1
|
Goetz MJ, Park KS, Joshi M, Gottlieb AP, Dowling DJ, Mitragotri S. An ionic liquid-based adjuvant for modulating cellular and humoral immune responses. J Control Release 2024; 376:632-645. [PMID: 39437967 DOI: 10.1016/j.jconrel.2024.10.038] [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: 07/25/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Vaccination is an important strategy for the prevention of infectious diseases worldwide. Adjuvants can be incorporated in vaccine formulations to enhance the resultant immune response and subsequently confer more robust protection upon natural infection. While adjuvants have exciting potential to improve vaccination, the landscape of materials employed in clinical adjuvants is small and its expansion is needed to facilitate vaccine development against current and future infectious diseases. This study introduces the first ionic liquid (IL) adjuvant comprised of choline and sorbic acid (ChoSorb) to produce an antigen-specific cellular as well as humoral immune response against multiple antigens. The abilities of ChoSorb as a vaccine adjuvant is evaluated and characterized through material analysis, innate immune responses, and adaptive responses to both a model and clinical grade antigen. With the robust immune responses generated by ChoSorb and the accompanying mechanistic insights, this study introduces ILs as a new class of adjuvant materials for future vaccine design.
Collapse
Affiliation(s)
- Morgan J Goetz
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA
| | - Kyung Soo Park
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA
| | - Maithili Joshi
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA
| | - Alexander P Gottlieb
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA.
| |
Collapse
|
2
|
Oboge H, Riitho V, Nyamai M, Omondi GP, Lacasta A, Githaka N, Nene V, Aboge G, Thumbi SM. Safety and efficacy of toll-like receptor agonists as therapeutic agents and vaccine adjuvants for infectious diseases in animals: a systematic review. Front Vet Sci 2024; 11:1428713. [PMID: 39355141 PMCID: PMC11442433 DOI: 10.3389/fvets.2024.1428713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/20/2024] [Indexed: 10/03/2024] Open
Abstract
Introduction Strengthening global health security relies on adequate protection against infectious diseases through vaccination and treatment. Toll-like receptor (TLR) agonists exhibit properties that can enhance immune responses, making them potential therapeutic agents or vaccine adjuvants. Methods We conducted an extensive systematic review to assess the efficacy of TLR agonists as therapeutic agents or vaccine adjuvants for infectious diseases and their safety profile in animals, excluding rodents and cold-blooded animals. We collected qualitative and available quantitative data on the efficacy and safety outcomes of TLR agonists and employed descriptive analysis to summarize the outcomes. Results Among 653 screened studies, 51 met the inclusion criteria. In this review, 82% (42/51) of the studies used TLR agonists as adjuvants, while 18% (9/51) applied TLR agonist as therapeutic agents. The predominant TLR agonists utilized in animals against infectious diseases was CpG ODN, acting as a TLR9 agonist in mammals, and TLR21 agonists in chickens. In 90% (46/51) of the studies, TLR agonists were found effective in stimulating specific and robust humoral and cellular immune responses, thereby enhancing the efficacy of vaccines or therapeutics against infectious diseases in animals. Safety outcomes were assessed in 8% (4/51) of the studies, with one reporting adverse effects. Discussion Although TLR agonists are efficacious in enhancing immune responses and the protective efficacy of vaccines or therapeutic agents against infectious diseases in animals, a thorough evaluation of their safety is imperative to in-form future clinical applications in animal studies. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=323122.
Collapse
Affiliation(s)
- Harriet Oboge
- Department of Public Health Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - Victor Riitho
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - Mutono Nyamai
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - George P Omondi
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
- Department of Clinical Studies, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Anna Lacasta
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - Naftaly Githaka
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - Vishvanath Nene
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - Gabriel Aboge
- Department of Public Health Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - S M Thumbi
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
3
|
Cui Y, Ho M, Hu Y, Shi Y. Vaccine adjuvants: current status, research and development, licensing, and future opportunities. J Mater Chem B 2024; 12:4118-4137. [PMID: 38591323 PMCID: PMC11180427 DOI: 10.1039/d3tb02861e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Vaccines represent one of the most significant inventions in human history and have revolutionized global health. Generally, a vaccine functions by triggering the innate immune response and stimulating antigen-presenting cells, leading to a defensive adaptive immune response against a specific pathogen's antigen. As a key element, adjuvants are chemical materials often employed as additives to increase a vaccine's efficacy and immunogenicity. For over 90 years, adjuvants have been essential components in many human vaccines, improving their efficacy by enhancing, modulating, and prolonging the immune response. Here, we provide a timely and comprehensive review of the historical development and the current status of adjuvants, covering their classification, mechanisms of action, and roles in different vaccines. Additionally, we perform systematic analysis of the current licensing processes and highlights notable examples from clinical trials involving vaccine adjuvants. Looking ahead, we anticipate future trends in the field, including the development of new adjuvant formulations, the creation of innovative adjuvants, and their integration into the broader scope of systems vaccinology and vaccine delivery. The article posits that a deeper understanding of biochemistry, materials science, and vaccine immunology is crucial for advancing vaccine technology. Such advancements are expected to lead to the future development of more effective vaccines, capable of combating emerging infectious diseases and enhancing public health.
Collapse
Affiliation(s)
- Ying Cui
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Megan Ho
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Yongjie Hu
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
4
|
Criado M, Reyes LE, Marín JFG, Gutiérrez-Expósito D, Zapico D, Espinosa J, Pérez V. Adjuvants influence the immune cell populations present at the injection site granuloma induced by whole-cell inactivated paratuberculosis vaccines in sheep. Front Vet Sci 2024; 11:1284902. [PMID: 38352038 PMCID: PMC10861745 DOI: 10.3389/fvets.2024.1284902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/04/2024] [Indexed: 02/16/2024] Open
Abstract
Vaccination is the most effective tool for paratuberculosis control. Currently, available vaccines prevent the progression of clinical disease in most animals but do not fully protect them against infection and induce the formation of an injection site granuloma. The precise mechanisms that operate in response to vaccination and granuloma development, as well as the effect that adjuvants could trigger, have not been fully investigated. Therefore, this study aimed to investigate the injection site granulomas induced by two inactivated paratuberculosis vaccines, which differ in the adjuvant employed. Two groups of 45-day-old lambs were immunized with two commercially available vaccines-one (n = 4) with Gudair® and the other (n = 4) with Silirum®. A third group (n = 4) was not vaccinated and served as control. The peripheral humoral response was assessed throughout the study by a commercial anti-Mycobacterium avium subspecies paratuberculosis (Map) antibody indirect ELISA, and the cellular immune response was assessed similarly by the IFN-γ release and comparative intradermal tests. The injection site granulomas were measured during the experiment and sampled at 75 days post-vaccination (dpv) when the animals were euthanized. The tissue damage, antigen and adjuvant distribution, and the presence and amount of immune cells were then determined and assessed by immunohistochemical methods. Antibodies against Map antigens; a general macrophage marker (Iba1), M1 (iNOS), and M2 (CD204) macrophages; T (CD3), B (CD20), and γδ T lymphocytes, proteins MHC-II and NRAMP1, and cytokines IL-4, IL-10, TNF, and IFN-γ were employed. Silirum® elicited a stronger peripheral cellular immune response than Gudair®, while the latter induced larger granulomas and more tissue damage at the site of injection. Additionally, adjuvant and Map antigen distribution throughout the granulomatous inflammatory infiltrate, as well as the NRAMP1 cell expression, which is linked to antigen phagocytosis, were highly irregular. In Silirum® induced granulomas, a higher number of MHC-II and TNF-expressing cells and a lower number of M2 macrophages suggested an improved antigen presentation, which could be due to the better antigen distribution and reduced tissue damage induced by this vaccine.
Collapse
Affiliation(s)
- Miguel Criado
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas-Grulleros, León, Spain
| | - Luis E. Reyes
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Juan F. García Marín
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas-Grulleros, León, Spain
| | - Daniel Gutiérrez-Expósito
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas-Grulleros, León, Spain
| | - David Zapico
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas-Grulleros, León, Spain
| | - José Espinosa
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas-Grulleros, León, Spain
| | - Valentín Pérez
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, León, Spain
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas-Grulleros, León, Spain
| |
Collapse
|
5
|
Gholami S, Korosec CS, Farhang-Sardroodi S, Dick DW, Craig M, Ghaemi MS, Ooi HK, Heffernan JM. A mathematical model of protein subunits COVID-19 vaccines. Math Biosci 2023; 358:108970. [PMID: 36773843 PMCID: PMC9911981 DOI: 10.1016/j.mbs.2023.108970] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 02/12/2023]
Abstract
We consider a general mathematical model for protein subunit vaccine with a focus on the MF59-adjuvanted spike glycoprotein-clamp vaccine for SARS-CoV-2, and use the model to study immunological outcomes in the humoral and cell-mediated arms of the immune response from vaccination. The mathematical model is fit to vaccine clinical trial data. We elucidate the role of Interferon-γ and Interleukin-4 in stimulating the immune response of the host. Model results, and results from a sensitivity analysis, show that a balance between the TH1 and TH2 arms of the immune response is struck, with the TH1 response being dominant. The model predicts that two-doses of the vaccine at 28 days apart will result in approximately 85% humoral immunity loss relative to peak immunity approximately 6 months post dose 1.
Collapse
Affiliation(s)
- Samaneh Gholami
- Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada.
| | - Chapin S Korosec
- Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada
| | - Suzan Farhang-Sardroodi
- Department of Mathematics, University of Manitoba, Winnipeg, Manitoba, Canada; Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada
| | - David W Dick
- Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada
| | - Morgan Craig
- Sainte-Justine University Hospital Research Centre and Department of Mathematics and Statistics, Université de Montréal, Montreal, Quebec, Canada
| | - Mohammad Sajjad Ghaemi
- Digital Technologies Research Centre, National Research Council Canada, Toronto, ON, Canada
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, Toronto, ON, Canada
| | - Jane M Heffernan
- Modelling Infection & Immunity Lab, Centre for Disease Modelling, Mathematics & Statistics, York University, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Nooraei S, Sarkar Lotfabadi A, Akbarzadehmoallemkolaei M, Rezaei N. Immunogenicity of Different Types of Adjuvants and Nano-Adjuvants in Veterinary Vaccines: A Comprehensive Review. Vaccines (Basel) 2023; 11:vaccines11020453. [PMID: 36851331 PMCID: PMC9962389 DOI: 10.3390/vaccines11020453] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Vaccination is the best way to prevent and reduce the damage caused by infectious diseases in animals and humans. So, several vaccines are used for prophylactic purposes before the pathogen infects, while therapeutic vaccines strengthen the immune system after infection with the pathogen. Adjuvants are molecules, compounds, or macromolecules that enhance non-specific immunity and, in collaboration with antigen(s), can improve the body's immune responses and change the type of immune response. The potential and toxicity of adjuvants must be balanced to provide the safest stimulation with the fewest side effects. In order to overcome the limitations of adjuvants and the effective and controlled delivery of antigens, attention has been drawn to nano-carriers that can be a promising platform for better presenting and stimulating the immune system. Some studies show that nanoparticles have a more remarkable ability to act as adjuvants than microparticles. Because nano-adjuvants inactively target antigen-presenting cells (APCs) and change their chemical surface, nanoparticles also perform better in targeted antigen delivery because they cross biological barriers more easily. We collected and reviewed various types of nano-adjuvants with their specific roles in immunogenicity as a prominent strategy used in veterinary vaccines in this paper.
Collapse
Affiliation(s)
- Soren Nooraei
- Faculty of Veterinary Medicine, Shahrekord University, Shahrekord 8818634141, Iran
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Alireza Sarkar Lotfabadi
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Milad Akbarzadehmoallemkolaei
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Nima Rezaei
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran 1419733151, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417653761, Iran
- Correspondence:
| |
Collapse
|
7
|
Kim KR, Lee AS, Kim SM, Heo HR, Kim CS. Virus-like nanoparticles as a theranostic platform for cancer. Front Bioeng Biotechnol 2023; 10:1106767. [PMID: 36714624 PMCID: PMC9878189 DOI: 10.3389/fbioe.2022.1106767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Virus-like nanoparticles (VLPs) are natural polymer-based nanomaterials that mimic viral structures through the hierarchical assembly of viral coat proteins, while lacking viral genomes. VLPs have received enormous attention in a wide range of nanotechnology-based medical diagnostics and therapies, including cancer therapy, imaging, and theranostics. VLPs are biocompatible and biodegradable and have a uniform structure and controllable assembly. They can encapsulate a wide range of therapeutic and diagnostic agents, and can be genetically or chemically modified. These properties have led to sophisticated multifunctional theranostic platforms. This article reviews the current progress in developing and applying engineered VLPs for molecular imaging, drug delivery, and multifunctional theranostics in cancer research.
Collapse
Affiliation(s)
- Kyeong Rok Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Ae Sol Lee
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Su Min Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Hye Ryoung Heo
- Senotherapy-Based Metabolic Disease Control Research Center, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| | - Chang Sup Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea,School of Chemistry and Biochemistry, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| |
Collapse
|
8
|
Campra NA, Reinoso EB, Montironi ID, Moliva MV, Raviolo J, Ruiz Moreno F, Marin C, Camacho NM, Paredes AJ, Morán MC, Estein SM, Maletto BA, Palma SD, Cariddi LN. Spray-drying-microencapsulated Minthostachys verticillata essential oil and limonene as innovative adjuvant strategy to bovine mastitis vaccines. Res Vet Sci 2022; 149:136-150. [PMID: 35792419 DOI: 10.1016/j.rvsc.2022.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 11/23/2022]
Abstract
Design of innovative adjuvant strategies with an appropriate safety profile is relevant to developed subunit or inactivated microorganism vaccines for bovine mastitis. Minthostachys verticillata essential oil (EO) has demonstrated ability to stimulate the innate immune response and adjuvant effect similar to Al(OH)3. Here we evaluated the adjuvant effect of EO and its metabolite, limonene (L) alone and microencapsulated by spray-drying, using an inactivated Enterococcus faecium strain bovine-mastitis inducer. The gas chromatography-mass spectrometry analysis showed that microencapsulation process did not alter the EO or L chemistry. Microencapsulated EO (McEO) or L (McL) (2.0, 2.5 and 5.0 mg/ml) decreased the viability of bovine mammary gland epithelial cells in a dose-dependent way. Balb/c mice (n = 32) were subcutaneously inoculated (day 0) and revaccinated (day 14 and 28) with saline solution, inactivated bacteria alone or combined with Incomplete Freund's Adjuvant; EO or L (2.5 mg/ml); McEO or McL (5.0 mg/ml); or microcapsule wall material (Mc) alone (2.5 mg/ml). EO, L, McEO and McL stimulated E. faecium-specific IgG (IgG1 or IgG2a) with opsonizing capacity and increased the proportion of CD4+ and CD8+ T cells producers of IFN-γ. Microencapsulation was an effective strategy to increase the adjuvant potential of EO or L. These new adjuvants deserve further study to evaluate their incorporation into vaccines for bovine mastitis.
Collapse
Affiliation(s)
- Noelia Anahí Campra
- Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Río Cuarto, Córdoba, Argentina; Instituto de Biotecnología Ambiental y Salud (INBIAS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Río Cuarto, Córdoba, Argentina
| | - Elina Beatriz Reinoso
- Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Río Cuarto, Córdoba, Argentina; Instituto de Biotecnología Ambiental y Salud (INBIAS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Río Cuarto, Córdoba, Argentina
| | - Ivana Dalila Montironi
- Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Río Cuarto, Córdoba, Argentina; Instituto de Biotecnología Ambiental y Salud (INBIAS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Río Cuarto, Córdoba, Argentina
| | - Melina Vanesa Moliva
- Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Río Cuarto, Córdoba, Argentina; Instituto de Biotecnología Ambiental y Salud (INBIAS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Río Cuarto, Córdoba, Argentina
| | - José Raviolo
- Departamento de Producción Animal, Universidad Nacional de Río Cuarto, Facultad de Agronomía y Veterinaria, Río Cuarto, Córdoba, Argentina
| | - Federico Ruiz Moreno
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
| | - Constanza Marin
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
| | - Nahuel Matías Camacho
- Departamento de Ciencias Farmacéuticas, Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Alejandro Javier Paredes
- Departamento de Ciencias Farmacéuticas, Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina; School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland, UK
| | - María Celeste Morán
- Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET-CICPBA), Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Buenos Aires, Argentina
| | - Silvia Marcela Estein
- Centro de Investigación Veterinaria Tandil (CIVETAN-CONICET-CICPBA), Facultad de Ciencias Veterinarias (FCV), Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Tandil, Buenos Aires, Argentina
| | - Belkys Angélica Maletto
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
| | - Santiago Daniel Palma
- Departamento de Ciencias Farmacéuticas, Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Laura Noelia Cariddi
- Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Río Cuarto, Córdoba, Argentina; Instituto de Biotecnología Ambiental y Salud (INBIAS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Río Cuarto, Córdoba, Argentina.
| |
Collapse
|
9
|
Khorattanakulchai N, Srisutthisamphan K, Shanmugaraj B, Manopwisedjaroen S, Rattanapisit K, Panapitakkul C, Kemthong T, Suttisan N, Malaivijitnond S, Thitithanyanont A, Jongkaewwattana A, Phoolcharoen W. A recombinant subunit vaccine candidate produced in plants elicits neutralizing antibodies against SARS-CoV-2 variants in macaques. FRONTIERS IN PLANT SCIENCE 2022; 13:901978. [PMID: 36247553 PMCID: PMC9555276 DOI: 10.3389/fpls.2022.901978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/29/2022] [Indexed: 06/16/2023]
Abstract
Since the outbreak of the coronavirus disease (COVID) pandemic in 2019, the development of effective vaccines to combat the infection has been accelerated. With the recent emergence of highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOC), there are concerns regarding the immune escape from vaccine-induced immunity. Hence an effective vaccine against VOC with a potent immune response is required. Our previous study confirmed that the two doses of the plant-produced receptor-binding domain (RBD) of SARS-CoV-2 fused with the Fc region of human IgG1, namely Baiya SARS-CoV-2 Vax 1, showed high immunogenicity in mice and monkeys. Here, we aimed to evaluate the immunogenicity of a three-dose intramuscular injection of Baiya SARS-CoV-2 Vax 1 on days 0, 21, and 133 in cynomolgus monkeys. At 14 days after immunization, blood samples were collected to determine RBD-specific antibody titer, neutralizing antibody, and pseudovirus neutralizing antibody titers. Immunized monkeys developed significantly high levels of antigen-specific antibodies against SARS-CoV-2 compared to the control group. Interestingly, the sera collected from immunized monkeys also showed a neutralizing antibody response against the SARS-CoV-2 VOCs; Alpha, Beta, Gamma, Delta, and Omicron. These findings demonstrate that a three-dose regimen of Baiya SARS-CoV-2 Vax 1 vaccine elicits neutralizing immune response against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Narach Khorattanakulchai
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Kanjana Srisutthisamphan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | | | | | | | - Chalisa Panapitakkul
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Taratorn Kemthong
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi, Thailand
| | - Nutchanat Suttisan
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi, Thailand
| | | | | | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
10
|
Angrand L, Masson JD, Rubio-Casillas A, Nosten-Bertrand M, Crépeaux G. Inflammation and Autophagy: A Convergent Point between Autism Spectrum Disorder (ASD)-Related Genetic and Environmental Factors: Focus on Aluminum Adjuvants. TOXICS 2022; 10:toxics10090518. [PMID: 36136483 PMCID: PMC9502677 DOI: 10.3390/toxics10090518] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 05/10/2023]
Abstract
Autism spectrum disorder (ASD), schizophrenia, and bipolar disorder are genetically complex and heterogeneous neurodevelopmental disorders (NDDs) resulting from genetic factors and gene-environment (GxE) interactions for which onset occurs in early brain development. Recent progress highlights the link between ASD and (i) immunogenetics, neurodevelopment, and inflammation, and (ii) impairments of autophagy, a crucial neurodevelopmental process involved in synaptic pruning. Among various environmental factors causing risk for ASD, aluminum (Al)-containing vaccines injected during critical periods have received special attention and triggered relevant scientific questions. The aim of this review is to discuss the current knowledge on the role of early inflammation, immune and autophagy dysfunction in ASD as well as preclinical studies which question Al adjuvant impacts on brain and immune maturation. We highlight the most recent breakthroughs and the lack of epidemiological, pharmacokinetic and pharmacodynamic data constituting a "scientific gap". We propose additional research, such as genetic studies that could contribute to identify populations at genetic risk, improving diagnosis, and potentially the development of new therapeutic tools.
Collapse
Affiliation(s)
- Loïc Angrand
- Univ Paris Est Créteil, INSERM, IMRB, F-94010 Créteil, France; (L.A.); (J.-D.M.)
- Ecole Nationale Vétérinaire d’Alfort IMRB, F-94700 Maisons-Alfort, France
- INSERM UMR-S 1270, 75005 Paris, France;
- Sorbonne Université, Campus Pierre et Marie Curie, 75005 Paris, France
- Institut du Fer à Moulin, 75005 Paris, France
| | - Jean-Daniel Masson
- Univ Paris Est Créteil, INSERM, IMRB, F-94010 Créteil, France; (L.A.); (J.-D.M.)
- Ecole Nationale Vétérinaire d’Alfort IMRB, F-94700 Maisons-Alfort, France
| | - Alberto Rubio-Casillas
- Biology Laboratory, Autlán Regional Preparatory School, University of Guadalajara, Autlán 48900, Jalisco, Mexico;
- Autlán Regional Hospital, Health Secretariat, Autlán 48900, Jalisco, Mexico
| | - Marika Nosten-Bertrand
- INSERM UMR-S 1270, 75005 Paris, France;
- Sorbonne Université, Campus Pierre et Marie Curie, 75005 Paris, France
- Institut du Fer à Moulin, 75005 Paris, France
| | - Guillemette Crépeaux
- Univ Paris Est Créteil, INSERM, IMRB, F-94010 Créteil, France; (L.A.); (J.-D.M.)
- Ecole Nationale Vétérinaire d’Alfort IMRB, F-94700 Maisons-Alfort, France
- Correspondence:
| |
Collapse
|
11
|
Díaz-Dinamarca DA, Salazar ML, Castillo BN, Manubens A, Vasquez AE, Salazar F, Becker MI. Protein-Based Adjuvants for Vaccines as Immunomodulators of the Innate and Adaptive Immune Response: Current Knowledge, Challenges, and Future Opportunities. Pharmaceutics 2022; 14:1671. [PMID: 36015297 PMCID: PMC9414397 DOI: 10.3390/pharmaceutics14081671] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
New-generation vaccines, formulated with subunits or nucleic acids, are less immunogenic than classical vaccines formulated with live-attenuated or inactivated pathogens. This difference has led to an intensified search for additional potent vaccine adjuvants that meet safety and efficacy criteria and confer long-term protection. This review provides an overview of protein-based adjuvants (PBAs) obtained from different organisms, including bacteria, mollusks, plants, and humans. Notably, despite structural differences, all PBAs show significant immunostimulatory properties, eliciting B-cell- and T-cell-mediated immune responses to administered antigens, providing advantages over many currently adopted adjuvant approaches. Furthermore, PBAs are natural biocompatible and biodegradable substances that induce minimal reactogenicity and toxicity and interact with innate immune receptors, enhancing their endocytosis and modulating subsequent adaptive immune responses. We propose that PBAs can contribute to the development of vaccines against complex pathogens, including intracellular pathogens such as Mycobacterium tuberculosis, those with complex life cycles such as Plasmodium falciparum, those that induce host immune dysfunction such as HIV, those that target immunocompromised individuals such as fungi, those with a latent disease phase such as Herpes, those that are antigenically variable such as SARS-CoV-2 and those that undergo continuous evolution, to reduce the likelihood of outbreaks.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
| | - Michelle L. Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Byron N. Castillo
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Augusto Manubens
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| | - Abel E. Vasquez
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Providencia, Santiago 8320000, Chile
| | - Fabián Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, UK
| | - María Inés Becker
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| |
Collapse
|
12
|
Techawiwattanaboon T, Courant T, Brunner L, Sathean-anan-kun S, Krangvichian P, Iadsee N, Nakornpakdee Y, Sangjun N, Komanee P, Collin N, Ruxrungtham K, Patarakul K. Designing Adjuvant Formulations to Promote Immunogenicity and Protective Efficacy of Leptospira Immunoglobulin-Like Protein A Subunit Vaccine. Front Cell Infect Microbiol 2022; 12:918629. [PMID: 35782116 PMCID: PMC9243587 DOI: 10.3389/fcimb.2022.918629] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022] Open
Abstract
The leptospirosis burden on humans, especially in high-risk occupational groups and livestock, leads to public health and economic problems. Leptospirosis subunit vaccines have been under development and require further improvement to provide complete protection. Adjuvants can be used to enhance the amplitude, quality, and durability of immune responses. Previously, we demonstrated that LMQ adjuvant (neutral liposomes containing monophosphoryl lipid A (MPL) and Quillaja saponaria derived QS21 saponin) promoted protective efficacy of LigAc vaccine against Leptospira challenge. To promote immunogenicity and protective efficacy of the subunit vaccines, three alternative adjuvants based on neutral liposomes or squalene-in-water emulsion were evaluated in this study. LQ and LQuil adjuvants combined the neutral liposomes with the QS21 saponin or Quillaja saponaria derived QuilA® saponin, respectively. SQuil adjuvant combined a squalene-in-water emulsion with the QuilA® saponin. The immunogenicity and protective efficacy of LigAc (20 µg) formulated with the candidate adjuvants were conducted in golden Syrian hamsters. Hamsters were vaccinated three times at a 2-week interval, followed by a homologous challenge of L. interrogans serovar Pomona. The results showed that LigAc combined with LQ, LQuil, or SQuil adjuvants conferred substantial antibody responses and protective efficacy (survival rate, pathological change, and Leptospira renal colonization) comparable to LMQ adjuvant. The LigAc+LQ formulation conferred 62.5% survival but was not significantly different from LigAc+LMQ, LigAc+LQuil, and LigAc+SQuil formulations (50% survival). This study highlights the potential of saponin-containing adjuvants LMQ, LQ, LQuil, and SQuil for both human and animal leptospirosis vaccines.
Collapse
Affiliation(s)
- Teerasit Techawiwattanaboon
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
| | - Thomas Courant
- Vaccine Formulation Institute, Plan-Les-Ouates, Switzerland
| | - Livia Brunner
- Vaccine Formulation Institute, Plan-Les-Ouates, Switzerland
| | - Suwitra Sathean-anan-kun
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
| | - Pratomporn Krangvichian
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
- Medical Microbiology, Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Nutta Iadsee
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
- Medical Microbiology, Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Yaowarin Nakornpakdee
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Noppadon Sangjun
- Laboratory Animal Section, Analysis Division, Armed Force Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Pat Komanee
- Laboratory Animal Section, Analysis Division, Armed Force Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Nicolas Collin
- Vaccine Formulation Institute, Plan-Les-Ouates, Switzerland
| | - Kiat Ruxrungtham
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
| | - Kanitha Patarakul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
- *Correspondence: Kanitha Patarakul, ;
| |
Collapse
|
13
|
Aghamiri S, Noofeli M, Saffarian P, Salehi Najafabadi Z, Goudarzi HR. Investigating preparation and characterisation of diphtheria toxoid-loaded on sodium alginate nanoparticles. IET Nanobiotechnol 2022; 16:199-209. [PMID: 35610737 PMCID: PMC9178656 DOI: 10.1049/nbt2.12088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/18/2022] [Accepted: 05/05/2022] [Indexed: 11/30/2022] Open
Abstract
This paper aims to investigate the preparation and characterisation of the alginate nanoparticles (NPs) as antigen delivery system loaded by diphtheria toxoid (DT). For this purpose, both the loading capacity (LC) and Loading efficiency (LE) of the alginate NPs burdened by DT are evaluated. Moreover, the effects of different concentrations of sodium alginate and calcium chloride on the NPs physicochemical characteristics are surveyed in addition to other physical conditions such as homogenization time and rate. To do so, the NPs are characterised using particle size and distribution, zeta potential, scanning electron microscopy, encapsulation efficiency, in vitro release study and FT‐IR spectroscopy. Subsequently, the effects of homogenization time and rate on the NPs are assessed. At the meantime, the NPs LC and efficiency in several DT concentrations are estimated. The average size of the NPs was 400.7 and 276.6 nm for unloaded and DT loaded, respectively. According to the obtained results, the zeta potential of the blank and DT loaded NPs are estimated as −23.7 mV and −21.2 mV, respectively. Whereas, the LC and LE were >80% and >90%, in that order. Furthermore, 95% of the releasing DT loaded NPs occurs at 140 h in the sustained mode without any bursting release. It can be concluded that the features of NPs such as morphology and particle size are strongly depended on the calcium chloride, sodium alginate concentrations and physicochemical conditions in the NPs formation process. In addition, appropriate concentrations of the sodium alginate and calcium ions would lead to obtaining the desirable NPs formation associated with the advantageous LE, LC (over 80%) and sustained in vitro release profile. Ultimately, the proposed NPs can be employed in vaccine formulation for the targeted delivery, controlled and slow antigen release associated with the improved antigen stability.
Collapse
Affiliation(s)
- Samira Aghamiri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mojtaba Noofeli
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agriculture Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Parvaneh Saffarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Salehi Najafabadi
- Department of Human Bacterial Vaccine, Razi Vaccine and Serum Research Institute, Agriculture Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Hamid Reza Goudarzi
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agriculture Research Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
14
|
Gorse GJ, Grimes S, Buck H, Mulla H, White P, Hill H, May J, Frey SE, Blackburn P. A phase 1 dose-sparing, randomized clinical trial of seasonal trivalent inactivated influenza vaccine combined with MAS-1, a novel water-in-oil adjuvant/delivery system. Vaccine 2022; 40:1271-1281. [PMID: 35125219 DOI: 10.1016/j.vaccine.2022.01.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND New influenza vaccines are needed to increase vaccine efficacy. Adjuvants may allow hemagglutinin (HA) dose-sparing with enhanced immunogenicity. MAS-1 is an investigational low viscosity, free-flowing, water-in-oil emulsion-based adjuvant/delivery system comprised of stable nanoglobular aqueous droplets. METHODS A phase 1, double-blind, safety and immunogenicity, HA dose escalation, randomized clinical trial was conducted. MAS-1 adjuvant with 1, 3, 5 or 9 µg per HA derived from licensed seasonal trivalent high dose inactivated influenza vaccine (IIV, Fluzone HD 60 µg per HA) in a 0.3 mL dose were compared to standard dose IIV (Fluzone SD, 15 µg per HA). Safety was measured by reactogenicity, adverse events, and clinical laboratory tests. Serum hemagglutination inhibition (HAI) antibody titers were measured for immunogenicity. RESULTS Seventy-two subjects, aged 18-47 years, received one dose of either 0.3 mL adjuvanted vaccine or SD IIV intramuscularly. Common injection site and systemic reactions post-vaccination were mild tenderness, induration, pain, headache, myalgia, malaise and fatigue. All reactions resolved within 14 days post-vaccination. Safety laboratory measures were not different between groups. Geometric mean antibody titers, geometric mean fold increases in antibody titer, seroconversion rates and seroprotection rates against vaccine strains were in general higher and of longer duration (day 85 and 169 visits) with MAS-1-adjuvanted IIV at all doses of HA compared with SD IIV. Adjuvanted vaccine induced higher antibody responses against a limited number of non-study vaccine influenza B and A/H3N2 viruses including ones from subsequent years. CONCLUSION MAS-1 adjuvant in a 0.3 mL dose volume provided HA dose-sparing effects without safety concerns and induced higher HAI antibody and seroconversion responses through at least 6 months, demonstrating potential to provide greater vaccine efficacy throughout an influenza season in younger adults. In summary, MAS-1 may provide enhanced, more durable and broader protective immunity compared with non-adjuvanted SD IIV. Clinical Trial Registry: ClinicalTrials.gov # NCT02500680.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sharon E Frey
- Saint Louis University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
15
|
Cao X, Cordova AF, Li L. Therapeutic Interventions Targeting Innate Immune Receptors: A Balancing Act. Chem Rev 2021; 122:3414-3458. [PMID: 34870969 DOI: 10.1021/acs.chemrev.1c00716] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The innate immune system is an organism's first line of defense against an onslaught of internal and external threats. The downstream adaptive immune system has been a popular target for therapeutic intervention, while there is a relative paucity of therapeutics targeting the innate immune system. However, the innate immune system plays a critical role in many human diseases, such as microbial infection, cancer, and autoimmunity, highlighting the need for ongoing therapeutic research. In this review, we discuss the major innate immune pathways and detail the molecular strategies underpinning successful therapeutics targeting each pathway as well as previous and ongoing efforts. We will also discuss any recent discoveries that could inform the development of novel therapeutic strategies. As our understanding of the innate immune system continues to develop, we envision that therapies harnessing the power of the innate immune system will become the mainstay of treatment for a wide variety of human diseases.
Collapse
|
16
|
Cai T, Liu H, Zhang S, Hu J, Zhang L. Delivery of nanovaccine towards lymphoid organs: recent strategies in enhancing cancer immunotherapy. J Nanobiotechnology 2021; 19:389. [PMID: 34823541 PMCID: PMC8620195 DOI: 10.1186/s12951-021-01146-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/14/2021] [Indexed: 01/15/2023] Open
Abstract
With the in-depth exploration on cancer therapeutic nanovaccines, increasing evidence shows that the poor delivery of nanovaccines to lymphoid organs has become the culprit limiting the rapid induction of anti-tumor immune response. Unlike the conventional prophylactic vaccines that mainly form a depot at the injection site to gradually trigger durable immune response, the rapid proliferation of tumors requires an efficient delivery of nanovaccines to lymphoid organs for rapid induction of anti-tumor immunity. Optimization of the physicochemical properties of nanovaccine (e.g., size, shape, charge, colloidal stability and surface ligands) is an effective strategy to enhance their accumulation in lymphoid organs, and nanovaccines with dynamic structures are also designed for precise targeted delivery of lymphoid organs or their subregions. The recent progress of these nanovaccine delivery strategies is highlighted in this review, and the challenges and future direction are also discussed. ![]()
Collapse
Affiliation(s)
- Ting Cai
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315010, China
| | - Huina Liu
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315010, China
| | - Shun Zhang
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315010, China
| | - Jing Hu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China. .,Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 211200, China.
| | - Lingxiao Zhang
- Ningbo Clinical Research Center for Digestive System Tumors, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China. .,Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China. .,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315010, China. .,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
17
|
Johnson W, Boyer I, Bergfeld WF, Belsito DV, Hill RA, Klaassen CD, Liebler DC, Marks JG, Shank RC, Slaga TJ, Snyder PW, Gill LJ, Heldreth B. Safety Assessment of Phosphoric Acid and Its Salts as Used in Cosmetics. Int J Toxicol 2021; 40:34S-85S. [PMID: 34259064 DOI: 10.1177/10915818211014499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Expert Panel for Cosmetic Ingredient Safety (Panel) assessed the safety of Phosphoric Acid and its salts (31 ingredients), which are reported to function as buffering agents, corrosion inhibitors, chelating agents, and pH adjusters in cosmetic products. The Panel reviewed data relating to the safety of these ingredients and concluded that Phosphoric Acid and its salts are safe in the present practices of use and concentration in cosmetics when formulated to be nonirritating.
Collapse
Affiliation(s)
- Wilbur Johnson
- Cosmetic Ingredient Review Senior Scientific Analyst/Writer
| | - Ivan Boyer
- Cosmetic Ingredient Review Former Senior Toxicologist
| | | | | | - Ronald A Hill
- Expert Panel for Cosmetic Ingredient Safety Former Member
| | | | | | - James G Marks
- Expert Panel for Cosmetic Ingredient Safety Former Member
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
![]()
The development of
lipopeptides (lipidated peptides) for vaccines
is discussed, including their role as antigens and/or adjuvants. Distinct
classes of lipopeptide architectures are covered including simple
linear and ligated constructs and lipid core peptides. The design,
synthesis, and immunological responses of the important class of glycerol-based
Toll-like receptor agonist lipopeptides such as Pam3CSK4, which contains three palmitoyl chains and a CSK4 hexapeptide sequence, and many derivatives of this model immunogenic
compound are also reviewed. Self-assembled lipopeptide structures
including spherical and worm-like micelles that have been shown to
act as vaccine agents are also described. The work discussed includes
examples of lipopeptides developed with model antigens, as well as
for immunotherapies to treat many infectious diseases including malaria,
influenza, hepatitis, COVID-19, and many others, as well as cancer
immunotherapies. Some of these have proceeded to clinical development.
The research discussed highlights the huge potential of, and diversity
of roles for, lipopeptides in contemporary and future vaccine development.
Collapse
Affiliation(s)
- Ian W Hamley
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| |
Collapse
|
19
|
Cabaña-Brunod M, Herrera PA, Márquez-Miranda V, Llancalahuen FM, Duarte Y, González-Nilo D, Fuentes JA, Vilos C, Velásquez L, Otero C. Development of a PHBV nanoparticle as a peptide vehicle for NOD1 activation. Drug Deliv 2021; 28:1020-1030. [PMID: 34060399 PMCID: PMC8174487 DOI: 10.1080/10717544.2021.1923862] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
NOD1 is an intracellular receptor that, when activated, induces gene expression of pro-inflammatory factors promoting macrophages and neutrophils recruitment at the infection site. However, iE-DAP, the dipeptide agonist that promotes this receptor's activation, cannot permeate cell membranes. To develop a nanocarrier capable of achieving a high and prolonged activation over time, iE-DAP was encapsulated in nanoparticles (NPs) made of poly (3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV). The physicochemical properties, colloidal stability, encapsulation efficiency, and cellular uptake of iE-DAP-loaded PHVB NPs were analyzed. Results evidenced that physicochemical properties of iE-DAP-loaded NPs remained stable over time, and NPs were efficiently internalized into cells, a process that depends on time and concentration. Moreover, our results showed that NPs elicited a controlled cargo release in vitro, and the encapsulated agonist response was higher than its free form, suggesting the possibility of activating intracellular receptors triggering an immune response through the release of NOD1 agonist.
Collapse
Affiliation(s)
- Mauricio Cabaña-Brunod
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Pablo A Herrera
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Valeria Márquez-Miranda
- Center for Applied Nanotechnology, Faculty of Sciences, Universidad Mayor, Huechuraba, Santiago, Chile
| | - Felipe M Llancalahuen
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Yorley Duarte
- Laboratorio de Fisiopatología Integrativa, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Danilo González-Nilo
- Laboratorio de Fisiopatología Integrativa, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Juan A Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Cristián Vilos
- Laboratory of Nanomedicine and Targeted Delivery, School of Medicine, Universidad de Talca, Talca, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Luis Velásquez
- Instituto de Investigación Interdisciplinar en Ciencias Biomédicas SEK, Facultad de Ciencias de la Salud, Universidad SEK, Santiago, Chile
| | - Carolina Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
20
|
Bartlett S, Skwarczynski M, Toth I. Lipids as Activators of Innate Immunity in Peptide Vaccine Delivery. Curr Med Chem 2020; 27:2887-2901. [PMID: 30362416 DOI: 10.2174/0929867325666181026100849] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 05/16/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Innate immune system plays an important role in pathogen detection and the recognition of vaccines, mainly through pattern recognition receptors (PRRs) that identify pathogen components (danger signals). One of the typically recognised bacterial components are lipids in conjugation with peptides, proteins and saccharides. Lipidic compounds are readily recognised by the immune system, and thus are ideal candidates for peptide- based vaccine delivery. Thus, bacterial or synthetic lipids mixed with, or conjugated to, antigens have shown adjuvant properties. These systems have many advantages over traditional adjuvants, including low toxicity and good efficacy for stimulating mucosal and systemic immune responses. METHODS The most recent literature on the role of lipids in stimulation of immune responses was selected for this review. The vast majority of reviewed papers were published in the last decade. Older but significant findings are also cited. RESULTS This review focuses on the development of lipopeptide vaccine systems including application of palmitic acid, bacterial lipopeptides, glycolipids and the lipid core peptide and their routes of administration. The use of liposomes as a delivery system that incorporates lipopeptides is discussed. The review also includes a brief description of immune system in relation to vaccinology and discussion on vaccine delivery routes. CONCLUSION Lipids and their conjugates are an ideal frontrunner in the development of safe and efficient vaccines for different immunisation routes.
Collapse
Affiliation(s)
- Stacey Bartlett
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD, 4072, Australia
| | - Mariusz Skwarczynski
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD, 4072, Australia
| | - Istvan Toth
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD, 4072, Australia.,The University of Queensland, School of Pharmacy, Woolloongabba, QLD 4102, Australia.,The University of Queensland, Institute for Molecular Bioscience, St Lucia, QLD 4072, Australia
| |
Collapse
|
21
|
Zmarowski A, Ballin JD, Sharits J, Carrico K, Novak J, Shearer J, Blauth B, Ionin B, Reece J, Savransky V. Repeat Dose Toxicity Study of the AV7909 Anthrax Vaccine Candidate in Juvenile Rats. Int J Toxicol 2020; 39:1091581820941412. [PMID: 32691648 DOI: 10.1177/1091581820941412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AV7909 is a next-generation anthrax vaccine candidate indicated for post-exposure prophylaxis of exposure to Bacillus anthracis. AV7909 consists of the Anthrax Vaccine Adsorbed (AVA) bulk drug substance and the immunostimulatory Toll-like receptor 9 agonist oligodeoxynucleotide adjuvant, CPG 7909. Safety testing for pediatric population is warranted to support the potential emergency use of AV7909 in children. This study was conducted to investigate the local tolerance and potential systemic toxicity and their reversibility in juvenile rats by repeat intramuscular injections of the AV7909 vaccine candidate. Animals were dosed on postnatal day (PND) 21 (at weaning), PND 28, and PND 35, with the test article (AV7909), the adjuvant alone (Alhydrogel + CPG 7909), or sterile water for injection. Core group animals were necropsied on PND 37 and recovery group on PND 49. Study end points included survival, clinical observations, injection site observations, body weights, clinical pathology (hematology, coagulation, and clinical chemistry), pro-inflammatory biomarker analysis (alpha-2 macroglobulin [A2M] and alpha-1 acid glycoprotein [AGP]), and anatomic pathology. Immune response to vaccination was measured using the high-throughput anthrax lethal toxin neutralization assay (htpTNA). The AV7909 vaccine candidate produced no apparent systemic or local toxicity. The AGP and A2M levels were elevated in both the adjuvant-alone and AV7909 groups at the end of treatment but were comparable to control levels by the end of the recovery period. All animals in the AV7909 group demonstrated a robust neutralizing antibody response. The results indicate that AV7909 has a favorable safety profile in juvenile rats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bruna Blauth
- Emergent BioSolutions Inc, Gaithersburg, MD, USA
| | - Boris Ionin
- Emergent BioSolutions Inc, Gaithersburg, MD, USA
| | - Joshua Reece
- Emergent BioSolutions Inc, Gaithersburg, MD, USA
| | | |
Collapse
|
22
|
The Construction and Immunoadjuvant Activities of the Oral Interleukin-17B Expressed by Lactobacillus plantarum NC8 Strain in the Infectious Bronchitis Virus Vaccination of Chickens. Vaccines (Basel) 2020; 8:vaccines8020282. [PMID: 32517220 PMCID: PMC7350006 DOI: 10.3390/vaccines8020282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023] Open
Abstract
Interleukin-17B (IL-17B) is a protective cytokine of the IL-17 family and plays an essential role in the regulation of mucosal inflammation. However, little is known about the role of IL-17B in the control of viral infections. In this study, a recombinant Lactobacillus plantarum, designated as NC8-ChIL17B, was constructed to express the chicken IL-17B (ChIL-17B) gene. The recombinant ChIL17B (rChIL17B) protein was about 14 kDa and was anchored to the surface of NC8 cells. In vitro, it was found that the rChIL17B protein inhibited the proliferation of the infectious bronchitis virus (IBV) through activation of nuclear factor kappa B (NF-κB) and the JAK (Janus kinase)-STAT (signal transducers and activators of transcription) signaling. Moreover, to evaluate the immunoadjuvant activities of NC8-ChIL17B, 40 three-day-old specific pathogen-free (SPF) chickens were divided into four groups. Three groups were orally vaccinated with fresh NC8, NC8-ChIL17B, and phosphate buffered saline (PBS), along with the infectious bronchitis virus vaccine, and the other group was the PBS-negative control. The results of the IBV-specific antibody titer and the concentration of the cytokines IL-2, IL-4, IL-6, and interferon gamma (IFN-γ) in sera, as well as the concentration of secretory immunoglobulin A (sIgA) in the tracheal and small intestinal mucosa, the number of cluster of differentiation 4 positive (CD4+) and cluster of differentiation 8 positive (CD8+) T cells in the blood, and the expression of immune-related genes all indicated that NC8-ChIL17B efficiently enhanced the humoral and cellular immune responses to IBV vaccine. Moreover, the viral loads in the NC8-ChIL17B- and IBV-vaccinated group were significantly lower than in the control groups, suggesting a significant promotion of the immunoprotection of IBV vaccination against the virulent IBV strain. Therefore, ChIL-17B is a promising, effective adjuvant candidate for chicken virus vaccines.
Collapse
|
23
|
Xu H, Niu Y, Hong W, Liu W, Zuo X, Bao X, Guo C, Lu Y, Deng B. Development of a water-in-oil-in-water adjuvant for foot-and-mouth disease vaccine based on ginseng stem-leaf saponins as an immune booster. Comp Immunol Microbiol Infect Dis 2020; 71:101499. [PMID: 32505765 DOI: 10.1016/j.cimid.2020.101499] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/15/2020] [Accepted: 05/28/2020] [Indexed: 11/24/2022]
Abstract
There has been an increasing interest in finding new formulations that qualify as vaccine adjuvants, which must be safe, stable, and have the capacity to stimulate a strong immune response. In this study, a basic formulation of a water-in-oil-in-water (W/O/W) adjuvant CV13 was developed, and ginseng stem-leaf saponins (GSLS) were added as an immune booster into oil phase. The physicochemical properties of the adjuvant were tested. Furthermore, the immune activity and the adjuvant effects, as indicated by the foot-and-mouth disease virus (FMDV) antigen were evaluated. The results showed that CV13 was similar in appearance to ISA 206 and could package FMDV antigen into a stable W/O/W emulsion. The FMD vaccine prepared with CV13 alone or CV13 containing GSLS achieved pharmaceutical characteristics comparable to a vaccine prepared with ISA 206, moreover the structural stability of the CV 13 vaccine was found to be better. Mice that were immunized with the FMD vaccine prepared with CV13 containing GSLS presented a significantly higher LPBE antibody titer and splenocyte proliferation rate than those immunized with a vaccine prepared with CV13 alone (p < 0.05). In addition, there was no significant difference between the groups that were immunized with FMD vaccine prepared with CV13 containing GSLS and ISA206 in terms of cellular and humoral immune response. In this paper, CV13 containing GSLS shows excellent immunologic adjuvant effect in mice model, and this new adjuvant may provide a potential choice for FMD vaccine production in the future.
Collapse
Affiliation(s)
- Hai Xu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, 225300, Jiangsu Province, PR China; Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Science, Nanjing, 210014, Jiangsu Province, PR China
| | - Yale Niu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Science, Nanjing, 210014, Jiangsu Province, PR China
| | - Weiming Hong
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, 225300, Jiangsu Province, PR China
| | - Weixin Liu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Science, Nanjing, 210014, Jiangsu Province, PR China
| | - Xiaoxin Zuo
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Science, Nanjing, 210014, Jiangsu Province, PR China
| | - Xi Bao
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Science, Nanjing, 210014, Jiangsu Province, PR China
| | - Changming Guo
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, 225300, Jiangsu Province, PR China
| | - Yu Lu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Science, Nanjing, 210014, Jiangsu Province, PR China; School of Pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, PR China
| | - Bihua Deng
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Science, Nanjing, 210014, Jiangsu Province, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu province, PR China.
| |
Collapse
|
24
|
Flórez-Álvarez L, Ruiz-Perez L, Taborda N, Hernandez JC. Toll-like receptors as a therapeutic target in cancer, infections and inflammatory diseases. Immunotherapy 2020; 12:311-322. [PMID: 32237938 DOI: 10.2217/imt-2019-0096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Toll-like receptors (TLRs) are widely expressed pattern recognition receptors that bind to conserved molecular patterns expressed by pathogens and damaged cells. After recognition, activated TLRs induce the expression of various proinflammatory and antiviral molecules. Thus, TLRs are potential targets for treatment strategies aimed at boosting the adaptive immune response to vaccines, controlling infections, enhancing immune responses during tumor treatment and attenuating immune responses in inflammatory disorders. This Special Report examines the potential of TLRs as targets for the treatment of cancer, infections and inflammatory diseases. Here, we make a particular emphasis on molecules capable of modulating TLRs and their therapeutic applications.
Collapse
Affiliation(s)
- Lizdany Flórez-Álvarez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia.,Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA; Calle 70 No. 52-21, Medellín, Colombia
| | - Lanie Ruiz-Perez
- School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth WA 6845, Australia
| | - Natalia Taborda
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, UdeA; Calle 70 No. 52-21, Medellín, Colombia.,Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| | - Juan C Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| |
Collapse
|
25
|
Savransky V, Lacy M, Ionin B, Skiadopoulos MH, Shearer J. Repeat-Dose Toxicity Study of a Lyophilized Recombinant Protective Antigen-Based Anthrax Vaccine Adjuvanted With CpG 7909. Int J Toxicol 2020; 38:163-172. [PMID: 31179828 DOI: 10.1177/1091581819848722] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A recombinant protective antigen (rPA) anthrax vaccine candidate (rPA7909) was developed as a next-generation vaccine indicated for postexposure prophylaxis of disease resulting from suspected or confirmed Bacillus anthracis exposure. The lyophilized form of rPA7909-vaccinated candidate contains 75 µg purified rPA, 750 µg aluminum (as Alhydrogel adjuvant), and 250 µg of an immunostimulatory Toll-like receptor 9 agonist oligodeoxynucleotide CpG 7909 in a 0.5 mL phosphate-buffered suspension. General toxicity and local reactogenicity were evaluated in Sprague Dawley rats vaccinated with the full human dose of rPA7909 by intramuscular injection. Animals were immunized on study days 1, 15, and 29. Control groups were administered diluent only or adjuvant control (excipients, CpG 7909, and Alhydrogel adjuvant in diluent) intramuscularly at the same dose volume and according to the same schedule used for rPA7909. Toxicity was assessed based on the results of clinical observations, physical examinations, body weights, injection site reactogenicity, ophthalmology, clinical pathology (hematology, coagulation, and serum chemistry), organ weights, and macroscopic and microscopic pathology evaluation. The immune response to rPA7909 vaccination was confirmed by measuring serum anti-PA immunoglobulin G levels. The rPA7909 vaccine produced no apparent systemic toxicity and only transient reactogenicity at the injection site. The injection site reaction from animals receiving the adjuvant control was very similar to those receiving rPA7909 with respect to the inflammation. The inflammatory response observed in the injection site and the draining lymph nodes was consistent with expected immune stimulation. The overall results indicated a favorable safety profile for rPA7909.
Collapse
Affiliation(s)
| | - Michael Lacy
- 1 Emergent BioSolutions Inc, Gaithersburg, MD, USA
| | - Boris Ionin
- 1 Emergent BioSolutions Inc, Gaithersburg, MD, USA
| | | | | |
Collapse
|
26
|
Felix CR, Siedler BS, Barbosa LN, Timm GR, McFadden J, McBride AJA. An overview of human leptospirosis vaccine design and future perspectives. Expert Opin Drug Discov 2019; 15:179-188. [PMID: 31777290 DOI: 10.1080/17460441.2020.1694508] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: It's been 20 years since the first report of a recombinant vaccine that protected against leptospirosis. Since then, numerous recombinant vaccines have been evaluated; however, no recombinant vaccine candidate has advanced to clinical trials. With the ever-increasing burden of leptospirosis, there is an urgent need for a universal vaccine against leptospirosis.Areas covered: This review covers the most promising vaccine candidates that induced significant, reproducible, protection and how advances in the field of bioinformatics has led to the discovery of hundreds of novel protein targets. The authors also discuss the most recent findings regarding the innate immune response and host-pathogen interactions and their impact on the discovery of novel vaccine candidates. In addition, the authors have identified what they believe are the most challenging problems for the discovery and development of a universal vaccine and their potential solutions.Expert opinion: A universal vaccine for leptospirosis will likely only be achieved using a recombinant vaccine as the bacterins are of limited use due to the lack of a cross-protective immune response. Although there are hundreds of novel targets, due to the lack of immune correlates and the need for more research into the basic microbiology of Leptospira spp., a universal vaccine is 10-15 years away.
Collapse
Affiliation(s)
- Carolina R Felix
- Biotechnology Department, Centre for Technological Development, Federal University of Pelotas, Pelotas, Brazil
| | - Bianca S Siedler
- Biotechnology Department, Centre for Technological Development, Federal University of Pelotas, Pelotas, Brazil.,School of Biosciences and Medicine, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Liana N Barbosa
- Biotechnology Department, Centre for Technological Development, Federal University of Pelotas, Pelotas, Brazil
| | - Gabriana R Timm
- Biotechnology Department, Centre for Technological Development, Federal University of Pelotas, Pelotas, Brazil
| | - Johnjoe McFadden
- School of Biosciences and Medicine, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Alan J A McBride
- Biotechnology Department, Centre for Technological Development, Federal University of Pelotas, Pelotas, Brazil
| |
Collapse
|
27
|
One-pot synthesis and characterization of ovalbumin-conjugated gold nanoparticles: A comparative study of adjuvanticity against the physical mixture of ovalbumin and gold nanoparticles. Int J Pharm 2019; 571:118704. [PMID: 31536763 DOI: 10.1016/j.ijpharm.2019.118704] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/09/2019] [Accepted: 09/15/2019] [Indexed: 11/21/2022]
Abstract
Only few adjuvants are licensed for use in humans and there is a need to develop safe and improved vaccine adjuvants. In this study, we report the one-pot synthesis of antigen ovalbumin (OVA)-conjugated gold nanoparticles (OVA@GNPs). A systematical study was performed by comparing OVA@GNPs with the simple mixture of OVA and gold nanoparticles (OVA+GNPs), including their physiochemical properties through spectrometric and electrophoretic analysis, in vitro stability, cytotoxicity and cellular uptake, and in vivo humoral immune responses following subcutaneous and transcutaneous immunization in mice. The results demonstrate a much stronger interaction between protein and GNPs in OVA@GNPs than OVA+GNPs, which makes OVA@GNPs more stable under in vitro conditions than OVA+GNPs with the ability to induce 4 times higher OVA-specific serum IgG titers following subcutaneous immunization. We also show the dose sparing of OVA@GNPs, as the dosage for aluminum adjuvant required to reach to an equivalent OVA-specific antibody titer was almost five times higher than OVA@GNPs. However, we found that the co-administration of small-sized GNPs had a limited ability for the transcutaneous delivery of OVA. These results demonstrate the potential application of one-pot synthesis approach for producing antigen protein-conjugated gold nanoparticles for vaccine delivery.
Collapse
|
28
|
Adjuvants as Delivery Systems in Antigen-Specific Immunotherapies. J Pharm Sci 2019; 108:3831-3841. [PMID: 31526814 DOI: 10.1016/j.xphs.2019.09.011] [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: 03/29/2019] [Revised: 07/23/2019] [Accepted: 09/11/2019] [Indexed: 11/24/2022]
Abstract
Combining autoantigens with immune-modulating drugs has emerged as an attractive approach to selectively reinstate tolerance in autoimmune diseases. The disparate properties of autoantigens and small-molecule immunosuppressants commonly used to treat autoimmune diseases can confound efforts to co-deliver these therapies. However, both components may be co-delivered with adjuvants which have been successful in delivering antigens to immune cells. We evaluated several common adjuvants as vehicles to co-deliver a model antigen and immunosuppressant, ovalbumin (OVA) and dexamethasone (DEX), respectively. Formulations were developed, and the release of DEX from adjuvants was investigated. Next, the effect of adjuvant, DEX, and OVA was tested in vitro using a DC line. A MF59-analog (MF59a) formulation was advanced to more sophisticated co-culture studies using OVA-primed bone marrow-derived dendritic cells and splenocytes or T-cells from OT-II mice. Most of these studies indicated MF59a-based antigen-specific immunotherapies could diminish the markers of inflammation associated with OVA recognition. We rationalized MF59a co-delivery of antigen and drug could reduce the risk of side effects typically associated with these drugs and reinstate immune tolerance, thus prompting continued investigation of emulsion adjuvants as delivery vehicles for antigen-specific immunotherapy of autoimmune diseases.
Collapse
|
29
|
Hay JA, Laurie K, White M, Riley S. Characterising antibody kinetics from multiple influenza infection and vaccination events in ferrets. PLoS Comput Biol 2019; 15:e1007294. [PMID: 31425503 PMCID: PMC6715255 DOI: 10.1371/journal.pcbi.1007294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/29/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022] Open
Abstract
The strength and breadth of an individual's antibody repertoire is an important predictor of their response to influenza infection or vaccination. Although progress has been made in understanding qualitatively how repeated exposures shape the antibody mediated immune response, quantitative understanding remains limited. We developed a set of mathematical models describing short-term antibody kinetics following influenza infection or vaccination and fit them to haemagglutination inhibition (HI) titres from 5 groups of ferrets which were exposed to different combinations of trivalent inactivated influenza vaccine (TIV with or without adjuvant), A/H3N2 priming inoculation and post-vaccination A/H1N1 inoculation. We fit models with various immunological mechanisms that have been empirically observed but have not previously been included in mathematical models of antibody landscapes, including: titre ceiling effects, antigenic seniority and exposure-type specific cross reactivity. Based on the parameter estimates of the best supported models, we describe a number of key immunological features. We found quantifiable differences in the degree of homologous and cross-reactive antibody boosting elicited by different exposure types. Infection and adjuvanted vaccination generally resulted in strong, broadly reactive responses whereas unadjuvanted vaccination resulted in a weak, narrow response. We found that the order of exposure mattered: priming with A/H3N2 improved subsequent vaccine response, and the second dose of adjuvanted vaccination resulted in substantially greater antibody boosting than the first. Either antigenic seniority or a titre ceiling effect were included in the two best fitting models, suggesting a role for a mechanism describing diminishing antibody boosting with repeated exposures. Although there was considerable uncertainty in our estimates of antibody waning parameters, our results suggest that both short and long term waning were present and would be identifiable with a larger set of experiments. These results highlight the potential use of repeat exposure animal models in revealing short-term, strain-specific immune dynamics of influenza.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Viral/blood
- Computational Biology
- Cross Reactions
- Disease Models, Animal
- Ferrets/immunology
- Humans
- Immunization, Secondary
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Kinetics
- Models, Immunological
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Vaccines, Inactivated/administration & dosage
Collapse
Affiliation(s)
- James A. Hay
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| | - Karen Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Seqirus, 63 Poplar Road, Parkville, Victoria, Australia
| | - Michael White
- Malaria: Parasites and Hosts, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Steven Riley
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
30
|
Ratnapriya S, Keerti, Sahasrabuddhe AA, Dube A. Visceral leishmaniasis: An overview of vaccine adjuvants and their applications. Vaccine 2019; 37:3505-3519. [PMID: 31103364 DOI: 10.1016/j.vaccine.2019.04.092] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/08/2019] [Accepted: 04/30/2019] [Indexed: 11/25/2022]
Abstract
Although there has been an extensive research on vaccine development over the last decade and some vaccines have been commercialized for canine visceral leishmaniasis (CVL), but as yet no effective vaccine is available for anthroponotic VL which may partly be due to the absence of an appropriate adjuvant system. Vaccines alone yield poor immunity hence requiring an adjuvant which can boost the immunosuppressed state of VL infected individuals by eliciting adaptive immune responses to achieve required immunological enhancement. Recent studies have documented the continuous efforts that are being made in the field of adjuvants research in an attempt to render vaccines more effective. This review article focuses on adjuvants, particularly particulate and non-particulate ones, which have been assessed with VL vaccine candidates in several preclinical and clinical trials outlining the induction of immune responses obtained from these studies. Moreover, we have emphasized the applicability of multiple adjuvants combination for an improvement in the potential of a VL vaccine.
Collapse
Affiliation(s)
- Sneha Ratnapriya
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Keerti
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Amogh A Sahasrabuddhe
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Anuradha Dube
- Division of Parasitology, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India.
| |
Collapse
|
31
|
Shabani SH, Zakeri S, Mortazavi Y, Mehrizi AA. Immunological evaluation of two novel engineered Plasmodium vivax circumsporozoite proteins formulated with different human-compatible vaccine adjuvants in C57BL/6 mice. Med Microbiol Immunol 2019; 208:731-745. [DOI: 10.1007/s00430-019-00606-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/01/2019] [Indexed: 12/28/2022]
|
32
|
Si-doping increases the adjuvant activity of hydroxyapatite nanorods. Colloids Surf B Biointerfaces 2019; 174:300-307. [DOI: 10.1016/j.colsurfb.2018.11.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/16/2018] [Accepted: 11/13/2018] [Indexed: 11/23/2022]
|
33
|
Filipić B, Stojić-Vukanić Z. Adjuvants in vaccines registered for human use. ARHIV ZA FARMACIJU 2019. [DOI: 10.5937/arhfarm1906406f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
34
|
Thakkar SG, Warnken ZN, Alzhrani RF, Valdes SA, Aldayel AM, Xu H, Williams RO, Cui Z. Intranasal immunization with aluminum salt-adjuvanted dry powder vaccine. J Control Release 2018; 292:111-118. [PMID: 30339906 PMCID: PMC6328263 DOI: 10.1016/j.jconrel.2018.10.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/09/2018] [Accepted: 10/15/2018] [Indexed: 02/08/2023]
Abstract
Intranasal vaccination using dry powder vaccine formulation represents an attractive, non-invasive vaccination modality with better storage stability and added protection at the mucosal surfaces. Herein we report that it is feasible to induce specific mucosal and systemic antibody responses by intranasal immunization with a dry powder vaccine adjuvanted with an insoluble aluminum salt. The dry powder vaccine was prepared by thin-film freeze-drying of a model antigen, ovalbumin, adsorbed on aluminum (oxy)hydroxide as an adjuvant. Special emphasis was placed on the characterization of the dry powder vaccine formulation that can be realistically used in humans by a nasal dry powder delivery device. The vaccine powder was found to have "passable" to "good" flow properties, and the vaccine was uniformly distributed in the dry powder. An in vitro nasal deposition study using nasal casts of adult humans showed that around 90% of the powder was deposited in the nasal cavity. Intranasal immunization of rats with the dry powder vaccine elicited a specific serum antibody response as well as specific IgA responses in the nose and lung secretions of the rats. This study demonstrates the generation of systemic and mucosal immune responses by intranasal immunization using a dry powder vaccine adjuvanted with an aluminum salt.
Collapse
Affiliation(s)
- Sachin G Thakkar
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Zachary N Warnken
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Riyad F Alzhrani
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Solange A Valdes
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Abdulaziz M Aldayel
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States; Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), NGHA, Riyadh, 11426, Saudi Arabia
| | - Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Robert O Williams
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States.
| |
Collapse
|
35
|
Chen W, Zuo H, Rolfe B, Schembri MA, Cobbold RN, Zhang B, Mahony TJ, Xu ZP. Clay nanoparticles co-deliver three antigens to promote potent immune responses against pathogenic Escherichia coli. J Control Release 2018; 292:196-209. [DOI: 10.1016/j.jconrel.2018.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/02/2018] [Accepted: 11/04/2018] [Indexed: 01/02/2023]
|
36
|
Batista-Duharte A, Martínez DT, Carlos IZ. Efficacy and safety of immunological adjuvants. Where is the cut-off? Biomed Pharmacother 2018; 105:616-624. [DOI: 10.1016/j.biopha.2018.06.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/05/2018] [Accepted: 06/05/2018] [Indexed: 12/21/2022] Open
|
37
|
Kuznetsova TA, Persiyanova EV, Ermakova SP, Khotimchenko MY, Besednova NN. The Sulfated Polysaccharides of Brown Algae and Products of Their Enzymatic Transformation as Potential Vaccine Adjuvants. Nat Prod Commun 2018. [DOI: 10.1177/1934578x1801300837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The review is devoted to critical analysis of literature data, deal with effects and mechanisms of action of sulfated polysaccharides (PSs) – fucoidans from brown algae and products of their enzymatic transformation as potential adjuvants for enhancement of anti-infective and antitumor immune response. Numerous experimental data indicate that sulfated PSs demonstrate properties of vaccine adjuvants. Application perspectiveness of fucoidans as vaccine adjuvants is defined by their high biocompatibility, low-toxicity, safety and good tolerance by macroorganism, and also mechanisms of their immunomodulatory action. In particular, fucoidans are agonists of receptors of innate immunity and strong inducers of cellular and humoral immune response. At presenting the data of structural - functional interrelations, attention focused to the defining role of degree of sulfation, uronic acids and polyphenols contents, and also molecular mass in actions of fucoidans to innate and adaptive immunity cells. Insufficiency of literary data on studying of correlation of structure – physicochemical characteristics with adjuvanticities of the sulfated PSs, and also the problem of standardization of their active fractions are noted. Special attention is paid to the analysis of immunomodulatory and adjuvant activity of fucoidan oligosaccharides. Presented here results of experimental trial indicate that, despite the difficulties due to preparation of highly purified structurally characterized fractions and complex structure of fucoidans, these substances can be used as safe and effective adjuvants in vaccines against various pathogens including viruses, and also in antitumor vaccines.
Collapse
Affiliation(s)
- Tatyana A. Kuznetsova
- Federal State Budgetary Scientific Institution «Research Somov Institute of Epidemiology and Microbiology», Sel'skaya street, 1, 690087, Vladivostok, Russian Federation
- Far Eastern Federal University, School of Biomedicine, bldg. M25 FEFU Campus, Ajax Bay, Russky Isl., 690922 Vladivostok, Russian Federation
| | - Elena V. Persiyanova
- Federal State Budgetary Scientific Institution «Research Somov Institute of Epidemiology and Microbiology», Sel'skaya street, 1, 690087, Vladivostok, Russian Federation
| | - Svetlana P. Ermakova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Pr. 100-letya Vladivostoka 159, 690022, Vladivostok, Russian Federation
| | - Maxim Yu. Khotimchenko
- Far Eastern Federal University, School of Biomedicine, bldg. M25 FEFU Campus, Ajax Bay, Russky Isl., 690922 Vladivostok, Russian Federation
| | - Natalya N. Besednova
- Federal State Budgetary Scientific Institution «Research Somov Institute of Epidemiology and Microbiology», Sel'skaya street, 1, 690087, Vladivostok, Russian Federation
| |
Collapse
|
38
|
Jin Y, Li P, Wang F. β-glucans as potential immunoadjuvants: A review on the adjuvanticity, structure-activity relationship and receptor recognition properties. Vaccine 2018; 36:5235-5244. [PMID: 30049632 DOI: 10.1016/j.vaccine.2018.07.038] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/03/2018] [Accepted: 07/15/2018] [Indexed: 12/18/2022]
Abstract
β-glucans, a group of polysaccharides exist in many organism species such as mushrooms, yeasts, oats, barley, seaweed, but not mammalians, have a variety of biological activities and applications in drugs and other healthcare products. In recent years, β-glucans have been studied as adjuvants in anti-infection vaccines as well as immunomodulators in anti-cancer immunotherapy. β-glucans can regulate immune responses when administered alone and can connect innate and adaptive immunity to improve immunogenicity of vaccines. When β-glucans act as immunostimulants or adjuvants, a set of receptors have been revealed to recognize β-glucans, including dectin-1, complement receptor 3 (CR3), CD5, lactosylceramide, and so on. Therefore, this review is mainly focused on the application of β-glucans as immune adjuvants, the receptors of β-glucans, as well as their structure and activity relationship which will benefit future research of β-glucans.
Collapse
Affiliation(s)
- Yiming Jin
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhuaxi Road, Jinan 250012, China
| | - Pingli Li
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Road, Jinan 250012, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, No. 44 Wenhuaxi Road, Jinan 250012, China.
| |
Collapse
|
39
|
Li X, Wang X, Ito A. Tailoring inorganic nanoadjuvants towards next-generation vaccines. Chem Soc Rev 2018; 47:4954-4980. [PMID: 29911725 DOI: 10.1039/c8cs00028j] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vaccines, one of the most effective and powerful public health measures, have saved countless lives over the past century and still have a tremendous global impact. As an indispensable component of modern vaccines, adjuvants play a critical role in strengthening and/or shaping a specific immune response against infectious diseases as well as malignancies. The application of nanotechnology provides the possibility of precisely tailoring the building blocks of nanoadjuvants towards modern vaccines with the desired immune response. The last decade has witnessed great academic progress in inorganic nanomaterials for vaccine adjuvants in terms of nanometer-scale synthesis, structure control, and functionalization design. Inorganic adjuvants generally facilitate the delivery of antigens, allowing them to be released in a sustained manner, enhance immunogenicity, deliver antigens efficiently to specific targets, and induce a specific immune response. In particular, the recent discovery of the intrinsic immunomodulatory function of inorganic nanomaterials further allows us to shape the immune response towards the desired type and increase the efficacy of vaccines. In this article, we comprehensively review state-of-the-art research on the use of inorganic nanomaterials as vaccine adjuvants. Attention is focused on the physicochemical properties of versatile inorganic nanoadjuvants, such as composition, size, morphology, shape, hydrophobicity, and surface charge, to effectively stimulate cellular immunity, considering that the clinically used alum adjuvants can only induce strong humoral immunity. In addition, the efforts made to date to expand the application of inorganic nanoadjuvants in cancer vaccines are summarized. Finally, we discuss the future prospects and our outlook on tailoring inorganic nanoadjuvants towards next-generation vaccines.
Collapse
Affiliation(s)
- Xia Li
- Health Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.
| | | | | |
Collapse
|
40
|
Oliveira TL, Schuch RA, Inda GR, Roloff BC, Neto ACPS, Amaral M, Dellagostin OA, Hartwig DD. LemA and Erp Y-like recombinant proteins from Leptospira interrogans protect hamsters from challenge using AddaVax™ as adjuvant. Vaccine 2018; 36:2574-2580. [PMID: 29625765 DOI: 10.1016/j.vaccine.2018.03.078] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/24/2018] [Accepted: 03/28/2018] [Indexed: 11/18/2022]
Abstract
BACKGROUND Recombinant subunit vaccines have been extensively evaluated as promising alternatives against leptospirosis. Here, we evaluated two proteins in formulations containing the adjuvant AddaVax™ as vaccine candidates for prevention and control of leptospirosis. METHODS Recombinant proteins rErp Y-like and rLemA were characterized by ELISA to assess their ability to bind extracellular matrix (ECM) components and fibrinogen. Groups of eight hamsters were immunized intramuscularly with rErp Y-like or rLemA mixed with a squalene-based adjuvant (AddaVax), and then vaccine efficacy was determined in terms of protection against a lethal challenge. The humoral immune response was determined by ELISA, and the evidence of sub-lethal infection was evaluated by histopathology and kidney culture. RESULTS rLemA protein binds laminin, fibrinogen, and collagen type IV, while rErp Y-like interacts with fibrinogen. Significant protection was achieved for rLemA and rErp Y-like vaccines, which showed 87.5% and 62.5% survivals, respectively. On day 28, the humoral immune response was significantly greater in the vaccine groups as compared to that in the control group, and the response was predominantly based on IgG2/3. The surviving animals showed negative results in culture isolation but presented with tissue lesions in the lungs and kidneys. CONCLUSION Cumulatively, our findings suggest that LemA and Erp Y-like proteins act as adhesins and are able to protect against mortality, but not against tissue lesions. Moreover, AddaVax is a novel adjuvant with potential for improving the immunogenicity of leptospiral vaccines.
Collapse
Affiliation(s)
- Thaís Larré Oliveira
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Rodrigo Andrade Schuch
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Guilherme Roig Inda
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Bárbara Couto Roloff
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Amilton Clair Pinto Seixas Neto
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Marta Amaral
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Odir Antonio Dellagostin
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Daiane Drawanz Hartwig
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil; Departamento de Microbiologia e Parasitologia, Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
41
|
Gobec M, Tomašič T, Štimac A, Frkanec R, Trontelj J, Anderluh M, Mlinarič-Raščan I, Jakopin Ž. Discovery of Nanomolar Desmuramylpeptide Agonists of the Innate Immune Receptor Nucleotide-Binding Oligomerization Domain-Containing Protein 2 (NOD2) Possessing Immunostimulatory Properties. J Med Chem 2018. [PMID: 29543461 DOI: 10.1021/acs.jmedchem.7b01052] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Muramyl dipeptide (MDP), a fragment of bacterial peptidoglycan, has long been known as the smallest fragment possessing adjuvant activity, on the basis of its agonistic action on the nucleotide-binding oligomerization domain-containing protein 2 (NOD2). There is a pressing need for novel adjuvants, and NOD2 agonists provide an untapped source of potential candidates. Here, we report the design, synthesis, and characterization of a series of novel acyl tripeptides. A pivotal structural element for molecular recognition by NOD2 has been identified, culminating in the discovery of compound 9, the most potent desmuramylpeptide NOD2 agonist to date. Compound 9 augmented pro-inflammatory cytokine release from human peripheral blood mononuclear cells in synergy with lipopolysaccharide. Furthermore, it was able to induce ovalbumin-specific IgG titers in a mouse model of adjuvancy. These findings provide deeper insights into the structural requirements of desmuramylpeptides for NOD2-activation and highlight the potential use of NOD2 agonists as adjuvants for vaccines.
Collapse
Affiliation(s)
- Martina Gobec
- Faculty of Pharmacy , University of Ljubljana , Aškerčeva 7 , SI-1000 Ljubljana , Slovenia
| | - Tihomir Tomašič
- Faculty of Pharmacy , University of Ljubljana , Aškerčeva 7 , SI-1000 Ljubljana , Slovenia
| | - Adela Štimac
- Centre for Research and Knowledge Transfer in Biotechnology , University of Zagreb , Rockefellerova 10 , 10000 Zagreb , Croatia
| | - Ruža Frkanec
- Centre for Research and Knowledge Transfer in Biotechnology , University of Zagreb , Rockefellerova 10 , 10000 Zagreb , Croatia
| | - Jurij Trontelj
- Faculty of Pharmacy , University of Ljubljana , Aškerčeva 7 , SI-1000 Ljubljana , Slovenia
| | - Marko Anderluh
- Faculty of Pharmacy , University of Ljubljana , Aškerčeva 7 , SI-1000 Ljubljana , Slovenia
| | - Irena Mlinarič-Raščan
- Faculty of Pharmacy , University of Ljubljana , Aškerčeva 7 , SI-1000 Ljubljana , Slovenia
| | - Žiga Jakopin
- Faculty of Pharmacy , University of Ljubljana , Aškerčeva 7 , SI-1000 Ljubljana , Slovenia
| |
Collapse
|
42
|
Fernández Cotrina J, Iniesta V, Monroy I, Baz V, Hugnet C, Marañon F, Fabra M, Gómez-Nieto LC, Alonso C. A large-scale field randomized trial demonstrates safety and efficacy of the vaccine LetiFend® against canine leishmaniosis. Vaccine 2018. [PMID: 29525281 DOI: 10.1016/j.vaccine.2018.02.111] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Canine leishmaniosis is a zoonotic disease caused by Leishmania infantum. Extensive research is currently ongoing to develop safe and effective vaccines to protect from disease development. The European Commission has granted a marketing authorization for LetiFend®, a new vaccine containing recombinant Protein Q. The efficacy of LetiFend® vaccination in a large-scale dog population of both sexes, different breeds and ages in endemic areas is reported in this multicenter, randomized, double-blind, placebo-controlled field trial. Dogs (n = 549) living in France and Spain were randomly selected to receive a single subcutaneous dose of LetiFend® or placebo per year, and were naturally exposed to two L. infantum transmission seasons. Clinical examinations, blood and lymphoid organ sampling to evaluate serological, parasitological and disease status of the dogs were performed at different time points during the study. LetiFend® was very well tolerated and clearly reduced the incidence of clinical signs related to leishmaniosis. The number of confirmed cases of leishmaniosis was statistically significantly lower in the vaccine group. The number of dogs with parasites was close to be significantly reduced in the vaccine group (p = 0.0564). Re-vaccination of seropositive dogs demonstrated to be safe and not to worsen the course of the disease. The likelihood that a dog vaccinated with LetiFend® develops a confirmed case or clinical signs of leishmaniosis in areas with high pressure is, respectively, 5 and 9.8 time less than that for an unvaccinated dog. Thus, the overall efficacy of the LetiFend® vaccine in the prevention of confirmed cases of leishmaniosis in endemic areas with high disease pressure was shown to be 72%. In conclusion, this field trial demonstrates that LetiFend® is a novel, safe and effective vaccine for the active immunization of non-infected dogs from 6 months of age in reducing the risk of developing clinical leishmaniosis after natural infection with Leishmania infantum.
Collapse
Affiliation(s)
- Javier Fernández Cotrina
- Unidad de Parasitología, Facultad de Veterinaria, Universidad de Extremadura, Avda. de la Universidad, s/n, 10003 Cáceres, Spain.
| | - Virginia Iniesta
- Unidad de Parasitología, Facultad de Veterinaria, Universidad de Extremadura, Avda. de la Universidad, s/n, 10003 Cáceres, Spain.
| | - Isabel Monroy
- Unidad de Parasitología, Facultad de Veterinaria, Universidad de Extremadura, Avda. de la Universidad, s/n, 10003 Cáceres, Spain.
| | - Victoria Baz
- Unidad de Parasitología, Facultad de Veterinaria, Universidad de Extremadura, Avda. de la Universidad, s/n, 10003 Cáceres, Spain.
| | - Christophe Hugnet
- Clinique Vétérinaire des Lavandes, Quartier Boulagne 26160, La Begude de Mazenc, France.
| | - Francisco Marañon
- Animal Health Unit, Laboratorios LETI S.L.U., Gran Vía de les Corts Catalanes, 184, 08038 Barcelona, Spain.
| | - Mercedes Fabra
- Animal Health Unit, Laboratorios LETI S.L.U., Gran Vía de les Corts Catalanes, 184, 08038 Barcelona, Spain.
| | - Luis Carlos Gómez-Nieto
- Unidad de Parasitología, Facultad de Veterinaria, Universidad de Extremadura, Avda. de la Universidad, s/n, 10003 Cáceres, Spain.
| | - Carlos Alonso
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain.
| |
Collapse
|
43
|
Bernau M, Liesner BG, Schwanitz S, Kraus AS, Falkenau A, Leipig-Rudolph M, Hermanns W, Scholz AM. Vaccine safety testing using magnetic resonance imaging in suckling pigs. Vaccine 2018; 36:1789-1795. [DOI: 10.1016/j.vaccine.2018.02.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 12/14/2017] [Accepted: 02/02/2018] [Indexed: 11/29/2022]
|
44
|
Intraperitoneal administration of aluminium-based adjuvants produces severe transient systemic adverse events in mice. Eur J Pharm Sci 2018; 115:362-368. [DOI: 10.1016/j.ejps.2018.01.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 11/19/2022]
|
45
|
Alshanqiti FM, Al-Masaudi SB, Al-Hejin AM, El-Baky NA, Redwan EM. Development of nanoparticle adjuvants to potentiate the immune response against diphtheria toxoid. Hum Antibodies 2018; 26:75-85. [PMID: 29171990 DOI: 10.3233/hab-170324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Over the years, diphtheria was known as contagious fatal infection caused by Corynebacterium diphtheria that affects upper respiratory system. The spread of diphtheria epidemic disease is best prevented by vaccination with diphtheria toxoid vaccine. Aluminum adjuvants were reported to stimulate the immune responses to killed and subunit vaccines. OBJECTIVE Our study aimed to minimize adjuvant particles size, to gain insight of resulting immunity titer and impact on immune response antibody subtypes. METHODS Aluminum salts and calcium phosphate adjuvants were prepared, followed by micro/nanoparticle adjuvants preparation. After formulation of diphtheria vaccine from diphtheria toxoid and developed adjuvants, we evaluated efficacy of these prepared vaccines based on their impact on immune response via measuring antibodies titer, antibodies isotyping and cytokines profile in immunized mice. RESULTS A noteworthy increase in immunological parameters was observed; antibodies titer was higher in serum of mice injected with nanoparticle adjuvants-containing vaccine than mice injected with standard adjuvant-containing vaccine and commercial vaccine. Aluminum compounds adjuvants (nanoparticles and microparticles formulation) and microparticles calcium phosphate adjuvant induce TH2 response, while nanoparticles calcium phosphate and microparticles aluminum compounds adjuvants stimulate TH1 response. CONCLUSIONS Different treatments to our adjuvant preparations (nanoparticles and microparticles formulation) had a considerable impact on vaccine immunogenicity.
Collapse
Affiliation(s)
- Fatimah M Alshanqiti
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Saad Berki Al-Masaudi
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed M Al-Hejin
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nawal Abd El-Baky
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab 21934, Alexandria, Egypt
| | - Elrashdy M Redwan
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.,Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab 21934, Alexandria, Egypt
| |
Collapse
|
46
|
Efficient induction of comprehensive immune responses to control pathogenic E. coli by clay nano-adjuvant with the moderate size and surface charge. Sci Rep 2017; 7:13367. [PMID: 29042573 PMCID: PMC5645426 DOI: 10.1038/s41598-017-13570-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/26/2017] [Indexed: 02/06/2023] Open
Abstract
In recent decades, diseases caused by pathogenic Escherichia coli (E. coli), enterohaemorrhagic E. coli (EHEC) O26 have been increasingly reported worldwide, which are as severe as those caused by EHEC strain O157:H7 and require effective intervention strategies. Herein, we report the application of clay nanoparticles, i.e. hectorites as effective nano-adjuvants for vaccination against EHEC O26 colonization. We show that medium size HEC (hectorite, around 73~77 nm diameter) is able to induce efficient humoral and cellular immune responses against EHEC antigen - intimin β (IB), which are significantly higher than those triggered by commercially used adjuvants - QuilA and Alum. We also demonstrate that mice immunized with IB adjuvanted with HEC nanoparticles elicit sufficient secretion of mucosal IgA, capable of providing effective protection against EHEC O26 binding to ruminant and human cells. In addition, we demonstrate for the first time that hectorites are able to initiate maturation of RAW 264.7 macrophages, inducing expression of co-stimulatory cytokines at a low nanoparticle concentration (10 μg/mL). Together these data strongly suggest that hectorite with optimized size is a highly efficient vaccine nano-adjuvant.
Collapse
|
47
|
Vaccine adjuvants: smart components to boost the immune system. Arch Pharm Res 2017; 40:1238-1248. [PMID: 29027637 DOI: 10.1007/s12272-017-0969-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023]
Abstract
Vaccination is an effective approach to prevent the consequences of infectious diseases. Vaccines strengthen immunity and make individuals resistant to infections with pathogens. Although conventional vaccines are highly immunogenic, they are associated with some safety issues. Subunit vaccines are safe, but they require adjuvants to stimulate the immune system because of their weaker immunogenicity. Adjuvants are entities incorporated into vaccines to increase the immunogenic responses of antigens. They play a crucial role in increasing the potency and efficacy of vaccines. Different adjuvants have different modes of action; therefore, a better understanding of their immunology could provide guidance for the development of novel adjuvants. Numerous studies have been conducted using different types of adjuvants to characterize their potency and safety; however, in practice, only few are used in human or animal vaccines. This review aims to introduce the different modes of action of adjuvants and give insight into the types of adjuvants that possess the greatest potential for adjuvanticity.
Collapse
|
48
|
Bernau M, Kremer-Rücker PV, Kreuzer LS, Schwanitz S, Cussler K, Hoffmann A, Scholz AM. Magnetic resonance imaging to detect local tissue reactions after vaccination in sheep in vivo. Vet Rec Open 2017; 4:e000200. [PMID: 29018531 PMCID: PMC5623326 DOI: 10.1136/vetreco-2016-000200] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 06/19/2017] [Accepted: 06/26/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Vaccination is one of the most effective methods to keep up the health status in humans and in livestock. Therefore, farm animals are vaccinated several times during their lifetime. Although vaccines are being checked regarding their local reactogenicity, side effects occur frequently-especially in the case of the application of adjuvanted products. Many reports exist about local reactions in sheep. The present study aimed at testing MRI as a method to document injection site reactions three-dimensionally. DESIGN Two groups of Merino lambs (n=16 each) were vaccinated subcutaneously into the left neck side. Two different, licensed inactivated vaccines were used. Both groups of lambs were anaesthetised and scanned using MRI at days 1, 3, 8, 15, 22 and 29 after vaccination. SETTING The study was performed on a commercial-like farm. PARTICIPANTS Thirty-two Merino lambs entered the experiment, 16 male and 16 female ones (one animal died at day 22 after vaccination). At first examination day they were approximately three months old. PRIMARY AND SECONDARY OUTCOME MEASURES Volume differences were measured between vaccination and control neck side to evaluate the time pattern of local tissue reactions. RESULTS Local tissue reactions were visible on the skin surface and also appeared in deeper tissue layers on MRI. These deeper reactions would not have been found without MRI or, alternatively, without sacrificing the animals. Some of these extensive local reactions lasted for more than 29 days. CONCLUSIONS The in vivo MRI results proved suitable to record local tissue reactions in terms of three-dimensional extent over a longer period of time in large farm animals without the need to sacrifice test animals. A three-dimensional MRI examination of the injection site during regulatory licensing studies offers an objective evaluation that could be used in a benefit-risk assessment of veterinary vaccines. TRIAL REGISTRATION NUMBER District Government of Upper Bavaria:55.2-1-54-2532-2-13.
Collapse
Affiliation(s)
- Maren Bernau
- Livestock Center Oberschleissheim, Veterinary Faculty of the Ludwig-Maximilians-University Munich, Oberschleissheim, Germany
| | | | - Lena Sophie Kreuzer
- Livestock Center Oberschleissheim, Veterinary Faculty of the Ludwig-Maximilians-University Munich, Oberschleissheim, Germany
| | - Sebastian Schwanitz
- Livestock Center Oberschleissheim, Veterinary Faculty of the Ludwig-Maximilians-University Munich, Oberschleissheim, Germany
| | - Klaus Cussler
- Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Andreas Hoffmann
- Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Armin Manfred Scholz
- Livestock Center Oberschleissheim, Veterinary Faculty of the Ludwig-Maximilians-University Munich, Oberschleissheim, Germany
| |
Collapse
|
49
|
Portuondo DL, Batista-Duharte A, Ferreira LS, de Andrade CR, Quinello C, Téllez-Martínez D, de Aguiar Loesch ML, Carlos IZ. Comparative efficacy and toxicity of two vaccine candidates against Sporothrix schenckii using either Montanide™ Pet Gel A or aluminum hydroxide adjuvants in mice. Vaccine 2017; 35:4430-4436. [PMID: 28687406 DOI: 10.1016/j.vaccine.2017.05.046] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/19/2017] [Accepted: 05/15/2017] [Indexed: 12/14/2022]
Abstract
Sporotrichosis is an important zoonosis in Brazil and the most frequent subcutaneous mycosis in Latin America, caused by different Sporothrix species. Currently, there is no effective vaccine available to prevent this disease. In this study, the efficacy and toxicity of the adjuvant Montanide™ Pet Gel A (PGA) formulated with S. schenckii cell wall proteins (ssCWP) was evaluated and compared with that of aluminum hydroxide (AH). Balb/c mice received two subcutaneous doses (1st and 14th days) of either the unadjuvanted or adjuvanted vaccine candidates. On the 21st day, anti-ssCWP antibody levels (ELISA), the phagocytic index, as well as the ex vivo release of IFN-γ, IL-4, and IL-17 by splenocytes and IL-12 by peritoneal macrophages were assessed. Cytotoxicity of the vaccine formulations was evaluated in vitro and by histopathological analysis of the inoculation site. Both adjuvanted vaccine formulations increased anti-ssCWP IgG, IgG1, IgG2a, and IgG3 levels, although IgG2a levels were higher in response to PGA+CWP100, probably contributing to the increase in S. schenckii yeast phagocytosis by macrophages in the opsonophagocytosis assay when using serum from PGA+CWP100-immunized mice. Immunization with AH+CWP100 led to a mixed Th1/Th2/Th17 ex vivo cytokine release profile, while PGA+CWP100 stimulated a preferential Th1/Th2 profile. Moreover, PGA+CWP100 was less cytotoxic in vitro, caused less local toxicity and led to a similar reduction in fungal load in the liver and spleen of S. schenckii- or S. brasiliensis-challenged mice as compared with AH+CWP100. These results suggest that PGA may be an effective and safe adjuvant for a future sporotrichosis vaccine.
Collapse
Affiliation(s)
- Deivys Leandro Portuondo
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, SP, Brazil.
| | - Alexander Batista-Duharte
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, SP, Brazil.
| | - Lucas Souza Ferreira
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, SP, Brazil.
| | - Cleverton Roberto de Andrade
- São Paulo State University (UNESP), School of Dentistry, Department of Physiology & Pathology, Araraquara, SP, Brazil.
| | - Camila Quinello
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, SP, Brazil.
| | - Damiana Téllez-Martínez
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, SP, Brazil.
| | - Maria Luiza de Aguiar Loesch
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, SP, Brazil.
| | - Iracilda Zeppone Carlos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Department of Clinical Analysis, Araraquara, SP, Brazil.
| |
Collapse
|
50
|
Alshanqiti FM, Al-Masaudi SB, Al-Hejin AM, Redwan EM. Adjuvants for Clostridium tetani and Clostridium diphtheriae vaccines updating. Hum Antibodies 2017; 25:23-29. [PMID: 27858706 DOI: 10.3233/hab-160302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It's known that diphtheria and tetanus are a contagious lethal diseases over the years, they caused by pathogenic microbes corynebacterium diphtheria and Clostridium tetani, respectively. The diseases result from the production of bacterial toxin. Vaccination with bacterial toxoid vaccines adsorbed on particulates adjuvants still are the best way to prevent this epidemic diseases from spread. The particulate vaccines have been shown to be more efficient than soluble one for the induction of the immune responses. Nanoparticles can be engineered to enhance the immune responses. As well known the immune response to inactivate killed and subunit vaccine enhances by alum adjuvants. The adjuvants examined and tested after reducing its size to particle size, thus mimic size of viruses which is considered smallest units can derive the immune system. The major issue is minimizing the adjuvant particles, to gain insight of resulting immunity types and impact on immune response. The adjuvant effect of micro/nanoparticles appears to largely be a consequence of their uptake into antigen presenting cells.
Collapse
Affiliation(s)
- Fatimah M Alshanqiti
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saad B Al-Masaudi
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed M Al-Hejin
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Elrashdy M Redwan
- Biological Sciences Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| |
Collapse
|