1
|
Yan R, Zou C, Yang X, Zhuang W, Huang Y, Zheng X, Hu J, Liao L, Yao Y, Sun X, Hu WW. Nebulized inhalation drug delivery: clinical applications and advancements in research. J Mater Chem B 2025; 13:821-843. [PMID: 39652178 DOI: 10.1039/d4tb01938e] [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: 01/03/2025]
Abstract
Nebulized inhalation administration refers to the dispersion of drugs into small droplets suspended in the gas through a nebulized device, which are deposited in the respiratory tract by inhalation, to achieve the local therapeutic effect of the respiratory tract. Compared with other drug delivery methods, nebulized inhalation has the advantages of fast effect, high local drug concentration, less dosage, convenient application and less systemic adverse reactions, and has become one of the main drug delivery methods for the treatment of respiratory diseases. In this review, we first discuss the characteristics of nebulized inhalation, including its principles and influencing factors. Next, we compare the advantages and disadvantages of different types of nebulizers. Finally, we explore the clinical applications and recent research developments of nebulized inhalation therapy. By delving into these aspects, we aim to gain a deeper understanding of its pivotal role in contemporary medical treatment.
Collapse
Affiliation(s)
- Ruyi Yan
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Chang Zou
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xiaohang Yang
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Weihua Zhuang
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yushi Huang
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xiuli Zheng
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jie Hu
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Lingni Liao
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yongchao Yao
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xuping Sun
- High Altitude Medical Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Wenchuang Walter Hu
- Department of Laboratory Medicine, Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
2
|
Pöpperl P, Stoff M, Beineke A. Alveolar Macrophages in Viral Respiratory Infections: Sentinels and Saboteurs of Lung Defense. Int J Mol Sci 2025; 26:407. [PMID: 39796262 PMCID: PMC11721917 DOI: 10.3390/ijms26010407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Respiratory viral infections continue to cause pandemic and epidemic outbreaks in humans and animals. Under steady-state conditions, alveolar macrophages (AlvMϕ) fulfill a multitude of tasks in order to maintain tissue homeostasis. Due to their anatomic localization within the deep lung, AlvMϕ are prone to detect and react to inhaled viruses and thus play a role in the early pathogenesis of several respiratory viral infections. Here, detection of viral pathogens causes diverse antiviral and proinflammatory reactions. This fact not only makes them promising research targets, but also suggests them as potential targets for therapeutic and prophylactic approaches. This review aims to give a comprehensive overview of the current knowledge about the role of AlvMϕ in respiratory viral infections of humans and animals.
Collapse
Affiliation(s)
- Pauline Pöpperl
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Melanie Stoff
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| |
Collapse
|
3
|
Vera-Peralta H, Ruffié C, Najburg V, Brione M, Combredet C, Frantz P, Tournier JN, Tangy F, Mura M. Induction of tissue resident memory T cells by measles vaccine vector. Hum Vaccin Immunother 2024; 20:2436241. [PMID: 39693193 DOI: 10.1080/21645515.2024.2436241] [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: 08/22/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
Measles live attenuated vaccine (MV) induces strong humoral and cellular systemic memory responses allowing the successful control of measles since decades. MV has also been adapted into a promising vaccine platform with several vaccine candidates in clinical development. To understand and document the tissue-scaled memory response induced by MV, we explored the specific induction and persistence of resident memory T cells (Trm) in the lungs and the liver, two critical targeted tissues for vaccine development against several diseases. Trm are a subset of non-circulating highly specialized T cells. They are found at multiple barrier and mucosal sites, conveniently positioned to rapidly react against pathogens. The induction of Trm in different tissues is therefore critical for vaccine development. We demonstrated in mice the rapid generation of MV-specific and vectorized antigen-specific Trm in the liver and the lungs after a single dose, whatever the route of immunization. The intranasal route induced more Trm in the lungs than other routes, confirming the potential of intranasal vaccine administration of replicative viral vectors to generate a strong pulmonary immune response. MV-specific Trm cells were functionally active, with CD8+ Trm secreting granzyme B upon in vitro restimulation and CD4+ Trm cells secreting IFN-γ and TNF-α. We confirmed in human lymphocytes this tissue tropism by showing an overexpression of homing receptors directing them to epithelial and inflamed tissues. Vaccination strategies able to induce Trm cells at key sites represent a promising field to improve current vaccines, prioritize vaccine platforms and design future vaccines with enhanced protective efficacy.
Collapse
Affiliation(s)
- Heidy Vera-Peralta
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
- Interactions hôte-pathogène, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Claude Ruffié
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Valérie Najburg
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Matthias Brione
- Interactions hôte-pathogène, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Chantal Combredet
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Phanramphoei Frantz
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Jean-Nicolas Tournier
- Division recherche et innovation, Académie du Service de santé des armées, Paris, France
| | - Frédéric Tangy
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Marie Mura
- Interactions hôte-pathogène, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| |
Collapse
|
4
|
Wu L, Xu W, Jiang H, Yang M, Cun D. Respiratory delivered vaccines: Current status and perspectives in rational formulation design. Acta Pharm Sin B 2024; 14:5132-5160. [PMID: 39807330 PMCID: PMC11725141 DOI: 10.1016/j.apsb.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/20/2024] [Accepted: 08/18/2024] [Indexed: 01/16/2025] Open
Abstract
The respiratory tract is susceptible to various infections and can be affected by many serious diseases. Vaccination is one of the most promising ways that prevent infectious diseases and treatment of some diseases such as malignancy. Direct delivery of vaccines to the respiratory tract could mimic the natural process of infection and shorten the delivery path, therefore unique mucosal immunity at the first line might be induced and the efficiency of delivery can be high. Despite considerable attempts at the development of respiratory vaccines, the rational formulation design still warrants attention, i.e., how the formulation composition, particle properties, formulation type (liquid or solid), and devices would influence the immune outcome. This article reviews the recent advances in the formulation design and development of respiratory vaccines. The focus is on the state of the art of delivering antigenic compounds through the respiratory tract, overcoming the pulmonary bio-barriers, enhancing delivery efficiencies of respiratory vaccines as well as maintaining the stability of vaccines during storage and use. The choice of devices and the influence of deposition sites on vaccine efficiencies were also reviewed.
Collapse
Affiliation(s)
- Lan Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Wenwen Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Huiyang Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
- School of Food and Drug, Shenzhen Polytechnic University, China, Shenzhen 518055, China
| |
Collapse
|
5
|
Seefeld ML, Templeton EL, Lehtinen JM, Sinclair N, Yadav D, Hartwell BL. Harnessing the potential of the NALT and BALT as targets for immunomodulation using engineering strategies to enhance mucosal uptake. Front Immunol 2024; 15:1419527. [PMID: 39286244 PMCID: PMC11403286 DOI: 10.3389/fimmu.2024.1419527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.
Collapse
Affiliation(s)
- Madison L Seefeld
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Erin L Templeton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Justin M Lehtinen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Noah Sinclair
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Daman Yadav
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Brittany L Hartwell
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Zhang Y, Chamblee M, Xu J, Qu P, Shamseldin MM, Yoo SJ, Misny J, Thongpan I, Kc M, Hall JM, Gupta YA, Evans JP, Lu M, Ye C, Hsu CC, Liang X, Martinez-Sobrido L, Yount JS, Boyaka PN, Liu SL, Dubey P, Peeples ME, Li J. Three SARS-CoV-2 spike protein variants delivered intranasally by measles and mumps vaccines are broadly protective. Nat Commun 2024; 15:5589. [PMID: 38961063 PMCID: PMC11222507 DOI: 10.1038/s41467-024-49443-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/29/2024] [Indexed: 07/05/2024] Open
Abstract
As the new SARS-CoV-2 Omicron variants and subvariants emerge, there is an urgency to develop intranasal, broadly protective vaccines. Here, we developed highly efficacious, intranasal trivalent SARS-CoV-2 vaccine candidates (TVC) based on three components of the MMR vaccine: measles virus (MeV), mumps virus (MuV) Jeryl Lynn (JL1) strain, and MuV JL2 strain. Specifically, MeV, MuV-JL1, and MuV-JL2 vaccine strains, each expressing prefusion spike (preS-6P) from a different variant of concern (VoC), were combined to generate TVCs. Intranasal immunization of IFNAR1-/- mice and female hamsters with TVCs generated high levels of S-specific serum IgG antibodies, broad neutralizing antibodies, and mucosal IgA antibodies as well as tissue-resident memory T cells in the lungs. The immunized female hamsters were protected from challenge with SARS-CoV-2 original WA1, B.1.617.2, and B.1.1.529 strains. The preexisting MeV and MuV immunity does not significantly interfere with the efficacy of TVC. Thus, the trivalent platform is a promising next-generation SARS-CoV-2 vaccine candidate.
Collapse
Affiliation(s)
- Yuexiu Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Michelle Chamblee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Jiayu Xu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Panke Qu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Mohamed M Shamseldin
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Department of Microbiology, The Ohio State University, Columbus, OH, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan, Egypt
| | - Sung J Yoo
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Jack Misny
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Ilada Thongpan
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Mahesh Kc
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Jesse M Hall
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Yash A Gupta
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - John P Evans
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Cheng Chih Hsu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Xueya Liang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | | | - Jacob S Yount
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Shan-Lu Liu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, USA
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA.
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
7
|
Heida R, Frijlink HW, Hinrichs WLJ. Inhalation of vaccines and antiviral drugs to fight respiratory virus infections: reasons to prioritize the pulmonary route of administration. mBio 2023; 14:e0129523. [PMID: 37768057 PMCID: PMC10653782 DOI: 10.1128/mbio.01295-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023] Open
Abstract
Many of the current pandemic threats are caused by viruses that infect the respiratory tract. Remarkably though, the majority of vaccines and antiviral drugs are administered via alternative routes. In this perspective, we argue that the pulmonary route of administration deserves more attention in the search for novel therapeutic strategies against respiratory virus infections. Firstly, vaccines administered at the viral portal of entry can induce a broader immune response, employing the mucosal arm of the immune system; secondly, direct administration of antiviral drugs at the target site leads to superior bioavailability, enabling lower dosing and reducing the chance of side effects. We further elaborate on why the pulmonary route may induce a superior effect compared to the intranasal route of administration and provide reasons why dry powder formulations for inhalation have significant advantages over standard liquid formulations.
Collapse
Affiliation(s)
- Rick Heida
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Henderik W. Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Wouter L. J. Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
8
|
Jeyanathan M, Afkhami S, Kang A, Xing Z. Viral-vectored respiratory mucosal vaccine strategies. Curr Opin Immunol 2023; 84:102370. [PMID: 37499279 DOI: 10.1016/j.coi.2023.102370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023]
Abstract
Increasing global concerns of pandemic respiratory viruses highlight the importance of developing optimal vaccination strategies that encompass vaccine platform, delivery route, and regimens. The decades-long effort to develop vaccines to combat respiratory infections such as influenza, respiratory syncytial virus, and tuberculosis has met with challenges, including the inability of systemically administered vaccines to induce respiratory mucosal (RM) immunity. In this regard, ample preclinical and available clinical studies have demonstrated the superiority of RM vaccination to induce RM immunity over parenteral route of vaccination. A great stride has been made in developing vaccines for RM delivery against respiratory pathogens, including M. tuberculosis and SARS-CoV-2. In particular, inhaled aerosol delivery of adenoviral-vectored vaccines has shown significant promise.
Collapse
Affiliation(s)
- Mangalakumari Jeyanathan
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alisha Kang
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre and Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
9
|
Komalla V, Wong CYJ, Sibum I, Muellinger B, Nijdam W, Chaugule V, Soria J, Ong HX, Buchmann NA, Traini D. Advances in soft mist inhalers. Expert Opin Drug Deliv 2023; 20:1055-1070. [PMID: 37385962 DOI: 10.1080/17425247.2023.2231850] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/01/2023]
Abstract
INTRODUCTION Soft mist inhalers (SMIs) are propellant-free inhalers that utilize mechanical power to deliver single or multiple doses of inhalable drug aerosols in the form of a slow mist to patients. Compared to traditional inhalers, SMIs allow for a longer and slower release of aerosol with a smaller ballistic effect, leading to a limited loss in the oropharyngeal area, whilst requiring little coordination of actuation and inhalation by patients. Currently, the Respimat® is the only commercially available SMI, with several others in different stages of preclinical and clinical development. AREAS COVERED The primary purpose of this review is to critically assess recent advances in SMIs for the delivery of inhaled therapeutics. EXPERT OPINION Advanced particle formulations, such as nanoparticles which target specific areas of the lung, Biologics, such as vaccines, proteins, and antibodies (which are sensitive to aerosolization), are expected to be generally delivered by SMIs. Furthermore, repurposed drugs are expected to constitute a large share of future formulations to be delivered by SMIs. SMIs can also be employed for the delivery of formulations that target systemic diseases. Finally, digitalizing SMIs would improve patient adherence and provide clinicians with fundamental insights into patients' treatment progress.
Collapse
Affiliation(s)
- Varsha Komalla
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, Australia
| | - Chun Yuen Jerry Wong
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, Australia
- Macquarie Medical School, Department of Biological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | | | | | | | - Vishal Chaugule
- Laboratory for Turbulence Research in Aerospace and Combustion (LTRAC), Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Australia
| | - Julio Soria
- Laboratory for Turbulence Research in Aerospace and Combustion (LTRAC), Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, Australia
- Macquarie Medical School, Department of Biological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | | | - D Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, Australia
- Macquarie Medical School, Department of Biological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| |
Collapse
|
10
|
Dowell AC, Lancaster T, Bruton R, Ireland G, Bentley C, Sylla P, Zuo J, Scott S, Jadir A, Begum J, Roberts T, Stephens C, Ditta S, Shepherdson R, Powell AA, Brent AJ, Brent B, Baawuah F, Okike I, Beckmann J, Ahmad S, Aiano F, Garstang J, Ramsay ME, Azad R, Waiblinger D, Willett B, Wright J, Ladhani SN, Moss P. Immunological imprinting of humoral immunity to SARS-CoV-2 in children. Nat Commun 2023; 14:3845. [PMID: 37386081 PMCID: PMC10310754 DOI: 10.1038/s41467-023-39575-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023] Open
Abstract
Omicron variants of SARS-CoV-2 are globally dominant and infection rates are very high in children. We measure immune responses following Omicron BA.1/2 infection in children aged 6-14 years and relate this to prior and subsequent SARS-CoV-2 infection or vaccination. Primary Omicron infection elicits a weak antibody response with poor functional neutralizing antibodies. Subsequent Omicron reinfection or COVID-19 vaccination elicits increased antibody titres with broad neutralisation of Omicron subvariants. Prior pre-Omicron SARS-CoV-2 virus infection or vaccination primes for robust antibody responses following Omicron infection but these remain primarily focussed against ancestral variants. Primary Omicron infection thus elicits a weak antibody response in children which is boosted after reinfection or vaccination. Cellular responses are robust and broadly equivalent in all groups, providing protection against severe disease irrespective of SARS-CoV-2 variant. Immunological imprinting is likely to act as an important determinant of long-term humoral immunity, the future clinical importance of which is unknown.
Collapse
Affiliation(s)
- Alexander C Dowell
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Tara Lancaster
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rachel Bruton
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Georgina Ireland
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, UK
| | - Christopher Bentley
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Panagiota Sylla
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jianmin Zuo
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sam Scott
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Azar Jadir
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jusnara Begum
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Thomas Roberts
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Christine Stephens
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Shabana Ditta
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Rebecca Shepherdson
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Annabel A Powell
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, UK
| | - Andrew J Brent
- Oxford University Hospitals NHS Foundation Trust, Old Road, Oxford, UK
- University of Oxford, Wellington Square, Oxford, OX1 2JD, UK
| | - Bernadette Brent
- Oxford University Hospitals NHS Foundation Trust, Old Road, Oxford, UK
| | - Frances Baawuah
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, UK
| | - Ifeanyichukwu Okike
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, UK
- University Hospitals of Derby and Burton NHS Foundation Trust, Uttoxeter New Road, Derby, UK
| | - Joanne Beckmann
- East London NHS Foundation Trust, 9 Allie Street, London, UK
| | - Shazaad Ahmad
- Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Felicity Aiano
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, UK
| | - Joanna Garstang
- Birmingham Community Healthcare NHS Trust, Holt Street, Aston, UK
| | - Mary E Ramsay
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, UK
| | - Rafaq Azad
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Dagmar Waiblinger
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Brian Willett
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Shamez N Ladhani
- Immunisation Department, UK Health Security Agency, 61 Colindale Avenue, London, UK.
| | - Paul Moss
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
11
|
Brandimarte B, Di Rienzo Businco L, Cappello F, Fiore R, Bastone G, Gualdi G, Sollaku S, Casciani E, Tortorella F, Longo P, Centanini E, Pavaci S, Sangiuolo F, Patrizi MP, Miersch S, Sidhu SS, Sacchini V, Novelli G. Nebulization of pharmacological solutions with an innovative medical device based on microvaporization. Heliyon 2023; 9:e14673. [PMID: 37020941 PMCID: PMC10068108 DOI: 10.1016/j.heliyon.2023.e14673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
The currently available nebulization devices have a slow aerosol flow and produce vapor with large microdrops. Improved devices that achieve higher airflow and produce smaller microdrops are needed to improve the clinical care of patients. To address this critical need, we developed a novel system for the molecular vaporization of liquids. This device vaporizes an active pharmacological substance dissolved in water, alcohol, or a mixture of water and alcohol using two energy sources at the same time: high-frequency ultrasound and thermal induction. Application of energy to a solution contained in the device's tank allows, within tens of seconds, for the vaporization of the solution itself, with the generation of a vapor consisting of microdrops of very small diameter (0.2-0.3 μm). In this article, we illustrate the technology used, the main verification tests performed, and the primary fields of application for this device. In particular, the advantages of both the aerosol delivery system and the administration system are highlighted.
Collapse
|
12
|
Schmitz KS, Eblé PL, van Gennip RGP, Maris-Veldhuis MA, de Vries RD, van Keulen LJM, de Swart RL, van Rijn PA. Pathogenesis of wild-type- and vaccine-based recombinant peste des petits ruminants virus (PPRV) expressing EGFP in experimentally infected domestic goats. J Gen Virol 2023; 104. [PMID: 36757863 DOI: 10.1099/jgv.0.001828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) is a highly contagious morbillivirus related to measles and canine distemper virus, mostly affecting small ruminants. The corresponding PPR disease has a high clinical impact in goats and is characterized by fever, oral and nasal erosions, diarrhoea and pneumonia. In addition, massive infection of lymphoid tissues causes lymphopaenia and immune suppression. This results in increased susceptibility to secondary bacterial infections, explaining the observed high mortality in some outbreaks. We studied the pathogenesis of PPR by experimental inoculation of Dutch domestic goats with a recombinant virulent PPRV strain modified to express EGFP and compared it to an EGFP-expressing vaccine strain of PPRV. After intratracheal inoculation with virulent PPRV, animals developed fever, viraemia and leucopaenia, and shed virus from the respiratory and gastro-intestinal tracts. Macroscopic evaluation of fluorescence at the peak of infection 7 days post-inoculation (dpi) showed prominent PPRV infection of the respiratory tract, lymphoid tissues, gastro-intestinal tract, mucosae and skin. Flow cytometry of PBMCs collected over time demonstrated a cell-associated viraemia mediated by infected lymphocytes. At 14 dpi, pathognomonic zebra stripes were detected in the mucosa of the large intestine. In contrast, vaccine strain-inoculated goats remained largely macroscopically fluorescence negative and did not present clinical signs. A low-level viraemia was detected by flow cytometry, but at necropsy no histological lesions were observed. Animals from both groups seroconverted as early as 7 dpi and sera efficiently neutralized virulent PPRV in vitro. Combined, this work presents a study of the pathogenesis of wild type- and vaccine-based PPRV in its natural host. This study shows the strength of recombinant EGFP-expressing viruses in fluorescence-guided pathogenesis studies.
Collapse
Affiliation(s)
| | - Phaedra L Eblé
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, Netherlands
| | - René G P van Gennip
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, Netherlands
| | | | - Rory D de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
| | - Lucien J M van Keulen
- Department of Infection Biology, Wageningen Bioveterinary Research, Lelystad, Netherlands
| | - Rik L de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, Netherlands.,Department of Virology, Wageningen Bioveterinary Research, Lelystad, Netherlands
| | - Piet A van Rijn
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, Netherlands.,Department of Biochemistry, Centre of Human Metabolomics, North-West University, Potchefstroom, South Africa
| |
Collapse
|
13
|
Sievers BL, Sievers RE, Sievers EL. Incentivized self-vaccination for global measles eradication. J Virus Erad 2022; 8:100310. [PMID: 36578361 PMCID: PMC9791812 DOI: 10.1016/j.jve.2022.100310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Measles-we've become inured to its cruel, insidious impact as it kills over 100,000 children yearly because of suboptimal vaccination coverage. It does not have to be this way. A familiar, safe, exceptionally effective measles vaccine saves lives and permanent, global measles eradication is within reach. But now we need to be clever and courageously explore new strategies to save lives. Firstly, let us enable people to vaccinate themselves, not with a needle and syringe, but with a quick inhaled puff of dry powder vaccine. Secondly, let us provide micro-payments using digital currency to incentivize those who vaccinate themselves. Thirdly, let us leverage learnings from how our social networks guide our behaviors to further encourage self-vaccination. Fourthly, let us inspire friendly regional competition among communities vying for the highest proportion of citizens who show measles neutralizing antibodies in spot saliva samples. With global cooperation and relentless determination, we eradicated smallpox. Next up? Measles.
Collapse
Affiliation(s)
| | - Robert E. Sievers
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO, 80309, USA
| | | |
Collapse
|
14
|
Jeyanathan V, Afkhami S, D’Agostino MR, Zganiacz A, Feng X, Miller MS, Jeyanathan M, Thompson MR, Xing Z. Differential Biodistribution of Adenoviral-Vectored Vaccine Following Intranasal and Endotracheal Deliveries Leads to Different Immune Outcomes. Front Immunol 2022; 13:860399. [PMID: 35757753 PMCID: PMC9231681 DOI: 10.3389/fimmu.2022.860399] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/11/2022] [Indexed: 11/22/2022] Open
Abstract
Infectious diseases of the respiratory tract are one of the top causes of global morbidity and mortality with lower respiratory tract infections being the fourth leading cause of death. The respiratory mucosal (RM) route of vaccine delivery represents a promising strategy against respiratory infections. Although both intranasal and inhaled aerosol methods have been established for human application, there is a considerable knowledge gap in the relationship of vaccine biodistribution to immune efficacy in the lung. Here, by using a murine model and an adenovirus-vectored model vaccine, we have compared the intranasal and endotracheal delivery methods in their biodistribution, immunogenicity and protective efficacy. We find that compared to intranasal delivery, the deepened and widened biodistribution in the lung following endotracheal delivery is associated with much improved vaccine-mediated immunogenicity and protection against the target pathogen. Our findings thus support further development of inhaled aerosol delivery of vaccines over intranasal delivery for human application.
Collapse
Affiliation(s)
- Vidthiya Jeyanathan
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Michael R. D’Agostino
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Anna Zganiacz
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Xueya Feng
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew S. Miller
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Michael R. Thompson
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada,*Correspondence: Zhou Xing,
| |
Collapse
|
15
|
Bianchi FP, Stefanizzi P, Trerotoli P, Tafuri S. Sex and age as determinants of the seroprevalence of anti-measles IgG among European healthcare workers: A systematic review and meta-analysis. Vaccine 2022; 40:3127-3141. [PMID: 35491343 DOI: 10.1016/j.vaccine.2022.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/14/2022] [Accepted: 04/04/2022] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The international literature shows good evidence of a significant rate of measles susceptibility among healthcare workers (HCWs). As such, they are an important public health issue. METHODS We conducted a systematic review and meta-analysis to estimate the prevalence of susceptible HCWs in EU/EEA countries and in the UK and to explore the characteristics (sex and age differences) and management of those found to be susceptible. RESULTS Nineteen studies were included in the meta-analysis. The prevalence of measles-susceptible HCWs was 13.3% (95 %CI: 10.0-17.0%). In a comparison of serosusceptible female vs. male HCWs, the RR was 0.92 (95 %CI = 0.83-1.03), and in a comparison of age classes (born after vs. before 1980) the RR was 2.78 (95 %CI = 2.20-3.50). The most recent studies proposed the mandatory vaccination of HCWs. DISCUSSION According to our meta-analysis, the prevalence of serosusceptible European HCWs is 13%; HCWs born in the post-vaccination era seem to be at higher risk. Healthcare professionals susceptible to measles are a serious epidemiological concern. Greater efforts should therefore be made to identify those who have yet to be vaccinated and actively encourage their vaccination.
Collapse
Affiliation(s)
| | - Pasquale Stefanizzi
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Italy
| | - Paolo Trerotoli
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Italy
| | - Silvio Tafuri
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, Italy.
| |
Collapse
|
16
|
Hassan J, Haigh C, Ahmed T, Uddin MJ, Das DB. Potential of Microneedle Systems for COVID-19 Vaccination: Current Trends and Challenges. Pharmaceutics 2022; 14:1066. [PMID: 35631652 PMCID: PMC9144974 DOI: 10.3390/pharmaceutics14051066] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/27/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
To prevent the coronavirus disease 2019 (COVID-19) pandemic and aid restoration to prepandemic normality, global mass vaccination is urgently needed. Inducing herd immunity through mass vaccination has proven to be a highly effective strategy for preventing the spread of many infectious diseases, which protects the most vulnerable population groups that are unable to develop immunity, such as people with immunodeficiencies or weakened immune systems due to underlying medical or debilitating conditions. In achieving global outreach, the maintenance of the vaccine potency, transportation, and needle waste generation become major issues. Moreover, needle phobia and vaccine hesitancy act as hurdles to successful mass vaccination. The use of dissolvable microneedles for COVID-19 vaccination could act as a major paradigm shift in attaining the desired goal to vaccinate billions in the shortest time possible. In addressing these points, we discuss the potential of the use of dissolvable microneedles for COVID-19 vaccination based on the current literature.
Collapse
Affiliation(s)
- Jasmin Hassan
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
| | - Charlotte Haigh
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK;
| | - Tanvir Ahmed
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
| | - Md Jasim Uddin
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
- Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK;
| |
Collapse
|
17
|
Abstract
Measles is a highly contagious, potentially fatal, but vaccine-preventable disease caused by measles virus. Symptoms include fever, maculopapular rash, and at least one of cough, coryza, or conjunctivitis, although vaccinated individuals can have milder or even no symptoms. Laboratory diagnosis relies largely on the detection of specific IgM antibodies in serum, dried blood spots, or oral fluid, or the detection of viral RNA in throat or nasopharyngeal swabs, urine, or oral fluid. Complications can affect many organs and often include otitis media, laryngotracheobronchitis, pneumonia, stomatitis, and diarrhoea. Neurological complications are uncommon but serious, and can occur during or soon after the acute disease (eg, acute disseminated encephalomyelitis) or months or even years later (eg, measles inclusion body encephalitis and subacute sclerosing panencephalitis). Patient management mainly involves supportive therapy, such as vitamin A supplementation, monitoring for and treatment of secondary bacterial infections with antibiotics, and rehydration in the case of severe diarrhoea. There is no specific antiviral therapy for the treatment of measles, and disease control largely depends on prevention. However, despite the availability of a safe and effective vaccine, measles is still endemic in many countries and causes considerable morbidity and mortality, especially among children in resource-poor settings. The low case numbers reported in 2020, after a worldwide resurgence of measles between 2017 and 2019, have to be interpreted cautiously, owing to the effect of the COVID-19 pandemic on disease surveillance. Disrupted vaccination activities during the pandemic increase the potential for another resurgence of measles in the near future, and effective, timely catch-up vaccination campaigns, strong commitment and leadership, and sufficient resources will be required to mitigate this threat.
Collapse
Affiliation(s)
- Judith M Hübschen
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.
| | - Ionela Gouandjika-Vasilache
- Laboratoire des Virus Entériques et de la Rougeole, Institut Pasteur de Bangui, Bangui, Central African Republic
| | - Julia Dina
- Virology Department, Normandie University, UNICAEN, INSERM U1311 DynaMicURe, Caen University Hospital, Caen, France
| |
Collapse
|
18
|
Meenakshi S, Kumar VU, Dhingra S, Murti K. Nasal vaccine as a booster shot: a viable solution to restrict pandemic? Clin Exp Vaccine Res 2022; 11:184-192. [PMID: 35799869 PMCID: PMC9200647 DOI: 10.7774/cevr.2022.11.2.184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/03/2022] [Indexed: 01/23/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic revolutionized the vaccine market and initiated the momentum for alternative routes of administration for vaccines. The intranasal route of immunization is one such possibility that appears to be the most promising since it has some significant advantages, particularly in the prevention of respiratory infection. To analyze and summarize the role of nasal vaccines over conventional vaccines during COVID-19 and the need for the nasal vaccine as a booster shot. In this narrative review, the required data was retrieved using keywords “COVID-19,” “Intranasal,” “Immunity,” “Nasal spray,” and “Mucosal” in databases including PubMed, Scopus, Embase, Science Direct, and Web of Sciences. The results of the study showed that the nasal vaccines were both effective and protective according to the current researches approaching during the COVID-19 period and the preclinical and clinical phase trials prove the intranasal vaccination elicits more robust and cross-protective immunity than conventional vaccines. In this narrative review article, mechanisms across the nasal mucosa will be briefly presented and the current status of nasal vaccines during the COVID-19 pandemic is summarized, and advantages over traditional vaccines are provided. Furthermore, after exploring the primary benefits and kinetics of nasal vaccine, the potential for consideration of nasal vaccine as a booster dose is also discussed.
Collapse
Affiliation(s)
- Sarasa Meenakshi
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - V. Udaya Kumar
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Sameer Dhingra
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| |
Collapse
|
19
|
Vatzia E, Allen ER, Manjegowda T, Morris S, McNee A, Martini V, Kaliath R, Ulaszewska M, Boyd A, Paudyal B, Carr VB, Chrun T, Maze E, MacLoughlin R, van Diemen PM, Everett HE, Lambe T, Gilbert SC, Tchilian E. Respiratory and Intramuscular Immunization With ChAdOx2-NPM1-NA Induces Distinct Immune Responses in H1N1pdm09 Pre-Exposed Pigs. Front Immunol 2021; 12:763912. [PMID: 34804053 PMCID: PMC8595216 DOI: 10.3389/fimmu.2021.763912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/13/2021] [Indexed: 01/12/2023] Open
Abstract
There is a critical need to develop superior influenza vaccines that provide broader protection. Influenza vaccines are traditionally tested in naive animals, although humans are exposed to influenza in the first years of their lives, but the impact of prior influenza exposure on vaccine immune responses has not been well studied. Pigs are an important natural host for influenza, are a source of pandemic viruses, and are an excellent model for human influenza. Here, we investigated the immunogenicity of the ChAdOx2 viral vectored vaccine, expressing influenza nucleoprotein, matrix protein 1, and neuraminidase in H1N1pdm09 pre-exposed pigs. We evaluated the importance of the route of administration by comparing intranasal, aerosol, and intramuscular immunizations. Aerosol delivery boosted the local lung T-cell and antibody responses, while intramuscular immunization boosted peripheral blood immunity. These results will inform how best to deliver vaccines in order to harness optimal protective immunity.
Collapse
Affiliation(s)
- Eleni Vatzia
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Elizabeth R Allen
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Tanuja Manjegowda
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Susan Morris
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Adam McNee
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Veronica Martini
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Reshma Kaliath
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Marta Ulaszewska
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Amy Boyd
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Basudev Paudyal
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Veronica B Carr
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Tiphany Chrun
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Emmanuel Maze
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | | | | | - Helen E Everett
- Animal and Plant Health Agency (APHA)-Weybridge, Addlestone, United Kingdom
| | - Teresa Lambe
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah C Gilbert
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Elma Tchilian
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| |
Collapse
|
20
|
Doornekamp L, Comvalius AD, GeurtsvanKessel CH, Slobbe L, Scherbeijn SMJ, van Genderen PJJ, van Binnendijk RS, van Gorp ECM, de Swart RL, Goeijenbier M. Measles seroprevalence among Dutch travelling families. Travel Med Infect Dis 2021; 44:102194. [PMID: 34728385 DOI: 10.1016/j.tmaid.2021.102194] [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: 08/04/2020] [Revised: 10/05/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND While measles vaccination is widely implemented in national immunisation programmes, measles incidence rates are increasing worldwide. Dutch inhabitants who were born between 1965-1975 may have fallen between two stools, lacking protection from a natural infection, and having missed the introduction of the measles vaccination schedule. With this study we aim to find the measles seroprevalence in travellers born between 1965 and 1975, compared to those born before 1965 and after 1975. METHODS Families travelling to Eastern Europe or outside Europe during the preceding year were recruited via Dutch secondary schools between 2016 and 2018. Their vaccination status was assessed using questionnaires, vaccination records and measles serology in dried blood spot (DBS) eluates. Measles virus antibody concentrations were determined with an ELISA (EUROIMMUNE®) and a subset was retested with a focus reduction neutralization assay (FRNT). RESULTS In 188 (79%) of the 239 available DBS eluates, the ELISA could detect sufficient measles virus-specific IgG antibodies. Of the negative samples that were retested with FRNT, 85% remained negative, resulting in an overall seroprevalence of 82% [95% CI 76-86]. Children had a lower seroprevalence (72%) than adults (87%). Travellers born between 1965 and 1975 were protected in 89%. CONCLUSIONS In this study, we report a measles seroprevalence of 82% among Dutch travelling families. Remarkably, seroprevalence rates were lowest in children (12-18 years) instead of travellers born between 1965 and 1975. Although a fraction of people without detectable antibodies may be protected by other immune mechanisms, these data suggest that measles (re)vaccination should be considered for travellers to endemic regions.
Collapse
Affiliation(s)
- Laura Doornekamp
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Travel Clinic, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Anouskha D Comvalius
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Corine H GeurtsvanKessel
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Lennert Slobbe
- Travel Clinic, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Institute for Tropical Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Sandra M J Scherbeijn
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Perry J J van Genderen
- Travel Clinic, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Institute for Tropical Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Rob S van Binnendijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Eric C M van Gorp
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Travel Clinic, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Rik L de Swart
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marco Goeijenbier
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
21
|
Menon I, Bagwe P, Gomes KB, Bajaj L, Gala R, Uddin MN, D’Souza MJ, Zughaier SM. Microneedles: A New Generation Vaccine Delivery System. MICROMACHINES 2021; 12:435. [PMID: 33919925 PMCID: PMC8070939 DOI: 10.3390/mi12040435] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/17/2022]
Abstract
Transdermal vaccination route using biodegradable microneedles is a rapidly progressing field of research and applications. The fear of painful needles is one of the primary reasons most people avoid getting vaccinated. Therefore, developing an alternative pain-free method of vaccination using microneedles has been a significant research area. Microneedles comprise arrays of micron-sized needles that offer a pain-free method of delivering actives across the skin. Apart from being pain-free, microneedles provide various advantages over conventional vaccination routes such as intramuscular and subcutaneous. Microneedle vaccines induce a robust immune response as the needles ranging from 50 to 900 μm in length can efficiently deliver the vaccine to the epidermis and the dermis region, which contains many Langerhans and dendritic cells. The microneedle array looks like band-aid patches and offers the advantages of avoiding cold-chain storage and self-administration flexibility. The slow release of vaccine antigens is an important advantage of using microneedles. The vaccine antigens in the microneedles can be in solution or suspension form, encapsulated in nano or microparticles, and nucleic acid-based. The use of microneedles to deliver particle-based vaccines is gaining importance because of the combined advantages of particulate vaccine and pain-free immunization. The future of microneedle-based vaccines looks promising however, addressing some limitations such as dosing inadequacy, stability and sterility will lead to successful use of microneedles for vaccine delivery. This review illustrates the recent research in the field of microneedle-based vaccination.
Collapse
Affiliation(s)
- Ipshita Menon
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (I.M.); (P.B.); (K.B.G.); (L.B.); (M.N.U.); (M.J.D.)
| | - Priyal Bagwe
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (I.M.); (P.B.); (K.B.G.); (L.B.); (M.N.U.); (M.J.D.)
| | - Keegan Braz Gomes
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (I.M.); (P.B.); (K.B.G.); (L.B.); (M.N.U.); (M.J.D.)
| | - Lotika Bajaj
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (I.M.); (P.B.); (K.B.G.); (L.B.); (M.N.U.); (M.J.D.)
| | - Rikhav Gala
- Biotechnology Division, Center for Mid-Atlantic (CMA), Fraunhofer USA, Newark, DE 19711, USA;
| | - Mohammad N. Uddin
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (I.M.); (P.B.); (K.B.G.); (L.B.); (M.N.U.); (M.J.D.)
| | - Martin J. D’Souza
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (I.M.); (P.B.); (K.B.G.); (L.B.); (M.N.U.); (M.J.D.)
| | - Susu M. Zughaier
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2731, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2731, Qatar
| |
Collapse
|
22
|
Martini V, Hinchcliffe M, Blackshaw E, Joyce M, McNee A, Beverley P, Townsend A, MacLoughlin R, Tchilian E. Distribution of Droplets and Immune Responses After Aerosol and Intra-Nasal Delivery of Influenza Virus to the Respiratory Tract of Pigs. Front Immunol 2020; 11:594470. [PMID: 33193445 PMCID: PMC7653178 DOI: 10.3389/fimmu.2020.594470] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/07/2020] [Indexed: 11/13/2022] Open
Abstract
Recent evidence indicates that local immune responses and tissue resident memory T cells (TRM) are critical for protection against respiratory infections but there is little information on the contributions of upper and lower respiratory tract (URT and LRT) immunity. To provide a rational basis for designing methods for optimal delivery of vaccines to the respiratory tract in a large animal model, we investigated the distribution of droplets generated by a mucosal atomization device (MAD) and two vibrating mesh nebulizers (VMNs) and the immune responses induced by delivery of influenza virus by MAD in pigs. We showed that droplets containing the drug albuterol, a radiolabel (99mTc-DTPA), or a model influenza virus vaccine (S-FLU) have similar aerosol characteristics. 99mTc-DTPA scintigraphy showed that VMNs deliver droplets with uniform distribution throughout the lungs as well as the URT. Surprisingly MAD administration (1ml/nostril) also delivered a high proportion of the dose to the lungs, albeit concentrated in a small area. After MAD administration of influenza virus, antigen specific T cells were found at high frequency in nasal turbinates, trachea, broncho-alveolar lavage, lungs, tracheobronchial nodes, and blood. Anti-influenza antibodies were detected in serum, BAL and nasal swabs. We conclude that the pig is useful for investigating optimal targeting of vaccines to the respiratory tract.
Collapse
Affiliation(s)
- Veronica Martini
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom.,Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Elaine Blackshaw
- Radiological Sciences, School of Medicine, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | | | - Adam McNee
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom.,School of Veterinary Medicine, Daphne Jackson Road, University of Surrey, Guildford, United Kingdom
| | - Peter Beverley
- National Heart and Lung Institute, St Mary's Campus, Imperial College, London, United Kingdom
| | - Alain Townsend
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Elma Tchilian
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| |
Collapse
|
23
|
McCarthy SD, González HE, Higgins BD. Future Trends in Nebulized Therapies for Pulmonary Disease. J Pers Med 2020; 10:E37. [PMID: 32397615 PMCID: PMC7354528 DOI: 10.3390/jpm10020037] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Aerosol therapy is a key modality for drug delivery to the lungs of respiratory disease patients. Aerosol therapy improves therapeutic effects by directly targeting diseased lung regions for rapid onset of action, requiring smaller doses than oral or intravenous delivery and minimizing systemic side effects. In order to optimize treatment of critically ill patients, the efficacy of aerosol therapy depends on lung morphology, breathing patterns, aerosol droplet characteristics, disease, mechanical ventilation, pharmacokinetics, and the pharmacodynamics of cell-drug interactions. While aerosol characteristics are influenced by drug formulations and device mechanisms, most other factors are reliant on individual patient variables. This has led to increased efforts towards more personalized therapeutic approaches to optimize pulmonary drug delivery and improve selection of effective drug types for individual patients. Vibrating mesh nebulizers (VMN) are the dominant device in clinical trials involving mechanical ventilation and emerging drugs. In this review, we consider the use of VMN during mechanical ventilation in intensive care units. We aim to link VMN fundamentals to applications in mechanically ventilated patients and look to the future use of VMN in emerging personalized therapeutic drugs.
Collapse
Affiliation(s)
- Sean D. McCarthy
- Anaesthesia, School of Medicine, National University of Ireland Galway, H91 TK33 Galway, Ireland; (S.D.M.); (H.E.G.)
- Lung Biology Group, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Héctor E. González
- Anaesthesia, School of Medicine, National University of Ireland Galway, H91 TK33 Galway, Ireland; (S.D.M.); (H.E.G.)
- Lung Biology Group, Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Brendan D. Higgins
- Physiology, School of Medicine, National University of Ireland Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
24
|
Measles pathogenesis, immune suppression and animal models. Curr Opin Virol 2020; 41:31-37. [PMID: 32339942 DOI: 10.1016/j.coviro.2020.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 02/09/2023]
Abstract
Measles virus causes a disease with seemingly innocent symptoms, such as fever and rash. However, measles immune suppression causes increased susceptibility to opportunistic infections that are responsible for the majority of over 100000 yearly fatalities. The pathogenesis of measles is complex, because measles virus uses multiple receptors to infect different cell types in different phases of the disease. Experimental morbillivirus infections with wild-type viruses in natural host species have demonstrated that direct infection and depletion of memory immune cells causes immune amnesia. This was confirmed in studies of a measles outbreak in unvaccinated children and provides an explanation for epidemiological observations of long-term increases in morbidity and mortality after measles.
Collapse
|
25
|
Mahapatra M, Selvaraj M, Parida S. Comparison of Immunogenicity and Protective Efficacy of PPR Live Attenuated Vaccines (Nigeria 75/1 and Sungri 96) Administered by Intranasal and Subcutaneous Routes. Vaccines (Basel) 2020; 8:vaccines8020168. [PMID: 32268574 PMCID: PMC7349158 DOI: 10.3390/vaccines8020168] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/16/2022] Open
Abstract
Following the successful eradication of rinderpest, the World Organization of Animal Health (OIE) and the Food and Agriculture Organization (FAO) have set a goal to eradicate peste des petits ruminants (PPR) globally by 2030. Vaccination is being taken forward as the key strategy along with epidemiological surveillance to target vaccination efforts and eradicate the disease. PPR is highly contagious and is generally spread by aerosolized droplets and close contact. Currently, two live attenuated vaccines (Nigeria 75/1 and Sungri 96) are in use, and administered subcutaneously to prevent transmission of PPR and protect vaccinated animals. Though the target cells that support primary replication of PPR vaccine strains are largely unknown, it is hypothesized that the immune response could be intensified following intranasal vaccine delivery as this route mimics the natural route of infection. This study aims to compare the immunogenicity and protective efficacy of the two currently available live attenuated PPR vaccines following subcutaneous and intranasal routes of vaccination in target species. Groups of five goats were vaccinated with live attenuated PPR vaccines (Nigeria 75/1 and Sungri 96) by either the subcutaneous or intranasal route, and 28 days later challenged intranasally with virulent PPR virus. All vaccinated animals regardless of vaccination route produced PPRV-specific antibodies post-vaccination. Following challenge, all goats were protected from clinical disease, and vaccination was considered to have induced sterilizing immunity. This study demonstrates that the intranasal route of vaccination is as effective as the subcutaneous route of vaccination when using available live attenuated PPR vaccines.
Collapse
|
26
|
Yenkoidiok-Douti L, Jewell CM. Integrating Biomaterials and Immunology to Improve Vaccines Against Infectious Diseases. ACS Biomater Sci Eng 2020; 6:759-778. [PMID: 33313391 PMCID: PMC7725244 DOI: 10.1021/acsbiomaterials.9b01255] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite the success of vaccines in preventing many infectious diseases, effective vaccines against pathogens with ongoing challenges - such as HIV, malaria, and tuberculosis - remain unavailable. The emergence of new pathogen variants, the continued prevalence of existing pathogens, and the resurgence of yet other infectious agents motivate the need for new, interdisciplinary approaches to direct immune responses. Many current and candidate vaccines, for example, are poorly immunogenic, provide only transient protection, or create risks of regaining pathogenicity in certain immune-compromised conditions. Recent advances in biomaterials research are creating new potential to overcome these challenges through improved formulation, delivery, and control of immune signaling. At the same time, many of these materials systems - such as polymers, lipids, and self-assembly technologies - may achieve this goal while maintaining favorable safety profiles. This review highlights ways in which biomaterials can advance existing vaccines to safer, more efficacious technologies, and support new vaccines for pathogens that do not yet have vaccines. Biomaterials that have not yet been applied to vaccines for infectious disease are also discussed, and their potential in this area is highlighted.
Collapse
Affiliation(s)
- Lampouguin Yenkoidiok-Douti
- Fischell Department of Bioengineering, University of Maryland, College Park, 8278 Paint Branch Drive, College Park, MD, 20742, United States
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Rockville, MD, 20852, United States
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, 8278 Paint Branch Drive, College Park, MD, 20742, United States
- Department of Veterans Affairs, VA Maryland Health Care System, 10. N Green Street, Baltimore, MD 21201, USA
- Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD 20742, United States
- Department of Microbiology and Immunology, University of Maryland Medical School, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, United States
- Marlene and Stewart Greenebaum Cancer Center, 22 S. Greene Street, Suite N9E17, Baltimore, MD 21201, United States
| |
Collapse
|
27
|
Sécher T, Mayor A, Heuzé-Vourc'h N. Inhalation of Immuno-Therapeutics/-Prophylactics to Fight Respiratory Tract Infections: An Appropriate Drug at the Right Place! Front Immunol 2019; 10:2760. [PMID: 31849954 PMCID: PMC6896187 DOI: 10.3389/fimmu.2019.02760] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/12/2019] [Indexed: 01/18/2023] Open
Affiliation(s)
- Thomas Sécher
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Tours, France.,Centre d'Etude des Pathologies Respiratoires, Université de Tours, Tours, France
| | - Alexie Mayor
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Tours, France.,Centre d'Etude des Pathologies Respiratoires, Université de Tours, Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Tours, France.,Centre d'Etude des Pathologies Respiratoires, Université de Tours, Tours, France
| |
Collapse
|
28
|
Mina MJ, Kula T, Leng Y, Li M, de Vries RD, Knip M, Siljander H, Rewers M, Choy DF, Wilson MS, Larman HB, Nelson AN, Griffin DE, de Swart RL, Elledge SJ. Measles virus infection diminishes preexisting antibodies that offer protection from other pathogens. Science 2019; 366:599-606. [PMID: 31672891 PMCID: PMC8590458 DOI: 10.1126/science.aay6485] [Citation(s) in RCA: 254] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/30/2019] [Indexed: 12/25/2022]
Abstract
Measles virus is directly responsible for more than 100,000 deaths yearly. Epidemiological studies have associated measles with increased morbidity and mortality for years after infection, but the reasons why are poorly understood. Measles virus infects immune cells, causing acute immune suppression. To identify and quantify long-term effects of measles on the immune system, we used VirScan, an assay that tracks antibodies to thousands of pathogen epitopes in blood. We studied 77 unvaccinated children before and 2 months after natural measles virus infection. Measles caused elimination of 11 to 73% of the antibody repertoire across individuals. Recovery of antibodies was detected after natural reexposure to pathogens. Notably, these immune system effects were not observed in infants vaccinated against MMR (measles, mumps, and rubella), but were confirmed in measles-infected macaques. The reduction in humoral immune memory after measles infection generates potential vulnerability to future infections, underscoring the need for widespread vaccination.
Collapse
Affiliation(s)
- Michael J Mina
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA 02115, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tomasz Kula
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yumei Leng
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Mamie Li
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 CN, Rotterdam, Netherlands
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, 00014 Helsinki, Finland
| | - Heli Siljander
- Children's Hospital, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, 00014 Helsinki, Finland
| | - Marian Rewers
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Denver, CO 80045, USA
| | - David F Choy
- Genentech Inc., South San Francisco, CA 94080, USA
| | | | - H Benjamin Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ashley N Nelson
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rik L de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 CN, Rotterdam, Netherlands
| | - Stephen J Elledge
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA 02115, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
29
|
Haralambieva IH, Kennedy RB, Ovsyannikova IG, Schaid DJ, Poland GA. Current perspectives in assessing humoral immunity after measles vaccination. Expert Rev Vaccines 2019; 18:75-87. [PMID: 30585753 PMCID: PMC6413513 DOI: 10.1080/14760584.2019.1559063] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/10/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Repeated measles outbreaks in countries with relatively high vaccine coverage are mainly due to failure to vaccinate and importation; however, cases in immunized individuals exist raising questions about suboptimal measles vaccine-induced humoral immunity and/or waning immunity in a low measles-exposure environment. AREAS COVERED The plaque reduction neutralization measurement of functional measles-specific antibodies correlates with protection is the gold standard in measles serology, but it does not assess cellular-immune or other parameters that may be associated with durable and/or protective immunity after vaccination. Additional correlates of protection and long-term immunity and new determinants/signatures of vaccine responsiveness such as specific CD46 and IFI44L genetic variants associated with neutralizing antibody titers after measles vaccination are under investigation. Current and future systems biology studies, coupled with new technology/assays and analytical approaches, will lead to an increasingly sophisticated understanding of measles vaccine-induced humoral immunity and will identify 'signatures' of protective and durable immune responses. EXPERT OPINION This will translate into the development of highly predictive assays of measles vaccine efficacy, effectiveness, and durability for prospective identification of potential low/non-responders and susceptible individuals who require additional vaccine doses. Such new advances may drive insights into the development of new/improved vaccine formulations and delivery systems.
Collapse
Affiliation(s)
| | - Richard B Kennedy
- a Mayo Clinic Vaccine Research Group , Mayo Clinic , Rochester , MN , USA
| | | | - Daniel J Schaid
- a Mayo Clinic Vaccine Research Group , Mayo Clinic , Rochester , MN , USA
- b Department of Health Sciences Research , Mayo Clinic , Rochester , MN , USA
| | - Gregory A Poland
- a Mayo Clinic Vaccine Research Group , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
30
|
Studies into the mechanism of measles-associated immune suppression during a measles outbreak in the Netherlands. Nat Commun 2018; 9:4944. [PMID: 30470742 PMCID: PMC6251901 DOI: 10.1038/s41467-018-07515-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/07/2018] [Indexed: 01/08/2023] Open
Abstract
Measles causes a transient immune suppression, leading to increased susceptibility to opportunistic infections. In experimentally infected non-human primates (NHPs) measles virus (MV) infects and depletes pre-existing memory lymphocytes, causing immune amnesia. A measles outbreak in the Dutch Orthodox Protestant community provided a unique opportunity to study the pathogenesis of measles immune suppression in unvaccinated children. In peripheral blood mononuclear cells (PBMC) of prodromal measles patients, we detected MV-infected memory CD4+ and CD8+ T cells and naive and memory B cells at similar levels as those observed in NHPs. In paired PBMC collected before and after measles we found reduced frequencies of circulating memory B cells and increased frequencies of regulatory T cells and transitional B cells after measles. These data support our immune amnesia hypothesis and offer an explanation for the previously observed long-term effects of measles on host resistance. This study emphasises the importance of maintaining high measles vaccination coverage. The mechanisms by which measles virus infection induces transient immune suppression in humans are poorly understood. Here, Laksono and colleagues characterise the pathogenesis of measles-associated immune suppression in unvaccinated children, and shed new light on the long-term effects of measles on the host.
Collapse
|
31
|
O’Malley KJ, Bowling JL, Stinson E, Cole KS, Mann BJ, Namjoshi P, Hazlett KRO, Barry EM, Reed DS. Aerosol prime-boost vaccination provides strong protection in outbred rabbits against virulent type A Francisella tularensis. PLoS One 2018; 13:e0205928. [PMID: 30346998 PMCID: PMC6197691 DOI: 10.1371/journal.pone.0205928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
Tularemia, also known as rabbit fever, is a severe zoonotic disease in humans caused by the gram-negative bacterium Francisella tularensis (Ft). While there have been a number of attempts to develop a vaccine for Ft, few candidates have advanced beyond experiments in inbred mice. We report here that a prime-boost strategy with aerosol delivery of recombinant live attenuated candidate Ft S4ΔaroD offers significant protection (83% survival) in an outbred animal model, New Zealand White rabbits, against aerosol challenge with 248 cfu (11 LD50) of virulent type A Ft SCHU S4. Surviving rabbits given two doses of the attenuated strains by aerosol did not exhibit substantial post-challenge fevers, changes in erythrocyte sedimentation rate or in complete blood counts. At a higher challenge dose (3,186 cfu; 139 LD50), protection was still good with 66% of S4ΔaroD-vaccinated rabbits surviving while 50% of S4ΔguaBA vaccinated rabbits also survived challenge. Pre-challenge plasma IgG titers against Ft SCHU S4 corresponded with survival time after challenge. Western blot analysis found that plasma antibody shifted from predominantly targeting Ft O-antigen after the prime vaccination to other antigens after the boost. These results demonstrate the superior protection conferred by a live attenuated derivative of virulent F. tularensis, particularly when given in an aerosol prime-boost regimen.
Collapse
Affiliation(s)
- Katherine J. O’Malley
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Jennifer L. Bowling
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Elizabeth Stinson
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kelly S. Cole
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Barbara J. Mann
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, United States of America
| | - Prachi Namjoshi
- Department for Immunology & Microbial Diseases, Albany Medical College, Albany, NY, United States of America
| | - Karsten R. O. Hazlett
- Department for Immunology & Microbial Diseases, Albany Medical College, Albany, NY, United States of America
| | - Eileen M. Barry
- Center for Vaccine Development, University of Maryland Baltimore, Baltimore, MD, United States of America
| | - Douglas S. Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
32
|
Macrophages and Dendritic Cells Are the Predominant Cells Infected in Measles in Humans. mSphere 2018; 3:3/3/e00570-17. [PMID: 29743202 PMCID: PMC5956143 DOI: 10.1128/msphere.00570-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/19/2018] [Indexed: 01/08/2023] Open
Abstract
Characterization of human measles cases is essential in order to better assess the data generated in model systems of morbillivirus infection. To this end, we collected formalin-fixed tissue samples from 23 natural measles cases from different areas in the world and different phases of disease ranging from prodromal and acute measles to a persistent infection in an immunocompromised subject. We show that the vast majority of measles virus (MV)-infected cells in epithelia were intraepithelial immune cells that were, in most cases, positive for the CD11c myeloid cell marker. Small numbers of measles virus-infected cytokeratin-positive epithelial cells were also detected in bronchial and appendix epithelia. Dissolution and disruption of uninfected and MV-infected alveolar and bronchial epithelia were prominent features of the measles cases, especially in the established and late phases of the disease. In some instances, this was associated with the formation of MV-infected multinucleated giant cells which expressed CD11c and/or macrophage cell marker 68, a pathological feature also prominently observed in closely associated mucosa-associated lymphoid tissue. Collectively, these data show that resident and inflammatory infiltrating immune cells alter the architecture of respiratory tract epithelia and highlight the necessity for additional research into the function(s) and expression of nectin-4 in human tissues.IMPORTANCE We have brought together a unique collection of 23 human cases of measles infection and studied the types of cells that are infected. This work has not been done with modern technologies such as double labeling with antibodies and confocal microscopy in human cases primarily due to the fact that it is difficult to obtain the material because, fortunately, measles is fatal in only a very small fraction of infected patients. During the past decades, the receptors for measles virus have been elucidated and monkey models have been developed. We found that, in most cases, independently of whether the tissues were obtained early or later in the infection, the primary cell types that were infected were those of the immune system such as lymphocytes, macrophages, and dendritic cells. A very small number of epithelial cells were also found to be infected.
Collapse
|
33
|
Isomura M, Yamada K, Noguchi K, Nishizono A. Near-infrared fluorescent protein iRFP720 is optimal for in vivo fluorescence imaging of rabies virus infection. J Gen Virol 2017; 98:2689-2698. [PMID: 29039733 DOI: 10.1099/jgv.0.000950] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In vivo imaging is a noninvasive method that enables real-time monitoring of viral infection dynamics in a small animal, which allows a better understanding of viral pathogenesis. In vivo bioluminescence imaging of virus infection is widely used but, despite its advantage over bioluminescence that no substrate administration is required, fluorescence imaging is not used because of severe autofluorescence. Recently, several far-red and near-infrared (NIR) fluorescent proteins (FPs) have been developed and shown to be useful for whole-body fluorescence imaging. Here, we report comparative testing of far-red and NIR FPs in the imaging of rabies virus (RABV) infection. Using the highly neuroinvasive 1088 strain, we generated recombinant RABV that expressed FPs such as Katushka2S, E2-Crimson, iRFP670 or iRFP720. After intracerebral inoculation to nude mice, the 1088 strain expressing iRFP720, the most red-shifted FP, was detected the earliest with the highest signal-to-noise ratio using a filter set for >700 nm, in which the background signal level was very low. Furthermore, we could also track viral dissemination from the spinal cord to the brain in nude mice after intramuscular inoculation of iRFP720-expressing 1088 into the hind limb. Hence, we conclude that the NIR FP iRFP720 used with a filter set for >700 nm is useful for in vivo fluorescence imaging not only for RABV infection but also for other virus infections. Our findings will also be useful for developing dual-optical imaging of virus-host interaction dynamics using bioluminescence reporter mice for inflammation imaging.
Collapse
Affiliation(s)
- Minori Isomura
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu city, Oita, Japan
| | - Kentaro Yamada
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu city, Oita, Japan.,Research Promotion Institute, Oita University, Yufu city, Oita, Japan
| | - Kazuko Noguchi
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu city, Oita, Japan.,Present address: Department of Food Science and Technology, Minami Kyusyu University, Miyazaki city, Miyazaki, Japan
| | - Akira Nishizono
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu city, Oita, Japan
| |
Collapse
|
34
|
|