351
|
Nestola P, Peixoto C, Villain L, Alves PM, Carrondo MJT, Mota JPB. Rational development of two flowthrough purification strategies for adenovirus type 5 and retro virus-like particles. J Chromatogr A 2015; 1426:91-101. [PMID: 26643723 DOI: 10.1016/j.chroma.2015.11.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 11/10/2015] [Accepted: 11/10/2015] [Indexed: 12/22/2022]
Abstract
We report on the rational design and implementation of flowthrough (FT) platforms for purification of virus vectors (VVs) and virus-like particles (VLPs), combining anion-exchange polyallylamine membranes (Sartobind STIC) and core-shell octylamine resins (CaptoCore 700). In one configuration, the VV bulk is concentrated and conditioned with appropriate buffer in a ultra/diafiltration (UF/DF) unit prior to injection into the STIC chromatography membrane. The FT pool and an intermediate cut of the elution pool of the STIC membrane are admixed and directed to a second UF/DF. Finally, the retentate is injected into a CC700 packed bed adsorber where the purified VVs are collected in the FT pool, whereas the residual amount of DNA and host cell protein (HCP) are discarded in the eluate. The experimental recovery achieved with this downstream processing (DSP) platform is close to 100%, the DNA clearance is roughly a 4-log reduction, and the HCP level is reduced by 5 logs. The platform developed for VLP purification is simpler than the previous one, as the STIC membrane adsorber and CC700 bed are connected in series with no UF/DF unit in between. Experimentally, the FT scheme for VLP purification gave a recovery yield of 45% in the chromatography train; the experimental log reduction of DNA and HCP were 2.0 and 3.5, respectively. These results are in line with other purification strategies in the specific field of enveloped VLPs. Both DSP platforms were successfully developed from an initial design space of the binding of the major contaminant (DNA) to the two ligands, determined by surface plasmon resonance, which was subsequently scaled up and confirmed experimentally.
Collapse
Affiliation(s)
- Piergiuseppe Nestola
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade NOVA de Lisboa, 2780-157 Oeiras, Portugal
| | - Cristina Peixoto
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade NOVA de Lisboa, 2780-157 Oeiras, Portugal
| | - Louis Villain
- Sartorius Stedim Biotech GmbH, Spindler-Strasse11, 37079 Gottingen, Germany
| | - Paula M Alves
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade NOVA de Lisboa, 2780-157 Oeiras, Portugal
| | - Manuel J T Carrondo
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - José P B Mota
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.
| |
Collapse
|
352
|
Abstract
Nanoscale engineering is revolutionizing the way we prevent, detect, and treat diseases. Viruses have played a special role in these developments because they can function as prefabricated nanoscaffolds that have unique properties and are easily modified. The interiors of virus particles can encapsulate and protect sensitive compounds, while the exteriors can be altered to display large and small molecules in precisely defined arrays. These properties of viruses, along with their innate biocompatibility, have led to their development as actively targeted drug delivery systems that expand on and improve current pharmaceutical options. Viruses are naturally immunogenic, and antigens displayed on their surface have been used to create vaccines against pathogens and to break self-tolerance to initiate an immune response to dysfunctional proteins. Densely and specifically aligned imaging agents on viruses have allowed for high-resolution and noninvasive visualization tools to detect and treat diseases earlier than previously possible. These and future applications of viruses have created an exciting new field within the disciplines of both nanotechnology and medicine.
Collapse
Affiliation(s)
| | | | - Marianne Manchester
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093
| | - Nicole F Steinmetz
- Departments of 2Biomedical Engineering
- Radiology
- Materials Science and Engineering, and
- Macromolecular Science and Engineering, Case Western Reserve University, Schools of Medicine and Engineering, Cleveland, Ohio 44106;
| |
Collapse
|
353
|
Jaberolansar N, Toth I, Young PR, Skwarczynski M. Recent advances in the development of subunit-based RSV vaccines. Expert Rev Vaccines 2015; 15:53-68. [PMID: 26506139 DOI: 10.1586/14760584.2016.1105134] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections causing pneumonia and bronchiolitis in infants. RSV also causes serious illness in elderly populations, immunocompromised patients and individuals with pulmonary or cardiac problems. The significant morbidity and mortality associated with RSV infection have prompted interest in RSV vaccine development. In the 1960s, a formalin-inactivated vaccine trial failed to protect children, and indeed enhanced pathology when naturally infected later with RSV. Hence, an alternative approach to traditional killed virus vaccines, which can induce protective immunity without serious adverse events, is desired. Several strategies have been explored in attempts to produce effective vaccine candidates including gene-based and subunit vaccines. Subunit-based vaccine approaches have shown promising efficacy in animal studies and several have reached clinical trials. The current stage of development of subunit-based vaccines against RSV is reviewed in this article.
Collapse
Affiliation(s)
- Noushin Jaberolansar
- a School of Chemistry and Molecular Biosciences , The University of Queensland , St Lucia , Queensland , Australia
| | - Istvan Toth
- a School of Chemistry and Molecular Biosciences , The University of Queensland , St Lucia , Queensland , Australia.,b Institute for Molecular Bioscience , The University of Queensland , St Lucia , Queensland , Australia.,c School of Pharmacy , The University of Queensland , Woolloongabba , Queensland , Australia
| | - Paul R Young
- a School of Chemistry and Molecular Biosciences , The University of Queensland , St Lucia , Queensland , Australia.,b Institute for Molecular Bioscience , The University of Queensland , St Lucia , Queensland , Australia.,d Australian Infectious Diseases Research Centre , The University of Queensland , St Lucia , Queensland , Australia
| | - Mariusz Skwarczynski
- a School of Chemistry and Molecular Biosciences , The University of Queensland , St Lucia , Queensland , Australia
| |
Collapse
|
354
|
Lee HJ, Kim JY, Kye SJ, Seul HJ, Jung SC, Choi KS. Efficient self-assembly and protective efficacy of infectious bursal disease virus-like particles by a recombinant baculovirus co-expressing precursor polyprotein and VP4. Virol J 2015; 12:177. [PMID: 26502988 PMCID: PMC4621879 DOI: 10.1186/s12985-015-0403-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/14/2015] [Indexed: 12/17/2022] Open
Abstract
Background Virus-like particle (VLP) technology is considered one of the most promising approaches in animal vaccines, due to the intrinsic immunogenic properties as well as high safety profile of VLPs. In this study, we developed a VLP vaccine against infectious bursal disease virus (IBDV), which causes morbidity and mortality in chickens, by expressing a baculovirus in insect cells. Methods To improve the self-proteolytic processing of precursor polyprotein (PP), we constructed a recombinant baculovirus transfer vector that co-expresses PP and the VP4 protease gene of IBDV. Results Expression and VLP assembly of recombinant proteins and antigenicity of the VLP were examined by Western blotting, ELISA, and transmission electron microscopy. In animal experiments, vaccination with the recombinant VLP induced strong and uniform humoral immunity and provided complete protection against challenge with very virulent (vv) IBDV in SPF chickens (n = 12). As determined by the bursa of Fabricius (BF)/body weight (B/BW) ratio, the protection against post-challenge bursal atrophy was significantly higher (P < 0.001) in VLP-vaccinated birds than in non-vaccinated controls. Conclusions Since the protective efficacy of the VLP vaccine was comparable to that of a commercially available inactivated vaccine, the recombinant VLP merits further investigation as an alternative means of protection against vvIBD.
Collapse
Affiliation(s)
- Hyun-Jeong Lee
- Avian Disease Division, Animal and Plant Quarantine Agency, 175 Anyangro, Anyang, Gyeonggi, 430-757, Republic of Korea.
| | - Ji-Ye Kim
- Avian Disease Division, Animal and Plant Quarantine Agency, 175 Anyangro, Anyang, Gyeonggi, 430-757, Republic of Korea.
| | - Soo-Jeong Kye
- Avian Disease Division, Animal and Plant Quarantine Agency, 175 Anyangro, Anyang, Gyeonggi, 430-757, Republic of Korea.
| | - Hee-Jung Seul
- Avian Disease Division, Animal and Plant Quarantine Agency, 175 Anyangro, Anyang, Gyeonggi, 430-757, Republic of Korea.
| | - Suk-Chan Jung
- Avian Disease Division, Animal and Plant Quarantine Agency, 175 Anyangro, Anyang, Gyeonggi, 430-757, Republic of Korea.
| | - Kang-Seuk Choi
- Avian Disease Division, Animal and Plant Quarantine Agency, 175 Anyangro, Anyang, Gyeonggi, 430-757, Republic of Korea.
| |
Collapse
|
355
|
Comparative efficacy of virus like particle (VLP) vaccine of foot-and-mouth-disease virus (FMDV) type O adjuvanted with poly I:C or CpG in guinea pigs. Biologicals 2015; 43:437-43. [PMID: 26493006 DOI: 10.1016/j.biologicals.2015.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 04/15/2015] [Accepted: 09/23/2015] [Indexed: 11/23/2022] Open
Abstract
Foot-and-mouth disease (FMD) is one of the most contagious and economically important diseases of cloven-hoofed livestock. Currently used inactivated FMD vaccines have short lived immunity besides risk of handling live virus. We studied recombinant FMD virus like particles (VLPs) encoded by FMDV type O/IND/R2/75 polyprotein genes expressed in Sf9 cells and adjuvanted with CpG or Poly I:C in inducing protective immune response in guinea pigs. Guinea pigs immunized with VLP + CpG vaccine had shown markedly higher cell mediated immunity (CMI) in comparison to the conventional vaccine group as evident from higher levels of IgG2 than IgG1. Although the humoral response was less in VLP + CpG compared to conventional vaccine, the lymphocyte stimulation index was more in VLP + CpG compared to conventional and VLP + Poly I:C vaccine groups. Finally the challenge experiments on 28 and 56 dpv had shown 75% protection in VLP + CpG immunized guinea pigs primary and boosted animals, while 50% and 62% protection in VLP + Poly I:C in primary and boosted animals, respectively. In conclusion, CpG adjuvant was found to be superior followed by ISA206 and Poly I:C in eliciting protection in VLP based FMD vaccines in guinea pigs.
Collapse
|
356
|
Xu PW, Wu X, Wang HN, Ma BC, Ding MD, Yang X. Assembly and immunogenicity of baculovirus-derived infectious bronchitis virus-like particles carrying membrane, envelope and the recombinant spike proteins. Biotechnol Lett 2015; 38:299-304. [PMID: 26463372 PMCID: PMC7087761 DOI: 10.1007/s10529-015-1973-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 09/28/2015] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To assemble infectious bronchitis virus (IBV)-like particles bearing the recombinant spike protein and investigate the humoral immune responses in chickens. RESULTS IBV virus-like particles (VLPs) were generated through the co-infection with three recombinant baculoviruses separately encoding M, E or the recombinant S genes. The recombinant S protein was sufficiently flexible to retain the ability to self-assemble into VLPs. The size and morphology of the VLPs were similar to authentic IBV particles. In addition, the immunogenicity of IBV VLPs had been investigated. The results demonstrated that the efficiency of the newly generated VLPs was comparable to that of the inactivated M41 viruses in eliciting IBV-specific antibodies and neutralizing antibodies in chickens via subcutaneous inoculation. CONCLUSIONS This work provides basic information for the mechanism of IBV VLP formation and develops a platform for further designing IBV VLP-based vaccines against IBV or other viruses.
Collapse
Affiliation(s)
- Peng-wei Xu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, School of Life Science, Sichuan University, Chengdu, Sichuan Province, 610064, China.
| | - Xuan Wu
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, School of Life Science, Sichuan University, Chengdu, Sichuan Province, 610064, China.
| | - Hong-ning Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, School of Life Science, Sichuan University, Chengdu, Sichuan Province, 610064, China.
| | - Bing-cun Ma
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, School of Life Science, Sichuan University, Chengdu, Sichuan Province, 610064, China.
| | - Meng-die Ding
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, School of Life Science, Sichuan University, Chengdu, Sichuan Province, 610064, China.
| | - Xin Yang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, School of Life Science, Sichuan University, Chengdu, Sichuan Province, 610064, China.
| |
Collapse
|
357
|
The application of virus-like particles as vaccines and biological vehicles. Appl Microbiol Biotechnol 2015; 99:10415-32. [PMID: 26454868 PMCID: PMC7080154 DOI: 10.1007/s00253-015-7000-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/01/2015] [Accepted: 09/04/2015] [Indexed: 01/04/2023]
Abstract
Virus-like particles (VLPs) can be spontaneously self-assembled by viral structural proteins under appropriate conditions in vitro while excluding the genetic material and potential replication probability. In addition, VLPs possess several features including can be rapidly produced in large quantities through existing expression systems, highly resembling native viruses in terms of conformation and appearance, and displaying repeated cluster of epitopes. Their capsids can be modified via genetic insertion or chemical conjugation which facilitating the multivalent display of a homologous or heterogeneous epitope antigen. Therefore, VLPs are considered as a safe and effective candidate of prophylactic and therapeutic vaccines. VLPs, with a diameter of approximately 20 to 150 nm, also have the characteristics of nanometer materials, such as large surface area, surface-accessible amino acids with reactive moieties (e.g., lysine and glutamic acid residues), inerratic spatial structure, and good biocompatibility. Therefore, assembled VLPs have great potential as a delivery system for specifically carrying a variety of materials. This review summarized recent researches on VLP development as vaccines and biological vehicles, which demonstrated the advantages and potential of VLPs in disease control and prevention and diagnosis. Then, the prospect of VLP biology application in the future is discussed as well.
Collapse
|
358
|
Zhao B, Pan X, Teng Y, Xia W, Wang J, Wen Y, Chen Y. Rotavirus VP7 epitope chimeric proteins elicit cross-immunoreactivity in guinea pigs. Virol Sin 2015; 30:363-70. [PMID: 26459269 PMCID: PMC8200902 DOI: 10.1007/s12250-015-3620-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/28/2015] [Indexed: 10/23/2022] Open
Abstract
VP7 of group A rotavirus (RVA) contains major neutralizing epitopes. Using the antigenic protein VP6 as the vector, chimeric proteins carrying foreign epitopes have been shown to possess good immunoreactivity and immunogenicity. In the present study, using modified VP6 as the vector, three chimeric proteins carrying epitopes derived from VP7 of RVA were constructed. The results showed that the chimeric proteins reacted with anti-VP6 and with SA11 and Wa virus strains. Antibodies from guinea pigs inoculated with the chimeric proteins recognized VP6 and VP7 of RVA and protected mammalian cells from SA11 and Wa infection in vitro. The neutralizing activities of the antibodies against the chimeric proteins were significantly higher than those against the vector protein VP6F. Thus, development of chimeric vaccines carrying VP7 epitopes using VP6 as a vector could be a promising alternative to enhance immunization against RVAs.
Collapse
Affiliation(s)
- Bingxin Zhao
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Xiaoxia Pan
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming, 650500, China
| | - Yumei Teng
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Wenyue Xia
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Jing Wang
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yuling Wen
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yuanding Chen
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.
| |
Collapse
|
359
|
Jain NK, Sahni N, Kumru OS, Joshi SB, Volkin DB, Russell Middaugh C. Formulation and stabilization of recombinant protein based virus-like particle vaccines. Adv Drug Deliv Rev 2015; 93:42-55. [PMID: 25451136 DOI: 10.1016/j.addr.2014.10.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 10/15/2014] [Accepted: 10/18/2014] [Indexed: 02/06/2023]
Abstract
Vaccine formulation development has traditionally focused on improving antigen storage stability and compatibility with conventional adjuvants. More recently, it has also provided an opportunity to modify the interaction and presentation of an antigen/adjuvant to the immune system to better stimulate the desired immune responses for maximal efficacy. In the last decade, there has been a paradigm shift in vaccine antigen and formulation design involving an improved physical understanding of antigens and a better understanding of the immune system. In addition, the discovery of novel adjuvants and delivery systems promises to further improve the design of new, more effective vaccines. Here we describe some of the fundamental aspects of formulation design applicable to virus-like-particle based vaccine antigens (VLPs). Case studies are presented for commercially approved VLP vaccines as well as some investigational VLP vaccine candidates. An emphasis is placed on the biophysical analysis of vaccines to facilitate formulation and stabilization of these particulate antigens.
Collapse
|
360
|
Uribe-Campero L, Monroy-García A, Durán-Meza AL, Villagrana-Escareño MV, Ruíz-García J, Hernández J, Núñez-Palenius HG, Gómez-Lim MA. Plant-based porcine reproductive and respiratory syndrome virus VLPs induce an immune response in mice. Res Vet Sci 2015; 102:59-66. [PMID: 26412521 DOI: 10.1016/j.rvsc.2015.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 07/10/2015] [Accepted: 07/19/2015] [Indexed: 01/14/2023]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) significantly affects the swine industry worldwide. An efficient, protective vaccine is still lacking. Here, we report for the first time the generation and purification of PRRSV virus like particles (VLPs) by expressing GP5, M and N genes in Nicotiana silvestris plants. The particles were clearly visible by transmission electron microscopy (TEM) with a size of 60-70 nm. Hydrodynamic diameter of the particles was obtained and it was confirmed that the VLPs had the appropriate size for PRRS virions and that the VLPs were highly pure. By measuring the Z potential we described the electrophoretic mobility behavior of VLPs and the best conditions for stability of the VLPs were determined. The particles were immunogenic in mice. A western blot of purified particles allowed detection of three coexpressed genes. These VLPs may serve as a platform to develop efficient PRRSV vaccines.
Collapse
Affiliation(s)
- Laura Uribe-Campero
- Departamento de Ingeniería Genética, CINVESTAV-IPN, Km 9.6 Carretera Irapuato-León, C.P. 36821 Irapuato, Guanajuato, México.
| | - Alberto Monroy-García
- Unidad de Investigación Médica en Enfermedades Oncológicas, IMSS, CMN SXXI, México, D.F., México; Laboratorio de Inmunobiología, Lab, 3PB, Unidad de Investigación en Diferenciación Celular y Cáncer, Facultad de Estudios Superiores Zaragoza, UMIEZ, Campus II, UNAM, Batalla 5 de mayo s/n, Col. E. Oriente, Esquina Fuerte Loreto, Iztapalapa, CP 09230 México, D.F., México.
| | - Ana L Durán-Meza
- Laboratorio de Física Biológica, Instituto de Física, Universidad Autónoma de San Luis Potosí, Álvaro Obregón 64, San Luis Potosí, SLP 78000, México.
| | - María V Villagrana-Escareño
- Laboratorio de Física Biológica, Instituto de Física, Universidad Autónoma de San Luis Potosí, Álvaro Obregón 64, San Luis Potosí, SLP 78000, México.
| | - Jaime Ruíz-García
- Laboratorio de Física Biológica, Instituto de Física, Universidad Autónoma de San Luis Potosí, Álvaro Obregón 64, San Luis Potosí, SLP 78000, México.
| | - Jesús Hernández
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo A.C., Carretera a La Victoria km 0.6, Hermosillo, Sonora C.P. 83304, México.
| | - Héctor G Núñez-Palenius
- División de Ciencias de la Vida, Campus Irapuato-Salamanca, Universidad de Guanajuato, Exhacienda El Copal s/n, A.P. 311, Irapuato, Gto. C.P. 36500, México.
| | - Miguel A Gómez-Lim
- Departamento de Ingeniería Genética, CINVESTAV-IPN, Km 9.6 Carretera Irapuato-León, C.P. 36821 Irapuato, Guanajuato, México.
| |
Collapse
|
361
|
Ruiz V, Mozgovoj MV, Dus Santos MJ, Wigdorovitz A. Plant-produced viral bovine vaccines: what happened during the last 10 years? PLANT BIOTECHNOLOGY JOURNAL 2015; 13:1071-1077. [PMID: 26250843 DOI: 10.1111/pbi.12440] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/05/2015] [Accepted: 06/27/2015] [Indexed: 06/04/2023]
Abstract
Vaccination has proved to be an efficient strategy to deal with viral infections in both human and animal species. However, protection of cattle against viral infections is still a major concern in veterinary science. During the last two decades, the development of efficient plant-based expression strategies for recombinant proteins prompted the application of this methodology for veterinary vaccine purposes. The main goals of viral bovine vaccines are to improve the health and welfare of cattle and increase the production of livestock, in a cost-effective manner. This review explores some of the more prominent recent advances in plant-made viral bovine vaccines against foot-and-mouth disease virus (FMDV), bovine rotavirus (BRV), bovine viral diarrhoea virus (BVDV), bluetongue virus (BTV) and bovine papillomavirus (BPV), some of which are considered to be the most important viral causative agents of economic loss in cattle production.
Collapse
Affiliation(s)
- Vanesa Ruiz
- Instituto de Virología, CICVyA, INTA, Hurlingham, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Marina V Mozgovoj
- Instituto de Virología, CICVyA, INTA, Hurlingham, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - María José Dus Santos
- Instituto de Virología, CICVyA, INTA, Hurlingham, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Andrés Wigdorovitz
- Instituto de Virología, CICVyA, INTA, Hurlingham, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
362
|
Encapsulation as a Strategy for the Design of Biological Compartmentalization. J Mol Biol 2015; 428:916-27. [PMID: 26403362 DOI: 10.1016/j.jmb.2015.09.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/16/2015] [Accepted: 09/03/2015] [Indexed: 02/06/2023]
Abstract
Compartmentalization is one of the defining features of life. Through intracellular spatial control, cells are able to organize and regulate their metabolism. One of the most broadly used organizational principles in nature is encapsulation. Cellular processes can be encapsulated within either membrane-bound organelles or proteinaceous compartments that create distinct microenvironments optimized for a given task. Further challenges addressed through intracellular compartmentalization are toxic or volatile pathway intermediates, slow turnover rates and competing side reactions. This review highlights a selection of naturally occurring membrane- and protein-based encapsulation systems in microbes and their recent applications and emerging opportunities in synthetic biology. We focus on examples that use engineered cellular organization to control metabolic pathway flux for the production of useful compounds and materials.
Collapse
|
363
|
Young KR, Arthus-Cartier G, Yam KK, Lavoie PO, Landry N, D'Aoust MA, Vézina LP, Couture MMJ, Ward BJ. Generation and characterization of a trackable plant-made influenza H5 virus-like particle (VLP) containing enhanced green fluorescent protein (eGFP). FASEB J 2015; 29:3817-27. [PMID: 26038124 DOI: 10.1096/fj.15-270421] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/18/2015] [Indexed: 12/17/2022]
Abstract
Medicago, Inc. has developed an efficient virus-like particle (VLP) vaccine production platform using the Nicotiana benthamiana expression system, and currently has influenza-based products targeting seasonal/pandemic hemagglutinin (HA) proteins in advanced clinical trials. We wished to generate a trackable HA-based VLP that would allow us to study both particle assembly in plants and VLP interactions within the mammalian immune system. To this end, a fusion protein was designed, composed of H5 (from influenza A/Indonesia/05/2005 [H5N1]) with enhanced green fluorescent protein (eGFP). Expression of H5-eGFP in N. benthamiana produced brightly fluorescent ∼160 nm particles resembling H5-VLPs. H5-eGFP-VLPs elicited anti-H5 serologic responses in mice comparable to those elicited by H5-VLPs in almost all assays tested (hemagglutination inhibition/IgG(total)/IgG1/IgG2b/IgG2a:IgG1 ratio), as well as a superior anti-GFP IgG response (mean optical density = 2.52 ± 0.16 sem) to that elicited by soluble GFP (mean optical density = 0.12 ± 0.06 sem). Confocal imaging of N. benthamiana cells expressing H5-eGFP displayed large fluorescent accumulations at the cell periphery, and draining lymph nodes from mice given H5-eGFP-VLPs via footpad injection demonstrated bright fluorescence shortly after administration (10 min), providing proof of concept that the H5-eGFP-protein/VLPs could be used to monitor both VLP assembly and immune trafficking. Given these findings, this novel fluorescent reagent will be a powerful tool to gain further fundamental insight into the biology of influenza VLP vaccines.
Collapse
Affiliation(s)
- Katie R Young
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| | - Guillaume Arthus-Cartier
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| | - Karen K Yam
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| | - Pierre-Olivier Lavoie
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| | - Nathalie Landry
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| | - Marc-André D'Aoust
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| | - Louis-Philippe Vézina
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| | - Manon M-J Couture
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| | - Brian J Ward
- *Research Institute of McGill University Health Centre and Department of Experimental Medicine, McGill University, Montréal, Québec, Canada; and Medicago, Incorporated, Québec, Québec, Canada
| |
Collapse
|
364
|
Hassani-Mehraban A, Creutzburg S, van Heereveld L, Kormelink R. Feasibility of Cowpea chlorotic mottle virus-like particles as scaffold for epitope presentations. BMC Biotechnol 2015; 15:80. [PMID: 26311254 PMCID: PMC4551372 DOI: 10.1186/s12896-015-0180-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 06/29/2015] [Indexed: 11/23/2022] Open
Abstract
Background & Methods Within the last decade Virus-Like Particles (VLPs) have increasingly received attention from scientists for their use as a carrier of (peptide) molecules or as scaffold to present epitopes for use in subunit vaccines. To test the feasibility of Cowpea chlorotic mottle virus (CCMV) particles as a scaffold for epitope presentation and identify sites for epitope fusion or insertion that would not interfere with virus-like-particle formation, chimeric CCMV coat protein (CP) gene constructs were engineered, followed by expression in E. coli and assessment of VLP formation. Various constructs were made encoding a 6x-His-tag, or selected epitopes from Influenza A virus [IAV] (M2e, HA) or Foot and Mouth Disease Virus [FMDV] (VP1 and 2C). The epitopes were either inserted 1) in predicted exposed loop structures of the CCMV CP protein, 2) fused to the amino- (N) or carboxyl-terminal (C) ends, or 3) to a N-terminal 24 amino acid (aa) deletion mutant (N∆24-CP) of the CP protein. Results High levels of insoluble protein expression, relative to proteins from the entire cell lysate, were obtained for CCMV CP and all chimeric derivatives. A straightforward protocol was used that, without the use of purification columns, successfully enabled CCMV CP protein solubilization, reassembly and subsequent collection of CCMV CP VLPs. While insertions of His-tag or M2e (7-23 aa) into the predicted external loop structures did abolish VLP formation, high yields of VLPs were obtained with all fusions of His-tag or various epitopes (13- 27 aa) from IAV and FMDV at the N- or C-terminal ends of CCMV CP or N∆24-CP. VLPs derived from CCMV CP still encapsulated RNA, while those from CCMV CP-chimera containing a negatively charged N-terminal domain had lost this ability. The usefulness and rapid ease of exploitation of CCMV VLPs for the production of potential subunit vaccines was demonstrated with the synthesis of chimeric CCMV VLPs containing selected sequences from the GN and GC glycoproteins of the recently emerged Schmallenberg orthobunyavirus at both termini of the CP protein. Conclusions CCMV VLPs can be successfully exploited as scaffold for epitope fusions up to 31 aa at the N- and C-terminus, and at a N-terminal 24 amino acid (aa) deletion mutant (N∆24-CP) of the CP protein.
Collapse
Affiliation(s)
- Afshin Hassani-Mehraban
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Sjoerd Creutzburg
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Luc van Heereveld
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Richard Kormelink
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| |
Collapse
|
365
|
Trovato M, Berardinis PD. Novel antigen delivery systems. World J Virol 2015; 4:156-168. [PMID: 26279977 PMCID: PMC4534807 DOI: 10.5501/wjv.v4.i3.156] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 06/23/2015] [Accepted: 08/03/2015] [Indexed: 02/05/2023] Open
Abstract
Vaccines represent the most relevant contribution of immunology to human health. However, despite the remarkable success achieved in the past years, many vaccines are still missing in order to fight important human pathologies and to prevent emerging and re-emerging diseases. For these pathogens the known strategies for making vaccines have been unsuccessful and thus, new avenues should be investigated to overcome the failure of clinical trials and other important issues including safety concerns related to live vaccines or viral vectors, the weak immunogenicity of subunit vaccines and side effects associated with the use of adjuvants. A major hurdle of developing successful and effective vaccines is to design antigen delivery systems in such a way that optimizes antigen presentation and induces broad protective immune responses. Recent advances in vector delivery technologies, immunology, vaccinology and system biology, have led to a deeper understanding of the molecular and cellular mechanisms by which vaccines should stimulate both arms of the adaptive immune responses, offering new strategies of vaccinations. This review is an update of current strategies with respect to live attenuated and inactivated vaccines, DNA vaccines, viral vectors, lipid-based carrier systems such as liposomes and virosomes as well as polymeric nanoparticle vaccines and virus-like particles. In addition, this article will describe our work on a versatile and immunogenic delivery system which we have studied in the past decade and which is derived from a non-pathogenic prokaryotic organism: the “E2 scaffold” of the pyruvate dehydrogenase complex from Geobacillus stearothermophilus.
Collapse
|
366
|
Tekewe A, Connors NK, Sainsbury F, Wibowo N, Lua LH, Middelberg AP. A rapid and simple screening method to identify conditions for enhanced stability of modular vaccine candidates. Biochem Eng J 2015. [DOI: 10.1016/j.bej.2015.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
367
|
Bitrus Y, Andrew JN, Owolodun OA, Luka PD, Umaru DA. The reoccurrence of H5N1 outbreaks necessitates the development of safe and effective influenza vaccine technologies for the prevention and control of avian influenza in Sub-Saharan Africa. ACTA ACUST UNITED AC 2015. [DOI: 10.5897/bmbr2015.0246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
368
|
Rodrigues AF, Soares HR, Guerreiro MR, Alves PM, Coroadinha AS. Viral vaccines and their manufacturing cell substrates: New trends and designs in modern vaccinology. Biotechnol J 2015. [PMID: 26212697 PMCID: PMC7161866 DOI: 10.1002/biot.201400387] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vaccination is one of the most effective interventions in global health. The worldwide vaccination programs significantly reduced the number of deaths caused by infectious agents. A successful example was the eradication of smallpox in 1979 after two centuries of vaccination campaigns. Since the first variolation administrations until today, the knowledge on immunology has increased substantially. This knowledge combined with the introduction of cell culture and DNA recombinant technologies revolutionized vaccine design. This review will focus on vaccines against human viral pathogens, recent developments on vaccine design and cell substrates used for their manufacture. While the production of attenuated and inactivated vaccines requires the use of the respective permissible cell substrates, the production of recombinant antigens, virus‐like particles, vectored vaccines and chimeric vaccines requires the use – and often the development – of specific cell lines. Indeed, the development of novel modern viral vaccine designs combined with, the stringent safety requirements for manufacture, and the better understanding on animal cell metabolism and physiology are increasing the awareness on the importance of cell line development and engineering areas. A new era of modern vaccinology is arriving, offering an extensive toolbox to materialize novel and creative ideas in vaccine design and its manufacture.
Collapse
Affiliation(s)
- Ana F Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Hugo R Soares
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Miguel R Guerreiro
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana S Coroadinha
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal. .,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
369
|
Koho T, Ihalainen TO, Stark M, Uusi-Kerttula H, Wieneke R, Rahikainen R, Blazevic V, Marjomäki V, Tampé R, Kulomaa MS, Hytönen VP. His-tagged norovirus-like particles: A versatile platform for cellular delivery and surface display. Eur J Pharm Biopharm 2015; 96:22-31. [PMID: 26170162 DOI: 10.1016/j.ejpb.2015.07.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/01/2015] [Accepted: 07/06/2015] [Indexed: 11/26/2022]
Abstract
In addition to vaccines, noninfectious virus-like particles (VLPs) that mimic the viral capsid show an attractive possibility of presenting immunogenic epitopes or targeting molecules on their surface. Here, functionalization of norovirus-derived VLPs by simple non-covalent conjugation of various molecules is shown. By using the affinity between a surface-exposed polyhistidine-tag and multivalent tris-nitrilotriacetic acid (trisNTA), fluorescent dye molecules and streptavidin-biotin conjugated to trisNTA are displayed on the VLPs to demonstrate the use of these VLPs as easily modifiable nanocarriers as well as a versatile vaccine platform. The VLPs are able to enter and deliver surface-displayed fluorescent dye into HEK293T cells via a surface-attached cell internalization peptide (VSV-G). The ease of manufacturing, the robust structure of these VLPs, and the straightforward conjugation provide a technology, which can be adapted to various applications in biomedicine.
Collapse
Affiliation(s)
- Tiia Koho
- BioMediTech, University of Tampere, Biokatu 6, FI-33520 Tampere, Finland
| | - Teemu O Ihalainen
- BioMediTech, University of Tampere, Biokatu 6, FI-33520 Tampere, Finland
| | - Marie Stark
- Department of Biological and Environmental Science/Nanoscience Center, Survontie 9, FI-40500 Jyväskylä, Finland
| | - Hanni Uusi-Kerttula
- Vaccine Research Center, Medical School, University of Tampere, Biokatu 10, FI-33520 Tampere, Finland
| | - Ralph Wieneke
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue-Strasse 9, D-60438 Frankfurt am Main, Germany
| | - Rolle Rahikainen
- BioMediTech, University of Tampere, Biokatu 6, FI-33520 Tampere, Finland
| | - Vesna Blazevic
- Vaccine Research Center, Medical School, University of Tampere, Biokatu 10, FI-33520 Tampere, Finland
| | - Varpu Marjomäki
- Department of Biological and Environmental Science/Nanoscience Center, Survontie 9, FI-40500 Jyväskylä, Finland
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue-Strasse 9, D-60438 Frankfurt am Main, Germany
| | - Markku S Kulomaa
- BioMediTech, University of Tampere, Biokatu 6, FI-33520 Tampere, Finland
| | - Vesa P Hytönen
- BioMediTech, University of Tampere, Biokatu 6, FI-33520 Tampere, Finland; Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland.
| |
Collapse
|
370
|
Adsorption of virus-like particles on ion exchange surface: Conformational changes at different pH detected by dual polarization interferometry. J Chromatogr A 2015; 1408:161-8. [PMID: 26189208 DOI: 10.1016/j.chroma.2015.07.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 06/29/2015] [Accepted: 07/05/2015] [Indexed: 11/21/2022]
Abstract
Disassembling of virus-like particles (VLPs) like hepatitis B virus surface antigen (HB-VLPs) during chromatographic process has been identified as a major cause of loss of antigen activity. In this study, dual polarization interferometry (DPI) measurement, together with chromatography experiments, were performed to study the adsorption and conformational change of HB-VLPs on ion exchange surface at three different pHs. Changes in pH values of buffer solution showed only minimal effect on the HB-VLPs assembly and antigen activity, while significantly different degree of HB-VLPs disassembling was observed after ion exchange chromatography (IEC) at different pHs, indicating the conformational change of HB-VLPs caused mainly by its interactions with the adsorbent surface. By creating an ion exchange surface on chip surface, the conformational changes of HB-VLPs during adsorption to the surface were monitored in real time by DPI for the first time. As pH increased from 7.0 to 9.0, strong electrostatic interactions between oppositely charged HB-VLPs and the ion exchange surface make the HB-VLPs spread thinly or even adsorbed in disassembled formation on the surface as revealed by significant decrease in thickness of the adsorbed layer measured by DPI. Such findings were consistent with the results of IEC experiments operated at different pHs, that more disassembled HB-VLPs were detected in the eluted proteins at pH 9.0. At low pH like pH 5.0, however, possible bi-layer adsorption was involved as evidenced by an adsorbed layer thickness higher than average diameter of the HB-VLPs. The "lateral" protein-protein interactions might be unfavorable and would make additional contribution to the disassembling of HB-VLPs besides the primary mechanism related to the protein-surface interactions; therefore, the lowest antigen activity was observed after IEC at pH 5.0. Such real-time information on conformational change of VLPs is helpful for better understanding the real mechanism for the disassembling of VLPs on the solid-liquid interface.
Collapse
|
371
|
Lung CD8+ T Cell Impairment Occurs during Human Metapneumovirus Infection despite Virus-Like Particle Induction of Functional CD8+ T Cells. J Virol 2015; 89:8713-26. [PMID: 26063431 DOI: 10.1128/jvi.00670-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/04/2015] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Human metapneumovirus (HMPV) is a major cause of respiratory disease in infants, the elderly, and immunocompromised individuals worldwide. There is currently no licensed HMPV vaccine. Virus-like particles (VLPs) are an attractive vaccine candidate because they are noninfectious and elicit a neutralizing antibody response. However, studies show that serum neutralizing antibodies are insufficient for complete protection against reinfection and that adaptive T cell immunity is important for viral clearance. HMPV and other respiratory viruses induce lung CD8(+) T cell (TCD8) impairment, mediated by programmed death 1 (PD-1). In this study, we generated HMPV VLPs by expressing the fusion and matrix proteins in mammalian cells and tested whether VLP immunization induces functional HMPV-specific TCD8 responses in mice. C57BL/6 mice vaccinated twice with VLPs and subsequently challenged with HMPV were protected from lung viral replication for at least 20 weeks postimmunization. A single VLP dose elicited F- and M-specific lung TCD8s with higher function and lower expression of PD-1 and other inhibitory receptors than TCD8s from HMPV-infected mice. However, after HMPV challenge, lung TCD8s from VLP-vaccinated mice exhibited inhibitory receptor expression and functional impairment similar to those of mice experiencing secondary infection. HMPV challenge of VLP-immunized μMT mice also elicited a large percentage of impaired lung TCD8s, similar to mice experiencing secondary infection. Together, these results indicate that VLPs are a promising vaccine candidate but do not prevent lung TCD8 impairment upon HMPV challenge. IMPORTANCE Human metapneumovirus (HMPV) is a leading cause of acute respiratory disease for which there is no licensed vaccine. Virus-like particles (VLPs) are an attractive vaccine candidate and induce antibodies, but T cell responses are less defined. Moreover, HMPV and other respiratory viruses induce lung CD8(+) T cell (TCD8) impairment mediated by programmed death 1 (PD-1). In this study, HMPV VLPs containing viral fusion and matrix proteins elicited epitope-specific TCD8s that were functional with low PD-1 expression. Two VLP doses conferred sterilizing immunity in C57BL/6 mice and facilitated HMPV clearance in antibody-deficient μMT mice without enhancing lung pathology. However, regardless of whether responding lung TCD8s had previously encountered HMPV antigens in the context of VLPs or virus, similar proportions were impaired and expressed comparable levels of PD-1 upon viral challenge. These results suggest that VLPs are a promising vaccine candidate but do not prevent lung TCD8 impairment upon HMPV challenge.
Collapse
|
372
|
Doll TAPF, Neef T, Duong N, Lanar DE, Ringler P, Müller SA, Burkhard P. Optimizing the design of protein nanoparticles as carriers for vaccine applications. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1705-13. [PMID: 26051652 PMCID: PMC4587294 DOI: 10.1016/j.nano.2015.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 03/02/2015] [Accepted: 05/19/2015] [Indexed: 01/19/2023]
Abstract
Successful vaccine development remains a huge challenge for infectious diseases such as malaria, HIV and influenza. As a novel way to present antigenic epitopes to the immune system, we have developed icosahedral self-assembling protein nanoparticles (SAPNs) to serve as a prototypical vaccine platform for infectious diseases. Here we examine some biophysical factors that affect the self-assembly of these nanoparticles, which have as basic building blocks coiled-coil oligomerization domains joined by a short linker region. Relying on in silico computer modeling predictions, we selected five different linker regions from the RCSB protein database that connect oligomerization domains, and then further studied the self-assembly and stability of in vitro produced nanoparticles through biophysical characterization of formed particles. One design in particular, T2i88, revealed excellent self-assembly and homogeneity thus paving the way toward a more optimized nanoparticle for vaccine applications. From the Clinical Editor Despite the widespread use of vaccines worldwide, successful development of vaccines against some diseases remains a challenge still. In this article, the authors investigated the physic-chemical and biological properties of icosahedral self-assembling protein nanoparticles (SAPNs), which mimic viral particles, in order to utilize this technology as potential platform for future design of vaccines.
Collapse
Affiliation(s)
- Tais A P F Doll
- Institute of Materials Science, University of Connecticut, Storrs, CT, USA
| | - Tobias Neef
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Nha Duong
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - David E Lanar
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, MD, USA
| | - Philippe Ringler
- Center for Cellular Imaging and Nano Analytics (C-CINA), Biozentrum, University of Basel, Mattenstrasse 26, Basel, Switzerland
| | - Shirley A Müller
- Center for Cellular Imaging and Nano Analytics (C-CINA), Biozentrum, University of Basel, Mattenstrasse 26, Basel, Switzerland
| | - Peter Burkhard
- Institute of Materials Science, University of Connecticut, Storrs, CT, USA; Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
373
|
Abstract
Antigen- and adjuvant-based bioconjugates that can stimulate the immune system play an important role in vaccine applications. Bioconjugates have demonstrated unique physicochemical and biological properties, enabling vaccines to be delivered to key immune cells, to target specific intracellular pathways, or to mimic immunogenic properties of natural pathogens. In this Review we highlight recent advances in such molecular immunomodulators, with an emphasis on the structure-function relationships that provide the foundation for rational design of safe and effective vaccines and immunotherapies.
Collapse
Affiliation(s)
- Haipeng Liu
- †Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, United States
- ‡Department of Oncology, Wayne State University, Detroit, Michigan 48201, United States
- §Tumor Biology and Microenvironment Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| | - Darrell J Irvine
- ▼Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
| |
Collapse
|
374
|
Pitoiset F, Vazquez T, Bellier B. Enveloped virus-like particle platforms: vaccines of the future? Expert Rev Vaccines 2015; 14:913-5. [PMID: 25968245 DOI: 10.1586/14760584.2015.1046440] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The techniques to produce effective vaccines have evolved, and the early vaccines (live, inactivated, subunit...) are no longer considered as the most appropriate for new vaccine development. We question here what will be the future vaccines, and argue that virus-like particle (VLP)-based vaccines are promising candidates. In addition to being effective vaccines against analogous viruses from which they are derived, VLPs can also be used to present foreign epitopes to the immune system. The achievement of this strategy can be illustrated by the recent development of malaria candidate vaccine. We point out recent VLP-based vaccine developments and discuss future perspectives.
Collapse
Affiliation(s)
- Fabien Pitoiset
- Department of Inflammation-Immunopathology-Biotherapy (I2B), Clinical Investigation Center for Biotherapies (CIC-BTi), Hôpital Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | | | | |
Collapse
|
375
|
Jia Y, Krishnan L, Omri A. Nasal and pulmonary vaccine delivery using particulate carriers. Expert Opin Drug Deliv 2015; 12:993-1008. [PMID: 25952104 DOI: 10.1517/17425247.2015.1044435] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Many human pathogens cause respiratory illness by colonizing and invading the respiratory mucosal surfaces. Preventing infection at local sites via mucosally active vaccines is a promising and rational approach for vaccine development. However, stimulating mucosal immunity is often challenging. Particulate adjuvants that can specifically target mucosal immune cells offer a promising opportunity to stimulate local immunity at the nasal and/or pulmonary mucosal surfaces. AREAS COVERED This review analyzes the common causes of respiratory infections, the challenges in the induction of mucosal and systemic responses and current pulmonary and nasal mucosal vaccination strategies. The ability of various particulate adjuvant formulations, including lipid-based particles, polymers and other particulate systems, to be effectively utilized for mucosal vaccine delivery is discussed. EXPERT OPINION Induction of antibody and cell-mediated mucosal immunity that can effectively combat respiratory pathogens remains a challenge. Particulate delivery systems can be developed to target mucosal immune cells and effectively present antigen to evoke a rapid and long-term local immunity in the respiratory mucosa. In particular, particulate delivery systems offer the versatility of being formulated with multiple adjuvants and antigenic cargo, and can be tailored to effectively prime immune responses across the mucosal barrier. The opportunity for rational design of novel subunit particulate vaccines is emerging.
Collapse
Affiliation(s)
- Yimei Jia
- National Research Council of Canada-Human Health Therapeutics , Ottawa, Ontario K1A 0R6 , Canada +1 613 991 3210 ;
| | | | | |
Collapse
|
376
|
Adhikary RR, More P, Banerjee R. Smart nanoparticles as targeting platforms for HIV infections. NANOSCALE 2015; 7:7520-7534. [PMID: 25874901 DOI: 10.1039/c5nr01285f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
While Human Immunodeficiency Virus (HIV) infections are reducing in incidence with the advent of Highly Active Anti-retroviral Therapy (HAART), there remain a number of challenges including the existence of reservoirs, drug resistance and anatomical barriers to antiretroviral therapy. To overcome these, smart nanoparticles with stimuli responsive release are proposed for delivery of anti-retroviral agents. The paper highlights the strategic similarities between the design of smart antiretroviral nanocarriers and those optimized for cancer chemotherapy. This includes the development of nanoparticles capable of passive and active targeting as well as those that are responsive to various internal and external triggers. For antiretroviral therapy, the relevant triggers for stimuli responsive release of drugs include semen, enzymes, endosomal escape, temperature and magnetic field. Deriving from the experience of cancer chemotherapy, additional potential triggers are light and ultrasound which remain hitherto unexplored in HIV therapy. In addition, the roles of nanomicrobicides (nanogels) and virus mimetic nanoparticles are discussed from the point of view of prevention of HIV transmission. The challenges associated with translation of smart nanoparticles for HIV infections to realize the Millennium Development Goal of combating HIV infections are discussed.
Collapse
Affiliation(s)
- Rishi Rajat Adhikary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| | | | | |
Collapse
|
377
|
Crooks ET, Tong T, Chakrabarti B, Narayan K, Georgiev IS, Menis S, Huang X, Kulp D, Osawa K, Muranaka J, Stewart-Jones G, Destefano J, O’Dell S, LaBranche C, Robinson JE, Montefiori DC, McKee K, Du SX, Doria-Rose N, Kwong PD, Mascola JR, Zhu P, Schief WR, Wyatt RT, Whalen RG, Binley JM. Vaccine-Elicited Tier 2 HIV-1 Neutralizing Antibodies Bind to Quaternary Epitopes Involving Glycan-Deficient Patches Proximal to the CD4 Binding Site. PLoS Pathog 2015; 11:e1004932. [PMID: 26023780 PMCID: PMC4449185 DOI: 10.1371/journal.ppat.1004932] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/04/2015] [Indexed: 12/28/2022] Open
Abstract
Eliciting broad tier 2 neutralizing antibodies (nAbs) is a major goal of HIV-1 vaccine research. Here we investigated the ability of native, membrane-expressed JR-FL Env trimers to elicit nAbs. Unusually potent nAb titers developed in 2 of 8 rabbits immunized with virus-like particles (VLPs) expressing trimers (trimer VLP sera) and in 1 of 20 rabbits immunized with DNA expressing native Env trimer, followed by a protein boost (DNA trimer sera). All 3 sera neutralized via quaternary epitopes and exploited natural gaps in the glycan defenses of the second conserved region of JR-FL gp120. Specifically, trimer VLP sera took advantage of the unusual absence of a glycan at residue 197 (present in 98.7% of Envs). Intriguingly, removing the N197 glycan (with no loss of tier 2 phenotype) rendered 50% or 16.7% (n = 18) of clade B tier 2 isolates sensitive to the two trimer VLP sera, showing broad neutralization via the surface masked by the N197 glycan. Neutralizing sera targeted epitopes that overlap with the CD4 binding site, consistent with the role of the N197 glycan in a putative "glycan fence" that limits access to this region. A bioinformatics analysis suggested shared features of one of the trimer VLP sera and monoclonal antibody PG9, consistent with its trimer-dependency. The neutralizing DNA trimer serum took advantage of the absence of a glycan at residue 230, also proximal to the CD4 binding site and suggesting an epitope similar to that of monoclonal antibody 8ANC195, albeit lacking tier 2 breadth. Taken together, our data show for the first time that strain-specific holes in the glycan fence can allow the development of tier 2 neutralizing antibodies to native spikes. Moreover, cross-neutralization can occur in the absence of protecting glycan. Overall, our observations provide new insights that may inform the future development of a neutralizing antibody vaccine.
Collapse
Affiliation(s)
- Ema T. Crooks
- San Diego Biomedical Research Institute, San Diego, California, United States of America
| | - Tommy Tong
- San Diego Biomedical Research Institute, San Diego, California, United States of America
| | - Bimal Chakrabarti
- International AIDS Vaccine Initiative (IAVI) Neutralizing Antibody Center at The Scripps Research Institute, Department of Immunology and Microbial Science, La Jolla, California, United States of America
| | - Kristin Narayan
- Altravax, Inc., Sunnyvale, California, United States of America
| | - Ivelin S. Georgiev
- Vaccine Research Center, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Sergey Menis
- International AIDS Vaccine Initiative (IAVI) Neutralizing Antibody Center at The Scripps Research Institute, Department of Immunology and Microbial Science, La Jolla, California, United States of America
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
| | - Xiaoxing Huang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Daniel Kulp
- International AIDS Vaccine Initiative (IAVI) Neutralizing Antibody Center at The Scripps Research Institute, Department of Immunology and Microbial Science, La Jolla, California, United States of America
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
| | - Keiko Osawa
- San Diego Biomedical Research Institute, San Diego, California, United States of America
| | | | - Guillaume Stewart-Jones
- Vaccine Research Center, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Joanne Destefano
- International AIDS Vaccine Initiative, Design and Development Laboratory, Brooklyn, New York, United States of America
| | - Sijy O’Dell
- Vaccine Research Center, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Celia LaBranche
- Department of Surgery, Duke University, Duke University Medical Center, Durham, North Carolina, United States of America
| | - James E. Robinson
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - David C. Montefiori
- Department of Surgery, Duke University, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Krisha McKee
- Vaccine Research Center, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Sean X. Du
- Altravax, Inc., Sunnyvale, California, United States of America
| | - Nicole Doria-Rose
- Vaccine Research Center, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Peter D. Kwong
- Vaccine Research Center, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - John R. Mascola
- Vaccine Research Center, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Ping Zhu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - William R. Schief
- International AIDS Vaccine Initiative (IAVI) Neutralizing Antibody Center at The Scripps Research Institute, Department of Immunology and Microbial Science, La Jolla, California, United States of America
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Richard T. Wyatt
- International AIDS Vaccine Initiative (IAVI) Neutralizing Antibody Center at The Scripps Research Institute, Department of Immunology and Microbial Science, La Jolla, California, United States of America
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, California, United States of America
| | | | - James M. Binley
- San Diego Biomedical Research Institute, San Diego, California, United States of America
| |
Collapse
|
378
|
Effio CL, Hubbuch J. Next generation vaccines and vectors: Designing downstream processes for recombinant protein-based virus-like particles. Biotechnol J 2015; 10:715-27. [PMID: 25880158 DOI: 10.1002/biot.201400392] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/11/2015] [Accepted: 03/19/2015] [Indexed: 12/28/2022]
Abstract
In recent years, the development of novel recombinant virus-like particles (VLPs) has been generating new perspectives for the prevention of untreated and arising infectious diseases. However, cost-reduction and acceleration of manufacturing processes for VLP-based vaccines or vectors are key challenges for the global health system. In particular, the design of rapid and cost-efficient purification processes is a critical bottleneck. In this review, we describe and evaluate new concepts, development strategies and unit operations for the downstream processing of VLPs. A special focus is placed on purity requirements and current trends, as well as chances and limitations of novel technologies. The discussed methods and case studies demonstrate the advances and remaining challenges in both rational process development and purification tools for large biomolecules. The potential of a new era of VLP-based products is highlighted by the progress of various VLPs in clinical phases.
Collapse
Affiliation(s)
- Christopher Ladd Effio
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | | |
Collapse
|
379
|
Immunogenic Display of Purified Chemically Cross-Linked HIV-1 Spikes. J Virol 2015; 89:6725-45. [PMID: 25878116 DOI: 10.1128/jvi.03738-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/11/2015] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED HIV-1 envelope glycoprotein (Env) spikes are prime vaccine candidates, at least in principle, but suffer from instability, molecular heterogeneity and a low copy number on virions. We anticipated that chemical cross-linking of HIV-1 would allow purification and molecular characterization of trimeric Env spikes, as well as high copy number immunization. Broadly neutralizing antibodies bound tightly to all major quaternary epitopes on cross-linked spikes. Covalent cross-linking of the trimer also stabilized broadly neutralizing epitopes, although surprisingly some individual epitopes were still somewhat sensitive to heat or reducing agent. Immunodepletion using non-neutralizing antibodies to gp120 and gp41 was an effective method for removing non-native-like Env. Cross-linked spikes, purified via an engineered C-terminal tag, were shown by negative stain EM to have well-ordered, trilobed structure. An immunization was performed comparing a boost with Env spikes on virions to spikes cross-linked and captured onto nanoparticles, each following a gp160 DNA prime. Although differences in neutralization did not reach statistical significance, cross-linked Env spikes elicited a more diverse and sporadically neutralizing antibody response against Tier 1b and 2 isolates when displayed on nanoparticles, despite attenuated binding titers to gp120 and V3 crown peptides. Our study demonstrates display of cross-linked trimeric Env spikes on nanoparticles, while showing a level of control over antigenicity, purity and density of virion-associated Env, which may have relevance for Env based vaccine strategies for HIV-1. IMPORTANCE The envelope spike (Env) is the target of HIV-1 neutralizing antibodies, which a successful vaccine will need to elicit. However, native Env on virions is innately labile, as well as heterogeneously and sparsely displayed. We therefore stabilized Env spikes using a chemical cross-linker and removed non-native Env by immunodepletion with non-neutralizing antibodies. Fixed native spikes were recognized by all classes of known broadly neutralizing antibodies but not by non-neutralizing antibodies and displayed on nanoparticles in high copy number. An immunization experiment in rabbits revealed that cross-linking Env reduced its overall immunogenicity; however, high-copy display on nanoparticles enabled boosting of antibodies that sporadically neutralized some relatively resistant HIV-1 isolates, albeit at a low titer. This study describes the purification of stable and antigenically correct Env spikes from virions that can be used as immunogens.
Collapse
|
380
|
Zhang C, Ku Z, Liu Q, Wang X, Chen T, Ye X, Li D, Jin X, Huang Z. High-yield production of recombinant virus-like particles of enterovirus 71 in Pichia pastoris and their protective efficacy against oral viral challenge in mice. Vaccine 2015; 33:2335-41. [PMID: 25820068 DOI: 10.1016/j.vaccine.2015.03.034] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 02/23/2015] [Accepted: 03/12/2015] [Indexed: 11/27/2022]
Abstract
Enterovirus 71 (EV71) is one of the major causative pathogens of hand, foot and mouth disease (HFMD), which is highly prevalent in the Asia-Pacific regions. Severe HFMD cases with neurological complications and even death are often associated with EV71 infections. However, no licensed EV71 vaccine is currently available. Recombinant virus-like particles (VLPs) of EV71 have been produced and shown to be a promising vaccine candidate in preclinical studies. However, the performance of current recombinant expression systems for EV71 VLP production remains unsatisfactory with regard to VLP yield and manufacturing procedure, and thus hinders further product development. In this study, we evaluated the expression of EV71 VLPs in Pichia pastoris and determined their protective efficacy in mouse models of EV71 infections. We showed that EV71 VLPs could be produced at high levels up to 4.9% of total soluble protein in transgenic P. pastoris yeast co-expressing P1 and 3CD proteins of EV71. The resulting yeast-produced VLPs potently induced neutralizing antibodies against homologous and heterologous EV71 strains in mice. More importantly, maternal immunization with VLPs protected neonatal mice in both intraperitoneal and oral challenge experiments. Collectively, these results demonstrated the success of simple, high-yield production of EV71 VLPs in transgenic P. pastoris, thus lifting the major roadblock in commercial development of VLP-based EV71 vaccines.
Collapse
Affiliation(s)
- Chao Zhang
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zhiqiang Ku
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qingwei Liu
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiaoli Wang
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Tan Chen
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiaohua Ye
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Dapeng Li
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xia Jin
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zhong Huang
- Vaccine Research Center, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| |
Collapse
|
381
|
Kang H, Qi Y, Wang H, Zheng X, Gao Y, Li N, Yang S, Xia X. Chimeric rabies virus-like particles containing membrane-anchored GM-CSF enhances the immune response against rabies virus. Viruses 2015; 7:1134-52. [PMID: 25768031 PMCID: PMC4379564 DOI: 10.3390/v7031134] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 02/13/2015] [Accepted: 02/24/2015] [Indexed: 12/17/2022] Open
Abstract
Rabies remains an important public health threat in most developing countries. To develop a more effective and safe vaccine against rabies, we have constructed a chimeric rabies virus-like particle (VLP), which containing glycoprotein (G) and matrix protein (M) of rabies virus (RABV) Evelyn-Rokitnicki-Abelseth (ERA) strain, and membrane-anchored granulocyte-macrophage colony-stimulating factor (GM-CSF), and it was named of EVLP-G. The immunogenicity and protective efficacy of EVLP-G against RABV were evaluated by intramuscular administration in a mouse model. The EVLP-G was successfully produced in insect cells by coinfection with three recombinant baculoviruses expressing G, M, and GM-CSF, respectively. The membrane-anchored GM-CSF possesses a strong adjuvant activity. More B cells and dendritic cells (DCs) were recruited and/or activated in inguinal lymph nodes in mice immunized with EVLP-G. EVLP-G was found to induce a significantly increased RABV-specific virus-neutralizing antibody and elicit a larger and broader antibody subclass responses compared with the standard rabies VLP (sRVLP, consisting of G and M). The EVLP-G also elicited significantly more IFN-γ- or IL-4-secreting CD4+ and CD8+ T cells than the sRVLP. Moreover, the immune responses induced by EVLP-G protect all vaccinated mice from lethal challenge with RABV. These results suggest that EVLP-G has the potential to be developed as a novel vaccine candidate for the prevention and control of animal rabies.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Female
- Glycoproteins/genetics
- Glycoproteins/immunology
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Injections, Intramuscular
- Interferon-gamma/metabolism
- Interleukin-4/metabolism
- Mice, Inbred BALB C
- Rabies/prevention & control
- Rabies Vaccines/administration & dosage
- Rabies Vaccines/genetics
- Rabies Vaccines/immunology
- Rabies virus/genetics
- Rabies virus/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/immunology
Collapse
Affiliation(s)
- Hongtao Kang
- College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China.
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, 666 Liuying West Road, Changchun 130122, China.
| | - Yinglin Qi
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, 666 Liuying West Road, Changchun 130122, China.
- College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun 130062, China.
| | - Hualei Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, 666 Liuying West Road, Changchun 130122, China.
| | - Xuexing Zheng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, 666 Liuying West Road, Changchun 130122, China.
| | - Yuwei Gao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, 666 Liuying West Road, Changchun 130122, China.
| | - Nan Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, 666 Liuying West Road, Changchun 130122, China.
| | - Songtao Yang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, 666 Liuying West Road, Changchun 130122, China.
- College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun 130062, China.
| | - Xianzhu Xia
- College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Guangzhou 510642, China.
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, 666 Liuying West Road, Changchun 130122, China.
- College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun 130062, China.
| |
Collapse
|
382
|
Seth A, Ritchie FK, Wibowo N, Lua LHL, Middelberg APJ. Non-carrier nanoparticles adjuvant modular protein vaccine in a particle-dependent manner. PLoS One 2015; 10:e0117203. [PMID: 25756283 PMCID: PMC4355484 DOI: 10.1371/journal.pone.0117203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/20/2014] [Indexed: 11/18/2022] Open
Abstract
Nanoparticles are increasingly used to adjuvant vaccine formulations due to their biocompatibility, ease of manufacture and the opportunity to tailor their size, shape, and physicochemical properties. The efficacy of similarly-sized silica (Si-OH), poly (D,L-lactic-co-glycolic acid) (PLGA) and poly caprolactone (PCL) nanoparticles (nps) to adjuvant recombinant capsomere presenting antigenic M2e modular peptide from Influenza A virus (CapM2e) was investigated in vivo. Formulation of CapM2e with Si-OH or PLGA nps significantly boosted the immunogenicity of modular capsomeres, even though CapM2e was not actively attached to the nanoparticles prior to injection (i.e., formulation was by simple mixing). In contrast, PCL nps showed no significant adjuvant effect using this simple-mixing approach. The immune response induced by CapM2e alone or formulated with nps was antibody-biased with very high antigen-specific antibody titer and less than 20 cells per million splenocytes secreting interferon gamma. Modification of silica nanoparticle surface properties through amine functionalization and pegylation did not lead to significant changes in immune response. This study confirms that simple mixing-based formulation can lead to effective adjuvanting of antigenic protein, though with antibody titer dependent on nanoparticle physicochemical properties.
Collapse
Affiliation(s)
- Arjun Seth
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD, Australia
| | - Fiona K Ritchie
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD, Australia
| | - Nani Wibowo
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD, Australia
| | - Linda H L Lua
- The University of Queensland, Protein Expression Facility, St Lucia, QLD, Australia
| | - Anton P J Middelberg
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD, Australia
| |
Collapse
|
383
|
Wi GR, Hwang JY, Kwon MG, Kim HJ, Kang HA, Kim HJ. Protective immunity against nervous necrosis virus in convict grouper Epinephelus septemfasciatus following vaccination with virus-like particles produced in yeast Saccharomyces cerevisiae. Vet Microbiol 2015; 177:214-8. [PMID: 25759291 DOI: 10.1016/j.vetmic.2015.02.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 02/14/2015] [Accepted: 02/17/2015] [Indexed: 10/23/2022]
Abstract
Infection with nervous necrosis virus (NNV) causes viral nervous necrosis, which inflicts serious economic losses in marine fish cultivation. Virus-like particles (VLPs) are protein complexes consisting of recombinant virus capsid proteins, whose shapes are similar to native virions. VLPs are considered a novel vaccine platform because they are not infectious and have the ability to induce neutralizing antibodies efficiently. However, there have been few studies of protective immune responses employing virus challenge following immunization with NNV VLPs, and this is important for evaluating the utility of the vaccine. In the present study, we produced red-spotted grouper (Epinephelus akaara) NNV (RGNNV) VLPs in Saccharomyces cerevisiae and investigated protective immune responses in convict grouper (Epinephelus septemfasciatus) following intraperitoneal injection and oral immunization with the RGNNV VLPs. The parenterally administered VLPs elicited neutralizing antibody with high efficacy, and provided the fish with full protection against RGNNV challenge: 100% of the immunized fish survived compared with only 37% of the control fish receiving phosphate-buffered saline. RGNNV VLPs administered orally provoked neutralizing antibody systemically and conferred protective immunity against virus challenge: however only 57% of the fish survived. Our results demonstrate that RGNNV VLP produced in yeast has great potential as vaccine in fish.
Collapse
Affiliation(s)
- Ga Ram Wi
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 156-756, South Korea
| | - Jee Youn Hwang
- Fish Pathology Division, National Fisheries Research and Development Institute, Busan 619-902, South Korea
| | - Mun-Gyeong Kwon
- Fish Pathology Division, National Fisheries Research and Development Institute, Busan 619-902, South Korea
| | - Hyoung Jin Kim
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 156-756, South Korea
| | - Hyun Ah Kang
- Department of Life Science, College of Natural Science, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 156-756, South Korea
| | - Hong-Jin Kim
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 156-756, South Korea.
| |
Collapse
|
384
|
Ayithan N, Bradfute SB, Anthony SM, Stuthman KS, Bavari S, Bray M, Ozato K. Virus-like particles activate type I interferon pathways to facilitate post-exposure protection against Ebola virus infection. PLoS One 2015; 10:e0118345. [PMID: 25719445 PMCID: PMC4342244 DOI: 10.1371/journal.pone.0118345] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/14/2015] [Indexed: 12/19/2022] Open
Abstract
Ebola virus (EBOV) causes a severe hemorrhagic disease with high fatality. Virus-like particles (VLPs) are a promising vaccine candidate against EBOV. We recently showed that VLPs protect mice from lethal EBOV infection when given before or after viral infection. To elucidate pathways through which VLPs confer post-exposure protection, we investigated the role of type I interferon (IFN) signaling. We found that VLPs lead to accelerated induction of IFN stimulated genes (ISGs) in liver and spleen of wild type mice, but not in Ifnar-/- mice. Accordingly, EBOV infected Ifnar-/- mice, unlike wild type mice succumbed to death even after VLP treatment. The ISGs induced in wild type mice included anti-viral proteins and negative feedback factors known to restrict viral replication and excessive inflammatory responses. Importantly, proinflammatory cytokine/chemokine expression was much higher in WT mice without VLPs than mice treated with VLPs. In EBOV infected Ifnar-/- mice, however, uninhibited viral replication and elevated proinflammatory factor expression ensued, irrespective of VLP treatment, supporting the view that type I IFN signaling helps to limit viral replication and attenuate inflammatory responses. Further analyses showed that VLP protection requires the transcription factor, IRF8 known to amplify type I IFN signaling in dendritic cells and macrophages, the probable sites of initial EBOV infection. Together, this study indicates that VLPs afford post-exposure protection by promoting expeditious initiation of type I IFN signaling in the host.
Collapse
Affiliation(s)
- Natarajan Ayithan
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, United States of America
| | - Steven B. Bradfute
- United States Army Medical Institute of Infectious Diseases, Fort Detrick, MD, United States of America
| | - Scott M. Anthony
- United States Army Medical Institute of Infectious Diseases, Fort Detrick, MD, United States of America
| | - Kelly S. Stuthman
- United States Army Medical Institute of Infectious Diseases, Fort Detrick, MD, United States of America
| | - Sina Bavari
- United States Army Medical Institute of Infectious Diseases, Fort Detrick, MD, United States of America
| | - Mike Bray
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institute of Health, Fort Detrick, MD, United States of America
| | - Keiko Ozato
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, United States of America
- * E-mail:
| |
Collapse
|
385
|
Kumar R, Sreenivasa BP, Tamilselvan RP. Construction and characterization of recombinant human adenovirus type 5 expressing foot-and-mouth disease virus capsid proteins of Indian vaccine strain, O/IND/R2/75. Vet World 2015; 8:147-55. [PMID: 27047064 PMCID: PMC4774695 DOI: 10.14202/vetworld.2015.147-155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/18/2014] [Accepted: 12/27/2014] [Indexed: 12/20/2022] Open
Abstract
AIM Generation of recombinant human adenovirus type 5 expressing foot-and-mouth disease virus (FMDV) capsid protein genes along with full-length 2B, 3B and 3C(pro) and its characterization. MATERIALS AND METHODS FMD viral RNA isolation, cDNA synthesis, and polymerase chain reaction were performed to synthesize expression cassettes (P1-2AB3BC(wt) and P1-2AB3BC(m)) followed by cloning in pShuttle-CMV vector. Chemically competent BJ5183-AD-1 cells were transformed with the recombinant pShuttle-CMV to produce recombinant adenoviral plasmids. HEK-293 cells were transfected with the recombinant adenoviral plasmids to generate recombinant adenoviruses (hAd5/P1-2AB3BC(wt) and hAd5/P1-2AB3BC(m)). Expression of the target proteins was analyzed by sandwich ELISA and indirect immunofluorescence assay. The recombinant adenoviruses were purified and concentrated by CsCl density gradient ultracentrifugation. Growth kinetics and thermostability of the recombinant adenoviruses were compared with that of non-recombinant replication-defective adenovirus (dAd5). RESULTS The recombinant adenoviruses containing capsid protein genes of the FMDV O/IND/R2/75 were generated and amplified in HEK-293 cells. The titer of the recombinant adenoviruses was approximately 10(8), 10(9.5) and 10(11) TCID50/ml in supernatant media, cell lysate and CsCl purified preparation, respectively. Expression of the FMDV capsid protein was detectable in sandwich ELISA and confirmed by immunofluorescence assay. Growth kinetics of the recombinant adenoviruses did not reveal a significant difference when compared with that of dAd5. A decrement of up to 10-fold at 4°C and 21-fold at 37°C was recorded in the virus titers during 60 h incubation period and found to be statistically significant (p<0.01). CONCLUSION Recombinant adenoviruses expressing capsid proteins of the FMDV O/IND/R2/75 were constructed and produced in high titers. In vitro expression of the target proteins in the adenovirus vector system was detected by sandwich ELISA and immunofluorescence assay.
Collapse
Affiliation(s)
- Ramesh Kumar
- FMD Research Centre, Indian Veterinary Research Institute, Bangalore - 560 024, India
| | - B P Sreenivasa
- FMD Research Centre, Indian Veterinary Research Institute, Bangalore - 560 024, India
| | - R P Tamilselvan
- FMD Research Centre, Indian Veterinary Research Institute, Bangalore - 560 024, India
| |
Collapse
|
386
|
Prokhnevsky A, Mamedov T, Leffet B, Rahimova R, Ghosh A, Mett V, Yusibov V. Development of a single-replicon miniBYV vector for co-expression of heterologous proteins. Mol Biotechnol 2015; 57:101-10. [PMID: 25280556 DOI: 10.1007/s12033-014-9806-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In planta production of recombinant proteins, including vaccine antigens and monoclonal antibodies, continues gaining acceptance. With the broadening range of target proteins, the need for vectors with higher performance is increasing. Here, we have developed a single-replicon vector based on beet yellows virus (BYV) that enables co-delivery of two target genes into the same host cell, resulting in transient expression of each target. This BYV vector maintained genetic stability during systemic spread throughout the host plant, Nicotiana benthamiana. Furthermore, we have engineered a miniBYV vector carrying the sequences encoding heavy and light chains of a monoclonal antibody (mAb) against protective antigen (PA) of Bacillius anthracis, and achieved the expression of the full-length functional anti-PA mAb at ~300 mg/kg of fresh leaf tissue. To demonstrate co-expression and functionality of two independent proteins, we cloned the sequences of the Pfs48/45 protein of Plasmodium falciparum and endoglycosidase F (PNGase F) from Flavobacterium meningosepticum into the miniBYV vector under the control of two subgenomic RNA promoters. Agroinfiltration of N. benthamiana with this miniBYV vector resulted in accumulation of biologically active Pfs48/45 that was devoid of N-linked glycosylation and had correct conformation and epitope display. Overall, our findings demonstrate that the new BYV-based vector is capable of co-expressing two functionally active recombinant proteins within the same host cell.
Collapse
Affiliation(s)
- Alex Prokhnevsky
- Fraunhofer USA Center for Molecular Biotechnology, 9 Innovation Way, Suite 200, Newark, DE, 19711, USA
| | | | | | | | | | | | | |
Collapse
|
387
|
|
388
|
Shawky H, Maghraby AS, Solliman MED, El-Mokadem MT, Sherif MM, Arafa A, Bahgat MM. Expression, immunogenicity and diagnostic value of envelope proteins from an Egyptian hepatitis C virus isolate. Arch Virol 2015; 160:945-58. [PMID: 25631616 DOI: 10.1007/s00705-015-2334-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/02/2015] [Indexed: 12/28/2022]
Abstract
The present work aimed at 1) characterization of the E1 and E2 proteins (HCV-E) from an Egyptian hepatitis C virus genotype 4a (HCV-4a) isolate at the molecular and immunological level, 2) in silico identification of the B- and T-cell epitopes responsible for the immunogenicity of HCV-E, and 3) evaluation of the diagnostic potential of both the recombinant HCV-E and antibodies raised using mammalian expression constructs encoding the protein. The region encoding the E1 and E2 proteins was amplified by RT-PCR from RNA isolated from blood of a human infected with HCV-4 and cloned into the pSC-TA plasmid, and the sequence was verified and used to construct a neighbor-joining phylogenetic tree. The translated nucleotide sequence was used to predict the HCV-E secondary structure using the PREDICT-PROTEIN server and PSI-PRED. A 3D model of HCV-E was generated using the online tool 3Dpro. B- and T-cell epitopes were predicted using the online tools BCPred and Epijen v1.0, respectively. The HCV-E-encoding sequence was later subcloned into the mammalian expression plasmid pQE, and the constructs that were generated were used to immunize mice in the absence and presence of adjuvants of plant origin. The maximum sequence identity obtained by nucleotide and protein BLAST analysis with previously published HCV-E sequences was 85 and 77 %, respectively. The B-cell epitope CFTPSPVVV at position 203 and the T-cell epitope ALSTGLIHL at position 380 were found to be highly conserved among all HCV genotypes. Both ELISA and Western blotting experiments on crude and purified recombinant HCV envelope proteins using mouse antisera raised using the HCV-E mammalian expression construct confirmed the specific antigenicity of the expressed protein. The antibodies raised in mice using the HCV-E-encoding construct could efficiently capture circulating antigens in patients' sera with good sensitivity that correlated with liver enzyme levels (r = 0.4052, P < 0.0001 for ALT; r = -0.5439, P = 0.0019 for AST). Moreover, combining the HCV-E-encoding construct with extracts prepared from Echinacea purpurea and Nigella sativa prior to immunizing mice significantly (P < 0.05) increased both the humoral (14.9- to 20-fold increase in antibodies) and the cellular (CD4(+) and cytotoxic CD8(+)- T lymphocytes) responses compared to mice that received the DNA construct alone or PBS-treated mice. Both recombinant HCV-E protein preparations and antibodies raised using the HCV-E-encoding mammalian expression construct represent useful diagnostic tools that can report on active HCV infection. Also, the immunostimulatory effects induced by the two plant extracts used at the cellular and humoral level highlight the potential of natural products for inducing protection against HCV infection. The neutralizing capacity of the induced antibodies is a subject of future investigations. Furthermore, the predicted B- and T-cell epitopes may be useful for tailoring future diagnostics and candidate vaccines against various HCV genotypes.
Collapse
Affiliation(s)
- Heba Shawky
- The Immunology and Infectious Diseases Laboratory, Therapeutic Chemistry Department, The Center of Excellence for Advanced Sciences, The National Research Centre, Dokki, Giza, 12622, Egypt,
| | | | | | | | | | | | | |
Collapse
|
389
|
Ladd Effio C, Wenger L, Ötes O, Oelmeier SA, Kneusel R, Hubbuch J. Downstream processing of virus-like particles: single-stage and multi-stage aqueous two-phase extraction. J Chromatogr A 2015; 1383:35-46. [PMID: 25637013 DOI: 10.1016/j.chroma.2015.01.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 12/31/2022]
Abstract
The demand for vaccines against untreated diseases has enforced the research and development of virus-like particle (VLP) based vaccine candidates in recent years. Significant progress has been made in increasing VLP titres during upstream processing in bacteria, yeast and insect cells. Considering downstream processing, the separation of host cell impurities is predominantly achieved by time-intensive ultracentrifugation processes or numerous chromatography and filtration steps. In this work, we evaluate the potential of an alternative separation technology for VLPs: aqueous two-phase extraction (ATPE). The benefits of ATPE have been demonstrated for various biomolecules, but capacity and separation efficiency were observed to be low for large biomolecules such as VLPs or viruses. Both performance parameters were examined in detail in a case study on human B19 parvovirus-like particles derived from Spodoptera frugiperda Sf9 insect cells. A solubility-guided approach enabled the design of polyethylene (PEG) salt aqueous two-phase systems with a high capacity of up to 4.1mg/mL VLPs. Unique separation efficiencies were obtained by varying the molecular weight of PEG, the pH value and by using neutral salt additives. Further improvement of the separation of host cell impurities was achieved by multi-stage ATPE on a centrifugal partition chromatography (CPC) device in 500mL scale. While single-stage ATPE enabled a DNA clearance of 99.6%, multi-stage ATPE improved the separation of host cell proteins (HCPs). The HPLC purity ranged from 16.8% (100% VLP recovery) for the single-stage ATPE to 69.1% (40.1% VLP recovery) for the multi-stage ATPE. An alternative two-step downstream process is presented removing the ATPS forming polymer, cell debris and 99.77% DNA with a HPLC purity of 90.6% and a VLP recovery of 63.9%.
Collapse
Affiliation(s)
- Christopher Ladd Effio
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | - Lukas Wenger
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | - Ozan Ötes
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | - Stefan A Oelmeier
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany; Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Jürgen Hubbuch
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany.
| |
Collapse
|
390
|
Chen Q, Sun Q, Molino NM, Wang SW, Boder ET, Chen W. Sortase A-mediated multi-functionalization of protein nanoparticles. Chem Commun (Camb) 2015; 51:12107-10. [DOI: 10.1039/c5cc03769g] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new strategy was developed to create multi-functionalizaton of protein nanoparticles using Sortase A-mediated ligation, resulting in modified protein nanoparticles that are both thermally responsive and catalytic active.
Collapse
Affiliation(s)
- Qi Chen
- Department of Chemical and Biomolecular Engineering
- University of Delaware
- Newark
- USA
| | - Qing Sun
- Department of Chemical and Biomolecular Engineering
- University of Delaware
- Newark
- USA
| | - Nicholas M. Molino
- Department of Chemical Engineering and Materials Science
- University of California
- Irvine
- USA
| | - Szu-Wen Wang
- Department of Chemical Engineering and Materials Science
- University of California
- Irvine
- USA
| | - Eric T. Boder
- Department of Chemical and Biomolecular Engineering
- University of Tennessee
- Knoxville
- USA
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering
- University of Delaware
- Newark
- USA
| |
Collapse
|
391
|
Zhang L, Lua LHL, Middelberg APJ, Sun Y, Connors NK. Biomolecular engineering of virus-like particles aided by computational chemistry methods. Chem Soc Rev 2015; 44:8608-18. [DOI: 10.1039/c5cs00526d] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multi-scale investigation of VLP self-assembly aided by computational methods is facilitating the design, redesign, and modification of functionalized VLPs.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072, People's Republic of China
| | - Linda H. L. Lua
- Protein Expression Facility
- The University of Queensland
- Brisbane, Australia
| | - Anton P. J. Middelberg
- Australian Institute for Bioengineering and Nanotechnology
- The University of Queensland
- Brisbane, Australia
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072, People's Republic of China
| | - Natalie K. Connors
- Australian Institute for Bioengineering and Nanotechnology
- The University of Queensland
- Brisbane, Australia
| |
Collapse
|
392
|
Abstract
This review describes nanoparticles made from protein by self-assembly or desolvation as carriers for the delivery of therapeutic proteins.
Collapse
Affiliation(s)
- L. P. Herrera Estrada
- School of Chemical & Biomolecular Engineering. Georgia Institute of Technology
- Atlanta
- USA
| | - J. A. Champion
- School of Chemical & Biomolecular Engineering. Georgia Institute of Technology
- Atlanta
- USA
| |
Collapse
|
393
|
Sun X, Wang Y, Dong C, Hu J, Yang L. High copy numbers and N terminal insertion position of influenza A M2E fused with hepatitis B core antigen enhanced immunogenicity. Biosci Trends 2015; 9:221-7. [DOI: 10.5582/bst.2015.01060] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Xincheng Sun
- Basic Medical School of Zhengzhou University
- College of Food and Biological Engineering, Zhengzhou University of Light Industry
| | - Yunlong Wang
- Basic Medical School of Zhengzhou University
- Bioengineering Research Center of Henan Province
- Henan Biotechnology Research Centre
| | - Caiwen Dong
- College of Food and Biological Engineering, Zhengzhou University of Light Industry
| | - Jinqiang Hu
- College of Food and Biological Engineering, Zhengzhou University of Light Industry
| | - Liping Yang
- Basic Medical School of Henan University of Traditional Chinese Medicine
| |
Collapse
|
394
|
Structural basis for the development of avian virus capsids that display influenza virus proteins and induce protective immunity. J Virol 2014; 89:2563-74. [PMID: 25520499 DOI: 10.1128/jvi.03025-14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Bioengineering of viruses and virus-like particles (VLPs) is a well-established approach in the development of new and improved vaccines against viral and bacterial pathogens. We report here that the capsid of a major avian pathogen, infectious bursal disease virus (IBDV), can accommodate heterologous proteins to induce protective immunity. The structural units of the ~70-nm-diameter T=13 IBDV capsid are trimers of VP2, which is made as a precursor (pVP2). The pVP2 C-terminal domain has an amphipathic α helix that controls VP2 polymorphism. In the absence of the VP3 scaffolding protein, 466-residue pVP2 intermediates bearing this α helix assemble into genuine VLPs only when expressed with an N-terminal His6 tag (the HT-VP2-466 protein). HT-VP2-466 capsids are optimal for protein insertion, as they are large enough (cargo space, ~78,000 nm(3)) and are assembled from a single protein. We explored HT-VP2-466-based chimeric capsids initially using enhanced green fluorescent protein (EGFP). The VLP assembly yield was efficient when we coexpressed EGFP-HT-VP2-466 and HT-VP2-466 from two recombinant baculoviruses. The native EGFP structure (~240 copies/virion) was successfully inserted in a functional form, as VLPs were fluorescent, and three-dimensional cryo-electron microscopy showed that the EGFP molecules incorporated at the inner capsid surface. Immunization of mice with purified EGFP-VLPs elicited anti-EGFP antibodies. We also inserted hemagglutinin (HA) and matrix (M2) protein epitopes derived from the mouse-adapted A/PR/8/34 influenza virus and engineered several HA- and M2-derived chimeric capsids. Mice immunized with VLPs containing the HA stalk, an M2 fragment, or both antigens developed full protection against viral challenge. IMPORTANCE Virus-like particles (VLPs) are multimeric protein cages that mimic the infectious virus capsid and are potential candidates as nonliving vaccines that induce long-lasting protection. Chimeric VLPs can display or include foreign antigens, which could be a conserved epitope to elicit broadly neutralizing antibodies or several variable epitopes effective against a large number of viral strains. We report the biochemical, structural, and immunological characterization of chimeric VLPs derived from infectious bursal disease virus (IBDV), an important poultry pathogen. To test the potential of IBDV VLPs as a vaccine vehicle, we used the enhanced green fluorescent protein and two fragments derived from the hemagglutinin and the M2 matrix protein of the human murine-adapted influenza virus. The IBDV capsid protein fused to influenza virus peptides formed assemblies able to protect mice against viral challenge. Our studies establish the basis for a new generation of multivalent IBDV-based vaccines.
Collapse
|
395
|
Yusibov V, Kushnir N, Streatfield SJ. Advances and challenges in the development and production of effective plant-based influenza vaccines. Expert Rev Vaccines 2014; 14:519-35. [PMID: 25487788 DOI: 10.1586/14760584.2015.989988] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Influenza infections continue to present a major threat to public health. Traditional modes of influenza vaccine manufacturing are failing to satisfy the global demand because of limited scalability and long production timelines. In contrast, subunit vaccines (SUVs) can be produced in heterologous expression systems in shorter times and at higher quantities. Plants are emerging as a promising platform for SUV production due to time efficiency, scalability, lack of harbored mammalian pathogens and possession of the machinery for eukaryotic post-translational protein modifications. So far, several organizations have utilized plant-based transient expression systems to produce SUVs against influenza, including vaccines based on virus-like particles. Plant-produced influenza SUV candidates have been extensively evaluated in animal models and some have shown safety and immunogenicity in clinical trials. Here, the authors review ongoing efforts and challenges to producing influenza SUV candidates in plants and discuss the likelihood of bringing these products to the market.
Collapse
Affiliation(s)
- Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, 9 Innovation Way, Suite 200, Newark, DE 19711, USA
| | | | | |
Collapse
|
396
|
Palomares O, Crameri R, Rhyner C. The contribution of biotechnology toward progress in diagnosis, management, and treatment of allergic diseases. Allergy 2014; 69:1588-601. [PMID: 25307026 DOI: 10.1111/all.12533] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2014] [Indexed: 12/18/2022]
Abstract
'Biotechnology' has been intuitively used by humans since thousands of years for the production of foods, beverages, and drugs based on the experience without any scientific background. However, the golden era of this discipline emerged only during the second half of the last century. Incredible progresses have been achieved on all fields starting from the industrialization of the production of foods to the discovery of antibiotics, the decipherment of the genetic code, and rational approaches to understand and define the status we now call 'healthy'. The extremely complex interactions between genetic background, life style, and environmental factors influencing our continuously increasing life span have become more and more evident and steadily generate new questions which are only partly answered. Here, we try to summarize the contribution of biotechnology to our understanding, control, and cure of IgE-mediated allergic diseases. We are aware that a review of such a vast topic can never cover all aspects of the progress achieved in the different fields.
Collapse
Affiliation(s)
- O. Palomares
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | - R. Crameri
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Davos Switzerland
| | - C. Rhyner
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Davos Switzerland
| |
Collapse
|
397
|
Updates in inducible transgene expression using viral vectors: from transient to stable expression. Curr Opin Biotechnol 2014; 32:85-92. [PMID: 25437638 DOI: 10.1016/j.copbio.2014.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 10/29/2014] [Accepted: 11/10/2014] [Indexed: 01/19/2023]
Abstract
The prospect of economically producing useful biologics in plants has greatly increased with the advent of viral vectors. The ability of viral vectors to amplify transgene expression has seen them develop into robust transient platforms for the high-level, rapid production of recombinant proteins. To adapt these systems to stably transformed plants, new ways of deconstructing the virus machinery and linking its expression and replication to chemically controlled promoters have been developed. The more advanced of these stable, inducible hyper-expression vectors provide both activated and amplified heterologous transgene expression. Such systems could be deployed in broad acre crops and provide a pathway to fully exploit the advantages of plants as a platform for the manufacture of a wide spectrum of products.
Collapse
|
398
|
Shoji Y, Prokhnevsky A, Leffet B, Vetter N, Tottey S, Satinover S, Musiychuk K, Shamloul M, Norikane J, Jones RM, Chichester JA, Green BJ, Streatfield SJ, Yusibov V. Immunogenicity of H1N1 influenza virus-like particles produced in Nicotiana benthamiana. Hum Vaccin Immunother 2014; 11:118-23. [PMID: 25483524 PMCID: PMC4514423 DOI: 10.4161/hv.34365] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/09/2014] [Indexed: 12/15/2022] Open
Abstract
The H1N1 influenza pandemic of 2009 stimulated interest in developing safe and effective subunit influenza vaccines using rapid and cost-effective recombinant technologies that can avoid dependence on hens' eggs supply and live viruses for production. Among alternative approaches to subunit vaccine development, virus-like particles (VLPs) represent an attractive strategy due to their safety and immunogenicity. Previously, we have produced a recombinant monomeric hemagglutinin (HA) protein derived from the A/California/04/09 (H1N1) strain of influenza virus in a plant-based transient expression system and demonstrated immunogenicity and safety of this monomeric HA in animal models and human volunteers. In an effort to produce higher potency influenza vaccine in plants, we have designed and generated enveloped VLPs using the ectodomain of HA from the A/California/04/09 strain and heterologous sequences. The resulting H1 HA VLPs (HAC-VLPs) elicited robust hemagglutination inhibition antibody responses in mice at doses lower than 1 µg in the presence or absence of Alhydrogel adjuvant. These results suggest enhanced immunogenicity of recombinant HA in the form of an enveloped VLP over soluble antigen.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Aluminum Hydroxide/administration & dosage
- Animals
- Antibodies, Viral/blood
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Hemagglutination Inhibition Tests
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/isolation & purification
- Mice, Inbred BALB C
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/metabolism
- Nicotiana/genetics
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/isolation & purification
Collapse
Affiliation(s)
- Yoko Shoji
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Alex Prokhnevsky
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Brett Leffet
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Nancy Vetter
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Stephen Tottey
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Shama Satinover
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | - R Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Brian J Green
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology; Newark, DE USA
| |
Collapse
|
399
|
Rodrigues AF, Carrondo MJT, Alves PM, Coroadinha AS. Cellular targets for improved manufacturing of virus-based biopharmaceuticals in animal cells. Trends Biotechnol 2014; 32:602-7. [PMID: 25450042 DOI: 10.1016/j.tibtech.2014.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 09/18/2014] [Accepted: 09/26/2014] [Indexed: 12/24/2022]
Abstract
The past decade witnessed the entry into the market of new virus-based biopharmaceuticals produced in animal cells such as oncolytic vectors, virus-like particle vaccines, and gene transfer vectors. Therefore, increased attention and investment to optimize cell culture processes towards enhanced manufacturing of these bioproducts is anticipated. Herein, we review key findings on virus-host interactions that have been explored in cell culture optimization. Approaches supporting improved productivity or quality of vector preparations are discussed, mainly focusing on medium design and genetic manipulation. This review provides an integrated outline for current and future efforts in exploring cellular targets for the optimization of cell culture manufacturing of virus-based biopharmaceuticals.
Collapse
Affiliation(s)
- Ana F Rodrigues
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Manuel J T Carrondo
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; FCT-UNL, P-2829-516 Caparica, Portugal
| | - Paula M Alves
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana S Coroadinha
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
400
|
Moon H, Lee J, Min J, Kang S. Developing Genetically Engineered Encapsulin Protein Cage Nanoparticles as a Targeted Delivery Nanoplatform. Biomacromolecules 2014; 15:3794-801. [DOI: 10.1021/bm501066m] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Hyojin Moon
- Department of Biological
Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, Korea
| | - Jisu Lee
- Department of Biological
Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, Korea
| | - Junseon Min
- Department of Biological
Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, Korea
| | - Sebyung Kang
- Department of Biological
Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, Korea
| |
Collapse
|