1
|
Yan K, Mao L, Lan J, Xiao Z. Advancements in dengue vaccines: A historical overview and pro-spects for following next-generation candidates. J Microbiol 2025; 63:e2410018. [PMID: 40044132 DOI: 10.71150/jm.2410018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/06/2025] [Indexed: 05/13/2025]
Abstract
Dengue, caused by four serotypes of dengue viruses (DENV-1 to DENV-4), is the most prevalent and widely mosquito-borne viral disease affecting humans. Dengue virus (DENV) infection has been reported in over 100 countries, and approximately half of the world's population is now at risk. The paucity of universally licensed DENV vaccines highlights the urgent need to address this public health concern. Action and atten-tion to antibody-dependent enhancement increase the difficulty of vaccine development. With the worsen-ing dengue fever epidemic, Dengvaxia® (CYD-TDV) and Qdenga® (TAK-003) have been approved for use in specific populations in affected areas. However, these vaccines do not provide a balanced immune response to all four DENV serotypes and the vaccination cannot cover all populations. There is still a need to develop a safe, broad-spectrum, and effective vaccine to address the increasing number of dengue cases worldwide. This review provides an overview of the existing DENV vaccines, as well as potential candidates for future studies on DENV vaccine development, and discusses the challenges and possible solutions in the field.
Collapse
Affiliation(s)
- Kai Yan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Lingjing Mao
- CAS Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection Chinese Academy of Sciences, Shanghai, P. R. China
- University of the Chinese Academy of Sciences, Beijing, P. R. China
| | - Jiaming Lan
- CAS Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection Chinese Academy of Sciences, Shanghai, P. R. China
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
2
|
Kim MY, Mason HS, Ma JKC, Reljic R. Recombinant immune complexes as vaccines against infectious diseases. Trends Biotechnol 2024; 42:1427-1438. [PMID: 38825437 DOI: 10.1016/j.tibtech.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 06/04/2024]
Abstract
New vaccine technologies are needed to combat many existing infections and prepare better for those that may emerge in the future. The conventional technologies that rely on protein-based vaccines are still severely restricted by the sparsity and poor accessibility of available adjuvants. One possible solution to this problem is to enhance antigen immunogenicity by a more natural means by complexing it with antibodies in the form of immune complexes (ICs). However, natural ICs are impractical as vaccines, and significant research efforts have been made to generate them in recombinant form, with plant bioengineering being at the forefront of these efforts. Here, we describe the challenges and progress made to date to make recombinant IC vaccines applicable to humans.
Collapse
Affiliation(s)
- Mi-Young Kim
- St. George's University of London, London, UK; Jeonbuk National University, Jeonju, South Korea
| | - Hugh S Mason
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | | | | |
Collapse
|
3
|
Kim M, Vergara E, Tran A, Paul MJ, Kwon T, Ma JK, Jang Y, Reljic R. Marked enhancement of the immunogenicity of plant-expressed IgG-Fc fusion proteins by inclusion of cholera toxin non-toxic B subunit within the single polypeptide. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:1402-1416. [PMID: 38163285 PMCID: PMC11022806 DOI: 10.1111/pbi.14275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/23/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
Immunoglobulin G (IgG)-based fusion proteins have been widely exploited as a potential vaccine delivery platform but in the absence of exogenous adjuvants, the lack of robust immunity remains an obstacle. Here, we report on a key modification that overcomes that obstacle. Thus, we constructed an IgG-Fc vaccine platform for dengue, termed D-PCF, which in addition to a dengue antigen incorporates the cholera toxin non-toxic B subunit (CTB) as a molecular adjuvant, with all three proteins expressed as a single polypeptide. Following expression in Nicotiana benthamiana plants, the D-PCF assembled as polymeric structures of similar size to human IgM, a process driven by the pentamerization of CTB. A marked improvement of functional properties in vitro and immunogenicity in vivo over a previous iteration of the Fc-fusion protein without CTB [1] was demonstrated. These include enhanced antigen presenting cell binding, internalization and activation, complement activation, epithelial cell interactions and ganglioside binding, as well as more efficient polymerization within the expression host. Following immunization of mice with D-PCF by a combination of systemic and mucosal (intranasal) routes, we observed robust systemic and mucosal immune responses, as well as systemic T cell responses, significantly higher than those induced by a related Fc-fusion protein but without CTB. The induced antibodies could bind to the domain III of the dengue virus envelope protein from all four dengue serotypes. Finally, we also demonstrated feasibility of aerosolization of D-PCF as a prerequisite for vaccine delivery by the respiratory route.
Collapse
Affiliation(s)
- Mi‐Young Kim
- Department of Molecular BiologyJeonbuk National UniversityJeonjuRepublic of Korea
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Emil Vergara
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Andy Tran
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Matthew John Paul
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | | | - Julian K.C. Ma
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Yong‐Suk Jang
- Department of Molecular BiologyJeonbuk National UniversityJeonjuRepublic of Korea
| | - Rajko Reljic
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| |
Collapse
|
4
|
Akter R, Tasneem F, Das S, Soma MA, Georgakopoulos-Soares I, Juthi RT, Sazed SA. Approaches of dengue control: vaccine strategies and future aspects. Front Immunol 2024; 15:1362780. [PMID: 38487527 PMCID: PMC10937410 DOI: 10.3389/fimmu.2024.1362780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/08/2024] [Indexed: 03/17/2024] Open
Abstract
Dengue, caused by the dengue virus (DENV), affects millions of people worldwide every year. This virus has two distinct life cycles, one in the human and another in the mosquito, and both cycles are crucial to be controlled. To control the vector of DENV, the mosquito Aedes aegypti, scientists employed many techniques, which were later proved ineffective and harmful in many ways. Consequently, the attention shifted to the development of a vaccine; researchers have targeted the E protein, a surface protein of the virus and the NS1 protein, an extracellular protein. There are several types of vaccines developed so far, such as live attenuated vaccines, recombinant subunit vaccines, inactivated virus vaccines, viral vectored vaccines, DNA vaccines, and mRNA vaccines. Along with these, scientists are exploring new strategies of developing improved version of the vaccine by employing recombinant DNA plasmid against NS1 and also aiming to prevent the infection by blocking the DENV life cycle inside the mosquitoes. Here, we discussed the aspects of research in the field of vaccines until now and identified some prospects for future vaccine developments.
Collapse
Affiliation(s)
- Runa Akter
- Department of Pharmacy, Independent University Bangladesh, Dhaka, Bangladesh
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Faria Tasneem
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Shuvo Das
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | | | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rifat Tasnim Juthi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Saiful Arefeen Sazed
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
5
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
6
|
Xu Q, Ma F, Yang D, Li Q, Yan L, Ou J, Zhang L, Liu Y, Zhan Q, Li R, Wei Q, Hu H, Wang Y, Li X, Zhang S, Yang J, Chai S, Du Y, Wang L, Zhang E, Zhang G. Rice-produced classical swine fever virus glycoprotein E2 with herringbone-dimer design to enhance immune responses. PLANT BIOTECHNOLOGY JOURNAL 2023; 21:2546-2559. [PMID: 37572354 PMCID: PMC10651154 DOI: 10.1111/pbi.14152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 06/15/2023] [Accepted: 07/25/2023] [Indexed: 08/14/2023]
Abstract
Pestiviruses, including classical swine fever virus, remain a concern for global animal health and are responsible for major economic losses of livestock worldwide. Despite high levels of vaccination, currently available commercial vaccines are limited by safety concerns, moderate efficacy, and required high doses. The development of new vaccines is therefore essential. Vaccine efforts should focus on optimizing antigen presentation to enhance immune responses. Here, we describe a simple herringbone-dimer strategy for efficient vaccine design, using the classical swine fever virus E2 expressed in a rice endosperm as an example. The expression of rE2 protein was identified, with the rE2 antigen accumulating to 480 mg/kg. Immunological assays in mice, rabbits, and pigs showed high antigenicity of rE2. Two immunizations with 284 ng of the rE2 vaccine or one shot with 5.12 μg provided effective protection in pigs without interference from pre-existing antibodies. Crystal structure and small-angle X-ray scattering results confirmed the stable herringbone dimeric conformation, which had two fully exposed duplex receptor binding domains. Our results demonstrated that rice endosperm is a promising platform for precise vaccine design, and this strategy can be universally applied to other Flaviviridae virus vaccines.
Collapse
Affiliation(s)
- Qianru Xu
- School of Basic Medical SciencesHenan UniversityKaifengChina
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Fanshu Ma
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
- CAS Key Laboratory of Nano‐Bio Interface, Suzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhouChina
| | - Daichang Yang
- College of Life ScienceWuhan UniversityWuhanChina
- Wuhan Healthgen Biotechnology Corp.WuhanChina
| | - Qingmei Li
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Liming Yan
- Laboratory of Structural Biology, School of MedicineTsinghua UniversityBeijingChina
| | - Jiquan Ou
- Wuhan Healthgen Biotechnology Corp.WuhanChina
| | - Longxian Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
- Longhu LaboratoryZhengzhouChina
| | - Yunchao Liu
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Quan Zhan
- Wuhan Healthgen Biotechnology Corp.WuhanChina
| | - Rui Li
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Qiang Wei
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Hui Hu
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
| | - Yanan Wang
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Xueyang Li
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
| | - Shenli Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
| | - Jifei Yang
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Shujun Chai
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Yongkun Du
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
| | - Li Wang
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
| | - Erqin Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
- Longhu LaboratoryZhengzhouChina
| | - Gaiping Zhang
- International Joint Research Center of National Animal Immunology, College of Veterinary MedicineHenan Agriculture UniversityZhengzhouChina
- Key Laboratory of Animal ImmunologyHenan Academy of Agricultural SciencesZhengzhouChina
- Longhu LaboratoryZhengzhouChina
- School of Advanced Agricultural SciencesPeking UniversityBeijingChina
| |
Collapse
|
7
|
Jain S, Vimal N, Angmo N, Sengupta M, Thangaraj S. Dengue Vaccination: Towards a New Dawn of Curbing Dengue Infection. Immunol Invest 2023; 52:1096-1149. [PMID: 37962036 DOI: 10.1080/08820139.2023.2280698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dengue is an infectious disease caused by dengue virus (DENV) and is a serious global burden. Antibody-dependent enhancement and the ability of DENV to infect immune cells, along with other factors, lead to fatal Dengue Haemorrhagic Fever and Dengue Shock Syndrome. This necessitates the development of a robust and efficient vaccine but vaccine development faces a number of hurdles. In this review, we look at the epidemiology, genome structure and cellular targets of DENV and elaborate upon the immune responses generated by human immune system against DENV infection. The review further sheds light on various challenges in development of a potent vaccine against DENV which is followed by presenting a current account of different vaccines which are being developed or have been licensed.
Collapse
Affiliation(s)
- Sidhant Jain
- Independent Researcher, Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, India
| | - Neha Vimal
- Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi, India
| | - Nilza Angmo
- Maitreyi College, University of Delhi, Delhi, India
| | - Madhumita Sengupta
- Janki Devi Bajaj Government Girls College, University of Kota, Kota, India
| | - Suraj Thangaraj
- Swami Ramanand Teerth Rural Government Medical College, Maharashtra University of Health Sciences, Ambajogai, India
| |
Collapse
|
8
|
Diamos AG, Pardhe MD, Bergeman MH, Kamzina AS, DiPalma MP, Aman S, Chaves A, Lowe K, Kilbourne J, Hogue IB, Mason HS. A self-binding immune complex vaccine elicits strong neutralizing responses against herpes simplex virus in mice. Front Immunol 2023; 14:1085911. [PMID: 37205110 PMCID: PMC10186352 DOI: 10.3389/fimmu.2023.1085911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/18/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction It has been known for over half a century that mixing an antigen with its cognate antibody in an immune complex (IC) can enhance antigen immunogenicity. However, many ICs produce inconsistent immune responses, and the use of ICs in the development new vaccines has been limited despite the otherwise widespread success of antibody-based therapeutics. To address this problem, we designed a self-binding recombinant immune complex (RIC) vaccine which mimics the larger ICs generated during natural infection. Materials and methods In this study, we created two novel vaccine candidates: 1) a traditional IC targeting herpes simplex virus 2 (HSV-2) by mixing glycoprotein D (gD) with a neutralizing antibody (gD-IC); and 2) an RIC consisting of gD fused to an immunoglobulin heavy chain and then tagged with its own binding site, allowing self-binding (gD-RIC). We characterized the complex size and immune receptor binding characteristics in vitro for each preparation. Then, the in vivo immunogenicity and virus neutralization of each vaccine were compared in mice. Results gD-RIC formed larger complexes which enhanced C1q receptor binding 25-fold compared to gD-IC. After immunization of mice, gD-RIC elicited up to 1,000-fold higher gD-specific antibody titers compared to traditional IC, reaching endpoint titers of 1:500,000 after two doses without adjuvant. The RIC construct also elicited stronger virus-specific neutralization against HSV-2, as well as stronger cross-neutralization against HSV-1, although the proportion of neutralizing antibodies to total antibodies was somewhat reduced in the RIC group. Discussion This work demonstrates that the RIC system overcomes many of the pitfalls of traditional IC, providing potent immune responses against HSV-2 gD. Based on these findings, further improvements to the RIC system are discussed. RIC have now been shown to be capable of inducing potent immune responses to a variety of viral antigens, underscoring their broad potential as a vaccine platform.
Collapse
Affiliation(s)
- Andrew G. Diamos
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | | | | | | | | | | | | | | | - Ian B. Hogue
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Hugh S. Mason
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
9
|
Improving Protein Quantity and Quality—The Next Level of Plant Molecular Farming. Int J Mol Sci 2022; 23:ijms23031326. [PMID: 35163249 PMCID: PMC8836236 DOI: 10.3390/ijms23031326] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/15/2022] Open
Abstract
Plants offer several unique advantages in the production of recombinant pharmaceuticals for humans and animals. Although numerous recombinant proteins have been expressed in plants, only a small fraction have been successfully put into use. The hugely distinct expression systems between plant and animal cells frequently cause insufficient yield of the recombinant proteins with poor or undesired activity. To overcome the issues that greatly constrain the development of plant-produced pharmaceuticals, great efforts have been made to improve expression systems and develop alternative strategies to increase both the quantity and quality of the recombinant proteins. Recent technological revolutions, such as targeted genome editing, deconstructed vectors, virus-like particles, and humanized glycosylation, have led to great advances in plant molecular farming to meet the industrial manufacturing and clinical application standards. In this review, we discuss the technological advances made in various plant expression platforms, with special focus on the upstream designs and milestone achievements in improving the yield and glycosylation of the plant-produced pharmaceutical proteins.
Collapse
|
10
|
Izadi S, Jalali Javaran M, Rashidi Monfared S, Castilho A. Reteplase Fc-fusions produced in N. benthamiana are able to dissolve blood clots ex vivo. PLoS One 2021; 16:e0260796. [PMID: 34847186 PMCID: PMC8631678 DOI: 10.1371/journal.pone.0260796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/16/2021] [Indexed: 11/19/2022] Open
Abstract
Thrombolytic and fibrinolytic therapies are effective treatments to dissolve blood clots in stroke therapy. Thrombolytic drugs activate plasminogen to its cleaved form plasmin, a proteolytic enzyme that breaks the crosslinks between fibrin molecules. The FDA-approved human tissue plasminogen activator Reteplase (rPA) is a non-glycosylated protein produced in E. coli. rPA is a deletion mutant of the wild-type Alteplase that benefits from an extended plasma half-life, reduced fibrin specificity and the ability to better penetrate into blood clots. Different methods have been proposed to improve the production of rPA. Here we show for the first time the transient expression in Nicotiana benthamiana of rPA fused to the immunoglobulin fragment crystallizable (Fc) domain on an IgG1, a strategy commonly used to improve the stability of therapeutic proteins. Despite our success on the expression and purification of dimeric rPA-Fc fusions, protein instability results in high amounts of Fc-derived degradation products. We hypothesize that the "Y"- shape of dimeric Fc fusions cause steric hindrance between protein domains and leads to physical instability. Indeed, mutations of critical residues in the Fc dimerization interface allowed the expression of fully stable rPA monomeric Fc-fusions. The ability of rPA-Fc to convert plasminogen into plasmin was demonstrated by plasminogen zymography and clot lysis assay shows that rPA-Fc is able to dissolve blood clots ex vivo. Finally, we addressed concerns with the plant-specific glycosylation by modulating rPA-Fc glycosylation towards serum-like structures including α2,6-sialylated and α1,6-core fucosylated N-glycans completely devoid of plant core fucose and xylose residues.
Collapse
Affiliation(s)
- Shiva Izadi
- Department of Applied Genetics and Cell Biology, Natural Resources and Life Sciences, Vienna, Austria
- Faculty of Agriculture, Department of Plant Genetics and Breeding, Tarbiat Modares University, Tehran, Iran
| | - Mokhtar Jalali Javaran
- Faculty of Agriculture, Department of Agricultural Biotechnology, Tarbiat Modares University, Tehran, Iran
| | - Sajad Rashidi Monfared
- Faculty of Agriculture, Department of Agricultural Biotechnology, Tarbiat Modares University, Tehran, Iran
| | - Alexandra Castilho
- Department of Applied Genetics and Cell Biology, Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
11
|
He W, Baysal C, Lobato Gómez M, Huang X, Alvarez D, Zhu C, Armario‐Najera V, Blanco Perera A, Cerda Bennaser P, Saba‐Mayoral A, Sobrino‐Mengual G, Vargheese A, Abranches R, Alexandra Abreu I, Balamurugan S, Bock R, Buyel JF, da Cunha NB, Daniell H, Faller R, Folgado A, Gowtham I, Häkkinen ST, Kumar S, Sathish Kumar R, Lacorte C, Lomonossoff GP, Luís IM, K.‐C. Ma J, McDonald KA, Murad A, Nandi S, O’Keef B, Parthiban S, Paul MJ, Ponndorf D, Rech E, Rodrigues JC, Ruf S, Schillberg S, Schwestka J, Shah PS, Singh R, Stoger E, Twyman RM, Varghese IP, Vianna GR, Webster G, Wilbers RHP, Christou P, Oksman‐Caldentey K, Capell T. Contributions of the international plant science community to the fight against infectious diseases in humans-part 2: Affordable drugs in edible plants for endemic and re-emerging diseases. PLANT BIOTECHNOLOGY JOURNAL 2021; 19:1921-1936. [PMID: 34181810 PMCID: PMC8486237 DOI: 10.1111/pbi.13658] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 05/05/2023]
Abstract
The fight against infectious diseases often focuses on epidemics and pandemics, which demand urgent resources and command attention from the health authorities and media. However, the vast majority of deaths caused by infectious diseases occur in endemic zones, particularly in developing countries, placing a disproportionate burden on underfunded health systems and often requiring international interventions. The provision of vaccines and other biologics is hampered not only by the high cost and limited scalability of traditional manufacturing platforms based on microbial and animal cells, but also by challenges caused by distribution and storage, particularly in regions without a complete cold chain. In this review article, we consider the potential of molecular farming to address the challenges of endemic and re-emerging diseases, focusing on edible plants for the development of oral drugs. Key recent developments in this field include successful clinical trials based on orally delivered dried leaves of Artemisia annua against malarial parasite strains resistant to artemisinin combination therapy, the ability to produce clinical-grade protein drugs in leaves to treat infectious diseases and the long-term storage of protein drugs in dried leaves at ambient temperatures. Recent FDA approval of the first orally delivered protein drug encapsulated in plant cells to treat peanut allergy has opened the door for the development of affordable oral drugs that can be manufactured and distributed in remote areas without cold storage infrastructure and that eliminate the need for expensive purification steps and sterile delivery by injection.
Collapse
Affiliation(s)
- Wenshu He
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Can Baysal
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Maria Lobato Gómez
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Xin Huang
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Derry Alvarez
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Changfu Zhu
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Victoria Armario‐Najera
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Aamaya Blanco Perera
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Pedro Cerda Bennaser
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Andrea Saba‐Mayoral
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | | | - Ashwin Vargheese
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Rita Abranches
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Isabel Alexandra Abreu
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Shanmugaraj Balamurugan
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Ralph Bock
- Max Planck Institute of Molecular Plant PhysiologyPotsdam‐GolmGermany
| | - Johannes F. Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachenGermany
- Institute for Molecular BiotechnologyRWTH Aachen UniversityAachenGermany
| | - Nicolau B. da Cunha
- Centro de Análise Proteômicas e Bioquímicas de BrasíliaUniversidade Católica de BrasíliaBrasíliaBrazil
| | - Henry Daniell
- School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Roland Faller
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
| | - André Folgado
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Iyappan Gowtham
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Suvi T. Häkkinen
- Industrial Biotechnology and Food SolutionsVTT Technical Research Centre of Finland LtdEspooFinland
| | - Shashi Kumar
- International Centre for Genetic Engineering and BiotechnologyNew DelhiIndia
| | - Ramalingam Sathish Kumar
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Cristiano Lacorte
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | | | - Ines M. Luís
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Julian K.‐C. Ma
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Karen A. McDonald
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Global HealthShare InitiativeUniversity of California, DavisDavisCAUSA
| | - Andre Murad
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | - Somen Nandi
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Global HealthShare InitiativeUniversity of California, DavisDavisCAUSA
| | - Barry O’Keef
- Division of Cancer Treatment and DiagnosisMolecular Targets ProgramCenter for Cancer ResearchNational Cancer Institute, and Natural Products Branch, Developmental Therapeutics ProgramNational Cancer Institute, NIHFrederickMDUSA
| | - Subramanian Parthiban
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Mathew J. Paul
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Daniel Ponndorf
- Department of Biological ChemistryJohn Innes CentreNorwich Research Park, NorwichUK
| | - Elibio Rech
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | - Julio C.M. Rodrigues
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | - Stephanie Ruf
- Max Planck Institute of Molecular Plant PhysiologyPotsdam‐GolmGermany
| | - Stefan Schillberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachenGermany
- Institute for PhytopathologyJustus‐Liebig‐University GiessenGiessenGermany
| | - Jennifer Schwestka
- Institute of Plant Biotechnology and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Priya S. Shah
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Department of Microbiology and Molecular GeneticsUniversity of California, DavisDavisCAUSA
| | - Rahul Singh
- School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eva Stoger
- Institute of Plant Biotechnology and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | | | - Inchakalody P. Varghese
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Giovanni R. Vianna
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | - Gina Webster
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Ruud H. P. Wilbers
- Laboratory of NematologyPlant Sciences GroupWageningen University and ResearchWageningenThe Netherlands
| | - Paul Christou
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
- ICREACatalan Institute for Research and Advanced StudiesBarcelonaSpain
| | | | - Teresa Capell
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| |
Collapse
|
12
|
Abstract
Introduction: Dengue virus is a global health threat, with approximately 390 million dengue infections annually. Efficient vaccines for dengue prevention are currently lacking. This review aims to summarize the current progress in dengue vaccine development.Area covered: This article discusses recent dengue vaccine developments based on the published literature and ClinicalTrials.gov website up to December 2020.Expert opinion: The first live-attenuated chimeric yellow-fever/tetravalent dengue vaccine (CYD-TDV), Dengvaxia, has been licensed in several countries. However, the low efficacy of this vaccine was observed in children and dengue-naïve individuals. It also increased the risk of severe dengue in people who had not been exposed to dengue. The heterologous prime-boost regimen of sequential immunization with DENVax and Dengvaxia covers four serotypes of immunogenicity, eliminating the effect of ADE. Moreover, a heterologous prime-boost regimen that combines inactivated vaccines with alum and live attenuated vaccines might increase the immunogenic response. The lack of an ideal animal model is an obstacle to the development of dengue vaccines, and the macaque model may be considered for similar immunologic responses in humans.
Collapse
Affiliation(s)
- Chung-Hao Huang
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Infection Control Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Te Tsai
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Seng-Fan Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Hung Wang
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Hsu Chen
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan.,College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
13
|
So KK, Chun J, Luong NN, Seo HW, Kim DH. Expression of an immunocomplex consisting of Fc fragment fused with a consensus dengue envelope domain III in Saccharomyces cerevisiae. Biotechnol Lett 2021; 43:1895-1904. [PMID: 34245387 PMCID: PMC8272446 DOI: 10.1007/s10529-021-03161-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 06/28/2021] [Indexed: 11/25/2022]
Abstract
Objectives To explore Saccharomyces cerevisiae as an expression platform for dengue oral immune complex vaccine development. Results Molecular engineering was applied to create a fusion gene construct (scEDIII-PIGS) consisting of a yeast codon optimized sequence encoding for a synthetic consensus dengue envelope domain III (scEDIII) followed by a modified IgG Fc domain (PIGS). Northern blot showed transcription of the target gene, with a temporal expression pattern similar to those from previous work. Western blot showed assembly of various immune complexes from monomer to hexamer. Partial purification of scEDIII-PIGS was also attempted to demonstrate the feasibility of yeast system for immune complex vaccine development. Approximately 1 mg of scEDIII-PIGS can be produced from 1 l culture. Conclusion This work demonstrated for the first time that various immunocomplex structures of our target protein could be efficiently produced in S. cerevisiae for future application in developing oral and injectable vaccines against various pathogens. Supplementary Information The online version contains supplementary material available at 10.1007/s10529-021-03161-7.
Collapse
Affiliation(s)
- Kum-Kang So
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Jeonbuk National University, Jeollabuk-do, Jeonju, 54896, Republic of Korea
| | - Jeesun Chun
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Jeonbuk National University, Jeollabuk-do, Jeonju, 54896, Republic of Korea
| | - Nguyen Ngoc Luong
- Department of Biology, College of Sciences, Hue University, Hue, Vietnam
| | - Hee-Won Seo
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Jeonbuk National University, Jeollabuk-do, Jeonju, 54896, Republic of Korea
| | - Dae-Hyuk Kim
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Jeonbuk National University, Jeollabuk-do, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
14
|
Tien NQD, Yang MS, Jang YS, Kwon TH, Reljic R, Kim MY. Systemic and Oral Immunogenicity of Porcine Epidemic Diarrhea Virus Antigen Fused to Poly-Fc of Immunoglobulin G and Expressed in ΔXT/FT Nicotiana benthamiana Plants. Front Pharmacol 2021; 12:653064. [PMID: 33995068 PMCID: PMC8120289 DOI: 10.3389/fphar.2021.653064] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/16/2021] [Indexed: 11/28/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), a member of the Coronaviridae family has become increasingly probelmatic in the pig farming industry. Currently, there are no effective, globally applicable vaccines against PEDV. Here, we tested a recombinant PEDV vaccine candidate based on the expression of the core neutralising epitope (COE) of PEDV conjugated to polymeric immunoglobulin G scaffold (PIGS) in glycoengineered Nicotiana benthamiana plants. The biological activity of COE-PIGS was demonstrated by binding to C1q component of the complement system, as well as the surface of antigen-presenting cells (APCs) in vitro. The recombinant COE-PIGS induced humoral and cellular immune responses specific for PEDV after both systemic and mucosal vaccination. Altogether, the data indicated that PEDV antigen fusion to poly-Fc could be a promising vaccine platform against respiratory PEDV infection.
Collapse
Affiliation(s)
- Nguyen-Quang-Duc Tien
- Department of Molecular Biology, Jeonbuk National University, Jeonju-Si, South Korea.,University of Sciences, Hue University, Hue City, Viet Nam
| | - Moon-Sik Yang
- Department of Molecular Biology, Jeonbuk National University, Jeonju-Si, South Korea.,NBM Inc., Wanjusandan 5-ro, Bongdong-eup, Wanju-Gun, Jeollabuk-do, South Korea
| | - Yong-Suk Jang
- Department of Molecular Biology, Jeonbuk National University, Jeonju-Si, South Korea
| | - Tae-Ho Kwon
- NBM Inc., Wanjusandan 5-ro, Bongdong-eup, Wanju-Gun, Jeollabuk-do, South Korea
| | - Rajko Reljic
- Institute for Infection and Immunity, St George's University of London, London, UK
| | - Mi-Young Kim
- Department of Molecular Biology, Jeonbuk National University, Jeonju-Si, South Korea.,Institute for Infection and Immunity, St George's University of London, London, UK
| |
Collapse
|
15
|
Dendritic Cell Tumor Vaccination via Fc Gamma Receptor Targeting: Lessons Learned from Pre-Clinical and Translational Studies. Vaccines (Basel) 2021; 9:vaccines9040409. [PMID: 33924183 PMCID: PMC8074394 DOI: 10.3390/vaccines9040409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
Despite significant recent improvements in the field of immunotherapy, cancer remains a heavy burden on patients and healthcare systems. In recent years, immunotherapies have led to remarkable strides in treating certain cancers. However, despite the success of checkpoint inhibitors and the advent of cellular therapies, novel strategies need to be explored to (1) improve treatment in patients where these approaches fail and (2) make such treatments widely and financially accessible. Vaccines based on tumor antigens (Ag) have emerged as an innovative strategy with the potential to address these areas. Here, we review the fundamental aspects relevant for the development of cancer vaccines and the critical role of dendritic cells (DCs) in this process. We first offer a general overview of DC biology and routes of Ag presentation eliciting effective T cell-mediated immune responses. We then present new therapeutic avenues specifically targeting Fc gamma receptors (FcγR) as a means to deliver antigen selectively to DCs and its effects on T-cell activation. We present an overview of the mechanistic aspects of FcγR-mediated DC targeting, as well as potential tumor vaccination strategies based on preclinical and translational studies. In particular, we highlight recent developments in the field of recombinant immune complex-like large molecules and their potential for DC-mediated tumor vaccination in the clinic. These findings go beyond cancer research and may be of relevance for other disease areas that could benefit from FcγR-targeted antigen delivery, such as autoimmunity and infectious diseases.
Collapse
|
16
|
Gattinger P, Izadi S, Grünwald-Gruber C, Kallolimath S, Castilho A. The Instability of Dimeric Fc-Fusions Expressed in Plants Can Be Solved by Monomeric Fc Technology. FRONTIERS IN PLANT SCIENCE 2021; 12:671728. [PMID: 34305971 PMCID: PMC8299721 DOI: 10.3389/fpls.2021.671728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/18/2021] [Indexed: 05/18/2023]
Abstract
The potential therapeutic value of many proteins is ultimately limited by their rapid in vivo clearance. One strategy to limit clearance by metabolism and excretion, and improving the stability of therapeutic proteins, is their fusion to the immunoglobulin fragment crystallizable region (Fc). The Fc region plays multiple roles in (i) dimerization for the formation of "Y"-shaped structure of Ig, (ii) Fc-mediated effector functions, (iii) extension of serum half-life, and (iv) a cost-effective purification tag. Plants and in particular Nicotiana benthamiana have proven to be suitable expression platforms for several recombinant therapeutic proteins. Despite the enormous success of their use for the production of full-length monoclonal antibodies, the expression of Fc-fused therapeutic proteins in plants has shown limitations. Many Fc-fusion proteins expressed in plants show different degrees of instability resulting in high amounts of Fc-derived degradation products. To address this issue, we used erythropoietin (EPO) as a reporter protein and evaluated the efforts to enhance the expression of full-length EPO-Fc targeted to the apoplast of N. benthamiana. Our results show that the instability of the fusion protein is independent from the Fc origin or IgG subclass and from the peptide sequence used to link the two domains. We also show that a similar instability occurs upon the expression of individual heavy chains of monoclonal antibodies and ScFv-Fc that mimic the "Y"-shape of antibodies but lack the light chain. We propose that in this configuration, steric hindrance between the protein domains leads to physical instability. Indeed, mutations of critical residues located on the Fc dimerization interface allowed the expression of fully stable EPO monomeric Fc-fusion proteins. We discuss the limitations of Fc-fusion technology in N. benthamiana transient expression systems and suggest strategies to optimize the Fc-based scaffolds on their folding and aggregation resistance in order to improve the stability.
Collapse
Affiliation(s)
- Pia Gattinger
- Department of Applied Genetics and Cell Biology, Institute for Plant Biotechnology and Cell Biology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Shiva Izadi
- Department of Applied Genetics and Cell Biology, Institute for Plant Biotechnology and Cell Biology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
- Department of Plant Genetics and Breeding, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Clemens Grünwald-Gruber
- Division of Biochemistry, Department of Chemistry, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Somanath Kallolimath
- Department of Applied Genetics and Cell Biology, Institute for Plant Biotechnology and Cell Biology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Alexandra Castilho
- Department of Applied Genetics and Cell Biology, Institute for Plant Biotechnology and Cell Biology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
- *Correspondence: Alexandra Castilho,
| |
Collapse
|
17
|
Zaheer T, Pal K, Zaheer I. Topical review on nano-vaccinology: Biochemical promises and key challenges. Process Biochem 2021; 100:237-244. [PMID: 33013180 PMCID: PMC7521878 DOI: 10.1016/j.procbio.2020.09.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022]
Abstract
Nanomaterials have wide-ranging biomedical applications in prevention, treatment and control of diseases. Nanoparticle based vaccines have proven prodigious prophylaxis of various infectious and non-infectious diseases of human and animal concern. Nano-vaccines outnumber the conventional vaccines by virtue of plasticity in physio-chemical properties and ease of administration. The efficacy of nano-based vaccines may be attributed to the improved antigen stability, minimum immuno-toxicity, sustained release, enhanced immunogenicity and the flexibility of physical features of nanoparticles. Based on these, the nano-based vaccines have potential to evoke both cellular and humoral immune responses. Targeted and highly specific immunological pathways required for solid and long lasting immunity may be achieved with specially engineered nano-vaccines. This review presents an insight into the prevention of infectious diseases (of bacterial, viral and parasitic origin) and non-infectious diseases (cancer, auto-immune diseases) using nano-vaccinology. Additionally, key challenges to the effective utilization of nano-vaccines from bench to clinical settings have been highlighted as research domains for future.
Collapse
Key Words
- CAPN, calcium-phosphate nanoparticles
- CNT, carbon nanotube
- COVID-19, Corona virus disease-2019
- Chi-Alg, chitosan alginate
- HIV, human immune deficiency virus
- HPV, human papilloma virus
- ISCOMS, immune stimulating complexes
- IgA, immunoglobulin A
- Immunity
- MERS, Middle-East respiratory syndrome
- MRSA, methcillin resistant Staphylococcus aureus
- NMVs, nano multilamellar lipid vesicles
- Nanoparticles
- PLGA, poly(lactic-co-glycolic acid)
- PSNP, polystyrene nanoparticles
- Pathogens
- Prevention
- SAPN, Self-Assembling Protein Nanoparticle
- SARS-CoV-1, severe acute respiratory syndrome Coronavirus-1
- VLP, virus like particles
- Vaccine
Collapse
Affiliation(s)
- Tean Zaheer
- Department of Parasitology, University of Agriculture, Faisalabad, Faisalabad 38040, Pakistan
| | - Kaushik Pal
- Federal University of Rio de Janeiro, Cidade Universitária, Rio de Janeiro RJ, 21941-901, Brazil
- Wuhan University, 8 East Lake South Road, Wuchang 430072, Hubei Province, China
| | - Iqra Zaheer
- Department of Pathology, University of Agriculture, Faisalabad, Faisalabad 38040, Pakistan
| |
Collapse
|
18
|
Deng SQ, Yang X, Wei Y, Chen JT, Wang XJ, Peng HJ. A Review on Dengue Vaccine Development. Vaccines (Basel) 2020; 8:E63. [PMID: 32024238 PMCID: PMC7159032 DOI: 10.3390/vaccines8010063] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Dengue virus (DENV) has become a global health threat with about half of the world's population at risk of infection. Although the disease caused by DENV is self-limiting in the first infection, the antibody-dependent enhancement (ADE) effect increases the mortality in the second infection with a heterotypic virus. Since there is no specific efficient medicine in treatment, it is urgent to develop vaccines to prevent infection and disease progression. Currently, only a live attenuated vaccine, chimeric yellow fever 17D-tetravalent dengue vaccine (CYD-TDV), has been licensed for clinical use in some countries, and many candidate vaccines are still under research and development. This review discusses the progress, strengths, and weaknesses of the five types of vaccines including live attenuated vaccine, inactivated virus vaccine, recombinant subunit vaccine, viral vectored vaccine, and DNA vaccine.
Collapse
Affiliation(s)
- Sheng-Qun Deng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.-Q.D.); (X.Y.); (Y.W.); (J.-T.C.)
| | - Xian Yang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.-Q.D.); (X.Y.); (Y.W.); (J.-T.C.)
| | - Yong Wei
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.-Q.D.); (X.Y.); (Y.W.); (J.-T.C.)
| | - Jia-Ting Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.-Q.D.); (X.Y.); (Y.W.); (J.-T.C.)
| | - Xiao-Jun Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Medical University, Dongguan 523808, China;
| | - Hong-Juan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.-Q.D.); (X.Y.); (Y.W.); (J.-T.C.)
| |
Collapse
|
19
|
Murad S, Fuller S, Menary J, Moore C, Pinneh E, Szeto T, Hitzeroth I, Freire M, Taychakhoonavudh S, Phoolcharoen W, Ma JKC. Molecular Pharming for low and middle income countries. Curr Opin Biotechnol 2019; 61:53-59. [PMID: 31751895 DOI: 10.1016/j.copbio.2019.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/14/2019] [Indexed: 01/02/2023]
Abstract
Interest in applications and benefits that Molecular Pharming might offer to Low and Middle Income Countries has always been a potent driver for the research discipline, and a major reason why many scientists entered the field. Although enthusiasm remains high, the reality is that such a game-changing innovation would always take longer than traditional uptake of new technology in developed countries, and be complicated by external factors beyond technical feasibility. Excitingly, signs of increasing interest by LMICS in Molecular Pharming are now emerging. Here, three case studies from Thailand, South Africa and Brazil are used to identify some of the key issues when a new investment into Molecular Pharming manufacturing capacity is under consideration. At present, academic research is not necessarily addressing these issues. Only by understanding the concerns, can members of the academic community contribute to helping the development of Molecular Pharming for LMICs by focusing their research efforts appropriately.
Collapse
Affiliation(s)
- Sheeba Murad
- Institute for Infection and Immunity, St. George's University of London, SW18 0RE, London, UK
| | - Sebastian Fuller
- Institute for Infection and Immunity, St. George's University of London, SW18 0RE, London, UK
| | - Jonathan Menary
- Institute for Infection and Immunity, St. George's University of London, SW18 0RE, London, UK
| | - Cathy Moore
- Institute for Infection and Immunity, St. George's University of London, SW18 0RE, London, UK
| | - Elizabeth Pinneh
- Institute for Infection and Immunity, St. George's University of London, SW18 0RE, London, UK
| | - Tim Szeto
- Institute for Infection and Immunity, St. George's University of London, SW18 0RE, London, UK
| | - Inga Hitzeroth
- Biopharming Research Unit, Molecular & Cell Biology Department, University of Cape Town, Cape Town, South Africa
| | - Marcos Freire
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Suthira Taychakhoonavudh
- Department of Social and Administrative Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Waranyoo Phoolcharoen
- Research Unit for Plant-produced Pharmaceuticals, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Julian K-C Ma
- Institute for Infection and Immunity, St. George's University of London, SW18 0RE, London, UK.
| |
Collapse
|
20
|
Schiavinato M, Strasser R, Mach L, Dohm JC, Himmelbauer H. Genome and transcriptome characterization of the glycoengineered Nicotiana benthamiana line ΔXT/FT. BMC Genomics 2019; 20:594. [PMID: 31324144 PMCID: PMC6642603 DOI: 10.1186/s12864-019-5960-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/08/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The allotetraploid tobacco species Nicotiana benthamiana native to Australia has become a popular host for recombinant protein production. Although its usage grows every year, little is known on this plant's genomic and transcriptomic features. Most N. benthamiana accessions currently used in research lack proper documentation of their breeding history and provenance. One of these, the glycoengineered N. benthamiana line ΔXT/FT is increasingly used for the production of biopharmaceutical proteins. RESULTS Based on an existing draft assembly of the N. benthamiana genome we predict 50,516 protein -encoding genes (62,216 transcripts) supported by expression data derived from 2.35 billion mRNA-seq reads. Using single-copy core genes we show high completeness of the predicted gene set. We functionally annotate more than two thirds of the gene set through sequence homology to genes from other Nicotiana species. We demonstrate that the expression profiles from leaf tissue of ΔXT/FT and its wild type progenitor only show minimal differences. We identify the transgene insertion sites in ΔXT/FT and show that one of the transgenes was inserted inside another predicted gene that most likely lost its function upon insertion. Based on publicly available mRNA-seq data, we confirm that the N. benthamiana accessions used by different research institutions most likely derive from a single source. CONCLUSIONS This work provides gene annotation of the N. benthamiana genome, a genomic and transcriptomic characterization of a transgenic N. benthamiana line in comparison to its wild-type progenitor, and sheds light onto the relatedness of N. benthamiana accessions that are used in laboratories around the world.
Collapse
Affiliation(s)
- Matteo Schiavinato
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Richard Strasser
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Lukas Mach
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Juliane C. Dohm
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Heinz Himmelbauer
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
21
|
Pang EL, Peyret H, Ramirez A, Loh HS, Lai KS, Fang CM, Rosenberg WM, Lomonossoff GP. Epitope Presentation of Dengue Viral Envelope Glycoprotein Domain III on Hepatitis B Core Protein Virus-Like Particles Produced in Nicotiana benthamiana. FRONTIERS IN PLANT SCIENCE 2019; 10:455. [PMID: 31057572 PMCID: PMC6477658 DOI: 10.3389/fpls.2019.00455] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/26/2019] [Indexed: 05/07/2023]
Abstract
Dengue fever is currently ranked as the top emerging tropical disease, driven by increased global travel, urbanization, and poor hygiene conditions as well as global warming effects which facilitate the spread of Aedes mosquitoes beyond their current distribution. Today, more than 100 countries are affected most of which are tropical Asian and Latin American nations with limited access to medical care. Hence, the development of a dengue vaccine that is dually cost-effective and able to confer a comprehensive protection is ultimately needed. In this study, a consensus sequence of the antigenic dengue viral glycoprotein domain III (cEDIII) was used aiming to provide comprehensive coverage against all four circulating dengue viral serotypes and potential clade replacement event. Utilizing hepatitis B tandem core technology, the cEDIII sequence was inserted into the immunodominant c/e1 loop region so that it could be displayed on the spike structures of assembled particles. The tandem core particles displaying cEDIII epitopes (tHBcAg-cEDIII) were successfully produced in Nicotiana benthamiana via Agrobacterium-mediated transient expression strategy to give a protein of ∼54 kDa, detected in both soluble and insoluble fractions of plant extracts. The assembled tHBcAg-cEDIII virus-like particles (VLPs) were also visualized from transmission electron microscopy. These VLPs had diameters that range from 32 to 35 nm, presenting an apparent size increment as compared to tHBcAg control particles without cEDIII display (namely tEL). Mice immunized with tHBcAg-cEDIII VLPs showed a positive seroconversion to cEDIII antigen, thereby signifying that the assembled tHBcAg-cEDIII VLPs have successfully displayed cEDIII antigen to the immune system. If it is proven to be successful, tHBcAg-cEDIII has the potential to be developed as a cost-effective vaccine candidate that confers a simultaneous protection against all four infecting dengue viral serotypes.
Collapse
Affiliation(s)
- Ee Leen Pang
- School of Biosciences, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Hadrien Peyret
- Department of Biological Chemistry, John Innes Centre, Norwich, United Kingdom
| | | | - Hwei-San Loh
- School of Biosciences, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Kok-Song Lai
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, Semenyih, Malaysia
| | | | | |
Collapse
|
22
|
Webster GR, van Dolleweerd C, Guerra T, Stelter S, Hofmann S, Kim M, Teh AY, Diogo GR, Copland A, Paul MJ, Hart P, Reljic R, Ma JK. A polymeric immunoglobulin-antigen fusion protein strategy for enhancing vaccine immunogenicity. PLANT BIOTECHNOLOGY JOURNAL 2018; 16:1983-1996. [PMID: 29682888 PMCID: PMC6230950 DOI: 10.1111/pbi.12932] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/15/2018] [Accepted: 03/21/2018] [Indexed: 05/06/2023]
Abstract
In this study, a strategy based on polymeric immunoglobulin G scaffolds (PIGS) was used to produce a vaccine candidate for Mycobacterium tuberculosis. A genetic fusion construct comprising genes encoding the mycobacterial Ag85B antigen, an immunoglobulin γ-chain fragment and the tailpiece from immunoglobulin μ chain was engineered. Expression was attempted in Chinese Hamster Ovary (CHO) cells and in Nicotiana benthamiana. The recombinant protein assembled into polymeric structures (TB-PIGS) in N. benthamiana, similar in size to polymeric IgM. These complexes were subsequently shown to bind to the complement protein C1q and FcγRs with increased affinity. Modification of the N-glycans linked to TB-PIGS by removal of xylose and fucose residues that are normally found in plant glycosylated proteins also resulted in increased affinity for low-affinity FcγRs. Immunization studies in mice indicated that TB-PIGS are highly immunogenic with and without adjuvant. However, they did not improve protective efficacy in mice against challenge with M. tuberculosis compared to conventional vaccination with BCG, suggesting that additional or alternative antigens may be needed to protect against this disease. Nevertheless, these results establish a novel platform for producing polymeric antigen-IgG γ-chain molecules with inherent functional characteristics that are desirable in vaccines.
Collapse
Affiliation(s)
- Gina R. Webster
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | | | - Thais Guerra
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Szymon Stelter
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Sven Hofmann
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Mi‐Young Kim
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Audrey Y‐H. Teh
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Gil Reynolds Diogo
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Alastair Copland
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Mathew J. Paul
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Peter Hart
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Rajko Reljic
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| | - Julian K‐C. Ma
- Institute for Infection and ImmunitySt. George's University of LondonLondonUK
| |
Collapse
|
23
|
Bal J, Jung HY, Nguyen LN, Park J, Jang YS, Kim DH. Evaluation of cell-surface displayed synthetic consensus dengue EDIII cells as a potent oral vaccine candidate. Microb Cell Fact 2018; 17:146. [PMID: 30217208 PMCID: PMC6138890 DOI: 10.1186/s12934-018-0994-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/10/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Dengue is a rapidly spreading mosquito borne tropical viral disease affecting hundreds of millions of people across the globe annually. The dengue virus (DENV) includes four genetically distinct serotypes that cause serious life-threatening infections, including dengue hemorrhagic fever/dengue shock syndrome. Dengue vaccine development is complicated by the possibility of vaccine-enhanced severe dengue disease due to antibody-dependent enhancement by pre-existing cross-reactivity, as well as homotypic antibodies. Thus, the development of an efficacious dengue vaccine conferring simultaneous and durable immunity to each of the four DENV serotypes has not yet been developed despite years of research. For mass immunization in deeply affected resource-limited countries, oral vaccination is considered more beneficial than conventional approaches. Therefore, in a continuing effort towards designing economical and potent vaccine candidates, the current study applied yeast surface display technology to develop an oral dengue vaccine candidate using whole recombinant yeast cells displaying the recombinant fusion protein of M cell targeting ligand Co1 fused to the synthetic consensus dengue envelope domain III (scEDIII). Female Balb/c mice were orally fed with recombinant yeast cells and immunogenicity in terms of systemic and mucosal immune responses was monitored. RESULTS Immunofluorescence microscopy with dengue specific antibody and fluorescein isothiocyanate-conjugated anti-mouse IgG antibody clearly showed that recombinant protein Co1-scEDIII-AGA was localized on the cell surface of the respective clones in comparison with scEDIII-Co1 and Mock cells with no fluorescence. Oral dosage applications of surface displayed Co1-scEDIII-AGA stimulated a systemic humoral immune response in the form of dengue-specific serum IgG, as well as a mucosal immune response in the form of secretory immunoglobulin A (sIgA). Antigen-specific B cell responses in isolated lymphoid cells from the spleen and Peyer's patches further supported an elevated mucosal immune response. In addition, surface displayed Co1-scEDIII-AGA feeding elicited strong immune responses in comparison with scEDIII-Co1 and Mock following intraperitoneal booster with purified scEDIII antigen. CONCLUSIONS Surface displayed preparations of Co1-scEDIII-AGA induced strong immunogenicity compared with non-displayed scEDIII-Co1. Prior studies have supported the neutralization potential of scEDIII constructs against all four serotypes. Thus, the oral administration of genetically engineered yeast whole cells displaying biologically active Co1-scEDIII fusion protein without any further processing shows prospective as a potent oral vaccine candidate against dengue viral infection.
Collapse
Affiliation(s)
- Jyotiranjan Bal
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Hee-Young Jung
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Luong Ngoc Nguyen
- Department of Biology, College of Sciences, Hue University, Hue, Vietnam
| | - Jisang Park
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Yong-Suk Jang
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Dae-Hyuk Kim
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| |
Collapse
|
24
|
Tripathi NK, Shrivastava A. Recent Developments in Recombinant Protein-Based Dengue Vaccines. Front Immunol 2018; 9:1919. [PMID: 30190720 PMCID: PMC6115509 DOI: 10.3389/fimmu.2018.01919] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 08/03/2018] [Indexed: 12/11/2022] Open
Abstract
Recombinant proteins are gaining enormous importance these days due to their wide application as biopharmaceutical products and proven safety record. Various recombinant proteins of therapeutic and prophylactic importance have been successfully produced in microbial and higher expression host systems. Since there is no specific antiviral therapy available against dengue, the prevention by vaccination is the mainstay in reducing the disease burden. Therefore, efficacious vaccines are needed to control the spread of dengue worldwide. Dengue is an emerging viral disease caused by any of dengue virus 1-4 serotypes that affects the human population around the globe. Dengue virus is a single stranded RNA virus encoding three structural proteins (capsid protein, pre-membrane protein, and envelope protein) and seven non-structural proteins (NS1, NS2a, NS2b, NS3, NS4a, NS4b, NS5). As the only licensed dengue vaccine (Dengvaxia) is unable to confer balanced protection against all the serotypes, therefore various approaches for development of dengue vaccines including tetravalent live attenuated, inactivated, plasmid DNA, virus-vectored, virus-like particles, and recombinant subunit vaccines are being explored. These candidates are at different stages of vaccine development and have their own merits and demerits. The promising subunit vaccines are mainly based on envelope or its domain and non-structural proteins of dengue virus. These proteins have been produced in different hosts and are being investigated for development of a successful dengue vaccine. Novel immunogens have been designed employing various strategies like protein engineering and fusion of antigen with various immunostimulatory motif to work as self-adjuvant. Moreover, recombinant proteins can be formulated with novel adjuvants to enhance the immunogenicity and thus conferring better protection to the vaccinees. With the advent of newer and safer host systems, these recombinant proteins can be produced in a cost effective manner at large scale for vaccine studies. In this review, we summarize recent developments in recombinant protein based dengue vaccines that could lead to a good number of efficacious vaccine candidates for future human use and ultimately alternative dengue vaccine candidates.
Collapse
Affiliation(s)
- Nagesh K. Tripathi
- Bioprocess Scale Up Facility, Defence Research and Development Establishment, Gwalior, India
| | - Ambuj Shrivastava
- Division of Virology, Defence Research and Development Establishment, Gwalior, India
| |
Collapse
|
25
|
Rosales-Mendoza S, Nieto-Gómez R. Green Therapeutic Biocapsules: Using Plant Cells to Orally Deliver Biopharmaceuticals. Trends Biotechnol 2018; 36:1054-1067. [PMID: 29980327 DOI: 10.1016/j.tibtech.2018.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 12/18/2022]
Abstract
The use of innovative platforms to produce biopharmaceuticals cheaply and deliver them through noninvasive routes could expand their social benefits. Coverage should increase as a consequence of lower cost and higher patient compliance due to painless administration. For more than two decades of research, oral therapies that rely on genetically engineered plants for the production of biopharmaceuticals have been explored to treat or prevent high-impact diseases. Recent reports on the successful oral delivery of plant-made biopharmaceuticals raise new hopes for the field. Several candidates have shown protection in animal models, and efforts to establish their production on an industrial scale are ongoing. These advances and perspectives for the field are analyzed.
Collapse
Affiliation(s)
- Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, Mexico; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Avenue Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí, 78210, Mexico.
| | - Ricardo Nieto-Gómez
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, Mexico; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Avenue Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí, 78210, Mexico
| |
Collapse
|
26
|
Kim MY, Copland A, Nayak K, Chandele A, Ahmed MS, Zhang Q, Diogo GR, Paul MJ, Hofmann S, Yang M, Jang Y, Ma JK, Reljic R. Plant-expressed Fc-fusion protein tetravalent dengue vaccine with inherent adjuvant properties. PLANT BIOTECHNOLOGY JOURNAL 2018; 16:1283-1294. [PMID: 29223138 PMCID: PMC5999314 DOI: 10.1111/pbi.12869] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/23/2017] [Accepted: 12/03/2017] [Indexed: 05/07/2023]
Abstract
Dengue is a major global disease requiring improved treatment and prevention strategies. The recently licensed Sanofi Pasteur Dengvaxia vaccine does not protect children under the age of nine, and additional vaccine strategies are thus needed to halt this expanding global epidemic. Here, we employed a molecular engineering approach and plant expression to produce a humanized and highly immunogenic poly-immunoglobulin G scaffold (PIGS) fused to the consensus dengue envelope protein III domain (cEDIII). The immunogenicity of this IgG Fc receptor-targeted vaccine candidate was demonstrated in transgenic mice expressing human FcγRI/CD64, by induction of neutralizing antibodies and evidence of cell-mediated immunity. Furthermore, these molecules were able to prime immune cells from human adenoid/tonsillar tissue ex vivo as evidenced by antigen-specific CD4+ and CD8+ T-cell proliferation, IFN-γ and antibody production. The purified polymeric fraction of dengue PIGS (D-PIGS) induced stronger immune activation than the monomeric form, suggesting a more efficient interaction with the low-affinity Fcγ receptors on antigen-presenting cells. These results show that the plant-expressed D-PIGS have the potential for translation towards a safe and easily scalable single antigen-based tetravalent dengue vaccine.
Collapse
Affiliation(s)
- Mi Young Kim
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
- Department of Molecular Biology and the Institute for Molecular Biology and GeneticsChonbuk National UniversityJeonjuKorea
| | - Alastair Copland
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Kaustuv Nayak
- ICGEB‐Emory Vaccine CenterInternational Center for Genetic Engineering and BiotechnologyNew DelhiIndia
| | - Anmol Chandele
- ICGEB‐Emory Vaccine CenterInternational Center for Genetic Engineering and BiotechnologyNew DelhiIndia
| | - Muhammad S. Ahmed
- Department of Clinical Infection, Microbiology and ImmunologyInstitute of Infection and Global HealthUniversity of LiverpoolLiverpoolUK
| | - Qibo Zhang
- Department of Clinical Infection, Microbiology and ImmunologyInstitute of Infection and Global HealthUniversity of LiverpoolLiverpoolUK
| | - Gil R. Diogo
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Matthew J. Paul
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Sven Hofmann
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Moon‐Sik Yang
- Department of Molecular Biology and the Institute for Molecular Biology and GeneticsChonbuk National UniversityJeonjuKorea
| | - Yong‐Suk Jang
- Department of Molecular Biology and the Institute for Molecular Biology and GeneticsChonbuk National UniversityJeonjuKorea
| | - Julian K‐C. Ma
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Rajko Reljic
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| |
Collapse
|
27
|
Frey S, Castro A, Arsiwala A, Kane RS. Bionanotechnology for vaccine design. Curr Opin Biotechnol 2018; 52:80-88. [PMID: 29597075 DOI: 10.1016/j.copbio.2018.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/10/2018] [Indexed: 01/30/2023]
Abstract
There have been significant advances in the design of nanostructured scaffolds for eliciting robust immune responses. One method to produce strong immune responses is to emulate the appearance of a pathogen. Since pathogens such as viruses and bacteria often display multiple copies of ligands on their surfaces, the immune system is particularly sensitive towards multivalent displays of antigens. Consequently, when designing a vaccine, it is advantageous to decorate a nanostructured surface with multiple copies of an antigen. This review highlights the design and efficacy of a diverse set of recently developed nanostructured vaccine scaffolds.
Collapse
Affiliation(s)
- Steven Frey
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ana Castro
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ammar Arsiwala
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ravi S Kane
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
28
|
Hart P, Copland A, Diogo GR, Harris S, Spallek R, Oehlmann W, Singh M, Basile J, Rottenberg M, Paul MJ, Reljic R. Nanoparticle-Fusion Protein Complexes Protect against Mycobacterium tuberculosis Infection. Mol Ther 2018; 26:822-833. [PMID: 29518353 PMCID: PMC5910664 DOI: 10.1016/j.ymthe.2017.12.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 11/26/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of death from infectious disease, and the current vaccine, Bacillus Calmette-Guerin (BCG), is inadequate. Nanoparticles (NPs) are an emerging vaccine technology, with recent successes in oncology and infectious diseases. NPs have been exploited as antigen delivery systems and also for their adjuvantic properties. However, the mechanisms underlying their immunological activity remain obscure. Here, we developed a novel mucosal TB vaccine (Nano-FP1) based upon yellow carnauba wax NPs (YC-NPs), coated with a fusion protein consisting of three Mycobacterium tuberculosis (Mtb) antigens: Acr, Ag85B, and HBHA. Mucosal immunization of BCG-primed mice with Nano-FP1 significantly enhanced protection in animals challenged with low-dose, aerosolized Mtb. Bacterial control by Nano-FP1 was associated with dramatically enhanced cellular immunity compared to BCG, including superior CD4+ and CD8+ T cell proliferation, tissue-resident memory T cell (Trm) seeding in the lungs, and cytokine polyfunctionality. Alongside these effects, we also observed potent humoral responses, such as the generation of Ag85B-specific serum IgG and respiratory IgA. Finally, we found that YC-NPs were able to activate antigen-presenting cells via an unconventional IRF-3-associated activation signature, without the production of potentially harmful inflammatory mediators, providing a mechanistic framework for vaccine efficacy and future development.
Collapse
Affiliation(s)
- Peter Hart
- St George's Medical School, University of London, London SW17 0RE, UK
| | - Alastair Copland
- St George's Medical School, University of London, London SW17 0RE, UK
| | | | - Shane Harris
- St George's Medical School, University of London, London SW17 0RE, UK
| | | | | | | | | | | | - Matthew John Paul
- St George's Medical School, University of London, London SW17 0RE, UK
| | - Rajko Reljic
- St George's Medical School, University of London, London SW17 0RE, UK.
| |
Collapse
|
29
|
Bal J, Luong NN, Park J, Song KD, Jang YS, Kim DH. Comparative immunogenicity of preparations of yeast-derived dengue oral vaccine candidate. Microb Cell Fact 2018; 17:24. [PMID: 29452594 PMCID: PMC5815244 DOI: 10.1186/s12934-018-0876-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/09/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Dengue is listed as a neglected tropical disease by the Center for Disease Control and Preservation, as there are insufficient integrated surveillance strategies, no effective treatment, and limited licensed vaccines. Consisting of four genetically distinct serotypes, dengue virus (DENV) causes serious life-threatening infections due to its complexity. Antibody-dependent enhancement by pre-existing cross-reactive as well as homotypic antibodies further worsens the clinical symptoms of dengue. Thus, a vaccine conferring simultaneous and durable immunity to each of the four DENV serotypes is essential to restrict its escalation. In deeply affected resource-limited countries, oral vaccination using food-grade organisms is considered to be a beneficial approach in terms of costs, patient comfort, and simple logistics for mass immunization. The current study used a mouse model to explore the immunogenicity of an oral dengue vaccine candidate prepared using whole recombinant yeast cells (WC) and cell-free extracts (CFE) from cells expressing recombinant Escherichia coli heat-labile toxin protein B-subunit (LTB) fused to the consensus dengue envelope domain III (scEDIII). Mice were treated orally with recombinant WC and CFE vaccines in 2-week intervals for 4 weeks and changes in systemic and mucosal immune responses were monitored. RESULTS Both WC and CFE dosage applications of LTB-scEDIII stimulated a systemic humoral immune response in the form of dengue-specific serum IgG as well as mucosal immune response in the form of secretory sIgA. Antigen-specific B cell responses in isolated lymphoid cells from the spleen and Peyer's patches further indicated an elevated mucosal immune response. Cellular immune response estimated through lymphocyte proliferation assay indicated higher levels in CFE than WC dosage. Furthermore, sera obtained after both oral administrations successfully neutralized DENV-1, whereas CFE formulation only neutralized DENV-2 serotype, two representative serotypes which cause severe dengue infection. Sera from mice that were fed CFE preparations demonstrated markedly higher neutralizing titers compared to those from WC-fed mice. However, WC feeding elicited strong immune responses, which were similar to the levels induced by CFE feeding after intraperitoneal booster with purified scEDIII antigen. CONCLUSIONS CFE preparations of LTB-scEDIII produced strong immunogenicity with low processing requirements, signifying that this fusion protein shows promise as a potent oral vaccine candidate against dengue viral infection.
Collapse
Affiliation(s)
- Jyotiranjan Bal
- Department of Molecular Biology, Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
| | - Nguyen Ngoc Luong
- Department of Biology, College of Sciences, Hue University, Hue, Vietnam
| | - Jisang Park
- Department of Molecular Biology, Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
| | - Ki-Duk Song
- Department of Animal Biotechnology, The Animal Molecular Genetics and Breeding Center, Chonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
| | - Yong-Suk Jang
- Department of Molecular Biology, Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
| | - Dae-Hyuk Kim
- Department of Molecular Biology, Department of Bioactive Material Sciences, Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea.
| |
Collapse
|