Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Breast Cancer Res ; 26(1): 63, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38605414

RESUMEN

BACKGROUND: Chitinase-like proteins (CLPs) play a key role in immunosuppression under inflammatory conditions such as cancer. CLPs are enzymatically inactive and become neutralized upon binding of their natural ligand chitin, potentially reducing CLP-driven immunosuppression. We investigated the efficacy of chitin treatment in the context of triple-negative breast cancer (TNBC) using complementary mouse models. We also evaluated the immunomodulatory influence of chitin on immune checkpoint blockade (ICB) and compared its efficacy as general CLP blocker with blockade of a single CLP, i.e. chitinase 3-like 1 (CHI3L1). METHODS: Female BALB/c mice were intraductally injected with luciferase-expressing 4T1 or 66cl4 cells and systemically treated with chitin in combination with or without anti-programmed death (PD)-1 ICB. For single CLP blockade, tumor-bearing mice were treated with anti-CHI3L1 antibodies. Metastatic progression was monitored through bioluminescence imaging. Immune cell changes in primary tumors and lymphoid organs (i.e. axillary lymph nodes and spleen) were investigated through flow cytometry, immunohistochemistry, cytokine profiling and RNA-sequencing. CHI3L1-stimulated RAW264.7 macrophages were subjected to 2D lymphatic endothelial cell adhesion and 3D lymphatic integration in vitro assays for studying macrophage-mediated lymphatic remodeling. RESULTS: Chitin significantly reduced primary tumor progression in the 4T1-based model by decreasing the high production of CLPs that originate from tumor-associated neutrophils (TANs) and Stat3 signaling, prominently affecting the CHI3L1 and CHI3L3 primary tumor levels. It reduced immunosuppressive cell types and increased anti-tumorigenic T-cells in primary tumors as well as axillary lymph nodes. Chitin also significantly reduced CHI3L3 primary tumor levels and immunosuppression in the 66cl4-based model. Compared to anti-CHI3L1, chitin enhanced primary tumor growth reduction and anti-tumorigenicity. Both treatments equally inhibited lymphatic adhesion and integration of macrophages, thereby hampering lymphatic tumor cell spreading. Upon ICB combination therapy, chitin alleviated anti-PD-1 resistance in both TNBC models, providing a significant add-on reduction in primary tumor and lung metastatic growth compared to chitin monotherapy. These add-on effects occurred through additional increase in CD8α+ T-cell infiltration and activation in primary tumor and lymphoid organs. CONCLUSIONS: Chitin, as a general CLP blocker, reduces CLP production, enhances anti-tumor immunity as well as ICB responses, supporting its potential clinical relevance in immunosuppressed TNBC patients.


Asunto(s)
Quitina , Quitinasas , Neoplasias de la Mama Triple Negativas , Animales , Femenino , Humanos , Ratones , Línea Celular Tumoral , Quitina/farmacología , Quitina/uso terapéutico , Quitinasas/uso terapéutico , Terapia de Inmunosupresión , Metástasis Linfática , Proteínas/uso terapéutico , Neoplasias de la Mama Triple Negativas/patología
2.
Small ; 20(10): e2306892, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37867244

RESUMEN

Poly(I:C) is a synthetic analogue of dsRNA capable of activating both TLR3 and RLRs, such as MDA-5 and RIG-I, as pathogen recognition receptors. While poly(I:C) is known to provoke a robust type I IFN, type III IFN, and Th1 cytokine response, its therapeutic use as a vaccine adjuvant is limited due to its vulnerability to nucleases and poor uptake by immune cells. is encapsulated poly(I:C) into lipid nanoparticles (LNPs) containing an ionizable cationic lipid that can electrostatically interact with poly(I:C). LNP-formulated poly(I:C) triggered both lysosomal TLR3 and cytoplasmic RLRs, in vitro and in vivo, whereas poly(I:C) in an unformulated soluble form only triggered endosomal-localized TLR3. Administration of LNP-formulated poly(I:C) in mouse models led to efficient translocation to lymphoid tissue and concurrent innate immune activation following intramuscular (IM) administration, resulting in a significant increase in innate immune activation compared to unformulated soluble poly(I:C). When used as an adjuvant for recombinant full-length SARS-CoV-2 spike protein, LNP-formulated poly(I:C) elicited potent anti-spike antibody titers, surpassing those of unformulated soluble poly(I:C) by orders of magnitude and offered complete protection against a SARS-CoV-2 viral challenge in vivo, and serum from these mice are capable of significantly reducing viral infection in vitro.


Asunto(s)
Liposomas , Nanopartículas , Poli I-C , Glicoproteína de la Espiga del Coronavirus , Receptor Toll-Like 3 , Animales , Ratones , Humanos , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Adyuvantes Inmunológicos/farmacología
3.
Mol Ther ; 30(9): 2968-2983, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35450821

RESUMEN

Self-amplifying RNA vaccines may induce equivalent or more potent immune responses at lower doses compared to non-replicating mRNA vaccines via amplified antigen expression. In this paper, we demonstrate that 1 µg of an LNP-formulated dual-antigen self-amplifying RNA vaccine (ZIP1642), encoding both the S-RBD and N antigen, elicits considerably higher neutralizing antibody titers against Wuhan-like Beta B.1.351 and Delta B.1.617.2 SARS-CoV-2 variants compared to those of convalescent patients. In addition, ZIP1642 vaccination in mice expanded both S- and N-specific CD3+CD4+ and CD3+CD8+ T cells and caused a Th1 shifted cytokine response. We demonstrate that the induction of such dual antigen-targeted cell-mediated immune response may provide better protection against variants displaying highly mutated Spike proteins, as infectious viral loads of both Wuhan-like and Beta variants were decreased after challenge of ZIP1642 vaccinated hamsters. Supported by these results, we encourage redirecting focus toward the induction of multiple antigen-targeted cell-mediated immunity in addition to neutralizing antibody responses to bypass waning antibody responses and attenuate infectious breakthrough and disease severity of future SARS-CoV-2 variants.


Asunto(s)
COVID-19 , Vacunas Virales , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Linfocitos T CD8-positivos , COVID-19/prevención & control , Vacunas contra la COVID-19 , Cricetinae , Humanos , Inmunidad Celular , Inmunidad Humoral , Ratones , Ratones Endogámicos BALB C , ARN , SARS-CoV-2/genética , Vacunación , Vacunas Sintéticas , Vacunas de ARNm
4.
Angew Chem Int Ed Engl ; 62(23): e202301102, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-36896730

RESUMEN

Polyethylene glycol (PEG) is considered as the gold standard for colloidal stabilization of nanomedicines, yet PEG is non-degradable and lacks functionality on the backbone. Herein, we introduce concomitantly PEG backbone functionality and degradability via a one-step modification with 1,2,4-triazoline-3,5-diones (TAD) under green light. The TAD-PEG conjugates are degradable in aqueous medium under physiological conditions, with the rate of hydrolysis depending on pH and temperature. Subsequently, a PEG-lipid is modified with TAD-derivatives and successfully used for messenger RNA (mRNA) lipid nanoparticle (LNP) delivery, thereby improving mRNA transfection efficiency on multiple cell cultures in vitro. In vivo, in mice, mRNA LNP formulation exhibited a similar tissue distribution as common LNPs, with a slight decrease in transfection efficiency. Our findings pave the road towards the design of degradable, backbone-functionalized PEG for applications in nanomedicine and beyond.


Asunto(s)
Nanopartículas , Polietilenglicoles , Animales , Ratones , ARN Mensajero/genética , Liposomas , Lípidos
5.
Mol Ther ; 29(4): 1370-1381, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33484964

RESUMEN

Synthetic mRNAs are an appealing platform with multiple biomedical applications ranging from protein replacement therapy to vaccination. In comparison with conventional mRNA, synthetic self-amplifying mRNAs (sa-mRNAs) are gaining interest because of their higher and longer-lasting expression. However, sa-mRNAs also elicit an innate immune response, which may complicate their clinical application. Approaches to reduce the innate immunity of sa-mRNAs have not been studied in detail. Here we investigated, in vivo, the effect of several innate immune inhibitors and a novel cellulose-based mRNA purification approach on the type I interferon (IFN) response and the translation and vaccination efficacy of our formerly developed sa-mRNA vaccine against Zika virus. Among the investigated inhibitors, we found that corticosteroids and especially topical application of clobetasol at the sa-mRNA injection site was the most efficient in suppressing the type I IFN response and increasing the translation of sa-mRNA. However, clobetasol prevented formation of antibodies against sa-mRNA-encoded antigens and should therefore be avoided in a vaccination context. Residual dsRNA by-products of the in vitro transcription reaction are known inducers of immediate type I IFN responses. We additionally demonstrate a drastic reduction of these dsRNA by-products upon cellulose-based purification, reducing the innate immune response and improving sa-mRNA vaccination efficacy.


Asunto(s)
Inmunidad Innata/genética , ARN Mensajero/genética , Vacunación , Infección por el Virus Zika/tratamiento farmacológico , Corticoesteroides/química , Celulosa/química , Clobetasol/farmacología , Regulación de la Expresión Génica/genética , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/inmunología , ARN Mensajero/síntesis química , ARN Mensajero/química , ARN Mensajero/farmacología , Virus Zika/efectos de los fármacos , Virus Zika/patogenicidad , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
6.
Mol Ther ; 29(3): 1164-1173, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33186690

RESUMEN

Synthetic mRNA therapeutics have the potential to revolutionize healthcare, as they enable patients to produce therapeutic proteins inside their own bodies. However, convenient methods that allow external control over the timing and magnitude of protein production after in vivo delivery of synthetic mRNA are lacking. In this study, we validate the in vivo utility of a synthetic self-amplifying mRNA (RNA replicon) whose expression can be turned off using a genetic switch that responds to oral administration of trimethoprim (TMP), a US Food and Drug Administration (FDA)-approved small-molecule drug. After intramuscular electroporation, the engineered RNA replicon exhibited dose-dependent and reversible expression of its encoded protein upon TMP administration. The TMP serum level needed for maximal downregulation of protein translation was approximately 45-fold below that used in humans for therapeutic purposes. To demonstrate the therapeutic potential of the technology, we injected mice with a TMP-responsive RNA replicon encoding erythropoietin (EPO) and successfully controlled the timing and magnitude of EPO production as well as changes in hematocrit. This work demonstrates the feasibility of controlling mRNA kinetics in vivo, thereby broadly expanding the clinical versatility of mRNA therapeutics.


Asunto(s)
Eritropoyetina/metabolismo , Antagonistas del Ácido Fólico/administración & dosificación , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Replicón , Trimetoprim/administración & dosificación , Animales , Electroporación , Eritropoyetina/genética , Femenino , Terapia Genética , Inyecciones Intramusculares , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética
7.
J Am Chem Soc ; 143(26): 9872-9883, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34166595

RESUMEN

Small-molecular Toll-like receptor 7/8 (TLR7/8) agonists hold promise as immune modulators for a variety of immune therapeutic purposes including cancer therapy or vaccination. However, due to their rapid systemic distribution causing difficult-to-control inflammatory off-target effects, their application is still problematic, in particular systemically. To address this problem, we designed and robustly fabricated pH-responsive nanogels serving as versatile immunodrug nanocarriers for safe delivery of TLR7/8-stimulating imidazoquinolines after intravenous administration. To this aim, a primary amine-reactive methacrylamide monomer bearing a pendant squaric ester amide is introduced, which is polymerized under controlled RAFT polymerization conditions. Corresponding PEG-derived squaric ester amide block copolymers self-assemble into precursor micelles in polar protic solvents. Their cores are amine-reactive and can sequentially be transformed by acid-sensitive cross-linkers, dyes, and imidazoquinolines. Remaining squaric ester amides are hydrophilized affording fully hydrophilic nanogels with profound stability in human plasma but stimuli-responsive degradation upon exposure to endolysosomal pH conditions. The immunomodulatory behavior of the imidazoquinolines alone or conjugated to the nanogels was demonstrated by macrophages in vitro. In vivo, however, we observed a remarkable impact of the nanogel: After intravenous injection, a spatially controlled immunostimulatory activity was evident in the spleen, whereas systemic off-target inflammatory responses triggered by the small-molecular imidazoquinoline analogue were absent. These findings underline the potential of squaric ester-based, pH-degradable nanogels as a promising platform to permit intravenous administration routes of small-molecular TLR7/8 agonists and, thus, the opportunity to explore their adjuvant potency for systemic vaccination or cancer immunotherapy purposes.


Asunto(s)
Adyuvantes Inmunológicos/química , Ésteres/química , Nanogeles/química , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Portadores de Fármacos/química , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Inmunoterapia , Ratones Endogámicos BALB C , Micelas , Imagen Óptica , Polimerizacion , Polímeros/química
8.
Angew Chem Int Ed Engl ; 60(17): 9467-9473, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33464672

RESUMEN

The search for vaccines that protect from severe morbidity and mortality because of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19) is a race against the clock and the virus. Here we describe an amphiphilic imidazoquinoline (IMDQ-PEG-CHOL) TLR7/8 adjuvant, consisting of an imidazoquinoline conjugated to the chain end of a cholesterol-poly(ethylene glycol) macromolecular amphiphile. It is water-soluble and exhibits massive translocation to lymph nodes upon local administration through binding to albumin, affording localized innate immune activation and reduction in systemic inflammation. The adjuvanticity of IMDQ-PEG-CHOL was validated in a licensed vaccine setting (quadrivalent influenza vaccine) and an experimental trimeric recombinant SARS-CoV-2 spike protein vaccine, showing robust IgG2a and IgG1 antibody titers in mice that could neutralize viral infection in vitro and in vivo in a mouse model.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Vacunas contra la COVID-19/uso terapéutico , COVID-19/prevención & control , Imidazoles/uso terapéutico , Inmunidad Innata/efectos de los fármacos , Quinolinas/uso terapéutico , Animales , Vacunas contra la COVID-19/inmunología , Colesterol/análogos & derivados , Colesterol/inmunología , Colesterol/uso terapéutico , Femenino , Humanos , Imidazoles/inmunología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/prevención & control , Glicoproteínas de Membrana/agonistas , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Polietilenglicoles/uso terapéutico , Quinolinas/inmunología , Proteínas Recombinantes/inmunología , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/inmunología , Tensoactivos/uso terapéutico , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas
9.
J Am Chem Soc ; 142(28): 12133-12139, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32524819

RESUMEN

Synthetic immune-stimulatory drugs such as agonists of the Toll-like receptors (TLR) 7/8 are potent activators of antigen-presenting cells (APCs), however, they also induce severe side effects due to leakage from the site of injection into systemic circulation. Here, we report on the design and synthesis of an amphiphilic polymer-prodrug conjugate of an imidazoquinoline TLR7/8 agonist that in aqueous medium forms vesicular structures of 200 nm. The conjugate contains an endosomal enzyme-responsive linker enabling degradation of the vesicles and release of the TLR7/8 agonist in native form after endocytosis, which results in high in vitro TLR agonist activity. In a mouse model, locally administered vesicles provoke significantly more potent and long-lasting immune stimulation in terms of interferon expression at the injection site and in draining lymphoid tissue compared to a nonamphiphilic control and the native TLR agonist. Moreover, the vesicles induce robust activation of dendritic cells in the draining lymph node in vivo.


Asunto(s)
Imidazoles/farmacología , Glicoproteínas de Membrana/agonistas , Profármacos/farmacología , Quinolinas/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , beta-Galactosidasa/inmunología , Animales , Imidazoles/química , Imidazoles/metabolismo , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Estructura Molecular , Tamaño de la Partícula , Polietilenglicoles/química , Polietilenglicoles/metabolismo , Polietilenglicoles/farmacología , Profármacos/química , Profármacos/metabolismo , Quinolinas/química , Quinolinas/metabolismo , Propiedades de Superficie , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología , beta-Galactosidasa/química , beta-Galactosidasa/metabolismo
10.
Crit Rev Microbiol ; 46(1): 100-119, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32093536

RESUMEN

This review summarises major highlights on the structural biology of the chlamydial envelope. Chlamydiae are obligate intracellular bacteria, characterised by a unique biphasic developmental cycle. Depending on the stage of their lifecycle, they appear in the form of elementary or reticulate bodies. Since these particles have distinctive functions, it is not surprising that their envelope differs in lipid as well as in protein content. Vice versa, by identifying surface proteins, specific characteristics of the particles such as rigidity or immunogenicity may be deduced. Detailed information on the bacterial membranes will increase our understanding on the host-pathogen interactions chlamydiae employ to survive and grow and might lead to new strategies to battle chlamydial infections.


Asunto(s)
Membrana Celular/metabolismo , Chlamydia/metabolismo , Lípidos de la Membrana/metabolismo , Chlamydia/crecimiento & desarrollo , Infecciones por Chlamydia/patología , Interacciones Huésped-Patógeno , Humanos , Porinas/metabolismo
11.
Nat Chem Biol ; 14(11): 1043-1050, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30327560

RESUMEN

Synthetic mRNA is an attractive vehicle for gene therapies because of its transient nature and improved safety profile over DNA. However, unlike DNA, broadly applicable methods to control expression from mRNA are lacking. Here we describe a platform for small-molecule-based regulation of expression from modified RNA (modRNA) and self-replicating RNA (replicon) delivered to mammalian cells. Specifically, we engineer small-molecule-responsive RNA binding proteins to control expression of proteins from RNA-encoded genetic circuits. Coupled with specific modRNA dosages or engineered elements from a replicon, including a subgenomic promoter library, we demonstrate the capability to externally regulate the timing and level of protein expression. These control mechanisms facilitate the construction of ON, OFF, and two-output switches, with potential therapeutic applications such as inducible cancer immunotherapies. These circuits, along with other synthetic networks that can be developed using these tools, will expand the utility of synthetic mRNA as a therapeutic modality.


Asunto(s)
Redes Reguladoras de Genes , Terapia Genética/métodos , Regiones Promotoras Genéticas , ARN Mensajero/química , Proteínas de Unión al ARN/química , ARN/química , Animales , Línea Celular , Cricetinae , ADN/química , Biblioteca de Genes , Ingeniería Genética , Células HEK293 , Humanos , Inmunoterapia , Ratones , ARN Interferente Pequeño/metabolismo , Biología Sintética
12.
Angew Chem Int Ed Engl ; 58(43): 15390-15395, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31397948

RESUMEN

Uncontrolled systemic inflammatory immune triggering has hampered the clinical translation of several classes of small-molecule immunomodulators, such as imidazoquinoline TLR7/8 agonists for vaccine design and cancer immunotherapy. By taking advantage of the inherent serum-protein-binding property of lipid motifs and their tendency to accumulate in lymphoid tissue, we designed amphiphilic lipid-polymer conjugates that suppress systemic inflammation but provoke potent lymph-node immune activation. This work provides a rational basis for the design of lipid-polymer amphiphiles for optimized lymphoid targeting.


Asunto(s)
Inmunidad Innata , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Colesterol/química , Imidazoles/química , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/química , Factores Inmunológicos/metabolismo , Factores Inmunológicos/farmacología , Lípidos/química , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , FN-kappa B/metabolismo , Polímeros/química , Quinolinas/química , Quinolinas/farmacología , Células RAW 264.7 , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo
13.
J Am Chem Soc ; 140(43): 14300-14307, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30277761

RESUMEN

Small molecule immuno-modulators such as agonists of Toll-like receptors (TLRs) are attractive compounds to stimulate innate immune cells toward potent antiviral and antitumor responses. However, small molecules rapidly enter the systemic circulation and cause "wasted inflammation". Hence, synthetic strategies to confine their radius of action to lymphoid tissue are of great relevance, to both enhance their efficacy and concomitantly limit toxicity. Here, we demonstrate that covalent conjugation of a small molecule TLR7/8 agonist immunomodulatory to a micelle-forming amphiphilic block copolymer greatly alters the pharmacokinetic profile, resulting in highly efficient lymphatic delivery. Moreover, we designed amphiphilic block copolymers in such a way to form thermodynamically stable micelles through π-π stacking between aromatic moieties, and we engineered the block copolymers to undergo an irreversible amphiphilic to hydrophilic transition in response to the acidic endosomal pH.


Asunto(s)
Ganglios Linfáticos/efectos de los fármacos , Polímeros/farmacología , Tensoactivos/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Humanos , Concentración de Iones de Hidrógeno , Ganglios Linfáticos/inmunología , Micelas , Modelos Moleculares , Estructura Molecular , Polímeros/química , Tensoactivos/química , Termodinámica , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología
14.
Mol Pharm ; 15(2): 377-384, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29297692

RESUMEN

Synthetic mRNA is becoming increasingly popular as an alternative to pDNA-based gene therapy. Currently, multiple synthetic mRNA platforms have been developed. In this study we investigated the expression kinetics and the changes in mRNA encoding cytokine and chemokine levels following intradermal electroporation in pigs of pDNA, self-replicating mRNA, and modified and unmodified mRNA. The self-replicating mRNA tended to induce the highest protein expression, followed by pDNA, modified mRNA, and unmodified mRNA. Interestingly, the self-replicating mRNA was able to maintain its high expression levels during at least 12 days. In contrast, the expression of pDNA and the nonreplicating mRNAs dropped after respectively one and two days. Six days after intradermal electroporation a dose-dependent expression was observed for all vectors. Again, also at lower doses, the self-replicating mRNA tended to show the highest expression. All the mRNA vectors, including the modified mRNA, induced elevated levels of mRNA encoding cytokines and chemokines in the porcine skin after intradermal electroporation, while no such response was noticed after intradermal electroporation of the pDNA vector.


Asunto(s)
ADN Circular/administración & dosificación , Técnicas de Transferencia de Gen , Inmunidad/genética , ARN Mensajero/administración & dosificación , Animales , Quimiocinas/inmunología , Quimiocinas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , ADN Circular/genética , ADN Circular/metabolismo , Electroporación/métodos , Femenino , Terapia Genética/métodos , Vectores Genéticos/genética , Cinética , Ratones Endogámicos BALB C , Modelos Animales , Plásmidos/administración & dosificación , Plásmidos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Piel/metabolismo , Sus scrofa
16.
Cancer Immunol Immunother ; 66(12): 1545-1555, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28776079

RESUMEN

In this study, a xenogeneic DNA vaccine encoding for human vascular endothelial growth factor receptor-2 (hVEGFR-2) was evaluated in two murine tumor models, the B16-F10 melanoma and the EO771 breast carcinoma model. The vaccine was administered by intradermal injection followed by electroporation. The immunogenicity and the biological efficacy of the vaccine were tested in (1) a prophylactic setting, (2) a therapeutic setting, and (3) a therapeutic setting combined with surgical removal of the primary tumor. The tumor growth, survival, and development of an immune response were followed. The cellular immune response was measured by a bioluminescence-based cytotoxicity assay with vascular endothelial growth factor-2 (VEGFR-2)-expressing target cells. Humoral immune responses were quantified by enzyme-linked immunosorbent assay (ELISA). Ex vivo bioluminescence imaging and immunohistological observation of organs were used to detect (micro)metastases. A cellular and humoral immune response was present in prophylactically and therapeutically vaccinated mice, in both tumor models. Nevertheless, survival in prophylactically vaccinated mice was only moderately increased, and no beneficial effect on survival in therapeutically vaccinated mice could be demonstrated. An influx of CD3+ cells and a slight decrease in VEGFR-2 were noticed in the tumors of vaccinated mice. Unexpectedly, the vaccine caused an increased quantity of early micrometastases in the liver. Lung metastases were not increased by the vaccine. These early liver micrometastases did however not grow into macroscopic metastases in either control or vaccinated mice when allowed to develop further after surgical removal of the primary tumor.


Asunto(s)
Neoplasias de la Mama/genética , Melanoma/genética , Vacunas de ADN/inmunología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
18.
Mol Ther ; 29(3): 893-894, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33549165
19.
Mol Ther ; 24(9): 1686-96, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27434590

RESUMEN

DNA vaccination holds great promise for the prevention and treatment of cancer and infectious diseases. However, the clinical ability of DNA vaccines is still controversial due to the limited immune response initially observed in humans. We hypothesized that electroporation of a plasmid encoding the HIV-1 Gag viral capsid protein would enhance cancer DNA vaccine potency. DNA electroporation used to deliver plasmids in vivo, induced type I interferons, thereby supporting the activation of innate immunity. The coadministration of ovalbumin (OVA) and HIV-1 Gag encoding plasmids modulated the adaptive immune response. This strategy favored antigen-specific Th1 immunity, delayed B16F10-OVA tumor growth and improved mouse survival in both prophylactic and therapeutic vaccination approaches. Similarly, a prophylactic DNA immunization against the melanoma-associated antigen gp100 was enhanced by the codelivery of the HIV-1 Gag plasmid. The adjuvant effect was not driven by the formation of HIV-1 Gag virus-like particles. This work highlights the ability of both electroporation and the HIV-1 Gag plasmid to stimulate innate immunity for enhancing cancer DNA vaccine immunogenicity and demonstrates interesting tracks for the design of new translational genetic adjuvants to overcome the current limitations of DNA vaccines in humans.


Asunto(s)
Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Plásmidos/genética , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Animales , Línea Celular , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Interferón Tipo I/biosíntesis , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Plásmidos/administración & dosificación , Modelos de Riesgos Proporcionales , Células TH1/inmunología , Células TH1/metabolismo , Transfección , Resultado del Tratamiento , Carga Tumoral , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/ultraestructura
20.
Mol Ther ; 24(11): 2012-2020, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27506450

RESUMEN

Given their high potential to evoke cytolytic T cell responses, tumor antigen-encoding messenger RNA (mRNA) vaccines are now being intensively explored as therapeutic cancer vaccines. mRNA vaccines clearly benefit from wrapping the mRNA into nano-sized carriers such as lipoplexes that protect the mRNA from degradation and increase its uptake by dendritic cells in vivo. Nevertheless, the early innate host factors that regulate the induction of cytolytic T cells to mRNA lipoplex vaccines have remained unresolved. Here, we demonstrate that mRNA lipoplexes induce a potent type I interferon (IFN) response upon subcutaneous, intradermal and intranodal injection. Regardless of the route of immunization applied, these type I IFNs interfered with the generation of potent cytolytic T cell responses. Most importantly, blocking type I IFN signaling at the site of immunization through the use of an IFNAR blocking antibody greatly enhanced the prophylactic and therapeutic antitumor efficacy of mRNA lipoplexes in the highly aggressive B16 melanoma model. As type I IFN induction appears to be inherent to the mRNA itself rather than to unique properties of the mRNA lipoplex formulation, preventing type I IFN induction and/or IFNAR signaling at the site of immunization might constitute a widely applicable strategy to improve the potency of mRNA vaccination.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Interferón Tipo I/metabolismo , Melanoma Experimental/tratamiento farmacológico , ARN Mensajero/administración & dosificación , Linfocitos T Citotóxicos/metabolismo , Animales , Anticuerpos/administración & dosificación , Vacunas contra el Cáncer/inmunología , Humanos , Inyecciones Intradérmicas , Inyecciones Subcutáneas , Liposomas , Melanoma Experimental/inmunología , Ratones , ARN Mensajero/inmunología , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA