1
|
Jung J, Kim TH, Park JY, Kwon S, Sung JS, Kang MJ, Jose J, Lee M, Shin HJ, Pyun JC. SARS-CoV-2 vaccine based on ferritin complexes with screened immunogenic sequences from the Fv-antibody library. J Mater Chem B 2025; 13:1383-1394. [PMID: 39668674 DOI: 10.1039/d4tb01595a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
In this study, the vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was developed using ferritin complexes with the immunogenic sequences screened against the SARS-CoV-2 spike protein (SP) from the Fv-antibody library. The Fv-antibody library was prepared on the outer membrane of E. coli by the expression of the VH region of immunoglobulin G (IgG) with a randomized complementarity-determining region 3 (CDR3). Four Fv-antibodies to the receptor-binding domain (RBD) were screened from the Fv-antibody library, which had a comparable binding constant (KD) between SARS-CoV-2 SP and the angiotensin-converting enzyme 2 (ACE2) receptor. The binding sites of screened Fv-antibodies on the RBD were analyzed using a docking analysis, and these binding sites were used as immunogenic sequences for the vaccine. The four immunogenic sequences were modified and co-expressed as a part of ferritin which was assembled into a ferritin complex. After the vaccination of ferritin complexes to mice, the anti-sera were analyzed to have a high enough titer. Additionally, the immune responses were found to be activated by vaccination, such as the expression of IgG subclasses and the increased level of cytokines. The neutralizing activity of the anti-sera was estimated using a cell-based infection assay based on pseudo-virus expressing the SP of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Tae-Hun Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Jae-Yeon Park
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Soonil Kwon
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul 02456, Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, University of Munster, Münster (48149), Germany
| | - Misu Lee
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Hyun-Jin Shin
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| |
Collapse
|
2
|
Bonfiglio R, Giacobbi E, Palumbo V, Casciardi S, Sisto R, Servadei F, Scioli MP, Schiaroli S, Cornella E, Cervelli G, Sica G, Candi E, Melino G, Mauriello A, Scimeca M. Aluminum Concentration Is Associated with Tumor Mutational Burden and the Expression of Immune Response Biomarkers in Colorectal Cancers. Int J Mol Sci 2024; 25:13388. [PMID: 39769153 PMCID: PMC11676456 DOI: 10.3390/ijms252413388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Environmental pollution poses a significant risk to public health, as demonstrated by the bioaccumulation of aluminum (Al) in colorectal cancer (CRC). This study aimed to investigate the potential mutagenic effect of Al bioaccumulation in CRC samples, linking it to the alteration of key mediators of cancer progression, including immune response biomarkers. Aluminum levels in 20 CRC biopsy samples were analyzed using inductively coupled plasma mass spectrometry (ICP-MS). The results indicated that Al bioaccumulation occurred in 100% of the cases. A correlation between Al levels and tumor mutation burden was observed. Furthermore, RNA sequencing revealed a significant association between Al concentration and the expression of the immune checkpoint molecule CTLA-4. Although correlations with PD-1 and PD-L1 were not statistically significant, a trend was observed. Additionally, a correlation between Al levels and both the presence of myeloid cells and IFNγ expression was detected, linking Al exposure to inflammatory responses within the tumor microenvironment. These findings suggested that Al can play a role in CRC progression by promoting both genetic mutations and immune evasion. Given the ubiquitous presence of Al in industrial and consumer products, dietary sources, and environmental pollutants, these results underscored the need for stricter regulatory measures to control Al exposure.
Collapse
Affiliation(s)
- Rita Bonfiglio
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Erica Giacobbi
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Valeria Palumbo
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Stefano Casciardi
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Monte Porzio Catone, 00078 Rome, Italy; (S.C.); (R.S.)
| | - Renata Sisto
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Monte Porzio Catone, 00078 Rome, Italy; (S.C.); (R.S.)
| | - Francesca Servadei
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Maria Paola Scioli
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Stefania Schiaroli
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Elena Cornella
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Giulio Cervelli
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Giuseppe Sica
- Department of Surgery, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Gerry Melino
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| | - Manuel Scimeca
- Department of Experimental Medicine, Tor Vergata Oncoscience Research (TOR), University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.B.); (E.G.); (V.P.); (F.S.); (M.P.S.); (S.S.); (E.C.)
| |
Collapse
|
3
|
Gogoi H, Mani R, Bhatnagar R. Re-inventing traditional aluminum-based adjuvants: Insight into a century of advancements. Int Rev Immunol 2024; 44:58-81. [PMID: 39310923 DOI: 10.1080/08830185.2024.2404095] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 06/01/2023] [Accepted: 08/31/2024] [Indexed: 02/22/2025]
Abstract
Aluminum salt-based adjuvants like alum, alhydrogel and Adju-Phos are by far the most favored clinically approved vaccine adjuvants. They have demonstrated excellent safety profile and currently used in vaccines against diphtheria, tetanus, pertussis, hepatitis B, anthrax etc. These vaccinations cause minimal side effects like local inflammation at the injection site. Aluminum salt-based adjuvants primarily stimulate CD4+ T cells and B cell mediated Th2 immune response leading to generate a robust antibody response. In this review article, we have compiled the role of physio-chemical role of the two commonly used aluminum salt-based adjuvants alhydrogel and Adju-Phos, and the effect of surface properties, buffer composition, and adjuvant dosage on the immune response. After being studied for almost a century, researchers have come up with various mechanism by which these aluminum adjuvants activate the immune system. Firstly, we have covered the initial works of Glenny and his "repository effect" which paved the work for his successors to explore the involvement of cytokines, chemokines, recruitment of innate immune cells, enhanced antigen uptake by antigen presenting cells, and formation of NLRP3 inflammasome complex in mediating the immune response. It has been reported that aluminum adjuvants activate multiple immunological pathways which synergistically activates the immune system. We later discuss the recent developments in nanotechnology-based preparations of next generation aluminum based adjuvants which has enabled precise size control and morphology of the traditional aluminum adjuvants thereby manipulating the immune response as per our desire.
Collapse
Affiliation(s)
- Himanshu Gogoi
- Amity Institute of Microbial Technology, Amity University Rajasthan, Jaipur, India
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad - Gurgaon Expressway, Faridabad, Haryana, India
| | - Rajesh Mani
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Department of Microbiology, Immunology and Molecular Genetics, University Kentucky College of Medicine, Lexington, KY, USA
| | - Rakesh Bhatnagar
- Amity Institute of Microbial Technology, Amity University Rajasthan, Jaipur, India
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
4
|
Raison-Peyron N, El Gana M, Amelot F, Bessis D. Post-vaccine nodules with prurigo in a child successfully treated by dupilumab. Pediatr Dermatol 2024; 41:932-933. [PMID: 38697666 DOI: 10.1111/pde.15615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/24/2024] [Indexed: 05/05/2024]
Abstract
We report the case of a 7-year-old girl with severe generalized prurigo associated with diffuse persistent pruritic nodules due to sensitization to aluminum-adsorbed vaccines. Treatment with dupilumab resulted in an excellent therapeutic response.
Collapse
Affiliation(s)
- Nadia Raison-Peyron
- Department of Dermatology, University Hospital of Montpellier and University of Montpellier, Montpellier, France
| | - Marwa El Gana
- Department of Dermatology, University Hospital of Montpellier and University of Montpellier, Montpellier, France
| | | | - Didier Bessis
- Department of Dermatology, University Hospital of Montpellier and University of Montpellier, Montpellier, France
| |
Collapse
|
5
|
Shams N, Jaydari A, Najafi H, Hataminejad M, Khanizadeh S, Pouladi I. An Overview of the Types of Adjuvants Used in the Vaccination Industry And Their Mechanisms of Action. Viral Immunol 2024; 37:324-336. [PMID: 39172659 DOI: 10.1089/vim.2024.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
The widespread use of efficient vaccines against infectious diseases is regarded as one of the most significant advancements in public health and techniques for preventing and protecting against infectious diseases and cancer. Because the purpose of vaccination is to elicit an appropriate, powerful, and long-lasting immune response against the pathogen, compounds such as adjuvants must be used to enhance these responses. Adjuvants have been widely used since their discovery to boost immune responses, prevent diseases, and activate protective immunity. Today, several types of adjuvants with varying properties are available for specific applications. Adjuvants are supramolecular substances or complexes that strengthen and prolong the immune response to antigens. These compounds have long-term immunological effects and are low in toxicity. They also lower the amount of antigen or the number of immunogenic reactions needed to improve vaccine efficacy and are used in specific populations. This article provides an overview of the adjuvants commonly used in the vaccination industry, their respective mechanisms of action, and discusses how they function to stimulate the immune system. Understanding the mechanisms of action of adjuvants is crucial for the development of effective and safe vaccines.
Collapse
Affiliation(s)
- Nemat Shams
- Department of Microbiology and Food Hygiene, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Amin Jaydari
- Department of Microbiology and Food Hygiene, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Hamideh Najafi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Hataminejad
- Department of Parasitology and Mycology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Sayyad Khanizadeh
- Hepatitis Research Center, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Iman Pouladi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
6
|
Oladejo M, Tijani AO, Puri A, Chablani L. Adjuvants in cutaneous vaccination: A comprehensive analysis. J Control Release 2024; 369:475-492. [PMID: 38569943 DOI: 10.1016/j.jconrel.2024.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Skin is the body's largest organ and serves as a protective barrier from physical, thermal, and mechanical environmental challenges. Alongside, the skin hosts key immune system players, such as the professional antigen-presenting cells (APCs) like the Langerhans cells in the epidermis and circulating macrophages in the blood. Further, the literature supports that the APCs can be activated by antigen or vaccine delivery via multiple routes of administration through the skin. Once activated, the stimulated APCs drain to the associated lymph nodes and gain access to the lymphatic system. This further allows the APCs to engage with the adaptive immune system and activate cellular and humoral immune responses. Thus, vaccine delivery via skin offers advantages such as reliable antigen delivery, superior immunogenicity, and convenient delivery. Several preclinical and clinical studies have demonstrated the significance of vaccine delivery using various routes of administration via skin. However, such vaccines often employ adjuvant/(s), along with the antigen of interest. Adjuvants augment the immune response to a vaccine antigen and improve the therapeutic efficacy. Due to these reasons, adjuvants have been successfully used with infectious disease vaccines, cancer immunotherapy, and immune-mediated diseases. To capture these developments, this review will summarize preclinical and clinical study results of vaccine delivery via skin in the presence of adjuvants. A focused discussion regarding the FDA-approved adjuvants will address the experiences of using such adjuvant-containing vaccines. In addition, the challenges and regulatory concerns with these adjuvants will be discussed. Finally, the review will share the prospects of adjuvant-containing vaccines delivered via skin.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA.
| | - Lipika Chablani
- Wegmans School of Pharmacy, St. John Fisher University, 3690 East Ave, Rochester, NY 14618, USA.
| |
Collapse
|
7
|
Galloway DR, Li J, Nguyen NX, Falkenberg FW, Henning L, Krile R, Chou YL, Herron JN, Hale JS, Williamson ED. Co-formulation of the rF1V plague vaccine with depot-formulated cytokines enhances immunogenicity and efficacy to elicit protective responses against aerosol challenge in mice. Front Immunol 2024; 15:1277526. [PMID: 38605961 PMCID: PMC11007139 DOI: 10.3389/fimmu.2024.1277526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/05/2024] [Indexed: 04/13/2024] Open
Abstract
This study evaluated a depot-formulated cytokine-based adjuvant to improve the efficacy of the recombinant F1V (rF1V) plague vaccine and examined the protective response following aerosol challenge in a murine model. The results of this study showed that co-formulation of the Alhydrogel-adsorbed rF1V plague fusion vaccine with the depot-formulated cytokines recombinant human interleukin 2 (rhuIL-2) and/or recombinant murine granulocyte macrophage colony-stimulating factor (rmGM-CSF) significantly enhances immunogenicity and significant protection at lower antigen doses against a lethal aerosol challenge. These results provide additional support for the co-application of the depot-formulated IL-2 and/or GM-CSF cytokines to enhance vaccine efficacy.
Collapse
Affiliation(s)
- Darrell R. Galloway
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, United States
| | - Jiahui Li
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, United States
| | - Nguyen X. Nguyen
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | | | - Lisa Henning
- Battelle Biomedical Research Center, Columbus, OH, United States
| | - Robert Krile
- Battelle Biomedical Research Center, Columbus, OH, United States
| | - Ying-Liang Chou
- Battelle Biomedical Research Center, Columbus, OH, United States
| | - James N. Herron
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, United States
| | - J. Scott Hale
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | - E. Diane Williamson
- Chemical Biological Radiological Division, Defense Science and Technology Laboratory (DSTL), Porton Down, Salisbury, United Kingdom
| |
Collapse
|
8
|
Zhang Y, Yang F, Sun D, Xu L, Shi Y, Qin L, Zhao L, Wang L, Sun W, Wu H, Lu D, Zhang W, Luo P, Cheng P, Zou Q, Zeng H. rFSAV promotes Staphylococcus aureus-infected bone defect healing via IL-13- mediated M2 macrophage polarization. Clin Immunol 2023; 255:109747. [PMID: 37634854 DOI: 10.1016/j.clim.2023.109747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Staphylococcus aureus (S. aureus) contamination commonly occurs in orthopedic internal fixation operations, leading to a delayed healing of the defected bone tissue. However, antibiotic treatments are ineffective in dealing with S. aureus bone infections due to the rise in multiple antimicrobial resistances. Here, we reported the protective effects of a recombinant five-antigen S. aureus vaccine (rFSAV) in an S. aureus infected bone defect model. In this study, we found the number of M2 macrophages markedly increased in the defect site and played a critical role in the healing of defected bone mediated by rFSAV. Mechanistically, rFSAV mediated increased level of IL-13 in bone defect site predominant M2 macrophage polarization. In summary, our study reveals a key role of M2 macrophage polarization in the bone regeneration process in S. aureus infection induced bone defect, which provide a promising application of rFSAV for the treatment of bone infection for orthopedic applications.
Collapse
Affiliation(s)
- Yanhao Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Feng Yang
- Chengdu Olymvax Biopharmaceuticals Inc., Chengdu, Sichuan 611731, PR China
| | - Dong Sun
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Limin Xu
- Chengdu Olymvax Biopharmaceuticals Inc., Chengdu, Sichuan 611731, PR China
| | - Yaojia Shi
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Leilei Qin
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400038, China
| | - Liqun Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Wei Sun
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Hongri Wu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China.
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China.
| |
Collapse
|
9
|
Goudarzi T, Abkar M, Zamanzadeh Z, Fasihi-Ramandi M. Immunization of mice with chimeric protein-loaded aluminum hydroxide and selenium nanoparticles induces reduction of Brucella melitensis infection in mice. Clin Exp Vaccine Res 2023; 12:304-312. [PMID: 38025913 PMCID: PMC10655149 DOI: 10.7774/cevr.2023.12.4.304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Due to the many problems with commercially available vaccines, the production of effective vaccines against brucellosis is a necessity. The aim of this study was to evaluate the immune responses caused by the chimeric protein consisting of trigger factor, Bp26, and Omp31 (TBO) along with aluminum hydroxide (AH/TBO) and selenium (Se/TBO) nanoparticles (NPs) as adjuvants in mouse model. Materials and Methods Recombinant antigen expression was induced in Escherichia coli BL21 (DE3) bacteria using IPTG (isopropyl-d-1-thiogalactopyranoside). Purification and characterization of recombinant protein was conducted through NiFe3O4 NPs, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and Western blot. NP characteristics, including morphology and particle size, were measured in vitro. The recombinant TBO was loaded on to AH and Se NPs and were administered subcutaneously. After mice immunization, measurement of antibody titter and protection assay was performed. Results The average sizes of AH and Se NPs were about 60 nm and 150 nm, respectively. The enzyme-linked immunosorbent assay results showed that the serum of mice immunized by subcutaneous injection with both nanovaccines produced significant immunoglobulin G (IgG) responses against the chimeric antigen. The results of TBO-specific IgG isotype (IgG2a/IgG1) analysis showed that both AH and Se NPs induced a type to T-helper immune response. In addition, the results of the challenge with the pathogenic strain of Brucella melitensis 16M showed that vaccinated mice with AH/TBO NPs indicated a higher reduction of bacterial culture than immunized mice with Se/TBO NPs and TBO alone. Conclusion The results showed that AH NPs carrying chimeric antigen can be a promising vaccine candidate against brucellosis by producing protective immunity.
Collapse
Affiliation(s)
- Tahereh Goudarzi
- Department of Genetics, Faculty of Biological Sciences and Technology, Shahid Ashrafi Esfahani University, Isfahan, Iran
| | - Morteza Abkar
- Department of Genetics, Faculty of Biological Sciences and Technology, Shahid Ashrafi Esfahani University, Isfahan, Iran
| | - Zahra Zamanzadeh
- Department of Genetics, Faculty of Biological Sciences and Technology, Shahid Ashrafi Esfahani University, Isfahan, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Zhang Y, Jiang M, Du G, Zhong X, He C, Qin M, Hou Y, Liu R, Sun X. An antigen self-assembled and dendritic cell-targeted nanovaccine for enhanced immunity against cancer. Acta Pharm Sin B 2023; 13:3518-3534. [PMID: 37655327 PMCID: PMC10465870 DOI: 10.1016/j.apsb.2022.03.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022] Open
Abstract
The rise of nanotechnology has opened new horizons for cancer immunotherapy. However, most nanovaccines fabricated with nanomaterials suffer from carrier-related concerns, including low drug loading capacity, unpredictable metabolism, and potential systemic toxicity, which bring obstacles for their clinical translation. Herein, we developed an antigen self-assembled nanovaccine, which was resulted from a simple acryloyl modification of the antigen to induce self-assembly. Furthermore, a dendritic cell targeting head mannose monomer and a mevalonate pathway inhibitor zoledronic acid (Zol) were integrated or absorbed onto the nanoparticles (denoted as MEAO-Z) to intensify the immune response. The synthesized nanovaccine with a diameter of around 70 nm showed successful lymph node transportation, high dendritic cell internalization, promoted costimulatory molecule expression, and preferable antigen cross-presentation. In virtue of the above superiorities, MEAO-Z induced remarkably higher titers of serum antibody, stronger cytotoxic T lymphocyte immune responses and IFN-γ secretion than free antigen and adjuvants. In vivo, MEAO-Z significantly suppressed EG7-OVA tumor growth and prolonged the survival time of tumor-bearing mice. These results indicated the translation promise of our self-assembled nanovaccine for immune potentiation and cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaofang Zhong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chunting He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ming Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yingying Hou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Zhao T, Cai Y, Jiang Y, He X, Wei Y, Yu Y, Tian X. Vaccine adjuvants: mechanisms and platforms. Signal Transduct Target Ther 2023; 8:283. [PMID: 37468460 PMCID: PMC10356842 DOI: 10.1038/s41392-023-01557-7] [Citation(s) in RCA: 284] [Impact Index Per Article: 142.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023] Open
Abstract
Adjuvants are indispensable components of vaccines. Despite being widely used in vaccines, their action mechanisms are not yet clear. With a greater understanding of the mechanisms by which the innate immune response controls the antigen-specific response, the adjuvants' action mechanisms are beginning to be elucidated. Adjuvants can be categorized as immunostimulants and delivery systems. Immunostimulants are danger signal molecules that lead to the maturation and activation of antigen-presenting cells (APCs) by targeting Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs) to promote the production of antigen signals and co-stimulatory signals, which in turn enhance the adaptive immune responses. On the other hand, delivery systems are carrier materials that facilitate antigen presentation by prolonging the bioavailability of the loaded antigens, as well as targeting antigens to lymph nodes or APCs. The adjuvants' action mechanisms are systematically summarized at the beginning of this review. This is followed by an introduction of the mechanisms, properties, and progress of classical vaccine adjuvants. Furthermore, since some of the adjuvants under investigation exhibit greater immune activation potency than classical adjuvants, which could compensate for the deficiencies of classical adjuvants, a summary of the adjuvant platforms under investigation is subsequently presented. Notably, we highlight the different action mechanisms and immunological properties of these adjuvant platforms, which will provide a wide range of options for the rational design of different vaccines. On this basis, this review points out the development prospects of vaccine adjuvants and the problems that should be paid attention to in the future.
Collapse
Affiliation(s)
- Tingmei Zhao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yulong Cai
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yujie Jiang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yifan Yu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Laera D, HogenEsch H, O'Hagan DT. Aluminum Adjuvants-'Back to the Future'. Pharmaceutics 2023; 15:1884. [PMID: 37514070 PMCID: PMC10383759 DOI: 10.3390/pharmaceutics15071884] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Aluminum-based adjuvants will continue to be a key component of currently approved and next generation vaccines, including important combination vaccines. The widespread use of aluminum adjuvants is due to their excellent safety profile, which has been established through the use of hundreds of millions of doses in humans over many years. In addition, they are inexpensive, readily available, and are well known and generally accepted by regulatory agencies. Moreover, they offer a very flexible platform, to which many vaccine components can be adsorbed, enabling the preparation of liquid formulations, which typically have a long shelf life under refrigerated conditions. Nevertheless, despite their extensive use, they are perceived as relatively 'weak' vaccine adjuvants. Hence, there have been many attempts to improve their performance, which typically involves co-delivery of immune potentiators, including Toll-like receptor (TLR) agonists. This approach has allowed for the development of improved aluminum adjuvants for inclusion in licensed vaccines against HPV, HBV, and COVID-19, with others likely to follow. This review summarizes the various aluminum salts that are used in vaccines and highlights how they are prepared. We focus on the analytical challenges that remain to allowing the creation of well-characterized formulations, particularly those involving multiple antigens. In addition, we highlight how aluminum is being used to create the next generation of improved adjuvants through the adsorption and delivery of various TLR agonists.
Collapse
Affiliation(s)
- Donatello Laera
- Technical Research & Development, Drug Product, GSK, 53100 Siena, Italy
- Global Manufacturing Division, Corporate Industrial Analytics, Chiesi Pharmaceuticals, 43122 Parma, Italy
| | - Harm HogenEsch
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | | |
Collapse
|
13
|
Zhang T, He P, Guo D, Chen K, Hu Z, Zou Y. Research Progress of Aluminum Phosphate Adjuvants and Their Action Mechanisms. Pharmaceutics 2023; 15:1756. [PMID: 37376204 DOI: 10.3390/pharmaceutics15061756] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/14/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Although hundreds of different adjuvants have been tried, aluminum-containing adjuvants are by far the most widely used currently. It is worth mentioning that although aluminum-containing adjuvants have been commonly applied in vaccine production, their acting mechanism remains not completely clear. Thus far, researchers have proposed the following mechanisms: (1) depot effect, (2) phagocytosis, (3) activation of pro-inflammatory signaling pathway NLRP3, (4) host cell DNA release, and other mechanisms of action. Having an overview on recent studies to increase our comprehension on the mechanisms by which aluminum-containing adjuvants adsorb antigens and the effects of adsorption on antigen stability and immune response has become a mainstream research trend. Aluminum-containing adjuvants can enhance immune response through a variety of molecular pathways, but there are still significant challenges in designing effective immune-stimulating vaccine delivery systems with aluminum-containing adjuvants. At present, studies on the acting mechanism of aluminum-containing adjuvants mainly focus on aluminum hydroxide adjuvants. This review will take aluminum phosphate as a representative to discuss the immune stimulation mechanism of aluminum phosphate adjuvants and the differences between aluminum phosphate adjuvants and aluminum hydroxide adjuvants, as well as the research progress on the improvement of aluminum phosphate adjuvants (including the improvement of the adjuvant formula, nano-aluminum phosphate adjuvants and a first-grade composite adjuvant containing aluminum phosphate). Based on such related knowledge, determining optimal formulation to develop effective and safe aluminium-containing adjuvants for different vaccines will become more substantiated.
Collapse
Affiliation(s)
- Ting Zhang
- Sinovac Biotech Sciences Co., Ltd., Beijing 102601, China
| | - Peng He
- Division of Hepatitis Virus & Enterovirus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing 102619, China
| | - Dejia Guo
- Sinovac Life Sciences Co., Ltd., Beijing 102601, China
| | - Kaixi Chen
- Sinovac Life Sciences Co., Ltd., Beijing 102601, China
| | - Zhongyu Hu
- Division of Hepatitis Virus & Enterovirus Vaccines, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing 102619, China
| | - Yening Zou
- Sinovac Life Sciences Co., Ltd., Beijing 102601, China
| |
Collapse
|
14
|
Dong H, Li Q, Zhang Y, Ding M, Teng Z, Mou Y. Biomaterials Facilitating Dendritic Cell-Mediated Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301339. [PMID: 37088780 PMCID: PMC10288267 DOI: 10.1002/advs.202301339] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has exhibited remarkable clinical prospects because DCs play a central role in initiating and regulating adaptive immune responses. However, the application of traditional DC-mediated immunotherapy is limited due to insufficient antigen delivery, inadequate antigen presentation, and high levels of immunosuppression. To address these challenges, engineered biomaterials have been exploited to enhance DC-mediated immunotherapeutic effects. In this review, vital principal components that can enhance DC-mediated immunotherapeutic effects are first introduced. The parameters considered in the rational design of biomaterials, including targeting modifications, size, shape, surface, and mechanical properties, which can affect biomaterial optimization of DC functions, are further summarized. Moreover, recent applications of various engineered biomaterials in the field of DC-mediated immunotherapy are reviewed, including those serve as immune component delivery platforms, remodel the tumor microenvironment, and synergistically enhance the effects of other antitumor therapies. Overall, the present review comprehensively and systematically summarizes biomaterials related to the promotion of DC functions; and specifically focuses on the recent advances in biomaterial designs for DC activation to eradicate tumors. The challenges and opportunities of treatment strategies designed to amplify DCs via the application of biomaterials are discussed with the aim of inspiring the clinical translation of future DC-mediated cancer immunotherapies.
Collapse
Affiliation(s)
- Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Qiang Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Yu Zhang
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Meng Ding
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Yongbin Mou
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| |
Collapse
|
15
|
Lajiness JD, Cook-Mills JM. Catching Our Breath: Updates on the Role of Dendritic Cell Subsets in Asthma. Adv Biol (Weinh) 2023; 7:e2200296. [PMID: 36755197 PMCID: PMC10293089 DOI: 10.1002/adbi.202200296] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/04/2023] [Indexed: 02/10/2023]
Abstract
Dendritic cells (DCs), as potent antigen presenting cells, are known to play a central role in the pathophysiology of asthma. The understanding of DC biology has evolved over the years to include multiple subsets of DCs with distinct functions in the initiation and maintenance of asthma. Furthermore, asthma is increasingly recognized as a heterogeneous disease with potentially diverse underlying mechanisms. The goal of this review is to summarize the role of DCs and the various subsets therein in the pathophysiology of asthma and highlight some of the crucial animal models shaping the field today. Potential future avenues of investigation to address existing gaps in knowledge are discussed.
Collapse
Affiliation(s)
- Jacquelyn D Lajiness
- Department of Pediatrics, Division of Neonatology, Indiana University School of Medicine, 1030 West Michigan Street, Suite C 4600, Indianapolis, IN, 46202-5201, USA
| | - Joan M Cook-Mills
- Department of Pediatrics, Department of Microbiology and Immunology, Pediatric Pulmonary, Asthma, and Allergy Basic Research Program, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut Street, R4-202A, Indianapolis, IN, 46202, USA
| |
Collapse
|
16
|
Thanvi R, Nada S, Dissanayake R, Vartak A, Sebilleau CO, Alom NE, Prestwich EG, Wall KA, Sucheck SJ. Synthesis and Evaluation of a Self-Adjuvanting Pseudomonal Vaccine Based on Major Outer Membrane Porin OprF Epitopes Formulated with Low-Toxicity QS-21-Containing Liposomes. Bioconjug Chem 2023; 34:893-910. [PMID: 37092892 PMCID: PMC10723056 DOI: 10.1021/acs.bioconjchem.3c00103] [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: 04/25/2023]
Abstract
Pseudomonas aeruginosa (PA) is a Gram-negative pathogen that the World Health Organization has ranked as a priority 1 (critical) threat. One potential prophylactic approach to preventing or reducing the incidence of PA would be development of a long sought-after vaccine. Both antibody and CD4+ T-cell responses have been noted as playing key roles in protection against infection. In these studies, we have designed a prototype vaccine consisting of several known linear B-cell epitopes derived from an outer membrane porin F (OprF). The resulting thiol-containing protein was conjugated to a version of the lipopeptide-based Toll-like receptor agonist Pam3CysSK4Mal (10) containing a maleimide moiety and formulated into dipalmitoylphosphatidylcholine (DPPC)/cholesterol (Chol) liposomes. Mice immunized with the resulting vaccine generated antibodies that bound PA14 (serotype O10) in vitro and induced opsonization in the presence of rabbit complement and murine macrophage RAW264.7 cells. The liposome was optimized to contain 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1,2-dimyristoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (DMPG), Chol, Pam3CysSK4-OprF (12) and the Quillaja saponaria-derived saponin adjuvant QS-21. The resulting vaccine formulation produced significantly higher antibody titers, increased the IgG2a antibody isotype, and increased the number of IgG-producing B-cells as well as splenic primed T-cells. In summary, the liposomal vaccine platform was found highly useful for the generation of a robust and balanced TH1/TH2 response.
Collapse
Affiliation(s)
- Radhika Thanvi
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Shadia Nada
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43614, United States
| | - Ravindika Dissanayake
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43614, United States
| | - Abhishek Vartak
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Chloé Olayinka Sebilleau
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Nur-E Alom
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Erin G Prestwich
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43614, United States
| | - Katherine A Wall
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43614, United States
| | - Steven J Sucheck
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| |
Collapse
|
17
|
Advances on the early cellular events occurring upon exposure of human macrophages to aluminum oxyhydroxide adjuvant. Sci Rep 2023; 13:3198. [PMID: 36823452 PMCID: PMC9950428 DOI: 10.1038/s41598-023-30336-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Aluminum compounds are the most widely used adjuvants in veterinary and human vaccines. Despite almost a century of use and substantial advances made in recent decades about their fate and biological effects, the exact mechanism of their action has been continuously debated, from the initial "depot-theory" to the direct immune system stimulation, and remains elusive. Here we investigated the early in vitro response of primary human PBMCs obtained from healthy individuals to aluminum oxyhydroxide (the most commonly used adjuvant) and a whole vaccine, in terms of internalization, conventional and non-conventional autophagy pathways, inflammation, ROS production, and mitochondrial metabolism. During the first four hours of contact, aluminum oxyhydroxide particles, with or without adsorbed vaccine antigen, (1) were quickly recognized and internalized by immune cells; (2) increased and balanced two cellular clearance mechanisms, i.e. canonical autophagy and LC3-associated phagocytosis; (3) induced an inflammatory response with TNF-α production as an early event; (4) and altered mitochondrial metabolism as assessed by both decreased maximal oxygen consumption and reduced mitochondrial reserve, thus potentially limiting further adaptation to other energetic requests. Further studies should consider a multisystemic approach of the cellular adjuvant mechanism involving interconnections between clearance mechanism, inflammatory response and mitochondrial respiration.
Collapse
|
18
|
Liao Y, Sun L, Nie M, Li J, Huang X, Heng S, Zhang W, Xia T, Guo Z, Zhao Q, Zhang LJ. Modulation of Skin Inflammatory Responses by Aluminum Adjuvant. Pharmaceutics 2023; 15:pharmaceutics15020576. [PMID: 36839900 PMCID: PMC9966661 DOI: 10.3390/pharmaceutics15020576] [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: 01/07/2023] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Aluminum salt (AS), one of the most commonly used vaccine adjuvants, has immuno-modulatory activity, but how the administration of AS alone may impact the activation of the skin immune system under inflammatory conditions has not been investigated. Here, we studied the therapeutic effect of AS injection on two distinct skin inflammatory mouse models: an imiquimod (IMQ)-induced psoriasis-like model and an MC903 (calcipotriol)-induced atopic dermatitis-like model. We found that injection of a high dose of AS not only suppressed the IMQ-mediated development of T-helper 1 (Th1) and T-helper 17 (Th17) immune responses but also inhibited the IMQ-mediated recruitment and/or activation of neutrophils and macrophages. In contrast, AS injection enhanced MC903-mediated development of the T-helper 2 (Th2) immune response and neutrophil recruitment. Using an in vitro approach, we found that AS treatment inhibited Th1 but promoted Th2 polarization of primary lymphocytes, and inhibited activation of peritoneal macrophages but not bone marrow derived neutrophils. Together, our results suggest that the injection of a high dose of AS may inhibit Th1 and Th17 immune response-driven skin inflammation but promote type 2 immune response-driven skin inflammation. These results may provide a better understanding of how vaccination with an aluminum adjuvant alters the skin immune response to external insults.
Collapse
Affiliation(s)
- Yanhang Liao
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361002, China
| | - Lixiang Sun
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361002, China
| | - Meifeng Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Jiacheng Li
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361002, China
| | - Xiaofen Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Shujun Heng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361002, China
| | - Wenlu Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361002, China
| | - Tian Xia
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361002, China
| | - Zhuolin Guo
- Department of Dermatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Correspondence: (Q.Z.); (L.-j.Z.)
| | - Ling-juan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361002, China
- Correspondence: (Q.Z.); (L.-j.Z.)
| |
Collapse
|
19
|
Gatt Z, Gunes U, Raponi A, da Rosa LC, Brewer JM. Review: Unravelling the Role of DNA Sensing in Alum Adjuvant Activity. DISCOVERY IMMUNOLOGY 2022; 2:kyac012. [PMID: 38567066 PMCID: PMC10917177 DOI: 10.1093/discim/kyac012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/11/2022] [Accepted: 12/28/2022] [Indexed: 04/04/2024]
Abstract
Public interest in vaccines is at an all-time high following the SARS-CoV-2 global pandemic. Currently, over 6 billion doses of various vaccines are administered globally each year. Most of these vaccines contain Aluminium-based adjuvants (alum), which have been known and used for almost 100 years to enhance vaccine immunogenicity. However, despite the historical use and importance of alum, we still do not have a complete understanding of how alum works to drive vaccine immunogenicity. In this article, we critically review studies investigating the mechanisms of action of alum adjuvants, highlighting some of the misconceptions and controversies within the area. Although we have emerged with a clearer understanding of how this ubiquitous adjuvant works, we have also highlighted some of the outstanding questions in the field. While these may seem mainly of academic interest, developing a more complete understanding of these mechanisms has the potential to rationally modify and improve the immune response generated by alum-adjuvanted vaccines.
Collapse
Affiliation(s)
- Zara Gatt
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland
| | - Utku Gunes
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland
| | - Arianna Raponi
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland
| | - Larissa Camargo da Rosa
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland
| | - James M Brewer
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland
| |
Collapse
|
20
|
Wang Q, Song Z, Yang J, He Q, Mao Q, Bai Y, Liu J, An C, Yan X, Cui B, Song L, Liu D, Xu M, Liang Z. Transcriptomic analysis of the innate immune signatures of a SARS-CoV-2 protein subunit vaccine ZF2001 and an mRNA vaccine RRV. Emerg Microbes Infect 2022; 11:1145-1153. [PMID: 35343384 PMCID: PMC9037177 DOI: 10.1080/22221751.2022.2059404] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022]
Abstract
Analysis of large-scale gene expression post vaccination can provide an overview of immune responses. We used transcriptional approaches to comprehensively analyze the innate immune response signatures elicited by protein subunit (PS) vaccine ZF2001 and an mRNA vaccine named RRV. A fine-grained time-dependent dissection of large-scale gene expression post immunization revealed that ZF001 induced MHC class II-related genes, including cd74 and H2-Aa, more expeditiously than the RRV. Notably, the RRV induced MHC class I-related genes such as Tap1/2, B2m, and H2-D1/K1. At day 21 post immunization, the titres of binding and neutralization antibody (NAb) induced by both vaccines were comparable, which were accordant with the expression level of genes essential to BCR/TCR signalling transduction and B/T cells activation at day 7. However, compared to ZF2001, the early responses of RRV were more robust, including the activation of pattern recognition receptors (PRRs), expression of genes involved in RNA degradation, and transcription inhibition, which are directly related to anti-viral signals. This pattern also coincided with the induction of cytokines by the RRV. Generally, the transcriptomic patterns of two very different vaccines mapped here provide a framework for establishing correlates between the induction of genes and protection, which can be tailored for evoking specific and potent immune responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Qian Wang
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
| | - Ziyang Song
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
- Shanghai Institute of Biological Products Co., Ltd., China National Biotec Group, Shanghai, People's Republic of China
| | - Jinghuan Yang
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
- Beijing Institute of Biological Products Co., Ltd., China National Biotec Group, Beijing, People's Republic of China
| | - Qian He
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
| | - Qunying Mao
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
| | - Yu Bai
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
| | - Jianyang Liu
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
| | - Chaoqiang An
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
- Minhai Biotechnology Co., Ltd., Beijing, People’s Republic of China
| | - Xujia Yan
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
- Changchun Institute of Biological Products Co., Ltd., China National Biotec Group, Changchun, People's Republic of China
| | - Bopei Cui
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
- Wuhan Institute of Biological Products Co., Ltd., China National Biotec Group, Wuhan, People's Republic of China
| | - Lifang Song
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
| | - Dong Liu
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
- Changchun Institute of Biological Products Co., Ltd., China National Biotec Group, Changchun, People's Republic of China
| | - Miao Xu
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
| | - Zhenglun Liang
- Hepatitis virus and enterovirus vaccines Division, National Institutes for Food and Drug Control, Beijing, People’s Republic of China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, People's Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, People's Republic of China
| |
Collapse
|
21
|
Zhuang W, Zhang L, Zheng Y, Liu B, Ma C, Zhao W, Liu S, Liu F, Gao C. USP3 deubiquitinates and stabilizes the adapter protein ASC to regulate inflammasome activation. Cell Mol Immunol 2022; 19:1141-1152. [PMID: 36050480 PMCID: PMC9508167 DOI: 10.1038/s41423-022-00917-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/06/2022] [Indexed: 11/08/2022] Open
Abstract
Inflammasomes are essential components of the innate immune system and its defense against infections, whereas the dysregulation of inflammasome activation has a detrimental effect on human health. The activation of inflammasomes is subjected to tight regulation to maintain immune homeostasis, yet the underlying mechanism remains elusive. Here, we identify USP3 as a direct deubiquitinating enzyme (DUB) for ASC, the central adapter mediating the assembly and activation of most inflammasomes. USP3 removes the K48-linked ubiquitination on ASC and strengthens its stability by blocking proteasomal degradation. Additionally, USP3 promotes inflammasome activation, and this function was confirmed in mouse models of aluminum (Alum)-induced peritonitis, F. novicida infection and flagellin-induced pneumonia in vivo. Our work unveils that USP3 functions as a key regulator of ASC ubiquitination and maintains the physiological role of ASC in mediating inflammasome activation, and we propose a new mechanism by which the ubiquitination of ASC regulates inflammasome activation.
Collapse
Affiliation(s)
- Wanxin Zhuang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Chunhong Ma
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Suxia Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Feng Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China.
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China.
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
22
|
Heine S, Aguilar-Pimentel A, Russkamp D, Alessandrini F, Gailus-Durner V, Fuchs H, Ollert M, Bredehorst R, Ohnmacht C, Zissler UM, Hrabě de Angelis M, Schmidt-Weber CB, Blank S. Thermosensitive PLGA–PEG–PLGA Hydrogel as Depot Matrix for Allergen-Specific Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14081527. [PMID: 35893787 PMCID: PMC9329805 DOI: 10.3390/pharmaceutics14081527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Allergen-specific immunotherapy (AIT) is the only currently available curative treatment option for allergic diseases. AIT often includes depot-forming and immunostimulatory adjuvants, to prolong allergen presentation and to improve therapeutic efficacy. The use of aluminium salts in AIT, which are commonly used as depot-forming adjuvants, is controversially discussed, due to health concerns and Th2-promoting activity. Therefore, there is the need for novel delivery systems in AIT with similar therapeutic efficacy compared to classical AIT strategies. In this study, a triblock copolymer (hydrogel) was assessed as a delivery system for AIT in a murine model of allergic asthma. We show that the hydrogel combines the advantages of both depot function and biodegradability at the same time. We further demonstrate the suitability of hydrogel to release different bioactive compounds in vitro and in vivo. AIT delivered with hydrogel reduces key parameters of allergic inflammation, such as inflammatory cell infiltration, mucus hypersecretion, and allergen-specific IgE, in a comparable manner to standard AIT treatment. Additionally, hydrogel-based AIT is superior in inducing allergen-specific IgG antibodies with potentially protective functions. Taken together, hydrogel represents a promising delivery system for AIT that is able to combine therapeutic allergen administration with the prolonged release of immunomodulators at the same time.
Collapse
Affiliation(s)
- Sonja Heine
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Antonio Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.-P.); (V.G.-D.); (H.F.); (M.H.d.A.)
| | - Dennis Russkamp
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Francesca Alessandrini
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.-P.); (V.G.-D.); (H.F.); (M.H.d.A.)
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.-P.); (V.G.-D.); (H.F.); (M.H.d.A.)
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 4354 Esch-Sur-Alzette, Luxembourg;
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, 5000 Odense, Denmark
| | - Reinhard Bredehorst
- Institute of Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany;
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Ulrich M. Zissler
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.-P.); (V.G.-D.); (H.F.); (M.H.d.A.)
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
- Correspondence: ; Tel.: +49-89-318-726-25
| |
Collapse
|
23
|
Masson JD, Angrand L, Badran G, de Miguel R, Crépeaux G. Clearance, biodistribution, and neuromodulatory effects of aluminum-based adjuvants. Systematic review and meta-analysis: what do we learn from animal studies? Crit Rev Toxicol 2022; 52:403-419. [PMID: 36112128 DOI: 10.1080/10408444.2022.2105688] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Aluminum (Al) salts are commonly used as adjuvants in human and veterinary vaccines for almost a century. Despite this long history of use and the very large number of exposed individuals, data in the literature concerning the fate of these molecules after injection and their potential effects on the nervous system is limited. In the context of (i) an increase of exposure to Al salts through vaccination; (ii) the absence of safety values determined by health regulators; (iii) the lack of robustness of the studies used as references to officially claim Al adjuvant innocuity; (iv) the publication of several animal studies investigating Al salts clearance/biopersistence and neurotoxicity; we have examined in this review all published studies performed on animals and assessing Al adjuvants kinetics, biodistribution, and neuromodulation since the first work of A. Glenny in the 1920s. The diversity of methodological approaches, results, and potential weaknesses of the 31 collected studies are exposed. A large range of protocols has been used, including a variety of exposure schedule and analyses methods, making comparisons between studies uneasy. Nevertheless, published data highlight that when biopersistence, translocation, or neuromodulation were assessed, they were documented whatever the different in vivo models and methods used. Moreover, the studies pointed out the crucial importance of the different Al adjuvant physicochemical properties and host genetic background on their kinetics, biodistribution, and neuromodulatory effects. Regarding the state of the art on this key public health topic, further studies are clearly needed to determine the exact safety level of Al salts.
Collapse
Affiliation(s)
- J-D Masson
- INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - L Angrand
- INSERM, IMRB, Univ Paris Est Créteil, Créteil, France.,École Nationale Vétérinaire d'Alfort, IMRB, Maisons-Alfort, France
| | - G Badran
- INSERM, IMRB, Univ Paris Est Créteil, Créteil, France.,Laboratoire SABNP, Université d'Evry Val d'Essonne, Paris, France
| | - R de Miguel
- Department of Animal Pathology, University of Zaragoza, Zaragoza, Spain
| | - G Crépeaux
- INSERM, IMRB, Univ Paris Est Créteil, Créteil, France.,École Nationale Vétérinaire d'Alfort, IMRB, Maisons-Alfort, France
| |
Collapse
|
24
|
Meena J, Singhvi P, Srichandan S, Dandotiya J, Verma J, Singh M, Ahuja R, Panwar N, Wani TQ, Khatri R, Siddiqui G, Gupta A, Samal S, Panda AK. RBD decorated PLA nanoparticle admixture with aluminum hydroxide elicit robust and long lasting immune response against SARS-CoV-2. Eur J Pharm Biopharm 2022; 176:43-53. [PMID: 35589003 PMCID: PMC9110063 DOI: 10.1016/j.ejpb.2022.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/02/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022]
Abstract
Nanoparticles-based multivalent antigen display has the capability of mimicking natural virus infection characteristics, making it useful for eliciting potent long-lasting immune response. Several vaccines are developed against global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However these subunit vaccines use mammalian expression system, hence mass production with rapid pace is a bigger challenge. In contrast E. coli based subunit vaccine production circumvents these limitations.The objective of the present investigation was to develop nanoparticle vaccine with multivalent display of receptor binding domain (RBD) of SARS-CoV-2 expressed in E. coli. Results showed that RBD entrapped PLA (Poly lactic acid) nanoparticle in combination with aluminum hydroxide elicited 9-fold higher immune responses as compared to RBD adsorbed aluminum hydroxide, a common adjuvant used for human immunization. It was interesting to note that RBD entrapped PLA nanoparticle with aluminum hydroxide not only generated robust and long-lasting antibody response but also provided Th1 and Th2 balanced immune response. Moreover, challenge with 1 µg of RBD alone was able to generate secondary antibody response, suggesting that immunization with RBD-PLA nanoparticleshas the ability to elicit memory antibody against RBD. Plaque assay revealed that the antibody generated using the polymeric formulation was able to neutralize SARS-CoV-2.The RBD entrapped PLA nanoparticles blended with aluminum hydroxide thus has potential to develop asa subunit vaccine against COVID-19.
Collapse
Affiliation(s)
- Jairam Meena
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India; Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh 221005, India.
| | - Priyank Singhvi
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sudeepa Srichandan
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Jyotsna Dandotiya
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Juhi Verma
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Mamta Singh
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Rahul Ahuja
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Neha Panwar
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Tabiya Qayoom Wani
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Ritika Khatri
- Infection and Immunology Laboratory, Translational Health Science & Technology Institute, Gurgaon-Faridabad, India
| | - Gazala Siddiqui
- Infection and Immunology Laboratory, Translational Health Science & Technology Institute, Gurgaon-Faridabad, India
| | - Anuradha Gupta
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sweety Samal
- Infection and Immunology Laboratory, Translational Health Science & Technology Institute, Gurgaon-Faridabad, India
| | - Amulya Kumar Panda
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
25
|
|
26
|
Kooijman S, Vrieling H, Verhagen L, de Ridder J, de Haan A, van Riet E, Heck AJ, Kersten GF, Pennings JL, Metz B, Meiring HD. Aluminum Hydroxide and Aluminum Phosphate Adjuvants Elicit a Different Innate Immune Response. J Pharm Sci 2022; 111:982-990. [DOI: 10.1016/j.xphs.2022.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 12/21/2022]
|
27
|
Xu H, Cai L, Hufnagel S, Cui Z. Intranasal vaccine: Factors to consider in research and development. Int J Pharm 2021; 609:121180. [PMID: 34637935 DOI: 10.1016/j.ijpharm.2021.121180] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
Most existing vaccines for human use are administered by needle-based injection. Administering vaccines needle-free intranasally has numerous advantages over by needle-based injection, but there are only a few intranasal vaccines that are currently approved for human use, and all of them are live attenuated influenza virus vaccines. Clearly, there are immunological as well as non-immunological challenges that prevent vaccine developers from choosing the intranasal route of administration. We reviewed current approved intranasal vaccines and pipelines and described the target of intranasal vaccines, i.e. nose and lymphoid tissues in the nasal cavity. We then analyzed factors unique to intranasal vaccines that need to be considered when researching and developing new intranasal vaccines. We concluded that while the choice of vaccine formulations, mucoadhesives, mucosal and epithelial permeation enhancers, and ligands that target M-cells are important, safe and effective intranasal mucosal vaccine adjuvants are needed to successfully develop an intranasal vaccine that is not based on live-attenuated viruses or bacteria. Moreover, more effective intranasal vaccine application devices that can efficiently target a vaccine to lymphoid tissues in the nasal cavity as well as preclinical animal models that can better predict intranasal vaccine performance in clinical trials are needed to increase the success rate of intranasal vaccines in clinical trials.
Collapse
Affiliation(s)
- Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Lucy Cai
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephanie Hufnagel
- 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
|
28
|
Choy CH, He L, Tulumello D, Gajewska B, Terebiznik MR, Botelho RJ, Azizi A. Aggregation and Size Attributes Analysis of Unadsorbed and Adjuvant-adsorbed Antigens using a Multispectral Imaging Flow Cytometer Platform. J Pharm Sci 2021; 111:672-679. [PMID: 34742727 DOI: 10.1016/j.xphs.2021.10.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/30/2021] [Accepted: 10/30/2021] [Indexed: 11/26/2022]
Abstract
Various vaccine quality attributes should be monitored to ensure consistency, potency, purity, and safety of vaccine products prior to lot release. Vaccine particle size and protein antigen aggregation are two important considerations for particle-adsorbed vaccines. In this study, we evaluated the use of imaging flow cytometry as a potential all-in-one platform to measure adjuvant particle size and to detect protein aggregates through a combination of brightfield microscopy, side scatter detection, and fluorescence microscopy. An aluminum phosphate adjuvant was analyzed for size using the brightfield function, and the size measurement was compared against laser diffraction. Heat-induced protein aggregates of either unadsorbed antigens or aluminum phosphate adjuvant-adsorbed antigens were stained with the fluorescent ProteoStat aggregation dye, followed by detection and analysis using a combination of the brightfield and fluorescence microscopy functions. The change in aggregation of unadsorbed antigens was confirmed using dynamic light scattering. These results demonstrate the versatility of the imaging flow cytometry platform for the evaluation of multiple vaccine quality characteristics.
Collapse
Affiliation(s)
- Christopher H Choy
- Immunology platform, Analytical Sciences North America, Sanofi Pasteur, 1755 Steeles Avenue West, Toronto, ON, M2R 3T4, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Liwei He
- Immunology platform, Analytical Sciences North America, Sanofi Pasteur, 1755 Steeles Avenue West, Toronto, ON, M2R 3T4, Canada
| | - David Tulumello
- Biochemistry platform, Analytical Sciences North America, Sanofi Pasteur, 1755 Steeles Avenue West, Toronto, Ontario, M2R 3T4, Canada
| | - Beata Gajewska
- Immunology platform, Analytical Sciences North America, Sanofi Pasteur, 1755 Steeles Avenue West, Toronto, ON, M2R 3T4, Canada
| | - Mauricio R Terebiznik
- Department of Biological Sciences, University of Toronto at Scarborough, Toronto, Ontario, M1C 1A4, Canada
| | - Roberto J Botelho
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Ali Azizi
- Immunology platform, Analytical Sciences North America, Sanofi Pasteur, 1755 Steeles Avenue West, Toronto, ON, M2R 3T4, Canada.
| |
Collapse
|
29
|
Heterologous administration of HPV16 E7 epitope-loaded nanocomplexes inhibits tumor growth in mouse model. Int Immunopharmacol 2021; 101:108298. [PMID: 34739928 DOI: 10.1016/j.intimp.2021.108298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/02/2023]
Abstract
The nanostructured complexes can result in enhanced vaccine efficacy by facilitating the distribution and uptake of antigens by antigen-presenting cells (APCs), thereby stimulating immune responses. Here, we hypothesized that either directly coating of nanoadjuvants including aluminum phosphate (AlPO4) and adenovirus (Ad) with a modified HPV16 E7 MHC-I specific epitope, RAHYNIVTF49-57, or mixing the CpG oligodeoxynucleotide (CpG-ODN) with the cationic epitope to form nanocomlexes, and their combinational therapy would enhance their anti-tumor effects in a TC-1 mouse model. The positively-charged HPV16 E7 epitope was attracted to the oppositely-charged adjuvants by electrostatic interaction to generate epitope/adjuvant nanocomplexes. We showed that coating the nanosized adjuvants with the cationic epitope increased the particles' surface charge without significant change in their size. We then tested the cellular immunogenicity and therapeutic efficacy of nanocomplexes by measuring IL-10 and IFN-γ production, the expression of CD107a as a marker of CTL response, and tumor growth inhibition. The nanocomplexes were administered either in homologous or heterologous prime-boost regimens, and heterologous immunizations including Ad/Pep-CpG/Pep, CpG/Pep-Ad/Pep, Ad/Pep-Alum/Pep, and Alum/Pep-Ad/Pep induced significantly higher levels of IL-10, IFN-γ, and CD107a-expressing CD8 T cells compared with homologous administrations. Furthermore, the tumor growth was significantly suppressed in mice receiving nanostructured complexes in the heterologous immunizations. Our study highlights the potential of the heterologous prime-boost administration of the epitope-coated nanostructures as an effective immunization strategy.
Collapse
|
30
|
Ong GH, Lian BSX, Kawasaki T, Kawai T. Exploration of Pattern Recognition Receptor Agonists as Candidate Adjuvants. Front Cell Infect Microbiol 2021; 11:745016. [PMID: 34692565 PMCID: PMC8526852 DOI: 10.3389/fcimb.2021.745016] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
Adjuvants are used to maximize the potency of vaccines by enhancing immune reactions. Components of adjuvants include pathogen-associated molecular patterns (PAMPs) and damage-associate molecular patterns (DAMPs) that are agonists for innate immune receptors. Innate immune responses are usually activated when pathogen recognition receptors (PRRs) recognize PAMPs derived from invading pathogens or DAMPs released by host cells upon tissue damage. Activation of innate immunity by PRR agonists in adjuvants activates acquired immune responses, which is crucial to enhance immune reactions against the targeted pathogen. For example, agonists for Toll-like receptors have yielded promising results as adjuvants, which target PRR as adjuvant candidates. However, a comprehensive understanding of the type of immunological reaction against agonists for PRRs is essential to ensure the safety and reliability of vaccine adjuvants. This review provides an overview of the current progress in development of PRR agonists as vaccine adjuvants, the molecular mechanisms that underlie activation of immune responses, and the enhancement of vaccine efficacy by these potential adjuvant candidates.
Collapse
Affiliation(s)
- Guang Han Ong
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Benedict Shi Xiang Lian
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Takumi Kawasaki
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| |
Collapse
|
31
|
Zhang L, Ko CJ, Li Y, Jie Z, Zhu L, Zhou X, Xie X, Gao T, Liu T, Cheng X, Sun SC. Peli1 facilitates NLRP3 inflammasome activation by mediating ASC ubiquitination. Cell Rep 2021; 37:109904. [PMID: 34706239 PMCID: PMC12011377 DOI: 10.1016/j.celrep.2021.109904] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/25/2021] [Accepted: 10/07/2021] [Indexed: 11/23/2022] Open
Abstract
Inflammasomes are crucial for innate immunity against infections and, when deregulated, also contribute to inflammatory diseases. Here, we identify a critical function of the E3 ubiquitin ligase Peli1 in regulating the activation of NLRP3 inflammasome. Peli1 deficiency impairs induction of interleukin-1β (IL-1β) secretion by different NLRP3 inducers, but not by inducers of the Aim2, NLRP1, and NLRC4 inflammasomes. Peli1-deficient mice have alleviated peritonitis induction by alum and display increased resistance to lipopolysaccharide (LPS) endotoxin shock, coupled with decreased serum concentration of IL-1β. Peli1 is required for NLRP3-induced caspase-1 activation and IL-1β maturation. Mechanistically, Peli1 conjugates K63 ubiquitin chain to lysine 55 of the inflammasome adaptor apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), which in turn facilitates ASC/NLRP3 interaction and ASC oligomerization, thereby contributing to inflammasome activation. Peli1 deficiency impairs the ubiquitination of ASC and inhibits inflammasome activation. Our findings establish Peli1 as an important inflammasome regulator and suggest a mechanism by which Peli1 mediates inflammatory responses.
Collapse
Affiliation(s)
- Lingyun Zhang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Chun-Jung Ko
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Yanchuan Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Zuliang Jie
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Lele Zhu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Xiaofei Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Xiaoping Xie
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Tianxiao Gao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Ting Liu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Xuhong Cheng
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX, USA; MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
32
|
Alum Pickering Emulsion as Effective Adjuvant to Improve Malaria Vaccine Efficacy. Vaccines (Basel) 2021; 9:vaccines9111244. [PMID: 34835175 PMCID: PMC8624716 DOI: 10.3390/vaccines9111244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/27/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Malaria is a life-threatening global epidemic disease and has caused more than 400,000 deaths in 2019. To control and prevent malaria, the development of a vaccine is a potential method. An effective malaria vaccine should either combine antigens from all stages of the malaria parasite’s life cycle, or epitopes of multiple key antigens due to the complexity of the Plasmodium parasite. Malaria’s random constructed antigen-1 (M.RCAg-1) is one of the recombinant vaccines, which was selected from a DNA library containing thousands of diverse multi-epitope chimeric antigen genes. Moreover, besides selecting an antigen, using an adjuvant is another important procedure for most vaccine development procedures. Freund’s adjuvant is considered an effective vaccine adjuvant for malaria vaccine, but it cannot be used in clinical settings because of its serious side effects. Traditional adjuvants, such as alum adjuvant, are limited by their unsatisfactory immune effects in malaria vaccines, hence there is an urgent need to develop a novel, safe and efficient adjuvant. In recent years, Pickering emulsions have attracted increasing attention as novel adjuvant. In contrast to classical emulsions, Pickering emulsions are stabilized by solid particles instead of surfactant, having pliability and lateral mobility. In this study, we selected aluminum hydroxide gel (termed as “alum”) as a stabilizer to prepare alum-stabilized Pickering emulsions (ALPE) as a malaria vaccine adjuvant. In addition, monophosphoryl lipid A (MPLA) as an immunostimulant was incorporated into the Pickering emulsion (ALMPE) to further enhance the immune response. In vitro tests showed that, compared with alum, ALPE and ALMPE showed higher antigen load rates and could be effectively endocytosed by J774a.1 cells. In vivo studies indicated that ALMPE could induce as high antibody titers as Freund’s adjuvant. The biocompatibility study also proved ALMPE with excellent biocompatibility. These results suggest that ALMPE is a potential adjuvant for a malaria vaccine.
Collapse
|
33
|
Meshkini E, Aminpour A, Hazrati Tappeh K, Seyyedi S, Shokri M. Evaluation of Adjuvant Effectiveness of Alum-Propranolol Mixture on the Immunogenicity of Excreted/Secreted Antigens of Toxoplasma gondii RH Strain. Adv Pharm Bull 2021; 11:570-577. [PMID: 34513633 PMCID: PMC8421635 DOI: 10.34172/apb.2021.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/09/2020] [Accepted: 07/07/2020] [Indexed: 11/09/2022] Open
Abstract
Purpose: The introduction of novel adjuvants is an important step in attempts to develop a safe and more efficient vaccine. The present study was performed to determine whether the use of a mixed beta-adrenergic receptor antagonist propranolol (PRP) and aluminum (alum), as an adjuvant, have efficacy for Toxoplasma gondii vaccine to induce protective immunity in a mouse model. Methods: Female BALB/c mice divided into five groups were immunized with excretorys-ecretory antigens (ESA) vaccine, alum-ESA vaccine, PRP-ESA vaccine, and alum-PRP ESA vaccine, as well as with phosphate buffered saline (PBS), as a negative control group. The immune responses were evaluated by lymphocyte proliferation assay for measuring delayedtype hypersensitivity (DTH) response and by cytokine assay for evaluating IFN-γ and IL-5 levels. The survival rate of mice in all groups was assessed during a three-week monitoring period after an intraperitoneal challenge with T. gondii tachyzoites. Results: The results showed that mice immunized with PRP, as an adjuvant, could secret a higher level of IFN-γ, which was significant in comparison to other groups. However, mice vaccinated with alum-precipitated ESA antigen had ability to produce an elevated level of IL-5 compared to other mouse groups (P ≤ 0.05). Moreover, alum-PRP co-administration together with ESA vaccine resulted in the longer survival of mice. Conclusion: The findings of this study revealed that the combination of alum-PRP adjuvants and ESA vaccine of T. gondii elicits both humoral and cellular immune responses, which are comparable to either alum or PRP alone.
Collapse
Affiliation(s)
- Elyar Meshkini
- Department of Parasitology & Mycology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Arash Aminpour
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Parasitology & Mycology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Khosrow Hazrati Tappeh
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Parasitology & Mycology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahram Seyyedi
- Department of Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Meysam Shokri
- Department of Parasitology & Mycology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
34
|
Shardlow E, Linhart C, Connor S, Softely E, Exley C. The measurement and full statistical analysis including Bayesian methods of the aluminium content of infant vaccines. J Trace Elem Med Biol 2021; 66:126762. [PMID: 33887692 DOI: 10.1016/j.jtemb.2021.126762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Aluminium salts are the most common adjuvants in infant vaccines. The aluminium content of a vaccine is provided by the manufacturer and is indicated on the patient information leaflet. There is no independent verification, for example by the European Medicines Agency, of the aluminium content of infant vaccines. METHODS We have measured the aluminium content of thirteen infant vaccines using microwave-assisted acid and peroxide digestion followed by transversely heated graphite furnace atomic absorption spectrometry. Our data are compared with manufacturer's data using full statistical analyses including Bayesian methods. RESULTS We found that only three vaccines contained the amount of aluminium indicated by the manufacturer. Six vaccines contained a statistically significant (P < 0.05) greater quantity while four vaccines contained a statistically significant (P < 0.05) lower quantity. The range of content for any single vaccine varied considerably, for example, from 0.172 to 0.602 mg/vaccine for Havrix. CONCLUSIONS The data have raised specific questions about the significance of the aluminium content of vaccines and identified areas of extremely limited information. Since aluminium is a known toxin in humans and specifically a neurotoxin, its content in vaccines should be accurate and independently monitored to ensure both efficacy and safety.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom
| | - Caroline Linhart
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom
| | - Sameerah Connor
- Life Sciences, The Huxley Building, Keele University, Staffordshire, United Kingdom
| | - Erin Softely
- Life Sciences, The Huxley Building, Keele University, Staffordshire, United Kingdom
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom.
| |
Collapse
|
35
|
Targeting Antigens for Universal Influenza Vaccine Development. Viruses 2021; 13:v13060973. [PMID: 34073996 PMCID: PMC8225176 DOI: 10.3390/v13060973] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Traditional influenza vaccines generate strain-specific antibodies which cannot provide protection against divergent influenza virus strains. Further, due to frequent antigenic shifts and drift of influenza viruses, annual reformulation and revaccination are required in order to match circulating strains. Thus, the development of a universal influenza vaccine (UIV) is critical for long-term protection against all seasonal influenza virus strains, as well as to provide protection against a potential pandemic virus. One of the most important strategies in the development of UIVs is the selection of optimal targeting antigens to generate broadly cross-reactive neutralizing antibodies or cross-reactive T cell responses against divergent influenza virus strains. However, each type of target antigen for UIVs has advantages and limitations for the generation of sufficient immune responses against divergent influenza viruses. Herein, we review current strategies and perspectives regarding the use of antigens, including hemagglutinin, neuraminidase, matrix proteins, and internal proteins, for universal influenza vaccine development.
Collapse
|
36
|
Guerrini G, Vivi A, Gioria S, Ponti J, Magrì D, Hoeveler A, Medaglini D, Calzolai L. Physicochemical Characterization Cascade of Nanoadjuvant-Antigen Systems for Improving Vaccines. Vaccines (Basel) 2021; 9:vaccines9060544. [PMID: 34064212 PMCID: PMC8224364 DOI: 10.3390/vaccines9060544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/09/2021] [Accepted: 05/17/2021] [Indexed: 01/25/2023] Open
Abstract
Adjuvants have been used for decades to enhance the immune response to vaccines, in particular for the subunit-based adjuvants. Physicochemical properties of the adjuvant-protein antigen complexes, such as size, morphology, protein structure and binding, influence the overall efficacy and safety of the vaccine. Here we show how to perform an accurate physicochemical characterization of the nanoaluminum-ovalbumin complex. Using a combination of existing techniques, we developed a multi-staged characterization strategy based on measurements of increased complexity. This characterization cascade has the advantage of being very flexible and easily adaptable to any adjuvant-protein antigen combinations. It will contribute to control the quality of antigen-adjuvant complexes and immunological outcomes, ultimately leading to improved vaccines.
Collapse
Affiliation(s)
- Giuditta Guerrini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.G.); (A.V.); (D.M.)
| | - Antonio Vivi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.G.); (A.V.); (D.M.)
| | - Sabrina Gioria
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.G.); (J.P.); (D.M.); (A.H.)
| | - Jessica Ponti
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.G.); (J.P.); (D.M.); (A.H.)
| | - Davide Magrì
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.G.); (J.P.); (D.M.); (A.H.)
| | - Arnd Hoeveler
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.G.); (J.P.); (D.M.); (A.H.)
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.G.); (A.V.); (D.M.)
| | - Luigi Calzolai
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.G.); (J.P.); (D.M.); (A.H.)
- Correspondence:
| |
Collapse
|
37
|
de Sá-Rocha LC, Demarchi LMMF, Postol E, Sampaio RO, de Alencar RE, Kalil J, Guilherme L. StreptInCor, a Group A Streptococcal Adsorbed Vaccine: Evaluation of Repeated Intramuscular Dose Toxicity Testing in Rats. Front Cardiovasc Med 2021; 8:643317. [PMID: 34046438 PMCID: PMC8144318 DOI: 10.3389/fcvm.2021.643317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/22/2021] [Indexed: 01/18/2023] Open
Abstract
Streptococcus pyogenes infections continue to be a worldwide public health problem, causing various diseases in humans, with rheumatic fever and rheumatic heart disease being the most harmful manifestations. Impetigo and post-streptococcal glomerulonephritis are also important sequelae of skin infections. We have developed a candidate vaccine epitope (StreptInCor) that presents promising results in diverse animal models. To assess whether the StreptInCor alum-adsorbed vaccine could induce undesirable effects, a certified independent company conducted a repeated intramuscular dose toxicity evaluation in Wistar rats, a choice model for toxicity studies. We did not observe significant alterations in clinical, hematological, biochemical, anatomical, or histopathological parameters due to vaccine administration, even when the animals received the highest dose. In conclusion, repeated intramuscular doses did not show signs of macroscopic or other significant changes in the clinical or histopathological parameters, indicating that StreptInCor can be considered a safe candidate vaccine.
Collapse
Affiliation(s)
- Luiz Carlos de Sá-Rocha
- Neuroimmunology Laboratory School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Edilberto Postol
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Immunology Investigation Institute, National Institute for Science and Technology, University of São Paulo, São Paulo, Brazil
| | - Roney Orismar Sampaio
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Raquel Elaine de Alencar
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Immunology Investigation Institute, National Institute for Science and Technology, University of São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Immunology Investigation Institute, National Institute for Science and Technology, University of São Paulo, São Paulo, Brazil
| | - Luiza Guilherme
- Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Immunology Investigation Institute, National Institute for Science and Technology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Negm II, Ragab YM, Mohamed AF. Outer membrane proteins of Salmonella typhimurium as an adjuvant in rabies vaccine. Clin Exp Vaccine Res 2021; 10:132-140. [PMID: 34222125 PMCID: PMC8217580 DOI: 10.7774/cevr.2021.10.2.132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/26/2021] [Indexed: 11/15/2022] Open
Abstract
PURPOSE The objective of the present study was to evaluate the immune-enhancing potential of Salmonella typhimurium outer membrane protein (OMP) and alum as adjuvants towards inactivated Vero cells rabies vaccine (FRV/K2). MATERIALS AND METHODS Six groups of female Sprague Dawley albino rats (10/group) were used in the evaluation of immunogenicity and safety of vaccines and adjuvants. Total immunoglobulin G secreted interferon-gamma (IFN-γ), and the percentage of proliferated CD4+ and CD8+ T cells were measured. Biochemical analysis and histopathological examination were used to test safety profiles. RESULTS OMP adjuvanted rabies vaccine (FRV/K2+OMP) (OMP combined locally prepared vaccine) induced significantly higher neutralizing antibodies on day 21 post-vaccination relative to free (FRV/K2) vaccine and alum adsorbed vaccine (FRV/K2+alum) (alum adsorbed locally prepared vaccine). (FRV/K2+OMP) induced a significantly higher level of IFN-γ on day 14 post-vaccination. CD8+ T cells were significantly higher post-vaccination with reference (RV), free (FRV/K2), and (FRV/K2+OMP) than (FRV/K2+alum). On the contrary, CD4+ T cells were significantly elevated post-vaccination with (FRV/K2+alum) at p<0.05. Biochemical analysis and histopathological examination revealed that OMP could be used safely as an adjuvant for the development of more effective rabies vaccines. CONCLUSION Outer membrane proteins adjuvanted rabies vaccines would be beneficial to induce rapid neutralizing antibodies and essential cytokines.
Collapse
Affiliation(s)
- Iman Ibrahim Negm
- The Egyptian Holding Company for the Production of Vaccines, Sera and Drugs, Cairo, Egypt
| | - Yasser M. Ragab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aly Fahmy Mohamed
- International Center for Training and Advanced Researches (ICTAR-Egypt), Cairo, Egypt
| |
Collapse
|
39
|
Pirahmadi S, Zakeri S, Djadid ND, Mehrizi AA. A review of combination adjuvants for malaria vaccines: a promising approach for vaccine development. Int J Parasitol 2021; 51:699-717. [PMID: 33798560 DOI: 10.1016/j.ijpara.2021.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/18/2020] [Accepted: 01/28/2021] [Indexed: 01/16/2023]
Abstract
It is obvious that there is a critical need for an efficient malaria vaccine to accelerate malaria eradication. Currently, recombinant subunit vaccination against malaria using proteins and peptides is gaining attention. However, one of the major drawbacks of this approach is the lack of an efficient and durable immune response. Therefore, subunit vaccines require adjuvants to make the vaccine sufficiently immunogenic. Considering the history of the RTS,S vaccine, it seems likely that no single adjuvant is capable of eliciting all the protective immune responses required in many malarial subunit vaccines and the use of combination adjuvants will be increasingly important as the science of malaria vaccines advances. In light of this, it appears that identifying the most effective mixture of adjuvants with minimal adverse effects offers tremendous opportunities in improving the efficacy of vaccines against malaria. Owing to the importance of a multi-adjuvanted approach in subunit malaria vaccine development, this review paper outlines some of the best known combination adjuvants used in malaria subunit vaccines, focusing on their proposed mechanisms of action, their immunological properties, and their notable results. The aim of the present review is to consolidate these findings to aid the application of these combination adjuvants in experimental malaria vaccines.
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Navid D Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram A Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
40
|
Activation of the NLRP3 Inflammasome by Particles from the Echinococcus granulosus Laminated Layer. Infect Immun 2020; 88:IAI.00190-20. [PMID: 32571988 PMCID: PMC7440765 DOI: 10.1128/iai.00190-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/14/2020] [Indexed: 12/14/2022] Open
Abstract
The interaction of dendritic cells and macrophages with a variety of rigid noncellular particles triggers activation of the NLRP3 inflammasome and consequent secretion of interleukin 1β (IL-1β). Noncellular particles can also be generated in the context of helminth infection, since these large pathogens often shed their outermost structures during growth and/or molting. One such structure is the massive, mucin-based, soft, flexible laminated layer (LL), which protects the larval stages of cestodes of the genus Echinococcus. The interaction of dendritic cells and macrophages with a variety of rigid noncellular particles triggers activation of the NLRP3 inflammasome and consequent secretion of interleukin 1β (IL-1β). Noncellular particles can also be generated in the context of helminth infection, since these large pathogens often shed their outermost structures during growth and/or molting. One such structure is the massive, mucin-based, soft, flexible laminated layer (LL), which protects the larval stages of cestodes of the genus Echinococcus. We show that particles from the Echinococcus granulosus LL (pLL) trigger NLRP3- and caspase-1-dependent IL-1β in lipopolysaccharide (LPS)-primed mouse bone marrow-derived dendritic cells (BMDC). This response can be elicited by pLL too large for phagocytosis and nonetheless requires actin dynamics, Syk, and phosphatidylinositol 3-kinase (PI3K). These three requirements had already been observed in our previous study on the alteration by pLL of CD86, CD40, IL-10, and IL-12 responses to LPS in BMDC; however, we now show that these alterations are independent of NLRP3 and caspase-1. In other words, an initial interaction with particles requiring actin dynamics, Syk, and PI3K, but not phagocytosis, elicits both NLRP3-dependent and NLRP3-independent responses. Intraperitoneal injection of pLL induced IL-1β, suggesting that contact with LL materials induces IL-1β in the E. granulosus infection setting. Our results extend our understanding of NLRP3 inflammasome activation by noncellular particulate materials both to helminth-derived materials and to flexible/soft materials.
Collapse
|
41
|
Co-delivery of antigen and dual adjuvants by aluminum hydroxide nanoparticles for enhanced immune responses. J Control Release 2020; 326:120-130. [PMID: 32585230 DOI: 10.1016/j.jconrel.2020.06.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022]
Abstract
Adjuvants that contain pathogen-associated molecular patterns (PAMPs) can enhance vaccination efficacy by binding to pattern recognition receptors (PRRs), thereby stimulating immune responses. Particularly effective may be the combination of multiple PAMPs that activate different PRRs, which occurs with natural pathogens. Here we hypothesized the enhanced effects would occur in two adjuvants that stimulate different PRRs: CpG oligodeoxynucleotide (CpG-ODN), which is Toll-like receptor 9 agonist; and 5'-triphosphate, short, double-stranded RNA (3pRNA), which activates retinoic acid-inducible gene I (RIG-I). The model antigen ovalbumin (OVA) was loaded and adjuvants were surface-adsorbed to aluminum hydroxide nanoparticles (hereafter NP-3pRNA-CpG) by electrostatic interaction with an average size of 120 nm and a negative surface charge for targeting lymph nodes. These nanoparticles were efficiently internalized by antigen-presenting cells (APCs) in the lymph nodes, and the resulting APC activation and antigen cross-presentation generated strong humoral immunity and cytotoxic T lymphocyte responses and IFN-γ secretion. NP-3pRNA-CpG significantly suppressed B16-OVA tumor growth and prolonged survival of tumor-bearing mice in therapeutic and prophylactic models, illustrating the enhanced effects of CpG-ODN and 3pRNA. Our study highlights the potential of combining multiple adjuvants for effective vaccine design.
Collapse
|
42
|
Nguyen QT, Kim E, Yang J, Lee C, Ha DH, Lee CG, Lee YR, Poo H. E. coli-Produced Monophosphoryl Lipid a Significantly Enhances Protective Immunity of Pandemic H1N1 Vaccine. Vaccines (Basel) 2020; 8:vaccines8020306. [PMID: 32560094 PMCID: PMC7350214 DOI: 10.3390/vaccines8020306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 11/16/2022] Open
Abstract
Emerging influenza viruses pose an extreme global risk to human health, resulting in an urgent need for effective vaccination against influenza infection. Adjuvants are vital components that can improve vaccine efficacy, yet only a few adjuvants have been licensed in human vaccines. Here, we investigate the adjuvant effects of Escherichia coli-produced monophosphoryl lipid A (MPL), named EcML, in enhancing the immunogenicity and efficacy of an influenza vaccine. Similar to MPL, EcML activated dendritic cells and enhanced the antigen processing of cells in vitro. Using ovalbumin (OVA) as a model antigen, EcML increased OVA-specific antibody production, cytotoxic T lymphocyte activity. The safety of EcML was demonstrated as being similar to that of MPL by showing not significant in vitro cell cytotoxicity but transient systemic inflammatory responses within 24 h in OVA immunized mice. Importantly, mice vaccinated with pandemic H1N1 (pH1N1) vaccine antigen, combined with EcML, were fully protected from pH1N1 virus infection by enhanced influenza-specific antibody titers, hemagglutination inhibition titers, and IFN-γ- secreting cells. Taken together, our results strongly suggest that EcML might be a promising vaccine adjuvant for preventing influenza virus infection.
Collapse
Affiliation(s)
- Quyen Thi Nguyen
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (Q.T.N.); (E.K.); (J.Y.)
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon 34113, Korea
| | - Eunjin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (Q.T.N.); (E.K.); (J.Y.)
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Jihyun Yang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (Q.T.N.); (E.K.); (J.Y.)
| | - Chankyu Lee
- Eubiologics. Co., Ltd., V Plant, Gangwon-do 24410, Korea; (C.L.); (D.H.H.); (C.G.L.); (Y.R.L.)
| | - Da Hui Ha
- Eubiologics. Co., Ltd., V Plant, Gangwon-do 24410, Korea; (C.L.); (D.H.H.); (C.G.L.); (Y.R.L.)
| | - Choon Geun Lee
- Eubiologics. Co., Ltd., V Plant, Gangwon-do 24410, Korea; (C.L.); (D.H.H.); (C.G.L.); (Y.R.L.)
| | - Ye Ram Lee
- Eubiologics. Co., Ltd., V Plant, Gangwon-do 24410, Korea; (C.L.); (D.H.H.); (C.G.L.); (Y.R.L.)
| | - Haryoung Poo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (Q.T.N.); (E.K.); (J.Y.)
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: ; Tel.: +82-42-860-4157
| |
Collapse
|
43
|
Collins MK, Shotland AM, Wade MF, Atif SM, Richards DK, Torres-Llompart M, Mack DG, Martin AK, Fontenot AP, McKee AS. A role for TNF-α in alveolar macrophage damage-associated molecular pattern release. JCI Insight 2020; 5:134356. [PMID: 32255768 DOI: 10.1172/jci.insight.134356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/01/2020] [Indexed: 01/22/2023] Open
Abstract
Chronic beryllium disease (CBD) is a metal hypersensitivity/autoimmune disease in which damage-associated molecular patterns (DAMPs) promote a break in T cell tolerance and expansion of Be2+/self-peptide-reactive CD4+ T cells. In this study, we investigated the mechanism of cell death induced by beryllium particles in alveolar macrophages (AMs) and its impact on DAMP release. We found that phagocytosis of Be led to AM cell death independent of caspase, receptor-interacting protein kinases 1 and 3, or ROS activity. Before cell death, Be-exposed AMs secreted TNF-α that boosted intracellular stores of IL-1α followed by caspase-8-dependent fragmentation of DNA. IL-1α and nucleosomal DNA were subsequently released from AMs upon loss of plasma membrane integrity. In contrast, necrotic AMs released only unfragmented DNA and necroptotic AMs released only IL-1α. In mice exposed to Be, TNF-α promoted release of DAMPs and was required for the mobilization of immunogenic DCs, the expansion of Be-reactive CD4+ T cells, and pulmonary inflammation in a mouse model of CBD. Thus, early autocrine effects of particle-induced TNF-α on AMs led to a break in peripheral tolerance. This potentially novel mechanism may underlie the known relationship between fine particle inhalation, TNF-α, and loss of peripheral tolerance in T cell-mediated autoimmune disease and hypersensitivities.
Collapse
Affiliation(s)
- Morgan K Collins
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine
| | - Abigail M Shotland
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine
| | - Morgan F Wade
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine
| | - Shaikh M Atif
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine
| | | | | | - Douglas G Mack
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine
| | - Allison K Martin
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine
| | - Andrew P Fontenot
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine.,Department of Immunology and Microbiology, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amy S McKee
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine.,Department of Immunology and Microbiology, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
44
|
Leal VNC, Reis EC, Pontillo A. Inflammasome in HIV infection: Lights and shadows. Mol Immunol 2019; 118:9-18. [PMID: 31835091 DOI: 10.1016/j.molimm.2019.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/27/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
The importance of inflammasome, and related cytokines IL-1ß and IL-18, in host defense against pathogens is well documented, however, at the same time, dysregulation of inflammasome has been associated to multifactorial diseases characterized by chronic inflammation (i.e.: metabolic disorders, cardiovascular diseases, neurodegenerative diseases, autoimmunity, cancer). Inflammasome activation has been described in response to HIV-1 and possibly contributes to the resistance against virus establishment, however, on the other hand, when viral infection becomes chronic, independently from antiretroviral therapy, the increase constitutive activation of inflammasome has been eventually associated to a worse prognosis, raising the question about the role played by inflammasome and/or some specific receptors in this context. Due to the chance to imply targeted therapies that inhibit inflammasome activation and/or cytokines release, it will be important to define the impact of the complex in the pathogenesis of HIV. The purpose of this review is to depict the double-faced inflammasome role in HIV-1 infection, trying to unveil whether besides its role in first line defense against the virus, it exerts a harmful effect during the chronic phase of infection.
Collapse
Affiliation(s)
- Vinicius Nunes Cordeiro Leal
- Laboratorio de Imunogenetica, Departamento de Imunologia, Instituto de Ciencias Biomedicas (ICB), Universidade de Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Edione Cristina Reis
- Laboratorio de Imunogenetica, Departamento de Imunologia, Instituto de Ciencias Biomedicas (ICB), Universidade de Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Alessandra Pontillo
- Laboratorio de Imunogenetica, Departamento de Imunologia, Instituto de Ciencias Biomedicas (ICB), Universidade de Sao Paulo (USP), Sao Paulo, SP, Brazil.
| |
Collapse
|
45
|
Abstract
Metals are essential components in all forms of life required for the function of nearly half of all enzymes and are critically involved in virtually all fundamental biological processes. Especially, the transition metals iron (Fe), zinc (Zn), manganese (Mn), nickel (Ni), copper (Cu) and cobalt (Co) are crucial micronutrients known to play vital roles in metabolism as well due to their unique redox properties. Metals carry out three major functions within metalloproteins: to provide structural support, to serve as enzymatic cofactors, and to mediate electron transportation. Metal ions are also involved in the immune system from metal allergies to nutritional immunity. Within the past decade, much attention has been drawn to the roles of metal ions in the immune system, since increasing evidence has mounted to suggest that metals are critically implicated in regulating both the innate immune sensing of and the host defense against invading pathogens. The importance of ions in immunity is also evidenced by the identification of various immunodeficiencies in patients with mutations in ion channels and transporters. In addition, cancer immunotherapy has recently been conclusively demonstrated to be effective and important for future tumor treatment, although only a small percentage of cancer patients respond to immunotherapy because of inadequate immune activation. Importantly, metal ion-activated immunotherapy is becoming an effective and potential way in tumor therapy for better clinical application. Nevertheless, we are still in a primary stage of discovering the diverse immunological functions of ions and mechanistically understanding the roles of these ions in immune regulation. This review summarizes recent advances in the understanding of metal-controlled immunity. Particular emphasis is put on the mechanisms of innate immune stimulation and T cell activation by the essential metal ions like calcium (Ca2+), zinc (Zn2+), manganese (Mn2+), iron (Fe2+/Fe3+), and potassium (K+), followed by a few unessential metals, in order to draw a general diagram of metalloimmunology.
Collapse
Affiliation(s)
- Chenguang Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Rui Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xiaoming Wei
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Mengze Lv
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
46
|
Shardlow E, Mold M, Exley C. The interaction of aluminium-based adjuvants with THP-1 macrophages in vitro: Implications for cellular survival and systemic translocation. J Inorg Biochem 2019; 203:110915. [PMID: 31751817 DOI: 10.1016/j.jinorgbio.2019.110915] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023]
Abstract
Within clinical vaccinations, recombinant antigens are routinely entrapped inside or adsorbed onto the surface of aluminium salts in order to increase their immunological potency in vivo. The efficacy of these immunisations is highly dependent upon the recognition and uptake of these complexes by professional phagocytes and their subsequent delivery to the draining lymph nodes for further immunological processing. While monocytes have been shown to internalise aluminium adjuvants and their adsorbates, the role of macrophages in this respect has not been fully established. Furthermore, this study explored the interaction of THP-1 macrophages with aluminium-based adjuvants (ABAs) and how this relationship influenced the survival of such cells in vitro. THP-1 macrophages were exposed to low concentrations of ABAs (1.7 μg/mL Al) for a maximum of seven days. ABA uptake was determined using lumogallion staining and cell viability by both DAPI (4',6-diamidino-2-phenylindole) staining and LDH (lactate dehydrogenase) assay. Evidence of ABA particle loading was identified within cells at early junctures following treatment and appeared to be quite prolific (>90% cells positive for Al signal after 24 h). Total sample viability (% LDH release) in treated samples was predominantly similar to untreated cells and low levels of cellular death were consistently observed in populations positive for Al uptake. It can thus be concluded that aluminium salts can persist for some time within the intracellular environment of these cells without adversely affecting their viability. These results imply that macrophages may play a role in the systemic translocation of ABAs once administered in the form of an inoculation.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK
| | - Matthew Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG, UK.
| |
Collapse
|
47
|
Zhong H, Zhao C, Luo S. HLA in myasthenia gravis: From superficial correlation to underlying mechanism. Autoimmun Rev 2019; 18:102349. [DOI: 10.1016/j.autrev.2019.102349] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 03/01/2019] [Indexed: 12/26/2022]
|
48
|
Vrieling H, Kooijman S, de Ridder JW, Thies-Weesie DME, Soema PC, Jiskoot W, van Riet E, Heck AJR, Philipse AP, Kersten GFA, Meiring HD, Pennings JL, Metz B. Activation of Human Monocytes by Colloidal Aluminum Salts. J Pharm Sci 2019; 109:750-760. [PMID: 31449816 DOI: 10.1016/j.xphs.2019.08.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/10/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023]
Abstract
Subunit vaccines often contain colloidal aluminum salt-based adjuvants to activate the innate immune system. These aluminum salts consist of micrometer-sized aggregates. It is well-known that particle size affects the adjuvant effect of particulate adjuvants. In this study, the activation of human monocytes by hexagonal-shaped gibbsite (ø = 210 ± 40 nm) and rod-shaped boehmite (ø = 83 ± 827 nm) was compared with classical aluminum oxyhydroxide adjuvant (alum). To this end, human primary monocytes were cultured in the presence of alum, gibbsite, or boehmite. The transcriptome and proteome of the monocytes were investigated by using quantitative polymerase chain reaction and mass spectrometry. Human monocytic THP-1 cells were used to investigate the effect of the particles on cellular maturation, differentiation, activation, and cytokine secretion, as measured by flow cytometry and enzyme-linked immunosorbent assay. Each particle type resulted in a specific gene expression profile. IL-1ß and IL-6 secretion was significantly upregulated by boehmite and alum. Of the 7 surface markers investigated, only CD80 was significantly upregulated by alum and none by gibbsite or boehmite. Gibbsite hardly activated the monocytes. Boehmite activated human primary monocytes equally to alum, but induced a much milder stress-related response. Therefore, boehmite was identified as a promising adjuvant candidate.
Collapse
Affiliation(s)
- Hilde Vrieling
- Intravacc (Institute for Translational Vaccinology), Bilthoven, the Netherlands; Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Sietske Kooijman
- Intravacc (Institute for Translational Vaccinology), Bilthoven, the Netherlands; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, the Netherlands
| | - Justin W de Ridder
- Intravacc (Institute for Translational Vaccinology), Bilthoven, the Netherlands
| | - Dominique M E Thies-Weesie
- Van 't Hoff Laboratory for Physical and Colloid Chemistry, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, the Netherlands
| | - Peter C Soema
- Intravacc (Institute for Translational Vaccinology), Bilthoven, the Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Elly van Riet
- Intravacc (Institute for Translational Vaccinology), Bilthoven, the Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Centre, Utrecht, the Netherlands
| | - Albert P Philipse
- Van 't Hoff Laboratory for Physical and Colloid Chemistry, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, the Netherlands
| | - Gideon F A Kersten
- Intravacc (Institute for Translational Vaccinology), Bilthoven, the Netherlands; Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, the Netherlands
| | - Hugo D Meiring
- Intravacc (Institute for Translational Vaccinology), Bilthoven, the Netherlands
| | - Jeroen L Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Bernard Metz
- Intravacc (Institute for Translational Vaccinology), Bilthoven, the Netherlands.
| |
Collapse
|
49
|
Entwicklung der subkutanen Allergen-Immuntherapie (Teil 2): präventive Aspekte der SCIT und Innovationen. ALLERGO JOURNAL 2019. [DOI: 10.1007/s15007-019-1847-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
50
|
Kumar S, Sunagar R, Gosselin E. Bacterial Protein Toll-Like-Receptor Agonists: A Novel Perspective on Vaccine Adjuvants. Front Immunol 2019; 10:1144. [PMID: 31191528 PMCID: PMC6549121 DOI: 10.3389/fimmu.2019.01144] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
Adjuvants have been used in vaccines for over a century, however, the search for safe and effective vaccine adjuvants continues. In recent decades toll-like-receptor (TLR) agonists have been investigated as potential vaccine adjuvants. In this regard, the majority of the currently investigated TLR agonists are non-protein microbial components such as lipopolysaccharides, oligonucleotides, and lipopeptides. On the other hand, a growing number of studies reveal that TLR signaling and immune responses can be activated by numerous bacterial proteins. However, their potential roles as adjuvants have been somewhat overlooked. Herein, we discuss several such bacterial proteins which exhibit adjuvant properties, including the activation of TLR signaling, antigen presenting cell maturation, pro-inflammatory cytokine production and adaptive immune response. The protein nature of these TLR agonists presents several unique features not shared by non-protein TLR agonists. These properties include the amenability for modifying the structure and function as necessary for optimal immunogenicity and minimal toxicity. Protein adjuvants can be genetically fused to protein antigens which ensure the co-delivery of adjuvant-antigen not only into the same cell but also in the same endocytic cargo, leading to more effective activation of innate and adaptive immune response.
Collapse
Affiliation(s)
- Sudeep Kumar
- Department of Immunology and Microbial Diseases, Albany Medical College, Albany, NY, United States
| | - Raju Sunagar
- Ella Foundation, Genome Valley, Hyderabad, India
| | - Edmund Gosselin
- Department of Immunology and Microbial Diseases, Albany Medical College, Albany, NY, United States
| |
Collapse
|