1
|
Grau K, Lillie-Jaschniski K, Graaf-Rau A, Harder T, Eddicks M, Zöls S, Zablotski Y, Ritzmann M, Stadler J. Effect of stabilizers on the detection of swine influenza A virus (swIAV) in spiked oral fluids over time. Porcine Health Manag 2024; 10:49. [PMID: 39529191 PMCID: PMC11552184 DOI: 10.1186/s40813-024-00386-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/16/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Aggregated samples such as oral fluids (OFs) display an animal friendly and time and cost-efficient sample type for swine Influenza A virus (swIAV) monitoring. However, further molecular and biological characterization of swIAV is of particular significance. The reportedly inferior suitability of aggregated samples for subtyping of swIAV presents a major drawback compared to nasal swabs, still considered the most appropriate sample type for this purpose (Garrido-Mantilla et al. BMC Vet Res 15(1):61, 2019). In addition, the viral load in the original sample, storage conditions and characteristics of different swIAV strains might further compromise the eligibility of aggregated samples for molecular detection and subtyping. Therefore, the present study aimed to evaluate the suitability of stabilizing media to minimize the degradation of viral RNA and thus increase the detection and subtyping rate of swIAV by RT-qPCR in spiked OFs under different conditions (virus strain, storage temperature and viral load in the original sample) over a time span of 14 days. RESULTS The use of stabilizing media in spiked OFs resulted in a significant higher probability to detect swIAV RNA compared to OFs without stabilizers (OR = 46.1, p < 0.001). In addition, swIAV degradation over time was significantly reduced in samples suspended with stabilizer (OR = 5.80, p < 0.001), in samples stored at 4 °C (OR = 2.53, p < 0.001) and in samples spiked with the avian derived H1N2 subtype (OR = 2.26, p < 0.01). No significant differences in swIAV RNA detection and degradation of swIAV RNA in spiked OFs over time were observed between the three different stabilizing media. CONCLUSION Addition of stabilizers and storage of samples under cooled conditions significantly improved detection and subtyping of swIAV in spiked OFs.
Collapse
Affiliation(s)
- K Grau
- Clinic for Swine at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | | | - A Graaf-Rau
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald, Insel-Riems, Germany
- Helmholtz Institute for One Health, Greifswald, Germany
| | - T Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald, Insel-Riems, Germany
| | - M Eddicks
- Clinic for Swine at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - S Zöls
- Clinic for Swine at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - Y Zablotski
- Clinic for Swine at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - M Ritzmann
- Clinic for Swine at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - J Stadler
- Clinic for Swine at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Oberschleißheim, Germany.
| |
Collapse
|
2
|
Graaf-Rau A, Schmies K, Breithaupt A, Ciminski K, Zimmer G, Summerfield A, Sehl-Ewert J, Lillie-Jaschniski K, Helmer C, Bielenberg W, Grosse Beilage E, Schwemmle M, Beer M, Harder T. Reassortment incompetent live attenuated and replicon influenza vaccines provide improved protection against influenza in piglets. NPJ Vaccines 2024; 9:127. [PMID: 39003272 PMCID: PMC11246437 DOI: 10.1038/s41541-024-00916-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024] Open
Abstract
Swine influenza A viruses (swIAV) cause an economically important respiratory disease in modern pig production. Continuous virus transmission and antigenic drift are difficult to control in enzootically infected pig herds. Here, antibody-positive piglets from a herd enzootically infected with swIAV H1N2 (clade 1 A.3.3.2) were immunized using a homologous prime-boost vaccination strategy with novel live attenuated influenza virus (LAIV) based on a reassortment-incompetent bat influenza-swIAV chimera or a vesicular stomatitis virus-based replicon vaccine. Challenge infection of vaccinated piglets by exposure to H1N2 swIAV-infected unvaccinated seeder pigs showed that both LAIV and replicon vaccine markedly reduced virus replication in the upper and lower respiratory tract, respectively, compared to piglets immunized with commercial heterologous or autologous adjuvanted whole-inactivated virus vaccines. Our novel vaccines may aid in interrupting continuous IAV transmission chains in large enzootically infected pig herds, improve the health status of the animals, and reduce the risk of zoonotic swIAV transmission.
Collapse
Affiliation(s)
- Annika Graaf-Rau
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Greifswald, Germany.
| | - Kathrin Schmies
- Field Station for Epidemiology, University of Veterinary Medicine Hannover, Foundation, Bakum, Germany
| | - Angele Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler- Institut, Greifswald, Insel Riems, Germany
| | - Kevin Ciminski
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gert Zimmer
- Institute of Virology and Immunology, Bern & Mittelhaeusern, Switzerland, and Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern & Mittelhaeusern, Switzerland, and Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Julia Sehl-Ewert
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler- Institut, Greifswald, Insel Riems, Germany
| | | | - Carina Helmer
- SAN Group Biotech Germany GmbH, Hoeltinghausen, Germany
| | | | - Elisabeth Grosse Beilage
- Field Station for Epidemiology, University of Veterinary Medicine Hannover, Foundation, Bakum, Germany
| | - Martin Schwemmle
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Greifswald, Germany
| | - Timm Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Greifswald, Germany
| |
Collapse
|
3
|
Studniski M, Stumvoll K, Kromm M, Ssematimba A, Marusak R, Xing Z, Halvorson D, Culhane M, Cardona C. Vaccination of Poultry Against Influenza. Avian Dis 2024; 67:402-409. [PMID: 38300659 DOI: 10.1637/aviandiseases-d-23-99995] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 02/02/2024]
Abstract
The complexity of influenza A virus (IAV) infections in avian hosts leads to equally complex scenarios for the vaccination of poultry. Vaccination against avian influenza strains can be used to prevent infections from sources with a single strain of IAV. It has been used as a part of outbreak control strategies as well as a way to maintain production for both low and high pathogenicity outbreaks. Unlike other viral pathogens of birds, avian influenza vaccination when used against highly pathogenic avian influenza virus, is tied to international trade and thus is not freely available for use without specific permission.
Collapse
Affiliation(s)
| | | | | | - Amos Ssematimba
- Department of Mathematics, Gulu University, Gulu, Uganda
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Rosemary Marusak
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Zheng Xing
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Dave Halvorson
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Marie Culhane
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Carol Cardona
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108,
| |
Collapse
|
4
|
Keay S, Poljak Z, Alberts F, O’Connor A, Friendship R, O’Sullivan TL, Sargeant JM. Does Vaccine-Induced Maternally-Derived Immunity Protect Swine Offspring against Influenza a Viruses? A Systematic Review and Meta-Analysis of Challenge Trials from 1990 to May 2021. Animals (Basel) 2023; 13:3085. [PMID: 37835692 PMCID: PMC10571953 DOI: 10.3390/ani13193085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
It is unclear if piglets benefit from vaccination of sows against influenza. For the first time, methods of evidence-based medicine were applied to answer the question: "Does vaccine-induced maternally-derived immunity (MDI) protect swine offspring against influenza A viruses?". Challenge trials were reviewed that were published from 1990 to April 2021 and measured at least one of six outcomes in MDI-positive versus MDI-negative offspring (hemagglutination inhibition (HI) titers, virus titers, time to begin and time to stop shedding, risk of infection, average daily gain (ADG), and coughing) (n = 15). Screening and extraction of study characteristics was conducted in duplicate by two reviewers, with data extraction and assessment for risk of bias performed by one. Homology was defined by the antigenic match of vaccine and challenge virus hemagglutinin epitopes. Results: Homologous, but not heterologous MDI, reduced virus titers in piglets. There was no difference, calculated as relative risks (RR), in infection incidence risk over the entire study period; however, infection hazard (instantaneous risk) was decreased in pigs with MDI (log HR = -0.64, 95% CI: -1.13, -0.15). Overall, pigs with MDI took about a ½ day longer to begin shedding virus post-challenge (MD = 0.51, 95% CI: 0.03, 0.99) but the hazard of infected pigs ceasing to shed was not different (log HR = 0.32, 95% CI: -0.29, 0.93). HI titers were synthesized qualitatively and although data on ADG and coughing was extracted, details were insufficient for conducting meta-analyses. Conclusion: Homology of vaccine strains with challenge viruses is an important consideration when assessing vaccine effectiveness. Herd viral dynamics are complex and may include concurrent or sequential exposures in the field. The practical significance of reduced weaned pig virus titers is, therefore, not known and evidence from challenge trials is insufficient to make inferences on the effects of MDI on incidence risk, time to begin or to cease shedding virus, coughing, and ADG. The applicability of evidence from single-strain challenge trials to field practices is limited. Despite the synthesis of six outcomes, challenge trial evidence does not support or refute vaccination of sows against influenza to protect piglets. Additional research is needed; controlled trials with multi-strain concurrent or sequential heterologous challenges have not been conducted, and sequential homologous exposure trials were rare. Consensus is also warranted on (1) the selection of core outcomes, (2) the sizing of trial populations to be reflective of field populations, (3) the reporting of antigenic characterization of vaccines, challenge viruses, and sow exposure history, and (4) on the collection of non-aggregated individual pig data.
Collapse
Affiliation(s)
- Sheila Keay
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (Z.P.); (F.A.); (R.F.); (T.L.O.); (J.M.S.)
| | - Zvonimir Poljak
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (Z.P.); (F.A.); (R.F.); (T.L.O.); (J.M.S.)
| | - Famke Alberts
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (Z.P.); (F.A.); (R.F.); (T.L.O.); (J.M.S.)
| | - Annette O’Connor
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA;
| | - Robert Friendship
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (Z.P.); (F.A.); (R.F.); (T.L.O.); (J.M.S.)
| | - Terri L. O’Sullivan
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (Z.P.); (F.A.); (R.F.); (T.L.O.); (J.M.S.)
| | - Jan M. Sargeant
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (Z.P.); (F.A.); (R.F.); (T.L.O.); (J.M.S.)
- Centre for Public Health and Zoonoses, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
5
|
Setta A, Yehia N, Shakak AO, Al-Quwaie DA, Al- Otaibi AM, El-Saadony MT, El-Tarabily KA, Salem H. Molecular detection of highly pathogenic avian influenza H5N8 in commercial broiler chicken farms from 2019 to 2022. Poult Sci 2023; 102:102639. [PMID: 37104901 PMCID: PMC10165147 DOI: 10.1016/j.psj.2023.102639] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/14/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Highly pathogenic avian influenza (HPAI) is a serious viral infection that causes massive economic losses in poultry. The current study investigated the HPAI virus prevalence in commercial broiler chicken flocks from 2019 to 2022. Organ samples, including trachea, cecal tonsils, spleen, brain, as well as tracheal and cloacal swabs, were harvested from 111 problematic broiler chicken flocks that suffered from variable mortalities accompanied with respiratory signs (103 H5-vaccinated and 8 nonvaccinated flocks) in Egypt during the observation duration. Molecular tools were used to analyze the samples, including real-time reverse transcription-polymerase chain reaction (rRT-PCR) and sequence analysis of some PCR positive strains. The results indicated that 24 flocks were positive for HPAI H5N8, representing 21.6%, with 22.3% (23/103) prevalence and 12.5% (1/8) detection in vaccinated and nonvaccinated flocks, respectively, and they were almost detected in the autumn and winter seasons. Phylogenetic evaluation of the hemagglutinin (HA) gene showed that the 6 Egyptian strains were clustered in clade 2.3.4.4b and allocated into 2 groups (I and II). The samples recovered in 2019 were clustered in new subgroup A, and samples recovered in 2020 to 2022 were clustered in new subgroup B with 10 nucleotide mutations (R72S, A83D, T140A). In conclusion, HPAI H5N8 is a serious threat even in vaccinated birds; to control such problems, periodic molecular monitoring with vaccine efficacy evaluation and the use of preventive strategies are recommended.
Collapse
|
6
|
Niqueux É, Flodrops M, Allée C, Lebras MO, Pierre I, Louboutin K, Guillemoto C, Le Prioux A, Le Bouquin-Leneveu S, Keïta A, Amelot M, Martenot C, Massin P, Cherbonnel-Pansart M, Briand FX, Schmitz A, Cazaban C, Dauphin G, Delquigny T, Lemière S, Watier JM, Mogler M, Tarpey I, Grasland B, Eterradossi N. Evaluation of three hemagglutinin-based vaccines for the experimental control of a panzootic clade 2.3.4.4b A(H5N8) high pathogenicity avian influenza virus in mule ducks. Vaccine 2023; 41:145-158. [PMID: 36411134 DOI: 10.1016/j.vaccine.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/17/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022]
Abstract
In France during winter 2016-2017, 487 outbreaks of clade 2.3.4.4b H5N8 subtype high pathogenicity (HP) avian influenza A virus (AIV) infections were detected in poultry and captive birds. During this epizootic, HPAIV A/decoy duck/France/161105a/2016 (H5N8) was isolated and characterized in an experimental infection transmission model in conventional mule ducks. To investigate options to possibly protect such ducks against this HPAIV, three vaccines were evaluated in controlled conditions. The first experimental vaccine was derived from the hemagglutinin gene of another clade 2.3.4.4b A(H5N8) HPAIV. It was injected at three weeks of age, either alone (Vac1) or after a primer injection at day-old (Vac1 + boost). The second vaccine (Vac2) was a commercial bivalent adjuvanted vaccine containing an expressed hemagglutinin modified from a clade 2.3.2 A(H5N1) HPAIV. Vac2 was administered as a single injection at two weeks of age. The third experimental vaccine (Vac3) also incorporated a homologous 2.3.4.4b H5 HA gene and was administered as a single injection at three weeks of age. Ducks were challenged with HPAIV A/decoy duck/France/161105a/2016 (H5N8) at six weeks of age. Post-challenge virus excretion was monitored in vaccinated and control birds every 2-3 days for two weeks using real-time reverse-transcription polymerase chain reaction and serological analyses (haemagglutination inhibition test against H5N8, H5 ELISA and AIV ELISA) were performed. Vac1 abolished oropharyngeal and cloacal shedding to almost undetectable levels, whereas Vac3 abolished cloacal shedding only (while partially reducing respiratory shedding) and Vac2 only partly reduced the respiratory and intestinal excretion of the challenge virus. These results provided relevant insights in the immunogenicity of recombinant H5 vaccines in mule ducks, a rarely investigated hybrid between Pekin and Muscovy duck species that has played a critical role in the recent H5 HPAI epizootics in France.
Collapse
Affiliation(s)
- Éric Niqueux
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Marion Flodrops
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Chantal Allée
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Marie-Odile Lebras
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Isabelle Pierre
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Katell Louboutin
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Carole Guillemoto
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Aurélie Le Prioux
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Sophie Le Bouquin-Leneveu
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Epidemiology Health and Welfare Unit, BP53, 22440 Ploufragan, France
| | - Alassane Keïta
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian Experimentation and Breeding Service, BP53, 22440 Ploufragan, France
| | - Michel Amelot
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian Experimentation and Breeding Service, BP53, 22440 Ploufragan, France
| | - Claire Martenot
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Pascale Massin
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Martine Cherbonnel-Pansart
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - François-Xavier Briand
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | - Audrey Schmitz
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| | | | - Gwenaëlle Dauphin
- Ceva Santé Animale, 10 Avenue de la Ballastière, 33500 Libourne, France
| | - Thomas Delquigny
- Boehringer Ingelheim Animal Health, 29 avenue Tony Garnier, 69007 Lyon, France
| | - Stéphane Lemière
- Boehringer Ingelheim Animal Health, 29 avenue Tony Garnier, 69007 Lyon, France
| | - Jean-Marie Watier
- MSD Santé Animale, 7 rue Olivier de Serres, BP 17144, 49071 Beaucouzé Cedex, France
| | - Mark Mogler
- Merck Animal Health, Ames, IA 50010, United States of America
| | - Ian Tarpey
- MSD Animal Health, Walton Manor, Milton Keynes MK7 7AJ, United Kingdom
| | - Béatrice Grasland
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France.
| | - Nicolas Eterradossi
- Anses (French Agency for Food, Environmental and Occupational Health & Safety), Ploufragan-Plouzané-Niort Laboratory, Avian and Rabbit Virology Immunology and Parasitology Unit, National Reference Laboratory for Avian Influenza, BP53, 22440 Ploufragan, France
| |
Collapse
|
7
|
Graaf A, Petric PP, Sehl-Ewert J, Henritzi D, Breithaupt A, King J, Pohlmann A, Deutskens F, Beer M, Schwemmle M, Harder T. Cold-passaged isolates and bat-swine influenza a chimeric viruses as modified live-attenuated vaccines against influenza a viruses in pigs. Vaccine 2022; 40:6255-6270. [PMID: 36137904 DOI: 10.1016/j.vaccine.2022.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
Swine influenza A virus (swIAV) infections in pig populations cause considerable morbidity and economic losses. Frequent reverse zoonotic incursions of human IAV boost reassortment opportunities with authentic porcine and avian-like IAV in swine herds potentially enhancing zoonotic and even pre-pandemic potential. Vaccination using adjuvanted inactivated full virus vaccines is frequently used in attempting control of swIAV infections. Accelerated antigenic drift of swIAV in large swine holdings and interference of maternal antibodies with vaccine in piglets can compromise these efforts. Potentially more efficacious modified live-attenuated vaccines (MLVs) bear the risk of reversion of MLV to virulence. Here we evaluated new MLV candidates based on cold-passaged swIAV or on reassortment-incompetent bat-IAV-swIAV chimeric viruses. Serial cold-passaging of various swIAV subtypes did not yield unambiguously temperature-sensitive mutants although safety studies in mice and pigs suggested some degree of attenuation. Chimeric bat-swIAV expressing the hemagglutinin and neuraminidase of an avian-like H1N1, in contrast, proved to be safe in mice and pigs, and a single nasal inoculation induced protective immunity against homologous challenge in pigs. Reassortant-incompetent chimeric bat-swIAV vaccines could aid in reducing the amount of swIAV circulating in pig populations, thereby increasing animal welfare, limiting economic losses and lowering the risk of zoonotic swIAV transmission.
Collapse
Affiliation(s)
- Annika Graaf
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany.
| | - Philipp P Petric
- Institute of Virology, Medical Center, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Julia Sehl-Ewert
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Dinah Henritzi
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Angele Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Jacqueline King
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Anne Pohlmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | | | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Martin Schwemmle
- Institute of Virology, Medical Center, University of Freiburg, 79104 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Timm Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| |
Collapse
|
8
|
Alqazlan N, Astill J, Raj S, Sharif S. Strategies for enhancing immunity against avian influenza virus in chickens: A review. Avian Pathol 2022; 51:211-235. [PMID: 35297706 DOI: 10.1080/03079457.2022.2054309] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Poultry infection with avian influenza viruses (AIV) is a continuous source of concern for poultry production and human health. Uncontrolled infection and transmission of AIV in poultry increases the potential for viral mutation and reassortment, possibly resulting in the emergence of zoonotic viruses. To this end, implementing strategies to disrupt the transmission of AIVs in poultry, including a wide array of traditional and novel methods, is much needed. Vaccination of poultry is a targeted approach to reduce clinical signs and shedding in infected birds. Strategies aimed at enhancing the effectiveness of AIV vaccines are multi-pronged and include methods directed towards eliciting immune responses in poultry. Strategies include producing vaccines of greater immunogenicity via vaccine type and adjuvant application and increasing bird responsiveness to vaccines by modification of the gastrointestinal tract (GIT) microbiome and dietary interventions. This review provides an in-depth discussion of recent findings surrounding novel AIV vaccines for poultry, including reverse genetics vaccines, vectors, protein vaccines and virus like particles, highlighting their experimental efficacy among other factors such as safety and potential for use in the field. In addition to the type of vaccine employed, vaccine adjuvants also provide an effective way to enhance AIV vaccine efficacy, therefore, research on different types of vaccine adjuvants and vaccine adjuvant delivery strategies is discussed. Finally, the poultry gastrointestinal microbiome is emerging as an important factor in the effectiveness of prophylactic treatments. In this regard, current findings on the effects of the chicken GIT microbiome on AIV vaccine efficacy are summarized here.
Collapse
Affiliation(s)
- Nadiyah Alqazlan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jake Astill
- Artemis Technologies Inc., Guelph, ON, N1L 1E3, Canada
| | - Sugandha Raj
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| |
Collapse
|
9
|
Sączyńska V, Romanik-Chruścielewska A, Florys-Jankowska K, Cecuda-Adamczewska V, Kęsik-Brodacka M. Chitosan-based formulation of hemagglutinin antigens for oculo-nasal booster vaccination of chickens against influenza viruses. Vet Immunol Immunopathol 2022; 247:110406. [DOI: 10.1016/j.vetimm.2022.110406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 11/30/2022]
|
10
|
A new self-attenuated therapeutic influenza vaccine that uses host cell-restricted attenuation by artificial microRNAs. Int J Pharm 2022; 612:121325. [PMID: 34883209 PMCID: PMC8871448 DOI: 10.1016/j.ijpharm.2021.121325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 01/27/2023]
Abstract
New strategies are urgently needed for developing vaccines and/or anti-viral drugs against influenza viruses, because antigenic shift and drift inevitably occurs in circulating strains each year, and new strains resistant to anti-viral drugs have recently emerged. In our study, we designed and incorporated artificial microRNAs (amiRNAs) into the NA segment of rescued influenza viruses to separately target two host genes, Cdc2-like kinase 1 (CLK1) and SON DNA binding protein (SON), which were found to play an essential role in virus replication. Mouse epithelial fibroblast (MEF) or human lung carcinoma A549 cells infected with engineered influenza PR8 viruses expressing amiR-30CLK1 (PR8-amiR-30CLK1) or amiR-93SON (PR8-amiR-93SON) had reduced expression of host proteins CLK1 and SON, respectively. All engineered influenza viruses functioned as attenuated vaccines, induced significantly higher antibody responses, and provided greater protective efficacy. In addition, they were found to be safe, based on the mouse weight changes and clinical signs observed. In contrast to the engineered viruses targeting SON, mice treated with engineered viruses targeting CLK1 recovered from weight loss and survived lethal infection by 6 h after lethal-dose PR8 infection, suggesting that our PR8-amiR-30CLK1 self-attenuated influenza virus (SAIV) could be used as a new therapeutic influenza vaccine.
Collapse
|
11
|
Salvesen HA, Whitelaw CBA. Current and prospective control strategies of influenza A virus in swine. Porcine Health Manag 2021; 7:23. [PMID: 33648602 PMCID: PMC7917534 DOI: 10.1186/s40813-021-00196-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/21/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Influenza A Viruses (IAV) are endemic pathogens of significant concern in humans and multiple keystone livestock species. Widespread morbidity in swine herds negatively impacts animal welfare standards and economic performance whilst human IAV pandemics have emerged from pigs on multiple occasions. To combat the rising prevalence of swine IAV there must be effective control strategies available. MAIN BODY The most basic form of IAV control on swine farms is through good animal husbandry practices and high animal welfare standards. To control inter-herd transmission, biosecurity considerations such as quarantining of pigs and implementing robust health and safety systems for workers help to reduce the likelihood of swine IAV becoming endemic. Closely complementing the physical on-farm practices are IAV surveillance programs. Epidemiological data is critical in understanding regional distribution and variation to assist in determining an appropriate response to outbreaks and understanding the nature of historical swine IAV epidemics and zoonoses. Medical intervention in pigs is restricted to vaccination, a measure fraught with the intrinsic difficulties of mounting an immune response against a highly mutable virus. It is the best available tool for controlling IAV in swine but is far from being a perfect solution due to its unreliable efficacy and association with an enhanced respiratory disease. Because IAV generally has low mortality rates there is a reticence in the uptake of vaccination. Novel genetic technologies could be a complementary strategy for IAV control in pigs that confers broad-acting resistance. Transgenic pigs with IAV resistance are useful as models, however the complexity of these reaching the consumer market limits them to research models. More promising are gene-editing approaches to prevent viral exploitation of host proteins and modern vaccine technologies that surpass those currently available. CONCLUSION Using the suite of IAV control measures that are available for pigs effectively we can improve the economic productivity of pig farming whilst improving on-farm animal welfare standards and avoid facing the extensive social and financial costs of a pandemic. Fighting 'Flu in pigs will help mitigate the very real threat of a human pandemic emerging, increase security of the global food system and lead to healthier pigs.
Collapse
Affiliation(s)
- Hamish A. Salvesen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh, UK
| | - C. Bruce A. Whitelaw
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh, UK
| |
Collapse
|
12
|
Bullard BL, Corder BN, DeBeauchamp J, Rubrum A, Korber B, Webby RJ, Weaver EA. Epigraph hemagglutinin vaccine induces broad cross-reactive immunity against swine H3 influenza virus. Nat Commun 2021; 12:1203. [PMID: 33619277 PMCID: PMC7900167 DOI: 10.1038/s41467-021-21508-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 01/27/2021] [Indexed: 01/09/2023] Open
Abstract
Influenza A virus infection in swine impacts the agricultural industry in addition to its zoonotic potential. Here, we utilize epigraph, a computational algorithm, to design a universal swine H3 influenza vaccine. The epigraph hemagglutinin proteins are delivered using an Adenovirus type 5 vector and are compared to a wild type hemagglutinin and the commercial inactivated vaccine, FluSure. In mice, epigraph vaccination leads to significant cross-reactive antibody and T-cell responses against a diverse panel of swH3 isolates. Epigraph vaccination also reduces weight loss and lung viral titers in mice after challenge with three divergent swH3 viruses. Vaccination studies in swine, the target species for this vaccine, show stronger levels of cross-reactive antibodies and T-cell responses after immunization with the epigraph vaccine compared to the wild type and FluSure vaccines. In both murine and swine models, epigraph vaccination shows superior cross-reactive immunity that should be further investigated as a universal swH3 vaccine.
Collapse
Affiliation(s)
- Brianna L Bullard
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln, NE, USA
| | - Brigette N Corder
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln, NE, USA
| | | | - Adam Rubrum
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bette Korber
- Los Alamos National Laboratory, Los Alamos, NM, USA
| | | | - Eric A Weaver
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska, Lincoln, NE, USA.
| |
Collapse
|
13
|
Vaccines That Reduce Viral Shedding Do Not Prevent Transmission of H1N1 Pandemic 2009 Swine Influenza A Virus Infection to Unvaccinated Pigs. J Virol 2021; 95:JVI.01787-20. [PMID: 33268518 PMCID: PMC7851569 DOI: 10.1128/jvi.01787-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/15/2020] [Indexed: 01/19/2023] Open
Abstract
Swine influenza A virus (swIAV) infection causes substantial economic loss and disease burden in humans and animals. The 2009 pandemic H1N1 (pH1N1) influenza A virus is now endemic in both populations. In this study, we evaluated the efficacy of different vaccines in reducing nasal shedding in pigs following pH1N1 virus challenge. We also assessed transmission from immunized and challenged pigs to naive, directly in-contact pigs. Pigs were immunized with either adjuvanted, whole inactivated virus (WIV) vaccines or virus-vectored (ChAdOx1 and MVA) vaccines expressing either the homologous or heterologous influenza A virus hemagglutinin (HA) glycoprotein, as well as an influenza virus pseudotype (S-FLU) vaccine expressing heterologous HA. Only two vaccines containing homologous HA, which also induced high hemagglutination inhibitory antibody titers, significantly reduced virus shedding in challenged animals. Nevertheless, virus transmission from challenged to naive, in-contact animals occurred in all groups, although it was delayed in groups of vaccinated animals with reduced virus shedding.IMPORTANCE This study was designed to determine whether vaccination of pigs with conventional WIV or virus-vectored vaccines reduces pH1N1 swine influenza A virus shedding following challenge and can prevent transmission to naive in-contact animals. Even when viral shedding was significantly reduced following challenge, infection was transmissible to susceptible cohoused recipients. This knowledge is important to inform disease surveillance and control strategies and to determine the vaccine coverage required in a population, thereby defining disease moderation or herd protection. WIV or virus-vectored vaccines homologous to the challenge strain significantly reduced virus shedding from directly infected pigs, but vaccination did not completely prevent transmission to cohoused naive pigs.
Collapse
|
14
|
Reemers S, Verstegen I, Basten S, Hubers W, van de Zande S. A broad spectrum HVT-H5 avian influenza vector vaccine which induces a rapid onset of immunity. Vaccine 2021; 39:1072-1079. [PMID: 33483211 DOI: 10.1016/j.vaccine.2021.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/15/2020] [Accepted: 01/04/2021] [Indexed: 11/25/2022]
Abstract
Current methods to combat highly pathogenic avian influenza (HPAI) outbreaks in poultry rely on stamping out and preventive culling, which can lead to high economic losses and invoke ethical resistance. Emergency vaccination could be an alternative as vaccination is one of the most efficient and cost-effective measures to protect poultry from HPAI infection, preventing spreading to other poultry and greatly reducing the potential transmission to humans. Current conventional inactivated AI vaccines may be useful for combating AI outbreaks, but do not fulfil all targets of an ideal AI vaccine, including mass applicability and rapid onset of immunity. We aimed to further investigate the potential of Herpesvirus of Turkeys (HVT) as a vector containing a recombinant H5 hemagglutinin of HPAI H5N1. This HVT-H5 vector was analysed in vitro, tested for onset of immunity against AI challenge, breadth of protection, reduction of virus shedding, and induction of both antibody and cellular responses in SPF layers or broiler chicks containing maternal derived antibodies (MDA+). In SPF layers HVT-H5 provided full protection to lethal challenges with 4 antigenically diverse HPAI H5N1 strains from 2 weeks post vaccination (w.p.v.), while in MDA+ birds full protection was provided from 3 w.p.v. to homologous challenge. Also shedding of challenge virus was reduced in both SPF and MDA+ birds. HVT-H5 induced a protective HI titre (≥4) to 11 HPAI H5N1 strains at 3 w.p.v. in 3-week-old SPF layers and to HPAI H5N8 A/ch/Neth/14015531/2014. Besides inducing a protective antibody response HVT-H5 also induced an influenza-specific T cell response. This data demonstrates that HVT-H5 vaccine appears to fulfil many of the criteria for an ideal AI vaccine including early onset of immunity, a broad protection, reduced virus shedding, protection in presence of AI-MDA and could be a useful tool in the combat of AI outbreaks worldwide.
Collapse
Affiliation(s)
- Sylvia Reemers
- MSD Animal Health, Wim de Körverstraat 35, 5831 AN Boxmeer, the Netherlands.
| | - Iwan Verstegen
- MSD Animal Health, Wim de Körverstraat 35, 5831 AN Boxmeer, the Netherlands
| | - Stephanie Basten
- MSD Animal Health, Wim de Körverstraat 35, 5831 AN Boxmeer, the Netherlands
| | - Willem Hubers
- MSD Animal Health, Wim de Körverstraat 35, 5831 AN Boxmeer, the Netherlands
| | | |
Collapse
|
15
|
Nassif S, Zaki F, Mourad A, Fouad E, Saad A, Setta A, Felföldi B, Mató T, Kiss I, Palya V. Herpesvirus of turkey-vectored avian influenza vaccine offers cross-protection against antigenically drifted H5Nx highly pathogenic avian influenza virus strains. Avian Pathol 2020; 49:547-556. [PMID: 32615785 DOI: 10.1080/03079457.2020.1790502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Among the different vaccines used to control highly pathogenic avian influenza, an HVT vector-based live recombinant avian influenza vaccine, expressing the haemagglutinin gene of an H5N1 HPAI virus, has been used by the poultry industry since 2012. The objective of the study presented in this paper was to test the efficacy of the commercially available HVT-based recombinant H5 vaccine against antigenically drifted H5N1, H5N8 and H5N2 HPAI virus circulating in Egypt recently. Groups of SPF chicks vaccinated at day-old with the HVT-based recombinant H5 vaccine were challenged, along with non-vaccinated controls, with 106 EID50 each of H5N1, H5N2 or H5N8 HPAI virus at 28 days of age. The birds were monitored for clinical protection and virus shedding during a 10-day postchallenge period. Clinical protection levels were 90%, 90% and 80% following challenge with the H5N1, H5N2 and H5N8 field isolates, respectively. Challenge virus shedding was significantly reduced in vaccinated groups, with up to 40%, 30% and 20% of non-shedders, and 3.8, 3.3 and 2.8 log10 reduction in the amount of excreted virus following challenge with H5N1, H5N2 and H5N8 viruses, respectively. Analyses of the amino acid sequences of the HA proteins of challenge viruses and serological relatedness with the vaccine insert revealed significant antigenic divergences between the vaccine and the challenge viruses. These results provide further evidence of the potential of HVT-based recombinant H5 vaccine to provide cross-protection against antigenically drifted HPAI H5Nx viruses with strong control on virus shedding.
Collapse
Affiliation(s)
- Samir Nassif
- The Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agriculture Research Centre (ARC), Cairo, Egypt
| | - Farid Zaki
- The Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agriculture Research Centre (ARC), Cairo, Egypt
| | - Ahlam Mourad
- The Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agriculture Research Centre (ARC), Cairo, Egypt
| | - Esraa Fouad
- The Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agriculture Research Centre (ARC), Cairo, Egypt
| | - Asem Saad
- The Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agriculture Research Centre (ARC), Cairo, Egypt
| | - Ahmed Setta
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.,Ceva-Phylaxia, Ceva Sante Animale, Cairo, Egypt
| | | | - Tamás Mató
- Ceva-Phylaxia, Ceva Sante Animale, Budapest, Hungary
| | - Istvan Kiss
- Ceva-Phylaxia, Ceva Sante Animale, Budapest, Hungary
| | - Vilmos Palya
- Ceva-Phylaxia, Ceva Sante Animale, Budapest, Hungary
| |
Collapse
|
16
|
Yu L, Pan J, Cao G, Jiang M, Zhang Y, Zhu M, Liang Z, Zhang X, Hu X, Xue R, Gong C. AIV polyantigen epitope expressed by recombinant baculovirus induces a systemic immune response in chicken and mouse models. Virol J 2020; 17:121. [PMID: 32758272 PMCID: PMC7403573 DOI: 10.1186/s12985-020-01388-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The protective efficacy of avian influenza virus (AIV) vaccines is unsatisfactory due to the presence of various serotypes generated by genetic reassortment. Thus, immunization with a polyantigen chimeric epitope vaccine may be an effective strategy for protecting poultry from infection with different AIV subtypes. METHODS Baculovirus has recently emerged as a novel and attractive gene delivery vehicle for animal cells. In the present study, a recombinant baculovirus BmNPV-CMV/THB-P10/CTLT containing a fused codon-optimized sequence (CTLT) of T lymphocyte epitopes from H1HA, H9HA, and H7HA AIV subtypes, and another fused codon-optimized sequence (THB) of Th and B cell epitopes from H1HA, H9HA, and H7HA AIV subtypes, driven by a baculovirus P10 promoter and cytomegalovirus CMV promoter, respectively, was constructed. RESULTS Western blotting and cellular immunofluorescence demonstrated that the CTLT (THB) can be expressed in rBac-CMV/THB-P10/CTLT-infected silkworm cells (mammalian HEK293T cells). Furthermore, the recombinant virus, rBac-CMV-THB-CTLT, was used to immunize both chickens and mice. CONCLUSIONS The results of an indirect ELISA, immunohistochemistry, and T lymphocyte proliferation assay indicated that specific humoral and cellular responses were detected in both chicken and mice. These results suggest that rBac-CMV/THB-P10/CTLT can be developed as a potential vaccine against different AIV subtypes.
Collapse
Affiliation(s)
- Lei Yu
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
| | - Jun Pan
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
| | - Guangli Cao
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, 215123, China
| | - Mengsheng Jiang
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
| | - Yunshan Zhang
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
| | - Min Zhu
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
| | - Zi Liang
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
| | - Xing Zhang
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, 215123, China
| | - Xiaolong Hu
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, 215123, China
| | - Renyu Xue
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, 215123, China
| | - Chengliang Gong
- School of Biology and Basic Medical Sciences, Soochow University, No.199 Ren'ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park, Suzhou, 215123, P.R. China.
- Agricultural Biotechnology Research Institute, Agricultural biotechnology and Ecological Research Institute, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
17
|
Keay S, Poljak Z, Klapwyk M, O’Connor A, Friendship RM, O’Sullivan TL, Sargeant JM. Influenza A virus vaccine research conducted in swine from 1990 to May 2018: A scoping review. PLoS One 2020; 15:e0236062. [PMID: 32673368 PMCID: PMC7365442 DOI: 10.1371/journal.pone.0236062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/27/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Influenza A viruses of swine (IAV-S) are a global zoonotic and economic concern. Primary control is through vaccination yet a formal evidence map summarizing vaccine research conducted in pigs is not available. OBJECTIVE Ten characteristics of English language primary IAV-S vaccine research, conducted at the level of the pig or higher, were charted to identify research gaps, topics for systematic review, and coverage across different publication types. DESIGN Six online databases and grey literature were searched, without geographic, population, or study type restrictions, and abstracts screened independently and in duplicate for relevant research published between 1990 and May 2018. Full text data was charted by a single reviewer. RESULTS Over 11,000 unique citations were screened, identifying 376 for charting, including 175 proceedings from 60 conferences, and 170 journal articles from 51 journals. Reported outcomes were heterogeneous with measures of immunity (86%, n = 323) and virus detection (65%, n = 246) reported far more than production metrics (9%, n = 32). Study of transmissibility under conditions of natural exposure (n = 7), use of mathematical modelling (n = 11), and autogenous vaccine research reported in journals (n = 7), was limited. CONCLUSIONS Most research used challenge trials (n = 219) and may have poor field relevance or suitability for systematic review if the purpose is to inform clinical decisions. Literature on vaccinated breeding herds (n = 89) and weaned pigs (n = 136) is potentially sufficient for systematic review. Research under field conditions is limited, disproportionately reported in conference proceedings versus journal articles, and may be insufficient to support systematic review.
Collapse
Affiliation(s)
- Sheila Keay
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Zvonimir Poljak
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Mackenzie Klapwyk
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Annette O’Connor
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Robert M. Friendship
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Terri L. O’Sullivan
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jan M. Sargeant
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
- Centre for Public Health and Zoonoses, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
18
|
Magiri RB, Lai KJ, Mutwiri GK, Wilson HL. Experimental PCEP-Adjuvanted Swine Influenza H1N1 Vaccine Induced Strong Immune Responses but Did Not Protect Piglets against Heterologous H3N2 Virus Challenge. Vaccines (Basel) 2020; 8:E235. [PMID: 32443540 PMCID: PMC7349969 DOI: 10.3390/vaccines8020235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 01/07/2023] Open
Abstract
Vaccination is the most efficient method of protection against influenza infections. However, the rapidly mutating viruses and development of new strains make it necessary to develop new influenza vaccines annually. Hence, vaccines that stimulate cross-protection against multiple influenza subtypes are highly sought. Recent evidence suggests that adjuvants such as PCEP that promote Th1-type T cell and Th2-type T cell immune responses and broad-spectrum immune responses may confer cross-protection against heterologous influenza strains. In this study, we evaluated whether the immunogenic and protective potential of PCEP-adjuvanted inactivated swine influenza virus H1N1 vaccine can protect pigs immunized against live H3N2 virus. Piglets were vaccinated via the intradermal route with PCEP-adjuvanted inactivated swine influenza virus (SIV) H1N1 vaccine, boosted at day 21 with the same vaccines then challenged with infectious SIV H3N2 virus at day 35 via the tracheobronchial route. The pigs showed significant anti-H1N1 SIV specific antibody titres and H1N1 SIV neutralizing antibody titres, and these serum titres remained after the challenge with the H3N2 virus. In contrast, vaccination with anti-H1N1 SIV did not trigger anti-H3N2 SIV antibody titres or neutralizing antibody titres and these titres remained low until pigs were challenged with H3N2 SIV. At necropsy (six days after challenge), we collected prescapular lymph nodes and tracheobronchial draining the vaccination sites and challenge site, respectively. ELISPOTs from lymph node cells restimulated ex vivo with inactivated SIV H1N1 showed significant production of IFN-γ in the tracheobronchial cells, but not the prescapular lymph nodes. In contrast, lymph node cells restimulated ex vivo with inactivated SIV H1N1 showed significantly higher IL-13 and IL-17A in the prescapular lymph nodes draining the vaccination sites relative to unchallenged animals. Lung lesion scores show that intradermal vaccination with H1N1 SIV plus PCEP did not prevent lesions when the animals were challenged with H3N2. These results confirm previous findings that PCEP is effective as a vaccine adjuvant in that it induces strong immune responses and protects against homologous swine influenza H1N1 virus, but the experimental H1N1 vaccine failed to cross-protect against heterologous H3N2 virus.
Collapse
Affiliation(s)
- Royford Bundi Magiri
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada; (R.B.M.); (K.J.L.); (G.K.M.)
- Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada
- College of Agriculture, Fisheries and Forestry, Fiji National University, Suva 7222, Fiji
| | - Ken John Lai
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada; (R.B.M.); (K.J.L.); (G.K.M.)
| | - George Kiremu Mutwiri
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada; (R.B.M.); (K.J.L.); (G.K.M.)
- Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada
| | - Heather Lynne Wilson
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada; (R.B.M.); (K.J.L.); (G.K.M.)
- Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada
| |
Collapse
|
19
|
DNA vaccine based on conserved HA-peptides induces strong immune response and rapidly clears influenza virus infection from vaccinated pigs. PLoS One 2019; 14:e0222201. [PMID: 31553755 PMCID: PMC6760788 DOI: 10.1371/journal.pone.0222201] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/24/2019] [Indexed: 01/08/2023] Open
Abstract
Swine influenza virus (SIVs) infections cause a significant economic impact to the pork industry. Moreover, pigs may act as mixing vessel favoring genome reassortment of diverse influenza viruses. Such an example is the pandemic H1N1 (pH1N1) virus that appeared in 2009, harboring a combination of gene segments from avian, pig and human lineages, which rapidly reached pandemic proportions. In order to confront and prevent these possible emergences as well as antigenic drift phenomena, vaccination remains of vital importance. The present work aimed to evaluate a new DNA influenza vaccine based on distinct conserved HA-peptides fused with flagellin and applied together with Diluvac Forte as adjuvant using a needle-free device (IntraDermal Application of Liquids, IDAL®). Two experimental pig studies were performed to test DNA-vaccine efficacy against SIVs in pigs. In the first experiment, SIV-seronegative pigs were vaccinated with VC4-flagellin DNA and intranasally challenged with a pH1N1. In the second study, VC4-flagellin DNA vaccine was employed in SIV-seropositive animals and challenged intranasally with an H3N2 SIV-isolate. Both experiments demonstrated a reduction in the viral shedding after challenge, suggesting vaccine efficacy against both the H1 and H3 influenza virus subtypes. In addition, the results proved that maternally derived antibodies (MDA) did not constitute an obstacle to the vaccine approach used. Moreover, elevated titers in antibodies both against H1 and H3 proteins in serum and in bronchoalveolar lavage fluids (BALFs) was detected in the vaccinated animals along with a markedly increased mucosal IgA response. Additionally, vaccinated animals developed stronger neutralizing antibodies in BALFs and higher inhibiting hemagglutination titers in sera against both the pH1N1 and H3N2 influenza viruses compared to unvaccinated, challenged-pigs. It is proposed that the described DNA-vaccine formulation could potentially be used as a multivalent vaccine against SIV infections.
Collapse
|
20
|
Gebauer M, Hürlimann HC, Behrens M, Wolff T, Behrens SE. Subunit vaccines based on recombinant yeast protect against influenza A virus in a one-shot vaccination scheme. Vaccine 2019; 37:5578-5587. [PMID: 31399274 DOI: 10.1016/j.vaccine.2019.07.094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/22/2019] [Accepted: 07/27/2019] [Indexed: 01/03/2023]
Abstract
Here we report on new subunit vaccines based on recombinant yeast of the type Kluyveromyces lactis (K. lactis), which protect mice from a lethal influenza A virus infection. Applying a genetic system that enables the rapid generation of transgenic yeast, we have developed K. lactis strains that express the influenza A virus hemagglutinin, HA, either individually or in combination with the viral M1 matrix protein. Subcutaneous application of the inactivated, but otherwise non-processed yeast material shows a complete protection of BALB/c mice in prime/boost and even one-shot/single dose vaccination schemes against a subsequent, lethal challenge with the cognate influenza virus. The yeast vaccines induce titers of neutralizing antibodies that are readily comparable to those induced by an inactivated virus vaccine. These data suggest that HA and M1 are produced with a high antigenicity in the yeast cells. Based on these findings, multivalent, DIVA-capable, yeast-based subunit vaccines may be developed as promising alternatives to conventional virus-based anti-flu vaccines for veterinary applications.
Collapse
Affiliation(s)
- Mandy Gebauer
- Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Institute of Biochemistry and Biotechnology, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Hans C Hürlimann
- Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Institute of Biology, Weinbergweg 10, 06120 Halle (Saale), Germany
| | - Martina Behrens
- Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Institute of Biochemistry and Biotechnology, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Thorsten Wolff
- Robert Koch Institute, Unit 17 "Influenza and Other Respiratory Viruses", Seestr. 10, 13353 Berlin, Germany
| | - Sven-Erik Behrens
- Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Institute of Biochemistry and Biotechnology, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany.
| |
Collapse
|
21
|
Mamerow S, Scheffter R, Röhrs S, Stech O, Blohm U, Schwaiger T, Schröder C, Ulrich R, Schinköthe J, Beer M, Mettenleiter TC, Stech J. Double-attenuated influenza virus elicits broad protection against challenge viruses with different serotypes in swine. Vet Microbiol 2019; 231:160-168. [PMID: 30955804 DOI: 10.1016/j.vetmic.2019.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022]
Abstract
Influenza A viruses (IAV) have caused seasonal epidemics and severe pandemics in humans. Novel pandemic strains as in 2009 may emerge from pigs, serving as perpetual virus reservoir. However, reliably effective vaccination has remained a key issue for humans and swine. Here, we generated a novel double-attenuated influenza live vaccine by reverse genetics and subjected immunized mice and pigs to infection with the homologous wild-type, another homosubtypic H1N1, or a heterosubtypic H3N2 virus to address realistic challenge constellations. This attenuated mutant contains an artificial, strictly elastase-dependent hemagglutinin cleavage site and a C-terminally truncated NS1 protein from the IAV A/Bayern/74/2009 (H1N1pdm09). Prior to challenge, we immunized mice once and pigs twice intranasally. In vitro, the double-attenuated mutant replicated strictly elastase-dependently. Immunized mice and pigs developed neither clinical symptoms nor detectable virus replication after homologous challenge. In pigs, we observed considerably reduced clinical signs and no nasal virus shedding after homosubtypic and reduced viral loads in respiratory tracts after heterosubtypic infection. Protection against homosubtypic challenge suggests that an optimized backbone strain may require less frequent updates with recent HA and NA genes and still induce robust protection in relevant IAV hosts against drifted viruses.
Collapse
Affiliation(s)
- Svenja Mamerow
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Robert Scheffter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Susanne Röhrs
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Olga Stech
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Ulrike Blohm
- Institute of Immunology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Theresa Schwaiger
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Charlotte Schröder
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Reiner Ulrich
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Jan Schinköthe
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Thomas C Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Jürgen Stech
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany.
| |
Collapse
|
22
|
Palya V, Tatár-Kis T, Walkóné Kovács E, Kiss I, Homonnay Z, Gardin Y, Kertész K, Dán Á. Efficacy of a Recombinant Turkey Herpesvirus AI (H5) Vaccine in Preventing Transmission of Heterologous Highly Pathogenic H5N8 Clade 2.3.4.4b Challenge Virus in Commercial Broilers and Layer Pullets. J Immunol Res 2018; 2018:3143189. [PMID: 30584541 PMCID: PMC6280313 DOI: 10.1155/2018/3143189] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/24/2018] [Accepted: 09/04/2018] [Indexed: 12/31/2022] Open
Abstract
Outbreaks caused by the highly pathogenic avian influenza virus (HPAIV) H5N8 subtype clade 2.3.4.4 were first reported in 2014 in South Korea then spread very rapidly in Asia, to Europe, and for the first time, to North America. Efficacy of a recombinant HVT-AI (H5) vaccine (rHVT-H5) to provide clinical protection as well as to significantly reduce the shedding of an H5N8 challenge virus has already been demonstrated in SPF chickens. The aim of our studies was to test the efficacy of the same rHVT-H5 vaccine in controlling the transmission of a recent Hungarian HPAIV H5N8 challenge virus in commercial chickens. Broilers and layers were vaccinated at day old according to the manufacturer's recommendation and then challenged with a 2017 Hungarian HPAIV H5N8 (2.3.4.4b) isolate at 5 or 7 weeks of age, respectively. Evaluation of clinical protection, reduction of challenge virus shedding, and transmission to vaccinated contact birds was done on the basis of clinical signs/mortality, detection, and quantitation of challenge virus in oronasal and cloacal swabs (regularly between 1 and 14 days postchallenge). Measurement of seroconversion to AIV nucleoprotein was used as an indicator of infection and replication of challenge virus. Our results demonstrated that rHVT-H5 vaccination could prevent the development of clinical disease and suppress shedding very efficiently, resulting in the lack of challenge virus transmission to vaccinated contact chickens, regardless the type of birds. Single immunization with the tested rHVT-H5 vaccine proved to be effective to stop HPAIV H5N8 (2.3.4.4b) transmission within vaccinated poultry population under experimental conditions.
Collapse
Affiliation(s)
- Vilmos Palya
- Scientific Support and Investigation Unit, Ceva-Phylaxia, Ceva Animal Health, Budapest 1107, Hungary
| | - Tímea Tatár-Kis
- Scientific Support and Investigation Unit, Ceva-Phylaxia, Ceva Animal Health, Budapest 1107, Hungary
| | - Edit Walkóné Kovács
- Scientific Support and Investigation Unit, Ceva-Phylaxia, Ceva Animal Health, Budapest 1107, Hungary
| | - István Kiss
- Scientific Support and Investigation Unit, Ceva-Phylaxia, Ceva Animal Health, Budapest 1107, Hungary
| | - Zalán Homonnay
- Scientific Support and Investigation Unit, Ceva-Phylaxia, Ceva Animal Health, Budapest 1107, Hungary
| | | | | | - Ádám Dán
- Veterinary Diagnostic Directorate, National Food Chain Safety Office (NEBIH), Budapest 1149, Hungary
| |
Collapse
|
23
|
MicroRNA-Based Attenuation of Influenza Virus across Susceptible Hosts. J Virol 2018; 92:JVI.01741-17. [PMID: 29093096 DOI: 10.1128/jvi.01741-17] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022] Open
Abstract
Influenza A virus drives significant morbidity and mortality in humans and livestock. Annual circulation of the virus in livestock and waterfowl contributes to severe economic disruption and increases the risk of zoonotic transmission of novel strains into the human population, where there is no preexisting immunity. Seasonal vaccinations in humans help prevent infection and can reduce symptoms when infection does occur. However, current vaccination regimens available for livestock are limited in part due to safety concerns regarding reassortment/recombination with circulating strains. Therefore, inactivated vaccines are used instead of the more immunostimulatory live attenuated vaccines. MicroRNAs (miRNAs) have been used previously to generate attenuated influenza A viruses for use as a vaccine. Here, we systematically targeted individual influenza gene mRNAs using the same miRNA to determine the segment(s) that yields maximal attenuation potential. This analysis demonstrated that targeting of NP mRNA most efficiently ablates replication. We further increased the plasticity of miRNA-mediated attenuation of influenza A virus by exploiting a miRNA, miR-21, that is ubiquitously expressed across influenza-susceptible hosts. In order to construct this targeted virus, we used CRISPR/Cas9 to eliminate the universally expressed miR-21 from MDCK cells. miR-21-targeted viruses were attenuated in human, mouse, canine, and avian cells and drove protective immunity in mice. This strategy has the potential to enhance the safety of live attenuated vaccines in humans and zoonotic reservoirs.IMPORTANCE Influenza A virus circulates annually in both avian and human populations, causing significant morbidity, mortality, and economic burden. High incidence of zoonotic infections greatly increases the potential for transmission to humans, where no preexisting immunity or vaccine exists. There is a critical need for new vaccine strategies to combat emerging influenza outbreaks. MicroRNAs were used previously to attenuate influenza A viruses. We propose the development of a novel platform to produce live attenuated vaccines that are highly customizable, efficacious across a broad species range, and exhibit enhanced safety over traditional vaccination methods. This strategy exploits a microRNA that is expressed abundantly in influenza virus-susceptible hosts. By eliminating this ubiquitous microRNA from a cell line, targeted viruses that are attenuated across susceptible strains can be generated. This approach greatly increases the plasticity of the microRNA targeting approach and enhances vaccine safety.
Collapse
|
24
|
Yoo SJ, Kwon T, Lyoo YS. Challenges of influenza A viruses in humans and animals and current animal vaccines as an effective control measure. Clin Exp Vaccine Res 2018; 7:1-15. [PMID: 29399575 PMCID: PMC5795040 DOI: 10.7774/cevr.2018.7.1.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 12/07/2017] [Accepted: 12/12/2017] [Indexed: 11/28/2022] Open
Abstract
Influenza A viruses (IAVs) are genetically diverse and variable pathogens that share various hosts including human, swine, and domestic poultry. Interspecies and intercontinental viral spreads make the ecology of IAV more complex. Beside endemic IAV infections, human has been exposed to pandemic and zoonotic threats from avian and swine influenza viruses. Animal health also has been threatened by high pathogenic avian influenza viruses (in domestic poultry) and reverse zoonosis (in swine). Considering its dynamic interplay between species, prevention and control against IAV should be conducted effectively in both humans and animal sectors. Vaccination is one of the most efficient tools against IAV. Numerous vaccines against animal IAVs have been developed by a variety of vaccine technologies and some of them are currently commercially available. We summarize several challenges in control of IAVs faced by human and animals and discuss IAV vaccines for animal use with those application in susceptible populations.
Collapse
Affiliation(s)
- Sung J. Yoo
- College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Taeyong Kwon
- College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Young S. Lyoo
- College of Veterinary Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
25
|
Kapczynski DR, Sylte MJ, Killian ML, Torchetti MK, Chrzastek K, Suarez DL. Protection of commercial turkeys following inactivated or recombinant H5 vaccine application against the 2015U.S. H5N2 clade 2.3.4.4 highly pathogenic avian influenza virus. Vet Immunol Immunopathol 2017; 191:74-79. [PMID: 28895870 DOI: 10.1016/j.vetimm.2017.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/25/2017] [Accepted: 08/03/2017] [Indexed: 01/20/2023]
Abstract
Between December 2014 and June 2015, North America experienced the largest recorded foreign animal disease outbreak with over 47 million poultry dead or euthanized from viral exposure to a clade 2.3.4.4 H5 highly pathogenic avian influenza (HPAI) epizootic. Soon after the epizootic began, the U.S. Department of Agriculture (USDA) began testing the efficacy of different vaccines as a possible future control strategy. The aim of these studies were to evaluate the efficacy three H5 vaccines to aid in control of HPAI in commercial turkeys. Three different vaccine technologies were evaluated for efficacy: 1) inactivated reverse genetic laboratory-generated virus encoding a clade 2.3.4.4 H5 hemagglutinin (HA) gene (rgH5), 2) recombinant turkey herpesvirus encoding a clade 2.2. H5 HA (rHVT-AI), and 3) recombinant replication-deficient alphavirus RNA particle vaccine encoding a clade 2.3.4.4 H5 HA (RP-H5). All vaccines tested significantly (P<0.01) increased survival rates between vaccinated and sham vaccinated groups of poults challenged with A/turkey/Minnesota/12582/2015 clade 2.3.4.4 H5N2 HPAI. The rgH5 vaccine had detectable serum hemagglutination inhibition (HI) antibody against the challenge virus, and significantly reduced the frequency and level of viral shedding from oropharyngeal and cloacal swabs at days 2 and 4 post-challenge. Vaccination with only rHVT-AI or RP-H5 was not 100% protective, and failed to significantly reduce viral shedding post-challenge. A combined prime and boost strategy with the rHVT-AI and RP-H5, or rHVT-AI and rgH5, was 100% protective against lethal H5N2 HPAI challenge. Results of these studies led to USDA conditional approval of commercially available recombinant vaccines for use in turkeys as a control measure for clade 2.3.4.4 H5 HPAI epizootics.
Collapse
Affiliation(s)
- Darrell R Kapczynski
- Exotic and Emerging Avian Viral Diseases Research Unit, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA, 30605, United States.
| | - Matthew J Sylte
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, 1920 Dayton Avenue, Ames, IA, 50010, United States
| | - Mary L Killian
- National Veterinary Services Laboratories, Veterinary Services, Animal and Plant Health Inspection Service, U.S. Department of Agriculture, Ames, IA 50010, United States
| | - Mia K Torchetti
- National Veterinary Services Laboratories, Veterinary Services, Animal and Plant Health Inspection Service, U.S. Department of Agriculture, Ames, IA 50010, United States
| | - Klaudia Chrzastek
- Exotic and Emerging Avian Viral Diseases Research Unit, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA, 30605, United States
| | - David L Suarez
- Exotic and Emerging Avian Viral Diseases Research Unit, U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, 934 College Station Road, Athens, GA, 30605, United States
| |
Collapse
|
26
|
Bassano I, Ong SH, Lawless N, Whitehead T, Fife M, Kellam P. Accurate characterization of the IFITM locus using MiSeq and PacBio sequencing shows genetic variation in Galliformes. BMC Genomics 2017; 18:419. [PMID: 28558694 PMCID: PMC5450142 DOI: 10.1186/s12864-017-3801-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 05/16/2017] [Indexed: 01/16/2023] Open
Abstract
Background Interferon inducible transmembrane (IFITM) proteins are effectors of the immune system widely characterized for their role in restricting infection by diverse enveloped and non-enveloped viruses. The chicken IFITM (chIFITM) genes are clustered on chromosome 5 and to date four genes have been annotated, namely chIFITM1, chIFITM3, chIFITM5 and chIFITM10. However, due to poor assembly of this locus in the Gallus Gallus v4 genome, accurate characterization has so far proven problematic. Recently, a new chicken reference genome assembly Gallus Gallus v5 was generated using Sanger, 454, Illumina and PacBio sequencing technologies identifying considerable differences in the chIFITM locus over the previous genome releases. Methods We re-sequenced the locus using both Illumina MiSeq and PacBio RS II sequencing technologies and we mapped RNA-seq data from the European Nucleotide Archive (ENA) to this finalized chIFITM locus. Using SureSelect probes capture probes designed to the finalized chIFITM locus, we sequenced the locus of a different chicken breed, namely a White Leghorn, and a turkey. Results We confirmed the Gallus Gallus v5 consensus except for two insertions of 5 and 1 base pair within the chIFITM3 and B4GALNT4 genes, respectively, and a single base pair deletion within the B4GALNT4 gene. The pull down revealed a single amino acid substitution of A63V in the CIL domain of IFITM2 compared to Red Jungle fowl and 13, 13 and 11 differences between IFITM1, 2 and 3 of chickens and turkeys, respectively. RNA-seq shows chIFITM2 and chIFITM3 expression in numerous tissue types of different chicken breeds and avian cell lines, while the expression of the putative chIFITM1 is limited to the testis, caecum and ileum tissues. Conclusions Locus resequencing using these capture probes and RNA-seq based expression analysis will allow the further characterization of genetic diversity within Galliformes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3801-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Irene Bassano
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Division of Infectious Diseases, Department of Medicine, Imperial College Faculty of Medicine, Wright Fleming Wing, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
| | - Swee Hoe Ong
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Nathan Lawless
- The Pirbright Institute, Pirbright Laboratory, Ash Road, Woking, GU24 0NF, UK
| | - Thomas Whitehead
- The Pirbright Institute, Pirbright Laboratory, Ash Road, Woking, GU24 0NF, UK
| | - Mark Fife
- The Pirbright Institute, Pirbright Laboratory, Ash Road, Woking, GU24 0NF, UK
| | - Paul Kellam
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK. .,Division of Infectious Diseases, Department of Medicine, Imperial College Faculty of Medicine, Wright Fleming Wing, St Mary's Campus, Norfolk Place, London, W2 1PG, UK.
| |
Collapse
|
27
|
Tchilian E, Holzer B. Harnessing Local Immunity for an Effective Universal Swine Influenza Vaccine. Viruses 2017; 9:v9050098. [PMID: 28475122 PMCID: PMC5454411 DOI: 10.3390/v9050098] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 02/06/2023] Open
Abstract
Influenza A virus infections are a global health threat to humans and are endemic in pigs, contributing to decreased weight gain and suboptimal reproductive performance. Pigs are also a source of new viruses of mixed swine, avian, and human origin, potentially capable of initiating human pandemics. Current inactivated vaccines induce neutralising antibody against the immunising strain but rapid escape occurs through antigenic drift of the surface glycoproteins. However, it is known that prior infection provides a degree of cross-protective immunity mediated by cellular immune mechanisms directed at the more conserved internal viral proteins. Here we review new data that emphasises the importance of local immunity in cross-protection and the role of the recently defined tissue-resident memory T cells, as well as locally-produced, and sometimes cross-reactive, antibody. Optimal induction of local immunity may require aerosol delivery of live vaccines, but it remains unclear how long protective local immunity persists. Nevertheless, a universal vaccine might be extremely useful for disease prevention in the face of a pandemic. As a natural host for influenza A viruses, pigs are both a target for a universal vaccine and an excellent model for developing human influenza vaccines.
Collapse
Affiliation(s)
- Elma Tchilian
- The Pirbright Institute, Woking, Surrey GU24 0NF, UK.
| | | |
Collapse
|
28
|
Hu CMJ, Chien CY, Liu MT, Fang ZS, Chang SY, Juang RH, Chang SC, Chen HW. Multi-antigen avian influenza a (H7N9) virus-like particles: particulate characterizations and immunogenicity evaluation in murine and avian models. BMC Biotechnol 2017; 17:2. [PMID: 28061848 PMCID: PMC5219756 DOI: 10.1186/s12896-016-0321-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 12/07/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Human infection with avian influenza A virus (H7N9) was first reported in China in March 2013. Since then, hundreds of cases have been confirmed showing severe symptoms with a high mortality rate. The virus was transmitted from avian species to humans and has spread to many neighboring areas, raising serious concerns over its pandemic potential. Towards containing the disease, the goal of this study is to prepare a virus-like particle (VLP) that consists of hemagglutinin (HA), neuraminidase (NA) and matrix protein 1 (M1) derived from the human isolate A/Taiwan/S02076/2013(H7N9) for potential vaccine development. RESULTS Full length HA, NA, and M1 protein genes were cloned and expressed using a baculoviral expression system, and the VLPs were generated by co-infecting insect cells with three respective recombinant baculoviruses. Nanoparticle tracking analysis and transmission electron microscopy were applied to verify the VLPs' structure and antigenicity, and the multiplicity of infection of the recombinant baculoviruses was adjusted to achieve the highest hemagglutination activity. In animal experiments, BALB/c mice and specific-pathogen-free chickens receiving the VLP immunization showed elevated hemagglutination inhibition serum titer and antibodies against NA and M1 proteins. In addition, examination of cellular immunity showed the VLP-immunized mice and chickens exhibited an increased splenic antigen-specific cytokines production. CONCLUSIONS The H7N9 VLPs possess desirable immunogenicity in vivo and may serve as a candidate for vaccine development against avian influenza A (H7N9) infection.
Collapse
Affiliation(s)
- Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Research Center for Nanotechnology and Infectious Diseases, Taipei, Taiwan
| | - Chu-Yang Chien
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Tsan Liu
- Center for Research, Diagnostics and Vaccine Development, Centers for Disease Control, Taipei, Taiwan
| | - Zih-Syun Fang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Rong-Huay Juang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Shih-Chung Chang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Hui-Wen Chen
- Research Center for Nanotechnology and Infectious Diseases, Taipei, Taiwan. .,Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
29
|
Swine and Avian Influenza Outbreaks in Recent Times. EMERGING ZOONOSES 2017. [PMCID: PMC7119929 DOI: 10.1007/978-3-319-50890-0_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Influenza A is a zoonotic virus and wild waterfowls are the main reservoir of avian influenza viruses, which are precursors of human influenza A viruses. Through mutations and gene reassortment, some strains of avian influenza viruses establish stable lineages in poultry species, pigs, horses, and humans. The first zoonotic influenza pandemic of the twenty-first century, the swine H1N1 pandemic of 2009, originated from Mexico, and fortunately the virus was only of modest virulence. However, lessons have been learned on the shortcomings of the global preparedness for influenza pandemic, and this should be considered as a valuable experience for the preparation of the next major outbreak. Of more concern is the emergence of the highly pathogenic avian influenza A [H5N1], ongoing since 1996, and the low pathogenic avian influenza A [H7N9], since 2013, which have crossed the species barrier to humans in China. Risks of a H5N1 pandemic appear to be receding with declining human cases, and the H7N9 influenza virus is now the leading candidate as the next pandemic influenza virus. However, influenza pandemics are unpredictable in their timing, specific strain of virus, and origin. Most experts predict that the next influenza pandemic will arise from Asia, especially China, and will be directly of avian origin. Continued influenza surveillance in animals and humans globally with prompt reporting to the WHO and the World Animal Health Organization with sharing of data promptly between countries is essential. Long-term solutions to prevent cross-species transmission of zoonotic influenza viruses to humans and development of more effective, longer-lasting vaccines against emerging avian influenza viruses are needed. Currently there is no evidence of an impending zoonotic or avian influenza pandemic, and the viruses of interest, H5N1 and H7N9 avian influenza A viruses, have not mutated to allow for easy transmission to humans nor human to human.
Collapse
|
30
|
Kapczynski DR, Tumpey TM, Hidajat R, Zsak A, Chrzastek K, Tretyakova I, Pushko P. Vaccination with virus-like particles containing H5 antigens from three H5N1 clades protects chickens from H5N1 and H5N8 influenza viruses. Vaccine 2016; 34:1575-1581. [PMID: 26868083 DOI: 10.1016/j.vaccine.2016.02.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/22/2016] [Accepted: 02/01/2016] [Indexed: 12/28/2022]
Abstract
Highly pathogenic avian influenza (HPAI) viruses, especially H5N1 strains, represent a public health threat and cause widespread morbidity and mortality in domestic poultry. Recombinant virus-like particles (VLPs) represent a promising novel vaccine approach to control avian influenza including HPAI strains. Influenza VLPs contain viral hemagglutinin (HA), which can be expressed in cell culture within highly immunogenic VLPs that morphologically and antigenically resemble influenza virions, except VLPs are non-infectious. Here we describe a recombinant VLP containing HA proteins derived from three distinct clades of H5N1 viruses as an experimental, broadly protective H5 avian influenza vaccine. A baculovirus vector was configured to co-express the H5 genes from recent H5N1 HPAI isolates A/chicken/Germany/2014 (clade 2.3.4.4), A/chicken/West Java/Subang/29/2007 (clade 2.1.3) and A/chicken/Egypt/121/2012 (clade 2.2.1). Co-expression of these genes in Sf9 cells along with influenza neuraminidase (NA) and retrovirus gag genes resulted in production of triple-clade H555 VLPs that exhibited hemagglutination activity and morphologically resembled influenza virions. Vaccination of chickens with these VLPs resulted in induction of serum antibody responses and efficient protection against experimental challenges with three different viruses including the recent U.S. H5N8 HPAI isolate. We conclude that these novel triple-clade VLPs represent a feasible strategy for simultaneously evoking protective antibodies against multiple variants of H5 influenza virus.
Collapse
Affiliation(s)
| | - Terrence M Tumpey
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Road N.E., Atlanta, GA, USA
| | - Rachmat Hidajat
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA
| | | | | | | | - Peter Pushko
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA.
| |
Collapse
|
31
|
Tretyakova I, Hidajat R, Hamilton G, Horn N, Nickols B, Prather RO, Tumpey TM, Pushko P. Preparation of quadri-subtype influenza virus-like particles using bovine immunodeficiency virus gag protein. Virology 2016; 487:163-71. [PMID: 26529299 PMCID: PMC4679414 DOI: 10.1016/j.virol.2015.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/02/2015] [Accepted: 10/03/2015] [Indexed: 01/13/2023]
Abstract
Influenza VLPs comprised of hemagglutinin (HA), neuraminidase (NA), and matrix (M1) proteins have been previously used for immunological and virological studies. Here we demonstrated that influenza VLPs can be made in Sf9 cells by using the bovine immunodeficiency virus gag (Bgag) protein in place of M1. We showed that Bgag can be used to prepare VLPs for several influenza subtypes including H1N1 and H10N8. Furthermore, by using Bgag, we prepared quadri-subtype VLPs, which co-expressed within the VLP the four HA subtypes derived from avian-origin H5N1, H7N9, H9N2 and H10N8 viruses. VLPs showed hemagglutination and neuraminidase activities and reacted with specific antisera. The content and co-localization of each HA subtype within the quadri-subtype VLP were evaluated. Electron microscopy showed that Bgag-based VLPs resembled influenza virions with the diameter of 150-200nm. This is the first report of quadri-subtype design for influenza VLP and the use of Bgag for influenza VLP preparation.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Cell Line
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunodeficiency Virus, Bovine/genetics
- Immunodeficiency Virus, Bovine/immunology
- Influenza A Virus, H10N8 Subtype/genetics
- Influenza A Virus, H10N8 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H9N2 Subtype/genetics
- Influenza A Virus, H9N2 Subtype/immunology
- Insecta
- Neuraminidase/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Sf9 Cells
- Spodoptera
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
| | - Rachmat Hidajat
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA
| | | | - Noah Horn
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA
| | - Brian Nickols
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA
| | | | - Terrence M Tumpey
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Road N.E., Atlanta, GA, USA
| | - Peter Pushko
- Medigen, Inc., 8420 Gas House Pike, Suite S, Frederick, MD, USA.
| |
Collapse
|