Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Eur J Immunol ; 54(5): e2250133, 2024 May.
Article in English | MEDLINE | ID: mdl-38571392

ABSTRACT

Live-attenuated yellow fever vaccine (YF17D) was developed in the 1930s as the first ever empirically derived human vaccine. Ninety years later, it is still a benchmark for vaccines made today. YF17D triggers a particularly broad and polyfunctional response engaging multiple arms of innate, humoral and cellular immunity. This unique immunogenicity translates into an extraordinary vaccine efficacy and outstanding longevity of protection, possibly by single-dose immunization. More recently, progress in molecular virology and synthetic biology allowed engineering of YF17D as a powerful vector and promising platform for the development of novel recombinant live vaccines, including two licensed vaccines against Japanese encephalitis and dengue, even in paediatric use. Likewise, numerous chimeric and transgenic preclinical candidates have been described. These include prophylactic vaccines against emerging viral infections (e.g. Lassa, Zika and SARS-CoV-2) and parasitic diseases (e.g. malaria), as well as therapeutic applications targeting persistent infections (e.g. HIV and chronic hepatitis), and cancer. Efforts to overcome historical safety concerns and manufacturing challenges are ongoing and pave the way for wider use of YF17D-based vaccines. In this review, we summarize recent insights regarding YF17D as vaccine platform, and how YF17D-based vaccines may complement as well as differentiate from other emerging modalities in response to unmet medical needs and for pandemic preparedness.


Subject(s)
Vaccines, Attenuated , Yellow Fever Vaccine , Yellow fever virus , Humans , Yellow Fever Vaccine/immunology , Yellow fever virus/immunology , Vaccines, Attenuated/immunology , Animals , Yellow Fever/prevention & control , Yellow Fever/immunology , Vaccination/methods
2.
Funct Integr Genomics ; 24(5): 143, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39192058

ABSTRACT

The greatest obstacle for scientists is to develop an effective HIV vaccine. An effective vaccine represents the last hope for halting the unstoppable global spread of HIV and its catastrophic clinical consequences. Creating this vaccine has been challenging due to the virus's extensive genetic variability and the unique role of cytotoxic T lymphocytes (CTL) in containing it. Innovative methods to stimulate CTL have demonstrated significant therapeutic advantages in nonhuman primate model systems, unlike traditional vaccination techniques that are not expected to provide safe and efficient protection against HIV. Human clinical trials are currently evaluating these vaccination strategies, which involve plasmid DNA and live recombinant vectors. This review article covers the existing vaccines and ongoing trial vaccines. It also explores the different approaches used in developing HIV vaccines, including their molecular mechanisms, target site effectiveness, and potential side effects.


Subject(s)
AIDS Vaccines , Clinical Trials as Topic , HIV Infections , Humans , AIDS Vaccines/immunology , HIV Infections/prevention & control , HIV Infections/immunology , Animals , T-Lymphocytes, Cytotoxic/immunology , Vaccine Development , HIV-1/immunology , HIV-1/genetics
3.
J Med Virol ; 96(9): e29900, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39234788

ABSTRACT

Despite remarkable progress in the treatment of hepatitis C virus (HCV) infection, it remains a significant global health burden, necessitating the development of an effective prophylactic vaccine. This review paper presents the current landscape of HCV vaccine candidates and approaches, including more traditional, based on inactivated virus, and more modern, such as subunit protein, vectored, based on nucleic acids (DNA and mRNA) and virus-like particles. The concept of the HCV vaccine is first put in the context of viral genetic diversity and adaptive responses to HCV infection, an understanding of which is crucial in guiding the development of an effective vaccine against such a complex virus. Because ethical dimensions are also significant in vaccine research, development, and potential deployment, we also address them in this paper. The road to a safe and effective vaccine to prevent HCV infection remains bumpy due to the genetic variation of HCV and its ability to evade immune responses. The progress in cell-culture systems allowed for the production of an inactivated HCV vaccine candidate, which can induce cross-neutralizing antibodies in vitro, but whether this could prevent infection in humans is unknown. Subunit protein vaccine candidates that entered clinical trials elicited HCV-specific humoral and cellular responses, though it remains to be shown whether they translate into effective prevention of HCV infection or progression of infection to a chronic state. Such responses were also induced by a clinically tested vector-based vaccine candidate, which decreased the viral HCV load but did not prevent chronic HCV infection. These disappointments were not readily predicted from preclinical animal studies. The vaccine platforms employing virus-like particles, DNA, and mRNA provide opportunities for the HCV vaccine, but their potential in this context has yet to be shown. Ensuring the designed vaccine is based on conserved epitope(s) and elicits broadly neutralizing immune responses is also essential. Given failures in developing a prophylactic HCV vaccine, it is crucial to continue supporting national strategies, including funding for screening and treatment programs. However, these actions are likely insufficient to permanently control the HCV burden, encouraging further mobilization of significant resources for HCV vaccine research as a missing element in the elimination of viral hepatitis as a global public health.


Subject(s)
Hepacivirus , Hepatitis C , Vaccine Development , Viral Hepatitis Vaccines , Humans , Viral Hepatitis Vaccines/immunology , Hepatitis C/prevention & control , Hepatitis C/immunology , Hepacivirus/immunology , Hepacivirus/genetics , Antibodies, Neutralizing/immunology , Vaccines, Subunit/immunology , Animals , Vaccines, Inactivated/immunology
4.
Anaerobe ; 89: 102895, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39122140

ABSTRACT

INTRODUCTION: Producing commercial bacterins/toxoids against Clostridium spp. is laborious and hazardous. Conversely, developing prototype vaccines using purified recombinant toxoids, though safe and effective, is both laborious and costly for application in production animals. OBJECTIVE: Considering that inactivated recombinant Escherichiacoli (bacterin) is a simple, cost-effective, and to be safe solution, we evaluated, for the first time, a pentavalent formulation of recombinant bacterins containing the alpha, beta, and epsilon toxins of Clostridiumperfringens and C and D neurotoxins of Clostridiumbotulinum in sheep. METHODS: Subcutaneously, 18 Texel sheep received two doses (200 µg of each antigen) of recombinant bacterin (n = 7) or purified recombinant antigens (n = 6) on days 0 and 28, while the control group (n = 5) did not receive an immunization. Sera samples from days 0 (before the 1st dose), 28 (before the 2nd dose), and 56, 84, and 112 were used for measuring IgG (indirect ELISA) and neutralizing antibodies (mouse serum neutralization). RESULTS: Both formulations induced significant levels of IgG against all five toxins (p < 0.05) up to day 112, with peaks at days 28 and 56 post-immunization. The expected booster effect occurred only for the botulinum toxins. The neutralizing antibody titers were satisfactory against ETX (≥2 IU/ml for both formulations) and BoNT-D [5 IU/ml (bacterin) and 10 IU/ml (purified)]. CONCLUSION: While adjustments are required, the recombinant bacterin platform holds great potential for polyvalent vaccines due to its straightforward, safe, and cost-effective production, establishing it as a user-friendly technology for the veterinary immunobiological industry.


Subject(s)
Antibodies, Bacterial , Antibodies, Neutralizing , Bacterial Vaccines , Botulism , Enterotoxemia , Animals , Botulism/prevention & control , Botulism/veterinary , Botulism/immunology , Sheep , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Antibodies, Bacterial/blood , Enterotoxemia/prevention & control , Enterotoxemia/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Sheep Diseases/prevention & control , Sheep Diseases/immunology , Sheep Diseases/microbiology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Immunoglobulin G/blood , Escherichia coli/genetics , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Female
5.
Int J Mol Sci ; 24(22)2023 Nov 09.
Article in English | MEDLINE | ID: mdl-38003300

ABSTRACT

Herpesviruses are large DNA viruses that have long been used as powerful gene therapy tools. In recent years, the ability of herpesviruses to stimulate both innate and adaptive immune responses has led to their transition to various applications as vaccine vectors. This vaccinology branch is growing at an unprecedented and accelerated rate. To date, human herpesvirus-based vectors have been used in vaccines to combat a variety of infectious agents, including the Ebola virus, foot and mouth disease virus, and human immunodeficiency viruses. Additionally, these vectors are being tested as potential vaccines for cancer-associated antigens. Thanks to advances in recombinant DNA technology, immunology, and genomics, numerous steps in vaccine development have been greatly improved. A better understanding of herpesvirus biology and the interactions between these viruses and the host cells will undoubtedly foster the use of herpesvirus-based vaccine vectors in clinical settings. To overcome the existing drawbacks of these vectors, ongoing research is needed to further advance our knowledge of herpesvirus biology and to develop safer and more effective vaccine vectors. Advanced molecular virology and cell biology techniques must be used to better understand the mechanisms by which herpesviruses manipulate host cells and how viral gene expression is regulated during infection. In this review, we cover the underlying molecular structure of herpesviruses and the strategies used to engineer their genomes to optimize capacity and efficacy as vaccine vectors. Also, we assess the available data on the successful application of herpesvirus-based vaccines for combating diseases such as viral infections and the potential drawbacks and alternative approaches to surmount them.


Subject(s)
Herpesviridae , Viral Vaccines , Virus Diseases , Humans , Herpesviridae/genetics , Simplexvirus/genetics , Genetic Vectors/genetics
6.
Virol J ; 18(1): 50, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33663531

ABSTRACT

BACKGROUND: Zika virus (ZIKV) has been declared a public health emergency that requires development of an effective vaccine, as it might represent an international threat. METHODS: Here, two novel DNA-based (pVAXzenv) and fowlpox-based (FPzenv) recombinant putative vaccine candidates were constructed that contained the cPrME genes of ZIKV. The env gene inserted into the fowlpox vector was verified for correct transgene expression by Western blotting and by immunofluorescence in different cell lines. The production of virus-like particles as a result of env gene expression was also demonstrated by electron microscopy. BALB/c mice were immunosuppressed with dexamethasone and immunized following a prime-boost strategy in a heterologous protocol where pVAXzenv was followed by FPzenv, to evaluate the immunogenicity of the Env protein. The mice underwent a challenge with an epidemic ZIKV after the last boost. RESULTS: These data show that the ZIKV Env protein was correctly expressed in both normal human lung fibroblasts (MRC-5 cells) and green monkey kidney (Vero) cells infected with FPzenv, and that the transgene expression lasted for more than 2 weeks. After mucosal administration of FPzenv, the immunized mice showed specific and significantly higher humoral responses compared to the control mice. However, virus neutralizing antibodies were not detected using plaque reduction assays. CONCLUSIONS: Although BALB/c mice appear to be an adequate model for ZIKV infection, as it mimics the natural mild infection in human beings, inadequate immune suppression seemed to occur by dexamethasone and different immune suppression strategies should be applied before challenge to reveal any protection of the mice.


Subject(s)
Avipoxvirus , Genes, env , Viral Vaccines , Zika Virus Infection , Animals , Antibodies, Neutralizing , Antibodies, Viral , Chlorocebus aethiops , Dexamethasone , Fibroblasts , Gene Products, env , Humans , Immunity, Humoral , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/genetics , Vero Cells , Viral Vaccines/genetics , Zika Virus/genetics , Zika Virus Infection/prevention & control
7.
Avian Pathol ; 50(6): 490-499, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34463588

ABSTRACT

Md5-BAC-REV-LTR is a recombinant Marek's disease virus (MDV), with an insertion of the long terminal repeat (LTR) of reticuloendotheliosis virus (REV) into the genome of the highly virulent MDV strain rMd5. It has been shown that Md5-BAC-REV-LTR does not induce tumours and confers high protection against challenge with MDV in 15 × 7 chickens. The objective of the present study was to evaluate the protection and safety (in terms of oncogenicity and immunosuppression) of Md5-BAC-REV-LTR in commercial meat-type chickens bearing maternal antibodies against MDV. Our results show that sub-cutaneous administration of Md5-BAC-REV-LTR at 1 day of age conferred high protection (protection index PI = 84.2) against an early challenge (1 day) by contact exposure to shedder birds infected with the vv+ MDV 648A strain. In such stringent challenge conditions, Md5-BAC-REV-LTR was more protective than a commercial CVI988 (PI = 12.4) and similar to the experimental vaccine Md5-BACΔmeq (PI = 92.4). Furthermore, Md5-BAC-REV-LTR did not induce either tumours or immunosuppression in this study. Immunosuppression was evaluated by the relative lymphoid organ weights and also by the ability of the vaccine to induce late-MDV-induced immunosuppression associated with reactivation of the virus. This study shows that Md5-BAC-REV-LTR has the potential to be used as a MD vaccine and is highly protective against early challenge with vv+ MDV.RESEARCH HIGHLIGHTSMd5-BAC-REV-LTR is highly protective against early challenge with vv+ MDV in commercial meat-type chickens.Md5-BAC-REV-LTR does not cause early immunosuppression.Md5-BAC-REV-LTR does not cause late immunosuppression.Unlike other serotype 1 vaccines, Md5-BAC-REV-LTR is not detected in feather pulp at 7 days post vaccination.


Subject(s)
Herpesvirus 2, Gallid , Marek Disease Vaccines , Reticuloendotheliosis virus , Animals , Chickens , Immunosuppression Therapy/veterinary , Marek Disease Vaccines/genetics , Meat , Terminal Repeat Sequences/genetics
8.
Microbiology (Reading) ; 166(6): 501-509, 2020 06.
Article in English | MEDLINE | ID: mdl-32324529

ABSTRACT

A range of bacteria and archaea produce gas vesicles as a means to facilitate flotation. These gas vesicles have been purified from a number of species and their applications in biotechnology and medicine are reviewed here. Halobacterium sp. NRC-1 gas vesicles have been engineered to display antigens from eukaryotic, bacterial and viral pathogens. The ability of these recombinant nanoparticles to generate an immune response has been quantified both in vitro and in vivo. These gas vesicles, along with those purified from Anabaena flos-aquae and Bacillus megaterium, have been developed as an acoustic reporter system. This system utilizes the ability of gas vesicles to retain gas within a stable, rigid structure to produce contrast upon exposure to ultrasound. The susceptibility of gas vesicles to collapse when exposed to excess pressure has also been proposed as a biocontrol mechanism to disperse cyanobacterial blooms, providing an environmental function for these structures.


Subject(s)
Bacillus megaterium/metabolism , Biotechnology/methods , Halobacterium/metabolism , Nanotechnology/methods , Organelles/metabolism , Animals , Bacillus megaterium/genetics , Biotechnology/instrumentation , Environment , Gases/metabolism , Halobacterium/genetics , Humans , Medicine , Nanotechnology/instrumentation , Organelles/genetics
9.
BMC Biotechnol ; 20(1): 7, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31992276

ABSTRACT

BACKGROUND: Clostridium perfringens is the causative agent of several diseases and enteric infections in animals and humans. The virulence of C. perfringens is largely attributable to the production of numerous toxins; of these, the alpha toxin (CPA) plays a crucial role in histotoxic infections (gas gangrene). CPA toxin consists of two domains, i.e., the phospholipase C active site, which lies in the N-terminal domain amino acid (aa residues 1-250), and the C-terminal region (aa residues 251-370), which is responsible for the interaction of the toxin with membrane phospholipids in the presence of calcium ions. All currently produced clostridial vaccines contain toxoids derived from culture supernatants that are inactivated, mostly using formalin. The CPA is an immunogenic antigen; recently, it has been shown that mice that were immunized with the C-terminal domain of the toxin produced in E. coli were protected against C. perfringens infections and the anti-sera produced were able to inhibit the CPA activity. Monoclonal and polyclonal antibodies were produced only against full-length CPA and not against the truncated forms. RESULTS: In the present study, we have reported for the first time; about the generation of a recombinant baculovirus capable of producing a deleted rCPA toxin (rBacCPA250-363H6) lacking the N-terminal domain and the 28 amino acids (aa) of the putative signal sequence. The insertion of the L21 consensus sequence upstream of the translational start codon ATG, drastically increases the yield of recombinant protein in the baculovirus-based expression system. The protein was purified by Ni-NTA affinity chromatography and the lack of toxicity in vitro was confirmed in CaCo-2 cells. Polyclonal antibodies and eight hybridoma-secreting Monoclonal antibodies were generated and tested to assess specificity and reactivity. The anti-sera obtained against the fragment rBacCPA250-363H6 neutralized the phospholipase C activity of full-length PLC. CONCLUSIONS: The L21 leader sequence enhanced the expression of atoxic C-terminal recombinant CPA protein produced in insect cells. The monoclonal and polyclonal antibodies obtained were specific and highly reactive. The availability of these biologicals could contribute to the development of diagnostic assays and/or new recombinant protein vaccines.


Subject(s)
Antibodies, Bacterial/metabolism , Bacterial Toxins/genetics , Baculoviridae/growth & development , Calcium-Binding Proteins/genetics , Clostridium Infections/prevention & control , Clostridium perfringens/metabolism , Recombinant Proteins/administration & dosage , Type C Phospholipases/genetics , Animals , Antibodies, Monoclonal/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Baculoviridae/genetics , Baculoviridae/metabolism , Caco-2 Cells , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Clostridium Infections/metabolism , Clostridium perfringens/genetics , Clostridium perfringens/immunology , Consensus Sequence , Humans , Immunization , Mice , Protein Domains , Protein Engineering , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Type C Phospholipases/chemistry , Type C Phospholipases/immunology , Type C Phospholipases/metabolism
10.
Microb Pathog ; 142: 104045, 2020 Feb 06.
Article in English | MEDLINE | ID: mdl-32035105

ABSTRACT

Peste des petits ruminants (PPR) is a highly contagious and fatal disease of small ruminants, particularly sheep and goats. This disease leads to high morbidity and mortality of small ruminants, thus resulting in devastating economic loss to the livestock industry globally. The severe disease impact has prompted the Food and Agriculture Organization of the United Nations (FAO) and the World Organization for Animal Health (OIE) to develop a global strategy for the control and eradication of PPR by 2030. Over the past decades, the control of PPR is mainly achieved through vaccinating the animals with live-attenuated vaccines, e.g., rinderpest vaccines. As a closely related disease to PPR of large ruminants, rinderpest was eradicated in 2011 and its vaccines subsequently got banned in order to keep rinderpest-free zones. Consequently, it is desirable to develop homologous PPR vaccines to control the disease. The present review summarizes the objectives of PPR control and eradication by focusing on the homologous PPR vaccines.

11.
Biotechnol Lett ; 42(11): 2223-2230, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32500473

ABSTRACT

OBJECTIVES: Earlier studies have demonstrated the use of inactivated recombinant E. coli (bacterins), to protect against Clostridium spp. in vaccinated animals. These bacterins have a simpler, safer, and faster production process. However, these bacterins carry expression plasmids, containing antibiotic resistance gene, which could be assimilate accidentally by environmental microorganisms. Considering this, we aimed to impair this plasmids using formaldehyde at different concentrations. RESULTS: This compound inactivated the highest density of cells in 24 h. KanR cassette amplification was found to be impaired with 0.8% for 24 h or 0.4% for 72 h. Upon electroporation, E. coli DH5α ultracompetent cells were unable to acquire the plasmids extracted from the bacterins after inactivation procedure. Formaldehyde-treated bacterins were incubated with other viable strains of E. coli, leading to no detectable gene transfer. CONCLUSIONS: We found that this compound is effective as an inactivation agent. Here we demonstrate the biosafety involving antibiotic resistance gene of recombinant E. coli vaccines allowing to industrial production and animal application.


Subject(s)
Escherichia coli/genetics , Formaldehyde/pharmacology , Kanamycin Resistance/drug effects , Plasmids/drug effects , Escherichia coli/drug effects , Escherichia coli Vaccines/adverse effects , Escherichia coli Vaccines/genetics , Gene Transfer, Horizontal/drug effects , Plasmids/genetics , Vaccines, Inactivated , Vaccines, Synthetic
12.
Avian Pathol ; 48(3): 209-220, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30640536

ABSTRACT

Infectious laryngotracheitis (ILT) is an acute respiratory disease of chickens controlled through vaccination with live-modified attenuated vaccines, the chicken embryo origin (CEO) vaccines and the tissue-culture origin (TCO) vaccines. Recently, novel recombinant vaccines have been developed using herpesvirus of turkey (HVT) and fowl pox virus (FPV) as vectors to express ILTV immunogens for protection against ILT. The objective of this study was to assess the protection efficacy against ILT induced by recombinants, live-modified attenuated, and inactivated virus vaccines when administered alone or in combination. Commercial layer pullets were vaccinated with one or more vaccines and challenged at 35 (35 WCH) or 74 weeks of age (74 WCH). Protection was assessed by scoring clinical signs; and by determining the challenge viral load in the trachea at five days post-challenge. The FPV-LT vaccinated birds were not protected when challenged at 35 weeks; the HVT-LT and TCO vaccines in combination provided protection similar to that observed in chickens vaccinated with either HVT-LT or TCO vaccines when challenged at 35 weeks, whereas protection induced by vaccination with HVT-LT followed by TCO was superior in the 74 WCH group compared with the 35 WCH group. Birds given the inactivated ILT vaccine had fewer clinical signs and/or lower viral replication at 74 WCH when combined with TCO or HVT-LT, but not when given alone. Finally, the CEO-vaccinated birds had top protection as indicated by reduction of clinical signs and viral replication when challenged at 35 weeks (74 weeks not done). These results suggest that certain vaccine combinations may be successful to produce long-term protection up to 74 weeks of age against ILT.


Subject(s)
Chickens/immunology , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/immunology , Poultry Diseases/prevention & control , Vaccination/veterinary , Viral Vaccines/immunology , Animals , Chickens/virology , Female , Fowlpox virus/genetics , Genetic Vectors/genetics , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesvirus 1, Meleagrid/genetics , Poultry Diseases/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage
13.
Avian Pathol ; 48(6): 573-581, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31304770

ABSTRACT

The chicken embryo origin (CEO) infectious laryngotracheitis (ILT) live attenuated vaccines, although capable of protecting against disease and reducing challenge virus replication, can regain virulence. Recombinant ILT vaccines do not regain virulence but are partially successful at blocking challenge virus replication. The objective of this study was to evaluate the effect of rHVT-LT vaccination on CEO replication and how this vaccination strategy enhances protection and limits challenge virus transmission to naïve contact chickens. The rHVT-LT vaccine was administered at 1 day of age subcutaneously and the CEO vaccine was administered at 6 weeks of age via eye-drop or drinking water. CEO vaccine replication post vaccination, challenge virus replication and transmission post challenge were evaluated. After vaccination, only the group that received the CEO via eye-drop developed transient conjunctivitis. A significant decrease in CEO replication was detected for the rHVT-LT + CEO groups as compared to groups that received CEO alone. After challenge, reduction in clinical signs and challenge virus replication were observed in all vaccinated groups. However, among the vaccinated groups, the rHVT-LT group presented higher clinical signs and challenge virus replication. Transmission of the challenge virus to naïve contact chickens was only observed in the rHVT-LT vaccinated group of chickens. Overall, this study found that priming with rHVT-LT reduced CEO virus replication and the addition of a CEO vaccination provided a more robust protection than rHVT alone. Therefore, rHVT-LT + CEO vaccination strategy constitutes an alternative approach to gain better control of the disease.


Subject(s)
Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/immunology , Poultry Diseases/prevention & control , Tracheitis/veterinary , Vaccination/veterinary , Viral Vaccines/immunology , Animals , Chick Embryo , Chickens , Female , Herpesviridae Infections/prevention & control , Herpesviridae Infections/transmission , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/physiology , Poultry Diseases/transmission , Poultry Diseases/virology , Tracheitis/prevention & control , Tracheitis/virology , Turkeys , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Virus Replication
14.
Acta Microbiol Immunol Hung ; 66(2): 219-234, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30484328

ABSTRACT

Immunogenic potency of the recombinant Erp, HspR, LppX, MmaA4, and OmpA proteins from Mycobacterium tuberculosis (MTB), formulated with Montanide ISA 720 VG adjuvant, was evaluated in BALB/c mice for the first time in this study. The five vaccine formulations, adjuvant, and BCG vaccine were subcutaneously injected into mice, and the sera were collected at days 0, 15, 30, 41, and 66. The humoral and cellular immune responses against vaccine formulations were determined by measuring serum IgG and serum interferon-gamma (IFN-γ) and interleukin-12 (IL-12) levels, respectively. All formulations significantly increased IgG levels post-vaccination. The highest increase in IFN-γ level was provided by MmaA4 formulation. The Erp, HspR, and LppX formulations were as effective as BCG in enhancement of IFN-γ level. The most efficient vaccine boosting the IL-12 level was HspR formulation, especially at day 66. Erp formulation also increased the IL-12 level more than BCG at days 15 and 30. The IL-12 level boosted by MmaA4 formulation was found to be similar to that by BCG. OmpA formulation was inefficient in enhancement of cellular immune responses. This study showed that MmaA4, HspR, and Erp proteins from MTB are successful in eliciting both humoral and cellular immune responses in mice.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Heat-Shock Proteins/immunology , Membrane Proteins/immunology , Methyltransferases/immunology , Mixed Function Oxygenases/immunology , Repressor Proteins/immunology , Tuberculosis Vaccines/immunology , Animals , Antibodies, Bacterial/blood , BCG Vaccine/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Cytokines/immunology , Female , Heat-Shock Proteins/genetics , Immunity, Cellular , Immunity, Humoral , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Membrane Proteins/genetics , Methyltransferases/genetics , Mice , Mice, Inbred BALB C , Mixed Function Oxygenases/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Oleic Acids/administration & dosage , Repressor Proteins/genetics , Tuberculosis/microbiology , Tuberculosis/prevention & control , Vaccination , Vaccines, Synthetic/immunology
15.
Biotechnol Appl Biochem ; 65(3): 306-322, 2018 May.
Article in English | MEDLINE | ID: mdl-28972297

ABSTRACT

Since its introduction in 1982, biopharmaceutical drugs have revolutionized the treatment of a broad spectrum of diseases and are increasingly used in nearly all branches of medicine. In recent years, the biopharmaceuticals market has developed much faster than the market for all drugs and is believed to have great potential for further dynamic growth because of the tremendous demand for these drugs. Biobetters, which contain altered active pharmaceutical ingredients with enhanced efficacy, will play an important role in the development of biopharmaceuticals. Another significant group of biopharmaceuticals are biosimilars. Their introduction in the European Union and, recently, the Unites States markets will reduce the costs of biopharmaceutical treatment. This review highlights recent progress in the field of biopharmaceutical development and issues concerning the registration of innovative biopharmaceuticals and biosimilars. The leading class of biopharmaceuticals, the current biopharmaceuticals market, and forecasts are also discussed.


Subject(s)
Biopharmaceutics , Pharmaceutical Preparations/chemical synthesis , Humans , Pharmaceutical Preparations/chemistry
16.
Protein Expr Purif ; 136: 52-57, 2017 Aug.
Article in English | MEDLINE | ID: mdl-26578115

ABSTRACT

Plasmodium vivax is dependent on interaction with the Duffy antigen receptor for chemokines (DARC) for invasion of human erythrocytes. The P. vivax Duffy binding protein (PvDBP) mediates interaction of P. vivax merozoites with DARC. The DARC receptor-binding domain lies in a conserved N-terminal cysteine-rich region of PvDBP referred to as region II (PvDBPII). PvDBPII is an attractive vaccine candidate since antibodies raised against PvDBPII block erythrocyte invasion by P. vivax. Here, we describe methods to produce recombinant PvDBPII in its correctly folded conformation. A synthetic gene optimized for expression of PvDBPII in Escherichia coli and fed batch fermentation process based on exponential feeding strategy was used to achieve high levels of expression of recombinant PvDBPII. Recombinant PvDBPII was isolated from inclusion bodies, refolded by rapid dilution and purified by ion exchange chromatography. Purified recombinant PvDBPII was characterized for identity, purity and functional activity using standardized release assays. Recombinant PvDBPII formulated with various human compatible adjuvants including glycosylpyranosyl lipid A-stable emulsion (GLA-SE) and alhydrogel was used for immunogenicity studies in small animals to downselect a suitable formulation for clinical development. Sera collected from immunized animals were tested for recognition of PvDBPII and inhibition of PvDBPII-DARC binding. GLA-SE formulations of PvDBPII yielded higher ELISA and binding inhibition titres compared to PvDBPII formulated with alhydrogel. These data support further development of a recombinant vaccine for P. vivax based on PvDBPII formulated with GLA-SE.


Subject(s)
Antigens, Protozoan , Immunogenicity, Vaccine , Malaria Vaccines , Plasmodium vivax/genetics , Protozoan Proteins , Receptors, Cell Surface , Animals , Antigens, Protozoan/biosynthesis , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/isolation & purification , Humans , Malaria Vaccines/biosynthesis , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria Vaccines/isolation & purification , Mice , Mice, Inbred BALB C , Plasmodium vivax/immunology , Protein Domains , Protozoan Proteins/biosynthesis , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/isolation & purification , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
17.
Parasite Immunol ; 38(11): 663-669, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27512980

ABSTRACT

We demonstrated recently that immunization with recombinant Neospora caninum profilin (rNcPRO) induces limited protection and a regulatory T-cell response in mice. The aim of this study was to evaluate the immune response elicited by rNcPRO in cattle and assess a strategy to enhance its immunogenicity, combining the addition of T-cell epitopes and immune modulators. We developed a chimeric recombinant profilin fused to functional T-cell epitopes present in the N-terminal sequence of vesicular stomatitis virus (VSV) glycoprotein G (rNcPRO/G). Groups of three cattle were immunized with two doses (2 weeks apart) of rNcPRO or rNcPRO/G formulated with alum hydroxide or a nanoparticulated soya-based adjuvant enriched with Toll-like receptor (TLR) 2 and TLR9 agonists, aimed to tackle the MyD88 pathway (AVECplus). rNcPRO induced only a primary immune response (IgM mediated), while antibodies in rNcPRO/G-vaccinated animals switched to IgG1 after the booster. The vaccine formulated with rNcPRO/G and AVECplus improved the production of systemic IFN-γ and induced long-term recall B-cell responses. Overall, our study provides data supporting the use of T-cell epitopes from VSV glycoprotein G and TLR agonists to enhance and modulate immunity to peptide antigens in bovines, particularly when using small proteins from parasites for which immune responses are usually feeble.


Subject(s)
Cattle Diseases/immunology , Coccidiosis/veterinary , Neospora/physiology , Protozoan Vaccines/immunology , Toll-Like Receptors/agonists , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/immunology , B-Lymphocytes/immunology , Cattle , Coccidiosis/immunology , Epitopes, T-Lymphocyte , Female , Immunoglobulin G , Interferon-gamma/immunology , Mice , Profilins/analysis , Profilins/genetics , Protozoan Vaccines/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Proteins/metabolism , T-Lymphocytes, Regulatory/immunology
18.
Appl Microbiol Biotechnol ; 100(1): 125-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26386688

ABSTRACT

Two new vaccine candidates against dengue virus (DENV) infection were generated by fusing the coding sequences of the self-budding Z protein from Junin virus (Z-JUNV) to those of two cryptic peptides (Z/DENV-P1 and Z/DENV-P2) conserved on the envelope protein of all serotypes of DENV. The capacity of these chimeras to generate virus-like particles (VLPs) and to induce virus-neutralizing antibodies in mice was determined. First, recombinant proteins that displayed reactivity with a Z-JUNV-specific serum by immunofluorescence were detected in HEK-293 cells transfected with each of the two plasmids and VLP formation was also observed by transmission electron microscopy. Next, we determined the presence of antibodies against the envelope peptides of DENV in the sera of immunized C57BL/6 mice. Results showed that those animals that received Z/DENV-P2 DNA coding sequences followed by a boost with DENV-P2 synthetic peptides elicited significant specific antibody titers (≥6.400). Finally, DENV plaque-reduction neutralization tests (PRNT) were performed. Although no significant protective effect was observed when using sera of Z/DENV-P1-immunized animals, antibodies raised against vaccine candidate Z/DENV-P2 (diluted 1:320) were able to reduce in over 50 % the number of viral plaques generated by infectious DENV particles. This reduction was comparable to that of the 4G2 DENV-specific monoclonal cross-reactive (all serotypes) neutralizing antibody. We conclude that Z-JUNV-VLP is a valid carrier to induce antibody-mediated immune responses in mice and that Z/DENV-P2 is not only immunogenic but also protective in vitro against infection of cells with DENV, deserving further studies. On the other side, DENV's fusion peptide-derived chimera Z/DENV-P1 did not display similar protective properties.


Subject(s)
Antibodies, Neutralizing/blood , Dengue Vaccines/immunology , Dengue Virus/genetics , Drug Carriers , Junin virus/genetics , Vaccines, Virus-Like Particle/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Viral/blood , Dengue Vaccines/administration & dosage , Dengue Vaccines/genetics , Mice, Inbred C57BL , Neutralization Tests , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Viral Envelope Proteins/genetics , Viral Plaque Assay
19.
Front Vet Sci ; 11: 1413237, 2024.
Article in English | MEDLINE | ID: mdl-39193370

ABSTRACT

African swine fever is a hemorrhagic disease of pigs with high mortality rates. Since its first characterization in 1921, there has been sufficient information about African swine fever virus (ASFV) and related diseases. The virus has been found and maintained in the sylvatic cycle involving ticks and domestic and wild boars in affected regions. The ASFV is spread through direct and indirect contact with infected pigs, their products and carrier vectors especially Ornithodoros ticks. Severe economic losses and a decline in pig production have been observed in ASFV affected countries, particularly in sub-Saharan Africa and Europe. At the end of 2018, the ASFV adversely affected China, the world's leading pork-producer. Control strategies for the disease remained challenging due to the unavailability of effective vaccines and the lack of successful therapeutic measures. However, considerable efforts have been made in recent years to understand the biology of the virus, surveillance and effective control measures. This review emphasizes and summarizes the current state of information regarding the knowledge of etiology, epidemiology, transmission, and vaccine-based control measures against ASFV.

20.
One Health ; 18: 100674, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39010962

ABSTRACT

Hepatitis E virus (HEV) is a major cause of acute viral hepatitis worldwide. Up to now, no approved treatment nor a globally licensed vaccine is available. Several recombinant HEV vaccines have been developed to protect against HEV infection in humans, including the commercially available Hecolin vaccine, which are mainly based on HEV genotype 1. However, the efficacy of these vaccines against other HEV genotypes, especially genotype 3 is unknown. In this study, we evaluated the protective efficacy of Hecolin® and a novel genotype 3-based vaccine p239(gt3) against HEV-3 in a pig infection model. Pigs were divided into three groups: one group was vaccinated with Hecolin®, the second group was vaccinated with p239(gt3), and the control group received no vaccine. All pigs were subsequently challenged with HEV genotype 3 to assess the effectiveness of the vaccines. Although all immunised animals developed a high titer of neutralizing antibodies, the results showed that both vaccine applications could not provide complete protection against HEV (gt3) infection: Two out of four animals of the Hecolin® group displayed even virus shedding, and viral RNA could be detected in bile and/or liver of three out of four animals in both vaccination groups. Only one out of four animals in each group was fully protected. Neither Hecolin® nor the novel p239(gt3) vaccine provided sufficient protection against genotype 3 infection. While Hecolin® only partial protected pigs from HEV shedding, the novel p239(gt3) vaccine was at least able to prevent infected pigs from virus shedding. The results highlight the need for further development of HEV vaccines that exhibit broad protection against multiple HEV genotypes and the use of appropriate animal infection models.

SELECTION OF CITATIONS
SEARCH DETAIL