1
|
Honda-Okubo Y, Bart Tarbet E, Hurst BL, Petrovsky N. An Advax-CpG adjuvanted recombinant H5 hemagglutinin vaccine protects mice against lethal influenza infection. Vaccine 2023; 41:5730-5741. [PMID: 37567799 DOI: 10.1016/j.vaccine.2023.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/11/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
There is a major unmet need for strategies to improve the immunogenicity of vaccines to protect against highly pathogenic avian influenza strains with pandemic potential. This study tested the ability of adjuvants based on delta inulin (Advax™) alone or combined with a TLR9 agonist (Advax-CpG™) to enhance the immunogenicity of recombinant H5 hemagglutinin antigen expressed in insect cells (rH5HA) to protect mice against lethal influenza infection. The Advax-adjuvanted rH5HA induced high serum hemagglutination inhibition activity, as well as Th1 and Th2 cytokine secreting CD4 and CD8 T cells. Immunization protected mice against a lethal heterosubtypic H5N1 virus challenge. Mice immunized with an Advax-adjuvanted rHA2 stem antigen prepared by enzymatic cleavage of rH5HA produced serum antibodies devoid of hemagglutination inhibition activity, but these anti-HA2 antibodies were nevertheless able to transfer protection against lethal H1N1 or H3N2 infections to naïve mice. We hypothesize that the enhanced protection afforded by Advax-adjuvanted rH5HA may be mediated by the combination of neutralizing antibodies directed at the HA head, anti-HA2 stem antibodies plus memory CD4 + and CD8 + T cells. This outcome supports further development of the Advax-adjuvanted rH5 pandemic influenza vaccine platform.
Collapse
Affiliation(s)
- Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, Warradale, Adelaide, SA 5046, Australia; Flinders University, Bedford Park, Adelaide, SA 5042, Australia
| | - E Bart Tarbet
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, UT 84322, USA
| | - Brett L Hurst
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, UT 84322, USA
| | | |
Collapse
|
2
|
Carnet F, Perrin-Cocon L, Paillot R, Lotteau V, Pronost S, Vidalain PO. An inventory of adjuvants used for vaccination in horses: the past, the present and the future. Vet Res 2023; 54:18. [PMID: 36864517 PMCID: PMC9983233 DOI: 10.1186/s13567-023-01151-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/27/2023] [Indexed: 03/04/2023] Open
Abstract
Vaccination is one of the most widely used strategies to protect horses against pathogens. However, available equine vaccines often have limitations, as they do not always provide effective, long-term protection and booster injections are often required. In addition, research efforts are needed to develop effective vaccines against emerging equine pathogens. In this review, we provide an inventory of approved adjuvants for equine vaccines worldwide, and discuss their composition and mode of action when available. A wide range of adjuvants are used in marketed vaccines for horses, the main families being aluminium salts, emulsions, polymers, saponins and ISCOMs. We also present veterinary adjuvants that are already used for vaccination in other species and are currently evaluated in horses to improve equine vaccination and to meet the expected level of protection against pathogens in the equine industry. Finally, we discuss new adjuvants such as liposomes, polylactic acid polymers, inulin, poly-ε-caprolactone nanoparticles and co-polymers that are in development. Our objective is to help professionals in the horse industry understand the composition of marketed equine vaccines in a context of mistrust towards vaccines. Besides, this review provides researchers with a list of adjuvants, either approved or at least evaluated in horses, that could be used either alone or in combination to develop new vaccines.
Collapse
Affiliation(s)
- Flora Carnet
- grid.508204.bLABÉO, 14280 Saint-Contest, France ,grid.412043.00000 0001 2186 4076BIOTARGEN, Normandie University, UNICAEN, 14280 Saint-Contest, France
| | - Laure Perrin-Cocon
- grid.462394.e0000 0004 0450 6033CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, 69007 Lyon, France
| | - Romain Paillot
- grid.451003.30000 0004 0387 5232School of Equine and Veterinary Physiotherapy, Writtle University College, Lordship Road, Writtle, Chelmsford, CM1 3RR UK
| | - Vincent Lotteau
- grid.462394.e0000 0004 0450 6033CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, 69007 Lyon, France
| | - Stéphane Pronost
- LABÉO, 14280, Saint-Contest, France. .,BIOTARGEN, Normandie University, UNICAEN, 14280, Saint-Contest, France.
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, 69007, Lyon, France.
| |
Collapse
|
3
|
Kumar A, Sharma A, Tirpude NV, Padwad Y, Hallan V, Kumar S. Plant-derived immuno-adjuvants in vaccines formulation: a promising avenue for improving vaccines efficacy against SARS-CoV-2 virus. Pharmacol Rep 2022; 74:1238-1254. [PMID: 36125739 PMCID: PMC9487851 DOI: 10.1007/s43440-022-00418-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 12/13/2022]
Abstract
The SARS-CoV-2 outbreak has posed a plethora of problems for the global healthcare system and socioeconomic burden. Despite valiant efforts to contain the COVID-19 outbreak, the situation has deteriorated to the point that there are no viable preventive therapies to treat this disease. The case count has skyrocketed globally due to the newly evolved variants. Despite vaccination drives, the re-occurrence of recent pandemic waves has reinforced the importance of innovation/utilization of immune-booster to achieve appropriate long-term vaccine protection. Plant-derived immuno-adjuvants, which have multifaceted functions, can impede infections by boosting the immune system. Many previous studies have shown that formulation of vaccines using plant-derived adjuvant results in long-lasting immunity may overcome the natural tendency of coronavirus immunity to wane quickly. Plant polysaccharides, glycosides, and glycoprotein extracts have reportedly been utilized as enticing adjuvants in experimental vaccines, such as Advax, Matrix-M, and Mistletoe lectin, which have been shown to be highly immunogenic and safe. When employed in vaccine formulation, Advax and Matrix-M generate long-lasting antibodies, a balanced robust Th1/Th2 cytokine profile, and the stimulation of cytotoxic T cells. Thus, the use of adjuvants derived from plants may increase the effectiveness of vaccines, resulting in the proper immunological response required to combat COVID-19. A few have been widely used in epidemic outbreaks, including SARS and H1N1 influenza, and their use could also improve the efficacy of COVID-19 vaccines. In this review, the immunological adjuvant properties of plant compounds as well as their potential application in anti-COVID-19 therapy are thoroughly discussed.
Collapse
Affiliation(s)
- Arbind Kumar
- COVID-19 Testing facility, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Aashish Sharma
- COVID-19 Testing facility, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Narendra Vijay Tirpude
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Yogendra Padwad
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Vipin Hallan
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| | - Sanjay Kumar
- CSIR-Institute of Himalayan Bioresource Technology (IHBT), Palampur, India
| |
Collapse
|
4
|
Crispell G, Williams K, Zielinski E, Iwami A, Homas Z, Thomas K. Method comparison for Japanese encephalitis virus detection in samples collected from the Indo-Pacific region. Front Public Health 2022; 10:1051754. [PMID: 36504937 PMCID: PMC9730272 DOI: 10.3389/fpubh.2022.1051754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Japanese encephalitis virus (JEV) is a mosquito-borne viral pathogen, which is becoming a growing public health concern throughout the Indo-Pacific. Five genotypes of JEV have been identified. Current vaccines are based on genotype III and provide a high degree of protection for four of the five known genotypes. Methods RT-PCR, Magpix, Twist Biosciences Comprehensive Viral Research Panel (CVRP), and SISPA methods were used to detect JEV from mosquito samples collected in South Korea during 2021. These methods were compared to determine which method would be most effective for biosurveillance in the Indo-Pacific region. Results Our data showed that RT-PCR, Twist CVRP, and SISPA methods were all able to detect JEV genotype I, however, the proprietary Magpix panel was only able to detect JEV genotype III. Use of minION sequencing for pathogen detection in arthropod samples will require further method development. Conclusion Biosurveillance of vectorborne pathogens remains an area of concern throughout the Indo-Pacific. RT-PCR was the most cost effective method used in the study, but TWIST CVRP allows for the identification of over 3,100 viral genomes. Further research and comparisons will be conducted to ensure optimal methods are used for large scale biosurveillance.
Collapse
|
5
|
Shoushtari M, Roohvand F, Salehi-Vaziri M, Arashkia A, Bakhshi H, Azadmanesh K. Adenovirus vector-based vaccines as forefront approaches in fighting the battle against flaviviruses. Hum Vaccin Immunother 2022; 18:2079323. [PMID: 35714271 PMCID: PMC9481145 DOI: 10.1080/21645515.2022.2079323] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Flaviviruses are arthropod-borne viruses (arboviruses) that have been recently considered among the significant public health problems in defined geographical regions. In this line, there have been vaccines approved for some flaviviruses including dengue virus (DENV), Japanese encephalitis virus (JEV), yellow fever virus (YFV), and tick-borne encephalitis virus (TBEV), although the efficiency of such vaccines thought to be questionable. Surprisingly, there are no effective vaccine for many other hazardous flaviviruses, including West Nile and Zika viruses. Furthermore, in spite of approved vaccines for some flaviviruses, for example DENV, alternative prophylactic vaccines seem to be still needed for the protection of a broader population, and it originates from the unsatisfying safety, and the efficacy of vaccines that have been introduced. Thus, adenovirus vector-based vaccine candidates are suggested to be effective, safe, and reliable. Interestingly, recent widespread use of adenovirus vector-based vaccines for the COVID-19 pandemic have highlighted the importance and feasibility of their widespread application. In this review, the applicability of adenovirus vector-based vaccines, as promising approaches to harness the diseases caused by Flaviviruses, is discussed.
Collapse
Affiliation(s)
| | - Farzin Roohvand
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Salehi-Vaziri
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Hasan Bakhshi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Kayhan Azadmanesh
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Akin I, Akdas S, Ceylan MN, Altiner S, Aribal Ayral P, Yazihan N. Evaluation of the safety and efficacy of Advax TM as an adjuvant: A systematic review and meta-analysis. Adv Med Sci 2022; 67:10-17. [PMID: 34562856 DOI: 10.1016/j.advms.2021.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 07/06/2021] [Accepted: 09/03/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Developing a vaccine with improved immunogenicity is still a growing priority for many diseases. Different types of adjuvants may be beneficial to initiate and maintain the long-lasting immunogenicity of vaccines. Evidence has shown that polysaccharide adjuvants are efficient in improving immunological mechanisms with their biocompatibility and biodegradability characteristics. In this study, we aimed to investigate the safety and efficacy of AdvaxTM an adjuvant derived from delta inulin. METHODS A systematic research was performed in Pubmed, Web of Science, and Scopus databases for the following keywords; "AdvaxTM" OR "delta inulin" until December 14th, 2020. RevMan 5.4.1 software was used for cumulative meta-analysis and bias analysis. We also used GraphPad Prism 6 software for the figures. RESULTS In the cumulative meta-analysis, it was found that seroconversion and geometric mean titers (GMT) levels significantly increased in AdvaxTM-adjuvanted group (mean difference: 12.31, 95% Cl [4.14, 20.47], p = 0.003; 17.10, 95% Cl [4.35, 29.85], p = 0.009, respectively). We also observed that AdvaxTM could be effective in improving immunogenicity by inducing T-cell responses and plasmablast generation in viral vaccines. CONCLUSIONS In this study, it was shown that AdvaxTM is a safe and well-tolerated adjuvant. AdvaxTM could be a potent adjuvant in increasing the protection and immunogenicity of different vaccines without safety issues. However, further studies are needed to verify these effects of AdvaxTM adjuvant.
Collapse
Affiliation(s)
- Irem Akin
- Institute of Health Sciences, Interdisciplinary Food, Metabolism and Clinical Nutrition Department, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sevginur Akdas
- Institute of Health Sciences, Interdisciplinary Food, Metabolism and Clinical Nutrition Department, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Merve Nur Ceylan
- Institute of Health Sciences, Interdisciplinary Food, Metabolism and Clinical Nutrition Department, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Seda Altiner
- Allergy and Clinical Immunology Clinic, Necip Fazil City Hospital, Kahramanmaras, Ankara, Turkey
| | - Pelin Aribal Ayral
- Institute of Health Sciences, Interdisciplinary Food, Metabolism and Clinical Nutrition Department, Ankara University Faculty of Medicine, Ankara, Turkey; Department of Pathophysiology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Nuray Yazihan
- Institute of Health Sciences, Interdisciplinary Food, Metabolism and Clinical Nutrition Department, Ankara University Faculty of Medicine, Ankara, Turkey; Department of Pathophysiology, Faculty of Medicine, Ankara University, Ankara, Turkey.
| |
Collapse
|
7
|
Ali Dahhas M, Alsenaidy MA. Role of site-directed mutagenesis and adjuvants in the stability and potency of anthrax protective antigen. Saudi Pharm J 2022; 30:595-604. [PMID: 35693445 PMCID: PMC9177452 DOI: 10.1016/j.jsps.2022.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/21/2022] [Indexed: 11/03/2022] Open
Abstract
Anthrax is a zoonotic infection caused by the gram-positive, aerobic, spore-forming bacterium Bacillus anthracis. Depending on the origin of the infection, serious health problems or mortality is possible. The virulence of B. anthracis is reliant on three pathogenic factors, which are secreted upon infection: protective antigen (PA), lethal factor (LF), and edema factor (EF). Systemic illness results from LF and EF entering cells through the formation of a complex with the heptameric form of PA, bound to the membrane of infected cells through its receptor. The currently available anthrax vaccines have multiple drawbacks, and recombinant PA is considered a promising second-generation vaccine candidate. However, the inherent chemical instability of PA through Asn deamidation at multiple sites prevents its use after long-term storage owing to loss of potency. Moreover, there is a distinct possibility of B. anthracis being used as a bioweapon; thus, the developed vaccine should remain efficacious and stable over the long-term. Second-generation anthrax vaccines with appropriate adjuvant formulations for enhanced immunogenicity and safety are desired. In this article, using protein engineering approaches, we have reviewed the stabilization of anthrax vaccine candidates that are currently licensed or under preclinical and clinical trials. We have also proposed a formulation to enhance recombinant PA vaccine potency via adjuvant formulation.
Collapse
|
8
|
Firdaus FZ, Skwarczynski M, Toth I. Developments in Vaccine Adjuvants. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2412:145-178. [PMID: 34918245 DOI: 10.1007/978-1-0716-1892-9_8] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vaccines, including subunit, recombinant, and conjugate vaccines, require the use of an immunostimulator/adjuvant for maximum efficacy. Adjuvants not only enhance the strength and longevity of immune responses but may also influence the type of response. In this chapter, we review the adjuvants that are available for use in human vaccines, such as alum, MF59, AS03, and AS01. We extensively discuss their composition, characteristics, mechanism of action, and effects on the immune system. Additionally, we summarize recent trends in adjuvant discovery, providing a brief overview of saponins, TLRs agonists, polysaccharides, nanoparticles, cytokines, and mucosal adjuvants.
Collapse
Affiliation(s)
- Farrhana Ziana Firdaus
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,Institute of Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
| |
Collapse
|
9
|
Li H, Monslow MA, Freed DC, Chang D, Li F, Gindy M, Wang D, Vora K, Espeseth AS, Petrovsky N, Fu TM. Novel adjuvants enhance immune responses elicited by a replication-defective human cytomegalovirus vaccine in nonhuman primates. Vaccine 2021; 39:7446-7456. [PMID: 34852943 DOI: 10.1016/j.vaccine.2021.10.075] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/13/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022]
Abstract
Adjuvants have long been explored to enhance vaccine efficacy. Current adjuvants approved for human vaccines are mostly studied for their ability to improve antibody responses. There remains a need for development of novel adjuvants, especially those able to enhance cell-mediated immunity (CMI). In this preclinical study we assessed the effect of two novel adjuvants, a delta inulin microparticle Advax formulated with or without a toll-like receptor 9 (TLR9) agonist CpG oligonucleotide, and a Merck & Co., Inc., Kenilworth, NJ, USA proprietary lipid nanoparticle (LNP), on immune responses elicited by V160, an experimental replication-defective human cytomegalovirus vaccine. Adult rhesus macaques were immunized with a low dose of V160 (10 units) either alone or in combination with the adjuvants as compared to those immunized with a high dose of V160 alone (100 units). While neither adjuvant conferred a significant benefit to vaccine-elicited humoral immune responses at the dose tested, both enhanced cellular immune responses to V160, where Advax promoted both CD4+ and CD8+ T cells and LNP predominantly impacted the CD4+ T cell response. Transcriptome analyses of peripheral blood samples demonstrated different modes of action for these adjuvants. One day post vaccination, LNP induced upregulation of a large number of genes involved in the innate immune response similar to those triggered by viral infection. In contrast, Advax did not activate any known inflammatory pathways and did not significantly impact gene expression pattern until day 7 post administration, suggesting a unique, non-inflammatory mechanism. These data warrant further exploration of Advax and LNP as adjuvants in clinical trials for vaccines desiring to elicit both humoral and T cell responses.
Collapse
Affiliation(s)
- Hualin Li
- Merck & Co., Inc., Kenilworth, NJ, USA.
| | | | | | - Dan Chang
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - Dai Wang
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - Nikolai Petrovsky
- Vaxine Pty Ltd, Flinders University, Bedford Park SA 5042, Australia
| | | |
Collapse
|
10
|
Kumbhar PS, Pandya AK, Manjappa AS, Disouza JI, Patravale VB. Carbohydrates-based diagnosis, prophylaxis and treatment of infectious diseases: Special emphasis on COVID-19. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2021. [PMCID: PMC7935400 DOI: 10.1016/j.carpta.2021.100052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
COVID-19 pandemic is taking a dangerous turn due to unavailability of approved and effective vaccines and therapy. Currently available diagnostic techniques are time-consuming, expensive, and maybe impacted by the mutations produced in the virus. Therefore, investigation of novel, rapid, and economic diagnosis techniques, prophylactic vaccines and targeted efficacious drug delivery systems as treatment strategy is imperative. Carbohydrates are essential biomolecules which also act as markers in the realization of immune systems. Moreover, they exhibit antiviral, antimicrobial, and antifungal properties. Carbohydrate-based vaccines and therapeutics including stimuli sensitive systems can be developed successfully and used effectively to fight COVID-19. Thus, carbohydrate-based diagnostic, prophylactic and therapeutic alternatives could be promising to defeat COVID-19 propitiously. Morphology of SARS-CoV-2 and its relevance in devising combat strategies has been discussed. Carbohydrate-based approaches for tackling infectious diseases and their importance in the design of various diagnostic and treatment modalities have been reviewed.
Collapse
|
11
|
An Advax-Adjuvanted Inactivated Cell-Culture Derived Japanese Encephalitis Vaccine Induces Broadly Neutralising Anti-Flavivirus Antibodies, Robust Cellular Immunity and Provides Single Dose Protection. Vaccines (Basel) 2021; 9:vaccines9111235. [PMID: 34835166 PMCID: PMC8618450 DOI: 10.3390/vaccines9111235] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 01/24/2023] Open
Abstract
ccJE+Advax is an inactivated cell culture Japanese encephalitis (JE) vaccine formulated with Advax, a novel polysaccharide adjuvant based on delta inulin. This vaccine has previously shown promise in murine and equine studies and the current study sought to better understand its mechanism of action and assess the feasibility of single dose vaccine protection. Mice immunised with ccJE+Advax had higher serum neutralisation titres than those immunised with ccJE alone or with alum adjuvant. ccJE+Advax induced extraordinarily broad cross-neutralising antibodies against multiple flaviviruses including West Nile virus (WNV), Murray Valley encephalitis virus (MVEV), St Louis encephalitis virus (SLEV) and Dengue virus-1 and -2 (DENV-1 and -2). Notably, the DENV-2 cross-neutralising antibodies from ccJE+Advax immunised mice uniquely had no DENV-2 antibody-dependent infection enhancement (ADIE) activity, in contrast to high ADIE activity seen with DENV-1 cross-reactive antibodies induced by mbJE or ccJE alone or with alum adjuvant. JEV-stimulated splenocytes from ccJE+Advax immunised mice showed increased IL-17 and IFN-γ production, consistent with a mixed Th1 and Th17 response, whereas ccJE-alum was associated with production of mainly Th2 cytokines. In a mouse lethal challenge study against highly virulent JaTH160 JEV strain, ccJE+Advax conferred complete protection in a two-dose schedule with 50 ng of vaccine antigen and near complete protection after a single 200 ng dose of vaccine antigen. There is an ongoing lack of human vaccines against particular flaviviruses, including WNV, SLEV and MVEV. Given its ability to provide single-dose JEV protection and induce broadly neutralising antibodies devoid of ADIE activity, ccJE+Advax vaccine could be useful in situations where rapid protection is desirable, e.g., during a local outbreak or for use in travellers or armies requiring rapid deployment to JEV endemic regions.
Collapse
|
12
|
Enhanced dengue vaccine virus replication and neutralizing antibody responses in immune primed rhesus macaques. NPJ Vaccines 2021; 6:77. [PMID: 34021159 PMCID: PMC8140083 DOI: 10.1038/s41541-021-00339-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/15/2021] [Indexed: 11/13/2022] Open
Abstract
Antibody-dependent enhancement (ADE) is suspected to influence dengue virus (DENV) infection, but the role ADE plays in vaccination strategies incorporating live attenuated virus components is less clear. Using a heterologous prime-boost strategy in rhesus macaques, we examine the effect of priming with DENV purified inactivated vaccines (PIVs) on a tetravalent live attenuated vaccine (LAV). Sera exhibited low-level neutralizing antibodies (NAb) post PIV priming, yet moderate to high in vitro ADE activity. Following LAV administration, the PIV primed groups exhibited DENV-2 LAV peak viremias up to 1,176-fold higher than the mock primed group, and peak viremia correlated with in vitro ADE. Furthermore, PIV primed groups had more balanced and higher DENV-1–4 NAb seroconversion and titers than the mock primed group following LAV administration. These results have implications for the development of effective DENV vaccine prime-boost strategies and for our understanding of the role played by ADE in modulating DENV replication.
Collapse
|
13
|
Luczo JM, Bousse T, Johnson SK, Jones CA, Pearce N, Neiswanger CA, Wang MX, Miller EA, Petrovsky N, Wentworth DE, Bronshtein V, Papania M, Tompkins SM. Intranasal powder live attenuated influenza vaccine is thermostable, immunogenic, and protective against homologous challenge in ferrets. NPJ Vaccines 2021; 6:59. [PMID: 33883559 PMCID: PMC8060263 DOI: 10.1038/s41541-021-00320-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Influenza viruses cause annual seasonal epidemics and sporadic pandemics; vaccination is the most effective countermeasure. Intranasal live attenuated influenza vaccines (LAIVs) are needle-free, mimic the natural route of infection, and elicit robust immunity. However, some LAIVs require reconstitution and cold-chain requirements restrict storage and distribution of all influenza vaccines. We generated a dry-powder, thermostable LAIV (T-LAIV) using Preservation by Vaporization technology and assessed the stability, immunogenicity, and efficacy of T-LAIV alone or combined with delta inulin adjuvant (Advax™) in ferrets. Stability assays demonstrated minimal loss of T-LAIV titer when stored at 25 °C for 1 year. Vaccination of ferrets with T-LAIV alone or with delta inulin adjuvant elicited mucosal antibody and robust serum HI responses in ferrets, and was protective against homologous challenge. These results suggest that the Preservation by Vaporization-generated dry-powder vaccines could be distributed without refrigeration and administered without reconstitution or injection. Given these significant advantages for vaccine distribution and delivery, further research is warranted.
Collapse
Affiliation(s)
- Jasmina M Luczo
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Emory-UGA Centers of Excellence for Influenza Research and Surveillance (CEIRS), Athens, Georgia, 30602, USA
| | - Tatiana Bousse
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Scott K Johnson
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Cheryl A Jones
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Nicholas Pearce
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Carlie A Neiswanger
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Min-Xuan Wang
- Universal Stabilization Technologies, Inc., San Diego, California, USA
| | - Erin A Miller
- Universal Stabilization Technologies, Inc., San Diego, California, USA
| | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
- Vaxine Pty Ltd, Warradale, South Australia, Australia
| | - David E Wentworth
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Victor Bronshtein
- Universal Stabilization Technologies, Inc., San Diego, California, USA
| | - Mark Papania
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Stephen M Tompkins
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA.
- Emory-UGA Centers of Excellence for Influenza Research and Surveillance (CEIRS), Athens, Georgia, 30602, USA.
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
14
|
Advax-CpG Adjuvant Provides Antigen Dose-Sparing and Enhanced Immunogenicity for Inactivated Poliomyelitis Virus Vaccines. Pathogens 2021; 10:pathogens10050500. [PMID: 33919442 PMCID: PMC8143488 DOI: 10.3390/pathogens10050500] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/17/2022] Open
Abstract
Global immunization campaigns have resulted in a major decline in the global incidence of polio cases, with wild-type poliovirus remaining endemic in only two countries. Live oral polio vaccine (OPV) played a role in the reduction in polio case numbers; however, the risk of OPV developing into circulating vaccine-derived poliovirus makes it unsuitable for eradication programs. Trivalent inactivated polio virus (TIPV) vaccines which contain formalin-inactivated antigens produced from virulent types 1, 2 and 3 reference polio strains grown in Vero monkey kidney cells have been advocated as a replacement for OPV; however, TIPVs have weak immunogenicity and multiple boosts are required before peak neutralizing titers are reached. This study examined whether the incorporation of the novel polysaccharide adjuvant, Advax-CpG, could boost the immunogenicity of two TIPV vaccines, (i) a commercially available polio vaccine (IPOL®, Sanofi Pasteur) and (ii) a new TIPV formulation developed by Statens Serum Institut (SSI). Mice were immunized intramuscularly based on recommended vaccine dosage schedules and serum antibody titers were followed for 12 months post-immunization. Advax-CpG significantly enhanced the long-term immunogenicity of both TIPV vaccines and had at least a 10-fold antigen dose-sparing effect. An exception was the poor ability of the SSI TIPV to induce serotype type 1 neutralizing antibodies. Immunization with monovalent IPVs suggested that the low type 1 response to TIPV may be due to antigen competition when the type 1 antigen was co-formulated with the type 2 and 3 antigens. This study provides valuable insights into the complexity of the formulation of multivalent polio vaccines and supports the further development of adjuvanted antigen-sparing TIPV vaccines in the fight to eradicate polio.
Collapse
|
15
|
Behzadi M, Vakili B, Ebrahiminezhad A, Nezafat N. Iron nanoparticles as novel vaccine adjuvants. Eur J Pharm Sci 2021; 159:105718. [PMID: 33465476 DOI: 10.1016/j.ejps.2021.105718] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022]
Abstract
The poor immunogenicity of peptide vaccines compared to conventional ones re usually improved by applying different adjuvants. As chemical or biological substances, adjuvants are added to vaccines to enhance and prolong the immune response. According to considerable investigations over the recent years in the context of finding new adjuvants, a handful of vaccine adjuvants have been licensed for human use. Recently, engineered nanoparticles (NPs) have been introduced as novel alternatives to traditional vaccine adjuvant. Metallic nanoparticles (MeNPs) are among the most promising NPs used for vaccine adjuvant as well as the delivery system that can improve immune responses against pathogens. Iron NPs, as an important class of MeNPs, have gained increasing attention as novel vaccine adjuvants. These particles have shown acceptable results in preclinical studies. Hence, understanding the physicochemical properties of iron NPs, including size, surface properties, charge and route of administration, is of substantial importance. The aim of this review is to provide an overview of the immunomodulatory effects of iron NPs as novel adjuvants. Furthermore, physicochemical properties of these NPs were also discussed.
Collapse
Affiliation(s)
- Maryam Behzadi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Vakili
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Ebrahiminezhad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
16
|
Abstract
A DNA vaccine is a plasmid encoding a vaccine antigen together with an efficient eukaryotic promoter to drive protein expression. The chief problem of DNA vaccines has been their suboptimal immunogenicity in humans. Many different flaviviruses infect and cause serious illness and even death in humans, but human vaccines are not available against most of the relevant flaviviruses with the exception of Japanese encephalitis virus. DNA vaccines are easy and fast to produce at relatively low cost, do not require handling of dangerous pathogens, are stable at room temperature allowing for low-cost storage and transportation, and are highly versatile, allowing for rapid changes in coding sequence design and synthesis. This makes a DNA vaccine approach ideally suited for development as a broad-based flavivirus vaccine platform. However, to be useful as a flavivirus prophylactic vaccine platform in humans, a method would need to be found to enhance DNA vaccine immunogenicity without the need for the cumbersome and expensive equipment involved with electroporation. We describe here a protocol used to test different adjuvants with flavivirus DNA vaccines to determine an optimal formulation. An optimal regimen involving a DNA adjuvanted vaccine prime followed by an adjuvanted protein vaccine boost is described and can be applied by readers to solve barriers to the development of other DNA vaccines where immunogenicity is a problem.
Collapse
|
17
|
Gupta T, Gupta SK. Potential adjuvants for the development of a SARS-CoV-2 vaccine based on experimental results from similar coronaviruses. Int Immunopharmacol 2020; 86:106717. [PMID: 32585611 PMCID: PMC7301105 DOI: 10.1016/j.intimp.2020.106717] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
The extensive efforts around the globe are being made to develop a suitable vaccine against COVID-19 (Coronavirus Disease-19) caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus-2). An effective vaccine should be able to induce high titers of neutralizing antibodies to prevent the virus from attaching to the host cell receptors. However, to elicit the protective levels of antibodies, a vaccine may require multiple doses or assistance from other immunostimulatory molecules. Further, the vaccine should be able to induce protective levels of antibodies rapidly with the least amount of antigen used. This decreases the cost of a vaccine and makes it affordable. As the pandemic has hit most countries across the globe, there will be an overwhelming demand for the vaccine in a quick time. Incorporating a suitable adjuvant in a SARS-CoV-2 vaccine may address these requirements. This review paper will discuss the experimental results of the adjuvanted vaccine studies with similar coronaviruses (CoVs) which might be useful to select an appropriate adjuvant for a vaccine against rapidly emergingSARS-CoV-2. We also discuss the current progress in the development of adjuvanted vaccines against the disease.
Collapse
Affiliation(s)
- Tania Gupta
- Dr GC Negi College of Veterinary and Animal Sciences, Palampur 176062, Himachal Pradesh, India.
| | - Shishir K Gupta
- CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| |
Collapse
|
18
|
Li N, Zhang ZR, Zhang YN, Liu J, Deng CL, Shi PY, Yuan ZM, Ye HQ, Zhang B. A replication-defective Japanese encephalitis virus (JEV) vaccine candidate with NS1 deletion confers dual protection against JEV and West Nile virus in mice. NPJ Vaccines 2020; 5:73. [PMID: 32802412 PMCID: PMC7406499 DOI: 10.1038/s41541-020-00220-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/14/2020] [Indexed: 11/09/2022] Open
Abstract
In our previous study, we have demonstrated in the context of WNV-ΔNS1 vaccine (a replication-defective West Nile virus (WNV) lacking NS1) that the NS1 trans-complementation system may offer a promising platform for the development of safe and efficient flavivirus vaccines only requiring one dose. Here, we produced high titer (107 IU/ml) replication-defective Japanese encephalitis virus (JEV) with NS1 deletion (JEV-ΔNS1) in the BHK-21 cell line stably expressing NS1 (BHKNS1) using the same strategy. JEV-ΔNS1 appeared safe with a remarkable genetic stability and high degrees of attenuation of in vivo neuroinvasiveness and neurovirulence. Meanwhile, it was demonstrated to be highly immunogenic in mice after a single dose, providing similar degrees of protection to SA14-14-2 vaccine (a most widely used live attenuated JEV vaccine), with healthy condition, undetectable viremia and gradually rising body weight. Importantly, we also found JEV-ΔNS1 induced robust cross-protective immune responses against the challenge of heterologous West Nile virus (WNV), another important member in the same JEV serocomplex, accounting for up to 80% survival rate following a single dose of immunization relative to mock-vaccinated mice. These results not only support the identification of the NS1-deleted flavivirus vaccines with a satisfied balance between safety and efficacy, but also demonstrate the potential of the JEV-ΔNS1 as an alternative vaccine candidate against both JEV and WNV challenge.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Zhe-Rui Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Ya-Nan Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Jing Liu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Zhi-Ming Yuan
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Han-Qing Ye
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,Drug Discovery Center for Infectious Disease, Nankai University, 300350 Tianjin, China
| |
Collapse
|
19
|
Wang L, Song Y, Parikh A, Joyce P, Chung R, Liu L, Afinjuomo F, Hayball JD, Petrovsky N, Barclay TG, Garg S. Doxorubicin-Loaded Delta Inulin Conjugates for Controlled and Targeted Drug Delivery: Development, Characterization, and In Vitro Evaluation. Pharmaceutics 2019; 11:pharmaceutics11110581. [PMID: 31698755 PMCID: PMC6920814 DOI: 10.3390/pharmaceutics11110581] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
Delta inulin, also known as microparticulate inulin (MPI), was modified by covalently attaching doxorubicin to its nanostructured surface for use as a targeted drug delivery vehicle. MPI is readily endocytosed by monocytes, macrophages, and dendritic cells and in this study, we sought to utilize this property to develop a system to target anti-cancer drugs to lymphoid organs. We investigated, therefore, whether MPI could be used as a vehicle to deliver doxorubicin selectively, thereby reducing the toxicity of this antibiotic anthracycline drug. Doxorubicin was covalently attached to the surface of MPI using an acid–labile linkage to enable pH-controlled release. The MPI-doxorubicin conjugate was characterized using FTIR and SEM, confirming covalent attachment and indicating doxorubicin coupling had no obvious impact on the physical nanostructure, integrity, and cellular uptake of the MPI particles. To simulate the stability of the MPI-doxorubicin in vivo, it was stored in artificial lysosomal fluid (ALF, pH 4.5). Although the MPI-doxorubicin particles were still visible after 165 days in ALF, 53% of glycosidic bonds in the inulin particles were hydrolyzed within 12 days in ALF, reflected by the release of free glucose into solution. By contrast, the fructosidic bonds were much more stable. Drug release studies of the MPI-doxorubicin in vitro, demonstrated a successful pH-dependent controlled release effect. Confocal laser scanning microscopy studies and flow cytometric analysis confirmed that when incubated with live cells, MPI-doxorubicin was efficiently internalized by immune cells. An assay of cell metabolic activity demonstrated that the MPI carrier alone had no toxic effects on RAW 264.7 murine monocyte/macrophage-like cells, but exhibited anti-cancer effects against HCT116 human colon cancer cells. MPI-doxorubicin had a greater anti-cancer cell effect than free doxorubicin, particularly when at lower concentrations, suggesting a drug-sparing effect. This study establishes that MPI can be successfully modified with doxorubicin for chemotherapeutic drug delivery.
Collapse
Affiliation(s)
- Lixin Wang
- Centre for Pharmaceutical Innovation and Development, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5000, Australia; (L.W.); (Y.S.); (A.P.); (R.C.); (F.A.); (T.G.B.)
| | - Yunmei Song
- Centre for Pharmaceutical Innovation and Development, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5000, Australia; (L.W.); (Y.S.); (A.P.); (R.C.); (F.A.); (T.G.B.)
| | - Ankit Parikh
- Centre for Pharmaceutical Innovation and Development, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5000, Australia; (L.W.); (Y.S.); (A.P.); (R.C.); (F.A.); (T.G.B.)
| | - Paul Joyce
- Division of Biological Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden;
| | - Rosa Chung
- Centre for Pharmaceutical Innovation and Development, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5000, Australia; (L.W.); (Y.S.); (A.P.); (R.C.); (F.A.); (T.G.B.)
| | - Liang Liu
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Adelaide SA 5000, Australia; (L.L.); (J.D.H.)
| | - Franklin Afinjuomo
- Centre for Pharmaceutical Innovation and Development, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5000, Australia; (L.W.); (Y.S.); (A.P.); (R.C.); (F.A.); (T.G.B.)
| | - John D. Hayball
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Adelaide SA 5000, Australia; (L.L.); (J.D.H.)
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide SA 5005, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Bedford Park, Adelaide 5042, Australia;
- Department of Diabetes and Endocrinology, Flinders University, Adelaide 5042, Australia
| | - Thomas G. Barclay
- Centre for Pharmaceutical Innovation and Development, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5000, Australia; (L.W.); (Y.S.); (A.P.); (R.C.); (F.A.); (T.G.B.)
| | - Sanjay Garg
- Centre for Pharmaceutical Innovation and Development, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5000, Australia; (L.W.); (Y.S.); (A.P.); (R.C.); (F.A.); (T.G.B.)
- Correspondence: ; Tel.: +61-8-8302-1067
| |
Collapse
|
20
|
Gene-edited vero cells as rotavirus vaccine substrates. Vaccine X 2019; 3:100045. [PMID: 31660537 PMCID: PMC6806661 DOI: 10.1016/j.jvacx.2019.100045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/30/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022] Open
Abstract
Background Rotavirus (RV) is a leading cause of severe gastroenteritis globally and can cause substantial morbidity associated with gastroenteritis in children <5 years of age. Orally administered live-attenuated RV vaccines offer protection against disease but vaccination efforts have been hampered by high manufacturing costs and the need to maintain a cold chain. Methods A subset of Vero cell host genes was identified by siRNA that when knocked down increased RV replication and these anti-viral host genes were individually deleted using CRISPR-Cas9. Results Fully-sequenced gene knockout Vero cell substrates were assessed for increased RV replication and RV vaccine antigen expression compared to wild type Vero cells. The results showed that RV replication and antigen production were logs higher in Vero cells having an EMX2 gene deletion compared to other Vero cell substrates tested. Conclusions We used siRNAs to screen for host genes that negatively affected RV replication, then CRISPR-Cas9 gene editing to delete select genes. The gene editing led to the development of enhanced RV vaccine substrates supporting a potential path forward for improving RV vaccine production.
Collapse
|
21
|
Afinjuomo F, Barclay TG, Parikh A, Song Y, Chung R, Wang L, Liu L, Hayball JD, Petrovsky N, Garg S. Design and Characterization of Inulin Conjugate for Improved Intracellular and Targeted Delivery of Pyrazinoic Acid to Monocytes. Pharmaceutics 2019; 11:E243. [PMID: 31121836 PMCID: PMC6572292 DOI: 10.3390/pharmaceutics11050243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/10/2019] [Accepted: 05/16/2019] [Indexed: 11/16/2022] Open
Abstract
The propensity of monocytes to migrate into sites of mycobacterium tuberculosis (TB) infection and then become infected themselves makes them potential targets for delivery of drugs intracellularly to the tubercle bacilli reservoir. Conventional TB drugs are less effective because of poor intracellular delivery to this bacterial sanctuary. This study highlights the potential of using semicrystalline delta inulin particles that are readily internalised by monocytes for a monocyte-based drug delivery system. Pyrazinoic acid was successfully attached covalently to the delta inulin particles via a labile linker. The formation of new conjugate and amide bond was confirmed using zeta potential, Proton Nuclear Magnetic Resonance (1HNMR) and Fourier transform infrared spectroscopy (FTIR). Scanning electron microscopy (SEM) confirmed that no significant change in size after conjugation which is an important parameter for monocyte targeting. Thermogravimetric analysis (TGA) and differential scanning calorimetry (DSC) were used to establish the change in thermal properties. The analysis of in-vitro release demonstrated pH-triggered drug cleavage off the delta inulin particles that followed a first-order kinetic process. The efficient targeting ability of the conjugate for RAW 264.7 monocytic cells was supported by cellular uptake studies. Overall, our finding confirmed that semicrystalline delta inulin particles (MPI) can be modified covalently with drugs and such conjugates allow intracellular drug delivery and uptake into monocytes, making this system potentially useful for the treatment of TB.
Collapse
Affiliation(s)
- Franklin Afinjuomo
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Thomas G Barclay
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Ankit Parikh
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Yunmei Song
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Rosa Chung
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Lixin Wang
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Liang Liu
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - John D Hayball
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| | - Nikolai Petrovsky
- Vaxine Pty. Ltd., Adelaide, SA 5042, Australia.
- Department of Endocrinology, Flinders University, Adelaide, SA 5042, Australia.
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia.
| |
Collapse
|
22
|
Weir C, Oksa A, Millar J, Alexander M, Kynoch N, Walton-Weitz Z, Mackenzie-Wood P, Tam F, Richards H, Naylor R, Cheng K, Bennett P, Petrovsky N, Allavena R. The Safety of an Adjuvanted Autologous Cancer Vaccine Platform in Canine Cancer Patients. Vet Sci 2018; 5:vetsci5040087. [PMID: 30322015 PMCID: PMC6313922 DOI: 10.3390/vetsci5040087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 10/02/2018] [Accepted: 10/10/2018] [Indexed: 12/01/2022] Open
Abstract
Canine cancer rates are similar to humans, though the therapeutic options might be limited. Inducing a patient’s own immune system to have an anti-tumor response is an attractive approach to cancer therapy. In this safety study, autologous tumor vaccines produced specifically for each canine patient were combined with Advax™, a novel non-inflammatory immunomodulator and vaccine adjuvant and were tested for safety in a diverse range of patient presentations alone or in combination with other treatments. Canine patients had their tumor biopsied, debulked or resected and the tumor antigens were processed into an autologous vaccine formulated with Advax™ adjuvant with or without rhizavidin as an additional immune stimulant. Patients treated early in the trial received two intramuscular (IM) doses, 2 weeks apart. As the study progressed and no issues of safety were observed, the protocol was changed to weekly vaccinations for 4 weeks followed by monthly booster shots. Over the 150 I.M injections delivered to date, the vaccine was found to be very safe and no significant adverse reactions were observed. These results justify ongoing development and future controlled studies of this autologous vaccine approach.
Collapse
Affiliation(s)
- Chris Weir
- Northern Blood Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital and the Sydney Medical School, University of Sydney, Sydney NSW 2065, Australia.
| | - Annika Oksa
- School of Veterinary Science, The University of Queensland, Gatton Campus, Gatton, QLD 4343 Australia.
| | | | | | - Nicola Kynoch
- Willougby Veterinary Hospital, Sydney, NSW 2068, Australia.
| | | | | | - Felicia Tam
- Castle Hill Veterinary Hospital, Sydney, NSW 2154, Australia.
| | - Hope Richards
- Willougby Veterinary Hospital, Sydney, NSW 2068, Australia.
| | - Richard Naylor
- Castle Hill Veterinary Hospital, Sydney, NSW 2154, Australia.
| | - Katrina Cheng
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, NSW 2006, Australia.
| | - Peter Bennett
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, NSW 2006, Australia.
| | - Nikolai Petrovsky
- Flinders University Bedford Park, Adelaide, SA 5042, Australia.
- Vaxine Pty Ltd., Bedford Park, Adelaide, SA 5042, Australia.
| | - Rachel Allavena
- School of Veterinary Science, The University of Queensland, Gatton Campus, Gatton, QLD 4343 Australia.
| |
Collapse
|
23
|
Barzon L, Palù G. Recent developments in vaccines and biological therapies against Japanese encephalitis virus. Expert Opin Biol Ther 2018; 18:851-864. [DOI: 10.1080/14712598.2018.1499721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Luisa Barzon
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
24
|
Nerome K, Yamaguchi R, Fuke N, Izzati UZ, Maegawa K, Sugita S, Kawasaki K, Kuroda K, Nerome R. Development of a Japanese encephalitis virus genotype V virus-like particle vaccine in silkworms. J Gen Virol 2018; 99:897-907. [PMID: 29877787 DOI: 10.1099/jgv.0.001081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
To counter the spread of multiple Japanese encephalitis virus (JEV) variants harboured in alternative host species and highly neurotoxic variants with new antigenicity, such as genotype V (Muar), methods for developing more effective and low-cost vaccines against a variety of epidemic JEV strains are required. Here, we successfully synthesized large amounts of a Muar virus-like particle (MVLP) vaccine for JEV in silkworm pupae by using a Bombyx mori nuclear polyhedrosis virus recombinant consisting of JEV codon-optimized envelope (E) DNA. In particular, histopathological examination suggested that MVLP was efficiently synthesized in body fat tissues as well as epithelial cells. Quantitative analysis indicated that one silkworm pupa produced 724.8 µg of E protein in the MVLP vaccine. Electron microscopic examination of purified MVLP vaccine defined a typical MVLP morphological structure. Detailed MVLP antigen assessment by immune-electron microscopy revealed that the majority of MVLPs were covered with approximately 10 nm projections. Boosted immunization with MVLP antigens in mice and rabbits tended to show improved plaque inhibition potency against homologous Muar and heterologous Nakayama, but less potency to Beijing-1 strains. Notably, mixed immune rabbit antisera against Nakayama and Muar VLP antigens led to an increase in the low antibody reaction to Beijing-1. Additionally, a stopgap divalent JEV vaccine consisting of MVLP and Nakayama VLP and its immune mouse serum significantly increased plaque inhibition titre against Muar, Nakayama and Beijing-1 strains. These findings suggested that low-cost MVLP vaccines prepared in silkworm pupae are suitable for providing simultaneous protection of individuals in developing countries against various JEV strains.
Collapse
Affiliation(s)
| | - Ryoji Yamaguchi
- Laboratory of Veterinary Pathology, Department of Veterinary, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Naoyuki Fuke
- Laboratory of Veterinary Pathology, Department of Veterinary, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Uda Zahli Izzati
- Laboratory of Veterinary Pathology, Department of Veterinary, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | | | - Shigeo Sugita
- Equine Research Institute, Japan Racing Association, Tochigi, Japan
| | - Kazunori Kawasaki
- National Institute of Advanced Science and Technology (AIST), Osaka, Japan
| | - Kazumichi Kuroda
- Division of Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Reiko Nerome
- The Institute of Biological Resources, Okinawa, Japan
| |
Collapse
|
25
|
Counoupas C, Pinto R, Nagalingam G, Britton WJ, Petrovsky N, Triccas JA. Delta inulin-based adjuvants promote the generation of polyfunctional CD4 + T cell responses and protection against Mycobacterium tuberculosis infection. Sci Rep 2017; 7:8582. [PMID: 28819247 PMCID: PMC5561132 DOI: 10.1038/s41598-017-09119-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/19/2017] [Indexed: 01/16/2023] Open
Abstract
There is an urgent need for the rational design of safe and effective vaccines to protect against chronic bacterial pathogens such as Mycobacterium tuberculosis. Advax™ is a novel adjuvant based on delta inulin microparticles that enhances immunity with a minimal inflammatory profile and has entered human trials to protect against viral pathogens. In this report we determined if Advax displays broad applicability against important human pathogens by assessing protective immunity against infection with M. tuberculosis. The fusion protein CysVac2, comprising the M. tuberculosis antigens Ag85B (Rv1886c) and CysD (Rv1285) formulated with Advax provided significant protection in the lungs of M. tuberculosis-infected mice. Protection was associated with the generation of CysVac2-specific multifunctional CD4+ T cells (IFN-γ+TNF+IL-2+). Addition to Advax of the TLR9 agonist, CpG oligonucleotide (AdvaxCpG), improved both the immunogenicity and protective efficacy of CysVac2. Immunisation with CysVac2/AdvaxCpG resulted in heightened release of the chemoattractants, CXCL1, CCL3, and TNF, and rapid influx of monocytes and neutrophils to the site of vaccination, with pronounced early priming of CysVac2-specific CD4+ T cells. As delta inulin adjuvants have shown an excellent safety and tolerability profile in humans, CysVac2/AdvaxCpG is a strong candidate for further preclinical evaluation for progression to human trials.
Collapse
Affiliation(s)
- Claudio Counoupas
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia
| | - Rachel Pinto
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia
| | - Gayathri Nagalingam
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia
| | - Warwick J Britton
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia
| | - Nikolai Petrovsky
- Department of Endocrinology, Flinders University, Adelaide, Australia
- Vaxine Pty Ltd, Flinders Medical Centre, Adelaide, Australia
| | - James A Triccas
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia.
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia.
| |
Collapse
|
26
|
Investigation of the biodistribution, breakdown and excretion of delta inulin adjuvant. Vaccine 2017; 35:4382-4388. [PMID: 28676380 DOI: 10.1016/j.vaccine.2017.06.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 01/08/2023]
Abstract
Insoluble, nanostructured delta inulin particles enhance the immunogenicity of co-administered protein antigens and consequently are used as a vaccine adjuvant (Advax™). To better understand their immunomodulatory properties, the in vitro hydrolysis and in vivo distribution of delta inulin particles were investigated. Delta inulin particle hydrolysis under bio-relevant acidic conditions resulted in no observable change to the bulk morphology using SEM, and HPLC results showed that only 6.1% of the inulin was hydrolysed over 21days. However, 65% of the terminal glucose groups were released, showing that acid hydrolysis relatively rapidly releases surface bound chemistries. This was used to explain in vivo biodistribution results in which delta inulin particles surface-labelled with fluorescein-5-thiosemicabizide were administered to mice using intramuscular (I.M.) or subcutaneous (S.C.) routes. Comparison analysis of the fluorescence of soluble inulin in the supernatants of homogenised tissues maintained at room temperature or heated to 100°C to solubilise particulate inulin was used to distinguish between fluorescent probe on soluble inulin and probe bound to inulin within particles. Following both I.M. and S.C. injection delta inulin exhibited a depot behaviour with local injection site residence for several weeks. Over this time, as injection site inulin reduced, there was measurable transport of intact delta inulin particles by macrophages to secondary lymphoid organs and the liver. Ultimately, the injected delta inulin became solubilised resulting in its detection in the plasma and in the urine. Thus injected delta inulin particles are initially taken up by macrophages at the site of injection, trafficked to secondary lymphoid tissue and the liver, and hydrolysed resulting in their becoming soluble and diffusing into the blood stream, from whence they are glomerularly filtered and excreted into the urine. These results provide important insights into the biodistribution of I.M. or S.C. injected delta inulin particles when used as vaccine adjuvants and their method of excretion.
Collapse
|
27
|
Matsuda S, Nerome R, Maegawa K, Kotaki A, Sugita S, Kawasaki K, Kuroda K, Yamaguchi R, Takasaki T, Nerome K. Development of a Japanese encephalitis virus-like particle vaccine in silkworms using codon-optimised prM and envelope genes. Heliyon 2017; 3:e00286. [PMID: 28435908 PMCID: PMC5390689 DOI: 10.1016/j.heliyon.2017.e00286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/03/2017] [Accepted: 03/28/2017] [Indexed: 11/29/2022] Open
Abstract
We have successfully prepared a Japanese encephalitis virus (JEV) - Nakayama virus like particle (NVLP) vaccine using synthetic codon-optimized prM and E genes. The expression of the recombinant JEV Nakayama-BmNPV (JEV-NNPV) virus was determined in infected silkworm Bm-N cells by fluorescence and Western blot analysis. The recombinant was inoculated into silkworm pupae and the yield of Nakayama VLP (NVLP) reached a peak in the homogenates after 3 days. Additionally, in the peptide analysis of infected pupae homogenate, it appeared approximately 300-500 μg E protein/pupa were produced. When purified the above eluates on the discontinuous sucrose density gradient centrifugation, NVLP showed a strong hemagglutination (HA) activity by using chicken red blood cell in phosphate-buffered saline (PBS) free from Mg++ and Ca++ ions. The immune antisera against NVLP strain could efficiently neutralize the plaque formation of Nakayama, Beijing-1 and Muar strains, showing tendency of much higher reaction with heterologous Muar strain than homologous Nakayama strain. Our findings suggest that the JEV-NVLP may be useful for JEV epidemic control in many endemic areas of Asian countries as a widely effective and less expensive JE vaccine.
Collapse
Affiliation(s)
- Sayaka Matsuda
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Reiko Nerome
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Kenichi Maegawa
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Akira Kotaki
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Shigeo Sugita
- Equine Research Institute, Japan Racing Association, 1400-4, Shiba, Shimotsuke-shi, Tochigi 329-0412, Japan
| | - Kazunori Kawasaki
- National Institute of Advanced Science and Technology (AIST), 1-8-31, Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Kazumichi Kuroda
- Division of Microbiology, Nihon University School of Medicine, 30-1, Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Ryoji Yamaguchi
- Laboratory of Veterinary Pathology, Department of Veterinary, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuenkibanadai-Nishi, Miyazaki 889-2192, Japan
| | - Tomohiko Takasaki
- Department of Virology 1, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Kuniaki Nerome
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| |
Collapse
|
28
|
|
29
|
Vaccine Adjuvant Nanotechnologies. MICRO AND NANOTECHNOLOGY IN VACCINE DEVELOPMENT 2017. [PMCID: PMC7151801 DOI: 10.1016/b978-0-323-39981-4.00007-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
The increasing sophistication of vaccine adjuvant design has been driven by improved understanding of the importance of nanoscale features of adjuvants to their immunological function. Newly available advanced nanomanufacturing techniques now allow very precise control of adjuvant particle size, shape, texture, and surface chemistry. Novel adjuvant concepts include self-assembling particles and targeted immune delivery. These individual concepts can be combined to create a single integrated vaccine nanoparticle-combining antigen, adjuvants, and DC-targeting elements. In the process, the concept of an adjuvant has broadened to include not only immune-stimulatory substances but also any design features that enhance the immune response against the relevant vaccine antigen. The modern definition of an adjuvant includes not only classical immune stimulators but also any aspects of particle size, shape, and surface chemistry that enhance vaccine immunogenicity. It even includes purely physical processes such as texturing of particle surfaces to maximize immunogenicity. Looking forward, adjuvants will increasingly be seen not as separate add-on items but as wholly integrated elements of a complete vaccine delivery package. Hence, vaccine systems will increasingly approach the complexity and sophistication of pathogens themselves, incorporating highly specific particle properties, contents, and behaviors, all designed to maximize immune system recognition and drive the immune response in the specific direction that affords maximal protection.
Collapse
|
30
|
Wong TM, Petrovsky N, Bissel SJ, Wiley CA, Ross TM. Delta inulin-derived adjuvants that elicit Th1 phenotype following vaccination reduces respiratory syncytial virus lung titers without a reduction in lung immunopathology. Hum Vaccin Immunother 2016; 12:2096-2105. [PMID: 27215855 PMCID: PMC4994749 DOI: 10.1080/21645515.2016.1162931] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/17/2016] [Accepted: 03/02/2016] [Indexed: 10/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a significant cause of lower respiratory tract infections resulting in bronchiolitis and even mortality in the elderly and young children/infants. Despite the impact of this virus on human health, no licensed vaccine exists. Unlike many other viral infections, RSV infection or vaccination does not induce durable protective antibodies in humans. In order to elicit high titer, neutralizing antibodies against RSV, we investigated the use of the adjuvant Advax™, a novel polysaccharide adjuvant based on delta inulin microparticles, to enhance antibody titers following vaccination. BALB/c mice were vaccinated intramuscularly with live RSV as a vaccine antigen in combination with one of two formulations of Advax™. Advax-1 was comprised of the standard delta inulin adjuvant and Advax-2 was formulated delta inulin plus CpG oligodendronucleotides (ODNs). An additional group of mice were either mock vaccinated, immunized with vaccine only, or administered vaccine plus Imject Alum. Following 3 vaccinations, mice had neutralizing antibody titers that correlated with reduction in viral titers in the lungs. Advax-1 significantly enhanced serum RSV-specific IgG1 levels at week 6 indicative of a Th2 response, similar to titers in mice administered vaccine plus Imject Alum. In contrast, mice vaccinated with vaccine plus Advax-2 had predominately IgG2a titers indicative of a Th1 response that was maintained during the entire study. Interestingly, regardless of which AdvaxTM adjuvant was used, the neutralizing titers were similar between groups, but the viral lung titers were significantly lower (∼10E+3pfu/g) in mice administered vaccine with either AdvaxTM adjuvant compared to mice administered adjuvants only. The lung pathology in vaccinated mice with AdvaxTM was similar to Imject Alum. Overall, RSV vaccine formulated with AdvaxTM had high neutralizing antibody titers with low lung viral titers, but exacerbated lung pathology compared to unvaccinated mice.
Collapse
Affiliation(s)
- Terianne M. Wong
- Center for Vaccines and Immunology, Department of Infectious Diseases, University of Georgia, Athens, GA USA
| | | | | | - Clayton A. Wiley
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, Department of Infectious Diseases, University of Georgia, Athens, GA USA
| |
Collapse
|
31
|
Rosales-Mendoza S, Salazar-González JA, Decker EL, Reski R. Implications of plant glycans in the development of innovative vaccines. Expert Rev Vaccines 2016; 15:915-25. [DOI: 10.1586/14760584.2016.1155987] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, Mexico
| | - Jorge A. Salazar-González
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, Mexico
| | - Eva L. Decker
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, Freiburg, Germany
| | - Ralf Reski
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, Freiburg, Germany
- BIOSS – Centre for Biological Signalling Studies, Freiburg, Germany
- FRIAS – Freiburg Institute for Advanced Studies, Freiburg, Germany
| |
Collapse
|
32
|
Higuchi A, Toriniwa H, Komiya T, Nakayama T. Recombinant Measles AIK-C Vaccine Strain Expressing the prM-E Antigen of Japanese Encephalitis Virus. PLoS One 2016; 11:e0150213. [PMID: 26930411 PMCID: PMC4773129 DOI: 10.1371/journal.pone.0150213] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 02/10/2016] [Indexed: 01/04/2023] Open
Abstract
An inactivated Japanese encephalitis virus (JEV) vaccine, which induces neutralizing antibodies, has been used for many years in Japan. In the present study, the JEV prM-E protein gene was cloned, inserted at the P/M junction of measles AIK-C cDNA, and an infectious virus was recovered. The JEV E protein was expressed in B95a cells infected with the recombinant virus. Cotton rats were inoculated with recombinant virus. Measles PA antibodies were detected three weeks after immunization. Neutralizing antibodies against JEV developed one week after inoculation, and EIA antibodies were detected three weeks after immunization. The measles AIK-C-based recombinant virus simultaneously induced measles and JEV immune responses, and may be a candidate for infant vaccines. Therefore, the present strategy of recombinant viruses based on a measles vaccine vector would be applicable to the platform for vaccine development.
Collapse
Affiliation(s)
- Akira Higuchi
- Kitasato-Daiichi Sankyo Vaccine, Division of Vaccine Production, Kitamoto City, Saitama Prefecture, 364–0026, Japan
| | - Hiroko Toriniwa
- Kitasato-Daiichi Sankyo Vaccine, Division of Vaccine Development, Kitamoto City, Saitama Prefecture, 364–0026, Japan
| | - Tomoyoshi Komiya
- Kitasato-Daiichi Sankyo Vaccine, Division of Vaccine Development, Kitamoto City, Saitama Prefecture, 364–0026, Japan
| | - Tetsuo Nakayama
- Kitasato Institute of Life Sciences, Laboratory of Viral Infection, Tokyo, 108–8641, Japan
- * E-mail:
| |
Collapse
|
33
|
Abstract
Given periodic outbreaks of fatal human infections caused by coronaviruses, development of an optimal coronavirus vaccine platform capable of rapid production is an ongoing priority. This chapter describes the use of an insect cell expression system for rapid production of a recombinant vaccine against severe acute respiratory syndrome coronavirus (SARS). Detailed methods are presented for expression, purification, and release testing of SARS recombinant spike protein antigen, followed by adjuvant formulation and animal testing. The methods herein described for rapid development of a highly protective SARS vaccine are equally suited to rapid development of vaccines against other fatal human coronavirus infections, e.g., the MERS coronavirus.
Collapse
|
34
|
Qian P, Zhi X, Wang B, Zhang H, Chen H, Li X. Construction and immune efficacy of recombinant pseudorabies virus expressing PrM-E proteins of Japanese encephalitis virus genotype І. Virol J 2015; 12:214. [PMID: 26651827 PMCID: PMC4676090 DOI: 10.1186/s12985-015-0449-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/04/2015] [Indexed: 01/24/2023] Open
Abstract
Background Japanese encephalitis (JE) is an arboviral disease with high case fatality rates and neurologic or psychiatric sequelae among survivors in Asia, western Pacific countries and northern Australia. Japanese encephalitis virus (JEV) is the cause of JE and the emergence of genotype І (GI) JEV has displaced genotype III (GIII) as the dominant strains circulating in some Asian regions. The currently available JE vaccines are safe and effective in preventing this disease, but they are developed based on the GIII JEV strains. Methods The recombinant virus PRV TK−/gE−/PrM-E+ which expressed the premembrane (prM) and envelope (E) proteins of JEV SX09S-01 strain (genotype I, GI) was constructed by homologous recombination between the genome of PRV TK−/gE−/LacZ+ digested with EcoRI and plasmid pIE-CAG-PrM-E-BGH. Expression of JEV PrM and E proteins was analyzed by Western blot analysis. Immune efficacy of PRV TK−/gE−/PrM-E+ was further evaluated in mouse model. Results A recombinant pseudorabies virus (PRV TK−/gE−/PrM-E+) was successfully constructed. Mice experiments showed that PRV TK−/gE−/PrM-E+ could induce a high level of ELISA antibodies against PRV and JEV, as well as high titer of PRV neutralizing antibodies. After challenge with 1 × 107 PFU virulent JEV SX09S-01 strain, the time of death was delayed and the survival rate was improved in PRV TK−/gE−/PrM-E+ vaccinated mice. Conclusions PRV TK−/gE−/PrM-E+ is a potential vaccine candidate against PRV and JEV GI infection in the future.
Collapse
Affiliation(s)
- Ping Qian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Key Laboratory of development of veterinary diagnostic products, Ministry of Agriculture, Wuhan, 430070, P.R China.
| | - Xianwei Zhi
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, P.R. China.
| | - Bo Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, P.R. China.
| | - Huawei Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, P.R. China.
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Key Laboratory of development of veterinary diagnostic products, Ministry of Agriculture, Wuhan, 430070, P.R China.
| | - Xiangmin Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, P.R. China. .,Key Laboratory of development of veterinary diagnostic products, Ministry of Agriculture, Wuhan, 430070, P.R China.
| |
Collapse
|
35
|
Petrovsky N, Cooper PD. Advax™, a novel microcrystalline polysaccharide particle engineered from delta inulin, provides robust adjuvant potency together with tolerability and safety. Vaccine 2015; 33:5920-6. [PMID: 26407920 PMCID: PMC4639457 DOI: 10.1016/j.vaccine.2015.09.030] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/06/2015] [Accepted: 09/11/2015] [Indexed: 12/19/2022]
Abstract
There is an ongoing need for new adjuvants to facilitate development of vaccines against HIV, tuberculosis, malaria and cancer, amongst many others. Unfortunately, the most potent adjuvants are often associated with toxicity and safety issues. Inulin, a plant-derived polysaccharide, has no immunological activity in its native soluble form but when crystallized into a stable microcrystalline particulate from (delta inulin) acquires potent adjuvant activity. Delta inulin has been shown to enhance humoral and cellular immune responses against a broad range of co-administered viral, bacterial, parasitic and toxin antigens. Inulin normally crystallizes as large heterogeneous particles with a broad size distribution and variable solubility temperatures. To ensure reproducible delta inulin particles with a consistent size distribution and temperature of solubility, a current Good Manufacturing Practice (cGMP) process was designed to produce Advax™ adjuvant. In its cCMP form, Advax™ adjuvant has proved successful in human trials of vaccines against seasonal and pandemic influenza, hepatitis B and insect sting anaphylaxis, enhancing antibody and T-cell responses while being safe and well tolerated. Advax™ adjuvant represents a novel human adjuvant that enhances both humoral and cellular immunity. This review describes the discovery and development of Advax™ adjuvant and research into its unique mechanism of action.
Collapse
Affiliation(s)
- Nikolai Petrovsky
- Vaxine Pty Ltd, Flinders Medical Centre, Adelaide, SA 5042, Australia; Department of Endocrinology, Flinders Medical Centre and Flinders University, Adelaide 5042, Australia.
| | - Peter D Cooper
- Vaxine Pty Ltd, Flinders Medical Centre, Adelaide, SA 5042, Australia; John Curtin School of Medical Research, Australian National University, Canberra 2061, Australia
| |
Collapse
|
36
|
Abstract
Use of highly pure antigens to improve vaccine safety has led to reduced vaccine immunogenicity and efficacy. This has led to the need to use adjuvants to improve vaccine immunogenicity. The ideal adjuvant should maximize vaccine immunogenicity without compromising tolerability or safety. Unfortunately, adjuvant research has lagged behind other vaccine areas such as antigen discovery, with the consequence that only a very limited number of adjuvants based on aluminium salts, monophosphoryl lipid A and oil emulsions are currently approved for human use. Recent strategic initiatives to support adjuvant development by the National Institutes of Health should translate into greater adjuvant choices in the future. Mechanistic studies have been valuable for better understanding of adjuvant action, but mechanisms of adjuvant toxicity are less well understood. The inflammatory or danger-signal model of adjuvant action implies that increased vaccine reactogenicity is the inevitable price for improved immunogenicity. Hence, adjuvant reactogenicity may be avoidable only if it is possible to separate inflammation from adjuvant action. The biggest remaining challenge in the adjuvant field is to decipher the potential relationship between adjuvants and rare vaccine adverse reactions, such as narcolepsy, macrophagic myofasciitis or Alzheimer's disease. While existing adjuvants based on aluminium salts have a strong safety record, there are ongoing needs for new adjuvants and more intensive research into adjuvants and their effects.
Collapse
Affiliation(s)
- Nikolai Petrovsky
- Department of Endocrinology and Diabetes, Flinders University, Adelaide, SA, 5042, Australia.
- Vaxine Pty Ltd, Adelaide, SA, Australia.
| |
Collapse
|
37
|
Hu J, Qiu L, Wang X, Zou X, Lu M, Yin J. Carbohydrate-based vaccine adjuvants - discovery and development. Expert Opin Drug Discov 2015; 10:1133-44. [PMID: 26372693 DOI: 10.1517/17460441.2015.1067198] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The addition of a suitable adjuvant to a vaccine can generate significant effective adaptive immune responses. There is an urgent need for the development of novel po7tent and safe adjuvants for human vaccines. Carbohydrate molecules are promising adjuvants for human vaccines due to their high biocompatibility and good tolerability in vivo. AREAS COVERED The present review covers a few promising carbohydrate-based adjuvants, lipopolysaccharide, trehalose-6,6'-dibehenate, QS-21 and inulin as examples, which have been extensively studied in human vaccines in a number of preclinical and clinical studies. The authors discuss the current status, applications and strategies of development of each adjuvant and different adjuvant formulation systems. This information gives insight regarding the exciting prospect in the field of carbohydrate-based adjuvant research. EXPERT OPINION Carbohydrate-based adjuvants are promising candidates as an alternative to the Alum salts for human vaccines development. Furthermore, combining two or more adjuvants in one formulation is one of the effective strategies in adjuvant development. However, further research efforts are needed to study and develop novel adjuvants systems, which can be more stable, potent and safe. The development of synthetic carbohydrate chemistry can improve the study of carbohydrate-based adjuvants.
Collapse
Affiliation(s)
- Jing Hu
- a 1 Jiangnan University, Wuxi Medical School , Lihu Avenue 1800, 214122, Wuxi, China
| | - Liying Qiu
- a 1 Jiangnan University, Wuxi Medical School , Lihu Avenue 1800, 214122, Wuxi, China
| | - Xiaoli Wang
- b 2 Jiangnan University, The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology , Lihu Avenue 1800, 214122, Wuxi, China +86 51 085 328 229 ; +86 51 085 328 229 ;
| | - Xiaopeng Zou
- b 2 Jiangnan University, The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology , Lihu Avenue 1800, 214122, Wuxi, China +86 51 085 328 229 ; +86 51 085 328 229 ;
| | - Mengji Lu
- c 3 University Hospital Essen, Institute of Virology , Hufelandstr, 55, 45122 Essen, Germany +49 2 017 233 530 ; +49 2 017 235 929 ;
| | - Jian Yin
- b 2 Jiangnan University, The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology , Lihu Avenue 1800, 214122, Wuxi, China +86 51 085 328 229 ; +86 51 085 328 229 ;
| |
Collapse
|
38
|
Honda-Okubo Y, Ong CH, Petrovsky N. Advax delta inulin adjuvant overcomes immune immaturity in neonatal mice thereby allowing single-dose influenza vaccine protection. Vaccine 2015; 33:4892-900. [PMID: 26232344 PMCID: PMC4562881 DOI: 10.1016/j.vaccine.2015.07.051] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/10/2015] [Accepted: 07/17/2015] [Indexed: 01/16/2023]
Abstract
A single dose of Advax-adjuvanted influenza vaccine in 7-day-old pups protected against lethal influenza infection. Advax adjuvant enhanced both B-cell and T-cell memory in neonates. Influenza protection in Advax-immunized neonates was dependent on memory B-cells. Advax adjuvant confirmed to be safe and well tolerated in neonates.
Neonates are at high risk for influenza morbidity and mortality due to immune immaturity and lack of priming by prior influenza virus exposure. Inactivated influenza vaccines are ineffective in infants under six months and to provide protection in older children generally require two doses given a month apart. This leaves few options for rapid protection of infants, e.g. during an influenza pandemic. We investigated whether Advax™, a novel polysaccharide adjuvant based on delta inulin microparticles could help overcome neonatal immune hypo-responsiveness. We first tested whether it was possible to use Advax to obtain single-dose vaccine protection of neonatal pups against lethal influenza infection. Inactivated influenza A/H1N1 vaccine (iH1N1) combined with Advax™ adjuvant administered as a single subcutaneous immunization to 7-day-old mouse pups significantly enhanced serum influenza-specific IgM, IgG1, IgG2a and IgG2b levels and was associated with a 3–4 fold increase in the frequency of splenic influenza-specific IgM and IgG antibody secreting cells. Pups immunized with Advax had significantly higher splenocyte influenza-stimulated IFN-γ, IL-2, IL-4, and IL-10 production by CBA and a 3–10 fold higher frequency of IFN-γ, IL-2, IL-4 or IL-17 secreting T cells by ELISPOT. Immunization with iH1N1 + Advax induced robust protection of pups against virus challenge 3 weeks later, whereas pups immunized with iH1N1 antigen alone had no protection. Protection by Advax-adjuvanted iH1N1 was dependent on memory B cells rather than memory T cells, with no protection in neonatal μMT mice that are B-cell deficient. Hence, Advax adjuvant overcame neonatal immune hypo-responsiveness and enabled single-dose protection of pups against otherwise lethal influenza infection, thereby supporting ongoing development of Advax™ as a neonatal vaccine adjuvant.
Collapse
Affiliation(s)
- Yoshikazu Honda-Okubo
- Vaxine Pty Ltd., Bedford Park, Australia; Flinders Medical Centre, Adelaide 5042, Australia
| | - Chun Hao Ong
- Vaxine Pty Ltd., Bedford Park, Australia; Flinders Medical Centre, Adelaide 5042, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Bedford Park, Australia; Flinders Medical Centre, Adelaide 5042, Australia; Department of Endocrinology, Flinders University, Adelaide 5042, Australia.
| |
Collapse
|
39
|
Li L, Honda-Okubo Y, Li C, Sajkov D, Petrovsky N. Delta Inulin Adjuvant Enhances Plasmablast Generation, Expression of Activation-Induced Cytidine Deaminase and B-Cell Affinity Maturation in Human Subjects Receiving Seasonal Influenza Vaccine. PLoS One 2015; 10:e0132003. [PMID: 26177480 PMCID: PMC4503308 DOI: 10.1371/journal.pone.0132003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 06/08/2015] [Indexed: 12/29/2022] Open
Abstract
There is a major need for new adjuvants to improve the efficacy of seasonal and pandemic influenza vaccines. Advax is a novel polysaccharide adjuvant based on delta inulin that has been shown to enhance the immunogenicity of influenza vaccine in animal models and human clinical trials. To better understand the mechanism for this enhancement, we sought to assess its effect on the plasmablast response in human subjects. This pilot study utilised cryopreserved 7 day post-vaccination (7dpv) peripheral blood mononuclear cell samples obtained from a subset of 25 adult subjects from the FLU006-12 trial who had been immunized intramuscularly with a standard dose of 2012 trivalent inactivated influenza vaccine (TIV) alone (n=9 subjects) or combined with 5mg (n=8) or 10mg (n=8) of Advax adjuvant. Subjects receiving Advax adjuvant had increased 7dpv plasmablasts, which in turn exhibited a 2-3 fold higher rate of non-silent mutations in the B-cell receptor CDR3 region associated with higher expression of activation-induced cytidine deaminase (AID), the major enzyme controlling BCR affinity maturation. Together, these data suggest that Advax adjuvant enhances influenza immunity in immunized subjects via multiple mechanisms including increased plasmablast generation, AID expression and CDR3 mutagenesis resulting in enhanced BCR affinity maturation and increased production of high avidity antibody. How Advax adjuvant achieves these beneficial effects on plasmablasts remains the subject of ongoing investigation.
Collapse
Affiliation(s)
- Lei Li
- Vaxine Pty Ltd, Flinders Medical Centre, Bedford Park, Adelaide, Australia
| | | | - Connie Li
- Vaxine Pty Ltd, Flinders Medical Centre, Bedford Park, Adelaide, Australia
| | - Dimitar Sajkov
- Australian Respiratory and Sleep Medicine Institute, Flinders Medical Centre, Bedford Park, Adelaide, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Flinders Medical Centre, Bedford Park, Adelaide, Australia
- Department of Endocrinology, Flinders Medical Centre/Flinders University, Adelaide, Australia
- * E-mail:
| |
Collapse
|
40
|
Calderon-Gonzalez R, Tobes R, Pareja E, Frande-Cabanes E, Petrovsky N, Alvarez-Dominguez C. Identification and characterisation of T-cell epitopes for incorporation into dendritic cell-delivered Listeria vaccines. J Immunol Methods 2015; 424:111-9. [PMID: 26031451 PMCID: PMC7127673 DOI: 10.1016/j.jim.2015.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 01/05/2023]
Abstract
Dendritic cells loaded with antigenic peptides, because of their safety and robust immune stimulation, would be ideal for induction of immunity to protect against listeriosis. However, there is no currently accepted method to predict which peptides derived from the Listeria proteome might confer protection. While elution of peptides from MHC molecules after Listeria infection yields high-affinity immune-dominant epitopes, these individual epitopes did not reliably confer Listeria protection. Instead we applied bioinformatic predictions of MHC class I and II epitopes to generate antigenic peptides that were then formulated with Advax™, a novel polysaccharide particulate adjuvant able to enhance cross-presentation prior to being screened for their ability to induce protective T-cell responses. A combination of at least four intermediate strength MHC-I binding epitopes and one weak MHC-II binding epitope when expressed in a single peptide sequence and formulated with Advax adjuvant induced a potent T-cell response and high TNF-α and IL-12 production by dendritic cells resulting in robust listeriosis protection in susceptible mice. This T-cell vaccine approach might be useful for the design of vaccines to protect against listeriosis or other intracellular infections.
Collapse
Affiliation(s)
- Ricardo Calderon-Gonzalez
- Grupo de Genómica, Proteómica y Vacunas, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Raquel Tobes
- Information Technologies Research Group, Era7 Bioinformatics, Granada, Spain
| | - Eduardo Pareja
- Information Technologies Research Group, Era7 Bioinformatics, Granada, Spain
| | - Elisabet Frande-Cabanes
- Grupo de Genómica, Proteómica y Vacunas, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Nikolai Petrovsky
- Department of Diabetes and Endocrinology, Flinders University, Adelaide, Australia; Vaxine Pty Ltd, Flinders Medical Center, Adelaide, Australia
| | - Carmen Alvarez-Dominguez
- Grupo de Genómica, Proteómica y Vacunas, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain.
| |
Collapse
|
41
|
Rodriguez-Del Rio E, Marradi M, Calderon-Gonzalez R, Frande-Cabanes E, Penadés S, Petrovsky N, Alvarez-Dominguez C. A gold glyco-nanoparticle carrying a Listeriolysin O peptide and formulated with Advax™ delta inulin adjuvant induces robust T-cell protection against listeria infection. Vaccine 2015; 33:1465-73. [PMID: 25659269 DOI: 10.1016/j.vaccine.2015.01.062] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/12/2015] [Accepted: 01/25/2015] [Indexed: 01/17/2023]
Abstract
In the search for an effective vaccine against the human pathogen, Listeria monocytogenes (Listeria), gold glyconanoparticles (GNP) loaded with a listeriolysin O peptide LLO91-99 (GNP-LLO) were used to immunise mice, initially using a dendritic cell (DC) vaccine approach, but subsequently using a standard parenteral immunisation approach. To enhance vaccine immunogenicity a novel polysaccharide adjuvant based on delta inulin (Advax™) was also co-formulated with the GNP vaccine. Confirming previous results, DC loaded in vitro with GNP-LLO provided better protection against listeriosis than DC loaded in vitro using free LLO peptide. The immunogenicity of GNP-LLO loaded DC vaccines was further increased by addition of Advax™ adjuvant. However, as DC vaccines are expensive and impracticable for prophylactic use, we next asked whether the same GNP-LLO antigen could be used to directly target DC in vivo. Immunisation of mice with GNP-LLO plus Advax™ adjuvant induced LLO-specific T-cell immunity and protection against Listeria challenge. Protection correlated with an increased frequency of splenic CD4(+) and CD8(+) T cells, NK cells and CD8α(+) DC, and Th1 cytokine production (IL-12, IFN-γ, TNF-α, and MCP-1), post-challenge. Enhanced T-cell epitope recruitment post-challenge was seen in the groups that received Advax™ adjuvant. Immunisation with GNP-LLO91-99 plus Advax™ adjuvant provided equally robust Listeria protection as the best DC vaccine strategy but without the complexity and cost, making this a highly promising strategy for development of a prophylactic vaccine against listeriosis.
Collapse
Affiliation(s)
- Estela Rodriguez-Del Rio
- Grupo de Genómica, Proteómica y Vacunas, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Marco Marradi
- CIC biomaGUNE, P° de Miramón 182, San Sebastian, Gipúzcoa, Spain; CIBER-BBN, P° de Miramón 182, San Sebastian, Gipúzcoa, Spain
| | - Ricardo Calderon-Gonzalez
- Grupo de Genómica, Proteómica y Vacunas, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Elisabet Frande-Cabanes
- Grupo de Genómica, Proteómica y Vacunas, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Soledad Penadés
- CIC biomaGUNE, P° de Miramón 182, San Sebastian, Gipúzcoa, Spain; CIBER-BBN, P° de Miramón 182, San Sebastian, Gipúzcoa, Spain
| | - Nikolai Petrovsky
- Department of Diabetes and Endocrinology, Flinders University, Adelaide 5042, SA, Australia; Vaxine Pty Ltd, Flinders Medical Centre, Adelaide 5042, SA, Australia
| | - Carmen Alvarez-Dominguez
- Grupo de Genómica, Proteómica y Vacunas, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain.
| |
Collapse
|
42
|
Ashraf U, Ye J, Ruan X, Wan S, Zhu B, Cao S. Usutu virus: an emerging flavivirus in Europe. Viruses 2015; 7:219-38. [PMID: 25606971 PMCID: PMC4306835 DOI: 10.3390/v7010219] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/13/2015] [Indexed: 12/24/2022] Open
Abstract
Usutu virus (USUV) is an African mosquito-borne flavivirus belonging to the Japanese encephalitis virus serocomplex. USUV is closely related to Murray Valley encephalitis virus, Japanese encephalitis virus, and West Nile virus. USUV was discovered in South Africa in 1959. In Europe, the first true demonstration of circulation of USUV was reported in Austria in 2001 with a significant die-off of Eurasian blackbirds. In the subsequent years, USUV expanded to neighboring countries, including Italy, Germany, Spain, Hungary, Switzerland, Poland, England, Czech Republic, Greece, and Belgium, where it caused unusual mortality in birds. In 2009, the first two human cases of USUV infection in Europe have been reported in Italy, causing meningoencephalitis in immunocompromised patients. This review describes USUV in terms of its life cycle, USUV surveillance from Africa to Europe, human cases, its cellular tropism and pathogenesis, its genetic relationship with other flaviviruses, genetic diversity among USUV strains, its diagnosis, and a discussion of the potential future threat to Asian countries.
Collapse
Affiliation(s)
- Usama Ashraf
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Jing Ye
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Xindi Ruan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Shengfeng Wan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Bibo Zhu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Shengbo Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
43
|
Affiliation(s)
- Pingli Li
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University
| | - Fengshan Wang
- National Glycoengineering Research Center, Shandong University
- Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University
| |
Collapse
|
44
|
Bielefeldt-Ohmann H, Prow NA, Wang W, Tan CSE, Coyle M, Douma A, Hobson-Peters J, Kidd L, Hall RA, Petrovsky N. Safety and immunogenicity of a delta inulin-adjuvanted inactivated Japanese encephalitis virus vaccine in pregnant mares and foals. Vet Res 2014; 45:130. [PMID: 25516480 PMCID: PMC4268807 DOI: 10.1186/s13567-014-0130-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/04/2014] [Indexed: 11/10/2022] Open
Abstract
In 2011, following severe flooding in Eastern Australia, an unprecedented epidemic of equine encephalitis occurred in South-Eastern Australia, caused by Murray Valley encephalitis virus (MVEV) and a new variant strain of Kunjin virus, a subtype of West Nile virus (WNVKUN). This prompted us to assess whether a delta inulin-adjuvanted, inactivated cell culture-derived Japanese encephalitis virus (JEV) vaccine (JE-ADVAX™) could be used in horses, including pregnant mares and foals, to not only induce immunity to JEV, but also elicit cross-protective antibodies against MVEV and WNVKUN. Foals, 74–152 days old, received two injections of JE-ADVAX™. The vaccine was safe and well-tolerated and induced a strong JEV-neutralizing antibody response in all foals. MVEV and WNVKUN antibody cross-reactivity was seen in 33% and 42% of the immunized foals, respectively. JE-ADVAX™ was also safe and well-tolerated in pregnant mares and induced high JEV-neutralizing titers. The neutralizing activity was passively transferred to their foals via colostrum. Foals that acquired passive immunity to JEV via maternal antibodies then were immunized with JE-ADVAX™ at 36–83 days of age, showed evidence of maternal antibody interference with low peak antibody titers post-immunization when compared to immunized foals of JEV-naïve dams. Nevertheless, when given a single JE-ADVAX™ booster immunization as yearlings, these animals developed a rapid and robust JEV-neutralizing antibody response, indicating that they were successfully primed to JEV when immunized as foals, despite the presence of maternal antibodies. Overall, JE-ADVAX™ appears safe and well-tolerated in pregnant mares and young foals and induces protective levels of JEV neutralizing antibodies with partial cross-neutralization of MVEV and WNVKUN.
Collapse
Affiliation(s)
- Helle Bielefeldt-Ohmann
- School of Veterinary Science, University of Queensland, Gatton Campus, Gatton 4343, Qld, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mahalakshmi N, Aparnaa R, Kaliraj P. Evaluation of immune response elicited by inulin as an adjuvant with filarial antigens in mice model. Scand J Immunol 2014; 80:261-70. [PMID: 25041426 DOI: 10.1111/sji.12208] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 06/25/2014] [Indexed: 11/30/2022]
Abstract
Filariasis caused by infectious parasitic nematodes has been identified as the second leading source of permanent and long-term disability in Sub-Saharan Africa, Asia and Latin America. Several vaccine candidates were identified from infective third-stage larvae (L3) which involves in the critical transition from arthropod to human. Hitherto studies of these antigens in combination with alum adjuvant have shown to elicit its characteristic Th2 responses. Inulin is a safe, non-toxic adjuvant that principally stimulates the innate immune response through the alternative complement pathway. In the present study, the immune response elicited by inulin and alum as adjuvants were compared with filarial antigens from different aetiological agents: secreted larval acidic protein 1 (SLAP1) from Onchocerca volvulus and venom allergen homologue (VAH) from Brugia malayi as single or as cocktail vaccines in mice model. The study revealed that inulin can induce better humoral response against these antigens than alum adjuvant. Antibody isotyping disclosed inulin's ability to elevate the levels of IgG2a and IgG3 antibodies which mediates in complement-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity (ADCC), respectively, in mice. Splenocyte analysis showed that T cells prestimulated with inulin have higher stimulation index (P < 0.05) than alum except for BmVAH antigen. In vitro ADCC assay showed that inulin formulation had induced higher cytotoxicity with filarial antigens (as single P < 0.01 and as cocktail P < 0.05, respectively) than alum. The results had confirmed the capability of inulin to deplete the levels of Treg and brought a balance in Th1/Th2 arms against filarial antigens in mice.
Collapse
Affiliation(s)
- N Mahalakshmi
- Centre for Biotechnology, Anna University, Guindy, Chennai, Tamilnadu, India
| | | | | |
Collapse
|
46
|
Gordon D, Kelley P, Heinzel S, Cooper P, Petrovsky N. Immunogenicity and safety of Advax™, a novel polysaccharide adjuvant based on delta inulin, when formulated with hepatitis B surface antigen: a randomized controlled Phase 1 study. Vaccine 2014; 32:6469-77. [PMID: 25267153 PMCID: PMC4253909 DOI: 10.1016/j.vaccine.2014.09.034] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 12/19/2022]
Abstract
There is a need for additional safe and effective human vaccine adjuvants. Advax™ is a novel adjuvant produced from semi-crystalline particles of delta inulin. In animal studies Advax enhanced humoral and cellular immunity to hepatitis B surface antigen (HBsAg) without inducing local or systemic reactogenicity. This first-in-man Phase 1 clinical trial tested the safety and tolerability of three intramuscular doses of HBsAg formulated with Advax in a group of healthy adult subjects. Advax was well tolerated with injection site pain scores not significantly different to subjects receiving HBsAg alone and no adverse events were reported in subjects that received Advax. Seroprotection and HBsAb geometric mean titers (GMT) after three immunizations were higher in the Advax 5mg (seroprotection 5/6, 83.3%, GMT 40.7, 95% CI 11.9-139.1) and 10mg (seroprotection 4/5, 80%, GMT 51.6, 95% CI 10.0-266.2) groups versus HBsAg alone (seroprotection 1/5, 20%, GMT 4.1, 95% CI 1.3-12.8). Similarly the proportion of subjects with positive CD4 T-cell responses to HBsAg was higher in the Advax 5mg (4/6, 67%) and Advax 10mg (4/5, 80%) groups versus HBsAg alone (1/5, 20%). These results confirm the safety, tolerability and immunogenicity of Advax adjuvant observed in preclinical studies. Advax may represent a suitable replacement for alum adjuvants in prophylactic human vaccines subject to confirmation of current results in larger studies. Australia and New Zealand Clinical Trial Registry: ACTRN12607000598482.
Collapse
Affiliation(s)
- David Gordon
- Department of Microbiology and Infectious Diseases, Flinders Medical Centre/Flinders University, Adelaide 5042, Australia
| | - Peter Kelley
- Department of Microbiology and Infectious Diseases, Flinders Medical Centre/Flinders University, Adelaide 5042, Australia
| | | | - Peter Cooper
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia; Cancer Research Laboratory, Australian National University Medical School at The Canberra Hospital, Garran, ACT 2605, Australia; John Curtin School of Medical Research, Australian National University, Acton, ACT 2601, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia; Department of Endocrinology, Flinders Medical Centre/Flinders University, Adelaide 5042, Australia.
| |
Collapse
|
47
|
Honda-Okubo Y, Kolpe A, Li L, Petrovsky N. A single immunization with inactivated H1N1 influenza vaccine formulated with delta inulin adjuvant (Advax™) overcomes pregnancy-associated immune suppression and enhances passive neonatal protection. Vaccine 2014; 32:4651-9. [PMID: 24958701 DOI: 10.1016/j.vaccine.2014.06.057] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 05/21/2014] [Accepted: 06/11/2014] [Indexed: 01/14/2023]
Abstract
Pregnant women and neonates represent high-risk groups for influenza infection, and in general have suppressed responses to standard influenza vaccines due to pregnancy-associated immune suppression and immune system immaturity, respectively. We therefore wished to test whether addition of Advax™, a polysaccharide adjuvant based on delta inulin, to an inactivated influenza vaccine (A/H1N1/PR8) administered during pregnancy would safely enhance vaccine immunogenicity and thereby provide improved protection of pregnant mothers and their newborns. Pregnant mice received a single intramuscular injection of β-propiolactone-inactivated H1N1 antigen alone or with Advax adjuvant. Pregnant dams receiving Advax-adjuvanted vaccine exhibited significantly increased serum and breast milk anti-influenza IgG titers. This translated into higher serum anti-influenza IgG titers in the pups of these dams. Complete protection was seen in pups of dams that received Advax-adjuvanted vaccine whereas no survival was seen in pups of control mothers or mothers immunized with unadjuvanted influenza vaccine. Cross-fostering studies confirmed that enhanced protection of pups of dams that received Advax-adjuvanted vaccine was mediated by enhanced transfer of maternal IgG to the pups via breast-feeding. The delta inulin adjuvant was not associated with any reproductive or developmental adverse effects. This study shows that Advax adjuvant was safe when administered with influenza vaccine during pregnancy and provided protection of pups via enhanced breast milk transfer of anti-influenza antibodies, not seen with administration of unadjuvanted vaccine.
Collapse
Affiliation(s)
| | | | - Lei Li
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, SA, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, SA, Australia; Department of Diabetes and Endocrinology, Flinders Medical Centre/Flinders University, Adelaide 5042, SA, Australia.
| |
Collapse
|
48
|
Yang DK, Nah JJ, Kim HH, Song JY. Inactivated genotype 1 Japanese encephalitis vaccine for swine. Clin Exp Vaccine Res 2014; 3:212-9. [PMID: 25003095 PMCID: PMC4083074 DOI: 10.7774/cevr.2014.3.2.212] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 04/14/2014] [Accepted: 04/20/2014] [Indexed: 11/15/2022] Open
Abstract
PURPOSE Japanese encephalitis is a reproductive disorder caused by Japanese encephalitis virus (JEV) in swine. Recent genotype (G) shift phenomenon (G3 to G1) in the Asia-wide has posed a challenge for proper prevention by the current vaccine strain. Thus, new kinds of JEV G1 vaccines with enhanced immunogenicity have been required for pigs. MATERIALS AND METHODS Recombinant porcine granulocyte monocyte-colony stimulating factor (reporGM-CSF) protein was expressed in Spodoptera frugiperda (Sf-9) cells using baculovirus expression system. Two kinds of trials with inactivated JEV vaccines containing IMS1313 adjuvant (Seppic, France) were prepared with or without reporGM-CSF protein. Safety and immunogenicity of the pigs inoculated with the JEV vaccines via intramuscular route was evaluated for 28 days after inoculation. RESULTS Mice, guinea pigs, and fattening pigs inoculated with the inactivated vaccine showed no signs for 14 and 21 days. Both hemagglutination inhibition and plaque reduction neutralizing antibody titers were significantly higher in pigs immunized with the vaccine containing reporGM-CSF protein after boosting. However, on the side of vaccine efficacy, most mice (87%) immunized with the inactivated JEV vaccine survived after virulent JEV challenge. Whereas the group with the vaccine containing reporGM-CSF protein showed lower protective effects than the vaccine alone for the biological activity of the GM-CSF depending on species specific. CONCLUSION Our data indicate that animals inoculated with the JEV vaccines was safe and pigs inoculated with inactivated JEV vaccine containing reporGM-CSF protein showed higher humoral immune responses than that of inactivated JEV vaccine without reporGM-CSF protein.
Collapse
Affiliation(s)
- Dong-Kun Yang
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs (MAFRA), Anyang, Korea
| | - Jin-Ju Nah
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs (MAFRA), Anyang, Korea
| | - Ha-Hyun Kim
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs (MAFRA), Anyang, Korea
| | - Jae-Young Song
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs (MAFRA), Anyang, Korea
| |
Collapse
|
49
|
Rosales-Mendoza S, Salazar-González JA. Immunological aspects of using plant cells as delivery vehicles for oral vaccines. Expert Rev Vaccines 2014; 13:737-49. [DOI: 10.1586/14760584.2014.913483] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
50
|
Lee HJ, Min KI, Park KH, Choi HJ, Kim MK, Ahn CY, Hong YJ, Kim YB. Comparison of JEV neutralization assay using pseudotyped JEV with the conventional plaque-reduction neutralization test. J Microbiol 2014; 52:435-40. [PMID: 24610332 PMCID: PMC7090846 DOI: 10.1007/s12275-014-3529-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/13/2013] [Accepted: 12/16/2013] [Indexed: 11/25/2022]
Abstract
We previously reported the development of a neutralization assay system for evaluating Japanese Encephalitis Virus (JEV) neutralizing antibody (NAb) using pseudotyped-JEV (JEV-PV). JEV-PV-based neutralization assay offers several advantages compared with the current standard plaque-reduction neutralization test (PRNT), including simplicity, safety, and speed. To evaluate the suitability of the JEV-PV assay as new replacement neutralization assay, we compared its repeatability, reproducibility, specificity, and correlated its results with those obtained using the PRNT. These analyses showed a close correlation between the results obtained with the JEV-PV assay and the PRNT, using the 50% plaque reduction method as a standard for measuring NAb titers to JEV. The validation results met all analytical acceptance criteria. These results suggest that the JEV-PV assay could serve as a safe and simple method for measuring NAb titer against JEV and could be used as an alternative approach for assaying the potency of JEV neutralization.
Collapse
Affiliation(s)
- Hee-Jung Lee
- Department of Bio-industrial technologies, Konkuk University, Seoul, 143-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|