Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 187(12): 2919-2934.e20, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38761800

RESUMO

A critical roadblock to HIV vaccine development is the inability to induce B cell lineages of broadly neutralizing antibodies (bnAbs) in humans. In people living with HIV-1, bnAbs take years to develop. The HVTN 133 clinical trial studied a peptide/liposome immunogen targeting B cell lineages of HIV-1 envelope (Env) membrane-proximal external region (MPER) bnAbs (NCT03934541). Here, we report MPER peptide-liposome induction of polyclonal HIV-1 B cell lineages of mature bnAbs and their precursors, the most potent of which neutralized 15% of global tier 2 HIV-1 strains and 35% of clade B strains with lineage initiation after the second immunization. Neutralization was enhanced by vaccine selection of improbable mutations that increased antibody binding to gp41 and lipids. This study demonstrates proof of concept for rapid vaccine induction of human B cell lineages with heterologous neutralizing activity and selection of antibody improbable mutations and outlines a path for successful HIV-1 vaccine development.


Assuntos
Vacinas contra a AIDS , Anticorpos Neutralizantes , Linfócitos B , Anticorpos Anti-HIV , HIV-1 , Humanos , Vacinas contra a AIDS/imunologia , HIV-1/imunologia , Anticorpos Neutralizantes/imunologia , Linfócitos B/imunologia , Anticorpos Anti-HIV/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Linhagem da Célula , Lipossomos , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Mutação , Proteína gp41 do Envelope de HIV/imunologia
2.
Nature ; 594(7864): 553-559, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33971664

RESUMO

Betacoronaviruses caused the outbreaks of severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome, as well as the current pandemic of SARS coronavirus 2 (SARS-CoV-2)1-4. Vaccines that elicit protective immunity against SARS-CoV-2 and betacoronaviruses that circulate in animals have the potential to prevent future pandemics. Here we show that the immunization of macaques with nanoparticles conjugated with the receptor-binding domain of SARS-CoV-2, and adjuvanted with 3M-052 and alum, elicits cross-neutralizing antibody responses against bat coronaviruses, SARS-CoV and SARS-CoV-2 (including the B.1.1.7, P.1 and B.1.351 variants). Vaccination of macaques with these nanoparticles resulted in a 50% inhibitory reciprocal serum dilution (ID50) neutralization titre of 47,216 (geometric mean) for SARS-CoV-2, as well as in protection against SARS-CoV-2 in the upper and lower respiratory tracts. Nucleoside-modified mRNAs that encode a stabilized transmembrane spike or monomeric receptor-binding domain also induced cross-neutralizing antibody responses against SARS-CoV and bat coronaviruses, albeit at lower titres than achieved with the nanoparticles. These results demonstrate that current mRNA-based vaccines may provide some protection from future outbreaks of zoonotic betacoronaviruses, and provide a multimeric protein platform for the further development of vaccines against multiple (or all) betacoronaviruses.


Assuntos
Anticorpos Neutralizantes/imunologia , Betacoronavirus/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , Resfriado Comum/prevenção & controle , Reações Cruzadas/imunologia , Pandemias , Vacinas Virais/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Animais , COVID-19/epidemiologia , Vacinas contra COVID-19/imunologia , Resfriado Comum/imunologia , Resfriado Comum/virologia , Modelos Animais de Doenças , Feminino , Humanos , Macaca/imunologia , Masculino , Modelos Moleculares , Nanopartículas/química , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/imunologia , Traqueia , Vacinação
3.
Semin Immunol ; 39: 22-29, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30366662

RESUMO

Lipopolysaccharide (LPS) is a well-defined agonist of Toll-like receptor (TLR) 4 that activates innate immune responses and influences the development of the adaptive response during infection with Gram-negative bacteria. Many years ago, Dr. Edgar Ribi separated the adjuvant activity of LPS from its toxic effects, an effort that led to the development of monophosphoryl lipid A (MPL). MPL, derived from Salmonella minnesota R595, has progressed through clinical development and is now used in various product-enabling formulations to support the generation of antigen-specific responses in several commercial and preclinical vaccines. We have generated several synthetic lipid A molecules, foremost glucopyranosyl lipid adjuvant (GLA) and second-generation lipid adjuvant (SLA), and have advanced these to clinical trial for various indications. In this review we summarize the potential and current positioning of TLR4-based adjuvant formulations in approved and emerging vaccines.


Assuntos
Adjuvantes Imunológicos/farmacologia , Compostos de Alúmen/farmacologia , Glucosídeos/farmacologia , Imunogenicidade da Vacina , Lipídeo A/análogos & derivados , Tuberculose/prevenção & controle , Adjuvantes Imunológicos/química , Compostos de Alúmen/química , Animais , Glucosídeos/química , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Leishmaniose/imunologia , Leishmaniose/parasitologia , Leishmaniose/prevenção & controle , Hanseníase/imunologia , Hanseníase/parasitologia , Hanseníase/prevenção & controle , Lipídeo A/química , Lipídeo A/farmacologia , Lipossomos/administração & dosagem , Lipossomos/química , Lipossomos/imunologia , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Camundongos , Esquistossomose/imunologia , Esquistossomose/parasitologia , Esquistossomose/prevenção & controle , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/microbiologia , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Tuberculose/imunologia , Tuberculose/microbiologia , Vacinas/administração & dosagem , Vacinas/química , Vacinas/imunologia
4.
J Immunol ; 201(1): 98-112, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29769270

RESUMO

The involvement of innate receptors that recognize pathogen- and danger-associated molecular patterns is critical to programming an effective adaptive immune response to vaccination. The synthetic TLR4 agonist glucopyranosyl lipid adjuvant (GLA) synergizes with the squalene oil-in-water emulsion (SE) formulation to induce strong adaptive responses. Although TLR4 signaling through MyD88 and TIR domain-containing adapter inducing IFN-ß are essential for GLA-SE activity, the mechanisms underlying the synergistic activity of GLA and SE are not fully understood. In this article, we demonstrate that the inflammasome activation and the subsequent release of IL-1ß are central effectors of the action of GLA-SE, as infiltration of innate cells into the draining lymph nodes and production of IFN-γ are reduced in ASC-/- animals. Importantly, the early proliferation of Ag-specific CD4+ T cells was completely ablated after immunization in ASC-/- animals. Moreover, numbers of Ag-specific CD4+ T and B cells as well as production of IFN-γ, TNF-α, and IL-2 and Ab titers were considerably reduced in ASC-/-, NLRP3-/-, and IL-1R-/- mice compared with wild-type mice and were completely ablated in TLR4-/- animals. Also, extracellular ATP, a known trigger of the inflammasome, augments Ag-specific CD4+ T cell responses, as hydrolyzing it with apyrase diminished adaptive responses induced by GLA-SE. These data thus demonstrate that GLA-SE adjuvanticity acts through TLR4 signaling and NLRP3 inflammasome activation to promote robust Th1 and B cell responses to vaccine Ags. The findings suggest that engagement of both TLR and inflammasome activators may be a general paradigm for induction of robust CD4 T cell immunity with combination adjuvants such as GLA-SE.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos/imunologia , Linfócitos B/imunologia , Inflamassomos/imunologia , Células Th1/imunologia , Receptor 4 Toll-Like/imunologia , Vacinas/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Feminino , Glucosídeos/imunologia , Imunidade Humoral , Interferon beta/imunologia , Interferon gama/imunologia , Interleucina-1beta/metabolismo , Interleucina-2/imunologia , Lipídeo A/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Receptores Tipo I de Interleucina-1/genética , Esqualeno/imunologia , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/imunologia , Vacinação
5.
J Virol ; 92(20)2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30089691

RESUMO

Toward the goal of developing an effective HIV vaccine that can be administered in infancy to protect against postnatal and lifelong sexual HIV transmission risks, the current pilot study was designed to compare the effect of novel adjuvants on the induction of HIV Env-specific antibody responses in infant macaques. Aligning our studies with the adjuvanted proteins evaluated in a prime-boost schedule with ALVAC in the ongoing HVTN (HIV Vaccine Trials Network) 702 efficacy trial, we selected the bivalent clade C Env immunogens gp120 C.1086 and gp120 TV1 in combination with the MF59 adjuvant. However, we hypothesized that the adjuvant system AS01, that is included in the pediatric RTS,S malaria vaccine, would promote Env-specific antibody responses superior to those of the oil-in-water MF59 emulsion adjuvant. In a second study arm, we compared two emulsions, glucopyranosyl lipid adjuvant formulated in a stable emulsion (GLA-SE) and 3M-052-SE, containing Toll-like receptor 4 (TLR4) and TLR7/TLR8 (TLR7/8) ligand, respectively. The latter adjuvant had been previously demonstrated to be especially effective in activating neonatal antigen-presenting cells. Our results demonstrate that different adjuvants drive quantitatively or qualitatively distinct responses to the bivalent Env vaccine. AS01 induced higher Env-specific plasma IgG antibody levels than the antigen in MF59 and promoted improved antibody function in infants, and 3M-052-SE outperformed GLA-SE by inducing the highest breadth and functionality of antibody responses. Thus, distinct adjuvants are likely to be required for maximizing vaccine-elicited immune responses in infants, particularly when immunization in infancy aims to elicit both perinatal and lifelong immunity against challenging pathogens such as HIV.IMPORTANCE Alum remains the adjuvant of choice for pediatric vaccines. Yet the distinct nature of the developing immune system in infants likely requires novel adjuvants targeted specifically at the pediatric population to reach maximal vaccine efficacy with an acceptable safety profile. The current study supports the idea that additional adjuvants for pediatric vaccines should be, and need to be, tested in infants for their potential to enhance immune responses. Using an infant macaque model, our results suggest that both AS01 and 3M-052-SE can significantly improve and better sustain HIV Env-specific antibody responses than alum. Despite the limited number of animals, the results revealed interesting differences that warrant further testing of promising novel adjuvant candidates in larger preclinical and clinical studies to define the mechanisms leading to adjuvant-improved antibody responses and to identify targets for adjuvant and vaccine optimization.


Assuntos
Vacinas contra a AIDS/imunologia , Adjuvantes Imunológicos/administração & dosagem , Formação de Anticorpos , Anticorpos Anti-HIV/sangue , Proteína gp120 do Envelope de HIV/imunologia , Vacinas contra a AIDS/administração & dosagem , Animais , Animais Recém-Nascidos , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Proteína gp120 do Envelope de HIV/administração & dosagem , Imunoglobulina G/sangue , Macaca mulatta
6.
J Virol ; 92(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29793957

RESUMO

We developed a method of simultaneous vaccination with DNA and protein resulting in robust and durable cellular and humoral immune responses with efficient dissemination to mucosal sites and protection against simian immunodeficiency virus (SIV) infection. To further optimize the DNA-protein coimmunization regimen, we tested a SIVmac251-based vaccine formulated with either of two Toll-like receptor 4 (TLR4) ligand-based liposomal adjuvant formulations (TLR4 plus TLR7 [TLR4+7] or TLR4 plus QS21 [TLR4+QS21]) in macaques. Although both vaccines induced humoral responses of similar magnitudes, they differed in their functional quality, including broader neutralizing activity and effector functions in the TLR4+7 group. Upon repeated heterologous SIVsmE660 challenge, a trend of delayed viral acquisition was found in vaccinees compared to controls, which reached statistical significance in animals with the TRIM-5α-resistant (TRIM-5α R) allele. Vaccinees were preferentially infected by an SIVsmE660 transmitted/founder virus carrying neutralization-resistant A/K mutations at residues 45 and 47 in Env, demonstrating a strong vaccine-induced sieve effect. In addition, the delay in virus acquisition directly correlated with SIVsmE660-specific neutralizing antibodies. The presence of mucosal V1V2 IgG binding antibodies correlated with a significantly decreased risk of virus acquisition in both TRIM-5α R and TRIM-5α-moderate/sensitive (TRIM-5α M/S) animals, although this vaccine effect was more prominent in animals with the TRIM-5α R allele. These data support the combined contribution of immune responses and genetic background to vaccine efficacy. Humoral responses targeting V2 and SIV-specific T cell responses correlated with viremia control. In conclusion, the combination of DNA and gp120 Env protein vaccine regimens using two different adjuvants induced durable and potent cellular and humoral responses contributing to a lower risk of infection by heterologous SIV challenge.IMPORTANCE An effective AIDS vaccine continues to be of paramount importance for the control of the pandemic, and it has been proven to be an elusive target. Vaccine efficacy trials and macaque challenge studies indicate that protection may be the result of combinations of many parameters. We show that a combination of DNA and protein vaccinations applied at the same time provides rapid and robust cellular and humoral immune responses and evidence for a reduced risk of infection. Vaccine-induced neutralizing antibodies and Env V2-specific antibodies at mucosal sites contribute to the delay of SIVsmE660 acquisition, and genetic makeup (TRIM-5α) affects the effectiveness of the vaccine. These data are important for the design of better vaccines and may also affect other vaccine platforms.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Produtos do Gene env , Imunidade Humoral , Vacinas contra a SAIDS , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Vacinas de DNA , Adjuvantes Imunológicos/farmacologia , Substituição de Aminoácidos , Animais , Produtos do Gene env/genética , Produtos do Gene env/imunologia , Produtos do Gene env/farmacologia , Imunização , Macaca , Mutação de Sentido Incorreto , Vacinas contra a SAIDS/genética , Vacinas contra a SAIDS/imunologia , Vacinas contra a SAIDS/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/imunologia , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/farmacologia
7.
Mol Ther ; 26(10): 2507-2522, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30078765

RESUMO

Since the first demonstration of in vivo gene expression from an injected RNA molecule almost two decades ago,1 the field of RNA-based therapeutics is now taking significant strides, with many cancer and infectious disease targets entering clinical trials.2 Critical to this success has been advances in the knowledge and application of delivery formulations. Currently, various lipid nanoparticle (LNP) platforms are at the forefront,3 but the encapsulation approach underpinning LNP formulations offsets the synthetic and rapid-response nature of RNA vaccines.4 Second, limited stability of LNP formulated RNA precludes stockpiling for pandemic readiness.5 Here, we show the development of a two-vialed approach wherein the delivery formulation, a highly stable nanostructured lipid carrier (NLC), can be manufactured and stockpiled separate from the target RNA, which is admixed prior to administration. Furthermore, specific physicochemical modifications to the NLC modulate immune responses, either enhancing or diminishing neutralizing antibody responses. We have combined this approach with a replicating viral RNA (rvRNA) encoding Zika virus (ZIKV) antigens and demonstrated a single dose as low as 10 ng can completely protect mice against a lethal ZIKV challenge, representing what might be the most potent approach to date of any Zika vaccine.


Assuntos
Antígenos Virais/administração & dosagem , Lipídeos/administração & dosagem , Nanopartículas/administração & dosagem , Infecção por Zika virus/terapia , Animais , Antígenos Virais/genética , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Humanos , Lipídeos/química , Camundongos , Nanopartículas/química , RNA Viral/genética , RNA Viral/imunologia , Replicação Viral/efeitos dos fármacos , Zika virus/genética , Zika virus/patogenicidade , Infecção por Zika virus/genética , Infecção por Zika virus/virologia
8.
J Immunol ; 197(11): 4351-4359, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27794001

RESUMO

Designing modern vaccine adjuvants depends on understanding the cellular and molecular events that connect innate and adaptive immune responses. The synthetic TLR4 agonist glycopyranosyl lipid adjuvant (GLA) formulated in a squalene-in-water emulsion (GLA-SE) augments both cellular and humoral immune responses to vaccine Ags. This adjuvant is currently included in several vaccines undergoing clinical evaluation including those for tuberculosis, leishmaniasis, and influenza. Delineation of the mechanisms of adjuvant activity will enable more informative evaluation of clinical trials. Early after injection, GLA-SE induces substantially more Ag-specific B cells, higher serum Ab titers, and greater numbers of T follicular helper (TFH) and Th1 cells than alum, the SE alone, or GLA without SE. GLA-SE augments Ag-specific B cell differentiation into germinal center and memory precursor B cells as well as preplasmablasts that rapidly secrete Abs. CD169+ SIGNR1+ subcapsular medullary macrophages are the primary cells to take up GLA-SE after immunization and are critical for the innate immune responses, including rapid IL-18 production, induced by GLA-SE. Depletion of subcapsular macrophages (SCMф) or abrogation of IL-18 signaling dramatically impairs the Ag-specific B cell and Ab responses augmented by GLA-SE. Depletion of SCMф also drastically reduces the Th1 but not the TFH response. Thus the GLA-SE adjuvant operates through interaction with IL-18-producing SCMф for the rapid induction of B cell expansion and differentiation, Ab secretion, and Th1 responses, whereas augmentation of TFH numbers by GLA-SE is independent of SCMф.


Assuntos
Adjuvantes Imunológicos/farmacologia , Linfócitos B/imunologia , Diferenciação Celular/efeitos dos fármacos , Glucosídeos/farmacologia , Interleucina-18/imunologia , Lipídeo A/farmacologia , Linfonodos/imunologia , Macrófagos/imunologia , Receptor 4 Toll-Like/agonistas , Animais , Linfócitos B/citologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Diferenciação Celular/imunologia , Feminino , Glucosídeos/farmacocinética , Interleucina-18/genética , Subunidade alfa de Receptor de Interleucina-18/genética , Subunidade alfa de Receptor de Interleucina-18/imunologia , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Lipídeo A/farmacocinética , Linfonodos/citologia , Macrófagos/citologia , Camundongos , Camundongos Knockout , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Th1/citologia , Células Th1/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
9.
J Immunol ; 197(12): 4663-4673, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27849170

RESUMO

The HIV-1 envelope protein (Env) has evolved to subvert the host immune system, hindering viral control by the host. The tryptophan metabolic enzyme kynureninase (KYNU) is mimicked by a portion of the HIV Env gp41 membrane proximal region (MPER) and is cross-reactive with the HIV broadly neutralizing Ab (bnAb) 2F5. Molecular mimicry of host proteins by pathogens can lead to autoimmune disease. In this article, we demonstrate that neither the 2F5 bnAb nor HIV MPER-KYNU cross-reactive Abs elicited by immunization with an MPER peptide-liposome vaccine in 2F5 bnAb VHDJH and VLJL knock-in mice and rhesus macaques modified KYNU activity or disrupted tissue tryptophan metabolism. Thus, molecular mimicry by HIV-1 Env that promotes the evasion of host anti-HIV-1 Ab responses can be directed toward nonfunctional host protein epitopes that do not impair host protein function. Therefore, the 2F5 HIV Env gp41 region is a key and safe target for HIV-1 vaccine development.


Assuntos
Vacinas contra a AIDS/imunologia , Proteína gp41 do Envelope de HIV/metabolismo , Infecções por HIV/imunologia , HIV-1/imunologia , Hidrolases/metabolismo , Peptídeos/metabolismo , Triptofano/metabolismo , Animais , Anticorpos Neutralizantes/metabolismo , Reações Cruzadas , Anticorpos Anti-HIV/metabolismo , Proteína gp41 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/imunologia , Interações Hospedeiro-Patógeno , Humanos , Hidrolases/genética , Hidrolases/imunologia , Evasão da Resposta Imune , Macaca mulatta , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mimetismo Molecular , Peptídeos/genética , Peptídeos/imunologia , Vacinação , Vacinas de Subunidades Antigênicas
10.
Infect Immun ; 85(2)2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27895131

RESUMO

Recent studies have shown that immune responses against the cell-traversal protein for Plasmodium ookinetes and sporozoites (CelTOS) can inhibit parasite infection. While these studies provide important evidence toward the development of vaccines targeting this protein, it remains unknown whether these responses could engage the Plasmodium falciparum CelTOS in vivo Using a newly developed rodent malaria chimeric parasite expressing the P. falciparum CelTOS (PfCelTOS), we evaluated the protective effect of in vivo immune responses elicited by vaccination and assessed the neutralizing capacity of monoclonal antibodies specific against PfCelTOS. Mice immunized with recombinant P. falciparum CelTOS in combination with the glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE) or glucopyranosyl lipid adjuvant-liposome-QS21 (GLA-LSQ) adjuvant system significantly inhibited sporozoite hepatocyte infection. Notably, monoclonal antibodies against PfCelTOS strongly inhibited oocyst development of P. falciparum and Plasmodium berghei expressing PfCelTOS in Anopheles gambiae mosquitoes. Taken together, our results demonstrate that anti-CelTOS responses elicited by vaccination or passive immunization can inhibit sporozoite and ookinete infection and impair vector transmission.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Modelos Animais de Doenças , Hepatócitos/efeitos dos fármacos , Hepatócitos/parasitologia , Imunização , Imunização Passiva , Estágios do Ciclo de Vida , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Camundongos , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/genética , Proteínas Recombinantes
11.
AAPS PharmSciTech ; 18(6): 2077-2084, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28000085

RESUMO

Although substantial effort has been made in the development of next-generation recombinant vaccine systems, maintenance of a cold chain is still typically required and remains a critical challenge in effective vaccine distribution. The ability to engineer alternative containment systems that improve distribution and administration represents potentially significant enhancements to vaccination strategies. In this work, we evaluate the ability to successfully lyophilize a previously demonstrated thermostable tuberculosis vaccine formulation (ID93 + GLA-SE) in a cartridge format compared to a traditional vial container format. Due to differences in the shape of the container formats, a novel apparatus was developed to facilitate lyophilization in a cartridge. Following lyophilization, the lyophilizate was assessed visually, by determining residual moisture content, and by collecting melting profiles. Reconstituted formulations were assayed for particle size, protein presence, and GLA content. Based on assessment of the lyophilizate, the multicomponent vaccine was successfully lyophilized in both formats. Also, the physicochemical properties of the major components in the formulation, including antigen and adjuvant, were retained after lyophilization in either format. Ultimately, this study demonstrates that complex formulations can be lyophilized in alternative container formats to the standard pharmaceutical glass vial, potentially helping to increase the distribution of vaccines.


Assuntos
Adjuvantes Imunológicos/síntese química , Química Farmacêutica/instrumentação , Mycobacterium tuberculosis , Vacinas contra a Tuberculose/síntese química , Química Farmacêutica/métodos , Liofilização/métodos , Preparações Farmacêuticas
12.
Eur J Immunol ; 45(2): 407-17, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25367751

RESUMO

The synthetic TLR4 agonist glucopyranosyl lipid adjuvant (GLA) is a potent Th1-response-inducing adjuvant when formulated in a squalene oil-in-water emulsion (SE). While the innate signals triggered by TLR4 engagement are well studied, the contribution of SE remains unclear. To better understand the effect of SE on the adjuvant properties of GLA-SE, we compared the innate and adaptive immune responses elicited by immunization with different formulations: GLA without oil, SE alone or the combination, GLA-SE, in mice. Within the innate response to adjuvants, only GLA-SE displayed features of inflammasome activation, evidenced by early IL-18 secretion and IFN-γ production in memory CD8(+) T cells and neutrophils. Such early IFN-γ production was ablated in caspase-1/11(-/-) mice and in IL-18R1(-/-) mice. Furthermore, caspase-1/11 and IL-18 were also required for full Th1 CD4(+) T-cell induction via GLA-SE. Thus, we demonstrate that IL-18 and caspase-1/11 are components of the response to immunization with the TLR4 agonist/squalene oil-in-water based adjuvant, GLA-SE, providing implications for other adjuvants that combine oils with TLR agonists.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Caspase 1/imunologia , Caspases/imunologia , Interferon gama/imunologia , Interleucina-18/imunologia , Esqualeno/administração & dosagem , Receptor 4 Toll-Like/agonistas , Imunidade Adaptativa/efeitos dos fármacos , Adjuvantes Imunológicos/síntese química , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Caspase 1/genética , Caspases/genética , Caspases Iniciadoras , Emulsões , Feminino , Expressão Gênica , Imunidade Inata/efeitos dos fármacos , Imunização , Memória Imunológica , Inflamassomos/efeitos dos fármacos , Interferon gama/biossíntese , Interleucina-18/biossíntese , Lipídeos/administração & dosagem , Lipídeos/síntese química , Lipídeos/imunologia , Camundongos , Camundongos Knockout , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Receptores de Interleucina-18/genética , Receptores de Interleucina-18/imunologia , Esqualeno/química , Esqualeno/imunologia , Células Th1/citologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
13.
Biomacromolecules ; 17(1): 165-72, 2016 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-26652915

RESUMO

The potential to replace shark-derived squalene in vaccine adjuvant applications with synthetic squalene/poly(isoprene) oligomers, synthesized by the controlled oligomerization of isoprene is demonstrated. Following on from our previous work regarding the synthesis of poly(isoprene) oligomers, we demonstrate the ability to tune the molecular weight of the synthetic poly(isoprene) material beyond that of natural squalene, while retaining a final backbone structure that contained a minimum of 75% of 1,4 addition product and an acceptable polydispersity. The synthesis was successfully scaled from the 2 g to the 40 g scale both in the bulk (i.e., solvent free) and with the aid of additional solvent by utilizing catalytic chain transfer polymerization (CCTP) as the control method, such that the target molecular weight, acceptable dispersity levels, and the desired level of 1,4 addition in the backbone structure and an acceptable yield (∼60%) are achieved. Moreover, the stability and in vitro bioactivity of nanoemulsion adjuvant formulations manufactured with the synthetic poly(isoprene) material are evaluated in comparison to emulsions made with shark-derived squalene. Emulsions containing the synthetic poly(isoprene) achieved smaller particle size and equivalent or enhanced bioactivity (stimulation of cytokine production in human whole blood) compared to corresponding shark squalene emulsions. However, as opposed to the shark squalene-based emulsions, the poly(isoprene) emulsions demonstrated reduced long-term size stability and induced hemolysis at high concentrations. Finally, we demonstrate that the synthetic oligomeric poly(isoprene) material could successfully be hydrogenated such that >95% of the double bonds were successfully removed to give a representative poly(isoprene)-derived squalane mimic.


Assuntos
Adjuvantes Imunológicos/química , Butadienos/química , Hemiterpenos/química , Pentanos/química , Esqualeno/análogos & derivados , Esqualeno/química , Vacinas/química , Química Farmacêutica/métodos , Estabilidade de Medicamentos , Emulsões/química , Tamanho da Partícula , Polimerização
14.
Immunol Rev ; 239(1): 178-96, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21198672

RESUMO

Our improved understanding of how innate immune responses can be initiated and how they can shape adaptive B- and T-cell responses is having a significant impact on vaccine development by directing the development of defined adjuvants. Experience with first generation vaccines, as well as rapid advances in developing defined vaccines containing Toll-like receptor ligands (TLRLs), indicate that an expanded number of safe and effective vaccines containing such molecules will be available in the future. In this review, we outline current knowledge regarding TLRs, detailing the different cell types that express TLRs, the various signaling pathways TLRs utilize, and the currently known TLRLs. We then discuss the current status of TLRLs within vaccine development programs, including the importance of appropriate formulation, and how recent developments can be used to better define the mechanisms of action of vaccines. Finally, we introduce the possibility of using TLRLs, either in combination or with non-TLRLs, to synergistically potentiate vaccine-induced responses to provide not only prophylactic, but therapeutic protection against infectious diseases and cancer.


Assuntos
Adjuvantes Imunológicos , Receptores Toll-Like/imunologia , Vacinas/imunologia , Imunidade Adaptativa , Animais , Células Dendríticas/imunologia , Humanos , Imunidade Inata , Ligantes , Camundongos , Transdução de Sinais , Linfócitos T/imunologia , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
15.
J Nanobiotechnology ; 12: 17, 2014 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-24766820

RESUMO

BACKGROUND: Recent reports that TLR4 and TLR7 ligands can synergistically trigger Th1 biased immune responses suggest that an adjuvant that contains both ligands would be an excellent candidate for co-administration with vaccine antigens for which heavily Th1 biased responses are desired. Ligands of each of these TLRs generally have disparate biochemical properties, however, and straightforward co-formulation may represent an obstacle. RESULTS: We show here that the TLR7 ligand, imiquimod, and the TLR4 ligand, GLA, synergistically trigger responses in human whole blood. We combined these ligands in an anionic liposomal formulation where the TLR7 ligand is in the interior of the liposome and the TLR4 ligand intercalates into the lipid bilayer. The new liposomal formulations are stable for at least a year and have an attractive average particle size of around 140 nm allowing sterile filtration. The synergistic adjuvant biases away from Th2 responses, as seen by significantly reduced IL-5 and enhanced interferon gamma production upon antigen-specific stimulation of cells from immunized mice, than any of the liposomal formulations with only one TLR agonist. Qualitative alterations in antibody responses in mice demonstrate that the adjuvant enhances Th1 adaptive immune responses above any adjuvant containing only a single TLR ligand as well. CONCLUSION: We now have a manufacturable, synergistic TLR4/TLR7 adjuvant that is made with excipients and agonists that are pharmaceutically acceptable and will have a straightforward path into human clinical trials.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Aminoquinolinas/administração & dosagem , Sinergismo Farmacológico , Lipossomos/química , Receptor 4 Toll-Like/imunologia , Receptor 7 Toll-Like/imunologia , Adjuvantes Imunológicos/farmacologia , Aminoquinolinas/farmacologia , Animais , Feminino , Humanos , Imiquimode , Lipídeos/administração & dosagem , Lipídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Células Th1/efeitos dos fármacos , Células Th1/imunologia
16.
Nurs Clin North Am ; 59(2): 219-233, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38670691

RESUMO

People living with HIV (PLWH) have a risk of cardiovascular disease (CVD) that is 1.5 to 2 times higher than the general population owing to traditional risk factors, HIV-mediated factors like chronic inflammation and immune dysfunction, and exposure to antiretroviral therapy. Currently available CVD risk estimation calculators tend to underestimate risk in PLWH but can be useful when an individual's HIV history is considered. Improving modifiable risks is the primary intervention for reducing CVD risk in PLWH. Statin therapy is important for specific individuals, but attention should be given to drug interactions with antiretroviral agents used to treat HIV.


Assuntos
Doenças Cardiovasculares , Infecções por HIV , Prevenção Primária , Humanos , Doenças Cardiovasculares/prevenção & controle , Infecções por HIV/tratamento farmacológico , Infecções por HIV/complicações , Infecções por HIV/prevenção & controle , Prevenção Primária/métodos , Fatores de Risco , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Fármacos Anti-HIV/uso terapêutico
17.
Hum Vaccin Immunother ; 20(1): 2374147, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-39090779

RESUMO

Entamoeba histolytica, the causative agent of amebiasis, is one of the top three parasitic causes of mortality worldwide. However, no vaccine exists against amebiasis. Using a lead candidate vaccine containing the LecA fragment of Gal-lectin and GLA-3M-052 liposome adjuvant, we immunized rhesus macaques via intranasal or intramuscular routes. The vaccine elicited high-avidity functional humoral responses as seen by the inhibition of amebic attachment to mammalian target cells by plasma and stool antibodies. Importantly, antigen-specific IFN-γ-secreting peripheral blood mononuclear cells (PBMCs) and IgG/IgA memory B cells (BMEM) were detected in immunized animals. Furthermore, antigen-specific antibody and cellular responses were maintained for at least 8 months after the final immunization as observed by robust LecA-specific BMEM as well as IFN-γ+ PBMC responses. Overall, both intranasal and intramuscular immunizations elicited a durable and functional response in systemic and mucosal compartments, which supports advancing the LecA+GLA-3M-052 liposome vaccine candidate to clinical testing.


Assuntos
Administração Intranasal , Anticorpos Antiprotozoários , Entamoeba histolytica , Entamebíase , Interferon gama , Leucócitos Mononucleares , Lipossomos , Macaca mulatta , Vacinas Protozoárias , Animais , Entamoeba histolytica/imunologia , Lipossomos/imunologia , Lipossomos/administração & dosagem , Vacinas Protozoárias/imunologia , Vacinas Protozoárias/administração & dosagem , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Leucócitos Mononucleares/imunologia , Entamebíase/prevenção & controle , Entamebíase/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Injeções Intramusculares , Imunogenicidade da Vacina , Adjuvantes de Vacinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Linfócitos B/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina A/imunologia , Imunoglobulina A/sangue , Antígenos de Protozoários/imunologia , Imunidade Humoral , Memória Imunológica , Proteínas de Protozoários/imunologia
18.
Sci Transl Med ; 16(758): eadn6605, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39083589

RESUMO

Authorization of the Matrix-M (MM)-adjuvanted R21 vaccine by three countries and its subsequent endorsement by the World Health Organization for malaria prevention in children are a milestone in the fight against malaria. Yet, our understanding of the innate and adaptive immune responses elicited by this vaccine remains limited. Here, we compared three clinically relevant adjuvants [3M-052 + aluminum hydroxide (Alum) (3M), a TLR7/8 agonist formulated in Alum; GLA-LSQ, a TLR4 agonist formulated in liposomes with QS-21; and MM, the now-approved adjuvant for R21] for their capacity to induce durable immune responses to R21 in macaques. R21 adjuvanted with 3M on a 0, 8, and 23-week schedule elicited anti-circumsporozoite antibody responses comparable in magnitude to the R21/MM vaccine administered using a 0-4-8-week regimen and persisted up to 72 weeks with a half-life of 337 days. A booster dose at 72 weeks induced a recall response similar to the R21/MM vaccination. In contrast, R21/GLA-LSQ immunization induced a lower, short-lived response at the dose used. Consistent with the durable serum antibody responses, MM and 3M induced long-lived plasma cells in the bone marrow and other tissues, including the spleen. Furthermore, whereas 3M stimulated potent and persistent antiviral transcriptional and cytokine signatures after primary and booster immunizations, MM induced enhanced expression of interferon- and TH2-related signatures more highly after the booster vaccination. Collectively, these findings provide a resource on the immune responses of three clinically relevant adjuvants with R21 and highlight the promise of 3M as another adjuvant for malarial vaccines.


Assuntos
Adjuvantes Imunológicos , Vacinas Antimaláricas , Animais , Vacinas Antimaláricas/imunologia , Adjuvantes Imunológicos/farmacologia , Macaca mulatta , Adjuvantes de Vacinas , Anticorpos Antiprotozoários/imunologia , Citocinas/metabolismo
19.
Sci Adv ; 10(27): eadg3747, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38959314

RESUMO

Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.


Assuntos
Adjuvantes Imunológicos , Pirimidinas , Receptor 7 Toll-Like , Receptor 8 Toll-Like , Humanos , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/metabolismo , Animais , Camundongos , Adjuvantes Imunológicos/farmacologia , Receptor 7 Toll-Like/agonistas , Pirimidinas/farmacologia , Pirimidinas/química , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/imunologia , Imidazóis/farmacologia , Imidazóis/química , Células THP-1 , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/imunologia , COVID-19/virologia , COVID-19/imunologia , NF-kappa B/metabolismo , Feminino , Descoberta de Drogas/métodos , Imunidade Inata/efeitos dos fármacos
20.
medRxiv ; 2024 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-38562833

RESUMO

Background: HIV-1 vaccine development is a global health priority. Broadly neutralizing antibodies (bnAbs) which target the HIV-1 gp41 membrane-proximal external region (MPER) have some of the highest neutralization breadth. An MPER peptide-liposome vaccine has been found to expand bnAb precursors in monkeys. Methods: The HVTN133 phase 1 clinical trial (NCT03934541) studied the MPER-peptide liposome immunogen in 24 HIV-1 seronegative individuals. Participants were recruited between 15 July 2019 and 18 October 2019 and were randomized in a dose-escalation design to either 500 mcg or 2000 mcg of the MPER-peptide liposome or placebo. Four intramuscular injections were planned at months 0, 2, 6, and 12. Results: The trial was stopped prematurely due to an anaphylaxis reaction in one participant ultimately attributed to vaccine-associated polyethylene glycol. The immunogen induced robust immune responses, including MPER+ serum and blood CD4+ T-cell responses in 95% and 100% of vaccinees, respectively, and 35% (7/20) of vaccine recipients had blood IgG memory B cells with MPER-bnAb binding phenotype. Affinity purification of plasma MPER+ IgG demonstrated tier 2 HIV-1 neutralizing activity in two of five participants after 3 immunizations. Conclusions: MPER-peptide liposomes induced gp41 serum neutralizing epitope-targeted antibodies and memory B-cell responses in humans despite the early termination of the study. These results suggest that the MPER region is a promising target for a candidate HIV vaccine.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA