201
|
Sheervalilou R, Shirvaliloo M, Sargazi S, Bahari S, Saravani R, Shahraki J, Shirvalilou S, Shahraki O, Nazarlou Z, Shams Z, Ghaznavi H. Convalescent Blood: Current Perspective on the Efficacy of a Legacy Approach in COVID-19 Treatment. Blood Purif 2021; 51:1-14. [PMID: 33789273 PMCID: PMC8089443 DOI: 10.1159/000513164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
Since early 2020, COVID-19 has wreaked havoc in many societies around the world. As of the present, the SARS-CoV-2-borne disease is propagating in almost all countries, affecting hundreds of thousands of people in an unprecedented way. As the name suggests, the novel coronavirus, widely known as SARS-CoV-2, is a new emerging human pathogen. A novel disease of relatively unknown origin, COVID-19 does not seem to be amenable to the currently available medicines since there is no specific cure for the disease. In the absence of any vaccine or effective antiviral medication, we have no tools at our disposal, but the method of quarantine, be it domestic or institutional, to hinder any further progression of this outbreak. However, there is a record of physicians in the past who practiced convalescent blood transfusion. To their awe, the method seemed to be useful. It is anticipated that these contemporary methods will outdo any other vaccination process in the time being, as blood transfusion is instead a cost-effective and time-friendly technique. Following a successful trial, this new approach of contemporary nature to a viral disease may serve as an emergency intervention to intercept infectious outbreaks and prevent an impending epidemic/pandemic. In this review, we document the most recent evidence regarding the efficiency of convalescent plasma and serum therapy on SARS, MERS, and particularly COVID-19, while discussing potential advantages and possible risks of such practice.
Collapse
Affiliation(s)
- Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Milad Shirvaliloo
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Soraiya Bahari
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramin Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Jafar Shahraki
- Department of Toxicology and Pharmacology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Sakine Shirvalilou
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Omolbanin Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ziba Nazarlou
- Material Engineering Department, College of Science Koç University, Istanbul, Turkey
| | - Zinat Shams
- Department of Biological Science, Kharazmi University, Tehran, Iran
| | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
202
|
Mazzocco G, Niemiec I, Myronov A, Skoczylas P, Kaczmarczyk J, Sanecka-Duin A, Gruba K, Król P, Drwal M, Szczepanik M, Pyrc K, Stȩpniak P. AI Aided Design of Epitope-Based Vaccine for the Induction of Cellular Immune Responses Against SARS-CoV-2. Front Genet 2021; 12:602196. [PMID: 33841493 PMCID: PMC8027494 DOI: 10.3389/fgene.2021.602196] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/28/2021] [Indexed: 12/17/2022] Open
Abstract
The heavy burden imposed by the COVID-19 pandemic on our society triggered the race toward the development of therapies or preventive strategies. Among these, antibodies and vaccines are particularly attractive because of their high specificity, low probability of drug-drug interaction, and potentially long-standing protective effects. While the threat at hand justifies the pace of research, the implementation of therapeutic strategies cannot be exempted from safety considerations. There are several potential adverse events reported after the vaccination or antibody therapy, but two are of utmost importance: antibody-dependent enhancement (ADE) and cytokine storm syndrome (CSS). On the other hand, the depletion or exhaustion of T-cells has been reported to be associated with worse prognosis in COVID-19 patients. This observation suggests a potential role of vaccines eliciting cellular immunity, which might simultaneously limit the risk of ADE and CSS. Such risk was proposed to be associated with FcR-induced activation of proinflammatory macrophages (M1) by Fu et al. (2020) and Iwasaki and Yang (2020). All aspects of the newly developed vaccine (including the route of administration, delivery system, and adjuvant selection) may affect its effectiveness and safety. In this work we use a novel in silico approach (based on AI and bioinformatics methods) developed to support the design of epitope-based vaccines. We evaluated the capabilities of our method for predicting the immunogenicity of epitopes. Next, the results of our approach were compared with other vaccine-design strategies reported in the literature. The risk of immuno-toxicity was also assessed. The analysis of epitope conservation among other Coronaviridae was carried out in order to facilitate the selection of peptides shared across different SARS-CoV-2 strains and which might be conserved in emerging zootic coronavirus strains. Finally, the potential applicability of the selected epitopes for the development of a vaccine eliciting cellular immunity for COVID-19 was discussed, highlighting the benefits and challenges of such an approach.
Collapse
Affiliation(s)
| | | | - Alexander Myronov
- Ardigen, Krakow, Poland
- Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | | | | | | | - Katarzyna Gruba
- Ardigen, Krakow, Poland
- Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | | | | | - Marian Szczepanik
- Department of Medical Biology, Faculty of Health Sciences, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | | |
Collapse
|
203
|
Influence of obesity on serum levels of SARS-CoV-2-specific antibodies in COVID-19 patients. PLoS One 2021; 16:e0245424. [PMID: 33760825 PMCID: PMC7990309 DOI: 10.1371/journal.pone.0245424] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
SARS-CoV-2 (Severe Acute Respiratory Syndrome Corona Virus-2), cause of COVID-19 (Coronavirus Disease of 2019), represents a significant risk to people living with pre-existing conditions associated with exacerbated inflammatory responses and consequent dysfunctional immunity. In this paper, we have evaluated the influence of obesity, a condition associated with chronic systemic inflammation, on the secretion of SARS-CoV-2-specific IgG antibodies in the blood of COVID-19 patients. Our hypothesis is that obesity is associated with reduced amounts of specific IgG antibodies. Results have confirmed our hypothesis and have shown that SARS-CoV-2 IgG antibodies are negatively associated with Body Mass Index (BMI) in COVID-19 obese patients, as expected based on the known influence of obesity on humoral immunity. Antibodies in COVID-19 obese patients are also negatively associated with serum levels of pro-inflammatory and metabolic markers of inflammaging and pulmonary inflammation, such as SAA (serum amyloid A protein), CRP (C-reactive protein), and ferritin, but positively associated with NEFA (nonesterified fatty acids). These results altogether could help to identify an inflammatory signature with strong predictive value for immune dysfunction. Inflammatory markers identified may subsequently be targeted to improve humoral immunity in individuals with obesity and in individuals with other chronic inflammatory conditions.
Collapse
|
204
|
Jafari Porzani S, Konur O, Nowruzi B. Cyanobacterial natural products as sources for antiviral drug discovery against COVID-19. J Biomol Struct Dyn 2021; 40:7629-7644. [PMID: 33749496 DOI: 10.1080/07391102.2021.1899050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The recent Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), positive-sense RNA viruses, originated from Wuhan City in December 2019 and propagated widely globally. Hence, the disease caused by this virus has been declared as a global pandemic by the WHO. As of 18th February 2021, at least seven different vaccines across three platforms have been rolled out in countries and more than 200 additional vaccine candidates have been in development, of which more than 60 are at the stage of the clinical development. So far, Most of the approved vaccine manufacturers are Pfizer, AstraZeneca, and Serum Institute of India, which have been finalized by WHO. Synthetic drug-associated complications have evoked scientific attention for natural product-based drugs. There has been a surge in the antiviral compounds from natural resources along with some therapies. Cyanobacteria are the fruitful reservoir of many metabolites like sulfated polysaccharides and lectins that possess strong antiviral activities and immunity boosting effects. However, the research in this field has been relatively under-developed. The current research highlights important features of cyanobacterial antiviral biomaterials, benefits and drawbacks of cyanobacterial drugs, challenges, future perspectives as well as overview of drugs against COVID-19. In addition, we have described mutated variants and transmission rate of coronaviruses. The current research suggests that cyanobacterial species and their extracts have promising applications as potentially antiviral drug biomaterials against COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Samaneh Jafari Porzani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ozcan Konur
- Formerly, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Bahareh Nowruzi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
205
|
Rahman MM, Zhou N, Huang J. An Overview on the Development of mRNA-Based Vaccines and Their Formulation Strategies for Improved Antigen Expression In Vivo. Vaccines (Basel) 2021; 9:244. [PMID: 33799516 PMCID: PMC8001631 DOI: 10.3390/vaccines9030244] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022] Open
Abstract
The mRNA-based vaccine approach is a promising alternative to traditional vaccines due to its ability for prompt development, high potency, and potential for secure administration and low-cost production. Nonetheless, the application has still been limited by the instability as well as the ineffective delivery of mRNA in vivo. Current technological improvements have now mostly overcome these concerns, and manifold mRNA vaccine plans against various forms of malignancies and infectious ailments have reported inspiring outcomes in both humans and animal models. This article summarizes recent mRNA-based vaccine developments, advances of in vivo mRNA deliveries, reflects challenges and safety concerns, and future perspectives, in developing the mRNA vaccine platform for extensive therapeutic use.
Collapse
Affiliation(s)
- Md. Motiar Rahman
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (N.Z.); (J.H.)
| | - Nan Zhou
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (N.Z.); (J.H.)
| | - Jiandong Huang
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (N.Z.); (J.H.)
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
206
|
Bertoglio F, Meier D, Langreder N, Steinke S, Rand U, Simonelli L, Heine PA, Ballmann R, Schneider KT, Roth KDR, Ruschig M, Riese P, Eschke K, Kim Y, Schäckermann D, Pedotti M, Kuhn P, Zock-Emmenthal S, Wöhrle J, Kilb N, Herz T, Becker M, Grasshoff M, Wenzel EV, Russo G, Kröger A, Brunotte L, Ludwig S, Fühner V, Krämer SD, Dübel S, Varani L, Roth G, Čičin-Šain L, Schubert M, Hust M. SARS-CoV-2 neutralizing human recombinant antibodies selected from pre-pandemic healthy donors binding at RBD-ACE2 interface. Nat Commun 2021; 12:1577. [PMID: 33707427 PMCID: PMC7952403 DOI: 10.1038/s41467-021-21609-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/28/2021] [Indexed: 12/20/2022] Open
Abstract
COVID-19 is a severe acute respiratory disease caused by SARS-CoV-2, a new recently emerged sarbecovirus. This virus uses the human ACE2 enzyme as receptor for cell entry, recognizing it with the receptor binding domain (RBD) of the S1 subunit of the viral spike protein. We present the use of phage display to select anti-SARS-CoV-2 spike antibodies from the human naïve antibody gene libraries HAL9/10 and subsequent identification of 309 unique fully human antibodies against S1. 17 antibodies are binding to the RBD, showing inhibition of spike binding to cells expressing ACE2 as scFv-Fc and neutralize active SARS-CoV-2 virus infection of VeroE6 cells. The antibody STE73-2E9 is showing neutralization of active SARS-CoV-2 as IgG and is binding to the ACE2-RBD interface. Thus, universal libraries from healthy human donors offer the advantage that antibodies can be generated quickly and independent from the availability of material from recovering patients in a pandemic situation.
Collapse
Affiliation(s)
- Federico Bertoglio
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Doris Meier
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Nora Langreder
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Stephan Steinke
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Ulfert Rand
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Luca Simonelli
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Philip Alexander Heine
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Rico Ballmann
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Kai-Thomas Schneider
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Kristian Daniel Ralph Roth
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Maximilian Ruschig
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Peggy Riese
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kathrin Eschke
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Yeonsu Kim
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dorina Schäckermann
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Mattia Pedotti
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | | | | | | | | | | | - Marlies Becker
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Martina Grasshoff
- Helmholtz Centre for Infection Research, Research Group Innate Immunity and Infection, Braunschweig, Germany
| | - Esther Veronika Wenzel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Giulio Russo
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Andrea Kröger
- Helmholtz Centre for Infection Research, Research Group Innate Immunity and Infection, Braunschweig, Germany
| | - Linda Brunotte
- Westfälische Wilhelms-Universität Münster, Institut für Virologie (IVM), Münster, Germany
| | - Stephan Ludwig
- Westfälische Wilhelms-Universität Münster, Institut für Virologie (IVM), Münster, Germany
| | - Viola Fühner
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | | | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Luca Varani
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland.
| | | | - Luka Čičin-Šain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
- Centre for Individualised Infection Medicine (CIIM), a joint venture of Helmholtz Centre for Infection Research and Medical School Hannover, Braunschweig, Germany.
| | - Maren Schubert
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany.
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany.
| |
Collapse
|
207
|
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: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [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
|
208
|
A comparative review of pathogenesis and host innate immunity evasion strategies among the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), severe acute respiratory syndrome coronavirus (SARS-CoV) and the Middle East respiratory syndrome coronavirus (MERS-CoV). Arch Microbiol 2021; 203:1943-1951. [PMID: 33682075 PMCID: PMC7937358 DOI: 10.1007/s00203-021-02265-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 02/12/2021] [Accepted: 02/24/2021] [Indexed: 12/09/2022]
Abstract
COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has put the global public health at its highest threat around the world. Previous epidemic caused by the acute respiratory syndrome coronavirus (SARS-CoV) in 2002 is also considered since both the coronaviruses resulted in the similar clinical complications. The outbreak caused by the Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012 had a low rate of disease transmission and death cases. Modes of entry by MERS and SARS coronaviruses are similar to that of SARS-CoV-2, except MERS-CoV utilize different receptor. They all belong to the lineage C of β-coronavirus. Based on the information from the previous reports, the present review is mainly focused on the mechanisms of disease progression by each of these viruses in association to their strategies to escape the host immunity. The viral entry is the first step of pathogenesis associated with attachment of viral spike protein with host receptor help releasing the viral RNA into the host cell. Models of molecular pathogenesis are outlined with virus strategies escaping the host immunity along with the role of various inflammatory cytokines and chemokines in the process. The molecular aspects of pathogenesis have also been discussed.
Collapse
|
209
|
Gai J, Ma L, Li G, Zhu M, Qiao P, Li X, Zhang H, Zhang Y, Chen Y, Ji W, Zhang H, Cao H, Li X, Gong R, Wan Y. A potent neutralizing nanobody against SARS-CoV-2 with inhaled delivery potential. MedComm (Beijing) 2021; 2:101-113. [PMID: 33821254 PMCID: PMC8013425 DOI: 10.1002/mco2.60] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/28/2021] [Accepted: 01/31/2021] [Indexed: 12/17/2022] Open
Abstract
The coronavirus disease 2019 (COVID‐19) pandemic has become a serious burden on global public health. Although therapeutic drugs against COVID‐19 have been used in many countries, their efficacy is still limited. We here reported nanobody (Nb) phage display libraries derived from four camels immunized with the SARS‐CoV‐2 spike receptor‐binding domain (RBD), from which 381 Nbs were identified to recognize SARS‐CoV‐2‐RBD. Furthermore, seven Nbs were shown to block interaction of human angiotensin‐converting enzyme 2 (ACE2) with SARS‐CoV‐2‐RBD variants and two Nbs blocked the interaction of human ACE2 with bat‐SL‐CoV‐WIV1‐RBD and SARS‐CoV‐1‐RBD. Among these candidates, Nb11‐59 exhibited the highest activity against authentic SARS‐CoV‐2 with 50% neutralizing dose (ND50) of 0.55 μg/ml. Nb11‐59 can be produced on large scale in Pichia pastoris, with 20 g/L titer and 99.36% purity. It also showed good stability profile, and nebulization did not impact its stability. Overall, Nb11‐59 might be a promising prophylactic and therapeutic molecule against COVID‐19, especially through inhalation delivery.
Collapse
Affiliation(s)
- Junwei Gai
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Linlin Ma
- Shanghai Key Laboratory of Molecular Imaging Shanghai University of Medicine and Health Sciences Shanghai China
| | - Guanghui Li
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Min Zhu
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Peng Qiao
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Xiaofei Li
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Haiwei Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science Chinese Academy of Sciences Wuhan China
| | - Yanmin Zhang
- Laboratory of Molecular Design and Drug Discovery, School of Science China Pharmaceutical University Nanjing China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science China Pharmaceutical University Nanjing China
| | - Weiwei Ji
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Hao Zhang
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Huanhuan Cao
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Xionghui Li
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science Chinese Academy of Sciences Wuhan China
| | - Yakun Wan
- Shanghai Novamab Biopharmaceuticals Co., Ltd Shanghai China
| |
Collapse
|
210
|
Su S, Du L, Jiang S. Learning from the past: development of safe and effective COVID-19 vaccines. Nat Rev Microbiol 2021; 19:211-219. [PMID: 33067570 PMCID: PMC7566580 DOI: 10.1038/s41579-020-00462-y] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2020] [Indexed: 01/29/2023]
Abstract
The rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has elicited an equally rapid response aiming to develop a COVID-19 vaccine. These efforts are encouraging; however, comprehensive efficacy and safety evaluations are essential in the development of a vaccine, and we can learn from previous vaccine development campaigns. In this Perspective, we summarize examples of vaccine-associated disease enhancement in the history of developing vaccines against respiratory syncytial virus, dengue virus, SARS-CoV and Middle East respiratory syndrome coronavirus, which highlight the importance of a robust safety and efficacy profile, and present recommendations for preclinical and clinical evaluation of COVID-19 vaccine candidates as well as for vaccine design and optimization.
Collapse
Affiliation(s)
- Shan Su
- Key Laboratory of Medical Molecular Virology (MOE/MOH/CAM), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/MOH/CAM), School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA.
| |
Collapse
|
211
|
Riedel RN, Pérez-Pérez A, Sánchez-Margalet V, Varone CL, Maymó JL. Stem cells and COVID-19: are the human amniotic cells a new hope for therapies against the SARS-CoV-2 virus? Stem Cell Res Ther 2021; 12:155. [PMID: 33648582 PMCID: PMC7919997 DOI: 10.1186/s13287-021-02216-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
A new coronavirus respiratory disease (COVID-19) caused by the SARS-CoV-2 virus, surprised the entire world, producing social, economic, and health problems. The COVID-19 triggers a lung infection with a multiple proinflammatory cytokine storm in severe patients. Without effective and safe treatments, COVID-19 has killed thousands of people, becoming a pandemic. Stem cells have been suggested as a therapy for lung-related diseases. In particular, mesenchymal stem cells (MSCs) have been successfully tested in some clinical trials in patients with COVID-19. The encouraging results positioned MSCs as a possible cell therapy for COVID-19. The amniotic membrane from the human placenta at term is a valuable stem cell source, including human amniotic epithelial cells (hAECs) and human mesenchymal stromal cells (hAMSCs). Interestingly, amnion cells have immunoregulatory, regenerative, and anti-inflammatory properties. Moreover, hAECs and hAMSCs have been used both in preclinical studies and in clinical trials against respiratory diseases. They have reduced the inflammatory response and restored the pulmonary tissue architecture in lung injury in vivo models. Here, we review the existing data about the stem cells use for COVID-19 treatment, including the ongoing clinical trials. We also consider the non-cellular therapies that are being applied. Finally, we discuss the human amniotic membrane cells use in patients who suffer from immune/inflammatory lung diseases and hypothesize their possible use as a successful treatment against COVID-19.
Collapse
Affiliation(s)
- Rodrigo N Riedel
- Instituto de Química Biológica (IQUIBICEN), CONICET- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria Pabellón 2, 4° piso, 1428, Buenos Aires, Argentina
| | - Antonio Pérez-Pérez
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Avenida Sánchez Pizjuán 4, 41009, Sevilla, España
| | - Víctor Sánchez-Margalet
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Avenida Sánchez Pizjuán 4, 41009, Sevilla, España
| | - Cecilia L Varone
- Instituto de Química Biológica (IQUIBICEN), CONICET- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria Pabellón 2, 4° piso, 1428, Buenos Aires, Argentina
| | - Julieta L Maymó
- Instituto de Química Biológica (IQUIBICEN), CONICET- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria Pabellón 2, 4° piso, 1428, Buenos Aires, Argentina.
| |
Collapse
|
212
|
Harapan H, Ryan M, Yohan B, Abidin RS, Nainu F, Rakib A, Jahan I, Emran TB, Ullah I, Panta K, Dhama K, Sasmono RT. Covid-19 and dengue: Double punches for dengue-endemic countries in Asia. Rev Med Virol 2021; 31:e2161. [PMID: 32946149 PMCID: PMC7536968 DOI: 10.1002/rmv.2161] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022]
Abstract
The coronavirus disease 2019 (Covid-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is an international public health crisis with devastating effects. In particular, this pandemic has further exacerbated the burden in tropical and subtropical regions of the world, where dengue fever, caused by dengue virus (DENV), is already endemic to the population. The similar clinical manifestations shared by Covid-19 and dengue fever have raised concerns, especially in dengue-endemic countries with limited resources, leading to diagnostic challenges. In addition, cross-reactivity of the immune responses in these infections is an emerging concern, as pre-existing DENV-antibodies might potentially affect Covid-19 through antibody-dependent enhancement. In this review article, we aimed to raise the issue of Covid-19 and dengue fever misdiagnosis, not only in a clinical setting but also with regards to cross-reactivity between SARS-CoV-2 and DENV antibodies. We also have discussed the potential consequences of overlapping immunological cascades between dengue and Covid-19 on disease severity and vaccine development.
Collapse
Affiliation(s)
- Harapan Harapan
- Medical Research Unit, School of MedicineUniversitas Syiah KualaBanda AcehIndonesia
- Tropical Disease Centre, School of MedicineUniversitas Syiah KualaBanda AcehIndonesia
- Department of Microbiology, School of MedicineUniversitas Syiah KualaBanda AcehIndonesia
| | - Mirza Ryan
- Medical Research Unit, School of MedicineUniversitas Syiah KualaBanda AcehIndonesia
| | | | | | - Firzan Nainu
- Faculty of PharmacyHasanuddin UniversityMakassarIndonesia
| | - Ahmed Rakib
- Department of Pharmacy, Faculty of Biological SciencesUniversity of ChittagongChittagongBangladesh
| | - Israt Jahan
- Department of PharmacyInternational Islamic University ChittagongChittagongBangladesh
| | - Talha Bin Emran
- Department of PharmacyBGC Trust University BangladeshChittagongBangladesh
| | - Irfan Ullah
- Kabir Medical CollegeGandhara UniversityPeshawarPakistan
| | - Kritu Panta
- School of Biomedical SciencesThe University of Western AustraliaCrawleyAustralia
| | - Kuldeep Dhama
- Division of PathologyICAR‐Indian Veterinary Research InstituteBareillyIndia
| | | |
Collapse
|
213
|
Bhardwaj A, Sapra L, Saini C, Azam Z, Mishra PK, Verma B, Mishra GC, Srivastava RK. COVID-19: Immunology, Immunopathogenesis and Potential Therapies. Int Rev Immunol 2021; 41:171-206. [PMID: 33641587 PMCID: PMC7919479 DOI: 10.1080/08830185.2021.1883600] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/09/2020] [Accepted: 01/21/2021] [Indexed: 02/08/2023]
Abstract
The Coronavirus Disease-2019 (COVID-19) imposed public health emergency and affected millions of people around the globe. As of January 2021, 100 million confirmed cases of COVID-19 along with more than 2 million deaths were reported worldwide. SARS-CoV-2 infection causes excessive production of pro-inflammatory cytokines thereby leading to the development of "Cytokine Storm Syndrome." This condition results in uncontrollable inflammation that further imposes multiple-organ-failure eventually leading to death. SARS-CoV-2 induces unrestrained innate immune response and impairs adaptive immune responses thereby causing tissue damage. Thus, understanding the foremost features and evolution of innate and adaptive immunity to SARS-CoV-2 is crucial in anticipating COVID-19 outcomes and in developing effective strategies to control the viral spread. In the present review, we exhaustively discuss the sequential key immunological events that occur during SARS-CoV-2 infection and are involved in the immunopathogenesis of COVID-19. In addition to this, we also highlight various therapeutic options already in use such as immunosuppressive drugs, plasma therapy and intravenous immunoglobulins along with various novel potent therapeutic options that should be considered in managing COVID-19 infection such as traditional medicines and probiotics.
Collapse
Affiliation(s)
- Asha Bhardwaj
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Leena Sapra
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Chaman Saini
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Zaffar Azam
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Pradyumna K. Mishra
- Department of Molecular Biology, ICMR-NIREH, Nehru Hospital Building, Gandhi Medical College Campus, Bhopal, India
| | - Bhupendra Verma
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Gyan C. Mishra
- Lab # 1, National Centre for Cell Science (NCCS), Savitribai Phule Pune University Campus, Pune, India
| | - Rupesh K. Srivastava
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
214
|
Ricke DO. Two Different Antibody-Dependent Enhancement (ADE) Risks for SARS-CoV-2 Antibodies. Front Immunol 2021; 12:640093. [PMID: 33717193 PMCID: PMC7943455 DOI: 10.3389/fimmu.2021.640093] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/03/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 (SARS-CoV-2) disease severity and stages varies from asymptomatic, mild flu-like symptoms, moderate, severe, critical, and chronic disease. COVID-19 disease progression include lymphopenia, elevated proinflammatory cytokines and chemokines, accumulation of macrophages and neutrophils in lungs, immune dysregulation, cytokine storms, acute respiratory distress syndrome (ARDS), etc. Development of vaccines to severe acute respiratory syndrome (SARS), Middle East Respiratory Syndrome coronavirus (MERS-CoV), and other coronavirus has been difficult to create due to vaccine induced enhanced disease responses in animal models. Multiple betacoronaviruses including SARS-CoV-2 and SARS-CoV-1 expand cellular tropism by infecting some phagocytic cells (immature macrophages and dendritic cells) via antibody bound Fc receptor uptake of virus. Antibody-dependent enhancement (ADE) may be involved in the clinical observation of increased severity of symptoms associated with early high levels of SARS-CoV-2 antibodies in patients. Infants with multisystem inflammatory syndrome in children (MIS-C) associated with COVID-19 may also have ADE caused by maternally acquired SARS-CoV-2 antibodies bound to mast cells. ADE risks associated with SARS-CoV-2 has implications for COVID-19 and MIS-C treatments, B-cell vaccines, SARS-CoV-2 antibody therapy, and convalescent plasma therapy for patients. SARS-CoV-2 antibodies bound to mast cells may be involved in MIS-C and multisystem inflammatory syndrome in adults (MIS-A) following initial COVID-19 infection. SARS-CoV-2 antibodies bound to Fc receptors on macrophages and mast cells may represent two different mechanisms for ADE in patients. These two different ADE risks have possible implications for SARS-CoV-2 B-cell vaccines for subsets of populations based on age, cross-reactive antibodies, variabilities in antibody levels over time, and pregnancy. These models place increased emphasis on the importance of developing safe SARS-CoV-2 T cell vaccines that are not dependent upon antibodies.
Collapse
Affiliation(s)
- Darrell O. Ricke
- Biological and Chemical Technologies, Massachusetts Institute of Technology Lincoln Laboratory, Biotechnology and Human Systems, Lexington, MA, United States
| |
Collapse
|
215
|
Silveira MM, Moreira GMSG, Mendonça M. DNA vaccines against COVID-19: Perspectives and challenges. Life Sci 2021; 267:118919. [PMID: 33352173 PMCID: PMC7749647 DOI: 10.1016/j.lfs.2020.118919] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/01/2020] [Accepted: 12/13/2020] [Indexed: 12/23/2022]
Abstract
The coronavirus disease 2019 (COVID-19) is caused by a novel coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is associated with several fatal cases worldwide. The rapid spread of this pathogen and the increasing number of cases highlight the urgent development of vaccines. Among the technologies available for vaccine development, DNA vaccination is a promising alternative to conventional vaccines. Since its discovery in the 1990s, it has been of great interest because of its ability to elicit both humoral and cellular immune responses while showing relevant advantages regarding producibility, stability, and storage. This review aimed to summarize the current knowledge and advancements on DNA vaccines against COVID-19, particularly those in clinical trials.
Collapse
Affiliation(s)
| | | | - Marcelo Mendonça
- Federal University of Agreste of Pernambuco, Veterinary Medicine Course, Garanhuns, Pernambuco, Brazil
| |
Collapse
|
216
|
Koenig PA, Das H, Liu H, Kümmerer BM, Gohr FN, Jenster LM, Schiffelers LDJ, Tesfamariam YM, Uchima M, Wuerth JD, Gatterdam K, Ruetalo N, Christensen MH, Fandrey CI, Normann S, Tödtmann JMP, Pritzl S, Hanke L, Boos J, Yuan M, Zhu X, Schmid-Burgk JL, Kato H, Schindler M, Wilson IA, Geyer M, Ludwig KU, Hällberg BM, Wu NC, Schmidt FI. Structure-guided multivalent nanobodies block SARS-CoV-2 infection and suppress mutational escape. Science 2021; 371:eabe6230. [PMID: 33436526 PMCID: PMC7932109 DOI: 10.1126/science.abe6230] [Citation(s) in RCA: 268] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
The pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread, with devastating consequences. For passive immunization efforts, nanobodies have size and cost advantages over conventional antibodies. In this study, we generated four neutralizing nanobodies that target the receptor binding domain of the SARS-CoV-2 spike protein. We used x-ray crystallography and cryo-electron microscopy to define two distinct binding epitopes. On the basis of these structures, we engineered multivalent nanobodies with more than 100 times the neutralizing activity of monovalent nanobodies. Biparatopic nanobody fusions suppressed the emergence of escape mutants. Several nanobody constructs neutralized through receptor binding competition, whereas other monovalent and biparatopic nanobodies triggered aberrant activation of the spike fusion machinery. These premature conformational changes in the spike protein forestalled productive fusion and rendered the virions noninfectious.
Collapse
MESH Headings
- Amino Acid Substitution
- Angiotensin-Converting Enzyme 2/metabolism
- Animals
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/metabolism
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- Antibodies, Viral/metabolism
- Antibody Affinity
- Antigens, Viral/immunology
- Binding Sites, Antibody
- COVID-19/immunology
- COVID-19/virology
- Cell Line
- Cryoelectron Microscopy
- Epitopes
- Humans
- Membrane Fusion
- Mutation
- Protein Binding
- Protein Conformation
- Protein Domains
- Receptors, Coronavirus/metabolism
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/physiology
- Single-Domain Antibodies/chemistry
- Single-Domain Antibodies/immunology
- Single-Domain Antibodies/metabolism
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Virus Replication
Collapse
Affiliation(s)
- Paul-Albert Koenig
- Core Facility Nanobodies, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Hrishikesh Das
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Hejun Liu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Beate M Kümmerer
- Institute of Virology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, 53127 Bonn, Germany
| | - Florian N Gohr
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Lea-Marie Jenster
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Lisa D J Schiffelers
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Yonas M Tesfamariam
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Miki Uchima
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Jennifer D Wuerth
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Karl Gatterdam
- Institute of Structural Biology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Natalia Ruetalo
- Institute for Medical Virology and Epidemiology, Section Molecular Virology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Maria H Christensen
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Caroline I Fandrey
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Sabine Normann
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Jan M P Tödtmann
- Core Facility Nanobodies, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Steffen Pritzl
- Core Facility Nanobodies, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Leo Hanke
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Jannik Boos
- Institute of Human Genetics, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jonathan L Schmid-Burgk
- Institute for Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Hiroki Kato
- Institute of Cardiovascular Immunology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Michael Schindler
- Institute for Medical Virology and Epidemiology, Section Molecular Virology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Matthias Geyer
- Institute of Structural Biology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Kerstin U Ludwig
- Institute of Human Genetics, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - B Martin Hällberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden.
- Centre for Structural Systems Biology (CSSB) and Karolinska Institutet VR-RÅC, Notkestrasse 85, 22607 Hamburg, Germany
| | - Nicholas C Wu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Florian I Schmidt
- Core Facility Nanobodies, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
217
|
Xu Y, He Y, Momben-Abolfath S, Eller N, Norton M, Zhang P, Scott D, Struble EB. Entry and Disposition of Zika Virus Immune Complexes in a Tissue Culture Model of the Maternal-Fetal Interface. Vaccines (Basel) 2021; 9:vaccines9020145. [PMID: 33670199 PMCID: PMC7916977 DOI: 10.3390/vaccines9020145] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/16/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) infections have been associated with an increased incidence of severe microcephaly and other neurodevelopmental disorders in newborn babies. Passive immunization with anti-ZIKV neutralizing antibodies has the potential to become a feasible treatment or prophylaxis option during pregnancy. Prior to clinical use, such antibodies should be assessed for their ability to block ZIKV passage to the fetus. We used human placental and mammalian cell monolayers that express FcRn and laboratory preparations of anti-ZIKV antibodies as a model system to investigate the disposition of ZIKV/antibody immune complexes (ICs) at the maternal-fetal interface. We further characterized solution properties of the ICs to evaluate whether these are related to in vitro effects. We found that both ZIKV and ZIKV envelope glycoprotein can enter and passage through epithelial cells, especially those that overexpress FcRn. In the presence of ZIKV antibodies, Zika virus entry was bimodal, with reduced entry at the lowest (0.3–3 ng/mL) and highest (µg/mL) antibody concentrations. Intermediate concentrations attenuated inhibition or enhanced viral entry. With respect to anti-ZIKV antibodies, we found that their degradation was accelerated when presented as ICs containing increased amounts of ZIKV immunogen. Of the two monoclonal antibodies tested, the preparation with higher aggregation also exhibited higher degradation. Our studies confirm that intact Zika virus and its envelope immunogen have the potential to enter and be transferred across placental and other epithelial cells that express FcRn. Presence of anti-ZIKV IgG antibodies can either block or enhance cellular entry, with the antibody concentration playing a complex role in this process. Physicochemical properties of IgG antibodies can influence their degradation in vitro.
Collapse
|
218
|
Yoo JH. What We Do Know and Do Not Yet Know about COVID-19 Vaccines as of the Beginning of the Year 2021. J Korean Med Sci 2021; 36:e54. [PMID: 33559409 PMCID: PMC7870421 DOI: 10.3346/jkms.2021.36.e54] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), which started at the end of 2019 and has spread worldwide, has remained unabated in 2021. Since non-pharmaceutical interventions including social distancing are facing limitations in controlling COVID-19, additional absolute means to change the trend are necessary. To this end, coronavirus-specific antiviral drugs and vaccines are urgently needed, but for now, the priority is to promote herd immunity through extensive nationwide vaccination campaign. In addition to the vaccines based on the conventional technology such inactivated or killed virus or protein subunit vaccines, several vaccines on the new technological platforms, for example, nucleic acids-based vaccines delivered by viral carriers, nanoparticles, or plasmids as a medium were introduced in this pandemic. In addition to achieving sufficient herd immunity with vaccination, the development of antiviral treatments that work specifically against COVID-19 will also be necessary to terminate the epidemic completely.
Collapse
Affiliation(s)
- Jin Hong Yoo
- Division of Infectious Diseases, Department of Internal Medicine, Bucheon St. Mary's Hospital, Bucheon, Korea
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
219
|
Bhatta M, Nandi S, Dutta S, Saha MK. Coronavirus (SARS-CoV-2): a systematic review for potential vaccines. Hum Vaccin Immunother 2021; 18:1865774. [PMID: 33545014 PMCID: PMC8920137 DOI: 10.1080/21645515.2020.1865774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
COVID-19 is an international public health emergency in need of effective and safe vaccines for SARS-CoV-2. A systematic review has been done to analyze the availability, development and status of new COVID-19 vaccine candidates as well as the status of vaccines for other diseases that might be effective against SARS-CoV-2 infection. PubMed, MEDLINE, EMBASE, Science Direct, Google Scholar, Cochrane library, ClinicalTrials.gov, Web of Science and different trial registries were searched for currently available and probable future vaccines. Articles and ongoing clinical trials are included to ascertain the availability and developmental approaches of new vaccines that could limit the present and future outbreaks. Pharmaceutical companies and institutions are at different stages of developing new vaccines, and extensive studies and clinical trials are still required.
Collapse
Affiliation(s)
- Mihir Bhatta
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Srijita Nandi
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Malay Kumar Saha
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
220
|
Wang R, Luo X, Liu F, Luo S. Confronting the threat of SARS-CoV-2: Realities, challenges and therapeutic strategies (Review). Exp Ther Med 2021; 21:155. [PMID: 33456522 PMCID: PMC7807638 DOI: 10.3892/etm.2020.9587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
The novel coronavirus (SARS-CoV-2) appeared in2019 in Wuhan, China, and rapidly developed into a global pandemic. The disease has affected not only health care systems and economies worldwide but has also changed the lifestyles and habits of the majority of the world's population. Among the potential targets for SARS-CoV-2 therapy, the viral spike glycoprotein has been studied most intensely, due to its key role in mediating viral entry into target cells and inducing a protective antibody response in infected individuals. In the present manuscript the molecular mechanisms that are responsible for SARS-CoV-2 infection are described and a progress report on the status of SARS-CoV-2 research is provided. A brief review of the clinical symptoms of the condition and current diagnostic methods and treatment plans for SARS-CoV-2 are also presented and the progress of preclinical research into medical intervention against SARS-CoV-2 infection are discussed.
Collapse
Affiliation(s)
- Ruixue Wang
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong 528000, P.R. China
| | - Xiaoshan Luo
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong 528000, P.R. China
| | - Fang Liu
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong 528000, P.R. China
| | - Shuhong Luo
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong 528000, P.R. China
| |
Collapse
|
221
|
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
|
222
|
Sharifi M, Hasan A, Haghighat S, Taghizadeh A, Attar F, Bloukh SH, Edis Z, Xue M, Khan S, Falahati M. Rapid diagnostics of coronavirus disease 2019 in early stages using nanobiosensors: Challenges and opportunities. Talanta 2021; 223:121704. [PMID: 33303154 PMCID: PMC7521920 DOI: 10.1016/j.talanta.2020.121704] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 01/08/2023]
Abstract
The rapid outbreak of coronavirus disease 2019 (COVID-19) around the world is a tragic and shocking event that demonstrates the unpreparedness of humans to develop quick diagnostic platforms for novel infectious diseases. In fact, statistical reports of diagnostic tools show that their accuracy, specificity and sensitivity in the detection of COVID hampered by some challenges that can be eliminated by using nanoparticles (NPs). In this study, we aimed to present an overview on the most important ways to diagnose different kinds of viruses followed by the introduction of nanobiosensors. Afterward, some methods of COVID-19 detection such as imaging, laboratory and kit-based diagnostic tests are surveyed. Furthermore, nucleic acids/protein- and immunoglobulin (Ig)-based nanobiosensors for the COVID-19 detection infection are reviewed. Finally, current challenges and future perspective for the development of diagnostic or monitoring technologies in the control of COVID-19 are discussed to persuade the scientists in advancing their technologies beyond imagination. In conclusion, it can be deduced that as rapid COVID-19 detection infection can play a vital role in disease control and treatment, this review may be of great help for controlling the COVID-19 outbreak by providing some necessary information for the development of portable, accurate, selectable and simple nanobiosensors.
Collapse
Affiliation(s)
- Majid Sharifi
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Akbar Taghizadeh
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Farnoosh Attar
- Department of Food Toxicology, Research Center of Food Technology and Agricultural Products, Standard Research Institute (SRI), Karaj, Iran
| | - Samir Haj Bloukh
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, PO Box 346, Ajman, United Arab Emirates
| | - Zehra Edis
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, PO Box 346, Ajman, United Arab Emirates
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
223
|
Sajna KV, Kamat S. Antibodies at work in the time of severe acute respiratory syndrome coronavirus 2. Cytotherapy 2021; 23:101-110. [PMID: 32988772 PMCID: PMC7458058 DOI: 10.1016/j.jcyt.2020.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022]
Abstract
In view of devastating effects of COVID-19 on human life, there is an urgent need for the licened vaccines or therapeutics for the SARS-CoV-2 infection. Age-old passive immunization with protective antibodies to neutralize the virus is one of the strategies for emergency prophylaxis and therapy for coronavirus disease 2019 (COVID-19). In this review, the authors discuss up-to-date advances in immune-based therapy for COVID-19. The use of convalescent plasma therapy as the first line of defense to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been established, with encouraging results. Monoclonal antibodies (mAbs) that bind to the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein or block the interaction between SARS-CoV-2 RBD and the human angiotensin-converting enzyme 2 receptor have been found to be very promising as a countermeasure for tackling the SARS-CoV-2 infection, and clinical trials are underway. Considering the counterproductive antibody-dependent enhancement of the virus, mAbs therapy that is safe and efficacious, even in people with underlying conditions, will be a significant breakthrough. In addition, emerging immunotherapeutic interventions using nanobodies and cellular immunotherapy are promising avenues for tackling the COVID-19 pandemic. The authors also discuss the implication of mAbs as mediators of cytokine storm syndrome to modify the immune response of COVID-19 patients, thus reducing the fatality rate of COVID-19 infection.
Collapse
Affiliation(s)
| | - Siya Kamat
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| |
Collapse
|
224
|
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: 0.5] [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
|
225
|
Moore KM, Suthar MS. Comprehensive analysis of COVID-19 during pregnancy. Biochem Biophys Res Commun 2021; 538:180-186. [PMID: 33384142 PMCID: PMC7759124 DOI: 10.1016/j.bbrc.2020.12.064] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022]
Abstract
The COVID-19 pandemic resulting from the emergence of the coronavirus SARS-CoV-2 remains a major global health concern. Pregnant individuals are more likely to develop severe COVID-19 and a number of pregnancy complications have been observed in COVID-19 patients. To date, little is known about the impact of COVID-19 on pregnancy. In this review, we examine key aspects of pregnancy that may be impacted by COVID-19 and summarize the current literature on SARS-CoV-2 infection of the placenta and in utero vertical transmission. Furthermore, we highlight recent studies exploring the role of the maternal antibody response to SARS-CoV-2 during pregnancy and the passive transfer of maternal antibodies from mothers with COVID-19 to fetus.
Collapse
Affiliation(s)
- Kathryn M Moore
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, 30322, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Atlanta, GA, 30329, USA
| | - Mehul S Suthar
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, 30322, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Atlanta, GA, 30329, USA.
| |
Collapse
|
226
|
Carrillo J, Izquierdo-Useros N, Ávila-Nieto C, Pradenas E, Clotet B, Blanco J. Humoral immune responses and neutralizing antibodies against SARS-CoV-2; implications in pathogenesis and protective immunity. Biochem Biophys Res Commun 2021; 538:187-191. [PMID: 33187644 PMCID: PMC7648524 DOI: 10.1016/j.bbrc.2020.10.108] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
The magnitude and the quality of humoral responses against SARS-CoV-2 have been associated with clinical outcome. Although the elicitation of humoral responses against different viral proteins is rapid and occurs in most infected individuals, its magnitude is highly variable among them and positively correlates with COVID-19 disease severity. This rapid response is characterized by the almost concomitant appearance of virus-specific IgG, IgA and IgM antibodies that contain neutralizing antibodies directed against different epitopes of the Spike glycoprotein. Of particularly interest, the antibodies against domain of the Spike that interacts with the cellular receptor ACE2, known as the receptor binding domain (RBD), are present in most infected individuals and are block viral entry and infectivity. Such neutralizing antibodies protect different animal species when administered before virus exposure; therefore, its elicitation is the main target of current vaccine approaches and their clinical use as recombinant monoclonal antibodies (mAbs) is being explored. Yet, little information exists on the duration of humoral responses during natural infection. This is a key issue that will impact the management of the pandemic and determine the utility of seroconversion studies and the level of herd immunity. Certainly, several cases of reinfection have been reported, suggesting that immunity could be transient, as reported for other coronaviruses. In summary, although the kinetics of the generation of antibodies against SASR-CoV-2 and their protective activity have been clearly defined, their role in COVID-19 pathogenesis and the length of these responses are still open questions.
Collapse
Affiliation(s)
- Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,Corresponding author. Institut de Recerca de la Sida, IrsiCaixa Hospital Universitari Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916, Badalona, Barcelona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Carlos Ávila-Nieto
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Edwards Pradenas
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,Infectious Diseases Department, Germans Trias i Pujol Hospital, Badalona, Catalonia, Spain,University of Vic (UVic-UCC), Vic, Catalonia, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Catalonia, Spain,University of Vic (UVic-UCC), Vic, Catalonia, Spain,Corresponding author. Institut de Recerca de la Sida, IrsiCaixa Hospital Universitari Germans Trias i Pujol, Ctra. de Canyet s/n. 2a Planta Maternal, 08916, Badalona, Barcelona, Spain
| |
Collapse
|
227
|
Trinité B, Tarrés-Freixas F, Rodon J, Pradenas E, Urrea V, Marfil S, Rodríguez de la Concepción ML, Ávila-Nieto C, Aguilar-Gurrieri C, Barajas A, Ortiz R, Paredes R, Mateu L, Valencia A, Guallar V, Ruiz L, Grau E, Massanella M, Puig J, Chamorro A, Izquierdo-Useros N, Segalés J, Clotet B, Carrillo J, Vergara-Alert J, Blanco J. SARS-CoV-2 infection elicits a rapid neutralizing antibody response that correlates with disease severity. Sci Rep 2021; 11:2608. [PMID: 33510275 PMCID: PMC7843981 DOI: 10.1038/s41598-021-81862-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
The protective effect of neutralizing antibodies in SARS-CoV-2 infected individuals is not yet well defined. To address this issue, we have analyzed the kinetics of neutralizing antibody responses and their association with disease severity. Between March and May 2020, the prospective KING study enrolled 72 COVID-19+ participants grouped according to disease severity. SARS-CoV-2 infection was diagnosed by serological and virological tests. Plasma neutralizing responses were assessed against replicative virus and pseudoviral particles. Multiple regression and non-parametric tests were used to analyze dependence of parameters. The magnitude of neutralizing titers significantly increased with disease severity. Hospitalized individuals developed higher titers compared to mild-symptomatic and asymptomatic individuals, which together showed titers below the detection limit in 50% of cases. Longitudinal analysis confirmed the strong differences in neutralizing titers between non-hospitalized and hospitalized participants and showed rapid kinetics of appearance of neutralizing antibodies (50% and 80% of maximal activity reached after 11 and 17 days after symptoms onset, respectively) in hospitalized patients. No significant impact of age, gender or treatment on the neutralizing titers was observed in this limited cohort. These data identify a clear association of humoral immunity with disease severity and point to immune mechanisms other than antibodies as relevant players in COVID-19 protection.
Collapse
Affiliation(s)
- Benjamin Trinité
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Ferran Tarrés-Freixas
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Jordi Rodon
- IRTA Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la UAB, 08193, Bellaterra, Catalonia, Spain
| | - Edwards Pradenas
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Víctor Urrea
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Silvia Marfil
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - María Luisa Rodríguez de la Concepción
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Carlos Ávila-Nieto
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Carmen Aguilar-Gurrieri
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Ana Barajas
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Raquel Ortiz
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Roger Paredes
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain.,Infectious Diseases Department, Fight Against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Badalona, Catalonia, Spain
| | - Lourdes Mateu
- Infectious Diseases Department, Fight Against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Badalona, Catalonia, Spain
| | | | - Víctor Guallar
- Barcelona Supercomputing Center, Barcelona, Catalonia, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| | - Lidia Ruiz
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Eulàlia Grau
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Marta Massanella
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Jordi Puig
- Infectious Diseases Department, Fight Against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Badalona, Catalonia, Spain
| | - Anna Chamorro
- Infectious Diseases Department, Fight Against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Badalona, Catalonia, Spain
| | - Nuria Izquierdo-Useros
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Joaquim Segalés
- IRTA Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la UAB, 08193, Bellaterra, Catalonia, Spain.,UAB, CReSA (IRTA-UAB), Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Catalonia, Spain
| | - Bonaventura Clotet
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain.,Infectious Diseases Department, Fight Against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Badalona, Catalonia, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
| | - Jorge Carrillo
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain
| | - Júlia Vergara-Alert
- IRTA Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la UAB, 08193, Bellaterra, Catalonia, Spain
| | - Julià Blanco
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Hospital Universitari Germans Trias I Pujol, Can Ruti Campus, Ctra, de Canyet s/n, 2a Planta Maternal, 08916, Badalona, Catalonia, Spain. .,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain.
| |
Collapse
|
228
|
Lin JE, Asfour A, Sewell TB, Hooe B, Pryce P, Earley C, Shen MY, Kerner-Rossi M, Thakur KT, Vargas WS, Silver WG, Geneslaw AS. Neurological issues in children with COVID-19. Neurosci Lett 2021; 743:135567. [PMID: 33352286 PMCID: PMC7831718 DOI: 10.1016/j.neulet.2020.135567] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022]
Abstract
Coronavirus disease 2019 (COVID-19) usually leads to a mild infectious disease course in children, but serious complications may occur in conjunction with both acute infection and associated phenomena such as the multisystem inflammatory syndrome in children (MIS-C). Neurological symptoms, which have been predominantly reported in adults, range from mild headache to seizure, peripheral neuropathy, stroke, demyelinating disorders, and encephalopathy. Similar to respiratory and cardiac manifestations of COVID-19, neurological complications present differently based on age and underlying comorbidities. This review provides a concise overview of the neurological conditions seen in the context of COVID-19, as well as potential mechanisms and long-term implications of COVID-19 in the pediatric population from literature reviews and primary data collected at NewYork-Presbyterian Morgan Stanley Children's Hospital.
Collapse
Affiliation(s)
- Jieru E Lin
- Department of Neurology, Division of Child Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Pediatrics, Columbia University Irving Medical Center, Morgan Stanley Children's Hospital, New York, NY 10032, United States
| | - Arsenoi Asfour
- Department of Neurology, Division of Child Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Pediatrics, Columbia University Irving Medical Center, Morgan Stanley Children's Hospital, New York, NY 10032, United States
| | - Taylor B Sewell
- Department of Pediatrics, Division of Pediatric Critical Care and Hospital Medicine, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Benjamin Hooe
- Department of Pediatrics, Division of Pediatric Critical Care and Hospital Medicine, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Patrice Pryce
- Department of Pediatrics, Division of Pediatric Critical Care and Hospital Medicine, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Chelsea Earley
- Department of Neurology, Division of Child Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Pediatrics, Columbia University Irving Medical Center, Morgan Stanley Children's Hospital, New York, NY 10032, United States
| | - Min Ye Shen
- Department of Neurology, Division of Child Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Pediatrics, Columbia University Irving Medical Center, Morgan Stanley Children's Hospital, New York, NY 10032, United States
| | - Mallory Kerner-Rossi
- Department of Neurology, Division of Child Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States; Department of Pediatrics, Columbia University Irving Medical Center, Morgan Stanley Children's Hospital, New York, NY 10032, United States
| | - Kiran T Thakur
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Wendy S Vargas
- Department of Neurology, Division of Child Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Wendy G Silver
- Department of Neurology, Division of Child Neurology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Andrew S Geneslaw
- Department of Pediatrics, Division of Pediatric Critical Care and Hospital Medicine, Columbia University Irving Medical Center, New York, NY 10032, United States.
| |
Collapse
|
229
|
Pinotti F, Wikramaratna PS, Obolski U, Paton RS, Damineli DSC, Alcantara LCJ, Giovanetti M, Gupta S, Lourenço J. Potential impact of individual exposure histories to endemic human coronaviruses on age-dependent severity of COVID-19. BMC Med 2021; 19:19. [PMID: 33430856 PMCID: PMC7801230 DOI: 10.1186/s12916-020-01887-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/11/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Cross-reactivity to SARS-CoV-2 from exposure to endemic human coronaviruses (eHCoV) is gaining increasing attention as a possible driver of both protection against infection and COVID-19 severity. Here we explore the potential role of cross-reactivity induced by eHCoVs on age-specific COVID-19 severity in a mathematical model of eHCoV and SARS-CoV-2 transmission. METHODS We use an individual-based model, calibrated to prior knowledge of eHCoV dynamics, to fully track individual histories of exposure to eHCoVs. We also model the emergent dynamics of SARS-CoV-2 and the risk of hospitalisation upon infection. RESULTS We hypothesise that primary exposure with any eHCoV confers temporary cross-protection against severe SARS-CoV-2 infection, while life-long re-exposure to the same eHCoV diminishes cross-protection, and increases the potential for disease severity. We show numerically that our proposed mechanism can explain age patterns of COVID-19 hospitalisation in EU/EEA countries and the UK. We further show that some of the observed variation in health care capacity and testing efforts is compatible with country-specific differences in hospitalisation rates under this model. CONCLUSIONS This study provides a "proof of possibility" for certain biological and epidemiological mechanisms that could potentially drive COVID-19-related variation across age groups. Our findings call for further research on the role of cross-reactivity to eHCoVs and highlight data interpretation challenges arising from health care capacity and SARS-CoV-2 testing.
Collapse
Affiliation(s)
| | | | - Uri Obolski
- School of Public Health, Tel Aviv University, Tel Aviv, Israel
- Porter School of the Environment and Earth Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Daniel S C Damineli
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz C J Alcantara
- Laboratório de Genética Celular e Molecular, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratório de Flavivírus, Instituto Oswaldo Cruz Fiocruz, Rio de Janeiro, Brazil
| | - Marta Giovanetti
- Laboratório de Genética Celular e Molecular, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratório de Flavivírus, Instituto Oswaldo Cruz Fiocruz, Rio de Janeiro, Brazil
| | - Sunetra Gupta
- Department of Zoology, University of Oxford, Oxford, UK
| | - José Lourenço
- Department of Zoology, University of Oxford, Oxford, UK
| |
Collapse
|
230
|
Development and structural basis of a two-MAb cocktail for treating SARS-CoV-2 infections. Nat Commun 2021; 12:264. [PMID: 33431876 PMCID: PMC7801428 DOI: 10.1038/s41467-020-20465-w] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/03/2020] [Indexed: 01/10/2023] Open
Abstract
The ongoing pandemic of coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Neutralizing antibodies against SARS-CoV-2 are an option for drug development for treating COVID-19. Here, we report the identification and characterization of two groups of mouse neutralizing monoclonal antibodies (MAbs) targeting the receptor-binding domain (RBD) on the SARS-CoV-2 spike (S) protein. MAbs 2H2 and 3C1, representing the two antibody groups, respectively, bind distinct epitopes and are compatible in formulating a noncompeting antibody cocktail. A humanized version of the 2H2/3C1 cocktail is found to potently neutralize authentic SARS-CoV-2 infection in vitro with half inhibitory concentration (IC50) of 12 ng/mL and effectively treat SARS-CoV-2-infected mice even when administered at as late as 24 h post-infection. We determine an ensemble of cryo-EM structures of 2H2 or 3C1 Fab in complex with the S trimer up to 3.8 Å resolution, revealing the conformational space of the antigen–antibody complexes and MAb-triggered stepwise allosteric rearrangements of the S trimer, delineating a previously uncharacterized dynamic process of coordinated binding of neutralizing antibodies to the trimeric S protein. Our findings provide important information for the development of MAb-based drugs for preventing and treating SARS-CoV-2 infections. Here, the authors identify and characterize two mouse-derived monoclonal antibodies against SARS-CoV-2 spike protein that target different epitopes in RBD and block the interaction S/ACE2 and show that a formulated humanized version cocktail exhibits prophylaxis and therapeutic antiviral effects in an hACE2-adenovector expressed mouse model.
Collapse
|
231
|
Shahrajabian MH, Sun W, Cheng Q. Product of natural evolution (SARS, MERS, and SARS-CoV-2); deadly diseases, from SARS to SARS-CoV-2. Hum Vaccin Immunother 2021; 17:62-83. [PMID: 32783700 PMCID: PMC7872062 DOI: 10.1080/21645515.2020.1797369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/24/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
SARS-CoV-2, the virus causing COVID-19, is a single-stranded RNA virus belonging to the order Nidovirales, family Coronaviridae, and subfamily Coronavirinae. SARS-CoV-2 entry to cellsis initiated by the binding of the viral spike protein (S) to its cellular receptor. The roles of S protein in receptor binding and membrane fusion makes it a prominent target for vaccine development. SARS-CoV-2 genome sequence analysis has shown that this virus belongs to the beta-coronavirus genus, which includes Bat SARS-like coronavirus, SARS-CoV and MERS-CoV. A vaccine should induce a balanced immune response to elicit protective immunity. In this review, we compare and contrast these three important CoV diseases and how they inform on vaccine development.
Collapse
Affiliation(s)
| | - Wenli Sun
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qi Cheng
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
- College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, China
- Global Alliance of HeBAU-CLS&HeQiS for BioAl-Manufacturing, Baoding, Hebei, China
| |
Collapse
|
232
|
Bonetti TCDS, Latini FRM, Invitti AL, Fonseca MCM, Scorza FA, Saldanha MG, Bellucco FT, Bacarov NB, Soane MM, Girão MJBC. Antibody indexes in COVID-19 convalescent plasma donors: Unanswered questions. Clinics (Sao Paulo) 2021; 76:e2818. [PMID: 34468538 PMCID: PMC8366902 DOI: 10.6061/clinics/2021/e2818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is characterized by high contagiousness, as well as variable clinical manifestations and immune responses. The antibody response to SARS-CoV-2 is directly related to viral clearance and the antibodies' ability to neutralize the virus and confer long-term immunity. Nevertheless, the response can also be associated with disease severity and evolution. This study correlated the clinical characteristics of convalescent COVID-19 patients with immunoglobulin A (IgA) and IgG anti-SARS-CoV-2 antibodies. METHODS This study included 51 COVID-19 health care professionals who were candidates for convalescent plasma donation from April to June 2020. The subjects had symptomatic COVID-19 with a polymerase chain reaction-confirmed diagnosis. We measured anti-SARS-CoV-2 IgA and IgG antibodies after symptom recovery, and the subjects were classified as having mild, moderate, or severe symptoms. RESULTS Anti-SARS-CoV-2 antibodies were positive in most patients (90.2%). The antibody indexes for IgA and IgG did not differ significantly between patients presenting with mild or moderate symptoms. However, they were significantly higher in patients with severe symptoms. CONCLUSIONS Our study showed an association between higher antibody indexes and severe COVID-19 cases, and several hypotheses regarding the association of the antibody dynamics and severity of the disease in SARS-CoV-2 infection have been raised, although many questions remain unanswered.
Collapse
Affiliation(s)
- Tatiana Carvalho de Souza Bonetti
- Laboratorio de Ginecologia Molecular, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
- Centro de Neurociencias e Saude da Mulher “Professor Geraldo Rodrigues de Lima”, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
- Corresponding author. E-mail:
| | | | - Adriana Lukow Invitti
- Laboratorio de Ginecologia Molecular, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
- Centro de Neurociencias e Saude da Mulher “Professor Geraldo Rodrigues de Lima”, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
- Associacao Beneficente de Coleta de Sangue (COLSAN), Sao Paulo, SP, BR
| | - Marcelo Cunio Machado Fonseca
- Laboratorio de Ginecologia Molecular, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
- Centro de Neurociencias e Saude da Mulher “Professor Geraldo Rodrigues de Lima”, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
| | - Fulvio Alexandre Scorza
- Centro de Neurociencias e Saude da Mulher “Professor Geraldo Rodrigues de Lima”, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
- Disciplina de Neurociencia, Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
| | - Maira Garcia Saldanha
- Laboratorio de Ginecologia Molecular, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
| | | | | | | | - Manoel João Batista Castelo Girão
- Laboratorio de Ginecologia Molecular, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
- Centro de Neurociencias e Saude da Mulher “Professor Geraldo Rodrigues de Lima”, Departamento de Ginecologia, Escola Paulista de Medicina/Universidade Federal de Sao Paulo (EPM/UNIFESP), Sao Paulo, SP, BR
- Associacao Beneficente de Coleta de Sangue (COLSAN), Sao Paulo, SP, BR
| |
Collapse
|
233
|
Rothan HA, Byrareddy SN. The potential threat of multisystem inflammatory syndrome in children during the COVID-19 pandemic. Pediatr Allergy Immunol 2021; 32:17-22. [PMID: 32897642 PMCID: PMC7887110 DOI: 10.1111/pai.13361] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/15/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) during the COVID-19 pandemic raised a global alert from the Centers for Disease Control and Prevention's Health Alert Network. The main manifestations of MIS-C (also known as pediatric MIS (PMIS)) in the setting of a severe inflammatory state include fever, diarrhea, shock, and variable presence of rash, conjunctivitis, extremity edema, and mucous membrane changes. In some cases, these symptoms progressed to multi-organ failure. The low percentage of children with asymptomatic cases compared with mild illness and moderate illness could be correlated with the rare cases of MIS-C. One potential explanation for the progression to severe MIS-C disease despite the presence of readily detectable anti-SARS-CoV-2 antibodies could be due to the potential role of antibody-dependent enhancement (ADE). We reason that the incidence of the ADE phenomenon whereby the pathogen-specific antibodies can promote pathology should be considered in vaccine development against SARS-CoV-2.
Collapse
Affiliation(s)
- Hussin A. Rothan
- Department of Biology, College of Arts and Sciences, Georgia State University, GA, USA
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Centre, Omaha, NE, USA
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Centre, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Centre, Omaha, NE, USA
| |
Collapse
|
234
|
Wang Y, Li J, Li H, Lei P, Shen G, Yang C. Persistence of SARS-CoV-2-specific antibodies in COVID-19 patients. Int Immunopharmacol 2021; 90:107271. [PMID: 33310664 PMCID: PMC7724312 DOI: 10.1016/j.intimp.2020.107271] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 11/16/2022]
Abstract
To better understand humoral immunity following SARS-CoV-2 infection, 114 hospitalised COVID-19 patients with antibody monitored over 8 weeks from symptom onset were retrospectively investigated. A total of 445 serum samples were assessed via chemiluminescence immunoassay. Positive rate of virus-specific IgM reached up to over 80% from the second week to the eighth week after symptom onset, then declined quickly to below 30% in the twelfth week. Concentrations of IgG remained high for at least 3 months before subsequently declining. As compared with the non-severe group, serum IgM level from week 3 to week 8 was significantly higher among the patients with severe clinical symptoms (P = 0.012) but not IgG (P = 0.053). Serum IgM level from week 3 to week 8 was correlated with positive virus RNA test (r = 0.201, P = 0.044), albumin level (r = -0.295, P = 0.003), lactic dehydrogenase (LDH) level (r = 0.292, P = 0.003), alkaline phosphatase (ALP) level (r = 0.254, P = 0.010), C-reactive protein (CRP) level (r = 0.281, P = 0.004) during the same course, while serum IgG level was correlated with age (r = 0.207, P = 0.038). This presented results provide insight into duration of SARS-CoV-2 antibodies and interaction between the virus and host systems.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Li
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huijun Li
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Lei
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunguang Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
235
|
A review on the immune responses against novel emerging coronavirus (SARS-CoV-2). Immunol Res 2021; 69. [PMID: 33928531 PMCID: PMC8084416 DOI: 10.1007/s12026-021-09198-0&n933034=v971361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a highly contagious disease caused by a newly identified coronavirus called the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) which was initially emerged in Wuhan, China in late December 2019 and then rapidly extended to other countries worldwide. COVID-19 is now known as a pandemic threat to global public health. It possesses a wide spectrum of clinical manifestations, ranging from asymptomatic infection to mild, moderate, and ultimately severe pneumonia accompanied by multi-organ system dysfunction that can cause the death of the afflicted patients. The host immune system plays a critical role in defending against potentially pathogenic microorganisms such as coronaviruses, and it eliminates and eradicates these invading agents by triggering effective immune responses. However, there exists evidence indicating that in critically ill cases of the COVID-19, dysregulated immune responses and hyper-inflammation lead to acute respiratory distress syndrome (ARDS) and multi-organ failure. Achieving a profound understanding of the pathological immune responses involved in the pathogenesis of COVID-19 will boost our comprehending of disease pathogenesis and its progression toward severe form, contributing to the identification and rational design of effective therapies. In this review, we have tried to summarize the current knowledge regarding the role of immune responses against SARS-CoV-2 and also give a glimpse of the immune evasion strategies of this virus.
Collapse
|
236
|
Chen Y, Klein SL, Garibaldi BT, Li H, Wu C, Osevala NM, Li T, Margolick JB, Pawelec G, Leng SX. Aging in COVID-19: Vulnerability, immunity and intervention. Ageing Res Rev 2021; 65:101205. [PMID: 33137510 PMCID: PMC7604159 DOI: 10.1016/j.arr.2020.101205] [Citation(s) in RCA: 555] [Impact Index Per Article: 138.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic was first reported in Wuhan, China in December 2019, moved across the globe at an unprecedented speed, and is having a profound and yet still unfolding health and socioeconomic impacts. SARS-CoV-2, a β-coronavirus, is a highly contagious respiratory pathogen that causes a disease that has been termed the 2019 coronavirus disease (COVID-19). Clinical experience thus far indicates that COVID-19 is highly heterogeneous, ranging from being asymptomatic and mild to severe and causing death. Host factors including age, sex, and comorbid conditions are key determinants of disease severity and progression. Aging itself is a prominent risk factor for severe disease and death from COVID-19. We hypothesize that age-related decline and dysregulation of immune function, i.e., immunosenescence and inflammaging play a major role in contributing to heightened vulnerability to severe COVID-19 outcomes in older adults. Much remains to be learned about the immune responses to SARS-CoV-2 infection. We need to begin partitioning all immunological outcome data by age to better understand disease heterogeneity and aging. Such knowledge is critical not only for understanding of COVID-19 pathogenesis but also for COVID-19 vaccine development.
Collapse
Affiliation(s)
- Yiyin Chen
- Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Brian T Garibaldi
- Division of Pulmonary and Critical Care Medicine and Johns Hopkins Biocontainment Unit, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Huifen Li
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Cunjin Wu
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Geriatrics, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Nicole M Osevala
- Division of Geriatrics, Department of Medicine, Pennsylvania State University School of Medicine, Hershey, PA, United States
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Joseph B Margolick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Graham Pawelec
- Department of Immunology, University of Tübingen, Tübingen, Germany; Health Sciences North Research Institute, Sudbury, ON, Canada
| | - Sean X Leng
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States; Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
237
|
Xu L, Ma Z, Li Y, Pang Z, Xiao S. Antibody dependent enhancement: Unavoidable problems in vaccine development. Adv Immunol 2021; 151:99-133. [PMID: 34656289 PMCID: PMC8438590 DOI: 10.1016/bs.ai.2021.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In some cases, antibodies can enhance virus entry and replication in cells. This phenomenon is called antibody-dependent infection enhancement (ADE). ADE not only promotes the virus to be recognized by the target cell and enters the target cell, but also affects the signal transmission in the target cell. Early formalin-inactivated virus vaccines such as aluminum adjuvants (RSV and measles) have been shown to induce ADE. Although there is no direct evidence that there is ADE in COVID-19, this potential risk is a huge challenge for prevention and vaccine development. This article focuses on the virus-induced ADE phenomenon and its molecular mechanism. It also summarizes various attempts in vaccine research and development to eliminate the ADE phenomenon, and proposes to avoid ADE in vaccine development from the perspective of antigens and adjuvants.
Collapse
|
238
|
Ye CH, Hsu WL, Peng GR, Yu WC, Lin WC, Hu S, Yu SH. Role of the Immune Microenvironment in SARS-CoV-2 Infection. Cell Transplant 2021; 30:9636897211010632. [PMID: 33949207 PMCID: PMC8114753 DOI: 10.1177/09636897211010632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV-2) first emerged in December 2019 in Wuhan, China, and has since spread rapidly worldwide. As researchers seek to learn more about COVID-19, the disease it causes, this novel virus continues to infect and kill. Despite the socioeconomic impacts of SARS-CoV-2 infections and likelihood of future outbreaks of other pathogenic coronaviruses, options to prevent or treat coronavirus infections remain limited. In current clinical trials, potential coronavirus treatments focusing on killing the virus or on preventing infection using vaccines largely ignore the host immune response. The relatively small body of current research on the virus indicates pathological responses by the immune system as the leading cause for much of the morbidity and mortality caused by COVID-19. In this review, we investigated the host innate and adaptive immune responses against COVID-19, collated information on recent COVID-19 experimental data, and summarized the systemic immune responses to and histopathology of SARS-CoV-2 infection. Finally, we summarized the immune-related biomarkers to define patients with high-risk and worst-case outcomes, and identified the possible usefulness of inflammatory markers as potential immunotherapeutic targets. This review provides an overview of current knowledge on COVID-19 and the symptomatological differences between healthy, convalescent, and severe cohorts, while offering research directions for alternative immunoregulation therapeutic targets.
Collapse
Affiliation(s)
- Chih-Hung Ye
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Wen-Lin Hsu
- Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Cancer Center, Hualien, Taiwan
- Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Department of Radiation Oncology, Hualien, Taiwan
| | - Guan-Ru Peng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Wei-Chieh Yu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Wei-Chen Lin
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - SuiYun Hu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Shu-Han Yu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
239
|
Lotfi R, Kalmarzi RN, Roghani SA. A review on the immune responses against novel emerging coronavirus (SARS-CoV-2). Immunol Res 2021; 69:213-224. [PMID: 33928531 PMCID: PMC8084416 DOI: 10.1007/s12026-021-09198-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 01/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a highly contagious disease caused by a newly identified coronavirus called the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) which was initially emerged in Wuhan, China in late December 2019 and then rapidly extended to other countries worldwide. COVID-19 is now known as a pandemic threat to global public health. It possesses a wide spectrum of clinical manifestations, ranging from asymptomatic infection to mild, moderate, and ultimately severe pneumonia accompanied by multi-organ system dysfunction that can cause the death of the afflicted patients. The host immune system plays a critical role in defending against potentially pathogenic microorganisms such as coronaviruses, and it eliminates and eradicates these invading agents by triggering effective immune responses. However, there exists evidence indicating that in critically ill cases of the COVID-19, dysregulated immune responses and hyper-inflammation lead to acute respiratory distress syndrome (ARDS) and multi-organ failure. Achieving a profound understanding of the pathological immune responses involved in the pathogenesis of COVID-19 will boost our comprehending of disease pathogenesis and its progression toward severe form, contributing to the identification and rational design of effective therapies. In this review, we have tried to summarize the current knowledge regarding the role of immune responses against SARS-CoV-2 and also give a glimpse of the immune evasion strategies of this virus.
Collapse
Affiliation(s)
- Ramin Lotfi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, PO-Box: 6617713446, Sanandaj, Iran.
- Lung Diseases and Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Rasoul Nasiri Kalmarzi
- Lung Diseases and Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Seyed Askar Roghani
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
240
|
Al‐Riyami AZ, Schäfer R, van den Berg K, Bloch EM, Estcourt LJ, Goel R, Hindawi S, Josephson CD, Land K, McQuilten ZK, Spitalnik SL, Wood EM, Devine DV, So‐Osman C. Clinical use of Convalescent Plasma in the COVID-19 pandemic: a transfusion-focussed gap analysis with recommendations for future research priorities. Vox Sang 2021; 116:88-98. [PMID: 32542847 PMCID: PMC7891452 DOI: 10.1111/vox.12973] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVES Use of convalescent plasma for coronavirus disease 2019 (COVID-19) treatment has gained interest worldwide. However, there is lack of evidence on its dosing, safety and effectiveness. Until data from clinical studies are available to provide solid evidence for worldwide applicable guidelines, there is a need to provide guidance to the transfusion community and researchers on this emergent therapeutic option. This paper aims to identify existing key gaps in current knowledge in the clinical application of COVID-19 convalescent plasma (CCP). MATERIALS AND METHODS The International Society of Blood Transfusion (ISBT) initiated a multidisciplinary working group with worldwide representation from all six continents with the aim of reviewing existing practices on CCP use from donor, product and patient perspectives. A subgroup of clinical transfusion professionals was formed to draft a document for CCP clinical application to identify the gaps in knowledge in existing literature. RESULTS Gaps in knowledge were identified in the following main domains: study design, patient eligibility, CCP dose, frequency and timing of CCP administration, parameters to assess response to CCP treatment and long-term outcome, adverse events and CCP application in less-resourced countries as well as in paediatrics and neonates. CONCLUSION This paper outlines a framework of gaps in the knowledge of clinical deployment of CPP that were identified as being most relevant. Studies to address the identified gaps are required to provide better evidence on the effectiveness and safety of CCP use.
Collapse
Affiliation(s)
- Arwa Z. Al‐Riyami
- Department of HaematologySultan Qaboos University HospitalMuscatSultanate of Oman
| | - Richard Schäfer
- Institute for Transfusion Medicine and ImmunohaematologyGerman Red Cross Blood Donor Service Baden‐Württemberg‐Hessen gGmbHGoethe University HospitalFrankfurt am MainGermany
| | - Karin van den Berg
- Medical DivisionTranslational Research DepartmentSouth African National Blood ServicePort ElizabethSouth Africa
- Division Clinical HaematologyDepartment of MedicineUniversity of Cape TownCape TownSouth Africa
| | - Evan M. Bloch
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Lise J. Estcourt
- Radcliffe Department of MedicineUniversity of Oxford and NHS Blood and TransplantOxfordUK
| | - Ruchika Goel
- Division of Transfusion MedicineDepartment of PathologyJohns Hopkins HospitalBaltimoreMDUSA
- Division of Hematology/OncologySimmons Cancer Institute at SIU School of Medicine and Mississippi Valley Regional Blood CenterSpringfieldILUSA
| | - Salwa Hindawi
- Haematology DepartmentFaculty of MedicineKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Cassandra D. Josephson
- Department of Pathology and Laboratory MedicineCenter for Transfusion and Cellular TherapiesEmory University School of MedicineAtlantaGEUSA
- Department of PediatricsAflac Cancer Center and Blood DisordersChildren's Healthcare of AtlantaEmory University School of MedicineAtlantaGEUSA
| | - Kevin Land
- Corporate Medical AffairsVitalantPhoenixAZUSA
- Department of PathologyUT Health Science San AntonioSan AntonioTXUSA
| | - Zoe K. McQuilten
- Transfusion Research UnitSchool of Public Health and Preventive MedicineMonash UniversityMelbourneVICAustralia
- Department of Clinical HaematologyMonash HealthMelbourneVICAustralia
| | | | - Erica M. Wood
- Transfusion Research UnitSchool of Public Health and Preventive MedicineMonash UniversityMelbourneVICAustralia
- Department of Clinical HaematologyMonash HealthMelbourneVICAustralia
| | - Dana V. Devine
- Canadian Blood ServicesVancouverBCCanada
- Department of Pathology & Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Cynthia So‐Osman
- Department Unit Transfusion MedicineSanquin Blood Supply FoundationAmsterdamThe Netherlands
- Department HaematologyErasmus Medical CenterRotterdamThe Netherlands
| |
Collapse
|
241
|
Han J, Sun J, Zhang G, Chen H. DCs-based therapies: potential strategies in severe SARS-CoV-2 infection. Int J Med Sci 2021; 18:406-418. [PMID: 33390810 PMCID: PMC7757148 DOI: 10.7150/ijms.47706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/09/2020] [Indexed: 01/08/2023] Open
Abstract
Pneumonia caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is spreading globally. There have been strenuous efforts to reveal the mechanisms that the host defends itself against invasion by this virus. The immune system could play a crucial role in virus infection. Dendritic cell as sentinel of the immune system plays an irreplaceable role. Dendritic cells-based therapeutic approach may be a potential strategy for SARS-CoV-2 infection. In this review, the characteristics of coronavirus are described briefly. We focus on the essential functions of dendritic cell in severe SARS-CoV-2 infection. Basis of treatment based dendritic cells to combat coronavirus infections is summarized. Finally, we propose that the combination of DCs based vaccine and other therapy is worth further study.
Collapse
Affiliation(s)
- Jian Han
- General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine of Dalian Medical University, Dalian 116044, China
- Department of Pharmaceutical Sciences USF Health, Taneja College of Pharmacy University of South Florida, Tampa, FL, USA
| | - Jiazhi Sun
- Department of Pharmaceutical Sciences USF Health, Taneja College of Pharmacy University of South Florida, Tampa, FL, USA
| | - Guixin Zhang
- General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine of Dalian Medical University, Dalian 116044, China
| | - Hailong Chen
- General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine of Dalian Medical University, Dalian 116044, China
| |
Collapse
|
242
|
Sánchez-Zuno GA, Matuz-Flores MG, González-Estevez G, Nicoletti F, Turrubiates-Hernández FJ, Mangano K, Muñoz-Valle JF. A review: Antibody-dependent enhancement in COVID-19: The not so friendly side of antibodies. Int J Immunopathol Pharmacol 2021; 35:20587384211050199. [PMID: 34632844 PMCID: PMC8512237 DOI: 10.1177/20587384211050199] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 09/14/2021] [Indexed: 12/23/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), represents an unprecedented global public health emergency with economic and social consequences. One of the main concerns in the development of vaccines is the antibody-dependent enhancement phenomenon, better known as ADE. In this review, we provide an overview of SARS-CoV-2 infection as well as the immune response generated by the host. On the bases of this principle, we also describe what is known about the ADE phenomenon in various viral infections and its possible role as a limiting factor in the development of new vaccines and therapeutic strategies.
Collapse
Affiliation(s)
- Gabriela Athziri Sánchez-Zuno
- Instituto de Investigación en
Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de
Guadalajara, Guadalajara, México
| | - Mónica Guadalupe Matuz-Flores
- Instituto de Investigación en
Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de
Guadalajara, Guadalajara, México
| | - Guillermo González-Estevez
- Instituto de Investigación en
Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de
Guadalajara, Guadalajara, México
| | - Ferdinando Nicoletti
- Department of Biomedical and
Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Katia Mangano
- Department of Biomedical and
Biotechnological Sciences, University of Catania, Catania, Italy
| | - José Francisco Muñoz-Valle
- Instituto de Investigación en
Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de
Guadalajara, Guadalajara, México
| |
Collapse
|
243
|
Artificial intelligence predicts the immunogenic landscape of SARS-CoV-2 leading to universal blueprints for vaccine designs. Sci Rep 2020; 10:22375. [PMID: 33361777 PMCID: PMC7758335 DOI: 10.1038/s41598-020-78758-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/30/2020] [Indexed: 02/08/2023] Open
Abstract
The global population is at present suffering from a pandemic of Coronavirus disease 2019 (COVID-19), caused by the novel coronavirus Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). The goal of this study was to use artificial intelligence (AI) to predict blueprints for designing universal vaccines against SARS-CoV-2, that contain a sufficiently broad repertoire of T-cell epitopes capable of providing coverage and protection across the global population. To help achieve these aims, we profiled the entire SARS-CoV-2 proteome across the most frequent 100 HLA-A, HLA-B and HLA-DR alleles in the human population, using host-infected cell surface antigen presentation and immunogenicity predictors from the NEC Immune Profiler suite of tools, and generated comprehensive epitope maps. We then used these epitope maps as input for a Monte Carlo simulation designed to identify statistically significant “epitope hotspot” regions in the virus that are most likely to be immunogenic across a broad spectrum of HLA types. We then removed epitope hotspots that shared significant homology with proteins in the human proteome to reduce the chance of inducing off-target autoimmune responses. We also analyzed the antigen presentation and immunogenic landscape of all the nonsynonymous mutations across 3,400 different sequences of the virus, to identify a trend whereby SARS-COV-2 mutations are predicted to have reduced potential to be presented by host-infected cells, and consequently detected by the host immune system. A sequence conservation analysis then removed epitope hotspots that occurred in less-conserved regions of the viral proteome. Finally, we used a database of the HLA haplotypes of approximately 22,000 individuals to develop a “digital twin” type simulation to model how effective different combinations of hotspots would work in a diverse human population; the approach identified an optimal constellation of epitope hotspots that could provide maximum coverage in the global population. By combining the antigen presentation to the infected-host cell surface and immunogenicity predictions of the NEC Immune Profiler with a robust Monte Carlo and digital twin simulation, we have profiled the entire SARS-CoV-2 proteome and identified a subset of epitope hotspots that could be harnessed in a vaccine formulation to provide a broad coverage across the global population.
Collapse
|
244
|
Malone B, Simovski B, Moliné C, Cheng J, Gheorghe M, Fontenelle H, Vardaxis I, Tennøe S, Malmberg JA, Stratford R, Clancy T. Artificial intelligence predicts the immunogenic landscape of SARS-CoV-2 leading to universal blueprints for vaccine designs. Sci Rep 2020; 10:22375. [PMID: 33361777 DOI: 10.1101/2020.04.21.052084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/30/2020] [Indexed: 05/23/2023] Open
Abstract
The global population is at present suffering from a pandemic of Coronavirus disease 2019 (COVID-19), caused by the novel coronavirus Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). The goal of this study was to use artificial intelligence (AI) to predict blueprints for designing universal vaccines against SARS-CoV-2, that contain a sufficiently broad repertoire of T-cell epitopes capable of providing coverage and protection across the global population. To help achieve these aims, we profiled the entire SARS-CoV-2 proteome across the most frequent 100 HLA-A, HLA-B and HLA-DR alleles in the human population, using host-infected cell surface antigen presentation and immunogenicity predictors from the NEC Immune Profiler suite of tools, and generated comprehensive epitope maps. We then used these epitope maps as input for a Monte Carlo simulation designed to identify statistically significant "epitope hotspot" regions in the virus that are most likely to be immunogenic across a broad spectrum of HLA types. We then removed epitope hotspots that shared significant homology with proteins in the human proteome to reduce the chance of inducing off-target autoimmune responses. We also analyzed the antigen presentation and immunogenic landscape of all the nonsynonymous mutations across 3,400 different sequences of the virus, to identify a trend whereby SARS-COV-2 mutations are predicted to have reduced potential to be presented by host-infected cells, and consequently detected by the host immune system. A sequence conservation analysis then removed epitope hotspots that occurred in less-conserved regions of the viral proteome. Finally, we used a database of the HLA haplotypes of approximately 22,000 individuals to develop a "digital twin" type simulation to model how effective different combinations of hotspots would work in a diverse human population; the approach identified an optimal constellation of epitope hotspots that could provide maximum coverage in the global population. By combining the antigen presentation to the infected-host cell surface and immunogenicity predictions of the NEC Immune Profiler with a robust Monte Carlo and digital twin simulation, we have profiled the entire SARS-CoV-2 proteome and identified a subset of epitope hotspots that could be harnessed in a vaccine formulation to provide a broad coverage across the global population.
Collapse
Affiliation(s)
- Brandon Malone
- NEC Laboratories Europe GmbH, Kurfuersten-Anlage 36, 69115, Heidelberg, Germany
| | - Boris Simovski
- NEC OncoImmunity AS, Ullernchausseen 64/66, 0379, Oslo, Norway
| | - Clément Moliné
- NEC OncoImmunity AS, Ullernchausseen 64/66, 0379, Oslo, Norway
| | - Jun Cheng
- NEC Laboratories Europe GmbH, Kurfuersten-Anlage 36, 69115, Heidelberg, Germany
| | - Marius Gheorghe
- NEC OncoImmunity AS, Ullernchausseen 64/66, 0379, Oslo, Norway
| | | | | | - Simen Tennøe
- NEC OncoImmunity AS, Ullernchausseen 64/66, 0379, Oslo, Norway
| | | | | | - Trevor Clancy
- NEC OncoImmunity AS, Ullernchausseen 64/66, 0379, Oslo, Norway.
| |
Collapse
|
245
|
Lucas C, Klein J, Sundaram M, Liu F, Wong P, Silva J, Mao T, Oh JE, Tokuyama M, Lu P, Venkataraman A, Park A, Israelow B, Wyllie AL, Vogels CBF, Muenker MC, Casanovas-Massana A, Schulz WL, Zell J, Campbell M, Fournier JB, Grubaugh ND, Farhadian S, Wisnewski AV, Cruz CD, Omer S, Ko AI, Ring A, Iwasaki A. Kinetics of antibody responses dictate COVID-19 outcome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.12.18.20248331. [PMID: 33398304 DOI: 10.1101/2020.06.13.20130252] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Recent studies have provided insights into innate and adaptive immune dynamics in coronavirus disease 2019 (COVID-19). Yet, the exact feature of antibody responses that governs COVID-19 disease outcomes remain unclear. Here, we analysed humoral immune responses in 209 asymptomatic, mild, moderate and severe COVID-19 patients over time to probe the nature of antibody responses in disease severity and mortality. We observed a correlation between anti-Spike (S) IgG levels, length of hospitalization and clinical parameters associated with worse clinical progression. While high anti-S IgG levels correlated with worse disease severity, such correlation was time-dependent. Deceased patients did not have higher overall humoral response than live discharged patients. However, they mounted a robust, yet delayed response, measured by anti-S, anti-RBD IgG, and neutralizing antibody (NAb) levels, compared to survivors. Delayed seroconversion kinetics correlated with impaired viral control in deceased patients. Finally, while sera from 89% of patients displayed some neutralization capacity during their disease course, NAb generation prior to 14 days of disease onset emerged as a key factor for recovery. These data indicate that COVID-19 mortality does not correlate with the cross-sectional antiviral antibody levels per se , but rather with the delayed kinetics of NAb production.
Collapse
|
246
|
Galipeau Y, Greig M, Liu G, Driedger M, Langlois MA. Humoral Responses and Serological Assays in SARS-CoV-2 Infections. Front Immunol 2020; 11:610688. [PMID: 33391281 PMCID: PMC7775512 DOI: 10.3389/fimmu.2020.610688] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
In December 2019, the novel betacoronavirus Severe Acute Respiratory Disease Coronavirus 2 (SARS-CoV-2) was first detected in Wuhan, China. SARS-CoV-2 has since become a pandemic virus resulting in hundreds of thousands of deaths and deep socioeconomic implications worldwide. In recent months, efforts have been directed towards detecting, tracking, and better understanding human humoral responses to SARS-CoV-2 infection. It has become critical to develop robust and reliable serological assays to characterize the abundance, neutralization efficiency, and duration of antibodies in virus-exposed individuals. Here we review the latest knowledge on humoral immune responses to SARS-CoV-2 infection, along with the benefits and limitations of currently available commercial and laboratory-based serological assays. We also highlight important serological considerations, such as antibody expression levels, stability and neutralization dynamics, as well as cross-reactivity and possible immunological back-boosting by seasonal coronaviruses. The ability to accurately detect, measure and characterize the various antibodies specific to SARS-CoV-2 is necessary for vaccine development, manage risk and exposure for healthcare and at-risk workers, and for monitoring reinfections with genetic variants and new strains of the virus. Having a thorough understanding of the benefits and cautions of standardized serological testing at a community level remains critically important in the design and implementation of future vaccination campaigns, epidemiological models of immunity, and public health measures that rely heavily on up-to-date knowledge of transmission dynamics.
Collapse
Affiliation(s)
- Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew Greig
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - George Liu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | | | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- uOttawa Center for Infection, Immunity and Inflammation (CI3), Ottawa, ON, Canada
| |
Collapse
|
247
|
Harvala H, Robb ML, Watkins N, Ijaz S, Dicks S, Patel M, Supasa P, Wanwisa D, Liu C, Mongkolsapaya J, Bown A, Bailey D, Vipond R, Grayson N, Temperton N, Gupta S, Ploeg RJ, Bolton J, Fyfe A, Gopal R, Simmonds P, Screaton G, Thompson C, Brooks T, Zambon M, Miflin G, Roberts DJ. Convalescent plasma therapy for the treatment of patients with COVID-19: Assessment of methods available for antibody detection and their correlation with neutralising antibody levels. Transfus Med 2020; 31:167-175. [PMID: 33333627 PMCID: PMC8246874 DOI: 10.1111/tme.12746] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The lack of approved specific therapeutic agents to treat coronavirus disease (COVID-19) associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has led to the rapid implementation of convalescent plasma therapy (CPT) trials in many countries, including the United Kingdom. Effective CPT is likely to require high titres of neutralising antibody (nAb) in convalescent donations. Understanding the relationship between functional neutralising antibodies and antibody levels to specific SARS-CoV-2 proteins in scalable assays will be crucial for the success of a large-scale collection. We assessed whether neutralising antibody titres correlated with reactivity in a range of enzyme-linked immunosorbent assays (ELISA) targeting the spike (S) protein, the main target for human immune response. METHODS Blood samples were collected from 52 individuals with a previous laboratory-confirmed SARS-CoV-2 infection. These were assayed for SARS-CoV-2 nAbs by microneutralisation and pseudo-type assays and for antibodies by four different ELISAs. Receiver operating characteristic (ROC) analysis was used to further identify sensitivity and specificity of selected assays to identify samples containing high nAb levels. RESULTS All samples contained SARS-CoV-2 antibodies, whereas neutralising antibody titres of greater than 1:20 were detected in 43 samples (83% of those tested) and >1:100 in 22 samples (42%). The best correlations were observed with EUROimmun immunoglobulin G (IgG) reactivity (Spearman Rho correlation coefficient 0.88; p < 0.001). Based on ROC analysis, EUROimmun would detect 60% of samples with titres of >1:100 with 100% specificity using a reactivity index of 9.1 (13/22). DISCUSSION Robust associations between nAb titres and reactivity in several ELISA-based antibody tests demonstrate their possible utility for scaled-up production of convalescent plasma containing potentially therapeutic levels of anti-SARS-CoV-2 nAbs.
Collapse
Affiliation(s)
- Heli Harvala
- National Microbiology Services, NHS Blood and Transplant, London, UK
| | - Matthew L Robb
- Statistics and Clinical Studies, NHS Blood and Transplant, Bristol, UK
| | - Nick Watkins
- Department of Research and Development, NHS Blood and Transplant Cambridge, Cambridge, UK
| | - Samreen Ijaz
- Virology Reference Department, National Infection Service, Public Health England, London, UK
| | - Steven Dicks
- Virology Reference Department, National Infection Service, Public Health England, London, UK
| | - Monika Patel
- High Containment Microbiology, National Infection Service, Public Health England, London, UK
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Dejnirattisai Wanwisa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Chang Liu
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Juthathip Mongkolsapaya
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Abbie Bown
- Rare and Imported Pathogens Laboratory, Public Health England, Porton Down, Wiltshire, UK
| | - Daniel Bailey
- Rare and Imported Pathogens Laboratory, Public Health England, Porton Down, Wiltshire, UK
| | - Richard Vipond
- Rare and Imported Pathogens Laboratory, Public Health England, Porton Down, Wiltshire, UK
| | - Nicholas Grayson
- Department of Paediatric Medicine, University of Oxford, University of Oxford, Oxford, UK
| | | | - Sunetra Gupta
- Department of Zoology, University of Oxford, Oxford, UK
| | - Rutger J Ploeg
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Department of Transplant Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jai Bolton
- Department of Zoology, University of Oxford, Oxford, UK
| | - Alex Fyfe
- Department of Zoology, University of Oxford, Oxford, UK
| | - Robin Gopal
- Virology Reference Department, National Infection Service, Public Health England, London, UK
| | - Peter Simmonds
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Gavin Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Tim Brooks
- Rare and Imported Pathogens Laboratory, Public Health England, Porton Down, Wiltshire, UK
| | - Maria Zambon
- Virology Reference Department, National Infection Service, Public Health England, London, UK
| | - Gail Miflin
- Department of Chief Medical Officer, NHS Blood and Transplant, Bristol, UK
| | - David J Roberts
- NHS Blood and Transplant, Oxford, John Radcliffe Hospital, Oxford, UK.,Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
248
|
Ku MW, Bourgine M, Authié P, Lopez J, Nemirov K, Moncoq F, Noirat A, Vesin B, Nevo F, Blanc C, Souque P, Tabbal H, Simon E, Hardy D, Le Dudal M, Guinet F, Fiette L, Mouquet H, Anna F, Martin A, Escriou N, Majlessi L, Charneau P. Intranasal vaccination with a lentiviral vector protects against SARS-CoV-2 in preclinical animal models. Cell Host Microbe 2020; 29:236-249.e6. [PMID: 33357418 PMCID: PMC7738935 DOI: 10.1016/j.chom.2020.12.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/26/2020] [Accepted: 12/09/2020] [Indexed: 11/25/2022]
Abstract
To develop a vaccine candidate against coronavirus disease 2019 (COVID-19), we generated a lentiviral vector (LV) eliciting neutralizing antibodies against the Spike glycoprotein of SARS-CoV-2. Systemic vaccination by this vector in mice, in which the expression of the SARS-CoV-2 receptor hACE2 has been induced by transduction of respiratory tract cells by an adenoviral vector, confers only partial protection despite high levels of serum neutralizing activity. However, eliciting an immune response in the respiratory tract through an intranasal boost results in a >3 log10 decrease in the lung viral loads and reduces local inflammation. Moreover, both integrative and non-integrative LV platforms display strong vaccine efficacy and inhibit lung deleterious injury in golden hamsters, which are naturally permissive to SARS-CoV-2 replication and closely mirror human COVID-19 physiopathology. Our results provide evidence of marked prophylactic effects of LV-based vaccination against SARS-CoV-2 and designate intranasal immunization as a powerful approach against COVID-19.
Collapse
Affiliation(s)
- Min-Wen Ku
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Maryline Bourgine
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France; Molecular Virology and Vaccinology Unit, Virology Department, Institut Pasteur, Paris 75015, France
| | - Pierre Authié
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Jodie Lopez
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Kirill Nemirov
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Fanny Moncoq
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Amandine Noirat
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Benjamin Vesin
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Fabien Nevo
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Catherine Blanc
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Philippe Souque
- Molecular Virology and Vaccinology Unit, Virology Department, Institut Pasteur, Paris 75015, France
| | - Houda Tabbal
- Molecular Genetics of RNA Viruses Unit, Virology Department, Institut Pasteur, CNRS UMR3569, Université de Paris, Paris 75015, France
| | - Emeline Simon
- Molecular Genetics of RNA Viruses Unit, Virology Department, Institut Pasteur, CNRS UMR3569, Université de Paris, Paris 75015, France; Université de Paris, Paris 75006, France
| | - David Hardy
- Experimental Neuropathology Unit, Global Health Department, Institut Pasteur, Paris 75015, France
| | | | - Françoise Guinet
- Lymphocytes and Immunity Unit, Immunology Department, Institut Pasteur, Paris 75015, France
| | | | - Hugo Mouquet
- Laboratory of Humoral Immunology, Immunology Department, Institut Pasteur, INSERM U1222, Paris, France
| | - François Anna
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Annette Martin
- Molecular Genetics of RNA Viruses Unit, Virology Department, Institut Pasteur, CNRS UMR3569, Université de Paris, Paris 75015, France
| | - Nicolas Escriou
- Innovation Lab, Vaccines, Virology Department, Institut Pasteur, Paris 75015, France
| | - Laleh Majlessi
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France.
| | - Pierre Charneau
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France; Molecular Virology and Vaccinology Unit, Virology Department, Institut Pasteur, Paris 75015, France.
| |
Collapse
|
249
|
Carsetti R, Zaffina S, Piano Mortari E, Terreri S, Corrente F, Capponi C, Palomba P, Mirabella M, Cascioli S, Palange P, Cuccaro I, Milito C, Zumla A, Maeurer M, Camisa V, Vinci MR, Santoro A, Cimini E, Marchioni L, Nicastri E, Palmieri F, Agrati C, Ippolito G, Porzio O, Concato C, Onetti Muda A, Raponi M, Quintarelli C, Quinti I, Locatelli F. Different Innate and Adaptive Immune Responses to SARS-CoV-2 Infection of Asymptomatic, Mild, and Severe Cases. Front Immunol 2020; 11:610300. [PMID: 33391280 PMCID: PMC7772470 DOI: 10.3389/fimmu.2020.610300] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a novel coronavirus, not encountered before by humans. The wide spectrum of clinical expression of SARS-CoV-2 illness suggests that individual immune responses to SARS-CoV-2 play a crucial role in determining the clinical course after first infection. Immunological studies have focused on patients with moderate to severe disease, demonstrating excessive inflammation in tissues and organ damage. In order to understand the basis of the protective immune response in COVID-19, we performed a longitudinal follow-up, flow-cytometric and serological analysis of innate and adaptive immunity in 64 adults with a spectrum of clinical presentations: 28 healthy SARS-CoV-2-negative contacts of COVID-19 cases; 20 asymptomatic SARS-CoV-2-infected cases; eight patients with Mild COVID-19 disease and eight cases of Severe COVID-19 disease. Our data show that high frequency of NK cells and early and transient increase of specific IgA, IgM and, to a lower extent, IgG are associated with asymptomatic SARS-CoV-2 infection. By contrast, monocyte expansion and high and persistent levels of IgA and IgG, produced relatively late in the course of the infection, characterize severe disease. Modest increase of monocytes and different kinetics of antibodies are detected in mild COVID-19. The importance of innate NK cells and the short-lived antibody response of asymptomatic individuals and patients with mild disease suggest that only severe COVID-19 may result in protective memory established by the adaptive immune response.
Collapse
Affiliation(s)
- Rita Carsetti
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Salvatore Zaffina
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Eva Piano Mortari
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Sara Terreri
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Francesco Corrente
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Claudia Capponi
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Patrizia Palomba
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Mattia Mirabella
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Simona Cascioli
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paolo Palange
- Department of Public Health and Infectious Diseases Pulmonary Division, Policlinico Umberto I Hospital, Rome, Italy
| | - Ilaria Cuccaro
- Department of Public Health and Infectious Diseases Pulmonary Division, Policlinico Umberto I Hospital, Rome, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alimuddin Zumla
- Center for Clinical Microbiology, Division of Infection and Immunity, University College London, London, United Kingdom
- NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, United Kingdom
| | - Markus Maeurer
- Immunotherapy Programme, Champalimaud Foundation, Lisbon, Portugal
- Med Clinic, University of Mainz, Mainz, Germany
| | - Vincenzo Camisa
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Maria Rosaria Vinci
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Annapaola Santoro
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology Laboratory, INMI L Spallanzani, IRCCS, Rome, Italy
| | | | | | | | - Chiara Agrati
- Cellular Immunology Laboratory, INMI L Spallanzani, IRCCS, Rome, Italy
| | | | - Ottavia Porzio
- Medical Laboratory Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Concato
- Virology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Andrea Onetti Muda
- Department of Laboratories, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Massimiliano Raponi
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Concetta Quintarelli
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Pediatrics, Sapienza, University of Rome, Rome, Italy
| |
Collapse
|
250
|
Ma X, Zou F, Yu F, Li R, Yuan Y, Zhang Y, Zhang X, Deng J, Chen T, Song Z, Qiao Y, Zhan Y, Liu J, Zhang J, Zhang X, Peng Z, Li Y, Lin Y, Liang L, Wang G, Chen Y, Chen Q, Pan T, He X, Zhang H. Nanoparticle Vaccines Based on the Receptor Binding Domain (RBD) and Heptad Repeat (HR) of SARS-CoV-2 Elicit Robust Protective Immune Responses. Immunity 2020; 53:1315-1330.e9. [PMID: 33275896 PMCID: PMC7687490 DOI: 10.1016/j.immuni.2020.11.015] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
Various vaccine strategies have been proposed in response to the global COVID-19 pandemic, each with unique strategies for eliciting immune responses. Here, we developed nanoparticle vaccines by covalently conjugating the self-assembled 24-mer ferritin to the receptor binding domain (RBD) and/or heptad repeat (HR) subunits of the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) spike (S) protein. Compared to monomer vaccines, nanoparticle vaccines elicited more robust neutralizing antibodies and cellular immune responses. RBD and RBD-HR nanoparticle vaccinated hACE2 transgenic mice vaccinated with RBD and/or RBD-HR nanoparticles exhibited reduced viral load in the lungs after SARS-CoV-2 challenge. RBD-HR nanoparticle vaccines also promoted neutralizing antibodies and cellular immune responses against other coronaviruses. The nanoparticle vaccination of rhesus macaques induced neutralizing antibodies, and T and B cell responses prior to boost immunization; these responses persisted for more than three months. RBD- and HR-based nanoparticles thus present a promising vaccination approach against SARS-CoV-2 and other coronaviruses.
Collapse
Affiliation(s)
- Xiancai Ma
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Fan Zou
- Qianyang Biomedical Research Institute, Guangzhou, Guangdong, 510063, China
| | - Fei Yu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Rong Li
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yaochang Yuan
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yiwen Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiantao Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Jieyi Deng
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Tao Chen
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zheng Song
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yidan Qiao
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yikang Zhan
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Jun Liu
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Qianyang Biomedical Research Institute, Guangzhou, Guangdong, 510063, China
| | - Junsong Zhang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Xu Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhilin Peng
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yuzhuang Li
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yingtong Lin
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Liting Liang
- Qianyang Biomedical Research Institute, Guangzhou, Guangdong, 510063, China
| | - Guanwen Wang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Yingshi Chen
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Qier Chen
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ting Pan
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Center for Infection and Immunity Study, School of Medicine, Sun Yat-sen University Shenzhen, Guangdong, 518107, China
| | - Xin He
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Hui Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Engineering Research Center of Gene Vaccine of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|