1
|
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
|
2
|
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
|
3
|
Salamony A, Shamikh Y, Amer K, Elnagdy T, Elnakib M, Yehia AA, Hassan W, Abdelsalam M. Are Measles-Mumps-Rubella (MMR) Antibodies Friends or Foes for Covid-19 Disease? Arch Immunol Ther Exp (Warsz) 2023; 71:15. [PMID: 37341786 DOI: 10.1007/s00005-023-00680-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/18/2022] [Indexed: 06/22/2023]
Abstract
Many factors have been implicated in the pathogenesis and severity of COVID-19 pandemic. A wide variation in the susceptibility for SARS-CoV-2 infection among different population, gender and age has been observed. Multiple studies investigated the relationship between the antibody's titre of previously vaccinated individuals and the susceptibility of coronavirus infection, to find a rapid effective therapy for this pandemic. This study focused on the association between measles-mumps-rubella (MMR) antibodies titre and the severity of COVID-19 infection. We aimed to investigate the correlation between the antibody's titre of MMR and the SARS-CoV-2 infection susceptibility and disease severity, in a cohort of COVID-19 Egyptian patients, compared to a control group. MMR antibody titre was measured using enzyme Linked Immune Sorbent Assay; (ELISA) for 136 COVID-19 patients and 44 healthy individuals, as control group. There were high levels of measles and mumps antibodies titer in the deteriorating cases, which could not protect from SARS-CoV-2 infection. However, the rubella antibodies might protect from SARS-CoV-2 infection, but once the infection occurs, it may aggravate the risk of case deterioration. MMR antibodies could be used as a guideline for COVID-19 symptom-severity and, in turn, may be considered as an economic prognostic marker used for early protection from multiple autoimmune organ failure.
Collapse
Affiliation(s)
- Azza Salamony
- Egypt Centre for Research and Regenerative Medicine, ECRRM, Cairo, 11517, Egypt
- Microbiology and Immunology, Central Public Health Laboratories, CPHL, Ministry of Health, Cairo, 11613, Egypt
| | - Yara Shamikh
- Egypt Centre for Research and Regenerative Medicine, ECRRM, Cairo, 11517, Egypt
- Microbiology and Immunology, Central Public Health Laboratories, CPHL, Ministry of Health, Cairo, 11613, Egypt
| | - Khaled Amer
- Egypt Centre for Research and Regenerative Medicine, ECRRM, Cairo, 11517, Egypt
| | - Tarek Elnagdy
- Egypt Centre for Research and Regenerative Medicine, ECRRM, Cairo, 11517, Egypt
| | - Mostafa Elnakib
- Egypt Centre for Research and Regenerative Medicine, ECRRM, Cairo, 11517, Egypt
| | - Abd Allah Yehia
- Egypt Centre for Research and Regenerative Medicine, ECRRM, Cairo, 11517, Egypt
| | - Wael Hassan
- Egypt Centre for Research and Regenerative Medicine, ECRRM, Cairo, 11517, Egypt
| | - Maha Abdelsalam
- Egypt Centre for Research and Regenerative Medicine, ECRRM, Cairo, 11517, Egypt.
- Clinical Pathology Department, Faculty of Medicine, Immunology Unit, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
4
|
Babaeimarzangou SS, Zaker H, Soleimannezhadbari E, Gamchi NS, Kazeminia M, Tarighi S, Seyedian H, Tsatsakis A, Spandidos DA, Margina D. Vaccine development for zoonotic viral diseases caused by positive‑sense single‑stranded RNA viruses belonging to the Coronaviridae and Togaviridae families (Review). Exp Ther Med 2022; 25:42. [PMID: 36569444 PMCID: PMC9768462 DOI: 10.3892/etm.2022.11741] [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: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Outbreaks of zoonotic viral diseases pose a severe threat to public health and economies worldwide, with this currently being more prominent than it previously was human history. These emergency zoonotic diseases that originated and transmitted from vertebrates to humans have been estimated to account for approximately one billion cases of illness and have caused millions of deaths worldwide annually. The recent emergence of severe acute respiratory syndrome coronavirus-2 (coronavirus disease 2019) is an excellent example of the unpredictable public health threat causing a pandemic. The present review summarizes the literature data regarding the main vaccine developments in human clinical phase I, II and III trials against the zoonotic positive-sense single-stranded RNA viruses belonging to the Coronavirus and Alphavirus genera, including severe acute respiratory syndrome, Middle east respiratory syndrome, Venezuelan equine encephalitis virus, Semliki Forest virus, Ross River virus, Chikungunya virus and O'nyong-nyong virus. That there are neither vaccines nor effective antiviral drugs available against most of these viruses is undeniable. Therefore, new explosive outbreaks of these zoonotic viruses may surely be expected. The present comprehensive review provides an update on the status of vaccine development in different clinical trials against these viruses, as well as an overview of the present results of these trials.
Collapse
Affiliation(s)
- Seyed Sajjad Babaeimarzangou
- Division of Poultry Health and Diseases, Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Himasadat Zaker
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | | | - Naeimeh Shamsi Gamchi
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | - Masoud Kazeminia
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417935840, Iran
| | - Shima Tarighi
- Veterinary Office of West Azerbaijan Province, Urmia 5717617695, Iran
| | - Homayon Seyedian
- Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Department of Medicine, University of Crete, 71307 Heraklion, Greece,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 020956 Bucharest, Romania,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| |
Collapse
|
5
|
Ebenig A, Lange MV, Mühlebach MD. Versatility of live-attenuated measles viruses as platform technology for recombinant vaccines. NPJ Vaccines 2022; 7:119. [PMID: 36243743 PMCID: PMC9568972 DOI: 10.1038/s41541-022-00543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
Live-attenuated measles virus (MeV) has been extraordinarily effective in preventing measles infections and their often deadly sequelae, accompanied by remarkable safety and stability since their first licensing in 1963. The advent of recombinant DNA technologies, combined with systems to generate infectious negative-strand RNA viruses on the basis of viral genomes encoded on plasmid DNA in the 1990s, paved the way to generate recombinant, vaccine strain-derived MeVs. These live-attenuated vaccine constructs can encode and express additional foreign antigens during transient virus replication following immunization. Effective humoral and cellular immune responses are induced not only against the MeV vector, but also against the foreign antigen cargo in immunized individuals, which can protect against the associated pathogen. This review aims to present an overview of the versatility of this vaccine vector as platform technology to target various diseases, as well as current research and developmental stages, with one vaccine candidate ready to enter phase III clinical trials to gain marketing authorization, MV-CHIK.
Collapse
Affiliation(s)
- Aileen Ebenig
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Mona V Lange
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Michael D Mühlebach
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany.
| |
Collapse
|
6
|
Sepand MR, Bigdelou B, Ho JQ, Sharaf M, Lannigan AJ, Sullivan IM, da Silva AP, Barrett LO, McGoldrick S, Lnu Y, Lynch SE, Boisclair JM, Barnard-Pratt DD, Zanganeh S. Long-Term Immunity and Antibody Response: Challenges for Developing Efficient COVID-19 Vaccines. Antibodies (Basel) 2022; 11:35. [PMID: 35645208 PMCID: PMC9149948 DOI: 10.3390/antib11020035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/01/2022] [Accepted: 05/06/2022] [Indexed: 12/04/2022] Open
Abstract
Questions and concerns regarding the efficacy and immunogenicity of coronavirus disease 2019 (COVID-19) vaccines have plagued scientists since the BNT162b2 mRNA vaccine was introduced in late 2020. As a result, decisions about vaccine boosters based on breakthrough infection rates and the decline of antibody titers have commanded worldwide attention and research. COVID-19 patients have displayed continued severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-spike-protein-specific antibodies and neutralizing antibodies in longitudinal studies; in addition, cytokine activation has been detected at early steps following SARS-CoV-2 infection. Epitopes that are highly reactive and can mediate long-term antibody responses have been identified at the spike and ORF1ab proteins. The N-terminal domain of the S1 and S2 subunits is the location of important SARS-CoV-2 spike protein epitopes. High sequence identity between earlier and newer variants of SARS-CoV-2 and different degrees of sequence homology among endemic human coronaviruses have been observed. Understanding the extent and duration of protective immunity is consequential for determining the course of the COVID-19 pandemic. Further knowledge of memory responses to different variants of SARS-CoV-2 is needed to improve the design of the vaccine.
Collapse
Affiliation(s)
- Mohammad Reza Sepand
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Banafsheh Bigdelou
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Jim Q. Ho
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Mohammad Sharaf
- Department of Chemical and Biomolecular Engineering, New York University, New York, NY 10012, USA;
| | - Alexis J. Lannigan
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Ian M. Sullivan
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Alecsander P. da Silva
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Leland O. Barrett
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Scott McGoldrick
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Yuvraj Lnu
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Shannon E. Lynch
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Jared M. Boisclair
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Dakarai D. Barnard-Pratt
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| | - Steven Zanganeh
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, North Dartmouth, MA 02747, USA; (M.R.S.); (B.B.); (A.J.L.); (I.M.S.); (A.P.d.S.); (L.O.B.); (S.M.); (Y.L.); (S.E.L.); (J.M.B.); (D.D.B.-P.)
| |
Collapse
|
7
|
Lundstrom K. Self-replicating vehicles based on negative strand RNA viruses. Cancer Gene Ther 2022:10.1038/s41417-022-00436-7. [PMID: 35169298 PMCID: PMC8853047 DOI: 10.1038/s41417-022-00436-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/14/2022] [Accepted: 01/31/2022] [Indexed: 11/10/2022]
Abstract
Self-replicating RNA viruses have been engineered as efficient expression vectors for vaccine development for infectious diseases and cancers. Moreover, self-replicating RNA viral vectors, particularly oncolytic viruses, have been applied for cancer therapy and immunotherapy. Among negative strand RNA viruses, measles viruses and rhabdoviruses have been frequently applied for vaccine development against viruses such as Chikungunya virus, Lassa virus, Ebola virus, influenza virus, HIV, Zika virus, and coronaviruses. Immunization of rodents and primates has elicited strong neutralizing antibody responses and provided protection against lethal challenges with pathogenic viruses. Several clinical trials have been conducted. Ervebo, a vaccine based on a vesicular stomatitis virus (VSV) vector has been approved for immunization of humans against Ebola virus. Different types of cancers such as brain, breast, cervical, lung, leukemia/lymphoma, ovarian, prostate, pancreatic, and melanoma, have been the targets for cancer vaccine development, cancer gene therapy, and cancer immunotherapy. Administration of measles virus and VSV vectors have demonstrated immune responses, tumor regression, and tumor eradication in various animal models. A limited number of clinical trials have shown well-tolerated treatment, good safety profiles, and dose-dependent activity in cancer patients.
Collapse
|
8
|
Launay O, Artaud C, Lachâtre M, Ait-Ahmed M, Klein J, Luong Nguyen LB, Durier C, Jansen B, Tomberger Y, Jolly N, Grossmann A, Tabbal H, Brunet J, Gransagne M, Choucha Z, Batalie D, Delgado A, Müllner M, Tschismarov R, Berghmans PJ, Martin A, Ramsauer K, Escriou N, Gerke C. Safety and immunogenicity of a measles-vectored SARS-CoV-2 vaccine candidate, V591 / TMV-083, in healthy adults: results of a randomized, placebo-controlled Phase I study. EBioMedicine 2022; 75:103810. [PMID: 35045362 PMCID: PMC8761070 DOI: 10.1016/j.ebiom.2021.103810] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND V591 (TMV-083) is a live recombinant measles vector-based vaccine candidate expressing a pre-fusion stabilized SARS-CoV-2 spike protein. METHODS We performed a randomized, placebo-controlled Phase I trial with an unblinded dose escalation and a double-blind treatment phase at 2 sites in France and Belgium to evaluate the safety and immunogenicity of V591. Ninety healthy SARS-CoV-2 sero-negative adults (18-55 years of age) were randomized into 3 cohorts, each comprising 24 vaccinees and 6 placebo recipients. Participants received two intramuscular injections of a low dose vaccine (1 × 105 median Tissue Culture Infectious Dose [TCID50]), one or two injections of a high dose vaccine (1 × 106 TCID50), or placebo with a 28 day interval. Safety was assessed by solicited and unsolicited adverse events. Immunogenicity was measured by SARS-CoV-2 spike protein-binding antibodies, neutralizing antibodies, spike-specific T cell responses, and anti-measles antibodies. ClinicalTrials.gov, NCT04497298. FINDINGS Between Aug 10 and Oct 13, 2020, 148 volunteers were screened of whom 90 were randomized. V591 showed a good safety profile at both dose levels. No serious adverse events were reported. At least one treatment-related adverse event was reported by 15 (20.8%) participants receiving V591 vs. 6 (33.3%) of participants receiving placebo. Eighty-one percent of participants receiving two injections of V591 developed spike-binding antibodies after the second injection. However, neutralizing antibodies were detectable on day 56 only in 17% of participants receiving the low dose and 61% receiving the high dose (2 injections). Spike-specific T cell responses were not detected. Pre-existing anti-measles immunity had a statistically significant impact on the immune response to V591, which was in contrast to previous results with the measles vector-based chikungunya vaccine. INTERPRETATION While V591 was generally well tolerated, the immunogenicity was not sufficient to support further development. FUNDING Themis Bioscience GmbH, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, USA; Coalition for Epidemic Preparedness Innovations (CEPI).
Collapse
Affiliation(s)
- Odile Launay
- Université de Paris, CIC Cochin-Pasteur; APHP, Hôpital Cochin; INSERM CIC1417, Paris, France
| | - Cécile Artaud
- Institut Pasteur, Université de Paris, Centre de Recherche Translationnelle, Paris, France
| | - Marie Lachâtre
- Université de Paris, CIC Cochin-Pasteur; APHP, Hôpital Cochin; INSERM CIC1417, Paris, France
| | - Mohand Ait-Ahmed
- Institut Pasteur, Université de Paris, Centre de Recherche Translationnelle, Paris, France
| | - Jelle Klein
- SGS, Clinical Pharmacology Unit, Antwerpen, Belgium
| | - Liem Binh Luong Nguyen
- Université de Paris, CIC Cochin-Pasteur; APHP, Hôpital Cochin; INSERM CIC1417, Paris, France
| | | | | | - Yvonne Tomberger
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | - Nathalie Jolly
- Institut Pasteur, Université de Paris, Centre de Recherche Translationnelle, Paris, France
| | - Anna Grossmann
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | - Houda Tabbal
- Institut Pasteur, Université de Paris, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Jérémy Brunet
- Institut Pasteur, Université de Paris, Département de Santé Globale, Paris, France
| | - Marion Gransagne
- Institut Pasteur, Université de Paris, Département de Santé Globale, Paris, France
| | - Zaineb Choucha
- Institut Pasteur, Université de Paris, Département de Santé Globale, Paris, France
| | - Damien Batalie
- Institut Pasteur, Université de Paris, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | | | - Matthias Müllner
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | - Roland Tschismarov
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | | | - Annette Martin
- Institut Pasteur, Université de Paris, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Katrin Ramsauer
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | - Nicolas Escriou
- Institut Pasteur, Université de Paris, Département de Santé Globale, Paris, France.
| | - Christiane Gerke
- Institut Pasteur, Université de Paris, Innovation Office, Vaccine Programs, Paris, France.
| |
Collapse
|
9
|
Vanhoutte F, Liu W, Wiedmann RT, Haspeslagh L, Cao X, Boundy K, Aliprantis A, Davila M, Hartzel J, Li J, McGuire M, Ramsauer K, Tomberger Y, Tschismarov R, Brown DD, Xu W, Sachs JR, Russell K, Stoch SA, Lai E. Safety and immunogenicity of the measles vector-based SARS-CoV-2 vaccine candidate, V591, in adults: results from a phase 1/2 randomised, double-blind, placebo-controlled, dose-ranging trial. EBioMedicine 2022; 75:103811. [PMID: 35042081 PMCID: PMC8759950 DOI: 10.1016/j.ebiom.2021.103811] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND We report on the safety and immunogenicity of V591, a measles vector-based SARS-CoV-2 vaccine candidate. METHODS In this multicentre, randomised, placebo-controlled, double-blind, phase 1/2 trial, healthy adults with no history of COVID-19 disease were assigned to intramuscular injection of V591 or placebo (4:1 ratio). In part 1, younger adults (18-55 years) received V591 median tissue culture infectious dose (TCID50)-levels of 1×105 or 1×106 or placebo, 56 days apart. In part 2, younger and older (>55 years) adults received a single dose of one of four (104/105/106/107) or one of two (105/106) V591 TCID50 levels, respectively, or placebo. PRIMARY OUTCOME safety/tolerability. Secondary outcome: humoral immunogenicity. ClinicalTrials.gov: NCT04498247. FINDINGS From August-December 2020, 444 participants were screened and 263 randomised (210 V591; 53 placebo); 262 received at least one and 10 received two doses of V591 or placebo. Adverse events were experienced by 140/209 (67.0%) V591 dose-group participants and 37/53 (69.8%) placebo-group participants following injection 1; most frequent were fatigue (57 [27.3%] vs 20 [37.7%]), headache (57 [27.3%] vs 19 [35.8%]), myalgia (35 [16.7%] vs 10 [18.9%]), and injection-site pain (35 [16.7%] vs 4 [7.5%]). No deaths nor vaccine-related serious adverse events occurred. At Day 29, no anti-SARS-CoV-2 spike serum neutralising antibody and IgG-responses were identified in placebo or the three lower V591 dose-groups; responses were detected with V591 1×107 TCID50, although titres were lower than convalescent serum. INTERPRETATION V591 was generally well tolerated, but immunogenicity was insufficient to warrant continued development. FUNDING Merck Sharp & Dohme, Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA.
Collapse
Affiliation(s)
| | - Wen Liu
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - Xin Cao
- Merck & Co., Inc., Kenilworth, NJ, USA
| | - Keith Boundy
- Merck & Co., Inc., Kenilworth, NJ, USA; Currently at AlloVir, Inc., Cambridge, MA, USA
| | - Antonios Aliprantis
- Merck & Co., Inc., Kenilworth, NJ, USA; Currently at Flagship Pioneering, Boston, MA, USA
| | | | | | | | | | - Katrin Ramsauer
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Yvonne Tomberger
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Roland Tschismarov
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | | | | | | | - Eseng Lai
- Merck & Co., Inc., Kenilworth, NJ, USA.
| |
Collapse
|
10
|
A live measles-vectored COVID-19 vaccine induces strong immunity and protection from SARS-CoV-2 challenge in mice and hamsters. Nat Commun 2021; 12:6277. [PMID: 34725327 PMCID: PMC8560864 DOI: 10.1038/s41467-021-26506-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 10/05/2021] [Indexed: 11/08/2022] Open
Abstract
Several COVID-19 vaccines have now been deployed to tackle the SARS-CoV-2 pandemic, most of them based on messenger RNA or adenovirus vectors.The duration of protection afforded by these vaccines is unknown, as well as their capacity to protect from emerging new variants. To provide sufficient coverage for the world population, additional strategies need to be tested. The live pediatric measles vaccine (MV) is an attractive approach, given its extensive safety and efficacy history, along with its established large-scale manufacturing capacity. We develop an MV-based SARS-CoV-2 vaccine expressing the prefusion-stabilized, membrane-anchored full-length S antigen, which proves to be efficient at eliciting strong Th1-dominant T-cell responses and high neutralizing antibody titers. In both mouse and golden Syrian hamster models, these responses protect the animals from intranasal infectious challenge. Additionally, the elicited antibodies efficiently neutralize in vitro the three currently circulating variants of SARS-CoV-2.
Collapse
|
11
|
Sa-nguanmoo N, Namdee K, Khongkow M, Ruktanonchai U, Zhao Y, Liang XJ. Review: Development of SARS-CoV-2 immuno-enhanced COVID-19 vaccines with nano-platform. NANO RESEARCH 2021; 15:2196-2225. [PMID: 34659650 PMCID: PMC8501370 DOI: 10.1007/s12274-021-3832-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 05/04/2023]
Abstract
Vaccination is the most effective way to prevent coronavirus disease 2019 (COVID-19). Vaccine development approaches consist of viral vector vaccines, DNA vaccine, RNA vaccine, live attenuated virus, and recombinant proteins, which elicit a specific immune response. The use of nanoparticles displaying antigen is one of the alternative approaches to conventional vaccines. This is due to the fact that nano-based vaccines are stable, able to target, form images, and offer an opportunity to enhance the immune responses. The diameters of ultrafine nanoparticles are in the range of 1-100 nm. The application of nanotechnology on vaccine design provides precise fabrication of nanomaterials with desirable properties and ability to eliminate undesirable features. To be successful, nanomaterials must be uptaken into the cell, especially into the target and able to modulate cellular functions at the subcellular levels. The advantages of nano-based vaccines are the ability to protect a cargo such as RNA, DNA, protein, or synthesis substance and have enhanced stability in a broad range of pH, ambient temperatures, and humidity for long-term storage. Moreover, nano-based vaccines can be engineered to overcome biological barriers such as nonspecific distribution in order to elicit functions in antigen presenting cells. In this review, we will summarize on the developing COVID-19 vaccine strategies and how the nanotechnology can enhance antigen presentation and strong immunogenicity using advanced technology in nanocarrier to deliver antigens. The discussion about their safe, effective, and affordable vaccines to immunize against COVID-19 will be highlighted.
Collapse
Affiliation(s)
- Nawamin Sa-nguanmoo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Uracha Ruktanonchai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - YongXiang Zhao
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumour Theranostics and Therapy, Guangxi Medical University, Nanning, 530021 China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
12
|
Daian e Silva DSDO, da Fonseca FG. The Rise of Vectored Vaccines: A Legacy of the COVID-19 Global Crisis. Vaccines (Basel) 2021; 9:1101. [PMID: 34696209 PMCID: PMC8538930 DOI: 10.3390/vaccines9101101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
The COVID-19 pandemic represents a milestone in vaccine research and development in a global context. A worldwide effort, as never seen before, involved scientists from all over the world in favor of the fast, accurate and precise construction and testing of immunogens against the new coronavirus, SARS-CoV-2. Among all the vaccine strategies put into play for study and validation, those based on recombinant viral vectors gained special attention due to their effectiveness, ease of production and the amplitude of the triggered immune responses. Some of these new vaccines have already been approved for emergency/full use, while others are still in pre- and clinical trials. In this article we will highlight what is behind adeno-associated vectors, such as those presented by the immunogens ChaAdOx1, Sputnik, Convidecia (CanSino, Tianjin, China), and Janssen (Johnson & Johnson, New Jersey, EUA), in addition to other promising platforms such as Vaccinia virus MVA, influenza virus, and measles virus, among others.
Collapse
Affiliation(s)
- Danielle Soares de Oliveira Daian e Silva
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
- CT Vacinas, BH-TEC Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31310-260, Brazil
| |
Collapse
|
13
|
Shalash AO, Hussein WM, Skwarczynski M, Toth I. Key Considerations for the Development of Safe and Effective SARS-CoV-2 Subunit Vaccine: A Peptide-Based Vaccine Alternative. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100985. [PMID: 34176237 PMCID: PMC8373118 DOI: 10.1002/advs.202100985] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/18/2021] [Indexed: 05/14/2023]
Abstract
COVID-19 is disastrous to global health and the economy. SARS-CoV-2 infection exhibits similar clinical symptoms and immunopathological sequelae to SARS-CoV infection. Therefore, much of the developmental progress on SARS-CoV vaccines can be utilized for the development of SARS-CoV-2 vaccines. Careful antigen selection during development is always of utmost importance for the production of effective vaccines that do not compromise recipient safety. This holds especially true for SARS-CoV vaccines, as several immunopathological disorders are associated with the activity of structural and nonstructural proteins encoded in the virus's genetic material. Whole viral protein and RNA-encoding full-length proteins contain both protective and "dangerous" sequences, unless pathological fragments are deleted. In light of recent advances, peptide vaccines may present a very safe and effective alternative. Peptide vaccines can avoid immunopathological pro-inflammatory sequences, focus immune responses on neutralizing immunogenic epitopes, avoid off-target antigen loss, combine antigens with different protective roles or mechanisms, even from different viral proteins, and avoid mutant escape by employing highly conserved cryptic epitopes. In this review, an attempt is made to exploit the similarities between SARS-CoV and SARS-CoV-2 in vaccine antigen screening, with particular attention to the pathological and immunogenic properties of SARS proteins.
Collapse
Affiliation(s)
- Ahmed O. Shalash
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt. LuciaQLD4072Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt. LuciaQLD4072Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt. LuciaQLD4072Australia
| | - Istvan Toth
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt. LuciaQLD4072Australia
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaQLD4072Australia
- School of PharmacyThe University of QueenslandWoolloongabbaQLD4102Australia
| |
Collapse
|
14
|
Motamedi H, Ari MM, Dashtbin S, Fathollahi M, Hossainpour H, Alvandi A, Moradi J, Abiri R. An update review of globally reported SARS-CoV-2 vaccines in preclinical and clinical stages. Int Immunopharmacol 2021; 96:107763. [PMID: 34162141 PMCID: PMC8101866 DOI: 10.1016/j.intimp.2021.107763] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/21/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the rapidly spreading pandemic COVID-19 in the world. As an effective therapeutic strategy is not introduced yet and the rapid genetic variations in the virus, there is an emerging necessity to design, evaluate and apply effective new vaccines. An acceptable vaccine must elicit both humoral and cellular immune responses, must have the least side effects and the storage and transport systems should be available and affordable for all countries. These vaccines can be classified into different types: inactivated vaccines, live-attenuated virus vaccines, subunit vaccines, virus-like particles (VLPs), nucleic acid-based vaccines (DNA and RNA) and recombinant vector-based vaccines (replicating and non-replicating viral vector). According to the latest update of the WHO report on April 2nd, 2021, at least 85 vaccine candidates were being studied in clinical trial phases and 184 candidate vaccines were being evaluated in pre-clinical stages. In addition, studies have shown that other vaccines, including the Bacillus Calmette-Guérin (BCG) vaccine and the Plant-derived vaccine, may play a role in controlling pandemic COVID-19. Herein, we reviewed the different types of COVID-19 candidate vaccines that are currently being evaluated in preclinical and clinical trial phases along with advantages, disadvantages or adverse reactions, if any.
Collapse
Affiliation(s)
- Hamid Motamedi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shirin Dashtbin
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Matin Fathollahi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hadi Hossainpour
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amirhoushang Alvandi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran; Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Jale Moradi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ramin Abiri
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
15
|
Afify MA, Alqahtani RM, Alzamil MAM, Khorshid FA, Almarshedy SM, Alattas SG, Alrawaf TN, Bin-Jumah M, Abdel-Daim MM, Almohideb M. Correlation between polio immunization coverage and overall morbidity and mortality for COVID-19: an epidemiological study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:34611-34618. [PMID: 33651292 PMCID: PMC7923406 DOI: 10.1007/s11356-021-12861-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/04/2021] [Indexed: 05/30/2023]
Abstract
We conducted the current analysis to determine the potential role of polio vaccination in the context of the spread of COVID-19. Data were extracted from the World Health Organization's (WHO) Global Health Observatory data repository regarding the polio immunization coverage estimates and correlated to the overall morbidity and mortality for COVID-19 among different countries. Data were analyzed using R software version 4.0.2. Mean and standard deviation were used to represent continuous variables while we used frequencies and percentages to represent categorical variables. The Kruskal-Wallis H test was used for continuous variables since they were not normally distributed. Moreover, the Spearman rank correlation coefficient (rho) was used to determine the relationship between different variables. There was a significantly positive correlation between the vaccine coverage (%) and both of total cases per one million populations (rho = 0.37; p-value < 0.001) and deaths per one million populations (rho = 0.30; p-value < 0.001). Moreover, there was a significant correlation between different income groups and each of vaccine coverage (%) (rho = 0.71; p-value < 0.001), total cases per one million populations (rho = 0.50; p-value < 0.001), and deaths per one million populations (rho = 0.39; p-value < 0.001). All claims regarding the possible protective effect of Polio vaccination do not have any support when analyzing the related data. Polio vaccination efforts should be limited to eradicate the disease from endemic countries; however, there is no evidence to support the immunization with live-attenuated vaccines for the protection against COVID-19.
Collapse
Affiliation(s)
- Marwa Adel Afify
- Potion CRO, Integrative Medicine Company, Al Malqa, Riyadh, 13524 Saudi Arabia
| | - Rakan M. Alqahtani
- Department of Critical Care Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Faten Abdulrahman Khorshid
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
| | - Sumayyah Mohammad Almarshedy
- Division of Adult Neurology, Department of Internal Medicine, College of medicine, University of Hail, Hail, Saudi Arabia
| | - Sana Ghazi Alattas
- Biological Sciences Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - May Bin-Jumah
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed M. Abdel-Daim
- Department of Zoology, Science College, King Saud University, Riyadh, 11451 Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522 Egypt
| | - Mohammad Almohideb
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Smith CC, Olsen KS, Gentry KM, Sambade M, Beck W, Garness J, Entwistle S, Willis C, Vensko S, Woods A, Fini M, Carpenter B, Routh E, Kodysh J, O'Donnell T, Haber C, Heiss K, Stadler V, Garrison E, Sandor AM, Ting JPY, Weiss J, Krajewski K, Grant OC, Woods RJ, Heise M, Vincent BG, Rubinsteyn A. Landscape and selection of vaccine epitopes in SARS-CoV-2. Genome Med 2021; 13:101. [PMID: 34127050 PMCID: PMC8201469 DOI: 10.1186/s13073-021-00910-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 05/14/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Early in the pandemic, we designed a SARS-CoV-2 peptide vaccine containing epitope regions optimized for concurrent B cell, CD4+ T cell, and CD8+ T cell stimulation. The rationale for this design was to drive both humoral and cellular immunity with high specificity while avoiding undesired effects such as antibody-dependent enhancement (ADE). METHODS We explored the set of computationally predicted SARS-CoV-2 HLA-I and HLA-II ligands, examining protein source, concurrent human/murine coverage, and population coverage. Beyond MHC affinity, T cell vaccine candidates were further refined by predicted immunogenicity, sequence conservation, source protein abundance, and coverage of high frequency HLA alleles. B cell epitope regions were chosen from linear epitope mapping studies of convalescent patient serum, followed by filtering for surface accessibility, sequence conservation, spatial localization near functional domains of the spike glycoprotein, and avoidance of glycosylation sites. RESULTS From 58 initial candidates, three B cell epitope regions were identified. From 3730 (MHC-I) and 5045 (MHC-II) candidate ligands, 292 CD8+ and 284 CD4+ T cell epitopes were identified. By combining these B cell and T cell analyses, as well as a manufacturability heuristic, we proposed a set of 22 SARS-CoV-2 vaccine peptides for use in subsequent murine studies. We curated a dataset of ~ 1000 observed T cell epitopes from convalescent COVID-19 patients across eight studies, showing 8/15 recurrent epitope regions to overlap with at least one of our candidate peptides. Of the 22 candidate vaccine peptides, 16 (n = 10 T cell epitope optimized; n = 6 B cell epitope optimized) were manually selected to decrease their degree of sequence overlap and then synthesized. The immunogenicity of the synthesized vaccine peptides was validated using ELISpot and ELISA following murine vaccination. Strong T cell responses were observed in 7/10 T cell epitope optimized peptides following vaccination. Humoral responses were deficient, likely due to the unrestricted conformational space inhabited by linear vaccine peptides. CONCLUSIONS Overall, we find our selection process and vaccine formulation to be appropriate for identifying T cell epitopes and eliciting T cell responses against those epitopes. Further studies are needed to optimize prediction and induction of B cell responses, as well as study the protective capacity of predicted T and B cell epitopes.
Collapse
Affiliation(s)
- Christof C Smith
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Kelly S Olsen
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Kaylee M Gentry
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Maria Sambade
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Wolfgang Beck
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Jason Garness
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Sarah Entwistle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Caryn Willis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Steven Vensko
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Allison Woods
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Misha Fini
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Brandon Carpenter
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Eric Routh
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Julia Kodysh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy O'Donnell
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Erik Garrison
- Genomics Institute, University of California, Santa Cruz, CA, USA
| | - Adam M Sandor
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Jenny P Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
- Institute for Inflammatory Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jared Weiss
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Division of Medical Oncology, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA
| | - Krzysztof Krajewski
- Department of Biochemistry and Biophysics, UNC School of Medicine, Chapel Hill, NC, USA
| | - Oliver C Grant
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Robert J Woods
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Mark Heise
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
| | - Benjamin G Vincent
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA.
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA.
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, NC, USA.
- Division of Hematology, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA.
| | - Alex Rubinsteyn
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA.
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA.
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
17
|
Yengil E, Onlen Y, Ozer C, Hambolat M, Ozdogan M. Effectiveness of Booster Measles-Mumps-Rubella Vaccination in Lower COVID-19 Infection Rates: A Retrospective Cohort Study in Turkish Adults. Int J Gen Med 2021; 14:1757-1762. [PMID: 33994804 PMCID: PMC8113608 DOI: 10.2147/ijgm.s309022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/23/2021] [Indexed: 12/18/2022] Open
Abstract
Objective The aim of this study was to investigate the effectiveness of booster vaccination of adults with measles-mumps-rubella in the COVID-19 infection rates. Methods In order to investigate this hypothesis, we tested COVID-19 positivity rate through PCR assay on the participants (n=245; male), who had to share the same student accommodation together with the same dining hall to provide governmental service. Participants were divided into two groups based on their booster vaccination status with measles-mumps-rubella: the non-vaccinated group (n=207) and the vaccinated group (n=38). The rate of COVID-19 seropositivity, age, body mass index (BMI), active smoking and presence of comorbidity were also measured and recorded. Results All of the participants were healthy, and age distribution, comorbidity rates, active smoking status and BMI did not vary significantly among the two groups (p=0.305, p=0.594, p=0.280, and p=0.922, respectively). About 36.7% (n=90) of the participants were found to be COVID-19 positive by PCR among which the non-vaccinated cases had higher rates of COVID-19 seropositivity than the vaccinated cases (40.6% vs 15.8%) (OR=3.6, 95%CI: 1.5–9.0, p=0.004). Conclusion Based on these results, we cautiously predict that immunity produced by MMR vaccination boosters may provide some degree of protection against COVID-19 in the adult population.
Collapse
Affiliation(s)
- Erhan Yengil
- Department of Family Medicine, Hatay Mustafa Kemal University School of Medicine, Hatay, Turkey
| | - Yusuf Onlen
- Department of Infectious Disease and Clinical Microbiology, Hatay Mustafa Kemal University School of Medicine, Hatay, Turkey
| | - Cahit Ozer
- Department of Family Medicine, Hatay Mustafa Kemal University School of Medicine, Hatay, Turkey
| | | | - Mehmet Ozdogan
- Head of Provincial Public Health Department, Hatay, Turkey
| |
Collapse
|
18
|
Baldo A, Leunda A, Willemarck N, Pauwels K. Environmental Risk Assessment of Recombinant Viral Vector Vaccines against SARS-Cov-2. Vaccines (Basel) 2021; 9:453. [PMID: 34063733 PMCID: PMC8147846 DOI: 10.3390/vaccines9050453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/23/2021] [Accepted: 05/01/2021] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the coronavirus disease 2019 (COVID-19) pandemic. Over the past months, considerable efforts have been put into developing effective and safe drugs and vaccines against SARS-CoV-2. Various platforms are being used for the development of COVID-19 vaccine candidates: recombinant viral vectors, protein-based vaccines, nucleic acid-based vaccines, and inactivated/attenuated virus. Recombinant viral vector vaccine candidates represent a significant part of those vaccine candidates in clinical development, with two already authorised for use in the European Union and one currently under rolling review by the European Medicines Agency (EMA). Since recombinant viral vector vaccine candidates are considered as genetically modified organisms (GMOs), their regulatory oversight includes besides an assessment of their quality, safety and efficacy, also an environmental risk assessment (ERA). The present article highlights the main characteristics of recombinant viral vector vaccine (candidates) against SARS-CoV-2 in the pipeline and discusses their features from an environmental risk point of view.
Collapse
Affiliation(s)
- Aline Baldo
- Sciensano, Service Biosafety and Biotechnology, Rue Juliette Wytsmanstraat 14, B-1050 Brussels, Belgium; (A.L.); (N.W.); (K.P.)
| | | | | | | |
Collapse
|
19
|
Nagy A, Alhatlani B. An overview of current COVID-19 vaccine platforms. Comput Struct Biotechnol J 2021; 19:2508-2517. [PMID: 33936564 PMCID: PMC8076774 DOI: 10.1016/j.csbj.2021.04.061] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 12/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19) pandemic that emerged in December 2019 in Wuhan city, China. An effective vaccine is urgently needed to protect humans and to mitigate the economic and societal impacts of the pandemic. Despite standard vaccine development usually requiring an extensive process and taking several years to complete all clinical phases, there are currently 184 vaccine candidates in pre-clinical testing and another 88 vaccine candidates in clinical phases based on different vaccine platforms as of April 13, 2021. Moreover, three vaccine candidates have recently been granted an Emergency Use Authorization by the United States Food and Drug Administration (for Pfizer/BioNtech, Moderna mRNA vaccines, and Johnson and Johnson viral vector vaccine) and by the UK government (for University of Oxford/AstraZeneca viral vector vaccine). Here we aim to briefly address the current advances in reverse genetics system of SARS-CoV-2 and the use of this in development of SARS-CoV-2 vaccines. Additionally, we cover the essential points concerning the different platforms of current SARS-CoV-2 vaccine candidates and the advantages and drawbacks of these platforms. We also assess recommendations for controlling the COVID-19 pandemic and future pandemics using the benefits of genetic engineering technology to design effective vaccines against emerging and re-emerging viral diseases with zoonotic and/or pandemic potential.
Collapse
Affiliation(s)
- Abdou Nagy
- Department of Virology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia 44511, Egypt
| | - Bader Alhatlani
- Department of Applied Medical Sciences, Unayzah Community College, Qassim University, Unayzah, Saudi Arabia
| |
Collapse
|
20
|
A safe and highly efficacious measles virus-based vaccine expressing SARS-CoV-2 stabilized prefusion spike. Proc Natl Acad Sci U S A 2021; 118:2026153118. [PMID: 33688034 PMCID: PMC8000430 DOI: 10.1073/pnas.2026153118] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Measles virus (MeV) vaccine is one of the safest and most efficient vaccines with a track record in children. Here, we generated a panel of rMeV-based vaccines with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) S antigens inserted near 3′ of the MeV genome. The rMeV expressing a soluble stabilized, prefusion spike (preS) is much more potent in triggering SARS-CoV-2–specific neutralizing antibody than rMeV-based full-length S vaccine candidate. A single dose of rMeV-preS is sufficient to induce high levels of SARS-CoV-2 antibody in animals. Furthermore, rMeV-preS induces high levels of Th1-biased immunity. Hamsters immunized with rMeV-preS were completely protected against SARS-CoV-2 challenge. Our results demonstrate rMeV-preS is a safe and highly efficacious bivalent vaccine candidate for SARS-CoV-2 and MeV. The current pandemic of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) highlights an urgent need to develop a safe, efficacious, and durable vaccine. Using a measles virus (rMeV) vaccine strain as the backbone, we developed a series of recombinant attenuated vaccine candidates expressing various forms of the SARS-CoV-2 spike (S) protein and its receptor binding domain (RBD) and evaluated their efficacy in cotton rat, IFNAR−/−mice, IFNAR−/−-hCD46 mice, and golden Syrian hamsters. We found that rMeV expressing stabilized prefusion S protein (rMeV-preS) was more potent in inducing SARS-CoV-2–specific neutralizing antibodies than rMeV expressing full-length S protein (rMeV-S), while the rMeVs expressing different lengths of RBD (rMeV-RBD) were the least potent. Animals immunized with rMeV-preS produced higher levels of neutralizing antibody than found in convalescent sera from COVID-19 patients and a strong Th1-biased T cell response. The rMeV-preS also provided complete protection of hamsters from challenge with SARS-CoV-2, preventing replication in lungs and nasal turbinates, body weight loss, cytokine storm, and lung pathology. These data demonstrate that rMeV-preS is a safe and highly efficacious vaccine candidate, supporting its further development as a SARS-CoV-2 vaccine.
Collapse
|
21
|
Zhou P, Li Z, Xie L, An D, Fan Y, Wang X, Li Y, Liu X, Wu J, Li G, Li Q. Research progress and challenges to coronavirus vaccine development. J Med Virol 2021; 93:741-754. [PMID: 32936465 DOI: 10.1002/jmv.26517] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/29/2020] [Accepted: 08/06/2020] [Indexed: 01/07/2023]
Abstract
Coronaviruses (CoVs) are nonsegmented, single-stranded, positive-sense RNA viruses highly pathogenic to humans. Some CoVs are known to cause respiratory and intestinal diseases, posing a threat to the global public health. Against this backdrop, it is of critical importance to develop safe and effective vaccines against these CoVs. This review discusses human vaccine candidates in any stage of development and explores the viral characteristics, molecular epidemiology, and immunology associated with CoV vaccine development. At present, there are many obstacles and challenges to vaccine research and development, including the lack of knowledge about virus transmission, pathogenesis, and immune response, absence of the most appropriate animal models.
Collapse
Affiliation(s)
- Peiwen Zhou
- Department of Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Zonghui Li
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linqing Xie
- Department of Guangzhou Cyanvaccine Biotechnology Co, Ltd, Guangzhou, China
| | - Dong An
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Yaohua Fan
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiao Wang
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiwei Li
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong Liu
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianguo Wu
- Department of Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Geng Li
- Department of Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qin Li
- Department of Guangzhou Cyanvaccine Biotechnology Co, Ltd, Guangzhou, China
| |
Collapse
|
22
|
Rasker JJ, Linn-Rasker SP. VACCINATION WITH MMR MAY REDUCE DISEASE SEVERITY IN COVID-19 PATIENTS. CENTRAL ASIAN JOURNAL OF MEDICAL HYPOTHESES AND ETHICS 2021. [DOI: 10.47316/cajmhe.2020.1.2.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We hypothesise that MMR vaccination is possibly a safe, cheap, effective and readily available method to reduce the severity of COVID-19 disease course in health care workers, elderly patients and other people at risk. The evidence is based on relevant literature. Suggestions for further studies are given.
Collapse
|
23
|
Soleimanpour S, Yaghoubi A. COVID-19 vaccine: where are we now and where should we go? Expert Rev Vaccines 2021; 20:23-44. [PMID: 33435774 PMCID: PMC7898300 DOI: 10.1080/14760584.2021.1875824] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 01/11/2021] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has currently caused the pandemic with a high progressive speed and has been considered as the global public health crisis in 2020. This new member of the coronavirus family has created a potentially fatal disease, called coronavirus disease-2019 (COVID-19). Despite the continuous efforts of researchers to find effective vaccines and drugs for COVID-19, there is still no success in this matter. AREAS COVERED Here, the literature regarding the COVID-19 vaccine candidates currently in the clinical trials, as well as main candidates in pre-clinical stages for development and research, were reviewed. These candidates have been developed under five different major platforms, including live-attenuated vaccine, mRNA-based vaccine, DNA vaccines, inactivated virus, and viral-vector-based vaccine. EXPERT OPINION There are several limitations in the field of the rapid vaccine development against SARS-CoV-2, and other members of the coronavirus family such as SARS-CoV and MERS-CoV. The key challenges of designing an effective vaccine within a short time include finding the virulence ability of an emerging virus and potential antigen, choosing suitable experimental models and efficient route of administration, the immune-response study, designing the clinical trials, and determining the safety, as well as efficacy.
Collapse
Affiliation(s)
- Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atieh Yaghoubi
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Mathew S, Faheem M, Hassain NA, Benslimane FM, Al Thani AA, Zaraket H, Yassine HM. Platforms Exploited for SARS-CoV-2 Vaccine Development. Vaccines (Basel) 2020; 9:11. [PMID: 33375677 PMCID: PMC7824029 DOI: 10.3390/vaccines9010011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/18/2022] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the only zoonotic-origin coronavirus (CoV) that has reached the pandemic stage. The virus uses its spike (S) glycoprotein to attach to the host cells and initiate a cascade of events that leads to infection. It has sternly affected public health, economy, education, and social behavior around the world. Several scientific and medical communities have mounted concerted efforts to limit this pandemic and the subsequent wave of viral spread by developing preventative and potential vaccines. So far, no medicine or vaccine has been approved to prevent or treat coronavirus disease 2019 (COVID-19). This review describes the latest advances in the development of SARS-CoV-2 vaccines for humans, mainly focusing on the lead candidates in clinical trials. Moreover, we seek to provide both the advantages and the disadvantages of the leading platforms used in current vaccine development, based on past vaccine delivery efforts for non-SARS CoV-2 infections. We also highlight the population groups who should receive a vaccine against COVID-19 in a timely manner to eradicate the pandemic rapidly.
Collapse
Affiliation(s)
- Shilu Mathew
- Biomedical Research Center, Qatar University, Doha 2173, Qatar; (S.M.); (F.M.B.); (A.A.A.T.)
| | - Muhammed Faheem
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada;
| | - Neeraja A. Hassain
- Department of Biotechnology, Jamal Mohamed College, Tamil Nadu 620020, India;
| | - Fatiha M. Benslimane
- Biomedical Research Center, Qatar University, Doha 2173, Qatar; (S.M.); (F.M.B.); (A.A.A.T.)
| | - Asmaa A. Al Thani
- Biomedical Research Center, Qatar University, Doha 2173, Qatar; (S.M.); (F.M.B.); (A.A.A.T.)
- Department of Public Health, College of Health Sciences, Qatar University, Doha 2173, Qatar
| | - Hassan Zaraket
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 11-0236, Lebanon;
- Center for Infectious Diseases Research, American University of Beirut, Beirut 11-0236, Lebanon
| | - Hadi M. Yassine
- Biomedical Research Center, Qatar University, Doha 2173, Qatar; (S.M.); (F.M.B.); (A.A.A.T.)
- Department of Public Health, College of Health Sciences, Qatar University, Doha 2173, Qatar
| |
Collapse
|
25
|
Zhao J, Zhao S, Ou J, Zhang J, Lan W, Guan W, Wu X, Yan Y, Zhao W, Wu J, Chodosh J, Zhang Q. COVID-19: Coronavirus Vaccine Development Updates. Front Immunol 2020; 11:602256. [PMID: 33424848 PMCID: PMC7785583 DOI: 10.3389/fimmu.2020.602256] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/26/2020] [Indexed: 12/27/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), a newly emerged coronavirus, and has been pandemic since March 2020 and led to many fatalities. Vaccines represent the most efficient means to control and stop the pandemic of COVID-19. However, currently there is no effective COVID-19 vaccine approved to use worldwide except for two human adenovirus vector vaccines, three inactivated vaccines, and one peptide vaccine for early or limited use in China and Russia. Safe and effective vaccines against COVID-19 are in urgent need. Researchers around the world are developing 213 COVID-19 candidate vaccines, among which 44 are in human trials. In this review, we summarize and analyze vaccine progress against SARS-CoV, Middle-East respiratory syndrome Coronavirus (MERS-CoV), and SARS-CoV-2, including inactivated vaccines, live attenuated vaccines, subunit vaccines, virus like particles, nucleic acid vaccines, and viral vector vaccines. As SARS-CoV-2, SARS-CoV, and MERS-CoV share the common genus, Betacoronavirus, this review of the major research progress will provide a reference and new insights into the COVID-19 vaccine design and development.
Collapse
Affiliation(s)
- Jing Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shan Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Junxian Ou
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jing Zhang
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Wendong Lan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenyi Guan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaowei Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuqian Yan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jianguo Wu
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - James Chodosh
- Department of Ophthalmology, Howe Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Qiwei Zhang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| |
Collapse
|
26
|
Leber MF, Neault S, Jirovec E, Barkley R, Said A, Bell JC, Ungerechts G. Engineering and combining oncolytic measles virus for cancer therapy. Cytokine Growth Factor Rev 2020; 56:39-48. [PMID: 32718830 PMCID: PMC7333629 DOI: 10.1016/j.cytogfr.2020.07.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
Cancer immunotherapy using tumor-selective, oncolytic viruses is an emerging therapeutic option for solid and hematologic malignancies. A considerable variety of viruses ranging from small picornaviruses to large poxviruses are currently being investigated as potential candidates. In the early days of virotherapy, non-engineered wild-type or vaccine-strain viruses were employed. However, these viruses often did not fully satisfy the major criteria of safety and efficacy. Since the advent of reverse genetics systems for manipulating various classes of viruses, the field has shifted to developing genetically engineered viruses with an improved therapeutic index. In this review, we will summarize the concepts and strategies of multi-level genetic engineering of oncolytic measles virus, a prime candidate for cancer immunovirotherapy. Furthermore, we will provide a brief overview of measles virus-based multimodal combination therapies for improved tumor control and clinical efficacy.
Collapse
Affiliation(s)
- Mathias F Leber
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Virotherapy, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Heidelberg University Hospital, Department of Medical Oncology, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany; Ottawa Hospital Research Institute, Cancer Therapeutics Program, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
| | - Serge Neault
- Ottawa Hospital Research Institute, Cancer Therapeutics Program, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
| | - Elise Jirovec
- Ottawa Hospital Research Institute, Cancer Therapeutics Program, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
| | - Russell Barkley
- Ottawa Hospital Research Institute, Cancer Therapeutics Program, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
| | - Aida Said
- Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada; University of Ottawa, Faculty of Medicine, Department of Cellular and Molecular Medicine, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - John C Bell
- Ottawa Hospital Research Institute, Cancer Therapeutics Program, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
| | - Guy Ungerechts
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Virotherapy, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Heidelberg University Hospital, Department of Medical Oncology, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany; Ottawa Hospital Research Institute, Cancer Therapeutics Program, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
| |
Collapse
|
27
|
A highly immunogenic and effective measles virus-based Th1-biased COVID-19 vaccine. Proc Natl Acad Sci U S A 2020; 117:32657-32666. [PMID: 33257540 PMCID: PMC7768780 DOI: 10.1073/pnas.2014468117] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The COVID-19 pandemic has already caused over 1 million deaths. Therefore, effective vaccine concepts are urgently needed. In search of such a concept, we have analyzed a measles virus-based vaccine candidate targeting SARS-CoV-2. Using this well-known, safe vaccine backbone, we demonstrate here induction of functional immune responses in both arms of adaptive immunity, yielding antiviral efficacy in vivo with the desired immune bias. Consequently, no immunopathologies became evident during challenge experiments. Moreover, the candidate still induces immunity against the measles, recognized as a looming second menace, when countries are forced to stop routine vaccination campaigns in the face of COVID-19. Thus, a bivalent measles-based COVID-19 vaccine could be the solution for two significant public health threats. The COVID-19 pandemic is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and has spread worldwide, with millions of cases and more than 1 million deaths to date. The gravity of the situation mandates accelerated efforts to identify safe and effective vaccines. Here, we generated measles virus (MeV)-based vaccine candidates expressing the SARS-CoV-2 spike glycoprotein (S). Insertion of the full-length S protein gene in two different MeV genomic positions resulted in modulated S protein expression. The variant with lower S protein expression levels was genetically stable and induced high levels of effective Th1-biased antibody and T cell responses in mice after two immunizations. In addition to neutralizing IgG antibody responses in a protective range, multifunctional CD8+ and CD4+ T cell responses with S protein-specific killing activity were detected. Upon challenge using a mouse-adapted SARS-CoV-2, virus loads in vaccinated mice were significantly lower, while vaccinated Syrian hamsters revealed protection in a harsh challenge setup using an early-passage human patient isolate. These results are highly encouraging and support further development of MeV-based COVID-19 vaccines.
Collapse
|
28
|
Babaei F, Mirzababaei M, Nassiri-Asl M, Hosseinzadeh H. Review of registered clinical trials for the treatment of COVID-19. Drug Dev Res 2020; 82:474-493. [PMID: 33251593 PMCID: PMC7753306 DOI: 10.1002/ddr.21762] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/05/2020] [Accepted: 11/13/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID‐19) is a viral disease caused by severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2). The disease was first reported in December 2019 in Wuhan, China, but now more than 200 countries have been affected and the coronavirus pandemic is still ongoing. The severity of COVID‐19 symptoms can range from mild to severe. FDA approved remdesivir as a treatment of COVID‐19 so far. Various clinical trials are underway to find an effective method to treat patients with COVID‐19. This review aimed at summarizing 219 registered clinical trials in the ClinicalTrials.gov database with possible mechanisms, and novel findings of them, and other recent publications related to COVID‐19. According to our analyses, various treatment approaches and drugs are being investigated to find an effective drug to cure COVID‐19 and among all strategies, three important mechanisms are suggested to be important against COVID‐19 including antiviral, anti‐inflammatory, and immunomodulatory properties. Our review can help future studies get on the way to finding an effective drug for COVID‐19 treatment by providing ideas for similar researches.
Collapse
Affiliation(s)
- Fatemeh Babaei
- Department of Clinical Biochemistry, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Mirzababaei
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marjan Nassiri-Asl
- Department of Pharmacology and Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
29
|
Badgujar KC, Badgujar VC, Badgujar SB. Vaccine development against coronavirus (2003 to present): An overview, recent advances, current scenario, opportunities and challenges. Diabetes Metab Syndr 2020; 14:1361-1376. [PMID: 32755836 PMCID: PMC7371592 DOI: 10.1016/j.dsx.2020.07.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM The pandemic COVID-19 occurring due to novel emerging coronavirus-2019 (SARS-CoV-2) is severely affecting the worldwide public health, culture, economy and human social behaviour. Till date, there is no approved medicine/treatment to cure COVID-19, whereas, vaccine development efforts are going on high priority. This review aimed to provide an overview of prior art, recent advances, vaccine designing strategies, current scenario, opportunities and challenges related to development of coronavirus vaccine. METHOD A literature survey was conducted using Scopus, PubMed and Google Scholar with the search key as: coronavirus vaccine, SARS vaccine, MERS vaccine and COVID-19 vaccine. Articles related to above search query were retrieved, sorted, analyzed and developed into an easy-to-understand review. RESULTS The genome phylogenetic analysis suggested that genomic sequence of SARS-CoV-2 is almost 80% similar to that of SARS-CoV, further both these viruses bind to same host cell receptor ACE-2. Hence it is expected that, previously available literature data about coronavirus vaccine designing may play crucial role in development of rapid vaccine against COVID-19. In view of this, the present review discuss (i) existing information (from 2003 to present) about the type of vaccine, antigen, immunogenic response, animal model, route of administration, adjuvants and current scenario for designing of coronavirus vaccine (ii) potential factors and challenges related to rapid development of COVID-19 vaccine. CONCLUSION In conclusion, we discuss possible clues/ target sites for designing of vaccine against SARS-CoV-2 virus based on prior-art.
Collapse
Affiliation(s)
- Kirtikumar C Badgujar
- Assistant Professor, Department of Chemistry, SIES College of Arts, Science and Commerce, Near SION Hospital, Sion, Mumbai, 400022, Maharashtra, India.
| | - Vivek C Badgujar
- Assistant Professor, Department of Chemistry, Pratap College of Arts, Science and Commerce, Amalner, Dist Jalgaon, 425401, Maharashtra, India
| | - Shamkant B Badgujar
- Scientist, Laboratory of Native Antigens, Research and Development Division, Advy Chemical Private Limited, Thane, 400604, Maharashtra, India.
| |
Collapse
|
30
|
Bozkurt HB. Is the impact of childhood vaccines on coronavirus disease 2019, which is moderate in pediatric patients, possible? Clin Exp Vaccine Res 2020; 9:183-184. [PMID: 32864377 PMCID: PMC7445315 DOI: 10.7774/cevr.2020.9.2.183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/28/2020] [Indexed: 11/15/2022] Open
|
31
|
Frederiksen LSF, Zhang Y, Foged C, Thakur A. The Long Road Toward COVID-19 Herd Immunity: Vaccine Platform Technologies and Mass Immunization Strategies. Front Immunol 2020; 11:1817. [PMID: 32793245 PMCID: PMC7385234 DOI: 10.3389/fimmu.2020.01817] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
There is an urgent need for effective countermeasures against the current emergence and accelerating expansion of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Induction of herd immunity by mass vaccination has been a very successful strategy for preventing the spread of many infectious diseases, hence protecting the most vulnerable population groups unable to develop immunity, for example individuals with immunodeficiencies or a weakened immune system due to underlying medical or debilitating conditions. Therefore, vaccination represents one of the most promising counter-pandemic measures to COVID-19. However, to date, no licensed vaccine exists, neither for SARS-CoV-2 nor for the closely related SARS-CoV or Middle East respiratory syndrome-CoV. In addition, a few vaccine candidates have only recently entered human clinical trials, which hampers the progress in tackling COVID-19 infection. Here, we discuss potential prophylactic interventions for SARS-CoV-2 with a focus on the challenges existing for vaccine development, and we review pre-clinical progress and ongoing human clinical trials of COVID-19 vaccine candidates. Although COVID-19 vaccine development is currently accelerated via so-called fast-track programs, vaccines may not be timely available to have an impact on the first wave of the ongoing COVID-19 pandemic. Nevertheless, COVID-19 vaccines will be essential in the future for reducing morbidity and mortality and inducing herd immunity, if SARS-CoV-2 becomes established in the population like for example influenza virus.
Collapse
Affiliation(s)
| | - Yibang Zhang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Elhusseiny KM, Abd-Elhay FAE, Kamel MG. Possible therapeutic agents for COVID-19: a comprehensive review. Expert Rev Anti Infect Ther 2020; 18:1005-1020. [PMID: 32538209 DOI: 10.1080/14787210.2020.1782742] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has emerged in China. There are no available vaccines or antiviral drugs for COVID-19 patients. Herein, we represented possible therapeutic agents that may stand as a potential therapy against COVID-19. AREAS COVERED We searched PubMed, Google Scholar, and clinicaltrials.gov for relevant papers. We showed some agents with potentially favorable efficacy, acceptable safety as well as good pharmacokinetic profiles. Several therapies are under assessment to evaluate their efficacy and safety for COVID-19. However, some drugs were withdrawn due to their side effects after demonstrating some clinical efficacy. Indeed, the most effective therapies could be organ function support, convalescent plasma, anticoagulants, and immune as well as antiviral therapies, especially anti-influenza drugs due to the similarities between respiratory viruses regarding viral entry, uncoating, and replication. We encourage giving more attention to favipiravir, remdesivir, and measles vaccine. EXPERT OPINION A combination, at least dual or even triple therapy, of the aforementioned efficacious and safe therapies is greatly recommended for COVID-19. Further, patients should have a routine assessment for their coagulation and bleeding profiles as well as their inflammatory and cytokine concentrations.
Collapse
Affiliation(s)
- Khaled Mosaad Elhusseiny
- Faculty of Medicine, Al-Azhar University , Cairo, Egypt.,Sayed Galal University Hospital , Cairo, Egypt.,Egyptian Collaborative Research Team , Egypt
| | | | | |
Collapse
|
33
|
Smith CC, Entwistle S, Willis C, Vensko S, Beck W, Garness J, Sambade M, Routh E, Olsen K, Kodysh J, O’Donnell T, Haber C, Heiss K, Stadler V, Garrison E, Grant OC, Woods RJ, Heise M, Vincent BG, Rubinsteyn A. Landscape and Selection of Vaccine Epitopes in SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.06.04.135004. [PMID: 32577654 PMCID: PMC7302209 DOI: 10.1101/2020.06.04.135004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
There is an urgent need for a vaccine with efficacy against SARS-CoV-2. We hypothesize that peptide vaccines containing epitope regions optimized for concurrent B cell, CD4+ T cell, and CD8+ T cell stimulation would drive both humoral and cellular immunity with high specificity, potentially avoiding undesired effects such as antibody-dependent enhancement (ADE). Additionally, such vaccines can be rapidly manufactured in a distributed manner. In this study, we combine computational prediction of T cell epitopes, recently published B cell epitope mapping studies, and epitope accessibility to select candidate peptide vaccines for SARS-CoV-2. We begin with an exploration of the space of possible T cell epitopes in SARS-CoV-2 with interrogation of predicted HLA-I and HLA-II ligands, overlap between predicted ligands, protein source, as well as concurrent human/murine coverage. Beyond MHC affinity, T cell vaccine candidates were further refined by predicted immunogenicity, viral source protein abundance, sequence conservation, coverage of high frequency HLA alleles and co-localization of CD4+ and CD8+ T cell epitopes. B cell epitope regions were chosen from linear epitope mapping studies of convalescent patient serum, followed by filtering to select regions with surface accessibility, high sequence conservation, spatial localization near functional domains of the spike glycoprotein, and avoidance of glycosylation sites. From 58 initial candidates, three B cell epitope regions were identified. By combining these B cell and T cell analyses, as well as a manufacturability heuristic, we propose a set of SARS-CoV-2 vaccine peptides for use in subsequent murine studies and clinical trials.
Collapse
Affiliation(s)
- Christof C. Smith
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sarah Entwistle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Caryn Willis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Steven Vensko
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Wolfgang Beck
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jason Garness
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Maria Sambade
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Eric Routh
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kelly Olsen
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Julia Kodysh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Timothy O’Donnell
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Erik Garrison
- Genomics Institute, University of California, Santa Cruz, California
| | - Oliver C. Grant
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Robert J. Woods
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Mark Heise
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, UNC School of Medicine, Chapel Hill, North Carolina
| | - Benjamin G. Vincent
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, North Carolina
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, North Carolina
- Division of Hematology/Oncology, Department of Medicine, UNC School of Medicine, Chapel Hill, North Carolina
| | - Alex Rubinsteyn
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, UNC School of Medicine, Chapel Hill, North Carolina
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
34
|
Guo G, Ye L, Pan K, Chen Y, Xing D, Yan K, Chen Z, Ding N, Li W, Huang H, Zhang L, Li X, Xue X. New Insights of Emerging SARS-CoV-2: Epidemiology, Etiology, Clinical Features, Clinical Treatment, and Prevention. Front Cell Dev Biol 2020; 8:410. [PMID: 32574318 PMCID: PMC7256189 DOI: 10.3389/fcell.2020.00410] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023] Open
Abstract
Since the first reports that the novel coronavirus was showing human-to-human transmission characteristics and asymptomatic cases, the number of patients with associated pneumonia has continued to rise and the epidemic has grown. It now threatens the health and lives of people across the world. The governments of many countries have attached great importance to the prevention of SARS-CoV-2, via research into the etiology and epidemiology of this newly emerged disease. Clinical signs, treatment, and prevention characteristics of the novel coronavirus pneumonia have been receiving attention worldwide, especially from medical personnel. However, owing to the different experimental methods, sample sizes, sample sources, and research perspectives of various studies, results have been inconsistent, or relate to an isolated aspect of the virus or the disease it causes. Currently, systematic summary data on the novel coronavirus are limited. This review combines experimental and clinical evidence into a systematic analysis and summary of the current progress of research into SARS-CoV-2, from multiple perspectives, with the aim of gaining a better overall understanding of the disease. Our report provides important information for current clinicians, for the prevention and treatment of COVID-19 pneumonia.
Collapse
Affiliation(s)
- Gangqiang Guo
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Lele Ye
- Department of Gynecologic Oncology, Wenzhou Central Hospital, Wenzhou, China
| | - Kan Pan
- First Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Yu Chen
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Dong Xing
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Kejing Yan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhiyuan Chen
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ning Ding
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Wenshu Li
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hong Huang
- Center for Health Assessment, Wenzhou Medical University, Wenzhou, China
| | - Lifang Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Institute of Virology, Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Xue
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
- Institute of Virology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Tricistronic Plasmid Expressing the T7 RNA Polymerase and Measles Virus N and P Proteins for Rescue of Measles Virus AIK-C Strain. ARCHIVES OF PEDIATRIC INFECTIOUS DISEASES 2020. [DOI: 10.5812/pedinfect.100928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
: Up to now, several attenuated measles virus vaccine strains derived from the Edmonston B vaccine consisting of Schwarz/Moraten, Zagreb, and AIK-C have been introduced for the rescue of their relative viruses through reverse genetics. In most studies, the measles virus rescue was done by supplying a cell line that expresses T7 RNA polymerase and measles virus N and P proteins as accessory proteins. The present study aimed to evaluate the rescue efficiency of the recombinant measles virus AIK-C vaccine strain by using a tricistronic expression plasmid. In this study, the rescue of the recombinant measles virus AIK-C vaccine strain was performed by co-transfection of three plasmids, including the cloned antigenomic cDNA of measles virus, a tricistronic expression plasmid that contained T7 RNA polymerase and measles virus N and P genes, and measles virus L polymerase expression plasmid. To increase the rescue efficiency, the transfected HEK-293 cells were co-cultured with Vero cells. As a result, the use of tricistronic expression plasmid that concomitantly encoded three necessary genes for the rescue of the measles virus led to the viral cytopathic effect with high efficacy five days post-transfection. Finally, the co-culture of transfected HEK-293 cells with Vero cells showed a relatively fast induction of viral cytopathic effect.
Collapse
|
36
|
Zhang L, Liu Y. Potential interventions for novel coronavirus in China: A systematic review. J Med Virol 2020; 92:479-490. [PMID: 32052466 PMCID: PMC7166986 DOI: 10.1002/jmv.25707] [Citation(s) in RCA: 714] [Impact Index Per Article: 142.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 12/14/2022]
Abstract
An outbreak of a novel coronavirus (COVID‐19 or 2019‐CoV) infection has posed significant threats to international health and the economy. In the absence of treatment for this virus, there is an urgent need to find alternative methods to control the spread of disease. Here, we have conducted an online search for all treatment options related to coronavirus infections as well as some RNA‐virus infection and we have found that general treatments, coronavirus‐specific treatments, and antiviral treatments should be useful in fighting COVID‐19. We suggest that the nutritional status of each infected patient should be evaluated before the administration of general treatments and the current children's RNA‐virus vaccines including influenza vaccine should be immunized for uninfected people and health care workers. In addition, convalescent plasma should be given to COVID‐19 patients if it is available. In conclusion, we suggest that all the potential interventions be implemented to control the emerging COVID‐19 if the infection is uncontrollable.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
37
|
O’Connell AK, Douam F. Humanized Mice for Live-Attenuated Vaccine Research: From Unmet Potential to New Promises. Vaccines (Basel) 2020; 8:E36. [PMID: 31973073 PMCID: PMC7157703 DOI: 10.3390/vaccines8010036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 01/24/2023] Open
Abstract
Live-attenuated vaccines (LAV) represent one of the most important medical innovations in human history. In the past three centuries, LAV have saved hundreds of millions of lives, and will continue to do so for many decades to come. Interestingly, the most successful LAVs, such as the smallpox vaccine, the measles vaccine, and the yellow fever vaccine, have been isolated and/or developed in a purely empirical manner without any understanding of the immunological mechanisms they trigger. Today, the mechanisms governing potent LAV immunogenicity and long-term induced protective immunity continue to be elusive, and therefore hamper the rational design of innovative vaccine strategies. A serious roadblock to understanding LAV-induced immunity has been the lack of suitable and cost-effective animal models that can accurately mimic human immune responses. In the last two decades, human-immune system mice (HIS mice), i.e., mice engrafted with components of the human immune system, have been instrumental in investigating the life-cycle and immune responses to multiple human-tropic pathogens. However, their use in LAV research has remained limited. Here, we discuss the strong potential of LAVs as tools to enhance our understanding of human immunity and review the past, current and future contributions of HIS mice to this endeavor.
Collapse
Affiliation(s)
| | - Florian Douam
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA 02118, USA;
| |
Collapse
|
38
|
Zhou D, Zhu MY, Wang YL, Hao XQ, Zhou DM, Liu RX, Zhang CD, Qu CF, Zhao ZY. Attenuated MuV-S79 as vector stably expressing foreign gene. World J Pediatr 2019; 15:511-515. [PMID: 31377975 DOI: 10.1007/s12519-019-00287-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/02/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND To describe mumps virus (MuV) used as a vector to express enhanced green fluorescent protein (EGFP) or red fluorescent protein (RFP) genes. METHODS Molecular cloning technique was applied to establish the cDNA clones of recombinant mumps viruses (rMuVs). rMuVs were recovered based on our reverse genetic system of MuV-S79. The properties of rMuVs were determined by growth curve, plaque assay, fluorescent microscopy and determination of fluorescent intensity. RESULTS Three recombinant viruses replicated well in Vero cells and similarly as parental rMuV-S79, expressed heterologous genes in high levels, and were genetically stable in at least 15 passages. CONCLUSION rMuV-S79 is a promising platform to accommodate foreign genes like marker genes, other antigens and immunomodulators for addressing various diseases.
Collapse
Affiliation(s)
- Duo Zhou
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Meng-Ying Zhu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yi-Long Wang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Xiao-Qiang Hao
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dong-Ming Zhou
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Rong-Xian Liu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chu-Di Zhang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chu-Fan Qu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zheng-Yan Zhao
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
39
|
Gerke C, Frantz PN, Ramsauer K, Tangy F. Measles-vectored vaccine approaches against viral infections: a focus on Chikungunya. Expert Rev Vaccines 2019; 18:393-403. [PMID: 30601074 DOI: 10.1080/14760584.2019.1562908] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The large global burden of viral infections and especially the rapidly spreading vector-borne diseases and other emerging viral diseases show the need for new approaches in vaccine development. Several new vaccine technology platforms have been developed and are under evaluation. Areas covered: This article discusses the measles vector platform technology derived from the safe and highly efficacious measles virus vaccine. The pipeline of measles-vectored vaccine candidates against viral diseases is reviewed. Particular focus is given to the Chikungunya vaccine candidate as the first measles-vectored vaccine that demonstrated safety, immunogenicity, and functionality of the technology in humans even in the presence of pre-existing anti-measles immunity and thus achieved proof of concept for the technology. Expert commentary: Demonstrating no impact of pre-existing anti-measles immunity in humans on the response to the transgene was fundamental for the technology and indicates that the technology is suitable for large-scale immunization in measles pre-immune populations. The proof of concept in humans combined with a large preclinical track record of safety, immunogenicity, and efficacy for a variety of pathogens suggest the measles vector platform as promising plug-and-play vaccine platform technology for rapid development of effective preventive vaccines against viral and other infectious diseases.
Collapse
Affiliation(s)
| | - Phanramphoei N Frantz
- b Viral Genomics and Vaccination Unit, UMR-3569 CNRS, Department of Virology , Institut Pasteur , Paris , France.,c Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC) , National Science and Technology Development Agency , Pathumthani , Thailand
| | | | - Frédéric Tangy
- b Viral Genomics and Vaccination Unit, UMR-3569 CNRS, Department of Virology , Institut Pasteur , Paris , France
| |
Collapse
|
40
|
Frantz PN, Teeravechyan S, Tangy F. Measles-derived vaccines to prevent emerging viral diseases. Microbes Infect 2018; 20:493-500. [PMID: 29410084 PMCID: PMC7110469 DOI: 10.1016/j.micinf.2018.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 02/03/2023]
Abstract
Infectious disease epidemics match wars and natural disasters in their capacity to threaten lives and damage economies. Like SARS previously and Zika recently, the Ebola crisis in 2015 showed how vulnerable the world is to these epidemics, with over 11,000 people dying in the outbreak. In addition to causing immense human suffering, these epidemics particularly affect low- and middle-income countries. Many of these deadly infectious diseases that have epidemic potential can become global health emergencies in the absence of effective vaccines. But very few vaccines against these threats have been developed to create proven medical products. The measles vaccine is an efficient, live attenuated, replicating virus that has been safely administered to 2 billion children over the last 40 years, affording life-long protection after a single dose. Taking advantage of these characteristics, this attenuated virus was transformed into a versatile chimeric or recombinant vaccine vector with demonstrated proof-of-principle in humans and a preclinical track record of rapid adaptability and effectiveness for a variety of pathogens. Clinical trials have shown the safety and immunogenicity of this vaccine platform in individuals with preexisting immunity to measles. This review describes the potential of this platform to develop new vaccines against emerging viral diseases.
Collapse
Affiliation(s)
- Phanramphoei N Frantz
- Viral Genomics and Vaccination Unit, Department of Virology, Institut Pasteur, CNRS UMR 3965, Paris, France; Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Samaporn Teeravechyan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Frédéric Tangy
- Viral Genomics and Vaccination Unit, Department of Virology, Institut Pasteur, CNRS UMR 3965, Paris, France.
| |
Collapse
|
41
|
Abstract
The classic development of vaccines is lengthy, tedious, and may not necessarily be successful as demonstrated by the case of HIV. This is especially a problem for emerging pathogens that are newly introduced into the human population and carry the inherent risk of pandemic spread in a naïve population. For such situations, a considerable number of different platform technologies are under development. These are also under development for pathogens, where directly derived vaccines are regarded as too complicated or even dangerous due to the induction of inefficient or unwanted immune responses causing considerable side-effects as for dengue virus. Among platform technologies are plasmid-based DNA vaccines, RNA replicons, single-round infectious vector particles, or replicating vaccine-based vectors encoding (a) critical antigen(s) of the target pathogens. Among the latter, recombinant measles viruses derived from vaccine strains have been tested. Measles vaccines are among the most effective and safest life-attenuated vaccines known. Therefore, the development of Schwarz-, Moraten-, or AIK-C-strain derived recombinant vaccines against a wide range of mostly viral, but also bacterial pathogens was quite straightforward. These vaccines generally induce powerful humoral and cellular immune responses in appropriate animal models, i.e., transgenic mice or non-human primates. Also in the recent first clinical phase I trial, the results have been quite encouraging. The trial indicated the expected safety and efficacy also in human patients, interestingly independent from the level of prevalent anti-measles immunity before the trial. Thereby, recombinant measles vaccines expressing additional antigens are a promising platform for future vaccines.
Collapse
Affiliation(s)
- Michael D Mühlebach
- Product Testing of IVMPs, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, 63225, Langen, Germany.
| |
Collapse
|
42
|
Abstract
In 2013, a major chikungunya virus (CHIKV) epidemic reached the Americas. In the past 2 years, >1.7 million people have been infected. In light of the current epidemic, with millions of people in North and South America at risk, efforts to rapidly develop effective vaccines have increased. Here, we focus on CHIKV vaccines that use viral-vector technologies. This group of vaccine candidates shares an ability to potently induce humoral and cellular immune responses by use of highly attenuated and safe vaccine backbones. So far, well-described vectors such as modified vaccinia virus Ankara, complex adenovirus, vesicular stomatitis virus, alphavirus-based chimeras, and measles vaccine Schwarz strain (MV/Schw) have been described as potential vaccines. We summarize here the recent data on these experimental vaccines, with a focus on the preclinical and clinical activities on the MV/Schw-based candidate, which is the first CHIKV-vectored vaccine that has completed a clinical trial.
Collapse
Affiliation(s)
| | - Frédéric Tangy
- Viral Genomics and Vaccination Unit, CNRS UMR 3569, Institut Pasteur, Paris, France
| |
Collapse
|
43
|
Abstract
This chapter describes the development of recombinant measles virus (MV)-based vaccines starting from plasmid DNA. Live-attenuated measles vaccines are very efficient and safe. Since the availability of a reverse genetic system to manipulate MV genomes and to generate respective recombinant viruses, a considerable number of recombinant viruses has been generated that present antigens of foreign pathogens during MV replication. Thereby, robust humoral and cellular immune responses can be induced, which have shown protective capacity in a substantial number of experiments.For this purpose, the foreign antigen-encoding genes are cloned into additional transcription units of plasmid based full-length MV vaccine strain genomes, which in turn are used to rescue recombinant MV by providing both full-length viral RNA genomes respective anti-genomes together with all protein components of the viral ribonucleoprotein complex after transient transfection of the so-called rescue cells. Infectious centers form among these transfected cells, which allow clonal isolation of single recombinant viruses that are subsequently amplified, characterized in vitro, and then evaluated for their immunogenicity in appropriate preclinical animal models.
Collapse
Affiliation(s)
- Maureen C. Ferran
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York USA
| | - Gary R. Skuse
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York USA
| |
Collapse
|
44
|
Lauer KB, Borrow R, Blanchard TJ. Multivalent and Multipathogen Viral Vector Vaccines. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:e00298-16. [PMID: 27535837 PMCID: PMC5216423 DOI: 10.1128/cvi.00298-16] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The presentation and delivery of antigens are crucial for inducing immunity and, desirably, lifelong protection. Recombinant viral vectors-proven safe and successful in veterinary vaccine applications-are ideal shuttles to deliver foreign proteins to induce an immune response with protective antibody levels by mimicking natural infection. Some examples of viral vectors are adenoviruses, measles virus, or poxviruses. The required attributes to qualify as a vaccine vector are as follows: stable insertion of coding sequences into the genome, induction of a protective immune response, a proven safety record, and the potential for large-scale production. The need to develop new vaccines for infectious diseases, increase vaccine accessibility, reduce health costs, and simplify overloaded immunization schedules has driven the idea to combine antigens from the same or various pathogens. To protect effectively, some vaccines require multiple antigens of one pathogen or different pathogen serotypes/serogroups in combination (multivalent or polyvalent vaccines). Future multivalent vaccine candidates are likely to be required for complex diseases like malaria and HIV. Other novel strategies propose an antigen combination of different pathogens to protect against several diseases at once (multidisease or multipathogen vaccines).
Collapse
Affiliation(s)
- Katharina B Lauer
- University of Manchester, Institute of Inflammation and Repair, Manchester, United Kingdom
- University of Cambridge, Department of Pathology, Cambridge, United Kingdom
| | - Ray Borrow
- University of Manchester, Institute of Inflammation and Repair, Manchester, United Kingdom
- Vaccine Evaluation Unit, Public Health England, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Thomas J Blanchard
- University of Manchester, Institute of Inflammation and Repair, Manchester, United Kingdom
- Consultant in Infectious Diseases and Tropical Medicine, Royal Liverpool Hospital, Liverpool, United Kingdom
| |
Collapse
|
45
|
Enjuanes L, Zuñiga S, Castaño-Rodriguez C, Gutierrez-Alvarez J, Canton J, Sola I. Molecular Basis of Coronavirus Virulence and Vaccine Development. Adv Virus Res 2016; 96:245-286. [PMID: 27712626 PMCID: PMC7112271 DOI: 10.1016/bs.aivir.2016.08.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Virus vaccines have to be immunogenic, sufficiently stable, safe, and suitable to induce long-lasting immunity. To meet these requirements, vaccine studies need to provide a comprehensive understanding of (i) the protective roles of antiviral B and T-cell-mediated immune responses, (ii) the complexity and plasticity of major viral antigens, and (iii) virus molecular biology and pathogenesis. There are many types of vaccines including subunit vaccines, whole-inactivated virus, vectored, and live-attenuated virus vaccines, each of which featuring specific advantages and limitations. While nonliving virus vaccines have clear advantages in being safe and stable, they may cause side effects and be less efficacious compared to live-attenuated virus vaccines. In most cases, the latter induce long-lasting immunity but they may require special safety measures to prevent reversion to highly virulent viruses following vaccination. The chapter summarizes the recent progress in the development of coronavirus (CoV) vaccines, focusing on two zoonotic CoVs, the severe acute respiratory syndrome CoV (SARS-CoV), and the Middle East respiratory syndrome CoV, both of which cause deadly disease and epidemics in humans. The development of attenuated virus vaccines to combat infections caused by highly pathogenic CoVs was largely based on the identification and characterization of viral virulence proteins that, for example, interfere with the innate and adaptive immune response or are involved in interactions with specific cell types, such as macrophages, dendritic and epithelial cells, and T lymphocytes, thereby modulating antiviral host responses and viral pathogenesis and potentially resulting in deleterious side effects following vaccination.
Collapse
Affiliation(s)
- L Enjuanes
- National Center of Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain.
| | - S Zuñiga
- National Center of Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - C Castaño-Rodriguez
- National Center of Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - J Gutierrez-Alvarez
- National Center of Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - J Canton
- National Center of Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - I Sola
- National Center of Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
46
|
Gupta G, Giannino V, Rishi N, Glueck R. Immunogenicity of next-generation HPV vaccines in non-human primates: Measles-vectored HPV vaccine versus Pichia pastoris recombinant protein vaccine. Vaccine 2016; 34:4724-4731. [PMID: 27523740 PMCID: PMC7126718 DOI: 10.1016/j.vaccine.2016.07.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 07/16/2016] [Accepted: 07/27/2016] [Indexed: 01/12/2023]
Abstract
Human papillomavirus (HPV) infection is the most common sexually transmitted disease worldwide. HPVs are oncogenic small double-stranded DNA viruses that are the primary causal agent of cervical cancer and other types of cancers, including in the anus, oropharynx, vagina, vulva, and penis. Prophylactic vaccination against HPV is an attractive strategy for preventing cervical cancer and some other types of cancers. However, there are few safe and effective vaccines against HPV infections. Current first-generation commercial HPV vaccines are expensive to produce and deliver. The goal of this study was to develop an alternate potent HPV recombinant L1-based vaccines by producing HPV virus-like particles into a vaccine that is currently used worldwide. Live attenuated measles virus (MV) vaccines have a well-established safety and efficacy record, and recombinant MV (rMV) produced by reverse genetics may be useful for generating candidate HPV vaccines to meet the needs of the developing world. We studied in non-human primate rMV-vectored HPV vaccine in parallel with a classical alum adjuvant recombinant HPV16L1 and 18L1 protein vaccine produced in Pichia pastoris. A combined prime-boost approach using both vaccines was evaluated, as well as immune interference due to pre-existing immunity against the MV. The humoral immune response induced by the MV, Pichia-expressed vaccine, and their combination as priming and boosting approaches was found to elicit HPV16L1 and 18L1 specific total IgG and neutralizing antibody titres. Pre-existing antibodies against measles did not prevent the immune response against HPV16L1 and 18L1.
Collapse
Affiliation(s)
- Gaurav Gupta
- Department of Virology, Vaccine Technology Centre, Cadila Healthcare Ltd., Ahmedabad, India; Etna Biotech S.r.l., Stradale Vincenzo Lancia 57, 95121 Catania, Italy; Amity Institute of Virology and Immunology, Amity University, Noida, India.
| | - Viviana Giannino
- Etna Biotech S.r.l., Stradale Vincenzo Lancia 57, 95121 Catania, Italy.
| | - Narayan Rishi
- Amity Institute of Virology and Immunology, Amity University, Noida, India
| | - Reinhard Glueck
- Department of Virology, Vaccine Technology Centre, Cadila Healthcare Ltd., Ahmedabad, India; Etna Biotech S.r.l., Stradale Vincenzo Lancia 57, 95121 Catania, Italy
| |
Collapse
|
47
|
Middle East respiratory syndrome vaccines. Int J Infect Dis 2016; 47:23-8. [PMID: 27062985 PMCID: PMC4969153 DOI: 10.1016/j.ijid.2016.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 03/31/2016] [Accepted: 04/03/2016] [Indexed: 12/22/2022] Open
Abstract
Middle East respiratory syndrome (MERS), caused by a novel coronavirus, is a highly lethal respiratory disease. No vaccines or antiviral therapies are available. Camels are widely infected and may be good targets for vaccination. MERS monoclonal antibodies and human convalescent sera may be useful for prophylaxis and treatment. Active immunization strategies, based on the severe acute respiratory syndrome (SARS) experience, are under development.
The Middle East respiratory syndrome coronavirus (MERS-CoV) has infected over 1600 individuals with nearly 600 deaths since it was first identified in human populations in 2012. No antiviral therapies or vaccines are available for its treatment or prophylaxis. Approaches to the development of MERS vaccines are discussed herein, including a summary of previous efforts to develop vaccines useful against human and non-human coronaviruses. A striking feature of MERS is the important role that camels have in transmission. Camel vaccination may be a novel approach to preventing human infection.
Collapse
|
48
|
Baldo A, Galanis E, Tangy F, Herman P. Biosafety considerations for attenuated measles virus vectors used in virotherapy and vaccination. Hum Vaccin Immunother 2015; 12:1102-16. [PMID: 26631840 PMCID: PMC4963060 DOI: 10.1080/21645515.2015.1122146] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Attenuated measles virus (MV) is one of the most effective and safe vaccines available, making it attractive candidate vector to prevent infectious diseases. Attenuated MV have acquired the ability to use the complement regulator CD46 as a major receptor to mediate virus entry and intercellular fusion. Therefore, attenuated MV strains preferentially infect and destroy a wide variety of cancer cells making them also attractive oncolytic vectors. The use of recombinant MV vector has to comply with various regulatory requirements, particularly relating to the assessment of potential risks for human health and the environment. The present article highlights the main characteristics of MV and recombinant MV vectors used for vaccination and virotherapy and discusses these features from a biosafety point of view.
Collapse
Affiliation(s)
- Aline Baldo
- a Scientific Institute of Public Health (WIV-ISP), Biosafety and Biotechnology Unit , Brussels , Belgium
| | - Evanthia Galanis
- b Division of Medical Oncology , Mayo Clinic , Rochester , MN , USA
| | - Frédéric Tangy
- c Institut Pasteur, Viral Genomics and Vaccination Unit, CNRS UMR 3569 , Paris , France
| | - Philippe Herman
- a Scientific Institute of Public Health (WIV-ISP), Biosafety and Biotechnology Unit , Brussels , Belgium
| |
Collapse
|
49
|
Wang X, Chen J, Shi D, Shi H, Zhang X, Yuan J, Jiang S, Feng L. Immunogenicity and antigenic relationships among spike proteins of porcine epidemic diarrhea virus subtypes G1 and G2. Arch Virol 2015; 161:537-47. [PMID: 26611909 PMCID: PMC7087089 DOI: 10.1007/s00705-015-2694-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/16/2015] [Indexed: 11/01/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a coronavirus that infects cells lining the small intestine of swine, resulting in vomiting, diarrhea, and dehydration. The amino acid sequence of the spike (S) protein, which is the principal target recognized by host immune cells, has multiple mutations that distinguish the two PEDV genotypes, G1 and G2. To determine whether these mutations lead to changes in antigenicity, as suggested by the failure of PEDV vaccines in China, we first optimized the codons of typical S genes of the CV777 vaccine strain (G1 subtype) and LNCT2 strain (G2 subtype) and expressed the recombinant full-length sequence of the S protein in a eukaryotic expression system. The IgG antibody levels of serum from mice immunized with purified S protein were markedly high. Antigenicity was compared by detection of polyclonal antibodies (PAbs) against the virus and S protein using an enzyme-linked immunosorbent assay (ELISA), an indirect immunofluorescence assay (IFA), and a serum cross-neutralization (SN) assay. Reactivity with the PAbs revealed significant cross-reactivity between the two PEDV subtypes, although there was a twofold difference in the antigenic responses based on PAb titers in the ELISA and IFA. Consistent with the variation in the S gene sequences, the SN titer suggested differences in the neutralization activity of the S protein between the two subtypes, which could explain the antigenic variation between the PEDV subtypes G1 and G2.
Collapse
Affiliation(s)
- Xiaobo Wang
- Division of Swine Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, People's Republic of China
| | - Jianfei Chen
- Division of Swine Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, People's Republic of China
| | - Da Shi
- Division of Swine Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, People's Republic of China
| | - Hongyan Shi
- Division of Swine Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, People's Republic of China
| | - Xin Zhang
- Division of Swine Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, People's Republic of China
| | - Jing Yuan
- Division of Swine Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, People's Republic of China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Public Health Clinical Center and Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Li Feng
- Division of Swine Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, People's Republic of China.
| |
Collapse
|
50
|
Nakayama T, Sawada A, Yamaji Y, Ito T. Recombinant measles AIK-C vaccine strain expressing heterologous virus antigens. Vaccine 2015; 34:292-295. [PMID: 26562316 PMCID: PMC7115616 DOI: 10.1016/j.vaccine.2015.10.127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/14/2014] [Accepted: 12/18/2014] [Indexed: 12/14/2022]
Abstract
Further attenuated measles vaccines were developed more than 50 years ago and have been used throughout the world. Recombinant measles vaccine candidates have been developed and express several heterologous virus protective antigens. Immunogenicity and protective actions were confirmed using experimental animals: transgenic mice, cotton rats, and primates. The recent development of measles vaccine-based vectored vaccine candidates has been reviewed and some information on recombinant measles vaccines expressing respiratory syncytial virus proteins has been shown and discussed.
Collapse
Affiliation(s)
- Tetsuo Nakayama
- Kitasato Institute for Life Sciences, Laboratory of Viral Infection I, Minato-ku, Tokyo 108-8641, Japan.
| | - Akihito Sawada
- Kitasato Institute for Life Sciences, Laboratory of Viral Infection I, Minato-ku, Tokyo 108-8641, Japan
| | - Yoshiaki Yamaji
- Kitasato Institute for Life Sciences, Laboratory of Viral Infection I, Minato-ku, Tokyo 108-8641, Japan
| | - Takashi Ito
- Kitasato Institute for Life Sciences, Laboratory of Viral Infection I, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|