1
|
Treating allergies via skin - Recent advances in cutaneous allergen immunotherapy. Adv Drug Deliv Rev 2022; 190:114458. [PMID: 35850371 DOI: 10.1016/j.addr.2022.114458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 01/24/2023]
Abstract
Subcutaneous allergen immunotherapy has been practiced clinically for decades to treat airborne allergies. Recently, the cutaneous route, which exploits the immunocompetence of the skin has received attention, which is evident from attempts to use it to treat peanut allergy. Delivery of allergens into the skin is inherently impeded by the barrier imposed by stratum corneum, the top layer of the skin. While the stratum corneum barrier must be overcome for efficient allergen delivery, excessive disruption of this layer can predispose to development of allergic inflammation. Thus, the most desirable allergen delivery approach must provide a balance between the level of skin disruption and the amount of allergen delivered. Such an approach should aim to achieve high allergen delivery efficiency across various skin types independent of age and ethnicity, and optimize variables such as safety profile, allergen dosage, treatment frequency, application time and patient compliance. The ability to precisely quantify the amount of allergen being delivered into the skin is crucial since it can allow for allergen dose optimization and can promote consistency and reproducibility in treatment response. In this work we review prominent cutaneous delivery approaches, and offer a perspective on further improvisation in cutaneous allergen-specific immunotherapy.
Collapse
|
2
|
Geng J, Hu X, Zhang Z, Gu Z, Li Y, Mou X, Mao L, Ge Y, Yang X, Song Y, Liu H, Wang L, Wei Z, Wang Z, Xu H. Discovery and pharmacodynamic evaluation of the novel butene lactone derivative M355 against influenza A virus in vitro and in vivo. J Med Virol 2022; 94:4393-4405. [PMID: 35560068 DOI: 10.1002/jmv.27853] [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: 03/17/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 11/06/2022]
Abstract
A new series of butene lactone derivatives were designed according to an influenza neuraminidase target and their antiviral activities against H1N1 infection of MDCK cells were evaluated. Among them, a compound that was given the name M355 was identified as the most potent against H1N1 (EC50 = 14.7 μM) with low toxicity (CC50 = 538.13 μM). It also visibly reduced the virus-induced cytopathic effect. Time-of-addition analysis indicated that H1N1 was mostly suppressed by M355 at the late stage of its infectious cycle. M355 inhibited neuraminidase in a dose-dependent fashion to a similar extent as oseltamivir, which was also indicated by computer modeling experiment. In a mouse model, lung lesions and virus load were reduced and the expression of nucleoprotein was moderated by M355. The ELISA and qRT-PCR analyses revealed that the levels of IFN-γ, IRF-3, TLR-3, TNF-α, IL-1β, IL-6 and IL-8 were down-regulated in the M355-treated groups, whereas the levels of IL-10 and IL-13 were up-regulated. Similarly, IgG was found to be increased in infected mice plasma. These results demonstrate that M355 inhibit the expression of H1N1 in both cellular and animal models. Thus, M355 has the potential to be effective in the treatment of influenza A virus infection. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jingwei Geng
- Zhongyuan District Center for Disease Control and Prevention of Zhengzhou, Zhengzhou, 450006, China
| | - Xiaoning Hu
- Binzhou People's Hospital, Binzhou, 256610, Shandong Province, China
| | - Zhongmou Zhang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Zichen Gu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuanyuan Li
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaodong Mou
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China
| | - Lu Mao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongzhuang Ge
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Xinyu Yang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China
| | - Yihui Song
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China
| | - Hongmin Liu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Linqing Wang
- Zhengzhou Key Laboratory of molecular biology, Zhengzhou Normal University, Zhengzhou, 450044, China
| | - Zhanyong Wei
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Zhenya Wang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| | - Haiwei Xu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery & Development, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
3
|
Mansoor KA, Qadan F, Schmidt M, Qinna NA, Badr M, Matalka KZ. A Functional Food Mixture "Protector" Reinforces the Protective Immune Parameters against Viral Flu Infection in Mice. Nutrients 2018; 10:nu10060743. [PMID: 29890620 PMCID: PMC6024812 DOI: 10.3390/nu10060743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/29/2018] [Accepted: 06/06/2018] [Indexed: 11/24/2022] Open
Abstract
Background: Viral influenza infection causes serious health issues especially when an outbreak occurs. Although influenza virus vaccines are available and each year manufactures modify the vaccine depending on the expected mutated strain, it is still far from satisfactory, mainly in young children and older adults. Therefore, a product that can support and shape the immune system to protect against viral flu infections is highly essential. Methods: A functional food water-soluble mixture of pomegranate, red grape, dates, olive fruit, figs, and ginger extracts, termed herein “Protector”, was prepared and tested in stimulating/modulating the production of specific cytokines, and hemagglutinin inhibition (HAI) antibodies following viral flu vaccination in mice. Results: A single intraperitoneal or multiple oral administration for 1–7 days of “Protector” significantly increased the production of interferon (IFN)-γ and interleukin (IL)-12 in blood, spleen, and lungs of mice. When “Protector” was orally administered for one week following a single vaccine injection (primary immunization) or for two weeks (one week apart) following double vaccine injections (secondary immunization), mice significantly produced higher titers of HAI antibodies. This increase in HAI antibodies was associated with Pillow-inducing significant and different changes in vaccine-induced IFN-γ, IL-12, IL-6 and IL-22 following primary and secondary immunizations. Conclusions: “Protector” administration reinforces the protective immune parameters against viral flu infection. Therefore, after performing preclinical toxicology studies and ensuring its safety, “Protector” should be considered a potential product to be tested in clinical trials to conclude its efficacy in reducing the devastating effects of flu infection in humans and its outbreaks.
Collapse
Affiliation(s)
- Kenza A Mansoor
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, Jordan.
| | - Fadi Qadan
- Herbresearch Germany, Wartbergweg 15, D-86874 Mattsies, Germany.
| | - Mathias Schmidt
- Herbresearch Germany, Wartbergweg 15, D-86874 Mattsies, Germany.
| | - Nidal A Qinna
- University of Petra Pharmaceutical Center (UPPC), Amman, Jordan.
| | - Mujtaba Badr
- University of Petra Pharmaceutical Center (UPPC), Amman, Jordan.
| | | |
Collapse
|
4
|
|
5
|
Falkeborn T, Hinkula J, Olliver M, Lindberg A, Maltais AK. The intranasal adjuvant Endocine™ enhances both systemic and mucosal immune responses in aged mice immunized with influenza antigen. Virol J 2017; 14:44. [PMID: 28253901 PMCID: PMC5335733 DOI: 10.1186/s12985-017-0698-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/02/2017] [Indexed: 01/17/2023] Open
Abstract
Despite availability of annual influenza vaccines, influenza causes significant morbidity and mortality in the elderly. This is at least in part a result of immunosenescence; the age-dependent decrease in immunological competence that results in greater susceptibility to infections and reduced responses to vaccination. To improve protective immune responses in this age group, new vaccines strategies, such as the use of adjuvants, are needed. Here, we evaluated the mucosal vaccine adjuvant Endocine™, formulated with split influenza antigen and administered intranasally in aged (20-month old) mice. Humoral immune responses were assessed and compared to unadjuvanted intranasal and subcutaneous vaccines. We show that formulation with Endocine™ significantly enhances hemagglutination inhibition (HI) titers, as well as serum IgG and mucosal IgA antibody titers, compared to both types of unadjuvanted vaccines. Thus, our results indicate that intranasal vaccination with Endocine™ is a possible approach for the development of mucosal influenza vaccines for the elderly.
Collapse
Affiliation(s)
- Tina Falkeborn
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jorma Hinkula
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Marie Olliver
- Eurocine Vaccines AB, Karolinska Institutet Science Park, Solna, Sweden
| | - Alf Lindberg
- Eurocine Vaccines AB, Karolinska Institutet Science Park, Solna, Sweden
| | | |
Collapse
|
6
|
Transcutaneous Immunization Using Nano-sized Drug Carriers. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3121-7_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
7
|
Kreuzer D, Nikoopour E, Au BCY, Krougly O, Lee-Chan E, Summers KL, Haeryfar SMM, Singh B. Reduced interferon-α production by dendritic cells in type 1 diabetes does not impair immunity to influenza virus. Clin Exp Immunol 2015; 179:245-55. [PMID: 25286929 DOI: 10.1111/cei.12462] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2014] [Indexed: 12/30/2022] Open
Abstract
The increased risk and persistence of infections in diabetic condition is probably associated with defects in the cellular immune responses. We have previously shown a decrease in the production of interferon (IFN)-α by dendritic cells (DCs) in diabetic subjects. The basal level of IFN-α in splenic plasmacytoid DCs (pDCs) is also lower in non-obese diabetic (NOD) mice compared to prediabetic mice. The objective of this study was to analyse the ability of diabetic mice to mobilize innate and CD8(+) T cell-mediated immune response to influenza A virus (IAV) with the live influenza A/Puerto Rico/8/1934 H1N1 (PR8) strain or with its immunodominant CD8(+) T cell epitopes. We found that following immunization with IAV, the level of IFN-α in diabetic mice was increased to the level in prediabetic mice. Immunization of NOD mice with the immunodominant IAV PR8 peptide induced clonal expansion of IFN-γ-producing CD8(+) T cells similar to the response observed in prediabetic mice. Thus, diabetic and prediabetic NOD mice have a similar capacity for IFN-α and IFN-γ production by pDCs and CD8(+) T cells, respectively. Therefore, the DC-related immune defect in diabetic NOD mice does not impair their capacity to develop an effective immune response to IAV. Our results suggest that reduced IFN-α production by diabetic human and mouse DCs is not an impediment to an effective immunity to IAV in type 1 diabetic subjects vaccinated with live attenuated influenza vaccine.
Collapse
Affiliation(s)
- D Kreuzer
- Centre for Human Immunology, Department of Microbiology and Immunology and Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Dormitzer P, Tsai T, Del Giudice G. New technologies for influenza vaccines. Hum Vaccin Immunother 2014; 8:45-58. [DOI: 10.4161/hv.8.1.18859] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
9
|
Nafziger AN, Pratt DS. Seasonal influenza vaccination and technologies. J Clin Pharmacol 2014; 54:719-31. [PMID: 24691877 DOI: 10.1002/jcph.299] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/26/2014] [Indexed: 11/06/2022]
Abstract
Seasonal influenza is a serious respiratory illness that causes annual worldwide epidemics resulting in significant morbidity and mortality. Influenza pandemics occur about every 40 yrs, and may carry a greater burden of illness and death than seasonal influenza. Both seasonal influenza and pandemic influenza have profound economic consequences. The combination of current vaccine efficacy and viral antigenic drifts and shifts necessitates annual vaccination. New manufacturing technologies in influenza vaccine development employ cell culture and recombinant techniques. Both allow more rapid vaccine creation and production. In the past 5 years, brisk, highly creative activity in influenza vaccine research and development has begun. New vaccine technologies and vaccination strategies are addressing the need for viable alternatives to egg production methods and improved efficacy. At present, stubborn problems of sub-optimal efficacy and the need for annual immunization persist. There is an obvious need for more efficacious vaccines and improved vaccination strategies to make immunization easier for providers and patients. Mitigating this serious annual health threat remains an important public health priority.
Collapse
MESH Headings
- Animals
- Antigenic Variation
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/metabolism
- Health Priorities
- Humans
- Influenza A virus/immunology
- Influenza A virus/metabolism
- Influenza Vaccines/biosynthesis
- Influenza Vaccines/therapeutic use
- Influenza, Human/epidemiology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Betainfluenzavirus/immunology
- Betainfluenzavirus/metabolism
- Mass Vaccination
- Pandemics/prevention & control
- Seasons
- Technology, Pharmaceutical/trends
- Vaccines, Synthetic/chemistry
- Vaccines, Synthetic/metabolism
- Vaccines, Synthetic/therapeutic use
Collapse
Affiliation(s)
- Anne N Nafziger
- Bertino Consulting, Schenectady, NY, USA; Adjunct Research Professor, School of Pharmacy & Pharmaceutical Sciences, Department of Pharmacy Practice, University at Buffalo, State University of New York, Buffalo, NY, USA
| | | |
Collapse
|
10
|
Davtyan H, Ghochikyan A, Hovakimyan A, Petrushina I, Yu J, Flyer D, Madsen PJ, Pedersen LO, Cribbs DH, Agadjanyan MG. Immunostimulant patches containing Escherichia coli LT enhance immune responses to DNA- and recombinant protein-based Alzheimer's disease vaccines. J Neuroimmunol 2014; 268:50-7. [PMID: 24507620 PMCID: PMC3951952 DOI: 10.1016/j.jneuroim.2014.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 12/19/2013] [Accepted: 01/07/2014] [Indexed: 12/22/2022]
Abstract
Immunotherapeutic approaches to treating Alzheimer's disease (AD) using vaccination strategies must overcome the obstacle of achieving adequate responses to vaccination in the elderly. Here we demonstrate for the first time that application of the Escherichia coli heat-labile enterotoxin adjuvant-laden immunostimulatory patches (LT-IS) dramatically enhances the onset and magnitude of immune responses to DNA- and protein-based vaccines for Alzheimer's disease following intradermal immunization via gene gun and conventional needles, respectively. Our studies suggest that the immune activation mediated by LT-IS offers improved potency for generating AD-specific vaccination responses that should be investigated as an adjuvant in the clinical arena.
Collapse
Affiliation(s)
- Hayk Davtyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Jianmei Yu
- Intercell USA, Inc., Gaithersburg, MD 20878, USA
| | - David Flyer
- Intercell USA, Inc., Gaithersburg, MD 20878, USA
| | | | | | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA; Department of Neurology, University of California, Irvine, CA 92697, USA
| | - Michael G Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
11
|
Topical CpG adjuvantation of a protein-based vaccine induces protective immunity to Listeria monocytogenes. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:329-39. [PMID: 24391136 DOI: 10.1128/cvi.00734-13] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Robust CD8(+) T cell responses are essential for immune protection against intracellular pathogens. Using parenteral administration of ovalbumin (OVA) protein as a model antigen, the effect of the Toll-like receptor 9 (TLR9) agonist, CpG oligodeoxynucleotide (ODN) 1826, as an adjuvant delivered either topically, subcutaneously, or intramuscularly on antigen-specific CD8(+) T cell responses in a mouse model was evaluated. Topical CpG adjuvant increased the frequency of OVA-specific CD8(+) T cells in the peripheral blood and in the spleen. The more effective strategy to administer topical CpG adjuvant to enhance CD8(+) T cell responses was single-dose administration at the time of antigen injection with a prime-boost regimen. Topical CpG adjuvant conferred both rapid and long-lasting protection against systemic challenge with recombinant Listeria monocytogenes expressing the cytotoxic T lymphocyte (CTL) epitope of OVA(257-264) (strain Lm-OVA) in a TLR9-dependent manner. Topical CpG adjuvant induced a higher proportion of CD8(+) effector memory T cells than parenteral administration of the adjuvant. Although traditional vaccination strategies involve coformulation of antigen and adjuvant, split administration using topical adjuvant is effective and has advantages of safety and flexibility. Split administration of topical CpG ODN 1826 with parenteral protein antigen is superior to other administration strategies in enhancing both acute and memory protective CD8(+) T cell immune responses to subcutaneous protein vaccines. This vaccination strategy induces rapid and persistent protective immune responses against the intracellular organism L. monocytogenes.
Collapse
|
12
|
Skountzou I, Compans RW. Skin immunization with influenza vaccines. Curr Top Microbiol Immunol 2014; 386:343-69. [PMID: 25038939 DOI: 10.1007/82_2014_407] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Problems with existing influenza vaccines include the strain specificity of the immune response, resulting in the need for frequent reformulation in response to viral antigenic drift. Even in years when the same influenza strains are prevalent, the duration of immunity is limited, and results in the need for annual revaccination. The immunogenicity of the present split or subunit vaccines is also lower than that observed with whole inactivated virus, and the vaccines are not very effective in high risk groups such as the young or the elderly. Vaccine coverage is incomplete, due in part to concerns about the use of hypodermic needles for delivery. Alternative approaches for vaccination are being developed which address many of these concerns. Here we review new approaches which focus on skin immunization, including the development of needle-free delivery systems which use stable dry formulations and induce stronger and longer-lasting immune responses.
Collapse
Affiliation(s)
- Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, CNR Building, 1518 Clifton Road, Atlanta, GA, 30322, USA,
| | | |
Collapse
|
13
|
|
14
|
Abstract
The vertebrate immune system is comprised of numerous distinct and interdependent components. Every component has its own inherent protective value, and the final combination of them is likely to be related to an animal’s immunological history and evolutionary development. Vertebrate immune system consists of both systemic and mucosal immune compartments, but it is the mucosal immune system which protects the body from the first encounter of pathogens. According to anatomical location, the mucosa-associated lymphoid tissue, in teleost fish is subdivided into gut-, skin-, and gill-associated lymphoid tissue and most available studies focus on gut. The purpose of this paper is to summarise the current knowledge of the immunological defences present in skin mucosa as a very important part of the fish immune system, serving as an anatomical and physiological barrier against external hazards. Interest in defence mechanism of fish arises from a need to develop health management tools to support a growing finfish aquaculture industry, while at the same time addressing questions concerning origins and evolution of immunity in vertebrates. Increased knowledge of fish mucosal immune system will facilitate the development of novel vaccination strategies in fish.
Collapse
Affiliation(s)
- María Ángeles Esteban
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, Regional Campus of International Excellence “Campus Mare Nostrum”, 30100 Murcia, Spain
| |
Collapse
|
15
|
Zahran E, Seo JK, Noga EJ. The effect of adjuvant and microbial challenge on the expression of antimicrobial polypeptides in channel catfish (Ictalurus punctatus). FISH & SHELLFISH IMMUNOLOGY 2012; 33:168-173. [PMID: 22507196 DOI: 10.1016/j.fsi.2012.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 03/13/2012] [Accepted: 03/13/2012] [Indexed: 05/31/2023]
Abstract
While antimicrobial polypeptides (AMPPs) are increasingly recognized as one of the most important components of innate immunity, there is very little information in vertebrates that documents their upregulation to levels that are microbicidal in vivo. Here we demonstrate that intraperitoneal injection of either Freund's complete adjuvant (FCA) or live Tetrahymena pyriformis (a parasitic ciliate) upregulated AMPP expression in channel catfish skin. FCA induced significant upregulation of total antibacterial activity, anti-Edwardsiella ictaluri activity (the fraction of antibacterial activity active against E. ictaluri), and HLP-1 (the major AMPP in channel catfish skin). Tetrahymena induced a similar upregulation, except that HLP-1 was not significantly induced and the response appeared to be more transient than FCA immunostimulation. AMPP levels were increased up to five-fold higher than resting levels and levels expressed were well within concentrations known to be inhibitory to many important pathogens in vitro. These results provide encouragement that AMPP upregulation may be a promising tool in aquaculture for enhancing the resistance of fish to disease.
Collapse
Affiliation(s)
- Eman Zahran
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA
| | | | | |
Collapse
|
16
|
NS1-truncated live attenuated virus vaccine provides robust protection to aged mice from viral challenge. J Virol 2012; 86:10293-301. [PMID: 22787224 DOI: 10.1128/jvi.01131-12] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immunological changes associated with age contribute to the high rates of influenza virus morbidity and mortality in the elderly. Compounding this problem, aged individuals do not respond to vaccination as well as younger, healthy adults. Efforts to increase protection to this demographic group are of utmost importance, as the proportion of the population above the age of 65 is projected to increase in the coming decade. Using a live influenza virus with a truncated nonstructural protein 1 (NS1), we are able to stimulate cellular and humoral immune responses of aged mice comparable to levels seen in young mice. Impressively, a single vaccination provided protection following stringent lethal challenge in aged mice.
Collapse
|
17
|
Karande P, Mitragotri S. Transcutaneous immunization: an overview of advantages, disease targets, vaccines, and delivery technologies. Annu Rev Chem Biomol Eng 2012; 1:175-201. [PMID: 22432578 DOI: 10.1146/annurev-chembioeng-073009-100948] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Skin is an immunologically active tissue composed of specialized cells and agents that capture and process antigens to confer immune protection. Transcutaneous immunization takes advantage of the skin immune network by inducing a protective immune response against topically applied antigens. This mode of vaccination presents a novel and attractive approach for needle-free immunization that is safe, noninvasive, and overcomes many of the limitations associated with needle-based administrations. In this review we will discuss the developments in the field of transcutaneous immunization in the past decade with special emphasis on disease targets and vaccine delivery technologies. We will also briefly discuss the challenges that need to be overcome to translate early laboratory successes in transcutaneous immunization into the development of effective clinical prophylactics.
Collapse
Affiliation(s)
- Pankaj Karande
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.
| | | |
Collapse
|
18
|
Abstract
It is now clear that the epidermis has an active role in local immune responses in the skin. Keratinocytes are involved early in inflammation by providing first-line innate mechanisms and, in addition, can contribute to adaptive immune responses that may be associated with clinical disease. Moreover, keratinocytes are capable of enhancing and shaping the outcome of inflammation in response to stimuli and promoting particular types of immune bias. Through understanding the underlying mechanisms, the role of keratinocytes in disease pathogenesis will be further defined, which is likely to lead to the identification of potential targets for prophylactic or therapeutic intervention.
Collapse
Affiliation(s)
- Danuta Gutowska-Owsiak
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | |
Collapse
|
19
|
Li N, Peng LH, Chen X, Nakagawa S, Gao JQ. Transcutaneous vaccines: Novel advances in technology and delivery for overcoming the barriers. Vaccine 2011; 29:6179-90. [DOI: 10.1016/j.vaccine.2011.06.086] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Revised: 06/19/2011] [Accepted: 06/22/2011] [Indexed: 12/17/2022]
|
20
|
Distinctive immunomodulatory and inflammatory properties of the Escherichia coli type II heat-labile enterotoxin LT-IIa and its B pentamer following intradermal administration. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1243-51. [PMID: 21677110 DOI: 10.1128/cvi.00012-11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The type I and type II heat-labile enterotoxins (LT-I and LT-II) are strong mucosal adjuvants when they are coadministered with soluble antigens. Nonetheless, data on the parenteral adjuvant activities of LT-II are still limited. Particularly, no previous study has evaluated the adjuvant effects and induced inflammatory reactions of LT-II holotoxins or their B pentameric subunits after delivery via the intradermal (i.d.) route to mice. In the present report, the adjuvant and local skin inflammatory effects of LT-IIa and its B subunit pentamer (LT-IIaB(5)) were determined. When coadministered with ovalbumin (OVA), LT-IIa and, to a lesser extent, LT-IIaB(5) exhibited serum IgG adjuvant effects. In addition, LT-IIa but not LT-IIaB(5) induced T cell-specific anti-OVA responses, particularly in respect to induction of antigen-specific cytotoxic CD8(+) T cell responses. LT-IIa and LT-IIaB(5) induced differential tissue permeability and local inflammatory reactions after i.d. injection. Of particular interest was the reduced or complete lack of local reactions, such as edema and tissue induration, in mice i.d. inoculated with LT-IIa and LT-IIaB(5,) respectively, compared with mice immunized with LT-I. In conclusion, the present results show that LT-IIa and, to a lesser extent, LT-IIaB(5) exert adjuvant effects when they are delivered via the i.d. route. In addition, the low inflammatory effects of LT-IIa and LT-IIaB(5) in comparison to those of LT-I support the usefulness of LT-IIa and LT-IIaB(5) as parenterally delivered vaccine adjuvants.
Collapse
|
21
|
Alecsandru D, Valor L, Sánchez-Ramón S, Gil J, Carbone J, Navarro J, Rodríguez J, Rodríguez-Sainz C, Fernández-Cruz E. Sublingual therapeutic immunization with a polyvalent bacterial preparation in patients with recurrent respiratory infections: immunomodulatory effect on antigen-specific memory CD4+ T cells and impact on clinical outcome. Clin Exp Immunol 2011; 164:100–7. [PMID: 21391984 DOI: 10.1111/j.1365-2249.2011.04320.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Recurrent respiratory tract infections (RRTIs) are common clinical conditions in individuals with alterations of the immune function. A prospective open pilot study in a cohort of patients with RRTIs has been performed to assess whether sublingual immunization with a polyvalent bacterial vaccine could exert an immunomodulatory effect on the antigen-specific immunological responses and have an impact on the clinical outcome. Seventeen patients with RRTIs were recruited. An oral polyvalent bacterial preparation (Bactek®) was administered to all patients daily for 6 months. Immunological assessment was performed at baseline and at the end of immunization. Immunological measurements included: T cell-specific proliferations of CD3+CD4+ and CD3+CD8+ to Bactek® antigens, total immunoglobulin levels, antibodies to pneumococcal polysaccharide and tetanus toxoid and B, T and natural killer (NK) cell subsets. There was a significant increase in the proliferative capacity of CD3+CD4+ T cells specific to Bactek® antigens at month 6 in comparison to baseline (P < 0·0001). A significant increase in total CD3+ T cells was also observed (P < 0·05). No significant differences were observed between baseline and month 6 in levels of total immunoglobulins, specific antibodies and B, T or NK cell subsets. A significant reduction in the patient's rate of RRTIs was observed compared with 1 year prior to initiation of therapy (P < 0·0001). The results demonstrate that long-term administration of a sublingual polyvalent bacterial preparation in patients with RRTIs exerts an immune stimulating effect on CD4+ T helper cell responses to bacterial antigens which could be associated with clinical benefit.
Collapse
Affiliation(s)
- D Alecsandru
- Clinical Immunology Unit, Immunology, Department, Hospital General Universitario, Gregorio Marañón, Departamento de Microbiología I, Universidad Complutense de Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Schaefer JT, Patterson JW, Deacon DH, Smolkin ME, Petroni GR, Jackson EM, Slingluff CL. Dynamic changes in cellular infiltrates with repeated cutaneous vaccination: a histologic and immunophenotypic analysis. J Transl Med 2010; 8:79. [PMID: 20727190 PMCID: PMC2936306 DOI: 10.1186/1479-5876-8-79] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 08/20/2010] [Indexed: 01/23/2023] Open
Abstract
Background Melanoma vaccines have not been optimized. Adjuvants are added to activate dendritic cells (DCs) and to induce a favourable immunologic milieu, however, little is known about their cellular and molecular effects in human skin. We hypothesized that a vaccine in incomplete Freund's adjuvant (IFA) would increase dermal Th1 and Tc1-lymphocytes and mature DCs, but that repeated vaccination may increase regulatory cells. Methods During and after 6 weekly immunizations with a multipeptide vaccine, immunization sites were biopsied at weeks 0, 1, 3, 7, or 12. In 36 participants, we enumerated DCs and lymphocyte subsets by immunohistochemistry and characterized their location within skin compartments. Results Mature DCs aggregated with lymphocytes around superficial vessels, however, immature DCs were randomly distributed. Over time, there was no change in mature DCs. Increases in T and B-cells were noted. Th2 cells outnumbered Th1 lymphocytes after 1 vaccine 6.6:1. Eosinophils and FoxP3+ cells accumulated, especially after 3 vaccinations, the former cell population most abundantly in deeper layers. Conclusions A multipeptide/IFA vaccine may induce a Th2-dominant microenvironment, which is reversed with repeat vaccination. However, repeat vaccination may increase FoxP3+T-cells and eosinophils. These data suggest multiple opportunities to optimize vaccine regimens and potential endpoints for monitoring the effects of new adjuvants. Trail Registration ClinicalTrials.gov Identifier: NCT00705640
Collapse
Affiliation(s)
- Jochen T Schaefer
- Human Immune Therapy Center, University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Safety and immunogenicity of an influenza vaccine A/H5N1 (A/Vietnam/1194/2004) when coadministered with a heat-labile enterotoxin (LT) adjuvant patch. Vaccine 2009; 27 Suppl 6:G60-6. [DOI: 10.1016/j.vaccine.2009.10.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
24
|
Immunization by influenza virus-like particles protects aged mice against lethal influenza virus challenge. Antiviral Res 2009; 84:215-24. [PMID: 19772876 DOI: 10.1016/j.antiviral.2009.09.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 08/31/2009] [Accepted: 09/03/2009] [Indexed: 11/24/2022]
Abstract
Influenza virus-like particles (VLPs) were produced in Sf9 insect cells by co-expressing the matrix protein M1 and the surface glycoproteins hemagglutinin (HA) and neuraminidase (NA) using the recombinant baculovirus expression system. The VLPs were morphologically similar to influenza virions. Both HA and NA proteins were incorporated into VLPs and these proteins retained their functional activities. Further, influenza VLPs but not inactivated influenza viruses (IIV) stimulated secretion of inflammatory cytokines from mouse bone marrow-derived dendritic cells (BMDC). Immunogenicity of influenza VLPs and their protective efficacies against lethal influenza virus challenge were evaluated in young and aged mice. Immunization with influenza VLPs induced strong antibody responses against HA that inhibited hemagglutination by influenza virus, similar to IIV vaccines. Compared to young mice, antibody responses in aged mice immunized with a low dose of either influenza VLPs or IIV vaccines exhibited markedly reduced avidity for HA. However, immunization of aged mice with a high dose of influenza VLPs induced antibody responses with high avidity similar to those in young mice. Furthermore, all vaccinated animals survived a lethal challenge by a mouse-adapted influenza virus (A/PR/8/34), indicating that influenza VLPs are highly efficacious for protection against influenza virus infection in both young and aged mice.
Collapse
|
25
|
Mahe B, Vogt A, Liard C, Duffy D, Abadie V, Bonduelle O, Boissonnas A, Sterry W, Verrier B, Blume-Peytavi U, Combadiere B. Nanoparticle-Based Targeting of Vaccine Compounds to Skin Antigen-Presenting Cells By Hair Follicles and their Transport in Mice. J Invest Dermatol 2009; 129:1156-64. [DOI: 10.1038/jid.2008.356] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
26
|
Michel JP, Chidiac C, Grubeck-Loebenstein B, Johnson RW, Lambert PH, Maggi S, Moulias R, Nicholson K, Werner H. Advocating Vaccination of Adults Aged 60 Years and Older in Western Europe:. Rejuvenation Res 2009; 12:127-35. [DOI: 10.1089/rej.2008.0813] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jean-Pierre Michel
- Department of Rehabilitation and Geriatrics, Geneva Medical School and Univeristy Hospitals, Thonex-Geneve, Switzerland
| | - Christian Chidiac
- Department of Infectious and Tropical Diseaes, Hopital de La Croix Rousse, Lyon, France
| | | | | | | | | | | | - Karl Nicholson
- University Hospitals of Leicester NHS Trust, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Hans Werner
- Ev. Krankenhaus Elisabethenstift, Darmstadt, Germany
| |
Collapse
|
27
|
Combadière B, Mahé B. Particle-based vaccines for transcutaneous vaccination. Comp Immunol Microbiol Infect Dis 2008; 31:293-315. [PMID: 17915323 DOI: 10.1016/j.cimid.2007.07.015] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2007] [Indexed: 01/12/2023]
Abstract
Immunization concepts evolve with increasing knowledge of how the immune system works and the development of new vaccination methods. Traditional vaccines are made of live, attenuated, killed or fragmented pathogens. New vaccine strategies can take advantage of particulate compounds--microspheres or nanoparticles--to target antigen-presenting cells better, which must subsequently reach the secondary lymphoid organs, which are the sites of the immune response. The use of the skin as a target organ for vaccine delivery stems from the fact that immature dendritic cells (DCs), which are professional antigen-presenting cells can be found at high density in the epidermis and dermis of human or animal skin. This has led to design various methods of dermal or transcutaneous vaccination. The quality and duration of the humoral and cellular responses to vaccination depend on the appropriate targeting of antigen-presenting cells, of the vaccine dose, route of administration and use of adjuvant. In this review, we will focus on the use of micro- and nano-particles to target the skin antigen-presenting cells and will discuss recent advances in the field of transcutaneous vaccination in animal models and humans.
Collapse
Affiliation(s)
- Behazine Combadière
- Institut National de la Santé et de la Recherche Médicale (INSERM) U543, Université Pierre et Marie Curie-Paris6, 91 Boulevard de l'Hôpital, 75634 Paris, France.
| | | |
Collapse
|
28
|
Mkrtichyan M, Ghochikyan A, Movsesyan N, Karapetyan A, Begoyan G, Yu J, Glenn GM, Ross TM, Agadjanyan MG, Cribbs DH. Immunostimulant adjuvant patch enhances humoral and cellular immune responses to DNA immunization. DNA Cell Biol 2008; 27:19-24. [PMID: 17961074 PMCID: PMC2478559 DOI: 10.1089/dna.2007.0639] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The focus of this report is on the development of an improved DNA immunization protocol, which takes advantage of the strengths of DNA immunization, as well as those associated with adjuvant delivered by transcutaneous immunostimulatory (IS) patches. Because transcutaneous delivery of adjuvants to the skin at the vaccination site has been shown to amplify the immune response to protein antigens, we hypothesized that the same IS patch when placed on the skin at the site of DNA injection could further enhance the immune response to a DNA influenza vaccine. We have combined an influenza DNA vaccine, hemagglutinin fused with three copies of complement C3d, to enhance uptake and antigen presentation, with an IS patch containing heat-labile enterotoxin from Escherichia coli. Coadministration of a potent adjuvant in IS patches placed on the skin at the site of DNA vaccination dramatically amplifies anti-influenza antibody immune response. Supplementing DNA vaccines with IS patches may be a particularly valuable strategy because DNA vaccines can be rapidly modified in response to mutations in pathogens, and individuals with compromised immune systems such as transplant patients and the elderly will benefit from the enhanced antibody response induced by the IS patches.
Collapse
Affiliation(s)
- Mikayel Mkrtichyan
- Department of Immunology, The Institute for Molecular Medicine, Huntington Beach, California
| | - Anahit Ghochikyan
- Department of Immunology, The Institute for Molecular Medicine, Huntington Beach, California
| | - Nina Movsesyan
- The Institute for Brain Aging and Dementia, University of California, Irvine, California
| | - Adrine Karapetyan
- Department of Immunology, The Institute for Molecular Medicine, Huntington Beach, California
| | - Gayane Begoyan
- Department of Immunology, The Institute for Molecular Medicine, Huntington Beach, California
| | - Jianmei Yu
- IOMAI Corporation, Gaithersburg, Maryland
| | | | - Ted M. Ross
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael G. Agadjanyan
- Department of Immunology, The Institute for Molecular Medicine, Huntington Beach, California
- The Institute for Brain Aging and Dementia, University of California, Irvine, California
| | - David H. Cribbs
- The Institute for Brain Aging and Dementia, University of California, Irvine, California
- Department of Neurology, University of California, Irvine, California
| |
Collapse
|
29
|
|
30
|
Glenn GM, Flyer DC, Ellingsworth LR, Frech SA, Frerichs DM, Seid RC, Yu J. Transcutaneous immunization with heat-labile enterotoxin: development of a needle-free vaccine patch. Expert Rev Vaccines 2007; 6:809-19. [PMID: 17931160 DOI: 10.1586/14760584.6.5.809] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The skin is an attractive target for vaccine delivery. Adjuvants and antigens delivered into the skin can result in potent immune responses and an unmatched safety profile. The heat-labile enterotoxin (LT) from Escherichia coli, which acts both as antigen and adjuvant, has been shown to be delivered to human skin efficiently when used in a patch, resulting in strong immune responses. Iomai scientists have capitalized on these observations to develop late-stage products based on LT. This has encouraged commercial-level product development of a delivery system that is efficient, user-friendly and designed to address important medical needs. Over the past 2 years, extensive clinical testing and optimization has allowed the patch to evolve to a late-stage product. As a strategy for approval of a revolutionary vaccine-delivery system, the singular focus on optimization of LT delivery has enabled technical progress to extend patch-vaccine product development beyond LT. The field efficacy of the LT-based travelers' diarrhea vaccine has validated this approach. The discussion of transcutaneous immunization is unique, in that any consideration of the adjuvant must also include delivery, and the significant advances in a commercial patch application system are described. In this review, we integrate these concepts, update the clinical data and look to the future.
Collapse
Affiliation(s)
- Gregory M Glenn
- Iomai Corporation, 20 Firstfield Road, Suite 250, Gaithersburg, MD 20878, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Ershler WB, Gravenstein S, Geloo ZS. Thymosin Alpha 1 as an Adjunct to Influenza Vaccination in the Elderly: Rationale and Trial Summaries. Ann N Y Acad Sci 2007; 1112:375-84. [PMID: 17600281 DOI: 10.1196/annals.1415.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
From a clinical perspective, the immune deficiency of aging (immune senescence) is not profound. In fact, it may be of little clinical consequence. However, older people are prone to chronic and debilitating disorders which alone, or in concert with the medications used in their treatment, may add to the age effect and create a more clinically relevant immune deficiency. As a result, many older people are susceptible to infection. Furthermore, it is now well recognized that older people respond less well to immunization protocols. Protection against influenza by vaccination with hemagluttinin is the prototype example. Despite programs that have raised vaccination rates dramatically over the past three decades, influenza remains a major cause of morbidity and mortality in the elderly. This, in part, is due to the fact that vaccine responses are reduced in older recipients. Strategies are under development to enhance vaccine efficacy in this population and one such strategy is the adjuvant use of thymosin alpha 1 (Talpha1). In both animal experiments and human trials, there has been demonstrated enhancement of vaccine responses. The findings to date warrant additional efforts to further examine the role of Talpha1 in augmenting specific vaccine responses both in the elderly or in younger subjects in situations in which there are suboptimal quantities of immunizing antigen available.
Collapse
Affiliation(s)
- William B Ershler
- Clinical Research Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21225, USA.
| | | | | |
Collapse
|
32
|
Abstract
The skin is populated with Langerhans cells, thought to be efficient, potent antigen-presenting cells, that are capable of inducing protective immunity by targeting antigen delivery to the skin. Delivery to the skin may be accomplished by active delivery such as intradermal injection, use of patches or a combination of a universal adjuvant patch with injections. The robust immunity induced by skin targeting can lead to dose sparing, novel vaccines and immune enhancement in populations with poorly responsive immune systems, such as the elderly. Vaccine delivery with patches (transcutaneous immunization), may allow self-administration, ambient temperature stabilization and ease of storage for stockpiling, leading to a new level of efficient vaccine distribution in times of crisis such as a bioterror event or pandemic influenza outbreak. The use of an adjuvant (immunostimulant) patch with injected vaccines has been shown in clinical studies to enhance the immune response to an injected vaccine. This can be used for dose sparing in pandemic influenza vaccines in critically short supply or immune enhancement for poor responders to flu vaccines such as the elderly. Transcutaneous immunization offers a unique safety profile, as adjuvants are sequestered in the skin and only delivered systemically by Langerhans cells. This results in an excellent safety profile and allows use of extremely potent adjuvants. The combination of the skin immune system, safe use of potent adjuvants and ease of delivery suggests that skin delivery of vaccines can address multiple unmet needs for mass vaccination scenarios.
Collapse
Affiliation(s)
- G M Glenn
- Iomai Corporation, Gaithersburg, MD 20878, USA.
| | | |
Collapse
|
33
|
Fulop T, Larbi A, Wikby A, Mocchegiani E, Hirokawa K, Pawelec G. Dysregulation of T-cell function in the elderly : scientific basis and clinical implications. Drugs Aging 2006; 22:589-603. [PMID: 16038574 DOI: 10.2165/00002512-200522070-00005] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The function of the immune system is to maintain body integrity by defending against infections, cancers, autoimmune diseases and inflammation-related chronic diseases. The immune response is known to become defective with aging, leading to decreased longevity and appearance of age-related disease. The most important changes occur in T-cell immunity, and are manifested particularly as altered clonal expansion of cells of limited antigen specificity. The causes of these alterations are multifactorial, and include thymic involution, T-cell subset changes and signal transduction alterations. The clinical consequences of these changes are not well defined, except for their extremely important negative impact on defence against infections, especially by new pathogens, and decreased responses to vaccination. Considering the public health consequences of decreased immune competence in old age, strategies for immune response modulation are desirable to decrease the health burden for the elderly and improve their quality of life.
Collapse
Affiliation(s)
- Tamas Fulop
- Immunology Program, Geriatric Division, Research Center on Aging, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada.
| | | | | | | | | | | |
Collapse
|
34
|
Fingerut E, Gutter B, Goldway M, Eliahoo D, Pitcovski J. B subunit of E. coli enterotoxin as adjuvant and carrier in oral and skin vaccination. Vet Immunol Immunopathol 2006; 112:253-63. [PMID: 16701905 DOI: 10.1016/j.vetimm.2006.03.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Revised: 02/26/2006] [Accepted: 03/14/2006] [Indexed: 02/01/2023]
Abstract
Mucosal sites are one of the main natural ports of entry into the body. Stimulation of a local response by antibodies as the systemic protection may enhance the efficacy of non-living vaccines, and allow for vaccination by subunit vaccines without the need for injection. Mucosal or skin vaccination necessitates a suitable adjuvant and carrier. Escherichia coli heat-labile enterotoxin (LT) and its B subunit (LTB) have been found to be effective adjuvants. The aim of this study was to efficiently produce and purify recombinant LTB (brLTB), and examine its adjuvant and carrier properties. The gene encoding LTB was cloned and expressed in E. coli, and the product was found to have a pentameric form with the ability to bind the cell receptor, GM1 ganglioside. A one-step method for efficient purification and concentration of brLTB was developed. Both oral and intramuscular vaccination with purified brLTB yielded high antibody titers, which detected the whole toxin. In an attempt to test its adjuvant characteristics, brLTB was mixed with either BSA or a recombinant protein (rKnob of egg drop syndrome adenovirus) and delivered intramuscularly, orally or transcutaneously. The addition of brLTB significantly elevated the antibody response in groups vaccinated orally and transcutaneously, but had no influence in injected groups. Vaccination with another recombinant protein, (viral protein 2 of infectious bursal disease virus) supplemented with brLTB did not elevate the antibody response, as compared to vaccination with the antigen alone. These results demonstrate that the addition of brLTB makes oral and transcutaneous vaccination with protein antigens possible.
Collapse
Affiliation(s)
- E Fingerut
- Migal, P.O. Box 831, Kiryat Shmona 11016, Israel
| | | | | | | | | |
Collapse
|
35
|
Pedraza-Sánchez S, González-Hernández Y, Escobar-Gutiérrez A, Ramachandra L. The immunostimulant RU41740 from Klebsiella pneumoniae activates human cells in whole blood to potentially stimulate innate and adaptive immune responses. Int Immunopharmacol 2006; 6:635-46. [PMID: 16504927 DOI: 10.1016/j.intimp.2005.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2005] [Revised: 08/03/2005] [Accepted: 10/07/2005] [Indexed: 11/16/2022]
Abstract
The compound RU41740 from Klebsiella pneumoniae, when used as an immunostimulant, improves responses to bacterial and yeast infections in murine models and in human trials. The aim of this study was to determine in vitro, the capacity of RU41740 to stimulate human leukocytes in whole blood. Blood samples from healthy adult donors were incubated with RU41740 for 4 or 24 h and leukocytes were assessed for levels of activation markers and cytokine production by flow cytometry and ELISA. The early activation marker CD69 was induced at 4 h in NK cells > B cells > T cells > monocytes whereas at 24 h CD80 and CD86 levels were augmented on monocytes and IL-12 was induced; HLA-DR levels increased on both B cells and monocytes. The pro-inflammatory cytokines TNF-alpha and IL-6 were produced at 4 h at similar levels to that induced by LPS and monocytes appeared to be a source of TNF-alpha. IFN-gamma, was induced at 5 h just in NK cells. Activation induced by RU41740 was not abolished by polymixin B, ruling out the possible contamination with LPS. These data indicate that RU41740 can impact not only the innate immune responses but potentially enhance adaptive immune responses by up-regulating expression of molecules involved in antigen presentation on antigen presenting cells.
Collapse
Affiliation(s)
- Sigifredo Pedraza-Sánchez
- Departamento de Investigaciones Inmunológicas, Instituto de Diagnóstico y Referencia Epidemiológicos, Secretaría de Salud, México City.
| | | | | | | |
Collapse
|
36
|
Linton PJ, Lustgarten J, Thoman M. T cell function in the aged: Lessons learned from animal models. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/j.cair.2006.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
37
|
Frech SA, Kenney RT, Spyr CA, Lazar H, Viret JF, Herzog C, Glück R, Glenn GM. Improved immune responses to influenza vaccination in the elderly using an immunostimulant patch. Vaccine 2005; 23:946-50. [PMID: 15603897 DOI: 10.1016/j.vaccine.2004.06.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2003] [Revised: 06/07/2004] [Accepted: 06/11/2004] [Indexed: 10/26/2022]
Abstract
The elderly have greater morbidity and mortality due to influenza, and respond poorly to influenza vaccination compared to younger adults. This study was designed to determine if the adjuvant heat-labile enterotoxin from Escherichia coli (LT), administered as an immunostimulant (IS) patch on the skin with influenza vaccination, improves influenza immune responses in the elderly. Three weeks following vaccination, hemagglutination inhibition (HAI) responses in LT IS patch recipients showed improvement over those of elderly receiving vaccine alone, as demonstrated by significance or trends in fold rise [A/Panama (P = 0.004), A/New Caledonia (P = 0.09)], seroconversion [A/New Caledonia (63% versus 40%, P = 0.01), A/Panama (54% versus 36%, P = 0.08)] and seroprotection [26%, 20% and 16% greater for the patch group for A/New Caledonia, A/Panama and B/Shandong strains, respectively]. The data suggest that an LT IS patch may further enhance influenza vaccine immune responses in the elderly.
Collapse
Affiliation(s)
- Sarah A Frech
- IOMAI Corporation, 20 Firstfield Road, Suite 250, Gaithersburg, MD 20878, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Glynn A, Freytag LC, Clements JD. Effect of homologous and heterologous prime–boost on the immune response to recombinant plague antigens. Vaccine 2005; 23:1957-65. [PMID: 15734068 DOI: 10.1016/j.vaccine.2004.10.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2004] [Revised: 10/07/2004] [Accepted: 10/12/2004] [Indexed: 11/17/2022]
Abstract
Among the pathogens that have been identified as potential agents of biological warfare or bioterrorism, Yersinia pestis is one of the main concerns due to the severity and potential transmissibility of the pneumonic form of the disease in humans. There are no approved vaccines for protection against pneumonic plague, but a Y. pestis-derived fusion protein (F1-V) has shown great promise as a protective antigen in murine studies. In the current study, we examine different prime-boost regimens, including parenteral, mucosal, and transcutaneous delivery, in order to explore the effect of changing the route of prime and boost on the ability of recombinant F1-V to promote the development of long-lasting, high-titer antibodies. The most significant findings of the study reported here are that (1) intranasal and subcutaneous immunizations are both effective and essentially equivalent for induction of serum and bronchioalveolar anti-F1-V IgG1 responses when a single booster dose is administered by the same (homologous) route, (2) heterologous boosting can be as or more effective than homologous boosting for induction of either serum or bronchioalveolar anti-F1-V IgG1 responses, and (3) anti-F1 and anti-V total IgG responses were highest in animals primed intranasally and boosted by any route when compared to animals primed transcutaneously or subcutaneously. As with previously published studies, there were still significant levels of circulating anti-F1-V antibodies 1 year post-primary immunization. These studies provide important insights into the development of new-generation biodefense vaccines.
Collapse
Affiliation(s)
- Audrey Glynn
- Department of Microbiology and Immunology, Program in Molecular Pathogenesis and Immunity, 1430 Tulane Avenue, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
39
|
Pearson H. Flu patch may save elderly. Nature 2004. [DOI: 10.1038/news040719-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|