首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
A dosage equal to or greater than approximately 3.4 Dex (decimal exponent, log(10)) weanling mouse intracerebral 50% lethal dose (LD(50)) was sufficient to elicit a yellow fever antibody response, as determined by the plaque neutralization (PN) test, in better than 90% of vaccinated rhesus monkeys. Lower dosages were progressively less effective in terms of PN titers and the PN and hemagglutination-inhibition serological conversion rates observed. A dose of between 3.4 and 4.2 Dex weanling mouse intracerebral LD(50), or one-tenth to one times the dosage recommended for man, provided an optimal antibody response in monkeys. In rhesus monkeys, in contrast to the findings for man, pre-existing yellow fever antibody did not interfere with the antibody response to yellow fever vaccine. The PN test was felt to be a more sensitive and specific indicator of yellow fever antibody in rhesus monkeys after vaccination than the hemagglutination inhibition or complement fixation tests.  相似文献   

2.
To optimize the thermostability of lyophilized 17D vaccine, the authors investigated parameters important for the freeze-drying process. Six different stabilizers with different sugars and amino acids were analysed in a freeze-thaw cycle for their crystallization characteristics and their stabilizing effect under thermal treatment conditions of 37 degrees C for 28 days. This test indicated that three out of six stabilizers (B, C, F) kept the vaccine significantly more stable than the three others (A, D, E). Under storing conditions of 4 degrees C over 96 days stabilizers A, B and C produced the lowest decrease in titre of about 10% in contrast to stabilizers D, E and F with a higher decrease in infectivity titre. Analysing the stability of the 17D vaccine using five different reconstitution solutions, we found that 90% D2O shows the best stabilizing effect under thermal treatment of 37 degrees C up to 24 h.  相似文献   

3.
含2A片段的重组黄热病毒17D疫苗表达载体的构建   总被引:1,自引:0,他引:1  
黄热疫苗是一种减毒的黄热病毒17D(YF-17D)活疫苗,是现有疫苗中最安全、最有效的疫苗之一,适于发展为疫苗载体。用RT-PCR法扩增出覆盖YF-17D全长基因组的3个cDNA片段:5′cDNA(A)、3′cDNA(B)和中间cDNA(C),同时引入SP6增强子序列、酶切位点和重复序列。顺序将A和B同E.coli-yeast穿梭质粒pRS424连接,再与C共转染酵母菌,利用缺少色氨酸和尿嘧啶的选择性固体培养基筛选出含YF-17D全长基因组的cDNA质粒。以该质粒为模板,经过DNA重组和酵母同源重组,获得含有口蹄疫病毒蛋白水解酶2A片段的重组YF-17D表达载体。将该表达载体体外转录后,电击转染BHK-21细胞。间接免疫荧光检测结果表明,RNA转录体在BHK-21细胞中进行了稳定的表达;滴度测定与形态学观察结果表明,重组病毒在细胞中的生长曲线等特征同母本YF-17D十分相似。结果提示,利用酵母菌同源重组在2A部位引入异种抗原基因,重组YF-17D表达载体pRS-YF-2A1具有成为高效活疫苗表达载体的潜力。  相似文献   

4.
After failed attempts at producing bacteria-based vaccines, the discovery of a viral agent causing yellow fever and its isolation in monkeys opened new avenues of research. Subsequent advances were the attenuation of the virus in mice and later in tissue culture; the creation of the seed lot system to avoid spontaneous mutations; the ability to produce the vaccine on a large scale in eggs; and the removal of dangerous contaminants. An important person in the story is Max Theiler, who was Professor of Epidemiology and Public Health at Yale from 1964-67, and whose work on virus attenuation created the modern vaccine and earned him the Nobel Prize.  相似文献   

5.
The yellow fever (YF) 17D vaccine is one of the most effective human vaccines ever created. The YF vaccine has been produced since 1937 in embryonated chicken eggs inoculated with the YF 17D virus. Yet, little information is available about the infection mechanism of YF 17DD virus in this biological model. To better understand this mechanism, we infected embryos of Gallus gallus domesticus and analyzed their histopathology after 72 hours of YF infection. Some embryos showed few apoptotic bodies in infected tissues, suggesting mild focal infection processes. Confocal and super-resolution microscopic analysis allowed us to identify as targets of viral infection: skeletal muscle cells, cardiomyocytes, nervous system cells, renal tubular epithelium, lung parenchyma, and fibroblasts associated with connective tissue in the perichondrium and dermis. The virus replication was heaviest in muscle tissues. In all of these specimens, RT-PCR methods confirmed the presence of replicative intermediate and genomic YF RNA. This clearer characterization of cell targets in chicken embryos paves the way for future development of a new YF vaccine based on a new cell culture system.  相似文献   

6.
7.
8.
Arboviruses such as yellow fever virus (YFV) are transmitted between arthropod vectors and vertebrate hosts. While barriers limiting arbovirus population diversity have been observed in mosquitoes, whether barriers exist in vertebrate hosts is unclear. To investigate whether arboviruses encounter bottlenecks during dissemination in the vertebrate host, we infected immunocompetent mice and immune-deficient mice lacking alpha/beta interferon (IFN-α/β) receptors (IFNAR−/− mice) with a pool of genetically marked viruses to evaluate dissemination and host barriers. We used the live attenuated vaccine strain YFV-17D, which contains many mutations compared with virulent YFV. We found that intramuscularly injected immunocompetent mice did not develop disease and that viral dissemination was restricted. Conversely, 32% of intramuscularly injected IFNAR−/− mice developed disease. By following the genetically marked viruses over time, we found broad dissemination in IFNAR−/− mice followed by clearance. The patterns of viral dissemination were similar in mice that developed disease and mice that did not develop disease. Unlike our previous results with poliovirus, these results suggest that YFV-17D encounters no major barriers during dissemination within a vertebrate host in the absence of the type I IFN response.  相似文献   

9.
Attenuated yellow fever (YF) virus 17D/17DD vaccines are the only available protection from YF infection, which remains a significant source of morbidity and mortality in the tropical areas of the world. The attenuated YF virus vaccine, which is used worldwide, generates both long-lasting neutralizing antibodies and strong T-cell responses. However, on rare occasions, this vaccine has toxic side effects that can be fatal. This study presents the design of two non-viral DNA-based antigen formulations and the characterization of their expression and immunological properties. The two antigen formulations consist of DNA encoding the full-length envelope protein (p/YFE) or the full-length envelope protein fused to the lysosomal-associated membrane protein signal, LAMP-1 (pL/YFE), aimed at diverting antigen processing/presentation through the major histocompatibility complex II precursor compartments. The immune responses triggered by these formulations were evaluated in H2b and H2d backgrounds, corresponding to the C57Bl/6 and BALB/c mice strains, respectively. Both DNA constructs were able to induce very strong T-cell responses of similar magnitude against almost all epitopes that are also generated by the YF 17DD vaccine. The pL/YFE formulation performed best overall. In addition to the T-cell response, it was also able to stimulate high titers of anti-YF neutralizing antibodies comparable to the levels elicited by the 17DD vaccine. More importantly, the pL/YFE vaccine conferred 100% protection against the YF virus in intracerebrally challenged mice. These results indicate that pL/YFE DNA is an excellent vaccine candidate and should be considered for further developmental studies.  相似文献   

10.
Here we describe a novel vaccine vector for expressing human immunodeficiency virus (HIV) antigens. We show that recombinant attenuated yellow fever vaccine virus 17D expressing simian immunodeficiency virus SIVmac239 Gag sequences can be used as a vector to generate SIV-specific CD8+ T-cell responses in the rhesus macaque. Priming with recombinant BCG expressing SIV antigens increased the frequency of these SIV-specific CD8+ T-cell responses after recombinant YF17D boosting. These recombinant YF17D-induced SIV-specific CD8+ T cells secreted several cytokines, were largely effector memory T cells, and suppressed viral replication in CD4+ T cells.None of the vaccine regimens tested in human immunodeficiency virus (HIV) vaccine efficacy trials to date have either reduced the rate of HIV infection or reduced the level of HIV replication. Structural features and the enormous variability of the envelope glycoprotein have frustrated efforts to induce broadly reactive neutralizing antibodies against HIV (10). Investigators have therefore focused their attention on T-cell-based vaccines (40). Simian immunodeficiency virus (SIV) challenge of rhesus macaques vaccinated with T-cell-based vaccines has shown that it is possible to control virus replication after SIV infection (22, 41, 42). The recent STEP trial of a recombinant Ad5-vectored vaccine was widely seen as an important test of this concept (http://www.hvtn.org/media/pr/step111307.html) (9, 25). Unfortunately, vaccinees became infected at higher rates than the controls (9). While it is still not clear what caused the enhanced infection rate in the vaccinated group, future Ad5-based human vaccine trials may be difficult to justify. We therefore need to develop new vaccine vectors for delivering SIV and HIV genes. Several other viral vectors currently under consideration include nonreplicating adenovirus (Ad)-based vectors (1, 21, 22), Venezuelan equine encephalitis (VEE) virus (12, 20), adeno-associated virus (AAV) (19), modified vaccinia virus Ankara (MVA) (3, 4, 13, 15, 18, 38), NYVAC (6), cytomegalovirus (CMV) (16), and replicating Ad (30). However, only a few of these have shown promise in monkey trials using rigorous SIV challenges.We explored whether the small (11-kb) yellow fever vaccine flavivirus 17D (YF17D) might be a suitable vector for HIV vaccines. The YF17D vaccine is inexpensive, production and quality control protocols already exist, and it disseminates widely in vivo after a single dose (27). Importantly, methods for the manipulation of the YF17D genome were recently established (7, 8, 24, 28). This effective vaccine has been safely used on >400 million people in the last 70 years (27). Additionally, the YF17D strain elicits robust CD8+ T-cell responses in humans (26). Chimeric YF17D is presently being developed as a vaccine for other flaviviruses, such as Japanese encephalitis virus (28), dengue virus (14), and West Nile virus (29). Inserts expressing a malaria B-cell epitope have been engineered into the E protein of YF17D (7). In murine models, recombinant YF17D viruses have generated robust and specific responses to engineered antigens inserted between the 2B and NS3 proteins in vivo (24, 35).We first used the YF17D vaccine virus to infect four Mamu-A*01-positive macaques. The vaccine virus replicated in these four animals and induced neutralizing antibodies in all four macaques by 2 weeks postvaccination (Fig. 1A and B). To monitor the CD8+ T-cell immune response against YF17D, we scanned its proteome for peptides that might bind to Mamu-A*01 using the major histocompatibility complex (MHC) pathway algorithm (31). We synthesized the 52 YF17D-derived peptides most likely to bind to Mamu-A*01 based on their predicted affinity for this MHC class I molecule. We then used a gamma interferon (IFN-γ) enzyme-linked immunospot (ELISPOT) assay to screen these peptides in YF17D-immunized animals at several time points after vaccination and discovered that four Mamu-A*01-binding peptides, LTPVTMAEV (LV91285-1293), VSPGNGWMI (VI93250-3258), MSPKGISRM (MM92179-2187), and TTPFGQQRVF (TF102853-2862), were recognized in vivo (Fig. (Fig.1C).1C). Using a previously reported protocol (26), we also observed CD8+ T-cell activation in all four animals (Fig. 1D and E). Thus, as was observed previously, the YF17D vaccine virus replicates in Indian rhesus monkeys (36) and induces neutralizing antibodies, yellow fever 17D-specific Mamu-A*01-restricted CD8+ T-cell responses, and CD8+ T-cell activation.Open in a separate windowFIG. 1.YF17D replicates and induces neutralizing antibodies, virus-specific CD8+ T cells, and the activation of CD8+ T cells in rhesus macaques. (A) Replication of YF17D during the first 10 days after vaccination with two different doses, as measured by quantitative PCR (Q-PCR) using the following primers: forward primer YF-17D 10188 (5′-GCGGATCACTGATTGGAATGAC-3′), reverse primer YF-17D 10264 (5′-CGTTCGGATACGATGGATGACTA-3′), and probe 6-carboxyfluorescein (6Fam)-5′-AATAGGGCCACCTGGGCCTCCC-3′-6-carboxytetramethylrhodamine (TamraQ). (B) Titer of neutralizing antibodies determined at 2 and 5 weeks after YF17D vaccination. (C) Fresh PBMC from vaccinees (100,000 cells/well) were used in IFN-γ ELISPOT assays (41) to assess T-cell responses against YF17D. We used 4 epitopes (LTPVTMAEV [LV91285-1293], VSPGNGWMI [VI93250-3258], MSPKGISRM [MM92179-2187], and TTPFGQQRVF [TF102853-2862]) predicted to bind to Mamu-A*01 as defined by the MHC pathway algorithm (31). All IFN-γ ELISPOT results were considered positive if they were ≥50 SFC/106 PBMC and ≥2 standard deviations over the background. (D) Identification of activated CD8+ T cells after vaccination with YF17D based on the expression of the proliferation and proapoptotic markers Ki-67 and Bcl-2, respectively (26). We stained whole blood cells with antibodies against CD3 and CD8. We then permeabilized and subsequently labeled these cells with Bcl-2- and Ki-67-specific antibodies. The flow graphs were gated on CD3+ CD8+ lymphocytes. (E) Expression kinetics of Ki-67 and Bcl-2 in CD8+ T cells after vaccination with YF17D.We next engineered the YF17D vaccine virus to express amino acids 45 to 269 of SIVmac239 Gag (rYF17D/SIVGag45-269) by inserting a yellow fever codon-optimized sequence between the genes encoding the viral proteins E and NS1. This recombinant virus replicated and induced neutralizing antibodies in mice (data not shown). We then tested the rYF17D/SIVGag45-269 construct in six Mamu-A*01-positive Indian rhesus macaques. We found evidence for the viral replication of rYF17D/SIVGag45-269 for five of these six macaques (Fig. (Fig.2A).2A). However, neutralizing antibodies were evident for all six animals at 2 weeks postvaccination (Fig. (Fig.2B).2B). Furthermore, all animals developed SIV-specific CD8+ T cells after a single immunization with rYF17D/SIVGag45-269 (Fig. (Fig.2C).2C). To test whether a second dose of this vaccine could boost virus-specific T-cell responses, we administered rYF17D/SIVGag45-269 (2.0 × 105 PFU) to four macaques on day 28 after the first immunization and monitored cellular immune responses. With the exception of animal r04091, the rYF17D/SIVGag45-269 boost did not increase the frequency of the vaccine-induced T-cell responses. This recombinant vaccine virus also induced CD8+ T-cell activation in the majority of the vaccinated animals (Fig. (Fig.2D2D).Open in a separate windowFIG. 2.rYF17D/SIVGag45-269 replicates and induces neutralizing antibodies, virus-specific CD8+ T cells, and the activation of CD8+ T cells in rhesus macaques. (A) Replication of rYF17D/SIVGag45-269 during the first 10 days after vaccination with two different doses as measured by Q-PCR using the YF17D-specific primers described in the legend of Fig. Fig.1.1. (B) Titer of neutralizing antibodies determined at 2 and 5 weeks after rYF17D/SIVGag45-269 vaccination. The low levels of neutralization for animal r02013 were observed in three separate assays. (C) Fresh PBMC from vaccinees (100,000 cells/well) were used in IFN-γ ELISPOT assays to assess T-cell responses against the YF17D vector (red) and the SIV Gag(45-269) insert (black) at several time points postvaccination. We measured YF17D-specific responses using the same epitopes described in the legend of Fig. Fig.1.1. For SIV Gag-specific responses, we used 6 pools of 15-mers overlapping by 11 amino acids spanning the entire length of the SIVmac239 Gag insert. In addition, we measured Mamu-A*01-restricted responses against the dominant Gag181-189CM9 and subdominant Gag254-262QI9 epitopes. Four animals received a second dose of rYF17D/SIVGag45-269 on day 28 after the first vaccination (dashed line). (D) Expression kinetics of Ki-67 and Bcl-2 in CD8+ T cells after vaccination with rYF17D/SIVGag45-269. This assay was performed as described in the legend of Fig. Fig.11.We could not detect differences in vaccine-induced immune responses between the group of animals vaccinated with YF17D and the group vaccinated with rYF17D/SIVGag45-269. There was, however, considerable animal-to-animal variability. Animal r02034, which was vaccinated with YF17D, exhibited massive CD8+ T-cell activation (a peak of 35% at day 14) (Fig. (Fig.1E),1E), which was probably induced by the high levels of viral replication (16,800 copies/ml at day 5) (Fig. (Fig.1A).1A). It was difficult to see differences between the neutralizing antibody responses induced by YF17D and those induced by rYF17D/SIVGag45-269 (Fig. (Fig.1B1B and and2B).2B). However, neutralizing antibodies in animal r02013 decreased by 5 weeks postvaccination. It was also difficult to detect differences in the YF17D-specific CD8+ T-cell responses induced by these two vaccines. Peak Mamu-A*01-restricted CD8+ T-cell responses against YF17D ranged from barely detectable (animal r02110 at day 11) (Fig. (Fig.1C)1C) to 265 spot-forming cells (SFCs)/106 peripheral blood mononuclear cells (PBMC) (animal r02034 at day 28) (Fig. (Fig.1C).1C). Similarly, three of the rYF17D/SIVGag45-269-vaccinated animals (animals r04091, r04051, and r02013) made low-frequency CD8+ T-cell responses against the Mamu-A*01-bound YF17D peptides, whereas the other three animals (animals r03130, r02049, and r02042) recognized these epitopes with responses ranging from 50 to 200 SFCs/106 PBMC (Fig. (Fig.2C).2C). For almost every rYF17D/SIVGag45-269-vaccinated animal, the Gag181-189CM9-specific responses (range, 50 to 750 SFCs/106 PBMC) were higher than those generated against the Mamu-A*01-restricted YF17D epitopes (range, 0 to 175 SFCs/106 PBMC), suggesting that the recombinant virus replicated stably in vivo (Fig. (Fig.2C).2C). Thus, the recombinant YF17D virus replicated and induced both virus-specific neutralizing antibodies and CD8+ T cells that were not demonstrably different from those induced by YF17D alone.Most viral vectors are usually more efficient after a prime with DNA or recombinant BCG (rBCG) (4, 11, 15, 18). We therefore used rYF17D/SIVGag45-269 to boost two macaques that had been primed with rBCG expressing SIV proteins (Fig. (Fig.3A).3A). We detected no SIV-specific responses after either of the two priming rBCG vaccinations. Unfortunately, while the recombinant YF17D virus replicated well in animal r01056, we found evidence for only low levels of replication of rYF17D/SIVGag45-269 on day 5 postvaccination for animal r01108 (7 copies/ml) (Fig. (Fig.3B).3B). Both animals, however, generated neutralizing antibodies at 2 weeks postvaccination (Fig. (Fig.3C).3C). Encouragingly, we detected high-frequency CD8+ T-cell responses in the Mamu-A*01-positive macaque (animal r01056) after boosting with rYF17D/SIVGag45-269 (Fig. 3D to F). These responses were directed mainly against the Mamu-A*01-restricted Gag181-189CM9 epitope, which is contained in the peptide pool Gag E (Fig. (Fig.3D).3D). Furthermore, the boost induced a massive activation of animal r01056''s CD8+ T cells, peaking at 35% at 17 days postvaccination (Fig. (Fig.3E).3E). Of these activated CD8+ T cells, approximately 10% were directed against the Gag181-189CM9 epitope, with a frequency of 3.5% of CD8+ T cells (Fig. (Fig.3E).3E). These epitope-specific CD8+ T cells made IFN-γ, tumor necrosis factor alpha (TNF-α), macrophage inflammatory protein 1β (MIP-1β), and degranulated (Fig. (Fig.3F3F and data not shown). Thus, an rBCG prime followed by a recombinant yellow fever 17D boost induced polyfunctional antigen-specific CD8+ T cells.Open in a separate windowFIG. 3.rYF17D/SIVGag45-269 vaccination induced a robust expansion of Gag-specific responses in an rBCG-primed macaque. (A) Vaccination scheme. We immunized two rhesus macaques with rBCG intradermally (i.d.) (2.0 × 105 CFU), rBCG orally (107 CFU), and rYF17D/SIVGag45-269 subcutaneously (2.0 × 105 PFU) at 6-month intervals. rBCG was engineered to express 18 minigenes containing sequences of Gag, Vif, Nef, Rev, and Tat from SIVmac239. (B) Replication of rYF17D/SIVGag45-269 during the first 10 days after vaccination as measured by Q-PCR using the YF17D-specific primers described in the legend of Fig. Fig.1.1. (C) Titer of neutralizing antibodies determined at 2 and 5 weeks after rYF17D/SIVGag45-269 vaccination. (D) Fresh PBMC from animal r01056 (100,000 cells/well) were used in IFN-γ ELISPOT assays to assess T-cell responses against the YF17D vector (red) and the SIV Gag(45-269) insert (black) at several time points postvaccination. (E) Kinetics of CD8+ T-cell activation (as described in the legend of Fig. Fig.1)1) and expansion of Gag181-189CM9-specific CD8+ T cells in animal r01056 after vaccination with rYF17D/SIVGag45-269. (F) Vaccination with rYF17D/SIVGag45-269 induced robust CD8+ T-cell responses against Gag181-189CM9 in r01056. CD8+ T-cell activation (Ki-67+/Bcl-2) for baseline and day 13 are shown. Gag181-189CM9-specific responses were measured by tetramer staining and intracellular cytokine staining (ICS) with antibodies against MIP-1β and IFN-γ.Vaccine-induced CD8+ T cells are usually central memory T cells (TCM) or effector memory T cells (TEM). These two subsets of CD8+ T cells differ in function and surface markers (23). Repeated boosting drives CD8+ T cells toward the TEM subset (23). We therefore determined whether a rBCG prime followed by a rYF17D/SIVGag45-269 boost induced TCM or TEM CD8+ T cells. Staining of PBMC obtained on day 30 postvaccination revealed that the SIV-specific CD8+ T cells were largely TEM cells since the majority of them were CD28 negative (Fig. (Fig.4A).4A). Furthermore, these cells persisted with the same phenotype until day 60 after vaccination (Fig. (Fig.4B).4B). It was recently suggested that TEM cells residing in the mucosae can effectively control infection after a low-dose challenge with SIVmac239 (16).Open in a separate windowFIG. 4.rYF17D/SIVGag45-269 vaccination of animal r01056 induced effector memory Gag181-189CM9-specific CD8+ T cells that suppressed viral replication in CD4+ targets. (A and B) Frequency and memory phenotype of tetramer-positive Gag181-189-specific CD8+ T cells in animal r01056 on day 30 (A) and day 60 (B) after rYF17D/SIVGag45-269 vaccination. CD28 and CD95 expression profiles of tetramer-positive cells show a polarized effector memory phenotype. Cells were gated on CD3+ CD8+ lymphocytes. (C) Ex vivo Gag181-189CM9-specific CD8+ T cells from animal r01056 inhibit viral replication from SIVmac239-infected CD4+ T cells. Gag181-189CM9-specific CD8+ T cells from three SIV-infected Mamu-A*01-positive animals and rYF17D/SIVGag45-269-vaccinated animal r01056 were tested for their ability to suppress viral replication from SIV-infected CD4+ T cells (39). Forty-eight hours after the incubation of various ratios of SIV-infected CD4+ T cells and Gag181-189CM9-specific CD8+ T cells, the supernatant was removed and measured for viral RNA (vRNA) copies per ml by Q-PCR. We observed no suppression when effectors were incubated with CD4+ targets from Mamu-A*01-negative animals (data not shown). Animal rh2029 was infected with SIVmac239 (viral load, ∼105 vRNA copies/ml) containing mutations in 8 Mamu-B*08-restricted epitopes as part of another study (37). Animal r01080 was vaccinated with a DNA/Ad5 regimen expressing Gag, Rev, Tat, and Nef and later infected with SIVmac239 (viral load, ∼103 vRNA copies/ml) (42). Animal r95061 was vaccinated with a DNA/MVA regimen containing Gag181-189CM9 and was later challenged with SIVmac239 (undetectable viral load) (2).We then assessed whether rYF17D/SIVGag45-269-induced CD8+ T cells could recognize virally infected CD4+ T cells. We have shown that these vaccine-induced CD8+ T cells stain for tetramers and produce cytokines after stimulation with synthetic peptides (Fig. (Fig.3).3). None of these assays, however, tested whether these SIV-specific CD8+ T cells recognize SIV-infected cells and reduce viral replication. We therefore used a newly developed assay (39) to determine whether vaccine-induced CD8+ T cells can reduce viral replication in CD4+ T cells. We sorted tetramer-positive (Gag181-189CM9-specific) lymphocytes directly from fresh PBMC and incubated them for 48 h with SIVmac239-infected CD4+ T cells expressing Mamu-A*01. We assessed the percentage of CD4+ T cells that expressed SIV Gag p27 (data not shown) and the quantity of virus in the culture supernatant (Fig. (Fig.4C).4C). Vaccine-induced CD8+ T cells reduced viral replication to the same extent as that seen with Gag181-189CM9-specific CD8+ T cells purified from three SIVmac239-infected rhesus macaques, including an elite controller rhesus macaque, animal r95061 (Fig. (Fig.4C4C).The most encouraging aspect of this study is that rBCG primed a high-frequency CD8+ T-cell response after boosting with rYF17D/SIVGag45-269. These CD8+ T cells reached frequencies that were similar to those induced by an rBCG prime followed by an Ad5 boost (11). Even without the benefit of the rBCG prime, the levels of CD8+ T cells induced by a single rYF17D/SIVGag45-269 vaccination were equivalent to those induced by our best SIV vaccine, SIVmac239ΔNef. Recombinant YF17D generated an average of 195 SFCs/106 PBMC (range, 100 to 750 SFCs/106 PBMC) (n = 6), whereas SIVmac239ΔNef induced an average of 238 SFCs/106 PBMC (range, 150 to 320 SFCs/106 PBMC) (n = 3) (32). It is also possible that any YF17D/HIV recombinants would likely replicate better in humans than they have in rhesus macaques and thus induce more robust immune responses. Also, rBCG was shown previously to be effective in humans (5, 17, 33, 34) and may be more useful at priming T-cell responses in humans than it has been in our limited study with rhesus macaques. These two vectors have long-distinguished safety and efficacy histories in humans and may therefore be well suited for HIV vaccine development.  相似文献   

11.
Li  Dan  Ye  Jing-Long  Liu  Zhong-Yu 《中国病毒学》2021,36(6):1456-1464
Virologica Sinica - Yellow fever virus (YFV) is a re-emerging virus that can cause life-threatening yellow fever disease in humans. Despite the availability of an effective vaccine, little is known...  相似文献   

12.
13.
Protein content of 60 batches of Yellow Fever Vaccine was measured by Biuret method and was compared to the values obtained by Kjeldahl method. Statistical analysis did not show any difference between the two methods. The Biuret method is specific, easy to carry out and takes little time for protein estimation during production of Yellow Fever Vaccine.  相似文献   

14.
Avian leukosis virus (ALV)-free candidate primary and secondary seed lots were indistinguishable from corresponding ALV-contaminated lots with respect to (i) potency as measured by titration in newborn and weanling mice and in the MA-104 plaque system, (ii) degree of viscerotropism as measured by viremia in monkeys, (iii) neurotropism as determined by the monkey neurovirulence test, and (iv) potency as determined by antibody response in monkeys inoculated by the intracerebral route.  相似文献   

15.
Lucey  Daniel R.  Kent  Kristen R. 《中国病毒学》2019,34(3):235-239
<正>Yellow fever(YF) is an acute disease caused by a flavivirus that infects the liver. It can cause jaundice, bleeding, kidney damage, and death. No antiviral therapy exists.A vaccine does exist, however, and fortunately confers lifelong immunity after a single dose(Monath et al. 2016;WHO 2017 a, b).YF is transmitted by mosquitoes in two main cycles. In  相似文献   

16.
17.
18.
Rare serotype and chimeric recombinant adenovirus (rAd) vectors that evade anti-Ad5 immunity are currently being evaluated as potential vaccine vectors for human immunodeficiency virus type 1 and other pathogens. We have recently reported that a heterologous rAd prime-boost regimen expressing simian immunodeficiency virus (SIV) Gag afforded durable partial immune control of an SIV challenge in rhesus monkeys. However, single-shot immunization may ultimately be preferable for global vaccine delivery. We therefore evaluated the immunogenicity and protective efficacy of a single immunization of chimeric rAd5 hexon hypervariable region 48 (rAd5HVR48) vectors expressing SIV Gag, Pol, Nef, and Env against a homologous SIV challenge in rhesus monkeys. Inclusion of Env resulted in improved control of peak and set point SIV RNA levels following challenge. In contrast, DNA vaccine priming did not further improve the protective efficacy of rAd5HVR48 vectors in this system.Heterologous prime-boost vaccine regimens have proven substantially more immunogenic than single vector immunizations in a variety of experimental models, but a single-shot vaccine would presumably be ideal for eventual global delivery. The potential utility of single-shot vaccines against pathogenic simian immunodeficiency virus (SIV) challenges in rhesus monkeys has not been well characterized. We therefore evaluated the protective efficacy of a single immunization of recombinant chimeric adenovirus type 5 (rAd5) hexon hypervariable region 48 (rAd5HVR48) vectors (15) expressing SIV Gag, Pol, Nef, and Env against a pathogenic SIV challenge in rhesus monkeys. These vectors contain the HVRs of the rare Ad48 serotype and have been shown to evade dominant Ad5 hexon-specific neutralizing antibodies (NAbs) (15). We also assessed the potential utility of inclusion of Env as an immunogen (6, 7, 17) and the degree to which DNA vaccine priming would enhance the protective efficacy afforded by a single rAd5HVR48 immunization (2, 7, 18, 21).Thirty adult rhesus monkeys (n = 6/group) lacking the Mamu-A*01, Mamu-B*17, and Mamu-B*08 class I alleles were primed with plasmid DNA vaccines and boosted with rAd5HVR48 vectors as follows: (1) adjuvanted DNA prime, rAd5HVR48 boost; (2) DNA prime, rAd5HVR48 boost; (3) rAd5HVR48 alone; (4) rAd5HVR48 alone (excluding Env); and (5) sham controls. Monkeys in groups 1 to 3 received vectors expressing SIVmac239 Gag, Pol, Nef, and Env, whereas monkeys in group 4 received vectors expressing only Gag, Pol, and Nef. The DNA vaccine adjuvants in group 1 were plasmids expressing the rhesus chemokine MIP-1α and Flt3L, which have been shown to increase recruitment of dendritic cells and to improve DNA vaccine immunogenicity (20). Monkeys were primed intramuscularly with a total dose of 4 mg of DNA vaccines at weeks 0, 4, and 8. All animals then received a single intramuscular immunization of 4 × 1010 viral particles (vp) of rAd5HVR48 at week 24. At week 52, animals were challenged intravenously (i.v.) with 100 monkey infectious doses of SIVmac251 (7, 10).  相似文献   

19.
20.
The live attenuated yellow fever vaccine (YF-17D) has been successfully used for more than 70 years. It is generally considered a safe vaccine, however, recent reports of serious adverse events following vaccination have raised concerns and led to suggestions that even safer YF vaccines should be developed. Replication deficient adenoviruses (Ad) have been widely evaluated as recombinant vectors, particularly in the context of prophylactic vaccination against viral infections in which induction of CD8+ T-cell mediated immunity is crucial, but potent antibody responses may also be elicited using these vectors. In this study, we present two adenobased vectors targeting non-structural and structural YF antigens and characterize their immunological properties. We report that a single immunization with an Ad-vector encoding the non-structural protein 3 from YF-17D could elicit a strong CD8+ T-cell response, which afforded a high degree of protection from subsequent intracranial challenge of vaccinated mice. However, full protection was only observed using a vector encoding the structural proteins from YF-17D. This vector elicited virus-specific CD8+ T cells as well as neutralizing antibodies, and both components were shown to be important for protection thus mimicking the situation recently uncovered in YF-17D vaccinated mice. Considering that Ad-vectors are very safe, easy to produce and highly immunogenic in humans, our data indicate that a replication deficient adenovector-based YF vaccine may represent a safe and efficient alternative to the classical live attenuated YF vaccine and should be further tested.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号