101
|
Butani N, Xu Y, Pan S, Durocher Y, Ghosh R. A fast, efficient, and scalable method for purifying recombinant SARS-CoV-2 spike protein. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1215:123579. [PMID: 36603473 PMCID: PMC9810479 DOI: 10.1016/j.jchromb.2022.123579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Recombinant SARS-CoV-2 trimeric spike protein produced by mammalian cell culture is a potential candidate for a COVID-19 vaccine. However, this protein is much larger than most typical biopharmaceutical proteins and its large-scale manufacture is therefore challenging. Particularly, its purification using resin-based chromatography is difficult as the diffusive transport of this protein to and from its binding site within the pores of the stationary phase particles is slow. Therefore, very low flow rates need to be used during binding and elution, and this slows down the purification process. Also, due to its large size, the binding capacity of this protein on resin-based media is low. Membrane chromatography is an efficient and scalable technique for purifying biopharmaceuticals. The predominant mode of solute transport in a membrane is convective and hence it is considered better than resin-based chromatography for purifying large proteins. In this paper, we propose a membrane chromatography-based purification method for fast and scalable manufacture of recombinant SARS-CoV-2 trimeric spike protein. A combination of cation exchange z2 laterally-fed membrane chromatography and size exclusion chromatography was found to be suitable for obtaining a homogeneous spike protein sample from mammalian cell culture supernatant. The proposed method is both fast and scalable and could be explored as a method for manufacturing vaccine grade spike protein.
Collapse
Affiliation(s)
- Nikhila Butani
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L7, Canada
| | - Yating Xu
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L7, Canada
| | - Si Pan
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L7, Canada
| | - Yves Durocher
- National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Raja Ghosh
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L7, Canada.
| |
Collapse
|
102
|
Kovalenko A, Ryabchevskaya E, Evtushenko E, Nikitin N, Karpova O. Recombinant Protein Vaccines against Human Betacoronaviruses: Strategies, Approaches and Progress. Int J Mol Sci 2023; 24:1701. [PMID: 36675218 PMCID: PMC9863728 DOI: 10.3390/ijms24021701] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Betacoronaviruses have already troubled humanity more than once. In 2002-2003 and 2012, the SARS-CoV and MERS-CoV, respectively, caused outbreaks of respiratory syndromes with a fatal outcome. The spread of the SARS-CoV-2 coronavirus has become a pandemic. These three coronaviruses belong to the genus Betacoronavirus and have a zoonotic origin. The emergence of new coronavirus infections in the future cannot be ruled out, and vaccination is the main way to prevent the spread of the infection. Previous experience in the development of vaccines against SARS and MERS has helped to develop a number of vaccines against SARS-CoV-2 in a fairly short time. Among them, there are quite a few recombinant protein vaccines, which seem to be very promising in terms of safety, minimization of side effects, storage and transportation conditions. The problem of developing a universal betacoronavirus vaccine is also still relevant. Here, we summarize the information on the designing of vaccines based on recombinant proteins against highly pathogenic human betacoronaviruses SARS-CoV, MERS-CoV and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | | | - Nikolai Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | | |
Collapse
|
103
|
Naeem M, Alkhodairy HF, Ashraf I, Khalil AB. CRISPR/Cas System Toward the Development of Next-Generation Recombinant Vaccines: Current Scenario and Future Prospects. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2023; 48:1-11. [PMID: 36185592 PMCID: PMC9510529 DOI: 10.1007/s13369-022-07266-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/30/2022] [Indexed: 01/11/2023]
Abstract
The initially developed vaccines were relying mostly on attenuation and inactivation of pathogens. The use of recombinant DNA technology allows the targeting of immune responses focused against a few protective antigens. The conventional recombination methods for generating vaccines are time-consuming, laborious, and less efficient. To overcome these limitations, a new precise CRISPR/Cas9 with high efficacy, specificity, and low-cost properties has solved a lot of current problems of recombinant vaccines that intrigued the inspiration for novel recombinant vaccine development. CRISPR/Cas9 system was discovered as a bacterial adaptive immune system. In the domain of virology, CRISPR/Cas9 is used to engineer the virus genome to understand the fundamentals of viral pathogenesis, gene therapy, and virus-host interactions. One step ahead CRISPR/Cass9 bypassed the vaccine to precisely engineer the B-cells to secrete the specific antibodies against deadly viral pathogens. There is a critical literature review gap especially in the use of CRISPR/Cas9 to generate recombinant vaccines against viral diseases and its prospective application to engineering the B-cells in immunocompromised people. This review heights the application of CRISPR/Cas9 compared to conventional approaches for the development of recombinant vaccine vectors, editing the genes of B-cells, and challenges that need to be overcome. The factors affecting CRISPR/Cas9-edited recombinant vaccines and prospects in the context of viral genome editing for the development of vaccines will be discussed.
Collapse
Affiliation(s)
- Muhammad Naeem
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261 Saudi Arabia
| | - Hanoof Fahd Alkhodairy
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261 Saudi Arabia
| | - Iqra Ashraf
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Amjad Bajes Khalil
- Department of Bioengineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261 Saudi Arabia
| |
Collapse
|
104
|
Gao Y, Wang W, Yang Y, Zhao Q, Yang C, Jia X, Liu Y, Zhou M, Zeng W, Huang X, Chiu S, Jin T, Wu X. Developing Next-Generation Protein-Based Vaccines Using High-Affinity Glycan Ligand-Decorated Glyconanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204598. [PMID: 36398611 PMCID: PMC9839878 DOI: 10.1002/advs.202204598] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Major diseases, such as cancer and COVID-19, are frightening global health problems, and sustained action is necessary to develop vaccines. Here, for the first time, ethoxy acetalated dextran nanoparticles (Ace-Dex-NPs) are functionalized with 9-N-(4H-thieno[3,2-c]chromene-2-carbamoyl)-Siaα2-3Galβ1-4GlcNAc (TCC Sia-LacNAc) targeting macrophages as a universal vaccine design platform. First, azide-containing oxidized Ace-Dex-NPs are synthesized. After the NPs are conjugated with ovalbumin (OVA) and resiquimod (Rd), they are coupled to TCC Sia-LacNAc-DBCO to produce TCC Sia-Ace-Dex-OVA-Rd, which induce a potent, long-lasting OVA-specific cytotoxic T-lymphocyte (CTL) response and high anti-OVA IgG, providing mice with superior protection against tumors. Next, this strategy is exploited to develop vaccines against infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein is the main target for neutralizing antibodies. The TCC Sia-Ace-Dex platform is preferentially used for designing an RBD-based vaccine. Strikingly, the synthetic TCC Sia-Ace-Dex-RBD-Rd elicited potent RBD-neutralizing antibodies against live SARS-CoV-2 infected Vero E6 cells. To develop a universal SARS-CoV-2 vaccine, the TCC Sia-Ace-Dex-N-Rd vaccine carrying SARS-CoV-2 nucleocapsid protein (N) is also prepared, which is highly conserved among SARS-CoV-2 and its variants of concern (VOCs), including Omicron (BA.1 to BA.5); this vaccine can trigger strong N-specific CTL responses against target cells infected with SARS-CoV-2 and its VOCs.
Collapse
Affiliation(s)
- Yanan Gao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Wei Wang
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Yunru Yang
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Qingyu Zhao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Chendong Yang
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Xiaoying Jia
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Yang Liu
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
| | - Minmin Zhou
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Weihong Zeng
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Xuefei Huang
- Departments of Chemistry and Biomedical EngineeringInstitute for Quantitative Health Science and EngineeringMichigan State UniversityEast LansingMichigan48824United States
| | - Sandra Chiu
- Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Tengchuan Jin
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Xuanjun Wu
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
- Suzhou Research InstituteShandong UniversitySuzhouJiangsu215123China
| |
Collapse
|
105
|
Muacevic A, Adler JR. Herd Immunity to Fight Against COVID-19: A Narrative Review. Cureus 2023; 15:e33575. [PMID: 36779140 PMCID: PMC9909126 DOI: 10.7759/cureus.33575] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/09/2023] [Indexed: 01/11/2023] Open
Abstract
The advent of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its consequent illness, coronavirus disease 2019 (COVID-19), has revealed the severe impact of new, contagious pathogens on the population throughout the globe. Here, we describe the fundamental notions of herd immunity and discuss their consequences from the perspective of COVID-19, along with the obstacles to acquiring herd immunity. SARS-CoV-2 causes COVID-19, a contagious respiratory infection. It is a major global health issue, with more than 179 million positive cases and 3.8 million deaths globally. It has impacted more than 159 countries; hence, the World Health Organization designated it a pandemic. Different vaccines have been developed against coronavirus to slow the spread of this deadly virus. Immunizing people against coronavirus is the key to getting through this infectious virus. The central concept of this review article is the effect of vaccinating a large population to achieve herd immunity and the reasons for the delay in developing herd immunity. Herd immunity can prove highly beneficial for dealing with reinfection. Moreover, it can reduce the severity of the reinfection in many people who are twice infected with COVID-19. Herd immunity can prevent people in the high-risk group such as immunocompromised individuals; those on immunosuppressants; organ transplant recipients; particular age groups such as neonates, infants, toddlers, and elderly; those with impaired immunity; those with anaphylaxis reactions; and people with chronic diseases. However, due to repeated mutations of the virus, it is evolving into new strains with more severity. Its consequences on the immune system and response to a vaccine are still a big challenge to overcome. How new variants of COVID-19 impacted herd immunity needs to be investigated. The duration required for the development of herd immunity and how long it would last is still under research, along with the number of doses needed, booster doses, and the proportion of the population to be vaccinated.
Collapse
|
106
|
Hisham Y, Seo SM, Kim S, Shim S, Hwang J, Yoo ES, Kim NW, Song CS, Jhun H, Park HY, Lee Y, Shin KC, Han SY, Seong JK, Choi YK, Kim S. COVID-19 spike polypeptide vaccine reduces the pathogenesis and viral infection in a mouse model of SARS-CoV-2. Front Immunol 2023; 14:1098461. [PMID: 36936979 PMCID: PMC10020603 DOI: 10.3389/fimmu.2023.1098461] [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: 11/15/2022] [Accepted: 01/25/2023] [Indexed: 03/08/2023] Open
Abstract
The SARS-CoV-2 coronavirus, which causes a respiratory disease called COVID-19, has been declared a pandemic by the World Health Organization (WHO) and is still ongoing. Vaccination is the most important strategy to end the pandemic. Several vaccines have been approved, as evidenced by the ongoing global pandemic, but the pandemic is far from over and no fully effective vaccine is yet available. One of the most critical steps in vaccine development is the selection of appropriate antigens and their proper introduction into the immune system. Therefore, in this study, we developed and evaluated two proposed vaccines composed of single and multiple SARS-CoV-2 polypeptides derived from the spike protein, namely, vaccine A and vaccine B, respectively. The polypeptides were validated by the sera of COVID-19-vaccinated individuals and/or naturally infected COVID-19 patients to shortlist the starting pool of antigens followed by in vivo vaccination to hACE2 transgenic mice. The spike multiple polypeptide vaccine (vaccine B) was more potent to reduce the pathogenesis of organs, resulting in higher protection against the SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yasmin Hisham
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Sun-Min Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sinae Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Saerok Shim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Jihyeong Hwang
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Eun-Seon Yoo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Na-Won Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Chang-Seon Song
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyunjhung Jhun
- Food Industry Infrastructure Team, Korea Food Research Institute, Wanju, Republic of Korea
| | - Ho-Young Park
- Research Group of Functional Food Materials, Korea Food Research Institute, Wanju, Republic of Korea
| | - Youngmin Lee
- Department of Medicine, Pusan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Kyeong-Cheol Shin
- Center for Respiratory Disease, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Sun-Young Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center, Interdisciplinary Program for Bioinformatics, and BioMAX Institute, Seoul National University, Seoul, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
- *Correspondence: Yang-Kyu Choi, ; Soohyun Kim,
| | - Soohyun Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, Republic of Korea
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
- *Correspondence: Yang-Kyu Choi, ; Soohyun Kim,
| |
Collapse
|
107
|
Toledo-Romaní ME, García-Carmenate M, Valenzuela-Silva C, Baldoquín-Rodríguez W, Martínez-Pérez M, Rodríguez-González M, Paredes-Moreno B, Mendoza-Hernández I, González-Mujica Romero R, Samón-Tabio O, Velazco-Villares P, Bacallao-Castillo JP, Licea-Martín E, Rodríguez-Ortega M, Herrera-Marrero N, Caballero-González E, Egües-Torres L, Duartes-González R, García-Blanco S, Pérez-Cabrera S, Huete-Ferreira S, Idalmis-Cisnero K, Fonte-Galindo O, Meliá-Pérez D, Rojas-Remedios I, Doroud D, Gouya MM, Biglari A, Fernández-Castillo S, Climent-Ruiz Y, Valdes-Balbín Y, García-Rivera D, Van der Stuyft P, Verez-Bencomo V, SOBERANA Phase 3 team. Safety and efficacy of the two doses conjugated protein-based SOBERANA-02 COVID-19 vaccine and of a heterologous three-dose combination with SOBERANA-Plus: a double-blind, randomised, placebo-controlled phase 3 clinical trial. LANCET REGIONAL HEALTH. AMERICAS 2022; 18:100423. [PMID: 36618081 PMCID: PMC9803910 DOI: 10.1016/j.lana.2022.100423] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 01/01/2023]
Abstract
Background SOBERANA-02 is a COVID-19 conjugate vaccine (recombinant RBD conjugated to tetanus toxoid). Phases 1/2 clinical trials demonstrated high immunogenicity, promoting neutralising IgG and specific T-cell response. A third heterologous dose of SOBERANA-Plus (RBD-dimer) further increased neutralising antibodies. The aim of this study is to evaluate the safety and efficacy of two immunisation regimes: two doses of SOBERANA-02 and a heterologous three-dose combination with SOBERANA-Plus added to it. Methods From March 8th to June 24th, 2021 we conducted in Havana, Cuba a multicentre randomised, double-blind, placebo-controlled, phase-3 trial evaluating a two doses SOBERANA-02 scheme and a heterologous scheme with one dose SOBERANA-Plus added to it (RPCEC00000354). Participants 19-80 years were randomly assigned to receiving 28 days apart either the two or three dose scheme or placebo. The main endpoint was vaccine efficacy in preventing the occurrence of RT-PCR confirmed symptomatic COVID-19 at least 14 days after the second or third dose in the per-protocol population. We also assessed efficacy against severe disease and, in all participants receiving at least one vaccine/placebo dose, safety for 28 days after each dose. Findings We included 44,031 participants (52.0% female, 48.0% male; median age 50 years, range 19-80 years; 7.0% black, 24.0% mixed-race, 59.0% white) in a context of initial Beta VOC predominance, with this variant being partially replaced by Delta near the trial's end. Vaccine efficacy in the heterologous combination was 92.0% (95%CI 80.4-96.7) against symptomatic disease. There were no severe COVID-19 cases in the vaccine group against 6 in the placebo group. Two doses of SOBERANA-02 was 69.7% (95%CI 56.5-78.9) and 74.9% (95%CI 33.7-90.5) efficacious against symptomatic and severe COVID-19, respectively. The occurrence of serious and severe adverse events (AE) was very rare and equally distributed between placebo and vaccine groups. Solicited AEs were slightly more frequent in the vaccine group but predominantly local and mostly mild and transient. Interpretation Our results indicate that the straightforward to manufacture SOBERANA vaccines are efficacious in a context of Beta and Delta VOC circulation, have a favourable safety profile, and may represent an attractive option for use in COVID-19 vaccination programmes. Funding This study received funds from the National Fund for Science and Technology (FONCI-CITMA-Cuba, contract 2020-20) of the Ministry of Science, Technology and Environment of Cuba.
Collapse
Affiliation(s)
- María Eugenia Toledo-Romaní
- “Pedro Kourí” Tropical Medicine Institute. Av “Novia del Mediodía”, Kv 6 1/2, La Lisa, Habana, 11400, Cuba,Corresponding author.
| | | | - Carmen Valenzuela-Silva
- Cybernetics, Mathematics and Physics Institute. 15th St #55, Vedado, Plaza de la Revolución, Havana, 10400, Cuba
| | | | - Marisel Martínez-Pérez
- Finlay Vaccine Institute. 21st Ave. N° 19810 between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Meiby Rodríguez-González
- Finlay Vaccine Institute. 21st Ave. N° 19810 between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Beatriz Paredes-Moreno
- Finlay Vaccine Institute. 21st Ave. N° 19810 between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Ivis Mendoza-Hernández
- National Coordinating Center for Clinical Trials. 5th Ave and 62, Miramar, Playa, Havana, Cuba
| | | | | | | | | | - Ernesto Licea-Martín
- Finlay Vaccine Institute. 21st Ave. N° 19810 between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Misladys Rodríguez-Ortega
- “Pedro Kourí” Tropical Medicine Institute. Av “Novia del Mediodía”, Kv 6 1/2, La Lisa, Habana, 11400, Cuba
| | - Nuris Herrera-Marrero
- “Pedro Kourí” Tropical Medicine Institute. Av “Novia del Mediodía”, Kv 6 1/2, La Lisa, Habana, 11400, Cuba
| | | | | | | | | | | | | | | | | | | | | | - Delaram Doroud
- Pasteur Institute of Iran. No. 69, Pasteur Ave., Tehran 1316943551, Islamic Republic of Iran
| | | | - Alireza Biglari
- Pasteur Institute of Iran. No. 69, Pasteur Ave., Tehran 1316943551, Islamic Republic of Iran
| | | | - Yanet Climent-Ruiz
- Finlay Vaccine Institute. 21st Ave. N° 19810 between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Yury Valdes-Balbín
- Finlay Vaccine Institute. 21st Ave. N° 19810 between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Dagmar García-Rivera
- Finlay Vaccine Institute. 21st Ave. N° 19810 between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | | | - Vicente Verez-Bencomo
- Finlay Vaccine Institute. 21st Ave. N° 19810 between 198 and 200 St, Atabey, Playa, Havana, Cuba,Corresponding author.
| | | |
Collapse
|
108
|
Homma T, Nagata N, Hashimoto M, Iwata-Yoshikawa N, Seki NM, Shiwa-Sudo N, Ainai A, Dohi K, Nikaido E, Mukai A, Ukai Y, Nakagawa T, Shimo Y, Maeda H, Shirai S, Aoki M, Sonoyama T, Sato M, Fumoto M, Nagira M, Nakata F, Hashiguchi T, Suzuki T, Omoto S, Hasegawa H. Immune response and protective efficacy of the SARS-CoV-2 recombinant spike protein vaccine S-268019-b in mice. Sci Rep 2022; 12:20861. [PMID: 36460696 PMCID: PMC9718471 DOI: 10.1038/s41598-022-25418-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Vaccines that efficiently target severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent for coronavirus disease (COVID-19), are the best means for controlling viral spread. This study evaluated the efficacy of the COVID-19 vaccine S-268019-b, which comprises the recombinant full-length SARS-CoV-2 spike protein S-910823 (antigen) and A-910823 (adjuvant). In addition to eliciting both Th1-type and Th2-type cellular immune responses, two doses of S-910823 plus A-910823 induced anti-spike protein IgG antibodies and neutralizing antibodies against SARS-CoV-2. In a SARS-CoV-2 challenge test, S-910823 plus A-910823 mitigated SARS-CoV-2 infection-induced weight loss and death and inhibited viral replication in mouse lungs. S-910823 plus A-910823 promoted cytokine and chemokine at the injection site and immune cell accumulation in the draining lymph nodes. This led to the formation of germinal centers and the induction of memory B cells, antibody-secreting cells, and memory T cells. These findings provide fundamental property of S-268019-b, especially importance of A-910823 to elicit humoral and cellular immune responses.
Collapse
Affiliation(s)
- Tomoyuki Homma
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Masayuki Hashimoto
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Naomi M Seki
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Nozomi Shiwa-Sudo
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Keiji Dohi
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Eiji Nikaido
- Laboratory for Bio-Modality Research, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Akiko Mukai
- UMN Pharma Inc., 7F, Tekko Building, 1-8-2, Marunouchi, Chiyoda-ku, Tokyo, 100-0005, Japan
| | - Yuuta Ukai
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Takayuki Nakagawa
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Yusuke Shimo
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Hiroki Maeda
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Seiki Shirai
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Miwa Aoki
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Takuhiro Sonoyama
- Medical Science Department, Shionogi & Co., Ltd., 8F, Nissei East Building, 3-3-16, Imabashi, Chuo-ku, Osaka, 541-0032, Japan
| | - Mamoru Sato
- UMN Pharma Inc., 7F, Tekko Building, 1-8-2, Marunouchi, Chiyoda-ku, Tokyo, 100-0005, Japan
| | - Masataka Fumoto
- Laboratory for Bio-Modality Research, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Morio Nagira
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Fumihisa Nakata
- UMN Pharma Inc., 7F, Tekko Building, 1-8-2, Marunouchi, Chiyoda-ku, Tokyo, 100-0005, Japan
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Virology, Faculty of Medicine, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Shinya Omoto
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan.
| | - Hideki Hasegawa
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan.
| |
Collapse
|
109
|
Zhang J, He Q, Yan X, Liu J, Bai Y, An C, Cui B, Gao F, Mao Q, Wang J, Xu M, Liang Z. Mixed formulation of mRNA and protein-based COVID-19 vaccines triggered superior neutralizing antibody responses. MedComm (Beijing) 2022; 3:e188. [PMID: 36474858 PMCID: PMC9717706 DOI: 10.1002/mco2.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
Integrating different types of vaccines into a singular immunization regimen is an effective and accessible approach to strengthen and broaden the immunogenicity of existing coronavirus disease 2019 (COVID-19) vaccine candidates. To optimize the immunization strategy of the novel mRNA-based vaccine and recombinant protein subunit vaccine that attracted much attention in COVID-19 vaccine development, we evaluated the immunogenicity of different combined regimens with the mRNA vaccine (RNA-RBD) and protein subunit vaccine (PS-RBD) in mice. Compared with homologous immunization of RNA-RBD or PS-RBD, heterologous prime-boost strategies for mRNA and protein subunit vaccines failed to simultaneously enhance neutralizing antibody (NAb) and Th1 cellular response in this study, showing modestly higher serum neutralizing activity and antibody-dependent cell-mediated cytotoxicity for "PS-RBD prime, RNA-RBD boost" and robust Th1 type cellular response for "RNA-RBD prime, PS-RBD boost". Interestingly, immunizing the mice with the mixed formulation of the two aforementioned vaccines in various proportions further significantly enhanced the NAb responses against ancestral, Delta, and Omicron strains and manifested increased Th1-type responses, suggesting that a mixed formulation of mRNA and protein vaccines might be a more prospective vaccination strategy. This study provides basic research data on the combined vaccination strategies of mRNA and protein-based COVID-19 vaccines.
Collapse
Affiliation(s)
- Jialu Zhang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Qian He
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Xujia Yan
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Jianyang Liu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Yu Bai
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Chaoqiang An
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Bopei Cui
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Fan Gao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Qunying Mao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Junzhi Wang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Miao Xu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| | - Zhenglun Liang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug ControlNHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological ProductsBeijingPeople's Republic of China
| |
Collapse
|
110
|
Özcengiz E, Keser D, Özcengiz G, Çelik G, Özkul A, İnçeh FN. Two formulations of coronavirus disease-19 recombinant subunit vaccine candidate made up of S1 fragment protein P1, S2 fragment protein P2, and nucleocapsid protein elicit strong immunogenicity in mice. Immun Inflamm Dis 2022; 10:e748. [PMID: 36444622 PMCID: PMC9695085 DOI: 10.1002/iid3.748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/09/2022] [Accepted: 10/29/2022] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Coronavirus disease (COVID-19) is ongoing as a global epidemic and there is still a need to develop much safer and more effective new vaccines that can also be easily adapted to important variants of the pathogen. In the present study in this direction, we developed a new COVID-19 vaccine, composed of two critical antigenic fragments of the S1 and S2 region of severe acute respiratory syndrome coronavirus 2 as well as the whole nucleocapsid protein (N), which was formulated with either alum or alum plus monophosphoryl lipid A (MPLA) adjuvant combinations. METHODS From within the spike protein S1 region, a fragmented protein P1 (MW:33 kDa) which includes the receptor-binding domain (RBD), another fragment protein P2 (MW:17.6) which contains important antigenic epitopes within the spike protein S2 region, and N protein (MW:46 kDa) were obtained after recombinant expression of the corresponding gene regions in Escherichia coli BL21. For use in immunization studies, three proteins were adsorbed with aluminum hydroxide gel and with the combination of aluminum hydroxide gel plus MPLA. RESULTS Each of the three protein antigens produced strong reactions in enzyme-linked immunosorbent assays and Western blot analysis studies performed with convalescent COVID-19 patient sera. In mice, these combined protein vaccine candidates elicited high titer anti-P1, anti-P2, and anti-N IgG and IgG2a responses. These also induced highly neutralizing antibodies and elicited significant cell-mediated immunity as demonstrated by enhanced antigen-specific levels of interferon-γ (INF-γ) in the splenocytes of immunized mice. CONCLUSION The results of this study showed that formulations of the three proteins with Alum or Alum + MPLA are effective in terms of humoral and cellular responses. However, since the Alum + MPLA formulation appears to be superior in Th1 response, this vaccine candidate may be recommended mainly for the elderly and immunocompromised individuals. We also believe that the alum-only formulation will provide great benefits for adults, young adolescents, and children.
Collapse
Affiliation(s)
| | - Duygu Keser
- Vaccine R&D, Pharmada Pharmaceuticals, Ankara, Turkey
| | - Gülay Özcengiz
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Gözde Çelik
- Vaccine R&D, Pharmada Pharmaceuticals, Ankara, Turkey
| | - Aykut Özkul
- Department of Virology, School of Veterinary Medicine, Ankara University, Ankara, Turkey
| | | |
Collapse
|
111
|
mRNA Vaccines as an Efficient Approach for the Rapid and Robust Induction of Host Immunity Against SARS-CoV-2. SN COMPREHENSIVE CLINICAL MEDICINE 2022; 4:88. [PMID: 35402783 PMCID: PMC8975617 DOI: 10.1007/s42399-022-01168-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/24/2022] [Indexed: 11/11/2022]
Abstract
Among the currently used COVID-19 vaccines, the mRNA-based vaccines drew the interest of the scientists because of its potent and versatile nature in mitigating the disease efficiently through increased translation as well as the robust modulation of the innate and adaptive immune responses within the host. The naked or lipid encapsulated mRNAs are usually optimized in order to formulate the vaccine. One of the interesting advantage of using mRNA vaccines is that such platform can even be used to mitigate other infectious diseases like influenza, zika, and rabies. However, the leading COVID-19 mRNA vaccines, i.e., mRNA-1273 and BNT162b2, have already been noticed to possess around 95% efficacy in provoking both the humoral and cell mediated immunity against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, causing the ongoing COVID-19 pandemic.
Collapse
|
112
|
Sarangi MK, Padhi S, Rath G, Nanda SS, Yi DK. Success of nano-vaccines against COVID-19: a transformation in nanomedicine. Expert Rev Vaccines 2022; 21:1739-1761. [PMID: 36384360 DOI: 10.1080/14760584.2022.2148659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The vaccines being used against COVID-19 are composed of either non-viral or viral nanoparticles (NPs). Nanotechnology-based vaccine technology was studied for its potentially transformative advancement of medicine. AREAS COVERED NPs protect the encapsulated mRNA in vaccines, thereby enhancing the stability of the ribonucleic acids and facilitating their intact delivery to their specific targets. Compared to liposomes, lipid nanoparticles (LNPs) are unique and, through their rigid morphology and better cellular penetrability, render enhanced cargo stability. To explore nanotechnology-mediated vaccine delivery and its potential in future pandemics, we assessed articles from various databases, such as PubMed, Embase, and Scopus, including editorial/research notes, expert opinions, and collections of data from several clinical research trials. In the current review, we focus on the nanoparticulate approach of the different SARS-CoV-2 vaccines and explore their success against the pandemic. EXPERT OPINION The mRNA-based vaccines, with their tremendous efficacy of ~95% (under phase III-IV clinical trials) and distinct nanocarriers (LNPs), represent a new medical front alongside DNA and siRNA-based vaccines.
Collapse
Affiliation(s)
- Manoj Kumar Sarangi
- Department of Pharmacy, School of Pharmaceutical Sciences, Sardar Bhagwan Singh University, Dehradun, India
| | - Sasmita Padhi
- Department of Pharmacy, School of Pharmaceutical Sciences, Sardar Bhagwan Singh University, Dehradun, India
| | - Gautam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, India
| | | | - Dong Kee Yi
- Department of Chemistry, Myongji University, Yongin, South Korea
| |
Collapse
|
113
|
Bajoria S, Kaur K, Kumru OS, Van Slyke G, Doering J, Novak H, Rodriguez Aponte SA, Dalvie NC, Naranjo CA, Johnston RS, Silverman JM, Kleanthous H, Love JC, Mantis NJ, Joshi SB, Volkin DB. Antigen-adjuvant interactions, stability, and immunogenicity profiles of a SARS-CoV-2 receptor-binding domain (RBD) antigen formulated with aluminum salt and CpG adjuvants. Hum Vaccin Immunother 2022; 18:2079346. [PMID: 35666264 PMCID: PMC9621007 DOI: 10.1080/21645515.2022.2079346] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023] Open
Abstract
Low-cost, refrigerator-stable COVID-19 vaccines will facilitate global access and improve vaccine coverage in low- and middle-income countries. To this end, subunit-based approaches targeting the receptor-binding domain (RBD) of SARS-CoV-2 Spike protein remain attractive. Antibodies against RBD neutralize SARS-CoV-2 by blocking viral attachment to the host cell receptor, ACE2. Here, a yeast-produced recombinant RBD antigen (RBD-L452K-F490W or RBD-J) was formulated with various combinations of aluminum-salt (Alhydrogel®, AH; AdjuPhos®, AP) and CpG 1018 adjuvants. We assessed the effect of antigen-adjuvant interactions on the stability and mouse immunogenicity of various RBD-J preparations. While RBD-J was 50% adsorbed to AH and <15% to AP, addition of CpG resulted in complete AH binding, yet no improvement in AP adsorption. ACE2 competition ELISA analyses of formulated RBD-J stored at varying temperatures (4, 25, 37°C) revealed that RBD-J was destabilized by AH, an effect exacerbated by CpG. DSC studies demonstrated that aluminum-salt and CpG adjuvants decrease the conformational stability of RBD-J and suggest a direct CpG-RBD-J interaction. Although AH+CpG-adjuvanted RBD-J was the least stable in vitro, the formulation was most potent at eliciting SARS-CoV-2 pseudovirus neutralizing antibodies in mice. In contrast, RBD-J formulated with AP+CpG showed minimal antigen-adjuvant interactions, a better stability profile, but suboptimal immune responses. Interestingly, the loss of in vivo potency associated with heat-stressed RBD-J formulated with AH+CpG after one dose was abrogated by a booster. Our findings highlight the importance of elucidating the key interrelationships between antigen-adjuvant interactions, storage stability, and in vivo performance to enable successful formulation development of stable and efficacious subunit vaccines.
Collapse
Affiliation(s)
- Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Kawaljit Kaur
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Ozan S. Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Greta Van Slyke
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Jennifer Doering
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Hayley Novak
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Sergio A. Rodriguez Aponte
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Neil C. Dalvie
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Christopher A. Naranjo
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan S. Johnston
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | - J. Christopher Love
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Nicholas J. Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Sangeeta B. Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
114
|
Ouyang WO, Tan TJ, Lei R, Song G, Kieffer C, Andrabi R, Matreyek KA, Wu NC. Probing the biophysical constraints of SARS-CoV-2 spike N-terminal domain using deep mutational scanning. SCIENCE ADVANCES 2022; 8:eadd7221. [PMID: 36417523 PMCID: PMC9683733 DOI: 10.1126/sciadv.add7221] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
Increasing the expression level of the SARS-CoV-2 spike (S) protein has been critical for COVID-19 vaccine development. While previous efforts largely focused on engineering the receptor-binding domain (RBD) and the S2 subunit, the amino-terminal domain (NTD) has been long overlooked because of the limited understanding of its biophysical constraints. In this study, the effects of thousands of NTD single mutations on S protein expression were quantified by deep mutational scanning. Our results revealed that in terms of S protein expression, the mutational tolerability of NTD residues was inversely correlated with their proximity to the RBD and S2. We also identified NTD mutations at the interdomain interface that increased S protein expression without altering its antigenicity. Overall, this study not only advances the understanding of the biophysical constraints of the NTD but also provides invaluable insights into S-based immunogen design.
Collapse
Affiliation(s)
- Wenhao O. Ouyang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Timothy J.C. Tan
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ruipeng Lei
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ge Song
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Collin Kieffer
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kenneth A. Matreyek
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Nicholas C. Wu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
115
|
Valenzuela-Fernández A, Cabrera-Rodriguez R, Ciuffreda L, Perez-Yanes S, Estevez-Herrera J, González-Montelongo R, Alcoba-Florez J, Trujillo-González R, García-Martínez de Artola D, Gil-Campesino H, Díez-Gil O, Lorenzo-Salazar JM, Flores C, Garcia-Luis J. Nanomaterials to combat SARS-CoV-2: Strategies to prevent, diagnose and treat COVID-19. Front Bioeng Biotechnol 2022; 10:1052436. [PMID: 36507266 PMCID: PMC9732709 DOI: 10.3389/fbioe.2022.1052436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the associated coronavirus disease 2019 (COVID-19), which severely affect the respiratory system and several organs and tissues, and may lead to death, have shown how science can respond when challenged by a global emergency, offering as a response a myriad of rapid technological developments. Development of vaccines at lightning speed is one of them. SARS-CoV-2 outbreaks have stressed healthcare systems, questioning patients care by using standard non-adapted therapies and diagnostic tools. In this scenario, nanotechnology has offered new tools, techniques and opportunities for prevention, for rapid, accurate and sensitive diagnosis and treatment of COVID-19. In this review, we focus on the nanotechnological applications and nano-based materials (i.e., personal protective equipment) to combat SARS-CoV-2 transmission, infection, organ damage and for the development of new tools for virosurveillance, diagnose and immune protection by mRNA and other nano-based vaccines. All the nano-based developed tools have allowed a historical, unprecedented, real time epidemiological surveillance and diagnosis of SARS-CoV-2 infection, at community and international levels. The nano-based technology has help to predict and detect how this Sarbecovirus is mutating and the severity of the associated COVID-19 disease, thereby assisting the administration and public health services to make decisions and measures for preparedness against the emerging variants of SARS-CoV-2 and severe or lethal COVID-19.
Collapse
Affiliation(s)
- Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Romina Cabrera-Rodriguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Laura Ciuffreda
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Silvia Perez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Judith Estevez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | | | - Julia Alcoba-Florez
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Departamento de Análisis Matemático, Facultad de Ciencias, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | | | - Helena Gil-Campesino
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Oscar Díez-Gil
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - José M. Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Health Sciences, University of Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Jonay Garcia-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
116
|
Chavda VP, Vihol DR, Solanki HK, Apostolopoulos V. The Vaccine World of COVID-19: India’s Contribution. Vaccines (Basel) 2022; 10:vaccines10111943. [PMID: 36423038 PMCID: PMC9695423 DOI: 10.3390/vaccines10111943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) eruption has left not only illness and mortality in its wake, but also an overwhelming threat to health policy, human regality, food security, and struggle worldwide. The accessibility and potential distribution of a protective and successful vaccination to communities throughout the world are being considered now not just, as a potential of overcoming these hurdles, but also as an example of human perseverance in the face of catastrophe. A vaccine is the only tool that can efficaciously deal with the COVID-19 catastrophe. Currently, more than 47 vaccines are permitted for emergency use in distinct parts of the world. India will play a significant role in the development of the high-priced Moderna shots and Pfizer Inc, therefore assisting in the immunization of a large portion of the world. Moreover, many of the internationally researched and developed vaccine laboratories seek manufacturing in Indian firms and companies for efficient and low-cost production of vaccines intending to provide to the world, hence, making India, a major role player during these pandemic times. This review highlights the Indian contribution to the globe for COVID-19 management.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad 380008, Gujarat, India
- Correspondence: (V.P.C.); (V.A.)
| | - Disha R. Vihol
- Pharmacy Section, Griffith University, Gold Coast, QLD 4215, Australia
| | - Hetvi K. Solanki
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad 380008, Gujarat, India
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3021, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
- Correspondence: (V.P.C.); (V.A.)
| |
Collapse
|
117
|
Liu Y, Zhao D, Wang Y, Chen Z, Yang L, Li W, Gong Y, Gan C, Tang J, Zhang T, Tang D, Dong X, Yang Q, Valencia CA, Dai L, Qi S, Dong B, Chow HY, Li Y. A vaccine based on the yeast-expressed receptor-binding domain (RBD) elicits broad immune responses against SARS-CoV-2 variants. Front Immunol 2022; 13:1011484. [PMID: 36439096 PMCID: PMC9682237 DOI: 10.3389/fimmu.2022.1011484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/24/2022] [Indexed: 06/23/2024] Open
Abstract
Development of safe and efficient vaccines is still necessary to deal with the COVID-19 pandemic. Herein, we reported that yeast-expressed recombinant RBD proteins either from wild-type or Delta SARS-CoV-2 were able to elicit immune responses against SARS-CoV-2 and its variants. The wild-type RBD (wtRBD) protein was overexpressed in Pichia pastoris, and the purified protein was used as the antigen to immunize mice after formulating an aluminium hydroxide (Alum) adjuvant. Three immunization programs with different intervals were compared. It was found that the immunization with an interval of 28 days exhibited the strongest immune response to SARS-CoV-2 than the one with an interval of 14 or 42 days based on binding antibody and the neutralizing antibody (NAb) analyses. The antisera from the mice immunized with wtRBD were able to neutralize the Beta variant with a similar efficiency but the Delta variant with 2~2.5-fold decreased efficiency. However, more NAbs to the Delta variant were produced when the Delta RBD protein was used to immunize mice. Interestingly, the NAbs may cross react with the Omicron variant. To increase the production of NAbs, the adjuvant combination of Alum and CpG oligonucleotides was used. Compared with the Alum adjuvant alone, the NAbs elicited by the combined adjuvants exhibited an approximate 10-fold increase for the Delta and a more than 53-fold increase for the Omicron variant. This study suggested that yeast-derived Delta RBD is a scalable and an effective vaccine candidate for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Yu Liu
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Danhua Zhao
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yichang Wang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhian Chen
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wenjuan Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunmei Gan
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jieshi Tang
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Tizhong Zhang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Tang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuju Dong
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qingzhe Yang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - C. Alexander Valencia
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shiqian Qi
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Real & Best Biotech Co., Ltd., Chengdu, China
| | - Hoi Yee Chow
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhua Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
118
|
Marchese AM, Beyhaghi H, Orenstein WA. With established safe and effective use, protein vaccines offer another choice against COVID-19. Vaccine 2022; 40:6567-6569. [PMID: 36210248 PMCID: PMC9515329 DOI: 10.1016/j.vaccine.2022.09.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/12/2022] [Accepted: 09/21/2022] [Indexed: 01/27/2023]
Affiliation(s)
- Anthony M. Marchese
- Novavax, Inc., Gaithersburg, MD, United States,Corresponding author at: Novavax Inc, 21 Firstfield Rd, Gaithersburg, MD 20878, United States
| | | | | |
Collapse
|
119
|
Wang R, Huang X, Cao T, Sun C, Luo D, Qiu H, Wu M, Huang X, Yu C, Li J, Kong D, Ma J, Zhang X, Hu P, Zhang Y, Luo C, Zhao H, Li Y, Deng Y, Qin C, Xie L. Development of a thermostable SARS-CoV-2 variant-based bivalent protein vaccine with cross-neutralizing potency against Omicron subvariants. Virology 2022; 576:61-68. [PMID: 36174448 PMCID: PMC9486464 DOI: 10.1016/j.virol.2022.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/23/2022] [Accepted: 09/05/2022] [Indexed: 01/07/2023]
Abstract
SARS-CoV-2 variants have posed significant challenges to the hopes of using ancestral strain-based vaccines to address the risk of breakthrough infection by variants. We designed and developed a bivalent vaccine based on SARS-CoV-2 Alpha and Beta variants (named SCTV01C). SCTV01C antigens were stable at 25 oC for at least 6 months. In the presence of a squalene-based oil-in-water adjuvant SCT-VA02B, SCTV01C showed significant protection efficacy against antigen-matched Beta variant, with favorable safety profiles in rodents. Notably, SCTV01C exhibited cross-neutralization capacity against Omicron subvariants (BA.1, BA.1.1, BA.2, BA.3, and BA.4/5) in mice, superior to a WT (D614G)-based vaccine, which reinforced our previously published findings that SCTV01C exhibited broad-spectrum neutralizing potencies against over a dozen pre-Omicron variants and the Omicron BA.1 variant. In summary, variant-based multivalent protein vaccine could be a platform approach to address the challenging issues of emerging variants, vaccine hesitancy and the needs of affordable and thermal stable vaccines.
Collapse
Affiliation(s)
- Rui Wang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Xun Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Tianshu Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Chunyun Sun
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Dan Luo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Hongying Qiu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Mei Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Xingyao Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Chulin Yu
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Jing Li
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Desheng Kong
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Juan Ma
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Xiao Zhang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Ping Hu
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Yanjing Zhang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Chunxia Luo
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China
| | - Hui Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Yuchang Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Yongqiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Chengfeng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, 100176, China; Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
120
|
Tran KTM, Gavitt TD, Le TT, Graichen A, Lin F, Liu Y, Tulman ER, Szczepanek SM, Nguyen TD. A Single-Administration Microneedle Skin Patch for Multi-Burst Release of Vaccine against SARS-CoV-2. ADVANCED MATERIALS TECHNOLOGIES 2022; 8:2200905. [PMID: 36714215 PMCID: PMC9874724 DOI: 10.1002/admt.202200905] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/25/2022] [Indexed: 06/18/2023]
Abstract
The necessity for multiple injections and cold-chain storage has contributed to suboptimal vaccine utilization, especially in pandemic situations. Thermally-stable and single-administration vaccines hold a great potential to revolutionize the global immunization process. Here, a new approach to thermally stabilize protein-based antigens is presented and a new high-throughput antigen-loading process is devised to create a single-administration, pulsatile-release microneedle (MN) patch which can deliver a recombinant SARS-CoV-2 S1-RBD protein-a model for the COVID-19 vaccine. Nearly 100% of the protein antigen could be stabilized at temperatures up to 100 °C for at least 1 h and at an average human body temperature (37 °C) for up to 4 months. Arrays of the stabilized S1-RBD formulations can be loaded into the MN shells via a single-alignment assembly step. The fabricated MNs are administered at a single time into the skin of rats and induce antibody response which could neutralize authentic SARS-CoV-2 viruses, providing similar immunogenic effect to that induced by multiple bolus injections of the same antigen stored in conventional cold-chain conditions. The MN system presented herein could offer the key solution to global immunization campaigns by avoiding low patient compliance, the requirement for cold-chain storage, and the need for multiple booster injections.
Collapse
Affiliation(s)
- Khanh T. M. Tran
- Department of Biomedical EngineeringUniversity of Connecticut181 Auditorium RoadStorrs06269USA
| | - Tyler D. Gavitt
- Department of Pathobiology and Veterinary ScienceCenter of Excellence for Vaccine ResearchUniversity of Connecticut61 North Eagleville RoadStorrs06269USA
| | - Thinh T. Le
- Department of Mechanical EngineeringUniversity of Connecticut191 Auditorium RoadStorrs06269USA
| | - Adam Graichen
- Department of ChemistryUniversity of Connecticut55 North Eagleville RoadStorrs06269USA
| | - Feng Lin
- Department of Mechanical EngineeringUniversity of Connecticut191 Auditorium RoadStorrs06269USA
| | - Yang Liu
- Department of Mechanical EngineeringUniversity of Connecticut191 Auditorium RoadStorrs06269USA
| | - Edan R. Tulman
- Department of Pathobiology and Veterinary ScienceCenter of Excellence for Vaccine ResearchUniversity of Connecticut61 North Eagleville RoadStorrs06269USA
| | - Steven M. Szczepanek
- Department of Pathobiology and Veterinary ScienceCenter of Excellence for Vaccine ResearchUniversity of Connecticut61 North Eagleville RoadStorrs06269USA
| | - Thanh D. Nguyen
- Department of Biomedical EngineeringUniversity of Connecticut181 Auditorium RoadStorrs06269USA
- Department of Mechanical EngineeringUniversity of Connecticut191 Auditorium RoadStorrs06269USA
| |
Collapse
|
121
|
Tulimilli SV, Dallavalasa S, Basavaraju CG, Kumar Rao V, Chikkahonnaiah P, Madhunapantula SV, Veeranna RP. Variants of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and Vaccine Effectiveness. Vaccines (Basel) 2022; 10:1751. [PMID: 36298616 PMCID: PMC9607623 DOI: 10.3390/vaccines10101751] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
The incidence and death toll due to SARS-CoV-2 infection varied time-to-time; and depended on several factors, including severity (viral load), immune status, age, gender, vaccination status, and presence of comorbidities. The RNA genome of SARS-CoV-2 has mutated and produced several variants, which were classified by the SARS-CoV-2 Interagency Group (SIG) into four major categories. The first category; “Variant Being Monitored (VBM)”, consists of Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), Epsilon (B.1.427, B.1.429), Eta (B.1.525), Iota (B.1.526), Kappa (B.1.617.1), Mu (B.1.621), and Zeta (P.2); the second category; “Variants of Concern” consists of Omicron (B.1.1.529). The third and fourth categories include “Variants of Interest (VOI)”, and “Variants of High Consequence (VOHC)”, respectively, and contain no variants classified currently under these categories. The surge in VBM and VOC poses a significant threat to public health globally as they exhibit altered virulence, transmissibility, diagnostic or therapeutic escape, and the ability to evade the host immune response. Studies have shown that certain mutations increase the infectivity and pathogenicity of the virus as demonstrated in the case of SARS-CoV-2, the Omicron variant. It is reported that the Omicron variant has >60 mutations with at least 30 mutations in the Spike protein (“S” protein) and 15 mutations in the receptor-binding domain (RBD), resulting in rapid attachment to target cells and immune evasion. The spread of VBM and VOCs has affected the actual protective efficacy of the first-generation vaccines (ChAdOx1, Ad26.COV2.S, NVX-CoV2373, BNT162b2). Currently, the data on the effectiveness of existing vaccines against newer variants of SARS-CoV-2 are very scanty; hence additional studies are immediately warranted. To this end, recent studies have initiated investigations to elucidate the structural features of crucial proteins of SARS-CoV-2 variants and their involvement in pathogenesis. In addition, intense research is in progress to develop better preventive and therapeutic strategies to halt the spread of COVID-19 caused by variants. This review summarizes the structure and life cycle of SARS-CoV-2, provides background information on several variants of SARS-CoV-2 and mutations associated with these variants, and reviews recent studies on the safety and efficacy of major vaccines/vaccine candidates approved against SARS-CoV-2, and its variants.
Collapse
Affiliation(s)
- SubbaRao V. Tulimilli
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570004, Karnataka, India
| | - Siva Dallavalasa
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570004, Karnataka, India
| | - Chaithanya G. Basavaraju
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570004, Karnataka, India
| | - Vinay Kumar Rao
- Department of Medical Genetics, JSS Medical College & Hospital, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India
| | - Prashanth Chikkahonnaiah
- Department of Pulmonary Medicine, Mysore Medical College and Research Institute, Mysuru 570001, Karnataka, India
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570004, Karnataka, India
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570004, Karnataka, India
| | - Ravindra P. Veeranna
- Department of Biochemistry, Council of Scientific and Industrial Research (CSIR)-Central Food Technological Research Institute (CFTRI), Mysuru 570020, Karnataka, India
| |
Collapse
|
122
|
Helmy SA, El-Morsi RM, Helmy SAM, El-Masry SM. Towards novel nano-based vaccine platforms for SARS-CoV-2 and its variants of concern: Advances, challenges and limitations. J Drug Deliv Sci Technol 2022; 76:103762. [PMID: 36097606 PMCID: PMC9452404 DOI: 10.1016/j.jddst.2022.103762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/07/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022]
Abstract
Vaccination is the most effective tool available for fighting the spread of COVID-19. Recently, emerging variants of SARS-CoV-2 have led to growing concerns about increased transmissibility and decreased vaccine effectiveness. Currently, many vaccines are approved for emergency use and more are under development. This review highlights the ongoing advances in the design and development of different nano-based vaccine platforms. The challenges, limitations, and ethical consideration imposed by these nanocarriers are also discussed. Further, the effectiveness of the leading vaccine candidates against all SARS-CoV-2 variants of concern are highlighted. The review also focuses on the possibility of using an alternative non-invasive routes of vaccine administration using micro and nanotechnologies to enhance vaccination compliance and coverage.
Collapse
Affiliation(s)
- Sally A Helmy
- Department of Clinical and Hospital Pharmacy, Faculty of Pharmacy, Taibah University, AL-Madinah AL-Munawarah, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Rasha M El-Morsi
- Department of Microbiology and Immunology, Faculty of Pharmacy, Delta University for Science and Technology, Egypt
| | - Soha A M Helmy
- Department of Languages and Translation, College of Arts and Humanities, Taibah University, AL-Madinah AL-Munawarah, Saudi Arabia
- Department of Foreign Languages, Faculty of Education, Tanta University, Tanta, Egypt
| | - Soha M El-Masry
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| |
Collapse
|
123
|
Khorattanakulchai N, Srisutthisamphan K, Shanmugaraj B, Manopwisedjaroen S, Rattanapisit K, Panapitakkul C, Kemthong T, Suttisan N, Malaivijitnond S, Thitithanyanont A, Jongkaewwattana A, Phoolcharoen W. A recombinant subunit vaccine candidate produced in plants elicits neutralizing antibodies against SARS-CoV-2 variants in macaques. FRONTIERS IN PLANT SCIENCE 2022; 13:901978. [PMID: 36247553 PMCID: PMC9555276 DOI: 10.3389/fpls.2022.901978] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/29/2022] [Indexed: 06/16/2023]
Abstract
Since the outbreak of the coronavirus disease (COVID) pandemic in 2019, the development of effective vaccines to combat the infection has been accelerated. With the recent emergence of highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOC), there are concerns regarding the immune escape from vaccine-induced immunity. Hence an effective vaccine against VOC with a potent immune response is required. Our previous study confirmed that the two doses of the plant-produced receptor-binding domain (RBD) of SARS-CoV-2 fused with the Fc region of human IgG1, namely Baiya SARS-CoV-2 Vax 1, showed high immunogenicity in mice and monkeys. Here, we aimed to evaluate the immunogenicity of a three-dose intramuscular injection of Baiya SARS-CoV-2 Vax 1 on days 0, 21, and 133 in cynomolgus monkeys. At 14 days after immunization, blood samples were collected to determine RBD-specific antibody titer, neutralizing antibody, and pseudovirus neutralizing antibody titers. Immunized monkeys developed significantly high levels of antigen-specific antibodies against SARS-CoV-2 compared to the control group. Interestingly, the sera collected from immunized monkeys also showed a neutralizing antibody response against the SARS-CoV-2 VOCs; Alpha, Beta, Gamma, Delta, and Omicron. These findings demonstrate that a three-dose regimen of Baiya SARS-CoV-2 Vax 1 vaccine elicits neutralizing immune response against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Narach Khorattanakulchai
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Kanjana Srisutthisamphan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | | | | | | | - Chalisa Panapitakkul
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Taratorn Kemthong
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi, Thailand
| | - Nutchanat Suttisan
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi, Thailand
| | | | | | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
124
|
Selective reversed-phase high-performance liquid chromatography method for the determination of intact SARS-CoV-2 spike protein. J Chromatogr A 2022; 1680:463424. [PMID: 36007475 PMCID: PMC9378212 DOI: 10.1016/j.chroma.2022.463424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 08/08/2022] [Accepted: 08/14/2022] [Indexed: 01/07/2023]
Abstract
Protein-based vaccines are playing an increasingly important role in the COVID-19 pandemic. As late-stage clinical data are finalized and released, the number of protein-based vaccines expected to enter the market will increase significantly. Most protein-based COVID-19 vaccines are based on the SARS-CoV-2 spike protein (S-protein), which plays a major role in viral attachment to human cells and infection. As a result, in order to develop and manufacture quality vaccines consistently, it is imperative to have access to selective and efficient methods for the bioanalytical assessment of S-protein. In this study, samples of recombinant S-protein (hexS-protein) and commercial S-protein were used to develop a selective reversed-phase HPLC (RP-HPLC) method that enabled elution of the intact S-protein monomer as a single peak on a wide pore, C8-bonded chromatographic column. The S-protein subunits, S1 and S2 subunits, were clearly separated from intact S-protein and identified. The results of this study set the foundation for reversed-phase HPLC method development and analysis for selective and efficient separation of S-protein monomer from its subunits.
Collapse
|
125
|
Fahr S, Peña-Benavides SA, Thiel L, Sengoba C, Karacasulu K, Ihling N, Sosa-Hernández JE, Gilleskie G, Woodley JM, Parra-Saldivar R, Mansouri SS, Roh K. Mobile On Demand COVID-19 Vaccine Production Units for Developing Countries. Ind Eng Chem Res 2022. [DOI: 10.1021/acs.iecr.2c01217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Steffen Fahr
- Process Systems Engineering (AVT.SVT), RWTH Aachen University, 52074 Aachen, Germany
- Institute of Plant and Process Technology, Technical University of Munich, 85748 Garching, Germany
| | - Samantha Ayde Peña-Benavides
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Lukas Thiel
- Process Systems Engineering (AVT.SVT), RWTH Aachen University, 52074 Aachen, Germany
| | - Carl Sengoba
- Process Systems Engineering (AVT.SVT), RWTH Aachen University, 52074 Aachen, Germany
| | - Kaan Karacasulu
- Process Systems Engineering (AVT.SVT), RWTH Aachen University, 52074 Aachen, Germany
| | - Nina Ihling
- Biochemical Engineering (AVT.BioVT), RWTH Aachen University, 52074 Aachen, Germany
| | | | - Gary Gilleskie
- Golden LEAF Biomanufacturing Training and Education Center (BTEC), NC State University, Raleigh, North Carolina 27606, United States
| | - John M. Woodley
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | | | - Seyed Soheil Mansouri
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Kosan Roh
- Process Systems Engineering (AVT.SVT), RWTH Aachen University, 52074 Aachen, Germany
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, 34141 Daejeon, Republic of Korea
| |
Collapse
|
126
|
Beeraka NM, Sukocheva OA, Lukina E, Liu J, Fan R. Development of antibody resistance in emerging mutant strains of SARS CoV-2: Impediment for COVID-19 vaccines. Rev Med Virol 2022; 32:e2346. [PMID: 35416390 PMCID: PMC9111059 DOI: 10.1002/rmv.2346] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/28/2022] [Accepted: 03/06/2022] [Indexed: 02/05/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), a highly infectious agent associated with unprecedented morbidity and mortality. A failure to stop growth of COVID-19-linked morbidity rates is caused by SARS-CoV-2 mutations and the emergence of new highly virulent SARS-CoV-2 strains. Several acquired SARS-CoV-2 mutations reflect viral adaptations to host immune defence. Mutations in the virus Spike-protein were associated with the lowered effectiveness of current preventive therapies, including vaccines. Recent in vitro studies detected diminished neutralisation capacity of vaccine-induced antibodies, which are targeted to bind Spike receptor-binding and N-terminal domains in the emerging strains. Lower than expected inhibitory activity of antibodies was reported against viruses with E484K Spike mutation, including B.1.1.7 (UK), P.1 (Brazil), B.1.351 (South African), and new Omicron variant (B.1.1.529) with E484A mutation. The vaccine effectiveness is yet to be examined against new mutant strains of SARS-CoV-2 originating in Europe, Nigeria, Brazil, South Africa, and India. To prevent the loss of anti-viral protection in vivo, often defined as antibody resistance, it is required to target highly conserved viral sequences (including Spike protein) and enhance the potency of antibody cocktails. In this review, we assess the reported mutation-acquiring potential of coronaviruses and compare efficacies of current COVID-19 vaccines against 'parent' and 'mutant' strains of SARS-CoV-2 (Kappa (B.1.617.1), Delta (B.1.617.2), and Omicron (B.1.1.529)).
Collapse
Affiliation(s)
- Narasimha M. Beeraka
- Department of Radiation OncologyCancer CenterThe First Affiliated Hospital of ZhengzhouZhengzhouChina
- Department of Human AnatomyI.M. Sechenov First Moscow State Medical University (Sechenov University)MoscowRussian Federation
| | - Olga A. Sukocheva
- Discipline of Health SciencesCollege of Nursing and Health SciencesFlinders University of South AustraliaBedford ParkAustralia
| | - Elena Lukina
- Discipline of BiologyCollege of SciencesFlinders University of South AustraliaBedford ParkAustralia
| | - Junqi Liu
- Department of Radiation OncologyCancer CenterThe First Affiliated Hospital of ZhengzhouZhengzhouChina
| | - Ruitai Fan
- Department of Radiation OncologyCancer CenterThe First Affiliated Hospital of ZhengzhouZhengzhouChina
| |
Collapse
|
127
|
Khorattanakulchai N, Manopwisedjaroen S, Rattanapisit K, Panapitakkul C, Kemthong T, Suttisan N, Srisutthisamphan K, Malaivijitnond S, Thitithanyanont A, Jongkaewwattana A, Shanmugaraj B, Phoolcharoen W. Receptor binding domain proteins of SARS-CoV-2 variants produced in Nicotiana benthamiana elicit neutralizing antibodies against variants of concern. J Med Virol 2022; 94:4265-4276. [PMID: 35615895 PMCID: PMC9348024 DOI: 10.1002/jmv.27881] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 11/15/2022]
Abstract
The constantly emerging severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) variants of concerns (VOCs) with mutations in the receptor-binding domain (RBD) spread rapidly and has become a severe public health problem worldwide. Effective vaccines and optimized booster vaccination strategies are thus highly required. Here, the gene encoding six different RBD (Alpha, Beta, Gamma, Kappa, Delta, and Epsilon variants) along with the Fc fragment of human IgG1 (RBD-Fc) was cloned into plant expression vector and produced in Nicotiana benthamiana by transient expression. Further, the immunogenicity of plant-produced variant RBD-Fc fusion proteins were tested in cynomolgus monkeys. Each group of cynomolgus monkeys was immunized three times intramuscularly with variant RBD-Fc vaccines at Day 0, 21, 42, and neutralizing antibody responses were evaluated against ancestral (Wuhan), Alpha, Beta, Gamma, and Delta variants. The results showed that three doses of the RBD-Fc vaccine significantly enhanced the immune response against all tested SARS-CoV-2 variants. In particular, the vaccines based on Delta and Epsilon mutant RBD elicit broadly neutralizing antibodies against ancestral (Wuhan), Alpha, and Delta SARS-CoV-2 variants whereas Beta and Gamma RBD-Fc vaccines elicit neutralizing antibodies against their respective SARS-CoV-2 strains. The Delta and Epsilon RBD-Fc based vaccines displayed cross-reactive immunogenicity and might be applied as a booster vaccine to induce broadly neutralizing antibodies. These proof-of-concept results will be helpful for the development of plant-derived RBD-Fc-based vaccines against SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Narach Khorattanakulchai
- Center of Excellence in Plant‐produced PharmaceuticalsChulalongkorn UniversityBangkokThailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical SciencesChulalongkorn UniversityBangkokThailand
| | | | | | - Chalisa Panapitakkul
- Center of Excellence in Plant‐produced PharmaceuticalsChulalongkorn UniversityBangkokThailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical SciencesChulalongkorn UniversityBangkokThailand
| | - Taratorn Kemthong
- National Primate Research Center of ThailandChulalongkorn UniversitySaraburiThailand
| | - Nutchanat Suttisan
- National Primate Research Center of ThailandChulalongkorn UniversitySaraburiThailand
| | - Kanjana Srisutthisamphan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC)National Science and Technology Development AgencyPathumthaniThailand
| | | | | | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC)National Science and Technology Development AgencyPathumthaniThailand
| | | | - Waranyoo Phoolcharoen
- Center of Excellence in Plant‐produced PharmaceuticalsChulalongkorn UniversityBangkokThailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical SciencesChulalongkorn UniversityBangkokThailand
| |
Collapse
|
128
|
Rahbar Z, Nazarian S, Dorostkar R, Sotoodehnejadnematalahi F, Amani J. Recombinant expression of SARS-CoV-2 receptor binding domain (RBD) in Escherichia coli and its immunogenicity in mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1110-1116. [PMID: 36246069 PMCID: PMC9526882 DOI: 10.22038/ijbms.2022.65045.14333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022]
Abstract
Objectives The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), giving rise to the coronavirus disease 2019 (COVID-19), has become a danger to wellbeing worldwide. Thus, finding efficient and safe vaccines for COVID-19 is of great importance. As a basic step amid contamination, SARS-CoV-2 employs the receptor-binding domain (RBD) of the spike protein to lock in with the receptor angiotensin-converting enzyme 2 (ACE2) on host cells. SARS-CoV-2 receptor-binding domain (RBD) is the main human antibody target for developing vaccines and virus inhibitors, as well as neutralizing antibodies. A bacterial procedure was developed for the expression and purification of the SARS-CoV-2 spike protein receptor-binding domain. Materials and Methods In this research study, RBD was expressed by Escherichia coli and purified with Ni-NTA chromatography. Then it was affirmed by the western blot test. The immunogenicity and protective efficacy of RBD recombinant protein were assessed on BALB/c mice. Additionally, RBD recombinant protein was tested by ELISA utilizing sera of COVID-19 healing patients contaminated with SARS-CoV-2 wild type and Delta variation. Results Indirect ELISA was able to detect the protein RBD in serum of the immunized mouse expressed in E. coli. The inactive SARS-CoV2 was detected by antibodies within the serum of immunized mice. Serum antibodies from individuals recovered from Covid19 reacted to the expressed protein. Conclusion Our findings showed that RBD is of great importance in vaccine design and it can be used to develop recombinant vaccines through induction of antibodies against RBD.
Collapse
Affiliation(s)
- Zahra Rahbar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Ruhollah Dorostkar
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Iran
| | | | - Jafar Amani
- Applied Microbiology Research Center, System Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
129
|
Soto LF, Romaní AC, Jiménez-Avalos G, Silva Y, Ordinola-Ramirez CM, Lopez Lapa RM, Requena D. Immunoinformatic analysis of the whole proteome for vaccine design: An application to Clostridium perfringens. Front Immunol 2022; 13:942907. [PMID: 36110855 PMCID: PMC9469472 DOI: 10.3389/fimmu.2022.942907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/02/2022] [Indexed: 11/21/2022] Open
Abstract
Clostridium perfringens is a dangerous bacterium and known biological warfare weapon associated with several diseases, whose lethal toxins can produce necrosis in humans. However, there is no safe and fully effective vaccine against C. perfringens for humans yet. To address this problem, we computationally screened its whole proteome, identifying highly immunogenic proteins, domains, and epitopes. First, we identified that the proteins with the highest epitope density are Collagenase A, Exo-alpha-sialidase, alpha n-acetylglucosaminidase and hyaluronoglucosaminidase, representing potential recombinant vaccine candidates. Second, we further explored the toxins, finding that the non-toxic domain of Perfringolysin O is enriched in CTL and HTL epitopes. This domain could be used as a potential sub-unit vaccine to combat gas gangrene. And third, we designed a multi-epitope protein containing 24 HTL-epitopes and 34 CTL-epitopes from extracellular regions of transmembrane proteins. Also, we analyzed the structural properties of this novel protein using molecular dynamics. Altogether, we are presenting a thorough immunoinformatic exploration of the whole proteome of C. perfringens, as well as promising whole-protein, domain-based and multi-epitope vaccine candidates. These can be evaluated in preclinical trials to assess their immunogenicity and protection against C. perfringens infection.
Collapse
Affiliation(s)
- Luis F. Soto
- Escuela Profesional de Genética y Biotecnología, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Ana C. Romaní
- Escuela Profesional de Genética y Biotecnología, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Gabriel Jiménez-Avalos
- Departamento de Ciencias Celulares y Moleculares, Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Yshoner Silva
- Departamento de Salud Pública, Facultad de Ciencias de la Salud, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - Carla M. Ordinola-Ramirez
- Departamento de Salud Pública, Facultad de Ciencias de la Salud, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - Rainer M. Lopez Lapa
- Departamento de Salud Pública, Facultad de Ciencias de la Salud, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
- Instituto de Ganadería y Biotecnología, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - David Requena
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, United States
- *Correspondence: David Requena,
| |
Collapse
|
130
|
Shanmugaraj B, Khorattanakulchai N, Panapitakkul C, Malla A, Im-Erbsin R, Inthawong M, Sunyakumthorn P, Hunsawong T, Klungthong C, Reed MC, Kemthong T, Suttisan N, Malaivijitnond S, Srimangkornkaew P, Klinkhamhom A, Manopwisedjaroen S, Thitithanyanont A, Taychakhoonavudh S, Phoolcharoen W. Preclinical evaluation of a plant-derived SARS-CoV-2 subunit vaccine: Protective efficacy, immunogenicity, safety, and toxicity. Vaccine 2022; 40:4440-4452. [PMID: 35697573 PMCID: PMC9167921 DOI: 10.1016/j.vaccine.2022.05.087] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/06/2022] [Accepted: 05/31/2022] [Indexed: 01/01/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an acute respiratory illness caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The prevention of SARS-CoV-2 transmission has become a global priority. Previously, we showed that a protein subunit vaccine that was developed based on the fusion of the SARS-CoV-2 receptor-binding domain (RBD) to the Fc portion of human IgG1 (RBD-Fc), produced in Nicotiana benthamiana, and adjuvanted with alum, namely, Baiya SARS-CoV-2 Vax 1, induced potent immunological responses in both mice and cynomolgus monkeys. Hence, this study evaluated the protective efficacy, safety, and toxicity of Baiya SARS-CoV-2 Vax 1 in K18-hACE2 mice, monkeys and Wistar rats. Two doses of vaccine were administered three weeks apart on Days 0 and 21. The administration of the vaccine to K18-hACE2 mice reduced viral loads in the lungs and brains of the vaccinated animals and protected the mice against challenge with SARS-CoV-2. In monkeys, the results of safety pharmacology tests, general clinical observations, and a core battery of studies of three vital systems, namely, the central nervous, cardiovascular, and respiratory systems, did not reveal any safety concerns. The toxicology study of the vaccine in rats showed no vaccine-related pathological changes, and all the animals remained healthy under the conditions of this study. Furthermore, the vaccine did not cause any abnormal toxicity in rats and was clinically tolerated even at the highest tested concentration. In addition, general health status, body temperature, local toxicity at the administration site, hematology, and blood chemistry parameters were also monitored. Overall, this work presents the results of the first systematic study of the safety profile of a plant-derived vaccine, Baiya SARS-CoV-2 Vax 1; this approach can be considered a viable strategy for the development of vaccines against COVID-19.
Collapse
Affiliation(s)
| | - Narach Khorattanakulchai
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chalisa Panapitakkul
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Rawiwan Im-Erbsin
- Department of Veterinary Medicine, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Manutsanun Inthawong
- Department of Veterinary Medicine, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Piyanate Sunyakumthorn
- Department of Veterinary Medicine, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Taweewun Hunsawong
- Department of Virology, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Chonticha Klungthong
- Department of Virology, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Matthew C Reed
- Department of Veterinary Medicine, U.S. Army Medical Directorate-Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Taratorn Kemthong
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | - Nutchanat Suttisan
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | - Suchinda Malaivijitnond
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi 18110, Thailand
| | | | - Aekkarin Klinkhamhom
- National Laboratory Animal Center, Mahidol University, Nakorn Pathom 73170, Thailand
| | | | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Suthira Taychakhoonavudh
- Department of Social and Administrative Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
131
|
Quinlan EJ, Chubet R, Leonardi P. A novel SARS-CoV-2 subunit vaccine engineered on an immune-activating platform technology. Hum Vaccin Immunother 2022; 18:2062971. [PMID: 35801956 DOI: 10.1080/21645515.2022.2062971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
While there are several SARS-CoV-2 vaccines currently available, additional options must be provided that are safe, effective, and affordable for the entire global population. We have developed a novel immune activating platform technology that will fill this need. This recombinant platform protein is produced in insect cells using baculoviral expression technology similar to what is currently used for several other approved vaccines as well as employed by myriad GMP facilities globally. Thus, infrastructure exists for rapid scale up following initial optimizations. Here we report initial results for a SARS-CoV-2 vaccine (OMN008) based on our platform technology. Unadjuvanted OMN008 vaccination resulted in robust antigenicity and neutralization. Additionally, OMN008 vaccination induced a specific CD8 T-cell response. All of these results taken together indicate OMN008 may be an excellent candidate to fill gaps left by the currently available vaccines. Further testing is necessary to fully optimize production; however, overall cost of production should remain low given the simple formulation of this recombinant platform.
Collapse
Affiliation(s)
| | - Richard Chubet
- Research and Development, OmniCyte LLC, Guilford, CT, USA
| | - Peter Leonardi
- Research and Development, OmniCyte LLC, Guilford, CT, USA
| |
Collapse
|
132
|
Azali MA, Mohamed S, Harun A, Hussain FA, Shamsuddin S, Johan MF. Application of Baculovirus Expression Vector system (BEV) for COVID-19 diagnostics and therapeutics: a review. J Genet Eng Biotechnol 2022; 20:98. [PMID: 35792966 PMCID: PMC9259773 DOI: 10.1186/s43141-022-00368-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/20/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND The baculovirus expression vector system has been developed for expressing a wide range of proteins, including enzymes, glycoproteins, recombinant viruses, and vaccines. The availability of the SARS-CoV-2 genome sequence has enabled the synthesis of SARS-CoV2 proteins in a baculovirus-insect cell platform for various applications. The most cloned SARS-CoV-2 protein is the spike protein, which plays a critical role in SARS-CoV-2 infection. It is available in its whole length or as subunits like S1 or the receptor-binding domain (RBD). Non-structural proteins (Nsps), another recombinant SARS-CoV-2 protein generated by the baculovirus expression vector system (BEV), are used in the identification of new medications or the repurposing of existing therapies for the treatment of COVID-19. Non-SARS-CoV-2 proteins generated by BEV for SARS-CoV-2 diagnosis or treatment include moloney murine leukemia virus reverse transcriptase (MMLVRT), angiotensin converting enzyme 2 (ACE2), therapeutic proteins, and recombinant antibodies. The recombinant proteins were modified to boost the yield or to stabilize the protein. CONCLUSION This review covers the wide application of the recombinant protein produced using the baculovirus expression technology for COVID-19 research. A lot of improvements have been made to produce functional proteins with high yields. However, there is still room for improvement and there are parts of this field of research that have not been investigated yet.
Collapse
Affiliation(s)
- Muhammad Azharuddin Azali
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- School of Agriculture Science and Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, 22200, Besut, Terengganu, Malaysia
| | - Salmah Mohamed
- School of Agriculture Science and Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, 22200, Besut, Terengganu, Malaysia
| | - Azian Harun
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Faezahtul Arbaeyah Hussain
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Muhammad Farid Johan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
133
|
Muhar BK, Nehira J, Malhotra A, Kotchoni SO. The Race for COVID-19 Vaccines: The Various Types and Their Strengths and Weaknesses. J Pharm Pract 2022:8971900221097248. [PMID: 35723017 PMCID: PMC9207585 DOI: 10.1177/08971900221097248] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
SARS-CoV-2 causes the highly contagious coronavirus disease (COVID-19), first discovered in Wuhan, China, in December of 2019. As of August 21, 2021, over 211 million people have been diagnosed with COVID-19 and 4.42 million people have died from the disease worldwide. The COVID-19 pandemic has adversely affected world economies, global public health infrastructure, and social behaviors. Despite physical distancing and the advent of symptomatic and monoclonal antibody therapies, perhaps the most effective method to combat COVID-19 remains the creation of immunity through vaccines. Scientific communities globally have been diligently working to develop vaccines since the start of the pandemic. Though a few have been authorized for use, the Pfizer vaccine was the first to be given full approval in the United States in August 2021 – being the quickest vaccine to ever be developed. Although several vaccines produced via different approaches are in use, no mortality has been reported thus far from vaccine use. Here, we highlight the latest advances in the development of the COVID-19 vaccines, specifically the lead candidates that are in late-stage clinical trials or authorized for emergency use. As SARS-CoV-2 uses its spike protein to enter a host cell and cause infection, most vaccine candidates target this protein. This review describes the various COVID-19 vaccines - authorized and/or under development - and their composition, advantages, and potential limitations as the world continues to fight this devastating pandemic.
Collapse
Affiliation(s)
- Bahaar K Muhar
- College of Graduate Studies, 436933California Northstate University, Elk-Grove, CA, USA
| | - Jeffrey Nehira
- College of Pharmacy, 436933California Northstate University, Elk-Grove, CA, USA
| | - Ashim Malhotra
- College of Pharmacy, 436933California Northstate University, Elk-Grove, CA, USA
| | - Simeon O Kotchoni
- College of Graduate Studies, 436933California Northstate University, Elk-Grove, CA, USA.,College of Pharmacy, 436933California Northstate University, Elk-Grove, CA, USA.,College of Medicine, 436933California Northstate University, Elk-Grove, CA, USA
| |
Collapse
|
134
|
Pollet J, Strych U, Chen WH, Versteeg L, Keegan B, Zhan B, Wei J, Liu Z, Lee J, Kundu R, Adhikari R, Poveda C, Jose Villar M, Rani Thimmiraju S, Lopez B, Gillespie PM, Ronca S, Kimata JT, Reers M, Paradkar V, Hotez PJ, Elena Bottazzi M. Receptor-binding domain recombinant protein on alum-CpG induces broad protection against SARS-CoV-2 variants of concern. Vaccine 2022; 40:3655-3663. [PMID: 35568591 PMCID: PMC9080055 DOI: 10.1016/j.vaccine.2022.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 03/18/2022] [Accepted: 05/03/2022] [Indexed: 01/17/2023]
Abstract
We conducted preclinical studies in mice using a yeast-produced SARS-CoV-2 RBD subunit vaccine candidate formulated with aluminum hydroxide (alum) and CpG deoxynucleotides. This formulation is equivalent to the CorbevaxTM vaccine that recently received emergency use authorization by the Drugs Controller General ofIndia. We compared the immune response of mice vaccinated with RBD/alum to mice vaccinated with RBD/alum + CpG. We also evaluated mice immunized with RBD/alum + CpG and boosted with RBD/alum. Mice were immunized twice intramuscularly at a 21-day interval. Compared to two doses of the /alum formulation, the RBD/alum + CpG vaccine induced a stronger and more balanced Th1/Th2 cellular immune response, with high levels of neutralizing antibodies against the original Wuhan isolate of SARS-CoV-2 as well as the B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 and (Delta) variants. Neutralizing antibody titers against the B.1.1.529 (BA.1, Omicron) variant exceeded those in human convalescent plasma after Wuhan infection but were lower than against the other variants. Interestingly, the second dose did not benefit from the addition of CpG, possibly allowing dose-sparing of the adjuvant in the future. The data reported here reinforces that the RBD/alum + CpG vaccine formulation is suitable for inducing broadly neutralizing antibodies against SARS-CoV-2, including variants of concern.
Collapse
Affiliation(s)
- Jeroen Pollet
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Wen-Hsiang Chen
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Leroy Versteeg
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Brian Keegan
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Bin Zhan
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Junfei Wei
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Zhuyun Liu
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Jungsoon Lee
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Rahki Kundu
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Rakesh Adhikari
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Cristina Poveda
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Maria Jose Villar
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | | | - Brianna Lopez
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Portia M Gillespie
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Shannon Ronca
- Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Biology, Baylor University, Waco, TX, USA; James A. Baker III Institute for Public Policy, Rice University, Houston, TX, USA
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Biology, Baylor University, Waco, TX, USA.
| |
Collapse
|
135
|
Febrianti RA, Narulita E. In-silico analysis of recombinant protein vaccines based on the spike protein of Indonesian SARS-CoV-2 through a reverse vaccinology approach. J Taibah Univ Med Sci 2022; 17:467-478. [PMID: 35250426 PMCID: PMC8881762 DOI: 10.1016/j.jtumed.2022.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/04/2022] [Accepted: 02/09/2022] [Indexed: 01/23/2023] Open
Abstract
Objectives This study aimed to produce a recombinant protein vaccine candidate based on an epitope of spike protein from Indonesian SARS-CoV-2 to provide immunogenicity and protection against future infection. Methods A reverse vaccinology approach was used to identify potential vaccine candidates by screening the pathogen's genome through computational analyses. Results Epitope vaccine candidates with the amino acid sequence of FKNHTSPDV were selected. This peptide is hydrophilic, does not induce autoimmune and allergic reactions, is antigenic, is classified as a stable protein, and is predicted to be present in the cell membrane. The selected epitope sequences were inserted into the plasmid vector pcDNA3.1(+) N-GST (thrombin). Inclusion of additional features of the gene encoding glutathione-S transferase, which can increase antigen expression and solubility, and the genes encoding NSP 1-4 proteins, which are essential in replication, added value to the produced recombinant protein. Conclusion Recombinant protein vaccine candidates with the FKNHTSPDV epitope have parameters sufficient for production on a laboratory scale for further testing.
Collapse
Affiliation(s)
- Riska A. Febrianti
- Department of Biotechnology, Postgraduate Program, University of Jember, Indonesia
| | - Erlia Narulita
- Department of Biotechnology, Postgraduate Program, University of Jember, Indonesia
- Department of Biology Education, University of Jember, Indonesia
| |
Collapse
|
136
|
Li L, Honda-Okubo Y, Baldwin J, Bowen R, Bielefeldt-Ohmann H, Petrovsky N. Covax-19/Spikogen® vaccine based on recombinant spike protein extracellular domain with Advax-CpG55.2 adjuvant provides single dose protection against SARS-CoV-2 infection in hamsters. Vaccine 2022; 40:3182-3192. [PMID: 35465982 PMCID: PMC9013662 DOI: 10.1016/j.vaccine.2022.04.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/03/2022] [Accepted: 04/11/2022] [Indexed: 01/28/2023]
Abstract
COVID-19 presents an ongoing global health crisis. Protein-based COVID-19 vaccines that are well-tolerated, safe, highly-protective and convenient to manufacture remain of major interest. We therefore sought to compare the immunogenicity and protective efficacy of a number of recombinant SARS-CoV-2 spike protein candidates expressed in insect cells. By comparison to a full length (FL) spike protein detergent-extracted nanoparticle antigen, the soluble secreted spike protein extracellular domain (ECD) generated higher protein yields per liter of culture and when formulated with either Alum-CpG55.2 or Advax-CpG55.2 combination adjuvants elicited robust antigen-specific humoral and cellular immunity in mice. In hamsters, the spike ECD when formulated with either adjuvant induced high serum neutralizing antibody titers even after a single dose. When challenged with the homologous SARS-CoV-2 virus, hamsters immunized with the adjuvanted spike ECD exhibited reduced viral load in day 1-3 oropharyngeal swabs and day 3 nasal turbinate tissue and had no recoverable infectious virus in day 3 lung tissue. The reduction in lung viral load correlated with less weight loss and lower lung pathology scores. The formulations of spike ECD with Alum-CpG55.2 or Advax-CpG55.2 were protective even after just a single dose, although the 2-dose regimen performed better overall and required only half the total amount of antigen. Pre-challenge serum neutralizing antibody levels showed a strong correlation with lung protection, with a weaker correlation seen with nasal or oropharyngeal protection. This suggests that serum neutralizing antibody levels may correlate more closely with systemic, rather than mucosal, protection. The spike protein ECD with Advax-CpG55.2 formulation (Covax-19® vaccine) was selected for human clinical development.
Collapse
Affiliation(s)
- Lei Li
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia; College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | - Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia; College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | | | - Richard Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Helle Bielefeldt-Ohmann
- School of Chemistry & Molecular Biosciences, The University of Queensland, St. Lucia, Qld 4072, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia; College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia.
| |
Collapse
|
137
|
Alagheband Bahrami A, Azargoonjahromi A, Sadraei S, Aarabi A, Payandeh Z, Rajabibazl M. An overview of current drugs and prophylactic vaccines for coronavirus disease 2019 (COVID-19). Cell Mol Biol Lett 2022; 27:38. [PMID: 35562685 PMCID: PMC9100302 DOI: 10.1186/s11658-022-00339-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
Designing and producing an effective vaccine is the best possible way to reduce the burden and spread of a disease. During the coronavirus disease 2019 (COVID-19) pandemic, many large pharmaceutical and biotechnology companies invested a great deal of time and money in trying to control and combat the disease. In this regard, due to the urgent need, many vaccines are now available earlier than scheduled. Based on their manufacturing technology, the vaccines available for COVID-19 (severe acute respiratory syndrome coronavirus 2 (SAR-CoV2)) infection can be classified into four platforms: RNA vaccines, adenovirus vector vaccines, subunit (protein-based) vaccines, and inactivated virus vaccines. Moreover, various drugs have been deemed to negatively affect the progression of the infection via various actions. However, adaptive variants of the SARS-CoV-2 genome can alter the pathogenic potential of the virus and increase the difficulty of both drug and vaccine development. In this review, along with drugs used in COVID-19 treatment, currently authorized COVID-19 vaccines as well as variants of the virus are described and evaluated, considering all platforms.
Collapse
Affiliation(s)
- Armina Alagheband Bahrami
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Samin Sadraei
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aryan Aarabi
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Payandeh
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
138
|
Li M, Wang H, Tian L, Pang Z, Yang Q, Huang T, Fan J, Song L, Tong Y, Fan H. COVID-19 vaccine development: milestones, lessons and prospects. Signal Transduct Target Ther 2022; 7:146. [PMID: 35504917 PMCID: PMC9062866 DOI: 10.1038/s41392-022-00996-y] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
With the constantly mutating of SARS-CoV-2 and the emergence of Variants of Concern (VOC), the implementation of vaccination is critically important. Existing SARS-CoV-2 vaccines mainly include inactivated, live attenuated, viral vector, protein subunit, RNA, DNA, and virus-like particle (VLP) vaccines. Viral vector vaccines, protein subunit vaccines, and mRNA vaccines may induce additional cellular or humoral immune regulations, including Th cell responses and germinal center responses, and form relevant memory cells, greatly improving their efficiency. However, some viral vector or mRNA vaccines may be associated with complications like thrombocytopenia and myocarditis, raising concerns about the safety of these COVID-19 vaccines. Here, we systemically assess the safety and efficacy of COVID-19 vaccines, including the possible complications and different effects on pregnant women, the elderly, people with immune diseases and acquired immunodeficiency syndrome (AIDS), transplant recipients, and cancer patients. Based on the current analysis, governments and relevant agencies are recommended to continue to advance the vaccine immunization process. Simultaneously, special attention should be paid to the health status of the vaccines, timely treatment of complications, vaccine development, and ensuring the lives and health of patients. In addition, available measures such as mix-and-match vaccination, developing new vaccines like nanoparticle vaccines, and optimizing immune adjuvant to improve vaccine safety and efficacy could be considered.
Collapse
Affiliation(s)
- Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Han Wang
- Laboratory for Clinical Immunology, Harbin Children's Hospital, Harbin, China
| | - Lili Tian
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Qingkun Yang
- College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Tianqi Huang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lihua Song
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China. .,Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
139
|
Wambani J, Okoth P. Scope of SARS-CoV-2 variants, mutations, and vaccine technologies. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2022; 34:34. [PMID: 35368846 PMCID: PMC8962228 DOI: 10.1186/s43162-022-00121-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
Background The COVID-19 pandemic is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 is disseminated by respiratory aerosols. The virus uses the spike protein to target epithelial cells by binding to the ACE2 receptor on the host cells. As a result, effective vaccines must target the viral spike glycoprotein. However, the appearance of an Omicron variant with 32 mutations in its spike protein raises questions about the vaccine's efficacy. Vaccines are critical in boosting immunity, lowering COVID-19-related illnesses, reducing the infectious burden on the healthcare system, and reducing economic loss, according to current data. An efficient vaccination campaign is projected to increase innate and adaptive immune responses, offering better protection against SARS-CoV-2 variants. Main body The presence of altered SARS-CoV-2 variants circulating around the world puts the effectiveness of vaccines already on the market at risk. The problem is made even worse by the Omicron variant, which has 32 mutations in its spike protein. Experts are currently examining the potential consequences of commercial vaccines on variants. However, there are worries about the vaccines' safety, the protection they provide, and whether future structural changes are required for these vaccines to be more effective. As a result of these concerns, new vaccines based on modern technology should be developed to guard against the growing SARS-CoV-2 variations. Conclusion The choice of a particular vaccine is influenced by several factors including mode of action, storage conditions, group of the vaccinee, immune response mounted, cost, dosage protocol, age, and side effects. Currently, seven SARS-CoV-2 vaccine platforms have been developed. This comprises of inactivated viruses, messenger RNA (mRNA), DNA vaccines, protein subunits, nonreplicating and replicating vector viral-like particles (VLP), and live attenuated vaccines. This review focuses on the SARS-CoV-2 mutations, variants of concern (VOCs), and advances in vaccine technologies.
Collapse
Affiliation(s)
- Josephine Wambani
- Kenya Medical Research Institute (KEMRI) HIV Laboratory-Alupe, P.O Box 3-50400, Busia, Kenya
- Department of Medical Laboratory Sciences, School of Public Health, Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology, P.O Box 190, Kakamega, 50100 Kenya
| | - Patrick Okoth
- Department of Biological Sciences, School of Natural Sciences, Masinde Muliro University of Science and Technology, P. O Box 190, Kakamega, 50100 Kenya
| |
Collapse
|
140
|
Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol 2022; 15:28. [PMID: 35303904 PMCID: PMC8931585 DOI: 10.1186/s13045-022-01247-x] [Citation(s) in RCA: 390] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/03/2022] [Indexed: 02/08/2023] Open
Abstract
Research on tumor immunotherapy has made tremendous progress in the past decades, with numerous studies entering the clinical evaluation. The cancer vaccine is considered a promising therapeutic strategy in the immunotherapy of solid tumors. Cancer vaccine stimulates anti-tumor immunity with tumor antigens, which could be delivered in the form of whole cells, peptides, nucleic acids, etc. Ideal cancer vaccines could overcome the immune suppression in tumors and induce both humoral immunity and cellular immunity. In this review, we introduced the working mechanism of cancer vaccines and summarized four platforms for cancer vaccine development. We also highlighted the clinical research progress of the cancer vaccines, especially focusing on their clinical application and therapeutic efficacy, which might hopefully facilitate the future design of the cancer vaccine.
Collapse
Affiliation(s)
- Jian Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Minyang Fu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
141
|
Tragni V, Preziusi F, Laera L, Onofrio A, Mercurio I, Todisco S, Volpicella M, De Grassi A, Pierri CL. Modeling SARS-CoV-2 spike/ACE2 protein-protein interactions for predicting the binding affinity of new spike variants for ACE2, and novel ACE2 structurally related human protein targets, for COVID-19 handling in the 3PM context. EPMA J 2022; 13:149-175. [PMID: 35013687 PMCID: PMC8732965 DOI: 10.1007/s13167-021-00267-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022]
Abstract
Aims The rapid spread of new SARS-CoV-2 variants has highlighted the crucial role played in the infection by mutations occurring at the SARS-CoV-2 spike receptor binding domain (RBD) in the interactions with the human ACE2 receptor. In this context, it urgently needs to develop new rapid tools for quickly predicting the affinity of ACE2 for the SARS-CoV-2 spike RBD protein variants to be used with the ongoing SARS-CoV-2 genomic sequencing activities in the clinics, aiming to gain clues about the transmissibility and virulence of new variants, to prevent new outbreaks and to quickly estimate the severity of the disease in the context of the 3PM. Methods In our study, we used a computational pipeline for calculating the interaction energies at the SARS-CoV-2 spike RBD/ACE2 protein-protein interface for a selected group of characterized infectious variants of concern/interest (VoC/VoI). By using our pipeline, we built 3D comparative models of the SARS-CoV-2 spike RBD/ACE2 protein complexes for the VoC B.1.1.7-United Kingdom (carrying the mutations of concern/interest N501Y, S494P, E484K at the RBD), P.1-Japan/Brazil (RBD mutations: K417T, E484K, N501Y), B.1.351-South Africa (RBD mutations: K417N, E484K, N501Y), B.1.427/B.1.429-California (RBD mutations: L452R), the B.1.141 (RBD mutations: N439K), and the recent B.1.617.1-India (RBD mutations: L452R; E484Q) and the B.1.620 (RBD mutations: S477N; E484K). Then, we used the obtained 3D comparative models of the SARS-CoV-2 spike RBD/ACE2 protein complexes for predicting the interaction energies at the protein-protein interface. Results Along SARS-CoV-2 mutation database screening and mutation localization analysis, it was ascertained that the most dangerous mutations at VoC/VoI spike proteins are located mainly at three regions of the SARS-CoV-2 spike "boat-shaped" receptor binding motif, on the RBD domain. Notably, the P.1 Japan/Brazil variant present three mutations, K417T, E484K, N501Y, located along the entire receptor binding motif, which apparently determines the highest interaction energy at the SARS-CoV-2 spike RBD/ACE2 protein-protein interface, among those calculated. Conversely, it was also observed that the replacement of a single acidic/hydrophilic residue with a basic residue (E484K or N439K) at the "stern" or "bow" regions, of the boat-shaped receptor binding motif on the RBD, appears to determine an interaction energy with ACE2 receptor higher than that observed with single mutations occurring at the "hull" region or with other multiple mutants. In addition, our pipeline allowed searching for ACE2 structurally related proteins, i.e., THOP1 and NLN, which deserve to be investigated for their possible involvement in interactions with the SARS-CoV-2 spike protein, in those tissues showing a low expression of ACE2, or as a novel receptor for future spike variants. A freely available web-tool for the in silico calculation of the interaction energy at the SARS-CoV-2 spike RBD/ACE2 protein-protein interface, starting from the sequences of the investigated spike and/or ACE2 variants, was made available for the scientific community at: https://www.mitoairm.it/covid19affinities. Conclusion In the context of the PPPM/3PM, the employment of the described pipeline through the provided webservice, together with the ongoing SARS-CoV-2 genomic sequencing, would help to predict the transmissibility of new variants sequenced from future patients, depending on SARS-CoV-2 genomic sequencing activities and on the specific amino acid replacement and/or on its location on the SARS-CoV-2 spike RBD, to put in play all the possible counteractions for preventing the most deleterious scenarios of new outbreaks, taking into consideration that a greater transmissibility has not to be necessarily related to a more severe manifestation of the disease. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-021-00267-w.
Collapse
Affiliation(s)
- Vincenzo Tragni
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
| | - Francesca Preziusi
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
| | - Luna Laera
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
| | - Angelo Onofrio
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
| | - Ivan Mercurio
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
| | - Simona Todisco
- Department of Sciences, University of Basilicata, Viale dell’Ateneo Lucano, 10-85100 Potenza, Italy
| | - Mariateresa Volpicella
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
| | - Anna De Grassi
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
- BROWSer S.r.l. at Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70126 Bari, Italy
| | - Ciro Leonardo Pierri
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
- BROWSer S.r.l. at Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70126 Bari, Italy
| |
Collapse
|
142
|
Pack SM, Peters PJ. SARS-CoV-2-Specific Vaccine Candidates; the Contribution of Structural Vaccinology. Vaccines (Basel) 2022; 10:236. [PMID: 35214693 PMCID: PMC8877865 DOI: 10.3390/vaccines10020236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 11/17/2022] Open
Abstract
SARS-CoV-2 vaccine production has taken us by storm. We aim to fill in the history of concepts and the work of pioneers and provide a framework of strategies employing structural vaccinology. Cryo-electron microscopy became crucial in providing three-dimensional (3D) structures and creating candidates eliciting T and B cell-mediated immunity. It also determined structural changes in the emerging mutants in order to design new constructs that can be easily, quickly and safely added to the vaccines. The full-length spike (S) protein, the S1 subunit and its receptor binding domain (RBD) of the virus are the best candidates. The vaccine development to cease this COVID-19 pandemic sets a milestone for the pan-coronavirus vaccine's designing and manufacturing. By employing structural vaccinology, we propose that the mRNA and the protein sequences of the currently approved vaccines should be modified rapidly to keep up with the more infectious new variants.
Collapse
Affiliation(s)
| | - Peter J. Peters
- The Maastricht Multimodal Molecular Imaging Institute (M4i), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
143
|
Noor R, Shareen S, Billah M. COVID-19 vaccines: their effectiveness against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its emerging variants. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2022; 46:96. [PMID: 35431535 PMCID: PMC8991668 DOI: 10.1186/s42269-022-00787-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/31/2022] [Indexed: 05/08/2023]
Abstract
BACKGROUND The world has been suffering from the COVID-19 pandemic (officially declared by WHO in March 2020), caused by the severe acute respiratory β-coronavirus 2 (SARS-CoV-2) since the last week of December 2019. The disease was initially designated as a Public Health Emergency of International Concern on January 30, 2020. In order to protect the health of mass public, an array of research on drugs and vaccines against SARS-CoV-2 has been conducted globally. However, the emerging variants of SARS-CoV-2, i.e., Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), and Delta (B.1.617.2) variants which evolved in late 2020 and the Omicron variant (B.1.1.529) which emerged in November 2021 along with its subvariant BA.2 which was first identified in India and South Africa in late December 2021, have raised the doubt about the efficiency of the currently used vaccines especially in terms of the consistent potential to produce neutralizing antibodies targeting the viral spike (S) protein. MAIN BODY OF THE ABSTRACT The present review discussed the functional details of major vaccines regarding their efficiency against such variants during the pandemic. Overall, the mRNA vaccines have shown around 94% effectiveness; the adenovector vaccine showed approximately 70% efficacy, whereas Sputnik V vaccines showed around 92% effectiveness; the inactivated whole-virus vaccine CoronaVac/PiCoVacc and BBIBP-CorV showed a varying effectiveness of 65-86% according to the geographic locations; the subunit vaccine NVX-CoV2373 has shown 60-89% effectiveness along with the global regions against the wild-type SARS-CoV-2 strain. However, reduced effectiveness of these vaccines against the SARS-CoV-2 variants was noticed which is suggestive for the further administration of booster dose. SHORT CONCLUSION Maximum variants of SARS-CoV-2 emerged during the second wave of COVID-19; and extensive studies on the viral genomic sequences from all geographical locations around the world have been conducted by an array of groups to assess the possible occurrence of mutations(s) specially within the receptor binding domain of the viral spike (S) protein. Mutational similarities and the new or critical mutations within all variants have been clearly identified so far. The study of effectiveness of the currently used vaccines is also ongoing. The persistence of memory B cell action and the other immune components as well as the administration of booster dose is expected to mitigate the disease.
Collapse
Affiliation(s)
- Rashed Noor
- Department of Life Sciences (DLS), School of Environment and Life Sciences (SELS), Independent University, Bangladesh (IUB), Plot 16, Block B, Aftabuddin Ahmed Road, Bashundhara, Dhaka, 1229 Bangladesh
| | - Saadia Shareen
- Department of Life Sciences (DLS), School of Environment and Life Sciences (SELS), Independent University, Bangladesh (IUB), Plot 16, Block B, Aftabuddin Ahmed Road, Bashundhara, Dhaka, 1229 Bangladesh
| | - Muntasir Billah
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, St Leonards, NSW 2065 Australia
| |
Collapse
|
144
|
Santana-Mederos D, Perez-Nicado R, Climent Y, Rodriguez L, Ramirez BS, Perez-Rodriguez S, Rodriguez M, Labrada C, Hernandez T, Diaz M, Orosa I, Ramirez U, Oliva R, Garrido R, Cardoso F, Landys M, Martinez R, Gonzalez H, Hernandez T, Ochoa-Azze R, Perez JL, Enriquez J, Gonzalez N, Infante Y, Espinosa LA, Ramos Y, González LJ, Valenzuela C, Casadesus AV, Fernandez B, Rojas G, Pérez-Massón B, Tundidor Y, Bermudez E, Plasencia CA, Boggiano T, Ojito E, Chiodo F, Fernandez S, Paquet F, Fang C, Chen GW, Rivera DG, Valdes-Balbin Y, Garcia-Rivera D, Verez Bencomo V. A COVID-19 vaccine candidate composed of the SARS-CoV-2 RBD dimer and Neisseria meningitidis outer membrane vesicles. RSC Chem Biol 2022; 3:242-249. [PMID: 35360883 PMCID: PMC8826971 DOI: 10.1039/d1cb00200g] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023] Open
Abstract
Soberana01 is composed of the SARS-CoV-2 dimeric RBD and Neisseria meningitidis outer membrane vesicles (OMVs) adsorbed on alum. This vaccine induces a potent neutralizing immune response and shows potential against SARS-CoV-2 variants of concern.
Collapse
Affiliation(s)
| | | | - Yanet Climent
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Laura Rodriguez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | | | - Meybi Rodriguez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Claudia Labrada
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Tays Hernandez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Marianniz Diaz
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ivette Orosa
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ubel Ramirez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Reynaldo Oliva
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Raine Garrido
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Felix Cardoso
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Mario Landys
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | | | | | | | | | - Jose L. Perez
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
| | - Juliet Enriquez
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Nibaldo Gonzalez
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Yenicet Infante
- National Civil Defense Research Laboratory, Mayabeque 32700, Cuba
| | - Luis A. Espinosa
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Yassel Ramos
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Luis Javier González
- Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Havana 10600, Cuba
| | - Carmen Valenzuela
- Institute of Cybernetics, Mathematics and Physics, Havana 10400, Cuba
| | | | - Briandy Fernandez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Gertrudis Rojas
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | | | - Yaima Tundidor
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Ernesto Bermudez
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | | | - Tammy Boggiano
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Eduardo Ojito
- Center of Molecular Immunology, P.O. Box 16040, 216 St., Havana, Cuba
| | - Fabrizio Chiodo
- Finlay Vaccine Institute, 200 and 21 Street, Havana 11600, Cuba
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands and Institute of Biomolecular Chemistry, National Research Council (CNR), Pozzuoli, Napoli, Italy
| | | | - Françoise Paquet
- Centre de Biophysique Moléculaire, CNRS UPR 4301, rue Charles Sadron, F-45071, Orléans Cedex 2, France
| | - Cheng Fang
- Shanghai Fenglin Glycodrug Promotion Center, Shanghai 200032, China
| | - Guang-Wu Chen
- Chengdu Olisynn Biotech. Co. Ltd., and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Daniel G. Rivera
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata y G, Havana 10400, Cuba
| | | | | | | |
Collapse
|
145
|
Ranade D, Jena R, Sancheti S, Deore V, Dogar V, Gairola S. Rapid, high throughput protein estimation method for saponin and alhydrogel adjuvanted R21 VLP Malaria vaccine based on intrinsic fluorescence. Vaccine 2021; 40:601-611. [PMID: 34933766 DOI: 10.1016/j.vaccine.2021.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/30/2021] [Accepted: 12/09/2021] [Indexed: 11/27/2022]
Abstract
Protein content estimation of recombinant vaccines at drug product (DP) stage is a crucial lot release and stability indicating assay in biopharmaceutical industries. Regulatory bodies such as US-FDA and WHO necessitates the quantitation of protein content to assess process parameters as well as formulation losses. Estimation of protein content at DP stage in presence of adjuvants (e.g AlOOH, AlPO4, saponin and squalene) is quite challenging, and the challenge intensifies when the target protein is in Virus like particles (VLP) form, owing to its size and structural complexity. Methods available for protein estimation of adjuvanted vaccines mostly suffer from inaccuracy at lower protein concentrations and in most cases require antigen desorption before analysis. Present research work is based on the development of a rapid plate-based method for protein estimation through intrinsic fluorescence by using Malaria vaccine R21 VLP as a model protein. Present method exhibited linearity for protein estimation of R21, in the range of 5-30 µg/mL in Alhydrogel and 4-20 µg/mL for Matrix M adjuvant. The method was validated as per ICH guidelines. The limit of quantification was found to be 0.94 µg/mL for both Alhydrogel and Matrix M adjuvanted R21. The method was found specific, precise and repeatable. This method is superior in terms of less sample quantity requirement, multiple sample analysis, short turnaround time and is non-invasive. This method was found to be stability indicating, works for other proteins containing tryptophan residues and operates well even in presence of host cell proteins. Based on the study, present method can be used in vaccine industries for routine in-process sample analysis (both inline and offline), lot release of VLP based drug products in presence of Alhydrogel and saponin based adjuvant systems.
Collapse
Affiliation(s)
- Dnyanesh Ranade
- Quality Control Department, Serum Institute of India Pvt. Ltd, 212/2, Soli Poonawalla Rd, JJC Colony, Suryalok Nagari, Hadapsar, Pune, Maharashtra 411028, India
| | - Rajender Jena
- Quality Control Department, Serum Institute of India Pvt. Ltd, 212/2, Soli Poonawalla Rd, JJC Colony, Suryalok Nagari, Hadapsar, Pune, Maharashtra 411028, India
| | - Shubham Sancheti
- Quality Control Department, Serum Institute of India Pvt. Ltd, 212/2, Soli Poonawalla Rd, JJC Colony, Suryalok Nagari, Hadapsar, Pune, Maharashtra 411028, India
| | - Vicky Deore
- Quality Control Department, Serum Institute of India Pvt. Ltd, 212/2, Soli Poonawalla Rd, JJC Colony, Suryalok Nagari, Hadapsar, Pune, Maharashtra 411028, India
| | - Vikas Dogar
- Quality Control Department, Serum Institute of India Pvt. Ltd, 212/2, Soli Poonawalla Rd, JJC Colony, Suryalok Nagari, Hadapsar, Pune, Maharashtra 411028, India
| | - Sunil Gairola
- Quality Control Department, Serum Institute of India Pvt. Ltd, 212/2, Soli Poonawalla Rd, JJC Colony, Suryalok Nagari, Hadapsar, Pune, Maharashtra 411028, India.
| |
Collapse
|
146
|
Coronavirus disease 2019 vaccine: An overview of the progression and current use. North Clin Istanb 2021; 8:529-536. [PMID: 34909595 PMCID: PMC8630730 DOI: 10.14744/nci.2021.99075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022] Open
Abstract
On December 31, 2019; unidentified pneumonia cases were reported from China. It was soon announced that these cases were of viral origin and the cause was a new coronavirus (CoV). Initially, the virus was called "novel CoV " and then defined as "severe acute respiratory syndrome CoV 2 (SARS-CoV-2)" after more detailed investigations. The disease caused by SARS-CoV-2 was named CoV disease 2019 (COVID-19) by the World Health Organization. The rapid spread of the disease in a few months has resulted in a global pandemic and it continues. However, there are no specific effective anti-viral drugs for SARS-CoV-2 infection, some antiviral drugs are using in the therapy of COVID-19 with limited success. Currently, for the prevention of the pandemic, global vaccination seems to be important. Antiviral protection of vaccines is provided by the development of antibodies that can neutralize the virus. Antibody response develops against spike protein and nucleocapsid protein but neutralizing antibodies are formed against the receptor-binding domain of the spike protein. It has also been shown that most viral proteins are recognized in T-cell responses. Vaccine discovery trials for COVID-19 have begun all over the world since the outbreak began. More than 100 vaccine studies against COVID-19 have been published in the last year. Some of them were urgently approved and used worldwide. The current study aimed to review the progression and current use of COVID-19 vaccines.
Collapse
|
147
|
Bok M, Zhao ZJ, Hwang SH, Jeong Y, Ko J, Ahn J, Lee JH, Jeon S, Jeong JH. Biocompatible All-in-One Adhesive Needle-Free Cup Patch for Enhancing Transdermal Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2021; 13:58220-58228. [PMID: 34793117 DOI: 10.1021/acsami.1c18750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Patch-type drug delivery has garnered increased attention as an attractive alternative to the existing drug delivery techniques. Thus far, needle phobia and efficient drug delivery remain huge challenges. To address the issue of needle phobia and enhance drug delivery, we developed a needle-free and self-adhesive microcup patch that can be loaded with an ultrathin salmon DNA (SDNA) drug carrier film. This physically integrated system can facilitate efficient skin penetration of drugs loaded into the microcup patch. The system consists of three main components, namely, a cup that acts as a drug reservoir, an adhesive system that attaches the patch to the skin, and physical stimulants that can be used to increase the efficiency of drug delivery. In addition, an ultrathin SDNA/drug film allows the retention of the drug in the cup and its efficient release by dissolution in the presence of moisture. This latter feature has been validated using gelatin as a skin mimic. The cup design itself has been validated by comparing its deformation and displacement with those of a cylindrical structure. Integration of the self-adhesive microcup patch with both ultrasonic waves and an electric current allows the model drug to penetrate the stratum corneum of the skin barrier and the whole epidermis, thereby enhancing transdermal drug delivery and reducing skin irritation. This system can be used as a wearable biomedical device for efficient transdermal and needle-free drug delivery.
Collapse
Affiliation(s)
- Moonjeong Bok
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon 34103, South Korea
| | - Zhi-Jun Zhao
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon 34103, South Korea
| | - Soon Hyoung Hwang
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon 34103, South Korea
| | - Yongrok Jeong
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon 34103, South Korea
| | - Jiwoo Ko
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon 34103, South Korea
| | - Junseong Ahn
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon 34103, South Korea
| | - Ju Ho Lee
- Department of Applied Physics, Dankook University, Yongin 16890, South Korea
| | - Sohee Jeon
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon 34103, South Korea
| | - Jun-Ho Jeong
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon 34103, South Korea
| |
Collapse
|
148
|
Chung YH, Church D, Koellhoffer EC, Osota E, Shukla S, Rybicki EP, Pokorski JK, Steinmetz NF. Integrating plant molecular farming and materials research for next-generation vaccines. NATURE REVIEWS. MATERIALS 2021; 7:372-388. [PMID: 34900343 DOI: 10.1038/s41578-021-00399-395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 05/28/2023]
Abstract
Biologics - medications derived from a biological source - are increasingly used as pharmaceuticals, for example, as vaccines. Biologics are usually produced in bacterial, mammalian or insect cells. Alternatively, plant molecular farming, that is, the manufacture of biologics in plant cells, transgenic plants and algae, offers a cheaper and easily adaptable strategy for the production of biologics, in particular, in low-resource settings. In this Review, we discuss current vaccination challenges, such as cold chain requirements, and highlight how plant molecular farming in combination with advanced materials can be applied to address these challenges. The production of plant viruses and virus-based nanotechnologies in plants enables low-cost and regional fabrication of thermostable vaccines. We also highlight key new vaccine delivery technologies, including microneedle patches and material platforms for intranasal and oral delivery. Finally, we provide an outlook of future possibilities for plant molecular farming of next-generation vaccines and biologics.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering, University of California, San Diego, La Jolla, CA USA
| | - Derek Church
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
| | - Edward C Koellhoffer
- Department of Radiology, University of California, San Diego Health, La Jolla, CA USA
| | - Elizabeth Osota
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
- Biomedical Science Program, University of California, San Diego, La Jolla, CA USA
| | - Sourabh Shukla
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
| | - Edward P Rybicki
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Jonathan K Pokorski
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA USA
- Center for Nano-Immuno Engineering, University of California, San Diego, La Jolla, CA USA
| | - Nicole F Steinmetz
- Department of Bioengineering, University of California, San Diego, La Jolla, CA USA
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
- Department of Radiology, University of California, San Diego Health, La Jolla, CA USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA USA
- Center for Nano-Immuno Engineering, University of California, San Diego, La Jolla, CA USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA USA
| |
Collapse
|
149
|
Chung YH, Church D, Koellhoffer EC, Osota E, Shukla S, Rybicki EP, Pokorski JK, Steinmetz NF. Integrating plant molecular farming and materials research for next-generation vaccines. NATURE REVIEWS. MATERIALS 2021; 7:372-388. [PMID: 34900343 PMCID: PMC8647509 DOI: 10.1038/s41578-021-00399-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 05/04/2023]
Abstract
Biologics - medications derived from a biological source - are increasingly used as pharmaceuticals, for example, as vaccines. Biologics are usually produced in bacterial, mammalian or insect cells. Alternatively, plant molecular farming, that is, the manufacture of biologics in plant cells, transgenic plants and algae, offers a cheaper and easily adaptable strategy for the production of biologics, in particular, in low-resource settings. In this Review, we discuss current vaccination challenges, such as cold chain requirements, and highlight how plant molecular farming in combination with advanced materials can be applied to address these challenges. The production of plant viruses and virus-based nanotechnologies in plants enables low-cost and regional fabrication of thermostable vaccines. We also highlight key new vaccine delivery technologies, including microneedle patches and material platforms for intranasal and oral delivery. Finally, we provide an outlook of future possibilities for plant molecular farming of next-generation vaccines and biologics.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering, University of California, San Diego, La Jolla, CA USA
| | - Derek Church
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
| | - Edward C. Koellhoffer
- Department of Radiology, University of California, San Diego Health, La Jolla, CA USA
| | - Elizabeth Osota
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
- Biomedical Science Program, University of California, San Diego, La Jolla, CA USA
| | - Sourabh Shukla
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
| | - Edward P. Rybicki
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Jonathan K. Pokorski
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA USA
- Center for Nano-Immuno Engineering, University of California, San Diego, La Jolla, CA USA
| | - Nicole F. Steinmetz
- Department of Bioengineering, University of California, San Diego, La Jolla, CA USA
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA USA
- Department of Radiology, University of California, San Diego Health, La Jolla, CA USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA USA
- Center for Nano-Immuno Engineering, University of California, San Diego, La Jolla, CA USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA USA
| |
Collapse
|
150
|
Vu MN, Kelly HG, Kent SJ, Wheatley AK. Current and future nanoparticle vaccines for COVID-19. EBioMedicine 2021; 74:103699. [PMID: 34801965 PMCID: PMC8602808 DOI: 10.1016/j.ebiom.2021.103699] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/21/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 has become a major cause of global mortality and driven massive health and economic disruptions. Mass global vaccination offers the most efficient pathway towards ending the pandemic. The development and deployment of first-generation COVID-19 vaccines, encompassing mRNA or viral vectors, has proceeded at a phenomenal pace. Going forward, nanoparticle-based vaccines which deliver SARS-CoV-2 antigens will play an increasing role in extending or improving vaccination outcomes against COVID-19. At present, over 26 nanoparticle vaccine candidates have advanced into clinical testing, with ∼60 more in pre-clinical development. Here, we discuss the emerging promise of nanotechnology in vaccine design and manufacturing to combat SARS-CoV-2, and highlight opportunities and challenges presented by these novel vaccine platforms.
Collapse
Affiliation(s)
- Mai N Vu
- Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC 3052, Australia; Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Department of Pharmaceutics, Hanoi University of Pharmacy, Hanoi 10000, Vietnam
| | - Hannah G Kelly
- Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC 3052, Australia
| | - Stephen J Kent
- Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC 3052, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.
| | - Adam K Wheatley
- Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC 3052, Australia.
| |
Collapse
|