Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Semin Immunol ; 50: 101427, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-33277154

RESUMO

In recent years the global market for monoclonal antibodies (mAbs) became a multi-billion-dollar business. This success is mainly driven by treatments in the oncology and autoimmune space. Instead, development of effective mAbs against infectious diseases has been lagging behind. For years the high production cost and limited efficacy have blocked broader application of mAbs in the infectious disease space, which instead has been dominated for almost a century by effective and cheap antibiotics and vaccines. Only very few mAbs against RSV, anthrax, Clostridium difficile or rabies have reached the market. This is about to change. The development of urgently needed and highly effective mAbs as preventive and therapeutic treatments against a variety of pathogens is gaining traction. Vast advances in mAb isolation, engineering and production have entirely shifted the cost-efficacy balance. MAbs against devastating diseases like Ebola, HIV and other complex pathogens are now within reach. This trend is further accelerated by ongoing or imminent health crises like COVID-19 and antimicrobial resistance (AMR), where antibodies could be the last resort. In this review we will retrace the history of antibodies from the times of serum therapy to modern mAbs and lay out how the current run for effective treatments against COVID-19 will lead to a quantum leap in scientific, technological and health care system innovation around mAb treatments for infectious diseases.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , COVID-19/terapia , SARS-CoV-2/imunologia , COVID-19/imunologia , Doenças Transmissíveis/terapia , Humanos , Imunização Passiva/métodos , Soroterapia para COVID-19
2.
Sci Rep ; 9(1): 11520, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31395915

RESUMO

Self-emulsification is routinely used for oral delivery of lipophilic drugs in vivo, with the emulsion forming in vivo. We modified this technique to prepare novel oil-in-water emulsions of varying droplet size and composition on bench to enable adjuvanted vaccine delivery. We used these formulations to show that smaller droplets (20 nm) were much less effective as adjuvants for an influenza vaccine in mice than the emulsion droplet size of commercial influenza vaccine adjuvants (~160 nm). This was unexpected, given the many claims in the literature of the advantages of smaller particulates. We also undertook cell-recruitment mechanistic studies at site of injection and draining lymph nodes to directly address the question of why the smaller droplets were less effective. We discovered that emulsion droplet size and composition have a considerable impact on the ability to recruit immune cells to the injection site. We believe that further work is warranted to more extensively explore the question of whether, the smaller is not 'better', is a more common observation for particulate adjuvants.


Assuntos
Adjuvantes Farmacêuticos/administração & dosagem , Emulsões/química , Vacinas contra Influenza/imunologia , Animais , Anticorpos Antivirais/biossíntese , Composição de Medicamentos , Feminino , Imunidade Celular , Vacinas contra Influenza/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Tamanho da Partícula
3.
Curr Opin Immunol ; 59: 130-134, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31450054

RESUMO

Screening of single B cells from convalescent or vaccinated people allows the discovery of novel targets for infectious diseases and rapid production of engineered human monoclonal antibodies (mAbs) that can prevent or control infections by passive immunization. Here we propose that the development of human mAbs can also significantly accelerate vaccine development by anticipating some of the key biological and regulatory questions.


Assuntos
Anticorpos Monoclonais/imunologia , Antineoplásicos Imunológicos/uso terapêutico , Linfócitos B/fisiologia , Imunoterapia/métodos , Neoplasias/terapia , Vacinas/imunologia , Viroses/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Bioengenharia , Controle de Doenças Transmissíveis , Humanos , Neoplasias/imunologia , Análise de Célula Única
4.
Hum Vaccin Immunother ; 14(1): 45-58, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29172945

RESUMO

Despite high vaccination coverage worldwide, pertussis has re-emerged in many countries. This randomized, controlled, observer-blind phase I study and extension study in Belgium (March 2012-June 2015) assessed safety and immunogenicity of investigational acellular pertussis vaccines containing genetically detoxified pertussis toxin (PT) (NCT01529645; NCT02382913). 420 healthy adults (average age: 26.8 ± 5.5 years, 60% female) were randomized to 1 of 10 vaccine groups: 3 investigational aP vaccines (containing pertussis antigens PT, filamentous hemagglutinin [FHA] and pertactin [PRN] at different dosages), 6 investigational TdaP (additionally containing tetanus toxoid [TT] and diphtheria toxoid [DT]), and 1 TdaP comparator containing chemically inactivated PT. Antibody responses were evaluated on days 1, 8, 30, 180, 365, and approximately 3 years post-booster vaccination. Cell-mediated immune responses and PT neutralization were evaluated in a subset of participants in pre-selected groups. Local and systemic adverse events (AEs), and unsolicited AEs were collected through day 7 and 30, respectively; serious AEs and AEs leading to study withdrawal were collected through day 365 post-vaccination. Antibody responses against pertussis antigens peaked at day 30 post-vaccination and then declined but remained above baseline level at approximately 3 years post-vaccination. Responses to FHA and PRN were correlated to antigen dose. Antibody responses specific to PT, toxin neutralization activity and persistence induced by investigational formulations were similar or significantly higher than the licensed vaccine, despite lower PT doses. Of 15 serious AEs, none were considered vaccination-related; 1 led to study withdrawal (premature labor, day 364; aP4 group). This study confirmed the potential benefits of genetically detoxified PT antigen. All investigational study formulations were well tolerated.


Assuntos
Vacinas contra Difteria, Tétano e Coqueluche Acelular/administração & dosagem , Imunização Secundária/métodos , Toxina Pertussis/imunologia , Vacina contra Coqueluche/administração & dosagem , Vacinação/métodos , Coqueluche/prevenção & controle , Adulto , Anticorpos Antibacterianos/análise , Bélgica , Vacinas contra Difteria, Tétano e Coqueluche Acelular/efeitos adversos , Vacinas contra Difteria, Tétano e Coqueluche Acelular/genética , Vacinas contra Difteria, Tétano e Coqueluche Acelular/imunologia , Feminino , Humanos , Imunidade Celular , Imunogenicidade da Vacina , Masculino , Toxina Pertussis/genética , Vacina contra Coqueluche/efeitos adversos , Vacina contra Coqueluche/genética , Vacina contra Coqueluche/imunologia , Resultado do Tratamento , Coqueluche/sangue , Coqueluche/imunologia , Adulto Jovem
5.
Vaccine ; 35(39): 5256-5263, 2017 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-28823618

RESUMO

A resurgence of whooping cough (pertussis) has been observed in recent years in a number of developed countries, despite widespread vaccine coverage. Although the exact reasons of the recurrence of pertussis are not clear, there are a number of potential causes, like antigenic variation in the circulating strains of Bordetella pertussis, changes in surveillance and diagnostic tools, and potential differences in protection afforded by current acellular pertussis (aP) vaccines compared to more reactogenic whole cell (wP) vaccines, which they replaced. Studies in animal models have shown that induction of cellular as well as humoral immune responses are key to conferring effective and long lasting protection against B. pertussis. wP vaccines induce robust Th1/Th17 responses, which are associated with good protection against lung infection. In contrast, aP vaccines induce mixed Th2/Th17 responses. One research option is to modify current aP vaccines with the intention of inducing protective T cell responses, without compromising on their low reactogenicity profile. Here we found that formulation of an aP vaccine with a novel adjuvant based on a Toll-like receptor 7 agonist (TLR7a) adsorbed to aluminum hydroxide (alum) enhanced B. pertussis-specific Th1 and Th17 responses and serum IgG2a/b antibodies, which had greater functional capacity than those induced by aP formulated with alum alone. Furthermore, addition of a TLR7a enhanced the protective efficacy of the aP vaccine against B. pertussis aerosol challenge; protection was comparable to that of a wP vaccine. These findings suggest that alum-TLR7a is a promising adjuvant for clinical development of next generation pertussis vaccines.


Assuntos
Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/metabolismo , Vacina contra Coqueluche/uso terapêutico , Células Th1/metabolismo , Células Th17/metabolismo , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/metabolismo , Adjuvantes Imunológicos , Animais , Bordetella pertussis/imunologia , Bordetella pertussis/patogenicidade , Células CHO , Cricetulus , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imunidade Humoral/imunologia , Imunidade Humoral/fisiologia , Imunoensaio , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Th1/imunologia , Células Th17/imunologia , Vacinação/métodos
6.
Sci Transl Med ; 9(393)2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28592561

RESUMO

The innate immune mechanisms by which adjuvants enhance the potency and protection of vaccine-induced adaptive immunity are largely unknown. We introduce a model to delineate the steps of how adjuvant-driven innate immune activation leads to priming of vaccine responses using rhesus macaques. Fluorescently labeled HIV-1 envelope glycoprotein (Env) was administered together with the conventional aluminum salt (alum) adjuvant. This was compared to Env given with alum with preabsorbed Toll-like receptor 7 (TLR7) ligand (alum-TLR7) or the emulsion MF59 because they show superiority over alum for qualitatively and quantitatively improved vaccine responses. All adjuvants induced rapid and robust immune cell infiltration to the injection site in the muscle. This resulted in substantial uptake of Env by neutrophils, monocytes, and myeloid and plasmacytoid dendritic cells (DCs) and migration exclusively to the vaccine-draining lymph nodes (LNs). Although less proficient than monocytes and DCs, neutrophils were capable of presenting Env to memory CD4+ T cells. MF59 and alum-TLR7 showed more pronounced cell activation and overall higher numbers of Env+ cells compared to alum. This resulted in priming of higher numbers of Env-specific CD4+ T cells in the vaccine-draining LNs, which directly correlated with increased T follicular helper cell differentiation and germinal center formation. Thus, strong innate immune activation promoting efficient vaccine antigen delivery to infiltrating antigen-presenting cells in draining LNs is an important mechanism by which superior adjuvants enhance vaccine responses.


Assuntos
Vacinas contra a AIDS/imunologia , Adjuvantes Imunológicos/farmacologia , Antígenos/imunologia , Linfonodos/patologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Compostos de Alúmen/farmacologia , Animais , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Centro Germinativo/efeitos dos fármacos , Interferon-alfa/metabolismo , Macaca mulatta , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Músculos/efeitos dos fármacos , Músculos/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Fenótipo , Polissorbatos/farmacologia , Esqualeno/farmacologia , Receptor 7 Toll-Like/metabolismo
7.
Sci Rep ; 6: 38043, 2016 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-27901071

RESUMO

Staphylococcus aureus is the major cause of human septic arthritis and osteomyelitis, which deserve special attention due to their rapid evolution and resistance to treatment. The progression of the disease depends on both bacterial presence in situ and uncontrolled disruptive immune response, which is responsible for chronic disease. Articular and bone infections are often the result of blood bacteremia, with the knees and hips being the most frequently infected joints showing the worst clinical outcome. We report the development of a hematogenous model of septic arthritis in murine knees, which progresses from an acute to a chronic phase, similarly to what occurs in humans. Characterization of the local and systemic inflammatory and immune responses following bacterial infection brought to light specific signatures of disease. Immunization of mice with the vaccine formulation we have recently described (4C-Staph), induced a strong antibody response and specific CD4+ effector memory T cells, and resulted in reduced bacterial load in the knee joints, a milder general inflammatory state and protection against bacterial-mediated cellular toxicity. Possible correlates of protection are finally proposed, which might contribute to the development of an effective vaccine for human use.


Assuntos
Artrite Infecciosa , Articulação do Joelho , Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Staphylococcus aureus/imunologia , Vacinação , Animais , Artrite Infecciosa/imunologia , Artrite Infecciosa/microbiologia , Artrite Infecciosa/patologia , Artrite Infecciosa/prevenção & controle , Feminino , Articulação do Joelho/imunologia , Articulação do Joelho/microbiologia , Articulação do Joelho/patologia , Camundongos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/patologia , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/imunologia , Vacinas Antiestafilocócicas/farmacologia
8.
BMC Microbiol ; 16: 215, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27633511

RESUMO

BACKGROUND: Pertussis or whooping cough is an acute respiratory illness caused by the Gram-negative pathogen Bordetella pertussis. Despite high vaccination coverage whooping cough is currently re-emerging in many developed countries. Although the causes of pertussis resurgence are matter of debate, emerging evidences suggest that acellular vaccines efficiently protect against the hallmark symptoms of pertussis disease but fail to prevent colonization. This presumably impacts on increased risk of bacterial transmission and consequent spread throughout the population. These evidences suggest that improved vaccines may be required for efficient bacterial clearance in the upper respiratory tract. Consequently, there is a need for novel bioassays to evaluate at pre-clinical or clinical level the impact of different vaccines on B. pertussis colonization. RESULTS: We developed a high-throughput bacterial adhesion inhibition (BAI) assay based on human respiratory cell lines and on live bacteria chemically conjugated to a fluorescent dye. Employing A549 cells as model, we evaluated the impact of antibodies elicited by acellular (aP) and whole cell (wP) vaccines on B. pertussis adhesion in vitro. Moreover, we settled the method also on polarized Calu-3 cells grown at air-liquid interface (ALI), showing that this assay can be extended to more complex cell models mimicking the airway epithelium. CONCLUSIONS: We proved that this method is a sensitive, rapid and reproducible system to evaluate the anti-adhesive properties of vaccine-induced antibodies and can be employed to assess improved pertussis vaccines.


Assuntos
Adesinas Bacterianas/análise , Bordetella pertussis/efeitos dos fármacos , Células Epiteliais/microbiologia , Ensaios de Triagem em Larga Escala/métodos , Vacina contra Coqueluche/análise , Sistema Respiratório/microbiologia , Células A549/efeitos dos fármacos , Células A549/microbiologia , Anticorpos Antibacterianos/efeitos dos fármacos , Bordetella pertussis/patogenicidade , Técnicas de Cultura de Células , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/microbiologia , Imunofluorescência/métodos , Humanos , Modelos Biológicos , Vacina contra Coqueluche/uso terapêutico , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Vacinação , Vacinas Acelulares/análise , Vacinas Acelulares/uso terapêutico , Coqueluche/tratamento farmacológico , Coqueluche/microbiologia
9.
J Infect Dis ; 213(12): 1876-85, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-26908732

RESUMO

BACKGROUND: Most preclinical studies assess vaccine effectiveness in single-pathogen infection models. This is unrealistic given that humans are continuously exposed to different commensals and pathogens in sequential and mixed infections. Accordingly, complications from secondary bacterial infection are a leading cause of influenza-associated morbidity and mortality. New vaccination strategies are needed to control infections on simultaneous fronts. METHODS: We compared different anti-influenza vaccines for their protective potential in a model of viral infection with bacterial superinfection. Mice were immunized with H1N1/A/California/7/2009 subunit vaccines, formulated with different adjuvants inducing either T-helper type 1 (Th1) (MF59 plus CpG)-, Th1/2 (MF59)-, or Th17 (LTK63)-prone immune responses and were sequentially challenged with mouse-adapted influenza virus H1N1/A/Puerto Rico/8/1934 and Staphylococcus aureus USA300, a clonotype emerging as a leading contributor in postinfluenza pneumonia in humans. RESULTS: Unadjuvanted vaccine controlled single viral infection, yet mice had considerable morbidity from viral disease and bacterial superinfection. In contrast, all adjuvanted vaccines efficiently protected mice in both conditions. Interestingly, the Th1-inducing formulation was superior to Th1/2 or Th17 inducers. CONCLUSIONS: Our studies should help us better understand how differential immunity to influenza skews immune responses toward coinfecting bacteria and discover novel modes to prevent bacterial superinfections in the lungs of persons with influenza.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/imunologia , Superinfecção/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Animais , Toxinas Bacterianas/administração & dosagem , Enterotoxinas/administração & dosagem , Proteínas de Escherichia coli/administração & dosagem , Feminino , Humanos , Imunização , Vacinas contra Influenza/administração & dosagem , Influenza Humana/complicações , Influenza Humana/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos/administração & dosagem , Polissorbatos/administração & dosagem , Organismos Livres de Patógenos Específicos , Esqualeno/administração & dosagem , Infecções Estafilocócicas/complicações , Infecções Estafilocócicas/microbiologia , Superinfecção/microbiologia
10.
Int J Pharm ; 492(1-2): 169-76, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26149936

RESUMO

The successful approach of combining diphtheria, tetanus and pertussis antigens into a single vaccine has become a cornerstone of immunization programs. Yet, even if vaccination coverage is high, a resurgence of pertussis has been reported in many countries suggesting current vaccines may not provide adequate protection. To induce better tailored and more durable immune responses against pertussis vaccines different approaches have been proposed, including the use of novel adjuvants. Licensed aP vaccines contain aluminum salts, which mainly stimulate humoral immune responses and might not be ideal for protecting against Bordetella pertussis infection. Adjuvants inducing more balanced T-helper profiles or even Th1-prone responses might be more adequate. In this study, two adjuvants already approved for human use have been tested: MF59 emulsion and the combination of aluminum hydroxide with the Toll-Like Receptor 4 agonist MPLA. Adjuvanticity was evaluated in a mouse model using a TdaP vaccine containing three B. pertussis antigens: genetically detoxified pertussis toxin (PT-9K/129G), filamentous hemagglutinin (FHA) and pertactin (PRN) The physico-chemical compatibility of TdaP antigens with the proposed adjuvants, together with a quicker onset and changed quality of the antibody responses, fully supports the replacement of aluminum salts with a new adjuvant to enhance aP vaccines immunogenicity.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacina contra Difteria, Tétano e Coqueluche/administração & dosagem , Lipídeo A/análogos & derivados , Polissorbatos/administração & dosagem , Esqualeno/administração & dosagem , Adjuvantes Imunológicos/química , Compostos de Alúmen/administração & dosagem , Compostos de Alúmen/química , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Bordetella pertussis/imunologia , Células CHO , Linhagem Celular Tumoral , Chlorocebus aethiops , Cricetulus , Toxina Diftérica/imunologia , Vacina contra Difteria, Tétano e Coqueluche/química , Feminino , Humanos , Imunoglobulina G/sangue , Lipídeo A/administração & dosagem , Lipídeo A/química , Camundongos , Camundongos Endogâmicos BALB C , Toxina Pertussis/imunologia , Polissorbatos/química , Esqualeno/química , Células Vero
11.
Microsc Microanal ; 21(4): 791-5, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26223548

RESUMO

Adjuvants are substances that enhance adaptive immune responses when formulated in a vaccine. Alum and MF59 are two vaccine adjuvants licensed for human vaccination. Their mode of action has not been completely elucidated. Here we show the first ultrastructural visualization of Alum and MF59 interaction with immune cells in vitro and in vivo. We observed that Alum is engulfed by cells as inclusions of laminae that are detectable within draining lymph nodes. MF59 is instead engulfed by cells in vitro as low-electron-dense lipid-like inclusions that display a vesicle pattern, as confirmed by confocal microscopy using fluorescently labeled MF59. However, lipid-like inclusions with different high- and low-electron-dense content are detected within cells of draining lymph nodes when injecting MF59. As high-electron-dense lipid-like inclusions are also detected upon injection of Alum, our results suggest that the low-electron-dense inclusions are formed by engulfed MF59, whereas the high-electron-dense inclusions are proper lipid inclusions. Thus, we demonstrated that vaccine adjuvants are engulfed as inclusions by lymph node cells and hypothesize that adjuvant treatment may modify lipid metabolism.


Assuntos
Adjuvantes Imunológicos/farmacocinética , Compostos de Alúmen/farmacocinética , Polissorbatos/farmacocinética , Esqualeno/farmacocinética , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Endocitose , Corpos de Inclusão/ultraestrutura , Camundongos Endogâmicos C57BL , Microscopia , Polissorbatos/administração & dosagem , Esqualeno/administração & dosagem
12.
J Immunol ; 194(10): 4836-45, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25870238

RESUMO

The early life influenza disease burden calls for more effective vaccines to protect this vulnerable population. Influenza vaccines including the MF59 oil-in-water adjuvant induce higher, broader, and more persistent Ab responses in adults and particularly in young, through yet undefined mechanisms. In this study, we show that MF59 enhances adult murine IgG responses to influenza hemagglutinin (HA) by promoting a potent T follicular helper cells (TFH) response, which directly controls the magnitude of the germinal center (GC) B cell response. Remarkably, this enhancement of TFH and GC B cells is already fully functional in 3-wk-old infant mice, which were fully protected by HA/MF59 but not HA/PBS immunization against intranasal challenge with the homologous H1N1 (A/California/7/2009) strain. In 1-wk-old neonatal mice, MF59 recruits and activates APCs, efficiently induces CD4(+) effector T cells and primes for enhanced infant responses but induces few fully functional TFH cells, which are mostly follicular regulatory T cells, and poor GC and anti-HA responses. The B cell adjuvanticity of MF59 appears to be mediated by the potent induction of TFH cells which directly controls GC responses both in adult and early life, calling for studies assessing its capacity to enhance the efficacy of influenza immunization in young infants.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas contra Influenza/imunologia , Polissorbatos/farmacologia , Esqualeno/farmacologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Animais Recém-Nascidos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Centro Germinativo/imunologia , Imuno-Histoquímica , Vírus da Influenza A Subtipo H1N1/imunologia , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Esqualeno/imunologia
13.
Expert Rev Vaccines ; 13(10): 1191-204, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25183193

RESUMO

Pertussis toxin (PT) is one of the major virulence factors of Bordetella pertussis and the primary component of all pertussis vaccines available to date. Because of its various noxious effects the toxin needs to be detoxified. In all currently available vaccines, detoxification is achieved by treatment with high quantity of chemical agents such as formaldehyde, glutaraldehyde or hydrogen peroxide. Although effective in detoxification, this chemical treatment alters dramatically the immunological properties of the toxin. In contrast, PT genetically detoxified through the substitution of two residues necessary for its enzymatic activity maintains all functional and immunological properties. This review describes in detail the characteristics of this PT-9K/129G mutant and shows that it is non-toxic and a superior immunogen compared with chemically detoxified PT. Importantly, data from an efficacy trial show that the PT-9K/129G-based vaccine induces earlier and longer-lasting protection, further supporting the hypothesis that PT-9K/129G represents an ideal candidate for future pertussis vaccine formulations.


Assuntos
Toxina Pertussis/genética , Toxina Pertussis/imunologia , Vacina contra Coqueluche/genética , Vacina contra Coqueluche/imunologia , Animais , Bordetella pertussis/enzimologia , Bordetella pertussis/genética , Bordetella pertussis/imunologia , Humanos , Modelos Moleculares , Toxina Pertussis/química , Toxina Pertussis/fisiologia , Vacina contra Coqueluche/química , Vacina contra Coqueluche/metabolismo
14.
Mol Ther ; 22(12): 2118-2129, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25027661

RESUMO

Nucleic acid-based vaccines such as viral vectors, plasmid DNA, and mRNA are being developed as a means to address a number of unmet medical needs that current vaccine technologies have been unable to address. Here, we describe a cationic nanoemulsion (CNE) delivery system developed to deliver a self-amplifying mRNA vaccine. This nonviral delivery system is based on Novartis's proprietary adjuvant MF59, which has an established clinical safety profile and is well tolerated in children, adults, and the elderly. We show that nonviral delivery of a 9 kb self-amplifying mRNA elicits potent immune responses in mice, rats, rabbits, and nonhuman primates comparable to a viral delivery technology, and demonstrate that, relatively low doses (75 µg) induce antibody and T-cell responses in primates. We also show the CNE-delivered self-amplifying mRNA enhances the local immune environment through recruitment of immune cells similar to an MF59 adjuvanted subunit vaccine. Lastly, we show that the site of protein expression within the muscle and magnitude of protein expression is similar to a viral vector. Given the demonstration that self-amplifying mRNA delivered using a CNE is well tolerated and immunogenic in a variety of animal models, we are optimistic about the prospects for this technology.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Emulsões/administração & dosagem , Imunidade Celular , RNA Mensageiro/imunologia , RNA Viral/imunologia , Vacinas de DNA/administração & dosagem , Animais , Cátions , Emulsões/química , Feminino , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Ratos
15.
Proc Natl Acad Sci U S A ; 110(52): 21095-100, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24324152

RESUMO

Vaccines are the most effective agents to control infections. In addition to the pathogen antigens, vaccines contain adjuvants that are used to enhance protective immune responses. However, the molecular mechanism of action of most adjuvants is ill-known, and a better understanding of adjuvanticity is needed to develop improved adjuvants based on molecular targets that further enhance vaccine efficacy. This is particularly important for tuberculosis, malaria, AIDS, and other diseases for which protective vaccines do not exist. Release of endogenous danger signals has been linked to adjuvanticity; however, the role of extracellular ATP during vaccination has never been explored. Here, we tested whether ATP release is involved in the immune boosting effect of four common adjuvants: aluminum hydroxide, calcium phosphate, incomplete Freund's adjuvant, and the oil-in-water emulsion MF59. We found that intramuscular injection is always associated with a weak transient release of ATP, which was greatly enhanced by the presence of MF59 but not by all other adjuvants tested. Local injection of apyrase, an ATP-hydrolyzing enzyme, inhibited cell recruitment in the muscle induced by MF59 but not by alum or incomplete Freund's adjuvant. In addition, apyrase strongly inhibited influenza-specific T-cell responses and hemagglutination inhibition titers in response to an MF59-adjuvanted trivalent influenza vaccine. These data demonstrate that a transient ATP release is required for innate and adaptive immune responses induced by MF59 and link extracellular ATP with an enhanced response to vaccination.


Assuntos
Trifosfato de Adenosina/metabolismo , Adjuvantes Imunológicos/farmacologia , Linfócitos T CD4-Positivos/imunologia , Músculo Esquelético/metabolismo , Polissorbatos/farmacologia , Esqualeno/farmacologia , Vacinação/métodos , Hidróxido de Alumínio/imunologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Fosfatos de Cálcio/imunologia , Sinergismo Farmacológico , Ensaio de Imunoadsorção Enzimática , Adjuvante de Freund/imunologia , Lipídeos/imunologia , Medições Luminescentes , Camundongos , Camundongos Endogâmicos BALB C , Organismos Livres de Patógenos Específicos , Esqualeno/imunologia
16.
Vaccine ; 31(33): 3363-9, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23684834

RESUMO

MF59 is a safe and effective vaccine adjuvant that has been used in a licensed seasonal influenza vaccine for 15 years. The purpose of the present studies was to directly address a question that has been asked of us on many occasions: "which is the adjuvant active component of MF59?". Since we have recently gained a number of insights on how MF59 works as an adjuvant, we were able to use these approaches to evaluate if the individual components of MF59 (squalene oil, the surfactants Span 85 and Tween 80 or the citrate buffer) showed any direct immunostimulatory activity. We assessed the ability of the individual components to stimulate the innate and adaptive immune responses that we have shown to be indicative of MF59-mediated adjuvanticity. No immune stimulatory capacities could be attributed to squalene, Tween 80 or the citrate buffer alone. Instead, we found that the lipophilic surfactant Span 85 contributes to activation of the muscle transcriptome. However, despite this local activation, Span 85 alone - like the other single components of MF59 - is not sufficient to induce an adjuvant effect. Only the fully formulated MF59 emulsion induces all the established hallmarks of innate and adaptive immune activation, which includes activation of genes indicative of transendothelial cell migration, strong influx of immune cells into the injection site and their enhanced antigen uptake and transport to the lymph nodes. These observations may have important implications in the design of optimal emulsion-based vaccine adjuvants.


Assuntos
Adjuvantes Imunológicos/farmacologia , Emulsões/farmacologia , Polissorbatos/farmacologia , Esqualeno/farmacologia , Imunidade Adaptativa , Adjuvantes Imunológicos/química , Animais , Anticorpos Antivirais/sangue , Apresentação de Antígeno/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Emulsões/química , Feminino , Testes de Inibição da Hemaglutinação , Hexoses/farmacologia , Humanos , Imunidade Inata , Vacinas contra Influenza/farmacologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Ativação Linfocitária , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Polissorbatos/química , Esqualeno/química , Transcriptoma/efeitos dos fármacos
17.
PLoS One ; 7(7): e40411, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22848376

RESUMO

Streptococcus pyogenes is a major human pathogen worldwide, responsible for both local and systemic infections. These bacteria express the subtilisin-like protease SpyCEP which cleaves human IL-8 and related chemokines. We show that localization of SpyCEP is growth-phase and strain dependent. Significant shedding was observed only in a strain naturally overexpressing SpyCEP, and shedding was not dependent on SpyCEP autoproteolytic activity. Surface-bound SpyCEP in two different strains was capable of cleaving IL-8. To investigate SpyCEP action in vivo, we adapted the mouse air pouch model of infection for parallel quantification of bacterial growth, host immune cell recruitment and chemokine levels in situ. In response to infection, the predominant cells recruited were neutrophils, monocytes and eosinophils. Concomitantly, the chemokines KC, LIX, and MIP-2 in situ were drastically increased in mice infected with the SpyCEP knockout strain, and growth of this mutant strain was reduced compared to the wild type. SpyCEP has been described as a potential vaccine candidate against S. pyogenes, and we showed that surface-associated SpyCEP was recognized by specific antibodies. In vitro, such antibodies also counteracted the inhibitory effects of SpyCEP on chemokine mediated PMN recruitment. Thus, α-SpyCEP antibodies may benefit the host both directly by enabling opsonophagocytosis, and indirectly, by neutralizing an important virulence factor. The animal model we employed shows promise for broad application in the study of bacterial pathogenesis.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Peptídeo Hidrolases/metabolismo , Proteólise , Infecções Estreptocócicas/enzimologia , Streptococcus pyogenes/fisiologia , Animais , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Quimiocinas/genética , Quimiocinas/imunologia , Quimiocinas/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Leucócitos/imunologia , Leucócitos/metabolismo , Camundongos , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/imunologia , Vacinas Antiestafilocócicas/genética , Vacinas Antiestafilocócicas/imunologia , Infecções Estreptocócicas/genética , Infecções Estreptocócicas/imunologia
18.
J Immunol ; 188(7): 3088-98, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22351935

RESUMO

The innate immune pathways induced by adjuvants required to increase adaptive responses to influenza subunit vaccines are not well characterized. We profiled different TLR-independent (MF59 and alum) and TLR-dependent (CpG, resiquimod, and Pam3CSK4) adjuvants for the ability to increase the immunogenicity to a trivalent influenza seasonal subunit vaccine and to tetanus toxoid (TT) in mouse. Although all adjuvants boosted the Ab responses to TT, only MF59 and Pam3CSK4 were able to enhance hemagglutinin Ab responses. To identify innate immune correlates of adjuvanticity to influenza subunit vaccine, we investigated the gene signatures induced by each adjuvant in vitro in splenocytes and in vivo in muscle and lymph nodes using DNA microarrays. We found that flu adjuvanticity correlates with the upregulation of proinflammatory genes and other genes involved in leukocyte transendothelial migration at the vaccine injection site. Confocal and FACS analysis confirmed that MF59 and Pam3CSK4 were the strongest inducers of blood cell recruitment in the muscle compared with the other adjuvants tested. Even though it has been proposed that IFN type I is required for adjuvanticity to influenza vaccines, we found that MF59 and Pam3CSK4 were not good inducers of IFN-related innate immunity pathways. By contrast, resiquimod failed to enhance the adaptive response to flu despite a strong activation of the IFN pathway in muscle and lymph nodes. By blocking IFN type I receptor through a mAb, we confirmed that the adjuvanticity of MF59 and Pam3CSK4 to a trivalent influenza vaccine and to TT is IFN independent.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas contra Influenza/imunologia , Interferon-alfa/imunologia , Lipopeptídeos/farmacologia , Polissorbatos/farmacologia , Esqualeno/farmacologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Quimiotaxia/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Imunidade Inata/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza B/imunologia , Linfonodos/imunologia , Linfonodos/metabolismo , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Músculos/imunologia , Músculos/metabolismo , Organismos Livres de Patógenos Específicos , Baço/citologia , Baço/imunologia , Baço/metabolismo , Toxoide Tetânico/imunologia , Vacinas de Subunidades Antigênicas/imunologia
19.
Proc Natl Acad Sci U S A ; 108(27): 11169-74, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690334

RESUMO

Oil-in-water emulsions have been successfully used to increase the efficacy, immunogenicity, and cross-protection of human vaccines; however, their mechanism of action is still largely unknown. Nlrp3 inflammasome has been previously associated to the activity of alum, another adjuvant broadly used in human vaccines, and MyD88 adaptor protein is required for the adjuvanticity of most Toll-like receptor agonists. We compared the contribution of Nlrp3 and MyD88 to the adjuvanticity of alum, the oil-in-water emulsion MF59, and complete Freund's adjuvant in mice using a three-component vaccine against serogroup B Neisseria meningitidis (rMenB). Although the basal antibody responses to the nonadjuvanted rMenB vaccine were largely dependent on Nlrp3, the high-level antibody responses induced by alum, MF59, or complete Freund's adjuvant did not require Nlrp3. Surprisingly, we found that MF59 requires MyD88 to enhance bactericidal antibody responses to the rMenB vaccine. Because MF59 did not activate any of the Toll-like receptors in vitro, we propose that MF59 requires MyD88 for a Toll-like receptor-independent signaling pathway.


Assuntos
Adjuvantes Imunológicos/farmacologia , Proteínas de Transporte/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Polissorbatos/farmacologia , Esqualeno/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Anticorpos Antibacterianos/biossíntese , Vacinas Bacterianas/administração & dosagem , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Emulsões , Feminino , Adjuvante de Freund/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR , Neisseria meningitidis Sorogrupo B/imunologia , Polissorbatos/administração & dosagem , Transdução de Sinais , Esqualeno/administração & dosagem , Receptores Toll-Like/metabolismo , Vacinas Sintéticas/administração & dosagem
20.
Vaccine ; 29(29-30): 4728-34, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21571025

RESUMO

We previously investigated immunogenicity of meningococcal native outer membrane vesicle (NOMV) vaccines prepared from recombinant strains with attenuated endotoxin (ΔLpxL1) and over-expressed factor H binding protein (fHbp) in a mouse model. The vaccines elicited broad serum bactericidal antibody responses. While human toll-like receptor 4 (TLR-4) is mainly stimulated by wildtype meningococcal endotoxin, mouse TLR-4 is stimulated by both the wildtype and mutant endotoxin. An adjuvant effect in mice of the mutant endotoxin would be expected to be much less in humans, and may have contributed to the broad mouse bactericidal responses. Here we show that as previously reported for humans, rhesus primate peripheral blood mononuclear cells incubated with a NOMV vaccine from ΔLpxL1 recombinant strains had lower proinflammatory cytokine responses than with a control wildtype NOMV vaccine. The cytokine responses to the mutant vaccine were similar to those elicited by a detergent-treated, wildtype outer membrane vesicle vaccine that had been safely administered to humans. Monkeys (N=4) were immunized beginning at ages 2-3 months with three doses of a NOMV vaccine prepared from ΔLpxL1 recombinant strains with over-expressed fHbp in the variant 1 and 2 groups. The mutant NOMV vaccine elicited serum bactericidal titers≥1:4 against all 10 genetically diverse strains tested, including 9 with heterologous PorA to those in the vaccine. Negative-control animals had serum bactericidal titers<1:4. Thus, the mutant NOMV vaccine elicited broadly protective serum antibodies in a non-human infant primate model that is more relevant for predicting human antibody responses than mice.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Micropartículas Derivadas de Células/imunologia , Endotoxinas/imunologia , Vacinas Meningocócicas/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/administração & dosagem , Proteínas de Bactérias/administração & dosagem , Atividade Bactericida do Sangue , Citocinas/metabolismo , Endotoxinas/administração & dosagem , Imunização Secundária/métodos , Leucócitos Mononucleares/imunologia , Macaca mulatta , Vacinas Meningocócicas/administração & dosagem , Vacinação/métodos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...