1
|
Bliss CM, Nachbagauer R, Mariottini C, Cuevas F, Feser J, Naficy A, Bernstein DI, Guptill J, Walter EB, Berlanda-Scorza F, Innis BL, García-Sastre A, Palese P, Krammer F, Coughlan L. A chimeric haemagglutinin-based universal influenza virus vaccine boosts human cellular immune responses directed towards the conserved haemagglutinin stalk domain and the viral nucleoprotein. EBioMedicine 2024; 104:105153. [PMID: 38805853 PMCID: PMC11154122 DOI: 10.1016/j.ebiom.2024.105153] [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: 10/25/2023] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The development of a universal influenza virus vaccine, to protect against both seasonal and pandemic influenza A viruses, is a long-standing public health goal. The conserved stalk domain of haemagglutinin (HA) is a promising vaccine target. However, the stalk is immunosubdominant. As such, innovative approaches are required to elicit robust immunity against this domain. In a previously reported observer-blind, randomised placebo-controlled phase I trial (NCT03300050), immunisation regimens using chimeric HA (cHA)-based immunogens formulated as inactivated influenza vaccines (IIV) -/+ AS03 adjuvant, or live attenuated influenza vaccines (LAIV), elicited durable HA stalk-specific antibodies with broad reactivity. In this study, we sought to determine if these vaccines could also boost T cell responses against HA stalk, and nucleoprotein (NP). METHODS We measured interferon-γ (IFN-γ) responses by Enzyme-Linked ImmunoSpot (ELISpot) assay at baseline, seven days post-prime, pre-boost and seven days post-boost following heterologous prime:boost regimens of LAIV and/or adjuvanted/unadjuvanted IIV-cHA vaccines. FINDINGS Our findings demonstrate that immunisation with adjuvanted cHA-based IIVs boost HA stalk-specific and NP-specific T cell responses in humans. To date, it has been unclear if HA stalk-specific T cells can be boosted in humans by HA-stalk focused universal vaccines. Therefore, our study will provide valuable insights for the design of future studies to determine the precise role of HA stalk-specific T cells in broad protection. INTERPRETATION Considering that cHA-based vaccines also elicit stalk-specific antibodies, these data support the further clinical advancement of cHA-based universal influenza vaccine candidates. FUNDING This study was funded in part by the Bill and Melinda Gates Foundation (BMGF).
Collapse
Affiliation(s)
- Carly M Bliss
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Cancer & Genetics and Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Mariottini
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frans Cuevas
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jodi Feser
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Abdi Naficy
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - David I Bernstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffrey Guptill
- Duke Early Phase Clinical Research Unit, Duke Clinical Research Institute, Durham, NC, USA
| | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Bruce L Innis
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; University of Maryland School of Medicine, Department of Microbiology and Immunology, Baltimore, MD 21201, USA; University of Maryland School of Medicine, Center for Vaccine Development and Global Health (CVD), Baltimore, MD 21201, USA.
| |
Collapse
|
2
|
Janssens Y, Joye J, Waerlop G, Clement F, Leroux-Roels G, Leroux-Roels I. The role of cell-mediated immunity against influenza and its implications for vaccine evaluation. Front Immunol 2022; 13:959379. [PMID: 36052083 PMCID: PMC9424642 DOI: 10.3389/fimmu.2022.959379] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/27/2022] [Indexed: 12/25/2022] Open
Abstract
Influenza vaccines remain the most effective tools to prevent flu and its complications. Trivalent or quadrivalent inactivated influenza vaccines primarily elicit antibodies towards haemagglutinin and neuraminidase. These vaccines fail to induce high protective efficacy, in particular in older adults and immunocompromised individuals and require annual updates to keep up with evolving influenza strains (antigenic drift). Vaccine efficacy declines when there is a mismatch between its content and circulating strains. Current correlates of protection are merely based on serological parameters determined by haemagglutination inhibition or single radial haemolysis assays. However, there is ample evidence showing that these serological correlates of protection can both over- or underestimate the protective efficacy of influenza vaccines. Next-generation universal influenza vaccines that induce cross-reactive cellular immune responses (CD4+ and/or CD8+ T-cell responses) against conserved epitopes may overcome some of the shortcomings of the current inactivated vaccines by eliciting broader protection that lasts for several influenza seasons and potentially enhances pandemic preparedness. Assessment of cellular immune responses in clinical trials that evaluate the immunogenicity of these new generation vaccines is thus of utmost importance. Moreover, studies are needed to examine whether these cross-reactive cellular immune responses can be considered as new or complementary correlates of protection in the evaluation of traditional and next-generation influenza vaccines. An overview of the assays that can be applied to measure cell-mediated immune responses to influenza with their strengths and weaknesses is provided here.
Collapse
Affiliation(s)
- Yorick Janssens
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
| | - Jasper Joye
- Center for Vaccinology (CEVAC), Ghent University Hospital, Ghent, Belgium
| | - Gwenn Waerlop
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
| | - Frédéric Clement
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
| | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
- Center for Vaccinology (CEVAC), Ghent University Hospital, Ghent, Belgium
| | - Isabel Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
- Center for Vaccinology (CEVAC), Ghent University Hospital, Ghent, Belgium
- *Correspondence: Isabel Leroux-Roels,
| |
Collapse
|
3
|
Wang T, Wei F, Liu L, Sun Y, Song J, Wang M, Yang J, Li C, Liu J. Recombinant HA1-ΔfliC enhances adherence to respiratory epithelial cells and promotes the superiorly protective immune responses against H9N2 influenza virus in chickens. Vet Microbiol 2021; 262:109238. [PMID: 34560407 DOI: 10.1016/j.vetmic.2021.109238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/11/2021] [Indexed: 01/17/2023]
Abstract
H9N2 subtype avian influenza virus (AIV) is an ongoing threat causing substantial loss to the poultry industry and thus necessitating the development of safe and effective vaccines against AIV. Given that inactivated vaccines are less effective in activating the mucosal immune system, we aimed to generate a vaccine that can actively engage the mucosal immunity which is the front line of the immune system. We generated a group of flagellin-based hemagglutinin globular head (HA1) fusion proteins and characterized their immunogenicity and efficacy. We found that Salmonella typhimurium flagellin (fliC) lacking the hypervariable domain (called herein as HA1-ΔfliC) was recognized by TLR5 and induced a moderate innate immune response compared to N-terminus of fliC (HA1-fliC) and C-terminus of fliC (fliC-HA1). The HA1-ΔfliC protein had increased adherence to the nasal cavity and trachea than HA1-fliC and fliC-HA1 and significantly increased the HA-specific sIgA titers. Our in vivo results revealed that chickens treated with HA1-ΔfliC had a significantly reduced level of viral loads in the cloaca and throat compared with chickens treated with inactivated vaccine. Overall, these results revealed that HA1-ΔfliC can protect chickens against H9N2 AIV by eliciting the efficient mucosal immune responses.
Collapse
Affiliation(s)
- Tong Wang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Fanhua Wei
- College of Agriculture, Ningxia University, Yinchuan, 750021, China
| | - Litao Liu
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Yan Sun
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Jingwei Song
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Mingyang Wang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Jizhe Yang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China
| | - Chengye Li
- College of Agriculture, Ningxia University, Yinchuan, 750021, China
| | - Jinhua Liu
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100094, China.
| |
Collapse
|
4
|
N-2-Hydroxypropyl Trimethyl Ammonium Chloride Chitosan as Adjuvant Enhances the Immunogenicity of a VP2 Subunit Vaccine against Porcine Parvovirus Infection in Sows. Vaccines (Basel) 2021; 9:vaccines9091027. [PMID: 34579264 PMCID: PMC8472385 DOI: 10.3390/vaccines9091027] [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: 08/16/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 12/16/2022] Open
Abstract
Porcine parvovirus (PPV) is the most important infectious agent causing infertility in pigs, which can be prevented by routine vaccination. Successful vaccination depends on the association with potent adjuvants that can enhance the immunogenicity of antigen and activate the immune system. Polysaccharide adjuvant has low toxicity and high safety, and they can enhance the humoral, cellular and mucosal immune responses. In the present study, we prepared the VP2 protein subunit vaccine against PPV (PPV/VP2/N-2-HACC) using water-soluble N-2-Hydroxypropyl trimethyl ammonium chloride chitosan (N-2-HACC) as the vaccine adjuvant, and the ability of the PPV/VP2/N-2-HACC to induce immune responses and protect sows from PPV infection was evaluated. In vivo immunization showed that the sows immunized with the PPV/VP2/N-2-HACC by intramuscular injection produced higher HI antibody levels and long-term immune protection compared with the other groups, while the subunit vaccine did not stimulate the proliferation of CD4+ and CD8+ T lymphocytes to trigger the secretion of higher levels of IL-2, IL-4, IFN-α, IFN-β, and IFN-γ, indicating that the PPV/VP2/N-2-HACC mainly induced humoral immunity rather than cellular immunity. PPV was not detected in the viscera of the sows immunized with the PPV/VP2/N-2-HACC, and the protective efficacy was 100%. Collectively, our findings suggested that the N-2-HACC was a potential candidate adjuvant, and the PPV/VP2/N-2-HACC had immense application value for the control of PPV.
Collapse
|
5
|
Kerstetter LJ, Buckley S, Bliss CM, Coughlan L. Adenoviral Vectors as Vaccines for Emerging Avian Influenza Viruses. Front Immunol 2021; 11:607333. [PMID: 33633727 PMCID: PMC7901974 DOI: 10.3389/fimmu.2020.607333] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
It is evident that the emergence of infectious diseases, which have the potential for spillover from animal reservoirs, pose an ongoing threat to global health. Zoonotic transmission events have increased in frequency in recent decades due to changes in human behavior, including increased international travel, the wildlife trade, deforestation, and the intensification of farming practices to meet demand for meat consumption. Influenza A viruses (IAV) possess a number of features which make them a pandemic threat and a major concern for human health. Their segmented genome and error-prone process of replication can lead to the emergence of novel reassortant viruses, for which the human population are immunologically naïve. In addition, the ability for IAVs to infect aquatic birds and domestic animals, as well as humans, increases the likelihood for reassortment and the subsequent emergence of novel viruses. Sporadic spillover events in the past few decades have resulted in human infections with highly pathogenic avian influenza (HPAI) viruses, with high mortality. The application of conventional vaccine platforms used for the prevention of seasonal influenza viruses, such as inactivated influenza vaccines (IIVs) or live-attenuated influenza vaccines (LAIVs), in the development of vaccines for HPAI viruses is fraught with challenges. These issues are associated with manufacturing under enhanced biosafety containment, and difficulties in propagating HPAI viruses in embryonated eggs, due to their propensity for lethality in eggs. Overcoming manufacturing hurdles through the use of safer backbones, such as low pathogenicity avian influenza viruses (LPAI), can also be a challenge if incompatible with master strain viruses. Non-replicating adenoviral (Ad) vectors offer a number of advantages for the development of vaccines against HPAI viruses. Their genome is stable and permits the insertion of HPAI virus antigens (Ag), which are expressed in vivo following vaccination. Therefore, their manufacture does not require enhanced biosafety facilities or procedures and is egg-independent. Importantly, Ad vaccines have an exemplary safety and immunogenicity profile in numerous human clinical trials, and can be thermostabilized for stockpiling and pandemic preparedness. This review will discuss the status of Ad-based vaccines designed to protect against avian influenza viruses with pandemic potential.
Collapse
Affiliation(s)
- Lucas J. Kerstetter
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Stephen Buckley
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Carly M. Bliss
- Division of Cancer & Genetics, Division of Infection & Immunity, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
6
|
Wang C, Wang Z, Rosner GL, Huh WK, Roden RBS, Bae S. A batch-effect adjusted Simon's two-stage design for cancer vaccine clinical studies. Biometrics 2020; 77:1075-1088. [PMID: 32822525 DOI: 10.1111/biom.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/25/2020] [Accepted: 08/10/2020] [Indexed: 11/29/2022]
Abstract
In the development of cancer treatment vaccines, phase II clinical studies are conducted to examine the efficacy of a vaccine in order to screen out vaccines with minimal activity. Immune responses are commonly used as the primary endpoint for assessing vaccine efficacy. With respect to study design, Simon's two-stage design is a popular format for phase II cancer clinical studies because of its simplicity and ethical considerations. Nonetheless, it is not straightforward to apply Simon's two-stage design to cancer vaccine studies when performing immune assays in batches, as outcomes from multiple patients may be correlated with each other in the presence of batch effects. This violates the independence assumption of Simon's two-stage design. In this paper, we numerically explore the impact of batch effects on Simon's two-stage design, propose a batch-effect adjusted Simon's two-stage design, demonstrate the proposed design by both a simulation study and a therapeutic human papillomavirus vaccine trial, and briefly introduce a software that implements the proposed design.
Collapse
Affiliation(s)
- Chenguang Wang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Zhixin Wang
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gary L Rosner
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Warner K Huh
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Richard B S Roden
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Sejong Bae
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
7
|
Boccard M, Albert-Vega C, Mouton W, Durieu I, Brengel-Pesce K, Venet F, Trouillet-Assant S, Ader F. [Functional immunoassays in the setting of infectious risk and immunosuppressive therapy of non-HIV immunocompromised patients]. Rev Med Interne 2020; 41:545-551. [PMID: 32624260 DOI: 10.1016/j.revmed.2020.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/24/2020] [Accepted: 04/09/2020] [Indexed: 11/25/2022]
Abstract
The holistic approach of the human immune system is based on the study of its components collectively driving a functional response to an immunogenic stimulus. To appreciate a specific immune dysfunction, a condition is mimicked ex vivo and the immune response induced is assessed. The application field of such assays are broad and expanding, from the diagnosis of primary and secondary immunodeficiencies, immunotherapy for cancer to the management of patients at-risk for infections and vaccination. These assays are immune monitoring tools that may contribute to a personalised and precision medicine. The purpose of this review is to describe immune functional assays available in the setting of non-HIV acquired immune deficiency. First, we will address the use of theses assays in the diagnosis of opportunistic infections such as viral reactivation. Secondly, we will report the usefulness of these assays to assess vaccine efficacy and to manage immunosuppressive therapies.
Collapse
Affiliation(s)
- M Boccard
- Centre International de Recherche en Infectiologie (CIRI), Inserm 1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, F-69007, Lyon, France; Département de médecine interne et vasculaire, centre hospitalier Lyon Sud, Hospices civils de Lyon, 69310 Pierre-Bénite, France; Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France.
| | - C Albert-Vega
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France
| | - W Mouton
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France; Laboratoire virologie et pathologies humaines (VirPath), faculté de médecine Lyon Est, université Claude-Bernard Lyon 1, 69008 Lyon, France
| | - I Durieu
- Département de médecine interne et vasculaire, centre hospitalier Lyon Sud, Hospices civils de Lyon, 69310 Pierre-Bénite, France
| | - K Brengel-Pesce
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France
| | - F Venet
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France; Laboratoire d'immunologie, hôpital Édouard-Herriot, Hospices civils de Lyon, 69003 Lyon, France; EA7426 Pathophysiology of injury-induced immunosuppression, université Claude-Bernard Lyon 1, 69008 Lyon, France
| | - S Trouillet-Assant
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France; Laboratoire virologie et pathologies humaines (VirPath), faculté de médecine Lyon Est, université Claude-Bernard Lyon 1, 69008 Lyon, France
| | - F Ader
- Centre International de Recherche en Infectiologie (CIRI), Inserm 1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, F-69007, Lyon, France; Département des maladies infectieuses et tropicales, hôpital de la Croix-Rousse, Hospices civils de Lyon, 69004 Lyon, France
| |
Collapse
|
8
|
Obeid MA, Teeravatcharoenchai T, Connell D, Niwasabutra K, Hussain M, Carter K, Ferro VA. Examination of the effect of niosome preparation methods in encapsulating model antigens on the vesicle characteristics and their ability to induce immune responses. J Liposome Res 2020; 31:195-202. [PMID: 32396752 DOI: 10.1080/08982104.2020.1768110] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Niosome nanoparticles can be prepared using different methods, each of which can affect the size and homogeneity of the prepared particles. The aim of this study was to establish if the method of preparation impacted on the prepared vesicles when loaded with a model protein and the type of immune responses induced to the vaccine antigen. Niosomes were prepared using both the traditional thin film hydration (TFH) technique and the microfluidic mixing (MM) technique. Influenza antigen was then entrapped in the niosomes and formulations tested for their ability to induce in vivo immune responses in immunised BALB/c mice. Niosomes prepared by MM had a mean size of 157 ± 1.8 nm and were significantly more uniform compared with the niosomes prepared using TFH (mean size 388 ± 10 nm). Niosomes play a key role as an adjuvant to help raise high antibody immune responses. This was confirmed in this study since animals treated with both types of niosomes and antigen were more responsive than unentrapped (free) antigen. Cytokine analysis showed that the TFH niosomes induced a Th1 immune response by raising IgG2a and high levels of IFN-ɣ, while the MM niosomes induced a Th2 immune response by inducing IgG1 (p < .05). These results confirmed that the method of preparation of the niosomes nanoparticles induced different immune responses and the average particle size of the niosomes differed depending on the method of manufacture. This indicates that particle size and uniformity are of importance and should be taken into consideration when designing an oral based delivery system for vaccine delivery.
Collapse
Affiliation(s)
- Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | | | - David Connell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Kanidta Niwasabutra
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Muattaz Hussain
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Katharine Carter
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Valerie A Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
9
|
Zhang ZS, Gu Y, Liu BG, Tang H, Hua Y, Wang J. Oncogenic role of Tc17 cells in cervical cancer development. World J Clin Cases 2020; 8:11-19. [PMID: 31970165 PMCID: PMC6962079 DOI: 10.12998/wjcc.v8.i1.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/18/2019] [Accepted: 11/30/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND As one of the subsets of CD8+ T cells, Tc17 cells have recently been identified and are characterized by the secretion of interleukin (IL)-17, which is related to inflammatory diseases.
AIM To assess the status of Tc17 cells in cervical cancer and investigate the biological function of Tc17 cells in cervical cancer development.
METHODS Flow cytometry assay, immunohistochemistry, and immunofluorescence were performed to detect the levels and phenotype of Tc17 cells in blood and tumor samples from patients with cervical cancer. Prior to cell suspension culture, ELISA was carried out to measure the production of IL-6, IL-1β, IL-23, CXCL12, and IL-17 in tumor tissue supernatant and co-cultured supernatant of patients with cervical cancer. In addition, multivariate analysis was performed to identify factors associated with overall survival using the Cox proportional hazards model.
RESULTS Compared with normal tissues, Tc17 cells specifically accumulated in tumor tissues of cervical cancer patients. Cancer cells produced a greater amount of IL-6, IL-1β, and IL-23, which in turn promoted Tc17 cell polarization. Unlike the traditional cytotoxic CD8+ T cells, Tc17 cells secreted IL-17, which subsequently promoted CXCL12 expression in tumor cells, eventually enhancing the proliferation and migration of tumor cells. Thus, the ratio of tumor-infiltrating Tc17 cells was highly correlated with poor clinical outcome in patients with cervical cancer.
CONCLUSION Our data identified the oncogenic role of Tc17 cells in the development of cervical cancer. We propose that the ratio of Tc17 cells may be a useful index in the prognosis of patients with cervical cancer.
Collapse
Affiliation(s)
- Zun-Sheng Zhang
- Department of Obstetrics and Gynecology, Shanghai Seventh People’s Hospital, Shanghai 200120, China
| | - Ying Gu
- Department of Obstetrics and Gynecology, Shanghai Seventh People’s Hospital, Shanghai 200120, China
| | - Bing-Gang Liu
- Department of Obstetrics and Gynecology, Shanghai Seventh People’s Hospital, Shanghai 200120, China
| | - Hong Tang
- Department of Obstetrics and Gynecology, Shanghai Seventh People’s Hospital, Shanghai 200120, China
| | - Yu Hua
- Department of Obstetrics and Gynecology, Shanghai Seventh People’s Hospital, Shanghai 200120, China
| | - Jun Wang
- Department of Obstetrics and Gynecology, Shanghai Seventh People’s Hospital, Shanghai 200120, China
| |
Collapse
|
10
|
Ozkan B, Budama-Kilinc Y, Cakir-Koc R, Mese S, Badur S. Application of an immunoglobulin Y-alkaline phosphatase bioconjugate as a diagnostic tool for influenza A virus. Bioengineered 2019; 10:33-42. [PMID: 30913952 PMCID: PMC6527078 DOI: 10.1080/21655979.2019.1586054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The diagnosis of influenza A virus is essential since it can be confused with influenza A like illness and lead to inaccurate drug prescription. In this study, the M2e peptide, a strategic antigen that is conserved in all virus subtypes, was used as a diagnostic marker of influenza A. For the first time, M2e-specific IgY antibody was covalently conjugated to alkaline phosphatase (ALP) enzyme in the presence of glutaraldehyde. The antibody-enzyme bioconjugate was characterized by fluorescence and Fourier-transform infrared spectroscopy. Subsequently, the diagnostic value of this bioconjugate was evaluated by direct sandwich ELISA using nasopharyngeal swab samples positive/negative for H1N1 and H3N2, which were previously analyzed by rRT-PCR for influenza. In conclusion, the M2e-specific IgY-ALP bioconjugate demonstrated positive results for Influenza A in samples that were diagnosed as Influenza A via the RT-PCR method.
Collapse
Affiliation(s)
- Busra Ozkan
- a Department of Bioengineering, Chemical and Metallurgical Engineering Faculty , Yildiz Technical University , Istanbul , Turkey
| | - Yasemin Budama-Kilinc
- a Department of Bioengineering, Chemical and Metallurgical Engineering Faculty , Yildiz Technical University , Istanbul , Turkey
| | - Rabia Cakir-Koc
- a Department of Bioengineering, Chemical and Metallurgical Engineering Faculty , Yildiz Technical University , Istanbul , Turkey
| | - Sevim Mese
- b Department of Virology and Fundamental Immunology, Istanbul Medical Faculty , Istanbul University , Istanbul , Turkey
| | - Selim Badur
- c GlaxoSmithKline-Vaccine , Istanbul , Turkey
| |
Collapse
|
11
|
Gianchecchi E, Torelli A, Montomoli E. The use of cell-mediated immunity for the evaluation of influenza vaccines: an upcoming necessity. Hum Vaccin Immunother 2019; 15:1021-1030. [PMID: 30614754 PMCID: PMC6605831 DOI: 10.1080/21645515.2019.1565269] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Influenza vaccines are a fundamental tool for preventing the disease and reducing its consequences, particularly in specific high-risk groups. In order to be licensed, influenza vaccines have to meet strict criteria established by European Medicines Agency. Although the licensure of influenza vaccines started 65 years ago, Hemagglutination Inhibition and Single Radial Hemolysis are the only serological assays that can ascertain correlates of protection. However, they present evident limitations. The present review focuses on the evaluation of cell-mediated immunity (CMI), which plays an important role in the host immune response in protecting against virus-related illness and in the establishment of long-term immunological memory. Although correlates of protection are not currently available for CMI, it would be advisable to investigate this kind of immunological response for the evaluation of next-generation vaccines.
Collapse
Affiliation(s)
| | - A Torelli
- a VisMederi srl , Siena , Italy.,b Department of Life Sciences , University of Siena , Siena , Italy
| | - E Montomoli
- a VisMederi srl , Siena , Italy.,c Department of Molecular and Developmental Medicine , University of Siena , Siena , Italy
| |
Collapse
|
12
|
Highly conserved hemagglutinin peptides of H1N1 influenza virus elicit immune response. 3 Biotech 2018; 8:492. [PMID: 30498665 DOI: 10.1007/s13205-018-1509-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 11/09/2018] [Indexed: 01/24/2023] Open
Abstract
In the current study, two highly conserved (> 90%) H1N1 hemagglutinin peptides STDTVDTVLEKNVTVTHSVNL (H1) and KVNSVIEKMNTQFTAVGKEF (H2) containing multiple T-cell epitopes have been assessed for their immunogenic potential in vitro, subjecting peripheral blood mononuclear cells from healthy volunteers to repetitive stimulation of chemically synthesised H1 and H2 peptides, and measuring their interferon (IFN)-γ level (ELISA) and proliferation (MTT assay). Further, these peptides were analysed for their binding affinity with 18 different human leukocyte antigen (HLA) class I and II by means of molecular docking. All seven samples tested for H1- and H2-induced IFN-γ secretion were found to have enhanced IFN-γ production. Six (H1) and five (H2) samples have shown proliferative response compared to unstimulated cells. Peptide-induced IFN-γ secretion and proliferation in healthy samples represent the immunogenic potential of these peptides. Further, molecular docking results reveal that the peptides have comparable binding energy to that of native bound peptide for both HLA classes which indicates that these peptides have the capability to be presented by different HLA molecules required for T-cell response. Hence, these conserved immunogenic hemagglutinin peptides are potential candidates for influenza vaccine development.
Collapse
|
13
|
Yan LM, Li OTW, Poh CM, Perera RAPM, Valkenburg SA, Peiris M, Poon LLM. Combined use of live-attenuated and inactivated influenza vaccines to enhance heterosubtypic protection. Virology 2018; 525:73-82. [PMID: 30248524 DOI: 10.1016/j.virol.2018.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 01/04/2023]
Abstract
The limited protection of current commerical vaccines necessitates the investigation of novel vaccine strategies for unpredictable outbreaks. To investigate the feasibility of using vaccines derived from Group 1 influenza A virus to induce broadly cross-reactive immune responses against multiple influenza subtypes, we tested a panel of sequential 4-dose immunization regimens in mice. Mice were treated with inactivated (seasonal H1N1, pandemic H1N1 and H5N1) and vaccinia virus-based H5N1 live-attenuated vaccines in different combinations. Mice were then challenged by viruses of either Group 1 (H1N1) or Group 2 (H3N2, H7N7) influenza virus. All studied sequential 4-dose vaccinations could induce some degrees of heterosubtypic protection in mice. Amongst all these regimens, the combined use of inactivated and live-attenuated vaccines could achieve the best heterologous protection. These results highlight the synergistic effect of combining different vaccine platforms to enhance heterosubtypic protection against influenza viruses.
Collapse
Affiliation(s)
- Li-Meng Yan
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Olive T W Li
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Chek M Poh
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Ranawaka A P M Perera
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie A Valkenburg
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China; HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Malik Peiris
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Leo L M Poon
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
14
|
Bowyer G, Rampling T, Powlson J, Morter R, Wright D, Hill AVS, Ewer KJ. Activation-induced Markers Detect Vaccine-Specific CD4⁺ T Cell Responses Not Measured by Assays Conventionally Used in Clinical Trials. Vaccines (Basel) 2018; 6:vaccines6030050. [PMID: 30065162 PMCID: PMC6161310 DOI: 10.3390/vaccines6030050] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 12/27/2022] Open
Abstract
Immunogenicity of T cell-inducing vaccines, such as viral vectors or DNA vaccines and Bacillus Calmette-Guérin (BCG), are frequently assessed by cytokine-based approaches. While these are sensitive methods that have shown correlates of protection in various vaccine studies, they only identify a small proportion of the vaccine-specific T cell response. Responses to vaccination are likely to be heterogeneous, particularly when comparing prime and boost or assessing vaccine performance across diverse populations. Activation-induced markers (AIM) can provide a broader view of the total antigen-specific T cell response to enable a more comprehensive evaluation of vaccine immunogenicity. We tested an AIM assay for the detection of vaccine-specific CD4+ and CD8+ T cell responses in healthy UK adults vaccinated with viral vectored Ebola vaccine candidates, ChAd3-EBO-Z and MVA-EBO-Z. We used the markers, CD25, CD134 (OX40), CD274 (PDL1), and CD107a, to sensitively identify vaccine-responsive T cells. We compared the use of OX40+CD25+ and OX40+PDL1+ in CD4+ T cells and OX40+CD25+ and CD25+CD107a+ in CD8+ T cells for their sensitivity, specificity, and associations with other measures of vaccine immunogenicity. We show that activation-induced markers can be used as an additional method of demonstrating vaccine immunogenicity, providing a broader picture of the global T cell response to vaccination.
Collapse
Affiliation(s)
- Georgina Bowyer
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Tommy Rampling
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | | | - Richard Morter
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Daniel Wright
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
15
|
Flaxman A, Ewer KJ. Methods for Measuring T-Cell Memory to Vaccination: From Mouse to Man. Vaccines (Basel) 2018; 6:E43. [PMID: 30037078 PMCID: PMC6161152 DOI: 10.3390/vaccines6030043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/27/2022] Open
Abstract
The development of effective vaccines continues to be a key goal for public health bodies, governments, funding bodies and pharmaceutical companies. With new vaccines such as Shingrix targeting Shingles and Bexsero for Meningitis B, licensed in recent years, today's population can be protected from more infectious diseases than ever before. Despite this, we are yet to license vaccines for some of the deadliest endemic diseases affecting children, such as malaria. In addition, the threat of epidemics caused by emerging pathogens is very real as exemplified by the 2014⁻2016 Ebola outbreak. Most licensed vaccines provide efficacy through humoral immunity and correlates of protection often quantify neutralising antibody titre. The role of T-cells in vaccine efficacy is less well understood and more complex to quantify. Defining T-cell responses which afford protection also remains a challenge, although more sophisticated assays for assessing cell-mediated immunity with the potential for higher throughput and scalability are now available and warrant review. Here we discuss the benefits of multiparameter cytokine analysis and omics approaches compared with flow cytometric and ELISpot assays. We also review technical challenges unique to clinical trial studies, including assay validation across laboratories and availability of sample type. Measuring T-cell immunogenicity alongside humoral responses provides information on the breadth of immune responses induced by vaccination. Accurately enumerating and phenotyping T-cell immunogenicity to vaccination is key for the determination of immune correlates of protection. However, identifying such T-cell parameters remains challenging without a clear understanding of the immunological mechanisms by which a T-cell-mediated response induces protection.
Collapse
Affiliation(s)
- Amy Flaxman
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|
16
|
Keshavarz M, Dianat-Moghadam H, Sofiani VH, Karimzadeh M, Zargar M, Moghoofei M, Biglari H, Ghorbani S, Nahand JS, Mirzaei H. miRNA-based strategy for modulation of influenza A virus infection. Epigenomics 2018; 10:829-844. [DOI: 10.2217/epi-2017-0170] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Influenza A virus is known worldwide as a threat associated with human and livestock diseases. Hence, identification of physiological and molecular aspects of influenza A could contribute to better design of therapeutic approaches for reducing adverse effects associated with disease caused by this virus. miRNAs are epigenetic regulators playing important roles in many pathological processes that help in progression of influenza A. Besides miRNAs, exosomes have ememrged as other effective players in influenza A pathogenesis. Exosomes exert their effects via targeting their cargos (e.g., DNAs, mRNA, miRNAs and proteins) to recipient cells. Here, we summarized various roles of miRNAs and exosomes in influenza A pathogenesis. Moreover, we highlighted therapeutic applications of miRNAs and exosomes in influenza.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Dianat-Moghadam
- Department of Medical Biotechnology, Faculty of Advanced Medicine Sciences, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Mohammad Karimzadeh
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Zargar
- Department of Microbiology, Faculty of Science, Qom Branch, Islamic Azad University, Qom, Iran
| | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamed Biglari
- Department of Environmental Health Engineering, School of Public Health, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Saied Ghorbani
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Department of Biomaterials, Tissue Engineering & Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Montomoli E, Torelli A, Manini I, Gianchecchi E. Immunogenicity and Safety of the New Inactivated Quadrivalent Influenza Vaccine Vaxigrip Tetra: Preliminary Results in Children ≥6 Months and Older Adults. Vaccines (Basel) 2018; 6:E14. [PMID: 29518013 PMCID: PMC5874655 DOI: 10.3390/vaccines6010014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 02/26/2018] [Accepted: 03/05/2018] [Indexed: 12/31/2022] Open
Abstract
Since the mid-1980s, two lineages of influenza B viruses have been distinguished. These can co-circulate, limiting the protection provided by inactivated trivalent influenza vaccines (TIVs). This has prompted efforts to formulate quadrivalent influenza vaccines (QIVs), to enhance protection against circulating influenza B viruses. This review describes the results obtained from seven phase III clinical trials evaluating the immunogenicity, safety, and lot-to-lot consistency of a new quadrivalent split-virion influenza vaccine (Vaxigrip Tetra®) formulated by adding a second B strain to the already licensed TIV. Since Vaxigrip Tetra was developed by means of a manufacturing process strictly related to that used for TIV, the data on the safety profile of TIV are considered supportive of that of Vaxigrip Tetra. The safety and immunogenicity of Vaxigrip Tetra were similar to those of the corresponding licensed TIV. Moreover, the new vaccine elicits a superior immune response towards the additional strain, without affecting immunogenicity towards the other three strains. Vaxigrip Tetra is well tolerated, has aroused no safety concerns, and is recommended for the active immunization of individuals aged ≥6 months. In addition, preliminary data confirm its immunogenicity and safety even in children aged 6-35 months and its immunogenicity in older subjects (aged 66-80 years).
Collapse
Affiliation(s)
- Emanuele Montomoli
- VisMederi srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy.
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| | - Alessandro Torelli
- VisMederi srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy.
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| | - Elena Gianchecchi
- VisMederi srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy.
| |
Collapse
|
18
|
Ward BJ, Pillet S, Charland N, Trepanier S, Couillard J, Landry N. The establishment of surrogates and correlates of protection: Useful tools for the licensure of effective influenza vaccines? Hum Vaccin Immunother 2018; 14:647-656. [PMID: 29252098 PMCID: PMC5861778 DOI: 10.1080/21645515.2017.1413518] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The search for a test that can predict vaccine efficacy is an important part of any vaccine development program. Although regulators hesitate to acknowledge any test as a true ‘correlate of protection’, there are many precedents for defining ‘surrogate’ assays. Surrogates can be powerful tools for vaccine optimization, licensure, comparisons between products and development of improved products. When such tests achieve ‘reference’ status however, they can inadvertently become barriers to new technologies that do not work the same way as existing vaccines. This is particularly true when these tests are based upon circularly-defined ‘reference’ or, even worse, proprietary reagents. The situation with inactivated influenza vaccines is a good example of this phenomenon. The most frequently used tests to define vaccine-induced immunity are all serologic assays: hemagglutination inhibition (HI), single radial hemolysis (SRH) and microneutralization (MN). The first two, and particularly the HI assay, have achieved reference status and criteria have been established in many jurisdictions for their use in licensing new vaccines and to compare the performance of different vaccines. However, all of these assays are based on biological reagents that are notoriously difficult to standardize and can vary substantially by geography, by chance (i.e. developing reagents in eggs that may not antigenitically match wild-type viruses) and by intention (ie: choosing reagents that yield the most favorable results). This review describes attempts to standardize these assays to improve their performance as surrogates, the dangers of over-reliance on ‘reference’ serologic assays, the ways that manufacturers can exploit the existing regulatory framework to make their products ‘look good’ and the implications of this long-established system for the introduction of novel influenza vaccines.
Collapse
Affiliation(s)
- Brian J Ward
- a Research Institute of the McGill University Health Centre, Infectious Diseases Division , Montreal , QC , Canada.,b Medicago Inc , Québec , QC , Canada
| | - Stephane Pillet
- a Research Institute of the McGill University Health Centre, Infectious Diseases Division , Montreal , QC , Canada.,b Medicago Inc , Québec , QC , Canada
| | | | | | | | | |
Collapse
|
19
|
Ortiz JR, Hickling J, Jones R, Donabedian A, Engelhardt OG, Katz JM, Madhi SA, Neuzil KM, Rimmelzwaan GF, Southern J, Spiro DJ, Hombach J. Report on eighth WHO meeting on development of influenza vaccines that induce broadly protective and long-lasting immune responses: Chicago, USA, 23-24 August 2016. Vaccine 2017; 36:932-938. [PMID: 29221895 DOI: 10.1016/j.vaccine.2017.11.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/26/2022]
Abstract
In August 2016, the World Health Organization (WHO) convened the "Eighth meeting on development of influenza vaccines that induce broadly protective and long-lasting immune responses" to discuss the regulatory requirements and pathways for licensure of next-generation influenza vaccines, and to identify areas where WHO can promote the development of such vaccines. Participants included approximately 120 representatives of academia, the vaccine industry, research and development funders, and regulatory and public health agencies. They reviewed the draft WHO preferred product characteristics (PPCs) of vaccines that could address prioritized unmet public health needs and discussed the challenges facing the development of such vaccines, especially for low- and middle-income countries (LMIC). They defined the data desired by public-health decision makers globally and explored how to support the progression of promising candidates into late-stage clinical trials and for all countries. This report highlights the major discussions of the meeting.
Collapse
Affiliation(s)
- Justin R Ortiz
- Initiative for Vaccine Research, World Health Organization (WHO), Geneva, Switzerland.
| | - Julian Hickling
- Working in Tandem Ltd, Cambridge, Northern Ireland, United Kingdom.
| | - Rebecca Jones
- Working in Tandem Ltd, Cambridge, Northern Ireland, United Kingdom.
| | - Armen Donabedian
- Biomedical Advanced Research and Development Authority, United States Department of Health and Human Services, Washington DC, United States.
| | - Othmar G Engelhardt
- Division of Virology, National Institute for Biological Standards and Control, A Centre of the Medicines and Healthcare products Regulatory Agency, Potters Bar, Hertfordshire, United Kingdom.
| | - Jacqueline M Katz
- Influenza Division, Centers for Disease Control and Prevention (CDC), Atlanta, United States.
| | - Shabir A Madhi
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Kathleen M Neuzil
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, United States.
| | - Guus F Rimmelzwaan
- Erasmus Medical Center, Department of Viroscience, Rotterdam, The Netherlands.
| | - James Southern
- Advisor to Medicines Control Council, Simon's Town, South Africa.
| | - David J Spiro
- National Institutes of Health, Bethesda, United States.
| | - Joachim Hombach
- Initiative for Vaccine Research, World Health Organization (WHO), Geneva, Switzerland.
| |
Collapse
|
20
|
Won G, Lee JH. Salmonella Typhimurium, the major causative agent of foodborne illness inactivated by a phage lysis system provides effective protection against lethal challenge by induction of robust cell-mediated immune responses and activation of dendritic cells. Vet Res 2017; 48:66. [PMID: 29070065 PMCID: PMC5657113 DOI: 10.1186/s13567-017-0474-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/20/2017] [Indexed: 12/12/2022] Open
Abstract
Salmonella Typhimurium infection via foodborne transmission remains a major public health threat even in developed countries. Vaccines have been developed to reduce the disease burden at the pre-harvest stage, but the cell-mediated immune response against intracellular invasion of the pathogen is not sufficiently elicited by conventional killed Salmonella vaccines, which are safer than live vaccines. In this study, we developed a genetically inactivated vaccine candidate by introducing lysis plasmid pJHL454 harboring the λ phage holin-endolysin system into S. Typhimurium; we designated this vaccine JOL1950. In vitro expression of endolysin was validated by immunoblotting, and complete inactivation of JOL1950 cells was observed following 36 h of the lysis. Electron microscopic examinations by scanning electron microscopy and immunogold labeling transmission EM revealed conserved surface antigenic traits of the JOL1950 cells after lysis. An in vivo immunogenicity study in mice immunized with lysed cells showed significantly increased serum IgG, IgG1, and IgG2a levels. Further, we observed markedly increased in vitro cell proliferation and upregulation of Th1, Th2, and Th17 cytokines in the repulsed splenic T-cells of immunized mice. In dendritic cells (DCs) treated with lysed JOL1950, we observed a significant increase in dendritic cell activation, co-stimulatory molecule production, and levels of immunomodulatory cytokines. In addition, Th1 and Th17 cytokines were also released by naïve CD4+ T-cells pulsed with primed DCs. Lysed JOL1950 also protected against lethal challenge in immunized mice. Together, these results indicate that our vaccine candidate has great potential to induce cell-mediated immunity against S. Typhimurium by facilitating the activation of DCs.
Collapse
Affiliation(s)
- Gayeon Won
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Gobong-ro 79, Iksan, 54596, South Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, Gobong-ro 79, Iksan, 54596, South Korea.
| |
Collapse
|
21
|
Lohia N, Baranwal M. Immune responses to highly conserved influenza A virus matrix 1 peptides. Microbiol Immunol 2017; 61:225-231. [PMID: 28429374 DOI: 10.1111/1348-0421.12485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/12/2017] [Accepted: 04/18/2017] [Indexed: 01/07/2023]
Abstract
Influenza vaccine development is considered to be complicated and challenging. Constantly evolving influenza viruses require continuous global monitoring and reformulation of the vaccine strains. Peptides that are conserved among different strains and subtypes of influenza A virus are strongly considered to be attractive targets for development of cross protective influenza vaccines that stimulate cellular responses. In this study, three highly conserved (>90%) matrix 1 peptides that contain multiple T cell epitopes, ILGFVFTLTVPSERGLQRRRF (PM 1), LIRHENRMVLASTTAKA (PM 2) and LQAYQKRMGVQMQR (PM 3), were assessed for their immunogenic potential in vitro by subjecting peripheral blood mononuclear cells from healthy volunteers to repetitive stimulation with these chemically synthesised peptides and measuring their IFN-γ concentrations, proliferation by ELISA, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, respectively. Seven samples were screened for immunogenicity of PM 1 and PM 2, and six for that of PM 3. All six samples had positive responses (IFN-γ secretion) to PM 3 stimulation, as did five and three for PM 2 and PM 1 respectively. In contrast, seven (PM 1 and PM 2) and four (PM 3) samples showed proliferative response as compared with unstimulated cells. The encouraging immunogenic response generated by these highly conserved matrix 1 peptides indicates they are prospective candidates for development of broadly reactive influenza vaccines.
Collapse
Affiliation(s)
- Neha Lohia
- Department of Biotechnology, Thapar University, Patiala147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar University, Patiala147004, India
| |
Collapse
|
22
|
Schulze K, Ebensen T, Babiuk LA, Gerdts V, Guzman CA. Intranasal vaccination with an adjuvanted polyphosphazenes nanoparticle-based vaccine formulation stimulates protective immune responses in mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2169-2178. [PMID: 28579436 DOI: 10.1016/j.nano.2017.05.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/05/2017] [Accepted: 05/20/2017] [Indexed: 01/08/2023]
Abstract
The most promising strategy to sustainably prevent infectious diseases is vaccination. However, emerging as well as re-emerging diseases still constitute a considerable threat. Furthermore, lack of compliance and logistic constrains often result in the failure of vaccination campaigns. To overcome these hurdles, novel vaccination strategies need to be developed, which fulfill maximal safety requirements, show maximal efficiency and are easy to administer. Mucosal vaccines constitute promising non-invasive approaches able to match these demands. Here we demonstrate that nanoparticle (polyphosphazenes)-based vaccine formulations including c-di-AMP as adjuvant, cationic innate defense regulator peptides (IDR) and ovalbumin (OVA) as model antigen were able to stimulate strong humoral and cellular immune responses, which conferred protection against the OVA expressing influenza strain A/WSN/OVAI (H1N1). The presented results confirm the potency of nanoparticle-based vaccine formulations to deliver antigens across the mucosal barrier, but also demonstrate the necessity to include adjuvants to stimulate efficient antigen-specific immune responses.
Collapse
Affiliation(s)
- Kai Schulze
- Helmholtz Center for Infection Research (HZI), Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Thomas Ebensen
- Helmholtz Center for Infection Research (HZI), Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | | | - Volker Gerdts
- Vaccine and Infectious Disease Organization and Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Canada.
| | - Carlos A Guzman
- Helmholtz Center for Infection Research (HZI), Department of Vaccinology and Applied Microbiology, Braunschweig, Germany.
| |
Collapse
|
23
|
Vector-based genetically modified vaccines: Exploiting Jenner's legacy. Vaccine 2016; 34:6436-6448. [PMID: 28029542 PMCID: PMC7115478 DOI: 10.1016/j.vaccine.2016.06.059] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/02/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022]
Abstract
The global vaccine market is diverse while facing a plethora of novel developments. Genetic modification (GM) techniques facilitate the design of ’smarter’ vaccines. For many of the major infectious diseases of humans, like AIDS and malaria, but also for most human neoplastic disorders, still no vaccines are available. It may be speculated that novel GM technologies will significantly contribute to their development. While a promising number of studies is conducted on GM vaccines and GM vaccine technologies, the contribution of GM technology to newly introduced vaccines on the market is disappointingly limited. In this study, the field of vector-based GM vaccines is explored. Data on currently available, actually applied, and newly developed vectors is retrieved from various sources, synthesised and analysed, in order to provide an overview on the use of vector-based technology in the field of GM vaccine development. While still there are only two vector-based vaccines on the human vaccine market, there is ample activity in the fields of patenting, preclinical research, and different stages of clinical research. Results of this study revealed that vector-based vaccines comprise a significant part of all GM vaccines in the pipeline. This study further highlights that poxviruses and adenoviruses are among the most prominent vectors in GM vaccine development. After the approval of the first vectored human vaccine, based on a flavivirus vector, vaccine vector technology, especially based on poxviruses and adenoviruses, holds great promise for future vaccine development. It may lead to cheaper methods for the production of safe vaccines against diseases for which no or less perfect vaccines exist today, thus catering for an unmet medical need. After the introduction of Jenner’s vaccinia virus as the first vaccine more than two centuries ago, which eventually led to the recent eradication of smallpox, this and other viruses may now be the basis for constructing vectors that may help us control other major scourges of mankind.
Collapse
|
24
|
Sterilizing immunity to influenza virus infection requires local antigen-specific T cell response in the lungs. Sci Rep 2016; 6:32973. [PMID: 27596047 PMCID: PMC5011745 DOI: 10.1038/srep32973] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 08/17/2016] [Indexed: 12/29/2022] Open
Abstract
Sterilizing immunity is a unique immune status, which prevents effective virus infection into the host. It is different from the immunity that allows infection but with subsequent successful eradication of the virus. Pre-infection induces sterilizing immunity to homologous influenza virus challenge in ferret. In our antigen-specific experimental system, mice pre-infected with PR8 influenza virus through nasal route are likewise resistant to reinfection of the same strain of virus. The virus is cleared before establishment of effective infection. Intramuscular influenza virus injection confers protection against re-infection with facilitated virus clearance but not sterilizing immunity. Pre-infection and intramuscular injection generates comparable innate immunity and antibody response, but only pre-infection induces virus receptor reduction and efficient antigen-specific T cell response in the lungs. Pre-infection with nH1N1 influenza virus induces virus receptor reduction but not PR8-specific T cell immune response in the lungs and cannot prevent infection of PR8 influenza virus. Pre-infection with PR8 virus induced PR8-specific T cell response in the lungs but cannot prevent infection of nH1N1 virus either. These results reveal that antigen-specific T cell immunity is required for sterilizing immunity.
Collapse
|
25
|
Sester M, Leboeuf C, Schmidt T, Hirsch HH. The "ABC" of Virus-Specific T Cell Immunity in Solid Organ Transplantation. Am J Transplant 2016; 16:1697-706. [PMID: 26699950 DOI: 10.1111/ajt.13684] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/07/2015] [Accepted: 12/08/2015] [Indexed: 01/25/2023]
Abstract
Transplant patients are at increased risk of viral complications due to impaired control of viral replication, resulting from HLA mismatching between graft and host and the immunosuppression needed to avert alloimmune reactions. In the past decade, quantitative viral load measurements have become widely available to identify patients at risk and to inform treatment decisions with respect to immunosuppressive drugs and antiviral therapies. Because viral loads are viewed as the result of viral replication and virus-specific immune control, virus-specific T cell monitoring has been explored to optimize management of adenovirus, BK polyomavirus and cytomegalovirus ("ABC") in transplant patients. Although most studies are descriptive using different technologies, the overall results show that the quantity and quality of virus-specific T cells inversely correlate with viral replication, whereby strong cellular immune responses are associated with containment of viral replication. The key obstacles to the introduction of assays for virus-specific T cells into clinical practice is the definition of reliable cutoffs for clinical decision making, the poor negative predictive value of some assays, and the absence of interventional trials justifying changes of antiviral treatment or immunosuppression. More clinical research is needed using optimized assays and targets before standardization and commutability can be envisaged as achieved for viral load testing.
Collapse
Affiliation(s)
- M Sester
- Department of Transplant and Infection Immunology, Saarland University, Homburg, Germany
| | - C Leboeuf
- Transplantation & Clinical Virology, Department Biomedicine (Haus Petersplatz), University of Basel, Basel, Switzerland
| | - T Schmidt
- Department of Transplant and Infection Immunology, Saarland University, Homburg, Germany
| | - H H Hirsch
- Transplantation & Clinical Virology, Department Biomedicine (Haus Petersplatz), University of Basel, Basel, Switzerland.,Division Infection Diagnostics, Department Biomedicine (Haus Petersplatz), University of Basel, Basel, Switzerland.,Infectious Diseases & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
26
|
Trombetta CM, Montomoli E. Influenza immunology evaluation and correlates of protection: a focus on vaccines. Expert Rev Vaccines 2016; 15:967-76. [PMID: 26954563 DOI: 10.1586/14760584.2016.1164046] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Vaccination is the most effective method of controlling seasonal influenza infections and preventing possible pandemic events. Although influenza vaccines have been licensed and used for decades, the potential correlates of protection induced by these vaccines are still a matter of discussion. Currently, inactivated vaccines are the most common and the haemagglutination inhibition antibody titer is regarded as an immunological correlate of protection and the best available parameter for predicting protection from influenza infection. However, the assay shows some limitations, such as its low sensitivity to B and avian strains and inter-laboratory variability. Additional assays and next-generation vaccines have been evaluated to overcome the limitations of the traditional serological techniques and to elicit broad immune responses, underlining the need to revise the current correlates of protection. The aim of this review is to provide an overview of the current scenario regarding the immunological evaluation and correlates of protection of influenza vaccines.
Collapse
Affiliation(s)
| | - Emanuele Montomoli
- a Department of Molecular and Developmental Medicine , University of Siena , Siena , Italy.,b VisMederi srl , Enterprise of services in Life Sciences , Siena , Italy
| |
Collapse
|