Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Med Res Rev ; 40(3): 1117-1141, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31793026

RESUMEN

Since being discovered in 2008, the STING (stimulator of interferon genes) pathway has gradually been recognized as a central and promising target for immunotherapy. The STING pathway can be stimulated by cyclic dinucleotides (CDNs), leading to the type I interferons (IFN) production for immunotherapy for cancer or other diseases. However, the negative charges, hydrophilicity, and instability of CDNs have hindered their further applications. In addition, chronic activation of the STING pathway has been found to be involved in autoimmune diseases as IFN overproduction. Thus, research and development of STING agonists and inhibitors has been a hot field for the treatment of several diseases. The past several years, especially 2018, has seen increasingly rapid advances in this field. Here, this review summarizes the synthesis and modification of CDNs, the identification of nonnucleotide agonists, the recent progress in delivery systems and the medical applications, such as personalized vaccine adjuvants, in detail. In addition, in this review, we summarize the STING inhibitors' advances from two aspects, covalent, and noncovalent inhibitors.


Asunto(s)
Inmunoterapia/métodos , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/antagonistas & inhibidores , Animales , Cationes , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Citosol/metabolismo , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Humanos , Hidrogeles/química , Concentración de Iones de Hidrógeno , Liposomas/química , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Oligonucleótidos Fosforotioatos/química , Polímeros/química
2.
Chembiochem ; 21(8): 1150-1154, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-31702879

RESUMEN

The activation of toll-like receptors (TLRs) plays important roles in the immune response. The ability to control the activities of TLRs could be usable as a switch for immune response. Here we have rationally designed and synthesized a photoswitchable Pam3 CSK4 derivative-P10-to control the activation of TLR1/2. The ground-state trans-P10 was able to stimulate and activate antigen-presenting cells (APCs) by promoting TLR1/2 heterodimerization. However, cis-P10, derived from UV irradiation of trans-P10, reduced the activities of APCs by impeding the TLR1/2 heterodimerization. In the absence of UV radiation, the cis-P10 slowly returned to its ground trans state, restoring the activities of the APCs stimulation. Our results indicated that optical control of TLR1/2 heterodimerization mediated by the photoswitchable P10 offers the potential to regulate immune activation and inflammation.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Inmunidad/inmunología , Lipopéptidos/farmacología , Multimerización de Proteína , Receptor Toll-Like 1/agonistas , Receptor Toll-Like 2/agonistas , Rayos Ultravioleta , Animales , Células Presentadoras de Antígenos/metabolismo , Humanos , Ratones , Células RAW 264.7 , Transducción de Señal , Células THP-1 , Receptor Toll-Like 1/química , Receptor Toll-Like 2/química
3.
Bioconjug Chem ; 31(11): 2499-2503, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33147965

RESUMEN

Cyclic dinucleotides (CDNs), agonists of stimulator of interferon genes (STING), are promising agents for immunotherapy. However, the application of CDNs has been limited by their instability and low transmembrane efficiency. Here, we introduced a conjugated adjuvant of STING and TLR1/2, Pam3CSK4-CDGSF. Conjugating CDGSF with Pam3CSK4 increased the stability and intracellular delivery. In addition, by synergistically activating the STING and TLR pathways, Pam3CSK4-CDGSF was able to enhance immune activation. Both humoral and cellular immune responses were triggered by Pam3CSK4-CDGSF plus OVA (V4), and tumor growth was significantly inhibited after V4 administration. More importantly, V4 can also boost the antigen-specific CD8+ T cell response for cancer cell killing. Thus, the conjugated STING and TLR1/2 agonist Pam3CSK4-CDGSF can serve as a potent adjuvant for vaccine construction to augment antitumor immunotherapy.


Asunto(s)
Inmunoterapia , Lipopéptidos/farmacología , Proteínas de la Membrana/agonistas , Neoplasias/terapia , Receptor Toll-Like 2/agonistas , Animales , Formación de Anticuerpos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Inmunidad Celular , Ratones , Neoplasias/inmunología , Receptor Toll-Like 1/agonistas
4.
Mol Pharm ; 17(2): 417-425, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31841011

RESUMEN

Constructing an effective therapeutic cancer vaccine is very attractive and promising for cancer immunotherapy. However, the poor immunogenicity of tumor antigens and suppression of the immune system in the tumor microenvironment are two major obstacles for developing effective cancer vaccines. Invariant NKT cells (iNKT cells), which are essential bridges between the innate and adaptive immune systems, can be rapidly activated by their agonists and, consequently, evoke whole immune systems. Herein, we conjugated a potent agonist of the iNKT cell, α-galactosylceramide (α-GalCer), with the tumor-associated MUC1 glycopeptide antigens as novel self-adjuvanting cancer vaccines through click chemistry. Immunological studies revealed that the mouse immune system was potently evoked and that high levels of tumor-specific IgG antibodies were elicited by vaccine conjugates without an external adjuvant. The produced antibodies could specifically recognize and bind to antigen-expressing cancer cells and, subsequently, induce cytotoxicity through complement-dependent cytotoxicity. Thus, the insertion of α-GalCer significantly improved the immunogenicity of the MUC1 glycopeptide and induced strong antigen-specific antitumor responses, indicating that α-GalCer is an effective built-in adjuvant for constructing potent chemical synthetic antitumor vaccines.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cáncer/inmunología , Galactosilceramidas/administración & dosificación , Inmunización/métodos , Inmunogenicidad Vacunal , Células T Asesinas Naturales/inmunología , Vacunas Sintéticas/inmunología , Adyuvantes Inmunológicos/química , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/administración & dosificación , Química Clic/métodos , Células Dendríticas/inmunología , Femenino , Galactosilceramidas/química , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mucina-1/química , Mucina-1/genética , Transfección , Vacunas Sintéticas/administración & dosificación
5.
Adv Mater ; 36(16): e2308155, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38295870

RESUMEN

Following the success of the dendritic cell (DC) vaccine, the cell-based tumor vaccine shows its promise as a vaccination strategy. Except for DC cells, targeting other immune cells, especially myeloid cells, is expected to address currently unmet clinical needs (e.g., tumor types, safety issues such as cytokine storms, and therapeutic benefits). Here, it is shown that an in situ injected macroporous myeloid cell adoptive scaffold (MAS) not only actively delivers antigens (Ags) that are triggered by scaffold-infiltrating cell surface thiol groups but also releases granulocyte-macrophage colony-stimulating factor and other adjuvant combos. Consequently, this promotes cell differentiation, activation, and migration from the produced monocyte and DC vaccines (MASVax) to stimulate antitumor T-cell immunity. Neoantigen-based MASVax combined with immune checkpoint blockade induces rejection of established tumors and long-term immune protection. The combined depletion of immunosuppressive myeloid cells further enhances the efficacy of MASVax, indicating the potential of myeloid cell-based therapies for immune enhancement and normalization treatment of cancer.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Vacunación , Ingeniería Celular , Células Mieloides , Células Dendríticas
6.
Nano Res ; 15(7): 6328-6339, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35464625

RESUMEN

Immunostimulatory therapies based on pattern recognition receptors (PRRs) have emerged as an effective approach in the fight against cancer, with the ability to recruit tumor-specific lymphocytes in a low-immunogenicity tumor environment. The agonist cyclic dinucleotides (CDNs) of the stimulator of interferon gene (STING) are a group of very promising anticancer molecules that increase tumor immunogenicity by activating innate immunity. However, the tumor immune efficacy of CDNs is limited by several factors, including relatively narrow cytokine production, inefficient delivery to STING, and rapid clearance. In addition, a single adjuvant molecule is unable to elicit a broad cytokine response and thus cannot further amplify the anticancer effect. To address this problem, two or more agonist molecules are often used together to synergistically enhance immune efficacy. In this work, we found that a combination of the STING agonist CDGSF and the Toll-like receptor 7/8 (TLR7/8) agonist 522 produced a broader cytokine response. Subsequently, we developed multicomponent nanovaccines (MCNVs) consisting of a PC7A polymer as a nanocarrier encapsulating the antigen OVA and adjuvant molecules. These MCNVs activate bone marrow-derived dendritic cells (BMDCs) to produce multiple proinflammatory factors that promote antigen cross-presentation to stimulate specific antitumor T-cell responses. In in vivo experiments, we observed that MCNVs triggered a strong T-cell response in tumor-infiltrating lymphocytes, resulting in significant tumor regression and, notably, a 100% survival rate in mice through 25 days without other partnering therapies. These data suggest that our nanovaccines have great potential to advance cancer immunotherapy with increased durability and potency. Electronic Supplementary Material: Supplementary material (synthesis of CDGSF, 522, PC7A and OVA; preparation of MCNVs; representative gating strategies for flow cytometry) is available in the online version of this article at 10.1007/s12274-022-4282-x.

7.
Biomaterials ; 273: 120788, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33933912

RESUMEN

Intrinsic immune behaviors of nanomaterials and immune systems promote research on their adjuvanticity and the design of next generation nanovaccine-based immunotherapies. Herein, we report a promising multifunctional nanoadjuvant by exploring the immune-potentiating effects of black phosphorus nanosheets (BPs) in vitro and in vivo. The facile coating of BPs with phenylalanine-lysine-phenylalanine (FKF) tripeptide-modified antigen epitopes (FKF-OVAp@BP) enables the generation of a minimalized nanovaccine by integrating high loading capacity, efficient drug delivery, comprehensive dendritic cell (DC) activation, and biocompatibility for cancer immunotherapy. Systemic immunization elicits potent antitumor cellular immunity and significantly augments checkpoint blockade (CPB) against melanoma in a mouse model. Furthermore, near-infrared (NIR) photothermal effects of BPs create an immune-favorable microenvironment for improved local immunization. This study offers new insight into the integration of immunoactivity and photothermal effects for enhanced cancer immunotherapy by using a nanoadjuvant and thus potentially advances the design and application of multifunctional adjuvant materials for cancer nanotreatment.


Asunto(s)
Inmunoterapia , Fósforo , Adyuvantes Inmunológicos , Animales , Sistemas de Liberación de Medicamentos , Factores Inmunológicos , Ratones
8.
Chem Commun (Camb) ; 57(4): 504-507, 2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33331360

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra la COVID-19/administración & dosificación , COVID-19/prevención & control , Melanoma Experimental/tratamiento farmacológico , Proteínas de la Membrana/agonistas , Nucleótidos Cíclicos/administración & dosificación , Neoplasias Cutáneas/tratamiento farmacológico , Adyuvantes Inmunológicos/síntesis química , Hidróxido de Aluminio/administración & dosificación , Hidróxido de Aluminio/química , Animales , Anticuerpos Antivirales/biosíntesis , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/virología , COVID-19/inmunología , COVID-19/virología , Vacunas contra la COVID-19/química , Ensayo de Immunospot Ligado a Enzimas , Humanos , Inmunoterapia/métodos , Interferón gamma/biosíntesis , Melanoma Experimental/inmunología , Melanoma Experimental/mortalidad , Melanoma Experimental/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Nucleótidos Cíclicos/síntesis química , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Glicoproteína de la Espiga del Coronavirus/administración & dosificación , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Análisis de Supervivencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/virología , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Vacunación/métodos
9.
Front Chem ; 8: 601, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32850636

RESUMEN

Cancer is a life-threatening disease, and immunotherapies have been developed as a novel, potent treatment for cancer. Adjuvants, used alone or in combination with other agents, play crucial roles in immune activation. This is necessary for cancer immunotherapy, particularly in the construction of therapeutic cancer vaccines. Adjuvants activate antigen-presenting cells and promote the presentation of antigen epitopes on major histocompatibility complex molecules, further enhancing adaptive immune responses, including cytotoxic T lymphocytes, to elicit cancer-cell death. However, the applications of adjuvants are limited by their poor efficacy or insufficient safety. In recent studies, researchers attempted to develop safe, efficacious adjuvants for cancer immunotherapy, and many compounds (including inorganic compounds, organic molecules, polymers, and colloids) have been identified and optimized as agonists of various pathways. In this review, we focus on the discovery and structural design of emerging adjuvants and discuss how these findings benefit healthcare.

10.
Chem Commun (Camb) ; 56(83): 12632-12635, 2020 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-32960198

RESUMEN

We developed a late-stage modification strategy by a phospha-Michael addition reaction between various functional phosphines and unprotected dehydroalanine (Dha) peptides and proteins under mild conditions. This strategy was applied to generate a staple peptide to enhance its cell membrane penetrability, and it was also able to regulate α-synuclein aggregation properties and morphological characteristics with the addition of different charges.


Asunto(s)
Péptidos/química , Fosfinas/química , Fluoresceína/química , Humanos , Células MCF-7 , Microscopía Confocal , Péptidos/metabolismo , Agregado de Proteínas , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo
11.
Chem Commun (Camb) ; 54(69): 9655-9658, 2018 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-30101273
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA