1
|
Szachniewicz MM, Neustrup MA, van den Eeden SJF, van Meijgaarden KE, Franken KLMC, van Veen S, Koning RI, Limpens RWAL, Geluk A, Bouwstra JA, Ottenhoff THM. Evaluation of PLGA, lipid-PLGA hybrid nanoparticles, and cationic pH-sensitive liposomes as tuberculosis vaccine delivery systems in a Mycobacterium tuberculosis challenge mouse model - A comparison. Int J Pharm 2024; 666:124842. [PMID: 39424087 DOI: 10.1016/j.ijpharm.2024.124842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Tuberculosis (TB) continues to pose a global threat for millennia, currently affecting over 2 billion people and causing 10.6 million new cases and 1.3 million deaths annually. The only existing vaccine, Mycobacterium Bovis Bacillus Calmette-Guérin (BCG), provides highly variable and inadequate protection in adults and adolescents. This study explores newly developed subunit tuberculosis vaccines that use a multistage protein fusion antigen Ag85b-ESAT6-Rv2034 (AER). The protection efficacy, as well as in vivo induced immune responses, were compared for five vaccines: BCG; AER-CpG/MPLA mix; poly(D,L-lactic-co-glycolic acid) (PLGA); lipid-PLGA hybrid nanoparticles (NPs); and cationic pH-sensitive liposomes (the latter three delivering AER together with CpG and MPLA). All vaccines, except the AER-adjuvant mix, induced protection in Mycobacterium tuberculosis (Mtb)-challenged C57/Bl6 mice as indicated by a significant reduction in bacterial burden in lungs and spleens of the animals. Four AER-based vaccines significantly increased the number of circulating multifunctional CD4+ and CD8+ T-cells producing IL-2, IFNγ, and TNFα, exhibiting a central memory phenotype. Furthermore, AER-based vaccines induced an increase in CD69+ B-cell counts as well as high antigen-specific antibody titers. Unexpectedly, none of the observed immune responses were associated with the bacterial burden outcome, such that the mechanism responsible for the observed vaccine-induced protection of these vaccines remains unclear. These findings suggest the existence of non-classical protective mechanisms for Mtb infection, which could, once identified, provide interesting targets for novel vaccines.
Collapse
Affiliation(s)
- Mikołaj M Szachniewicz
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands.
| | - Malene A Neustrup
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - Susan J F van den Eeden
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Krista E van Meijgaarden
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Kees L M C Franken
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Suzanne van Veen
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Roman I Koning
- Electron Microscopy Facility, Leiden University Medical Center (LUMC), the Netherlands
| | - Ronald W A L Limpens
- Electron Microscopy Facility, Leiden University Medical Center (LUMC), the Netherlands
| | - Annemieke Geluk
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Joke A Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - Tom H M Ottenhoff
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| |
Collapse
|
2
|
Amini Y, Kabiri M, Jamehdar SA, Sankian M, Meshkat Z, Zare S, Soleimanpour S, Farsiani H, Moradi B, Tafaghodi M. Assessment of immunogenicity and protective efficiency of multi-epitope antigen-loaded in mannan decorated PLGA nanoparticles against tuberculosis. J Pharm Sci 2024:S0022-3549(24)00544-6. [PMID: 39631524 DOI: 10.1016/j.xphs.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
The antigen-targeting to dendritic cells (DCs) has gained increasing attention as the potential approach for immunotherapy in recent years due to the ability of DCs to regulate innate and adaptive immunity. In the present study, the immunogenicity and protective efficiency of mannan-decorated PLGA nanoparticles (NPs) loaded with multi-epitopes mycobacterium tuberculosis antigen (HspX-Ppe44-EsxV) were evaluated as a targeted delivery system to DCs. For this purpose, PLGA nanoparticle formulations were prepared and subsequently decorated by mannan. The physicochemical properties and level of mannan incorporation, as well as encapsulation efficiency and antigen release, were assessed. The potential of formulated NPs for antigen targeting to DCs, and immunogenicity against tuberculosis (TB) were investigated using immunofluorescence assay and in-vivo experiments. Mannan incorporation enhanced the uptake of fusion-loaded PLGA by DCs. The cytokine and antibody assays demonstrated that mannosylation of NPs and BCG-primed mice boosted by mannan-PLGA could significantly elevate Th1-biased immune responses relative to the BCG and non-modified PLGA NPs. Our findings also proved that the mannosylated vaccine in the presence of CpG could evoke Th1 and Th17 responses with appropriate protective efficiency against TB in mice. This result illustrated that the active targeting of DCs by mannan-PLGA NPs could induce a proper anti-tuberculosis response, which is essential for protection against tuberculosis.
Collapse
Affiliation(s)
- Yousef Amini
- Infectious Diseases and Tropical Medicine Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran; Department of Microbiology, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mona Kabiri
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeid Amel Jamehdar
- Microbiology and Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Meshkat
- Microbiology and Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirwan Zare
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Microbiology and Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Farsiani
- Microbiology and Virology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bagher Moradi
- Esfarayen Faculty of Medical Sciences, Esfarayen, Iran
| | - Mohsen Tafaghodi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Ahmad F, Ahmad S, Upadhyay TK, Singh S, Khubaib M, Singh J, Saeed M, Ahmad I, Al-Keridis LA, Sharma R. Rifabutin loaded inhalable β-glucan microparticle based drug delivery system for pulmonary TB. Sci Rep 2024; 14:16437. [PMID: 39013991 PMCID: PMC11253001 DOI: 10.1038/s41598-024-66634-5] [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: 03/22/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
Inhalable microparticle-based anti TB drug delivery systems are being investigated extensively for Tuberculosis [TB] treatment as they offer efficient and deep lung deposition with several advantages over conventional routes. It can reduce the drug dose, treatment duration and toxic effects and optimize the drug bioavailability. Yeast derived β-glucan is a β-[1-3/1-6] linked biocompatible polymer and used as carrier for various biomolecules. Due to presence of glucan chains, particulate glucans act as PAMP and thereby gets internalized via receptor mediated phagocytosis by the macrophages. In this study, β-glucan microparticles were prepared by adding l-leucine as excipient, and exhibited 70% drug [Rifabutin] loading efficiency. Further, the sizing and SEM data of particles revealed a size of 2-4 µm with spherical dimensions. The FTIR and HPLC data confirmed the β-glucan composition and drug encapsulations efficiency of the particles. The mass median aerodynamic diameter [MMAD] and geometric standard deviation [GSD] data indicated that these particles are inhalable in nature and have better thermal stability as per DSC thermogram. These particles were found to be non-toxic upto a concentration of 80 µg/ml and were found to be readily phagocytosed by human macrophage cells in-vitro as well as in-vivo by lung alveolar macrophage. This study provides a framework for future design of inhalable β-glucan particle based host-directed drug delivery system against pulmonary TB.
Collapse
Grants
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
- Small Research Group Project under Grant no. [R.G.P.1/226/44] Irfan Ahmad Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia; Email: irfancsmmu@gmail.com
Collapse
Affiliation(s)
- Firoz Ahmad
- IIRC-3 Immunobiochemistry Lab, Department of Biosciences, Integral University, Lucknow, UP, 226026, India
- Department of Clinical Immunology & Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, UP, 226014, India
| | - Shad Ahmad
- Department of Biochemistry, Dr. Ram Manohar Lohia Avadh University, Faizabad, UP, 224001, India
| | - Tarun Kumar Upadhyay
- Department of Life Sciences, Parul Institute of Applied Sciences & Research and Development Cell, Parul University, Vadodara, Gujarat, 391760, India
| | - Sanjay Singh
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, UP, 226201, India
| | - Mohd Khubaib
- IIRC-3 Immunobiochemistry Lab, Department of Biosciences, Integral University, Lucknow, UP, 226026, India
| | - Jyotsna Singh
- Inhalation Toxicology Facility, CSIR-Indian Institute of Toxicology Research, Lucknow, UP, 226008, India
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, 34464, Hail, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Lamya Ahmed Al-Keridis
- Department of Biology, Faculty of Science, Princess Nourah bint Abdulrahman University, P.O.Box 84428, 11671, Riyadh, Saudi Arabia
| | - Rolee Sharma
- Department of Life Sciences and Biotechnology, CSJM University, Kanpur, UP, 228024, India.
| |
Collapse
|
4
|
Dutt TS, Karger BR, Fox A, Youssef N, Dadhwal R, Ali MZ, Patterson J, Creissen E, Rampacci E, Cooper SK, Podell BK, Gonzalez-Juarrero M, Obregon-Henao A, Henao-Tamayo M. Mucosal exposure to non-tuberculous mycobacteria elicits B cell-mediated immunity against pulmonary tuberculosis. Cell Rep 2022; 41:111783. [PMID: 36516760 DOI: 10.1016/j.celrep.2022.111783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/09/2022] [Accepted: 11/15/2022] [Indexed: 12/15/2022] Open
Abstract
Bacille Calmette-Guerin (BCG) is the only licensed vaccine against Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB) disease. However, BCG has limited efficacy, necessitating the development of better vaccines. Non-tuberculous mycobacteria (NTMs) are opportunistic pathogens present ubiquitously in the environment. TB endemic countries experience higher exposure to NTMs, but previous studies have not elucidated the relationship between NTM exposure and BCG efficacy against TB. Therefore, we develop a mouse model (BCG + NTM) to simulate human BCG immunization regime and continuous NTM exposure. BCG + NTM mice exhibit superior and prolonged protection against pulmonary TB, with increased B cell influx and anti-Mtb antibodies in serum and airways, compared with BCG alone. Notably, spatial transcriptomics and immunohistochemistry reveal that BCG + NTM mice formed B cell aggregates with features of germinal center development, which correlate with reduced Mtb burden. Our studies suggest a direct relationship between NTM exposure and TB protection, with B cells playing a crucial role.
Collapse
Affiliation(s)
- Taru S Dutt
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA.
| | | | - Amy Fox
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | | | - Rhythm Dadhwal
- College of Business, Colorado State University, Fort Collins, CO, USA
| | - Malik Zohaib Ali
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA; Cell and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Johnathan Patterson
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Elizabeth Creissen
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Elisa Rampacci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Sarah K Cooper
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Brendan K Podell
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Mercedes Gonzalez-Juarrero
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Andres Obregon-Henao
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Marcela Henao-Tamayo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA.
| |
Collapse
|
5
|
de Figueiredo DB, Kaneko K, Rodrigues TDC, MacLoughlin R, Miyaji EN, Saleem I, Gonçalves VM. Pneumococcal Surface Protein A-Hybrid Nanoparticles Protect Mice from Lethal Challenge after Mucosal Immunization Targeting the Lungs. Pharmaceutics 2022; 14:pharmaceutics14061238. [PMID: 35745810 PMCID: PMC9230107 DOI: 10.3390/pharmaceutics14061238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/29/2022] [Accepted: 06/08/2022] [Indexed: 12/04/2022] Open
Abstract
Pneumococcal disease remains a global burden, with current conjugated vaccines offering protection against the common serotype strains. However, there are over 100 serotype strains, and serotype replacement is now being observed, which reduces the effectiveness of the current vaccines. Pneumococcal surface protein A (PspA) has been investigated as a candidate for new serotype-independent pneumococcal vaccines, but requires adjuvants and/or delivery systems to improve protection. Polymeric nanoparticles (NPs) are biocompatible and, besides the antigen, can incorporate mucoadhesive and adjuvant substances such as chitosans, which improve antigen presentation at mucosal surfaces. This work aimed to define the optimal NP formulation to deliver PspA into the lungs and protect mice against lethal challenge. We prepared poly(glycerol-adipate-co-ω-pentadecalactone) (PGA-co-PDL) and poly(lactic-co-glycolic acid) (PLGA) NPs using an emulsion/solvent evaporation method, incorporating chitosan hydrochloride (HCl-CS) or carboxymethyl chitosan (CM-CS) as hybrid NPs with encapsulated or adsorbed PspA. We investigated the physicochemical properties of NPs, together with the PspA integrity and biological activity. Furthermore, their ability to activate dendritic cells in vitro was evaluated, followed by mucosal immunization targeting mouse lungs. PGA-co-PDL/HCl-CS (291 nm) or CM-CS (281 nm) NPs produced smaller sizes compared to PLGA/HCl-CS (310 nm) or CM-CS (299 nm) NPs. Moreover, NPs formulated with HCl-CS possessed a positive charge (PGA-co-PDL +17 mV, PLGA + 13 mV) compared to those formulated with CM-CS (PGA-co-PDL -20 mV, PLGA -40 mV). PspA released from NPs formulated with HCl-CS preserved the integrity and biological activity, but CM-CS affected PspA binding to lactoferrin and antibody recognition. PspA adsorbed in PGA-co-PDL/HCl-CS NPs stimulated CD80+ and CD86+ cells, but this was lower compared to when PspA was encapsulated in PLGA/HCl-CS NPs, which also stimulated CD40+ and MHC II (I-A/I-E)+ cells. Despite no differences in IgG being observed between immunized animals, PGA-co-PDL/HCl-CS/adsorbed-PspA protected 83% of mice after lethal pneumococcal challenge, while 100% of mice immunized with PLGA/HCl-CS/encapsulated-PspA were protected. Therefore, this formulation is a promising vaccine strategy, which has beneficial properties for mucosal immunization and could potentially provide serotype-independent protection.
Collapse
Affiliation(s)
- Douglas Borges de Figueiredo
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
- Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo 05508-070, Brazil;
| | - Kan Kaneko
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK;
| | - Tasson da Costa Rodrigues
- Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo 05508-070, Brazil;
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen, IDA Business Park, H91 HE94 Galway, Ireland;
| | - Eliane Namie Miyaji
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Imran Saleem
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK;
- Correspondence: (I.S.); (V.M.G.); Tel.: +55-112-6279819 (V.M.G.)
| | - Viviane Maimoni Gonçalves
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
- Correspondence: (I.S.); (V.M.G.); Tel.: +55-112-6279819 (V.M.G.)
| |
Collapse
|
6
|
Liang Z, Li H, Qu M, Liu Y, Wang Y, Wang H, Dong Y, Chen Y, Ge X, Zhou X. Intranasal bovine β-defensin-5 enhances antituberculosis immunity in a mouse model by a novel protein-based respiratory mucosal vaccine. Virulence 2022; 13:949-962. [PMID: 35603910 PMCID: PMC9154763 DOI: 10.1080/21505594.2022.2080342] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Respiratory mucosal immunization is an effective immunization strategy against tuberculosis (TB), and effective mucosal vaccines require adjuvants that can promote protective immunity without deleterious inflammation. Mucosal BCG (Bacille Calmette-Guerin) is effective, but it causes a severe inflammatory response in the lung. A novel less cytotoxic mucosal vaccine AH-PB containing Mycobacterium tuberculosis (Mtb) cell surface antigens Ag85A and HspX (AH), as well as polyinosinic-polycytidylic acid (Poly IC) and bovine neutrophil β-defensin-5 (B5) adjuvants were prepared, with the overarching goal of protecting against TB. Then, the immunogenicity and protective efficacy of these vaccines via the intranasal route were evaluated in a mouse model. Results showed that intranasal AH-PB promoted tissue-resident memory T cells (TRMs) development in the lung, induced antigen-specific antibody response in airway, provided protection against Mycobacterium bovis (M. bovis), conferred better protection than parenteral BCG in the later stage of infection, and boosted the protective immunity generated by BCG in mice. Moreover, both B5 and Poly IC were indispensable for the protection generated by AH-PB. Furthermore, intranasal immunization with AH-B5 fusion vaccines also provided similar protection against M. bovis compared to AH-PB. Collectively, B5-based TB vaccine via the intranasal route is a promising immunization strategy against bovine TB, and this kind of immunization strategy may be applied to human TB vaccine development. These findings highlight the potential importance of B5 as a mucosal adjuvant used in TB vaccines or other respiratory disease vaccines.
Collapse
Affiliation(s)
- Zhengmin Liang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Mengjin Qu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yiduo Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yuanzhi Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haoran Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yuhui Dong
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yulan Chen
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xin Ge
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | | |
Collapse
|
7
|
Makled S, Boraie N, Nafee N. Nanoparticle-mediated macrophage targeting-a new inhalation therapy tackling tuberculosis. Drug Deliv Transl Res 2020; 11:1037-1055. [PMID: 32617866 DOI: 10.1007/s13346-020-00815-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Despite the potent clinical efficacy of linezolid (LNZ) against drug-resistant tuberculosis, its safety and tolerability remain of major concern. Our objective is to develop antitubercular inhalable LNZ nano-embedded microparticles. In this context, LNZ incorporated in non-structured lipid carriers (NLCs) was characterized in terms of colloidal, morphological, thermal, and release profiles. The potential of LNZ-NLCs to cross mucosal barriers and invade alveolar macrophages (AM, MH-S cells) was appraised. In vivo proof of concept was accomplished via orotracheal administration to mice. Respirable microparticles prepared by spray drying NLCs with diluents were assessed for their size, shape, flowability, aerosolization performance, and lung deposition pattern. NLCs (809-827 nm in size, zeta potential - 37.4 to - 58.9 mV) ensued 19% LNZ loading and pH-independent sustained release. Penetration studies revealed 73% LNZ crossing mucus within 1 h. Meanwhile, viability assay on A549 cells ensured an IC50 of 1.2 and 0.32 mg/mL for plain and LNZ-NLCs, respectively. CLSM confirmed phagocytosis of NLCs by MH-S macrophages, while H&E staining demonstrated NLC accumulation in murine AM in vivo with no signs of histopathological/biochemical changes. Bronchoalveolar lavage showed significantly low levels of LDH and total proteins (TP) for LNZ-NLCs highlighting their superior safety. Respirable microparticles embedding LNZ-NLCs ensured excellent aerosolization (MMAD 2 μm, FPF 93%) denoting perfect alveolar deposition. The developed inhalation therapy provided sustained LNZ release, mucus penetrability, potential safety in therapeutic doses, in vitro and in vivo macrophage targetability, and preferential deposition in the deep lung. Overall positive outcomes rely on reduced dose, dosing frequency, and per se superior safety circumventing systemic-associated life-threatening side effects. Graphical abstract.
Collapse
Affiliation(s)
- Shaimaa Makled
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Nabila Boraie
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Noha Nafee
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt. .,Department of Pharmaceutics, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait.
| |
Collapse
|
8
|
Duan Q, Xia P, Nandre R, Zhang W, Zhu G. Review of Newly Identified Functions Associated With the Heat-Labile Toxin of Enterotoxigenic Escherichia coli. Front Cell Infect Microbiol 2019; 9:292. [PMID: 31456954 PMCID: PMC6700299 DOI: 10.3389/fcimb.2019.00292] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Heat-labile toxin (LT) is a well-characterized powerful enterotoxin produced by enterotoxigenic Escherichia coli (ETEC). This toxin is known to contribute to diarrhea in young children in developing countries, international travelers, as well as many different species of young animals. Interestingly, it has also been revealed that LT is involved in other activities in addition to its role in enterotoxicity. Recent studies have indicated that LT toxin enhances enteric pathogen adherence and subsequent intestinal colonization. LT has also been shown to act as a powerful adjuvant capable of upregulating vaccine antigenicity; it also serves as a protein or antigenic peptide display platform for new vaccine development, and can be used as a naturally derived cell targeting and protein delivery tool. This review summarizes the epidemiology, secretion, delivery, and mechanisms of action of LT, while also highlighting new functions revealed by recent studies.
Collapse
Affiliation(s)
- Qiangde Duan
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Pengpeng Xia
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Rahul Nandre
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Weiping Zhang
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Guoqiang Zhu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
9
|
Kaushik H, Deshmukh SK, Solanki AK, Bhatia B, Tiwari A, Garg LC. Immunization with recombinant fusion of LTB and linear epitope (40-62) of epsilon toxin elicits protective immune response against the epsilon toxin of Clostridium perfringens type D. AMB Express 2019; 9:105. [PMID: 31300915 PMCID: PMC6626085 DOI: 10.1186/s13568-019-0824-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/26/2019] [Indexed: 11/10/2022] Open
Abstract
Epsilon toxin (Etx) produced by Clostridium perfringens types B and D, a major causative agent of enterotoxaemia causes significant economic losses to animal industry. Conventional vaccines against these pathogens generally employ formalin-inactivated culture supernatants. However, immunization with the culture supernatant and full length toxin subjects the animal to antigenic load and often have adverse effect due to incomplete inactivation of the toxins. In the present study, an epitope-based vaccine against Clostridium perfringens Etx, comprising 40-62 amino acid residues of the toxin in translational fusion with heat labile enterotoxin B subunit (LTB) of E. coli, was evaluated for its protective potential. The ability of the fusion protein rLTB.Etx40-62 to form pentamers and biologically active holotoxin with LTA of E. coli indicated that the LTB present in the fusion protein retained its biological activity. Antigenicity of both the components in the fusion protein was retained as anti-fusion protein antisera detected both the wild type Etx and LTB in Western blot analysis. Immunization of BALB/c mice with the fusion protein resulted in a significant increase in all isotypes, predominantly IgG1, IgG2a and IgG2b. Anti-fusion protein antisera neutralized the cytotoxicity of epsilon toxin both in vitro and in vivo. Thus, the results demonstrate the potential of rLTB.Etx40-62 as a candidate vaccine against C. perfringens.
Collapse
|
10
|
Yang AQ, Yang HY, Guo SJ, Xie YE. MF59 adjuvant enhances the immunogenicity and protective immunity of the OmpK/Omp22 fusion protein from Acineterbacter baumannii through intratracheal inoculation in mice. Scand J Immunol 2019; 90:e12769. [PMID: 31006127 DOI: 10.1111/sji.12769] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 04/14/2019] [Accepted: 04/15/2019] [Indexed: 01/10/2023]
Abstract
Acinetobacter baumannii (A baumannii) is an emerging nosocomial pathogenic bacterium which leads to hospital infections. The increase in drug-resistant A baumannii strains makes it difficult to control by using common antibiotics. The development of effective vaccines is an alternative means to avoid A baumannii infections. In the present study, Balb/c mice were inoculated intratracheally with 30 μg of OmpK/Omp22 fusion protein alone or OmpK/Omp22 formulated with MF59 adjuvant. After two times of boosting at day 14 and 21, the antigen-specific antibody levels and the protective immunity against A baumannii challenge were evaluated. The results showed that the OmpK/Omp22 formulated with MF59 immunized mice produced much higher level of antigen-specific antibodies compared to mice immunized with OmpK/Omp22 alone (P < 0.01). Mice immunized with 30 μg of OmpK/Omp22 formulated with MF59 also provided more potent protection post-challenge, which showed lower bacterial loads in the blood and lung tissue, lower level of blood inflammatory cytokines and higher survival rate (83.3%) than mice immunized with OmpK/Omp22 alone (P < 0.001). In conclusion, this study demonstrated that OmpK/Omp22 fusion protein adjuvanted with MF59 induced superior immune response and better protection than OmpK/Omp22 alone through intratracheal inoculation in mice.
Collapse
Affiliation(s)
- Ai-Qiong Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hai-Yan Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - San-Jun Guo
- Institute of Immunology and Molecular Biology, North Sichuan Medical College, Nanchong, China
| | - Yong-En Xie
- Institute of Immunology and Molecular Biology, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
11
|
Vanbever R, Loira-Pastoriza C, Dauguet N, Hérin C, Ibouraadaten S, Vanvarenberg K, Ucakar B, Tyteca D, Huaux F. Cationic Nanoliposomes Are Efficiently Taken up by Alveolar Macrophages but Have Little Access to Dendritic Cells and Interstitial Macrophages in the Normal and CpG-Stimulated Lungs. Mol Pharm 2019; 16:2048-2059. [DOI: 10.1021/acs.molpharmaceut.9b00033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Rita Vanbever
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Cristina Loira-Pastoriza
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Nicolas Dauguet
- de Duve Institute, Flow Cytometry and Cell Sorting Platform, Université catholique de Louvain, Brussels, Belgium
| | - Caroline Hérin
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Saloua Ibouraadaten
- Institut de Recherche Expérimentale et Clinique, Louvain Centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Brussels, Belgium
| | - Kevin Vanvarenberg
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Bernard Ucakar
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Donatienne Tyteca
- de Duve Institute, Cell Biology, Université catholique de Louvain, Brussels, Belgium
| | - François Huaux
- Institut de Recherche Expérimentale et Clinique, Louvain Centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
12
|
Alves CC, Araujo N, Bernardes WPDOS, Mendes MM, Oliveira SC, Fonseca CT. A Strong Humoral Immune Response Induced by a Vaccine Formulation Containing rSm29 Adsorbed to Alum Is Associated With Protection Against Schistosoma mansoni Reinfection in Mice. Front Immunol 2018; 9:2488. [PMID: 30450095 PMCID: PMC6224358 DOI: 10.3389/fimmu.2018.02488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 10/08/2018] [Indexed: 01/17/2023] Open
Abstract
The helminth Schistosoma mansoni is one of main causes of human schistosomiasis, a health and economic concern in some of the world's poorest countries. Current treatment regimens can lead to serious side effects and are not suitable for breastfeeding mothers. As such, efforts have been undertaken to develop a vaccine to prevent infection. Of these, Sm29 is a promising candidate that has been associated with resistance to infection/reinfection in humans and mice. Its ability to induce resistance to reinfection has also been recently demonstrated using a vaccine formulation containing Freund's adjuvant. However, Freund's adjuvant is unsuitable for use in human vaccines. We therefore evaluated the ability of Sm29 to induce protection against S. mansoni reinfection when formulated with either alum or MPLA as an adjuvant, both approved for human use. Our data demonstrate that, in contrast to Sm29 with MPLA, Sm29 with alum reduced parasite burden after reinfection compared to a control. We next investigated whether the immune response was involved in creating the differences between the protective (Sm29Alum) and non-protective (Sm29MPLA) vaccine formulations. We observed that both formulations induced a similar mixed-profile immune response, however, the Sm29 with alum formulation raised the levels of antibodies against Sm29. This suggests that there is an association between a reduction in worm burden and parasite-specific antibodies. In summary, our data show that Sm29 with an alum adjuvant can successfully protect against S. mansoni reinfection in mice, indicating a potentially effective vaccine formulation that could be applied in humans.
Collapse
Affiliation(s)
- Clarice Carvalho Alves
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Neusa Araujo
- Laboratório de Esquistossomose, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | | | - Mariana Moreira Mendes
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Sergio Costa Oliveira
- Laboratório de Imunologia de doenças Infeciosas, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Instituto Nacional de Ciências e Tecnologia em Doenças Tropicais, CNPq, MCT, Salvador, Brazil
| | - Cristina Toscano Fonseca
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| |
Collapse
|
13
|
Tada R, Yamanaka D, Ogasawara M, Saito M, Ohno N, Kiyono H, Kunisawa J, Aramaki Y. Polymeric Caffeic Acid Is a Safer Mucosal Adjuvant That Augments Antigen-Specific Mucosal and Systemic Immune Responses in Mice. Mol Pharm 2018; 15:4226-4234. [PMID: 30107747 DOI: 10.1021/acs.molpharmaceut.8b00648] [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] [Indexed: 01/08/2023]
Abstract
Infections remain a major threat to human lives. To overcome the threat caused by pathogens, mucosal vaccines are considered a promising strategy. However, no inactivated and/or subunit mucosal vaccine has been approved for human use, largely because of the lack of a safe and effective mucosal adjuvant. Here, we show that enzymatically synthesized polymeric caffeic acid (pCA) can act as a potent mucosal adjuvant in mice. Intranasal administration of ovalbumin (OVA) in combination with pCA resulted in the induction of OVA-specific mucosal IgA and serum IgG, especially IgG1. Importantly, pCA was synthesized from caffeic acid and horseradish peroxidase from coffee beans and horseradish, respectively, which are commonly consumed. Therefore, pCA is believed to be a highly safe material. In fact, administration of pCA did not show distinct toxicity in mice. These data indicate that pCA has merit for use as a mucosal adjuvant for nasal vaccine formulations.
Collapse
Affiliation(s)
| | | | | | | | | | - Hiroshi Kiyono
- Division of Mucosal Immunology and International Research and Development Center for Mucosal Vaccines, Department of Microbiology and Immunology, The Institute of Medical Science , The University of Tokyo , Tokyo 108-8639 , Japan
| | - Jun Kunisawa
- Division of Mucosal Immunology and International Research and Development Center for Mucosal Vaccines, Department of Microbiology and Immunology, The Institute of Medical Science , The University of Tokyo , Tokyo 108-8639 , Japan.,Laboratory of Vaccine Materials , National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) , Osaka 567-0085 , Japan
| | | |
Collapse
|
14
|
Corthésy B, Bioley G. Lipid-Based Particles: Versatile Delivery Systems for Mucosal Vaccination against Infection. Front Immunol 2018; 9:431. [PMID: 29563912 PMCID: PMC5845866 DOI: 10.3389/fimmu.2018.00431] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
Vaccination is the process of administering immunogenic formulations in order to induce or harness antigen (Ag)-specific antibody and T cell responses in order to protect against infections. Important successes have been obtained in protecting individuals against many deleterious pathological situations after parenteral vaccination. However, one of the major limitations of the current vaccination strategies is the administration route that may not be optimal for the induction of immunity at the site of pathogen entry, i.e., mucosal surfaces. It is now well documented that immune responses along the genital, respiratory, or gastrointestinal tracts have to be elicited locally to ensure efficient trafficking of effector and memory B and T cells to mucosal tissues. Moreover, needle-free mucosal delivery of vaccines is advantageous in terms of safety, compliance, and ease of administration. However, the quest for mucosal vaccines is challenging due to (1) the fact that Ag sampling has to be performed across the epithelium through a relatively limited number of portals of entry; (2) the deleterious acidic and proteolytic environment of the mucosae that affect the stability, integrity, and retention time of the applied Ags; and (3) the tolerogenic environment of mucosae, which requires the addition of adjuvants to elicit efficient effector immune responses. Until now, only few mucosally applicable vaccine formulations have been developed and successfully tested. In animal models and clinical trials, the use of lipidic structures such as liposomes, virosomes, immune stimulating complexes, gas-filled microbubbles and emulsions has proven efficient for the mucosal delivery of associated Ags and the induction of local and systemic immune reponses. Such particles are suitable for mucosal delivery because they protect the associated payload from degradation and deliver concentrated amounts of Ags via specialized sampling cells (microfold cells) within the mucosal epithelium to underlying antigen-presenting cells. The review aims at summarizing recent development in the field of mucosal vaccination using lipid-based particles. The modularity ensured by tailoring the lipidic design and content of particles, and their known safety as already established in humans, make the continuing appraisal of these vaccine candidates a promising development in the field of targeted mucosal vaccination.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Gilles Bioley
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
15
|
Jiang Y, Yang G, Wang Q, Wang Z, Yang W, Gu W, Shi C, Wang J, Huang H, Wang C. Molecular mechanisms underlying protection against H9N2 influenza virus challenge in mice by recombinant Lactobacillus plantarum with surface displayed HA2-LTB. J Biotechnol 2017; 259:6-14. [PMID: 28811215 DOI: 10.1016/j.jbiotec.2017.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/24/2017] [Accepted: 08/09/2017] [Indexed: 12/29/2022]
Abstract
It has been considered that the Avian influenza virus (AIV) causes severe threats to poultry industry. In this study, we constructed a series of recombinant Lactobacillus plantarum (L. plantarum) with surface displayed hemagglutinin subunit 2 (HA2) alone or together with heat-labile toxin B subunit (LTB) from enterotoxigenic Escherichia coli. Balb/c mice were used as model to evaluate the protective effects of recombinant L. plantarum strains against H9N2 subtype challenge. The results showed that the presence of LTB significantly increased the percentages of CD3+CD4+IL-4+, CD3+CD4+IFN-γ+ and CD3+CD4+IL-17+ T cells, as well as CD3+CD8+IFN-γ+ T cells in spleen and MLNs determined by Fluorescence-Activated Cell Sorting assay. Similar increased production of serum IFN-γ was also confirmed by enzyme linked immunosorbent assay (ELISA). The L. plantarum with surface displayed HA2-LTB also dramatically increased the percentages of B220+ IgA+ B cells in peyer patch, in consistent with elevated production of mucosal SIgA antibody determined by ELISA. Finally, the orally administrated HA2-LTB expressing strain efficiently protected mice against H9N2 subtype AIV challenge shown by increased survival percentages, body weight gains and decreased lung lesions in histopathologic analysis. In conclusion, this study provides more detail mechanisms underlying the adjuvant effects of LTB on heterologous antigen produced in recombinant lactic acid bacteria.
Collapse
Affiliation(s)
- Yanlong Jiang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Guilian Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Qi Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Zhannan Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wentao Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wei Gu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China; Shandong Baolai-Leelai Bio-Tech Co., LTD, Taian, Shandong Province, 171000, China
| | - Chunwei Shi
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jianzhong Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Haibin Huang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chunfeng Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
16
|
Mancha-Agresti P, de Castro CP, Dos Santos JSC, Araujo MA, Pereira VB, LeBlanc JG, Leclercq SY, Azevedo V. Recombinant Invasive Lactococcus lactis Carrying a DNA Vaccine Coding the Ag85A Antigen Increases INF-γ, IL-6, and TNF-α Cytokines after Intranasal Immunization. Front Microbiol 2017; 8:1263. [PMID: 28744263 PMCID: PMC5504179 DOI: 10.3389/fmicb.2017.01263] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/23/2017] [Indexed: 11/29/2022] Open
Abstract
Tuberculosis (TB) remains a major threat throughout the world and in 2015 it caused the death of 1.4 million people. The Bacillus Calmette-Guérin is the only existing vaccine against this ancient disease; however, it does not provide complete protection in adults. New vaccines against TB are eminently a global priority. The use of bacteria as vehicles for delivery of vaccine plasmids is a promising vaccination strategy. In this study, we evaluated the use of, an engineered invasive Lactococcus lactis (expressing Fibronectin-Binding Protein A from Staphylococcus aureus) for the delivery of DNA plasmid to host cells, especially to the mucosal site as a new DNA vaccine against tuberculosis. One of the major antigens documented that offers protective responses against Mycobacterium tuberculosis is the Ag85A. L. lactis FnBPA+ (pValac:Ag85A) which was obtained and used for intranasal immunization of C57BL/6 mice and the immune response profile was evaluated. In this study we observed that this strain was able to produce significant increases in the amount of pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-6) in the stimulated spleen cell supernatants, showing a systemic T helper 1 (Th1) cell response. Antibody production (IgG and sIgA anti-Ag85A) was also significantly increased in bronchoalveolar lavage, as well as in the serum of mice. In summary, these findings open new perspectives in the area of mucosal DNA vaccine, against specific pathogens using a Lactic Acid Bacteria such as L. lactis.
Collapse
Affiliation(s)
- Pamela Mancha-Agresti
- Laboratory of Cellular and Molecular Genetics, Department of General Biology, Instituto de Ciências Biológicas - Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Camila Prosperi de Castro
- Laboratory of Cellular and Molecular Genetics, Department of General Biology, Instituto de Ciências Biológicas - Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Janete S C Dos Santos
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel DiasBelo Horizonte, Brazil
| | - Maíra A Araujo
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel DiasBelo Horizonte, Brazil
| | - Vanessa B Pereira
- Laboratory of Cellular and Molecular Genetics, Department of General Biology, Instituto de Ciências Biológicas - Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Jean G LeBlanc
- Centro de Referencia para Lactobacilos - Consejo Nacional de Investigaciones Científicas y TécnicasSan Miguel de Tucumán, Argentina
| | - Sophie Y Leclercq
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel DiasBelo Horizonte, Brazil
| | - Vasco Azevedo
- Laboratory of Cellular and Molecular Genetics, Department of General Biology, Instituto de Ciências Biológicas - Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| |
Collapse
|
17
|
McKay PF, Mann JFS, Pattani A, Kett V, Aldon Y, King D, Malcolm RK, Shattock RJ. Intravaginal immunisation using a novel antigen-releasing ring device elicits robust vaccine antigen-specific systemic and mucosal humoral immune responses. J Control Release 2017; 249:74-83. [PMID: 28115243 PMCID: PMC5333785 DOI: 10.1016/j.jconrel.2017.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/23/2016] [Accepted: 01/10/2017] [Indexed: 01/28/2023]
Abstract
The generation of effective levels of antigen-specific immunity at the mucosal sites of pathogen entry is a key goal for vaccinologists. We explored topical vaginal application as an approach to initiate local antigen-specific immunity, enhance previously existing systemic immunity or re-target responses to the mucosae. To deliver a protein vaccine formulation to the vaginal mucosal surface, we used a novel vaginal ring device comprising a silicone elastomer body into which three freeze-dried, rod-shaped, hydroxypropylmethylcellulose inserts were incorporated. Each rod contained recombinant HIV-1 CN54gp140 protein (167μg)±R848 (167μg) adjuvant. The inserts were loaded into cavities within each ring such that only the ends of the inserts were initially exposed. Sheep received a prime-boost vaccination regime comprising intramuscular injection of 100μg CN54gp140+200μg R848 followed by three successive ring applications of one week duration and separated by one month intervals. Other sheep received only the ring devices without intramuscular priming. Serum and vaginal mucosal fluids were sampled every two weeks and analysed by CN54gp140 ELISA and antigen-specific B cells were measured by flow cytometry at necropsy. Vaccine antigen-specific serum antibody responses were detected in both the intramuscularly-primed and vaginal mucosally-primed groups. Those animals that received only vaginal vaccinations had identical IgG but superior IgA responses. Analysis revealed that all animals exhibited mucosal antigen-specific IgG and IgA with the IgA responses 30-fold greater than systemic levels. Importantly, very high numbers of antigen-specific B cells were detected in local genital draining lymph nodes. We have elicited local genital antigen-specific immune responses after topical application of an adjuvanted antigen formulation within a novel vaginal ring vaccine release device. This regimen and delivery method elicited high levels of antigen-specific mucosal IgA and large numbers of local antigen-reactive B cells, both likely essential for effective mucosal protection.
Collapse
Affiliation(s)
- Paul F McKay
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK.
| | - Jamie F S Mann
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK
| | - Aditya Pattani
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Vicky Kett
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Yoann Aldon
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK
| | - Deborah King
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK
| | - R Karl Malcolm
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Robin J Shattock
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
18
|
Agger EM. Novel adjuvant formulations for delivery of anti-tuberculosis vaccine candidates. Adv Drug Deliv Rev 2016; 102:73-82. [PMID: 26596558 DOI: 10.1016/j.addr.2015.11.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/30/2015] [Accepted: 11/16/2015] [Indexed: 01/18/2023]
Abstract
There is an urgent need for a new and improved vaccine against tuberculosis for controlling this disease that continues to pose a global health threat. The current research strategy is to replace the present BCG vaccine or boost BCG-immunity with subunit vaccines such as viral vectored- or protein-based vaccines. The use of recombinant proteins holds a number of production advantages including ease of scalability, but requires an adjuvant inducing cell-mediated immune responses. A number of promising novel adjuvant formulations have recently been designed and show evidence of induction of cellular immune responses in humans. A common trait of effective TB adjuvants including those already in current clinical testing is a two-component approach combining a delivery system with an appropriate immunomodulator. This review summarizes the status of current TB adjuvant research with a focus on the division of labor between delivery systems and immunomodulators.
Collapse
Affiliation(s)
- Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark.
| |
Collapse
|
19
|
Tomar J, Born PA, Frijlink HW, Hinrichs WLJ. Dry influenza vaccines: towards a stable, effective and convenient alternative to conventional parenteral influenza vaccination. Expert Rev Vaccines 2016; 15:1431-1447. [DOI: 10.1080/14760584.2016.1182869] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
20
|
Ma Y. Recent advances in nontoxicEscherichia coliheat-labile toxin and its derivative adjuvants. Expert Rev Vaccines 2016; 15:1361-1371. [DOI: 10.1080/14760584.2016.1182868] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
21
|
Siefert AL, Caplan MJ, Fahmy TM. Artificial bacterial biomimetic nanoparticles synergize pathogen-associated molecular patterns for vaccine efficacy. Biomaterials 2016; 97:85-96. [PMID: 27162077 DOI: 10.1016/j.biomaterials.2016.03.039] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/16/2016] [Accepted: 03/28/2016] [Indexed: 11/19/2022]
Abstract
Antigen-presenting cells (APCs) sense microorganisms via pathogen-associated molecular patterns (PAMPs) by both extra- and intracellular Toll-like Receptors (TLRs), initiating immune responses against invading pathogens. Bacterial PAMPs include extracellular lipopolysaccharides and intracellular unmethylated CpG-rich oligodeoxynucleotides (CpG). We hypothesized that a biomimetic approach involving antigen-loaded nanoparticles (NP) displaying Monophosphoryl Lipid A (MPLA) and encapsulating CpG may function as an effective "artificial bacterial" biomimetic vaccine platform. This hypothesis was tested in vitro and in vivo using NP assembled from biodegradable poly(lactic-co-glycolic acid) (PLGA) polymer, surface-modified with MPLA, and loaded with CpG and model antigen Ovalbumin (OVA). First, CpG potency, characterized by cytokine profiles, titers, and antigen-specific T cell responses, was enhanced when CpG was encapsulated in NP compared to equivalent concentrations of surface-presented CpG, highlighting the importance of biomimetic presentation of PAMPs. Second, NP synergized surface-bound MPLA with encapsulated CpG in vitro and in vivo, inducing greater pro-inflammatory, antigen-specific T helper 1 (Th1)-skewed cellular and antibody-mediated responses compared to single PAMPs or soluble PAMP combinations. Importantly, NP co-presentation of CpG and MPLA was critical for CD8(+) T cell responses, as vaccination with a mixture of NP presenting either CpG or MPLA failed to induce cellular immunity. This work demonstrates a rational methodology for combining TLR ligands in a context-dependent manner for synergistic nanoparticulate vaccines.
Collapse
Affiliation(s)
- Alyssa L Siefert
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Michael J Caplan
- Department of Molecular and Cellular Physiology, Yale University, New Haven, CT 06520, USA
| | - Tarek M Fahmy
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Department of Chemical and Environmental Engineering, Yale University, New Haven, CT 06520, USA; Department of Immunobiology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
22
|
Orr MT, Beebe EA, Hudson TE, Argilla D, Huang PWD, Reese VA, Fox CB, Reed SG, Coler RN. Mucosal delivery switches the response to an adjuvanted tuberculosis vaccine from systemic TH1 to tissue-resident TH17 responses without impacting the protective efficacy. Vaccine 2015; 33:6570-8. [PMID: 26541135 DOI: 10.1016/j.vaccine.2015.10.115] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/25/2015] [Accepted: 10/28/2015] [Indexed: 11/20/2022]
Abstract
Pulmonary tuberculosis (TB) remains one of the leading causes of infectious disease death despite widespread usage of the BCG vaccine. A number of new TB vaccines have moved into clinical evaluation to replace or boost the BCG vaccine including ID93+GLA-SE, an adjuvanted subunit vaccine. The vast majority of new TB vaccines in trials are delivered parenterally even though intranasal delivery can augment lung-resident immunity and protective efficacy in small animal models. Parenteral immunization with the adjuvanted subunit vaccine ID93+GLA-SE elicits robust TH1 immunity and protection against aerosolized Mycobacterium tuberculosis in mice and guinea pigs. Here we describe the immunogenicity and efficacy of this vaccine when delivered intranasally. Intranasal delivery switches the CD4 T cell response from a TH1 to a TH17 dominated tissue-resident response with increased frequencies of ID93-specific cells in both the lung tissue and at the lung surface. Surprisingly these changes do not affect the protective efficacy of ID93+GLA-SE. Unlike intramuscular immunization, ID93+GLA does not require the squalene-based oil-in-water emulsion SE to elicit protective CD4 T cells when delivered intranasally. Finally we demonstrate that TNF and the IL-17 receptor are dispensable for the efficacy of the intranasal vaccine suggesting an alternative mechanism of protection.
Collapse
Affiliation(s)
- Mark T Orr
- Infectious Disease Research Institute, Seattle, WA 98102, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA.
| | - Elyse A Beebe
- Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - Thomas E Hudson
- Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - David Argilla
- Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - Po-Wei D Huang
- Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - Valerie A Reese
- Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - Christopher B Fox
- Infectious Disease Research Institute, Seattle, WA 98102, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, WA 98102, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Rhea N Coler
- Infectious Disease Research Institute, Seattle, WA 98102, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
23
|
Ayala JC, Pimienta E, Rodríguez C, Sarzo M, Jones J, Vallín C, Guerrero A, Milanés MT, Anné J, Mellaert LV, Huygen K. Assessment of an ELISA for serodiagnosis of active pulmonary tuberculosis in a Cuban population. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2015. [DOI: 10.1016/s2222-1808(15)60943-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
24
|
Zhang L, Wang W, Wang S. Effect of vaccine administration modality on immunogenicity and efficacy. Expert Rev Vaccines 2015; 14:1509-23. [PMID: 26313239 DOI: 10.1586/14760584.2015.1081067] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The many factors impacting the efficacy of a vaccine can be broadly divided into three categories: features of the vaccine itself, including immunogen design, vaccine type, formulation, adjuvant and dosing; individual variations among vaccine recipients and vaccine administration-related parameters. While much literature exists related to vaccines, and recently systems biology has started to dissect the impact of individual subject variation on vaccine efficacy, few studies have focused on the role of vaccine administration-related parameters on vaccine efficacy. Parenteral and mucosal vaccinations are traditional approaches for licensed vaccines; novel vaccine delivery approaches, including needless injection and adjuvant formulations, are being developed to further improve vaccine safety and efficacy. This review provides a brief summary of vaccine administration-related factors, including vaccination approach, delivery route and method of administration, to gain a better understanding of their potential impact on the safety and immunogenicity of candidate vaccines.
Collapse
Affiliation(s)
- Lu Zhang
- a 1 Department of Infectious Diseases, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.,b 2 China-US Vaccine Research Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wei Wang
- c 3 Wang Biologics, LLC, Chesterfield, MO 63017, USA ; Current affiliation: Bayer HealthCare, Berkeley, CA 94710, USA
| | - Shixia Wang
- d 4 Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
25
|
Comparison of adjuvants for a spray freeze-dried whole inactivated virus influenza vaccine for pulmonary administration. Eur J Pharm Biopharm 2015; 93:231-41. [PMID: 25896446 DOI: 10.1016/j.ejpb.2015.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 03/25/2015] [Accepted: 04/09/2015] [Indexed: 02/02/2023]
Abstract
Stable vaccines administered to the lungs by inhalation could circumvent many of the problems associated with current immunizations against respiratory infections. We earlier provided proof of concept in mice that pulmonary delivered whole inactivated virus (WIV) influenza vaccine formulated as a stable dry powder effectively elicits influenza-specific antibodies in lung and serum. Yet, mucosal IgA, considered particularly important for protection at the site of virus entry, was poorly induced. Here we investigate the suitability of various Toll-like receptor (TLR) ligands and the saponin-derived compound GPI-0100 to serve as adjuvant for influenza vaccine administered to the lungs as dry powder. The TLR ligands palmitoyl-3-cysteine-serine-lysine-4 (Pam3CSK4), monophosphoryl lipid A (MPLA) and CpG oligodeoxynucleotides (CpG ODN) as well as GPI-0100 tolerated the process of spray freeze-drying well. While Pam3CSK4 had no effect on systemic antibody titers, all the other adjuvants significantly increased influenza-specific serum and lung IgG titers. Yet, only GPI-0100 also enhanced mucosal IgA titers. Moreover, only GPI-0100-adjuvanted WIV provided partial protection against heterologous virus challenge. Pulmonary immunization with GPI-0100-adjuvanted vaccine did not induce an overt inflammatory response since influx of neutrophils and production of inflammatory cytokines were moderate and transient and lung histology was normal. Our results indicate that a GPI-0100-adjuvanted dry powder influenza vaccine is a safe and effective alternative to current parenteral vaccines.
Collapse
|
26
|
Zhang N, Channappanavar R, Ma C, Wang L, Tang J, Garron T, Tao X, Tasneem S, Lu L, Tseng CTK, Zhou Y, Perlman S, Jiang S, Du L. Identification of an ideal adjuvant for receptor-binding domain-based subunit vaccines against Middle East respiratory syndrome coronavirus. Cell Mol Immunol 2015; 13:180-90. [PMID: 25640653 PMCID: PMC4786625 DOI: 10.1038/cmi.2015.03] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 11/09/2022] Open
Abstract
Middle East respiratory syndrome (MERS), an emerging infectious disease caused by MERS coronavirus (MERS-CoV), has garnered worldwide attention as a consequence of its continuous spread and pandemic potential, making the development of effective vaccines a high priority. We previously demonstrated that residues 377–588 of MERS-CoV spike (S) protein receptor-binding domain (RBD) is a very promising MERS subunit vaccine candidate, capable of inducing potent neutralization antibody responses. In this study, we sought to identify an adjuvant that optimally enhanced the immunogenicity of S377–588 protein fused with Fc of human IgG (S377–588-Fc). Specifically, we compared several commercially available adjuvants, including Freund's adjuvant, aluminum, Monophosphoryl lipid A, Montanide ISA51 and MF59 with regard to their capacity to enhance the immunogenicity of this subunit vaccine. In the absence of adjuvant, S377–588-Fc alone induced readily detectable neutralizing antibody and T-cell responses in immunized mice. However, incorporating an adjuvant improved its immunogenicity. Particularly, among the aforementioned adjuvants evaluated, MF59 is the most potent as judged by its superior ability to induce the highest titers of IgG, IgG1 and IgG2a subtypes, and neutralizing antibodies. The addition of MF59 significantly augmented the immunogenicity of S377–588-Fc to induce strong IgG and neutralizing antibody responses as well as protection against MERS-CoV infection in mice, suggesting that MF59 is an optimal adjuvant for MERS-CoV RBD-based subunit vaccines.
Collapse
Affiliation(s)
- Naru Zhang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | | | - Cuiqing Ma
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Lili Wang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Jian Tang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA.,State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China.,Xiang-Ya Medical College, Central South University, Changsha, China
| | - Tania Garron
- Department of Microbiology and Immunology and Center for Biodefense and Emerging Disease, University of Texas Medical Branch, Galveston, TX, USA
| | - Xinrong Tao
- Department of Microbiology and Immunology and Center for Biodefense and Emerging Disease, University of Texas Medical Branch, Galveston, TX, USA
| | - Sumaiya Tasneem
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai, Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
| | - Chien-Te K Tseng
- Department of Microbiology and Immunology and Center for Biodefense and Emerging Disease, University of Texas Medical Branch, Galveston, TX, USA
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China.,Xiang-Ya Medical College, Central South University, Changsha, China
| | - Stanley Perlman
- Department of Microbiology, University of Iowa, Iowa City, IA, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai, Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| |
Collapse
|
27
|
Sharma M, Dixit A. Identification and immunogenic potential of B cell epitopes of outer membrane protein OmpF of Aeromonas hydrophila in translational fusion with a carrier protein. Appl Microbiol Biotechnol 2015; 99:6277-91. [DOI: 10.1007/s00253-015-6398-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/08/2015] [Accepted: 01/08/2015] [Indexed: 01/10/2023]
|
28
|
Verwaerde C, Debrie AS, Dombu C, Legrand D, Raze D, Lecher S, Betbeder D, Locht C. HBHA vaccination may require both Th1 and Th17 immune responses to protect mice against tuberculosis. Vaccine 2014; 32:6240-50. [PMID: 25252198 DOI: 10.1016/j.vaccine.2014.09.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 08/12/2014] [Accepted: 09/08/2014] [Indexed: 11/19/2022]
Abstract
Almost one century after the discovery of the BCG vaccine, tuberculosis remains a major cause of global mortality and morbidity, emphasizing the urgent need to design more efficient vaccines. The heparin-binding haemagglutinin (HBHA) appears to be a promising vaccine candidate, as it was shown to afford protection to mice against a challenge infection with Mycobacterium tuberculosis when combined with the strong adjuvant DDA/MPL (dimethyldioctadecyl-ammonium bromide/monophosphoryl lipid A), a TLR4 ligand. In this study, we investigated the immunological response and protection of mice immunized with HBHA formulated in lipid-containing nanoparticles and adjuvanted with CpG, a TLR9 ligand. Subcutaneous immunization with this HBHA formulation led to a marked Th1 response, characterized by high IFN-γ levels, but no significant IL-17 production, both in spleen and lung, in contrast to DDA/MPL MPL-formulated HBHA, which induced both IFN-γ and IL-17. This cytokine profile was also observed in BCG-primed mice and persisted after M. tuberculosis infection. No significant protection was obtained against challenge infection after vaccination with the nanoparticle-CpG formulation, and this was associated with a failure to mount a memory immune response. These results suggest the importance of both Th1 and Th17 immune responses for vaccine-induced immunity.
Collapse
Affiliation(s)
- Claudie Verwaerde
- Inserm U1019, Lille, France; CNRS UMR8204, Lille, France; Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France; Univ Lille Nord de France, Lille, France.
| | - Anne-Sophie Debrie
- Inserm U1019, Lille, France; CNRS UMR8204, Lille, France; Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France; Univ Lille Nord de France, Lille, France
| | | | - Damien Legrand
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
| | - Dominique Raze
- Inserm U1019, Lille, France; CNRS UMR8204, Lille, France; Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France; Univ Lille Nord de France, Lille, France
| | - Sophie Lecher
- Inserm U1019, Lille, France; CNRS UMR8204, Lille, France; Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France; Univ Lille Nord de France, Lille, France
| | | | - Camille Locht
- Inserm U1019, Lille, France; CNRS UMR8204, Lille, France; Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France; Univ Lille Nord de France, Lille, France
| |
Collapse
|
29
|
Diogo GR, Reljic R. Development of a new tuberculosis vaccine: is there value in the mucosal approach? Immunotherapy 2014; 6:1001-13. [DOI: 10.2217/imt.14.62] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
TB is a global health problem, killing 1.5 million people every year. The only currently available vaccine, Mycobacterium bovis BCG, is effective against severe childhood forms, but it demonstrates a variable efficacy against the pulmonary form of TB in adults. Many of these adult TB cases result from the reactivation of an initially controlled, latent Mycobacterium tuberculosis infection. Effective prophylactic vaccination remains the key long-term strategy for combating TB. Continued belief in reaching this goal requires unrelenting innovation in the formulation and delivery of candidate vaccines. It is also based on the assumption, that the failure of recent human vaccine trials could have been due to a suboptimal vaccine design and delivery, and therefore should not erode the key principle that a TB vaccine is an attainable target. This report gives a brief overview of the mucosal immune system in the context of M. tuberculosis infection, and focuses on the most recent advances in the field of mucosal TB vaccine development, with a specific emphasis on subunit TB vaccines.
Collapse
Affiliation(s)
- Gil Reynolds Diogo
- St George's Hospital, Institute of Infection & Immunity, St George's University of London, London, SW17 0RE, UK
| | - Rajko Reljic
- St George's Hospital, Institute of Infection & Immunity, St George's University of London, London, SW17 0RE, UK
| |
Collapse
|
30
|
Moreno-Mendieta SA, Guillén D, Espitia C, Hernández-Pando R, Sanchez S, Rodríguez-Sanoja R. A novel antigen-carrier system: the Mycobacterium tuberculosis Acr protein carried by raw starch microparticles. Int J Pharm 2014; 474:241-8. [PMID: 25093695 DOI: 10.1016/j.ijpharm.2014.07.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 06/23/2014] [Accepted: 07/24/2014] [Indexed: 01/17/2023]
Abstract
Microparticles have been used as promising carriers for in vivo vaccine delivery. However, the processes for immobilizing peptides or proteins on microparticles usually require the use of undesirable compounds and complex protocols. In this work, we propose a new immobilization and delivery system with raw starch microparticles and a starch binding domain (SBD) tag fusion protein. The heat shock protein alpha crystallin from Mycobacterium tuberculosis was used as model. The immunogenicity of the system was investigated in BALB/c mice inoculated with purified Acr-SBDtag protein (pAcr-SBDtag) and starch immobilized Acr-SBDtag protein (μAcr-SBDtag) by oral and intranasal routes. We demonstrated mucosal immunization with the μAcr-SBDtag protein induced systemic antibodies that were predominantly immunoglobulin G2a (IgG2a). An analysis of the cytokines from spleen cells and lung homogenates revealed that loaded microparticles induced the secretion of interferon-γ (INF-γ), suggesting an adjuvant effect from the immobilization. The immune responses induced by immobilized protein were primarily affected by the route of administration. These results demonstrate that the system exhibits the necessary characteristics to improve antigen release and presentation to antigen presenting cells (APCs) in the mucosae. Because no extra adjuvants were used, we posit that the system may be suitable for delivery and presentation to the field of subunit vaccine development.
Collapse
Affiliation(s)
- S A Moreno-Mendieta
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P.70228, Ciudad Universitaria, México, DF 04510, Mexico; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P.70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - D Guillén
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P.70228, Ciudad Universitaria, México, DF 04510, Mexico; Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México (UNAM), A.P.70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - C Espitia
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P.70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - R Hernández-Pando
- Servicio de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Mexico
| | - S Sanchez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P.70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - R Rodríguez-Sanoja
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P.70228, Ciudad Universitaria, México, DF 04510, Mexico.
| |
Collapse
|
31
|
Tonnis WF, Lexmond AJ, Frijlink HW, de Boer AH, Hinrichs WLJ. Devices and formulations for pulmonary vaccination. Expert Opin Drug Deliv 2013; 10:1383-97. [DOI: 10.1517/17425247.2013.810622] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|