Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Control Release ; 370: 379-391, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38697317

RESUMO

Although various types of mRNA-based vaccines have been explored, the optimal conditions for induction of both humoral and cellular immunity remain rather unknown. In this study, mRNA vaccines of nucleoside-modified mRNA in lipoplexes (LPXs) or lipid nanoparticles (LNPs) were evaluated after administration in mice through different routes, assessing mRNA delivery, tolerability and immunogenicity. In addition, we investigated whether mRNA vaccines could benefit from the inclusion of the adjuvant alpha-galactosylceramide (αGC), an invariant Natural Killer T (iNKT) cell ligand. Intramuscular (IM) vaccination with ovalbumin (OVA)-encoding mRNA encapsulated in LNPs adjuvanted with αGC showed the highest antibody- and CD8+ T cell responses. Furthermore, we observed that addition of signal peptides and endocytic sorting signals of either LAMP1 or HLA-B7 in the OVA-encoding mRNA sequence further enhanced CD8+ T cell activation although reducing the induction of IgG antibody responses. Moreover, mRNA LNPs with the ionizable lipidoid C12-200 exhibited higher pro-inflammatory- and reactogenic activity compared to mRNA LNPs with SM-102, correlating with increased T cell activation and antitumor potential. We also observed that αGC could further enhance the cellular immunity of clinically relevant mRNA LNP vaccines, thereby promoting therapeutic antitumor potential. Finally, a Listeria monocytogenes mRNA LNP vaccine supplemented with αGC showed synergistic protective effects against listeriosis, highlighting a key advantage of co-activating iNKT cells in antibacterial mRNA vaccines. Taken together, our study offers multiple insights for optimizing the design of mRNA vaccines for disease applications, such as cancer and intracellular bacterial infections.


Assuntos
Vacinas Anticâncer , Galactosilceramidas , Camundongos Endogâmicos C57BL , Nanopartículas , Ovalbumina , Animais , Galactosilceramidas/administração & dosagem , Galactosilceramidas/química , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Feminino , Nanopartículas/química , Nanopartículas/administração & dosagem , Ovalbumina/imunologia , Ovalbumina/administração & dosagem , Vacinas de mRNA , Adjuvantes Imunológicos/administração & dosagem , Linfócitos T CD8-Positivos/imunologia , RNA Mensageiro/administração & dosagem , Camundongos , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Lipídeos/química , Lipossomos
2.
J Mol Biol ; 436(2): 168385, 2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-38065276

RESUMO

Throughout the last decades, mRNA vaccines have been developed as a cancer immunotherapeutic and the technology recently gained momentum during the COVID-19 pandemic. Recent promising results obtained from clinical trials investigating lipid-based mRNA vaccines in cancer therapy further highlighted the potential of this therapy. Interestingly, while the technologies being used in authorized mRNA vaccines for the prevention of COVID-19 are relatively similar, mRNA vaccines in clinical development for cancer vaccination show marked differences in mRNA modification, lipid carrier, and administration route. In this review, we describe findings on how these factors can impact the potency of mRNA vaccines in cancer therapy and provide insights into the complex interplay between them. We discuss how lipid carrier composition can affect passive targeting to immune cells to improve the efficacy and safety of mRNA vaccines. Finally, we summarize strategies that are established or still being explored to improve the efficacy of mRNA cancer vaccines and include next-generation vaccines that are on the horizon in clinical development.


Assuntos
Vacinas Anticâncer , Lipídeos , Neoplasias , Desenvolvimento de Vacinas , Vacinas de mRNA , Humanos , Neoplasias/terapia , Desenvolvimento de Vacinas/métodos
3.
Theranostics ; 13(15): 5483-5500, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37908728

RESUMO

Rationale: Although promising responses are obtained in patients treated with immune checkpoint inhibitors targeting programmed death ligand 1 (PD-L1) and its receptor programmed death-1 (PD-1), only a fraction of patients benefits from this immunotherapy. Cancer vaccination may be an effective approach to improve the response to immune checkpoint inhibitors anti-PD-L1/PD-1 therapy. However, there is a lack of research on the dynamics of PD-L1 expression in response to cancer vaccination. Methods: We performed non-invasive whole-body imaging to visualize PD-L1 expression at different timepoints after vaccination of melanoma-bearing mice. Mice bearing ovalbumin (OVA) expressing B16 tumors were i.v. injected with the Galsome mRNA vaccine: OVA encoding mRNA lipoplexes co-encapsulating a low or a high dose of the atypical adjuvant α-galactosylceramide (αGC) to activate invariant natural killer T (iNKT) cells. Serial non-invasive whole-body immune imaging was performed using a technetium-99m (99mTc)-labeled anti-PD-L1 nanobody, single-photon emission computerized tomography (SPECT) and X-ray computed tomography (CT) images were quantified. Additionally, cellular expression of PD-L1 was evaluated with flow cytometry. Results: SPECT/CT-imaging showed a rapid and systemic upregulation of PD-L1 after vaccination. PD-L1 expression could not be correlated to the αGC-dose, although we observed a dose-dependent iNKT cell activation. Dynamics of PD-L1 expression were organ-dependent and most pronounced in lungs and liver, organs to which the vaccine was distributed. PD-L1 expression in lungs increased immediately after vaccination and gradually decreased over time, whereas in liver, vaccination-induced PD-L1 upregulation was short-lived. Flow cytometric analysis of these organs further showed myeloid cells as well as non-immune cells with elevated PD-L1 expression in response to vaccination. SPECT/CT imaging of the tumor demonstrated that the expression of PD-L1 remained stable over time and was overall not affected by vaccination although flow cytometric analysis at the cellular level demonstrated changes in PD-L1 expression in various immune cell populations following vaccination. Conclusion: Repeated non-invasive whole-body imaging using 99mTc-labeled anti-PD-L1 nanobodies allows to document the dynamic nature of PD-L1 expression upon vaccination. Galsome vaccination rapidly induced systemic upregulation of PD-L1 expression with the most pronounced upregulation in lungs and liver while flow cytometry analysis showed upregulation of PD-L1 in the tumor microenvironment. This study shows that imaging using nanobodies may be useful for monitoring vaccine-mediated PD-L1 modulation in patients and could provide a rationale for combination therapy. To the best of our knowledge, this is the first report that visualizes PD-L1 expression upon cancer vaccination.


Assuntos
Melanoma , Células T Matadoras Naturais , Anticorpos de Domínio Único , Humanos , Camundongos , Animais , Antígeno B7-H1 , Células T Matadoras Naturais/metabolismo , Anticorpos de Domínio Único/metabolismo , Inibidores de Checkpoint Imunológico/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Linfócitos T CD8-Positivos , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X , Vacinas Sintéticas , Melanoma/diagnóstico por imagem , Melanoma/terapia , Microambiente Tumoral , Vacinas de mRNA
4.
Nat Commun ; 13(1): 6075, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-36241641

RESUMO

Listeria monocytogenes is a foodborne intracellular bacterial pathogen leading to human listeriosis. Despite a high mortality rate and increasing antibiotic resistance no clinically approved vaccine against Listeria is available. Attenuated Listeria strains offer protection and are tested as antitumor vaccine vectors, but would benefit from a better knowledge on immunodominant vector antigens. To identify novel antigens, we screen for Listeria peptides presented on the surface of infected human cell lines by mass spectrometry-based immunopeptidomics. In between more than 15,000 human self-peptides, we detect 68 Listeria immunopeptides from 42 different bacterial proteins, including several known antigens. Peptides presented on different cell lines are often derived from the same bacterial surface proteins, classifying these antigens as potential vaccine candidates. Encoding these highly presented antigens in lipid nanoparticle mRNA vaccine formulations results in specific CD8+ T-cell responses and induces protection in vaccination challenge experiments in mice. Our results can serve as a starting point for the development of a clinical mRNA vaccine against Listeria and aid to improve attenuated Listeria vaccines and vectors, demonstrating the power of immunopeptidomics for next-generation bacterial vaccine development.


Assuntos
Listeria monocytogenes , Listeria , Listeriose , Animais , Proteínas de Bactérias/genética , Vacinas Bacterianas/genética , Linfócitos T CD8-Positivos , Humanos , Epitopos Imunodominantes , Lipossomos , Listeria/genética , Listeria monocytogenes/genética , Listeriose/prevenção & controle , Proteínas de Membrana , Camundongos , Nanopartículas , Vacinas Atenuadas , Vacinas Sintéticas/genética , Vacinas de mRNA
5.
Mol Cancer ; 20(1): 48, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658037

RESUMO

mRNA therapeutics have become the focus of molecular medicine research. Various mRNA applications have reached major milestones at high speed in the immuno-oncology field. This can be attributed to the knowledge that mRNA is one of nature's core building blocks carrying important information and can be considered as a powerful vector for delivery of therapeutic proteins to the patient.For a long time, the major focus in the use of in vitro transcribed mRNA was on development of cancer vaccines, using mRNA encoding tumor antigens to modify dendritic cells ex vivo. However, the versatility of mRNA and its many advantages have paved the path beyond this application. In addition, due to smart design of both the structural properties of the mRNA molecule as well as pharmaceutical formulations that improve its in vivo stability and selective targeting, the therapeutic potential of mRNA can be considered as endless.As a consequence, many novel immunotherapeutic strategies focus on the use of mRNA beyond its use as the source of tumor antigens. This review aims to summarize the state-of-the-art on these applications and to provide a rationale for their clinical application.


Assuntos
Antígenos de Neoplasias/metabolismo , Neoplasias/imunologia , Vacinas Sintéticas/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Desenho de Fármacos , Humanos , Neoplasias/tratamento farmacológico , Microambiente Tumoral , Vacinas de mRNA
6.
Mol Ther Nucleic Acids ; 20: 777-787, 2020 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-32438313

RESUMO

To date, mRNA-based biologics have mainly been developed for prophylactic and therapeutic vaccination to combat infectious diseases or cancer. In the past years, optimization of the characteristics of in vitro transcribed mRNA has led to significant reduction of the inflammatory responses. Thanks to this, mRNA therapeutics have entered the field of passive immunization. Here, we established an mRNA treatment that is based on mRNA that codes for a bispecific single-domain antibody construct that can selectively recruit innate immune cells to cells infected with influenza A virus. The constructs consist of a single-domain antibody that binds to the ectodomain of the conserved influenza A matrix protein 2, while the other single-domain antibody binds to the activating mouse Fcγ receptor IV. Formulating the mRNA into DOTAP (1,2-dioleoyl-3-trimethylammonium-propane)/cholesterol nanoparticles and delivering these intratracheally to mice allowed the production of the bispecific single-domain antibody in the lungs, and administration of these mRNA-particles prior to influenza A virus infection was associated with a significant reduction in viral titers and a reduced morbidity in mice. Overall, our data provide evidence that the local delivery of mRNA encoding a bispecific single-domain antibody format in the lungs could be a promising pulmonary antiviral prophylactic treatment.

7.
ACS Nano ; 13(2): 1655-1669, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30742405

RESUMO

Messenger RNA encoding tumor antigens has the potential to evoke effective antitumor immunity. This study reports on a nanoparticle platform, named mRNA Galsomes, that successfully co-delivers nucleoside-modified antigen-encoding mRNA and the glycolipid antigen and immunopotentiator α-galactosylceramide (α-GC) to antigen-presenting cells after intravenous administration. By co-formulating low doses of α-GC, mRNA Galsomes induce a pluripotent innate and adaptive tumor-specific immune response in mice, with invariant natural killer T cells (iNKT) as a driving force. In comparison, mRNA Galsomes exhibit advantages over the state-of-the-art cancer vaccines using unmodified ovalbumin (OVA)-encoding mRNA, as we observed up to seven times more tumor-infiltrating antigen-specific cytotoxic T cells, combined with a strong iNKT cell and NK cell activation. In addition, the presence of suppressive myeloid cells (myeloid-derived suppressor cells and tumor-associated macrophages) in the tumor microenvironment was significantly lowered. Owing to these antitumor effects, OVA mRNA Galsomes significantly reduced tumor growth in established E.G7-OVA lymphoma, with a complete tumor rejection in 40% of the animals. Moreover, therapeutic vaccination with mRNA Galsomes enhanced the responsiveness to treatment with a PD-L1 checkpoint inhibitor in B16-OVA melanoma, as evidenced by a synergistic reduction of tumor outgrowth and a significantly prolonged median survival. Taken together, these data show that intravenously administered mRNA Galsomes can provide controllable, multifaceted, and effective antitumor immunity, especially when combined with checkpoint inhibition.


Assuntos
Vacinas Anticâncer/química , Vacinas Anticâncer/uso terapêutico , Células T Matadoras Naturais/metabolismo , RNA Mensageiro/química , Animais , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer/imunologia , Feminino , Galactosilceramidas/química , Imunidade Celular/fisiologia , Estimativa de Kaplan-Meier , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Lipossomos/química , Ativação Linfocitária/fisiologia , Linfoma/prevenção & controle , Melanoma/prevenção & controle , Melanoma Experimental/imunologia , Melanoma Experimental/prevenção & controle , Camundongos , Células T Matadoras Naturais/imunologia , Ovalbumina/química , Linfócitos T/imunologia , Linfócitos T/metabolismo
8.
J Control Release ; 266: 287-300, 2017 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-28987878

RESUMO

This study reports on the design of mRNA and adjuvant-loaded lipid nanoparticles for therapeutic cancer vaccination. The use of nucleoside-modified mRNA has previously been shown to improve the translational capacity and safety of mRNA-therapeutics, as it prevents the induction of type I interferons (IFNs). However, type I IFNs were identified as the key molecules that trigger the activation of antigen presenting cells, and as such drive T cell immunity. We demonstrate that nucleoside-modified mRNA can be co-delivered with the clinically approved TLR agonist monophosphoryl lipid A (MPLA). As such, we simultaneously allow high antigen expression in vivo while substituting the type I IFN response by a more controllable adjuvant. This strategy shows promise to induce effective antigen-specific T cell immunity and may be useful to enhance the safety of mRNA vaccines.


Assuntos
Citidina/análogos & derivados , Lipídeo A/análogos & derivados , Neoplasias/terapia , RNA Mensageiro/administração & dosagem , Receptores Toll-Like/agonistas , Animais , Vacinas Anticâncer , Citidina/administração & dosagem , Citidina/química , Células Dendríticas/imunologia , Feminino , Imunoterapia , Lipídeo A/administração & dosagem , Lipídeos/administração & dosagem , Lipídeos/química , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , RNA Mensageiro/química , RNA Mensageiro/imunologia
9.
Mol Ther Nucleic Acids ; 5(6): e326, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-27327138

RESUMO

Cancer vaccines based on mRNA are extensively studied. The fragile nature of mRNA has instigated research into carriers that can protect it from ribonucleases and as such enable its systemic use. However, carrier-mediated delivery of mRNA has been linked to production of type I interferon (IFN) that was reported to compromise the effectiveness of mRNA vaccines. In this study, we evaluated a cationic lipid for encapsulation of mRNA. The nanometer-sized, negatively charged lipid mRNA particles (LMPs) efficiently transfected dendritic cells and macrophages in vitro. Furthermore, i.v. delivery of LMPs resulted in rapid expression of the mRNA-encoded protein in spleen and liver, predominantly in CD11c(+) cells and to a minor extent in CD11b(+) cells. Intravenous immunization of mice with LMPs containing ovalbumin, human papilloma virus E7, and tyrosinase-related protein-2 mRNA, either combined or separately, elicited strong antigen-specific T-cell responses. We further showed the production of type I IFNs upon i.v. LMP delivery. Although this decreased the expression of the mRNA-encoded protein, it supported the induction of antigen-specific T-cell responses. These data question the current notion that type I IFNs hamper particle-mediated mRNA vaccines.

10.
J Control Release ; 195: 138-46, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24960224

RESUMO

Dendritic cell (DC)-based cancer vaccination has shown great potential in cancer immunotherapy. As a result, novel nanoparticles aiming to load DCs with tumor antigens are being developed and evaluated in vitro. For this, murine bone marrow-derived DCs (BM-DCs) are most commonly used as model DCs. However, many different protocols exist to generate these cells. Therefore, we investigated to what extent different BM-DC culture protocols impact on the immunobiology of the cells, as well as their response to particulate antigens. We evaluated 4 different BM-DC protocols with 2 main variables: bovine serum and cytokine combinations. Our results show distinct differences in yield, phenotypical maturation status and the production of immune stimulatory and immune suppressive cytokines by the different BM-DCs. Importantly, we demonstrate that the antigen-loading of these different BM-DCs via transfection with mRNA lipoplexes results in large differences in transfection efficiency as well as in the capacity of mRNA-transfected BM-DCs to stimulate antigen-specific T cells. Thus, it is clear that the BM-DC model can have significant confounding effects on the evaluation of novel nanoparticulate vaccines. To take this into account when testing novel particulate antigen-delivery systems in BM-DC models, we propose to (1) perform a thorough immunological characterization of the BM-DCs and to (2) not only judge a particle's potential for cancer vaccination based on transfection efficiency, but also to include an evaluation of functional end-points such as T cell activation.


Assuntos
Antígenos/genética , Células Dendríticas/imunologia , Nanopartículas/administração & dosagem , RNA Mensageiro/administração & dosagem , Vacinas/administração & dosagem , Animais , Antígeno B7-2/imunologia , Células da Medula Óssea/citologia , Antígenos CD40/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Citocinas/imunologia , Células Dendríticas/citologia , Genes MHC da Classe II/imunologia , Proteínas de Fluorescência Verde/genética , Lipossomos , Luciferases/genética , Camundongos Endogâmicos C57BL , Ovalbumina/genética , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA