Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 966
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36835479

RESUMO

The worldwide spread of COVID-19 continues to impact our lives and has led to unprecedented damage to global health and the economy. This highlights the need for an efficient approach to rapidly develop therapeutics and prophylactics against SARS-CoV-2. We modified a single-domain antibody, SARS-CoV-2 VHH, to the surface of the liposomes. These immunoliposomes demonstrated a good neutralizing ability, but could also carry therapeutic compounds. Furthermore, we used the 2019-nCoV RBD-SD1 protein as an antigen with Lip/cGAMP as the adjuvant to immunize mice. Lip/cGAMP enhanced the immunity well. It was demonstrated that the combination of RBD-SD1 and Lip/cGAMP was an effective preventive vaccine. This work presented potent therapeutic anti-SARS-CoV-2 drugs and an effective vaccine to prevent the spread of COVID-19.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19 , SARS-CoV-2 , Anticorpos de Domínio Único , Animais , Camundongos , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/química , Anticorpos Antivirais/uso terapêutico , COVID-19/terapia , Lipossomos/imunologia , SARS-CoV-2/imunologia , Anticorpos de Domínio Único/uso terapêutico
2.
J Med Chem ; 65(4): 3563-3574, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35108485

RESUMO

Safe and effective vaccines are the best method to defeat worldwide SARS-CoV-2 and its circulating variants. The SARS-CoV-2 S protein and its subunits are the most attractive targets for the development of protein-based vaccines. In this study, we evaluated three lipophilic adjuvants, monophosphoryl lipid A (MPLA), Toll-like receptor (TLR) 1/2 ligand Pam3CSK4, and α-galactosylceramide (α-GalCer), in liposomal and nonliposomal vaccines. The immunological results showed that the MPLA-adjuvanted liposomal vaccine induced the strongest humoral and cellular immunity. Therefore, we further performed a systematic comparison of S-trimer, S-ECD, S1, and RBD as antigens in MPLA-adjuvanted liposomes and found that, although these four vaccines all induced robust specific antibody responses, only S-trimer, S1, and RBD liposomes, but not S-ECD, elicited potent neutralizing antibody responses. Moreover, RBD, S-trimer, and S1 liposomes effectively neutralized variants (B.1.1.7/alpha, B.1.351/beta, P.1/gamma, B.1.617.2/delta, and B.1.1.529/omicron). These results provide important information for the subunit vaccine design against SARS-CoV-2 and its variants.


Assuntos
Anticorpos Antivirais/imunologia , Lipídeo A/análogos & derivados , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/química , Feminino , Lipídeo A/química , Lipídeo A/imunologia , Lipossomos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Vacinação , Vacinas de Subunidades Antigênicas/química
3.
Molecules ; 27(2)2022 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-35056718

RESUMO

The advancements in the field of nanotechnology have provided a great platform for the development of effective antiviral vaccines. Liposome-mediated delivery of antigens has been shown to induce the antigen-specific stimulation of the humoral and cell-mediated immune responses. Here, we prepared dried, reconstituted vesicles (DRVs) from DPPC liposomes and used them as the vaccine carrier system for the Middle East respiratory syndrome coronavirus papain-like protease (DRVs-MERS-CoV PLpro). MERS-CoV PLpro emulsified in the Incomplete Freund's Adjuvant (IFA-MERS-CoV PLpro) was used as a control. Immunization of mice with DRVs-MERS-CoV PLpro did not induce any notable toxicity, as revealed by the levels of the serum alanine transaminase (ALT), aspartate transaminase (AST), blood urea nitrogen (BUN) and lactate dehydrogenase (LDH) in the blood of immunized mice. Immunization with DRVs-MERS-CoV PLpro induced greater antigen-specific antibody titer and switching of IgG1 isotyping to IgG2a as compared to immunization with IFA-MERS-CoV PLpro. Moreover, splenocytes from mice immunized with DRVs-MERS-CoV PLpro exhibited greater proliferation in response to antigen stimulation. Moreover, splenocytes from DRVs-MERS-CoV PLpro-immunized mice secreted significantly higher IFN-γ as compared to splenocytes from IFA-MERS-CoV PLpro mice. In summary, DRVs-MERS-CoV PLpro may prove to be an effective prophylactic formulation to prevent MERS-CoV infection.


Assuntos
Proteases Semelhantes à Papaína de Coronavírus/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Animais , Proliferação de Células , Infecções por Coronavirus/prevenção & controle , Feminino , Imunidade Celular , Imunidade Humoral , Imunização/métodos , Imunoglobulina G/sangue , Interferon gama/metabolismo , Lipossomos/administração & dosagem , Lipossomos/química , Lipossomos/imunologia , Lipossomos/toxicidade , Linfócitos/metabolismo , Camundongos , Vacinas Virais/química , Vacinas Virais/toxicidade
4.
J Med Chem ; 65(3): 2558-2570, 2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35073081

RESUMO

Safe and effective vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants are the best approach to successfully combat the COVID-19 pandemic. The receptor-binding domain (RBD) of the viral spike protein is a major target to develop candidate vaccines. α-Galactosylceramide (αGalCer), a potent invariant natural killer T cell (iNKT) agonist, was site-specifically conjugated to the N-terminus of the RBD to form an adjuvant-protein conjugate, which was anchored on the liposome surface. This is the first time that an iNKT cell agonist was conjugated to the protein antigen. Compared to the unconjugated RBD/αGalCer mixture, the αGalCer-RBD conjugate induced significantly stronger humoral and cellular responses. The conjugate vaccine also showed effective cross-neutralization to all variants of concern (B.1.1.7/alpha, B.1.351/beta, P.1/gamma, B.1.617.2/delta, and B.1.1.529/omicron). These results suggest that the self-adjuvanting αGalCer-RBD has great potential to be an effective COVID-19 vaccine candidate, and this strategy might be useful for designing various subunit vaccines.


Assuntos
Vacinas contra COVID-19/uso terapêutico , COVID-19/terapia , Galactosilceramidas/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , SARS-CoV-2/imunologia , Vacinas Conjugadas/uso terapêutico , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/uso terapêutico , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vacinas contra COVID-19/química , Vacinas contra COVID-19/imunologia , Feminino , Galactosilceramidas/química , Galactosilceramidas/imunologia , Imunidade Humoral/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Interferon gama/metabolismo , Lipossomos/química , Lipossomos/imunologia , Lipossomos/uso terapêutico , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Domínios Proteicos , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/uso terapêutico , Vacinas Conjugadas/química , Vacinas Conjugadas/imunologia
5.
PLoS Pathog ; 18(1): e1010255, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35073387

RESUMO

Nucleoside modified mRNA combined with Acuitas Therapeutics' lipid nanoparticles (LNPs) has been shown to support robust humoral immune responses in many preclinical animal vaccine studies and later in humans with the SARS-CoV-2 vaccination. We recently showed that this platform is highly inflammatory due to the LNPs' ionizable lipid component. The inflammatory property is key to support the development of potent humoral immune responses. However, the mechanism by which this platform drives T follicular helper (Tfh) cells and humoral immune responses remains unknown. Here we show that lack of Langerhans cells or cDC1s neither significantly affected the induction of PR8 HA and SARS-CoV-2 RBD-specific Tfh cells and humoral immune responses, nor susceptibility towards the lethal challenge of influenza and SARS-CoV-2. However, the combined deletion of these two DC subsets led to a significant decrease in the induction of PR8 HA and SARS-CoV-2 RBD-specific Tfh cell and humoral immune responses. Despite these observed defects, these mice remained protected from lethal influenza and SARS-CoV-2 challenges. We further found that IL-6, unlike neutrophils, was required to generate normal Tfh cells and antibody responses, but not for protection from influenza challenge. In summary, here we bring evidence that the mRNA-LNP platform can support the induction of protective immune responses in the absence of certain innate immune cells and cytokines.


Assuntos
Vacinas contra COVID-19/imunologia , Células Dendríticas/imunologia , Vacinas contra Influenza/imunologia , Células de Langerhans/imunologia , Lipossomos/imunologia , Vacinas Sintéticas/imunologia , Vacinas de mRNA/imunologia , Animais , COVID-19/imunologia , Camundongos , Nanopartículas , Infecções por Orthomyxoviridae/imunologia , SARS-CoV-2/imunologia
6.
Drug Metab Pharmacokinet ; 41: 100424, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34757287

RESUMO

Nucleic acid therapeutics are developing into precise medicines that can manipulate specific genes. However, the development of safe and effective delivery system for the target cells has remained a challenge. Lipid nanoparticles (LNPs) have provided a revolutionary delivery system that can ensure multiple clinical translation of RNA-based candidates. In 2018, Patisiran (Onpattro) was first approved as an LNP-based siRNA drug. In 2020, during the coronavirus disease 2019 (COVID-19) outbreak, LNPs have enabled the development of two SARS-CoV-2 mRNA vaccines, Tozinameran (Comirnaty or Pfizer-BioNTech COVID-19 vaccine) and Elasomeran (Spikevax or COVID-19 vaccine Moderna) for conditional approval. Here, we reviewed the state-of-the-art LNP technology employed in three approved drugs (one siRNA-based and two mRNA-based drugs) and discussed the differences in their mode of action, formulation design, and biodistribution.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Lipossomos/imunologia , RNA Interferente Pequeno/imunologia , Vacinas Sintéticas/imunologia , Vacinas de mRNA/imunologia , Animais , Humanos , Nanopartículas , Tecnologia/métodos
7.
Int Immunopharmacol ; 101(Pt A): 108280, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34710845

RESUMO

The COVID-19 pandemic, caused by a highly virulent and transmissible pathogen, has proven to be devastating to society. Mucosal vaccines that can induce antigen-specific immune responses in both the systemic and mucosal compartments are considered an effective measure to overcome infectious diseases caused by pathogenic microbes. We have recently developed a nasal vaccine system using cationic liposomes composed of 1,2-dioleoyl-3-trimethylammonium-propane and cholesteryl 3ß-N-(dimethylaminoethyl)carbamate in mice. However, the comprehensive molecular mechanism(s), especially the host soluble mediator involved in this process, by which cationic liposomes promote antigen-specific mucosal immune responses, remain to be elucidated. Herein, we show that intranasal administration of cationic liposomes elicited interleukin-6 (IL-6) expression at the site of administration. Additionally, both nasal passages and splenocytes from mice nasally immunized with cationic liposomes plus ovalbumin (OVA) were polarized to produce IL-6 when re-stimulated with OVA in vitro. Furthermore, pretreatment with anti-IL-6R antibody, which blocks the biological activities of IL-6, attenuated the production of OVA-specific nasal immunoglobulin A (IgA) but not OVA-specific serum immunoglobulin G (IgG) responses. In this study, we demonstrated that IL-6, exerted by nasally administered cationic liposomes, plays a crucial role in antigen-specific IgA induction.


Assuntos
Imunidade nas Mucosas/imunologia , Imunoglobulina A/metabolismo , Interleucina-6/imunologia , Vacinas/imunologia , Administração Intranasal , Animais , Formação de Anticorpos/efeitos dos fármacos , Antígenos/imunologia , COVID-19/prevenção & controle , Cátions/imunologia , Cátions/uso terapêutico , Ácidos Graxos Monoinsaturados/imunologia , Ácidos Graxos Monoinsaturados/uso terapêutico , Feminino , Imunidade nas Mucosas/efeitos dos fármacos , Imunoglobulina G/sangue , Interleucina-6/antagonistas & inibidores , Interleucina-6/genética , Interleucina-6/metabolismo , Lipossomos/imunologia , Lipossomos/uso terapêutico , Camundongos , Mucosa Nasal/imunologia , Mucosa Nasal/metabolismo , Ovalbumina/imunologia , Compostos de Amônio Quaternário/imunologia , Compostos de Amônio Quaternário/uso terapêutico , Baço/metabolismo , Vacinas/administração & dosagem
8.
PLoS One ; 16(8): e0254628, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34339430

RESUMO

Most current clinical vaccines work primarily by inducing the production of neutralizing antibodies against pathogens. Vaccine adjuvants that efficiently induce T cell responses to protein antigens need to be developed. In this study, we developed a new combination adjuvant consisting of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), D35, and an aluminum salt. Among the various combinations tested, the DOTAP/D35/aluminum salt adjuvant induced strong T cell and antibody responses against the model protein antigen with a single immunization. Adjuvant component and model antigen interaction studies in vitro also revealed that the strong mutual interactions among protein antigens and other components were one of the important factors for this efficient immune induction by the novel combination adjuvant. In addition, in vivo imaging of the antigen distribution suggested that the DOTAP component in the combination adjuvant formulation elicited transient antigen accumulation at the draining lymph nodes, possibly by antigen uptake DC migration. These results indicate the potential of the new combination adjuvant as a promising vaccine adjuvant candidate to treat infectious diseases and cancers.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos/imunologia , Proteínas/imunologia , Linfócitos T/imunologia , Alumínio/farmacologia , Animais , Formação de Anticorpos/imunologia , Movimento Celular/imunologia , Ácidos Graxos Monoinsaturados/farmacologia , Humanos , Imunidade/imunologia , Lipossomos/imunologia , Linfonodos/imunologia , Camundongos , Compostos de Amônio Quaternário/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Vacinação/métodos , Vacinas/imunologia
9.
Int Immunopharmacol ; 99: 108068, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34426114

RESUMO

Oligomannose-coated liposomes (OMLs) comprised of dipalmitoylphosphatidylcholine, cholesterol and Man3-DPPE at a molar ratio of 1:1:0.1 and particle diameters of about 1000 nm can induce liposome-encased antigen-specific strong Th1 immunity. In this study, we evaluated the effect of particle sizes of OMLs on induction of Th1 immune responses in mice. Spleen cells obtained from mice immunized with antigen-encapsulating OMLs with 1000- and 800-nm diameters secreted remarkably high levels of IFN-γ upon in vitro stimulation. In addition, sera of mice that received these OMLs had significantly higher titers of antigen-specific IgG2a than those of IgG1, which are commonly associated with Th1 responses. In contrast, treatment with antigen-encapsulating OMLs with 400- and 200-nm diameters failed to induce IFN-γ secretion from spleen cells, although these OMLs did elicit elevation of antigen-specific IgGs. In addition, the titers of serum antigen-specific IgG2a were the same as those of IgG1 in mice that received 400-nm OMLs. Resident peritoneal mononuclear phagocytes (MNPs) treated with OMLs of diameter ≥ 600 nm secreted IL-12, which is essential for induction of Th1 immune responses, while those treated with OMLs of ≤ 400 nm failed to produce this cytokine. However, 400-nm OMLs did induce enhanced expression of MHC class II and costimulatory molecules on MNPs, similarly to OMLs of ≥ 600 nm. Taken together, these results strongly indicate that OMLs of diameter ≥ 600 nm are required to induce Th1 immune responses against OML-encased antigens, although OMLs of diameter ≤ 400 nm can activate MNPs.


Assuntos
Lipossomos/química , Lipossomos/imunologia , Manose/química , Manose/imunologia , Células Th1/imunologia , 1,2-Dipalmitoilfosfatidilcolina/imunologia , Animais , Antígenos/imunologia , Antígeno B7-2/metabolismo , Citocalasina D/farmacologia , Feminino , Antígenos de Histocompatibilidade Classe II/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe II/metabolismo , Sistema Imunitário , Imunoglobulina G/sangue , Interferon gama/efeitos dos fármacos , Interferon gama/metabolismo , Subunidade p35 da Interleucina-12/metabolismo , Camundongos , Tamanho da Partícula , Absorção Peritoneal/efeitos dos fármacos , Fagócitos/efeitos dos fármacos , Fagócitos/metabolismo , Fagocitose/efeitos dos fármacos , Baço/efeitos dos fármacos , Baço/metabolismo
10.
Nanomedicine ; 37: 102445, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34303841

RESUMO

Chikungunya virus (CHIKV) is responsible for a self-limited illness that can evolve into long-lasting painful joint inflammation. In this study, we report a novel experimental CHIKV vaccine formulation of lipid nanoparticles loaded with a recombinant protein derived from the E2 structural protein. This antigen fragment, designated ∆E2.1, maintained the antigenicity of the native viral protein and was specifically recognized by antibodies induced in CHIKV-infected patients. The antigen has been formulated into nanoparticles consisting of nano-multilamellar vesicles (NMVs) combined with the adjuvant monophosphoryl lipid A (MPLA). The vaccine formulation demonstrated a depot effect, leading to controlled antigen release, and induced strong antibody responses significantly higher than in mice immunized with the purified protein combined with the adjuvant. More relevantly, E2-specific antibodies raised in mice immunized with ∆E2.1-loaded NMV-MPLA neutralized CHIKV under in vitro conditions. Taken together, the results demonstrated that the new nanoparticle-based vaccine formulation represents a promising approach for the development of effective anti-CHIKV vaccines.


Assuntos
Febre de Chikungunya/imunologia , Vírus Chikungunya/imunologia , Lipossomos/imunologia , Proteínas do Envelope Viral/genética , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/efeitos dos fármacos , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/efeitos dos fármacos , Anticorpos Antivirais/imunologia , Febre de Chikungunya/terapia , Febre de Chikungunya/virologia , Vírus Chikungunya/patogenicidade , Humanos , Lipossomos/química , Lipossomos/farmacologia , Camundongos , Nanopartículas/química , Proteínas do Envelope Viral/farmacologia , Vacinas Virais/imunologia
11.
Front Immunol ; 12: 674048, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054859

RESUMO

Dendritic cells (DCs) are paramount in initiating and guiding immunity towards a state of activation or tolerance. This bidirectional capacity of DCs sets them at the center stage for treatment of cancer and autoimmune or allergic conditions. Accordingly, many clinical studies use ex vivo DC vaccination as a strategy to boost anti-tumor immunity or to suppress immunity by including vitamin D3, NF-κB inhibitors or retinoic acid to create tolerogenic DCs. As harvesting DCs from patients and differentiating these cells in vitro is a costly and cumbersome process, in vivo targeting of DCs has huge potential as nanoparticulate platforms equipped with activating or tolerogenic adjuvants can modulate DCs in their natural environment. There is a rapid expansion of the choices of nanoparticles and activation- or tolerance-promoting adjuvants for a therapeutic vaccine platform. In this review we highlight the most recent nanomedical approaches aimed at inducing immune activation or tolerance via targeting DCs, together with novel fundamental insights into the mechanisms inherent to fostering anti-tumor or tolerogenic immunity.


Assuntos
Células Dendríticas/imunologia , Tolerância Imunológica/imunologia , Lipossomos , Nanopartículas , Vacinas , Sistemas de Liberação de Medicamentos/métodos , Humanos , Lipossomos/química , Lipossomos/imunologia , Nanopartículas/química , Vacinas/química , Vacinas/imunologia
12.
J Virol ; 95(13): e0000521, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33883221

RESUMO

A major goal of HIV vaccine design is to elicit broadly neutralizing antibodies (bNAbs). Such bNAbs target HIV's trimeric, membrane-embedded envelope glycoprotein spikes (mEnv). Soluble Env (sEnv) trimers have been used as vaccines, but engineering sEnvs for stability, multivalency, and desired antigenicity is problematic and deletes key neutralizing epitopes on glycoprotein 41 (gp41) while creating neoepitopes that elicit unwanted antibodies. Meanwhile, multivalent mEnv vaccines are challenging to develop due to trimer instability and low mEnv copy number amid other extraneous proteins on virus-like particles. Here, we describe a multivalent mEnv vaccine platform that does not require protein engineering or extraneous proteins. mEnv trimers were fixed, purified, and combined with naked liposomes in mild detergent. On removal of detergent, mEnv spikes were observed embedded in liposome particles (mean diameter, 133 nm) in correct orientation. These particles were recognized by HIV bNAbs and not non-NAbs and are designated mEnv liposomes (MELs). Following a sequential immunization scheme in rabbits, MELs elicited antibodies that neutralized tier 2 HIV isolates. Analysis of serum antibody specificities, including those to epitopes involving a missing conserved N-glycosylation site at position 197 near the CD4 binding site on two of the immunogens, provides clues on how NAb responses can be improved with modified immunogens. In sum, MELs are a biochemically defined platform that enables rational immunization strategies to elicit HIV bNAbs using multimerized mEnv. IMPORTANCE A vaccine that induced broadly neutralizing antibodies against HIV would likely end the AIDS pandemic. Such antibodies target membrane-embedded envelope glycoprotein spikes (mEnv) that HIV uses to enter cells. Due to HIV Env's low expression and instability, soluble stabilized Env trimers have been used as vaccine candidates, but these have an altered base that disrupts targets of HIV broadly neutralizing antibodies that bind near the membrane and are not available for all HIV isolates. Here, we describe membrane Env liposomes (MELs) that display a multivalent array of stable mEnvs on liposome particles. MELs showed the expected antibody recognition properties, including targeting parts of mEnv missing on soluble Envs. Immunization with MELs elicited antibodies that neutralized diverse HIV isolates. The MEL platform facilitates vaccine development with potentially any HIV Env at high valency, and a similar approach may be useful for eliciting antibodies to membrane-embedded targets of therapeutic interest.


Assuntos
Vacinas contra a AIDS/imunologia , Anticorpos Amplamente Neutralizantes/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Proteína gp41 do Envelope de HIV/imunologia , HIV-1/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Epitopos/imunologia , Células HEK293 , Anticorpos Anti-HIV/imunologia , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Células HeLa , Humanos , Lipossomos/imunologia , Engenharia de Proteínas/métodos , Vacinação
13.
J Nanobiotechnology ; 19(1): 102, 2021 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-33849551

RESUMO

BACKGROUND: The immunomodulation of the antitumor response driven by immunocheckpoint inhibitors (ICIs) such as PD-L1 (Programmed Death Ligand-1) monoclonal antibody (α-PD-L1) have shown relevant clinical outcomes in a subset of patients. This fact has led to the search for rational combinations with other therapeutic agents such as Doxorubicin (Dox), which cytotoxicity involves an immune activation that may enhance ICI response. Therefore, this study aims to evaluate the combination of chemotherapy and ICI by developing Dox Immunoliposomes functionalized with monovalent-variable fragments (Fab') of α-PD-L1. RESULTS: Immunoliposomes were assayed in vitro and in vivo in a B16 OVA melanoma murine cell line over-expressing PD-L1. Here, immune system activation in tumor, spleen and lymph nodes, together with the antitumor efficacy were evaluated. Results showed that immunoliposomes bound specifically to PD-L1+ cells, yielding higher cell interaction and Dox internalization, and decreasing up to 30-fold the IC50, compared to conventional liposomes. This mechanism supported a higher in vivo response. Indeed, immunoliposomes promoted full tumor regression in 20% of mice and increased in 1 month the survival rate. This formulation was the only treatment able to induce significant (p < 0.01) increase of activated tumor specific cytotoxic T lymphocytes at the tumor site. CONCLUSION: PD-L1 targeted liposomes encapsulating Dox have proved to be a rational combination able to enhance the modulation of the immune system by blocking PD-L1 and selectively internalizing Dox, thus successfully providing a dual activity offered by both, chemo and immune therapeutic strategies.


Assuntos
Antineoplásicos/farmacologia , Antígeno B7-H1/metabolismo , Doxorrubicina/farmacologia , Imunidade/efeitos dos fármacos , Lipossomos/imunologia , Melanoma/tratamento farmacológico , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Liberação Controlada de Fármacos , Tratamento Farmacológico , Feminino , Imunoterapia/métodos , Melanoma Experimental/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL
14.
Pharm Res ; 38(3): 429-450, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33655395

RESUMO

PURPOSE: To develop immunoliposomes modified with monoclonal cancer-specific antibody (mAb) 2C5 and co-loaded with a combination of two chemotherapeutics, in order to simultaneously target bulk cancer cells using paclitaxel and cancer stem cells (CSCs) using salinomycin to prevent cancer growth and metastases. METHODS: Breast cancer cells (MDA-MB-231 and/or SK-BR-3) were chosen as models for all in vitro testing. Liposomes composed of natural phospholipids co-loaded with salinomycin and paclitaxel were prepared and physically characterized. Immunoliposomes modified with mAb 2C5 coupled to polymeric conjugate were prepared and characterized for specific targeting. Wound healing assay was performed using the combination of free drugs in vitro. In vitro studies on cellular interaction and uptake were followed by holographic imaging to study cell-killing, cell-division and proliferation inhibiting effects of the formulation. Ex-vivo study on hemolysis was investigated to check possible toxicity of the formulation. RESULTS: Physical characterization of the liposomes showed stable nanoparticles of consistent and desirable size range (170-220 nm), zeta potential (-13 mV to - 20 mV), polydispersity indices (<0.2) and drug encapsulation efficiencies (~150 µg per ml for salinomycin, ~210 µg/ml for paclitaxel and 1:1 for combination drug loaded liposomes). Combination therapy strongly affected cancer cell proliferation as shown by significant diminishing of artificial gap closure at the wound site on MDA-MB-231 cells in culture using wound healing assay. Quantitation of changes in wound widths showed ~219 µm for drug combination, ~104 µm for only paclitaxel, and ~ 7 µm for only salinomycin treatments. Statistically significant increase in cellular interaction and specific uptake of the targeted drug co-loaded liposomal nanopreparation (p value ≤ 0.05) by MDA-MB-231 and SK-BR-3 cells confirmed the effectiveness of the approach. Holographic imaging using MDA-MB-231 cells produced visible increase in cell-killing, proliferation and division in vitro. Ex-vivo experimentation showed reduced hemolysis correlating with low toxicity in athymic nude mice model. CONCLUSION: The results demonstrated the enhanced therapeutic efficacy of a combination of salinomycin and paclitaxel delivered by mAb 2C5-modified liposomal preparation in cancer therapy.


Assuntos
Anticorpos Monoclonais/química , Protocolos de Quimioterapia Combinada Antineoplásica/química , Portadores de Fármacos/química , Lipossomos/química , Paclitaxel/química , Piranos/química , Animais , Anticorpos Monoclonais/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Proliferação de Células/efeitos dos fármacos , Preparações de Ação Retardada/química , Composição de Medicamentos , Liberação Controlada de Fármacos , Feminino , Humanos , Lipossomos/imunologia , Camundongos Nus , Neoplasias Experimentais , Paclitaxel/farmacologia , Fosfatidilcolinas/química , Polietilenoglicóis/química , Piranos/farmacologia
16.
Int J Mol Sci ; 22(2)2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33467541

RESUMO

This study aims to provide guidelines to design and perform a robust and reliable physical-chemical characterization of liposome-based nanomaterials, and to support method development with a specific focus on their inflammation-inducing potential. Out of eight differently functionalized liposomes selected as "case-studies", three passed the physical-chemical characterization ( in terms of size-distribution, homogeneity and stability) and the screening for bacterial contamination (sterility and apyrogenicity). Although all three were non-cytotoxic when tested in vitro, they showed a different capacity to activate human blood cells. HSPC/CHOL-coated liposomes elicited the production of several inflammation-related cytokines, while DPPC/CHOL- or DSPC/CHOL-functionalized liposomes did not. This work underlines the need for accurate characterization at multiple levels and the use of reliable in vitro methods, in order to obtain a realistic assessment of liposome-induced human inflammatory response, as a fundamental requirement of nanosafety regulations.


Assuntos
Citocinas/imunologia , Imunidade Inata/imunologia , Mediadores da Inflamação/imunologia , Lipossomos/imunologia , Nanoestruturas/química , Pesquisa Translacional Biomédica/métodos , 1,2-Dipalmitoilfosfatidilcolina/química , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Colesterol/química , Citocinas/metabolismo , Células Hep G2 , Humanos , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Lipossomos/química , Lipossomos/farmacologia , Tamanho da Partícula , Fosfatidilcolinas/química
17.
Sci China Life Sci ; 64(7): 1097-1115, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33009993

RESUMO

As a potential cancer immunotherapeutic agent, chlorogenic acid (CHA) has entered phase II clinical trials in China as a lyophilized powder formulation for treating glioma. However, the in vivo instability of CHA necessitates daily intramuscular injections, resulting in patient noncompliance. In this study, CHA-phospholipid complex (PC)-containing PEGylated liposomes (CHA-PC PEG-Lipo, named as CPPL), with CHA-PC as the drug intermediate, were prepared to lower the administration frequency. CPPL demonstrated excellent physicochemical properties, enhanced tumor accumulation, and inhibited tumor growth even when the administration interval was prolonged to 4 days when compared to a CHA solution and CHA-PC loaded liposomes (CHA-PC Lipo, labeled as CPL), both of which only demonstrated antitumor efficacy with once-daily administration. Further evaluation of the in vivo antitumor immune mechanism suggested that the extended antitumor immune efficacy of CPPL could be attributed to its distinct immune-stimulating mechanism when compared with CHA solution and CPL, such as stimulating both CD4+ and CD8+ T cell infiltration, inhibiting myeloid-derived suppressor cell expression, reducing the expression of Th2 related factors, and notably, increasing the memory T cells in tumor tissues. This CHA-containing formulation could reduce the frequency of in vivo CHA administration during cancer treatment via T cells, especially memory T cell regulation.


Assuntos
Ácido Clorogênico/farmacologia , Glioma/tratamento farmacológico , Imunoterapia/métodos , Lipossomos/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Ácido Clorogênico/imunologia , Modelos Animais de Doenças , Glioma/imunologia , Lipossomos/imunologia , Ratos , Linfócitos T Reguladores/imunologia
18.
Mucosal Immunol ; 14(2): 523-536, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32807838

RESUMO

This is a proof-of-principle study demonstrating that the combination of a cholera toxin derived adjuvant, CTA1-DD, and lipid nanoparticles (LNP) can significantly improve the immunogenicity and protective capacity of an intranasal vaccine. We explored the self-adjuvanted universal influenza vaccine candidate, CTA1-3M2e-DD (FPM2e), linked to LNPs. We found that the combined vector greatly enhanced survival against a highly virulent PR8 strain of influenza virus as compared to when mice were immunized with FPM2e alone. The combined vaccine vector enhanced early endosomal processing and peptide presentation in dendritic cells and upregulated co-stimulation. The augmenting effect was CTA1-enzyme dependent. Whereas systemic anti-M2e antibody and CD4+ T-cell responses were comparable to those of the soluble protein, the local respiratory tract IgA and the specific Th1 and Th17 responses were strongly enhanced. Surprisingly, the lung tissue did not exhibit gross pathology upon recovery from infection and M2e-specific lung resident CD4+ T cells were threefold higher than in FPM2e-immunized mice. This study conveys optimism as to the protective ability of a combination vaccine based on LNPs and various forms of the CTA1-DD adjuvant platform, in general, and, more specifically, an important way forward to develop a universal vaccine against influenza.


Assuntos
Toxina da Cólera/imunologia , Vírus da Influenza A/fisiologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Lipossomos/imunologia , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Proteínas Recombinantes de Fusão/imunologia , Células Th1/imunologia , Células Th17/imunologia , Administração Intranasal , Animais , Apresentação de Antígeno , Células Cultivadas , Toxina da Cólera/metabolismo , Humanos , Imunogenicidade da Vacina , Imunoglobulina A/metabolismo , Vacinas contra Influenza/metabolismo , Lipossomos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas/metabolismo , Peptídeos Cíclicos , Proteínas Recombinantes de Fusão/metabolismo , Vacinação
19.
Nanomedicine ; 35: 102338, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33197626

RESUMO

DNA vaccine is an attractive immune platform for the prevention and treatment of infectious diseases, but existing disadvantages limit its use in preclinical and clinical assays, such as weak immunogenicity and short half-life. Here, we reported a novel liposome-polymer hybrid nanoparticles (pSFV-MEG/LNPs) consisting of a biodegradable core (mPEG-PLGA) and a hydrophilic shell (lecithin/PEG-DSPE-Mal 2000) for delivering a multi-epitope self-replication DNA vaccine (pSFV-MEG). The pSFV-MEG/LNPs with optimal particle size (161.61 ±â€¯15.63 nm) and high encapsulation efficiency (87.60 ±â€¯8.73%) induced a strong humoral (3.22-fold) and cellular immune responses (1.60-fold) compared to PBS. Besides, the humoral and cellular immune responses of pSFV-MEG/LNPs were 1.58- and 1.05-fold than that of pSFV-MEG. All results confirmed that LNPs was a very promising tool to enhance the humoral and cellular immune responses of pSFV-MEG. In addition, the rational design and delivery platform can be used for the development of DNA vaccines for other infectious diseases.


Assuntos
Replicação do DNA , Epitopos , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Nanopartículas/uso terapêutico , Vacinas de DNA , Animais , Epitopos/genética , Epitopos/imunologia , Lipossomos/imunologia , Lipossomos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/farmacologia
20.
Genes Dev ; 34(23-24): 1697-1712, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33184222

RESUMO

Deciphering the mechanisms that regulate the sensitivity of pathogen recognition receptors is imperative to understanding infection and inflammation. Here we demonstrate that the RNA triphosphatase dual-specificity phosphatase 11 (DUSP11) acts on both host and virus-derived 5'-triphosphate RNAs rendering them less active in inducing a RIG-I-mediated immune response. Reducing DUSP11 levels alters host triphosphate RNA packaged in extracellular vesicles and induces enhanced RIG-I activation in cells exposed to extracellular vesicles. Virus infection of cells lacking DUSP11 results in a higher proportion of triphosphorylated viral transcripts and attenuated virus replication, which is rescued by reducing RIG-I expression. Consistent with the activity of DUSP11 in the cellular RIG-I response, mice lacking DUSP11 display lower viral loads, greater sensitivity to triphosphorylated RNA, and a signature of enhanced interferon activity in select tissues. Our results reveal the importance of controlling 5'-triphosphate RNA levels to prevent aberrant RIG-I signaling and demonstrate DUSP11 as a key effector of this mechanism.


Assuntos
Proteína DEAD-box 58/imunologia , Fosfatases de Especificidade Dupla/imunologia , Fosfatases de Especificidade Dupla/metabolismo , RNA/imunologia , Viroses/imunologia , Animais , Linhagem Celular , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Interferons/metabolismo , Lipossomos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Polifosfatos , Vírus de RNA/fisiologia , RNA Viral/metabolismo , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...