1
|
Zang X, Li G, Zhu J, Dong X, Zhai Y. Evaluation of the adjuvant effect of imiquimod and CpG ODN 1826 in chimeric DNA vaccine against Japanese encephalitis. Int Immunopharmacol 2024; 140:112816. [PMID: 39083930 DOI: 10.1016/j.intimp.2024.112816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/15/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Vaccines represent a significant milestone in the history of human medical science and serve as the primary means for controlling infectious diseases. In recent years, the geographical distribution of Japanese encephalitis viruses (JEV) of various genotypes has become increasingly complex, which provides a rationale for the development of safer and more effective vaccines. The advent of subunit and nucleic acid vaccines, especially propelled by advancements in genetic engineering since the 1980s, has accelerated the application of novel adjuvants. These novel vaccine adjuvants have diversified into toll-like receptor (TLR) agonists, complex adjuvants, nanoparticles and so on. However, the efficacy of adjuvant combinations can vary depending on the host system, disease model, or vaccine formulation, sometimes resulting in competitive or counteractive effects. In our previous study, we constructed a pJME-LC3 chimeric DNA vaccine aimed at inducing an immune response through autophagy induction. Building on this, we investigated the impact of the TLR7/8 agonist imiquimod (IMQ) and the TLR9 agonist CpG ODN 1826 as adjuvants on the immunogenicity of the Japanese encephalitis chimeric DNA vaccine. Our findings indicate that the combination of the pJME-LC3 vaccine with IMQ and CpG ODN 1826 adjuvants enhanced the innate immune response, promoting the maturation and activation of antigen-presenting cells in the early immune response. Furthermore, it played a regulatory and optimizing role in subsequent antigen-specific immune responses, resulting in effective cellular and humoral immunity and providing prolonged immune protection. The synergistic effect of IMQ and CpG ODN 1826 as adjuvants offers a novel approach for the development of Japanese encephalitis nucleic acid vaccines.
Collapse
Affiliation(s)
- Xin Zang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Guozhen Li
- Department of Gastroenterology, Wuhan Red Cross Hospital, Wuhan 430015, China
| | - Junyao Zhu
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaoying Dong
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yongzhen Zhai
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
2
|
Kim M, Noh K, Kim P, Kim JH, Choi BW, Singh R, Choi JH, Han SB, Kim SS, Lee EY, Bae MA, Shin D, Kim M, Ahn JH. Design, Synthesis, and Biological Evaluation of New 2,6,7-Substituted Purine Derivatives as Toll-like Receptor 7 Agonists for Intranasal Vaccine Adjuvants. J Med Chem 2024; 67:9389-9405. [PMID: 38787938 DOI: 10.1021/acs.jmedchem.4c00489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
TLR7/8 agonists are versatile immune stimulators capable of treating various diseases such as viral infections, autoimmune, and cancer. Despite the structural similarity of TLR7/8, their immune stimulation mechanisms and time-course responses significantly differ. In this study, a new series of TLR7-selective agonists was synthesized utilizing the economical building block 2,6-dichloropurine. Compound 27b showed the most potent activity on hTLR7 with an EC50 of 17.53 nM and demonstrated high hTLR7 selectivity (224 folds against TLR8). 27b effectively stimulated the secretion of proinflammatory cytokines in mouse macrophages and enhanced intranasal vaccine efficacy against influenza A virus in vivo. Assessment of humoral and mucosal antibody titers confirmed that 27b elevates IgG and IgA levels, protecting against both homologous and heterologous influenza viral infections. These findings suggest that 27b is a promising candidate as a vaccine adjuvant to prevent viral infections or as a robust immunomodulator with prolonged activity for treating immune-suppressed diseases.
Collapse
Affiliation(s)
- Morgan Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Kyungseob Noh
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Pyeongkeun Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jae Ho Kim
- JD Bioscience, 208 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Byeong Wook Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Ravi Singh
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jun-Ho Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Soo Bong Han
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seong Soon Kim
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Eun-Young Lee
- JD Bioscience, 208 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Myung Ae Bae
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Daeho Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- JD Bioscience, 208 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| |
Collapse
|
3
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Recent Insights into the Molecular Mechanisms of the Toll-like Receptor Response to Influenza Virus Infection. Int J Mol Sci 2024; 25:5909. [PMID: 38892096 PMCID: PMC11172706 DOI: 10.3390/ijms25115909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Influenza A viruses (IAVs) pose a significant global threat to human health. A tightly controlled host immune response is critical to avoid any detrimental effects of IAV infection. It is critical to investigate the association between the response of Toll-like receptors (TLRs) and influenza virus. Because TLRs may act as a double-edged sword, a balanced TLR response is critical for the overall benefit of the host. Consequently, a thorough understanding of the TLR response is essential for targeting TLRs as a novel therapeutic and prophylactic intervention. To date, a limited number of studies have assessed TLR and IAV interactions. Therefore, further research on TLR interactions in IAV infection should be conducted to determine their role in host-virus interactions in disease causation or clearance of the virus. Although influenza virus vaccines are available, they have limited efficacy, which should be enhanced to improve their efficacy. In this study, we discuss the current status of our understanding of the TLR response in IAV infection and the strategies adopted by IAVs to avoid TLR-mediated immune surveillance, which may help in devising new therapeutic or preventive strategies. Furthermore, recent advances in the use of TLR agonists as vaccine adjuvants to enhance influenza vaccine efficacy are discussed.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
4
|
Pagh-Berendtsen N, Pavlovskyi A, Flores Téllez D, Egebjerg C, Kolmos MG, Justinussen J, Kornum BR. Downregulation of hypocretin/orexin after H1N1 Pandemrix vaccination of adolescent mice. Sleep 2024; 47:zsae014. [PMID: 38227834 DOI: 10.1093/sleep/zsae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/07/2023] [Indexed: 01/18/2024] Open
Abstract
Narcolepsy type 1 (NT1), characterized by the loss of hypocretin/orexin (HCRT) production in the lateral hypothalamus, has been linked to Pandemrix vaccination during the 2009 H1N1 pandemic, especially in children and adolescents. It is still unknown why this vaccination increased the risk of developing NT1. This study investigated the effects of Pandemrix vaccination during adolescence on Hcrt mRNA expression in mice. Mice received a primary vaccination (50 µL i.m.) during prepubescence and a booster vaccination during peri-adolescence. Hcrt expression was measured at three-time points after the vaccinations. Control groups included both a saline group and an undisturbed group of mice. Hcrt expression was decreased after both Pandemrix and saline injections, but 21 days after the second injection, the saline group no longer showed decreased Hcrt expression, while the Pandemrix group still exhibited a significant reduction of about 60% compared to the undisturbed control group. This finding suggests that Pandemrix vaccination during adolescence influences Hcrt expression in mice into early adulthood. The Hcrt mRNA level did not reach the low levels known to induce NT1 symptoms, instead, our finding supports the multiple-hit hypothesis of NT1 that states that several insults to the HCRT system may be needed to induce NT1 and that Pandemrix could be one such insult.
Collapse
Affiliation(s)
- Nicolai Pagh-Berendtsen
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Artem Pavlovskyi
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Daniel Flores Téllez
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Christine Egebjerg
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Mie Gunni Kolmos
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Jessica Justinussen
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| | - Birgitte Rahbek Kornum
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Denmark
| |
Collapse
|
5
|
Chen Y, Song W, Li C, Wang J, Liu F, Ye Z, Ren P, Tong Y, Li J, Ou Z, Lee ACY, Cai JP, Wong BHY, Chan JFW, Yuen KY, Zhang AJX, Chu H. COVID-19 mRNA vaccine protects against SARS-CoV-2 Omicron BA.1 infection in diet-induced obese mice through boosting host innate antiviral responses. EBioMedicine 2023; 89:104485. [PMID: 36857860 PMCID: PMC9970285 DOI: 10.1016/j.ebiom.2023.104485] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Obesity is a worldwide epidemic and is considered a risk factor of severe manifestation of Coronavirus Disease 2019 (COVID-19). The pathogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses to infection, re-infection, and vaccination in individuals with obesity remain incompletely understood. METHODS Using the diet-induced obese (DIO) mouse model, we studied SARS-CoV-2 Alpha- and Omicron BA.1-induced disease manifestations and host immune responses to infection, re-infection, and COVID-19 mRNA vaccination. FINDINGS Unlike in lean mice, Omicron BA.1 and Alpha replicated to comparable levels in the lungs of DIO mice and resulted in similar degree of tissue damages. Importantly, both T cell and B cell mediated adaptive immune responses to SARS-CoV-2 infection or COVID-19 mRNA vaccination are impaired in DIO mice, leading to higher propensity of re-infection and lower vaccine efficacy. However, despite the absence of neutralizing antibody, vaccinated DIO mice are protected from lung damage upon Omicron challenge, accompanied with significantly more IFN-α and IFN-β production in the lung tissue. Lung RNAseq and subsequent experiments indicated that COVID-19 mRNA vaccination in DIO mice boosted antiviral innate immune response, including the expression of IFN-α, when compared to the nonvaccinated controls. INTERPRETATION Our findings suggested that COVID-19 mRNA vaccination enhances host innate antiviral responses in obesity which protect the DIO mice to a certain degree when adaptive immunity is suboptimal. FUNDING A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Yanxia Chen
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Wenchen Song
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China
| | - Can Li
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China
| | - Jiaxuan Wang
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Feifei Liu
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Zhanhong Ye
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Peidi Ren
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Yihan Tong
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Junhua Li
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Zhihua Ou
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Andrew Chak-Yiu Lee
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China
| | - Jian-Piao Cai
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Bosco Ho-Yin Wong
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China
| | - Jasper Fuk-Woo Chan
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou, Hainan Province, People's Republic of China; and The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou, Hainan Province, People's Republic of China; and The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Anna Jin-Xia Zhang
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China.
| | - Hin Chu
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China.
| |
Collapse
|
6
|
Chen Y, Li C, Liu F, Ye Z, Song W, Lee ACY, Shuai H, Lu L, To KKW, Chan JFW, Zhang AJ, Chu H, Yuen KY. Age-associated SARS-CoV-2 breakthrough infection and changes in immune response in a mouse model. Emerg Microbes Infect 2022; 11:368-383. [PMID: 34989330 PMCID: PMC8794076 DOI: 10.1080/22221751.2022.2026741] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/05/2022] [Indexed: 02/05/2023]
Abstract
Older individuals are at higher risk of SARS-CoV-2 infection and severe outcomes, but the underlying mechanisms are incompletely understood. In addition, how age modulates SARS-CoV-2 re-infection and vaccine breakthrough infections remain largely unexplored. Here, we investigated age-associated SARS-CoV-2 pathogenesis, immune responses, and the occurrence of re-infection and vaccine breakthrough infection utilizing a wild-type C57BL/6N mouse model. We demonstrated that interferon and adaptive antibody response upon SARS-CoV-2 challenge are significantly impaired in aged mice compared to young mice, which results in more effective virus replications and severe disease manifestations in the respiratory tract. Aged mice also showed increased susceptibility to re-infection due to insufficient immune protection acquired during the primary infection. Importantly, two-dose COVID-19 mRNA vaccination conferred limited adaptive immune response among the aged mice, making them susceptible to SARS-CoV-2 infection. Collectively, our findings call for tailored and optimized treatments and prevention strategies against SARS-CoV-2 among older individuals.
Collapse
Affiliation(s)
- Yanxia Chen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Can Li
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, People’s Republic of China
| | - Feifei Liu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Zhanhong Ye
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Wenchen Song
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, People’s Republic of China
| | - Andrew C. Y. Lee
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, People’s Republic of China
| | - Huiping Shuai
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Lu Lu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
| | - Kelvin Kai-Wang To
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
- Academician Workstation of Hainan Province and Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou, People’s Republic of China
| | - Anna Jinxia Zhang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, People’s Republic of China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, People’s Republic of China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
- Academician Workstation of Hainan Province and Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou, People’s Republic of China
| |
Collapse
|
7
|
Garnica M, Aiello A, Ligotti ME, Accardi G, Arasanz H, Bocanegra A, Blanco E, Calabrò A, Chocarro L, Echaide M, Kochan G, Fernandez-Rubio L, Ramos P, Pojero F, Zareian N, Piñeiro-Hermida S, Farzaneh F, Candore G, Caruso C, Escors D. How Can We Improve the Vaccination Response in Older People? Part II: Targeting Immunosenescence of Adaptive Immunity Cells. Int J Mol Sci 2022; 23:9797. [PMID: 36077216 PMCID: PMC9456031 DOI: 10.3390/ijms23179797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
The number of people that are 65 years old or older has been increasing due to the improvement in medicine and public health. However, this trend is not accompanied by an increase in quality of life, and this population is vulnerable to most illnesses, especially to infectious diseases. Vaccination is the best strategy to prevent this fact, but older people present a less efficient response, as their immune system is weaker due mainly to a phenomenon known as immunosenescence. The adaptive immune system is constituted by two types of lymphocytes, T and B cells, and the function and fitness of these cell populations are affected during ageing. Here, we review the impact of ageing on T and B cells and discuss the approaches that have been described or proposed to modulate and reverse the decline of the ageing adaptive immune system.
Collapse
Affiliation(s)
- Maider Garnica
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Hugo Arasanz
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Ana Bocanegra
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Ester Blanco
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
- Division of Gene Therapy and Regulation of Gene Expression, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Anna Calabrò
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Luisa Chocarro
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Miriam Echaide
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Grazyna Kochan
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Leticia Fernandez-Rubio
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Pablo Ramos
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Fanny Pojero
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Nahid Zareian
- The Rayne Institute, School of Cancer and Pharmaceutical Sciences, King’s College London, London WC2R 2LS, UK
| | - Sergio Piñeiro-Hermida
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Farzin Farzaneh
- The Rayne Institute, School of Cancer and Pharmaceutical Sciences, King’s College London, London WC2R 2LS, UK
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - David Escors
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
8
|
Wang Z, Yuan Y, Chen C, Zhang C, Huang F, Zhou M, Chen H, Fu ZF, Zhao L. Colloidal Manganese Salt Improves the Efficacy of Rabies Vaccines in Mice, Cats, and Dogs. J Virol 2021; 95:e0141421. [PMID: 34495701 PMCID: PMC8577392 DOI: 10.1128/jvi.01414-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
Rabies, caused by rabies virus (RABV), remains a serious threat to public health in most countries worldwide. At present, the administration of rabies vaccines has been the most effective strategy to control rabies. Herein, we evaluate the effect of colloidal manganese salt (Mn jelly [MnJ]) as an adjuvant of rabies vaccine in mice, cats, and dogs. The results showed that MnJ promoted type I interferon (IFN-I) and cytokine production in vitro and the maturation of dendritic cells (DCs) in vitro and in vivo. Besides, MnJ serving as an adjuvant for rabies vaccines could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, plasma cells (PCs), and RABV-specific antibody-secreting cells (ASCs), consequently improve the immunogenicity of rabies vaccines, and provide better protection against virulent RABV challenge. Similarly, MnJ enhanced the humoral immune response in cats and dogs as well. Collectively, our results suggest that MnJ can facilitate the maturation of DCs during rabies vaccination, which can be a promising adjuvant candidate for rabies vaccines. IMPORTANCE Extending the humoral immune response by using adjuvants is an important strategy for vaccine development. In this study, a novel adjuvant, MnJ, supplemented in rabies vaccines was evaluated in mice, cats, and dogs. Our results in the mouse model revealed that MnJ increased the numbers of mature DCs, Tfh cells, GC B cells, PCs, and RABV-specific ASCs, resulting in enhanced immunogenicity and protection rate of rabies vaccines. We further found that MnJ had the same stimulative effect in cats and dogs. Our study provides the first evidence that MnJ serving as a novel adjuvant of rabies vaccines can boost the immune response in both a mouse and pet model.
Collapse
Affiliation(s)
- Zongmei Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yueming Yuan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chen Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengguang Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fei Huang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
9
|
Nassar A, Ibrahim IM, Amin FG, Magdy M, Elgharib AM, Azzam EB, Nasser F, Yousry K, Shamkh IM, Mahdy SM, Elfiky AA. A Review of Human Coronaviruses' Receptors: The Host-Cell Targets for the Crown Bearing Viruses. Molecules 2021; 26:6455. [PMID: 34770863 PMCID: PMC8587140 DOI: 10.3390/molecules26216455] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/19/2022] Open
Abstract
A novel human coronavirus prompted considerable worry at the end of the year 2019. Now, it represents a significant global health and economic burden. The newly emerged coronavirus disease caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the primary reason for the COVID-19 global pandemic. According to recent global figures, COVID-19 has caused approximately 243.3 million illnesses and 4.9 million deaths. Several human cell receptors are involved in the virus identification of the host cells and entering them. Hence, understanding how the virus binds to host-cell receptors is crucial for developing antiviral treatments and vaccines. The current work aimed to determine the multiple host-cell receptors that bind with SARS-CoV-2 and other human coronaviruses for the purpose of cell entry. Extensive research is needed using neutralizing antibodies, natural chemicals, and therapeutic peptides to target those host-cell receptors in extremely susceptible individuals. More research is needed to map SARS-CoV-2 cell entry pathways in order to identify potential viral inhibitors.
Collapse
Affiliation(s)
- Aaya Nassar
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| | - Ibrahim M. Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| | - Fatma G. Amin
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
- Physics Department, Faculty of Science, Alexandria University, Alexandria 21519, Egypt
| | - Merna Magdy
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| | - Ahmed M. Elgharib
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| | - Eman B. Azzam
- Physics Department, Medical Biophysics Division, Faculty of Science, Helwan University, Cairo 11511, Egypt;
| | - Filopateer Nasser
- Biochemistry Department, Faculty of Science, Cairo University, Giza 12511, Egypt;
| | - Kirllos Yousry
- Faculty of Medicine, Cairo University, Cairo 11511, Egypt;
| | | | - Samah M. Mahdy
- National Museum of Egyptian Civilization, Ain Elsira-Elfustat, Cairo 11511, Egypt;
| | - Abdo A. Elfiky
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| |
Collapse
|
10
|
Zhang R, Hung IFN. Approaches in broadening the neutralizing antibody response of the influenza vaccine. Expert Rev Vaccines 2021; 20:1539-1547. [PMID: 34549677 DOI: 10.1080/14760584.2021.1984887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Influenza vaccine is the mainstay for influenza prevention and elicits immune response and antigen-specific neutralizing antibodies against influenza virus. However, antigenic drift and shift can confer influenza virus to escape from the immune response induced by vaccine, and then reduce the vaccine effectiveness. AREAS COVERED To improve effect and neutralizing antibody response of vaccine for heterologous influenza virus, a literature review of preclinical and clinical studies published before August 2021 and searched in PubMed, which evaluated vaccine effectiveness improved by adjuvants and administration route. EXPERT OPINION The review showed that adjuvant, including imiquimod, GLA, MF59, and AS03, can improve the effectiveness of influenza vaccines by regulating immune system. Subjects receiving influenza vaccine combined with these adjuvants showed enhanced antibody response against homologous and heterologous virus strains compared to those vaccinated without adjuvant. This review also discussed the role of intradermal vaccination. In contrast to intramuscular vaccination, intradermal vaccination elicited a robust and prolonged antibody response against vaccine strains and drifted virus than intramuscular vaccination.
Collapse
Affiliation(s)
- Ruiqi Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
11
|
Li C, Chen YX, Liu FF, Lee ACY, Zhao Y, Ye ZH, Cai JP, Chu H, Zhang RQ, Chan KH, Chiu KHY, Lung DC, Sridhar S, Hung IFN, To KKW, Zhang AJX, Chan JFW, Yuen KY. Absence of Vaccine-enhanced Disease With Unexpected Positive Protection Against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by Inactivated Vaccine Given Within 3 Days of Virus Challenge in Syrian Hamster Model. Clin Infect Dis 2021; 73:e719-e734. [PMID: 33515458 PMCID: PMC7929057 DOI: 10.1093/cid/ciab083] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Mass vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is ongoing amidst widespread transmission during the coronavirus disease-2019 (COVID-19) pandemic. Disease phenotypes of SARS-CoV-2 exposure occurring around the time of vaccine administration have not been described. METHODS Two-dose (14 days apart) vaccination regimen with formalin-inactivated whole virion SARS-CoV-2 in golden Syrian hamster model was established. To investigate the disease phenotypes of a 1-dose regimen given 3 days prior (D-3), 1 (D1) or 2 (D2) days after, or on the day (D0) of virus challenge, we monitored the serial clinical severity, tissue histopathology, virus burden, and antibody response of the vaccinated hamsters. RESULTS The 1-dose vaccinated hamsters had significantly lower clinical disease severity score, body weight loss, lung histology score, nucleocapsid protein expression in lung, infectious virus titers in the lung and nasal turbinate, inflammatory changes in intestines, and a higher serum neutralizing antibody or IgG titer against the spike receptor-binding domain or nucleocapsid protein when compared to unvaccinated controls. These improvements were particularly noticeable in D-3, but also in D0, D1, and even D2 vaccinated hamsters to varying degrees. No increased eosinophilic infiltration was found in the nasal turbinate, lung, and intestine after virus challenge. Significantly higher serum titer of fluorescent foci microneutralization inhibition antibody was detected in D1 and D2 vaccinated hamsters at day 4 post-challenge compared to controls despite undetectable neutralizing antibody titer. CONCLUSIONS Vaccination just before or soon after exposure to SARS-CoV-2 does not worsen disease phenotypes and may even ameliorate infection.
Collapse
Affiliation(s)
- Can Li
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yan-Xia Chen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Fei-Fei Liu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Andrew Chak-Yiu Lee
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yan Zhao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Zhan-Hong Ye
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jian-Piao Cai
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Rui-Qi Zhang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Hung Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Hei-Yeung Chiu
- Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - David Christopher Lung
- Department of Pathology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region, China
| | - Siddharth Sridhar
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Kai-Wang To
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Anna Jin-Xia Zhang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China.,Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China.,Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
12
|
Hung IFN, Yap DYH, Yip TPS, Zhang RR, To KKW, Chan KH, Tang SCW, Lui SL, Levin Y, Kochba E, Lau JYN, Yuen MF, Chan TM, Yuen KY. A Double-blind, Randomized Phase 2 Controlled Trial of Intradermal Hepatitis B Vaccination With a Topical Toll-like Receptor 7 Agonist Imiquimod, in Patients on Dialysis. Clin Infect Dis 2021; 73:e304-e311. [PMID: 32556176 DOI: 10.1093/cid/ciaa804] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/13/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Patients on dialysis are hyporesponsive to the hepatitis B virus vaccines (HBVv). We examined intradermal (ID) HBVv Sci-B-Vac, with topical Toll-like receptor 7 (TLR7) agonist imiquimod pretreatment in dialysis patients. METHODS We enrolled and prospectively followed adult patients on dialysis between January 2016 and September 2018. Eligible patients were randomly allocated (1:1:1) into 1 treatment group, topical imiquimod cream followed by ID HBVv (IMQ + ID); and 2 control groups: topical aqueous cream (placebo) followed by ID HBVv (AQ + ID) or topical aqueous cream followed by intramuscular HBVv (AQ + IM). The primary endpoint was the seroprotection rate (hepatitis B surface antibody ≥10 mIU/mL) at 52 weeks. RESULTS Ninety-four patients were enrolled, among which 57.4% were previous nonresponders. Seroprotection rate was significantly better at week 52 for the IMQ + ID group with 96.9% compared to 74.2% and 48.4% for AQ + ID and AQ + IM groups, respectively (P < .0001). The geometric mean concentration was significantly higher at week 52 for the IMQ + ID group: 1135 (95% confidence interval [CI], 579.4-2218.2) mIU/mL, compared to 86.9 (95% CI, 18.5-409.3) mIU/mL and 7.2 (2.0-26.5) mIU/mL for the AQ + ID and AQ + IM groups, respectively (P < .0001). IMQ + ID vaccination (odds ratio, 3.70 [95% CI, 1.16-11.81]; P = .027) was the only factor independently associated with higher 52-week seroprotection rate. Adverse reaction was infrequent. CONCLUSIONS Pretreatment with topical imiquimod before ID HBVv Sci-B-Vac was safe with favorable seroprotection in dialysis patients. CLINICAL TRIALS REGISTRATION NCT02621112.
Collapse
Affiliation(s)
- Ivan Fan-Ngai Hung
- Division of Infectious Diseases, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China.,Carol Yu's Centre for Infection and Division of Infectious Diseases, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Desmond Yat-Hin Yap
- Division of Nephrology, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Terence Pok-Siu Yip
- Division of Nephrology, Department of Medicine, Tung Wah Hospital, Hong Kong Special Administrative Region, China
| | - Ricky Ruiqi Zhang
- Division of Infectious Diseases, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China.,Carol Yu's Centre for Infection and Division of Infectious Diseases, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Kai-Wang To
- Carol Yu's Centre for Infection and Division of Infectious Diseases, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Hung Chan
- Carol Yu's Centre for Infection and Division of Infectious Diseases, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Sydney Chi-Wai Tang
- Division of Nephrology, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Sing-Leung Lui
- Division of Nephrology, Department of Medicine, Tung Wah Hospital, Hong Kong Special Administrative Region, China
| | | | | | - Johnson Yiu-Nam Lau
- Carol Yu's Centre for Infection and Division of Infectious Diseases, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Man-Fung Yuen
- Division of Infectious Diseases, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Tak-Mao Chan
- Division of Nephrology, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- Carol Yu's Centre for Infection and Division of Infectious Diseases, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
13
|
Lee ACY, Zhang AJ, Li C, Chen Y, Liu F, Zhao Y, Chu H, Fong CHY, Wang P, Lau SY, To KKW, Chen H, Yuen KY. Intradermal vaccination of live attenuated influenza vaccine protects mice against homologous and heterologous influenza challenges. NPJ Vaccines 2021; 6:95. [PMID: 34349128 PMCID: PMC8339132 DOI: 10.1038/s41541-021-00359-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 07/12/2021] [Indexed: 12/31/2022] Open
Abstract
We previously developed a temperature-sensitive, and NS1 gene deleted live attenuated influenza vaccine (DelNS1-LAIV) and demonstrated its potent protective efficacy in intranasally vaccinated mice. Here we investigated whether intradermal (i.d.) vaccination induces protective immunity. Our results showed that DelNS1-LAIV intradermal vaccination conferred effective and long-lasting protection against lethal virus challenge in mice. A single intradermal injection of DelNS1-LAIV conferred 100% survival with no weight loss in mice after A(H1N1)09 influenza virus (H1N1/415742Md) challenge. DelNS1-LAIV injection resulted in a significant reduction of lung viral load and reduced airway epithelial cell death and lung inflammatory cytokine responses at day 2 and 4 post challenge. Full protections of mice lasted for 6 months after immunization. In vitro infection of DelNS1-LAIV in monocyte-derived dendritic cells (MoDCs) demonstrated activation of antigen-presenting cells at 33 °C, together with the results of abortive replication of DelNS1-LAIV in skin tissue and strong upregulation of inflammatory cytokines/chemokines expression, our results suggested the strong immunogenicity of this vaccine. Further, we demonstrate that the underlying protection mechanism induced by intradermal DelNS1-LAIV is mainly attributed to antibody responses. Together, this study opens up an alternative route for the administration of LAIV, which may benefit individuals not suitable for intranasal LAIV immunization.
Collapse
Affiliation(s)
- Andrew Chak-Yiu Lee
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anna Jinxia Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Can Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yanxia Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Feifei Liu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yan Zhao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hin Chu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Carol Ho-Yan Fong
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Pui Wang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Siu-Ying Lau
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kelvin Kai-Wang To
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Honglin Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China. .,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China. .,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
Mombelli M, Hoschler K, Cavassini M, Pascual M, Manuel O. Seasonal trivalent inactivated influenza vaccine with topical imiquimod in immunocompromised patients: A randomized controlled trial. J Infect 2021; 83:354-360. [PMID: 34298035 DOI: 10.1016/j.jinf.2021.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/31/2021] [Accepted: 07/07/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND The effect of the Toll-like receptor 7 agonist imiquimod before intradermal (ID) or intramuscular (IM) influenza vaccine in immunocompromised hosts is unknown. METHODS In this open-label randomized controlled trial, kidney transplant recipients (KT) and people living with HIV (PLWH) were randomized to receive IM trivalent inactivated influenza vaccine alone (IM), IM vaccine after topical imiquimod (imi+IM) or ID vaccine after topical imiquimod (imi+ID). Immunogenicity was assessed by hemagglutination inhibition assay. The primary outcome was vaccine response, defined as seroconversion to at least one viral strain at day 21. RESULTS Seventy patients (35 KT and 35 PLWH) received IM (24), imi+IM (22), or imi+ID (24) vaccine. Vaccine response was 61% (14/23) with IM, 59% (13/22) with imi+IM, and 65% (15/23) with imi+ID vaccine (P = 0.909). Vaccine response was associated with HIV infection compared to kidney transplantation (adjusted-OR 3.74, 95% CI 1.25 - 11.23, P = 0.019), but not with imiquimod application nor ID injection. After vaccination, seroprotection to all viral strains was 79% (19/24) with IM, 68% (15/22) with imi+IM, and 70% (16/23) with imi+ID (P = 0.657). We did not observe any vaccine-related severe adverse event. CONCLUSIONS In our study, topical imiquimod did not improve the immunogenicity of influenza vaccine in KT and in PLWH.
Collapse
Affiliation(s)
- Matteo Mombelli
- Transplantation Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; Service of Infectious Diseases, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Katja Hoschler
- Public Health England, Microbiology Services Colindale, London, United Kingdom
| | - Matthias Cavassini
- Service of Infectious Diseases, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Manuel Pascual
- Transplantation Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Oriol Manuel
- Transplantation Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; Service of Infectious Diseases, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Gadanec LK, McSweeney KR, Qaradakhi T, Ali B, Zulli A, Apostolopoulos V. Can SARS-CoV-2 Virus Use Multiple Receptors to Enter Host Cells? Int J Mol Sci 2021; 22:992. [PMID: 33498183 PMCID: PMC7863934 DOI: 10.3390/ijms22030992] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
The occurrence of the novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), responsible for coronavirus disease 2019 (COVD-19), represents a catastrophic threat to global health. Protruding from the viral surface is a densely glycosylated spike (S) protein, which engages angiotensin-converting enzyme 2 (ACE2) to mediate host cell entry. However, studies have reported viral susceptibility in intra- and extrapulmonary immune and non-immune cells lacking ACE2, suggesting that the S protein may exploit additional receptors for infection. Studies have demonstrated interactions between S protein and innate immune system, including C-lectin type receptors (CLR), toll-like receptors (TLR) and neuropilin-1 (NRP1), and the non-immune receptor glucose regulated protein 78 (GRP78). Recognition of carbohydrate moieties clustered on the surface of the S protein may drive receptor-dependent internalization, accentuate severe immunopathological inflammation, and allow for systemic spread of infection, independent of ACE2. Furthermore, targeting TLRs, CLRs, and other receptors (Ezrin and dipeptidyl peptidase-4) that do not directly engage SARS-CoV-2 S protein, but may contribute to augmented anti-viral immunity and viral clearance, may represent therapeutic targets against COVID-19.
Collapse
|
16
|
Lu L, Fong CHY, Zhang AJ, Wu WL, Li IC, Lee ACY, Dissanayake TK, Chen L, Hung IFN, Chan KH, Chu H, Kok KH, Yuen KY, To KKW. Repurposing of Miltefosine as an Adjuvant for Influenza Vaccine. Vaccines (Basel) 2020; 8:vaccines8040754. [PMID: 33322574 PMCID: PMC7768360 DOI: 10.3390/vaccines8040754] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
We previously reported that topical imiquimod can improve the immunogenicity of the influenza vaccine. This study investigated another FDA-approved drug, miltefosine (MTF), as a vaccine adjuvant. Mice immunized with an influenza vaccine with or without MTF adjuvant were challenged by a lethal dose of influenza virus 3 or 7 days after vaccination. Survival, body weight, antibody response, histopathological changes, viral loads, cytokine levels, and T cell frequencies were compared. The MTF-adjuvanted vaccine (MTF-VAC) group had a significantly better survival rate than the vaccine-only (VAC) group, when administered 3 days (80% vs. 26.7%, p = 0.0063) or 7 days (96% vs. 65%, p = 0.0041) before influenza virus challenge. Lung damage was significantly ameliorated in the MTF-VAC group. Antibody response was significantly augmented in the MTF-VAC group against both homologous and heterologous influenza strains. There was a greater T follicular helper cell (TFH) response and an enhanced germinal center (GC) reaction in the MTF-VAC group. MTF-VAC also induced both TH1 and TH2 antigen-specific cytokine responses. MTF improved the efficacy of the influenza vaccine against homologous and heterologous viruses by improving the TFH and antibody responses. Miltefosine may also be used for other vaccines, including the upcoming vaccines for COVID-19.
Collapse
Affiliation(s)
- Lu Lu
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Carol Ho-Yan Fong
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Anna Jinxia Zhang
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Wai-Lan Wu
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Iris Can Li
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Andrew Chak-Yiu Lee
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Thrimendra Kaushika Dissanayake
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Linlei Chen
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China;
| | - Kwok-Hung Chan
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Hin Chu
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Kin-Hang Kok
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China;
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; (L.L.); (C.H.-Y.F.); (A.J.Z.); (W.-L.W.); (I.C.L.); (A.C.-Y.L.); (T.K.D.); (L.C.); (K.-H.C.); (H.C.); (K.-H.K.); (K.-Y.Y.)
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China;
- Correspondence:
| |
Collapse
|
17
|
Lianhua-Qingwen Displays Antiviral and Anti-Inflammatory Activity and Synergistic Effects with Oseltamivir against Influenza B Virus Infection in the Mouse Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3196375. [PMID: 32565852 PMCID: PMC7293728 DOI: 10.1155/2020/3196375] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022]
Abstract
Influenza B virus (IBV) is one of the main pathogens of the annual influenza epidemic, and the disease burden is significant, especially among children and young teenagers. In this study, the antiviral and anti-inflammatory effects of a traditional Chinese medicine prescription, the Lianhua-Qingwen capsule, were evaluated. Our results showed that Lianhua-Qingwen capsule can inhibit both Victoria and Yamagata lineages, and the 50% inhibitive concentrations ranged from 0.228 ± 0.150 to 0.754 ± 0.161 mg/mL. The time course results demonstrated that IBV yields were reduced with treatment at 0–4 h after infection, and the mechanistic research verified that Lianhua-Qingwen capsule has hemagglutination inhibition activity against B/Guangzhou/0215/2012 but not A/California/04/2009. In addition to antiviral activity, Lianhua-Qingwen capsule can also inhibit excessive expression of RANTES, IL-6, IL-8, IP-10, TNF-α, MCP-1, MIP-1β, and IFN-λ at the mRNA level and prevent a severe inflammatory response. The in vivo results confirmed that orally administered Lianhua-Qingwen capsule (100–400 mg/kg/day) does not reduce IBV-induced lung viral load and mortality in mice. However, the pathological change in lungs was alleviated, and there were fewer inflammatory cells in the lungs of Lianhua-Qingwen capsule treated mice than those in controls. Further research confirmed that the combination treatment of 200 mg/kg/day of Lianhua-Qingwen capsule with 2 mg/kg/day of oseltamivir significantly reduced IBV infection over the individual administration of either alone in vivo. Our findings prove that Lianhua-Qingwen capsule could be used as an assistant medicine to enhance the effect of oseltamivir against influenza B virus infection.
Collapse
|
18
|
Kaushik D, Dhingra S, Patil MT, Piplani S, Khanna V, Honda-Okubo Y, Li L, Fung J, Sakala IG, Salunke DB, Petrovsky N. BBIQ, a pure TLR7 agonist, is an effective influenza vaccine adjuvant. Hum Vaccin Immunother 2020; 16:1989-1996. [PMID: 32298200 PMCID: PMC7482670 DOI: 10.1080/21645515.2019.1710409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Better adjuvants are needed for vaccines against seasonal influenza. TLR7 agonists are potent activators of innate immune responses and thereby may be promising adjuvants. Among the imidazoquinoline compounds, 1-benzyl-2-butyl-1H-imidazo[4,5-c]quinolin-4-amine (BBIQ) was reported to be a highly active TLR7 agonist but has remained relatively unexplored because of its commercial unavailability. Indeed, in silico molecular modeling studies predicted that BBIQ had a higher TLR7 docking score and binding free energy than imiquimod, the gold standard TLR7 agonist. To circumvent the availability issue, we developed an improved and higher yield method to synthesize BBIQ. Testing BBIQ on human and mouse TLR7 reporter cell lines confirmed it to be TLR7 specific with significantly higher potency than imiquimod. To test its adjuvant potential, BBIQ or imiquimod were admixed with recombinant influenza hemagglutinin protein and administered to mice as two intramuscular immunizations 2 weeks apart. Serum anti-influenza IgG responses assessed by ELISA 2 weeks after the second immunization confirmed that the mice that received vaccine admixed with BBIQ had significantly higher anti-influenza IgG1 and IgG2c responses than mice immunized with antigen alone or admixed with imiquimod. This confirmed BBIQ to be a TLR7-specific adjuvant able to enhance humoral immune responses.
Collapse
Affiliation(s)
- Deepender Kaushik
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University , Chandigarh, India
| | - Simran Dhingra
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University , Chandigarh, India
| | - Madhuri T Patil
- Department of Chemistry, Mehr Chand Mahajan DAV College for Women , Chandigarh, India
| | - Sakshi Piplani
- Vaxine Pty Ltd , Warradale, Australia.,College of Medicine and Public Health, Flinders University , Adelaide, Australia
| | - Varun Khanna
- Vaxine Pty Ltd , Warradale, Australia.,College of Medicine and Public Health, Flinders University , Adelaide, Australia
| | - Yoshikazu Honda-Okubo
- Vaxine Pty Ltd , Warradale, Australia.,College of Medicine and Public Health, Flinders University , Adelaide, Australia
| | - Lei Li
- Vaxine Pty Ltd , Warradale, Australia.,College of Medicine and Public Health, Flinders University , Adelaide, Australia
| | | | - Isaac G Sakala
- Vaxine Pty Ltd , Warradale, Australia.,College of Medicine and Public Health, Flinders University , Adelaide, Australia
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University , Chandigarh, India.,National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials (NICOVIA), Panjab University , Chandigarh, India
| | - Nikolai Petrovsky
- Vaxine Pty Ltd , Warradale, Australia.,College of Medicine and Public Health, Flinders University , Adelaide, Australia
| |
Collapse
|
19
|
Zhang AJX, Zhu H, Chen Y, Li C, Li C, Chu H, Gozali L, Lee ACY, To KKW, Hung IFN, Yuen KY. Prostaglandin E2-Mediated Impairment of Innate Immune Response to A(H1N1)pdm09 Infection in Diet-Induced Obese Mice Could Be Restored by Paracetamol. J Infect Dis 2019; 219:795-807. [PMID: 30202973 DOI: 10.1093/infdis/jiy527] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/31/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Obesity is associated with increased severity of influenza infection. However, the underlying mechanism is largely unknown. METHODS We employed a mouse model with diet-induced obesity (DIO) to study the innate immune responses induced by influenza virus. RESULTS The lungs of DIO mice were heavily affected by obesity-associated chronic systemic inflammation with a significant increase in inflammatory cytokines/chemokines. Concurrently, lipid immune mediator prostaglandin E2 (PGE2) was also significantly elevated in DIO mice. However, the DIO mice mounted a blunted and delayed upregulation of mRNA and protein concentrations of interferon-β and inflammatory cytokines/chemokines upon A(H1N1)pdm09 virus (H1N1/415742Md) challenge compared with those of lean mice. PGE2 concentrations were significantly higher in the lungs of DIO mice compared to that of lean mice postchallenge. Treatment with paracetamol in challenged DIO mice significantly enhanced the expression of interferon-α/β and cytokine genes at days 1 and 3 postinfection compared with that of untreated DIO mice. Furthermore, paracetamol treatment alone started 3 days before virus challenge and continued until 6 days postchallenge ameliorated the severity of a lethal H1N1/415742Md infection in DIO mice with improved survival. CONCLUSIONS Impaired innate response to influenza in DIO mice is associated with elevated PGE2, which could be restored to some degree by paracetamol treatment.
Collapse
Affiliation(s)
- Anna J X Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Houshun Zhu
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yanxia Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chuangen Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Can Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hin Chu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Leonardi Gozali
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Andrew C Y Lee
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kelvin K W To
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Ivan F N Hung
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence: A systems-level overview of immune cell biology and strategies for improving vaccine responses. Exp Gerontol 2019; 124:110632. [PMID: 31201918 DOI: 10.1016/j.exger.2019.110632] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/30/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023]
Abstract
Immunosenescence contributes to a decreased capacity of the immune system to respond effectively to infections or vaccines in the elderly. The full extent of the biological changes that lead to immunosenescence are unknown, but numerous cell types involved in innate and adaptive immunity exhibit altered phenotypes and function as a result of aging. These manifestations of immunosenescence at the cellular level are mediated by dysregulation at the genetic level, and changes throughout the immune system are, in turn, propagated by numerous cellular interactions. Environmental factors, such as nutrition, also exert significant influence on the immune system during aging. While the mechanisms that govern the onset of immunosenescence are complex, systems biology approaches allow for the identification of individual contributions from each component within the system as a whole. Although there is still much to learn regarding immunosenescence, systems-level studies of vaccine responses have been highly informative and will guide the development of new vaccine candidates, novel adjuvant formulations, and immunotherapeutic drugs to improve vaccine responses among the aging population.
Collapse
Affiliation(s)
- Stephen N Crooke
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA.
| | | | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA.
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
21
|
H7N9 influenza A virus activation of necroptosis in human monocytes links innate and adaptive immune responses. Cell Death Dis 2019; 10:442. [PMID: 31165725 PMCID: PMC6549191 DOI: 10.1038/s41419-019-1684-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/06/2019] [Accepted: 05/23/2019] [Indexed: 12/16/2022]
Abstract
We previously demonstrated that avian influenza A H7N9 virus preferentially infected CD14+ monocyte in human peripheral blood mononuclear cells (PBMCs), which led to apoptosis. To better understand H7N9 pathogenesis in relation to monocyte cell death, we showed here that extensive phosphorylation of mixed lineage kinase domain-like (MLKL) protein occurred concurrently with the activation of caspases-8, -9 and -3 in H7N9-infected monocytes at 6 h post infection (hpi), indicating that apoptosis and necroptosis pathways were simultaneously activated. The apoptotic morphology was readily observed in H7N9-infected monocytes with transmission electron microscopy (TEM), while the pan-caspase inhibitor, IDN6556 (IDN), accelerated cell death through necroptosis as evidenced by the increased level of pMLKL accompanied with cell swelling and plasma membrane rupture. Most importantly, H7N9-induced cell death could only be stopped by the combined treatment of IDN and necrosulfonamide (NSA), a pMLKL membrane translocation inhibitor, but not by individual inhibition of caspase or RIPK3. Our data further showed that activation of apoptosis and necroptosis pathways in monocytes differentially contributed to the immune response of monocytes upon H7N9 infection. Specifically, caspase inhibition significantly enhanced, while RIPK3 inhibition reduced the early expression of type I interferons and cytokine/chemokines in H7N9-infected monocytes. Moreover, culture supernatants from IDN-treated H7N9-infected monocyte promoted the expression of co-stimulatory molecule CD80, CD83 and CD86 on freshly isolated monocytes and monocyte-derived dendritic cells (MDCs) and enhanced the capacity of MDCs to induce CD3+ T-cell proliferation in vitro. In contrast, these immune stimulatory effects were abrogated by using culture supernatants from H7N9-infected monocyte with RIPK3 inhibition. In conclusion, our findings indicated that H7N9 infection activated both apoptosis and necroptosis in monocytes. An intact RIPK3 activity is required for upregulation of innate immune responses, while caspase activation suppresses the immune response.
Collapse
|
22
|
To EE, Erlich J, Liong F, Luong R, Liong S, Bozinovski S, Seow HJ, O'Leary JJ, Brooks DA, Vlahos R, Selemidis S. Intranasal and epicutaneous administration of Toll-like receptor 7 (TLR7) agonists provides protection against influenza A virus-induced morbidity in mice. Sci Rep 2019; 9:2366. [PMID: 30787331 PMCID: PMC6382773 DOI: 10.1038/s41598-019-38864-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/09/2019] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptor 7 (TLR7) is a pattern recognition receptor that recognizes viral RNA following endocytosis of the virus and initiates a powerful immune response characterized by Type I IFN production and pro-inflammatory cytokine production. Despite this immune response, the virus causes very significant pathology, which may be inflammation-dependent. In the present study, we examined the effect of intranasal delivery of the TLR7 agonist, imiquimod or its topical formulation Aldara, on the inflammation and pathogenesis caused by IAV infection. In mice, daily intranasal delivery of imiquimod prevented peak viral replication, bodyweight loss, airway and pulmonary inflammation, and lung neutrophils. Imiquimod treatment also resulted in a significant reduction in pro-inflammatory neutrophil chemotactic cytokines and prevented the increase in viral-induced lung dysfunction. Various antibody isotypes (IgG1, IgG2a, total IgG, IgE and IgM), which were increased in the BALF following influenza A virus infection, were further increased with imiquimod. While epicutaneous application of Aldara had a significant effect on body weight, it did not reduce neutrophil and eosinophil airway infiltration; indicating less effective drug delivery for this formulation. We concluded that intranasal imiquimod facilitates a more effective immune response, which can limit the pathology associated with influenza A virus infection.
Collapse
Affiliation(s)
- Eunice E To
- Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Jonathan Erlich
- Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Felicia Liong
- Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Raymond Luong
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Stella Liong
- Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Steven Bozinovski
- Airways Inflammation Research Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Huei Jiunn Seow
- Respiratory Research Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - John J O'Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland.,Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland.,Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin 8, Ireland.,CERVIVA research consortium, Trinity College Dublin, Dublin, Ireland
| | - Doug A Brooks
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, 5001, Australia
| | - Ross Vlahos
- Respiratory Research Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Stavros Selemidis
- Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia. .,Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
23
|
Caimi AT, Altube MJ, de Farias MA, Portugal RV, Perez AP, Romero EL, Morilla MJ. Novel imiquimod nanovesicles for topical vaccination. Colloids Surf B Biointerfaces 2018; 174:536-543. [PMID: 30500742 DOI: 10.1016/j.colsurfb.2018.11.031] [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: 09/10/2018] [Revised: 11/10/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022]
Abstract
Development of needle and pain free noninvasive immunization procedures is a top priority for public health agencies. In this work the topical adjuvant activity of the immunomodulator imiquimod (IMQ) carried by ultradeformable archaeosomes (UDA2) (nanovesicles containing sn-2,3 ether linked phytanyl saturated archaeolipids) was surveyed and compared with that of ultradeformable liposomes lacking archaeolipids (UDL2) and free IMQ, using the model antigen ovalbumin and a seasonal influenza vaccine in Balb/c mice. UDA2 (250 ± 94 nm, -26 ± 4 mV Z potential) induced higher IMQ accumulation in human skin and higher production of TNF-α and IL-6 by macrophages and keratinocytes than free IMQ and UDL2. Mixed with ovalbumin, UDA2 was more efficient at generating cellular response, as measured by an increase in serum IgG2a and INF-γ production by splenocytes, compared with free IMQ and UDL2. Moreover, mixed with a seasonal influenza vaccine UDA2 produced same IgG titers and IgG2a/IgG1 isotypes ratio (≈1) than the subcutaneously administered influenza vaccine. Topical UDA2 however, induced highest stimulation index and INF-γ levels by splenocytes. UDA2 might be a promising adjuvant for topical immunization, since it produced cell-biased systemic response with ≈ 13-fold lower IMQ dose than the delivered as the commercial IMQ cream, Aldara.
Collapse
Affiliation(s)
- Ayelen Tatiana Caimi
- Nanomedicine Research & Development Center, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Saenz Peña 352, Bernal, B1876BXD, Argentina
| | - Maria Julia Altube
- Nanomedicine Research & Development Center, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Saenz Peña 352, Bernal, B1876BXD, Argentina
| | - Marcelo Alexandre de Farias
- Brazilian Nanotechnology National Laboratory, CNPEM, Caixa Postal 6192, CEP 13.083-970, Campinas, São Paulo, Brazil
| | - Rodrigo Villares Portugal
- Brazilian Nanotechnology National Laboratory, CNPEM, Caixa Postal 6192, CEP 13.083-970, Campinas, São Paulo, Brazil
| | - Ana Paula Perez
- Nanomedicine Research & Development Center, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Saenz Peña 352, Bernal, B1876BXD, Argentina
| | - Eder Lilia Romero
- Nanomedicine Research & Development Center, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Saenz Peña 352, Bernal, B1876BXD, Argentina
| | - Maria Jose Morilla
- Nanomedicine Research & Development Center, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Saenz Peña 352, Bernal, B1876BXD, Argentina.
| |
Collapse
|
24
|
Li C, To KKW, Zhang AJX, Lee ACY, Zhu H, Mak WWN, Hung IFN, Yuen KY. Co-stimulation With TLR7 Agonist Imiquimod and Inactivated Influenza Virus Particles Promotes Mouse B Cell Activation, Differentiation, and Accelerated Antigen Specific Antibody Production. Front Immunol 2018; 9:2370. [PMID: 30369932 PMCID: PMC6194170 DOI: 10.3389/fimmu.2018.02370] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
Current influenza vaccines have relatively low effectiveness, especially against antigenically drifted strains, the effectiveness is even lower in the elderly and immunosuppressed individuals. We have previously shown in a randomized clinical trial that the topical application of a toll-like receptor 7 agonist, imiquimod, just before intradermal influenza vaccine could expedite and augment antibody response, including to antigenically-drifted strains. However, the mechanism of this vaccine and imiquimod combination approach is poorly understood. Here, we demonstrated that imiquimod alone directly activated purified mouse peritoneal B cells. When combined with inactivated H1N1/415742Md influenza virus particle (VP) as vaccine, co-stimulation of mouse peritoneal B cells in vitro induced stronger activation, proliferation, and production of virus-antigen specific IgM and IgG. Intraperitoneal injection of a combination of VP and imiquimod (VCI) was associated with an increased number of activated B cells with enhanced expression of CD86 in the mesenteric draining lymph nodes (mesLN) and the spleen at 18 h after injection. Three days after immunization with VCI, mouse spleen showed significantly more IgM and IgG secreting cells upon in vitro re-stimulation with inactivated virus, mouse sera were detected with viral neutralizing antibody. Transfer of these spleen B cells to naïve mice improved survival after lethal dose of H1N1/415742Md challenge. More importantly, the functional response of VCI-induced B cell activation was demonstrated by early challenge with a lethal dose of H1N1/415742Md influenza virus at 3 days after immunization. The spleen and mediastinal lymph nodes (mdLN) in mice immunized with VCI had germinal center formation, and significantly higher number of plasmablasts, plasma cells, and virus-antigen specific IgM and IgG secreting cells at only 3–4 days post virus challenge, compared with those of mice that have received imiquimod, inactivated virus alone or PBS. Serum virus-specific IgG2a, IgG2b, and IgG1 and bronchoalveolar lavage fluid (BALF) virus-specific IgA at 3 or 4 days post challenge were significantly higher in mice immunized with VCI, which had significantly reduced lung viral load and 100% survival. These findings suggested that imiquimod accelerates the vaccine-induced antibody production via inducing rapid differentiation of naïve B cells into antigen-specific antibody producing cells.
Collapse
Affiliation(s)
- Can Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong.,Carol Yu Centre for Infection, University of Hong Kong, Pokfulam, Hong Kong.,Research Centre of Infection and Immunology, University of Hong Kong, Pokfulam, Hong Kong
| | - Kelvin K W To
- Department of Microbiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong.,Carol Yu Centre for Infection, University of Hong Kong, Pokfulam, Hong Kong.,Research Centre of Infection and Immunology, University of Hong Kong, Pokfulam, Hong Kong
| | - Anna J X Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong.,Carol Yu Centre for Infection, University of Hong Kong, Pokfulam, Hong Kong.,Research Centre of Infection and Immunology, University of Hong Kong, Pokfulam, Hong Kong
| | - Andrew C Y Lee
- Department of Microbiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong.,Carol Yu Centre for Infection, University of Hong Kong, Pokfulam, Hong Kong.,Research Centre of Infection and Immunology, University of Hong Kong, Pokfulam, Hong Kong
| | - Houshun Zhu
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Winger W N Mak
- Department of Microbiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong.,Carol Yu Centre for Infection, University of Hong Kong, Pokfulam, Hong Kong.,Research Centre of Infection and Immunology, University of Hong Kong, Pokfulam, Hong Kong
| | - Ivan F N Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory for Emerging Infectious Diseases, University of Hong Kong, Pokfulam, Hong Kong.,Carol Yu Centre for Infection, University of Hong Kong, Pokfulam, Hong Kong.,Research Centre of Infection and Immunology, University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
25
|
Triple combination of FDA-approved drugs including flufenamic acid, clarithromycin and zanamivir improves survival of severe influenza in mice. Arch Virol 2018; 163:2349-2358. [PMID: 29736671 DOI: 10.1007/s00705-018-3852-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/10/2018] [Indexed: 12/20/2022]
Abstract
Seasonal influenza virus remains a common cause of mortality despite the use of neuraminidase inhibitors. This study evaluated the efficacy of a triple combination of zanamivir, clarithromycin and flufenamic acid (FFA) in the treatment of influenza virus A(H1N1) infection. An in vitro cell protection assay and a multiple-cycle growth assay showed that the antiviral activity of zanamivir was enhanced when combined with clarithromycin or FFA. A mouse challenge model was used here for the evaluation of the in vivo efficacy of the triple combination treatment. We found that mice receiving the triple combination of FFA, zanamivir, and clarithromycin had a significantly better survival rate than those receiving the double combination of zanamivir and clarithromycin (88% versus 44%, P = 0.0083) or zanamivir monotherapy (88% versus 26%, P = 0.0002). Mice in the FFA-zanamivir-clarithromycin triple combination group also exhibited significantly less body weight loss than those in the zanamivir-clarithromycin double combination group. There was no significant difference in the lung viral titers among the different groups from day 2 to day 6 postinfection. However, the levels of IL-1β, TNF-α and RANTES in the FFA-zanamivir-clarithromycin triple combination group were significantly lower than those in the zanamivir-clarithromycin double combination group, zanamivir monotherapy group, or solvent group on day 2 postinfection. Our findings showed that the FFA-zanamivir-clarithromycin triple combination improved the inflammatory markers and survival of severe influenza A(H1N1) infection in mice.
Collapse
|
26
|
Weinberger B. Adjuvant strategies to improve vaccination of the elderly population. Curr Opin Pharmacol 2018; 41:34-41. [PMID: 29677646 DOI: 10.1016/j.coph.2018.03.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/30/2018] [Indexed: 10/17/2022]
Abstract
Immunosenescence contributes to increased incidence and severity of many infections in old age and is responsible for impaired immunogenicity and efficacy of vaccines. Adjuvants are one strategy to enhance immunogenicity of vaccines. The oil-in-water emulsions MF59TM and AS03, as well as a virosomal vaccine have been licensed in seasonal or pandemic influenza vaccines and are/were used successfully in the elderly. AS01, a liposome-based adjuvant comprising two immunostimulants has recently been approved in a recombinant protein vaccine for older adults, which showed very high efficacy against herpes zoster in clinical trials. Several adjuvants for use in the older population are in clinical and preclinical development and will hopefully improve vaccines for this age group in the future.
Collapse
Affiliation(s)
- Birgit Weinberger
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria.
| |
Collapse
|
27
|
Hung IFN, Yuen KY. Immunogenicity, safety and tolerability of intradermal influenza vaccines. Hum Vaccin Immunother 2018; 14:565-570. [PMID: 28604266 PMCID: PMC5861844 DOI: 10.1080/21645515.2017.1328332] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/17/2017] [Accepted: 05/05/2017] [Indexed: 12/26/2022] Open
Abstract
Intradermal influenza vaccination has been studied for more than 80 y. The revived interest in this strategy of vaccination is a result of the innovative technologies in needle design allowing more precise injection and making the device easier to use. Furthermore, clinical trials on these novel devices have demonstrated significant dose sparing effects, improved immunogenicity and very few adverse effects. This review compares intradermal vaccination with various devices with subcutaneous and intramuscular vaccination. We also discussed the role of topical adjuvant before intradermal vaccination.
Collapse
Affiliation(s)
- Ivan F. N. Hung
- Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong Special Administrative Region, China
- Carol Yu Centre for Infection and Division of Infectious Disease, State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, the University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- Carol Yu Centre for Infection and Division of Infectious Disease, State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, the University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
28
|
Van Hoeven N, Fox CB, Granger B, Evers T, Joshi SW, Nana GI, Evans SC, Lin S, Liang H, Liang L, Nakajima R, Felgner PL, Bowen RA, Marlenee N, Hartwig A, Baldwin SL, Coler RN, Tomai M, Elvecrog J, Reed SG, Carter D. A Formulated TLR7/8 Agonist is a Flexible, Highly Potent and Effective Adjuvant for Pandemic Influenza Vaccines. Sci Rep 2017; 7:46426. [PMID: 28429728 PMCID: PMC5399443 DOI: 10.1038/srep46426] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 03/20/2017] [Indexed: 11/30/2022] Open
Abstract
Since 1997, highly pathogenic avian influenza viruses of the H5N1 subtype have been transmitted from avian hosts to humans. The severity of H5N1 infection in humans, as well as the sporadic nature of H5N1 outbreaks, both geographically and temporally, make generation of an effective vaccine a global public health priority. An effective H5N1 vaccine must ultimately provide protection against viruses from diverse clades. Toll-like receptor (TLR) agonist adjuvant formulations have a demonstrated ability to broaden H5N1 vaccine responses in pre-clinical models. However, many of these agonist molecules have proven difficult to develop clinically. Here, we describe comprehensive adjuvant formulation development of the imidazoquinoline TLR-7/8 agonist 3M-052, in combination with H5N1 hemagglutinin (HA) based antigens. We find that 3M-052 in multiple formulations protects both mice and ferrets from lethal H5N1 homologous virus challenge. Furthermore, we conclusively demonstrate the ability of 3M-052 adjuvant formulations to broaden responses to H5N1 HA based antigens, and show that this broadening is functional using a heterologous lethal virus challenge in ferrets. Given the extensive clinical use of imidazoquinoline TLR agonists for other indications, these studies identify multiple adjuvant formulations which may be rapidly advanced into clinical trials in an H5N1 vaccine.
Collapse
Affiliation(s)
- Neal Van Hoeven
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Christopher B Fox
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Brian Granger
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Tara Evers
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Sharvari W Joshi
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Ghislain I Nana
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Sarah C Evans
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Susan Lin
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Hong Liang
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Li Liang
- University of California Irvine, Department of Medicine, Irvine CA 92697, USA
| | - Rie Nakajima
- University of California Irvine, Department of Medicine, Irvine CA 92697, USA
| | - Philip L Felgner
- University of California Irvine, Department of Medicine, Irvine CA 92697, USA
| | - Richard A Bowen
- Colorado State University Department of Biomedical Sciences, Foothills Campus, Fort Collins, CO 80523, USA
| | - Nicole Marlenee
- Colorado State University Department of Biomedical Sciences, Foothills Campus, Fort Collins, CO 80523, USA
| | - Airn Hartwig
- Colorado State University Department of Biomedical Sciences, Foothills Campus, Fort Collins, CO 80523, USA
| | - Susan L Baldwin
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Rhea N Coler
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Mark Tomai
- 3M, Inc., St. Paul, Minnesota 55121, USA
| | | | - Steven G Reed
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| | - Darrick Carter
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle WA 98103, USA
| |
Collapse
|
29
|
Chan JFW, Zhang AJ, Chan CCS, Yip CCY, Mak WWN, Zhu H, Poon VKM, Tee KM, Zhu Z, Cai JP, Tsang JOL, Chik KKH, Yin F, Chan KH, Kok KH, Jin DY, Au-Yeung RKH, Yuen KY. Zika Virus Infection in Dexamethasone-immunosuppressed Mice Demonstrating Disseminated Infection with Multi-organ Involvement Including Orchitis Effectively Treated by Recombinant Type I Interferons. EBioMedicine 2016; 14:112-122. [PMID: 27884655 PMCID: PMC5161441 DOI: 10.1016/j.ebiom.2016.11.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/10/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Disseminated or fatal Zika virus (ZIKV) infections were reported in immunosuppressed patients. Existing interferon-signaling/receptor-deficient mouse models may not be suitable for evaluating treatment effects of recombinant interferons. METHODS We developed a novel mouse model for ZIKV infection by immunosuppressing BALB/c mice with dexamethasone. RESULTS Dexamethasone-immunosuppressed male mice (6-8weeks) developed disseminated infection as evidenced by the detection of ZIKV-NS1 protein expression and high viral loads in multiple organs. They had ≥10% weight loss and high clinical scores soon after dexamethasone withdrawal (10dpi), which warranted euthanasia at 12dpi. Viral loads in blood and most tissues at 5dpi were significantly higher than those at 12dpi (P<0.05). Histological examination revealed prominent inflammatory infiltrates in multiple organs, and CD45+ and CD8+ inflammatory cells were seen in the testis. These findings suggested that clinical deterioration occurred during viral clearance by host immune response. Type I interferon treatments improved clinical outcome of mice (100% vs 0% survival). CONCLUSIONS Besides virus dissemination, inflammation of various tissues, especially orchitis, may be potential complications of ZIKV infection with significant implications on disease transmission and male fertility. Interferon treatment should be considered in patients at high risks for ZIKV-associated complications when the potential benefits outweigh the side effects of treatment.
Collapse
Affiliation(s)
- Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Anna Jinxia Zhang
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chris Chung-Sing Chan
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Cyril Chik-Yan Yip
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Winger Wing-Nga Mak
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Houshun Zhu
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Vincent Kwok-Man Poon
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kah-Meng Tee
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Zheng Zhu
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jian-Piao Cai
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jessica Oi-Ling Tsang
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kenn Ka-Heng Chik
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Feifei Yin
- Department of Pathogen Biology, Hainan Medical University, Haikou, Hainan 571101, China
| | - Kwok-Hung Chan
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kin-Hang Kok
- Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Rex Kwok-Him Au-Yeung
- Department of Pathology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Microbiology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong Special Administrative Region, China; The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
30
|
Solid nanoemulsion as antigen and immunopotentiator carrier for transcutaneous immunization. Cell Immunol 2016; 308:35-43. [DOI: 10.1016/j.cellimm.2016.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 05/06/2016] [Accepted: 06/01/2016] [Indexed: 01/17/2023]
|
31
|
Shin WJ, Noh HJ, Noh YW, Kim S, Um SH, Lim YT. Hyaluronic acid-supported combination of water insoluble immunostimulatory compounds for anti-cancer immunotherapy. Carbohydr Polym 2016; 155:1-10. [PMID: 27702491 DOI: 10.1016/j.carbpol.2016.08.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/01/2016] [Accepted: 08/13/2016] [Indexed: 01/27/2023]
Abstract
A novel powder-form combination adjuvant system containing two immunostimulatory compounds was firstly developed and evaluated as a therapeutic intervention for cancer immunotherapy. With the help of hyaluronic acid (HA), water insoluble monophosphoryl lipid A (MPL), QS21 and imiquimod (R837), could be easily dispersed in aqueous solution and lyophilized as powder-form, which have an advantage in room-temperature storage stability compared with those conventional liquid formulation that requires cold storage. Two kinds of HA-based combination vaccine adjuvants (HA/MPL/QS21, HMQ and HA/MPL/R837, HMR) contributed to the increase of both humoral and cellular immunity, which is very important for efficient cancer immunotherapy. Through the challenge experiments in EG7-OVA (mouse lymphoma-expressing OVA) tumor-bearing mice model, we found out that the immunostimulatory effects of HMQ and HMR were successful in the inhibition of tumor proliferation. Taken together, both HA-based powder-form combination adjuvant systems are expected to be used as potent prophylactic and therapeutic cancer vaccine.
Collapse
Affiliation(s)
- Woo Jung Shin
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyun Jong Noh
- Nanomedical Systems Laboratory, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Young-Woock Noh
- Nanomedical Systems Laboratory, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sohyun Kim
- Nanomedical Systems Laboratory, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yong Taik Lim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Nanomedical Systems Laboratory, SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
32
|
Liu R, Wang J, Yang Y, Khan I, Zhu N. Rabies virus lipopeptide conjugated to a TLR7 agonist improves the magnitude and quality of the Th1-biased humoral immune response in mice. Virology 2016; 497:102-110. [PMID: 27449478 DOI: 10.1016/j.virol.2016.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/06/2016] [Accepted: 06/23/2016] [Indexed: 12/17/2022]
Abstract
In this study, we conjugated the rabies-derived lipopeptide CE536 to a TLR7 agonist, imiquimod, and evaluated its adjuvanticity. The synthetic construct (Lipo-I) targeted to TLR7, induced dendritic cell phenotypic maturation and production of both type I interferon and pro-inflammatory cytokines more efficiently than unconjugated TLR7 ligands or lipopeptide alone. The immunostimulatory effects of the conjugate were apparently the result of IκBα degradation and sustained p38 and JNK phosphorylation. The analysis of IgG isotypes and T cell differentiation showed that IgG2a dominant Th1-biased humoral and CD8(+) IFN-γ T cell responses were induced by Lipo-I. Lipo-I could facilitate the rabies vaccine to induce the production of an earlier and more vigorous rabies virus neutralizing antibody. In the post-exposure test, the Lipo-I adjuvanted vaccine provided a 73.3% survival rate, while the traditional vaccine bestowed only a 26.7% survival. Therefore, Lipo-I is a promising adjuvant for the development of more effective rabies vaccines.
Collapse
Affiliation(s)
- Rui Liu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Jingbo Wang
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yan Yang
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Inamullah Khan
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Naishuo Zhu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
33
|
Deng L, Pang P, Zheng K, Nie J, Xu H, Wu S, Chen J, Chen X. Forsythoside A Controls Influenza A Virus Infection and Improves the Prognosis by Inhibiting Virus Replication in Mice. Molecules 2016; 21:molecules21050524. [PMID: 27128889 PMCID: PMC6273045 DOI: 10.3390/molecules21050524] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/08/2016] [Accepted: 04/16/2016] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE The objective of this study was to observe the effects of forsythoside A on controlling influenza A virus (IAV) infection and improving the prognosis of IAV infection. METHODS Forty-eight SPF C57BL/6j mice were randomly divided into the following four groups: Group A: normal control group (normal con); Group B: IAV control group (V con); Group C: IAV+ oseltamivir treatment group (V oseltamivir; 0.78 mg/mL, 0.2 mL/mouse/day); Group D: IAV+ forsythoside A treatment group (V FTA; 2 μg/mL, 0.2 mL/mouse/day). Real-time fluorescence quantitative PCR (RT-qPCR) was used to measure mRNA expression of the TLR7, MyD88, TRAF6, IRAK4 and NF-κB p65 mRNA in TLR7 signaling pathway and the virus replication level in lung. Western blot was used to measure TLR7, MyD88 and NF-κB p65 protein. Flow cytometry was used to detect the proportion of the T cell subsets Th1/Th2 and Th17/Treg. RESULTS The body weight began to decrease after IAV infection, while FTA and oseltamivir could reduce the rate of body weight loss. The pathological damages in the FTA and oseltamivir group were less serious. TLR7, MyD88, TRAF6, IRAK4 and NF-κB p65 mRNA were up-regulated after virus infection (p < 0.01) while down-regulated after oseltamivir and FTA treatment (p < 0.01). The results of TLR7, MyD88 and NF-κB p65 protein consisted with correlative mRNA. Flow cytometry showed the Th1/Th2 differentiated towards Th2, and the Th17/Treg cells differentiated towards Treg after FTA treatment. CONCLUSIONS Our study suggests forsythoside A can control influenza A virus infection and improve the prognosis of IAV infection by inhibiting influenza A virus replication.
Collapse
Affiliation(s)
- Li Deng
- Department of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Peng Pang
- Department of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Ke Zheng
- Department of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Jiao Nie
- Department of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Huachong Xu
- Department of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Sizhi Wu
- Department of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Jia Chen
- Department of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Xiaoyin Chen
- Department of Traditional Chinese Medicine, School of Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
34
|
Lebold KM, Jacoby DB, Drake MG. Toll-Like Receptor 7-Targeted Therapy in Respiratory Disease. Transfus Med Hemother 2016; 43:114-9. [PMID: 27226793 DOI: 10.1159/000445324] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/09/2016] [Indexed: 01/01/2023] Open
Abstract
Allergic asthma and allergic rhinitis are inflammatory diseases of the respiratory tract characterized by an excessive type-2 T helper cell (Th2) immune response. Toll-like receptor 7 (TLR7) is a single-stranded viral RNA receptor expressed in the airway that initiates a Th1 immune response and has garnered interest as a novel therapeutic target for treatment of allergic airway diseases. In animal models, synthetic TLR7 agonists reduce airway hyperreactivity, eosinophilic inflammation, and airway remodeling while decreasing Th2-associated cytokines. Furthermore, activation of TLR7 rapidly relaxes airway smooth muscle via production of nitric oxide. Thus, TLR7 has dual bronchodilator and anti-inflammatory effects. Two TLR7 ligands with promising pharmacologic profiles have entered clinical trials for the treatment of allergic rhinitis. Moreover, TLR7 agonists are potential antiviral therapies against respiratory viruses. TLR7 agonists enhance influenza vaccine efficacy and also reduce viral titers when given during an active airway infection. In this review, we examine the current data supporting TLR7 as a therapeutic target in allergic airway diseases.
Collapse
Affiliation(s)
- Katie M Lebold
- Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, OR, USA
| | - David B Jacoby
- Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Matthew G Drake
- Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
35
|
Zang F, Chen Y, Lin Z, Cai Z, Yu L, Xu F, Wang J, Zhu W, Lu H. Autophagy is involved in regulating the immune response of dendritic cells to influenza A (H1N1) pdm09 infection. Immunology 2016; 148:56-69. [PMID: 26800655 DOI: 10.1111/imm.12587] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 01/11/2016] [Accepted: 01/18/2016] [Indexed: 12/14/2022] Open
Abstract
Autophagy can mediate antiviral immunity. However, it remains unknown whether autophagy regulates the immune response of dendritic cells (DCs) to influenza A (H1N1) pdm09 infection. In this study, we found that infection with the H1N1 virus induced DC autophagy in an endocytosis-dependent manner. Compared with autophagy-deficient Beclin-1(+/-) mice, we found that bone-marrow-derived DCs from wild-type mice (WT BMDCs) presented a more mature phenotype on H1N1 infection. Wild-type BMDCs secreted higher levels of interleukin-6 (IL-6), tumour necrosis factor- α (TNF-α), interferon-β (IFN-β), IL-12p70 and IFN-γ than did Beclin-1(+/-) BMDCs. In contrast to Beclin-1(+/-) BMDCs, H1N1-infected WT BMDCs exhibited increased activation of extracellular signal-regulated kinase, Jun N-terminal kinase, p38, and nuclear factor-κB as well as IFN regulatory factor 7 nuclear translocation. Blockade of autophagosomal and lysosomal fusion by bafilomycin A1 decreased the co-localization of H1N1 viruses, autophagosomes and lysosomes as well as the secretion of IL-6, TNF-α and IFN-β in H1N1-infected BMDCs. In contrast to Beclin-1(+/-) BMDCs, H1N1-infected WT BMDCs were more efficient in inducing allogeneic CD4(+) T-cell proliferation and driving T helper type 1, 2 and 17 cell differentiation while inhibiting CD4(+) Foxp3(+) regulatory T-cell differentiation. Moreover, WT BMDCs were more efficient at cross-presenting the ovalbumin antigen to CD8(+) T cells. We consistently found that Beclin-1(+/-) BMDCs were inferior in their inhibition of H1N1 virus replication and their induction of H1N1-specific CD4(+) and CD8(+) T-cell responses, which produced lower levels of IL-6, TNF-α and IFN-β in vivo. Our data indicate that autophagy is important in the regulation of the DC immune response to H1N1 infection, thereby extending our understanding of host immune responses to the virus.
Collapse
Affiliation(s)
- Farong Zang
- Department of Respiration, The People's Hospital of Changxing County, Huzhou, China
| | - Yinghu Chen
- Division of Infection Disease, Zhejiang Key Laboratory for Neonatal Diseases, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhendong Lin
- Departments of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijian Cai
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaoli Wang
- Department of Respiratory Medicine, Affiliated Hangzhou Hospital (Hangzhou First People's Hospital), Nanjing Medical University, Hangzhou, China
| | - Weiguo Zhu
- Institute of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huoquan Lu
- Department of Respiration, The People's Hospital of Changxing County, Huzhou, China
| |
Collapse
|
36
|
Hung IFN, Zhang AJ, To KKW, Chan JFW, Li P, Wong TL, Zhang R, Chan TC, Chan BCY, Wai HH, Chan LW, Fong HPY, Hui RKC, Kong KL, Leung ACF, Ngan AHT, Tsang LWK, Yeung APC, Yiu GCN, Yung W, Lau JYN, Chen H, Chan KH, Yuen KY. Topical imiquimod before intradermal trivalent influenza vaccine for protection against heterologous non-vaccine and antigenically drifted viruses: a single-centre, double-blind, randomised, controlled phase 2b/3 trial. THE LANCET. INFECTIOUS DISEASES 2016; 16:209-18. [DOI: 10.1016/s1473-3099(15)00354-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 10/22/2022]
|
37
|
Stein P, Radsak MP. The skin as an orchestrator of influenza immunity. THE LANCET. INFECTIOUS DISEASES 2016; 16:139-140. [PMID: 26559481 DOI: 10.1016/s1473-3099(15)00413-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 10/20/2015] [Indexed: 06/05/2023]
Affiliation(s)
- Pamela Stein
- University Medical Center Mainz, Mainz 55131, Germany
| | | |
Collapse
|
38
|
Suboptimal Humoral Immune Response against Influenza A(H7N9) Virus Is Related to Its Internal Genes. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:1235-43. [PMID: 26446420 DOI: 10.1128/cvi.00443-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/28/2015] [Indexed: 11/20/2022]
Abstract
Influenza A(H7N9) virus pneumonia is associated with a high case fatality rate in humans. Multiple viral factors have been postulated to account for the high virulence of the virus. It has been reported that patients with influenza A(H7N9) virus infection have relatively low titers of neutralizing antibodies compared to those with seasonal influenza virus infections. In this study, we compared serum hemagglutination inhibition (HI) and microneutralization (MN) antibody titers of mice challenged with wild-type A(H7N9) viruses [H7N9(Anhui) and H7N9(Zhejiang)], an A(H1N1)pdm09 virus [pH1N1(2009)], and a recombinant A(H7N9) virus with PR8/H1N1 internal genes (rg-PR8-H7-N9). All mice infected by H7N9(Anhui) and H7N9(Zhejiang) developed serum HI antibodies at 14 days postinfection (dpi) but no detectable MN antibodies, even at 28 dpi. A low level of neutralizing activity was detected in H7N9(Anhui)- and H7N9(Zhejiang)-infected mice using fluorescent focus MN assay, but convalescent-phase serum samples obtained from H7N9(Anhui)-infected mice did not reduce the mortality of naive mice after homologous virus challenge. Reinfection with homologous A(H7N9) virus induced higher HI and MN titers than first infection. In contrast, pH1N1(2009) virus infection induced robust HI and MN antibody responses, even during the first infection. Moreover, rg-PR8-H7-N9 induced significantly higher HI and MN antibody titers than H7N9(Zhejiang). In conclusion, the internal genes of A(H7N9) virus can affect the humoral immune response against homologous viral surface proteins, which may also contribute to the virulence of A(H7N9) virus.
Collapse
|
39
|
Zhang L, Wang W, Wang S. Effect of vaccine administration modality on immunogenicity and efficacy. Expert Rev Vaccines 2015; 14:1509-23. [PMID: 26313239 DOI: 10.1586/14760584.2015.1081067] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The many factors impacting the efficacy of a vaccine can be broadly divided into three categories: features of the vaccine itself, including immunogen design, vaccine type, formulation, adjuvant and dosing; individual variations among vaccine recipients and vaccine administration-related parameters. While much literature exists related to vaccines, and recently systems biology has started to dissect the impact of individual subject variation on vaccine efficacy, few studies have focused on the role of vaccine administration-related parameters on vaccine efficacy. Parenteral and mucosal vaccinations are traditional approaches for licensed vaccines; novel vaccine delivery approaches, including needless injection and adjuvant formulations, are being developed to further improve vaccine safety and efficacy. This review provides a brief summary of vaccine administration-related factors, including vaccination approach, delivery route and method of administration, to gain a better understanding of their potential impact on the safety and immunogenicity of candidate vaccines.
Collapse
Affiliation(s)
- Lu Zhang
- a 1 Department of Infectious Diseases, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.,b 2 China-US Vaccine Research Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wei Wang
- c 3 Wang Biologics, LLC, Chesterfield, MO 63017, USA ; Current affiliation: Bayer HealthCare, Berkeley, CA 94710, USA
| | - Shixia Wang
- d 4 Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
40
|
Xiong D, Song L, Zhai X, Geng S, Pan Z, Jiao X. A porcine reproductive and respiratory syndrome virus (PRRSV) vaccine candidate based on the fusion protein of PRRSV glycoprotein 5 and the Toll-like Receptor-5 agonist Salmonella Typhimurium FljB. BMC Vet Res 2015; 11:121. [PMID: 26001608 PMCID: PMC4489122 DOI: 10.1186/s12917-015-0439-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/15/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Porcine reproductive and respiratory syndrome (PRRS) is characterized by severe reproductive failure and severe pneumonia in neonatal pigs and is caused by PRRS virus (PRRSV). Glycoprotein 5 (GP5) from PRRSV is a key inducer of neutralizing antibodies. Flagellin, a toll-like receptor 5 (TLR-5) agonist, is an effective inducer of innate immune responses. This study presents a novel PRRSV vaccine candidate based on the adjuvant effect of Salmonella Typhimurium FljB fused with PRRSV GP5. RESULTS A truncated rGP5 gene lacking the signal peptide and transmembrane sequences was amplified and inserted into prokaryotic expression vectors, pColdI or pGEX-6p-1. Salmonella Typhimurium flagellin fljB was amplified and inserted into the plasmid pCold-rGP5, generating recombinant plasmid pCold-rGP5-fljB. Histidine (His)-tagged rGP5 and fusion protein rGP5-FljB were induced with isopropyl-β-d-thiogalactoside, verified by SDS-PAGE and western blotting, and purified via Ni-NTA affinity columns. The TLR-5-specific bioactivity of fusion protein rGP5-FljB was determined by detecting the expression levels of the cytokine IL-8 in HEK293-mTLR5 cells by sandwich ELISA. The purified endotoxin-free proteins were administered intraperitoneally in a C3H/HeJ mouse model. The results show that immunization with the fusion protein rGP5-FljB induced a significantly enhanced GP5-specific and PRRSV-specific IgG response that persisted for almost 5 weeks. Co-administration of the rGP5 with R848 or Alum also yielded a higher IgG response than administration of rGP5 alone. The IgG1/IgG2a ratio in the rGP5-FljB immunization group was significantly higher (9-fold) than that in the rGP5 alone group and was equivalent to the response in the rGP5 + Alum immunization group, suggesting a strong Th2 immune response was induced by the fusion protein. CONCLUSIONS Purified fusion protein rGP5-FljB is capable of activating the innate immune response, as demonstrated by the results of our TLR-5-specific bioactivity assay, and FljB has adjuvant activity, as shown by the results from our administration of rGP5-FljB in a mouse model. Our findings confirm that FljB could serve as an excellent adjuvant for the production of GP5-specific and PRRSV-specific IgG antibodies as part of an induction of a robust humoral immune response.
Collapse
Affiliation(s)
- Dan Xiong
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.
| | - Li Song
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.
| | - Xianyue Zhai
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.
| | - Shizhong Geng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.
| |
Collapse
|
41
|
To KKW, Zhang AJX, Chan ASF, Li C, Cai JP, Lau CCY, Li CG, Jahan AS, Wu WL, Li L, Tsang AKL, Chan KH, Chen H, Yuen KY. Recombinant influenza A virus hemagglutinin HA2 subunit protects mice against influenza A(H7N9) virus infection. Arch Virol 2015; 160:777-86. [PMID: 25616843 DOI: 10.1007/s00705-014-2314-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 12/14/2014] [Indexed: 01/23/2023]
Abstract
A novel avian influenza A(H7N9) virus has emerged to infect humans in eastern China since 2013. An effective vaccine is needed because of the high mortality despite antiviral treatment and intensive care. We sought to develop an effective vaccine for A(H7N9) virus. The HA2 subunit was chosen as the vaccine antigen because it is highly conserved among the human A(H7N9) virus strains. Moreover, in silico analysis predicted two immunogenic regions within the HA2 subunit that may contain potential human B-cell epitopes. The HA2 fragment was readily expressed in Escherichia coli. In BALB/c mice, intraperitoneal immunization with two doses of HA2 with imiquimod (2-dose-imiquimod) elicited the highest geometric mean titer (GMT) of anti-HA2 IgG (12699), which was greater than that of two doses of HA2 without imiquimod (2-dose-no-adjuvant) (6350), one dose of HA2 with imiquimod (1-dose-imiquimod) (2000) and one dose of HA2 without imiquimod (1-dose-no-adjuvant) (794). The titer of anti-HA2 IgG was significantly higher in the 1-dose-imiquimod group than the 1-dose-no-adjuvant group. Although both hemagglutination inhibition titers and microneutralization titers were below 10, serum from immunized mice showed neutralizing activity in a fluorescent focus microneutralization assay. In a viral challenge experiment, the 2-dose-imiquimod group had the best survival rate (100 %), followed by the 2-dose-no-adjuvant group (90 %), the 1-dose-imiquimod group (70 %) and the 1-dose-no-adjuvant group (40 %). The 2-dose-imiquimod group also had significantly lower mean pulmonary viral loads than the 1-dose-imiquimod, 1-dose-no-adjuvant and non-immunized groups. This recombinant A(H7N9)-HA2 vaccine should be investigated as a complement to egg- or cell-based live attenuated or subunit influenza vaccines.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Aminoquinolines/administration & dosage
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Disease Models, Animal
- Escherichia coli/genetics
- Gene Expression
- Hemagglutination Inhibition Tests
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Imiquimod
- Immunoglobulin G/blood
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Injections, Intraperitoneal
- Mice, Inbred BALB C
- Neutralization Tests
- Orthomyxoviridae Infections/prevention & control
- Protein Subunits/genetics
- Protein Subunits/immunology
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Survival Analysis
- Vaccination/methods
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Kelvin K W To
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Li C, Li C, Zhang AJX, To KKW, Lee ACY, Zhu H, Wu HWL, Chan JFW, Chen H, Hung IFN, Li L, Yuen KY. Avian influenza A H7N9 virus induces severe pneumonia in mice without prior adaptation and responds to a combination of zanamivir and COX-2 inhibitor. PLoS One 2014; 9:e107966. [PMID: 25232731 PMCID: PMC4169509 DOI: 10.1371/journal.pone.0107966] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/16/2014] [Indexed: 12/26/2022] Open
Abstract
Background Human infection caused by the avian influenza A H7N9 virus has a case-fatality rate of over 30%. Systematic study of the pathogenesis of avian H7N9 isolate and effective therapeutic strategies are needed. Methods BALB/c mice were inoculated intranasally with an H7N9 virus isolated from a chicken in a wet market epidemiologically linked to a fatal human case, (A/chicken/Zhejiang/DTID-ZJU01/2013 [CK1]), and with an H7N9 virus isolated from a human (A/Anhui/01/2013 [AH1]). The pulmonary viral loads, cytokine/chemokine profiles and histopathological changes of the infected mice were compared. The therapeutic efficacy of a non-steroidal anti-inflammatory drug (NSAID), celecoxib, was assessed. Results Without prior adaptation, intranasal inoculation of 106 plaque forming units (PFUs) of CK1 caused a mortality rate of 82% (14/17) in mice. Viral nucleoprotein and RNA expression were limited to the respiratory system and no viral RNA could be detected from brain, liver and kidney tissues. CK1 caused heavy alveolar inflammatory exudation and pulmonary hemorrhage, associated with high pulmonary levels of proinflammatory cytokines. In the mouse lung cell line LA-4, CK1 also induced high levels of interleukin-6 (IL-6) and cyclooxygenase-2 (COX-2) mRNA. Administration of the antiviral zanamivir did not significantly improve survival in mice infected with CK1, but co-administration of the non-steroidal anti-inflammatory drug (NSAID) celecoxib in combination with zanamivir improved survival and lung pathology. Conclusions Our findings suggested that H7N9 viruses isolated from chicken without preceding trans-species adaptation can cause lethal mammalian pulmonary infection. The severe proinflammatory responses might be a factor contributing to the mortality. Treatment with combination of antiviral and NSAID could ameliorate pulmonary inflammation and may improve survival.
Collapse
MESH Headings
- Adaptation, Physiological/immunology
- Animals
- Antiviral Agents/pharmacology
- Antiviral Agents/therapeutic use
- Cell Line
- Cyclooxygenase 2/metabolism
- Cyclooxygenase 2 Inhibitors/pharmacology
- Cyclooxygenase 2 Inhibitors/therapeutic use
- Cytokines/metabolism
- Drug Evaluation, Preclinical
- Drug Synergism
- Drug Therapy, Combination
- Female
- Humans
- Influenza A Virus, H7N9 Subtype/drug effects
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/physiology
- Lung/pathology
- Lung/virology
- Mice
- Mice, Inbred BALB C
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/immunology
- Pneumonia, Viral/virology
- Virus Replication
- Zanamivir/pharmacology
- Zanamivir/therapeutic use
Collapse
Affiliation(s)
- Can Li
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Chuangen Li
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Anna J. X. Zhang
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- * E-mail: (K-YY); (AJXZ)
| | - Kelvin K. W. To
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - Andrew C. Y. Lee
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Houshun Zhu
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hazel W. L. Wu
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Jasper F. W. Chan
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
| | - Honglin Chen
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- Zhejiang University, Hangzhou, China
| | - Ivan F. N. Hung
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- Zhejiang University, Hangzhou, China
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
- Zhejiang University, Hangzhou, China
- * E-mail: (K-YY); (AJXZ)
| |
Collapse
|
43
|
Hung IFN, Zhang AJ, To KKW, Chan JFW, Li C, Zhu HS, Li P, Li C, Chan TC, Cheng VCC, Chan KH, Yuen KY. Immunogenicity of intradermal trivalent influenza vaccine with topical imiquimod: a double blind randomized controlled trial. Clin Infect Dis 2014; 59:1246-55. [PMID: 25048848 DOI: 10.1093/cid/ciu582] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Imiquimod, a synthetic Toll-like receptor 7 agonist enhanced immunogenicity of influenza vaccine in a mouse model. We hypothesized that topical imiquimod before intradermal influenza vaccination (TIV) would produce similar effect in human. METHODS We performed a prospective 1-year follow-up, double-blind, randomized, controlled trial with adults with comorbidities. Participants were randomized to 1 of the following 3 vaccinations: topical 5% 250 mg imiquimod ointment followed by intradermal TIV, topical aqueous-cream followed by intradermal TIV, or topical aqueous-cream followed by intramuscular TIV. Patients and investigators were blinded to the type of topical treatment applied. Hemagglutination inhibition (HI) and microneutralization antibody titers were measured. The primary outcome was the day 7 seroconversion rate. RESULTS Ninety-one recruited participants completed the study. The median age was 73 years. On day 7, 27/30 (90%) patients who received imiquimod and intradermal TIV achieved seroconversion against the H1N1 strain by HI, compared with 4/30 (13.3%) who received aqueous-cream and intramuscular TIV (P < .001), and 12/31 (38.7%) who received aqueous-cream and intradermal TIV (P < .001). The seroconversion, seroprotection, and geometric mean titer-fold increase were met in all 3 strains in the imiquimod and intradermal TIV group 2 weeks earlier, and the better seroconversion rate was sustained from day 7 to year 1 (P ≤ .001). The better immunogenicity was associated with fewer hospitalizations for influenza or pneumonia (P < .05). All adverse reactions were self-limited. CONCLUSIONS Pretreatment with topical imiquimod significantly expedited, augmented, and prolonged the immunogenicity of influenza vaccination. This strategy for influenza immunization should be considered for the elderly population.
Collapse
Affiliation(s)
- Ivan F N Hung
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Anna J Zhang
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Kelvin K W To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Jasper F W Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Can Li
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Hou-Shun Zhu
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Patrick Li
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Clara Li
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Tuen-Ching Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Vincent C C Cheng
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Kwok-Hung Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| |
Collapse
|