and immunofluorescence staining study. The data demonstrated that Cy5.5-ICOS mAb could detect ICOS+-activated T cells with high specificity, and ICOS NIRF imaging is a promising strategy for predicting and monitoring immune response in CRC.


Asunto(s)
Neoplasias Colorrectales , Inmunoterapia , Humanos , Carbocianinas , Inmunoterapia/métodos , Diagnóstico por Imagen , Colorantes Fluorescentes , Neoplasias Colorrectales/diagnóstico por imagen , Neoplasias Colorrectales/tratamiento farmacológico
20.
Nano Lett ; 21(23): 9939-9950, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34779631

RESUMEN

Agonists of the stimulator of interferon gene (STING) are considered as promising therapeutics for cancer immunotherapy. However, drug-delivery barriers and adverse effects limit the clinical application of STING agonists. Therefore, it is an urgent need to develop an ideal delivery system to deliver STING agonists and avoid side effects. Here, we discovered that STING agonists significantly stimulated type I interferon (IFN) secretion in Clec9a+ dendritic cells (DCs). Then, we designed an engineered peptide-expressed biomimetic cancer cell membrane (EPBM)-coated nanovaccine drug-delivery system (PLGA/STING@EPBM) to deliver STING agonists and tumor antigens to Clec9a+ DCs. The PLGA/STING@EPBM nanovaccine significantly enhanced IFN-stimulated expression of genes and antigen cross-presentation of Clec9a+ DCs, thus eliciting strong antitumor effects in both anti-PD-1-responsive and -resistant tumor models without obvious cytotoxicity. Moreover, the PLGA/STING@EPBM nanovaccine combined with radiotherapy exhibited remarkable synergistic antitumor effects. Our work highlights the great potential of a EPBM-coated nanovaccine for systemic STING agonist delivery as an attractive tool for cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Proteínas de la Membrana , Neoplasias , Presentación de Antígeno , Antígenos de Neoplasias/farmacología , Humanos , Inmunoterapia , Lectinas Tipo C/genética , Proteínas de la Membrana/agonistas , Nanopartículas , Neoplasias/terapia , Receptores Mitogénicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA