1
|
Stepanova E, Isakova-Sivak I, Matyushenko V, Mezhenskaya D, Kudryavtsev I, Kostromitina A, Chistiakova A, Rak A, Bazhenova E, Prokopenko P, Kotomina T, Donina S, Novitskaya V, Sivak K, Karal-Ogly D, Rudenko L. Safety and Immunogenicity Study of a Bivalent Vaccine for Combined Prophylaxis of COVID-19 and Influenza in Non-Human Primates. Vaccines (Basel) 2024; 12:1099. [PMID: 39460266 PMCID: PMC11511058 DOI: 10.3390/vaccines12101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Influenza and SARS-CoV-2 viruses are two highly variable pathogens. We have developed a candidate bivalent live vaccine based on the strain of licensed A/Leningrad/17-based cold-adapted live attenuated influenza vaccine (LAIV) of H3N2 subtype, which expressed SARS-CoV-2 immunogenic T-cell epitopes. A cassette encoding fragments of S and N proteins of SARS-CoV-2 was inserted into the influenza NA gene using the P2A autocleavage site. In this study, we present the results of preclinical evaluation of the developed bivalent vaccine in a non-human primate model. METHODS Rhesus macaques (Macaca mulatta) (n = 3 per group) were immunized intranasally with 7.5 lg EID50 of the LAIV/CoV-2 bivalent vaccine, a control non-modified H3N2 LAIV or a placebo (chorioallantoic fluid) using a sprayer device, twice, with a 28-day interval. The blood samples were collected at days 0, 3, 28 and 35 for hematological and biochemical assessment. Safety was also assessed by monitoring body weight, body temperature and clinical signs of the disease. Immune responses to influenza virus were assessed both by determining serum antibody titers in hemagglutination inhibition assay, microneutralization assay and IgG ELISA. T-cell responses were measured both to influenza and SARS-CoV-2 antigens using ELISPOT and flow cytometry. Three weeks after the second immunization, animals were challenged with 105 PFU of Delta SARS-CoV-2. The body temperature, weight and challenge virus shedding were monitored for 5 days post-challenge. In addition, virus titers in various organs and histopathology were evaluated on day 6 after SARS-CoV-2 infection. RESULTS There was no toxic effect of the immunizations on the hematological and coagulation hemostasis of animals. No difference in the dynamics of the average weight and thermometry results were found between the groups of animals. Both LAIV and LAIV/CoV-2 variants poorly replicated in the upper respiratory tract of rhesus macaques. Nevertheless, despite this low level of virus shedding, influenza-specific serum IgG responses were detected in the group of monkeys immunized with the LAIV/CoV-2 bivalent but not in the LAIV group. Furthermore, T-cell responses to both influenza and SARS-CoV-2 viruses were detected in the LAIV/CoV-2 vaccine group only. The animals were generally resistant to SARS-CoV-2 challenge, with minimal virus shedding in the placebo and LAIV groups. Histopathological changes in vaccinated animals were decreased compared to the PBS group, suggesting a protective effect of the chimeric vaccine candidate. CONCLUSIONS The candidate bivalent vaccine was safe and immunogenic for non-human primates and warrants its further evaluation in clinical trials.
Collapse
Affiliation(s)
- Ekaterina Stepanova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Victoria Matyushenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Arina Kostromitina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Anna Chistiakova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Alexandra Rak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Ekaterina Bazhenova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Svetlana Donina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Vlada Novitskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint-Petersburg 197376, Russia;
| | - Dzhina Karal-Ogly
- Center of Preclinical Research, Research Institute of Medical Primatology, Sochi 354376, Russia;
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| |
Collapse
|
2
|
Dashti N, Golsaz-Shirazi F, Soltanghoraee H, Zarnani AH, Mohammadi M, Imani D, Jeddi-Tehrani M, Amiri MM, Shokri F. Preclinical assessment of a recombinant RBD-Fc fusion protein as SARS-CoV-2 candidate vaccine. Eur J Microbiol Immunol (Bp) 2024; 14:228-242. [PMID: 38753442 PMCID: PMC11393645 DOI: 10.1556/1886.2024.00045] [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: 03/29/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Background Waning immunity and emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlight the need for further research in vaccine development. Methods A recombinant fusion protein containing the receptor-binding domain (RBD) fused to the human IgG1 Fc (RBD-Fc) was produced in CHO-K1 cells. RBD-Fc was emulsified with four adjuvants to evaluate its immunogenicity. The RBD-specific humoral and cellular immune responses were assessed by ELISA. The virus neutralizing potency of the vaccine was investigated using four neutralization methods. Safety was studied in mice and rabbits, and Antibody-Dependent Enhancement (ADE) effects were investigated by flow cytometry. Results RBD-Fc emulsified in Alum induced a high titer of anti-RBD antibodies with remarkable efficacy in neutralizing both pseudotyped and live SARS-CoV-2 Delta variant. The neutralization potency dropped significantly in response to the Omicron variant. RBD-Fc induced both TH2 and particularly TH1 immune responses. Histopathologic examinations demonstrated no substantial pathologic changes in different organs. No changes in serum biochemical and hematologic parameters were observed. ADE effect was not observed following immunization with RBD-Fc. Conclusion RBD-Fc elicits highly robust neutralizing antibodies and cellular immune responses, with no adverse effects. Therefore, it could be considered a promising and safe subunit vaccine against SARS-CoV-2.
Collapse
Affiliation(s)
- Navid Dashti
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Golsaz-Shirazi
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Soltanghoraee
- 2Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- 3Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mehdi Mohammadi
- 4Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Danyal Imani
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- 5Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Mehdi Amiri
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fazel Shokri
- 1Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Dillard JA, Taft-Benz SA, Knight AC, Anderson EJ, Pressey KD, Parotti B, Martinez SA, Diaz JL, Sarkar S, Madden EA, De la Cruz G, Adams LE, Dinnon KH, Leist SR, Martinez DR, Schäfer A, Powers JM, Yount BL, Castillo IN, Morales NL, Burdick J, Evangelista MKD, Ralph LM, Pankow NC, Linnertz CL, Lakshmanane P, Montgomery SA, Ferris MT, Baric RS, Baxter VK, Heise MT. Adjuvant-dependent impact of inactivated SARS-CoV-2 vaccines during heterologous infection by a SARS-related coronavirus. Nat Commun 2024; 15:3738. [PMID: 38702297 PMCID: PMC11068739 DOI: 10.1038/s41467-024-47450-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 04/02/2024] [Indexed: 05/06/2024] Open
Abstract
Whole virus-based inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous coronavirus infection, the emergence of novel variants and the presence of large zoonotic reservoirs harboring novel heterologous coronaviruses provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes like vaccine-associated enhanced respiratory disease. Here, we use a female mouse model of coronavirus disease to evaluate inactivated vaccine performance against either homologous challenge with SARS-CoV-2 or heterologous challenge with a bat-derived coronavirus that represents a potential emerging disease threat. We show that inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide can cause enhanced respiratory disease during heterologous infection, while use of an alternative adjuvant does not drive disease and promotes heterologous viral clearance. In this work, we highlight the impact of adjuvant selection on inactivated vaccine safety and efficacy against heterologous coronavirus infection.
Collapse
Affiliation(s)
- Jacob A Dillard
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sharon A Taft-Benz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Audrey C Knight
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth J Anderson
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katia D Pressey
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Breantié Parotti
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sabian A Martinez
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer L Diaz
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sanjay Sarkar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily A Madden
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabriela De la Cruz
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lily E Adams
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth H Dinnon
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John M Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Boyd L Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Izabella N Castillo
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Noah L Morales
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jane Burdick
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Lauren M Ralph
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicholas C Pankow
- Pathology Services Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Colton L Linnertz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Premkumar Lakshmanane
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephanie A Montgomery
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Dallas Tissue Research, Farmers Branch, TX, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria K Baxter
- Department of Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Texas Biomedical Research Institute, San Antonio, TX, USA.
| | - Mark T Heise
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Gao R, Feng C, Sheng Z, Li F, Wang D. Research progress in Fc-effector functions against SARS-CoV-2. J Med Virol 2024; 96:e29638. [PMID: 38682662 DOI: 10.1002/jmv.29638] [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: 03/12/2024] [Revised: 03/31/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has caused more than 676 million cases in the global human population with approximately 7 million deaths and vaccination has been proved as the most effective countermeasure in reducing clinical complications and mortality rate of SARS-CoV-2 infection in people. However, the protective elements and correlation of protection induced by vaccination are still not completely understood. Various antibodies with multiple protective mechanisms can be induced simultaneously by vaccination in vivo, thereby complicating the identification and characterization of individual correlate of protection. Recently, an increasing body of observations suggests that antibody-induced Fc-effector functions play a crucial role in combating SARS-CoV-2 infections, including neutralizing antibodies-escaping variants. Here, we review the recent progress in understanding the impact of Fc-effector functions in broadly disarming SARS-CoV-2 infectivity and discuss various efforts in harnessing this conserved antibody function to develop an effective SARS-CoV-2 vaccine that can protect humans against infections by SARS-CoV-2 virus and its variants of concern.
Collapse
Affiliation(s)
- Rongyuan Gao
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Chenchen Feng
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Zizhang Sheng
- Zuckerman Mind Brian Behavior Institute, Columbia University, New York, New York, USA
| | - Feng Li
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| | - Dan Wang
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
5
|
Brindha S, Yoshizue T, Wongnak R, Takemae H, Oba M, Mizutani T, Kuroda Y. An Escherichia coli Expressed Multi-Disulfide Bonded SARS-CoV-2 RBD Shows Native-like Biophysical Properties and Elicits Neutralizing Antisera in a Mouse Model. Int J Mol Sci 2022; 23:15744. [PMID: 36555383 PMCID: PMC9779815 DOI: 10.3390/ijms232415744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
A large-scale Escherichia coli (E. coli) production of the receptor-binding domain (RBD) of the SARS-CoV-2 could yield a versatile and low-cost antigen for a subunit vaccine. Appropriately folded antigens can potentially elicit the production of neutralizing antisera providing immune protection against the virus. However, E. coli expression using a standard protocol produces RBDs with aberrant disulfide bonds among the RBD's eight cysteines resulting in the expression of insoluble and non-native RBDs. Here, we evaluate whether E. coli expressing RBD can be used as an antigen candidate for a subunit vaccine. The expressed RBD exhibited native-like structural and biophysical properties as demonstrated by analytical RP-HPLC, circular dichroism, fluorescence, and light scattering. In addition, our E. coli expressed RBD binds to hACE2, the host cell's receptor, with a binding constant of 7.9 × 10-9 M, as indicated by biolayer interferometry analysis. Our E. coli-produced RBD elicited a high IgG titer in Jcl:ICR mice, and the RBD antisera inhibited viral growth, as demonstrated by a pseudovirus-based neutralization assay. Moreover, the increased antibody level was sustained for over 15 weeks after immunization, and a high percentage of effector and central memory T cells were generated. Overall, these results show that E. coli-expressed RBDs can elicit the production of neutralizing antisera and could potentially serve as an antigen for developing an anti-SARS-CoV-2 subunit vaccine.
Collapse
Affiliation(s)
- Subbaian Brindha
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi 184-8588, Tokyo, Japan
| | - Takahiro Yoshizue
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi 184-8588, Tokyo, Japan
| | - Rawiwan Wongnak
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi 184-8588, Tokyo, Japan
| | - Hitoshi Takemae
- Center for Infectious Disease Epidemiology and Prevention Research, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Mami Oba
- Center for Infectious Disease Epidemiology and Prevention Research, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Tetsuya Mizutani
- Center for Infectious Disease Epidemiology and Prevention Research, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Yutaka Kuroda
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi 184-8588, Tokyo, Japan
| |
Collapse
|
6
|
Solis-Andrade KI, Gonzalez-Ortega O, Govea-Alonso DO, Comas-Garcia M, Rosales-Mendoza S. Production and Purification of LTB-RBD: A Potential Antigen for Mucosal Vaccine Development against SARS-CoV-2. Vaccines (Basel) 2022; 10:vaccines10101759. [PMID: 36298624 PMCID: PMC9609574 DOI: 10.3390/vaccines10101759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Most of the current SARS-CoV-2 vaccines are based on parenteral immunization targeting the S protein. Although protective, such vaccines could be optimized by inducing effective immune responses (neutralizing IgA responses) at the mucosal surfaces, allowing them to block the virus at the earliest stage of the infectious cycle. Herein a recombinant chimeric antigen called LTB-RBD is described, which comprises the B subunit of the heat-labile enterotoxin from E. coli and a segment of the RBD from SARS-CoV-2 (aa 439-504, carrying B and T cell epitopes) from the Wuhan sequence and the variant of concern (VOC)—delta. Since LTB is a mucosal adjuvant, targeting the GM1 receptor at the surface and facilitating antigen translocation to the submucosa, this candidate will help in designing mucosal vaccines (i.e., oral or intranasal formulations). LTB-RBD was produced in E. coli and purified to homogeneity by IMAC and IMAC-anionic exchange chromatography. The yields in terms of pure LTB-RBD were 1.2 mg per liter of culture for the Wuhan sequence and 3.5 mg per liter for the delta variant. The E. coli-made LTB-RBD induced seric IgG responses and IgA responses in the mouth and feces of mice when subcutaneously administered and intestinal and mouth IgA responses when administered nasally. The expression and purification protocols developed for LTB-RBD constitute a robust system to produce vaccine candidates against SARS-CoV-2 and its variants, offering a low-cost production system with no tags and with ease of adaptation to new variants. The E. coli-made LTB-RBD will be the basis for developing mucosal vaccine candidates capable of inducing sterilizing immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Karla I. Solis-Andrade
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico
| | - Omar Gonzalez-Ortega
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico
| | - Dania O. Govea-Alonso
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico
| | - Mauricio Comas-Garcia
- Sección de Microscopía de Alta Resolución, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico
- Sección de Genómica Médica, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, Av. Parque Chapultepec 1570, San Luis Potosí 78210, Mexico
| | - Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, Mexico
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico
- Correspondence: ; Tel./Fax: +52-444-826-2440
| |
Collapse
|
7
|
Morajkar RV, Kumar AS, Kunkalekar RK, Vernekar AA. Advances in nanotechnology application in biosafety materials: A crucial response to COVID-19 pandemic. BIOSAFETY AND HEALTH 2022; 4:347-363. [PMID: 35765656 PMCID: PMC9225943 DOI: 10.1016/j.bsheal.2022.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 11/07/2022] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) has adversely affected the public domain causing unprecedented cases and high mortality across the globe. This has brought back the concept of biosafety into the spotlight to solve biosafety problems in developing diagnostics and therapeutics to treat COVID-19. The advances in nanotechnology and material science in combination with medicinal chemistry have provided a new perspective to overcome this crisis. Herein, we discuss the efforts of researchers in the field of material science in developing personal protective equipment (PPE), detection devices, vaccines, drug delivery systems, and medical equipment. Such a synergistic approach of disciplines can strengthen the research to develop biosafety products in solving biosafety problems.
Collapse
Affiliation(s)
- Rasmi V Morajkar
- Inorganic and Physical Chemistry Laboratory, Council of Scientific and Industrial Research (CSIR)-Central Leather Research Institute (CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Akhil S Kumar
- Inorganic and Physical Chemistry Laboratory, Council of Scientific and Industrial Research (CSIR)-Central Leather Research Institute (CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Rohan K Kunkalekar
- School of Chemical Sciences, Goa University, Taleigao Plateau 403206, Goa, India
| | - Amit A Vernekar
- Inorganic and Physical Chemistry Laboratory, Council of Scientific and Industrial Research (CSIR)-Central Leather Research Institute (CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| |
Collapse
|
8
|
Bigay J, Le Grand R, Martinon F, Maisonnasse P. Vaccine-associated enhanced disease in humans and animal models: Lessons and challenges for vaccine development. Front Microbiol 2022; 13:932408. [PMID: 36033843 PMCID: PMC9399815 DOI: 10.3389/fmicb.2022.932408] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
The fight against infectious diseases calls for the development of safe and effective vaccines that generate long-lasting protective immunity. In a few situations, vaccine-mediated immune responses may have led to exacerbated pathology upon subsequent infection with the pathogen targeted by the vaccine. Such vaccine-associated enhanced disease (VAED) has been reported, or at least suspected, in animal models, and in a few instances in humans, for vaccine candidates against the respiratory syncytial virus (RSV), measles virus (MV), dengue virus (DENV), HIV-1, simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), and the Middle East respiratory syndrome coronavirus (MERS-CoV). Although alleviated by clinical and epidemiological evidence, a number of concerns were also initially raised concerning the short- and long-term safety of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is causing the ongoing COVID-19 pandemic. Although the mechanisms leading to this phenomenon are not yet completely understood, the individual and/or collective role of antibody-dependent enhancement (ADE), complement-dependent enhancement, and cell-dependent enhancement have been highlighted. Here, we review mechanisms that may be associated with the risk of VAED, which are important to take into consideration, both in the assessment of vaccine safety and in finding ways to define models and immunization strategies that can alleviate such concerns.
Collapse
Affiliation(s)
| | | | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud-INSERM U1184, CEA, Fontenay-Aux-Roses, France
| | | |
Collapse
|
9
|
Nguyen DC, Lamothe PA, Woodruff MC, Saini AS, Faliti CE, Sanz I, Lee FE. COVID-19 and plasma cells: Is there long-lived protection? Immunol Rev 2022; 309:40-63. [PMID: 35801537 PMCID: PMC9350162 DOI: 10.1111/imr.13115] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Infection with SARS-CoV-2, the etiology of the ongoing COVID-19 pandemic, has resulted in over 450 million cases with more than 6 million deaths worldwide, causing global disruptions since early 2020. Memory B cells and durable antibody protection from long-lived plasma cells (LLPC) are the mainstay of most effective vaccines. However, ending the pandemic has been hampered by the lack of long-lived immunity after infection or vaccination. Although immunizations offer protection from severe disease and hospitalization, breakthrough infections still occur, most likely due to new mutant viruses and the overall decline of neutralizing antibodies after 6 months. Here, we review the current knowledge of B cells, from extrafollicular to memory populations, with a focus on distinct plasma cell subsets, such as early-minted blood antibody-secreting cells and the bone marrow LLPC, and how these humoral compartments contribute to protection after SARS-CoV-2 infection and immunization.
Collapse
Affiliation(s)
- Doan C. Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Pedro A. Lamothe
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Matthew C. Woodruff
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ankur S. Saini
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Caterina E. Faliti
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ignacio Sanz
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Frances Eun‐Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
10
|
Fovet CM, Pimienta C, Galhaut M, Relouzat F, Nunez N, Cavarelli M, Sconosciuti Q, Dhooge N, Marzinotto I, Lampasona V, Tolazzi M, Scarlatti G, Ho Tsong Fang R, Naninck T, Dereuddre-Bosquet N, Van Wassenhove J, Gallouët AS, Maisonnasse P, Le Grand R, Menu E, Seddiki N. A Case Study to Dissect Immunity to SARS-CoV-2 in a Neonate Nonhuman Primate Model. Front Immunol 2022; 13:855230. [PMID: 35603150 PMCID: PMC9114777 DOI: 10.3389/fimmu.2022.855230] [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: 01/14/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Most children are less severely affected by coronavirus-induced disease 2019 (COVID-19) than adults, and thus more difficult to study progressively. Here, we provide a neonatal nonhuman primate (NHP) deep analysis of early immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in blood and mucosal tissues. In addition, we provide a comparison with SARS-CoV-2-infected adult NHP. Infection of the neonate resulted in a mild disease compared with adult NHPs that develop, in most cases, moderate lung lesions. In concomitance with the viral RNA load increase, we observed the development of an early innate response in the blood, as demonstrated by RNA sequencing, flow cytometry, and cytokine longitudinal data analyses. This response included the presence of an antiviral type-I IFN gene signature, a persistent and lasting NKT cell population, a balanced peripheral and mucosal IFN-γ/IL-10 cytokine response, and an increase in B cells that was accompanied with anti-SARS-CoV-2 antibody response. Viral kinetics and immune responses coincided with changes in the microbiota profile composition in the pharyngeal and rectal mucosae. In the mother, viral RNA loads were close to the quantification limit, despite the very close contact with SARS-CoV-2-exposed neonate. This pilot study demonstrates that neonatal NHPs are a relevant model for pediatric SARS-CoV-2 infection, permitting insights into the early steps of anti-SARS-CoV-2 immune responses in infants.
Collapse
Affiliation(s)
- Claire-Maëlle Fovet
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Camille Pimienta
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Mathilde Galhaut
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Francis Relouzat
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | | | - Mariangela Cavarelli
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Quentin Sconosciuti
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Nina Dhooge
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Ilaria Marzinotto
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Vito Lampasona
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Monica Tolazzi
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Raphaël Ho Tsong Fang
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Thibaut Naninck
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Jérôme Van Wassenhove
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Anne-Sophie Gallouët
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Pauline Maisonnasse
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Elisabeth Menu
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Nabila Seddiki
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| |
Collapse
|
11
|
Gupta D, Ahmed F, Tandel D, Parthasarathy H, Vedagiri D, Sah V, Krishna Mohan B, Khan RA, Kondiparthi C, Savari P, Jain S, Reddy S, Kumar JM, Khan N, Harshan KH. Equine immunoglobulin fragment F(ab') 2 displays high neutralizing capability against multiple SARS-CoV-2 variants. Clin Immunol 2022; 237:108981. [PMID: 35306171 PMCID: PMC8926440 DOI: 10.1016/j.clim.2022.108981] [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: 11/08/2021] [Revised: 02/02/2022] [Accepted: 03/12/2022] [Indexed: 01/04/2023]
Abstract
Neutralizing antibody-based passive immunotherapy could be an important therapeutic option against COVID-19. Herein, we demonstrate that equines hyper-immunized with chemically inactivated SARS-CoV-2 elicited high antibody titers with a strong virus-neutralizing potential, and F(ab')2 fragments purified from them displayed strong neutralization potential against five different SARS-CoV-2 variants. F(ab')2 fragments purified from the plasma of hyperimmunized horses showed high antigen-specific affinity. Experiments in rabbits suggested that the F(ab')2 displays a linear pharmacokinetics with approximate plasma half-life of 47 h. In vitro microneutralization assays using the purified F(ab')2 displayed high neutralization titers against five different variants of SARS-CoV-2 including the Delta variant, demonstrating its potential efficacy against the emerging viral variants. In conclusion, this study demonstrates that F(ab')2 generated against SARS-CoV-2 in equines have high neutralization titers and have broad target-range against the evolving variants, making passive immunotherapy a potential regimen against the existing and evolving SARS-CoV-2 variants in combating COVID-19.
Collapse
Affiliation(s)
- Divya Gupta
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India
| | - Farhan Ahmed
- School of Life Sciences, Department of Animal Biology, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Dixit Tandel
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India,Academy for Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | | | - Dhiviya Vedagiri
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India,Academy for Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vishal Sah
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India,Academy for Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | | | - Rafiq Ahmad Khan
- School of Life Sciences, Department of Animal Biology, University of Hyderabad, Hyderabad 500046, Telangana, India
| | | | | | - Sandesh Jain
- VINS Bio Products Limited, Hyderabad 500034, Telangana, India
| | - Shashikala Reddy
- Department of Microbiology, Osmania Medical College, Koti, Hyderabad 500096, Telangana, India
| | - Jerald Mahesh Kumar
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India
| | - Nooruddin Khan
- School of Life Sciences, Department of Animal Biology, University of Hyderabad, Hyderabad 500046, Telangana, India,Corresponding authors
| | - Krishnan Harinivas Harshan
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India,Academy for Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India,Corresponding authors
| |
Collapse
|
12
|
Ajmeriya S, Kumar A, Karmakar S, Rana S, Singh H. Neutralizing Antibodies and Antibody-Dependent Enhancement in COVID-19: A Perspective. J Indian Inst Sci 2022; 102:671-687. [PMID: 35136306 PMCID: PMC8814804 DOI: 10.1007/s41745-021-00268-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
Antibody-dependent enhancement (ADE) is an alternative route of viral entry in the susceptible host cell. In this process, antiviral antibodies enhance the entry access of virus in the cells via interaction with the complement or Fc receptors leading to the worsening of infection. SARS-CoV-2 variants pose a general concern for the efficacy of neutralizing antibodies that may fail to neutralize infection, raising the possibility of a more severe form of COVID-19. Data from various studies on respiratory viruses raise the speculation that antibodies elicited against SARS-CoV-2 and during COVID-19 recovery could potentially exacerbate the infection through ADE at sub-neutralizing concentrations; this may contribute to disease pathogenesis. It is, therefore, of utmost importance to study the effectiveness of the anti-SARS-CoV-2 antibodies in COVID-19-infected subjects. Theoretically, ADE remains a general concern for the efficacy of antibodies elicited during infection, most notably in convalescent plasma therapy and in response to vaccines where it could be counterproductive.
Collapse
Affiliation(s)
- Swati Ajmeriya
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Amit Kumar
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, AIIMS, Room no 3020, Ansari Nagar, New Delhi, 110029 India
| | - Shweta Rana
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Harpreet Singh
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| |
Collapse
|
13
|
Shukla AK, Misra S. Antibody-dependent enhancement of virus infection and disease: implications in COVID-19. J Basic Clin Physiol Pharmacol 2022; 33:13-16. [PMID: 34995021 DOI: 10.1515/jbcpp-2021-0264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/20/2021] [Indexed: 01/24/2023]
Abstract
Antibody-dependent enhancement (ADE) can be seen in a variety of viruses. It has a deleterious impact on antibody treatment of viral infection. This effect was first discovered in the dengue virus, and it has since been discovered in the coronavirus. Over 213 million people have been affected by the rapid spread of the newly emerging coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19). The new coronavirus offers a significant threat and has sparked widespread concern. ADE in dengue virus and other viruses are discussed with possible effect on COVID-19 treatment and vaccine development will need to consider this phenomenon to ensure it is mitigated and avoided altogether. In these case scenarios, the role of ADE and its clinical consequences remains to be explored for this newly detected virus.
Collapse
Affiliation(s)
| | - Saurav Misra
- Department of Pharmacology, AIIMS Bhopal, Bhopal, India
| |
Collapse
|
14
|
Gartlan C, Tipton T, Salguero FJ, Sattentau Q, Gorringe A, Carroll MW. Vaccine-Associated Enhanced Disease and Pathogenic Human Coronaviruses. Front Immunol 2022; 13:882972. [PMID: 35444667 PMCID: PMC9014240 DOI: 10.3389/fimmu.2022.882972] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/14/2022] [Indexed: 01/14/2023] Open
Abstract
Vaccine-associated enhanced disease (VAED) is a difficult phenomenon to define and can be confused with vaccine failure. Using studies on respiratory syncytial virus (RSV) vaccination and dengue virus infection, we highlight known and theoretical mechanisms of VAED, including antibody-dependent enhancement (ADE), antibody-enhanced disease (AED) and Th2-mediated pathology. We also critically review the literature surrounding this phenomenon in pathogenic human coronaviruses, including MERS-CoV, SARS-CoV-1 and SARS-CoV-2. Poor quality histopathological data and a lack of consistency in defining severe pathology and VAED in preclinical studies of MERS-CoV and SARS-CoV-1 vaccines in particular make it difficult to interrogate potential cases of VAED. Fortuitously, there have been only few reports of mild VAED in SARS-CoV-2 vaccination in preclinical models and no observations in their clinical use. We describe the problem areas and discuss methods to improve the characterisation of VAED in the future.
Collapse
Affiliation(s)
- Cillian Gartlan
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tom Tipton
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Francisco J Salguero
- Research and Evaluation, UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Quentin Sattentau
- The Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Andrew Gorringe
- Research and Evaluation, UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Miles W Carroll
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Development of a SARS-CoV-2 Vaccine Candidate Using Plant-Based Manufacturing and a Tobacco Mosaic Virus-like Nano-Particle. Vaccines (Basel) 2021; 9:vaccines9111347. [PMID: 34835278 PMCID: PMC8619098 DOI: 10.3390/vaccines9111347] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/19/2022] Open
Abstract
Stable, effective, easy-to-manufacture vaccines are critical to stopping the COVID-19 pandemic resulting from the coronavirus SARS-CoV-2. We constructed a vaccine candidate CoV-RBD121-NP, which is comprised of the SARS-CoV-2 receptor-binding domain (RBD) of the spike glycoprotein (S) fused to a human IgG1 Fc domain (CoV-RBD121) and conjugated to a modified tobacco mosaic virus (TMV) nanoparticle. In vitro, CoV-RBD121 bound to the host virus receptor ACE2 and to the monoclonal antibody CR3022, a neutralizing antibody that blocks S binding to ACE2. The CoV-RBD121-NP vaccine candidate retained key SARS-CoV-2 spike protein epitopes, had consistent manufacturing release properties of safety, identity, and strength, and displayed stable potency when stored for 12 months at 2–8 °C or 22–28 °C. Immunogenicity studies revealed strong antibody responses in C57BL/6 mice with non-adjuvanted or adjuvanted (7909 CpG) formulations. The non-adjuvanted vaccine induced a balanced Th1/Th2 response and antibodies that recognized both the S1 domain and full S protein from SARS2-CoV-2, whereas the adjuvanted vaccine induced a Th1-biased response. Both adjuvanted and non-adjuvanted vaccines induced virus neutralizing titers as measured by three different assays. Collectively, these data showed the production of a stable candidate vaccine for COVID-19 through the association of the SARS-CoV-2 RBD with the TMV-like nanoparticle.
Collapse
|
16
|
Development of an Inactivated Vaccine against SARS CoV-2. Vaccines (Basel) 2021; 9:vaccines9111266. [PMID: 34835197 PMCID: PMC8624180 DOI: 10.3390/vaccines9111266] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 12/15/2022] Open
Abstract
The rapid spread of SARS-CoV-2 with its mutating strains has posed a global threat to safety during this COVID-19 pandemic. Thus far, there are 123 candidate vaccines in human clinical trials and more than 190 candidates in preclinical development worldwide as per the WHO on 1 October 2021. The various types of vaccines that are currently approved for emergency use include viral vectors (e.g., adenovirus, University of Oxford/AstraZeneca, Gamaleya Sputnik V, and Johnson & Johnson), mRNA (Moderna and Pfizer-BioNTech), and whole inactivated (Sinovac Biotech and Sinopharm) vaccines. Amidst the emerging cases and shortages of vaccines for global distribution, it is vital to develop a vaccine candidate that recapitulates the severe and fatal progression of COVID-19 and further helps to cope with the current outbreak. Hence, we present the preclinical immunogenicity, protective efficacy, and safety evaluation of a whole-virion inactivated SARS-CoV-2 vaccine candidate (ERUCoV-VAC) formulated in aluminium hydroxide, in three animal models, BALB/c mice, transgenic mice (K18-hACE2), and ferrets. The hCoV-19/Turkey/ERAGEM-001/2020 strain was used for the safety evaluation of ERUCoV-VAC. It was found that ERUCoV-VAC was highly immunogenic and elicited a strong immune response in BALB/c mice. The protective efficacy of the vaccine in K18-hACE2 showed that ERUCoV-VAC induced complete protection of the mice from a lethal SARS-CoV-2 challenge. Similar viral clearance rates with the safety evaluation of the vaccine in upper respiratory tracts were also positively appreciable in the ferret models. ERUCoV-VAC has been authorized by the Turkish Medicines and Medical Devices Agency and has now entered phase 3 clinical development (NCT04942405). The name of ERUCoV-VAC has been changed to TURKOVAC in the phase 3 clinical trial.
Collapse
|
17
|
A live measles-vectored COVID-19 vaccine induces strong immunity and protection from SARS-CoV-2 challenge in mice and hamsters. Nat Commun 2021; 12:6277. [PMID: 34725327 PMCID: PMC8560864 DOI: 10.1038/s41467-021-26506-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 10/05/2021] [Indexed: 11/08/2022] Open
Abstract
Several COVID-19 vaccines have now been deployed to tackle the SARS-CoV-2 pandemic, most of them based on messenger RNA or adenovirus vectors.The duration of protection afforded by these vaccines is unknown, as well as their capacity to protect from emerging new variants. To provide sufficient coverage for the world population, additional strategies need to be tested. The live pediatric measles vaccine (MV) is an attractive approach, given its extensive safety and efficacy history, along with its established large-scale manufacturing capacity. We develop an MV-based SARS-CoV-2 vaccine expressing the prefusion-stabilized, membrane-anchored full-length S antigen, which proves to be efficient at eliciting strong Th1-dominant T-cell responses and high neutralizing antibody titers. In both mouse and golden Syrian hamster models, these responses protect the animals from intranasal infectious challenge. Additionally, the elicited antibodies efficiently neutralize in vitro the three currently circulating variants of SARS-CoV-2.
Collapse
|
18
|
In Silico Modeling as a Perspective in Developing Potential Vaccine Candidates and Therapeutics for COVID-19. COATINGS 2021. [DOI: 10.3390/coatings11111273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The potential of computational models to identify new therapeutics and repurpose existing drugs has gained significance in recent times. The current ‘COVID-19’ pandemic caused by the new SARS CoV2 virus has affected over 200 million people and caused over 4 million deaths. The enormity and the consequences of this viral infection have fueled the research community to identify drugs or vaccines through a relatively expeditious process. The availability of high-throughput datasets has cultivated new strategies for drug development and can provide the foundation towards effective therapy options. Molecular modeling methods using structure-based or computer-aided virtual screening can potentially be employed as research guides to identify novel antiviral agents. This review focuses on in-silico modeling of the potential therapeutic candidates against SARS CoVs, in addition to strategies for vaccine design. Here, we particularly focus on the recently published SARS CoV main protease (Mpro) active site, the RNA-dependent RNA polymerase (RdRp) of SARS CoV2, and the spike S-protein as potential targets for vaccine development. This review can offer future perspectives for further research and the development of COVID-19 therapies via the design of new drug candidates and multi-epitopic vaccines and through the repurposing of either approved drugs or drugs under clinical trial.
Collapse
|
19
|
Wu X, Cheng L, Fu M, Huang B, Zhu L, Xu S, Shi H, Zhang D, Yuan H, Nawaz W, Yang P, Hu Q, Liu Y, Wu Z. A potent bispecific nanobody protects hACE2 mice against SARS-CoV-2 infection via intranasal administration. Cell Rep 2021; 37:109869. [PMID: 34644535 PMCID: PMC8492916 DOI: 10.1016/j.celrep.2021.109869] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/08/2021] [Accepted: 09/29/2021] [Indexed: 12/18/2022] Open
Abstract
The dramatically expanding coronavirus disease 2019 (COVID-19) needs multiple effective countermeasures. Neutralizing nanobodies (Nbs) are a potential therapeutic strategy for treating COVID-19. Here, we characterize several receptor binding domain (RBD)-specific Nbs isolated from an Nb library derived from an alpaca immunized with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein (S); among them, three Nbs exhibit picomolar potency against SARS-CoV-2 live virus, pseudotyped viruses, and circulating SARS-CoV-2 variants. To improve their efficacy, various configurations of Nbs are engineered. Nb15-NbH-Nb15, a trimer constituted of three Nbs, is constructed to be bispecific for human serum albumin (HSA) and RBD of SARS-CoV-2. Nb15-NbH-Nb15 exhibits single-digit ng/ml neutralization potency against the wild-type and Delta variants of SARS-CoV-2 with a long half-life in vivo. In addition, we show that intranasal administration of Nb15-NbH-Nb15 provides effective protection for both prophylactic and therapeutic purposes against SARS-CoV-2 infection in transgenic hACE2 mice. Nb15-NbH-Nb15 is a potential candidate for both the prevention and treatment of SARS-CoV-2 through respiratory administration.
Collapse
Affiliation(s)
- Xilin Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, People's Republic of China; Department of Antibody, Abrev Biotechnology Co., Ltd., Nanjing, People's Republic of China
| | - Lin Cheng
- Institute for Hepatology, National Clinical Research Center for Infectious, Disease, Shenzhen Third People's Hospital, Shenzhen, People's Republic of China
| | - Ming Fu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, People's Republic of China; Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Bilian Huang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, People's Republic of China
| | - Linjing Zhu
- Department of Antibody, Abrev Biotechnology Co., Ltd., Nanjing, People's Republic of China
| | - Shijie Xu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, People's Republic of China; Department of Antibody, Abrev Biotechnology Co., Ltd., Nanjing, People's Republic of China
| | - Haixia Shi
- Department of Antibody, Y-clone Medical Science Co. Ltd., Suzhou, People's Republic of China
| | - Doudou Zhang
- Department of Antibody, Abrev Biotechnology Co., Ltd., Nanjing, People's Republic of China
| | - Huanyun Yuan
- Department of Antibody, Abrev Biotechnology Co., Ltd., Nanjing, People's Republic of China
| | - Waqas Nawaz
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, People's Republic of China
| | - Ping Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, People's Republic of China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, People's Republic of China; Institute for Infection and Immunity, St George's University of London, London SW17 0RE, UK
| | - Yalan Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, People's Republic of China.
| | - Zhiwei Wu
- School of Life Sciences, Ningxia University, Yinchuan, People's Republic of China; Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, People's Republic of China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, People's Republic of China.
| |
Collapse
|
20
|
Bisgin A, Sanlioglu AD, Eksi YE, Griffith TS, Sanlioglu S. Current Update on Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Development with a Special Emphasis on Gene Therapy Viral Vector Design and Construction for Vaccination. Hum Gene Ther 2021; 32:541-562. [PMID: 33858231 DOI: 10.1089/hum.2021.052] [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] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) is a newly emerging infectious disease (COVID-19) caused by the novel coronavirus SARS-coronavirus 2 (CoV-2). To combat the devastating spread of SARS-CoV-2, extraordinary efforts from numerous laboratories have focused on the development of effective and safe vaccines. Traditional live-attenuated or inactivated viral vaccines are not recommended for immunocompromised patients as the attenuated virus can still cause disease via phenotypic or genotypic reversion. Subunit vaccines require repeated dosing and adjuvant use to be effective, and DNA vaccines exhibit lower immune responses. mRNA vaccines can be highly unstable under physiological conditions. On the contrary, naturally antigenic viral vectors with well-characterized structure and safety profile serve as among the most effective gene carriers to provoke immune response via heterologous gene transfer. Viral vector-based vaccines induce both an effective cellular immune response and a humoral immune response owing to their natural adjuvant properties via transduction of immune cells. Consequently, viral vectored vaccines carrying the SARS-CoV-2 spike protein have recently been generated and successfully used to activate cytotoxic T cells and develop a neutralizing antibody response. Recent progress in SARS-CoV-2 vaccines, with an emphasis on gene therapy viral vector-based vaccine development, is discussed in this review.
Collapse
Affiliation(s)
- Atil Bisgin
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Department of Medical Genetics, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ahter D Sanlioglu
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Yunus Emre Eksi
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Thomas S Griffith
- The Department of Urology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Salih Sanlioglu
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
21
|
Blumental S, Debré P. Challenges and Issues of Anti-SARS-CoV-2 Vaccines. Front Med (Lausanne) 2021; 8:664179. [PMID: 34055838 PMCID: PMC8163222 DOI: 10.3389/fmed.2021.664179] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
At the beginning of 2021, anti-SARS-CoV-2 vaccination campaigns had been launched in almost 60 countries with more than 500 million doses having been distributed. In addition to the few vaccines already in use, many other candidates are in preclinical phases or experimental stages in humans. Despite the fact that the availability of anti-SARS-CoV-2 vaccine constitutes a major advance and appear to be the only way to control the pandemic, some investigation remains to be carried out, and this is notably concerning the impact on transmissibility, the duration of the conferred protection in the mid- and long term, the effectiveness against present and future viral mutants, or the ideal schedule that should be applied. In this paper, we review the circumstances that facilitated such a rapid development of anti-SARS-CoV-2 vaccines and summarize the different vaccine platforms under investigation as well as their present results and perspectives in different settings. We also discuss the indications of vaccination under special conditions, such as a history of previous COVID-19 infection or belonging to extreme age categories like children and elderly. Overall, this review highlights the multiple challenges to face if aiming to find a global solution to the pandemic through high vaccination coverage all over the world.
Collapse
Affiliation(s)
- Sophie Blumental
- Pediatric Infectious Disease Unit, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Patrice Debré
- Immunology Department, APHP, Sorbonne Université CIMI (Inserm U1135), Hôpital Pitie Salpêtrière, Paris, France
| |
Collapse
|
22
|
Guo Y, He W, Mou H, Zhang L, Chang J, Peng S, Ojha A, Tavora R, Parcells MS, Luo G, Li W, Zhong G, Choe H, Farzan M, Quinlan BD. An Engineered Receptor-Binding Domain Improves the Immunogenicity of Multivalent SARS-CoV-2 Vaccines. mBio 2021; 12:mBio.00930-21. [PMID: 33975938 DOI: 10.1101/2020.04.10.036418] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein mediates viral entry into cells expressing angiotensin-converting enzyme 2 (ACE2). The S protein engages ACE2 through its receptor-binding domain (RBD), an independently folded 197-amino-acid fragment of the 1,273-amino-acid S-protein protomer. The RBD is the primary SARS-CoV-2 neutralizing epitope and a critical target of any SARS-CoV-2 vaccine. Here, we show that this RBD conjugated to each of two carrier proteins elicited more potent neutralizing responses in immunized rodents than did a similarly conjugated proline-stabilized S-protein ectodomain. Nonetheless, the native RBD is expressed inefficiently, limiting its usefulness as a vaccine antigen. However, we show that an RBD engineered with four novel glycosylation sites (gRBD) is expressed markedly more efficiently and generates a more potent neutralizing responses as a DNA vaccine antigen than the wild-type RBD or the full-length S protein, especially when fused to multivalent carriers, such as a Helicobacter pylori ferritin 24-mer. Further, gRBD is more immunogenic than the wild-type RBD when administered as a subunit protein vaccine. Our data suggest that multivalent gRBD antigens can reduce costs and doses, and improve the immunogenicity, of all major classes of SARS-CoV-2 vaccines.IMPORTANCE All available vaccines for coronavirus disease 2019 (COVID-19) express or deliver the full-length SARS-CoV-2 spike (S) protein. We show that this antigen is not optimal, consistent with observations that the vast majority of the neutralizing response to the virus is focused on the S-protein receptor-binding domain (RBD). However, this RBD is not expressed well as an independent domain, especially when expressed as a fusion protein with a multivalent scaffold. We therefore engineered a more highly expressed form of the SARS-CoV-2 RBD by introducing four glycosylation sites into a face of the RBD normally occluded in the full S protein. We show that this engineered protein, gRBD, is more immunogenic than the wild-type RBD or the full-length S protein in both genetic and protein-delivered vaccines.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/immunology
- Animals
- Binding Sites
- COVID-19 Vaccines/chemistry
- COVID-19 Vaccines/immunology
- Female
- Genetic Engineering
- Glycosylation
- HEK293 Cells
- Humans
- Immunogenicity, Vaccine
- Mice
- Mice, Inbred BALB C
- Models, Molecular
- Protein Domains
- Rats
- Rats, Sprague-Dawley
- Receptors, Coronavirus/genetics
- Receptors, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Conjugate/genetics
- Vaccines, Conjugate/immunology
- Vaccines, Synthetic/chemistry
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Yan Guo
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Wenhui He
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Huihui Mou
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Lizhou Zhang
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Jing Chang
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Shoujiao Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Amrita Ojha
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Rubens Tavora
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Mark S Parcells
- Department of Animal and Food Sciences, University of Delaware, Newark, Delaware, USA
| | - Guangxiang Luo
- Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Wenhui Li
- National Institute of Biological Sciences, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Guocai Zhong
- Scripps Research SZBL Chemical Biology Institute, Shenzhen Bay Laboratory, Shenzhen, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Hyeryun Choe
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Michael Farzan
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Brian D Quinlan
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| |
Collapse
|
23
|
Zhan W, Muhuri M, Tai PWL, Gao G. Vectored Immunotherapeutics for Infectious Diseases: Can rAAVs Be The Game Changers for Fighting Transmissible Pathogens? Front Immunol 2021; 12:673699. [PMID: 34046041 PMCID: PMC8144494 DOI: 10.3389/fimmu.2021.673699] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Conventional vaccinations and immunotherapies have encountered major roadblocks in preventing infectious diseases like HIV, influenza, and malaria. These challenges are due to the high genomic variation and immunomodulatory mechanisms inherent to these diseases. Passive transfer of broadly neutralizing antibodies may offer partial protection, but these treatments require repeated dosing. Some recombinant viral vectors, such as those based on lentiviruses and adeno-associated viruses (AAVs), can confer long-term transgene expression in the host after a single dose. Particularly, recombinant (r)AAVs have emerged as favorable vectors, given their high in vivo transduction efficiency, proven clinical efficacy, and low immunogenicity profiles. Hence, rAAVs are being explored to deliver recombinant antibodies to confer immunity against infections or to diminish the severity of disease. When used as a vaccination vector for the delivery of antigens, rAAVs enable de novo synthesis of foreign proteins with the conformation and topology that resemble those of natural pathogens. However, technical hurdles like pre-existing immunity to the rAAV capsid and production of anti-drug antibodies can reduce the efficacy of rAAV-vectored immunotherapies. This review summarizes rAAV-based prophylactic and therapeutic strategies developed against infectious diseases that are currently being tested in pre-clinical and clinical studies. Technical challenges and potential solutions will also be discussed.
Collapse
Affiliation(s)
- Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Manish Muhuri
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Phillip W. L. Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
24
|
Szekely L, Bozoky B, Bendek M, Ostad M, Lavignasse P, Haag L, Wu J, Jing X, Gupta S, Saccon E, Sönnerborg A, Cao Y, Björnstedt M, Szakos A. Pulmonary stromal expansion and intra-alveolar coagulation are primary causes of COVID-19 death. Heliyon 2021; 7:e07134. [PMID: 34056141 PMCID: PMC8141733 DOI: 10.1016/j.heliyon.2021.e07134] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/04/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022] Open
Abstract
Most COVID-19 victims are old and die from unrelated causes. Here we present twelve complete autopsies, including two rapid autopsies of young patients where the cause of death was COVID-19 ARDS. The main virus induced pathology was in the lung parenchyma and not in the airways. Most coagulation events occurred in the intra-alveolar and not in the intra-vascular space and the few thrombi were mainly composed of aggregated thrombocytes. The dominant inflammatory response was the massive accumulation of CD163 + macrophages and the disappearance of T killer, NK and B-cells. The virus was replicating in the pneumocytes and macrophages but not in bronchial epithelium, endothelium, pericytes or stromal cells. The lung consolidations were produced by a massive regenerative response, stromal and epithelial proliferation and neovascularization. We suggest that thrombocyte aggregation inhibition, angiogenesis inhibition and general proliferation inhibition may have a roll in the treatment of advanced COVID-19 ARDS.
Collapse
Affiliation(s)
- Laszlo Szekely
- Department of Pathology/Cytology, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| | - Bela Bozoky
- Department of Pathology/Cytology, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| | - Matyas Bendek
- Department of Pathology/Cytology, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| | - Masih Ostad
- Department of Pathology/Cytology, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| | - Pablo Lavignasse
- Department of Pathology/Cytology, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| | - Lars Haag
- Department of Pathology/Cytology, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| | - Jieyu Wu
- Microbiology and Tumor Biology Center, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Xu Jing
- Microbiology and Tumor Biology Center, Karolinska Institutet, 171 77 Stockholm, Sweden
- The Second Hospital of Shandong University, Department of Clinical Laboratory, 250033 Jinan, China
| | - Soham Gupta
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Elisa Saccon
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Anders Sönnerborg
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Yihai Cao
- Microbiology and Tumor Biology Center, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mikael Björnstedt
- Department of Pathology/Cytology, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| | - Attila Szakos
- Department of Pathology/Cytology, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| |
Collapse
|
25
|
Oesch F, Oesch-Bartlomowicz B, Efferth T. Toxicity as prime selection criterion among SARS-active herbal medications. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153476. [PMID: 33593628 PMCID: PMC7840405 DOI: 10.1016/j.phymed.2021.153476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 05/06/2023]
Abstract
We present here a new selection criterion for prioritizing research on efficacious drugs for the fight against COVID-19: the relative toxicity versus safety of herbal medications, which were effective against SARS in the 2002/2003 epidemic. We rank these medicines according to their toxicity versus safety as basis for preferential rapid research on their potential in the treatment of COVID-19. The data demonstrate that from toxicological information nothing speaks against immediate investigation on, followed by rapid implementation of Lonicera japonica, Morus alba, Forsythia suspensa, and Codonopsis spec. for treatment of COVID-19 patients. Glycyrrhiza spec. and Panax ginseng are ranked in second priority and ephedrine-free Herba Ephedrae extract in third priority (followed by several drugs in lower preferences). Rapid research on their efficacy in the therapy - as well as safety under the specific circumstances of COVID-19 - followed by equally rapid implementation will provide substantial advantages to Public Health including immediate availability, enlargement of medicinal possibilities, in cases where other means are not successful (non-responders), not tolerated (sensitive individuals) or just not available (as is presently the case) and thus minimize sufferings and save lives. Moreover, their moderate costs and convenient oral application are especially advantageous for underprivileged populations in developing countries.
Collapse
Affiliation(s)
- Franz Oesch
- Institute of Toxicology, Johannes Gutenberg University, 55131 Mainz, Germany.
| | | | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128, Mainz, Germany
| |
Collapse
|
26
|
Yao YF, Wang ZJ, Jiang RD, Hu X, Zhang HJ, Zhou YW, Gao G, Chen Y, Peng Y, Liu MQ, Zhang YN, Min J, Lu J, Gao XX, Guo J, Peng C, Shen XR, Li Q, Zhao K, Yang L, Wan X, Zhang B, Wang WH, Wu J, Zhou P, Yang XL, Shen S, Shan C, Yuan ZM, Shi ZL. Protective Efficacy of Inactivated Vaccine against SARS-CoV-2 Infection in Mice and Non-Human Primates. Virol Sin 2021; 36:879-889. [PMID: 33835391 PMCID: PMC8034048 DOI: 10.1007/s12250-021-00376-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 02/08/2021] [Indexed: 12/21/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused more than 96 million infections and over 2 million deaths worldwide so far. However, there is no approved vaccine available for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the disease causative agent. Vaccine is the most effective approach to eradicate a pathogen. The tests of safety and efficacy in animals are pivotal for developing a vaccine and before the vaccine is applied to human populations. Here we evaluated the safety, immunogenicity, and efficacy of an inactivated vaccine based on the whole viral particles in human ACE2 transgenic mouse and in non-human primates. Our data showed that the inactivated vaccine successfully induced SARS-CoV-2-specific neutralizing antibodies in mice and non-human primates, and subsequently provided partial (in low dose) or full (in high dose) protection of challenge in the tested animals. In addition, passive serum transferred from vaccine-immunized mice could also provide full protection from SARS-CoV-2 infection in mice. These results warranted positive outcomes in future clinical trials in humans.
Collapse
Affiliation(s)
- Yan-Feng Yao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ze-Jun Wang
- Wuhan Institute of Biological Products Co. Ltd, Jiangxia District, Wuhan, 430024, China
| | - Ren-Di Jiang
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue Hu
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Hua-Jun Zhang
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yi-Wu Zhou
- Department of Forensic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ge Gao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ying Chen
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yun Peng
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Mei-Qin Liu
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ya-Nan Zhang
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Juan Min
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jia Lu
- Wuhan Institute of Biological Products Co. Ltd, Jiangxia District, Wuhan, 430024, China
| | - Xiao-Xiao Gao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jing Guo
- Wuhan Institute of Biological Products Co. Ltd, Jiangxia District, Wuhan, 430024, China
| | - Cheng Peng
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xu-Rui Shen
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian Li
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai Zhao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xin Wan
- Wuhan Institute of Biological Products Co. Ltd, Jiangxia District, Wuhan, 430024, China
| | - Bo Zhang
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Wen-Hui Wang
- Wuhan Institute of Biological Products Co. Ltd, Jiangxia District, Wuhan, 430024, China
| | - Jia Wu
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Peng Zhou
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xing-Lou Yang
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Shuo Shen
- Wuhan Institute of Biological Products Co. Ltd, Jiangxia District, Wuhan, 430024, China.
| | - Chao Shan
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Zhi-Ming Yuan
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Zheng-Li Shi
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
27
|
Winkler ES, Gilchuk P, Yu J, Bailey AL, Chen RE, Chong Z, Zost SJ, Jang H, Huang Y, Allen JD, Case JB, Sutton RE, Carnahan RH, Darling TL, Boon ACM, Mack M, Head RD, Ross TM, Crowe JE, Diamond MS. Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions for optimal therapeutic protection. Cell 2021; 184:1804-1820.e16. [PMID: 33691139 PMCID: PMC7879018 DOI: 10.1016/j.cell.2021.02.026] [Citation(s) in RCA: 264] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/17/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
SARS-CoV-2 has caused the global COVID-19 pandemic. Although passively delivered neutralizing antibodies against SARS-CoV-2 show promise in clinical trials, their mechanism of action in vivo is incompletely understood. Here, we define correlates of protection of neutralizing human monoclonal antibodies (mAbs) in SARS-CoV-2-infected animals. Whereas Fc effector functions are dispensable when representative neutralizing mAbs are administered as prophylaxis, they are required for optimal protection as therapy. When given after infection, intact mAbs reduce SARS-CoV-2 burden and lung disease in mice and hamsters better than loss-of-function Fc variant mAbs. Fc engagement of neutralizing antibodies mitigates inflammation and improves respiratory mechanics, and transcriptional profiling suggests these phenotypes are associated with diminished innate immune signaling and preserved tissue repair. Immune cell depletions establish that neutralizing mAbs require monocytes and CD8+ T cells for optimal clinical and virological benefit. Thus, potently neutralizing mAbs utilize Fc effector functions during therapy to mitigate lung infection and disease.
Collapse
Affiliation(s)
- Emma S Winkler
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jinsheng Yu
- Department of Genetics, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Adam L Bailey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Rita E Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Zhenlu Chong
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Seth J Zost
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hyesun Jang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - Ying Huang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - James D Allen
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Rachel E Sutton
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Tamarand L Darling
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
28
|
Vishwakarma P, Yadav N, Rizvi ZA, Khan NA, Chiranjivi AK, Mani S, Bansal M, Dwivedi P, Shrivastava T, Kumar R, Awasthi A, Ahmed S, Samal S. Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein Based Novel Epitopes Induce Potent Immune Responses in vivo and Inhibit Viral Replication in vitro. Front Immunol 2021; 12:613045. [PMID: 33841395 PMCID: PMC8032902 DOI: 10.3389/fimmu.2021.613045] [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] [Received: 10/01/2020] [Accepted: 02/09/2021] [Indexed: 11/13/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) initiates infection by attachment of the surface-exposed spike glycoprotein to the host cell receptors. The spike glycoprotein (S) is a promising target for inducing immune responses and providing protection; thus the ongoing efforts for the SARS-CoV-2 vaccine and therapeutic developments are mostly spiraling around S glycoprotein. The matured functional spike glycoprotein is presented on the virion surface as trimers, which contain two subunits, such as S1 (virus attachment) and S2 (virus fusion). The S1 subunit harbors the N-terminal domain (NTD) and the receptor-binding domain (RBD). The RBD is responsible for binding to host-cellular receptor angiotensin-converting enzyme 2 (ACE2). The NTD and RBD of S1, and the S2 of S glycoprotein are the major structural moieties to design and develop spike-based vaccine candidates and therapeutics. Here, we have identified three novel epitopes (20-amino acid peptides) in the regions NTD, RBD, and S2 domains, respectively, by structural and immunoinformatic analysis. We have shown as a proof of principle in the murine model, the potential role of these novel epitopes in-inducing humoral and cellular immune responses. Further analysis has shown that RBD and S2 directed epitopes were able to efficiently inhibit the replication of SARS-CoV-2 wild-type virus in vitro suggesting their role as virus entry inhibitors. Structural analysis revealed that S2-epitope is a part of the heptad repeat 2 (HR2) domain which might have plausible inhibitory effects on virus fusion. Taken together, this study discovered novel epitopes that might have important implications in the development of potential SARS-CoV-2 spike-based vaccine and therapeutics.
Collapse
Affiliation(s)
- Preeti Vishwakarma
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Naveen Yadav
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Zaigham Abbas Rizvi
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Naseem Ahmed Khan
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Adarsh Kumar Chiranjivi
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Shailendra Mani
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Manish Bansal
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Prabhanjan Dwivedi
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Tripti Shrivastava
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Rajesh Kumar
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Amit Awasthi
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Shubbir Ahmed
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Sweety Samal
- Translational Health Science & Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| |
Collapse
|
29
|
Prompetchara E, Ketloy C, Tharakhet K, Kaewpang P, Buranapraditkun S, Techawiwattanaboon T, Sathean-anan-kun S, Pitakpolrat P, Watcharaplueksadee S, Phumiamorn S, Wijagkanalan W, Patarakul K, Palaga T, Ruxrungtham K. DNA vaccine candidate encoding SARS-CoV-2 spike proteins elicited potent humoral and Th1 cell-mediated immune responses in mice. PLoS One 2021; 16:e0248007. [PMID: 33750975 PMCID: PMC7984610 DOI: 10.1371/journal.pone.0248007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/17/2021] [Indexed: 12/23/2022] Open
Abstract
More than 65 million people have been confirmed infection with SARS-CoV-2 and more than 1 million have died from COVID-19 and this pandemic remains critical worldwide. Effective vaccines are one of the most important strategies to limit the pandemic. Here, we report a construction strategy of DNA vaccine candidates expressing full length wild type SARS-CoV-2 spike (S) protein, S1 or S2 region and their immunogenicity in mice. All DNA vaccine constructs of pCMVkan-S, -S1 and -S2 induced high levels of specific binding IgG that showed a balance of IgG1/IgG2a response. However, only the sera from mice vaccinated with pCMKkan-S or -S1 DNA vaccines could inhibit viral RBD and ACE2 interaction. The highest neutralizing antibody (NAb) titer was found in pCMVkan-S group, followed by -S1, while -S2 showed the lowest PRNT50 titers. The geometric mean titers (GMTs) were 2,551, 1,005 and 291 for pCMVkan-S, -S1 and -S2, respectively. pCMVkan-S construct vaccine also induced the highest magnitude and breadth of T cells response. Analysis of IFN-γ positive cells after stimulation with SARS-CoV-2 spike peptide pools were 2,991, 1,376 and 1,885 SFC/106 splenocytes for pCMVkan-S, -S1 and -S2, respectively. Our findings highlighted that full-length S antigen is more potent than the truncated spike (S1 or S2) in inducing of neutralizing antibody and robust T cell responses.
Collapse
Affiliation(s)
- Eakachai Prompetchara
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Integrated Frontier Biotechnology for Emerging Disease, Chulalongkorn University, Bangkok, Thailand
| | - Chutitorn Ketloy
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Integrated Frontier Biotechnology for Emerging Disease, Chulalongkorn University, Bangkok, Thailand
| | - Kittipan Tharakhet
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Papatsara Kaewpang
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supranee Buranapraditkun
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Teerasit Techawiwattanaboon
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Suwitra Sathean-anan-kun
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Patrawadee Pitakpolrat
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supaporn Watcharaplueksadee
- Thai Red Cross Emerging Infectious Diseases-Health Science Centre, World Health Organization Collaborating Centre for Research and Training on Viral Zoonoses, King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supaporn Phumiamorn
- Institute of Biological Product, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | | | - Kanitha Patarakul
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Integrated Frontier Biotechnology for Emerging Disease, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tanapat Palaga
- Integrated Frontier Biotechnology for Emerging Disease, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Kiat Ruxrungtham
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Integrated Frontier Biotechnology for Emerging Disease, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
30
|
Wang S, Ye J, Kang Z, Peng H, Mackey V, Sun L. The COVID-19 pandemic and the potential treatment of the novel coronavirus SARS-CoV-2. Am J Transl Res 2021; 13:871-881. [PMID: 33841627 PMCID: PMC8014381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/04/2020] [Indexed: 06/12/2023]
Abstract
Coronavirus SARS-CoV-2 is a novel coronavirus and the seventh that can infect human beings and result in severe and acute respiratory syndrome and deaths. Currently, the world is undergoing a global health emergency due to the SARS-CoV-2 pandemic. As of May 18, SARS-CoV-2 has spread to over two hundred countries and infected more than 4.8 million people, resulting in over 300,000 deaths since the first case of a novel pneumonia (COVID-19) patient was discovered in Wuhan, China at the end of December 2019. Currently, there are no effective and/or approved targeting drugs for it though various supportive therapy drugs such as small molecule drugs, vaccines, antibodies and even Chinese herb medicines have been used in the treatment of the first-line patients. However, certain drugs such as remdesivir and S416 are under clinical investigation and may become therapeutic drugs. In this article, we review and discuss SARS-CoV-2, its person-to-person transmission, genomics and proteomics, and the potential for drug development.
Collapse
Affiliation(s)
- Shilei Wang
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd.Guangdong, China
| | - Jinlei Ye
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd.Guangdong, China
| | - Zhichao Kang
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd.Guangdong, China
| | - Hongmei Peng
- Scientific Research and Education Department, The First Peoples Hospital of Changde CityChangde 415000, Hunan, China
| | - Vienna Mackey
- Department of Medicine, School of Medicine, Tulane University Health Sciences CenterNew Orleans, USA
| | - Lichun Sun
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd.Guangdong, China
- Department of Medicine, School of Medicine, Tulane University Health Sciences CenterNew Orleans, USA
- Sino-US Innovative Bio-Medical Center and Hunan Beautide PharmaceuticalsXiangtan, Hunan, China
| |
Collapse
|
31
|
Maeda K, Higashi-Kuwata N, Kinoshita N, Kutsuna S, Tsuchiya K, Hattori SI, Matsuda K, Takamatsu Y, Gatanaga H, Oka S, Sugiyama H, Ohmagari N, Mitsuya H. Neutralization of SARS-CoV-2 with IgG from COVID-19-convalescent plasma. Sci Rep 2021; 11:5563. [PMID: 33692457 PMCID: PMC7946899 DOI: 10.1038/s41598-021-84733-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/19/2021] [Indexed: 12/28/2022] Open
Abstract
While there are various attempts to administer COVID-19-convalescent plasmas to SARS-CoV-2-infected patients, neither appropriate approach nor clinical utility has been established. We examined the presence and temporal changes of the neutralizing activity of IgG fractions from 43 COVID-19-convalescent plasmas using cell-based assays with multiple endpoints. IgG fractions from 27 cases (62.8%) had significant neutralizing activity and moderately to potently inhibited SARS-CoV-2 infection in cell-based assays; however, no detectable neutralizing activity was found in 16 cases (37.2%). Approximately half of the patients (~ 41%), who had significant neutralizing activity, lost the neutralization activity within ~ 1 month. Despite the rapid decline of neutralizing activity in plasmas, good amounts of SARS-CoV-2-S1-binding antibodies were persistently seen. The longer exposure of COVID-19 patients to greater amounts of SARS-CoV-2 elicits potent immune response to SARS-CoV-2, producing greater neutralization activity and SARS-CoV-2-S1-binding antibody amounts. The dilution of highly-neutralizing plasmas with poorly-neutralizing plasmas relatively readily reduced neutralizing activity. The presence of good amounts of SARS-CoV-2-S1-binding antibodies does not serve as a surrogate ensuring the presence of good neutralizing activity. In selecting good COVID-19-convalescent plasmas, quantification of neutralizing activity in each plasma sample before collection and use is required.
Collapse
Affiliation(s)
- Kenji Maeda
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan.
| | - Nobuyo Higashi-Kuwata
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan
| | | | - Satoshi Kutsuna
- Disease Control and Prevention Center (DCC), NCGM, Tokyo, Japan
| | | | - Shin-Ichiro Hattori
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan
| | - Kouki Matsuda
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan
| | - Yuki Takamatsu
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan
| | | | | | | | - Norio Ohmagari
- Disease Control and Prevention Center (DCC), NCGM, Tokyo, Japan
| | - Hiroaki Mitsuya
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan.
- Experimental Retrovirology Section, HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
- Department of Clinical Sciences, Kumamoto University Hospital, Kumamoto, Japan.
| |
Collapse
|
32
|
West R, Kobokovich A, Connell N, Gronvall GK. COVID-19 Antibody Tests: A Valuable Public Health Tool with Limited Relevance to Individuals. Trends Microbiol 2021; 29:214-223. [PMID: 33234439 PMCID: PMC7836413 DOI: 10.1016/j.tim.2020.11.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022]
Abstract
Antibody tests for detecting past infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have many uses for public health decision making, but demand has largely come from individual consumers. This review focuses on the individual relevance of antibody tests: their accuracy in detecting prior infection, what past SARS-CoV-2 infection can currently infer about future immunity or possible medical sequelae, and the potential future importance of antibody tests for vaccine selection and medical screening. Given uncertainty about the antibody tests (quality, accuracy level, positive predictive value) and what those tests might indicate immunologically (durability of antibodies and necessity for protection from reinfection), seropositive test results should not be used to inform individual decision making, and antibody testing should remain a tool of public health at this time.
Collapse
Affiliation(s)
- Rachel West
- Johns Hopkins Center for Health Security, Baltimore, MD, USA
| | | | - Nancy Connell
- Johns Hopkins Center for Health Security, Baltimore, MD, USA
| | | |
Collapse
|
33
|
Mehmood I, Ijaz M, Ahmad S, Ahmed T, Bari A, Abro A, Allemailem KS, Almatroudi A, Tahir ul Qamar M. SARS-CoV-2: An Update on Genomics, Risk Assessment, Potential Therapeutics and Vaccine Development. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041626. [PMID: 33567746 PMCID: PMC7915969 DOI: 10.3390/ijerph18041626] [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] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a great threat to public health, being a causative pathogen of a deadly coronavirus disease (COVID-19). It has spread to more than 200 countries and infected millions of individuals globally. Although SARS-CoV-2 has structural/genomic similarities with the previously reported SARS-CoV and MERS-CoV, the specific mutations in its genome make it a novel virus. Available therapeutic strategies failed to control this virus. Despite strict standard operating procedures (SOPs), SARS-CoV-2 has spread globally and it is mutating gradually as well. Diligent efforts, special care, and awareness are needed to reduce transmission among susceptible masses particularly elder people, children, and health care workers. In this review, we highlighted the basic genome organization and structure of SARS-CoV-2. Its transmission dynamics, symptoms, and associated risk factors are discussed. This review also presents the latest mutations identified in its genome, the potential therapeutic options being used, and a brief explanation of vaccine development efforts against COVID-19. The effort will not only help readers to understand the deadly SARS-CoV-2 virus but also provide updated information to researchers for their research work.
Collapse
Affiliation(s)
- Iqra Mehmood
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (I.M.); (M.I.)
| | - Munazza Ijaz
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (I.M.); (M.I.)
| | - Sajjad Ahmad
- Department of Microbiology and Pharmacy, Abasyn University, Peshawar 25000, Pakistan;
| | - Temoor Ahmed
- Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China;
| | - Amna Bari
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China;
| | - Asma Abro
- Department of Biotechnology, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology Engineering and Management Sciences, Quetta 87100, Pakistan;
| | - Khaled S. Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
- Correspondence: (A.A.); (M.T.u.Q.)
| | - Muhammad Tahir ul Qamar
- College of Life Science and Technology, Guangxi University, Nanning 530004, China
- Correspondence: (A.A.); (M.T.u.Q.)
| |
Collapse
|
34
|
Chandrasekar SS, Phanse Y, Hildebrand RE, Hanafy M, Wu CW, Hansen CH, Osorio JE, Suresh M, Talaat AM. Localized and Systemic Immune Responses against SARS-CoV-2 Following Mucosal Immunization. Vaccines (Basel) 2021; 9:132. [PMID: 33562141 PMCID: PMC7914464 DOI: 10.3390/vaccines9020132] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
The rapid transmission of SARS-CoV-2 in the USA and worldwide necessitates the development of multiple vaccines to combat the COVID-19 global pandemic. Previously, we showed that a particulate adjuvant system, quil-A-loaded chitosan (QAC) nanoparticles, can elicit robust immunity combined with plasmid vaccines when used against avian coronavirus. Here, we report on the immune responses elicited by mucosal homologous plasmid and a heterologous immunization strategy using a plasmid vaccine and a Modified Vaccinia Ankara (MVA) expressing SARS-CoV-2 spike (S) and nucleocapsid (N) antigens. Only the heterologous intranasal immunization strategy elicited neutralizing antibodies against SARS-CoV-2 in serum and bronchoalveolar lavage of mice, suggesting a protective vaccine. The same prime/boost strategy led to the induction of type 1 and type 17 T-cell responses and polyfunctional T-cells expressing multiple type 1 cytokines (e.g., IFN-γ, TNFα, IL-2) in the lungs and spleens of vaccinated mice. In contrast, the plasmid homologous vaccine strategy led to the induction of local mono and polyfunctional T-cells secreting IFN-γ. Outcomes of this study support the potential of QAC-nano vaccines to elicit significant mucosal immune responses against respiratory coronaviruses.
Collapse
Affiliation(s)
- Shaswath S. Chandrasekar
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | | | - Rachel E. Hildebrand
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | - Mostafa Hanafy
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Chia-Wei Wu
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | - Chungyi H. Hansen
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | - Jorge E. Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
- Colombia Wisconsin One Health Consortium, Universidad Nacional Medellín, Calle 75#79a 5, Colombia
| | - M. Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
| | - Adel M. Talaat
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (S.S.C.); (R.E.H.); (M.H.); (C.-W.W.); (C.H.H.); (J.E.O.); (M.S.)
- Pan Genome Systems, Madison, WI 53719, USA;
| |
Collapse
|
35
|
Abstract
Vaccines are urgently needed to control the coronavirus disease 2019 (COVID-19) pandemic and to help the return to pre-pandemic normalcy. A great many vaccine candidates are being developed, several of which have completed late-stage clinical trials and are reporting positive results. In this Progress article, we discuss which viral elements are used in COVID-19 vaccine candidates, why they might act as good targets for the immune system and the implications for protective immunity.
Collapse
Affiliation(s)
- Lianpan Dai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
36
|
Hong J, Jhun H, Choi YO, Taitt AS, Bae S, Lee Y, Song CS, Yeom SC, Kim S. Structure of SARS-CoV-2 Spike Glycoprotein for Therapeutic and Preventive Target. Immune Netw 2021; 21:e8. [PMID: 33728101 PMCID: PMC7937506 DOI: 10.4110/in.2021.21.e8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/08/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022] Open
Abstract
The global crisis caused by the coronavirus disease 2019 (COVID-19) led to the most significant economic loss and human deaths after World War II. The pathogen causing this disease is a novel virus called the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As of December 2020, there have been 80.2 million confirmed patients, and the mortality rate is known as 2.16% globally. A strategy to protect a host from SARS-CoV-2 is by suppressing intracellular viral replication or preventing viral entry. We focused on the spike glycoprotein that is responsible for the entry of SARS-CoV-2 into the host cell. Recently, the US Food and Drug Administration/EU Medicines Agency authorized a vaccine and antibody to treat COVID-19 patients by emergency use approval in the absence of long-term clinical trials. Both commercial and academic efforts to develop preventive and therapeutic agents continue all over the world. In this review, we present a perspective on current reports about the spike glycoprotein of SARS-CoV-2 as a therapeutic target.
Collapse
Affiliation(s)
- Jaewoo Hong
- Department of Physiology, Daegu Catholic University School of Medicine, Daegu 42472, Korea
- Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunjhung Jhun
- Technical Assistance Center, Korea Food Research Institute, Wanju 55365, Korea
| | - Yeo-Ok Choi
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Afeisha S. Taitt
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Suyoung Bae
- Department of Bioequivalence Division for Drug Evaluation, Ministry of Food and Drug Safety, Cheongju 28159, Korea
| | - Youngmin Lee
- Department of Medicine, Pusan Paik Hospital, College of Medicine, Inje University, Busan 47392, Korea
| | - Chang-seon Song
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang 25354, Korea
| | - Soohyun Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Veterinary Science Research Institute, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
37
|
Wang ZJ, Zhang HJ, Lu J, Xu KW, Peng C, Guo J, Gao XX, Wan X, Wang WH, Shan C, Zhang SC, Wu J, Yang AN, Zhu Y, Xiao A, Zhang L, Fu L, Si HR, Cai Q, Yang XL, You L, Zhou YP, Liu J, Pang DQ, Jin WP, Zhang XY, Meng SL, Sun YX, Desselberger U, Wang JZ, Li XG, Duan K, Li CG, Xu M, Shi ZL, Yuan ZM, Yang XM, Shen S. Low toxicity and high immunogenicity of an inactivated vaccine candidate against COVID-19 in different animal models. Emerg Microbes Infect 2021; 9:2606-2618. [PMID: 33241728 PMCID: PMC7733911 DOI: 10.1080/22221751.2020.1852059] [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] [Indexed: 01/21/2023]
Abstract
The ongoing COVID-19 pandemic is causing huge impact on health, life, and global economy, which is characterized by rapid spreading of SARS-CoV-2, high number of confirmed cases and a fatality/case rate worldwide reported by WHO. The most effective intervention measure will be to develop safe and effective vaccines to protect the population from the disease and limit the spread of the virus. An inactivated, whole virus vaccine candidate of SARS-CoV-2 has been developed by Wuhan Institute of Biological Products and Wuhan Institute of Virology. The low toxicity, immunogenicity, and immune persistence were investigated in preclinical studies using seven different species of animals. The results showed that the vaccine candidate was well tolerated and stimulated high levels of specific IgG and neutralizing antibodies. Low or no toxicity in three species of animals was also demonstrated in preclinical study of the vaccine candidate. Biochemical analysis of structural proteins and purity analysis were performed. The inactivated, whole virion vaccine was characterized with safe double-inactivation, no use of DNases and high purity. Dosages, boosting times, adjuvants, and immunization schedules were shown to be important for stimulating a strong humoral immune response in animals tested. Preliminary observation in ongoing phase I and II clinical trials of the vaccine candidate in Wuzhi County, Henan Province, showed that the vaccine is well tolerant. The results were characterized by very low proportion and low degree of side effects, high levels of neutralizing antibodies, and seroconversion. These results consistent with the results obtained from preclinical data on the safety.
Collapse
Affiliation(s)
- Ze-Jun Wang
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Hua-Jun Zhang
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Jia Lu
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Kang-Wei Xu
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Cheng Peng
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Jing Guo
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Xiao-Xiao Gao
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Xin Wan
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Wen-Hui Wang
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Chao Shan
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Su-Cai Zhang
- JOINN Laboratories (Beijing), Beijing, People's Republic of China
| | - Jie Wu
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - An-Na Yang
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Yan Zhu
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Ao Xiao
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Lei Zhang
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Lie Fu
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Hao-Rui Si
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Qian Cai
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Xing-Lou Yang
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Lei You
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Yan-Ping Zhou
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Jing Liu
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - De-Qing Pang
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Wei-Ping Jin
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Xiao-Yu Zhang
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Sheng-Li Meng
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Yun-Xia Sun
- JOINN Laboratories (Beijing), Beijing, People's Republic of China
| | - Ulrich Desselberger
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Jun-Zhi Wang
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Xin-Guo Li
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Kai Duan
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| | - Chang-Gui Li
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Miao Xu
- National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Zheng-Li Shi
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Zhi-Ming Yuan
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Wuhan, People's Republic of China
| | - Xiao-Ming Yang
- National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China.,China National Biotec Group Company Ltd, Beijing, People's Republic of China
| | - Shuo Shen
- Wuhan Institute of Biological Products Co. Ltd., Wuhan, People's Republic of China.,National Engineering Technology Research Center of Combined Vaccines, Wuhan, People's Republic of China
| |
Collapse
|
38
|
Awadasseid A, Wu Y, Tanaka Y, Zhang W. Current advances in the development of SARS-CoV-2 vaccines. Int J Biol Sci 2021; 17:8-19. [PMID: 33390829 PMCID: PMC7757035 DOI: 10.7150/ijbs.52569] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/22/2020] [Indexed: 01/01/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is now a global pandemic that has wreaked havoc globally, which has put a heavy toll on public health, lives, and the world economy. Vaccination is considered as one of the greatest successes in medical history. Based on prior experience with the development of SARS-CoV vaccines, all COVID-19 vaccines must be subjected to the tests for protective effects and harmful risks derived from antibody-dependent enhancement that may contribute to augmented infectivity and/or eosinophilic infiltration. The SARS-CoV-2 vaccine is now being developed urgently in several different ways. China is regarded as one of the world's leading countries in SARS-CoV-2 vaccine development, up to date the last inactivated vaccine international clinical (Phase III) trial was launched in the United Arab Emirates by Sinopharm China National Biotec Group (CNBG). In this review, we outline the current status of vaccine development against clinically relevant SARS-CoV-2 strains, anticipating that such attempts would help create efficacious and sage SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Annoor Awadasseid
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
- Department of Biochemistry & Food Sciences, University of Kordofan, El-Obeid, 51111, Sudan
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| |
Collapse
|
39
|
Soleimanpour S, Yaghoubi A. COVID-19 vaccine: where are we now and where should we go? Expert Rev Vaccines 2021; 20:23-44. [PMID: 33435774 PMCID: PMC7898300 DOI: 10.1080/14760584.2021.1875824] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 01/11/2021] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has currently caused the pandemic with a high progressive speed and has been considered as the global public health crisis in 2020. This new member of the coronavirus family has created a potentially fatal disease, called coronavirus disease-2019 (COVID-19). Despite the continuous efforts of researchers to find effective vaccines and drugs for COVID-19, there is still no success in this matter. AREAS COVERED Here, the literature regarding the COVID-19 vaccine candidates currently in the clinical trials, as well as main candidates in pre-clinical stages for development and research, were reviewed. These candidates have been developed under five different major platforms, including live-attenuated vaccine, mRNA-based vaccine, DNA vaccines, inactivated virus, and viral-vector-based vaccine. EXPERT OPINION There are several limitations in the field of the rapid vaccine development against SARS-CoV-2, and other members of the coronavirus family such as SARS-CoV and MERS-CoV. The key challenges of designing an effective vaccine within a short time include finding the virulence ability of an emerging virus and potential antigen, choosing suitable experimental models and efficient route of administration, the immune-response study, designing the clinical trials, and determining the safety, as well as efficacy.
Collapse
Affiliation(s)
- Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atieh Yaghoubi
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
40
|
Winkler ES, Gilchuk P, Yu J, Bailey AL, Chen RE, Zost SJ, Jang H, Huang Y, Allen JD, Case JB, Sutton RE, Carnahan RH, Darling TL, Boon ACM, Mack M, Head RD, Ross TM, Crowe JE, Diamond MS. Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions and monocytes for optimal therapeutic protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33398272 DOI: 10.1101/2020.12.28.424554] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 has caused the global COVID-19 pandemic. Although passively delivered neutralizing antibodies against SARS-CoV-2 show promise in clinical trials, their mechanism of action in vivo is incompletely understood. Here, we define correlates of protection of neutralizing human monoclonal antibodies (mAbs) in SARS-CoV-2-infected animals. Whereas Fc effector functions are dispensable when representative neutralizing mAbs are administered as prophylaxis, they are required for optimal protection as therapy. When given after infection, intact mAbs reduce SARS-CoV-2 burden and lung disease in mice and hamsters better than loss-of-function Fc variant mAbs. Fc engagement of neutralizing antibodies mitigates inflammation and improves respiratory mechanics, and transcriptional profiling suggests these phenotypes are associated with diminished innate immune signaling and preserved tissue repair. Immune cell depletions establish that neutralizing mAbs require monocytes for therapeutic efficacy. Thus, potently neutralizing mAbs require Fc effector functions for maximal therapeutic benefit during therapy to modulate protective immune responses and mitigate lung disease.
Collapse
|
41
|
Al-Ahmady ZS, Ali-Boucetta H. Nanomedicine & Nanotoxicology Future Could Be Reshaped Post-COVID-19 Pandemic. FRONTIERS IN NANOTECHNOLOGY 2020. [DOI: 10.3389/fnano.2020.610465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Since its first emergence in December 2019, the coronavirus-2 infection has quickly spread around the world and the severity of the pandemic has already re-shaped our lives. This review highlights the role of nanotechnology in the fight against this pandemic with a focus on the design of effective nano-based prevention and treatment options that overcome the limitations associated with conventional vaccines and other therapies. How nanotechnology could be utilized to understand the pathology of the ongoing pandemic is also discussed as well as how our knowledge about SARS-CoV-2 cellular uptake and toxicity could influence future nanotoxicological considerations and nanomedicine design of safe yet effective nanomaterials.
Collapse
|
42
|
Kim DS, Rowland-Jones S, Gea-Mallorquí E. Will SARS-CoV-2 Infection Elicit Long-Lasting Protective or Sterilising Immunity? Implications for Vaccine Strategies (2020). Front Immunol 2020; 11:571481. [PMID: 33362759 PMCID: PMC7756008 DOI: 10.3389/fimmu.2020.571481] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022] Open
Abstract
In December 2019, an outbreak of a novel coronavirus (SARS-CoV-2) in Wuhan, China resulted in the current COVID-19 global pandemic. The human immune system has not previously encountered this virus, raising the important question as to whether or not protective immunity is generated by infection. Growing evidence suggests that protective immunity can indeed be acquired post-infection-although a handful of reinfection cases have been reported. However, it is still unknown whether the immune response to SARS-CoV-2 leads to some degree of long-lasting protection against the disease or the infection. This review draws insights from previous knowledge regarding the nature and longevity of immunity to the related virus, SARS-CoV, to fill the gaps in our understanding of the immune response to SARS-CoV-2. Deciphering the immunological characteristics that give rise to protective immunity against SARS-CoV-2 is critical to guiding vaccine development and also predicting the course of the pandemic. Here we discuss the recent evidence that characterises the adaptive immune response against SARS-CoV-2 and its potential implications for the generation of memory responses and long-term protection.
Collapse
Affiliation(s)
- David S. Kim
- Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Sarah Rowland-Jones
- Viral Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ester Gea-Mallorquí
- Viral Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
43
|
Cimolai N. Applying Immune Instincts and Maternal Intelligence from Comparative Microbiology to COVID-19. ACTA ACUST UNITED AC 2020; 2:2670-2683. [PMID: 33195997 PMCID: PMC7652409 DOI: 10.1007/s42399-020-00634-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2020] [Indexed: 01/02/2023]
Abstract
New data specific to COVID-19 are emerging quickly on key issues of immunity and prevention, but past research in coronavirology and for other human pathogens (e.g., Mycoplasma pneumoniae) has been available and of great relevance. Considerable study of endemic human coronaviruses has shown that neutralizing antibody correlates with protection, but effective clinical protection is variable for subsequent virus exposure. Animal coronavirus research has emphasized the importance of local mucosal protection (especially IgA) and systemic responses. Animal model and human post-infection studies for SARS-CoV and MERS-CoV are largely corroborative. Whether for passive therapeutic strategies or vaccination, these findings provide a template for COVID-19. Many approaches to vaccination have emerged, and there may be more than one vaccine that will be applied, but individualized obstacles and concerns for administration, efficacy, and safety are inevitable. Regardless of safeguards or promises that may be understood from laboratory or vertebrate experiments, observations from large-scale human trials will ultimately prove to shape the medical future. Focus on common mucosal immunity can be underrated, and equally or more, focus on lactogenic immunity may be underestimated. In understanding both passive immunity and protection, the body is already primed to educate us with decisions of what constitutes protection and harm. This review provides key insights that drive hypotheses into how the instinct of immunity and the intelligence of the maternal component of the common mucosal immune system has already guided us and may continue to do so effectively into a bright and safe future.
Collapse
Affiliation(s)
- Nevio Cimolai
- Faculty of Medicine, The University of British Columbia, Vancouver, BC Canada
- Children’s and Women’s Health Centre of British Columbia, 4480 Oak Street, Vancouver, BC V6H3V4 Canada
| |
Collapse
|
44
|
Lambkin‐Williams R, DeVincenzo JP. A COVID-19 human viral challenge model. Learning from experience. Influenza Other Respir Viruses 2020; 14:747-756. [PMID: 32790065 PMCID: PMC7578316 DOI: 10.1111/irv.12797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 01/15/2023] Open
Abstract
The controlled human infection model and specifically the human viral challenge model are not dissimilar to standard clinical trials while adding another layer of complexity and safety considerations. The models deliberately infect volunteers, with an infectious challenge agent to determine the effect of the infection and the potential benefits of the experimental interventions. The human viral challenge model studies can shorten the time to assess the efficacy of a new vaccine or treatment by combining this with the assessment of safety. The newly emerging SARS-CoV-2 virus is highly contagious, and an urgent race is on to develop a new vaccine against this virus in a timeframe never attempted before. The use of the human viral challenge model has been proposed to accelerate the development of the vaccine. In the early 2000s, the authors successfully developed a pathogenic human viral challenge model for another virus for which there was no effective treatment and established it to evaluate potential therapies and vaccines against respiratory syncytial virus. Experience gained in the development of that model can help with the development of a COVID-19 HVCM and the authors describe it here.
Collapse
|
45
|
Haun BK, Lai CY, Williams CA, Wong TAS, Lieberman MM, Pessaint L, Andersen H, Lehrer AT. CoVaccine HT™ Adjuvant Potentiates Robust Immune Responses to Recombinant SARS-CoV-2 Spike S1 Immunization. Front Immunol 2020; 11:599587. [PMID: 33193454 PMCID: PMC7661386 DOI: 10.3389/fimmu.2020.599587] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022] Open
Abstract
The current COVID-19 pandemic has claimed hundreds of thousands of lives and its causative agent, SARS-CoV-2, has infected millions, globally. The highly contagious nature of this respiratory virus has spurred massive global efforts to develop vaccines at record speeds. In addition to enhanced immunogen delivery, adjuvants may greatly impact protective efficacy of a SARS-CoV-2 vaccine. To investigate adjuvant suitability, we formulated protein subunit vaccines consisting of the recombinant S1 domain of SARS-CoV-2 Spike protein alone or in combination with either CoVaccine HT™ or Alhydrogel. CoVaccine HT™ induced high titres of antigen-binding IgG after a single dose, facilitated affinity maturation and class switching to a greater extent than Alhydrogel and elicited potent cell-mediated immunity as well as virus neutralizing antibody titres. Data presented here suggests that adjuvantation with CoVaccine HT™ can rapidly induce a comprehensive and protective immune response to SARS-CoV-2.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Aluminum Hydroxide/administration & dosage
- Aluminum Hydroxide/immunology
- Animals
- Antibodies, Viral/immunology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- Female
- Humans
- Immunity, Cellular
- Immunity, Humoral
- Immunization
- Immunoglobulin G/immunology
- Male
- Mice
- Mice, Inbred BALB C
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/administration & dosage
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Brien K. Haun
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
- Cell and Molecular Biology Graduate Program, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Chih-Yun Lai
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Caitlin A. Williams
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Teri Ann S. Wong
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Michael M. Lieberman
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | | | | | - Axel T. Lehrer
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
- Cell and Molecular Biology Graduate Program, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| |
Collapse
|
46
|
Wang M, Fu T, Hao J, Li L, Tian M, Jin N, Ren L, Li C. A recombinant Lactobacillus plantarum strain expressing the spike protein of SARS-CoV-2. Int J Biol Macromol 2020; 160:736-740. [PMID: 32485251 PMCID: PMC7260514 DOI: 10.1016/j.ijbiomac.2020.05.239] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022]
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global pandemic in the past four months and causes respiratory disease in humans of almost all ages. Although several drugs have been announced to be partially effective treatments for this disease, no approved vaccine is available. Here, we described the construction of a recombinant Lactobacillus plantarum strain expressing the SARS-CoV-2 spike protein. The results showed that the spike gene with optimized codons could be efficiently expressed on the surface of recombinant L. plantarum and exhibited high antigenicity. The highest protein yield was obtained under the following conditions: cells were induced with 50 ng/mL SppIP at 37 °C for 6-10 h. The recombinant spike (S) protein was stable under normal conditions and at 50 °C, pH = 1.5, or a high salt concentration. Recombinant L. plantarum may provide a promising food-grade oral vaccine candidate against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Maopeng Wang
- Institute of Virology, Wenzhou University, Chashan University Town, Wenzhou, 325035, China; Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun 130122, China
| | - Tingting Fu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun 130122, China
| | - Jiayi Hao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun 130122, China
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun 130122, China
| | - Mingyao Tian
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun 130122, China
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun 130122, China
| | - Linzhu Ren
- Key Lab for Zoonoses Research, Ministry of Education, Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun 130122, China.
| |
Collapse
|
47
|
Osuchowski MF, Aletti F, Cavaillon JM, Flohé SB, Giamarellos-Bourboulis EJ, Huber-Lang M, Relja B, Skirecki T, Szabó A, Maegele M. SARS-CoV-2/COVID-19: Evolving Reality, Global Response, Knowledge Gaps, and Opportunities. Shock 2020; 54:416-437. [PMID: 32433217 PMCID: PMC7363382 DOI: 10.1097/shk.0000000000001565] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Approximately 3 billion people around the world have gone into some form of social separation to mitigate the current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. The uncontrolled influx of patients in need of emergency care has rapidly brought several national health systems to near-collapse with deadly consequences to those afflicted by Coronavirus Disease 2019 (COVID-19) and other critical diseases associated with COVID-19. Solid scientific evidence regarding SARS-CoV-2/COVID-19 remains scarce; there is an urgent need to expand our understanding of the SARS-CoV-2 pathophysiology to facilitate precise and targeted treatments. The capacity for rapid information dissemination has emerged as a double-edged sword; the existing gap of high-quality data is frequently filled by anecdotal reports, contradictory statements, and misinformation. This review addresses several important aspects unique to the SARS-CoV-2/COVID-19 pandemic highlighting the most relevant knowledge gaps and existing windows-of-opportunity. Specifically, focus is given on SARS-CoV-2 immunopathogenesis in the context of experimental therapies and preclinical evidence and their applicability in supporting efficacious clinical trial planning. The review discusses the existing challenges of SARS-CoV-2 diagnostics and the potential application of translational technology for epidemiological predictions, patient monitoring, and treatment decision-making in COVID-19. Furthermore, solutions for enhancing international strategies in translational research, cooperative networks, and regulatory partnerships are contemplated.
Collapse
Affiliation(s)
- Marcin F. Osuchowski
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Center, Vienna, Austria
| | - Federico Aletti
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | | | - Stefanie B. Flohé
- Department of Trauma, Hand, and Reconstructive Surgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Tomasz Skirecki
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Marc Maegele
- Department of Trauma and Orthopaedic Surgery, Cologne-Merheim Medical Center (CMMC), University of Witten/Herdecke, Cologne-Merheim Campus, Cologne, Germany
- Institute for Research in Operative Medicine (IFOM), University of Witten/Herdecke, Cologne-Merheim Campus, Cologne, Germany
| |
Collapse
|
48
|
Lee WS, Wheatley AK, Kent SJ, DeKosky BJ. Antibody-dependent enhancement and SARS-CoV-2 vaccines and therapies. Nat Microbiol 2020; 5:1185-1191. [PMID: 32908214 DOI: 10.1038/s41564-020-00789-5] [Citation(s) in RCA: 458] [Impact Index Per Article: 114.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Antibody-based drugs and vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are being expedited through preclinical and clinical development. Data from the study of SARS-CoV and other respiratory viruses suggest that anti-SARS-CoV-2 antibodies could exacerbate COVID-19 through antibody-dependent enhancement (ADE). Previous respiratory syncytial virus and dengue virus vaccine studies revealed human clinical safety risks related to ADE, resulting in failed vaccine trials. Here, we describe key ADE mechanisms and discuss mitigation strategies for SARS-CoV-2 vaccines and therapies in development. We also outline recently published data to evaluate the risks and opportunities for antibody-based protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Wen Shi Lee
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
- ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, Victoria, Australia.
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Brandon J DeKosky
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA.
- Department of Chemical Engineering, The University of Kansas, Lawrence, KS, USA.
- Bioengineering Graduate Program, The University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
49
|
Wen J, Cheng Y, Ling R, Dai Y, Huang B, Huang W, Zhang S, Jiang Y. Antibody-dependent enhancement of coronavirus. Int J Infect Dis 2020; 100:483-489. [PMID: 32920233 PMCID: PMC7483033 DOI: 10.1016/j.ijid.2020.09.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/28/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
Antibody-dependent enhancement (ADE) exists in several kinds of virus. It has a negative influence on antibody therapy for viral infection. This effect was first identified in dengue virus and has since also been described for coronavirus. To date, the rapid spread of the newly emerged coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing coronavirus disease 2019 (COVID-19), has affected over 3.8 million people across the globe. The novel coronavirus poses a great challenge and has caused a wave of panic. In this review, antibody-dependent enhancements in dengue virus and two kinds of coronavirus are summarized. Possible solutions for the effects are reported. We also speculate that ADE may exist in SARS-CoV-2.
Collapse
Affiliation(s)
- Jieqi Wen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Yifan Cheng
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Rongsong Ling
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Yarong Dai
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Boxuan Huang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Wenjie Huang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Siyan Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Yizhou Jiang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| |
Collapse
|
50
|
Gupta T, Gupta SK. Potential adjuvants for the development of a SARS-CoV-2 vaccine based on experimental results from similar coronaviruses. Int Immunopharmacol 2020; 86:106717. [PMID: 32585611 PMCID: PMC7301105 DOI: 10.1016/j.intimp.2020.106717] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
The extensive efforts around the globe are being made to develop a suitable vaccine against COVID-19 (Coronavirus Disease-19) caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus-2). An effective vaccine should be able to induce high titers of neutralizing antibodies to prevent the virus from attaching to the host cell receptors. However, to elicit the protective levels of antibodies, a vaccine may require multiple doses or assistance from other immunostimulatory molecules. Further, the vaccine should be able to induce protective levels of antibodies rapidly with the least amount of antigen used. This decreases the cost of a vaccine and makes it affordable. As the pandemic has hit most countries across the globe, there will be an overwhelming demand for the vaccine in a quick time. Incorporating a suitable adjuvant in a SARS-CoV-2 vaccine may address these requirements. This review paper will discuss the experimental results of the adjuvanted vaccine studies with similar coronaviruses (CoVs) which might be useful to select an appropriate adjuvant for a vaccine against rapidly emergingSARS-CoV-2. We also discuss the current progress in the development of adjuvanted vaccines against the disease.
Collapse
Affiliation(s)
- Tania Gupta
- Dr GC Negi College of Veterinary and Animal Sciences, Palampur 176062, Himachal Pradesh, India.
| | - Shishir K Gupta
- CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| |
Collapse
|