301
|
Qian J, Xu X, Ding J, Yin R, Sun Y, Xue C, Wang J, Ding C, Yu S, Liu X, Hu S, Wang C, Cong Y, Ding Z. Newcastle disease virus-like particles induce DC maturation through TLR4/NF-κB pathway and facilitate DC migration by CCR7-CCL19/CCL21 axis. Vet Microbiol 2017; 203:158-166. [PMID: 28619138 DOI: 10.1016/j.vetmic.2017.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 03/01/2017] [Accepted: 03/01/2017] [Indexed: 01/07/2023]
Abstract
Newcastle disease virus-like particles (NDV VLPs) are a potential candidate vaccine, as shown by eliciting specific immune response against NDV in mice and chickens. Activation of dendritic cells (DCs) is critical to initiate immune response. However, the mechanism of how NDV VLPs induce DC maturation and migration remains elusive. In this study, we found that NDV VLPs are efficient in DC activation by up-regulating surface MHC II and costimulatory molecules, and proinflammatory cytokines through the TLR4/NF-κB pathway. Furthermore, NDV VLPs elevated CCR7 expression on DCs, resulting in DC migration towards CCL19/CCL21 both in vitro and ex vivo. As a consequence of DC maturation and migration, CD4+ T cells were also activated in vivo, demonstrating increased intracellular IFN-γ and IL-4 levels. Together, these results present new insights for NDV VLPs induced DC maturation and migration, providing a better understanding of VLP-triggered innate immune responses.
Collapse
Affiliation(s)
- Jing Qian
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Xiaohong Xu
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Jiaxin Ding
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Renfu Yin
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Yixue Sun
- Engineering Research Center of Jilin Province for Animals Probiotics, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Cong Xue
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Jianzhong Wang
- Engineering Research Center of Jilin Province for Animals Probiotics, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Shengqing Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Chunfeng Wang
- Engineering Research Center of Jilin Province for Animals Probiotics, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Yanlong Cong
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China.
| | - Zhuang Ding
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China.
| |
Collapse
|
302
|
Wang D, Fan F, Li Z, Liu X, Song S, Wei S, He M, Lin Y, Li Z, Wei M, Yu H, Gu Y, Li S, Xia N. Stop codon mutagenesis for homogenous expression of human papillomavirus L1 protein in Escherichia coli. Protein Expr Purif 2017; 133:110-120. [PMID: 28267627 DOI: 10.1016/j.pep.2017.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/02/2017] [Indexed: 01/01/2023]
Abstract
Human papillomavirus (HPV) is widely accepted to be the major causative pathogen of cervical cancer, warts, and other epithelial tumors. Virus infection and subsequent disease development can be prevented by vaccination with HPV vaccines derived from eukaryotic expression systems. Here, we report the soluble expression of the major capsid protein L1 of HPV31, a dominant carcinogenic HPV genotype, in Escherichia coli. HPV31 L1 protein and its elongated form (L1+) were observed in SDS-PAGE and CE-SDS analysis, generated by the native HPV31 L1 gene with a TAA stop codon. Replacing the TAA with TAG but not TGA could completely terminate protein translation. Mass spectrometry sequencing showed that L1+ comprised L1 with a C-terminal extension of 38 amino acids (aa). RNA folding analysis revealed that the unfaithful L1+ expression may result from translational read-through, as TAG is more stable and accessible than the other stop codons. The 38-aa elongated fragment perturbs self-assembly of HPV31 L1+, as shown in size and morphology analyses. By 3D cryo-electron microscopy structure determination, we show self-assembly of purified HPV31 L1 (TAG) VLPs into T = 7 icosahedral symmetry particles, resembling the native HPV virion. Finally, through additional characterization and antigenicity/immunogenicity assays, we verified that the E.coli-derived HPV31 VLPs are an ideal immunogen for HPV vaccine development. Our findings outline a codon optimization stratagem for protein expression and provide a method for the in-depth investigation of prokaryotic translation regulation.
Collapse
Affiliation(s)
- Daning Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Fei Fan
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhihai Li
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Xinlin Liu
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Shuo Song
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Shuangping Wei
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Maozhou He
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yahua Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zhongyi Li
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Minxi Wei
- National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Hai Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Disease, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
303
|
Ho TT, Nguyen JT, Liu J, Stanczak P, Thompson AA, Yan YG, Chen J, Allerston CK, Dillard CL, Xu H, Shoger NJ, Cameron JS, Massari ME, Aertgeerts K. Method for rapid optimization of recombinant GPCR protein expression and stability using virus-like particles. Protein Expr Purif 2017; 133:41-49. [PMID: 28263854 DOI: 10.1016/j.pep.2017.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 10/20/2022]
Abstract
Recent innovative approaches to stabilize and crystallize GPCRs have resulted in an unprecedented breakthrough in GPCR crystal structures as well as application of the purified receptor protein in biophysical and biochemical ligand binding assays. However, the protein optimization process to enable these technologies is lengthy and requires iterative overexpression, solubilization, purification and functional analysis of tens to hundreds of protein variants. Here, we report a new and versatile method to screen in parallel hundreds of GPCR variants in HEK293 produced virus-like particles (VLPs) for protein yield, stability, functionality and ligand binding. This approach reduces the time and resources during GPCR construct optimization by eliminating lengthy protein solubilization and purification steps and by its adaptability to many binding assay formats (label or label-free detection). We exemplified the robustness of our VLP method by screening 210 GALR3-VLP variants in a radiometric agonist-based binding assay and a subset of 88 variants in a label-free antagonist-based assay. The resulting GALR3 agonist or antagonist stabilizing variants were then further used for recombinant protein expression in transfected insect cells. The final purified protein variants were successfully immobilized on a biosensor chip and used in a surface plasmon resonance binding assay.
Collapse
Affiliation(s)
- Thao T Ho
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Jasmine T Nguyen
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Juping Liu
- Department of Preclinical Development, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Pawel Stanczak
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Aaron A Thompson
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Yingzhuo G Yan
- Department of Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Jasmine Chen
- Department of Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Charles K Allerston
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Charles L Dillard
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Hao Xu
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Nicholas J Shoger
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Jill S Cameron
- Department of Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Mark E Massari
- Department of Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| | - Kathleen Aertgeerts
- Department of Structural Biology, Dart Neuroscience, 12278 Scripps Summit Drive, San Diego, CA 92131, USA.
| |
Collapse
|
304
|
Le Mauff F, Loutelier‐Bourhis C, Bardor M, Berard C, Doucet A, D'Aoust M, Vezina L, Driouich A, Couture MM, Lerouge P. Cell wall biochemical alterations during Agrobacterium-mediated expression of haemagglutinin-based influenza virus-like vaccine particles in tobacco. PLANT BIOTECHNOLOGY JOURNAL 2017; 15:285-296. [PMID: 27483398 PMCID: PMC5316917 DOI: 10.1111/pbi.12607] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/18/2016] [Accepted: 07/24/2016] [Indexed: 05/17/2023]
Abstract
Influenza virus-like particles (VLPs) have been shown to induce a safe and potent immune response through both humoral and cellular responses. They represent promising novel influenza vaccines. Plant-based biotechnology allows for the large-scale production of VLPs of biopharmaceutical interest using different model organisms, including Nicotiana benthamiana plants. Through this platform, influenza VLPs bud from the plasma membrane and accumulate between the membrane and the plant cell wall. To design and optimize efficient production processes, a better understanding of the plant cell wall composition of infiltrated tobacco leaves is a major interest for the plant biotechnology industry. In this study, we have investigated the alteration of the biochemical composition of the cell walls of N. benthamiana leaves subjected to abiotic and biotic stresses induced by the Agrobacterium-mediated transient transformation and the resulting high expression levels of influenza VLPs. Results show that abiotic stress due to vacuum infiltration without Agrobacterium did not induce any detectable modification of the leaf cell wall when compared to non infiltrated leaves. In contrast, various chemical changes of the leaf cell wall were observed post-Agrobacterium infiltration. Indeed, Agrobacterium infection induced deposition of callose and lignin, modified the pectin methylesterification and increased both arabinosylation of RG-I side chains and the expression of arabinogalactan proteins. Moreover, these modifications were slightly greater in plants expressing haemagglutinin-based VLP than in plants infiltrated with the Agrobacterium strain containing only the p19 suppressor of silencing.
Collapse
Affiliation(s)
- François Le Mauff
- Laboratoire Glyco‐MEV EA 4358UNIROUENNormandie UnivRouenFrance
- Medicago Inc.QuébecQCCanada
- Present address: Departments of Medicine, Microbiology and ImmunologyMcGill universityMontrealQCCanada
- Present address: Infectious Diseases in Global Health ProgramResearch Institute of the McGill University Health CentreMcGill UniversityMontrealQCCanada
| | | | - Muriel Bardor
- Laboratoire Glyco‐MEV EA 4358UNIROUENNormandie UnivRouenFrance
| | | | | | | | - Louis‐Philippe Vezina
- Medicago Inc.QuébecQCCanada
- Present address: Groupe TH Inc. 1327, avenue Maguire, suite 100QuébecQCG1T 1Z2Canada
| | | | | | - Patrice Lerouge
- Laboratoire Glyco‐MEV EA 4358UNIROUENNormandie UnivRouenFrance
| |
Collapse
|
305
|
Differential Antibody Responses to Conserved HIV-1 Neutralizing Epitopes in the Context of Multivalent Scaffolds and Native-Like gp140 Trimers. mBio 2017; 8:mBio.00036-17. [PMID: 28246356 PMCID: PMC5347340 DOI: 10.1128/mbio.00036-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Broadly neutralizing antibodies (bNAbs) have provided valuable insights into the humoral immune response to HIV-1. While rationally designed epitope scaffolds and well-folded gp140 trimers have been proposed as vaccine antigens, a comparative understanding of their antibody responses has not yet been established. In this study, we probed antibody responses to the N332 supersite and the membrane-proximal external region (MPER) in the context of heterologous protein scaffolds and native-like gp140 trimers. Ferritin nanoparticles and fragment crystallizable (Fc) regions were utilized as multivalent carriers to display scaffold antigens with grafted N332 and MPER epitopes, respectively. Trimeric scaffolds were also identified to stabilize the MPER-containing BG505 gp140.681 trimer in a native-like conformation. Following structural and antigenic evaluation, a subset of scaffold and trimer antigens was selected for immunization in BALB/c mice. Serum binding revealed distinct patterns of antibody responses to these two bNAb targets presented in different structural contexts. For example, the N332 nanoparticles elicited glycan epitope-specific antibody responses that could also recognize the native trimer, while a scaffolded BG505 gp140.681 trimer generated a stronger and more rapid antibody response to the trimer apex than its parent gp140.664 trimer. Furthermore, next-generation sequencing (NGS) of mouse splenic B cells revealed expansion of antibody lineages with long heavy-chain complementarity-determining region 3 (HCDR3) loops upon activation by MPER scaffolds, in contrast to the steady repertoires primed by N332 nanoparticles and a soluble gp140.664 trimer. These findings will facilitate the future development of a coherent vaccination strategy that combines both epitope-focused and trimer-based approaches.IMPORTANCE Both epitope-focused and trimer-based strategies are currently being explored in HIV-1 vaccine development, which aims to elicit broadly neutralizing antibodies (bNAbs) targeting conserved epitopes on the viral envelope (Env). However, little is known about the differences in antibody response to these bNAb targets presented by foreign scaffolds and native Env. In this study, a systematic effort was undertaken to design multivalent epitope scaffolds and soluble gp140.681 trimers with a complete antigenic surface, and to comparatively analyze the antibody responses elicited by these antigens to the N332 supersite and MPER in a mouse model. This study will inform both epitope-focused and trimer-based vaccine design and will facilitate integration of the two vaccine strategies.
Collapse
|
306
|
Jeong H, Seong BL. Exploiting virus-like particles as innovative vaccines against emerging viral infections. J Microbiol 2017; 55:220-230. [PMID: 28243941 PMCID: PMC7090582 DOI: 10.1007/s12275-017-7058-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 02/18/2017] [Accepted: 02/20/2017] [Indexed: 01/20/2023]
Abstract
Emerging viruses pose a major threat to humans and livestock with global public health and economic burdens. Vaccination remains an effective tool to reduce this threat, and yet, the conventional cell culture often fails to produce sufficient vaccine dose. As an alternative to cell-culture based vaccine, virus-like particles (VLPs) are considered as a highpriority vaccine strategy against emerging viruses. VLPs represent highly ordered repetitive structures via macromolecular assemblies of viral proteins. The particulate nature allows efficient uptake into antigen presenting cells stimulating both innate and adaptive immune responses towards enhanced vaccine efficacy. Increasing research activity and translation opportunity necessitate the advances in the design of VLPs and new bioprocessing modalities for efficient and cost-effective production. Herein, we describe major achievements and challenges in this endeavor, with respect to designing strategies to harnessing the immunogenic potential, production platforms, downstream processes, and some exemplary cases in developing VLP-based vaccines.
Collapse
Affiliation(s)
- Hotcherl Jeong
- Department of Pharmacy, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Baik Lin Seong
- Department of Biotechnology & Vaccine Translational Research Center, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
307
|
Meier AF, Suter M, Schraner EM, Humbel BM, Tobler K, Ackermann M, Laimbacher AS. Transfer of Anti-Rotavirus Antibodies during Pregnancy and in Milk Following Maternal Vaccination with a Herpes Simplex Virus Type-1 Amplicon Vector. Int J Mol Sci 2017; 18:E431. [PMID: 28212334 PMCID: PMC5343965 DOI: 10.3390/ijms18020431] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/01/2017] [Accepted: 02/09/2017] [Indexed: 11/16/2022] Open
Abstract
Rotaviruses (RVs) are important enteric pathogens of newborn humans and animals, causing diarrhea and in rare cases death, especially in very young individuals. Rotavirus vaccines presently used are modified live vaccines that lack complete biological safety. Previous work from our laboratory suggested that vaccines based on in situ produced, non-infectious rotavirus-like particles (RVLPs) are efficient while being entirely safe. However, using either vaccine, active mucosal immunization cannot induce protective immunity in newborns due to their immature immune system. We therefore hypothesized that offspring from vaccinated dams are passively immunized either by transfer of maternal antibodies during pregnancy or by taking up antibodies from milk. Using a codon optimized polycistronic gene expression cassette packaged into herpesvirus particles, the simultaneous expression of the RV capsid genes led to the intracellular formation of RVLPs in various cell lines. Vaccinated dams developed a strong RV specific IgG antibody response determined in sera and milk of both mother and pups. Moreover, sera of naïve pups nursed by vaccinated dams also had RV specific antibodies suggesting a lactogenic transfer of antibodies. Although full protection of pups was not achieved in this mouse model, our observations are important for the development of improved vaccines against RV in humans as well as in various animal species.
Collapse
Affiliation(s)
- Anita F Meier
- Institute of Virology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland.
| | - Mark Suter
- Immunology Division, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland.
| | - Elisabeth M Schraner
- Institutes of Veterinary Anatomy and Virology, University of Zurich, 8057 Zurich, Switzerland.
| | - Bruno M Humbel
- Electron Microscopy Facility, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Kurt Tobler
- Institute of Virology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland.
| | - Mathias Ackermann
- Institute of Virology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland.
| | - Andrea S Laimbacher
- Institute of Virology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
308
|
von Roemeling C, Jiang W, Chan CK, Weissman IL, Kim BY. Breaking Down the Barriers to Precision Cancer Nanomedicine. Trends Biotechnol 2017; 35:159-171. [DOI: 10.1016/j.tibtech.2016.07.006] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/01/2016] [Accepted: 07/15/2016] [Indexed: 02/04/2023]
|
309
|
Gause KT, Wheatley AK, Cui J, Yan Y, Kent SJ, Caruso F. Immunological Principles Guiding the Rational Design of Particles for Vaccine Delivery. ACS NANO 2017; 11:54-68. [PMID: 28075558 DOI: 10.1021/acsnano.6b07343] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Despite the immense public health successes of immunization over the past century, effective vaccines are still lacking for globally important pathogens such as human immunodeficiency virus, malaria, and tuberculosis. Exciting recent advances in immunology and biotechnology over the past few decades have facilitated a shift from empirical to rational vaccine design, opening possibilities for improved vaccines. Some of the most important advancements include (i) the purification of subunit antigens with high safety profiles, (ii) the identification of innate pattern recognition receptors (PRRs) and cognate agonists responsible for inducing immune responses, and (iii) developments in nano- and microparticle fabrication and characterization techniques. Advances in particle engineering now allow highly tunable physicochemical properties of particle-based vaccines, including composition, size, shape, surface characteristics, and degradability. Enhanced collaborative efforts between researchers in immunology and materials science are expected to rise to next-generation vaccines. This process will be significantly aided by a greater understanding of the immunological principles guiding vaccine antigenicity, immunogenicity, and efficacy. With specific emphasis on PRR-targeted adjuvants and particle physicochemical properties, this review aims to provide an overview of the current literature to guide and focus rational particle-based vaccine design efforts.
Collapse
Affiliation(s)
- Katelyn T Gause
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Adam K Wheatley
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity , Parkville, Victoria 3010, Australia
| | - Jiwei Cui
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Yan Yan
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Stephen J Kent
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity , Parkville, Victoria 3010, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| |
Collapse
|
310
|
Penumarthi A, Smooker PM. New approaches to VLP-based vaccines. MICROBIOLOGY AUSTRALIA 2017. [DOI: 10.1071/ma17038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Vaccination is a long and established field of research, and outputs from the research have saved countless millions of lives. The early vaccines were developed with scant regard for the immunological mechanisms at play, largely because they were unknown. We are now in a position to use our knowledge of immunology to rationally design vaccines. This article focusses on the use of virus-like particles (VLPs) as vaccines.
Collapse
|
311
|
Kim H, Kim HJ. Yeast as an expression system for producing virus-like particles: what factors do we need to consider? Lett Appl Microbiol 2016; 64:111-123. [DOI: 10.1111/lam.12695] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/11/2016] [Accepted: 11/04/2016] [Indexed: 12/16/2022]
Affiliation(s)
- H.J. Kim
- Laboratory of Virology; College of Pharmacy; Chung-Ang University; Seoul South Korea
| | - H.-J. Kim
- Laboratory of Virology; College of Pharmacy; Chung-Ang University; Seoul South Korea
| |
Collapse
|
312
|
Karch CP, Burkhard P. Vaccine technologies: From whole organisms to rationally designed protein assemblies. Biochem Pharmacol 2016; 120:1-14. [PMID: 27157411 PMCID: PMC5079805 DOI: 10.1016/j.bcp.2016.05.001] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/04/2016] [Indexed: 11/16/2022]
Abstract
Vaccines have been the single most significant advancement in public health, preventing morbidity and mortality in millions of people annually. Vaccine development has traditionally focused on whole organism vaccines, either live attenuated or inactivated vaccines. While successful for many different infectious diseases whole organisms are expensive to produce, require culture of the infectious agent, and have the potential to cause vaccine associated disease in hosts. With advancing technology and a desire to develop safe, cost effective vaccine candidates, the field began to focus on the development of recombinantly expressed antigens known as subunit vaccines. While more tolerable, subunit vaccines tend to be less immunogenic. Attempts have been made to increase immunogenicity with the addition of adjuvants, either immunostimulatory molecules or an antigen delivery system that increases immune responses to vaccines. An area of extreme interest has been the application of nanotechnology to vaccine development, which allows for antigens to be expressed on a particulate delivery system. One of the most exciting examples of nanovaccines are rationally designed protein nanoparticles. These nanoparticles use some of the basic tenants of structural biology, biophysical chemistry, and vaccinology to develop protective, safe, and easily manufactured vaccines. Rationally developed nanoparticle vaccines are one of the most promising candidates for the future of vaccine development.
Collapse
MESH Headings
- Adjuvants, Immunologic/adverse effects
- Adjuvants, Immunologic/chemistry
- Adjuvants, Immunologic/therapeutic use
- Allergy and Immunology/history
- Allergy and Immunology/trends
- Animals
- Antigens/adverse effects
- Antigens/chemistry
- Antigens/immunology
- Antigens/therapeutic use
- Biopharmaceutics/history
- Biopharmaceutics/methods
- Biopharmaceutics/trends
- Chemistry, Pharmaceutical/history
- Chemistry, Pharmaceutical/trends
- Communicable Disease Control/history
- Communicable Disease Control/trends
- Communicable Diseases/immunology
- Communicable Diseases/veterinary
- Drug Delivery Systems/adverse effects
- Drug Delivery Systems/trends
- Drug Delivery Systems/veterinary
- Drug Design
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Nanoparticles/adverse effects
- Nanoparticles/chemistry
- Nanoparticles/therapeutic use
- Protein Engineering/trends
- Protein Engineering/veterinary
- Protein Folding
- Recombinant Proteins/adverse effects
- Recombinant Proteins/chemistry
- Recombinant Proteins/immunology
- Recombinant Proteins/therapeutic use
- Vaccines/adverse effects
- Vaccines/chemistry
- Vaccines/immunology
- Vaccines/therapeutic use
- Vaccines, Subunit/adverse effects
- Vaccines, Subunit/chemistry
- Vaccines, Subunit/immunology
- Vaccines, Subunit/therapeutic use
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/chemistry
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/therapeutic use
- Veterinary Drugs/adverse effects
- Veterinary Drugs/chemistry
- Veterinary Drugs/immunology
- Veterinary Drugs/therapeutic use
Collapse
Affiliation(s)
- Christopher P Karch
- The Institute of Materials Science, 97 North Eagleville Road, Storrs, CT 06269, United States
| | - Peter Burkhard
- The Institute of Materials Science, 97 North Eagleville Road, Storrs, CT 06269, United States; Department of Molecular and Cell Biology, 93 North Eagleville Road, Storrs, CT 06269, United States.
| |
Collapse
|
313
|
Schwarz B, Morabito KM, Ruckwardt TJ, Patterson DP, Avera J, Miettinen HM, Graham BS, Douglas T. Viruslike Particles Encapsidating Respiratory Syncytial Virus M and M2 Proteins Induce Robust T Cell Responses. ACS Biomater Sci Eng 2016; 2:2324-2332. [PMID: 29367948 PMCID: PMC5777520 DOI: 10.1021/acsbiomaterials.6b00532] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Subunit vaccines provide a safe, focused alternative to conventional vaccines. However, these vaccines often require significant adjuvants and are particularly hard to target toward cytotoxic T lymphocyte (CTL) immunity. Viruslike particles (VLPs) provide biomaterial scaffolds with pathogen-like polyvalent structures making them useful platforms for biomimetic antigen delivery to the immune system. Encapsidation of antigens within VLPs has been shown to enhance antigen availability for CD8 T cell responses. Here, we examine the potential to generate complex responses to multiple subunit antigens localized within the same VLP particle. Two proteins of respiratory syncytial virus (RSV) with well-characterized CD8 T cell responses, the matrix (M) and matrix 2 (M2) proteins, were successfully coencapsidated within the P22 VLP. Upon intranasal administration in mice, the particles stimulated CD8 T cell memory responses against both antigens. In addition, vaccination elicited tissue-resident T cell populations. Upon subsequent RSV challenge, P22-M/M2-treated mice displayed significantly reduced lung viral titers. This demonstrates the utility of the P22 VLP in directing immune responses to multiple encapsidated viral antigens, demonstrating the potential of this technology to facilitate immunity to multiple targets simultaneously.
Collapse
Affiliation(s)
- Benjamin Schwarz
- Department of Chemistry, Indiana University, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Kaitlyn M. Morabito
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, District of Columbia 20007, United States
| | - Tracy J. Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dustin P. Patterson
- Department of Chemistry and Biochemistry, University of Texas at Tyler, 3900 University Boulevard, Tyler, Texas 75799, United States
| | - John Avera
- Department of Chemistry, Indiana University, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Heini M. Miettinen
- Department of Microbiology and Immunology, Montana State University, PO Box 173400, Bozeman, Montana 59717, United States
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Trevor Douglas
- Department of Chemistry, Indiana University, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| |
Collapse
|
314
|
Giddam AK, Reiman JM, Zaman M, Skwarczynski M, Toth I, Good MF. A semi-synthetic whole parasite vaccine designed to protect against blood stage malaria. Acta Biomater 2016; 44:295-303. [PMID: 27544810 DOI: 10.1016/j.actbio.2016.08.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Although attenuated malaria parasitized red blood cells (pRBCs) are promising vaccine candidates, their application in humans may be restricted for ethical and regulatory reasons. Therefore, we developed an organic microparticle-based delivery platform as a whole parasite malaria-antigen carrier to mimic pRBCs. Killed blood stage parasites were encapsulated within liposomes that are targeted to antigen presenting cells (APCs). Mannosylated lipid core peptides (MLCPs) were used as targeting ligands for the liposome-encapsulated parasite antigens. MLCP-liposomes, but not unmannosylated liposomes, were taken-up efficiently by APCs which then significantly upregulated expression of MHC-ll and costimulatory molecules, CD80 and CD86. Two such vaccines using rodent model systems were constructed - one with Plasmodium chabaudi and the other with P. yoelii. MLCP-liposome vaccines were able to control the parasite burden and extended the survival of mice. Thus, we have demonstrated an alternative delivery system to attenuated pRBCs with similar vaccine efficacy and added clinical advantages. Such liposomes are promising candidates for a human malaria vaccine. STATEMENT OF SIGNIFICANCE Attenuated whole parasite-based vaccines, by incorporating all parasite antigens, are very promising candidates, but issues relating to production, storage and safety concerns are significantly slowing their development. We therefore developed a semi-synthetic whole parasite malaria vaccine that is easily manufactured and stored. Two such prototype vaccines (a P. chabaudi and a P. yoelii vaccine) have been constructed. They are non-infectious, highly immunogenic and give good protection profiles. This semi-synthetic delivery platform is an exciting strategy to accelerate the development of a licensed malaria vaccine. Moreover, this strategy can be potentially applied to a wide range of pathogens.
Collapse
|
315
|
Zilker C, Kozlova D, Sokolova V, Yan H, Epple M, Überla K, Temchura V. Nanoparticle-based B-cell targeting vaccines: Tailoring of humoral immune responses by functionalization with different TLR-ligands. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:173-182. [PMID: 27593489 DOI: 10.1016/j.nano.2016.08.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/01/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
Induction of an appropriate type of humoral immune response during vaccination is essential for protection against viral and bacterial infections. We recently observed that biodegradable calcium phosphate (CaP) nanoparticles coated with proteins efficiently targeted and activated naïve antigen-specific B-cells in vitro. We now compared different administration routes for CaP-nanoparticles and demonstrated that intramuscular immunization with such CaP-nanoparticles induced stronger immune responses than immunization with monovalent antigen. Additional functionalization of the CaP-nanoparticles with TRL-ligands allowed modulating the IgG subtype response and the level of mucosal IgA antibodies. CpG-containing CaP-nanoparticles were as immunogenic as a virus-like particle vaccine. Functionalization of CaP-nanoparticles with T-helper cell epitopes or CpG also allowed overcoming lack of T-cell help. Thus, our results indicate that CaP-nanoparticle-based B-cell targeting vaccines functionalized with TLR-ligands can serve as a versatile platform for efficient induction and modulation of humoral immune responses in vivo.
Collapse
Affiliation(s)
- Claudia Zilker
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Diana Kozlova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| | - Viktoriya Sokolova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| | - Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| | - Klaus Überla
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany; University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Vladimir Temchura
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany; University Hospital Erlangen, Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
316
|
Rabbit hemorrhagic disease virus capsid, a versatile platform for foreign B-cell epitope display inducing protective humoral immune responses. Sci Rep 2016; 6:31844. [PMID: 27549017 PMCID: PMC4994115 DOI: 10.1038/srep31844] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/27/2016] [Indexed: 11/30/2022] Open
Abstract
Virus-like particles (VLPs), comprised of viral structural proteins devoid of genetic material, are tunable nanoparticles that can be chemically or genetically engineered, to be used as platforms for multimeric display of foreign antigens. Here, we report the engineering of chimeric VLPs, derived from rabbit hemorrhagic disease virus (RHDV) for presentation of foreign B-cell antigens to the immune system. The RHDV capsid comprises 180 copies of a single capsid subunit (VP60). To evaluate the ability of chimeric RHDV VLPs to elicit protective humoral responses against foreign antigens, we tested two B-cell epitopes: a novel neutralizing B-cell epitope, derived from feline calicivirus capsid protein, and a well characterized B-cell epitope from the extracellular domain of influenza A virus M2 protein (M2e). We generated sets of chimeric RHDV VLPs by insertion of the foreign B-cell epitopes at three different locations within VP60 protein (which involved different levels of surface accessibility) and in different copy numbers per site. The immunogenic potential of the chimeric VLPs was analyzed in the mouse model. The results presented here indicated that chimeric RHDV VLPs elicit potent protective humoral responses against displayed foreign B-cell epitopes, demonstrated by both, in vitro neutralization and in vivo protection against a lethal challenge.
Collapse
|
317
|
Choi B, Moon H, Hong SJ, Shin C, Do Y, Ryu S, Kang S. Effective Delivery of Antigen-Encapsulin Nanoparticle Fusions to Dendritic Cells Leads to Antigen-Specific Cytotoxic T Cell Activation and Tumor Rejection. ACS NANO 2016; 10:7339-50. [PMID: 27390910 DOI: 10.1021/acsnano.5b08084] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
In cancer immunotherapy, robust and efficient activation of cytotoxic CD8(+) T cell immune responses is a promising, but challenging task. Dendritic cells (DCs) are well-known professional antigen presenting cells that initiate and regulate antigen-specific cytotoxic CD8(+) T cells that kill their target cells directly as well as secrete IFN-γ, a cytokine critical in tumor rejection. Here, we employed recently established protein cage nanoparticles, encapsulin (Encap), as antigenic peptide nanocarriers by genetically incorporating the OT-1 peptide of ovalbumin (OVA) protein to the three different positions of the Encap subunit. With them, we evaluated their efficacy in activating DC-mediated antigen-specific T cell cytotoxicity and consequent melanoma tumor rejection in vivo. DCs efficiently engulfed Encap and its variants (OT-1-Encaps), which carry antigenic peptides at different positions, and properly processed them within phagosomes. Delivered OT-1 peptides were effectively presented by DCs to naïve CD8(+) T cells successfully, resulting in the proliferation of antigen-specific cytotoxic CD8(+) T cells. OT-1-Encap vaccinations in B16-OVA melanoma tumor bearing mice effectively activated OT-1 peptide specific cytotoxic CD8(+) T cells before or even after tumor generation, resulting in significant suppression of tumor growth in prophylactic as well as therapeutic treatments. A large number of cytotoxic CD8(+) T cells that actively produce both intracellular and secretory IFN-γ were observed in tumor-infiltrating lymphocytes collected from B16-OVA tumor masses originally vaccinated with OT-1-Encap-C upon tumor challenges. The approaches we describe herein may provide opportunities to develop epitope-dependent vaccination systems that stimulate and/or modulate efficient and epitope-specific cytotoxic T cell immune responses in nonpathogenic diseases.
Collapse
Affiliation(s)
- Bongseo Choi
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, 689-798, Korea
| | - Hyojin Moon
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, 689-798, Korea
| | - Sung Joon Hong
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, 689-798, Korea
| | - Changsik Shin
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, 689-798, Korea
| | - Yoonkyung Do
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, 689-798, Korea
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University , Cheonan, 336-745, Korea
| | - Sebyung Kang
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST) , Ulsan, 689-798, Korea
| |
Collapse
|
318
|
Zhang X, Wang K, Lin Q, Zheng M, Li Q, Li T, Hong Q, Zheng Q, Yu H, Gu Y, Li S, Xia N. A shared N-terminal hydrophobic tail for the formation of nanoparticulates. Nanomedicine (Lond) 2016; 11:2289-303. [PMID: 27499052 DOI: 10.2217/nnm-2016-0146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
AIM Nanoparticulate design is important for the production of nanotechnological materials and passive immunogens. Using lessons from our hepatitis E vaccine, we herein design protein-based nanoparticles through incorporation of an N-terminal hydrophobic tail (NHT, located on HEV ORF2 aa368-460). MATERIALS & METHODS Flu HA1, HIV gp41/gp120/p24, HBsAg and HPV16 L2 were fused with NHT, expressed in Escherichia coli and subjected to self-assembly in vitro. Nanosized particles were characterized by size-exclusion chromatography and negative electron microscopy. Immunogenicity was assessed in mice. RESULTS All the NHT-fused proteins spontaneously formed nanoparticulates and presented with immunogenicity approximately 2-log over their nonassembling forms. CONCLUSION Protein self-assembly provides an attractive means to create nanosized particles that bear specific antigens. Our strategy outlines a novel and shared method for the design of immunogenic nanoparticles.
Collapse
Affiliation(s)
- Xiao Zhang
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China.,National Institute of Diagnostics & Vaccine Development in Infectious Disease, School of Public Health, Xiamen University, Xiamen, China
| | - Kaihang Wang
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qingshan Lin
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China
| | - Minghua Zheng
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qiong Li
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China
| | - Tingting Li
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qiyang Hong
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qingbing Zheng
- National Institute of Diagnostics & Vaccine Development in Infectious Disease, School of Public Health, Xiamen University, Xiamen, China
| | - Hai Yu
- National Institute of Diagnostics & Vaccine Development in Infectious Disease, School of Public Health, Xiamen University, Xiamen, China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China.,National Institute of Diagnostics & Vaccine Development in Infectious Disease, School of Public Health, Xiamen University, Xiamen, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China.,National Institute of Diagnostics & Vaccine Development in Infectious Disease, School of Public Health, Xiamen University, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen, China.,National Institute of Diagnostics & Vaccine Development in Infectious Disease, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
319
|
Kim HJ, Kwag HL, Kim DG, Kang BK, Han SY, Moon H, Hwang JY, Kwon MG, Kang HA, Kim HJ. Assembly of the capsid protein of red-spotted grouper nervous necrosis virus during purification, and role of calcium ions in chromatography. BIOTECHNOL BIOPROC E 2016. [DOI: 10.1007/s12257-016-0256-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
320
|
Li Y, Yi X, Zhuang Y, Chu J. Regulation of porcine circovirus type 2-like particles expressed in baculovirus expression system. BIORESOUR BIOPROCESS 2016. [DOI: 10.1186/s40643-016-0114-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
321
|
Bobbala S, Hook S. Is There an Optimal Formulation and Delivery Strategy for Subunit Vaccines? Pharm Res 2016; 33:2078-97. [DOI: 10.1007/s11095-016-1979-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/21/2016] [Indexed: 12/16/2022]
|
322
|
Presenting native-like trimeric HIV-1 antigens with self-assembling nanoparticles. Nat Commun 2016; 7:12041. [PMID: 27349934 PMCID: PMC4931238 DOI: 10.1038/ncomms12041] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/24/2016] [Indexed: 01/01/2023] Open
Abstract
Structures of BG505 SOSIP.664 trimer in complex with broadly neutralizing antibodies (bNAbs) have revealed the critical role of trimeric context for immune recognition of HIV-1. Presentation of trimeric HIV-1 antigens on nanoparticles may thus provide promising vaccine candidates. Here we report the rational design, structural analysis and antigenic evaluation of HIV-1 trimer-presenting nanoparticles. We first demonstrate that both V1V2 and gp120 can be presented in native-like trimeric conformations on nanoparticles. We then design nanoparticles presenting various forms of stabilized gp140 trimer based on ferritin and a large, 60-meric E2p that displays 20 spikes mimicking virus-like particles (VLPs). Particle assembly is confirmed by electron microscopy (EM), while antigenic profiles are generated using representative bNAbs and non-NAbs. Lastly, we demonstrate high-yield gp140 nanoparticle production and robust stimulation of B cells carrying cognate VRC01 receptors by gp120 and gp140 nanoparticles. Together, our study provides an arsenal of multivalent immunogens for HIV-1 vaccine development.
Collapse
|
323
|
Scalable chromatography-based purification of virus-like particle carrier for epitope based influenza A vaccine produced in Escherichia coli. J Virol Methods 2016; 232:8-11. [DOI: 10.1016/j.jviromet.2016.02.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/14/2016] [Accepted: 02/18/2016] [Indexed: 01/06/2023]
|
324
|
Immunogenicity of Virus Like Particle Forming Baculoviral DNA Vaccine against Pandemic Influenza H1N1. PLoS One 2016; 11:e0154824. [PMID: 27149064 PMCID: PMC4858234 DOI: 10.1371/journal.pone.0154824] [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: 02/02/2016] [Accepted: 04/19/2016] [Indexed: 12/29/2022] Open
Abstract
An outbreak of influenza H1N1 in 2009, representing the first influenza pandemic of the 21st century, was transmitted to over a million individuals and claimed 18,449 lives. The current status in many countries is to prepare influenza vaccine using cell-based or egg-based killed vaccine. However, traditional influenza vaccine platforms have several limitations. To overcome these limitations, many researchers have tried various approaches to develop alternative production platforms. One of the alternative approach, we reported the efficacy of influenza HA vaccination using a baculoviral DNA vaccine (AcHERV-HA). However, the immune response elicited by the AcHERV-HA vaccine, which only targets the HA antigen, was lower than that of the commercial killed vaccine. To overcome the limitations of this previous vaccine, we constructed a human endogenous retrovirus (HERV) envelope-coated, baculovirus-based, virus-like-particle (VLP)–forming DNA vaccine (termed AcHERV-VLP) against pandemic influenza A/California/04/2009 (pH1N1). BALB/c mice immunized with AcHERV-VLP (1×107 FFU AcHERV-VLP, i.m.) and compared with mice immunized with the killed vaccine or mice immunized with AcHERV-HA. As a result, AcHERV-VLP immunization produced a greater humoral immune response and exhibited neutralizing activity with an intrasubgroup H1 strain (PR8), elicited neutralizing antibody production, a high level of interferon-γ secretion in splenocytes, and diminished virus shedding in the lung after challenge with a lethal dose of influenza virus. In conclusion, VLP-forming baculovirus DNA vaccine could be a potential vaccine candidate capable of efficiently delivering DNA to the vaccinee and VLP forming DNA eliciting stronger immunogenicity than egg-based killed vaccines.
Collapse
|
325
|
Adjuvants: Classification, Modus Operandi, and Licensing. J Immunol Res 2016; 2016:1459394. [PMID: 27274998 PMCID: PMC4870346 DOI: 10.1155/2016/1459394] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/02/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023] Open
Abstract
Vaccination is one of the most efficient strategies for the prevention of infectious diseases. Although safer, subunit vaccines are poorly immunogenic and for this reason the use of adjuvants is strongly recommended. Since their discovery in the beginning of the 20th century, adjuvants have been used to improve immune responses that ultimately lead to protection against disease. The choice of the adjuvant is of utmost importance as it can stimulate protective immunity. Their mechanisms of action have now been revealed. Our increasing understanding of the immune system, and of correlates of protection, is helping in the development of new vaccine formulations for global infections. Nevertheless, few adjuvants are licensed for human vaccines and several formulations are now being evaluated in clinical trials. In this review, we briefly describe the most well known adjuvants used in experimental and clinical settings based on their main mechanisms of action and also highlight the requirements for licensing new vaccine formulations.
Collapse
|
326
|
Lee MFX, Chan ES, Tan WS, Tam KC, Tey BT. Negative chromatography of hepatitis B virus-like particle: Comparative study of different adsorbent designs. J Chromatogr A 2016; 1445:1-9. [DOI: 10.1016/j.chroma.2016.03.066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/09/2016] [Accepted: 03/22/2016] [Indexed: 02/06/2023]
|
327
|
Koch C, Eber FJ, Azucena C, Förste A, Walheim S, Schimmel T, Bittner AM, Jeske H, Gliemann H, Eiben S, Geiger FC, Wege C. Novel roles for well-known players: from tobacco mosaic virus pests to enzymatically active assemblies. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2016; 7:613-29. [PMID: 27335751 PMCID: PMC4901926 DOI: 10.3762/bjnano.7.54] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/03/2016] [Indexed: 05/22/2023]
Abstract
The rod-shaped nanoparticles of the widespread plant pathogen tobacco mosaic virus (TMV) have been a matter of intense debates and cutting-edge research for more than a hundred years. During the late 19th century, their behavior in filtration tests applied to the agent causing the 'plant mosaic disease' eventually led to the discrimination of viruses from bacteria. Thereafter, they promoted the development of biophysical cornerstone techniques such as electron microscopy and ultracentrifugation. Since the 1950s, the robust, helically arranged nucleoprotein complexes consisting of a single RNA and more than 2100 identical coat protein subunits have enabled molecular studies which have pioneered the understanding of viral replication and self-assembly, and elucidated major aspects of virus-host interplay, which can lead to agronomically relevant diseases. However, during the last decades, TMV has acquired a new reputation as a well-defined high-yield nanotemplate with multivalent protein surfaces, allowing for an ordered high-density presentation of multiple active molecules or synthetic compounds. Amino acid side chains exposed on the viral coat may be tailored genetically or biochemically to meet the demands for selective conjugation reactions, or to directly engineer novel functionality on TMV-derived nanosticks. The natural TMV size (length: 300 nm) in combination with functional ligands such as peptides, enzymes, dyes, drugs or inorganic materials is advantageous for applications ranging from biomedical imaging and therapy approaches over surface enlargement of battery electrodes to the immobilization of enzymes. TMV building blocks are also amenable to external control of in vitro assembly and re-organization into technically expedient new shapes or arrays, which bears a unique potential for the development of 'smart' functional 3D structures. Among those, materials designed for enzyme-based biodetection layouts, which are routinely applied, e.g., for monitoring blood sugar concentrations, might profit particularly from the presence of TMV rods: Their surfaces were recently shown to stabilize enzymatic activities upon repeated consecutive uses and over several weeks. This review gives the reader a ride through strikingly diverse achievements obtained with TMV-based particles, compares them to the progress with related viruses, and focuses on latest results revealing special advantages for enzyme-based biosensing formats, which might be of high interest for diagnostics employing 'systems-on-a-chip'.
Collapse
Affiliation(s)
- Claudia Koch
- Institute of Biomaterials and Biomolecular Systems, Department of Molecular Biology and Plant Virology, University of Stuttgart, Pfaffenwaldring 57, Stuttgart, D-70550, Germany
| | - Fabian J Eber
- Institute of Biomaterials and Biomolecular Systems, Department of Molecular Biology and Plant Virology, University of Stuttgart, Pfaffenwaldring 57, Stuttgart, D-70550, Germany
| | - Carlos Azucena
- Institute of Functional Interfaces (IFG), Chemistry of Oxidic and Organic Interfaces, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Karlsruhe, D-76344, Germany
| | - Alexander Förste
- Institute of Nanotechnology (INT) and Karlsruhe Institute of Applied Physics (IAP) and Center for Functional Nanostructures (CFN), Karlsruhe Institute of Technology (KIT), INT: Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, D-76344, Germany, and IAP/CFN: Wolfgang-Gaede-Straße 1, Karlsruhe, D-76131 Germany
| | - Stefan Walheim
- Institute of Nanotechnology (INT) and Karlsruhe Institute of Applied Physics (IAP) and Center for Functional Nanostructures (CFN), Karlsruhe Institute of Technology (KIT), INT: Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, D-76344, Germany, and IAP/CFN: Wolfgang-Gaede-Straße 1, Karlsruhe, D-76131 Germany
| | - Thomas Schimmel
- Institute of Nanotechnology (INT) and Karlsruhe Institute of Applied Physics (IAP) and Center for Functional Nanostructures (CFN), Karlsruhe Institute of Technology (KIT), INT: Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, D-76344, Germany, and IAP/CFN: Wolfgang-Gaede-Straße 1, Karlsruhe, D-76131 Germany
| | - Alexander M Bittner
- CIC Nanogune, Tolosa Hiribidea 76, E-20018 Donostia-San Sebastián, Spain, and Ikerbasque, Maria Díaz de Haro 3, E-48013 Bilbao, Spain
| | - Holger Jeske
- Institute of Biomaterials and Biomolecular Systems, Department of Molecular Biology and Plant Virology, University of Stuttgart, Pfaffenwaldring 57, Stuttgart, D-70550, Germany
| | - Hartmut Gliemann
- Institute of Functional Interfaces (IFG), Chemistry of Oxidic and Organic Interfaces, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Karlsruhe, D-76344, Germany
| | - Sabine Eiben
- Institute of Biomaterials and Biomolecular Systems, Department of Molecular Biology and Plant Virology, University of Stuttgart, Pfaffenwaldring 57, Stuttgart, D-70550, Germany
| | - Fania C Geiger
- Institute of Biomaterials and Biomolecular Systems, Department of Molecular Biology and Plant Virology, University of Stuttgart, Pfaffenwaldring 57, Stuttgart, D-70550, Germany
| | - Christina Wege
- Institute of Biomaterials and Biomolecular Systems, Department of Molecular Biology and Plant Virology, University of Stuttgart, Pfaffenwaldring 57, Stuttgart, D-70550, Germany
| |
Collapse
|
328
|
Guo H, Zhu J, Tan Y, Li C, Chen Z, Sun S, Liu G. Self-assembly of virus-like particles of rabbit hemorrhagic disease virus capsid protein expressed in Escherichia coli and their immunogenicity in rabbits. Antiviral Res 2016; 131:85-91. [PMID: 27118636 DOI: 10.1016/j.antiviral.2016.04.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/09/2016] [Accepted: 04/18/2016] [Indexed: 12/23/2022]
Abstract
In this study, virus-like particles (VLPs) derived from rabbit hemorrhagic disease virus (RHDV) were evaluated for the development of a vaccine against RHDV infection. The VP60 gene was cloned and inserted into a pSMK expression vector containing a small ubiquitin-like modifier (SUMO) tag that can promote the soluble expression of heterologous proteins in Escherichia coli cells. After expression and purification of His-SUMO-VP60 and cleavage of the SUMO tag, we found that the RHDV VP60 protein had self-assembled into VLPs with a similar shape and smaller size compared with authentic RHDV capsid. Next, the antigenicity and immunogenicity of the VLPs were examined. The results showed that RHDV-specific responses were clearly induced in rabbits and that all rabbits in the VLP group survived while those in the negative control group died within 72 h post-infection. These results suggest that VLP-based RHDV could be a promising RHDV vaccine candidate.
Collapse
Affiliation(s)
- Huimin Guo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Jie Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Yonggui Tan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Chuanfeng Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Zongyan Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Shiqi Sun
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 730046, Gansu, China.
| | - Guangqing Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China.
| |
Collapse
|
329
|
Baltabekova AZ, Shagyrova ZS, Kamzina AS, Voykov M, Zhiyenbay Y, Ramanculov EM, Shustov AV. SplitCore Technology Allows Efficient Production of Virus-Like Particles Presenting a Receptor-Contacting Epitope of Human IgE. Mol Biotechnol 2016; 57:746-55. [PMID: 25837568 DOI: 10.1007/s12033-015-9867-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Immunoglobulin E (IgE) plays a central role in type I hypersensitivity including allergy and asthma. Novel treatment strategy envisages development of a therapeutic vaccine designed to elicit autologous blocking antibodies against the IgE. We sought to develop an IgE-epitope antigen that induces antibodies against a receptor-contacting epitope on human IgE molecule. We designed the VLP immunogens which utilize hepatitis B virus core protein (HBcAg) as a carrier, and present arrays of the receptor-contacting epitopes of the human IgE on their surfaces. FG loop from the IgE domain Cε3 was engineered into the HBcAg. Two constructs explore a well-established approach of insertion into a main immunodominant region of the HBcAg. Third construct is different in that the carrier is produced in a form of an assembly of two polypeptide chains which upon expression remain associated in a stable VLP-forming subunit (SplitCore technology). No VLPs were isolated from E.coli expressing the IgE-epitope antigens with contiguous sequences. On the contrary, the SplitCore antigen carrying the FG loop efficiently formed the VLPs. Immunization of mice with the VLPs presenting receptor-contacting epitope of the IgE elicited antibodies recognizing the human IgE in ELISA.
Collapse
Affiliation(s)
- A Zh Baltabekova
- National Center for Biotechnology, Valikhanova 13/1, 010000, Astana, Kazakhstan
| | | | | | | | | | | | | |
Collapse
|
330
|
Challenges and opportunities of using liquid chromatography and mass spectrometry methods to develop complex vaccine antigens as pharmaceutical dosage forms. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1032:23-38. [PMID: 27071526 DOI: 10.1016/j.jchromb.2016.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 12/22/2022]
Abstract
Liquid chromatographic methods, combined with mass spectrometry, offer exciting and important opportunities to better characterize complex vaccine antigens including recombinant proteins, virus-like particles, inactivated viruses, polysaccharides, and protein-polysaccharide conjugates. The current abilities and limitations of these physicochemical methods to complement traditional in vitro and in vivo vaccine potency assays are explored in this review through the use of illustrative case studies. Various applications of these state-of-the art techniques are illustrated that include the analysis of influenza vaccines (inactivated whole virus and recombinant hemagglutinin), virus-like particle vaccines (human papillomavirus and hepatitis B), and polysaccharide linked to protein carrier vaccines (pneumococcal). Examples of utilizing these analytical methods to characterize vaccine antigens in the presence of adjuvants, which are often included to boost immune responses as part of the final vaccine dosage form, are also presented. Some of the challenges of using chromatographic and LC-MS as physicochemical assays to routinely test complex vaccine antigens are also discussed.
Collapse
|
331
|
Shirbaghaee Z, Bolhassani A. Different applications of virus-like particles in biology and medicine: Vaccination and delivery systems. Biopolymers 2016; 105:113-32. [PMID: 26509554 PMCID: PMC7161881 DOI: 10.1002/bip.22759] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/25/2015] [Accepted: 10/25/2015] [Indexed: 12/17/2022]
Abstract
Virus-like particles (VLPs) mimic the whole construct of virus particles devoid of viral genome as used in subunit vaccine design. VLPs can elicit efficient protective immunity as direct immunogens compared to soluble antigens co-administered with adjuvants in several booster injections. Up to now, several prokaryotic and eukaryotic systems such as insect, yeast, plant, and E. coli were used to express recombinant proteins, especially for VLP production. Recent studies are also generating VLPs in plants using different transient expression vectors for edible vaccines. VLPs and viral particles have been applied for different functions such as gene therapy, vaccination, nanotechnology, and diagnostics. Herein, we describe VLP production in different systems as well as its applications in biology and medicine.
Collapse
Affiliation(s)
- Zeinab Shirbaghaee
- Department of Hepatitis and AIDSPasteur Institute of IranTehranIran
- Department of Immunology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Azam Bolhassani
- Department of Hepatitis and AIDSPasteur Institute of IranTehranIran
| |
Collapse
|
332
|
Zhu R, Liu J, Chen C, Ye X, Xu L, Wang W, Zhao Q, Zhu H, Cheng T, Xia N. A highly conserved epitope-vaccine candidate against varicella-zoster virus induces neutralizing antibodies in mice. Vaccine 2016; 34:1589-1596. [PMID: 26873057 DOI: 10.1016/j.vaccine.2016.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 01/22/2016] [Accepted: 02/01/2016] [Indexed: 10/22/2022]
Abstract
Varicella-zoster virus (VZV) is a highly infectious agent of varicella and herpes zoster (HZ). Vaccination is by far the most effective way to prevent these diseases. More safe, stable and efficient vaccines, such as epitope-based vaccines, now have been increasingly investigated by many researchers. However, only a few VZV neutralizing epitopes have been identified to date. We have previously identified a linear epitope between amino acid residues 121 and 135 of gE. In this study, we validated that this epitope is highly conserved amongst different VZV strains that covered five existing phylogenetic clades with an identity of 100%. We evaluated the immunogenicity of the recombinant hepatitis B virus core (HBc) virus-like particles (VLPs) which included amino acids (121-135). VZV-gE-specific antibodies were detected in immunized mouse serum using ELISA. The anti-peptide antiserum positively detected VZV via Western blot and immunofluorescent staining assays. More importantly, these peptides could neutralize VZV, indicating that these peptides represented neutralizing epitopes. These findings have important implications for the development of epitope-based protective VZV vaccines.
Collapse
Affiliation(s)
- Rui Zhu
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jian Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chunye Chen
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiangzhong Ye
- Beijing Wantai Biological Pharmacy Enterprise, Beijing 102206, China
| | - Longfa Xu
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wei Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Qinjian Zhao
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hua Zhu
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
333
|
Structural studies on chimeric Sesbania mosaic virus coat protein: Revisiting SeMV assembly. Virology 2016; 489:34-43. [DOI: 10.1016/j.virol.2015.11.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 11/27/2015] [Accepted: 11/28/2015] [Indexed: 01/25/2023]
|
334
|
Molino NM, Neek M, Tucker JA, Nelson EL, Wang SW. Viral-mimicking protein nanoparticle vaccine for eliciting anti-tumor responses. Biomaterials 2016; 86:83-91. [PMID: 26894870 DOI: 10.1016/j.biomaterials.2016.01.056] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 01/25/2016] [Indexed: 12/19/2022]
Abstract
The immune system is a powerful resource for the eradication of cancer, but to overcome the low immunogenicity of tumor cells, a sufficiently strong CD8(+) T cell-mediated adaptive immune response is required. Nanoparticulate biomaterials represent a potentially effective delivery system for cancer vaccines, as they can be designed to mimic viruses, which are potent inducers of cellular immunity. We have been exploring the non-viral pyruvate dehydrogenase E2 protein nanoparticle as a biomimetic platform for cancer vaccine delivery. Simultaneous conjugation of a melanoma-associated gp100 epitope and CpG to the E2 nanoparticle (CpG-gp-E2) yielded an antigen-specific increase in the CD8(+) T cell proliferation index and IFN-γ secretion by 1.5-fold and 5-fold, respectively, compared to an unbound peptide and CpG formulation. Remarkably, a single nanoparticle immunization resulted in a 120-fold increase in the frequency of melanoma epitope-specific CD8(+) T cells in draining lymph nodes and a 30-fold increase in the spleen, relative to free peptide with free CpG. Furthermore, in the very aggressive B16 melanoma murine tumor model, prophylactic immunization with CpG-gp-E2 delayed the onset of tumor growth by approximately 5.5 days and increased animal survival time by approximately 40%, compared to PBS-treated animals. These results show that by combining optimal particle size and simultaneous co-delivery of molecular vaccine components, antigen-specific anti-tumor immune responses can be significantly increased.
Collapse
Affiliation(s)
- Nicholas M Molino
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA
| | - Medea Neek
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA
| | - Jo Anne Tucker
- Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Edward L Nelson
- Department of Medicine, University of California, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, CA 92697, USA
| | - Szu-Wen Wang
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
335
|
Foerster J, Bachman M. Beyond passive immunization: toward a nanoparticle-based IL-17 vaccine as first in class of future immune treatments. Nanomedicine (Lond) 2016; 10:1361-9. [PMID: 25955128 DOI: 10.2217/nnm.14.215] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Nanoparticles occur naturally as part of repetitive molecular structures forming virus-like particles (VLPs). VLPs are powerful immune activators. Specifically, VLP can elicit a direct activation of B lymphocytes to trigger production of antibodies targeted at molecules chemically linked to the VLP. We here review recent data from genetics research, large-scale genomic sequencing, as well as clinical trials which suggest that a VLP-based vaccine against the signaling molecule IL-17 will be safe and effective in the common skin disease psoriasis, as well as other conditions. Active vaccination against IL-17 is capable of replacing the costly manufacture of antibodies currently in clinical use with huge implications for treatment availability and health economics.
Collapse
Affiliation(s)
- John Foerster
- College of Medicine, Dentistry, & Nursing, University of Dundee, Dundee, DD1 9SY, UK
| | | |
Collapse
|
336
|
Zhao C, Ao Z, Yao X. Current Advances in Virus-Like Particles as a Vaccination Approach against HIV Infection. Vaccines (Basel) 2016; 4:vaccines4010002. [PMID: 26805898 PMCID: PMC4810054 DOI: 10.3390/vaccines4010002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 12/31/2015] [Accepted: 01/18/2016] [Indexed: 12/16/2022] Open
Abstract
HIV-1 virus-like particles (VLPs) are promising vaccine candidates against HIV-1 infection. They are capable of preserving the native conformation of HIV-1 antigens and priming CD4+ and CD8+ T cell responses efficiently via cross presentation by both major histocompatibility complex (MHC) class I and II molecules. Progress has been achieved in the preclinical research of HIV-1 VLPs as prophylactic vaccines that induce broadly neutralizing antibodies and potent T cell responses. Moreover, the progress in HIV-1 dendritic cells (DC)-based immunotherapy provides us with a new vision for HIV-1 vaccine development. In this review, we describe updates from the past 5 years on the development of HIV-1 VLPs as a vaccine candidate and on the combined use of HIV particles with HIV-1 DC-based immunotherapy as efficient prophylactic and therapeutic vaccination strategies.
Collapse
Affiliation(s)
- Chongbo Zhao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
- Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China.
| |
Collapse
|
337
|
Molecular pharming - VLPs made in plants. Curr Opin Biotechnol 2016; 37:201-206. [PMID: 26773389 DOI: 10.1016/j.copbio.2015.12.007] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 12/08/2015] [Accepted: 12/10/2015] [Indexed: 10/22/2022]
Abstract
Plant-based expression offers a safe, inexpensive and potentially limitless way to produce therapeutics in a quick and flexible manner. Plants require only simple inorganic nutrients, water, carbon dioxide and sunlight for efficient growth. Virus-like particles (VLPs) are convincing look-alikes of viruses but without carrying infectious genomic material. However, they can still elicit a very potent immune response which makes them ideal vaccine candidates. In this review the different methods of plant expression are described together with the most recent developments in the field of transiently-expressed plant-made VLPs.
Collapse
|
338
|
Kivi G, Teesalu K, Parik J, Kontkar E, Ustav M, Noodla L, Ustav M, Männik A. HybriFree: a robust and rapid method for the development of monoclonal antibodies from different host species. BMC Biotechnol 2016; 16:2. [PMID: 26747451 PMCID: PMC4706699 DOI: 10.1186/s12896-016-0232-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/04/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The production of recombinant monoclonal antibodies in mammalian cell culture is of high priority in research and medical fields. A critical step in this process is the isolation of the antigen-binding domain sequences of antibodies possessing the desired properties. Many different techniques have been described to achieve this goal, but all have shortcomings; most techniques have problems with robustness, are time-consuming and costly, or have complications in the transfer from isolation to production phase. Here, we report a novel HybriFree technology for the development of monoclonal antibodies from different species that is robust, rapid, inexpensive and flexible and can be used for the subsequent production of antibodies in mammalian cell factories. RESULTS HybriFree technology is illustrated herein via detailed examples of isolating mouse, rabbit and chicken monoclonal antibody sequences from immunized animals. Starting from crude spleen samples, antigen capturing of specific B-cells is performed initially. cDNA of antibody variable domains is amplified from the captured cells and used a source material for simple and rapid restriction/ligation free cloning of expression vector library in order to produce scFv-Fc or intact IgG antibodies. The vectors can be directly used for screening purposes as well as for the subsequent production of the developed monoclonal antibodies in mammalian cell culture. The antibodies isolated by the method have been shown to be functional in different immunoassays, including ELISA, immunofluorescence and Western blot. In addition, we demonstrate that by using a modified method including a negative selection step, we can isolate specific antibodies targeting the desired epitope and eliminate antibodies directed to undesired off-targets. CONCLUSIONS HybriFree can be used for the reliable development of monoclonal antibodies and their subsequent production in mammalian cells. This simple protocol requires neither the culturing of B-cells nor single-cell manipulations, and only standard molecular biology laboratory equipment is needed. In principle, the method is applicable to any species for which antibody cDNA sequence information is available.
Collapse
Affiliation(s)
- Gaily Kivi
- Icosagen Cell Factory OÜ, Eerika tee 1, Õssu village, Ülenurme parish, Tartumaa, 61713, Estonia. .,University of Tartu, Institute of Technology, Nooruse1, Tartu, 50411, Estonia.
| | - Kaupo Teesalu
- Icosagen Cell Factory OÜ, Eerika tee 1, Õssu village, Ülenurme parish, Tartumaa, 61713, Estonia.
| | - Jüri Parik
- Estonian Biocentre, Evolutionary Biology Group, Tartu, 51010, Estonia.
| | - Elen Kontkar
- Icosagen Cell Factory OÜ, Eerika tee 1, Õssu village, Ülenurme parish, Tartumaa, 61713, Estonia.
| | - Mart Ustav
- University of Tartu, Institute of Technology, Nooruse1, Tartu, 50411, Estonia.
| | - Liis Noodla
- University of Tartu, Institute of Technology, Nooruse1, Tartu, 50411, Estonia.
| | - Mart Ustav
- Icosagen Cell Factory OÜ, Eerika tee 1, Õssu village, Ülenurme parish, Tartumaa, 61713, Estonia. .,University of Tartu, Institute of Technology, Nooruse1, Tartu, 50411, Estonia. .,Estonian Academy of Sciences, Kohtu 6, Tallinn, 10130, Estonia.
| | - Andres Männik
- Icosagen Cell Factory OÜ, Eerika tee 1, Õssu village, Ülenurme parish, Tartumaa, 61713, Estonia.
| |
Collapse
|
339
|
Yamaji H, Konishi E. Production of Japanese Encephalitis Virus-Like Particles Using Insect Cell Expression Systems. Methods Mol Biol 2016; 1404:365-375. [PMID: 27076311 DOI: 10.1007/978-1-4939-3389-1_25] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Virus-like particles (VLPs) can be produced via the expression of virus surface proteins that self-assemble into particulate structures in recombinant protein expression systems. Expression of the DNA fragment encoding the Japanese encephalitis (JE) virus prM signal peptide, the precursor (prM) of the viral membrane protein (M), and the envelope glycoprotein (E) allows the production of a secretory form of VLPs. Expression systems that use lepidopteran insect cells, such as the baculovirus-insect cell system and stably transformed insect cells, can be used for the efficient production of JE VLPs. This chapter describes the production of JE VLPs from stably transformed lepidopteran insect cells.
Collapse
Affiliation(s)
- Hideki Yamaji
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657-8501, Japan.
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand.,BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
340
|
King B, Temperton NJ, Grehan K, Scott SD, Wright E, Tarr AW, Daly JM. Technical considerations for the generation of novel pseudotyped viruses. Future Virol 2016. [DOI: 10.2217/fvl.15.106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A pseudotyped virus (PV) is a virus particle with an envelope protein originating from a different virus. The ability to dictate which envelope proteins are expressed on the surface has made pseudotyping an important tool for basic virological studies such as determining the cellular targets of the envelope protein of the virus as well as identification of potential antiviral compounds and measuring specific antibody responses. In this review, we describe the common methodologies employed to generate PVs, with a focus on approaches to improve the efficacy of PV generation.
Collapse
Affiliation(s)
- Barnabas King
- School of Life Sciences & NIHR Biomedical Research Unit in Gastrointestinal & Liver Diseases, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
- NIHR Biomedical Research Unit in Gastrointestinal & Liver Diseases, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Nigel J Temperton
- Viral Pseudotype Unit (Medway), School of Pharmacy, Anson Building, Central Avenue, Chatham Maritime, Chatham, Kent, ME4 4TB, UK
| | - Keith Grehan
- Viral Pseudotype Unit (Medway), School of Pharmacy, Anson Building, Central Avenue, Chatham Maritime, Chatham, Kent, ME4 4TB, UK
| | - Simon D Scott
- Viral Pseudotype Unit (Medway), School of Pharmacy, Anson Building, Central Avenue, Chatham Maritime, Chatham, Kent, ME4 4TB, UK
| | - Edward Wright
- Viral Pseudotype Unit (Fitzrovia), Faculty of Science & Technology, University of Westminster, 115 New Cavendish Street, London, W1W 6UW, UK
| | - Alexander W Tarr
- School of Life Sciences & NIHR Biomedical Research Unit in Gastrointestinal & Liver Diseases, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
- NIHR Biomedical Research Unit in Gastrointestinal & Liver Diseases, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Janet M Daly
- School of Veterinary Medicine & Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| |
Collapse
|
341
|
Besnard L, Fabre V, Fettig M, Gousseinov E, Kawakami Y, Laroudie N, Scanlan C, Pattnaik P. Clarification of vaccines: An overview of filter based technology trends and best practices. Biotechnol Adv 2016; 34:1-13. [DOI: 10.1016/j.biotechadv.2015.11.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 11/28/2015] [Accepted: 11/29/2015] [Indexed: 12/18/2022]
|
342
|
Tretyakova I, Hidajat R, Hamilton G, Horn N, Nickols B, Prather RO, Tumpey TM, Pushko P. Preparation of quadri-subtype influenza virus-like particles using bovine immunodeficiency virus gag protein. Virology 2016; 487:163-71. [PMID: 26529299 PMCID: PMC4679414 DOI: 10.1016/j.virol.2015.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/02/2015] [Accepted: 10/03/2015] [Indexed: 01/13/2023]
Abstract
Influenza VLPs comprised of hemagglutinin (HA), neuraminidase (NA), and matrix (M1) proteins have been previously used for immunological and virological studies. Here we demonstrated that influenza VLPs can be made in Sf9 cells by using the bovine immunodeficiency virus gag (Bgag) protein in place of M1. We showed that Bgag can be used to prepare VLPs for several influenza subtypes including H1N1 and H10N8. Furthermore, by using Bgag, we prepared quadri-subtype VLPs, which co-expressed within the VLP the four HA subtypes derived from avian-origin H5N1, H7N9, H9N2 and H10N8 viruses. VLPs showed hemagglutination and neuraminidase activities and reacted with specific antisera. The content and co-localization of each HA subtype within the quadri-subtype VLP were evaluated. Electron microscopy showed that Bgag-based VLPs resembled influenza virions with the diameter of 150-200nm. This is the first report of quadri-subtype design for influenza VLP and the use of Bgag for influenza VLP preparation.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Cell Line
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunodeficiency Virus, Bovine/genetics
- Immunodeficiency Virus, Bovine/immunology
- Influenza A Virus, H10N8 Subtype/genetics
- Influenza A Virus, H10N8 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H9N2 Subtype/genetics
- Influenza A Virus, H9N2 Subtype/immunology
- Insecta
- Neuraminidase/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Sf9 Cells
- Spodoptera
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
| | - Rachmat Hidajat
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA
| | | | - Noah Horn
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA
| | - Brian Nickols
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA
| | | | - Terrence M Tumpey
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Road N.E., Atlanta, GA, USA
| | - Peter Pushko
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA.
| |
Collapse
|
343
|
Koch C, Wabbel K, Eber FJ, Krolla-Sidenstein P, Azucena C, Gliemann H, Eiben S, Geiger F, Wege C. Modified TMV Particles as Beneficial Scaffolds to Present Sensor Enzymes. FRONTIERS IN PLANT SCIENCE 2015; 6:1137. [PMID: 26734040 PMCID: PMC4689848 DOI: 10.3389/fpls.2015.01137] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/30/2015] [Indexed: 05/22/2023]
Abstract
Tobacco mosaic virus (TMV) is a robust nanotubular nucleoprotein scaffold increasingly employed for the high density presentation of functional molecules such as peptides, fluorescent dyes, and antibodies. We report on its use as advantageous carrier for sensor enzymes. A TMV mutant with a cysteine residue exposed on every coat protein (CP) subunit (TMVCys) enabled the coupling of bifunctional maleimide-polyethylene glycol (PEG)-biotin linkers (TMVCys/Bio). Its surface was equipped with two streptavidin [SA]-conjugated enzymes: glucose oxidase ([SA]-GOx) and horseradish peroxidase ([SA]-HRP). At least 50% of the CPs were decorated with a linker molecule, and all thereof with active enzymes. Upon use as adapter scaffolds in conventional "high-binding" microtiter plates, TMV sticks allowed the immobilization of up to 45-fold higher catalytic activities than control samples with the same input of enzymes. Moreover, they increased storage stability and reusability in relation to enzymes applied directly to microtiter plate wells. The functionalized TMV adsorbed to solid supports showed a homogeneous distribution of the conjugated enzymes and structural integrity of the nanorods upon transmission electron and atomic force microscopy. The high surface-increase and steric accessibility of the viral scaffolds in combination with the biochemical environment provided by the plant viral coat may explain the beneficial effects. TMV can, thus, serve as a favorable multivalent nanoscale platform for the ordered presentation of bioactive proteins.
Collapse
Affiliation(s)
- Claudia Koch
- Department of Molecular Biology and Plant Virology, Institute of Biomaterials and Biomolecular Systems, University of StuttgartStuttgart, Germany
| | - Katrin Wabbel
- Department of Molecular Biology and Plant Virology, Institute of Biomaterials and Biomolecular Systems, University of StuttgartStuttgart, Germany
| | - Fabian J. Eber
- Department of Molecular Biology and Plant Virology, Institute of Biomaterials and Biomolecular Systems, University of StuttgartStuttgart, Germany
| | - Peter Krolla-Sidenstein
- Chemistry of Oxydic and Organic Interfaces, Karlsruhe Institute of Technology, Institute of Functional InterfacesKarlsruhe, Germany
| | - Carlos Azucena
- Chemistry of Oxydic and Organic Interfaces, Karlsruhe Institute of Technology, Institute of Functional InterfacesKarlsruhe, Germany
| | - Hartmut Gliemann
- Chemistry of Oxydic and Organic Interfaces, Karlsruhe Institute of Technology, Institute of Functional InterfacesKarlsruhe, Germany
| | - Sabine Eiben
- Department of Molecular Biology and Plant Virology, Institute of Biomaterials and Biomolecular Systems, University of StuttgartStuttgart, Germany
| | - Fania Geiger
- Department of New Materials and Biosystems, Max-Planck-Institute for Intelligent SystemsStuttgart, Germany
| | - Christina Wege
- Department of Molecular Biology and Plant Virology, Institute of Biomaterials and Biomolecular Systems, University of StuttgartStuttgart, Germany
| |
Collapse
|
344
|
Optimized production of HIV-1 virus-like particles by transient transfection in CAP-T cells. Appl Microbiol Biotechnol 2015; 100:3935-47. [PMID: 26685677 DOI: 10.1007/s00253-015-7213-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/26/2015] [Accepted: 12/01/2015] [Indexed: 02/07/2023]
Abstract
HIV-1 virus-like particles (VLPs) have great potential as new-generation vaccines. The novel CAP-T cell line is used for the first time to produce Gag-GFP HIV-1 VLPs by means of polyethylenimine (PEI)-mediated transient transfection. CAP-T cells are adapted to grow to high cell densities in serum-free medium, and are able to express complex recombinant proteins with human post-translational modifications. Furthermore, this cell line is easily transfected with PEI, which offers the flexibility to rapidly generate and screen a number of candidates in preclinical studies. Transient transfection optimization of CAP-T cells has been performed systematically in this work. It is determined that for optimal production, cells need to be growing at mid-exponential phase, Protein Expression Medium (PEM) medium has to be added post-transfection, and cells can be transfected by independent addition of DNA and PEI with no prior complexation. A Box-Behnken experimental design is used to optimize cell density at time of transfection, DNA/cell and PEI/cell ratios. The optimal conditions determined are transfection at a density of 3.3E + 06 cells/mL with 0.5 pg of DNA/cell and 3 pg of PEI/cell. Using the optimized protocol, 6 × 10(10) VLP/mL are obtained, demonstrating that CAP-T is a highly efficient cell line for the production of HIV-1 VLPs and potentially other complex viral-based biotherapeutics.
Collapse
|
345
|
Ladd Effio C, Baumann P, Weigel C, Vormittag P, Middelberg A, Hubbuch J. High-throughput process development of an alternative platform for the production of virus-like particles in Escherichia coli. J Biotechnol 2015; 219:7-19. [PMID: 26707548 DOI: 10.1016/j.jbiotec.2015.12.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/17/2015] [Accepted: 12/14/2015] [Indexed: 11/26/2022]
Abstract
The production of safe vaccines against untreatable or new diseases has pushed the research in the field of virus-like particles (VLPs). Currently, a large number of commercial VLP-based human vaccines and vaccine candidates are available or under development. A promising VLP production route is the controlled in vitro assembly of virus proteins into capsids. In the study reported here, a high-throughput screening (HTS) procedure was implemented for the upstream process development of a VLP platform in bacterial cell systems. Miniaturized cultivations were carried out in 48-well format in the BioLector system (m2p-Labs, Germany) using an Escherichia coli strain with a tac promoter producing the murine polyomavirus capsid protein (VP1). The screening procedure incorporated micro-scale cultivations, HTS cell disruption by sonication and HTS-compatible analytics by capillary gel electrophoresis. Cultivation temperatures, shaking speeds, induction and medium conditions were varied to optimize the product expression in E. coli. The most efficient system was selected based on an evaluation of soluble and insoluble product concentrations as well as on the percentage of product in the total soluble protein fraction. The optimized system was scaled up to cultivation 2.5L shaker flask scale and purified using an anion exchange chromatography membrane adsorber, followed by a size exclusion chromatography polishing procedure. For proof of concept, purified VP1 capsomeres were assembled under defined buffer conditions into empty capsids and characterized using transmission electron microscopy (TEM). The presented HTS procedure allowed for a fast development of an efficient production process of VLPs in E. coli. Under optimized cultivation conditions, the VP1 product totalled up to 43% of the total soluble protein fraction, yielding 1.63 mg VP1 per mL of applied cultivation medium. The developed production process strongly promotes the murine polyoma-VLP platform, moving towards an industrially feasible technology for new chimeric vaccines.
Collapse
Affiliation(s)
- Christopher Ladd Effio
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| | - Pascal Baumann
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| | - Claudia Weigel
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Philipp Vormittag
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Anton Middelberg
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Australia
| | - Jürgen Hubbuch
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| |
Collapse
|
346
|
Takeyama N, Kiyono H, Yuki Y. Plant-based vaccines for animals and humans: recent advances in technology and clinical trials. THERAPEUTIC ADVANCES IN VACCINES 2015; 3:139-54. [PMID: 26668752 DOI: 10.1177/2051013615613272] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It has been about 30 years since the first plant engineering technology was established. Although the concept of plant-based pharmaceuticals or vaccines motivates us to develop practicable commercial products using plant engineering, there are some difficulties in reaching the final goal: to manufacture an approved product. At present, the only plant-made vaccine approved by the United States Department of Agriculture is a Newcastle disease vaccine for poultry that is produced in suspension-cultured tobacco cells. The progress toward commercialization of plant-based vaccines takes much effort and time, but several candidate vaccines for use in humans and animals are in clinical trials. This review discusses plant engineering technologies and regulations relevant to the development of plant-based vaccines and provides an overview of human and animal vaccines currently under clinical trials.
Collapse
Affiliation(s)
- Natsumi Takeyama
- Division of Mucosal Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshikazu Yuki
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
347
|
Bolhassani A, Shirbaghaee Z, Agi E, Davoudi N. VLP production in Leishmania tarentolae : A novel expression system for purification and assembly of HPV16 L1. Protein Expr Purif 2015; 116:7-11. [DOI: 10.1016/j.pep.2015.08.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/16/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022]
|
348
|
Correia-Pinto J, Peleteiro M, Csaba N, González-Fernández Á, Alonso M. Multi-enveloping of particulated antigens with biopolymers and immunostimulant polynucleotides. J Drug Deliv Sci Technol 2015. [DOI: 10.1016/j.jddst.2015.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
349
|
Kumar M, Saravanan P, Jalali SK. Expression and purification of virus like particles (VLPs) of foot-and-mouth disease virus in Eri silkworm (Samia cynthia ricini) larvae. Virusdisease 2015; 27:84-90. [PMID: 26925448 DOI: 10.1007/s13337-015-0290-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/16/2015] [Indexed: 11/29/2022] Open
Abstract
Foot-and-mouth disease (FMD) is a highly contagious viral disease, which causes severe economic loss to livestock. Virus like particles (VLPs) produced by recombinant DNA technology are gaining importance because of their immunogenic properties and safety in developing a new vaccine for FMD. In the present study, a practical and economically feasible approach of expression, purification and characterization of VLPs of FMDV in Eri silkworm (Samia cynthia ricini) larvae was described. Although three lepidopteran insect larvae (Helicoverpa armigera, Spodoptera litura and Samia cynthia ricini) were tested for production of VLPs, expression was obtained only in Eri silkworm larvae. High titred recombinant baculovirus encoding the polyprotein P1-2A-3C of FMDV was prepared in Sf9 cells. Injection of recombinant baculovirus into hemocoel of Eri silkworm larvae resulted in increasing levels of expression of VLPs in the hemolymph from 3 to 7 days post infection (dpi) compared to low level expression by oral feeding. The VLPs reacted in Sandwich ELISA with serum raised against whole virus particles of FMDV type O/IND/R2/75 and protein banding pattern of 26, 37 and 47 kDa in Western blotting demonstrated their antigenic resemblance to native virus. Sucrose density gradient purified VLPs were used for immunization of rabbits and guinea pigs for assessing immunogenicity. Further, the reactivity of serum samples of rabbits and guinea pigs in Indirect-ELISA with titres (1.30-2.81 Log10) indicated that the VLPs were antigenic and immunogenic in nature. We demonstrate that Eri silkworm larvae could be used for production of VLPs of FMDV type O/IND/R2/75 for the first time. This approach could be useful for large scale production of recombinant VLPs for vaccine or diagnostic use in FMD control programme.
Collapse
Affiliation(s)
- Manoj Kumar
- Foot-and-Mouth Disease Vaccine Centre, Indian Veterinary Research Institute, Bangalore Campus, Hebbal, Bangalore, Karnataka 560024 India
| | - P Saravanan
- Foot-and-Mouth Disease Vaccine Centre, Indian Veterinary Research Institute, Bangalore Campus, Hebbal, Bangalore, Karnataka 560024 India
| | - S K Jalali
- National Bureau of Agricultural Insect Resources, H A Farm Post, Bellary Road, Hebbal, Bangalore, Karnataka 560024 India
| |
Collapse
|
350
|
Thrane S, Janitzek CM, Agerbæk MØ, Ditlev SB, Resende M, Nielsen MA, Theander TG, Salanti A, Sander AF. A Novel Virus-Like Particle Based Vaccine Platform Displaying the Placental Malaria Antigen VAR2CSA. PLoS One 2015; 10:e0143071. [PMID: 26599509 PMCID: PMC4657905 DOI: 10.1371/journal.pone.0143071] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/30/2015] [Indexed: 11/24/2022] Open
Abstract
Placental malaria caused by Plasmodium falciparum is a major cause of mortality and severe morbidity. Clinical testing of a soluble protein-based vaccine containing the parasite ligand, VAR2CSA, has been initiated. VAR2CSA binds to the human receptor chondroitin sulphate A (CSA) and is responsible for sequestration of Plasmodium falciparum infected erythrocytes in the placenta. It is imperative that a vaccine against malaria in pregnancy, if administered to women before they become pregnant, can induce a strong and long lasting immune response. While most soluble protein-based vaccines have failed during clinical testing, virus-like particle (VLP) based vaccines (e.g., the licensed human papillomavirus vaccines) have demonstrated high efficacy, suggesting that the spatial assembly of the vaccine antigen is a critical parameter for inducing an optimal long-lasting protective immune response. We have developed a VLP vaccine display platform by identifying regions of the HPV16 L1 coat protein where a biotin acceptor site (AviTagTM) can be inserted without compromising VLP-assembly. Subsequent biotinylation of Avi-L1 VLPs allow us to anchor monovalent streptavidin (mSA)-fused proteins to the biotin, thereby obtaining a dense and repetitive VLP-display of the vaccine antigen. The mSA-VAR2CSA antigen was delivered on the Avi-L1 VLP platform and tested in C57BL/6 mice in comparison to two soluble protein-based vaccines consisting of naked VAR2CSA and mSA-VAR2CSA. The mSA-VAR2CSA Avi-L1 VLP and soluble mSA-VAR2CSA vaccines induced higher antibody titers than the soluble naked VAR2CSA vaccine after three immunizations. The VAR2CSA Avi-L1 VLP vaccine induced statistically significantly higher endpoint titres compared to the soluble mSA-VAR2CSA vaccine, after 1st and 2nd immunization; however, this difference was not statistically significant after 3rd immunization. Importantly, the VLP-VAR2CSA induced antibodies were functional in inhibiting the binding of parasites to CSA. This study demonstrates that the described Avi-L1 VLP-platform may serve as a versatile system for facilitating optimal VLP-display of large and complex vaccine antigens.
Collapse
Affiliation(s)
- Susan Thrane
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christoph M. Janitzek
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mette Ø. Agerbæk
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sisse B. Ditlev
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mafalda Resende
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Morten A. Nielsen
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thor G. Theander
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Adam F. Sander
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|