Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Hum Vaccin ; 6(7): 585-94, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20421727

RESUMO

Mutations of influenza virus increase concerns of worldwide epidemics resulting from the newly emergent strains. Current influenza vaccines are inefficient and require annual vaccinations. W805EC adjuvant is an oil-in-water emulsion composed of nanodroplets with an average diameter of approximately 400 nm. The nanoemulsion adjuvant has been used successfully to stimulate the immune response when mixed with several other antigens in animal models. In this study, W805EC nanoemulsion adjuvant activity was evaluated using nasal influenza vaccination in a murine model. Five to twenty percent W805EC adjuvant was used to inactivate influenza A/Puerto Rico/8/34/05 (H1N1). Mice immunized with the nanoemulsion adjuvanted influenza virus intranasally showed a robust specific humoral immune response as demonstrated using ELISA and HAI assays. Serum HAI titers were more than 104 following two vaccinations. Vaccinated mice were also protected against challenge with an LD80 of live influenza virus. Splenocytes from vaccinated mice were assayed for cytokine production following virus stimulation. The cytokine profile demonstrated a robust cellular immune response with enhanced Th1 and Th17 immunity that provided balanced immunity against both intracellular and extracellular forms of the virus. Additionally, the vaccine preparations showed minimal protein degradation but remained potent when stored at 4°C for up to three months. This work demonstrates that W805EC nanoemulsion adjuvant can effectively enhance the immunogenicity of influenza hemagglutinin antigen. The nanoemulsion adjuvant can result in antigen sparing and cross-protection. The potential exists for a nasally administered influenza vaccine that may require little or no refrigerated storage.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Emulsões/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/imunologia , Adjuvantes Imunológicos/efeitos adversos , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Citocinas/metabolismo , Emulsões/efeitos adversos , Ensaio de Imunoadsorção Enzimática , Feminino , Testes de Inibição da Hemaglutinação , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Leucócitos Mononucleares/imunologia , Masculino , Camundongos , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/prevenção & controle , Porto Rico , Doenças dos Roedores/mortalidade , Doenças dos Roedores/prevenção & controle , Baço/imunologia , Análise de Sobrevida , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/efeitos adversos , Vacinas de Produtos Inativados/imunologia
2.
Vaccine ; 37(43): 6470-6477, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31515143

RESUMO

Genital herpes is a sexually transmitted disease representing a major global health concern. Currently, there is no approved vaccine and existing antiviral therapies exhibit limited efficacy. Herein, we describe an intranasal (IN) vaccine comprised of HSV-2 surface glycoproteins gD2 and gB2 formulated in a nanoemulsion adjuvant (NE01-gD2/gB2). Using the HSV-2 genital herpes guinea pig model, we demonstrate that IN NE01-gD2/gB2 induces higher levels of neutralizing antibody compared to a monovalent IN NE01-gD2 vaccine, but less than an intramuscular (IM) Alum/MPL-gD2 vaccine. Following intravaginal (IVag) challenge with HSV-2, the group immunized with IN NE01-gD2/gB2 exhibited significantly reduced acute and recurrent disease scores compared to placebo recipients. Significantly, latent virus was only detected in the dorsal root ganglia of 1 of 12 IN NE01-gD2/gB2-vaccinated animals compared to 11 of 12 placebo recipient. In the therapeutic model, IN NE01-gD2/gB2 immunized guinea pigs exhibited a significant reduction in the recurrent lesions scores (64%, p < 0.01), number of animal days with disease (64%, p < 0.01), number of animals with viral shedding (50%, p < 0.04) and reduction in virus positive vaginal swabs (56%, p < 0.04), These data suggests that the treatment may be effective in treating chronic disease and minimizing virus transmission. These results warrant advancing the development of IN NE01-gD2/gB2 as both a prophylactic and therapeutic vaccine against HSV-2.


Assuntos
Anticorpos Antivirais/sangue , Herpes Genital/prevenção & controle , Herpes Genital/terapia , Vacinas contra o Vírus do Herpes Simples/administração & dosagem , Vacinas contra o Vírus do Herpes Simples/uso terapêutico , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Emulsões/administração & dosagem , Emulsões/química , Feminino , Cobaias , Herpes Genital/imunologia , Herpesvirus Humano 2 , Nanopartículas/administração & dosagem , Nanopartículas/química , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/uso terapêutico , Eliminação de Partículas Virais
3.
Vaccine ; 35(37): 4983-4989, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28774560

RESUMO

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) is contracted via aerosol infection, typically affecting the lungs. Mycobacterium bovis bacillus Calmette-Guerin (BCG) is the only licensed vaccine and has variable efficacy in protecting against pulmonary TB. Additionally, chemotherapy is associated with low compliance contributing to development of multidrug-resistant (MDR) and extensively drug-resistant (XDR) Mtb. Thus, there is an urgent need for the design of more effective vaccines against TB. Experimental vaccines delivered through the mucosal route induce robust T helper type 17 (Th17)/ Interleukin (IL) -17 responses and provide superior protection against Mtb infection. Thus, the development of safe mucosal adjuvants for human use is critical. In this study, we demonstrate that nanoemulsion (NE)-based adjuvants when delivered intranasally along with Mtb specific immunodominant antigens (NE-TB vaccine) induce potent mucosal IL-17T-cell responses. Additionally, the NE-TB vaccine confers significant protection against Mtb infection, and when delivered along with BCG, is associated with decreased disease severity. These findings strongly support the development of a NE-TB vaccine as a novel, safe and effective, first-of-kind IL-17 inducing mucosal vaccine for potential use in humans.


Assuntos
Interleucina-17/metabolismo , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Vacinas contra a Tuberculose/imunologia , Vacinas contra a Tuberculose/uso terapêutico , Adjuvantes Imunológicos , Animais , Antígenos de Bactérias/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Tuberculose/imunologia , Tuberculose/prevenção & controle
4.
Hum Vaccin Immunother ; 11(12): 2904-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26307915

RESUMO

Respiratory Syncytial Virus is a leading cause of bronchiolitis and pneumonia in infants, the elderly and individuals with compromised immune systems. Despite decades of research, there is currently no available vaccine for RSV. Our group has previously demonstrated that intranasal immunization of mice with RSV inactivated by and adjuvanted with W805EC nanoemulsion elicits robust humoral and cellular immune responses, resulting in protection against RSV infection. This protection was achieved without the induction of airway hyper-reactivity or a Th2-skewed immune response. The cotton rat Sigmodon hispidus has been used for years as an excellent small animal model of RSV disease. Thus, we extended these rodent studies to the more permissive cotton rat model. Intranasal immunization of the nanoemulsion-adjuvanted RSV vaccines induced high antibody titers and a robust Th1-skewed cellular response. Importantly, vaccination provided sterilizing cross-protective immunity against a heterologous RSV challenge and did not induce marked or severe histological effects or eosinophilia in the lung after viral challenge. Overall, these data demonstrate that nanoemulsion-formulated whole RSV vaccines are both safe and effective for immunization in multiple animal models.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Emulsões/uso terapêutico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/uso terapêutico , Sigmodontinae/imunologia , Vacinas de Produtos Inativados/imunologia , Administração Intranasal , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Bronquiolite/imunologia , Bronquiolite/prevenção & controle , Bronquiolite/virologia , Proteção Cruzada/imunologia , Feminino , Pulmão/patologia , Pulmão/virologia , Pneumonia Viral/imunologia , Pneumonia Viral/prevenção & controle , Pneumonia Viral/virologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Sigmodontinae/virologia , Células Th1/imunologia , Vacinação , Proteínas Virais de Fusão/imunologia , Carga Viral/imunologia
5.
PLoS One ; 10(5): e0126120, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25962136

RESUMO

Vaccine adjuvants have been reported to induce both mucosal and systemic immunity when applied to mucosal surfaces and this dual response appears important for protection against certain pathogens. Despite the potential advantages, however, no mucosal adjuvants are currently approved for human use. Evaluating compounds as mucosal adjuvants is a slow and costly process due to the need for lengthy animal immunogenicity studies. We have constructed a library of 112 intranasal adjuvant candidate formulations consisting of oil-in-water nanoemulsions that contain various cationic and nonionic surfactants. To facilitate adjuvant development we first evaluated this library in a series of high-throughput, in vitro assays for activities associated with innate and adaptive immune activation in vivo. These in vitro assays screened for the ability of the adjuvant to bind to mucin, induce cytotoxicity, facilitate antigen uptake in epithelial and dendritic cells, and activate cellular pathways. We then sought to determine how these parameters related to adjuvant activity in vivo. While the in vitro assays alone were not enough to predict the in vivo adjuvant activity completely, several interesting relationships were found with immune responses in mice. Furthermore, by varying the physicochemical properties of the surfactant components (charge, surfactant polar head size and hydrophobicity) and the surfactant blend ratio of the formulations, the strength and type of the immune response generated (TH1, TH2, TH17) could be modulated. These findings suggest the possibility of using high-throughput screens to aid in the design of custom adjuvants with unique immunological profiles to match specific mucosal vaccine applications.


Assuntos
Adjuvantes Imunológicos/química , Vacinas/administração & dosagem , Vacinas/química , Adjuvantes Imunológicos/toxicidade , Administração Intranasal , Animais , Linhagem Celular , Química Farmacêutica , Citocinas/biossíntese , Emulsões , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Ensaios de Triagem em Larga Escala , Imunidade Celular , Imunidade Humoral , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Nanotecnologia
6.
Hum Vaccin Immunother ; 10(3): 615-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24326268

RESUMO

Respiratory Syncytial Virus (RSV) is a ubiquitous virus that infects almost all people by age two and is a major source of respiratory illness in infants, the elderly and others with compromised immune systems. Currently there is no available vaccine. Prior efforts using formalin-inactivated RSV (FI-RSV) were associated with enhanced respiratory disease upon viral exposure following clinical vaccine trials. Several researchers and pharmaceutical companies have utilized vector-associated live attenuated RSV vaccines in pre-clinical and clinical studies. Another attractive approach, however, is a subunit vaccine which would be easier to produce and quality control. Our group has previously demonstrated in a murine model of infection that intranasal immunization with nanoemulsion-inactivated and adjuvanted RSV induces humoral and cellular immune responses, resulting in protection against RSV infection. The present studies characterize the immune responses elicited by intranasal RSV F protein adjuvanted with nanoemulsion. Intranasal application of nanoemulsion adjuvanted F protein induced a rapid and robust systemic and mucosal antibody response, as well as protection against subsequent RSV challenge. Importantly, RSV challenge in immunized animals did not elicit airway hyper-reactivity, a Th2-skewed immune response or immunopathology associated with hypersensitivity reactions with formalin-inactivated vaccine. These results suggest that RSV F protein adjuvanted with nanoemulsion may be a good mucosal vaccine candidate. Formulating RSV F protein in nanoemulsion creates a well-defined and well-controlled vaccine that can be delivered intranasally to induce T cell mediated immunity without inducing enhanced disease associated with the mouse model of FI-RSV vaccination and infection.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Imunização/métodos , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/sangue , Sangue/imunologia , Modelos Animais de Doenças , Emulsões/administração & dosagem , Feminino , Imunidade nas Mucosas , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vírus Sinciciais Respiratórios/isolamento & purificação , Células Th2/imunologia , Resultado do Tratamento , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas Virais de Fusão/imunologia
7.
Vaccine ; 30(48): 6871-7, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-22989689

RESUMO

Currently available influenza vaccines provide suboptimal protection. In order to improve the quality of protective immune responses elicited following vaccination, we developed an oil-in-water nanoemulsion (NE)-based adjuvant for an intranasally-delivered inactivated influenza vaccine. Using a prime-boost vaccination regimen, we show that intranasal vaccines containing the W(80)5EC NE elicited higher titers of serum hemagglutination inhibiting (HAI) antibody and influenza-specific IgG and IgA titers compared to vaccines that did not contain the NE. Similarly, vaccines containing the W(80)5EC NE resulted in higher influenza-specific IgA levels in the bronchoalveolar lavage (BAL) fluid and nasal wash when compared to vaccines formulated without NE. The higher antibody titers in mice immunized with the NE-containing vaccines correlated with reduced viral loads in the lungs and nasal turbinates following a high dose viral challenge. Mice immunized with vaccines containing the W(80)5EC NE also showed a reduction in body weight loss following challenge compared to mice immunized with equivalent vaccines produced without NE. Taken together, our results show that the W(80)5EC NE substantially improves the magnitude of protective influenza-specific antibody responses and is a promising mucosal adjuvant for influenza vaccines and vaccines against other mucosal pathogens.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Emulsões/administração & dosagem , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Nanoestruturas/administração & dosagem , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/virologia , Modelos Animais de Doenças , Feminino , Testes de Inibição da Hemaglutinação , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Camundongos , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Infecções por Orthomyxoviridae/prevenção & controle , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia
8.
Clin Vaccine Immunol ; 18(7): 1167-75, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21543588

RESUMO

NB-1008 is a surfactant-stabilized soybean oil-in-water nanoemulsion (NE) adjuvant with influenza virus antigen incorporated into the NE by simple mixing. Intranasal administration of the antigen with NE adjuvant efficiently produces both mucosal and serum antibody responses as well as a robust cellular Th1 immune response. To demonstrate the adjuvant effect of the W(80)5EC NE, a killed commercial influenza vaccine for intramuscular administration (Fluzone or Fluvirin) was mixed with the W(80)5EC NE adjuvant and administered intranasally to naïve ferrets. After a single intranasal immunization, the adjuvanted influenza vaccine elicited elevated serum hemagglutination inhibition (HAI) geometric mean titers (GMTs) ranging from 196 to 905 for the three hemagglutinin (HA) antigens present in the vaccine, which are approximately 19- to 90-fold higher titers at 1/50 the standard intramuscular commercial nonadjuvanted influenza vaccine dose. Seroconversion rates of 67% to 100% were achieved against each of the three viral strains present. The adjuvanted nasal influenza vaccine also produced significant cross immunity to five other H3N2 influenza virus strains not present in the vaccine and produced sterile immunity after challenge with homologous live virus. No safety issues were observed in 249 ferrets receiving the adjuvanted influenza vaccine. These findings demonstrate the ability of W(80)5EC NE to adjuvant nasally administered influenza vaccine and provide a basis for studying the intranasal W(80)5EC-adjuvanted influenza vaccine in humans.


Assuntos
Imunização/métodos , Vacinas contra Influenza/administração & dosagem , Adjuvantes Imunológicos , Administração Intranasal , Animais , Formação de Anticorpos , Reações Cruzadas/imunologia , Emulsões , Furões , Imunidade Celular , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vacinas de Produtos Inativados
9.
PLoS One ; 6(7): e21823, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21789184

RESUMO

BACKGROUND: Respiratory syncytial virus (RSV) is a leading cause of bronchiolitis and pneumonia in young children worldwide, and no vaccine is currently available. Inactivated RSV vaccines tested in the 1960's led to vaccine-enhanced disease upon viral challenge, which has undermined RSV vaccine development. RSV infection is increasingly being recognized as an important pathogen in the elderly, as well as other individuals with compromised pulmonary immunity. A safe and effective inactivated RSV vaccine would be of tremendous therapeutic benefit to many of these populations. PRINCIPAL FINDINGS: In these preclinical studies, a mouse model was utilized to assess the efficacy of a novel, nanoemulsion-adjuvanted, inactivated mucosal RSV vaccine. Our results demonstrate that NE-RSV immunization induced durable, RSV-specific humoral responses, both systemically and in the lungs. Vaccinated mice exhibited increased protection against subsequent live viral challenge, which was associated with an enhanced Th1/Th17 response. In these studies, NE-RSV vaccinated mice displayed no evidence of Th2 mediated immunopotentiation, as has been previously described for other inactivated RSV vaccines. CONCLUSIONS: These studies indicate that nanoemulsion-based inactivated RSV vaccination can augment viral-specific immunity, decrease mucus production and increase viral clearance, without evidence of Th2 immune mediated pathology.


Assuntos
Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Células Th2/imunologia , Vacinas de Produtos Inativados/imunologia , Administração Intranasal , Animais , Especificidade de Anticorpos/imunologia , Hiper-Reatividade Brônquica/complicações , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/patologia , Citocinas/biossíntese , Emulsões , Eosinofilia/complicações , Eosinofilia/imunologia , Imunidade , Imunização , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Especificidade da Espécie , Carga Viral/imunologia , Inativação de Vírus
10.
Surgery ; 148(3): 499-509, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20189619

RESUMO

BACKGROUND: Nanoemulsions are broadly antimicrobial oil-in-water emulsions containing nanometer-sized droplets stabilized with surfactants. We hypothesize that topical application of a nanoemulsion compound (NB-201) can attenuate burn wound infection. In addition to reducing infection, nanoemulsion therapy may modulate dermal inflammatory signaling and thereby lessen inflammation following thermal injury. METHODS: Male Sprague-Dawley rats underwent a 20% total body surface area scald burn to create a partial-thickness burn injury. Animals were resuscitated with Ringer's lactate solution and the wound covered with an occlusive dressing. At 8 hours after injury, the burn wound was inoculated with 1 x 10(6) colony-forming units (CFUs) of Pseudomonas aeruginosa. NB-201, NB-201 placebo, 5% mafenide acetate solution, or 0.9% saline (control) was applied onto the wound at 16 and 24 hours after burn injury. Skin was harvested 32 hours postburn for quantitative wound culture and determination of inflammatory mediators in tissue homogenates. RESULTS: NB-201 decreased mean bacterial growth in the burn wound by 1,000-fold, with only 13% (3/23) of animals having P. aeruginosa counts greater than 10(5) CFU/g tissue versus 91% (29/32) in the control group (P < .0001). Treatment with NB-201 attenuated neutrophil sequestration in the treatment group as measured by myeloperoxidase assay and by histology. It also significantly decreased levels of proinflammatory cytokines (interleukin [IL]-1beta and IL-6) and the degree of hair follicle cell apoptosis in skin compared to saline-treated controls. CONCLUSION: Topical NB-201 substantially decreased bacterial growth in a partial-thickness burn model. This decrease in the level of wound infection was associated with an attenuation of the local dermal inflammatory response and diminished neutrophil sequestration. NB-201 represents a novel potent antimicrobial and anti-inflammatory treatment for use in burn wounds.


Assuntos
Infecções Bacterianas/prevenção & controle , Queimaduras/complicações , Emulsões/uso terapêutico , Inflamação/prevenção & controle , Infecções por Pseudomonas/prevenção & controle , Infecção dos Ferimentos/prevenção & controle , Animais , Apoptose , Infecções Bacterianas/enzimologia , Infecções Bacterianas/etiologia , Biomarcadores/metabolismo , Queimaduras/etiologia , Queimaduras/patologia , Temperatura Alta/efeitos adversos , Humanos , Inflamação/enzimologia , Inflamação/etiologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Peroxidase/metabolismo , Infecções por Pseudomonas/enzimologia , Pseudomonas aeruginosa , Ratos , Ratos Sprague-Dawley , Pele/efeitos dos fármacos , Pele/patologia , Infecção dos Ferimentos/enzimologia , Infecção dos Ferimentos/etiologia
11.
Mycopathologia ; 155(4): 195-201, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12650595

RESUMO

Surfactant nanoemulsions are water in oil preparations that proved to have a broad spectrum biocidal activity against a variety of microorganisms including Gram-positive and Gram-negative bacteria, spores and enveloped viruses. These preparations are non-toxic to the skin, mucous membrane and gastrointestinal tissues at biocidal concentrations. In this study, 0.1% of the nanoemulsion designated X8W60PC has shown fungicidal activity against yeast including Candida albicans and C. tropicalis in 15 minutes. C. tropicalis was more sensitive than C. albicans, which required a longer time or a higher concentration of the nanoemulsion to achieve killing. Neutral to slightly alkaline pH was more effective in killing the yeast cells than acidic pH. Using the minimum inhibitory concentration assay, 0.08% of the nanoemulsion was inhibitory to C. albicans, and parapsilosis and filamentous fungi including Microsporum gypseum, Trichophyton mentagrophytes, Trichophyton rubrum, Aspergillus fumigatus and Fusarium oxysporum. None of the individual ingredients was as effective a fungicidal as the nanoemulsion at equivalent concentration. This shows that the nanoemulsion structure is an important factor in the anti-fungal activity. The X8W60PC has great potential as a topical anti-fungal agent and further investigation into the mechanism of fungicidal action is warranted.


Assuntos
Antifúngicos/farmacologia , Candida albicans/efeitos dos fármacos , Candida tropicalis/efeitos dos fármacos , Tensoativos/farmacologia , Candida albicans/crescimento & desenvolvimento , Candida albicans/metabolismo , Candida tropicalis/crescimento & desenvolvimento , Candida tropicalis/metabolismo , Emulsões , Fungos/efeitos dos fármacos , Fungos/crescimento & desenvolvimento , Fungos/metabolismo , Testes de Sensibilidade Microbiana , Microscopia Eletrônica
12.
J Infect Dis ; 186(9): 1222-30, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12402191

RESUMO

Influenza A viral infection begins by hemagglutinin glycoproteins on the viral envelope binding to cell membrane sialic acid (SA). Free SA monomers cannot block hemagglutinin adhesion in vivo because of toxicity. Polyvalent, generation 4 (G4) SA-conjugated polyamidoamine (PAMAM) dendrimer (G4-SA) was evaluated as a means of preventing adhesion of 3 influenza A subtypes (H1N1, H2N2, and H3N2). In hemagglutination-inhibition assays, G4-SA was found to inhibit all H3N2 and 3 of 5 H1N1 influenza subtype strains at concentrations 32-170 times lower than those of SA monomers. In contrast, G4-SA had no ability to inhibit hemagglutination with H2N2 subtypes or 2 of 5 H1N1 subtype strains. In vivo experiments showed that G4-SA completely prevented infection by a H3N2 subtype in a murine influenza pneumonitis model but was not effective in preventing pneumonitis caused by an H2N2 subtype. Polyvalent binding inhibitors have potential as antiviral therapeutics, but issues related to strain specificity must be resolved.


Assuntos
Vírus da Influenza A/patogenicidade , Influenza Humana/complicações , Pneumonia/prevenção & controle , Pneumonia/virologia , Polímeros/uso terapêutico , Ácidos Siálicos/uso terapêutico , Animais , Sequência de Carboidratos , Desenho de Fármacos , Hemaglutinação , Humanos , Vírus da Influenza A/classificação , Pulmão/efeitos dos fármacos , Pulmão/virologia , Camundongos , Modelos Moleculares , Conformação Molecular , Dados de Sequência Molecular , Poliaminas , Polímeros/síntese química , Polímeros/química , Ácidos Siálicos/síntese química , Ácidos Siálicos/química
13.
Vaccine ; 21(25-26): 3801-14, 2003 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-12922114

RESUMO

Nanoemulsion, a water-in-oil formulation stabilized by small amounts of surfactant, is non-toxic to mucous membranes and produces biocidal activity against enveloped viruses. We evaluated nanoemulsion as an adjuvant for mucosal influenza vaccines. Mice (C3H/HeNHsd strain) were vaccinated intranasally with 5 x 10(5) plaque forming units (pfu) of influenza A virus (Ann Arbor/6/60 strain) and a nanoemulsion mixture. The mice were challenged on day 21 after immunization with an intranasal lethal dose of 2 x 10(5) pfu of virus. Animals vaccinated with the influenza A/nanoemulsion mixture were completely protected against infection, while animals vaccinated with either formaldehyde-killed virus or nanoemulsion alone developed viral pneumonitis and died by day 6 after the challenge. Mice vaccinated with virus/nanoemulsion mixture had rapid cytokine responses followed by high levels of specific anti-influenza immunoglobulin G (IgG) and immunoglobulin A (IgA) antibodies. Specificity of the immune response was confirmed by assessment of the proliferation and cytokine production in splenocytes. This paper demonstrates that nanoemulsion can be employed as a non-toxic mucosal adjuvant for influenza virus vaccine.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Pneumonia/prevenção & controle , Administração Intranasal , Animais , Especificidade de Anticorpos , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/virologia , Divisão Celular/fisiologia , Citocinas/biossíntese , Emulsões , Feminino , Citometria de Fluxo , Imunidade nas Mucosas/efeitos dos fármacos , Imunidade nas Mucosas/imunologia , Imunização , Imunoglobulina A/biossíntese , Imunoglobulina G/biossíntese , Vacinas contra Influenza/toxicidade , Camundongos , Camundongos Endogâmicos C3H , Pneumonia/imunologia , RNA Viral/análise , RNA Viral/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa