1
|
Rizatdinova SN, Ershova AE, Astrakhantseva IV. Pseudotyped Viruses: A Useful Platform for Pre-Clinical Studies Conducted in a BSL-2 Laboratory Setting. Biomolecules 2025; 15:135. [PMID: 39858529 PMCID: PMC11763035 DOI: 10.3390/biom15010135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The study of pathogenic viruses has always posed significant biosafety challenges. In particular, the study of highly pathogenic viruses requires methods with low biological risk but relatively high sensitivity and convenience in detection. In recent years, pseudoviruses, which consist of a backbone of one virus and envelope proteins of another virus, have become one of the most widely used tools for exploring the mechanisms of viruses binding to cells, membrane fusion and viral entry, as well as for screening the libraries of antiviral substances, evaluating the potential of neutralizing monoclonal antibodies, developing neutralization tests, and therapeutic platforms. During the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), pseudotyped virus-based assays played a pivotal role in advancing our understanding of virus-cell interactions and the role of its proteins in disease pathogenesis. Such tools facilitated the search for potential therapeutic agents and accelerated epidemiological studies on post-infection and post-vaccination humoral immunity. This review focuses on the use of pseudoviruses as a model for large-scale applications to study enveloped viruses.
Collapse
Affiliation(s)
| | | | - Irina V. Astrakhantseva
- Department of Immunobiology and Biomedicine, Sirius University of Science and Technology, 354349 Sirius, Krasnodarsky Krai, Russia; (S.N.R.); (A.E.E.)
| |
Collapse
|
2
|
Antonelli R, Forconi V, Molesti E, Semplici C, Piu P, Altamura M, Dapporto F, Temperton N, Montomoli E, Manenti A. A validated and standardized pseudotyped microneutralization assay as a safe and powerful tool to measure LASSA virus neutralising antibodies for vaccine development and comparison. F1000Res 2024; 13:534. [PMID: 39512237 PMCID: PMC11541077 DOI: 10.12688/f1000research.149578.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 11/15/2024] Open
Abstract
Background Over the past few decades, World Health Organization (WHO) has made massive efforts to promote the development of a vaccine against Lassa virus (LASV), one of the top ten priority pathogens for research and development under the WHO R&D Blueprint for Emerging Infections. To date, several vaccines are at different stages of development. In this scenario, a validated and standardised assay to measure LASV neutralising antibodies is urgently needed for vaccine development and comparison. Methods The neutralisation assay remains the gold standard for determining antibody efficacy. Here we have proposed a safe and validated pseudotyped neutralisation assay for LASV, taking advantage of the development of the first WHO International Standard and Reference Panel for Anti-Lassa Fever (NIBSC code 21/332). Results and Conclusions The proposed results demonstrate that the pseudotyped luciferase neutralisation assay is a specific serological test for the measurement of LASV neutralising antibodies without cross-reacting with standard sera specific for heterologous viral infections. In addition, the assay is accurate, precise, and linear according to criteria and statistical analyses defined and accepted by international guidelines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy,, University of Kent and Greenwich at Medway, Chatham, Kent, UK
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | |
Collapse
|
3
|
Thimmiraju SR, Villar MJ, Kimata JT, Strych U, Bottazzi ME, Hotez PJ, Pollet J. Optimization of Cellular Transduction by the HIV-Based Pseudovirus Platform with Pan-Coronavirus Spike Proteins. Viruses 2024; 16:1492. [PMID: 39339968 PMCID: PMC11437443 DOI: 10.3390/v16091492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past three years, new SARS-CoV-2 variants have continuously emerged, evolving to a point where an immune response against the original vaccine no longer provided optimal protection against these new strains. During this time, high-throughput neutralization assays based on pseudoviruses have become a valuable tool for assessing the efficacy of new vaccines, screening updated vaccine candidates against emerging variants, and testing the efficacy of new therapeutics such as monoclonal antibodies. Lentiviral vectors derived from HIV-1 are popular for developing pseudo and chimeric viruses due to their ease of use, stability, and long-term transgene expression. However, the HIV-based platform has lower transduction rates for pseudotyping coronavirus spike proteins than other pseudovirus platforms, necessitating more optimized methods. As the SARS-CoV-2 virus evolved, we produced over 18 variants of the spike protein for pseudotyping with an HIV-based vector, optimizing experimental parameters for their production and transduction. In this article, we present key parameters that were assessed to improve such technology, including (a) the timing and method of collection of pseudovirus supernatant; (b) the timing of host cell transduction; (c) cell culture media replenishment after pseudovirus adsorption; and (d) the centrifugation (spinoculation) parameters of the host cell+ pseudovirus mix, towards improved transduction. Additionally, we found that, for some pseudoviruses, the addition of a cationic polymer (polybrene) to the culture medium improved the transduction process. These findings were applicable across variant spike pseudoviruses that include not only SARS-CoV-2 variants, but also SARS, MERS, Alpha Coronavirus (NL-63), and bat-like coronaviruses. In summary, we present improvements in transduction efficiency, which can broaden the dynamic range of the pseudovirus titration and neutralization assays.
Collapse
Affiliation(s)
- Syamala Rani Thimmiraju
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maria Jose Villar
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Ulrich Strych
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jeroen Pollet
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
4
|
Elliott S, Olufemi OT, Daly JM. Systematic Review of Equine Influenza A Virus Vaccine Studies and Meta-Analysis of Vaccine Efficacy. Viruses 2023; 15:2337. [PMID: 38140577 PMCID: PMC10747572 DOI: 10.3390/v15122337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Vaccines against equine influenza have been available since the late 1960s, but outbreaks continue to occur periodically, affecting both vaccinated and unvaccinated animals. The aim of this study was to systematically evaluate the efficacy of vaccines against influenza A virus in horses (equine IAV). For this, PubMed, CAB abstracts, and Web of Science were searched for controlled trials of equine IAV vaccines published up to December 2020. Forty-three articles reporting equine IAV vaccination and challenge studies in previously naïve equids using an appropriate comparison group were included in a qualitative analysis of vaccine efficacy. A value for vaccine efficacy (VE) was calculated as the percentage reduction in nasopharyngeal virus shedding detected by virus isolation in embryonated hens' eggs from 38 articles. Among 21 studies involving commercial vaccines, the mean VE was 50.03% (95% CI: 23.35-76.71%), ranging from 0 to 100%. Among 17 studies reporting the use of experimental vaccines, the mean VE was 40.37% (95% CI: 19.64-62.44), and the range was again 0-100%. Overall, complete protection from virus shedding was achieved in five studies. In conclusion, although commercially available vaccines can, in some circumstances, offer complete protection from infection, the requirement for frequent vaccination in the field to limit virus shedding and hence transmission is apparent. Although most studies were conducted by a few centres, a lack of consistent study design made comparisons difficult.
Collapse
Affiliation(s)
| | | | - Janet M. Daly
- One Virology, Wolfson Centre for Global Virus Research, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington LE12 5RD, UK
| |
Collapse
|
5
|
Cantoni D, Wilkie C, Bentley EM, Mayora-Neto M, Wright E, Scott S, Ray S, Castillo-Olivares J, Heeney JL, Mattiuzzo G, Temperton NJ. Correlation between pseudotyped virus and authentic virus neutralisation assays, a systematic review and meta-analysis of the literature. Front Immunol 2023; 14:1184362. [PMID: 37790941 PMCID: PMC10544934 DOI: 10.3389/fimmu.2023.1184362] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Background The virus neutralization assay is a principal method to assess the efficacy of antibodies in blocking viral entry. Due to biosafety handling requirements of viruses classified as hazard group 3 or 4, pseudotyped viruses can be used as a safer alternative. However, it is often queried how well the results derived from pseudotyped viruses correlate with authentic virus. This systematic review and meta-analysis was designed to comprehensively evaluate the correlation between the two assays. Methods Using PubMed and Google Scholar, reports that incorporated neutralisation assays with both pseudotyped virus, authentic virus, and the application of a mathematical formula to assess the relationship between the results, were selected for review. Our searches identified 67 reports, of which 22 underwent a three-level meta-analysis. Results The three-level meta-analysis revealed a high level of correlation between pseudotyped viruses and authentic viruses when used in an neutralisation assay. Reports that were not included in the meta-analysis also showed a high degree of correlation, with the exception of lentiviral-based pseudotyped Ebola viruses. Conclusion Pseudotyped viruses identified in this report can be used as a surrogate for authentic virus, though care must be taken in considering which pseudotype core to use when generating new uncharacterised pseudotyped viruses.
Collapse
Affiliation(s)
- Diego Cantoni
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - Craig Wilkie
- School of Mathematics & Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Emma M. Bentley
- Medicines and Healthcare Products Regulatory Agency, South Mimms, United Kingdom
| | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| | - Edward Wright
- Viral Pseudotype Unit, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Simon Scott
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| | - Surajit Ray
- School of Mathematics & Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Cambridge, United Kingdom
| | - Jonathan Luke Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Cambridge, United Kingdom
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Giada Mattiuzzo
- Medicines and Healthcare Products Regulatory Agency, South Mimms, United Kingdom
| | - Nigel James Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| |
Collapse
|
6
|
Beirag N, Varghese PM, Neto MM, Al Aiyan A, Khan HA, Qablan M, Shamji MH, Sim RB, Temperton N, Kishore U. Complement Activation-Independent Attenuation of SARS-CoV-2 Infection by C1q and C4b-Binding Protein. Viruses 2023; 15:1269. [PMID: 37376569 DOI: 10.3390/v15061269] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
The complement system is a key component of the innate immune response to viruses and proinflammatory events. Exaggerated complement activation has been attributed to the induction of a cytokine storm in severe SARS-CoV-2 infection. However, there is also an argument for the protective role of complement proteins, given their local synthesis or activation at the site of viral infection. This study investigated the complement activation-independent role of C1q and C4b-binding protein (C4BP) against SARS-CoV-2 infection. The interactions of C1q, its recombinant globular heads, and C4BP with the SARS-CoV-2 spike and receptor binding domain (RBD) were examined using direct ELISA. In addition, RT-qPCR was used to evaluate the modulatory effect of these complement proteins on the SARS-CoV-2-mediated immune response. Cell binding and luciferase-based viral entry assays were utilised to assess the effects of C1q, its recombinant globular heads, and C4BP on SARS-CoV-2 cell entry. C1q and C4BP bound directly to SARS-CoV-2 pseudotype particles via the RBD domain of the spike protein. C1q via its globular heads and C4BP were found to reduce binding as well as viral transduction of SARS-CoV-2 spike protein expressing lentiviral pseudotypes into transfected A549 cells expressing human ACE2 and TMPRSS2. Furthermore, the treatment of the SARS-CoV-2 spike, envelope, nucleoprotein, and membrane protein expressing alphaviral pseudotypes with C1q, its recombinant globular heads, or C4BP triggered a reduction in mRNA levels of proinflammatory cytokines and chemokines such as IL-1β, IL-8, IL-6, TNF-α, IFN-α, and RANTES (as well as NF-κB) in A549 cells expressing human ACE2 and TMPRSS2. In addition, C1q and C4BP treatment also reduced SARS-CoV-2 pseudotype infection-mediated NF-κB activation in A549 cells expressing human ACE2 and TMPRSS2. C1q and C4BP are synthesised primarily by hepatocytes; however, they are also produced by macrophages, and alveolar type II cells, respectively, locally at the pulmonary site. These findings support the notion that the locally produced C1q and C4BP can be protective against SARS-CoV-2 infection in a complement activation-independent manner, offering immune resistance by inhibiting virus binding to target host cells and attenuating the infection-associated inflammatory response.
Collapse
Affiliation(s)
- Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Praveen M Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Martin Mayora Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, Kent ME4 4TB, UK
| | - Ahmad Al Aiyan
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Haseeb A Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 4545, Saudi Arabia
| | - Moneeb Qablan
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Department of Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Robert B Sim
- MRC Immunochemistry Unit, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, Kent ME4 4TB, UK
| | - Uday Kishore
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
7
|
Ravi G, Eerike M, Konda VR, Bisoi D, Raj GM, Priyadarshini R, Mali KR, Chaliserry LF. Efficacy and Safety of Anti-SARS-CoV-2 Monoclonal Antibodies: An Updated Review. Monoclon Antib Immunodiagn Immunother 2023; 42:77-94. [PMID: 37129306 DOI: 10.1089/mab.2022.0036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Monoclonal antibodies (mAbs) had received emergency use authorization for mild-to-moderate coronavirus disease 2019 (COVID-19) or for prophylaxis against COVID-19, including casirivimab plus imdevimab (C+I), bamlanivimab plus etesevimab (B+E), tixagevimab plus cilgavimab (T+CG), and sotrovimab (S) and bebtelovimab (BEB). This systematic review was done to assess the efficacy and safety of the same. PubMed, Embase, Scopus, medRxiv, bioRxiv, and FDA fact sheets were searched for the studies published between January 2021 and May 2022, and appropriate search terms related to the mentioned mAbs were used for data collection. Review included original research including randomized clinical trials and observational studies published or preprints. Studies included in the review had compared with placebo or standard of care or no treatment or mAbs with each other and also of various doses. Data extraction was done and reviewed the same for both efficacy and safety. Total of 20 studies were included in this review. The rate of hospitalization within 30 days showed ∼2% in comparison to ∼7% with placebo. Significant reduction in viral load was more observed with combination mAbs. Combination therapy showed faster virological cure against the Gamma variant. With C + I as postexposure prophylaxis (PEP), 29.0% of asymptomatic participants developed symptomatic COVID-19. Pre-exposure prophylaxis with T+CG reduced the incidence of infection by 77%. Infusion-related reaction was the most common adverse event (AE). The neutralizing mAbs reduced hospitalization in mild-to-moderate patients with infusion-related reactions as common AE. The response was better in the seronegative patients. Most of these studies were conducted in unvaccinated individuals and against Alpha, Gamma, and Delta variants.
Collapse
Affiliation(s)
- Gandham Ravi
- Department of Pharmacology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Madhavi Eerike
- Department of Pharmacology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Venugopala Rao Konda
- Department of Pharmacology, TRR Institute of Medical Sciences, Patancheru, India
| | - Debasis Bisoi
- Department of Pharmacology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Gerard Marshall Raj
- Department of Pharmacology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Rekha Priyadarshini
- Department of Pharmacology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Kalpana Ramanna Mali
- Department of Pharmacology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Leo Francis Chaliserry
- Department of Pharmacology, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| |
Collapse
|
8
|
Mestiri S, Merhi M, Inchakalody VP, Taib N, Smatti MK, Ahmad F, Raza A, Ali FH, Hydrose S, Fernandes Q, Ansari AW, Sahir F, Al-Zaidan L, Jalis M, Ghoul M, Allahverdi N, Al Homsi MU, Uddin S, Jeremijenko AM, Nimir M, Abu-Raddad LJ, Abid FB, Zaqout A, Alfheid SR, Saqr HMH, Omrani AS, Hssain AA, Al Maslamani M, Yassine HM, Dermime S. Persistence of spike-specific immune responses in BNT162b2-vaccinated donors and generation of rapid ex-vivo T cells expansion protocol for adoptive immunotherapy: A pilot study. Front Immunol 2023; 14:1061255. [PMID: 36817441 PMCID: PMC9933868 DOI: 10.3389/fimmu.2023.1061255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION The BNT162b2 mRNA-based vaccine has shown high efficacy in preventing COVID-19 infection but there are limited data on the types and persistence of the humoral and T cell responses to such a vaccine. METHODS Here, we dissect the vaccine-induced humoral and cellular responses in a cohort of six healthy recipients of two doses of this vaccine. RESULTS AND DISCUSSION Overall, there was heterogeneity in the spike-specific humoral and cellular responses among vaccinated individuals. Interestingly, we demonstrated that anti-spike antibody levels detected by a novel simple automated assay (Jess) were strongly correlated (r=0.863, P<0.0001) with neutralizing activity; thus, providing a potential surrogate for neutralizing cell-based assays. The spike-specific T cell response was measured with a newly modified T-spot assay in which the high-homology peptide-sequences cross-reactive with other coronaviruses were removed. This response was induced in 4/6 participants after the first dose, and all six participants after the second dose, and remained detectable in 4/6 participants five months post-vaccination. We have also shown for the first time, that BNT162b2 vaccine enhanced T cell responses also against known human common viruses. In addition, we demonstrated the efficacy of a rapid ex-vivo T cell expansion protocol for spike-specific T cell expansion to be potentially used for adoptive-cell therapy in severe COVID-19, immunocompromised individuals, and other high-risk groups. There was a 9 to 13.7-fold increase in the number of expanded T cells with a significant increase of anti-spike specific response showing higher frequencies of both activation and cytotoxic markers. Interestingly, effector memory T cells were dominant in all four participants' CD8+ expanded memory T cells; CD4+ T cells were dominated by effector memory in 2/4 participants and by central memory in the remaining two participants. Moreover, we found that high frequencies of CD4+ terminally differentiated memory T cells were associated with a greater reduction of spike-specific activated CD4+ T cells. Finally, we showed that participants who had a CD4+ central memory T cell dominance expressed a high CD69 activation marker in the CD4+ activated T cells.
Collapse
Affiliation(s)
- Sarra Mestiri
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Varghese P. Inchakalody
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Nassiba Taib
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maria K. Smatti
- Qatar University Biomedical Research Center, Qatar University, Doha, Qatar
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Afsheen Raza
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Fatma H. Ali
- Qatar University Biomedical Research Center, Qatar University, Doha, Qatar
| | - Shereena Hydrose
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Queenie Fernandes
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- College of Medicine, Qatar University, Doha, Qatar
| | - Abdul W. Ansari
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Fairooz Sahir
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Lobna Al-Zaidan
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Munir Jalis
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Mokhtar Ghoul
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Niloofar Allahverdi
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Mohammed U. Al Homsi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Mai Nimir
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | - Laith J. Abu-Raddad
- Infectious Disease Epidemiology Group, Weill Cornell Medicine–Qatar, Cornell University, Qatar Foundation–Education City, Doha, Qatar
- World Health Organization Collaborating Centre for Disease Epidemiology Analytics on HIV/AIDS, Sexually Transmitted Infections, and Viral Hepatitis, Weill Cornell Medicine–Qatar, Cornell University, Qatar Foundation–Education City, Doha, Qatar
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Fatma Ben Abid
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed Zaqout
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | | | | | - Ali S. Omrani
- College of Medicine, Qatar University, Doha, Qatar
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | | | - Hadi M. Yassine
- Qatar University Biomedical Research Center, Qatar University, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
9
|
Balmert SC, Ghozloujeh ZG, Carey CD, Williams LH, Zhang J, Shahi P, Amer M, Sumpter TL, Erdos G, Korkmaz E, Falo LD. A microarray patch SARS-CoV-2 vaccine induces sustained antibody responses and polyfunctional cellular immunity. iScience 2022; 25:105045. [PMID: 36062075 PMCID: PMC9425707 DOI: 10.1016/j.isci.2022.105045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 04/19/2022] [Accepted: 08/25/2022] [Indexed: 12/01/2022] Open
Abstract
Sustainable global immunization campaigns against COVID-19 and other emerging infectious diseases require effective, broadly deployable vaccines. Here, we report a dissolvable microarray patch (MAP) SARS-CoV-2 vaccine that targets the immunoresponsive skin microenvironment, enabling efficacious needle-free immunization. Multicomponent MAPs delivering both SARS-CoV-2 S1 subunit antigen and the TLR3 agonist Poly(I:C) induce robust antibody and cellular immune responses systemically and in the respiratory mucosa. MAP vaccine-induced antibodies bind S1 and the SARS-CoV-2 receptor-binding domain, efficiently neutralize the virus, and persist at high levels for more than a year. The MAP platform reduces systemic toxicity of the delivered adjuvant and maintains vaccine stability without refrigeration. When applied to human skin, MAP vaccines activate skin-derived migratory antigen-presenting cells, supporting the feasibility of human translation. Ultimately, this shelf-stable MAP vaccine improves immunogenicity and safety compared to traditional intramuscular vaccines and offers an attractive alternative for global immunization efforts against a range of infectious pathogens.
Collapse
Affiliation(s)
- Stephen C. Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Li’an H. Williams
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jiying Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Preeti Shahi
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Maher Amer
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L. Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| | - Louis D. Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
10
|
Wells DA, Cantoni D, Mayora‐Neto M, Genova CD, Sampson A, Ferrari M, Carnell G, Nadesalingam A, Smith P, Chan A, Raddi G, Castillo‐Olivares J, Baxendale H, Temperton N, Heeney JL. Human seasonal coronavirus neutralization and COVID-19 severity. J Med Virol 2022; 94:4820-4829. [PMID: 35705514 PMCID: PMC9349487 DOI: 10.1002/jmv.27937] [Citation(s) in RCA: 6] [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: 09/28/2021] [Revised: 05/28/2022] [Accepted: 06/13/2022] [Indexed: 11/08/2022]
Abstract
The virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), responsible for the global coronavirus disease-2019 (COVID-19) pandemic, spread rapidly around the world causing high morbidity and mortality. However, there are four known, endemic seasonal coronaviruses in humans (HCoVs), and whether antibodies for these HCoVs play a role in severity of COVID-19 disease has generated a lot of interest. Of these seasonal viruses NL63 is of particular interest as it uses the same cell entry receptor as SARS-CoV-2. We use functional, neutralizing assays to investigate cross-reactive antibodies and their relationship with COVID-19 severity. We analyzed the neutralization of SARS-CoV-2, NL63, HKU1, and 229E in 38 COVID-19 patients and 62 healthcare workers, and a further 182 samples to specifically study the relationship between SARS-CoV-2 and NL63. We found that although HCoV neutralization was very common there was little evidence that these antibodies neutralized SARS-CoV-2. Despite no evidence in cross-neutralization, levels of NL63 neutralizing antibodies become elevated after exposure to SARS-CoV-2 through infection or following vaccination.
Collapse
Affiliation(s)
- David A. Wells
- Department of Veterinary Medicine, Lab of Viral ZoonoticsUniversity of CambridgeCambridgeUK
- DIOSynVaxUniversity of CambridgeCambridgeUK
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of PharmacyUniversity of KentMedwayUK
| | - Martin Mayora‐Neto
- Viral Pseudotype Unit, Medway School of PharmacyUniversity of KentMedwayUK
| | - Cecilia Di Genova
- Viral Pseudotype Unit, Medway School of PharmacyUniversity of KentMedwayUK
| | - Alexander Sampson
- Department of Veterinary Medicine, Lab of Viral ZoonoticsUniversity of CambridgeCambridgeUK
| | - Matteo Ferrari
- Department of Veterinary Medicine, Lab of Viral ZoonoticsUniversity of CambridgeCambridgeUK
- DIOSynVaxUniversity of CambridgeCambridgeUK
| | - George Carnell
- Department of Veterinary Medicine, Lab of Viral ZoonoticsUniversity of CambridgeCambridgeUK
| | - Angalee Nadesalingam
- Department of Veterinary Medicine, Lab of Viral ZoonoticsUniversity of CambridgeCambridgeUK
| | - Peter Smith
- Department of Veterinary Medicine, Lab of Viral ZoonoticsUniversity of CambridgeCambridgeUK
| | - Andrew Chan
- Department of Veterinary Medicine, Lab of Viral ZoonoticsUniversity of CambridgeCambridgeUK
| | | | | | | | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of PharmacyUniversity of KentMedwayUK
| | - Jonathan L. Heeney
- Department of Veterinary Medicine, Lab of Viral ZoonoticsUniversity of CambridgeCambridgeUK
- DIOSynVaxUniversity of CambridgeCambridgeUK
| |
Collapse
|
11
|
Mullender C, da Costa KAS, Alrubayyi A, Pett SL, Peppa D. SARS-CoV-2 immunity and vaccine strategies in people with HIV. OXFORD OPEN IMMUNOLOGY 2022; 3:iqac005. [PMID: 36846557 PMCID: PMC9452103 DOI: 10.1093/oxfimm/iqac005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/24/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022] Open
Abstract
Current severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccines, based on the ancestral Wuhan strain, were developed rapidly to meet the needs of a devastating global pandemic. People living with Human Immunodeficiency Virus (PLWH) have been designated as a priority group for SARS-CoV-2 vaccination in most regions and varying primary courses (two- or three-dose schedule) and additional boosters are recommended depending on current CD4+ T cell count and/or detectable HIV viraemia. From the current published data, licensed vaccines are safe for PLWH, and stimulate robust responses to vaccination in those well controlled on antiretroviral therapy and with high CD4+ T cell counts. Data on vaccine efficacy and immunogenicity remain, however, scarce in PLWH, especially in people with advanced disease. A greater concern is a potentially diminished immune response to the primary course and subsequent boosters, as well as an attenuated magnitude and durability of protective immune responses. A detailed understanding of the breadth and durability of humoral and T cell responses to vaccination, and the boosting effects of natural immunity to SARS-CoV-2, in more diverse populations of PLWH with a spectrum of HIV-related immunosuppression is therefore critical. This article summarizes focused studies of humoral and cellular responses to SARS-CoV-2 infection in PLWH and provides a comprehensive review of the emerging literature on SARS-CoV-2 vaccine responses. Emphasis is placed on the potential effect of HIV-related factors and presence of co-morbidities modulating responses to SARS-CoV-2 vaccination, and the remaining challenges informing the optimal vaccination strategy to elicit enduring responses against existing and emerging variants in PLWH.
Collapse
Affiliation(s)
- Claire Mullender
- Centre for Clinical Research in Infection and Sexual Health, Institute for Global Health, University College London Institute for Global Health, London, UK
| | - Kelly A S da Costa
- Division of Infection and Immunity, University College London, London, UK
| | - Aljawharah Alrubayyi
- Division of Infection and Immunity, University College London, London, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sarah L Pett
- Centre for Clinical Research in Infection and Sexual Health, Institute for Global Health, University College London Institute for Global Health, London, UK
- Medical Research Council Clinical Trials Unit, Institute of Clinical Trials and Methodology, London, UK
| | - Dimitra Peppa
- Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
12
|
Kostin NN, Bobik TV, Skryabin GA, Simonova MA, Knorre VD, Abrikosova VA, Mokrushina YA, Smirnov IV, Aleshenko NL, Kruglova NA, Mazurov DV, Nikitin AE, Gabibov AG. An ELISA Platform for the Quantitative Analysis of SARS-CoV-2 RBD-neutralizing Antibodies As an Alternative to Monitoring of the Virus-Neutralizing Activity. Acta Naturae 2022; 14:109-119. [PMID: 36348715 PMCID: PMC9611858 DOI: 10.32607/actanaturae.11776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
Monitoring of the level of the virus-neutralizing activity of serum immunoglobulins ensures that one can reliably assess the effectiveness of any protection against the SARS-CoV-2 infection. For SARS-CoV-2, the RBD-ACE2 neutralizing activity of sera is almost equivalent to the virus-neutralizing activity of their antibodies and can be used to assess the level of SARS-CoV-2 neutralizing antibodies. We are proposing an ELISA platform for performing a quantitative analysis of SARS-CoV-2 RBD-neutralizing antibodies, as an alternative to the monitoring of the virus-neutralizing activity using pseudovirus or "live" virus assays. The advantage of the developed platform is that it can be adapted to newly emerging virus variants in a very short time (1-2 weeks) and, thereby, provide quantitative data on the activity of SARS-CoV-2 RBD-neutralizing antibodies. The developed platform can be used to (1) study herd immunity to SARS-CoV-2, (2) monitor the effectiveness of the vaccination drive (revaccination) in a population, and (3) select potential donors of immune plasma. The protective properties of the humoral immune response in hospitalized patients and outpatients, as well as after prophylaxis with the two most popular SARS-CoV-2 vaccines in Russia, were studied in detail using this platform. The highest RBD-neutralizing activity was observed in the group of hospitalized patients. The protective effect in the group of individuals vaccinated with Gam-COVID-Vac vaccine was 25% higher than that in outpatients and almost four times higher than that in individuals vaccinated with the CoviVac vaccine.
Collapse
Affiliation(s)
- N. N. Kostin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - T. V. Bobik
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - G. A. Skryabin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - M. A. Simonova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - V. D. Knorre
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - V. A. Abrikosova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - Y. A. Mokrushina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - I. V. Smirnov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - N. L. Aleshenko
- Central Clinical Hospital of the Russian Academy of Sciences, Moscow, 117593 Russia
| | - N. A. Kruglova
- Institute of Gene Biology Russian Academy of Sciences, Moscow, 119334 Russia
| | - D. V. Mazurov
- Institute of Gene Biology Russian Academy of Sciences, Moscow, 119334 Russia
| | - A. E. Nikitin
- Central Clinical Hospital of the Russian Academy of Sciences, Moscow, 117593 Russia
| | - A. G. Gabibov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| |
Collapse
|
13
|
Homogeneous surrogate virus neutralization assay to rapidly assess neutralization activity of anti-SARS-CoV-2 antibodies. Nat Commun 2022; 13:3716. [PMID: 35778399 PMCID: PMC9249905 DOI: 10.1038/s41467-022-31300-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 06/13/2022] [Indexed: 12/23/2022] Open
Abstract
The COVID-19 pandemic triggered the development of numerous diagnostic tools to monitor infection and to determine immune response. Although assays to measure binding antibodies against SARS-CoV-2 are widely available, more specific tests measuring neutralization activities of antibodies are immediately needed to quantify the extent and duration of protection that results from infection or vaccination. We previously developed a 'Serological Assay based on a Tri-part split-NanoLuc® (SATiN)' to detect antibodies that bind to the spike (S) protein of SARS-CoV-2. Here, we expand on our previous work and describe a reconfigured version of the SATiN assay, called Neutralization SATiN (Neu-SATiN), which measures neutralization activity of antibodies directly from convalescent or vaccinated sera. The results obtained with our assay and other neutralization assays are comparable but with significantly shorter preparation and run time for Neu-SATiN. As the assay is modular, we further demonstrate that Neu-SATiN enables rapid assessment of the effectiveness of vaccines and level of protection against existing SARS-CoV-2 variants of concern and can therefore be readily adapted for emerging variants.
Collapse
|
14
|
Patel R, Khare S, Mahajan VS. Alternative Methods to Detect Severe Acute Respiratory Syndrome Coronavirus 2 Antibodies. Clin Lab Med 2022; 42:57-73. [PMID: 35153048 PMCID: PMC8563348 DOI: 10.1016/j.cll.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The COVID-19 pandemic has resulted in the development, validation, and rapid adoption of multiple novel diagnostic approaches. Hundreds of SARS-CoV-2 serologic assays have been developed and deployed to contain the spread of the virus, and to supply timely and important health information. Most of these serologic assays were based on a conventional enzyme-linked immunosorbent assay or the lateral flow assay format. The immunoassays that were developed were based on alternative technologies and are highlighted in this article with a brief discussion of the assay principle and the pros and cons for each assay. Measurement of neutralizing antibodies is also discussed.
Collapse
Affiliation(s)
- Rashmi Patel
- Micelio Labs, 58, 15th Cross Road, 2nd Phase, J P Nagar, Bengaluru 560078, India
| | - Siddharth Khare
- Micelio Labs, 58, 15th Cross Road, 2nd Phase, J P Nagar, Bengaluru 560078, India
| | - Vinay S. Mahajan
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA,Department of Pathology, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02215, USA,Corresponding author. Ragon Institute of MGH, MIT and Harvard400 Technology Square, Cambridge, MA 02139, USA
| |
Collapse
|
15
|
Castillo-Olivares J, Wells DA, Ferrari M, Chan ACY, Smith P, Nadesalingam A, Paloniemi M, Carnell GW, Ohlendorf L, Cantoni D, Mayora-Neto M, Palmer P, Tonks P, Temperton NJ, Peterhoff D, Neckermann P, Wagner R, Doffinger R, Kempster S, Otter AD, Semper A, Brooks T, Albecka A, James LC, Page M, Schwaeble W, Baxendale H, Heeney JL. Analysis of Serological Biomarkers of SARS-CoV-2 Infection in Convalescent Samples From Severe, Moderate and Mild COVID-19 Cases. Front Immunol 2021; 12:748291. [PMID: 34867975 PMCID: PMC8640495 DOI: 10.3389/fimmu.2021.748291] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
Precision monitoring of antibody responses during the COVID-19 pandemic is increasingly important during large scale vaccine rollout and rise in prevalence of Severe Acute Respiratory Syndrome-related Coronavirus-2 (SARS-CoV-2) variants of concern (VOC). Equally important is defining Correlates of Protection (CoP) for SARS-CoV-2 infection and COVID-19 disease. Data from epidemiological studies and vaccine trials identified virus neutralising antibodies (Nab) and SARS-CoV-2 antigen-specific (notably RBD and S) binding antibodies as candidate CoP. In this study, we used the World Health Organisation (WHO) international standard to benchmark neutralising antibody responses and a large panel of binding antibody assays to compare convalescent sera obtained from: a) COVID-19 patients; b) SARS-CoV-2 seropositive healthcare workers (HCW) and c) seronegative HCW. The ultimate aim of this study is to identify biomarkers of humoral immunity that could be used to differentiate severe from mild or asymptomatic SARS-CoV-2 infections. Some of these biomarkers could be used to define CoP in further serological studies using samples from vaccination breakthrough and/or re-infection cases. Whenever suitable, the antibody levels of the samples studied were expressed in International Units (IU) for virus neutralisation assays or in Binding Antibody Units (BAU) for ELISA tests. In this work we used commercial and non-commercial antibody binding assays; a lateral flow test for detection of SARS-CoV-2-specific IgG/IgM; a high throughput multiplexed particle flow cytometry assay for SARS-CoV-2 Spike (S), Nucleocapsid (N) and Receptor Binding Domain (RBD) proteins); a multiplex antigen semi-automated immuno-blotting assay measuring IgM, IgA and IgG; a pseudotyped microneutralisation test (pMN) and an electroporation-dependent neutralisation assay (EDNA). Our results indicate that overall, severe COVID-19 patients showed statistically significantly higher levels of SARS-CoV-2-specific neutralising antibodies (average 1029 IU/ml) than those observed in seropositive HCW with mild or asymptomatic infections (379 IU/ml) and that clinical severity scoring, based on WHO guidelines was tightly correlated with neutralisation and RBD/S antibodies. In addition, there was a positive correlation between severity, N-antibody assays and intracellular virus neutralisation.
Collapse
Affiliation(s)
- Javier Castillo-Olivares
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - David A. Wells
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Matteo Ferrari
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Andrew C. Y. Chan
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Peter Smith
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Angalee Nadesalingam
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Minna Paloniemi
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - George W. Carnell
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Luis Ohlendorf
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Phil Palmer
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paul Tonks
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nigel J. Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - David Peterhoff
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Patrick Neckermann
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Sarah Kempster
- Division of Virology, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | | | - Amanda Semper
- UK Health Security Agency, Porton Down, United Kingdom
| | - Tim Brooks
- UK Health Security Agency, Porton Down, United Kingdom
| | - Anna Albecka
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Leo C. James
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Mark Page
- Division of Virology, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | - Wilhelm Schwaeble
- Complement Laboratory, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Helen Baxendale
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Jonathan L. Heeney
- Lab of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
16
|
Di Genova C, Sampson A, Scott S, Cantoni D, Mayora-Neto M, Bentley E, Mattiuzzo G, Wright E, Derveni M, Auld B, Ferrara BT, Harrison D, Said M, Selim A, Thompson E, Thompson C, Carnell G, Temperton N. Production, Titration, Neutralisation, Storage and Lyophilisation of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Lentiviral Pseudotypes. Bio Protoc 2021; 11:e4236. [PMID: 34859134 PMCID: PMC8595416 DOI: 10.21769/bioprotoc.4236] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 11/02/2022] Open
Abstract
This protocol details a rapid and reliable method for the production and titration of high-titre viral pseudotype particles with the SARS-CoV-2 spike protein (and D614G or other variants of concern, VOC) on a lentiviral vector core, and use for neutralisation assays in target cells expressing angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). It additionally provides detailed instructions on substituting in new spike variants via gene cloning, lyophilisation and storage/shipping considerations for wide deployment potential. Results obtained with this protocol show that SARS-CoV-2 pseudotypes can be produced at equivalent titres to SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV) pseudotypes, neutralised by human convalescent plasma and monoclonal antibodies, and stored at a range of laboratory temperatures and lyophilised for distribution and subsequent application.
Collapse
Affiliation(s)
- Cecilia Di Genova
- Viral Pseudotype Unit (VPU Kent), Medway School of Pharmacy, University of Kent and Greenwich at Medway, Chatham Maritime, Kent, UK
| | - Alex Sampson
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge, UK
| | - Simon Scott
- Viral Pseudotype Unit (VPU Kent), Medway School of Pharmacy, University of Kent and Greenwich at Medway, Chatham Maritime, Kent, UK
| | - Diego Cantoni
- Viral Pseudotype Unit (VPU Kent), Medway School of Pharmacy, University of Kent and Greenwich at Medway, Chatham Maritime, Kent, UK
| | - Martin Mayora-Neto
- Viral Pseudotype Unit (VPU Kent), Medway School of Pharmacy, University of Kent and Greenwich at Medway, Chatham Maritime, Kent, UK
| | - Emma Bentley
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), Potters Bar, Hertfordshire, UK
| | - Giada Mattiuzzo
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), Potters Bar, Hertfordshire, UK
| | - Edward Wright
- Viral Pseudotype Unit (VPU Sussex), School of Life Sciences, University of Sussex, Brighton, UK
| | - Mariliza Derveni
- Viral Pseudotype Unit (VPU Sussex), School of Life Sciences, University of Sussex, Brighton, UK
| | - Bethany Auld
- Viral Pseudotype Unit (VPU Sussex), School of Life Sciences, University of Sussex, Brighton, UK
| | - Bill T Ferrara
- School of Science, University of Greenwich, Chatham Maritime, Kent, UK
| | - Dale Harrison
- School of Science, University of Greenwich, Chatham Maritime, Kent, UK
| | - Mohamed Said
- School of Science, University of Greenwich, Chatham Maritime, Kent, UK
| | - Arwa Selim
- School of Science, University of Greenwich, Chatham Maritime, Kent, UK
| | - Elinor Thompson
- School of Science, University of Greenwich, Chatham Maritime, Kent, UK
| | | | - George Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, Cambridge University, Madingley Road, Cambridge, UK
| | - Nigel Temperton
- Viral Pseudotype Unit (VPU Kent), Medway School of Pharmacy, University of Kent and Greenwich at Medway, Chatham Maritime, Kent, UK
| |
Collapse
|
17
|
Kalkeri R, Cai Z, Lin S, Farmer J, Kuzmichev YV, Koide F. SARS-CoV-2 Spike Pseudoviruses: A Useful Tool to Study Virus Entry and Address Emerging Neutralization Escape Phenotypes. Microorganisms 2021; 9:1744. [PMID: 34442823 PMCID: PMC8398529 DOI: 10.3390/microorganisms9081744] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
SARS-CoV-2 genetic variants are emerging around the globe. Unfortunately, several SARS-CoV-2 variants, especially variants of concern (VOCs), are less susceptible to neutralization by the convalescent and post-vaccination sera, raising concerns of increased disease transmissibility and severity. Recent data suggests that SARS-CoV-2 neutralizing antibody levels are a reliable correlate of vaccine-mediated protection. However, currently used BSL3-based virus micro-neutralization (MN) assays are more laborious, time-consuming, and expensive, underscoring the need for BSL2-based, cost-effective neutralization assays against SARS-CoV-2 variants. In light of this unmet need, we have developed a BSL-2 pseudovirus-based neutralization assay (PBNA) in cells expressing the human angiotensin-converting enzyme-2 (hACE2) receptor for SARS-CoV-2. The assay is reproducible (R2 = 0.96), demonstrates a good dynamic range and high sensitivity. Our data suggest that the biological Anti-SARS-CoV-2 research reagents such as NIBSC 20/130 show lower neutralization against B.1.351 SA (South Africa) and B.1.1.7 UK (United Kingdom) VOC, whereas a commercially available monoclonal antibody MM43 retains activity against both these variants. SARS-CoV-2 spike PBNAs for VOCs would be useful tools to measure the neutralization ability of candidate vaccines in both preclinical models and clinical trials and would further help develop effective prophylactic countermeasures against emerging neutralization escape phenotypes.
Collapse
Affiliation(s)
- Raj Kalkeri
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Zhaohui Cai
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Shuling Lin
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - John Farmer
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 2000 Ninth Avenue South, Birmingham, AL 35205, USA;
| | - Yury V. Kuzmichev
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| | - Fusataka Koide
- Department of Infectious Disease Research, Drug Development Division, Southern Research, 431 Aviation Way, Frederick, MD 21701, USA; (Z.C.); (S.L.); (Y.V.K.)
| |
Collapse
|
18
|
Sampson AT, Heeney J, Cantoni D, Ferrari M, Sans MS, George C, Di Genova C, Mayora Neto M, Einhauser S, Asbach B, Wagner R, Baxendale H, Temperton N, Carnell G. Coronavirus Pseudotypes for All Circulating Human Coronaviruses for Quantification of Cross-Neutralizing Antibody Responses. Viruses 2021; 13:1579. [PMID: 34452443 PMCID: PMC8402765 DOI: 10.3390/v13081579] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/17/2021] [Accepted: 08/01/2021] [Indexed: 12/23/2022] Open
Abstract
The novel coronavirus SARS-CoV-2 is the seventh identified human coronavirus. Understanding the extent of pre-existing immunity induced by seropositivity to endemic seasonal coronaviruses and the impact of cross-reactivity on COVID-19 disease progression remains a key research question in immunity to SARS-CoV-2 and the immunopathology of COVID-2019 disease. This paper describes a panel of lentiviral pseudotypes bearing the spike (S) proteins for each of the seven human coronaviruses (HCoVs), generated under similar conditions optimized for high titre production allowing a high-throughput investigation of antibody neutralization breadth. Optimal production conditions and most readily available permissive target cell lines were determined for spike-mediated entry by each HCoV pseudotype: SARS-CoV-1, SARS-CoV-2 and HCoV-NL63 best transduced HEK293T/17 cells transfected with ACE2 and TMPRSS2, HCoV-229E and MERS-CoV preferentially entered HUH7 cells, and CHO cells were most permissive for the seasonal betacoronavirus HCoV-HKU1. Entry of ACE2 using pseudotypes was enhanced by ACE2 and TMPRSS2 expression in target cells, whilst TMPRSS2 transfection rendered HEK293T/17 cells permissive for HCoV-HKU1 and HCoV-OC43 entry. Additionally, pseudotype viruses were produced bearing additional coronavirus surface proteins, including the SARS-CoV-2 Envelope (E) and Membrane (M) proteins and HCoV-OC43/HCoV-HKU1 Haemagglutinin-Esterase (HE) proteins. This panel of lentiviral pseudotypes provides a safe, rapidly quantifiable and high-throughput tool for serological comparison of pan-coronavirus neutralizing responses; this can be used to elucidate antibody dynamics against individual coronaviruses and the effects of antibody cross-reactivity on clinical outcome following natural infection or vaccination.
Collapse
Affiliation(s)
- Alexander Thomas Sampson
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
| | - Jonathan Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
- DIOSynVax Ltd., Cambridge CB3 0ES, UK
| | - Diego Cantoni
- Viral Pseudotype Unit, University of Kent, Chatham ME4 4TB, UK; (D.C.); (C.D.G.); (M.M.N.); (N.T.)
| | - Matteo Ferrari
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
- DIOSynVax Ltd., Cambridge CB3 0ES, UK
| | - Maria Suau Sans
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
| | - Charlotte George
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
| | - Cecilia Di Genova
- Viral Pseudotype Unit, University of Kent, Chatham ME4 4TB, UK; (D.C.); (C.D.G.); (M.M.N.); (N.T.)
| | - Martin Mayora Neto
- Viral Pseudotype Unit, University of Kent, Chatham ME4 4TB, UK; (D.C.); (C.D.G.); (M.M.N.); (N.T.)
| | - Sebastian Einhauser
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (B.A.); (R.W.)
| | - Benedikt Asbach
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (B.A.); (R.W.)
| | - Ralf Wagner
- Institute for Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany; (S.E.); (B.A.); (R.W.)
- Institute for Clinical Microbiology and Hygiene, University Hospital, 93053 Regensburg, Germany
| | - Helen Baxendale
- Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0AY, UK;
| | - Nigel Temperton
- Viral Pseudotype Unit, University of Kent, Chatham ME4 4TB, UK; (D.C.); (C.D.G.); (M.M.N.); (N.T.)
| | - George Carnell
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; (J.H.); (M.F.); (M.S.S.); (C.G.); (G.C.)
| |
Collapse
|