51
|
Jones RM, Chichester JA, Mett V, Jaje J, Tottey S, Manceva S, Casta LJ, Gibbs SK, Musiychuk K, Shamloul M, Norikane J, Mett V, Streatfield SJ, van de Vegte-Bolmer M, Roeffen W, Sauerwein RW, Yusibov V. A plant-produced Pfs25 VLP malaria vaccine candidate induces persistent transmission blocking antibodies against Plasmodium falciparum in immunized mice. PLoS One 2013; 8:e79538. [PMID: 24260245 PMCID: PMC3832600 DOI: 10.1371/journal.pone.0079538] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/20/2013] [Indexed: 11/18/2022] Open
Abstract
Malaria transmission blocking vaccines (TBVs) are considered an effective means to control and eventually eliminate malaria. The Pfs25 protein, expressed predominantly on the surface of the sexual and sporogonic stages of Plasmodium falciparum including gametes, zygotes and ookinetes, is one of the primary targets for TBV. It has been demonstrated that plants are an effective, highly scalable system for the production of recombinant proteins, including virus-like particles (VLPs). We engineered VLPs (Pfs25-CP VLP) comprising Pfs25 fused to the Alfalfa mosaic virus coat protein (CP) and produced these non-enveloped hybrid VLPs in Nicotiana benthamiana plants using a Tobacco mosaic virus-based ‘launch’ vector. Purified Pfs25-CP VLPs were highly consistent in size (19.3±2.4 nm in diameter) with an estimated 20–30% incorporation of Pfs25 onto the VLP surface. Immunization of mice with one or two doses of Pfs25-CP VLPs plus Alhydrogel® induced serum antibodies with complete transmission blocking activity through the 6 month study period. These results support the evaluation of Pfs25-CP VLP as a potential TBV candidate and the feasibility of the ‘launch’ vector technology for the production of VLP-based recombinant vaccines against infectious diseases.
Collapse
Affiliation(s)
- R. Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Jessica A. Chichester
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Vadim Mett
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Jennifer Jaje
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Stephen Tottey
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Slobodanka Manceva
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Louis J. Casta
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Sandra K. Gibbs
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Konstantin Musiychuk
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Valentina Mett
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Stephen J. Streatfield
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | | | - Will Roeffen
- Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
- * E-mail:
| |
Collapse
|
52
|
Musiychuk K, Sivalenka R, Jaje J, Bi H, Flores R, Shaw B, Jones RM, Golovina T, Schnipper J, Khandker L, Sun R, Li C, Kang L, Voskinarian-Berse V, Zhang X, Streatfield S, Hambor J, Abbot S, Yusibov V. Plant-produced human recombinant erythropoietic growth factors support erythroid differentiation in vitro. Stem Cells Dev 2013; 22:2326-40. [PMID: 23517237 PMCID: PMC3730378 DOI: 10.1089/scd.2012.0489] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 03/21/2013] [Indexed: 01/11/2023] Open
Abstract
Clinically available red blood cells (RBCs) for transfusions are at high demand, but in vitro generation of RBCs from hematopoietic stem cells requires significant quantities of growth factors. Here, we describe the production of four human growth factors: erythropoietin (EPO), stem cell factor (SCF), interleukin 3 (IL-3), and insulin-like growth factor-1 (IGF-1), either as non-fused proteins or as fusions with a carrier molecule (lichenase), in plants, using a Tobacco mosaic virus vector-based transient expression system. All growth factors were purified and their identity was confirmed by western blotting and peptide mapping. The potency of these plant-produced cytokines was assessed using TF1 cell (responsive to EPO, IL-3 and SCF) or MCF-7 cell (responsive to IGF-1) proliferation assays. The biological activity estimated here for the cytokines produced in plants was slightly lower or within the range cited in commercial sources and published literature. By comparing EC50 values of plant-produced cytokines with standards, we have demonstrated that all four plant-produced growth factors stimulated the expansion of umbilical cord blood-derived CD34+ cells and their differentiation toward erythropoietic precursors with the same potency as commercially available growth factors. To the best of our knowledge, this is the first report on the generation of all key bioactive cytokines required for the erythroid development in a cost-effective manner using a plant-based expression system.
Collapse
Affiliation(s)
| | | | - Jennifer Jaje
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Hong Bi
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Rosemary Flores
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Brenden Shaw
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - R. Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Tatiana Golovina
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | | | - Ruiqiang Sun
- Celgene Cellular Therapeutics, Warren, New Jersey
| | - Chang Li
- Celgene Cellular Therapeutics, Warren, New Jersey
| | - Lin Kang
- Celgene Cellular Therapeutics, Warren, New Jersey
| | | | | | | | - John Hambor
- Celgene Cellular Therapeutics, Warren, New Jersey
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| |
Collapse
|
53
|
Neuhaus V, Schwarz K, Klee A, Seehase S, Förster C, Pfennig O, Jonigk D, Fieguth HG, Koch W, Warnecke G, Yusibov V, Sewald K, Braun A. Functional testing of an inhalable nanoparticle based influenza vaccine using a human precision cut lung slice technique. PLoS One 2013; 8:e71728. [PMID: 23967238 PMCID: PMC3742667 DOI: 10.1371/journal.pone.0071728] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 07/02/2013] [Indexed: 12/12/2022] Open
Abstract
Annual outbreaks of influenza infections, caused by new influenza virus subtypes and high incidences of zoonosis, make seasonal influenza one of the most unpredictable and serious health threats worldwide. Currently available vaccines, though the main prevention strategy, can neither efficiently be adapted to new circulating virus subtypes nor provide high amounts to meet the global demand fast enough. New influenza vaccines quickly adapted to current virus strains are needed. In the present study we investigated the local toxicity and capacity of a new inhalable influenza vaccine to induce an antigen-specific recall response at the site of virus entry in human precision-cut lung slices (PCLS). This new vaccine combines recombinant H1N1 influenza hemagglutinin (HAC1), produced in tobacco plants, and a silica nanoparticle (NP)-based drug delivery system. We found no local cellular toxicity of the vaccine within applicable concentrations. However higher concentrations of NP (≥103 µg/ml) dose-dependently decreased viability of human PCLS. Furthermore NP, not the protein, provoked a dose-dependent induction of TNF-α and IL-1β, indicating adjuvant properties of silica. In contrast, we found an antigen-specific induction of the T cell proliferation and differentiation cytokine, IL-2, compared to baseline level (152±49 pg/mg vs. 22±5 pg/mg), which could not be seen for the NP alone. Additionally, treatment with 10 µg/ml HAC1 caused a 6-times higher secretion of IFN-γ compared to baseline (602±307 pg/mg vs. 97±51 pg/mg). This antigen-induced IFN-γ secretion was further boosted by the adjuvant effect of silica NP for the formulated vaccine to a 12-fold increase (97±51 pg/mg vs. 1226±535 pg/mg). Thus we were able to show that the plant-produced vaccine induced an adequate innate immune response and re-activated an established antigen-specific T cell response within a non-toxic range in human PCLS at the site of virus entry.
Collapse
Affiliation(s)
- Vanessa Neuhaus
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Katharina Schwarz
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Anna Klee
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Sophie Seehase
- Research Center Borstel, Leibniz Center for Medicine and Biosciences Airway Reserach Center North (ARCN), Borstel, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | | | - Olaf Pfennig
- KRH Klinikum Oststadt-Heidehaus, Hannover, Germany
| | - Danny Jonigk
- Institute for Pathology, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | | | - Wolfgang Koch
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Gregor Warnecke
- Division of Cardiac, Thoracic, Transplantation, and Vascular Surgery, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Institute of Immunology, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
- * E-mail:
| |
Collapse
|
54
|
An Y, Rininger JA, Jarvis DL, Jing X, Ye Z, Aumiller JJ, Eichelberger M, Cipollo JF. Comparative glycomics analysis of influenza Hemagglutinin (H5N1) produced in vaccine relevant cell platforms. J Proteome Res 2013; 12:3707-20. [PMID: 23848607 DOI: 10.1021/pr400329k] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hemagglutinin (HA) is the major antigen in influenza vaccines, and glycosylation is known to influence its antigenicity. Embryonated hen eggs are traditionally used for influenza vaccine production, but vaccines produced in mammalian and insect cells were recently licensed. This raises the concern that vaccines produced with different cell systems might not be equivalent due to differences in their glycosylation patterns. Thus, we developed an analytical method to monitor vaccine glycosylation through a combination of nanoLC/MS(E) and quantitative MALDI-TOF MS permethylation profiling. We then used this method to examine glycosylation of HAs from two different influenza H5N1 strains produced in five different platforms, including hen eggs, three different insect cell lines (High Five, expresSF+ and glycoengineered expresSF+), and a human cell line (HEK293). Our results demonstrated that (1) sequon utilization is not necessarily equivalent in different cell types, (2) there are quantitative and qualitative differences in the overall N-glycosylation patterns and structures produced by different cell types, (3) ∼20% of the N-glycans on the HAs produced by High Five cells are core α1,3-fucosylated structures, which may be allergenic in humans, and (4) our method can be used to monitor differences in glycosylation during the cellular glycoengineering stages of vaccine development.
Collapse
Affiliation(s)
- Yanming An
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
55
|
Abstract
Influenza virus infects a wide variety of species including humans, pigs, horses, sea mammals and birds. Weight loss caused by influenza infection and/or co-infection with other infectious agents results in significant financial loss in swine herds. The emergence of pandemic H1N1 (A/CA/04/2009/H1N1) and H3N2 variant (H3N2v) viruses, which cause disease in both humans and livestock constitutes a concerning public health threat. Influenza virus contains eight single-stranded, negative-sense RNA genome segments. This genetic structure allows the virus to evolve rapidly by antigenic drift and shift. Antigen-specific antibodies induced by current vaccines provide limited cross protection to heterologous challenge. In pigs, this presents a major obstacle for vaccine development. Different strategies are under development to produce vaccines that provide better cross-protection for swine. Moreover, overriding interfering maternal antibodies is another goal for influenza vaccines in order to permit effective immunization of piglets at an early age. Herein, we present a review of influenza virus infection in swine, including a discussion of current vaccine approaches and techniques used for novel vaccine development.
Collapse
|
56
|
Chichester JA, Manceva SD, Rhee A, Coffin MV, Musiychuk K, Mett V, Shamloul M, Norikane J, Streatfield SJ, Yusibov V. A plant-produced protective antigen vaccine confers protection in rabbits against a lethal aerosolized challenge with Bacillus anthracis Ames spores. Hum Vaccin Immunother 2013; 9:544-52. [PMID: 23324615 PMCID: PMC3891710 DOI: 10.4161/hv.23233] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 11/21/2012] [Indexed: 11/19/2022] Open
Abstract
The potential use of Bacillus anthracis as a bioterrorism weapon threatens the security of populations globally, requiring the immediate availability of safe, efficient and easily delivered anthrax vaccine for mass vaccination. Extensive research efforts have been directed toward the development of recombinant subunit vaccines based on protective antigen (PA), the principal virulence factor of B. anthracis. Among the emerging technologies for the production of these vaccine antigens is our launch vector-based plant transient expression system. Using this system, we have successfully engineered, expressed, purified and characterized full-length PA (pp-PA83) in Nicotiana benthamiana plants using agroinfiltration. This plant-produced antigen elicited high toxin neutralizing antibody titers in mice and rabbits after two vaccine administrations with Alhydrogel. In addition, immunization with this vaccine candidate protected 100% of rabbits from a lethal aerosolized B. anthracis challenge. The vaccine effects were dose-dependent and required the presence of Alhydrogel adjuvant. In addition, the vaccine antigen formulated with Alhydrogel was stable and retained immunogenicity after two-week storage at 4°C, the conditions intended for clinical use. These results support the testing of this vaccine candidate in human volunteers and the utility of our plant expression system for the production of a recombinant anthrax vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Aerosols
- Aluminum Hydroxide/administration & dosage
- Animals
- Anthrax/immunology
- Anthrax/prevention & control
- Anthrax Vaccines/administration & dosage
- Anthrax Vaccines/immunology
- Antibodies, Bacterial/blood
- Antibodies, Neutralizing/blood
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/isolation & purification
- Bacterial Toxins/administration & dosage
- Bacterial Toxins/genetics
- Bacterial Toxins/immunology
- Bacterial Toxins/isolation & purification
- Disease Models, Animal
- Inhalation Exposure
- Mice, Inbred BALB C
- Plants, Genetically Modified/genetics
- Rabbits
- Survival Analysis
- Nicotiana/genetics
- Treatment Outcome
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
| | | | - Amy Rhee
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Megan V. Coffin
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Vadim Mett
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| |
Collapse
|
57
|
Shoji Y, Jones RM, Mett V, Chichester JA, Musiychuk K, Sun X, Tumpey TM, Green BJ, Shamloul M, Norikane J, Bi H, Hartman CE, Bottone C, Stewart M, Streatfield SJ, Yusibov V. A plant-produced H1N1 trimeric hemagglutinin protects mice from a lethal influenza virus challenge. Hum Vaccin Immunother 2013; 9:553-60. [PMID: 23296194 PMCID: PMC3891711 DOI: 10.4161/hv.23234] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/03/2012] [Indexed: 11/19/2022] Open
Abstract
The increased worldwide awareness of seasonal and pandemic influenza, including pandemic H1N1 virus, has stimulated interest in the development of economic platforms for rapid, large-scale production of safe and effective subunit vaccines. In recent years, plants have demonstrated their utility as such a platform and have been used to produce vaccine antigens against various infectious diseases. Previously, we have produced in our transient plant expression system a recombinant monomeric hemagglutinin (HA) protein (HAC1) derived from A/California/04/09 (H1N1) strain of influenza virus and demonstrated its immunogenicity and safety in animal models and human volunteers. In the current study, to mimic the authentic HA structure presented on the virus surface and to improve stability and immunogenicity of the HA antigen, we generated trimeric HA by introducing a trimerization motif from a heterologous protein into the HA sequence. Here, we describe the engineering, production in Nicotiana benthamiana plants, and characterization of the highly purified recombinant trimeric HA protein (tHA-BC) from A/California/04/09 (H1N1) strain of influenza virus. The results demonstrate the induction of serum hemagglutination inhibition antibodies by tHA-BC and its protective efficacy in mice against a lethal viral challenge. In addition, the immunogenic and protective doses of tHA-BC were much lower compared with monomeric HAC1. Further investigation into the optimum vaccine dose and/or regimen as well as the stability of trimerized HA is necessary to determine whether trimeric HA is a more potent vaccine antigen than monomeric HA.
Collapse
MESH Headings
- Animals
- Antibodies, Bacterial/blood
- Disease Models, Animal
- Hemagglutination Inhibition Tests
- Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/isolation & purification
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Mice, Inbred BALB C
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Plants, Genetically Modified/genetics
- Protein Engineering
- Protein Multimerization
- Survival Analysis
- Nicotiana/genetics
- Treatment Outcome
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Yoko Shoji
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - R. Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Vadim Mett
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | | | - Xiangjie Sun
- Influenza Division; National Center for Immunization and Respiratory Diseases; Centers for Disease Control and Prevention; Atlanta, GA USA
| | - Terrence M. Tumpey
- Influenza Division; National Center for Immunization and Respiratory Diseases; Centers for Disease Control and Prevention; Atlanta, GA USA
| | - Brian J. Green
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Hong Bi
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Cory Bottone
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Michelle Stewart
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| |
Collapse
|
58
|
Protective efficacy of baculovirus-derived influenza virus-like particles bearing H5 HA alone or in combination with M1 in chickens. Vet Microbiol 2012; 162:623-630. [PMID: 23265240 DOI: 10.1016/j.vetmic.2012.11.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 11/15/2012] [Accepted: 11/22/2012] [Indexed: 11/23/2022]
Abstract
Since 2003, the highly pathogenic avian influenza (HPAI) H5N1 has become a serious problem in animals and an increasing threat to public health. To develop effective vaccines for H5 HPAI in chickens, virus-like particles (VLP) were produced using a baculovirus expression system. The particles comprised hemagglutinin (HA) alone (HA-VLP) or HA in combination with a matrix protein (M1; HAM-VLP) derived from a recent clade 2.3.2.1 H5N1 HPAI virus. To compare the immunogenicity and protective efficacy of these VLPs, 10 μg HAM-VLP, the equivalent amounts of HA incorporated HA-VLP or whole inactivated virus (WIV), were emulsified with mineral oil and used to immunize chickens. The serum hemagglutination inhibition antibody levels induced by HA-VLP and HAM-VLP were comparable to WIV. Antibodies to nucleoprotein were detected only in the WIV group. Immunized chickens in each group survived and were protected against a lethal homologous virus challenge, showing no clinical signs of infection. The challenge virus was detected intermittently in some oropharyngeal swabs, but not in cloacal swabs or various organs, which means that VLPs and WIV provide protection against systemic but not local virus replication in chickens. After the challenge, the HA-VLP group showed significantly increased serum antibody levels compared to the HAM-VLP and WIV groups, and some chickens in the HA-VLP group seroconverted with respect to nucleoprotein. Taken together, these results suggest that VLPs may be an effective method for controlling HPAI in chickens. They could be applied to a differentiating infected from vaccinated animals (DIVA) strategy. In addition, it is likely that HAM-VLP is more efficacious than HA-VLP in chickens.
Collapse
|
59
|
Chichester JA, Jones RM, Green BJ, Stow M, Miao F, Moonsammy G, Streatfield SJ, Yusibov V. Safety and immunogenicity of a plant-produced recombinant hemagglutinin-based influenza vaccine (HAI-05) derived from A/Indonesia/05/2005 (H5N1) influenza virus: a phase 1 randomized, double-blind, placebo-controlled, dose-escalation study in healthy adults. Viruses 2012; 4:3227-44. [PMID: 23202523 PMCID: PMC3509691 DOI: 10.3390/v4113227] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 11/15/2012] [Accepted: 11/16/2012] [Indexed: 01/23/2023] Open
Abstract
Recently, we have reported [1,2] on a subunit influenza vaccine candidate based on the recombinant hemagglutinin protein from the A/Indonesia/05/2005 (H5N1) strain of influenza virus, produced it using 'launch vector'-based transient expression technology in Nicotiana benthamiana, and demonstrated its immunogenicity in pre-clinical studies. Here, we present the results of a first-in-human, Phase 1 randomized, double-blind, placebo-controlled study designed to investigate safety, reactogenicity and immunogenicity of three escalating dose levels of this vaccine, HAI-05, (15, 45 and 90 µg) adjuvanted with Alhydrogel® (0.75 mg aluminum per dose) and the 90 µg dose level without Alhydrogel®. Vaccine was administered intramuscularly in two injections three weeks apart to healthy adults of 18-49 years of age. At all dose levels the vaccine was generally safe and well tolerated, with no reported serious adverse events or dose-limiting toxicities. Mild local and systemic reactions were observed in all vaccine dose groups and the placebo group and their occurrence was not dose related. The incidence rates were higher in the groups receiving vaccine with Alhydrogel®. The immune response elicited by the HAI-05 vaccine was variable with respect to both hemagglutination-inhibition and virus microneutralization antibody titers, with the highest responses observed in the 90 µg unadjuvanted group.
Collapse
|
60
|
Oligomeric recombinant H5 HA1 vaccine produced in bacteria protects ferrets from homologous and heterologous wild-type H5N1 influenza challenge and controls viral loads better than subunit H5N1 vaccine by eliciting high-affinity antibodies. J Virol 2012; 86:12283-93. [PMID: 22951833 DOI: 10.1128/jvi.01596-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recombinant hemagglutinin from influenza viruses with pandemic potential can be produced rapidly in various cell substrates. In this study, we compared the functionality and immunogenicity of bacterially produced oligomeric or monomeric HA1 proteins from H5N1 (A/Vietnam/1203/04) with those of the egg-based licensed subunit H5N1 (SU-H5N1) vaccine in ferrets challenged with homologous or heterologous H5N1 highly pathogenic influenza strains. Ferrets were vaccinated twice with the oligomeric or monomeric rHA1 or with SU-H5N1 (Sanofi Pasteur) emulsified with Titermax adjuvant and were challenged with wild-type homologous (A/Vietnam/1203/04; clade 1) or heterologous (A/Whooperswan/Mongolia/244/2005; clade 2.2) virus. Only the oligomeric rHA1 (not the monomeric rHA1) immunogen and the SU-H5N1 vaccine provided protection against the lethality and morbidity of homologous and heterologous highly pathogenic H5N1. Oligomeric rHA1 generated more cross-neutralizing antibodies and higher levels of serum antibody binding to HA1, with stronger avidity and a better IgG/IgM ratio, than monomeric HA1 and SU-H5N1 vaccines, as determined by surface plasmon resonance (SPR). Importantly, viral loads after heterologous H5N1 challenge were more efficiently controlled in ferrets vaccinated with the oligomeric rHA1 immunogen than in SU-H5N1-vaccinated ferrets. The reduction of viral loads in the nasal washes correlated strongly with higher-avidity antibodies to oligomeric rHA1 derived from H5N1 clade 1 and clade 2.2 viruses, as measured by SPR. This is the first study to show the role of antibody avidity for the HA1 globular head domain in reduction of viral loads in the upper respiratory tract, which could significantly reduce viral transmission.
Collapse
|
61
|
Green factory: Plants as bioproduction platforms for recombinant proteins. Biotechnol Adv 2012; 30:1171-84. [DOI: 10.1016/j.biotechadv.2011.08.020] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 08/24/2011] [Accepted: 08/30/2011] [Indexed: 12/15/2022]
|
62
|
Santiago FW, Lambert Emo K, Fitzgerald T, Treanor JJ, Topham DJ. Antigenic and immunogenic properties of recombinant hemagglutinin proteins from H1N1 A/Brisbane/59/07 and B/Florida/04/06 when produced in various protein expression systems. Vaccine 2012; 30:4606-16. [PMID: 22609035 DOI: 10.1016/j.vaccine.2012.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 03/27/2012] [Accepted: 05/01/2012] [Indexed: 10/28/2022]
Abstract
Antibodies directed against the influenza hemagglutinin (HA) protein largely mediate virus neutralization and confer protection against infection. Consequently, many studies and assays of influenza vaccines are focused on HA-specific immune responses. Recombinant HA (rHA) proteins can be produced in a number of protein expression and cell culture systems. These range from baculovirus infection of insect cell cultures, to transient transfection of plants, to stably transfected human cell lines. Furthermore, the rHA proteins may contain genetic modifications, such as histidine tags or trimerization domains, intended to ease purification or enhance protein stability. However, no systematic study of these different forms of the HA protein have been conducted. It is not clear which, if any, of these different protein expression systems or structural modifications improve or diminish the biological behavior of the proteins as immunogens or antigens in immune assays. Therefore we set out to perform systematic evaluation of rHA produced in different proteins expression systems and with varied modifications. Five rHA proteins based on recent strains of seasonal influenza A and five based on influenza B HA were kindly provided by the Biodefense and Emerging Infections Reagent Repository (BEIR). These proteins were evaluated in a combination of biochemical and structural assays, in vitro humoral and cellular immune assays, and in an animal vaccination model. Marked differences in the behavior of the individual proteins was evident suggesting that they are not equal when being used to detect an immune response. They were, nevertheless, similar at eliciting neutralizing antibody responses.
Collapse
Affiliation(s)
- Felix W Santiago
- New York Influenza Center of Excellence, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 609, Rochester, NY 14642, USA. felix
| | | | | | | | | |
Collapse
|
63
|
Jul-Larsen Å, Madhun AS, Brokstad KA, Montomoli E, Yusibov V, Cox RJ. The human potential of a recombinant pandemic influenza vaccine produced in tobacco plants. Hum Vaccin Immunother 2012; 8:653-61. [PMID: 22634440 PMCID: PMC3495720 DOI: 10.4161/hv.19503] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Rapid production of influenza vaccine antigen is an important challenge when a new pandemic occurs. Production of recombinant antigens in plants is a quick, cost effective and up scalable new strategy for influenza vaccine production. In this study, we have characterized a recombinant influenza haemagglutinin antigen (HAC1) that was derived from the 2009 pandemic H1N1 (pdmH1N1) virus and expressed in tobacco plants. Volunteers vaccinated with the 2009 pdmH1N1 oil-in-water adjuvanted vaccine provided serum and lymphocyte samples that were used to study the immunogenic properties of the HAC1 antigen in vitro. By 7 d post vaccination, the vaccine fulfilled the licensing criteria for antibody responses to the HA detected by haemagglutination inhibition and single radial hemolysis. By ELISA and ELISPOT analysis we showed that HAC1 was recognized by specific serum antibodies and antibody secreting cells, respectively. We conducted a kinetic analysis and found a peak of serum HAC1 specific antibody response between day 14 and 21 post vaccination by ELISA. We also detected elevated production of IL-2 and IFNγ and low frequencies of CD4(+) T cells producing single or multiple Th1 cytokines after stimulating PBMCs (peripheral blood mononuclear cells) with the HAC1 antigen in vitro. This indicates that the antigen can interact with T cells, although confirming an effective adjuvant would be required to improve the T-cell stimulation of plant based vaccines. We conclude that the tobacco derived recombinant HAC1 antigen is a promising vaccine candidate recognized by both B- and T cells.
Collapse
MESH Headings
- Adult
- Antibodies, Viral/blood
- B-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/immunology
- Cytokines/metabolism
- Enzyme-Linked Immunosorbent Assay
- Enzyme-Linked Immunospot Assay
- Female
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Human Experimentation
- Humans
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/isolation & purification
- Influenza, Human/prevention & control
- Male
- Middle Aged
- Plants, Genetically Modified
- Th1 Cells/immunology
- Time Factors
- Nicotiana
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
Collapse
Affiliation(s)
- Åsne Jul-Larsen
- Influenza Centre, The Gade Institute, University of Bergen, Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
64
|
Shoji Y, Farrance CE, Bautista J, Bi H, Musiychuk K, Horsey A, Park H, Jaje J, Green BJ, Shamloul M, Sharma S, Chichester JA, Mett V, Yusibov V. A plant-based system for rapid production of influenza vaccine antigens. Influenza Other Respir Viruses 2012; 6:204-10. [PMID: 21974811 PMCID: PMC4941669 DOI: 10.1111/j.1750-2659.2011.00295.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Influenza virus is a globally important respiratory pathogen that causes a high degree of annual morbidity and mortality. Significant antigenic drift results in emergence of new, potentially pandemic, virus variants. The best prophylactic option for controlling emerging virus strains is to manufacture and administer pandemic vaccines in sufficient quantities and to do so in a timely manner without impacting the regular seasonal influenza vaccine capacity. Current, egg-based, influenza vaccine production is well established and provides an effective product, but has limited capacity and speed. OBJECTIVES To satisfy the additional global demand for emerging influenza vaccines, high-performance cost-effective technologies need to be developed. Plants have a potential as an economic and efficient large-scale production platform for vaccine antigens. METHODS In this study, a plant virus-based transient expression system was used to produce hemagglutinin (HA) proteins from the three vaccine strains used during the 2008-2009 influenza season, A/Brisbane/59/07 (H1N1), A/Brisbane/10/07 (H3N2), and B/Florida/4/06, as well as from the recently emerged novel H1N1 influenza A virus, A/California/04/09. RESULTS The recombinant plant-based HA proteins were engineered and produced in Nicotiana benthamiana plants within 2 months of obtaining the genetic sequences specific to each virus strain. These antigens expressed at the rate of 400-1300 mg/kg of fresh leaf tissue, with >70% solubility. Immunization of mice with these HA antigens induced serum anti-HA IgG and hemagglutination inhibition antibody responses at the levels considered protective against these virus infections. CONCLUSIONS These results demonstrate the feasibility of our transient plant expression system for the rapid production of influenza vaccine antigens.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Gene Expression
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Orthomyxoviridae/genetics
- Orthomyxoviridae/immunology
- Nicotiana/genetics
- Nicotiana/metabolism
Collapse
Affiliation(s)
- Yoko Shoji
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | | | - James Bautista
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Hong Bi
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | | | - April Horsey
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - HeeWoo Park
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Jennifer Jaje
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Brian J. Green
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Satish Sharma
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | | | - Vadim Mett
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| |
Collapse
|
65
|
Zhang S, Sherwood RW, Yang Y, Fish T, Chen W, McCardle JA, Jones RM, Yusibov V, May ER, Rose JKC, Thannhauser TW. Comparative characterization of the glycosylation profiles of an influenza hemagglutinin produced in plant and insect hosts. Proteomics 2012; 12:1269-88. [PMID: 22577028 PMCID: PMC4545245 DOI: 10.1002/pmic.201100474] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Accepted: 01/01/2012] [Indexed: 11/11/2022]
Abstract
The main objective of this study was to characterize the N-linked glycosylation profiles of recombinant hemagglutinin (HA) proteins expressed in either insect or plant hosts, and to develop a mass spectrometry based workflow that can be used in quality control to assess batch-to-batch reproducibility for recombinant HA glycosylation. HA is a surface glycoprotein of the influenza virus that plays a key role in viral infectivity and pathogenesis. Characterization of the glycans for plant recombinant HA from the viral strain A/California/04/09 (H1N1) has not yet been reported. In this study, N-linked glycosylation patterns of the recombinant HAs from both insect and plant hosts were characterized by precursor ion scan-driven data-dependent analysis followed by high-resolution MS/MS analysis of the deglycosylated tryptic peptides. Five glycosylation sites (N11, N23, N276, N287, and N481) were identified containing high mannose type glycans in plant-expressed HAs, and complex type glycoforms for the insect-expressed HA. More than 95% site occupancy was observed for all glycosylation sites except N11, which was 60% occupied. Multiple-reaction monitoring based quantitation analysis was developed for each glycopeptide isoform and the quantitative results indicate that the Man(8) GlcNAc(2) is the dominant glycan for all sites in plant-expressed HAs. The relative abundance of the glycoforms at each specific glycosylation site and the relative quantitation for each glycoform among three HAs were determined. Few differences in the glycosylation profiles were detected between the two batches of plant HAs studied, but there were significant differences between the glycosylation patterns in the HAs generated in plant and insect expression hosts.
Collapse
Affiliation(s)
- Sheng Zhang
- Institute for Biotechnology and Life Science Biotechnologies, Cornell University, Ithaca, NY 14853, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Iyer V, Liyanage MR, Shoji Y, Chichester JA, Jones RM, Yusibov V, Joshi SB, Middaugh CR. Formulation development of a plant-derived H1N1 influenza vaccine containing purified recombinant hemagglutinin antigen. Hum Vaccin Immunother 2012; 8:453-64. [PMID: 22370514 DOI: 10.4161/hv.19106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Influenza is a prevalent, highly contagious and sometimes fatal respiratory disease. Vaccination provides an effective approach to control the disease, but because of frequent changes in the structure of the major surface proteins, there is great need for a technology that permits rapid preparation of new forms of the vaccine each year in sufficient quantities. Recently, using a safe, simple, time- and cost-effective plant viral vector-based transient expression system, the hemagglutinin antigen of H1N1 influenza A strain (HAC1), an H1N1 influenza vaccine candidate, has been produced in Nicotiana benthamiana plants. As a step toward the generation of a commercially viable subunit influenza vaccine, we developed HAC1 formulations in the presence and absence of an aluminum salt adjuvant (Alhydrogel(®)), analyzed their properties, and assessed immunogenicity in an animal model. Biophysical properties of HAC1 were evaluated using several spectroscopic and light scattering techniques as a function of pH and temperature combined with data analysis using an empirical phase diagram approach. Excipients that were potent stabilizers of the recombinant protein were identified using intrinsic fluorescence spectroscopy. The adsorptive capacity and thermal stability of the protein on the surface of Alhydrogel(®) were then examined in the presence and absence of selected stabilizers using UV absorbance after centrifugation and intrinsic fluorescence spectroscopy, respectively. Immunogenicity studies conducted in mice demonstrated that the highest level of serum immune responses (hemagglutination-inhibiting antibody titers), with a 100% seropositive rates, were induced by HAC1 in the presence of Alhydrogel(®), and this response was elicited regardless of the solution conditions of the formulation.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Aluminum Hydroxide/administration & dosage
- Animals
- Antibodies, Viral/blood
- Chemistry, Pharmaceutical
- Excipients/chemistry
- Genetic Vectors
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hydrogen-Ion Concentration
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/chemistry
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Mice
- Plant Viruses/genetics
- Protein Stability
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Spectrum Analysis
- Temperature
- Nicotiana/genetics
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/chemistry
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/chemistry
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Vidyashankara Iyer
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, KS, USA
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Kanagarajan S, Tolf C, Lundgren A, Waldenström J, Brodelius PE. Transient expression of hemagglutinin antigen from low pathogenic avian influenza A (H7N7) in Nicotiana benthamiana. PLoS One 2012; 7:e33010. [PMID: 22442675 PMCID: PMC3307706 DOI: 10.1371/journal.pone.0033010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 02/02/2012] [Indexed: 01/18/2023] Open
Abstract
The influenza A virus is of global concern for the poultry industry, especially the H5 and H7 subtypes as they have the potential to become highly pathogenic for poultry. In this study, the hemagglutinin (HA) of a low pathogenic avian influenza virus of the H7N7 subtype isolated from a Swedish mallard Anas platyrhynchos was sequenced, characterized and transiently expressed in Nicotiana benthamiana. Recently, plant expression systems have gained interest as an alternative for the production of vaccine antigens. To examine the possibility of expressing the HA protein in N. benthamiana, a cDNA fragment encoding the HA gene was synthesized de novo, modified with a Kozak sequence, a PR1a signal peptide, a C-terminal hexahistidine (6×His) tag, and an endoplasmic retention signal (SEKDEL). The construct was cloned into a Cowpea mosaic virus (CPMV)-based vector (pEAQ-HT) and the resulting pEAQ-HT-HA plasmid, along with a vector (pJL3:p19) containing the viral gene-silencing suppressor p19 from Tomato bushy stunt virus, was agro-infiltrated into N. benthamiana. The highest gene expression of recombinant plant-produced, uncleaved HA (rHA0), as measured by quantitative real-time PCR was detected at 6 days post infiltration (dpi). Guided by the gene expression profile, rHA0 protein was extracted at 6 dpi and subsequently purified utilizing the 6×His tag and immobilized metal ion adsorption chromatography. The yield was 0.2 g purified protein per kg fresh weight of leaves. Further molecular characterizations showed that the purified rHA0 protein was N-glycosylated and its identity confirmed by liquid chromatography-tandem mass spectrometry. In addition, the purified rHA0 exhibited hemagglutination and hemagglutination inhibition activity indicating that the rHA0 shares structural and functional properties with native HA protein of H7 influenza virus. Our results indicate that rHA0 maintained its native antigenicity and specificity, providing a good source of vaccine antigen to induce immune response in poultry species.
Collapse
MESH Headings
- Animals
- Antigens, Viral/biosynthesis
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Influenza A Virus, H7N7 Subtype
- Influenza in Birds/genetics
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/immunology
- Plants, Genetically Modified/metabolism
- Poultry/immunology
- Poultry Diseases/genetics
- Poultry Diseases/immunology
- Poultry Diseases/prevention & control
- Recombinant Proteins/biosynthesis
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Nicotiana
Collapse
Affiliation(s)
- Selvaraju Kanagarajan
- Section for Biomaterials and Medicinal Chemistry, School of Natural Sciences, Linnaeus University, Kalmar, Sweden
| | - Conny Tolf
- Section for Zoonotic Ecology and Epidemiology, School of Natural Sciences, Linnaeus University, Kalmar, Sweden
| | - Anneli Lundgren
- Section for Biomaterials and Medicinal Chemistry, School of Natural Sciences, Linnaeus University, Kalmar, Sweden
| | - Jonas Waldenström
- Section for Zoonotic Ecology and Epidemiology, School of Natural Sciences, Linnaeus University, Kalmar, Sweden
| | - Peter E. Brodelius
- Section for Biomaterials and Medicinal Chemistry, School of Natural Sciences, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
68
|
Plant-made therapeutics: An emerging platform in South Africa. Biotechnol Adv 2012; 30:449-59. [DOI: 10.1016/j.biotechadv.2011.07.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 05/26/2011] [Accepted: 07/25/2011] [Indexed: 12/20/2022]
|
69
|
Fischer R, Schillberg S, Hellwig S, Twyman RM, Drossard J. GMP issues for recombinant plant-derived pharmaceutical proteins. Biotechnol Adv 2011; 30:434-9. [PMID: 21856403 DOI: 10.1016/j.biotechadv.2011.08.007] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 08/05/2011] [Accepted: 08/05/2011] [Indexed: 01/11/2023]
Abstract
Recombinant proteins can be produced in a diverse array of plant-based systems, ranging from whole plants growing in the soil to plant suspension cells growing in a fully-defined synthetic medium in a bioreactor. When the recombinant proteins are intended for medical use (plant-derived pharmaceutical proteins, PDPs) they fall under the same regulatory guidelines for manufacturing that cover drugs from all other sources, and when such proteins enter clinical development this includes the requirement for production according to good manufacturing practice (GMP). In principle, the well-characterized GMP regulations that apply to pharmaceutical proteins produced in bacteria and mammalian cells are directly transferrable to plants. In practice, the cell-specific terminology and the requirement for a contained, sterile environment mean that only plant cells in a bioreactor fully meet the original GMP criteria. Significant changes are required to adapt these regulations for proteins produced in whole-plant systems and it is only recently that the first GMP-compliant production processes using plants have been delivered.
Collapse
Affiliation(s)
- Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Aachen, Germany.
| | | | | | | | | |
Collapse
|
70
|
A plant-produced Pfs230 vaccine candidate blocks transmission of Plasmodium falciparum. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1351-7. [PMID: 21715576 DOI: 10.1128/cvi.05105-11] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Plasmodium falciparum is transmitted to a new host after completing its sexual cycle within a mosquito. Developing vaccines against the parasite sexual stages is a critical component in the fight against malaria. We are targeting multiple proteins of P. falciparum which are found only on the surfaces of the sexual forms of the parasite and where antibodies against these proteins have been shown to block the progression of the parasite's life cycle in the mosquito and thus block transmission to the next human host. We have successfully produced a region of the Pfs230 antigen in our plant-based transient-expression system and evaluated this vaccine candidate in an animal model. This plant-produced protein, 230CMB, is expressed at approximately 800 mg/kg in fresh whole leaf tissue and is 100% soluble. Administration of 230CMB with >90% purity induces strong immune responses in rabbits with high titers of transmission-blocking antibodies, resulting in a greater than 99% reduction in oocyst counts in the presence of complement, as determined by a standard membrane feeding assay. Our data provide a clear perspective on the clinical development of a Pfs230-based transmission-blocking malaria vaccine.
Collapse
|