1
|
van Tol S, Fletcher P, Feldmann F, Mukesh RK, Port JR, Gallogly S, Schulz JE, Rhoderick JF, Makinson R, Carmody A, Myers L, Lovaglio J, Smith BJ, Okumura A, Shaia C, Saturday G, Marzi A, Lambe T, Munster VJ, van Doremalen N. A Bivalent Adenovirus-Vectored Vaccine Induces a Robust Humoral Response, but Does Not Protect Cynomolgus Macaques Against a Lethal Challenge With Sudan Virus. J Infect Dis 2024; 230:1083-1092. [PMID: 38487996 DOI: 10.1093/infdis/jiae056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/30/2024] [Indexed: 11/16/2024] Open
Abstract
The most recent Sudan virus (SUDV) outbreak in Uganda was first detected in September 2022 and resulted in 164 laboratory-confirmed cases and 77 deaths. There are no approved vaccines against SUDV. Here, we investigated the protective efficacy of ChAdOx1-biEBOV in cynomolgus macaques using a prime or a prime-boost regimen. ChAdOx1-biEBOV is a replication-deficient simian adenovirus vector encoding SUDV and Ebola virus (EBOV) glycoproteins (GPs). Intramuscular vaccination induced SUDV and EBOV GP-specific immunoglobulin G responses and neutralizing antibodies. Upon challenge with SUDV, vaccinated animals showed signs of disease like those observed in control animals, and no differences in survival outcomes were measured among all 3 groups. Viral load in blood samples and in tissue samples obtained after necropsy were not significantly different between groups. Overall, this study highlights the importance of evaluating vaccines in multiple animal models and demonstrates the importance of understanding protective efficacy in both animal models and human hosts.
Collapse
MESH Headings
- Animals
- Macaca fascicularis
- Hemorrhagic Fever, Ebola/prevention & control
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/virology
- Ebolavirus/immunology
- Ebolavirus/genetics
- Antibodies, Viral/blood
- Genetic Vectors
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Immunity, Humoral
- Ebola Vaccines/immunology
- Ebola Vaccines/administration & dosage
- Immunoglobulin G/blood
- Disease Models, Animal
- Viral Load
- Adenoviruses, Simian/immunology
- Adenoviruses, Simian/genetics
- Vaccination
- Adenoviridae/genetics
- Adenoviridae/immunology
Collapse
Affiliation(s)
| | | | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | | | | | | | | | | | - Rebecca Makinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom
| | - Aaron Carmody
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Lara Myers
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Brian J Smith
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | | | - Carl Shaia
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | | | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom
| | | | | |
Collapse
|
2
|
Miranda MC, Kepl E, Navarro MJ, Chen C, Johnson M, Sprouse KR, Stewart C, Palser A, Valdez A, Pettie D, Sydeman C, Ogohara C, Kraft JC, Pham M, Murphy M, Wrenn S, Fiala B, Ravichandran R, Ellis D, Carter L, Corti D, Kellam P, Lee K, Walls AC, Veesler D, King NP. Potent neutralization of SARS-CoV-2 variants by RBD nanoparticle and prefusion-stabilized spike immunogens. NPJ Vaccines 2024; 9:184. [PMID: 39379400 PMCID: PMC11461925 DOI: 10.1038/s41541-024-00982-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024] Open
Abstract
We previously described a two-component protein nanoparticle vaccine platform that displays 60 copies of the SARS-CoV-2 spike protein RBD (RBD-NP). The vaccine, when adjuvanted with AS03, was shown to elicit robust neutralizing antibody and CD4 T cell responses in Phase I/II clinical trials, met its primary co-endpoints in a Phase III trial, and has been licensed by multiple regulatory authorities under the brand name SKYCovioneTM. Here we characterize the biophysical properties, stability, antigenicity, and immunogenicity of RBD-NP immunogens incorporating mutations from the B.1.351 (β) and P.1 (γ) variants of concern (VOCs) that emerged in 2020. We also show that the RBD-NP platform can be adapted to the Omicron strains BA.5 and XBB.1.5. We compare β and γ variant and E484K point mutant nanoparticle immunogens to the nanoparticle displaying the Wu-1 RBD, as well as to soluble prefusion-stabilized (HexaPro) spike trimers harboring VOC-derived mutations. We find the properties of immunogens based on different SARS-CoV-2 variants can differ substantially, which could affect the viability of variant vaccine development. Introducing stabilizing mutations in the linoleic acid binding site of the RBD-NPs resulted in increased physical stability compared to versions lacking the stabilizing mutations without deleteriously affecting immunogenicity. The RBD-NP immunogens and HexaPro trimers, as well as combinations of VOC-based immunogens, elicited comparable levels of neutralizing antibodies against distinct VOCs. Our results demonstrate that RBD-NP-based vaccines can elicit neutralizing antibody responses against SARS-CoV-2 variants and can be rapidly designed and stabilized, demonstrating the potential of two-component RBD-NPs as a platform for the development of broadly protective coronavirus vaccines.
Collapse
Affiliation(s)
- Marcos C Miranda
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Elizabeth Kepl
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Mary Jane Navarro
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Chengbo Chen
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
- Biological Physics Structure and Design Program, University of Washington, Seattle, WA, USA
| | - Max Johnson
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Kaitlin R Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Anne Palser
- Kymab Ltd., Babraham Research Campus, Cambridge, UK
| | - Adian Valdez
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Deleah Pettie
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Claire Sydeman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Cassandra Ogohara
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - John C Kraft
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Minh Pham
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Michael Murphy
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Sam Wrenn
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brooke Fiala
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Rashmi Ravichandran
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Daniel Ellis
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Paul Kellam
- Kymab Ltd., Babraham Research Campus, Cambridge, UK
- Department of Infectious Disease, Imperial College, London, UK
| | - Kelly Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
- Biological Physics Structure and Design Program, University of Washington, Seattle, WA, USA
| | - Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| |
Collapse
|
3
|
Stepanova E, Isakova-Sivak I, Matyushenko V, Mezhenskaya D, Kudryavtsev I, Kostromitina A, Chistiakova A, Rak A, Bazhenova E, Prokopenko P, Kotomina T, Donina S, Novitskaya V, Sivak K, Karal-Ogly D, Rudenko L. Safety and Immunogenicity Study of a Bivalent Vaccine for Combined Prophylaxis of COVID-19 and Influenza in Non-Human Primates. Vaccines (Basel) 2024; 12:1099. [PMID: 39460266 PMCID: PMC11511058 DOI: 10.3390/vaccines12101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Influenza and SARS-CoV-2 viruses are two highly variable pathogens. We have developed a candidate bivalent live vaccine based on the strain of licensed A/Leningrad/17-based cold-adapted live attenuated influenza vaccine (LAIV) of H3N2 subtype, which expressed SARS-CoV-2 immunogenic T-cell epitopes. A cassette encoding fragments of S and N proteins of SARS-CoV-2 was inserted into the influenza NA gene using the P2A autocleavage site. In this study, we present the results of preclinical evaluation of the developed bivalent vaccine in a non-human primate model. METHODS Rhesus macaques (Macaca mulatta) (n = 3 per group) were immunized intranasally with 7.5 lg EID50 of the LAIV/CoV-2 bivalent vaccine, a control non-modified H3N2 LAIV or a placebo (chorioallantoic fluid) using a sprayer device, twice, with a 28-day interval. The blood samples were collected at days 0, 3, 28 and 35 for hematological and biochemical assessment. Safety was also assessed by monitoring body weight, body temperature and clinical signs of the disease. Immune responses to influenza virus were assessed both by determining serum antibody titers in hemagglutination inhibition assay, microneutralization assay and IgG ELISA. T-cell responses were measured both to influenza and SARS-CoV-2 antigens using ELISPOT and flow cytometry. Three weeks after the second immunization, animals were challenged with 105 PFU of Delta SARS-CoV-2. The body temperature, weight and challenge virus shedding were monitored for 5 days post-challenge. In addition, virus titers in various organs and histopathology were evaluated on day 6 after SARS-CoV-2 infection. RESULTS There was no toxic effect of the immunizations on the hematological and coagulation hemostasis of animals. No difference in the dynamics of the average weight and thermometry results were found between the groups of animals. Both LAIV and LAIV/CoV-2 variants poorly replicated in the upper respiratory tract of rhesus macaques. Nevertheless, despite this low level of virus shedding, influenza-specific serum IgG responses were detected in the group of monkeys immunized with the LAIV/CoV-2 bivalent but not in the LAIV group. Furthermore, T-cell responses to both influenza and SARS-CoV-2 viruses were detected in the LAIV/CoV-2 vaccine group only. The animals were generally resistant to SARS-CoV-2 challenge, with minimal virus shedding in the placebo and LAIV groups. Histopathological changes in vaccinated animals were decreased compared to the PBS group, suggesting a protective effect of the chimeric vaccine candidate. CONCLUSIONS The candidate bivalent vaccine was safe and immunogenic for non-human primates and warrants its further evaluation in clinical trials.
Collapse
Affiliation(s)
- Ekaterina Stepanova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Victoria Matyushenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Daria Mezhenskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Arina Kostromitina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Anna Chistiakova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Alexandra Rak
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Ekaterina Bazhenova
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Polina Prokopenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Tatiana Kotomina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Svetlana Donina
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Vlada Novitskaya
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint-Petersburg 197376, Russia;
| | - Dzhina Karal-Ogly
- Center of Preclinical Research, Research Institute of Medical Primatology, Sochi 354376, Russia;
| | - Larisa Rudenko
- Institute of Experimental Medicine, Saint-Petersburg 197022, Russia; (I.I.-S.); (V.M.); (D.M.); (I.K.); (A.K.); (A.C.); (A.R.); (P.P.); (T.K.); (V.N.); (L.R.)
| |
Collapse
|
4
|
Fischer RJ, Bushmaker T, Williamson BN, Pérez-Pérez L, Feldmann F, Lovaglio J, Scott D, Saturday G, Feldmann H, Munster VJ, de Wit E, van Doremalen N. Compartmentalized SARS-CoV-2 Replication in the Upper vs Lower Respiratory Tract After Intranasal Inoculation or Aerosol Exposure. J Infect Dis 2024; 230:657-661. [PMID: 38261786 PMCID: PMC11420804 DOI: 10.1093/infdis/jiae018] [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: 09/08/2023] [Revised: 12/21/2023] [Accepted: 01/19/2024] [Indexed: 01/25/2024] Open
Abstract
Nonhuman primate models are essential for the development of vaccines and antivirals against infectious diseases. Rhesus macaques are a widely utilized infection model for SARS-CoV-2. We compared cellular tropism and virus replication in rhesus macaques inoculated with SARS-CoV-2 via the intranasal route or via exposure to aerosols. Intranasal inoculation resulted in replication in the upper respiratory tract with limited involvement in the lower respiratory tract, whereas exposure to aerosols resulted in infection throughout the respiratory tract. In comparison with multiroute inoculation, intranasal and aerosol inoculation resulted in reduced SARS-CoV-2 replication in the respiratory tract.
Collapse
Affiliation(s)
| | | | | | | | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Dana Scott
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | | | | | | | | |
Collapse
|
5
|
Onishi R, Ikemoto S, Shiota A, Tsukamoto T, Asayama A, Tachibana M, Sakurai F, Mizuguchi H. Development of a novel adenovirus serotype 35 vector vaccine possessing an RGD peptide in the fiber knob and the E4 orf 4, 6, and 6/7 regions of adenovirus serotype 5. Int J Pharm 2024; 662:124480. [PMID: 39038719 DOI: 10.1016/j.ijpharm.2024.124480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 06/23/2024] [Accepted: 07/14/2024] [Indexed: 07/24/2024]
Abstract
Adenovirus (Ad) vectors based on human adenovirus serotype 5 (Ad5) have attracted significant attention as vaccine vectors for infectious diseases. However, the effectiveness of Ad5 vectors as vaccines is often inhibited by the anti-Ad5 neutralizing antibodies retained by many adults. To overcome this drawback, we focused on human adenovirus serotype 35 (Ad35) vectors with low seroprevalence in adults. Although Ad35 vectors can circumvent anti-Ad5 neutralizing antibodies, vector yields of Ad35 vectors are often inferior to those of Ad5 vectors. In this study, we developed novel Ad35 vectors containing the Ad5 E4 orf 4, 6, and 6/7 or the Ad5 E4 orf 6 and 6/7 for efficient vector production, and compared their properties. These E4-modified Ad35 vectors efficiently propagated to a similar extent at virus titers comparable to those of Ad5 vectors. An Ad35 vector containing the Ad5 E4 orf 4, 6, and 6/7 mediated more efficient transduction than that containing the Ad5 E4 orf 6 and 6/7 in human cultured cells. Furthermore, insertion of an arginine-glycine-aspartate (RGD) peptide in the fiber region of an Ad35 vector containing the Ad5 E4 orf 4, 6, and 6/7 significantly improved the transgene product-specific antibody production following intramuscular administration in mice. The Ad35 vector containing the RGD peptide mediated efficient vaccine effects even in the mice pre-immunized with an Ad5.
Collapse
Affiliation(s)
- Rika Onishi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Sena Ikemoto
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Aoi Shiota
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Tomohito Tsukamoto
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akira Asayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Masashi Tachibana
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan; The Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan; Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
6
|
Ceccopieri C, Madej JP. Chicken Secondary Lymphoid Tissues-Structure and Relevance in Immunological Research. Animals (Basel) 2024; 14:2439. [PMID: 39199973 PMCID: PMC11350708 DOI: 10.3390/ani14162439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Recent discoveries have indicated the importance of developing modern strategies for vaccinations, more ethical research models, and effective alternatives to antibiotic treatment in farm animals. Chickens (Gallus gallus) play a crucial role in this context given the commercial and economic relevance of poultry production worldwide and the search for analogies between the immune systems of humans and birds. Specifically, chicken secondary lymphoid tissues share similar features to their human counterparts. Chickens have several secondary or peripheral lymphoid tissues that are the sites where the adaptive immune response is initiated. The more general classification of these organs divides them into the spleen and skin-, pineal-, or mucosa-associated lymphoid tissues. Each of these tissues is further subdivided into separate lymphoid structures that perform specific and different functions along the animal's body. A review summarizing the state of the art of research on chicken secondary lymphoid organs is of great relevance for the design of future studies.
Collapse
Affiliation(s)
| | - Jan P. Madej
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland;
| |
Collapse
|
7
|
Kim DH, Lee J, Lee DY, Lee SH, Jeong JH, Kim JY, Kim J, Choi YK, Lee JB, Park SY, Choi IS, Lee SW, Youk S, Song CS. Intranasal Administration of Recombinant Newcastle Disease Virus Expressing SARS-CoV-2 Spike Protein Protects hACE2 TG Mice against Lethal SARS-CoV-2 Infection. Vaccines (Basel) 2024; 12:921. [PMID: 39204044 PMCID: PMC11359043 DOI: 10.3390/vaccines12080921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), emerged as a global outbreak in 2019, profoundly affecting both human health and the global economy. Various vaccine modalities were developed and commercialized to overcome this challenge, including inactivated vaccines, mRNA vaccines, adenovirus vector-based vaccines, and subunit vaccines. While intramuscular vaccines induce high IgG levels, they often fail to stimulate significant mucosal immunity in the respiratory system. We employed the Newcastle disease virus (NDV) vector expressing the spike protein of the SARS-CoV-2 Beta variant (rK148/beta-S), and evaluated the efficacy of intranasal vaccination with rK148/beta-S in K18-hACE2 transgenic mice. Intranasal vaccination with a low dose (106.0 EID50) resulted in an 86% survival rate after challenge with the SARS-CoV-2 Beta variant. Administration at a high dose (107.0 EID50) led to a reduction in lung viral load and 100% survival against the SARS-CoV-2 Beta and Delta variants. A high level of the SARS-CoV-2 spike-specific IgA was also induced in vaccinated mice lungs following the SARS-CoV-2 challenge. Our findings suggest that rK148/beta-S holds promise as an intranasal vaccine candidate that effectively induces mucosal immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Deok-Hwan Kim
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jiho Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, U.S. Department of Agriculture-Agricultural Research Service, 934 College Station Road, Athens, GA 30605, USA
| | - Da-Ye Lee
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seung-Hun Lee
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jei-Hyun Jeong
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ji-Yun Kim
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jiwon Kim
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
| | - Joong-Bok Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Seung-Young Park
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - In-Soo Choi
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Sang-Won Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
| | - Sungsu Youk
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju 28160, Republic of Korea
- Biomedical Research Institute, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (D.-H.K.)
- KHAV Co., Ltd., 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
8
|
Zhao T, Liu S, Wang P, Zhang Y, Kang X, Pan X, Li L, Li D, Gao P, An Y, Song H, Liu K, Qi J, Zhao X, Dai L, Liu P, Wang P, Wu G, Zhu T, Xu K, Li Y, Gao GF. Protective RBD-dimer vaccines against SARS-CoV-2 and its variants produced in glycoengineered Pichia pastoris. PLoS Pathog 2024; 20:e1012487. [PMID: 39213280 PMCID: PMC11364227 DOI: 10.1371/journal.ppat.1012487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Protective vaccines are crucial for preventing and controlling coronavirus disease 2019 (COVID-19). Updated vaccines are needed to confront the continuously evolving and circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. These vaccines should be safe, effective, amenable to easily scalable production, and affordable. Previously, we developed receptor binding domain (RBD) dimer-based protein subunit vaccines (ZF2001 and updated vaccines) in mammalian cells. In this study, we explored a strategy for producing RBD-dimer immunogens in Pichia pastoris. We found that wild-type P. pastoris produced hyperglycosylated RBD-dimer protein containing four N-glycosylation sites in P. pastoris. Therefore, we engineered the wild type P. pastoris (GS strain) into GSΔOCH1pAO by deleting the OCH1 gene (encoding α-1,6-mannosyltransferase enzyme) to decrease glycosylation, as well as by overexpressing the HIS4 gene (encoding histidine dehydrogenase) to increase histidine synthesis for better growth. In addition, RBD-dimer protein was truncated to remove the R328/F329 cleavage sites in P. pastoris. Several homogeneous RBD-dimer proteins were produced in the GSΔOCH1pAO strain, demonstrating the feasibility of using the P. pastoris expression system. We further resolved the cryo-EM structure of prototype-Beta RBD-dimer complexed with the neutralizing antibody CB6 to reveal the completely exposed immune epitopes of the RBDs. In a murine model, we demonstrated that the yeast-produced RBD-dimer induces robust and protective antibody responses, which is suitable for boosting immunization. This study developed the yeast system for producing SARS-CoV-2 RBD-dimer immunogens, providing a promising platform and pipeline for the future continuous updating and production of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Tongxin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Sheng Liu
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, China
| | - Pengyan Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yanfang Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Xinrui Kang
- Medical School, University of Chinese Academy of Sciences (UCAS), Beijing, China
| | - Xiaoqian Pan
- Medical School, University of Chinese Academy of Sciences (UCAS), Beijing, China
| | - Linjie Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Dedong Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Ping Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yaling An
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Hao Song
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences (CAS), Beijing, China
| | - Kefang Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Lianpan Dai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Peipei Liu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Peiyi Wang
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, China
| | - Guizhen Wu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Taicheng Zhu
- Department of Microbial Physiological & Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Kun Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yin Li
- Department of Microbial Physiological & Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - George F. Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences (CAS), Beijing, China
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
9
|
Bouazzaoui A, Abdellatif AA. Vaccine delivery systems and administration routes: Advanced biotechnological techniques to improve the immunization efficacy. Vaccine X 2024; 19:100500. [PMID: 38873639 PMCID: PMC11170481 DOI: 10.1016/j.jvacx.2024.100500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/21/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Since the first use of vaccine tell the last COVID-19 pandemic caused by spread of SARS-CoV-2 worldwide, the use of advanced biotechnological techniques has accelerated the development of different types and methods for immunization. The last pandemic showed that the nucleic acid-based vaccine, especially mRNA, has an advantage in terms of development time; however, it showed a very critical drawback namely, the higher costs when compared to other strategies, and its inability to protect against new variants. This showed the need of more improvement to reach a better delivery and efficacy. In this review we will describe different vaccine delivery systems including, the most used viral vector, and also variable strategies for delivering of nucleic acid-based vaccines especially lipid-based nanoparticles formulation, polymersomes, electroporation and also the new powerful tools for the delivery of mRNA, which is based on the use of cell-penetrating peptides (CPPs). Additionally, we will also discuss the main challenges associated with each system. Finlay, the efficacy and safety of the vaccines depends not only on the formulations and delivery systems, but also the dosage and route of administration are also important players, therefore we will see the different routes for the vaccine administration including traditionally routes (intramuscular, Transdermal, subcutaneous), oral inhalation or via nasal mucosa, and will describe the advantages and disadvantage of each administration route.
Collapse
Affiliation(s)
- Abdellatif Bouazzaoui
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
- Science and Technology Unit, Umm Al Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Ahmed A.H. Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, 51452 Qassim, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, 71524 Assiut, Egypt
| |
Collapse
|
10
|
Beaudoin-Bussières G, Finzi A. Deciphering Fc-effector functions against SARS-CoV-2. Trends Microbiol 2024; 32:756-768. [PMID: 38365562 DOI: 10.1016/j.tim.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 02/18/2024]
Abstract
Major efforts were deployed to study the antibody response against SARS-CoV-2. Antibodies neutralizing SARS-CoV-2 have been extensively studied in the context of infections, vaccinations, and breakthrough infections. Antibodies, however, are pleiotropic proteins that have many functions in addition to neutralization. These include Fc-effector functions such as antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Although important to combat viral infections, these Fc-effector functions were less studied in the context of SARS-CoV-2 compared with binding and neutralization. This is partly due to the difficulty in developing reliable assays to measure Fc-effector functions compared to antibody binding and neutralization. Multiple assays have now been developed and can be used to measure different Fc-effector functions. Here, we review these assays and what is known regarding anti-SARS-CoV-2 Fc-effector functions. Overall, this review summarizes and updates our current state of knowledge regarding anti-SARS-CoV-2 Fc-effector functions.
Collapse
Affiliation(s)
- Guillaume Beaudoin-Bussières
- Centre de recherche du CHUM, Montréal, Québec H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Québec H2X 0A9, Canada
| | - Andrés Finzi
- Centre de recherche du CHUM, Montréal, Québec H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Québec H2X 0A9, Canada.
| |
Collapse
|
11
|
Tan Z, Yang C, Lin PH, Ramadan S, Yang W, Rashidi Z, Lang S, Shafieichaharberoud F, Gao J, Pan X, Soloff N, Wu X, Bolin S, Pyeon D, Huang X. Inducing Long Lasting B Cell and T Cell Immunity Against Multiple Variants of SARS-CoV-2 Through Mutant Bacteriophage Qβ-Receptor Binding Domain Conjugate. Adv Healthc Mater 2024; 13:e2302755. [PMID: 38733291 PMCID: PMC11305917 DOI: 10.1002/adhm.202302755] [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: 08/21/2023] [Revised: 05/04/2024] [Indexed: 05/13/2024]
Abstract
More than 3 years into the global pandemic, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a significant threat to public health. Immunities acquired from infection or current vaccines fail to provide long term protection against subsequent infections, mainly due to their fast-waning nature and the emergence of variants of concerns (VOCs) such as Omicron. To overcome these limitations, SARS-CoV-2 Spike protein receptor binding domain (RBD)-based epitopes are investigated as conjugates with a powerful carrier, the mutant bacteriophage Qβ (mQβ). The epitope design is critical to eliciting potent antibody responses with the full length RBD being superior to peptide and glycopeptide antigens. The full length RBD conjugated with mQβ activates both humoral and cellular immune systems in vivo, inducing broad spectrum, persistent, and comprehensive immune responses effective against multiple VOCs including Delta and Omicron variants, rendering it a promising vaccine candidate.
Collapse
Affiliation(s)
- Zibin Tan
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Canchai Yang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Po-Han Lin
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Sherif Ramadan
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Department of Chemistry, Benha University, Benha, 13518, Egypt
| | - Weizhun Yang
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
| | - Zahra Rashidi
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Shuyao Lang
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Fatemeh Shafieichaharberoud
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Jia Gao
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Xingling Pan
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Nachy Soloff
- Hatzalah of Michigan, 13650 Oak Park Blvd., Oak Park, MI, 48237, USA
| | - Xuanjun Wu
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, Shandong, 250100, China
| | - Steven Bolin
- Veterinary Diagnostic Laboratory, Michigan State University, East Lansing, MI, 48824, USA
| | - Dohun Pyeon
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
12
|
Mandolesi M, Das H, de Vries L, Yang Y, Kim C, Dhinakaran M, Castro Dopico X, Fischbach J, Kim S, Guryleva MV, Àdori M, Chernyshev M, Stålmarck A, Hanke L, McInerney GM, Sheward DJ, Corcoran M, Hällberg BM, Murrell B, Karlsson Hedestam GB. Multi-compartmental diversification of neutralizing antibody lineages dissected in SARS-CoV-2 spike-immunized macaques. Nat Commun 2024; 15:6338. [PMID: 39068149 PMCID: PMC11283548 DOI: 10.1038/s41467-024-50286-0] [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: 02/15/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
The continued evolution of SARS-CoV-2 underscores the need to understand qualitative aspects of the humoral immune response elicited by spike immunization. Here, we combine monoclonal antibody (mAb) isolation with deep B cell receptor (BCR) repertoire sequencing of rhesus macaques immunized with prefusion-stabilized spike glycoprotein. Longitudinal tracing of spike-sorted B cell lineages in multiple immune compartments demonstrates increasing somatic hypermutation and broad dissemination of vaccine-elicited B cells in draining and non-draining lymphoid compartments, including the bone marrow, spleen and, most notably, periaortic lymph nodes. Phylogenetic analysis of spike-specific monoclonal antibody lineages identified through deep repertoire sequencing delineates extensive intra-clonal diversification that shaped neutralizing activity. Structural analysis of the spike in complex with a broadly neutralizing mAb provides a molecular basis for the observed differences in neutralization breadth between clonally related antibodies. Our findings highlight that immunization leads to extensive intra-clonal B cell evolution where members of the same lineage can both retain the original epitope specificity and evolve to recognize additional spike variants not previously encountered.
Collapse
MESH Headings
- Animals
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Macaca mulatta
- Antibodies, Neutralizing/immunology
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- B-Lymphocytes/immunology
- Antibodies, Viral/immunology
- Phylogeny
- Antibodies, Monoclonal/immunology
- Epitopes/immunology
- COVID-19/immunology
- COVID-19/virology
- Humans
- COVID-19 Vaccines/immunology
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/genetics
- Somatic Hypermutation, Immunoglobulin
- Immunization
Collapse
Affiliation(s)
- Marco Mandolesi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Hrishikesh Das
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Liset de Vries
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yiqiu Yang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Changil Kim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Manojj Dhinakaran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Julian Fischbach
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sungyong Kim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mariia V Guryleva
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Monika Àdori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mark Chernyshev
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Aron Stålmarck
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Leo Hanke
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Gerald M McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Daniel J Sheward
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin Corcoran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - B Martin Hällberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ben Murrell
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
13
|
Leekha A, Saeedi A, Sefat KMSR, Kumar M, Martinez-Paniagua M, Damian A, Kulkarni R, Reichel K, Rezvan A, Masoumi S, Liu X, Cooper LJN, Sebastian M, Sands CM, Das VE, Patel NB, Hurst B, Varadarajan N. Multi-antigen intranasal vaccine protects against challenge with sarbecoviruses and prevents transmission in hamsters. Nat Commun 2024; 15:6193. [PMID: 39043645 PMCID: PMC11266618 DOI: 10.1038/s41467-024-50133-2] [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: 08/01/2023] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
Immunization programs against SARS-CoV-2 with commercial intramuscular vaccines prevent disease but are less efficient in preventing infections. Mucosal vaccines can provide improved protection against transmission, ideally for different variants of concern (VOCs) and related sarbecoviruses. Here, we report a multi-antigen, intranasal vaccine, NanoSTING-SN (NanoSTING-Spike-Nucleocapsid), eliminates virus replication in both the lungs and the nostrils upon challenge with the pathogenic SARS-CoV-2 Delta VOC. We further demonstrate that NanoSTING-SN prevents transmission of the SARS-CoV-2 Omicron VOC (BA.5) to vaccine-naïve hamsters. To evaluate protection against other sarbecoviruses, we immunized mice with NanoSTING-SN. We showed that immunization affords protection against SARS-CoV, leading to protection from weight loss and 100% survival in mice. In non-human primates, animals immunized with NanoSTING-SN show durable serum IgG responses (6 months) and nasal wash IgA responses cross-reactive to SARS-CoV-2 (XBB1.5), SARS-CoV and MERS-CoV antigens. These observations have two implications: (1) mucosal multi-antigen vaccines present a pathway to reducing transmission of respiratory viruses, and (2) eliciting immunity against multiple antigens can be advantageous in engineering pan-sarbecovirus vaccines.
Collapse
Affiliation(s)
- Ankita Leekha
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Arash Saeedi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - K M Samiur Rahman Sefat
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Monish Kumar
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Melisa Martinez-Paniagua
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Adrian Damian
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Rohan Kulkarni
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Kate Reichel
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Ali Rezvan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Shalaleh Masoumi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | | | | | | | - Vallabh E Das
- College of Optometry, University of Houston, Houston, TX, USA
| | - Nimesh B Patel
- College of Optometry, University of Houston, Houston, TX, USA
| | - Brett Hurst
- Institute of Antiviral Research, Utah State University, UT, Logan, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA.
| |
Collapse
|
14
|
Wagner JT, Müller-Schmucker SM, Wang W, Arnold P, Uhlig N, Issmail L, Eberlein V, Damm D, Roshanbinfar K, Ensser A, Oltmanns F, Peter AS, Temchura V, Schrödel S, Engel FB, Thirion C, Grunwald T, Wuhrer M, Grimm D, Überla K. Influence of AAV vector tropism on long-term expression and Fc-γ receptor binding of an antibody targeting SARS-CoV-2. Commun Biol 2024; 7:865. [PMID: 39009807 PMCID: PMC11250830 DOI: 10.1038/s42003-024-06529-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 07/02/2024] [Indexed: 07/17/2024] Open
Abstract
Long-acting passive immunization strategies are needed to protect immunosuppressed vulnerable groups from infectious diseases. To further explore this concept for COVID-19, we constructed Adeno-associated viral (AAV) vectors encoding the human variable regions of the SARS-CoV-2 neutralizing antibody, TRES6, fused to murine constant regions. An optimized vector construct was packaged in hepatotropic (AAV8) or myotropic (AAVMYO) AAV capsids and injected intravenously into syngeneic TRIANNI-mice. The highest TRES6 serum concentrations (511 µg/ml) were detected 24 weeks after injection of the myotropic vector particles and mean TRES6 serum concentrations remained above 100 µg/ml for at least one year. Anti-drug antibodies or TRES6-specific T cells were not detectable. After injection of the AAV8 particles, vector mRNA was detected in the liver, while the AAVMYO particles led to high vector mRNA levels in the heart and skeletal muscle. The analysis of the Fc-glycosylation pattern of the TRES6 serum antibodies revealed critical differences between the capsids that coincided with different binding activities to murine Fc-γ-receptors. Concomitantly, the vector-based immune prophylaxis led to protection against SARS-CoV-2 infection in K18-hACE2 mice. High and long-lasting expression levels, absence of anti-drug antibodies and favourable Fc-γ-receptor binding activities warrant further exploration of myotropic AAV vector-based delivery of antibodies and other biologicals.
Collapse
Affiliation(s)
- Jannik T Wagner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sandra M Müller-Schmucker
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Preclinical Validation, Leipzig, Germany
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Preclinical Validation, Leipzig, Germany
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Preclinical Validation, Leipzig, Germany
| | - Dominik Damm
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Armin Ensser
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Friederike Oltmanns
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Antonia Sophia Peter
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Vladimir Temchura
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Preclinical Validation, Leipzig, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, University of Heidelberg; BioQuant Center, BQ0030, University of Heidelberg; German Center for Infection Research (DZIF), German Center for Cardiovascular Research (DZHK), partner site, Heidelberg, Germany
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
15
|
Vadibeler S. The (unresolved) antibody paradox. J Clin Invest 2024; 134:e183886. [PMID: 39007262 PMCID: PMC11245148 DOI: 10.1172/jci183886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
|
16
|
Darpel KE, Corla A, Stedman A, Bellamy F, Flannery J, Rajko-Nenow P, Powers C, Wilson S, Charleston B, Baron MD, Batten C. Long-term trial of protection provided by adenovirus-vectored vaccine expressing the PPRV H protein. NPJ Vaccines 2024; 9:98. [PMID: 38830899 PMCID: PMC11148195 DOI: 10.1038/s41541-024-00892-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
A recombinant, replication-defective, adenovirus-vectored vaccine expressing the H surface glycoprotein of peste des petits ruminants virus (PPRV) has previously been shown to protect goats from challenge with wild-type PPRV at up to 4 months post vaccination. Here, we present the results of a longer-term trial of the protection provided by such a vaccine, challenging animals at 6, 9, 12 and 15 months post vaccination. Vaccinated animals developed high levels of anti-PPRV H protein antibodies, which were virus-neutralising, and the level of these antibodies was maintained for the duration of the trial. The vaccinated animals were largely protected against overt clinical disease from the challenge virus. Although viral genome was intermittently detected in blood samples, nasal and/or ocular swabs of vaccinated goats post challenge, viral RNA levels were significantly lower compared to unvaccinated control animals and vaccinated goats did not appear to excrete live virus. This protection, like the antibody response, was maintained at the same level for at least 15 months after vaccination. In addition, we showed that animals that have been vaccinated with the adenovirus-based vaccine can be revaccinated with the same vaccine after 12 months and showed an increased anti-PPRV antibody response after this boost vaccination. Such vaccines, which provide a DIVA capability, would therefore be suitable for use when the current live attenuated PPRV vaccines are withdrawn at the end of the ongoing global PPR eradication campaign.
Collapse
Affiliation(s)
- Karin E Darpel
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Amanda Corla
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK
| | - Anna Stedman
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK
- Veterinary Medicines Directorate, Woodham Lane, Addlestone, Surrey, KT15 3LS, UK
| | | | - John Flannery
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK
- Department of Pharmaceutical Sciences and Biotechnology, Technological University of the Shannon, Athlone, Ireland
| | - Paulina Rajko-Nenow
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK
- Department of Pharmaceutical Sciences and Biotechnology, Technological University of the Shannon, Athlone, Ireland
| | - Claire Powers
- Viral Vector Core Facility, Pandemic Sciences Institute, Oxford University, Oxford, UK
| | - Steve Wilson
- Global Alliance for Livestock Veterinary Medicines, Edinburgh, UK
| | - Bryan Charleston
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK
| | - Michael D Baron
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK
| | - Carrie Batten
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK.
| |
Collapse
|
17
|
Wellford SA, Moseman EA. Olfactory immunology: the missing piece in airway and CNS defence. Nat Rev Immunol 2024; 24:381-398. [PMID: 38097777 PMCID: PMC11560121 DOI: 10.1038/s41577-023-00972-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2023] [Indexed: 12/23/2023]
Abstract
The olfactory mucosa is a component of the nasal airway that mediates the sense of smell. Recent studies point to an important role for the olfactory mucosa as a barrier to both respiratory pathogens and to neuroinvasive pathogens that hijack the olfactory nerve and invade the CNS. In particular, the COVID-19 pandemic has demonstrated that the olfactory mucosa is an integral part of a heterogeneous nasal mucosal barrier critical to upper airway immunity. However, our insufficient knowledge of olfactory mucosal immunity hinders attempts to protect this tissue from infection and other diseases. This Review summarizes the state of olfactory immunology by highlighting the unique immunologically relevant anatomy of the olfactory mucosa, describing what is known of olfactory immune cells, and considering the impact of common infectious diseases and inflammatory disorders at this site. We will offer our perspective on the future of the field and the many unresolved questions pertaining to olfactory immunity.
Collapse
Affiliation(s)
- Sebastian A Wellford
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - E Ashley Moseman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
18
|
Göttig L, Schreiner S. E4orf1: The triple agent of adenovirus - Unraveling its roles in oncogenesis, infectious obesity and immune responses in virus replication and vector therapy. Tumour Virus Res 2024; 17:200277. [PMID: 38428735 PMCID: PMC10937242 DOI: 10.1016/j.tvr.2024.200277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
Human Adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous sub-types that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating cellular pathways such as PI3K-Akt-mTOR, Ras, the immune response and further HAdV replication stages than previously anticipated. In this review, we aim to explore the structure, molecular mechanisms, and biological functions of E4orf1, shedding light on its potentially multifaceted roles during HAdV infection, including metabolic diseases and oncogenesis. Furthermore, we discuss the role of functional E4orf1 in biotechnological applications such as Adenovirus (AdV) vaccine vectors and oncolytic AdV. By dissecting the intricate relationships between HAdV types and E4orf1 proteins, this review provides valuable insights into viral pathogenesis and points to promising areas of future research.
Collapse
Affiliation(s)
- Lilian Göttig
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
| | - Sabrina Schreiner
- Institute of Virology, School of Medicine, Technical University of Munich, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (Resolving Infection Susceptibility; EXC 2155), Hannover, Germany; Institute of Virology, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
19
|
King CR, Dodge MJ, MacNeil KM, Tessier TM, Mymryk JS, Mehle A. Expanding the adenovirus toolbox: reporter viruses for studying the dynamics of human adenovirus replication. J Virol 2024; 98:e0020724. [PMID: 38639487 PMCID: PMC11092356 DOI: 10.1128/jvi.00207-24] [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: 01/30/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
To streamline standard virological assays, we developed a suite of nine fluorescent or bioluminescent replication competent human species C5 adenovirus reporter viruses that mimic their parental wild-type counterpart. These reporter viruses provide a rapid and quantitative readout of various aspects of viral infection and replication based on EGFP, mCherry, or NanoLuc measurement. Moreover, they permit real-time non-invasive measures of viral load, replication dynamics, and infection kinetics over the entire course of infection, allowing measurements that were not previously possible. This suite of replication competent reporter viruses increases the ease, speed, and adaptability of standard assays and has the potential to accelerate multiple areas of human adenovirus research.IMPORTANCEIn this work, we developed a versatile toolbox of nine HAdV-C5 reporter viruses and validated their functions in cell culture. These reporter viruses provide a rapid and quantitative readout of various aspects of viral infection and replication based on EGFP, mCherry, or NanoLuc measurement. The utility of these reporter viruses could also be extended for use in 3D cell culture, organoids, live cell imaging, or animal models, and provides a conceptual framework for the development of new reporter viruses representing other clinically relevant HAdV species.
Collapse
Affiliation(s)
- Cason R. King
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mackenzie J. Dodge
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Katelyn M. MacNeil
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Tanner M. Tessier
- Division of Protective Immunity, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Joe S. Mymryk
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
- Department of Otolaryngology, University of Western Ontario, London, Ontario, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Andrew Mehle
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
20
|
Brunet J, Choucha Z, Gransagne M, Tabbal H, Ku MW, Buchrieser J, Fernandes P, Batalie D, Lopez J, Ma L, Dufour E, Simon E, Hardy D, Petres S, Guinet F, Strick-Marchand H, Monot M, Charneau P, Majlessi L, Duprex WP, Gerke C, Martin A, Escriou N. A measles-vectored vaccine candidate expressing prefusion-stabilized SARS-CoV-2 spike protein brought to phase I/II clinical trials: candidate selection in a preclinical murine model. J Virol 2024; 98:e0169323. [PMID: 38563763 PMCID: PMC11210269 DOI: 10.1128/jvi.01693-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
In the early COVID-19 pandemic with urgent need for countermeasures, we aimed at developing a replicating viral vaccine using the highly efficacious measles vaccine as vector, a promising technology with prior clinical proof of concept. Building on our successful pre-clinical development of a measles virus (MV)-based vaccine candidate against the related SARS-CoV, we evaluated several recombinant MV expressing codon-optimized SARS-CoV-2 spike glycoprotein. Candidate V591 expressing a prefusion-stabilized spike through introduction of two proline residues in HR1 hinge loop, together with deleted S1/S2 furin cleavage site and additional inactivation of the endoplasmic reticulum retrieval signal, was the most potent in eliciting neutralizing antibodies in mice. After single immunization, V591 induced similar neutralization titers as observed in sera of convalescent patients. The cellular immune response was confirmed to be Th1 skewed. V591 conferred long-lasting protection against SARS-CoV-2 challenge in a murine model with marked decrease in viral RNA load, absence of detectable infectious virus loads, and reduced lesions in the lungs. V591 was furthermore efficacious in an established non-human primate model of disease (see companion article [S. Nambulli, N. Escriou, L. J. Rennick, M. J. Demers, N. L. Tilston-Lunel et al., J Virol 98:e01762-23, 2024, https://doi.org/10.1128/jvi.01762-23]). Thus, V591 was taken forward into phase I/II clinical trials in August 2020. Unexpected low immunogenicity in humans (O. Launay, C. Artaud, M. Lachâtre, M. Ait-Ahmed, J. Klein et al., eBioMedicine 75:103810, 2022, https://doi.org/10.1016/j.ebiom.2021.103810) revealed that the underlying mechanisms for resistance or sensitivity to pre-existing anti-measles immunity are not yet understood. Different hypotheses are discussed here, which will be important to investigate for further development of the measles-vectored vaccine platform.IMPORTANCESARS-CoV-2 emerged at the end of 2019 and rapidly spread worldwide causing the COVID-19 pandemic that urgently called for vaccines. We developed a vaccine candidate using the highly efficacious measles vaccine as vector, a technology which has proved highly promising in clinical trials for other pathogens. We report here and in the companion article by Nambulli et al. (J Virol 98:e01762-23, 2024, https://doi.org/10.1128/jvi.01762-23) the design, selection, and preclinical efficacy of the V591 vaccine candidate that was moved into clinical development in August 2020, 7 months after the identification of SARS-CoV-2 in Wuhan. These unique in-human trials of a measles vector-based COVID-19 vaccine revealed insufficient immunogenicity, which may be the consequence of previous exposure to the pediatric measles vaccine. The three studies together in mice, primates, and humans provide a unique insight into the measles-vectored vaccine platform, raising potential limitations of surrogate preclinical models and calling for further refinement of the platform.
Collapse
Affiliation(s)
- Jérémy Brunet
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, Paris, France
| | - Zaineb Choucha
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, Paris, France
| | - Marion Gransagne
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, Paris, France
| | - Houda Tabbal
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Min-Wen Ku
- Institut Pasteur, Université Paris Cité, Pasteur-TheraVectys Joint Lab, Paris, France
| | - Julian Buchrieser
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Virus and Immunity Unit, Paris, France
| | - Priyanka Fernandes
- Institut Pasteur, Université Paris Cité, INSERM U1223, Innate Immunity Unit, Paris, France
| | - Damien Batalie
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Jodie Lopez
- Institut Pasteur, Université Paris Cité, Pasteur-TheraVectys Joint Lab, Paris, France
| | - Laurence Ma
- Institut Pasteur, Université Paris Cité, Biomics, C2RT, Paris, France
| | - Evelyne Dufour
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Production and Purification of Recombinant Proteins Technological Platform, Paris, France
| | - Emeline Simon
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - David Hardy
- Institut Pasteur, Université Paris Cité, Histopathology Platform, Paris, France
| | - Stéphane Petres
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Production and Purification of Recombinant Proteins Technological Platform, Paris, France
| | - Françoise Guinet
- Institut Pasteur, Université Paris Cité, INSERM U1223, Lymphocytes and Immunity Unit, Paris, France
| | - Helene Strick-Marchand
- Institut Pasteur, Université Paris Cité, INSERM U1223, Innate Immunity Unit, Paris, France
| | - Marc Monot
- Institut Pasteur, Université Paris Cité, Biomics, C2RT, Paris, France
| | - Pierre Charneau
- Institut Pasteur, Université Paris Cité, Pasteur-TheraVectys Joint Lab, Paris, France
| | - Laleh Majlessi
- Institut Pasteur, Université Paris Cité, Pasteur-TheraVectys Joint Lab, Paris, France
| | - W. Paul Duprex
- Center for Vaccine Research, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christiane Gerke
- Institut Pasteur, Université Paris Cité, Innovation Office, Vaccine Programs, Paris, France
| | - Annette Martin
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Nicolas Escriou
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, Paris, France
| |
Collapse
|
21
|
Nambulli S, Escriou N, Rennick LJ, Demers MJ, Tilston‑Lunel NL, McElroy AK, Barbeau DJ, Crossland NA, Hoehl RM, Schrauf S, White AG, Borish HJ, Tomko JA, Frye LJ, Scanga CA, Flynn JL, Martin A, Gerke C, Hartman AL, Duprex WP. A measles-vectored vaccine candidate expressing prefusion-stabilized SARS-CoV-2 spike protein brought to phase I/II clinical trials: protection of African green monkeys from COVID-19 disease. J Virol 2024; 98:e0176223. [PMID: 38563762 PMCID: PMC11092351 DOI: 10.1128/jvi.01762-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and is responsible for the largest human pandemic in 100 years. Thirty-four vaccines are currently approved for use worldwide, and approximately 67% of the world population has received a complete primary series of one, yet countries are dealing with new waves of infections, variant viruses continue to emerge, and breakthrough infections are frequent secondary to waning immunity. Here, we evaluate a measles virus (MV)-vectored vaccine expressing a stabilized prefusion SARS-CoV-2 spike (S) protein (MV-ATU3-S2PΔF2A; V591) with demonstrated immunogenicity in mouse models (see companion article [J. Brunet, Z. Choucha, M. Gransagne, H. Tabbal, M.-W. Ku et al., J Virol 98:e01693-23, 2024, https://doi.org/10.1128/jvi.01693-23]) in an established African green monkey model of disease. Animals were vaccinated with V591 or the control vaccine (an equivalent MV-vectored vaccine with an irrelevant antigen) intramuscularly using a prime/boost schedule, followed by challenge with an early pandemic isolate of SARS-CoV-2 at 56 days post-vaccination. Pre-challenge, only V591-vaccinated animals developed S-specific antibodies that had virus-neutralizing activity as well as S-specific T cells. Following the challenge, V591-vaccinated animals had lower infectious virus and viral (v) RNA loads in mucosal secretions and stopped shedding virus in these secretions earlier. vRNA loads were lower in these animals in respiratory and gastrointestinal tract tissues at necropsy. This correlated with a lower disease burden in the lungs as quantified by PET/CT at early and late time points post-challenge and by pathological analysis at necropsy.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the largest human pandemic in 100 years. Even though vaccines are currently available, countries are dealing with new waves of infections, variant viruses continue to emerge, breakthrough infections are frequent, and vaccine hesitancy persists. This study uses a safe and effective measles vaccine as a platform for vaccination against SARS-CoV-2. The candidate vaccine was used to vaccinate African green monkeys (AGMs). All vaccinated AGMs developed robust antigen-specific immune responses. After challenge, these AGMs produced less virus in mucosal secretions, for a shorter period, and had a reduced disease burden in the lungs compared to control animals. At necropsy, lower levels of viral RNA were detected in tissue samples from vaccinated animals, and the lungs of these animals lacked the histologic hallmarks of SARS-CoV-2 disease observed exclusively in the control AGMs.
Collapse
MESH Headings
- Animals
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Chlorocebus aethiops
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19/virology
- Measles virus/immunology
- Measles virus/genetics
- COVID-19 Vaccines/immunology
- Humans
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Genetic Vectors
- Vero Cells
- Pandemics/prevention & control
- Female
- Betacoronavirus/immunology
- Betacoronavirus/genetics
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/virology
- Pneumonia, Viral/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Coronavirus Infections/veterinary
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Disease Models, Animal
Collapse
Affiliation(s)
- Sham Nambulli
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicolas Escriou
- Département de Santé Globale, Institut Pasteur, Université de Paris Cite, Paris, France
| | - Linda J. Rennick
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Matthew J. Demers
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Natasha L. Tilston‑Lunel
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anita K. McElroy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dominique J. Barbeau
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicholas A. Crossland
- National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sabrina Schrauf
- Themis Bioscience GmbH, a subsidiary of Merck & Co., Inc., Rahway, New Jersey, USA
| | - Alexander G. White
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - H. Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jaime A. Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lonnie J. Frye
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Charles A. Scanga
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - JoAnne L. Flynn
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Annette Martin
- CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Institut Pasteur, Université de Paris, Paris, France
| | - Christiane Gerke
- Vaccine Programs, Institut Pasteur, Université de Paris Cite, Innovation Office, Paris, France
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - W. Paul Duprex
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
22
|
Nour D, Ismail MB, Osman M, Rafei R, Kasir D, Dabboussi F, Colson P, Hamze M. Evaluation of SARS-CoV-2 anti-Spike antibody levels and breakthrough infection risk among vaccinated adults in North Lebanon. PLoS One 2024; 19:e0302579. [PMID: 38722969 PMCID: PMC11081361 DOI: 10.1371/journal.pone.0302579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 04/04/2024] [Indexed: 05/13/2024] Open
Abstract
Since March 2020, the COVID-19 pandemic has swiftly propagated, triggering a competitive race among medical firms to forge vaccines that thwart the infection. Lebanon initiated its vaccination campaign on February 14, 2021. Despite numerous studies conducted to elucidate the characteristics of immune responses elicited by vaccination, the topic remains unclear. Here, we aimed to track the progression of anti-spike SARS-CoV-2 antibody titers at two-time points (T1: shortly after the second vaccination dose, T2: six months later) within a cohort of 201 adults who received Pfizer-BioNTech (BNT162b2), AstraZeneca, or Sputnik V vaccines in North Lebanon. Blood specimens were obtained from participants, and antibody titers against SARS-CoV-2 were quantified through the Elecsys-Anti-SARS-CoV-2 S assay (Roche Diagnostics, Switzerland). We used univariate analysis and multivariable logistic regression models to predict determinants influencing the decline in immune response and the occurrence of breakthrough infections among vaccinated patients. Among the 201 participants, 141 exhibited unchanging levels of antibody titers between the two sample collections, 55 displayed waning antibody titers, and only five participants demonstrated heightened antibody levels. Notably, age emerged as the sole variable significantly linked to the waning immune response. Moreover, the BNT162b2 vaccine exhibited significantly higher efficacy concerning the occurrence of breakthrough infections when compared with the AstraZeneca vaccine. Overall, our study reflected the immune status of a sample of vaccinated adults in North Lebanon. Further studies on a larger scale are needed at the national level to follow the immune response after vaccination, especially after the addition of the third vaccination dose.
Collapse
Affiliation(s)
- Dalal Nour
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Science & Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
- Aix-Marseille Univ., Institut de Recherche pour le Développement (IRD), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Mohamad Bachar Ismail
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Science & Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
- Faculty of Sciences, Lebanese University, Tripoli, Lebanon
| | - Marwan Osman
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States of America
| | - Rayane Rafei
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Science & Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Dalal Kasir
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Science & Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Fouad Dabboussi
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Science & Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Philippe Colson
- Aix-Marseille Univ., Institut de Recherche pour le Développement (IRD), Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
- IHU Méditerranée Infection, Marseille, France
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Marseille, France
| | - Monzer Hamze
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School of Science & Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| |
Collapse
|
23
|
Metz C, Haug V, Müller M, Amann R. Pharmacokinetic and Environmental Risk Assessment of Prime-2-CoV, a Non-Replicating Orf Virus-Based Vaccine against SARS-CoV-2. Vaccines (Basel) 2024; 12:492. [PMID: 38793743 PMCID: PMC11126055 DOI: 10.3390/vaccines12050492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Viral vector vaccines represent a substantial advancement in immunization technology, offering numerous benefits over traditional vaccine modalities. The Orf virus (ORFV) strain D1701-VrV is a particularly promising candidate for vaccine development due to its distinctive attributes, such as a good safety profile, the ability to elicit both humoral and cellular immunity, and its favorable genetic and thermal stability. Despite ORFV's theoretical safety advantages, such as its narrow host range and limited systemic spread post-inoculation, a critical gap persists between these theoretical benefits and the empirical evidence regarding its in vivo safety profile. This discrepancy underscores the need for comprehensive preclinical validations to bridge this knowledge gap, especially considering ORFV's use in humans. Our research introduces Prime-2-CoV, an innovative ORFV-based vaccine candidate against COVID-19, designed to elicit a robust immune response by expressing SARS-CoV-2 Nucleocapsid and Spike proteins. Currently under clinical trials, Prime-2-CoV marks the inaugural application of ORFV in human subjects. Addressing the aforementioned safety concerns, our extensive preclinical evaluation, including an environmental risk assessment (ERA) and detailed pharmacokinetic studies in rats and immunocompromised NOG mice, demonstrates Prime-2-CoV's favorable pharmacokinetic profile, negligible environmental impact, and minimal ERA risks. These findings not only affirm the vaccine's safety and efficacy but also pioneer the use of ORFV-based therapeutics, highlighting its potential for wider therapeutic applications.
Collapse
Affiliation(s)
- Carina Metz
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
- Institute for Tropical Medicine, Travel Medicine, and Human Parasitology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Verena Haug
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
- Institute for Tropical Medicine, Travel Medicine, and Human Parasitology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Melanie Müller
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
| | - Ralf Amann
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
| |
Collapse
|
24
|
Ramos JRC, Pinto J, Poiares-Oliveira G, Peeters L, Dumas P, Oliveira R. Deep hybrid modeling of a HEK293 process: Combining long short-term memory networks with first principles equations. Biotechnol Bioeng 2024; 121:1554-1568. [PMID: 38343176 DOI: 10.1002/bit.28668] [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: 08/21/2023] [Revised: 12/22/2023] [Accepted: 01/22/2024] [Indexed: 04/14/2024]
Abstract
The combination of physical equations with deep learning is becoming a promising methodology for bioprocess digitalization. In this paper, we investigate for the first time the combination of long short-term memory (LSTM) networks with first principles equations in a hybrid workflow to describe human embryonic kidney 293 (HEK293) culture dynamics. Experimental data of 27 extracellular state variables in 20 fed-batch HEK293 cultures were collected in a parallel high throughput 250 mL cultivation system in an industrial process development setting. The adaptive moment estimation method with stochastic regularization and cross-validation were employed for deep learning. A total of 784 hybrid models with varying deep neural network architectures, depths, layers sizes and node activation functions were compared. In most scenarios, hybrid LSTM models outperformed classical hybrid Feedforward Neural Network (FFNN) models in terms of training and testing error. Hybrid LSTM models revealed to be less sensitive to data resampling than FFNN hybrid models. As disadvantages, Hybrid LSTM models are in general more complex (higher number of parameters) and have a higher computation cost than FFNN hybrid models. The hybrid model with the highest prediction accuracy consisted in a LSTM network with seven internal states connected in series with dynamic material balance equations. This hybrid model correctly predicted the dynamics of the 27 state variables (R2 = 0.93 in the test data set), including biomass, key substrates, amino acids and metabolic by-products for around 10 cultivation days.
Collapse
Affiliation(s)
- João R C Ramos
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - José Pinto
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Gil Poiares-Oliveira
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | | | | | - Rui Oliveira
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| |
Collapse
|
25
|
Kaiser JA, Nelson CE, Liu X, Park HS, Matsuoka Y, Luongo C, Santos C, Ahlers LRH, Herbert R, Moore IN, Wilder-Kofie T, Moore R, Walker A, Yang L, Munir S, Teng IT, Kwong PD, Dowdell K, Nguyen H, Kim J, Cohen JI, Johnson RF, Garza NL, Via LE, Barber DL, Buchholz UJ, Le Nouën C. Mucosal prime-boost immunization with live murine pneumonia virus-vectored SARS-CoV-2 vaccine is protective in macaques. Nat Commun 2024; 15:3553. [PMID: 38670948 PMCID: PMC11053155 DOI: 10.1038/s41467-024-47784-6] [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: 08/19/2023] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Immunization via the respiratory route is predicted to increase the effectiveness of a SARS-CoV-2 vaccine. Here, we evaluate the immunogenicity and protective efficacy of one or two doses of a live-attenuated murine pneumonia virus vector expressing SARS-CoV-2 prefusion-stabilized spike protein (MPV/S-2P), delivered intranasally/intratracheally to male rhesus macaques. A single dose of MPV/S-2P is highly immunogenic, and a second dose increases the magnitude and breadth of the mucosal and systemic anti-S antibody responses and increases levels of dimeric anti-S IgA in the airways. MPV/S-2P also induces S-specific CD4+ and CD8+ T-cells in the airways that differentiate into large populations of tissue-resident memory cells within a month after the boost. One dose induces substantial protection against SARS-CoV-2 challenge, and two doses of MPV/S-2P are fully protective against SARS-CoV-2 challenge virus replication in the airways. A prime/boost immunization with a mucosally-administered live-attenuated MPV vector could thus be highly effective in preventing SARS-CoV-2 infection and replication.
Collapse
Affiliation(s)
- Jaclyn A Kaiser
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christine E Nelson
- T-Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Xueqiao Liu
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hong-Su Park
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yumiko Matsuoka
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cindy Luongo
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Celia Santos
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Laura R H Ahlers
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard Herbert
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, MD, USA
| | - Ian N Moore
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Temeri Wilder-Kofie
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Division of Assurances, Office of Laboratory Animal Welfare, National Institutes of Health, Bethesda, MD, USA
| | - Rashida Moore
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Emory National Primate Research Center, Environmental Health and Safety Office, Emory University, Atlanta, GA, USA
| | - April Walker
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lijuan Yang
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shirin Munir
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kennichi Dowdell
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hanh Nguyen
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - JungHyun Kim
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey I Cohen
- Medical Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Reed F Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicole L Garza
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Laura E Via
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel L Barber
- T-Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ursula J Buchholz
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Cyril Le Nouën
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
26
|
Sanchez-Martinez ZV, Alpuche-Lazcano SP, Stuible M, Durocher Y. CHO cells for virus-like particle and subunit vaccine manufacturing. Vaccine 2024; 42:2530-2542. [PMID: 38503664 DOI: 10.1016/j.vaccine.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
Chinese Hamster Ovary (CHO) cells, employed primarily for manufacturing monoclonal antibodies and other recombinant protein (r-protein) therapeutics, are emerging as a promising host for vaccine antigen production. This is exemplified by the recently approved CHO cell-derived subunit vaccines (SUV) against respiratory syncytial virus (RSV) and varicella-zoster virus (VZV), as well as the enveloped virus-like particle (eVLP) vaccine against hepatitis B virus (HBV). Here, we summarize the design, production, and immunogenicity features of these vaccine and review the most recent progress of other CHO-derived vaccines in pre-clinical and clinical development. We also discuss the challenges associated with vaccine production in CHO cells, with a focus on ensuring viral clearance for eVLP products.
Collapse
Affiliation(s)
- Zalma V Sanchez-Martinez
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC H2X 3Y7, Canada.
| |
Collapse
|
27
|
Zhu C, Pang S, Liu J, Duan Q. Current Progress, Challenges and Prospects in the Development of COVID-19 Vaccines. Drugs 2024; 84:403-423. [PMID: 38652356 DOI: 10.1007/s40265-024-02013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 04/25/2024]
Abstract
The COVID-19 pandemic has resulted in over 772 million confirmed cases, including nearly 7 million deaths, according to the World Health Organization (WHO). Leveraging rapid development, accelerated vaccine approval processes, and large-scale production of various COVID-19 vaccines using different technical platforms, the WHO declared an end to the global health emergency of COVID-19 on May 5, 2023. Current COVID-19 vaccines encompass inactivated, live attenuated, viral vector, protein subunit, nucleic acid (DNA and RNA), and virus-like particle (VLP) vaccines. However, the efficacy of these vaccines is diminishing due to the constant mutation of SARS-CoV-2 and the heightened immune evasion abilities of emerging variants. This review examines the impact of the COVID-19 pandemic, the biological characteristics of the virus, and its diverse variants. Moreover, the review underscores the effectiveness, advantages, and disadvantages of authorized COVID-19 vaccines. Additionally, it analyzes the challenges, strategies, and future prospects of developing a safe, broad-spectrum vaccine that confers sufficient and sustainable immune protection against new variants of SARS-CoV-2. These discussions not only offer insight for the development of next-generation COVID-19 vaccines but also summarize experiences for combating future emerging viruses.
Collapse
Affiliation(s)
- Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510000, China
| | - Shengmei Pang
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Jiaqi Liu
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Qiangde Duan
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
28
|
Hromić-Jahjefendić A, Lundstrom K, Adilović M, Aljabali AAA, Tambuwala MM, Serrano-Aroca Á, Uversky VN. Autoimmune response after SARS-CoV-2 infection and SARS-CoV-2 vaccines. Autoimmun Rev 2024; 23:103508. [PMID: 38160960 DOI: 10.1016/j.autrev.2023.103508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
The complicated relationships between autoimmunity, COVID-19, and COVID-19 vaccinations are described, giving insight into their intricacies. Antinuclear antibodies (ANA), anti-Ro/SSA, rheumatoid factor, lupus anticoagulant, and antibodies against interferon (IFN)-I have all been consistently found in COVID-19 patients, indicating a high prevalence of autoimmune reactions following viral exposure. Furthermore, the discovery of human proteins with structural similarities to SARS-CoV-2 peptides as possible autoantigens highlights the complex interplay between the virus and the immune system in initiating autoimmunity. An updated summary of the current status of COVID-19 vaccines is presented. We present probable pathways underpinning the genesis of COVID-19 autoimmunity, such as bystander activation caused by hyperinflammatory conditions, viral persistence, and the creation of neutrophil extracellular traps. These pathways provide important insights into the development of autoimmune-related symptoms ranging from organ-specific to systemic autoimmune and inflammatory illnesses, demonstrating the wide influence of COVID-19 on the immune system.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | | | - Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan.
| | - Murtaza M Tambuwala
- Lincoln Medical School, Brayford Pool Campus, University of Lincoln, Lincoln LN6 7TS, UK.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001, Valencia, Spain.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
29
|
Manfrini N, Notarbartolo S, Grifantini R, Pesce E. SARS-CoV-2: A Glance at the Innate Immune Response Elicited by Infection and Vaccination. Antibodies (Basel) 2024; 13:13. [PMID: 38390874 PMCID: PMC10885122 DOI: 10.3390/antib13010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/13/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
The COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has led to almost seven million deaths worldwide. SARS-CoV-2 causes infection through respiratory transmission and can occur either without any symptoms or with clinical manifestations which can be mild, severe or, in some cases, even fatal. Innate immunity provides the initial defense against the virus by sensing pathogen-associated molecular patterns and triggering signaling pathways that activate the antiviral and inflammatory responses, which limit viral replication and help the identification and removal of infected cells. However, temporally dysregulated and excessive activation of the innate immune response is deleterious for the host and associates with severe COVID-19. In addition to its defensive role, innate immunity is pivotal in priming the adaptive immune response and polarizing its effector function. This capacity is relevant in the context of both SARS-CoV-2 natural infection and COVID-19 vaccination. Here, we provide an overview of the current knowledge of the innate immune responses to SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- Nicola Manfrini
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Samuele Notarbartolo
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Renata Grifantini
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
- CheckmAb Srl, 20122 Milan, Italy
| | - Elisa Pesce
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
30
|
Rostami Abusaeedi S, Kheirkhah Vakilabad AA, Movahed E, Zareipour M, Mashayekhi V, Shafiei S, Sohrani Y, Rafeie H, Gheyabi F. Investigation of the Side Effects of COVID-19 Vaccines in Southern Iran: A Cross-Sectional Study. ARCHIVES OF RAZI INSTITUTE 2024; 79:145-153. [PMID: 39192959 PMCID: PMC11345472 DOI: 10.32592/ari.2024.79.1.145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/05/2023] [Indexed: 08/29/2024]
Abstract
The COVID-19 disease is a newly emerging disease, and the COVID-19 vaccine is one of the necessities to prevent this disease. The present study aimed to investigate the side effects of COVID-19 vaccines in southern Iran. We used convenience sampling to conduct this cross-sectional study on 647 people living in cities under coverage in Kerman province, southern Iran. The data collection tool was a researcher-made questionnaire of vaccine symptoms and signs. The results were analyzed using ANOVA and Chi-squared tests by SPSS software (version 24). The mean age of the participants was 40.19±15.20. The results indicated that 431 people (66.6%) reported post-vaccination side effects, with 18.23% of them having severe side effects. We noticed the most severe side effects in AstraZeneca, Sinopharm, Sputnik, and Bharat. Fever, headache, and pain at the injection site were the most common side effects after vaccination in descending order, which had a statistically significant relationship with all types of vaccines (P=0.001). The side effects differed in the types of vaccines, and most of the vaccines had mild to moderate side effects. People with the B blood type showed the most severe side effects, while those with the AB showed the lowest rate of side effects. Therefore, the injection of the AstraZeneca vaccine in blood group B should be done with more caution. More attention should also be paid to blood groups B and A in the injection of COVID-19 vaccines. Moreover, health officials and the government should plan appropriate educational strategies to increase public awareness of the importance of vaccines in eradicating viral infections.
Collapse
Affiliation(s)
- S Rostami Abusaeedi
- Department of Clinical Sciences, School of Medicine, Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - A A Kheirkhah Vakilabad
- Department of Internal Medicine, School of Medicine, Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - E Movahed
- Department of Public Health, School of Public Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - M Zareipour
- Department of Public Health, School of Health, Khoy University of Medical Sciences, Khoy, Iran
| | - V Mashayekhi
- Deputy of Health, Jiroft University of Medical University, Jiroft, Iran
| | - S Shafiei
- Department of Clinical Sciences, Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Y Sohrani
- Clinical Research Development Unit, Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - H Rafeie
- Clinical Research Development Unit, Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - F Gheyabi
- Department of Public Health, School of Public Health, Jiroft University of Medical Sciences, Jiroft, Iran
| |
Collapse
|
31
|
Zhang D, Kukkar D, Kim KH, Bhatt P. A comprehensive review on immunogen and immune-response proteins of SARS-CoV-2 and their applications in prevention, diagnosis, and treatment of COVID-19. Int J Biol Macromol 2024; 259:129284. [PMID: 38211928 DOI: 10.1016/j.ijbiomac.2024.129284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Exposure to severe acute respiratory syndrome-corona virus-2 (SARS-CoV-2) prompts humoral immune responses in the human body. As the auxiliary diagnosis of a current infection, the existence of viral proteins can be checked from specific antibodies (Abs) induced by immunogenic viral proteins. For people with a weakened immune system, Ab treatment can help neutralize viral antigens to resist and treat the disease. On the other hand, highly immunogenic viral proteins can serve as effective markers for detecting prior infections. Additionally, the identification of viral particles or the presence of antibodies may help establish an immune defense against the virus. These immunogenic proteins rather than SARS-CoV-2 can be given to uninfected people as a vaccination to improve their coping ability against COVID-19 through the generation of memory plasma cells. In this work, we review immunogenic and immune-response proteins derived from SARS-CoV-2 with regard to their classification, origin, and diverse applications (e.g., prevention (vaccine development), diagnostic testing, and treatment (via neutralizing Abs)). Finally, advanced immunization strategies against COVID-19 are discussed along with the contemporary circumstances and future challenges.
Collapse
Affiliation(s)
- Daohong Zhang
- College of Food Engineering, Ludong University, Yantai 264025, Shandong, China; Bio-Nanotechnology Research Institute, Ludong University, Yantai 264025, Shandong, China
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Ki-Hyun Kim
- Department of Civil & Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, Republic of Korea.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Center for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| |
Collapse
|
32
|
Wellford SA, Moseman EA. Olfactory immune response to SARS-CoV-2. Cell Mol Immunol 2024; 21:134-143. [PMID: 38143247 PMCID: PMC10806031 DOI: 10.1038/s41423-023-01119-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/04/2023] [Indexed: 12/26/2023] Open
Abstract
Numerous pathogens can infect the olfactory tract, yet the pandemic caused by SARS-CoV-2 has strongly emphasized the importance of the olfactory mucosa as an immune barrier. Situated in the nasal passages, the olfactory mucosa is directly exposed to the environment to sense airborne odorants; however, this also means it can serve as a direct route of entry from the outside world into the brain. As a result, olfactotropic infections can have serious consequences, including dysfunction of the olfactory system, CNS invasion, dissemination to the lower respiratory tract, and transmission between individuals. Recent research has shown that a distinctive immune response is needed to protect this neuronal and mucosal tissue. A better understanding of innate, adaptive, and structural immune barriers in the olfactory mucosa is needed to develop effective therapeutics and vaccines against olfactotropic microbes such as SARS-CoV-2. Here, we summarize the ramifications of SARS-CoV-2 infection of the olfactory mucosa, review the subsequent immune response, and discuss important areas of future research for olfactory immunity to infectious disease.
Collapse
Affiliation(s)
- Sebastian A Wellford
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - E Ashley Moseman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
33
|
Chen Z, Yuan Y, Hu Q, Zhu A, Chen F, Li S, Guan X, Lv C, Tang T, He Y, Cheng J, Zheng J, Hu X, Zhao J, Zhao J, Sun J. SARS-CoV-2 immunity in animal models. Cell Mol Immunol 2024; 21:119-133. [PMID: 38238440 PMCID: PMC10806257 DOI: 10.1038/s41423-023-01122-w] [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: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
The COVID-19 pandemic, which was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a worldwide health crisis due to its transmissibility. SARS-CoV-2 infection results in severe respiratory illness and can lead to significant complications in affected individuals. These complications encompass symptoms such as coughing, respiratory distress, fever, infectious shock, acute respiratory distress syndrome (ARDS), and even multiple-organ failure. Animal models serve as crucial tools for investigating pathogenic mechanisms, immune responses, immune escape mechanisms, antiviral drug development, and vaccines against SARS-CoV-2. Currently, various animal models for SARS-CoV-2 infection, such as nonhuman primates (NHPs), ferrets, hamsters, and many different mouse models, have been developed. Each model possesses distinctive features and applications. In this review, we elucidate the immune response elicited by SARS-CoV-2 infection in patients and provide an overview of the characteristics of various animal models mainly used for SARS-CoV-2 infection, as well as the corresponding immune responses and applications of these models. A comparative analysis of transcriptomic alterations in the lungs from different animal models revealed that the K18-hACE2 and mouse-adapted virus mouse models exhibited the highest similarity with the deceased COVID-19 patients. Finally, we highlighted the current gaps in related research between animal model studies and clinical investigations, underscoring lingering scientific questions that demand further clarification.
Collapse
Affiliation(s)
- Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yaochang Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Fenghua Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Shu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xin Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Chao Lv
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Tian Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yiyun He
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jinling Cheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jie Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xiaoyu Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518005, China.
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
34
|
Lundstrom K. COVID-19 Vaccines: Where Did We Stand at the End of 2023? Viruses 2024; 16:203. [PMID: 38399979 PMCID: PMC10893040 DOI: 10.3390/v16020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccine development against SARS-CoV-2 has been highly successful in slowing down the COVID-19 pandemic. A wide spectrum of approaches including vaccines based on whole viruses, protein subunits and peptides, viral vectors, and nucleic acids has been developed in parallel. For all types of COVID-19 vaccines, good safety and efficacy have been obtained in both preclinical animal studies and in clinical trials in humans. Moreover, emergency use authorization has been granted for the major types of COVID-19 vaccines. Although high safety has been demonstrated, rare cases of severe adverse events have been detected after global mass vaccinations. Emerging SARS-CoV-2 variants possessing enhanced infectivity have affected vaccine protection efficacy requiring re-design and re-engineering of novel COVID-19 vaccine candidates. Furthermore, insight is given into preparedness against emerging SARS-CoV-2 variants.
Collapse
|
35
|
Mayer L, Weskamm LM, Fathi A, Kono M, Heidepriem J, Krähling V, Mellinghoff SC, Ly ML, Friedrich M, Hardtke S, Borregaard S, Hesterkamp T, Loeffler FF, Volz A, Sutter G, Becker S, Dahlke C, Addo MM. MVA-based vaccine candidates encoding the native or prefusion-stabilized SARS-CoV-2 spike reveal differential immunogenicity in humans. NPJ Vaccines 2024; 9:20. [PMID: 38278816 PMCID: PMC10817990 DOI: 10.1038/s41541-023-00801-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/12/2023] [Indexed: 01/28/2024] Open
Abstract
In response to the COVID-19 pandemic, multiple vaccines were developed using platforms such as viral vectors and mRNA technology. Here, we report humoral and cellular immunogenicity data from human phase 1 clinical trials investigating two recombinant Modified Vaccinia virus Ankara vaccine candidates, MVA-SARS-2-S and MVA-SARS-2-ST, encoding the native and the prefusion-stabilized SARS-CoV-2 spike protein, respectively. MVA-SARS-2-ST was more immunogenic than MVA-SARS-2-S, but both were less immunogenic compared to licensed mRNA- and ChAd-based vaccines in SARS-CoV-2 naïve individuals. In heterologous vaccination, previous MVA-SARS-2-S vaccination enhanced T cell functionality and MVA-SARS-2-ST boosted the frequency of T cells and S1-specific IgG levels when used as a third vaccination. While the vaccine candidate containing the prefusion-stabilized spike elicited predominantly S1-specific responses, immunity to the candidate with the native spike was skewed towards S2-specific responses. These data demonstrate how the spike antigen conformation, using the same viral vector, directly affects vaccine immunogenicity in humans.
Collapse
Affiliation(s)
- Leonie Mayer
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.
| | - Leonie M Weskamm
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Anahita Fathi
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- First Department of Medicine, Division of Infectious Diseases, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Maya Kono
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Jasmin Heidepriem
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Verena Krähling
- Institute for Virology, Philipps University Marburg, Marburg, Germany
- German Centre for Infection Research, Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Sibylle C Mellinghoff
- Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Centre for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), German CLL Group (GCLLSG), University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - My Linh Ly
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Monika Friedrich
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Svenja Hardtke
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | | | - Thomas Hesterkamp
- German Centre for Infection Research, Translational Project Management Office, Brunswick, Germany
| | - Felix F Loeffler
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
- German Centre for Infection Research, Partner Site Hannover-Brunswick, Hanover, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Institute for Infectious Diseases and Zoonoses, LMU Munich, Munich, Germany
- German Centre for Infection Research, Partner Site Munich, Munich, Germany
| | - Stephan Becker
- Institute for Virology, Philipps University Marburg, Marburg, Germany
- German Centre for Infection Research, Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Christine Dahlke
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Marylyn M Addo
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
- German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.
| |
Collapse
|
36
|
Berry N, Mee ET, Almond N, Rose NJ. The Impact and Effects of Host Immunogenetics on Infectious Disease Studies Using Non-Human Primates in Biomedical Research. Microorganisms 2024; 12:155. [PMID: 38257982 PMCID: PMC10818626 DOI: 10.3390/microorganisms12010155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Understanding infectious disease pathogenesis and evaluating novel candidate treatment interventions for human use frequently requires prior or parallel analysis in animal model systems. While rodent species are frequently applied in such studies, there are situations where non-human primate (NHP) species are advantageous or required. These include studies of animals that are anatomically more akin to humans, where there is a need to interrogate the complexity of more advanced biological systems or simply reflect susceptibility to a specific infectious agent. The contribution of different arms of the immune response may be addressed in a variety of NHP species or subspecies in specific physiological compartments. Such studies provide insights into immune repertoires not always possible from human studies. However, genetic variation in outbred NHP models may confound, or significantly impact the outcome of a particular study. Thus, host factors need to be considered when undertaking such studies. Considerable knowledge of the impact of host immunogenetics on infection dynamics was elucidated from HIV/SIV research. NHP models are now important for studies of emerging infections. They have contributed to delineating the pathogenesis of SARS-CoV-2/COVID-19, which identified differences in outcomes attributable to the selected NHP host. Moreover, their use was crucial in evaluating the immunogenicity and efficacy of vaccines against COVID-19 and establishing putative correlates of vaccine protection. More broadly, neglected or highly pathogenic emerging or re-emergent viruses may be studied in selected NHPs. These studies characterise protective immune responses following infection or the administration of candidate immunogens which may be central to the accelerated licensing of new vaccines. Here, we review selected aspects of host immunogenetics, specifically MHC background and TRIM5 polymorphism as exemplars of adaptive and innate immunity, in commonly used Old and New World host species. Understanding this variation within and between NHP species will ensure that this valuable laboratory source is used most effectively to combat established and emerging virus infections and improve human health worldwide.
Collapse
Affiliation(s)
- Neil Berry
- Research & Development—Science, Research and Innovation, Medicines and Healthcare products Regulatory Agency, South Mimms, Hertfordshire EN6 3QG, UK; (E.T.M.); (N.A.); (N.J.R.)
| | | | | | | |
Collapse
|
37
|
Poria R, Kala D, Nagraik R, Dhir Y, Dhir S, Singh B, Kaushik NK, Noorani MS, Kaushal A, Gupta S. Vaccine development: Current trends and technologies. Life Sci 2024; 336:122331. [PMID: 38070863 DOI: 10.1016/j.lfs.2023.122331] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
Despite the effectiveness of vaccination in reducing or eradicating diseases caused by pathogens, there remain certain diseases and emerging infections for which developing effective vaccines is inherently challenging. Additionally, developing vaccines for individuals with compromised immune systems or underlying medical conditions presents significant difficulties. As well as traditional vaccine different methods such as inactivated or live attenuated vaccines, viral vector vaccines, and subunit vaccines, emerging non-viral vaccine technologies, including viral-like particle and nanoparticle vaccines, DNA/RNA vaccines, and rational vaccine design, offer new strategies to address the existing challenges in vaccine development. These advancements have also greatly enhanced our understanding of vaccine immunology, which will guide future vaccine development for a broad range of diseases, including rapidly emerging infectious diseases like COVID-19 and diseases that have historically proven resistant to vaccination. This review provides a comprehensive assessment of emerging non-viral vaccine production methods and their application in addressing the fundamental and current challenges in vaccine development.
Collapse
Affiliation(s)
- Renu Poria
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Deepak Kala
- Centera Laboratories, Institute of High Pressure Physics PAS, 01-142 Warsaw, Poland
| | - Rupak Nagraik
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
| | - Yashika Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Sunny Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Bharat Singh
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Naveen Kumar Kaushik
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, Uttar Pradesh, India
| | - Md Salik Noorani
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Ankur Kaushal
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India.
| | - Shagun Gupta
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India.
| |
Collapse
|
38
|
McKendry R, Lemm NM, Papargyris L, Chiu C. Human Challenge Studies with Coronaviruses Old and New. Curr Top Microbiol Immunol 2024; 445:69-108. [PMID: 35181805 DOI: 10.1007/82_2021_247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Coronavirus infections have been known to cause disease in animals since as early as the 1920s. However, only seven coronaviruses capable of causing human disease have been identified thus far. These Human Coronaviruses (HCoVs) include the causes of the common cold, but more recent coronaviruses that have emerged (i.e. SARS-CoV, MERS-CoV and SARS-CoV-2) are associated with much greater morbidity and mortality. HCoVs have been relatively under-studied compared to other common respiratory infections, as historically they have presented with mild symptoms. This has led to a relatively limited understanding of their animal reservoirs, transmission and determinants of immune protection. To address this, human infection challenge studies with HCoVs have been performed that enable a detailed clinical and immunological analysis of the host response at specific time points under controlled conditions with standardised viral inocula. Until recently, all such human challenge studies were conducted with common cold HCoVs, with the study of SARS-CoV and MERS-CoV unacceptable due to their greater pathogenicity. However, with the emergence of SARS-CoV-2 and the COVID-19 pandemic during which severe outcomes in young healthy adults have been rare, human challenge studies with SARS-CoV-2 are now being developed. Two SARS-CoV-2 human challenge studies in the UK studying individuals with and without pre-existing immunity are underway. As well as providing a platform for testing of antivirals and vaccines, such studies will be critical for understanding the factors associated with susceptibility to SARS-CoV-2 infection and thus developing improved strategies to tackle the current as well as future HCoV pandemics. Here, we summarise the major questions about protection and pathogenesis in HCoV infection that human infection challenge studies have attempted to answer historically, as well as the knowledge gaps that aim to be addressed with contemporary models.
Collapse
Affiliation(s)
- Richard McKendry
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nana-Marie Lemm
- Department of Infectious Disease, Imperial College London, London, UK
| | - Loukas Papargyris
- Department of Infectious Disease, Imperial College London, London, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
39
|
Joe CCD, Segireddy RR, Oliveira C, Berg A, Li Y, Doultsinos D, Scholze S, Ahmad A, Nestola P, Niemann J, Douglas AD. Accelerated and intensified manufacturing of an adenovirus-vectored vaccine to enable rapid outbreak response. Biotechnol Bioeng 2024; 121:176-191. [PMID: 37747758 DOI: 10.1002/bit.28553] [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: 04/20/2023] [Revised: 08/02/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023]
Abstract
The Coalition for Epidemic Preparedness Innovations' "100-day moonshot" aspires to launch a new vaccine within 100 days of pathogen identification, followed by large-scale vaccine availability within the "second hundred days." Here, we describe work to optimize adenoviral vector manufacturing for rapid response, by minimizing time to clinical trial and first large-scale supply, and maximizing output from the available manufacturing footprint. We describe a rapid virus seed expansion workflow that allows vaccine release to clinical trials within 60 days of antigen sequence identification, followed by vaccine release from globally distributed sites within a further 40 days. We also describe a perfusion-based upstream production process, designed to maximize output while retaining simplicity and suitability for existing manufacturing facilities. This improves upstream volumetric productivity of ChAdOx1 nCoV-19 by approximately fourfold and remains compatible with the existing downstream process, yielding drug substance sufficient for 10,000 doses from each liter of bioreactor capacity. This accelerated manufacturing process, along with other advantages such as thermal stability, supports the ongoing value of adenovirus-vectored vaccines as a rapidly adaptable and deployable platform for emergency response.
Collapse
Affiliation(s)
- Carina C D Joe
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Cathy Oliveira
- Clinical Biomanufacturing Facility, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Adam Berg
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Yuanyuan Li
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Dimitrios Doultsinos
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Asma Ahmad
- Repligen Corporation, Waltham, Massachusetts, USA
| | | | | | - Alexander D Douglas
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
40
|
Ao D, He X, Liu J, Xu L. Strategies for the development and approval of COVID-19 vaccines and therapeutics in the post-pandemic period. Signal Transduct Target Ther 2023; 8:466. [PMID: 38129394 PMCID: PMC10739883 DOI: 10.1038/s41392-023-01724-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/24/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in significant casualties and put immense strain on public health systems worldwide, leading to economic recession and social unrest. In response, various prevention and control strategies have been implemented globally, including vaccine and drug development and the promotion of preventive measures. Implementing these strategies has effectively curbed the transmission of the virus, reduced infection rates, and gradually restored normal social and economic activities. However, the mutations of SARS-CoV-2 have led to inevitable infections and reinfections, and the number of deaths continues to rise. Therefore, there is still a need to improve existing prevention and control strategies, mainly focusing on developing novel vaccines and drugs, expediting medical authorization processes, and keeping epidemic surveillance. These measures are crucial to combat the Coronavirus disease (COVID-19) pandemic and achieve sustained, long-term prevention, management, and disease control. Here, we summarized the characteristics of existing COVID-19 vaccines and drugs and suggested potential future directions for their development. Furthermore, we discussed the COVID-19-related policies implemented over the past years and presented some strategies for the future.
Collapse
Affiliation(s)
- Danyi Ao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Jian Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China
| | - Li Xu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
41
|
Hu S, Wu C, Wu X, Ma X, Shu C, Chen Q, Zheng A, Yang H, Lu J, Du P, Gao GF, Wang Q. Classification of five SARS-CoV-2 serotypes based on RBD antigenicities. Sci Bull (Beijing) 2023; 68:3003-3012. [PMID: 37919162 DOI: 10.1016/j.scib.2023.09.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/24/2023] [Accepted: 09/25/2023] [Indexed: 11/04/2023]
Abstract
The continuous evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in a significant number of variants, particularly with the emergence of Omicron with many sub-variants. These variants have exhibited increased immune escape, leading to reduced efficacy of existing vaccines and therapeutic antibodies. Given the diminished cross-neutralization observed among these variants, it is plausible that SARS-CoV-2 has developed multiple serotypes. As the major antigenic site, the receptor-binding domain (RBD) of viral spike (S) protein was chosen for serotyping. We selected 23 representative variants, including pre-Omicron variants and Omicron sub-variants, and classified them into five serotypes based on systematic evaluation of the antigenicities of their RBDs. Each serotype includes several genetically distinct variants. Serotype-I encompasses all pre-Omicron variants (with two subtypes), while the remaining four serotypes are all comprised of Omicron sub-variants at different stages of evolution. We propose that these serotypes can serve as a foundation for rapid classification of newly emerging SARS-CoV-2 variants, and guide the development of future broad-spectrum vaccines and neutralizing antibodies against the coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Shixiong Hu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chunli Wu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Xinkai Wu
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Xuehui Ma
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chang Shu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qian Chen
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Anqi Zheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huiting Yang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jian Lu
- School of Life Sciences, Peking University, Beijing 100871, China.
| | - Pei Du
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Qihui Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, Yunnan University, Kunming 650091, China; Faculty of Health Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
42
|
Zhang P, Luo S, Zou P, Deng Q, Wang C, Li J, Cai P, Zhang L, Li C, Li T. A novel simian adenovirus-vectored COVID-19 vaccine elicits effective mucosal and systemic immunity in mice by intranasal and intramuscular vaccination regimens. Microbiol Spectr 2023; 11:e0179423. [PMID: 37877750 PMCID: PMC10715068 DOI: 10.1128/spectrum.01794-23] [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: 04/28/2023] [Accepted: 09/19/2023] [Indexed: 10/26/2023] Open
Abstract
IMPORTANCE The essential goal of vaccination is to generate potent and long-term protection against diseases. Several factors including vaccine vector, delivery route, and boosting regimen influence the outcome of prime-boost immunization approaches. The immunization regimens by constructing a novel simian adenovirus-vectored COVID-19 vaccine and employing combination of intranasal and intramuscular inoculations could elicit mucosal neutralizing antibodies against five mutant strains in the respiratory tract and strong systemic immunity. Immune protection could last for more than 32 weeks. Vectored vaccine construction and immunization regimens have positively impacted respiratory disease prevention.
Collapse
Affiliation(s)
- Panli Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou , China
| | - Shengxue Luo
- Department of Pediatrics, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Peng Zou
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou , China
| | - Qitao Deng
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou , China
| | - Cong Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bai Rui Kang (BRK) Biological Science and Technology Limited Company, Guangzhou , China
| | - Jinfeng Li
- Shenzhen Bao'an District Central Blood Station, Shenzhen, China
| | - Peiqiao Cai
- Department of Bioengineering, School of Medicine and College of Engineering, University of Washington, Seattle, Washington, USA
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Tingting Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
Honda-Okubo Y, Bowen R, Barker M, Bielefeldt-Ohmann H, Petrovsky N. Advax-CpG55.2-adjuvanted monovalent or trivalent SARS-CoV-2 recombinant spike protein vaccine protects hamsters against heterologous infection with Beta or Delta variants. Vaccine 2023; 41:7116-7128. [PMID: 37863669 PMCID: PMC10873063 DOI: 10.1016/j.vaccine.2023.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023]
Abstract
The ongoing evolution of SARS-CoV-2 variants emphasizes the need for vaccines providing broad cross-protective immunity. This study was undertaken to assess the ability of Advax-CpG55.2 adjuvanted monovalent recombinant spike protein (Wuhan, Beta, Gamma) vaccines or a trivalent formulation to protect hamsters againstBeta or Delta virus infection. The ability of vaccines to block virus transmission to naïve co-housed animals was also assessed. In naïve hosts, the Beta variant induced higher virus loads than the Delta variant, and conversely the Delta variant caused more severe disease and was more likely to be associated with virus transmission. The trivalent vaccine formulation provided the best protection against both Beta and Delta infection and also completely prevented virus transmission. The next best performing vaccine was the original monovalent Wuhan-based vaccine. Notably, hamsters that received the monovalent Gamma spike vaccine had the highest viral loads and clinical disease of all the vaccine groups, a potential signal of antibody dependent-enhancement (ADE). These hamsters were also the most likely to transmit Delta virus to naïve recipients. In murine studies, the Gamma spike vaccine induced the highest total spike protein to RBD IgG ratio and the lowest levels of neutralizing antibody, a context that could predispose to ADE. Overall, the study results confirmed that the current SpikoGen® vaccine based on Wuhan spike protein was still able to protect against clinical disease caused by either the Beta or Delta virus variants but suggested additional protection may be obtained by combining it with extra variant spike proteins to make a multivalent formulation. This study highlights the complexity of optimizing vaccine protection against multiple SARS-CoV-2 variants and stresses the need to continue to pursue new and improved COVID-19 vaccines able to provide robust, long-lasting, and broadly cross-protective immunity against constantly evolving SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Yoshikazu Honda-Okubo
- Vaxine Pty Ltd., Bedford Park, Adelaide, SA 5042, Australia; College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Richard Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Mckinzee Barker
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Helle Bielefeldt-Ohmann
- School of Chemistry & Molecular Biosciences, The University of Queensland, St. Lucia, Qld 4072, Australia
| | | |
Collapse
|
44
|
Weklak D, Tisborn J, Mangold MH, Scheu R, Wodrich H, Hagedorn C, Jönsson F, Kreppel F. Insights from the Construction of Adenovirus-Based Vaccine Candidates against SARS-CoV-2: Expecting the Unexpected. Viruses 2023; 15:2155. [PMID: 38005833 PMCID: PMC10675337 DOI: 10.3390/v15112155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
To contain the spread of the SARS-CoV-2 pandemic, rapid development of vaccines was required in 2020. Rational design, international efforts, and a lot of hard work yielded the market approval of novel SARS-CoV-2 vaccines based on diverse platforms such as mRNA or adenovirus vectors. The great success of these technologies, in fact, contributed significantly to control the pandemic. Consequently, most scientific literature available in the public domain discloses the results of clinical trials and reveals data of efficaciousness. However, a description of processes and rationales that led to specific vaccine design is only partially available, in particular for adenovirus vectors, even though it could prove helpful for future developments. Here, we disclose our insights from the endeavors to design compatible functional adenoviral vector platform expression cassettes for the SARS-CoV-2 spike protein. We observed that contextualizing genes from an ssRNA virus into a DNA virus provides significant challenges. Besides affecting physical titers, expression cassette design of adenoviral vaccine candidates can affect viral propagation and spike protein expression. Splicing of mRNAs was affected, and fusogenicity of the spike protein in ACE2-overexpressing cells was enhanced when the ER retention signal was deleted.
Collapse
Affiliation(s)
- Denice Weklak
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany; (D.W.); (J.T.); (M.H.M.); (R.S.); (C.H.)
| | - Julian Tisborn
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany; (D.W.); (J.T.); (M.H.M.); (R.S.); (C.H.)
| | - Maurin Helen Mangold
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany; (D.W.); (J.T.); (M.H.M.); (R.S.); (C.H.)
| | - Raphael Scheu
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany; (D.W.); (J.T.); (M.H.M.); (R.S.); (C.H.)
| | - Harald Wodrich
- Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR 5234, Université de Bordeaux, 33076 Bordeaux, France;
| | - Claudia Hagedorn
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany; (D.W.); (J.T.); (M.H.M.); (R.S.); (C.H.)
| | - Franziska Jönsson
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany; (D.W.); (J.T.); (M.H.M.); (R.S.); (C.H.)
| | - Florian Kreppel
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58453 Witten, Germany; (D.W.); (J.T.); (M.H.M.); (R.S.); (C.H.)
| |
Collapse
|
45
|
Wang W, Wang H, Yao T, Li Y, Yi L, Gao Y, Lian J, Feng Y, Wang S. The top 100 most cited articles on COVID-19 vaccine: a bibliometric analysis. Clin Exp Med 2023; 23:2287-2299. [PMID: 36939968 PMCID: PMC10026222 DOI: 10.1007/s10238-023-01046-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/07/2023] [Indexed: 03/21/2023]
Abstract
This study aimed to uncover the current major topics regarding COVID-19 vaccine, and systematically evaluate the development trends for future research. The top 100 most cited original articles on COVID-19 vaccine from January 2020 to October 2022 were identified from Web of Science Core Collection database. CiteSpace (v6.1.R3) was adopted for bibliometric analysis with statistical and visual analysis. The number of citations ranged from 206 to 5881, with a median of 349.5. The USA (n = 56), England (n = 33), and China (n = 16) ranked the top three countries/regions in terms of the number of publications. Harvard Medical School (centrality = 0.71), Boston Children's Hospital (centrality = 0.67), and Public Health England (centrality = 0.57) were the top three institutions leading the way on COVID-19 vaccine research. The New England of medicine journal dominated with 22 articles in the 32 high-quality journals. The three most frequent keywords were immunization (centrality = 0.25), influenza vaccination (centrality = 0.21), and coronavirus (centrality = 0.18). Cluster analysis of keywords showed that the top four categories were protection efficacy, vaccine hesitancy, spike protein, and second vaccine dose (Q value = 0.535, S value = 0.879). Cluster analysis of cited references showed that top eight largest categories were Cov-2 variant, clinical trial, large integrated health system, COV-2 rhesus macaque, mRNA vaccine, vaccination intent, phase II study, and Cov-2 omicron variant (Q value = 0.672, S value = 0.794). The research on COVID-19 vaccine is currently the hottest topic in academic community. At present, COVID-19 vaccines researches have focused on vaccine efficacy, vaccine hesitancy, and the efficacy of current vaccines on omicron variants. However, how to increase vaccine uptake, focus on mutations in the spike protein, evaluate of the efficacy of booster vaccine, and how effective new vaccines under pre- and clinical development against omicron will be spotlight in the future.
Collapse
Affiliation(s)
- Weigang Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001 Shanxi China
- Shanxi Province Cancer Hospital, Taiyuan, China
| | - Hu Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001 Shanxi China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Tian Yao
- First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Yandi Li
- Department of Epidemiology, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001 Shanxi China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Linzhu Yi
- Department of Epidemiology, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001 Shanxi China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Ying Gao
- Department of Epidemiology, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001 Shanxi China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Jia Lian
- Department of Epidemiology, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001 Shanxi China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Yongliang Feng
- Department of Epidemiology, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001 Shanxi China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| | - Suping Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001 Shanxi China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
46
|
Gould S, Wrzesinski C, Stebbings R, Segal L. Report from an ICT 2022 workshop on toxicology for Covid19 vaccines: Industry, regulatory and CRO perspectives. Vaccine 2023; 41:5919-5924. [PMID: 37633751 DOI: 10.1016/j.vaccine.2023.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/28/2023]
Abstract
The Covid pandemic took the world by surprise in late 2019 and the need for rapid development of vaccines became paramount. The challenge was how to accelerate standard vaccine development times as much as possible. With knowledge of the genetic code of SARsCOV2, vaccine manufacturers throughout the world have risen to the challenge and several new vaccines were rapidly developed for emergency use. In March 2020, global Regulatory Authorities met to consider how to start early clinical trials and accept rolling submissions. Before use in clinical trials or any mass vaccination campaigns, the safety of the candidate vaccine needs to be evaluated. Non-clinical toxicology studies are required as an important part of vaccine safety evaluation. The extent of the toxicology evaluation prior to the start of clinical trials depended on several factors, including: the type of the candidate vaccine as well as already available supportive information with the candidate vaccine or similar vaccine types. For vaccine candidates with pre-existing data, this would save valuable time whilst a full toxicology evaluation was completed in parallel. For vaccines with more limited data, toxicology data was required before clinical development could start. This workshop examined the nonclinical toxicology studies for new Covid vaccines from the perspectives of: Vaccine manufacturers with different vaccine technologies, managing global regulatory submissions/responses; CROs, managing the urgency of conducting and reporting studies and supporting new players in the vaccine world; and Regulatory Authorities, in supporting the review process, juggling the need for safety and quality with mounting pressure to approve vaccines.
Collapse
|
47
|
Gao A, Chen Y, Liang H, Cui X, Zhang A, Cui D. Developing an efficient MGCR microneedle nanovaccine patch for eliciting Th 1 cellular response against the SARS-CoV-2 infection. Theranostics 2023; 13:4821-4835. [PMID: 37771766 PMCID: PMC10526668 DOI: 10.7150/thno.83390] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/18/2023] [Indexed: 09/30/2023] Open
Abstract
Rationale: Novel vaccine R&D is essential to interrupt the COVID-19 pandemic and other epidemics in the future. Subunit vaccines have received tremendous attention for their low cost and safety. To improve the immunogenicity of subunit vaccines, we developed a novel vaccine adjuvant system. Methods: Here we rationally designed a CpG 1018 and graphene oxide-based bi-adjuvant system to deliver the Receptor-Binding Domain (RBD) of the SARS-CoV-2 spike protein and obtained the graphene oxide-based complex adjuvant nanovaccine (GCR). Furthermore, we developed a microneedle patch vaccine (MGCR) based on the GCR vaccine. Results: GCR nanovaccine displayed superb antigen loading and encapsulation efficiency. Two dosages of vaccination of GCR nanovaccine could elicit adequate RBD-specific binding antibody response with 2.14-fold higher IgG titer than Alum adjuvant vaccine. The peptide pools assay demonstrated the robust RBD-specific Type 1 Cellular response induced by the GCR nanovaccine in CD8+ T cells. Furthermore, we prepared an MGCR microneedle patch, which generated a similar RBD-specific binding antibody response to the GCR vaccine, sustained a high antibody level above 16 weeks, and significantly elevated the Tcm proportion in mouse spleen. The MGCR microneedle patch vaccine also could be stably stored at room temperature for several months and administrated without medical staff, which maximizes the vaccine distribution efficiency. Conclusion: The vaccine system could significantly improve the vaccine distribution rate in low-income areas and offer a potential vaccination approach to fight against the SARS-Cov-2 infection and other pandemics occurred in the future.
Collapse
Affiliation(s)
- Ang Gao
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Instrument for Diagnosis and Therapy, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- National Engineering Research Center for Nanotechnology, 28 East Jiangchuan Road, Shanghai 200241, China
| | - Yunsheng Chen
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Instrument for Diagnosis and Therapy, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Radiology Department of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai 200025, China
| | - Hui Liang
- National Engineering Research Center for Nanotechnology, 28 East Jiangchuan Road, Shanghai 200241, China
| | - Xinyuan Cui
- Radiology Department of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai 200025, China
| | - Amin Zhang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Instrument for Diagnosis and Therapy, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- National Engineering Research Center for Nanotechnology, 28 East Jiangchuan Road, Shanghai 200241, China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Instrument for Diagnosis and Therapy, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- National Engineering Research Center for Nanotechnology, 28 East Jiangchuan Road, Shanghai 200241, China
| |
Collapse
|
48
|
Scutari R, Renica S, Cento V, Nava A, Sammartino JC, Ferrari A, Pani A, Merli M, Fanti D, Vismara C, Scaglione F, Puoti M, Bandera A, Gori A, Piralla A, Baldanti F, Perno CF, Alteri C. Quantitative SARS-CoV-2 subgenomic RNA as a surrogate marker for viral infectivity: Comparison between culture isolation and direct sgRNA quantification. PLoS One 2023; 18:e0291120. [PMID: 37656746 PMCID: PMC10473502 DOI: 10.1371/journal.pone.0291120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 08/22/2023] [Indexed: 09/03/2023] Open
Abstract
Detection of subgenomic (sg) SARS-CoV-2 RNAs are frequently used as a correlate of viral infectiousness, but few data about correlation between sg load and viable virus are available. Here, we defined concordance between culture isolation and E and N sgRNA quantification by ddPCR assays in 51 nasopharyngeal swabs collected from SARS-CoV-2 positive hospitalized patients. Among the 51 samples, 14 were SARS-CoV-2 culture-positive and 37 were negative. According to culture results, the sensitivity and specificity of E and N sgRNA assays were 100% and 100%, and 84% and 86%, respectively. ROC analysis showed that the best E and N cut-offs to predict positive culture isolation were 32 and 161 copies/mL respectively, with an AUC (95% CI) of 0.96 (0.91-1.00) and 0.96 (0.92-1.00), and a diagnostic accuracy of 88% and 92%, respectively. Even if no significant correlations were observed between sgRNA amount and clinical presentation, a higher number of moderate/severe cases and lower number of days from symptoms onset characterized patients with sgRNA equal to or higher than sgRNA cut-offs. Overall, this study suggests that SARS-CoV-2 sgRNA quantification could be helpful to estimate the replicative activity of SARS-CoV-2 and can represent a valid surrogate marker to efficiently recognize patients with active infection. The inclusion of this assay in available SARS-CoV-2 diagnostics procedure might help in optimizing fragile patients monitoring and management.
Collapse
Affiliation(s)
- Rossana Scutari
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
- Multimodal Research Area, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Silvia Renica
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Valeria Cento
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCSS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alice Nava
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Alessandro Ferrari
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Arianna Pani
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Marco Merli
- Infectious Diseases Unit, Azienda Socio-Sanitaria Territoriale (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Diana Fanti
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Chiara Vismara
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Francesco Scaglione
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
- Chemical-Clinical and Microbiological Analysis, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Massimo Puoti
- Infectious Diseases Unit, Azienda Socio-Sanitaria Territoriale (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Antonio Piralla
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fausto Baldanti
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | | | - Claudia Alteri
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
- Multimodal Research Area, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| |
Collapse
|
49
|
Tokunoh N, Tamiya S, Watanabe M, Okamoto T, Anindita J, Tanaka H, Ono C, Hirai T, Akita H, Matsuura Y, Yoshioka Y. A nasal vaccine with inactivated whole-virion elicits protective mucosal immunity against SARS-CoV-2 in mice. Front Immunol 2023; 14:1224634. [PMID: 37720231 PMCID: PMC10500122 DOI: 10.3389/fimmu.2023.1224634] [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: 05/18/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Introduction Vaccinations are ideal for reducing the severity of clinical manifestations and secondary complications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); however, SARS-CoV-2 continues to cause morbidity and mortality worldwide. In contrast to parenteral vaccines such as messenger RNA vaccines, nasal vaccines are expected to be more effective in preventing viral infections in the upper respiratory tract, the primary locus for viral infection and transmission. In this study, we examined the prospects of an inactivated whole-virion (WV) vaccine administered intranasally against SARS-CoV-2. Methods Mice were immunized subcutaneously (subcutaneous vaccine) or intranasally (nasal vaccine) with the inactivated WV of SARS-CoV-2 as the antigen. Results The spike protein (S)-specific IgA level was found to be higher upon nasal vaccination than after subcutaneous vaccination. The level of S-specific IgG in the serum was also increased by the nasal vaccine, although it was lower than that induced by the subcutaneous vaccine. The nasal vaccine exhibited a stronger defense against viral invasion in the upper respiratory tract than the subcutaneous vaccine and unimmunized control; however, both subcutaneous and nasal vaccines provided protection in the lower respiratory tract. Furthermore, we found that intranasally administered inactivated WV elicited robust production of S-specific IgA in the nasal mucosa and IgG in the blood of mice previously vaccinated with messenger RNA encoding the S protein. Discussion Overall, these results suggest that a nasal vaccine containing inactivated WV can be a highly effective means of protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nagisa Tokunoh
- Innovative Vaccine Research and Development Center, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shigeyuki Tamiya
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Department of Microbiology and Immunology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Masato Watanabe
- Innovative Vaccine Research and Development Center, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Toru Okamoto
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Jessica Anindita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Science, Chiba University, Chiba-shi, Chiba, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Chikako Ono
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Toshiro Hirai
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Yoshiharu Matsuura
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
| | - Yasuo Yoshioka
- Innovative Vaccine Research and Development Center, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
50
|
Wang S, Qin M, Xu L, Mu T, Zhao P, Sun B, Wu Y, Song L, Wu H, Wang W, Liu X, Li Y, Yang F, Xu K, He Z, Klein M, Wu K. Aerosol Inhalation of Chimpanzee Adenovirus Vectors (ChAd68) Expressing Ancestral or Omicron BA.1 Stabilized Pre-Fusion Spike Glycoproteins Protects Non-Human Primates against SARS-CoV-2 Infection. Vaccines (Basel) 2023; 11:1427. [PMID: 37766104 PMCID: PMC10535855 DOI: 10.3390/vaccines11091427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Current COVID-19 vaccines are effective countermeasures to control the SARS-CoV-2 virus pandemic by inducing systemic immune responses through intramuscular injection. However, respiratory mucosal immunization will be needed to elicit local sterilizing immunity to prevent virus replication in the nasopharynx, shedding, and transmission. In this study, we first compared the immunoprotective ability of a chimpanzee replication-deficient adenovirus-vectored COVID-19 vaccine expressing a stabilized pre-fusion spike glycoprotein from the ancestral SARS-CoV-2 strain Wuhan-Hu-1 (BV-AdCoV-1) administered through either aerosol inhalation, intranasal spray, or intramuscular injection in cynomolgus monkeys and rhesus macaques. Compared with intranasal administration, aerosol inhalation of BV-AdCoV-1 elicited stronger humoral and mucosal immunity that conferred excellent protection against SARS-CoV-2 infection in rhesus macaques. Importantly, aerosol inhalation induced immunity comparable to that obtained by intramuscular injection, although at a significantly lower dose. Furthermore, to address the problem of immune escape variants, we evaluated the merits of heterologous boosting with an adenovirus-based Omicron BA.1 vaccine (C68-COA04). Boosting rhesus macaques vaccinated with two doses of BV-AdCoV-1 with either the homologous or the heterologous C68-COA04 vector resulted in cross-neutralizing immunity against WT, Delta, and Omicron subvariants, including BA.4/5 stronger than that obtained by administering a bivalent BV-AdCoV-1/C68-COA04 vaccine. These results demonstrate that the administration of BV-AdCoV-1 or C68-COA04 via aerosol inhalation is a promising approach to prevent SARS-CoV-2 infection and transmission and curtail the pandemic spread.
Collapse
Affiliation(s)
- Shen Wang
- Regulatory and Medical Affairs Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (S.W.); (L.S.)
| | - Mian Qin
- Project Management Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (M.Q.); (L.X.)
| | - Long Xu
- Project Management Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (M.Q.); (L.X.)
| | - Ting Mu
- Innovative Discovery Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (T.M.); (B.S.)
| | - Ping Zhao
- Test Development Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (P.Z.); (Y.W.)
| | - Bing Sun
- Innovative Discovery Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (T.M.); (B.S.)
| | - Yue Wu
- Test Development Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (P.Z.); (Y.W.)
| | - Lingli Song
- Regulatory and Medical Affairs Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China; (S.W.); (L.S.)
| | - Han Wu
- Quality Control Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
| | - Weicheng Wang
- Pilot Production Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
| | - Xingwen Liu
- Quality Assurance Department, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
| | - Yanyan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650000, China; (Y.L.); (Z.H.)
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650000, China; (Y.L.); (Z.H.)
| | - Ke Xu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China;
- Executive Office, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650000, China; (Y.L.); (Z.H.)
| | - Michel Klein
- Executive Office, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
- Executive Office, Shanghai BravoBio Co., Ltd., Shanghai 200000, China
| | - Ke Wu
- Executive Office, Wuhan BravoVax Co., Ltd., Wuhan 430070, China;
- Executive Office, Shanghai BravoBio Co., Ltd., Shanghai 200000, China
| |
Collapse
|