Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38290009

ABSTRACT

BACKGROUNDMalaria transmission-blocking vaccines aim to interrupt the transmission of malaria from one person to another.METHODSThe candidates R0.6C and ProC6C share the 6C domain of the Plasmodium falciparum sexual-stage antigen Pfs48/45. R0.6C utilizes the glutamate-rich protein (GLURP) as a carrier, and ProC6C includes a second domain (Pfs230-Pro) and a short 36-amino acid circumsporozoite protein (CSP) sequence. Healthy adults (n = 125) from a malaria-endemic area of Burkina Faso were immunized with 3 intramuscular injections, 4 weeks apart, of 30 µg or 100 µg R0.6C or ProC6C each adsorbed to Alhydrogel (AlOH) adjuvant alone or in combination with Matrix-M (15 µg or 50 µg, respectively). The allocation was random and double-blind for this phase I trial.RESULTSThe vaccines were safe and well tolerated with no vaccine-related serious adverse events. A total of 7 adverse events, mild to moderate in intensity and considered possibly related to the study vaccines, were recorded. Vaccine-specific antibodies were highest in volunteers immunized with 100 µg ProC6C-AlOH with Matrix-M, and 13 of 20 (65%) individuals in the group showed greater than 80% transmission-reducing activity (TRA) when evaluated in the standard membrane feeding assay at 15 mg/mL IgG. In contrast, R0.6C induced sporadic TRA.CONCLUSIONAll formulations were safe and well tolerated in a malaria-endemic area of Africa in healthy adults. The ProC6C-AlOH/Matrix-M vaccine elicited the highest levels of functional antibodies, meriting further investigation.TRIAL REGISTRATIONPan-African Clinical Trials Registry (https://pactr.samrc.ac.za) PACTR202201848463189.FUNDINGThe study was funded by the European and Developing Countries Clinical Trials Partnership (grant RIA2018SV-2311).


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Malaria , Adult , Humans , Plasmodium falciparum , Protozoan Proteins , Adjuvants, Immunologic , Antigens, Protozoan , Aluminum Hydroxide , Antibodies, Protozoan
2.
Hum Vaccin Immunother ; 19(1): 2189885, 2023 12 31.
Article in English | MEDLINE | ID: mdl-37113023

ABSTRACT

Matrix-M™ adjuvant is a key component of several novel vaccine candidates. The Matrix-M adjuvant consists of two distinct fractions of saponins purified from the Quillaja saponaria Molina tree, combined with cholesterol and phospholipids to form 40-nm open cage-like nanoparticles, achieving potent adjuvanticity with a favorable safety profile. Matrix-M induces early activation of innate immune cells at the injection site and in the draining lymph nodes. This translates into improved magnitude and quality of the antibody response to the antigen, broadened epitope recognition, and the induction of a Th1-dominant immune response. Matrix-M-adjuvanted vaccines have a favorable safety profile and are well tolerated in clinical trials. In this review, we discuss the latest findings on the mechanisms of action, efficacy, and safety of Matrix-M adjuvant and other saponin-based adjuvants, with a focus on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) subunit vaccine candidate NVX-CoV2373 developed to prevent coronavirus disease 2019 (COVID-19).


Subject(s)
COVID-19 , Saponins , Vaccines , Humans , COVID-19/prevention & control , SARS-CoV-2 , Adjuvants, Immunologic
3.
J Vet Med Sci ; 84(12): 1595-1604, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36273875

ABSTRACT

Porcine rubulavirus (PRV) is a contagious virus that affects the Mexican swine industry. This work aimed to evaluate the immunogenicity of an recombinant hemagglutinin neuraminidase-Porcine rubulavirus (rHN-PorPV) candidate vaccine on pregnant sows, and the protective efficacy afforded to their 7-day-old suckling piglets against PRV lethal challenge. Three sows were immunized with rHN-PorPV formulated with immune-stimulating complex (ISCOMs) and two sows with rHN-PorPV protein alone as well as a mock-immunized pregnant sow (negative control). Quantitative ELISA detected a high concentration of anti-rHN-PorPV Immunoglobulin G (IgG) antibodies in sow sera after the second dose of vaccine administered on day 14 until farrowing, showing viral-neutralizing and cross-neutralization activity against different variants of PRV. Sera samples from piglets of immunized sows (with or without adjuvant), showed high concentrations of IgG antibodies. As expected, piglets from the negative control sow (n=5), exhibited severe signs of disease and 100% of mortality after PRV challenge study. Conversely, 75% and 87.5% of the piglets born from the rHN-PorPV and the rHN-PorPV-ISCOMs-immunized sows (n=8), survived, respectively, showing milder PRV clinical signs. Our data indicate that rHN-PorPV candidate vaccine produced in Escherichia coli induces efficient humoral response in pregnant sows and that the maternally derived immunity provides high protection to suckling piglets against PRV lethal challenge.


Subject(s)
Escherichia coli Infections , ISCOMs , Swine Diseases , Pregnancy , Animals , Swine , Female , Neuraminidase/genetics , Hemagglutinins , Escherichia coli/genetics , Antibodies, Viral , Viral Proteins , Escherichia coli Infections/veterinary , Immunoglobulin G , Colostrum
4.
Vaccine ; 35(45): 6208-6217, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28941620

ABSTRACT

The use of viral vectors in heterologous prime-boost regimens to induce potent T cell responses in addition to humoral immunity is a promising vaccination strategy in the fight against malaria. We conducted an open-label, first-in-human, controlled Phase I study evaluating the safety and immunogenicity of Matrix-M adjuvanted vaccination with a chimpanzee adenovirus serotype 63 (ChAd63) prime followed by a modified vaccinia Ankara (MVA) boost eight weeks later, both encoding the malaria ME-TRAP antigenic sequence (a multiple epitope string fused to thrombospondin-related adhesion protein). Twenty-two healthy adults were vaccinated intramuscularly with either ChAd63-MVA ME-TRAP alone (n=6) or adjuvanted with 25µg (n=8) or 50µg (n=8) Matrix-M. Vaccinations appeared to be safe and generally well tolerated, with the majority of local and systemic adverse events being mild in nature. The addition of Matrix-M to the vaccine did not increase local reactogenicity; however, systemic adverse events were reported more frequently by volunteers who received adjuvanted vaccine in comparison to the control group. T cell ELISpot responses peaked at 7-days post boost vaccination with MVA ME-TRAP in all three groups. TRAP-specific IgG responses were highest at 28-days post boost with MVA ME-TRAP in all three groups. There were no differences in cellular and humoral immunogenicity at any of the time points between the control group and the adjuvanted groups. We demonstrate that Matrix-M can be safely used in combination with ChAd63-MVA ME-TRAP heterologous prime-boost immunization without any reduction in cellular or humoral immunogenicity. Clinical Trials Registration NCT01669512.


Subject(s)
Immunization, Secondary/adverse effects , Immunogenicity, Vaccine/immunology , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Nanoparticles/adverse effects , Saponins/adverse effects , Saponins/immunology , Vaccination/adverse effects , Adenoviridae/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adolescent , Adult , Antibodies, Protozoan/immunology , Enzyme-Linked Immunospot Assay/methods , Epitopes/adverse effects , Epitopes/immunology , Female , Genetic Vectors/adverse effects , Genetic Vectors/immunology , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunoglobulin G/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Male , Middle Aged , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Vaccinia/immunology , Young Adult
5.
Vet Res ; 48(1): 30, 2017 05 22.
Article in English | MEDLINE | ID: mdl-28532492

ABSTRACT

Saponin-based adjuvants have been widely used to enhance humoral and cellular immune responses in many species, but their mode of action is not fully understood. A characterization of the porcine transcriptional response to Matrix-M was performed in vitro using lymphocytes, monocytes or monocyte-derived dendritic cells (MoDCs) and in vivo. The effect of Matrix-M was also evaluated in specific pathogen free (SPF) pigs exposed to conventionally reared pigs. The pro-inflammatory cytokine genes IL1B and CXCL8 were up-regulated in monocytes and lymphocytes after Matrix-M exposure. Matrix-M also induced IL12B, IL17A and IFNG in lymphocytes and IFN-α gene expression in MoDCs. Several genes were indicated as up-regulated by Matrix-M in blood 18 h after injection, of which the genes for IFN-α and TLR2 could be statistically confirmed. Respiratory disease developed in all SPF pigs mixed with conventional pigs within 1-3 days. Two out of four SPF pigs injected with saline prior to contact exposure displayed systemic symptoms that was not recorded for the four pigs administered Matrix-M. Granulocyte counts, serum amyloid A levels and transcription of IL18 and TLR2 coincided with disease progression in the pigs. These results support further evaluation of Matrix-M as a possible enhancer of innate immune responses during critical moments in pig management.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity, Innate/drug effects , Saponins/metabolism , Swine/immunology , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Leukocyte Count/veterinary , Male , Nanoparticles , Real-Time Polymerase Chain Reaction/veterinary , Saponins/pharmacology , Serum Amyloid A Protein/analysis , Specific Pathogen-Free Organisms/immunology
6.
Virol J ; 12: 210, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26643820

ABSTRACT

BACKGROUND: Influenza virus infections are responsible for significant morbidity worldwide and therefore it remains a high priority to develop more broadly protective vaccines. Adjuvation of current seasonal influenza vaccines has the potential to achieve this goal. METHODS: To assess the immune potentiating properties of Matrix-M™, mice were immunized with virosomal trivalent seasonal vaccine adjuvated with Matrix-M™. Serum samples were isolated to determine the hemagglutination inhibiting (HAI) antibody titers against vaccine homologous and heterologous strains. Furthermore, we assess whether adjuvation with Matrix-M™ broadens the protective efficacy of the virosomal trivalent seasonal vaccine against vaccine homologous and heterologous influenza viruses. RESULTS: Matrix-M™ adjuvation enhanced HAI antibody titers and protection against vaccine homologous strains. Interestingly, Matrix-M™ adjuvation also resulted in HAI antibody titers against heterologous influenza B strains, but not against the tested influenza A strains. Even though the protection against heterologous influenza A was induced by the adjuvated vaccine, in the absence of HAI titers the protection was accompanied by severe clinical scores and body weight loss. In contrast, in the presence of heterologous HAI titers full protection against the heterologous influenza B strain without any disease symptoms was obtained. CONCLUSION: The results of this study emphasize the promising potential of a Matrix-M™-adjuvated seasonal trivalent virosomal influenza vaccine. Adjuvation of trivalent virosomal vaccine does not only enhance homologous protection, but in addition induces protection against heterologous strains and thus provides overall more potent and broad protective immunity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza Vaccines/immunology , Orthomyxoviridae/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Viral/blood , Body Weight , Female , Hemagglutination Inhibition Tests , Immunity, Heterologous , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice, Inbred BALB C , Orthomyxoviridae/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Severity of Illness Index , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics
7.
Clin Vaccine Immunol ; 21(3): 443-52, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24451327

ABSTRACT

Bluetongue virus (BTV) causes bluetongue disease, a vector-borne disease of ruminants. The recent northerly spread of BTV serotype 8 in Europe resulted in outbreaks characterized by clinical signs in cattle, including unusual teratogenic effects. Vaccination has been shown to be crucial for controlling the spread of vector-borne diseases such as BTV. With the aim of developing a novel subunit vaccine targeting BTV-8 that allows differentiation of infected from vaccinated animals, five His-tagged recombinant proteins, VP2 and VP5 of BTV-8 and NS1, NS2, and NS3 of BTV-2, were expressed in baculovirus or Escherichia coli expression systems for further study. Optimized purification protocols were determined for VP2, NS1, NS2, and NS3, which remained stable for detection for at least 560 to 610 days of storage at +4°C or -80°C, and Western blotting using sera from vaccinated or experimentally infected cattle indicated that VP2 and NS2 were recognized by BTV-specific antibodies. To characterize murine immune responses to the four proteins, mice were subcutaneously immunized twice at a 4-week interval with one of three protein combinations plus immunostimulating complex ISCOM-Matrix adjuvant or with ISCOM-Matrix alone (n = 6 per group). Significantly higher serum IgG antibody titers specific for VP2 and NS2 were detected in immunized mice than were detected in controls. VP2, NS1, and NS2 but not NS3 induced specific lymphocyte proliferative responses upon restimulation of spleen cells from immunized mice. The data suggest that these recombinant purified proteins, VP2, NS1, and NS2, could be an important part of a novel vaccine design against BTV-8.


Subject(s)
Antigens, Viral/immunology , Antigens, Viral/isolation & purification , Bluetongue virus/immunology , Viral Proteins/immunology , Viral Proteins/isolation & purification , Viral Vaccines/isolation & purification , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , Antigens, Viral/chemistry , Baculoviridae/genetics , Cattle , Cell Proliferation , Cholesterol/administration & dosage , Drug Combinations , Escherichia coli/genetics , Gene Expression , Immunoglobulin G/blood , Injections, Subcutaneous , Lymphocytes/immunology , Mice , Phospholipids/administration & dosage , Protein Stability , Saponins/administration & dosage , Vaccination/methods , Vaccines, Marker/chemistry , Vaccines, Marker/immunology , Vaccines, Marker/isolation & purification , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification , Viral Proteins/chemistry , Viral Vaccines/chemistry , Viral Vaccines/immunology
8.
Vet Immunol Immunopathol ; 158(1-2): 53-61, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-23988177

ABSTRACT

The early inflammatory response to Matrix-M was evaluated in pigs. Adverse reactions measured as body temperature, appetite, activity level and reaction at the site of injection were not observed after s.c. injection with three doses of the adjuvant (75, 100 or 150µg) into one week old piglets. Analyses of the immediate cytokine response of PBMC after in vitro exposure to Matrix-M (AbISCO-100(®)) revealed only a low expression of mRNA for tumour necrosis factor-α (p<0.05) after 6h incubation. Histological examination revealed an infiltration of leukocytes, haemorrhage and necrosis in muscle 24h after i.m. injection of 150µg Matrix-M in pigs aged eleven weeks. At this time, different grades of reactive lymphoid hyperplasia were recorded in the draining lymph node that was enlarged in three of these six pigs injected with Matrix-M. The global transcriptional response at the site of injection and in the draining lymph node was analyzed using Affymetrix GeneChip Porcine Genome Array. A significant enrichment of gene signatures for the cell types described as "myeloid cells" and "plasmacytoid dendritic cells" was observed at the site of injection in Matrix-M injected pigs compared with pigs injected with saline. A number of genes encoding cytokines/chemokines or their receptors were upregulated at the injection site as well as in the draining lymph node. In the draining lymph node, a majority of the upregulated genes were interferon-regulated genes (IRGs). The expression of IFN-ß, but not IFN-α, was increased in the draining lymph nodes of a majority of the pigs exposed to Matrix-M. These IFN-ß expressing pigs also expressed increased levels of osteopontin (OPN) or stimulator of interferon genes (STING), two factors known to facilitate the expression of type I IFNs in response to viral infection. Thus, Matrix-M does not appear to induce any harmful inflammatory response in piglets whilst contributing to the innate immunity by activating the type I IFN system, possibly through several alternative signalling pathways.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cholesterol/pharmacology , Cytokines/immunology , Gene Expression Regulation/immunology , Inflammation/veterinary , Phospholipids/pharmacology , Saponins/pharmacology , Swine Diseases/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Animals, Newborn , Cholesterol/administration & dosage , Cytokines/genetics , Drug Combinations , Immunity, Innate/immunology , Inflammation/immunology , Leukocytes, Mononuclear , Lymph Nodes/immunology , Oligonucleotide Array Sequence Analysis/veterinary , Phospholipids/administration & dosage , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Saponins/administration & dosage , Specific Pathogen-Free Organisms , Statistics, Nonparametric , Swine
9.
Clin Vaccine Immunol ; 20(8): 1115-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23720365

ABSTRACT

Bluetongue virus (BTV), the causative agent of bluetongue in ruminants, is an emerging virus in northern Europe. The 2006 outbreak of BTV serotype 8 (BTV-8) in Europe was marked by an unusual teratogenic effect and a high frequency of clinical signs in cattle. Conventional control strategies targeting small ruminants were therefore extended to include cattle. Since cattle were not routinely vaccinated before 2006, the immune responses to BTV have not been studied extensively in this species. With the aims of developing a subunit vaccine against BTV-8 for differentiation between infected and vaccinated animals based on viral protein 7 (VP7) antibody detection and of improving the current understanding of the immunogenicity of BTV proteins in cattle, the immune responses induced by recombinant VP2 (BTV-8) and nonstructural protein 1 (NS1) and NS2 (BTV-2) were studied. Cows were immunized twice (with a 3-week interval) with the experimental vaccine, a commercial inactivated vaccine, or a placebo. The two vaccines induced similar neutralizing antibody responses to BTV-8. Furthermore, the antibody responses detected against VP2, NS1, and NS2 were strongest in the animals immunized with the experimental vaccine, and for the first time, a serotype cross-reactive antibody response to NS2 was shown in cattle vaccinated with the commercial vaccine. The two vaccines evoked measurable T cell responses against NS1, thereby supporting a bovine cross-reactive T cell response. Finally, VP7 seroconversion was observed after vaccination with the commercial vaccine, as in natural infections, but not after vaccination with the experimental vaccine, indicating that the experimental vaccine may allow the differentiation of vaccinated animals from infected animals regardless of BTV serotype. The experimental vaccine will be further evaluated during a virulent challenge in a high-containment facility.


Subject(s)
Antigens, Viral/immunology , Bluetongue virus/immunology , Bluetongue/immunology , Bluetongue/prevention & control , Vaccines, Marker/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/administration & dosage , Cattle , Cattle Diseases/immunology , Cattle Diseases/prevention & control , T-Lymphocytes/immunology , Vaccines, Marker/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Proteins/administration & dosage , Viral Proteins/immunology , Viral Vaccines/administration & dosage
10.
PLoS One ; 7(7): e41451, 2012.
Article in English | MEDLINE | ID: mdl-22844480

ABSTRACT

Saponin-based adjuvants are widely used to enhance humoral and cellular immune responses towards vaccine antigens, although it is not yet completely known how they mediate their stimulatory effects. The aim of this study was to elucidate the mechanism of action of adjuvant Matrix-M™ without antigen and Alum was used as reference adjuvant. Adjuvant Matrix-M™ is comprised of 40 nm nanoparticles composed of Quillaja saponins, cholesterol and phospholipid. BALB/c mice were subcutaneously injected once with, 3, 12 or 30 µg of Matrix-M™, resulting in recruitment of leukocytes to draining lymph nodes (dLNs) and spleen 48 h post treatment. Flow cytometry analysis identified CD11b(+) Gr-1(high) granulocytes as the cell population increasing most in dLNs and spleen. Additionally, dendritic cells, F4/80(int) cells, T-, B- and NK-cells were recruited to dLNs and in spleen the number of F4/80(int) cells, and to some extent, B cells and dendritic cells, increased. Elevated levels of early activation marker CD69 were detected on T-, B- and NK-cells, CD11b(+) Gr-1(high) cells, F4/80(int) cells and dendritic cells in dLNs. In spleen CD69 was mainly up-regulated on NK cells. B cells and dendritic cells in dLNs and spleen showed an increased expression of the co-stimulatory molecule CD86 and dendritic cells in dLNs expressed elevated levels of MHC class II. The high-dose (30 µg) of Matrix-M™ induced detectable serum levels of IL-6 and MIP-1ß 4 h post administration, most likely representing spillover of locally produced cytokines. A lesser increase of IL-6 in serum after administration of 12 µg Matrix-M™ was also observed. In conclusion, early immunostimulatory properties were demonstrated by Matrix-M™ alone, as therapeutic doses resulted in a local transient immune response with recruitment and activation of central immune cells to dLNs. These effects may play a role in enhancing uptake and presentation of vaccine antigens to elicit a competent immune response.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immune System/cytology , Immune System/immunology , Animals , CD11b Antigen/metabolism , Cell Count , Chemokine CCL4/blood , Dose-Response Relationship, Immunologic , Female , Granulocytes/immunology , Granulocytes/metabolism , Immune System/drug effects , Immune System/metabolism , Interleukin-6/blood , Kinetics , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Receptors, Chemokine/metabolism , Spleen/drug effects , Spleen/immunology , Vaccines
11.
Dev Comp Immunol ; 38(1): 17-26, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22426325

ABSTRACT

ISCOM vaccines induce a balanced Th1/Th2 response, long-lasting antibody responses and cytotoxic T lymphocytes. The mode of action for the adjuvant component, the ISCOM-Matrix, is known to some extent but questions remain regarding its mechanism of action. The Affymetrix GeneChip® Porcine Genome Array was applied to study the global transcriptional response to ISCOM-Matrix in pigs at the injection site and in the draining lymph node 24h after i.m. injection. Gene enrichment analysis revealed inflammation, innate immunity and antigen processing to be central in the ISCOM-Matrix response. At the injection site, 594 genes were differentially expressed, including up-regulation of the cytokines osteopontin (SPP1), IL-10 and IL-18 and the chemokines CCL2, CCL19 and CXCL16. Of the 362 genes differentially expressed in the lymph node, IL-1ß and CXCL11 were up-regulated whereas IL18, CCL15 and CXCL12 were down-regulated. ISCOM-Matrix also modulated genes for pattern recognition receptors at the injection site (TLR2, TLR4, MRC1, PTX3, LGALS3) and in the lymph node (TLR4, RIG-I, MDA5, OAS1, EIF2AK2, LGALS3). A high proportion of up-regulated interferon-regulated genes indicated an interferon response. Thus, several genes, genetic pathways and biological processes were identified that are likely to shape the early immune response elicited by ISCOM-based vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Gene Expression Profiling , ISCOMs/immunology , Lymph Nodes/metabolism , Sus scrofa/immunology , Animals , ISCOMs/administration & dosage , Injections, Intramuscular , Lymph Nodes/immunology , Male , Random Allocation , Specific Pathogen-Free Organisms
13.
Clin Vaccine Immunol ; 17(3): 420-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20107005

ABSTRACT

We determined the impact of mucosal prime/boost regimens and vaccine type (attenuated Wa human rotavirus [AttHRV] or nonreplicating Wa 2/6 rotavirus-like particles [VLP]) on protection and antibody-secreting cell (ASC) responses to HRV in a neonatal gnotobiotic pig disease model. Comparisons of delivery routes for AttHRV and evaluation of nonreplicating VLP vaccines are important as alternative vaccine approaches to overcome risks associated with live oral vaccines. Groups of neonatal gnotobiotic pigs were vaccinated using combinations of oral (PO) and intranasal (IN) inoculation routes as follows: (i) 3 oral doses of AttHRV (AttHRV3xPO); (ii) AttHRV3xIN; (iii) AttHRVPO, then 2/6VLP2xIN; (iv) AttHRVIN, then 2/6VLP2xIN; and (v) mock-inoculated controls. Subsets of pigs from each group were challenged with virulent Wa HRV [P1A(8) G1] (4 weeks post-primary inoculation) to assess protection. The AttHRVPO+2/6VLP2xIN pigs had the highest protection rates against virus shedding and diarrhea (71% each); however, these rates did not differ statistically among the vaccine groups, except for the AttHRVIN+2/6VLPIN group, which had a significantly lower protection rate (17%) against diarrhea. The isotype, magnitude, and tissue distribution of ASCs were analyzed by enzyme-linked immunospot assay. The highest mean numbers of virus-specific IgG and IgA ASCs were observed pre- and postchallenge in both intestinal and systemic lymphoid tissues of the AttHRVPO+2/6VLPIN group. Thus, the AttHRVPO+2/6VLPIN vaccine regimen using immunostimulating complexes (ISCOM) and multiple mucosal inductive sites, followed by AttHRV3xPO or IN regimens, were the most effective vaccine regimens, suggesting that either AttHRVPO+2/6VLPIN or AttHRV3xIN may be an alternative approach to AttHRV3xPO for inducing protective immunity against rotavirus diarrhea.


Subject(s)
Antibody-Producing Cells/immunology , Antigens, Viral/immunology , Capsid Proteins/immunology , Rotavirus Vaccines/administration & dosage , Administration, Intranasal , Administration, Oral , Animals , Animals, Newborn , Germ-Free Life , Humans , Immunization, Secondary , Rotavirus/immunology , Rotavirus Vaccines/immunology , Swine , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Virion/immunology
14.
Vaccine ; 25(18): 3658-68, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17280751

ABSTRACT

Recombinant NcSRS2, a major immunodominant surface antigen of the intracellular protozoan parasite Neospora caninum, was used as a model antigen to compare the immunogenicity of iscoms prepared according to three different methods. Two NcSRS2 fusion proteins were used, one that was biotinylated upon expression in Escherichia coli and linked to Ni2+-loaded iscom matrix (iscom without any protein) via a hexahistidyl (His6)-tagged streptavidin fusion protein, and another that contained both a His6-tag and streptavidin (His6-SA-SRS2') and was coupled to either Ni2+-loaded or biotinylated matrix. While all three iscom preparations induced N. caninum specific antibodies at similar levels, His6-SA-SRS2' coupled to biotinylated matrix generated the strongest cellular responses measured as in vitro proliferation and production of interferon-gamma and interleukin-4 after antigen stimulation of spleen cells. However, the relationship between the levels of these cytokines as well as between IgG1 and IgG2a titres in serum induced by the three iscom preparations were similar, indicating that the balance between Th1 and Th2 responses did not differ. After challenge infection, mice immunised with His6-SA-SRS2' coupled to biotinylated matrix had significantly lower amounts of parasite DNA in their brains compared to the other immunised groups. Possible reasons for the performance of the different iscom formulations are discussed.


Subject(s)
Antigens, Protozoan/immunology , Antigens, Surface/immunology , Coccidiosis/prevention & control , ISCOMs/immunology , Neospora/immunology , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Recombinant Proteins/immunology , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Antigens, Surface/genetics , Coccidiosis/immunology , Coccidiosis/parasitology , Female , ISCOMs/administration & dosage , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Neospora/genetics , Neospora/pathogenicity , Protozoan Proteins/genetics , Protozoan Vaccines/administration & dosage , Recombinant Proteins/genetics , Specific Pathogen-Free Organisms , Spleen/cytology , Spleen/immunology
15.
Vaccine ; 24(47-48): 6839-49, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-17050045

ABSTRACT

The difficulty in developing an effective vaccine to contain the HIV/AIDS epidemic coupled with the fact that primary HIV-1 infection typically occurs via mucosal sites has increased emphasis on vaccine approaches that protect at mucosal surfaces. In this study we employed HIV and simian-HIV (SHIV)-derived T helper (Th) and cytotoxic T lymphocyte (CTL) single epitopes incorporated into immuno-stimulating complexes (ISCOM) as a candidate immunogens. Immunized rhesus macaques (Macaca mulatta) were challenged with CCR5-tropic SHIV(SF162p4). On the day of challenge, low levels of virus-neutralizing antibodies (Ab) and CTLs were detected in ISCOM-immunized macaques. Greater than 10(5) viral RNA copies per ml of plasma in 2/5 immunized and 3/4 control macaques were detected within 3 weeks post-challenge. Depletion of CD4+ T cells from gut-associated lymphoid tissues (GALT) was observed by post-challenge day (PCD) 14 in all macaques regardless immunization. Nonetheless, lower viral loads and relatively better preservation of peripheral CD4+ T cells following the SHIV infection was observed in ISCOM-immunized macaques. We predict that if coadministered with additional epitopes and/or more efficacious mucosal delivery system or route, HIV/SIV-derived peptide vaccines may have potential to elicit heterologous protection.


Subject(s)
Adjuvants, Immunologic/pharmacology , Epitopes/immunology , Immunity, Mucosal/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , Antibody Formation/immunology , CD4-Positive T-Lymphocytes/immunology , Gene Products, gag/immunology , Immunity, Cellular/immunology , Immunization , Interferon-gamma/biosynthesis , Ki-67 Antigen/immunology , Macaca mulatta/immunology , Neutralization Tests , Phenotype , fas Receptor/immunology
16.
Expert Rev Vaccines ; 5(3): 395-403, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16827623

ABSTRACT

In recent years, several studies have been reported with the common aim of generating general expression systems for straightforward production and subsequent coupling of expressed antigens to an adjuvant system. Here, we describe a series of such efforts with a common theme of using gene fusion technology for association of recombinant antigens to immunostimulating complexes (iscoms). In the early stages of vaccine development, uniform antigen preparations are crucial to allow the comparison of immune responses to different antigens, or even subdomains thereof, and we believe that the described systems constitute an important development in this context.


Subject(s)
Antigens/immunology , ISCOMs/immunology , Vaccines, Subunit/immunology , Animals , Antigens/genetics , Bacterial Proteins/chemistry , Biotin/analogs & derivatives , Biotin/chemistry , ISCOMs/chemistry , Lipoproteins/chemistry , Lipoproteins/immunology , Lipoproteins/metabolism , Peptides/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism
17.
Clin Vaccine Immunol ; 13(4): 475-85, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16603615

ABSTRACT

We investigated maternal antibody (MatAb) effects on protection and immune responses to rotavirus vaccines. Gnotobiotic pigs were injected intraperitoneally at birth with pooled serum from sows hyperimmunized with human rotavirus (HRV); control pigs received no sow serum. Pigs with or without MatAbs received either sequential attenuated HRV (AttHRV) oral priming and intranasal boosting with VP2/VP6 virus-like particle (VLP)-immunostimulating complex (ISCOM) (AttHRV/VLP) or intranasal VLP-ISCOM prime/boost (VLP) vaccines at 3 to 5 days of age. Subsets of pigs were challenged at 28 or 42 days postinoculation with virulent Wa HRV to assess protection. Isotype-specific antibody-secreting cell (ASC) responses to HRV were quantitated by enzyme-linked immunospot assay to measure effector and memory B-cell responses in intestinal and systemic lymphoid tissues pre- and/or postchallenge. Protection rates against HRV challenge (contributed by active immunity and passive circulating MatAbs) were consistently (but not significantly) lower in the MatAb-AttHRV/VLP groups than in the corresponding groups without MatAbs. Intestinal B-cell responses in the MatAb-AttHRV/VLP group were most suppressed with significantly reduced or no intestinal immunoglobulin A (IgA) and IgG effector and memory B-cell responses or antibody titers pre- and postchallenge. This suppression was not alleviated but was enhanced after extending vaccination/challenge from 28 to 42 days. In pigs vaccinated with nonreplicating VLP alone that failed to induce protection, MatAb effects differed, with intestinal and systemic IgG ASCs and prechallenge memory B cells suppressed but the low intestinal IgA and IgM ASC responses unaffected. Thus, we demonstrate that MatAbs differentially affect both replicating and nonreplicating HRV vaccines and suggest mechanisms of MatAb interference. This information should facilitate vaccine design to overcome MatAb suppression.


Subject(s)
Antibodies, Viral/blood , B-Lymphocytes/immunology , ISCOMs/immunology , Immunity, Maternally-Acquired , Immunization, Secondary , Immunologic Memory , Rotavirus Vaccines/immunology , Virion/immunology , Animals , Animals, Newborn , Antibodies, Viral/administration & dosage , B-Lymphocytes/virology , Diarrhea/immunology , Diarrhea/prevention & control , Diarrhea/virology , Female , Humans , ISCOMs/administration & dosage , Infant , Infant, Newborn , Injections, Intraperitoneal , Rotavirus/immunology , Rotavirus Vaccines/administration & dosage , Swine , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
18.
Vaccine ; 24(13): 2302-16, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16361002

ABSTRACT

We investigated effects of low titer (Lo) circulating MatAb on protection and immunogenicity of attenuated (Att) human rotavirus (HRV) priming and 2/6-virus-like particle (VLP)-immunostimulating complex (ISCOM) boosting (AttHRV/VLP) or VLP-ISCOM alone vaccines. LoMatAb had both enhancing and suppressing effects on B cell responses, depending on tissue, antibody isotype and vaccine. Differential effects of LoMatAb on IgA responses in different tissues suggest that LoMatAb did not suppress induction of IgA effector and memory B cells but impaired homing of these cells to secondary lymphoid or effector tissues, reducing IgA antibody secreting cells and antibodies at these sites. The AttHRV/VLP vaccine partially overcame LoMatAb suppression, conferred moderate protection against virulent HRV (as measured by reduced viral shedding and diarrhea) and represents a new candidate for rotavirus vaccines for both humans and animals.


Subject(s)
Adjuvants, Immunologic/administration & dosage , B-Lymphocytes/immunology , ISCOMs/administration & dosage , Immunity, Maternally-Acquired , Rotavirus Vaccines/immunology , Virion/immunology , Animals , Antibodies, Viral/biosynthesis , Humans , Immunologic Memory , Injections, Intraperitoneal , Swine , Vaccines, Attenuated/immunology , Virus Shedding
19.
Vaccine ; 23(17-18): 2331-5, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15755622

ABSTRACT

We have previously reported strategies for Escherichia coli production of recombinant immunogens fused to hydrophobic peptides or lipid tags to improve their capacity to be incorporated into an adjuvant formulation, e.g., immunostimulating complexes (iscoms). Recently, we also explored the strong interaction between biotin and streptavidin to achieve iscom association of recombinant immunogens. Plasmodium falciparum,Toxoplasma gondii and Neospora caninum antigens have served as model immunogens in the different studies. Generated fusion proteins have been found to be successfully incorporated into iscoms and high-titer antigen-specific antibody responses have been obtained upon immunization of mice. We believe that the different concepts presented, utilizing either hydrophobic peptide or lipid tags, or the recently explored biotin-streptavidin principle, offer convenient methods to achieve efficient adjuvant incorporation of recombinant immunogens.


Subject(s)
Immunologic Factors/administration & dosage , Vaccines, Subunit/administration & dosage , Amino Acid Sequence , Animals , Antigens/administration & dosage , Antigens/chemistry , Antigens/genetics , Biotin , Escherichia coli/genetics , ISCOMs , Immunization , Immunologic Factors/chemistry , Lipids/chemistry , Mice , Molecular Sequence Data , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Streptavidin , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/genetics
20.
Biotechnol Appl Biochem ; 41(Pt 2): 163-74, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15296488

ABSTRACT

We have previously reported strategies for Escherichia coli production of recombinant immunogens fused to hydrophobic peptide or lipid tags to improve their capacity to be incorporated into an adjuvant formulation. In the present study, we have explored the strong interaction between biotin and SA (streptavidin) (K(D) approximately 10(-15) M) to couple recombinant immunogens to iscoms (immunostimulating complexes). Two different concepts were evaluated. In the first concept, a His(6)-tagged SA fusion protein (His(6)-SA) was bound to Ni(2+)-loaded iscom matrix (iscom without associated protein), and biotinylated immunogens were thereafter associated with the SA-coated iscoms. The immunogens were either biotinylated in vivo on E. coli expression or double biotinylated in vivo and in vitro. In the second concept, the recombinant immunogens were expressed as SA fusion proteins, which were directly bound to a biotinylated iscom matrix. A 53-amino-acid malaria peptide (M5), derived from the central repeat region of the Plasmodium falciparum blood-stage antigen Pf155/RESA, and a 232-amino-acid segment (SRS2') from the central region (from Pro-97 to Lys-328) of the major surface antigen NcSRS2 of the protozoan parasite Neospora caninum, served as model immunogens in the present study. All fusion proteins generated were found to be efficiently expressed and could be recovered to high purity using affinity chromatography. The association between the different immunogen-containing fusion proteins and the corresponding iscom matrix was demonstrated by analytical ultracentrifugation in a sucrose density gradient. However, some fusion proteins were, to a certain extent, also found to associate unspecifically with a regular iscom matrix. Furthermore, selected iscom fractions were demonstrated to induce high-titre antigen-specific antibody responses on immunization of mice. For the particular target immunogen SRS2', the induced antibodies demonstrated reactivity to the native antigen NcSRS2. We believe that the presented concepts offer convenient methods to achieve efficient adjuvant association of recombinant immunogens, and the advantages and disadvantages of the two concepts are discussed.


Subject(s)
Bacterial Proteins/metabolism , Biotin/analogs & derivatives , Vaccines, Synthetic/biosynthesis , Animals , Antibodies/metabolism , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Antigens, Surface/immunology , Antigens, Surface/metabolism , Bacterial Proteins/chemistry , Base Sequence , Biotin/chemistry , Biotin/metabolism , Female , Genetic Engineering , Immunization , Mice , Molecular Sequence Data , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...