Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 231
Filtrar
1.
J Infect Dev Ctries ; 18(8): 1281-1290, 2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39288397

RESUMO

INTRODUCTION: Rabies is a fatal infectious disease, that poses a major public health threat in developing countries. With an annual death toll of approximately 59,000, more than half of which are children, an urgent need exists for a safe, affordable, and effective preventive measure against rabies virus infection. METHODOLOGY: A recombinant rabies vaccine called Ad5-dRVG was constructed by introducing two copies of the rabies virus glycoprotein into a human adenoviral vector. Virus-neutralizing assays and virus challenge experiments were employed to evaluate the Ad5-dRVG vaccine. RESULTS: Our findings demonstrate that a single dose of Ad5-dRVG, administered either intramuscularly or orally, elicited significantly stronger immune responses than Ad5-RVG. Moreover, both vaccines provided complete protection in mice. Notably, the vaccine exhibited remarkable efficacy even at low doses, suggesting potential cost reduction in production. CONCLUSIONS: The development of the Ad5-dRVG recombinant rabies vaccine represents a significant advancement in rabies prevention. Its enhanced immunogenicity, demonstrated efficacy and potential cost savings make it a promising candidate for widespread use.


Assuntos
Vetores Genéticos , Glicoproteínas , Vacina Antirrábica , Vírus da Raiva , Raiva , Vacinas Sintéticas , Animais , Vacina Antirrábica/imunologia , Vacina Antirrábica/genética , Vacina Antirrábica/administração & dosagem , Raiva/prevenção & controle , Raiva/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/administração & dosagem , Glicoproteínas/imunologia , Glicoproteínas/genética , Camundongos , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Feminino , Anticorpos Antivirais/sangue , Adenoviridae/genética , Camundongos Endogâmicos BALB C , Injeções Intramusculares , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Humanos , Modelos Animais de Doenças , Administração Oral , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Eficácia de Vacinas
2.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39273091

RESUMO

Rabies is a fatal neurological infectious disease caused by rabies virus (RABV), which invades the central nervous system (CNS). RABV with varying virulence regulates chemokine expression, and the mechanisms of signaling pathway activation remains to be elucidated. The relationship between Toll-like receptors (TLRs) and immune response induced by RABV has not been fully clarified. Here, we investigated the role of TLR7 in the immune response induced by RABV, and one-way analysis of variance (ANOVA) was employed to evaluate the data. We found that different RABV strains (SC16, HN10, CVS-11) significantly increased CCL2, CXCL10 and IL-6 production. Blocking assays indicated that the TLR7 inhibitor reduced the expression of CCL2, CXCL10 and IL-6 (p < 0.01). The activation of the Myd88 pathway in BV-2 cells stimulated by RABV was TLR7-dependent, whereas the inhibition of Myd88 activity reduced the expression of CCL2, CXCL10 and IL-6 (p < 0.01). Meanwhile, the RABV stimulation of BV-2 cells resulted in TRL7-mediated activation of NF-κB and induced the nuclear translocation of NF-κB p65. CCL2, CXCL10 and IL-6 release was attenuated by the specific NF-κB inhibitor used (p < 0.01). The findings above demonstrate that RABV-induced expression of CCL2, CXCL10 and IL-6 involves Myd88 and NF-κB pathways via the TLR7 signal.


Assuntos
Fator 88 de Diferenciação Mieloide , NF-kappa B , Vírus da Raiva , Transdução de Sinais , Receptor 7 Toll-Like , Receptor 7 Toll-Like/metabolismo , Animais , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Vírus da Raiva/patogenicidade , Vírus da Raiva/imunologia , Camundongos , NF-kappa B/metabolismo , Linhagem Celular , Interleucina-6/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Quimiocina CXCL10/metabolismo , Quimiocina CXCL10/genética , Raiva/virologia , Raiva/metabolismo , Raiva/imunologia , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Inflamação/metabolismo
3.
Emerg Microbes Infect ; 13(1): 2389115, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39129566

RESUMO

Rabies is a lethal zoonotic disease that threatens human health. As the only viral surface protein, the rabies virus (RABV) glycoprotein (G) induces main neutralizing antibody (Nab) responses; however, Nab titre is closely correlated with the conformation of G. Virus-like particles (VLP) formed by the co-expression of RABV G and matrix protein (M) improve retention and antigen presentation, inducing broad, durable immune responses. RABV nucleoprotein (N) can elicit humoral and cellular immune responses. Hence, we developed a series of nucleoside-modified RABV mRNA vaccines encoding wild-type G, soluble trimeric RABV G formed by an artificial trimer motif (tG-MTQ), membrane-anchored prefusion-stabilized G (preG). Furthermore, we also developed RABV VLP mRNA vaccine co-expressing preG and M to generate VLPs, and VLP/N mRNA vaccine co-expressing preG, M, and N. The RABV mRNA vaccines induced higher humoral and cellular responses than inactivated rabies vaccine, and completely protected mice against intracerebral challenge. Additionally, the IgG and Nab titres in RABV preG, VLP and VLP/N mRNA groups were significantly higher than those in G and tG-MTQ groups. A single administration of VLP or VLP/N mRNA vaccines elicited protective Nab responses, the Nab titres were significantly higher than that in inactivated rabies vaccine group at day 7. Moreover, RABV VLP and VLP/N mRNA vaccines showed superior capacities to elicit potent germinal centre, long-lived plasma cell and memory B cell responses, which linked to high titre and durable Nab responses. In summary, our data demonstrated that RABV VLP and VLP/N mRNA vaccines could be promising candidates against rabies.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Imunidade Celular , Imunidade Humoral , Vacina Antirrábica , Vírus da Raiva , Raiva , Vacinas de Partículas Semelhantes a Vírus , Animais , Vacina Antirrábica/imunologia , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/genética , Raiva/prevenção & controle , Raiva/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Camundongos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/genética , Feminino , Vacinas de mRNA/imunologia , Camundongos Endogâmicos BALB C , Nucleosídeos/imunologia , Glicoproteínas/imunologia , Glicoproteínas/genética , Humanos , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/genética , Antígenos Virais/imunologia , Antígenos Virais/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , RNA Mensageiro/genética , RNA Mensageiro/imunologia
4.
Virol J ; 21(1): 154, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38978059

RESUMO

BACKGROUND: Rabies is a fatal zoonotic disease whose pathogenesis has not been fully elucidated, and vaccination is the only effective method for protecting against rabies virus infection. Most inactivated vaccines are produced using Vero cells, which are African green monkey kidney cells, to achieve large-scale production. However, there is a potential carcinogenic risk due to nonhuman DNA contamination. Thus, replacing Vero cells with human diploid cells may be a safer strategy. In this study, we developed a novel 2BS cell-adapted rabies virus strain and analysed its sequence, virulence and immunogenicity to determine its application potential as a human diploid cell inactivated vaccine. METHODS AND RESULTS: The 2BS cell-adapted rabies virus strain 2aG4-B40 was established by passage for 40 generations and selection of plaques in 2BS cells. RNA sequence analysis revealed that mutations in 2BS cell-adapted strains were not located at key sites that regulate the production of neutralizing antibodies or virulence in the aG strain (GQ412744.1). The gradual increase in virulence (remaining above 7.0 logLD50/ml from the 40th to 55th generation) and antigen further indicated that these mutations may increase the affinity of the adapted strains for human diploid cells. Identification tests revealed that the 2BS cell-adapted virus strain was neutralized by anti-rabies serum, with a neutralization index of 19,952. PrEP and PEP vaccination and the NIH test further indicated that the vaccine prepared with the 2aG4-B40 strain had high neutralizing antibody levels (2.24 to 46.67 IU/ml), immunogenicity (protection index 270) and potency (average 11.6 IU/ml). CONCLUSIONS: In this study, a 2BS cell-adapted strain of the 2aG4 rabies virus was obtained by passage for 40 generations. The results of sequencing analysis and titre determination of the adapted strain showed that the mutations in the adaptive process are not located at key sequence regions of the virus, and these mutations may enhance the affinity of the adapted strain for human diploid cells. Moreover, vaccines made from the adapted strain 2aG4-B40 had high potency and immunogenicity and could be an ideal candidate rabies virus strain for inactivated vaccine preparation.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vacina Antirrábica , Vírus da Raiva , Raiva , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Animais , Vacina Antirrábica/imunologia , Vacina Antirrábica/genética , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Raiva/prevenção & controle , Raiva/imunologia , Raiva/virologia , Humanos , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Chlorocebus aethiops , Virulência , Vacinas de Produtos Inativados/imunologia , Células Vero , China , Camundongos , Linhagem Celular , Mutação , Feminino , Imunogenicidade da Vacina
5.
Vaccine ; 42(22): 126018, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-38834432

RESUMO

BACKGROUND: SYN023 is an anti-rabies monoclonal antibody mixture administered as part of post-exposure prophylaxis regimens. The rabies virus neutralizing antibody (RVNA) concentration generally accepted as an adequate immune response to vaccination is ≥ 0.5 IU/mL. METHODS: Within 54 h of potential rabies exposure, 448 patients in two risk substrata of WHO Category III exposure were randomized to receive either 0.3 mg/kg SYN023 or 0.133 mL/kg human rabies immunoglobulin (HRIG) injected in and around the wound site(s) plus a course of rabies vaccination. Patients were followed for safety and absence of rabies for ≥ 365 days. RESULTS: GMT RVNA was higher with SYN023 throughout the 2-week post-treatment period. In the primary analysis group (n = 368), 99.4 % of SYN023 recipients versus 4.5 % of HRIG recipients had protective RVNA levels on Day 4. On Day 8, 98.1 % SYN023 versus 12.2 % HRIG recipients were protected. The SYN023:HRIG ratio of geometric mean titer of RVNA (RVNA GMTs) on Day 8 (19.42) exceeded the 10 % superiority margin (P < 0.0001) indicating higher Day 8 RVNA with SYN023. On Day 99, the SYN023:HRIG RVNA GMT ratio (0.66) was below the non-inferiority margin of 20 % (P = 0.9485) suggesting some moderation of vaccine immune response by SYN023 relative to HRIG. The ratio of percent SYN023:HRIG recipients achieving RVNA ≥ 0.5 IU/mL on Day 99 (0.98) met the non-inferiority margin of 20 % (P = 0.013) indicating anti-rabies immune response with SYN023 was non-inferior to HRIG despite this effect. There were no probable/confirmed rabies cases in any patient. Study regimens were well tolerated. CONCLUSIONS: SYN023 provided higher RVNA than HRIG soon after rabies exposure. By Day 99 post-treatment, GM RVNA with SYN023 was lower than HRIG, however, the percent of SYN023 recipients with a protective response was not inferior at this time point. No rabies cases were reported in the study. The SYN023 safety profile was acceptable. CLINICALTRIALS: gov ID: NCT03961555.


Assuntos
Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Profilaxia Pós-Exposição , Vacina Antirrábica , Vírus da Raiva , Raiva , Humanos , Raiva/prevenção & controle , Raiva/imunologia , Masculino , Feminino , Adulto , Anticorpos Antivirais/imunologia , Profilaxia Pós-Exposição/métodos , Pessoa de Meia-Idade , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Método Duplo-Cego , Adulto Jovem , Adolescente , Vírus da Raiva/imunologia , Vacina Antirrábica/imunologia , Vacina Antirrábica/efeitos adversos , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/uso terapêutico , Idoso , Criança
6.
Med Microbiol Immunol ; 213(1): 7, 2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38761268

RESUMO

The incidence of rabies in Thailand reached its peak in 2018 with 18 human deaths. Preexposure prophylaxis (PrEP) vaccination is thus recommended for high-risk populations. WHO has recently recommended that patients who are exposed to a suspected rabid animal and have already been immunized against rabies should receive a 1-site intradermal (ID) injection of 0.1 mL on days 0 and 3 as postexposure prophylaxis (PEP). In Thailand, village health and livestock volunteers tasked with annual dog vaccination typically receive only a single lifetime PrEP dose and subsequent boosters solely upon confirmed animal bites. However, the adequacy of a single PrEP dose for priming and maintaining immunity in this high-risk group has not been evaluated. Therefore, our study was designed to address two key questions: (1) sufficiency of single-dose PrEP-to determine whether a single ID PrEP dose provides adequate long-term immune protection for high-risk individuals exposed to numerous dogs during their vaccination duties. (2) Booster efficacy for immune maturation-to investigate whether one or two additional ID booster doses effectively stimulate a mature and sustained antibody response in this population. The level and persistence of the rabies antibody were determined by comparing the immunogenicity and booster efficacy among the vaccination groups. Our study demonstrated that rabies antibodies persisted for more than 180 days after cost-effective ID PrEP or the 1st or the 2nd single ID booster dose, and adequate antibody levels were detected in more than 95% of participants by CEE-cELISA and 100% by indirect ELISA. Moreover, the avidity maturation of rabies-specific antibodies occurred after the 1st single ID booster dose. This smaller ID booster regimen was sufficient for producing a sufficient immune response and enhancing the maturation of anti-rabies antibodies. This safe and effective PrEP regimen and a single visit involving a one-dose ID booster are recommended, and at least one one-dose ID booster regimen could be equitably implemented in at-risk people in Thailand and other developing countries. However, an adequate antibody level should be monitored before the booster is administered.


Assuntos
Anticorpos Antivirais , Imunização Secundária , Vacina Antirrábica , Raiva , Vacina Antirrábica/imunologia , Vacina Antirrábica/administração & dosagem , Raiva/prevenção & controle , Raiva/imunologia , Anticorpos Antivirais/sangue , Tailândia , Humanos , Injeções Intradérmicas , Animais , Feminino , Adulto , Masculino , Adulto Jovem , Afinidade de Anticorpos , Pessoa de Meia-Idade , Cães , Profilaxia Pré-Exposição/métodos , Adolescente , Profilaxia Pós-Exposição/métodos , Formação de Anticorpos/imunologia
7.
Microbes Infect ; 26(4): 105321, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38461968

RESUMO

Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.


Assuntos
Proteína DEAD-box 58 , Vírus da Raiva , Humanos , Linhagem Celular , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Receptores Imunológicos/metabolismo , RNA Viral/genética , Transdução de Sinais , Replicação Viral
8.
Viruses ; 13(11)2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34835093

RESUMO

Rabies is a lethal zoonotic disease caused by lyssaviruses, such as rabies virus (RABV), that results in nearly 100% mortality once clinical symptoms appear. There are no curable drugs available yet. RABV contains five structural proteins that play an important role in viral replication, transcription, infection, and immune escape mechanisms. In the past decade, progress has been made in research on the pathogenicity of RABV, which plays an important role in the creation of new recombinant RABV vaccines by reverse genetic manipulation. Here, we review the latest advances on the interaction between RABV proteins in the infected host and the applied development of rabies vaccines by using a fully operational RABV reverse genetics system. This article provides a background for more in-depth research on the pathogenic mechanism of RABV and the development of therapeutic drugs and new biologics.


Assuntos
Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Proteínas Estruturais Virais/imunologia , Animais , Humanos , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/genética , Vírus da Raiva/genética , Genética Reversa/métodos , Vacinas Atenuadas , Proteínas Estruturais Virais/genética , Replicação Viral
9.
PLoS Negl Trop Dis ; 15(6): e0009484, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34086672

RESUMO

The global spread of Zika virus (ZIKV), which caused a pandemic associated with Congenital Zika Syndrome and neuropathology in newborns and adults, prompted the pursuit of a safe and effective vaccine. Here, three kinds of recombinant rabies virus (RABV) encoding the prM-E protein of ZIKV were constructed: ZI-D (prM-E), ZI-E (transmembrane domain (TM) of prM-E replaced with RABV G) and ZI-F (signal peptide and TM domain of prM-E replaced with the region of RABV G). When the TM of prM-E was replaced with the region of RABV G (termed ZI-E), it promoted ZIKV E protein localization on the cell membrane and assembly on recombinant viruses. In addition, the change in the signal peptide with RABV G (termed ZI-F) was not conducive to foreign protein expression. The immunogenicity of recombinant viruses mixed with a complex adjuvant of ISA 201 VG and poly(I:C) was tested in BALB/c mice. After immunization with ZI-E, the anti-ZIKV IgG antibody lasted for at least 10 weeks. The titers of neutralizing antibodies (NAbs) against ZIKV and RABV at week 6 were all greater than the protective titers. Moreover, ZI-E stimulated the proliferation of splenic lymphocytes and promoted the secretion of cytokines. It also promoted the production of central memory T cells (TCMs) among CD4+/CD8+ T cells and stimulated B cell activation and maturation. These results indicate that ZI-E could induce ZIKV-specific humoral and cellular immune responses, which have the potential to be developed into a promising vaccine for protection against both ZIKV and RABV infections.


Assuntos
Vírus da Raiva/genética , Raiva/prevenção & controle , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Animais , Anticorpos Antivirais/imunologia , Feminino , Humanos , Imunidade Celular , Camundongos , Camundongos Endogâmicos BALB C , Raiva/imunologia , Raiva/virologia , Vírus da Raiva/imunologia , Linfócitos T/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Zika virus/genética , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
10.
Front Immunol ; 12: 622516, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679766

RESUMO

Rabies virus (RABV) is able to reach the central nervous system (CNS) without triggering a strong immune response, using multiple mechanisms to evade and suppress the host immune system. After infection via a bite or scratch from a rabid animal, RABV comes into contact with macrophages, which are the first antigen-presenting cells (APCs) that are recruited to the area and play an essential role in the onset of a specific immune response. It is poorly understood how RABV affects macrophages, and if the interaction contributes to the observed immune suppression. This study was undertaken to characterize the interactions between RABV and human monocyte-derived macrophages (MDMs). We showed that street RABV does not replicate in human MDMs. Using a recombinant trimeric RABV glycoprotein (rRABV-tG) we showed binding to the nicotinic acetylcholine receptor alpha 7 (nAChr α7) on MDMs, and confirmed the specificity using the nAChr α7 antagonist alpha-bungarotoxin (α-BTX). We found that this binding induced the cholinergic anti-inflammatory pathway (CAP), characterized by a significant decrease in tumor necrosis factor α (TNF-α) upon LPS challenge. Using confocal microscopy we found that induction of the CAP is associated with significant cytoplasmic retention of nuclear factor κB (NF-κB). Co-cultures of human MDMs exposed to street RABV and autologous T cells further revealed that the observed suppression of MDMs might affect their function as T cell activators as well, as we found a significant decrease in proliferation of CD8+ T cells and an increased production of the anti-inflammatory cytokine IL-10. Lastly, using flow cytometric analysis we observed a significant increase in expression of the M2-c surface marker CD163, hinting that street RABV might be able to affect macrophage polarization. Taken together, these results show that street RABV is capable of inducing an anti-inflammatory state in human macrophages, possibly affecting T cell functioning.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Macrófagos/imunologia , Vírus da Raiva/fisiologia , Raiva/imunologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Anti-Inflamatórios , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Diferenciação Celular , Células Cultivadas , Colinérgicos , Técnicas de Cocultura , Humanos , Interleucina-10/metabolismo , Ativação Linfocitária , NF-kappa B/metabolismo , Neuroimunomodulação , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Células Th2/imunologia
11.
J Immunol Methods ; 492: 112939, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33309752

RESUMO

The potency of all modern tissue culture human rabies vaccines is measured based on the National Institute of Health (NIH) potency test that is laborious, time-consuming, involves large test variations and requires sacrifice of large number of animals. To circumvent these limitations, several researchers and WHO expert working groups have discussed development of alternative in vitro methods to replace the NIH potency test. Although several immunochemical methods have been proposed to quantify rabies glycoprotein (G-protein) using multiple murine monoclonal antibodies, we report an In vitro competitive inhibition ELISA (CIA) method based on the use of a neutralizing rabies glycoprotein site III directed novel therapeutic human rabies monoclonal antibody (RAB1) that shows equivalence to the mice NIH potency test in recognition of neutralization site of the glycoprotein. In vitro potency testing of WHO 7th International Standard for rabies vaccine (IS) by CIA using RAB1 and In-house reference standard (IHRS) as a standard to assess its suitability for the assessment of validation parameters showed accurate and precise values with <15% coefficient variance. The method was validated using 5PL standard curve with linearity r2 > 0.98 and LLOQ of 0.125 IU/mL indicating sensitivity of the method. The method was found to be precise, robust and accurate to quantitate intact rabies glycoprotein in final vaccine and showed a strong correlation (Pearson's r = 0.81) with the NIH potency values of licensed Vero cell rabies vaccine. The CIA test using RAB1 was able to accurately quantitate degradation of rabies vaccine and assess loss in antigenicity of lyophilized and reconstituted liquid rabies vaccine under thermal stress conditions. The method was able to differentiate between potent and reduced potency vaccine samples. The new in vitro competitive inhibition ELISA method using RAB1 thus can be a valid alternative to the NIH test.


Assuntos
Antígenos Virais/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos Virais/administração & dosagem , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática/métodos , Mapeamento de Epitopos/métodos , Humanos , Profilaxia Pós-Exposição/métodos , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/administração & dosagem , Potência de Vacina , Células Vero , Proteínas do Envelope Viral/administração & dosagem
12.
J Vis Exp ; (160)2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32658185

RESUMO

Functional rabies surveillance systems are crucial to provide reliable data and increase the political commitment necessary for disease control. To date, animals suspected as rabies-positive must be submitted to a postmortem confirmation using classical or molecular laboratory methods. However, most endemic areas are in low- and middle-income countries where animal rabies diagnosis is restricted to central veterinary laboratories. Poor availability of surveillance infrastructure leads to serious disease underreporting from remote areas. Several diagnostic protocols requiring low technical expertise have been recently developed, providing opportunity to establish rabies diagnosis in decentralized laboratories. We present here a complete protocol for field postmortem diagnosis of animal rabies using a rapid immunochromatographic diagnostic test (RIDT), from brain biopsy sampling to the final interpretation. We complete the protocol by describing a further use of the device for molecular analysis and viral genotyping. RIDT easily detects rabies virus and other lyssaviruses in brain samples. The principle of such tests is simple: brain material is applied on a test strip where gold conjugated antibodies bind specifically to rabies antigens. The antigen-antibody complexes bind further to fixed antibodies on the test line, resulting in a clearly visible purple line. The virus is inactivated in the test strip, but viral RNA can be subsequently extracted. This allows the test strip, rather than the infectious brain sample, to be safely and easily sent to an equipped laboratory for confirmation and molecular typing. Based on a modification of the manufacturer's protocol, we found increased test sensitivity, reaching 98% compared to the gold standard reference method, the direct immunofluorescence antibody test. The advantages of the test are numerous: rapid, easy-to-use, low cost and no requirement for laboratory infrastructure, such as microscopy or cold-chain compliance. RIDTs represent a useful alternative for areas where reference diagnostic methods are not available.


Assuntos
Testes Diagnósticos de Rotina/métodos , Vírus da Raiva/imunologia , Raiva/imunologia , Animais , Diagnóstico , Imunoensaio , Raiva/veterinária
13.
Vet Microbiol ; 242: 108578, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32122589

RESUMO

Rabies is a highly lethal infectious zoonosis caused by rabies virus (RABV), and the mortality rate is almost 100 % once clinical symptoms appear, which poses a huge threat to public health security across the many parts of the word. Vaccination is reported to be the most effective approach to prevent the disease. G protein is the only protein present on the surface of RABV, it also could induce humoral immunity to produce virus neutralizing antibodies (VNA) and stimulate T cells to produce cellular immunity. Adeno-associated viruses (AAVs) have been used as vectors for gene therapy of different human diseases for its low immunogenicity, high safety and long-term stable expression. To develop a safe and effective vaccine, recombinant AAVs containing different kind of G gene were constructed. After intramuscular (i.m.) immunization in mice, all of these rAAV-G vaccines could induce the production of high levels of VNA and effective cellular immune response. Consistently, all of the rAAV-G vaccines could provide protection against lethal RABV challenge. Our results shown that the rAAV-G vaccines could be potential candidates used in the control of RABV infection. In addition, rAAV-G as a vaccine has many advantages of low preparation cost, simple storage and transportation conditions (4 °C storage and transportation), simple immunization program (only one immunization) and so on. Thence, the rAAV-G vaccines could be potential candidates used in the control of RABV infection.


Assuntos
Imunidade Celular , Imunidade Humoral , Glicoproteínas de Membrana/imunologia , Vacina Antirrábica/imunologia , Raiva/prevenção & controle , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Dependovirus/genética , Dependovirus/imunologia , Feminino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Raiva/imunologia , Vacina Antirrábica/genética , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Vacinação , Proteínas Virais/genética , Proteínas Virais/imunologia
14.
Viral Immunol ; 33(5): 367-377, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32159457

RESUMO

Antibody induction test (AIT) is a promising candidate as a refinement of the troublesome National institutes of Health (NIH) test in the sense of animal welfare 3R approach for determination of potency of inactivated rabies vaccines for veterinary and human use. In this study, we initially try to develop AIT as a suitable alternative to NIH test, to achieve a reduction of test duration and diminish animal suffering by omitting intracerebral CVS infection and measuring humoral immunity upon vaccination. Designs of both multi-dose and single-dose AIT were examined. Biological reference preparation, batch 5 with assigned titer of 10 IU/vial, was taken as both standard and test vaccine. Six consecutive AITs were performed and eight pools of sera in each AIT were tested in triplicate by rapid fluorescent focus inhibition test. We estimated the upper detection limit and calculated test variability for individual dilutions. For multi-dose AIT, we estimated the dose-response function and performed calculations of final test results and statistical validity parameters for both linear and sigmoidal model using CombiStats program. Sigmoidal 4-parameter dose-response model was found optimal. Presented design of multi-dose AIT showed a satisfactory detection limit for testing of inactivated rabies vaccines for both veterinary and human use. However, due to nonconformity of obtained results with statistical validity criteria, we concluded that the presented model of multi-dose AIT was unsuitable for introduction in routine practice. However, we concluded that there was a realistic option for introduction of two versions of single-dose AIT. The first version would be with two standard vaccine controls and could be introduced immediately, while the second version would include testing of the sample only and rely on comparison of the induced rabies antibody level with absolute cut-off limits set in advance.


Assuntos
Alternativas ao Uso de Animais/métodos , Anticorpos Antivirais/análise , Imunogenicidade da Vacina , Vacina Antirrábica/imunologia , Raiva/imunologia , Alternativas ao Uso de Animais/normas , Animais , Anticorpos Neutralizantes/análise , Linhagem Celular Tumoral , Feminino , Fluorescência , Masculino , Camundongos , National Institutes of Health (U.S.) , Neuroblastoma , Raiva/prevenção & controle , Testes Sorológicos , Estados Unidos , Vacinas de Produtos Inativados/imunologia
15.
J Wildl Dis ; 56(1): 224-228, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31567036

RESUMO

Oral rabies vaccination is the principal strategy used to control rabies in wildlife. No oral rabies vaccine is licensed for small Indian mongooses (Herpestes auropunctatus). The Ontario Rabies Vaccine Bait (ONRAB) is a human adenovirus type-5 rabies glycoprotein recombinant vaccine licensed for rabies control in striped skunks (Mephitis mephitis) in Canada and is under experimental evaluation in the US. We evaluated varying doses of ONRAB vaccine by direct instillation into the oral cavity with three groups of 10 mongooses: Group 1 received 109.5 TCID50, group 2 received 108.8 TCID50, and group 3 received 108.5 TCID50 of vaccine. Six control mongooses were sham-vaccinated with culture media. We collected a serum sample prior to vaccination and on days 14 and 30 postvaccination (PV). We quantified the level of rabies virus neutralizing antibodies (RVNA) from mongoose sera and compared titers among vaccinated groups and time points PV, where values greater than or equal to 0.1 IU/mL were considered positive. On day 14 PV, 87% (26 of 30, 95% confidence interval 70-95%) of vaccinates had seroconverted, whereas all vaccinates demonstrated RVNA by day 30 PV. There was a marginal effect of vaccine dose on group means of log-transformed RVNA titers at day 14 PV (F=2.5, P=0.099), but not day 30 PV. Sham-vaccinated animals were seronegative during all time points.


Assuntos
Anticorpos Antivirais/sangue , Herpestidae/sangue , Vacina Antirrábica/imunologia , Raiva/veterinária , Administração Oral , Animais , Feminino , Masculino , Raiva/imunologia , Raiva/prevenção & controle , Vacina Antirrábica/administração & dosagem
16.
Emerg Microbes Infect ; 8(1): 1584-1592, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31682199

RESUMO

The genetic and/or antigenic differences between street rabies virus (RABV) and vaccine strains could potentially affect effectiveness of rabies vaccines. As such, it is important to continue monitoring the glycoprotein (G) of the street isolates. All RABVG sequences in public database were retrieved and analysed. Using a pseudovirus system, we investigated 99 naturally occurring mutants for their reactivities to well-characterized neutralizing monoclonal antibodies (mAbs) and vaccine-induced antisera. A divergence in G sequences was found between vaccine strains and recent street isolates, with mutants demonstrating resistance to neutralizing mAbs and vaccine-induced antibodies. Moreover, antigenic variants were observed in a wide range of animal hosts and geographic locations, with most of them emerging since 2010. As the number of antigenic variants has increased in recent years, close monitoring on street isolates should be strengthened.


Assuntos
Variação Antigênica , Vírus da Raiva/imunologia , Raiva/virologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Feminino , Cobaias , Humanos , Testes de Neutralização , Raiva/imunologia , Raiva/prevenção & controle , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vírus da Raiva/química , Vírus da Raiva/genética , Vírus da Raiva/isolamento & purificação , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
17.
PLoS Negl Trop Dis ; 13(11): e0007800, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31725816

RESUMO

B cell activating factor (BAFF) is a member of the tumor necrosis factor (TNF) superfamily of cytokines that links innate with adaptive immunity. BAFF signals through receptors on B cells, making it an attractive molecule to potentiate vaccine-induced B cell responses. We hypothesized that a rabies virus (RABV)-based vaccine displaying both antigen and BAFF on the surface of the same virus particle would target antigen-specific B cells for activation and improve RABV-specific antibody responses. To test this hypothesis, we constructed a recombinant RABV-based vector expressing virus membrane-anchored murine BAFF (RABV-ED51-mBAFF). BAFF was incorporated into the RABV particle and determined to be biologically functional, as demonstrated by increased B cell survival of primary murine B cells treated ex-vivo with RABV-ED51-mBAFF. B cell survival was inhibited by pre-treating RABV-ED51-mBAFF with an antibody that blocks BAFF functions. RABV-ED51-mBAFF also activated primary murine B cells ex-vivo more effectively than RABV as shown by significant upregulation of CD69, CD40, and MHCII on the surface of infected B cells. In-vivo, RABV-ED51-mBAFF induced significantly faster and higher virus neutralizing antibody (VNA) titers than RABV while not adversely affecting the longevity of the vaccine-induced antibody response. Since BAFF was incorporated into the virus particle and genome replication was not required for BAFF expression in-vivo, we hypothesized that RABV-ED51-mBAFF would be effective as an inactivated vaccine. Mice immunized with 250 ng/mouse of ß-propriolactone-inactivated RABV-ED51-mBAFF showed faster and higher anti-RABV VNA titers compared to mice immunized with inactivated RABV. Together, this model stands as a potential foundation for exploring other virus membrane-anchored molecular adjuvants to make safer, more effective inactivated RABV-based vaccines.


Assuntos
Formação de Anticorpos/imunologia , Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Vacina Antirrábica/imunologia , Raiva/imunologia , Raiva/prevenção & controle , Vírion/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Antivirais/sangue , Citocinas/metabolismo , Feminino , Imunização , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Vacina Antirrábica/efeitos adversos , Vírus da Raiva/genética , Vírus da Raiva/crescimento & desenvolvimento , Vírus da Raiva/imunologia , Vacinação , Vacinas Atenuadas/imunologia , Vacinas de Produtos Inativados/efeitos adversos , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas
18.
Vaccine ; 37(47): 6951-6961, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31047679

RESUMO

A variety of Good Manufacturing Practice (GMP) compliant processes have been reported for production of non-replicating adenovirus vectors, but important challenges remain. Most clinical development of adenovirus vectors now uses simian adenoviruses or rare human serotypes, whereas reported manufacturing processes mainly use serotypes such as AdHu5 which are of questionable relevance for clinical vaccine development. Many clinically relevant vaccine transgenes interfere with adenovirus replication, whereas most reported process development uses selected antigens or even model transgenes such as fluorescent proteins which cause little such interference. Processes are typically developed for a single adenovirus serotype - transgene combination, requiring extensive further optimization for each new vaccine. There is a need for rapid production platforms for small GMP batches of non-replicating adenovirus vectors for early-phase vaccine trials, particularly in preparation for response to emerging pathogen outbreaks. Such platforms must be robust to variation in the transgene, and ideally also capable of producing adenoviruses of more than one serotype. It is also highly desirable for such processes to be readily implemented in new facilities using commercially available single-use materials, avoiding the need for development of bespoke tools or cleaning validation, and for them to be readily scalable for later-stage studies. Here we report the development of such a process, using single-use stirred-tank bioreactors, a transgene-repressing HEK293 cell - promoter combination, and fully single-use filtration and ion exchange components. We demonstrate applicability of the process to candidate vaccines against rabies, malaria and Rift Valley fever, each based on a different adenovirus serotype. We compare performance of a range of commercially available ion exchange media, including what we believe to be the first published use of a novel media for adenovirus purification (NatriFlo® HD-Q, Merck). We demonstrate the need for minimal process individualization for each vaccine, and that the product fulfils regulatory quality expectations. Cell-specific yields are at the upper end of those previously reported in the literature, and volumetric yields are in the range 1 × 1013 - 5 × 1013 purified virus particles per litre of culture, such that a 2-4 L process is comfortably adequate to produce vaccine for early-phase trials. The process is readily transferable to any GMP facility with the capability for mammalian cell culture and aseptic filling of sterile products.


Assuntos
Adenovirus dos Símios/imunologia , Vetores Genéticos/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linhagem Celular , Células HEK293 , Humanos , Raiva/imunologia , Vacina Antirrábica/imunologia , Sorogrupo , Transgenes/imunologia , Replicação Viral/imunologia
19.
Biomed Res Int ; 2019: 4518163, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31008105

RESUMO

Inactivation of rabies virus is essential for rabies vaccine preparation where the inactivating compound that is currently recommended for rabies vaccine preparation is ß-propiolactone (ß-PL). This compound is considered better than phenol and formalin but it is expensive and potentially carcinogenic. Data revealed that Ascorbic acid (AA) with cupric ions could yield complete and irreversible inactivation of rabies virus without adversely affecting its antigenicity. Additionally, the results of testing the vaccine potency with the selected inactivating compounds were comparable (P<0.05), and ED50 was higher than the recommended World Health Organization (WHO) limits. The use of HemaGel (plasma substitute) for testing vaccine stabilization was compared with the currently used vaccine stabilizers (human albumin and lactose). HemaGel yielded better stability than the other tested stabilizers. Monitoring of cellular and humoral immune responses indicated that both the total IgG level against rabies vaccine and the IFN and IL5 levels obtained with the HemaGel-stabilized vaccines were higher than those obtained with human albumin- and lactose-stabilized vaccine candidates.


Assuntos
Imunogenicidade da Vacina/efeitos dos fármacos , Propiolactona/farmacologia , Vacina Antirrábica/farmacologia , Raiva/prevenção & controle , Albuminas/farmacologia , Animais , Anticorpos Antivirais/efeitos dos fármacos , Anticorpos Antivirais/imunologia , Ácido Ascórbico/farmacologia , Chlorocebus aethiops , Humanos , Imunoglobulina G/imunologia , Interferons/imunologia , Interleucina-5 , Lactose/química , Propiolactona/química , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/química , Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Potência de Vacina , Células Vero/virologia
20.
Biologicals ; 59: 56-61, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30898479

RESUMO

The Rapid Fluorescent Focus Inhibition Test (RFFIT) is a standard assay used to detect and assess the titers of rabies virus neutralizing antibodies (RVNA) in blood sera. To simplify the multistep RFFIT procedure by eliminating the immunostaining step, we generated a new recombinant RV expressing a green fluorescent protein (rRV-GFP) and assess its suitability for quantifying RVNA. We rescued the rRV-GFP virus from plasmid DNA carrying a full-length genome of the CVS-N2c strain of RV in which the eGFP gene was inserted between the glycoprotein and RNA-polymerase genes. The recombinant virus was genetically stable and grew efficiently in appropriate cells expressing sufficient GFP fluorescence to detect directly 20 h post infection (hpi). We evaluated the feasibility of using rRV-GFP in RFFIT by comparing RVNA titers in 27 serum samples measured by conventional RFFIT and RFFIT-GFP. A linear regression analysis of the data demonstrated a good agreement between these two methods (r = 0.9776) including results with samples having RVNA titers close to the minimally acceptable vaccine potency threshold (0.5 IU/ml). Study results showed that the rRV-GFP virus could replace the CVS-11 challenge virus currently used in the conventional RFFIT and enabling more rapid, simpler, and less expensive detection and quantitation of RVNA.


Assuntos
Anticorpos Neutralizantes/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Animais , Anticorpos Neutralizantes/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Fluorescência , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Cobaias , Humanos , Medições Luminescentes/métodos , Camundongos , Testes de Neutralização , Coelhos , Raiva/prevenção & controle , Raiva/virologia , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/genética , Vírus da Raiva/metabolismo , Recombinação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA