1
|
Shah BA, Holden JA, Lenzo JC, Hadjigol S, O'Brien-Simpson NM. Multi-disciplinary approaches paving the way for clinically effective peptide vaccines for cancer. NPJ Vaccines 2025; 10:68. [PMID: 40204832 PMCID: PMC11982186 DOI: 10.1038/s41541-025-01118-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Cytotoxic CD8+ T lymphocyte (CTL) cells are central in mediating antitumor immunity. Induction of a robust CTL response requires, CTL interaction with professional antigen-presenting cells, such as dendritic cells, displaying onco-antigenic peptide, often derived from tumor-associated antigens (TAAs) or neoantigens, and costimulation via CD4+ T helper cells which then elicits an effector and memory immune response that targets and kills cancer cells. Despite the tumoricidal capacity of CTLs, cancer cells can escape immune surveillance and killing due to their immunosuppressive tumor microenvironment (TME). Therefore, to harness the CTL immune response and combat the effect of the TME, peptide-based T cell vaccines targeting specific onco-antigens, conjugated with adjuvants are a subject of ongoing research for cancer immunotherapy; particularly, multi-peptide vaccines, containing both CTL and CD4+ T helper cell epitopes along with an immunostimulant. Historically, peptide-based T cell vaccines have been investigated as a potential strategy for cancer immunotherapy. Despite initial enthusiasm, these peptide vaccines have not demonstrated success in clinical outcomes. However, recent advancements in our understanding of cancer immunology and the design of peptide vaccines targeting specific tumor antigens have paved the way for novel strategies in peptide-based immunotherapy. These advancements have reignited optimism surrounding the potential of peptide-based vaccines as a viable cancer therapeutic. This review explores the new strategies and discusses the exciting possibilities they offer. Specifically, this review develops an understanding of vaccine design and clinical outcomes, by discussing mechanisms of CTL effector and memory responses, and how peptide-based vaccines can induce and enhance these responses. It addresses the challenge of Major Histocompatibility Complex (MHC) restriction, which limits the effectiveness of traditional peptide vaccines in individuals with diverse MHC types. It also delves into the immunosuppressive tumor microenvironment and overcoming its inhibitory effects using peptide-based vaccines for efficient cancer cell elimination. The review aims to provide an understanding of the complexities faced by each field in vaccine design, enhancing dialogue and understanding among researchers by bringing together the chemistry of vaccine synthesis, cancer immunology, and clinical studies to support the development of a peptide-based vaccine.
Collapse
Affiliation(s)
- Bansari A Shah
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital, and The Graeme Clark Institute, The University of Melbourne, Carlton, VIC, Australia
| | - James A Holden
- Centre for Oral Health Research, Melbourne Dental School, Royal Dental Hospital, The University of Melbourn, Carlton, VIC, Australia
| | - Jason C Lenzo
- Western Australian Health Translation Network, Harry Perkins Institute of Medical Research, Level 6, Nedlands, Perth, WA, Australia
| | - Sara Hadjigol
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital, and The Graeme Clark Institute, The University of Melbourne, Carlton, VIC, Australia.
| | - Neil M O'Brien-Simpson
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital, and The Graeme Clark Institute, The University of Melbourne, Carlton, VIC, Australia.
| |
Collapse
|
2
|
Gharaei A, Rahdar M, Jorjani O, Saberi S, Beiromvand M, Feiz-Haddad MH. Evaluation of the immunogenicity of a DNA vaccine for Leishmania major based on the Leishmania-activated C kinase antigen using calcium phosphate and chitosan adjuvants. Trans R Soc Trop Med Hyg 2025; 119:266-273. [PMID: 39749969 DOI: 10.1093/trstmh/trae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 09/23/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Leishmaniasis represents a significant parasitic disease with global health implications, and the development of an affordable and effective vaccine could provide a valuable solution. This study aimed to evaluate the immunogenicity of a DNA vaccine targeting Leishmania major specifically based on the Leishmania-activated C kinase (LACK) antigen, utilizing calcium phosphate nanoparticles (CaPNs) and chitosan nanoparticles (ChitNs) as adjuvants. METHODS Seventy female BALB/c mice, aged 4-6 wk and weighing 20-22 g, were selected and divided into five groups, each consisting of 14 mice. The first group received the plasmid LACK vaccine (pcDN3+LACK), the second group received the pcDN3+LACK vaccine with the CaPN adjuvant (pcDN3+LACK+CaPN), the third group received the pcDN3+LACK vaccine with the ChitN adjuvant (pcDN3+LACK+ChitN), the fourth group was administered phosphate-buffered saline as a negative control and the fifth group did not receive any vaccine, serving as a positive control. The vaccination program involved two intramuscular injections at 3-wk intervals. Three weeks following the final vaccination, the mice were challenged with wild-type L. major promastigotes via intradermal injection at the base of their tails. Clinical signs and lesion sizes were evaluated biweekly using Vernier calipers. Immune responses, including levels of interferon-gamma (IFN-γ) and interleukin-4 (IL-4), were assessed using ELISA. RESULTS The groups receiving pcDN3+LACK+ChitN, pcDN3+LACK+CaPN and pcDN3+LACK exhibited the highest increases in IFN-γ titers and the most significant reductions in IL-4 titers. Furthermore, lesion sizes associated with Leishmania infection were reduced in the vaccinated groups, with the most favorable outcomes observed in the pcDN3+LACK+ChitN group. CONCLUSIONS These findings suggest that vaccination utilizing the LACK antigen in conjunction with CaPN and ChitN adjuvants may represent an effective strategy for the control of cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Abdolaziz Gharaei
- Infecti ous and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 15794-61357, Iran
- Department of Medical Parasitology, Medical school, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Mahmoud Rahdar
- Infecti ous and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 15794-61357, Iran
- Department of Medical Parasitology, Medical school, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Oghlniaz Jorjani
- Department of Medical Parasitology, Medical school, Golestan University of Medical Sciences, Gorgan 4918936316, Iran
| | - Sedigheh Saberi
- Department of Medical Parasitology, Medical school, Isfahan University of Medical Sciences, Isfahan 8174623461, Iran
| | - Molouk Beiromvand
- Department of Medical Parasitology, Medical school, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Mohammad Hossein Feiz-Haddad
- Department of Medical Parasitology, Medical school, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| |
Collapse
|
3
|
Demir C, Bakırdere BE, Zaman BT, Öner M, Bozyiğit GD, Ergenler A, Turan F, Nejati O, Öztürk AB, Çetin G, Bakırdere S. Synthesis of calcium phosphate nanomaterial from quail eggshell for cadmium removal from wastewater and its genotoxic/cytotoxic properties. ENVIRONMENTAL MONITORING AND ASSESSMENT 2024; 196:1214. [PMID: 39557696 DOI: 10.1007/s10661-024-13415-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
In recent years, pollutants released into the environment from various sources threaten environmental health. Rapid industrialization and the constantly increasing needs of people facilitate the release of more hazardous wastes into the ecosystem. The presence of pollutants in water resources causes a wide range of adverse effects. In this study, calcium phosphate nanomaterial (Ca3(PO4)2 NM) was synthesized from biological waste eggshells for the cadmium removal in synthetic domestic wastewater, and a treatment method was developed using these NMs. The Ca3(PO4)2 NMs were produced by using a biowaste which provides the synthesis procedure greener approach. The biogenic NMs were used to remove toxic cadmium ions from wastewater samples. Cytotoxicity and genotoxicity studies of the synthesized NMs were also carried out, and their possible effects on the health of living organisms and the ecology were examined. In the developed method, the parameters affecting the removal of cadmium from wastewater samples were optimized and the removal efficiency was calculated by determining cadmium in a flame atomic absorption spectrophotometer system (FAAS). Synthetic domestic wastewater samples were utilized for evaluating the applicability of the developed treatment strategy. In addition, the adsorption capacity of the material for Cd2+ ion was calculated and the values obtained were modeled by using Langmuir adsorption isotherm (LAI). The calculated LAI parameters were within the appropriate limits, which proved that the developed NM can be used as an effective material for cadmium removal. Moreover, a new, rapid, and feasible synthesis strategy for the synthesis of Ca3(PO4)2 NM was presented in the literature.
Collapse
Affiliation(s)
- Cansu Demir
- Department of Chemistry, Yildiz Technical University, 34220, Istanbul, Türkiye
- Neutec Pharmaceutical, Yildiz Technical University Teknopark, 34220, Istanbul, Türkiye
| | - Bengisu Ece Bakırdere
- Galatasaray High School, Kuloğlu Neighborhood, İstiklal Street No: 159, 34430, Beyoğlu, Istanbul, Türkiye
| | - Buse Tuğba Zaman
- Department of Chemistry, Yildiz Technical University, 34220, Istanbul, Türkiye
| | - Miray Öner
- Department of Chemistry, Yildiz Technical University, 34220, Istanbul, Türkiye
- Neutec Pharmaceutical, Yildiz Technical University Teknopark, 34220, Istanbul, Türkiye
| | - Gamze Dalgıç Bozyiğit
- Department of Environmental Engineering, Yildiz Technical University, 34220, Istanbul, Türkiye
| | - Ayşegül Ergenler
- Department of Marine Science and Technology, İskenderun Technical University, 31200, İskenderun, Hatay, Türkiye
| | - Funda Turan
- Department of Marine Science and Technology, İskenderun Technical University, 31200, İskenderun, Hatay, Türkiye
| | - Omid Nejati
- Department of Stem Cell and Tissue Engineering, İstinye University, 34010, Zeytinburnu, Istanbul, Türkiye
| | - Ayça Bal Öztürk
- Faculty of Pharmacy, İstinye University, 34010, Zeytinburnu, Istanbul, Türkiye
| | - Gülten Çetin
- Department of Chemistry, Yildiz Technical University, 34220, Istanbul, Türkiye
| | - Sezgin Bakırdere
- Department of Chemistry, Yildiz Technical University, 34220, Istanbul, Türkiye.
- Turkish Academy of Sciences (TÜBA), Vedat Dalokay Street, No: 112, Çankaya, 06670, Ankara, Türkiye.
| |
Collapse
|
4
|
Jia Z, Zhou X, Liu J, De X, Li Y, Yang Z, Duan H, Wang F, Ge J. Immune enhancement of rhamnolipid/manganese calcium phosphate mineralized nanoparticle: A promising subunit antigen delivery system. Int J Biol Macromol 2024; 282:137239. [PMID: 39491710 DOI: 10.1016/j.ijbiomac.2024.137239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
The use of biomimetic mineralization strategy is promising to solve the problem of poor stability and immune effect of subunit antigens. However, non-specifically inducing protein mineralization is still a challenge. we hypothesized that rhamnolipids with both protein and metal binding capacity could be used to develop more functional and biocompatible calcium mineralized nanoparticle (RMCP). The results show that rhamnolipids synergistically enhanced the mineralization of protein with manganese ions and improved 21 % the loading antigens of RMCP compared to manganese calcium phosphate nanoparticles. Transmission electron microscopy (TEM) and Dynamic Light Scattering (DLS) showed particle size of RMCP is 260 ± 12.1 nm with spherical morphology. In vitro experiments have shown that RMCP effectively activate immune cells through the cGAS-STING and NLRP3 pathways and demonstrated a higher level of cytokines in RAW264.7 Macrophages. In vivo, RMCP triggered an increased IgG titer with 16.5-fold IgG2a/IgG1 ratio compared to the aluminum adjuvant which improved the recovery status after challenge in mice. We used biological surfactants for the first time to enhance the biomimetic mineralization process of subunit antigen, which provides a new approach for constructing calcium-based biocompatible antigen delivery vectors, helping to develop a new generation of stable, efficient, and safe subunit vaccines.
Collapse
Affiliation(s)
- Zheng Jia
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Xinyao Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Jingjing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China; State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Xinqi De
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Yifan Li
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Zaixing Yang
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Haoyuan Duan
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China
| | - Fang Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150086, China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China; Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin 150036, China.
| |
Collapse
|
5
|
Sun Y, Liu Y, Li R, Zhang C, Wu M, Zhang X, Zheng A, Liao N, Zheng Y, Xu H, Zeng R, Zeng Y, Liu X. Multifunctional Biomimetic Nanocarriers for Dual-Targeted Immuno-Gene Therapy Against Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400951. [PMID: 38973319 PMCID: PMC11425963 DOI: 10.1002/advs.202400951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/22/2024] [Indexed: 07/09/2024]
Abstract
Growing evidences have proved that tumors evade recognition and attack by the immune system through immune escape mechanisms, and PDL1/Pbrm1 genes have a strong correlation with poor response or resistance to immune checkpoint blockade (ICB) therapy. Herein, a multifunctional biomimetic nanocarrier (siRNA-CaP@PD1-NVs) is developed, which can not only enhance the cytotoxic activity of immune cells by blocking PD1/PDL1 axis, but also reduce tumor immune escape via Pbrm1/PDL1 gene silencing, leading to a significant improvement in tumor immunosuppressive microenvironment. Consequently, the nanocarrier promotes DC cell maturation, enhances the infiltration and activity of CD8+ T cells, and forms long-term immune memory, which can effectively inhibit tumor growth or even eliminate tumors, and prevent tumor recurrence and metastasis. Overall, this study presents a powerful strategy for co-delivery of siRNA drugs, immune adjuvant, and immune checkpoint inhibitors, and holds great promise for improving the effectiveness and safety of current immunotherapy regimens.
Collapse
Affiliation(s)
- Yupeng Sun
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Yan Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- College of Biological Science and EngineeringFuzhou UniversityFuzhou350116P. R. China
| | - Rui Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- College of Biological Science and EngineeringFuzhou UniversityFuzhou350116P. R. China
| | - Cuilin Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Xiaolong Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Aixian Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Youshi Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Haipo Xu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Rui Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
- Liver Disease CenterThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350005P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
- CAS Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of Matter Chinese Academy of SciencesFuzhou350002P. R. China
| |
Collapse
|
6
|
Li M, Yao Z, Wang H, Ma Y, Yang W, Guo Y, Yu G, Shi W, Zhang N, Xu M, Li X, Zhao J, Zhang Y, Xue C, Sun B. Silicon or Calcium Doping Coordinates the Immunostimulatory Effects of Aluminum Oxyhydroxide Nanoadjuvants in Prophylactic Vaccines. ACS NANO 2024; 18:16878-16894. [PMID: 38899978 DOI: 10.1021/acsnano.4c02685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Aluminum salts still remain as the most popular adjuvants in marketed human prophylactic vaccines due to their capability to trigger humoral immune responses with a good safety record. However, insufficient induction of cellular immune responses limits their further applications. In this study, we prepare a library of silicon (Si)- or calcium (Ca)-doped aluminum oxyhydroxide (AlOOH) nanoadjuvants. They exhibit well-controlled physicochemical properties, and the dopants are homogeneously distributed in nanoadjuvants. By using Hepatitis B surface antigen (HBsAg) as the model antigen, doped AlOOH nanoadjuvants mediate higher antigen uptake and promote lysosome escape of HBsAg through lysosomal rupture induced by the dissolution of the dopant in the lysosomes in bone marrow-derived dendritic cells (BMDCs). Additionally, doped nanoadjuvants trigger higher antigen accumulation and immune cell activation in draining lymph nodes. In HBsAg and varicella-zoster virus glycoprotein E (gE) vaccination models, doped nanoadjuvants induce high IgG titer, activations of CD4+ and CD8+ T cells, cytotoxic T lymphocytes, and generations of effector memory T cells. Doping of aluminum salt-based adjuvants with biological safety profiles and immunostimulating capability is a potential strategy to mediate robust humoral and cellular immunity. It potentiates the applications of engineered adjuvants in the development of vaccines with coordinated immune responses.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Zhiying Yao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Huiyang Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Yubin Ma
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Wenqi Yang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Yiyang Guo
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Ge Yu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Wendi Shi
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Ning Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Muzhe Xu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Xin Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Jiashu Zhao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Yue Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Changying Xue
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| |
Collapse
|
7
|
Zhang X, Wei M, Zhang Z, Zeng Y, Zou F, Zhang S, Wang Z, Chen F, Xiong H, Li Y, Zhou L, Li T, Zheng Q, Yu H, Zhang J, Gu Y, Zhao Q, Li S, Xia N. Risedronate-functionalized manganese-hydroxyapatite amorphous particles: A potent adjuvant for subunit vaccines and cancer immunotherapy. J Control Release 2024; 367:13-26. [PMID: 38244843 DOI: 10.1016/j.jconrel.2024.01.033] [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: 08/31/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024]
Abstract
The cGAS-STING pathway and the Mevalonate Pathway are druggable targets for vaccine adjuvant discovery. Manganese (Mn) and bisphosphonates are known to exert adjuvant effects by targeting these two pathways, respectively. This study found the synergistic potential of the two pathways in enhancing immune response. Risedronate (Ris) significantly amplified the Mn adjuvant early antibody response by 166-fold and fortified its cellular immunity. However, direct combination of Mn2+ and Ris resulted in increased adjuvant toxicity (40% mouse mortality). By the combination of doping property of hydroxyapatite (HA) and its high affinity for Ris, we designed Ris-functionalized Mn-HA micro-nanoparticles as an organic-inorganic hybrid adjuvant, named MnHARis. MnHARis alleviated adjuvant toxicity (100% vs. 60% survival rate) and exhibited good long-term stability. When formulated with the varicella-zoster virus glycoprotein E (gE) antigen, MnHARis triggered a 274.3-fold increase in IgG titers and a 61.3-fold surge in neutralization titers while maintaining a better long-term humoral immunity compared to the aluminum adjuvant. Its efficacy spanned other antigens, including ovalbumin, HPV18 VLP, and SARS-CoV-2 spike protein. Notably, the cellular immunity elicited by the group of gE + MnHARis was comparable to the renowned Shingrix®. Moreover, intratumoral co-administration with an anti-trophoblast cell surface antigen 2 nanobody revealed synergistic antitumor capabilities. These findings underscore the potential of MnHARis as a potent adjuvant for augmenting vaccine immune responses and improving cancer immunotherapy outcomes.
Collapse
Affiliation(s)
- Xiuli Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Mingjing Wei
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Zhigang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Yarong Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Feihong Zou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Sibo Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Zhiping Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Fentian Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Hualong Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Yufang Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Lizhi Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Tingting Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Qingbing Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Hai Yu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Ying Gu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Collaborative Innovation Center of Biologic Products, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
8
|
Feng Y, Wang J, Cao J, Cao F, Chen X. Manipulating calcium homeostasis with nanoplatforms for enhanced cancer therapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230019. [PMID: 38854493 PMCID: PMC10867402 DOI: 10.1002/exp.20230019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/28/2023] [Indexed: 06/11/2024]
Abstract
Calcium ions (Ca2+) are indispensable and versatile metal ions that play a pivotal role in regulating cell metabolism, encompassing cell survival, proliferation, migration, and gene expression. Aberrant Ca2+ levels are frequently linked to cell dysfunction and a variety of pathological conditions. Therefore, it is essential to maintain Ca2+ homeostasis to coordinate body function. Disrupting the balance of Ca2+ levels has emerged as a potential therapeutic strategy for various diseases, and there has been extensive research on integrating this approach into nanoplatforms. In this review, the current nanoplatforms that regulate Ca2+ homeostasis for cancer therapy are first discussed, including both direct and indirect approaches to manage Ca2+ overload or inhibit Ca2+ signalling. Then, the applications of these nanoplatforms in targeting different cells to regulate their Ca2+ homeostasis for achieving therapeutic effects in cancer treatment are systematically introduced, including tumour cells and immune cells. Finally, perspectives on the further development of nanoplatforms for regulating Ca2+ homeostasis, identifying scientific limitations and future directions for exploitation are offered.
Collapse
Affiliation(s)
- Yanlin Feng
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Jianlin Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Jimin Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Fangfang Cao
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingaporeSingapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Agency for Science, Technology, and Research (A*STAR)Institute of Molecular and Cell BiologySingaporeSingapore
| |
Collapse
|
9
|
Sun B, Li M, Yao Z, Yu G, Ma Y. Advances in Vaccine Adjuvants: Nanomaterials and Small Molecules. Handb Exp Pharmacol 2024; 284:113-132. [PMID: 37059911 DOI: 10.1007/164_2023_652] [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: 04/16/2023]
Abstract
Adjuvants have been extensively and essentially formulated in subunits and certain inactivated vaccines for enhancing and prolonging protective immunity against infections and diseases. According to the types of infectious diseases and the required immunity, adjuvants with various acting mechanisms have been designed and applied in human vaccines. In this chapter, we introduce the advances in vaccine adjuvants based on nanomaterials and small molecules. By reviewing the immune mechanisms induced by adjuvants with different characteristics, we aim to establish structure-activity relationships between the physicochemical properties of adjuvants and their immunostimulating capability for the development of adjuvants for more effective preventative and therapeutic vaccines.
Collapse
Affiliation(s)
- Bingbing Sun
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering and Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, China.
| | - Min Li
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering and Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Zhiying Yao
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering and Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Ge Yu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering and Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Yubin Ma
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering and Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, China
| |
Collapse
|
10
|
Liu S, Hu M, Liu X, Liu X, Chen T, Zhu Y, Liang T, Xiao S, Li P, Ma X. Nanoparticles and Antiviral Vaccines. Vaccines (Basel) 2023; 12:30. [PMID: 38250843 PMCID: PMC10819235 DOI: 10.3390/vaccines12010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Viruses have threatened human lives for decades, causing both chronic and acute infections accompanied by mild to severe symptoms. During the long journey of confrontation, humans have developed intricate immune systems to combat viral infections. In parallel, vaccines are invented and administrated to induce strong protective immunity while generating few adverse effects. With advancements in biochemistry and biophysics, different kinds of vaccines in versatile forms have been utilized to prevent virus infections, although the safety and effectiveness of these vaccines are diverse from each other. In this review, we first listed and described major pathogenic viruses and their pandemics that emerged in the past two centuries. Furthermore, we summarized the distinctive characteristics of different antiviral vaccines and adjuvants. Subsequently, in the main body, we reviewed recent advances of nanoparticles in the development of next-generation vaccines against influenza viruses, coronaviruses, HIV, hepatitis viruses, and many others. Specifically, we described applications of self-assembling protein polymers, virus-like particles, nano-carriers, and nano-adjuvants in antiviral vaccines. We also discussed the therapeutic potential of nanoparticles in developing safe and effective mucosal vaccines. Nanoparticle techniques could be promising platforms for developing broad-spectrum, preventive, or therapeutic antiviral vaccines.
Collapse
Affiliation(s)
- Sen Liu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Meilin Hu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511400, China
| | - Xiaoqing Liu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xingyu Liu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
| | - Tao Chen
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511400, China
| | - Yiqiang Zhu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
| | - Taizhen Liang
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511400, China
| | - Shiqi Xiao
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
| | - Peiwen Li
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
| | - Xiancai Ma
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China; (S.L.); (M.H.); (X.L.); (X.L.); (T.C.); (Y.Z.); (T.L.); (S.X.); (P.L.)
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511400, China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
11
|
Deka NJ, Kalita DJ, Tamuly S, Sharma RK, Bora DP, Dutta R, Hazorika M, Chabukdhara P, George S. Calcium phosphate nanoparticles conjugated with outer membrane vesicle of Riemerella anatipestifer for vaccine development in ducklings. Microb Pathog 2023; 185:106446. [PMID: 37951409 DOI: 10.1016/j.micpath.2023.106446] [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: 08/12/2023] [Revised: 10/14/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Biodegradable calcium phosphate nanoparticles offer a viable substitute for traditional adjuvants such as aluminum in vaccine production. Calcium phosphate nanoparticle adjuvanted with outer membrane vesicle (OMV) of gram negative bacteria may induce efficient immune response in the host. The present study was carried out to evaluate the potential of a mucosal vaccine formulation of calcium phosphate (CAP) nanoparticle using OMV of Riemerella anatipestifer (RA) as antigen against New Duck disease in ducks. The work was initiated with isolation, identification of RA, followed by OMV production and extraction. The CAP-OMV nanoparticle was prepared and characterized. The efficacy of the vaccine formulation and toxicity were studied in ducks. The average OMV yield in terms of protein concentration was found to be 122.33 ± 3.48 mg per liter of BHI broth. In SDS-PAGE, isolated OMVs exhibited presence of 16 distinct protein bands with molecular weight ranging from 142.1 to 12.1 kDa. Seven protein bands of 74.1, 69.3, 55.5, 50.6, 45.6, 25.1 and 13.1 kDa were detected relatively more distinct. The major bands detected in our findings were 42 kDa, 37 kDa and 16 kDa that corresponds to OmpA, OmpH, P6 respectively. The mean zeta size (±SD) and potential of the nanoparticle were 246.20 ± 0.53 nm and -25.60 ± 5.97 respectively. In transmission electron microscopy (TEM), the nanoparticles exhibited an average diameter of 129.80 ± 11.10 nm and displayed spherical morphology. The median protective dose (PD50) of CAP-OMV nanoparticle was 1881.10 μg of protein. Group I ducks received 3762 μg of protein (entrapped protein in CAP-OMV nanoparticle) via intra nasal route and it showed the highest serum IgG and secretory IgA level than other immunized groups. All experimental ducks were challenged with 10 × LD50 on 35 days of post primary immunization. Group I showed 100 % survivability in the challenge study. No gross and biochemical indication of acute or chronic toxicity were recorded. In conclusion, our results suggest that CAP-OMV nanoparticle can be a safe and efficient mucosal vaccine delivery system for RA, eliciting strong immune response in the host.
Collapse
Affiliation(s)
- Naba Jyoti Deka
- Department of Veterinary Biochemistry, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati, Assam, 781022, India.
| | - Dhruba Jyoti Kalita
- Department of Veterinary Biochemistry, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati, Assam, 781022, India
| | - Shantanu Tamuly
- Department of Veterinary Biochemistry, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati, Assam, 781022, India
| | - Rajeev Kumar Sharma
- Department of Veterinary Microbiology, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati, Assam, 781022, India
| | - Durlav Prasad Bora
- Department of Veterinary Microbiology, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati, Assam, 781022, India
| | - Rupam Dutta
- Department of Animal Biotechnology, College of Veterinary Science, Assam Agricultural University, Khanapara, Assam, 781022, India
| | - Mousumi Hazorika
- Department of Veterinary Biochemistry, College of Veterinary Science, Assam Agricultural University, Khanapara, Guwahati, Assam, 781022, India
| | - Prasanta Chabukdhara
- Department of Veterinary Physiology & Biochemistry, Lakhimpur College of Veterinary Science, Assam Agricultural University, North Lakhimpur, Assam, 787 051, India
| | - Shiney George
- Department of Veterinary Microbiology, College of Veterinary Science, Assam Agricultural University, North Lakhimpur, Assam, 787051, India
| |
Collapse
|
12
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
13
|
Han S, Lee P, Choi HJ. Non-Invasive Vaccines: Challenges in Formulation and Vaccine Adjuvants. Pharmaceutics 2023; 15:2114. [PMID: 37631328 PMCID: PMC10458847 DOI: 10.3390/pharmaceutics15082114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Given the limitations of conventional invasive vaccines, such as the requirement for a cold chain system and trained personnel, needle-based injuries, and limited immunogenicity, non-invasive vaccines have gained significant attention. Although numerous approaches for formulating and administrating non-invasive vaccines have emerged, each of them faces its own challenges associated with vaccine bioavailability, toxicity, and other issues. To overcome such limitations, researchers have created novel supplementary materials and delivery systems. The goal of this review article is to provide vaccine formulation researchers with the most up-to-date information on vaccine formulation and the immunological mechanisms available, to identify the technical challenges associated with the commercialization of non-invasive vaccines, and to guide future research and development efforts.
Collapse
Affiliation(s)
| | | | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada; (S.H.); (P.L.)
| |
Collapse
|
14
|
Liu M, Zhao Y, Shi Z, Zink JI, Yu Q. Virus-like Magnetic Mesoporous Silica Particles as a Universal Vaccination Platform against Pathogenic Infections. ACS NANO 2023; 17:6899-6911. [PMID: 36961475 DOI: 10.1021/acsnano.3c00644] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Vaccination is the most important way of population protection from life-threatening pathogenic infections. However, its efficiency is frequently compromised by a failure of strong antigen presentation and immune activation. Herein, we developed virus-like magnetic mesoporous silica nanoparticles as a universal vaccination platform (termed MagParV) for preventing pathogenic infections. This platform was constructed by integrating synthetic biology-based endoplasmic reticulum-targeting vesicles with magnetic mesoporous silica particles. This platform exhibited high antigen-loading capacity, strongly targeting the endoplasmic reticulum and promoting antigen presentation in dendritic cells. After prime-boost vaccination, the antigen-loading MagParV with AMF drastically elicited specific antibody production against corresponding antigens of fungal, bacterial, and viral pathogens. A systemic infection model further revealed that the platform effectively protected the mice from severe fungal systemic infections. This study realized synthetic biology-facilitated green manufacturing of vaccines, which is promising for magnetism-activated vaccination against different kinds of pathogenic infections.
Collapse
Affiliation(s)
- Mingyang Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, People's Republic of China
| | - Yan Zhao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Zhishang Shi
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| | - Jeffrey I Zink
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- California Nano Systems Institute (CNSI), University of California, Los Angeles, California 90095, United States
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, People's Republic of China
| |
Collapse
|
15
|
Huang L, Liu Z, Wu C, Lin J, Liu N. Magnetic nanoparticles enhance the cellular immune response of dendritic cell tumor vaccines by realizing the cytoplasmic delivery of tumor antigens. Bioeng Transl Med 2023; 8:e10400. [PMID: 36925683 PMCID: PMC10013825 DOI: 10.1002/btm2.10400] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/02/2022] [Accepted: 08/16/2022] [Indexed: 11/08/2022] Open
Abstract
Dendritic cells (DCs)-based tumor vaccines have the advantages of high safety and rapid activation of T cells, and have been approved for clinical tumor treatment. However, the conventional DC vaccines have some severe problems, such as poor activation of DCs in vitro, low level of antigen presentation, reduced cell viability, and difficulty in targeting lymph nodes in vivo, resulting in poor clinical therapeutic effects. In this research, magnetic nanoparticles Fe3O4@Ca/MnCO3 were prepared and used to actively and efficiently deliver antigens to the cytoplasm of DCs, promote antigen cross-presentation and DC activation, and finally enhance the cellular immune response of DC vaccines. The results show that the magnetic nanoparticles can actively and quickly deliver antigens to the cytoplasm of DCs by regulating the magnetic field, and achieve cross-presentation of antigens. At the same time, the nanoparticles degradation product Mn2+ enhanced immune stimulation through the interferon gene stimulating protein (STING) pathway, and another degradation product Ca2+ ultimately promoted cellular immune response by increasing autophagy. The DC vaccine constructed with the magnetic nanoparticles can more effectively migrate to the lymph nodes, promote the proliferation of CD8+ T cells, prolong the time of immune memory, and produce higher antibody levels. Compared with traditional DC vaccines, cytoplasmic antigen delivery with the magnetic nanoparticles provides a new idea for the construction of novel DC vaccines.
Collapse
Affiliation(s)
- Linghong Huang
- Department of Biomedical EngineeringJinan UniversityGuangzhouChina
| | - Zonghua Liu
- Department of Biomedical EngineeringJinan UniversityGuangzhouChina
| | - Chongjie Wu
- Department of Bone and Joint SurgeryThe First Affiliated Hospital of Jinan University, Jinan UniversityGuangzhouChina
| | - Jiansheng Lin
- Department of AnatomyHunan University of Chinese MedicineChangshaChina
| | - Ning Liu
- Department of Bone and Joint SurgeryThe First Affiliated Hospital of Jinan University, Jinan UniversityGuangzhouChina
| |
Collapse
|
16
|
Peletta A, Lemoine C, Courant T, Collin N, Borchard G. Meeting vaccine formulation challenges in an emergency setting: Towards the development of accessible vaccines. Pharmacol Res 2023; 189:106699. [PMID: 36796463 DOI: 10.1016/j.phrs.2023.106699] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Vaccination is considered one of the most successful strategies to prevent infectious diseases. In the event of a pandemic or epidemic, the rapid development and distribution of the vaccine to the population is essential to reduce mortality, morbidity and transmission. As seen during the COVID-19 pandemic, the production and distribution of vaccines has been challenging, in particular for resource-constrained settings, essentially slowing down the process of achieving global coverage. Pricing, storage, transportation and delivery requirements of several vaccines developed in high-income countries resulted in limited access for low-and-middle income countries (LMICs). The capacity to manufacture vaccines locally would greatly improve global vaccine access. In particular, for the development of classical subunit vaccines, the access to vaccine adjuvants is a pre-requisite for more equitable access to vaccines. Vaccine adjuvants are agents required to augment or potentiate, and possibly target the specific immune response to such type of vaccine antigens. Openly accessible or locally produced vaccine adjuvants may allow for faster immunization of the global population. For local research and development of adjuvanted vaccines to expand, knowledge on vaccine formulation is of paramount importance. In this review, we aim to discuss the optimal characteristics of a vaccine developed in an emergency setting by focusing on the importance of vaccine formulation, appropriate use of adjuvants and how this may help overcome barriers for vaccine development and production in LMICs, achieve improved vaccine regimens, delivery and storage requirements.
Collapse
Affiliation(s)
- Allegra Peletta
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| | - Céline Lemoine
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Thomas Courant
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| |
Collapse
|
17
|
Biomedical applications of solid-binding peptides and proteins. Mater Today Bio 2023; 19:100580. [PMID: 36846310 PMCID: PMC9950531 DOI: 10.1016/j.mtbio.2023.100580] [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: 12/06/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Over the past decades, solid-binding peptides (SBPs) have found multiple applications in materials science. In non-covalent surface modification strategies, solid-binding peptides are a simple and versatile tool for the immobilization of biomolecules on a vast variety of solid surfaces. Especially in physiological environments, SBPs can increase the biocompatibility of hybrid materials and offer tunable properties for the display of biomolecules with minimal impact on their functionality. All these features make SBPs attractive for the manufacturing of bioinspired materials in diagnostic and therapeutic applications. In particular, biomedical applications such as drug delivery, biosensing, and regenerative therapies have benefited from the introduction of SBPs. Here, we review recent literature on the use of solid-binding peptides and solid-binding proteins in biomedical applications. We focus on applications where modulating the interactions between solid materials and biomolecules is crucial. In this review, we describe solid-binding peptides and proteins, providing background on sequence design and binding mechanism. We then discuss their application on materials relevant for biomedicine (calcium phosphates, silicates, ice crystals, metals, plastics, and graphene). Although the limited characterization of SBPs still represents a challenge for their design and widespread application, our review shows that SBP-mediated bioconjugation can be easily introduced into complex designs and on nanomaterials with very different surface chemistries.
Collapse
|
18
|
Nayeri T, Sarvi S, Fasihi-Ramandi M, Valadan R, Asgarian-Omran H, Ajami A, Khalilian A, Hosseininejad Z, Dodangeh S, Javidnia J, Daryani A. Enhancement of immune responses by vaccine potential of three antigens, including ROP18, MIC4, and SAG1 against acute toxoplasmosis in mice. Exp Parasitol 2022; 244:108427. [PMID: 36379272 DOI: 10.1016/j.exppara.2022.108427] [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: 09/05/2022] [Revised: 10/31/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Toxoplasma gondii (T. gondii) causes considerable financial losses in the livestock industry and can present serious threats to pregnant women, as well as immunocompromised patients. Therefore, it is required to design and produce an efficient vaccine for controlling toxoplasmosis. The present study aimed to evaluate the protective immunity induced by RMS protein (ROP18, MIC4, and SAG1) with Freund adjuvant, calcium phosphate nanoparticles (CaPNs), and chitosan nanoparticles (CNs) in BALB/c mice. The RMS protein was expressed in Escherichia coli (E. coli) and purified using a HisTrap HP column. Thereafter, cellular and humoral immunity was assessed by injecting RMS protein on days 0, 21, and 35 into four groups [RMS, RMS-chitosan nanoparticles (RMS-CNs), RMS-calcium phosphate nanoparticles (RMS-CaPNs), and RMS-Freund]. Phosphate buffered saline (PBS), CNs, CaPNs, and Freund served as the four control groups. The results displayed that vaccination with RMS protein and adjuvants significantly elicited the levels of specific IgG antibodies and cytokines against toxoplasmosis. There were high levels of total IgG, IgG2a, and IFN-γ in vaccinated mice, compared to those in the control groups, especially in the RMS-Freund, indicating a Th-1 type response. The vaccinated and control mice were challenged intraperitoneally with 1 × 103 tachyzoites of the T. gondii RH strain four weeks after the last injection, and in RMS-Freund and RMS-CaPNs groups, the highest increase in survival time was observed (15 days). The RMS can significantly increase Th1 and Th2 responses; moreover, multi-epitope vaccines with adjuvants can be a promising strategy for the production of a vaccine against toxoplasmosis.
Collapse
Affiliation(s)
- Tooran Nayeri
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Valadan
- Immunology Department, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Immunology Department, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abolghasem Ajami
- Immunology Department, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Antimicrobial Resistance Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Khalilian
- Department of Biostatistics and Community Medicine, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Hosseininejad
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Samira Dodangeh
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Javad Javidnia
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran; Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
19
|
Gong X, Gao Y, Shu J, Zhang C, Zhao K. Chitosan-Based Nanomaterial as Immune Adjuvant and Delivery Carrier for Vaccines. Vaccines (Basel) 2022; 10:1906. [PMID: 36423002 PMCID: PMC9696061 DOI: 10.3390/vaccines10111906] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 08/26/2023] Open
Abstract
With the support of modern biotechnology, vaccine technology continues to iterate. The safety and efficacy of vaccines are some of the most important areas of development in the field. As a natural substance, chitosan is widely used in numerous fields-such as immune stimulation, drug delivery, wound healing, and antibacterial procedures-due to its good biocompatibility, low toxicity, biodegradability, and adhesion. Chitosan-based nanoparticles (NPs) have attracted extensive attention with respect to vaccine adjuvants and delivery systems due to their excellent properties, which can effectively enhance immune responses. Here, we list the classifications and mechanisms of action of vaccine adjuvants. At the same time, the preparation methods of chitosan, its NPs, and their mechanism of action in the delivery system are introduced. The extensive applications of chitosan and its NPs in protein vaccines and nucleic acid vaccines are also introduced. This paper reviewed the latest research progress of chitosan-based NPs in vaccine adjuvant and drug delivery systems.
Collapse
Affiliation(s)
- Xiaochen Gong
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
- School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Yuan Gao
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
| | - Jianhong Shu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Hom-Sun Biotechnology Co., Ltd., Shaoxing 312366, China
| | - Chunjing Zhang
- School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Kai Zhao
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Hom-Sun Biotechnology Co., Ltd., Shaoxing 312366, China
| |
Collapse
|
20
|
Lee J, Ahn SY, Le CTT, Lee DH, Jung J, Ko EJ. Protective and vaccine dose-sparing efficacy of Poly I:C-functionalized calcium phosphate nanoparticle adjuvants in inactivated influenza vaccination. Int Immunopharmacol 2022; 112:109240. [PMID: 36115278 DOI: 10.1016/j.intimp.2022.109240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022]
Abstract
Adjuvants are required to increase the immunogenicity and efficacy of vaccination and enable vaccine dose sparing. Polyinosinic-polycytidylic acid (Poly I:C), a toll-like receptor 3 agonist, is a promising adjuvant candidate that can induce cell-mediated immune responses; however, it remains unlicensed owing to its low stability and toxicity. Calcium phosphate (CaP), a biocompatible and biodegradable nanoparticle, is widely used in biomedicine for stable and targeted drug delivery. In this study, we developed Poly I:C-functionalized CaP (Poly-CaP) and evaluated its vaccine adjuvant efficacy in vitro and in vivo. A half dose of Poly-CaP nanoparticles showed similar efficacy to a full dose of soluble Poly I:C in stimulating bone marrow-derived dendritic cells and macrophages to secrete proinflammatory cytokines and express their activation markers. Immunization with a half dose of inactivated influenza vaccine in the presence of Poly I:C or Poly-CaP adjuvants induced sufficient antigen-specific humoral responses after boost immunization. Immunization with Poly I:C, CaP, or Poly-CaP-adjuvanted with a half dose of influenza vaccine showed comparable protective efficacy against lethal virus infection, with lower weight loss and virus titer than a full dose of influenza vaccine. The Poly-CaP adjuvant was effective in stimulating antigen-specific CD4+ T cell proliferation in the lungs. Collectively, our results showed that the Poly-CaP adjuvant enhanced antigen-specific cell-mediated immunity and humoral immune responses with vaccine dose-sparing effects, suggesting its potential as a novel vaccine adjuvant candidate.
Collapse
Affiliation(s)
- Jueun Lee
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - So Yeon Ahn
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Chau Thuy Tien Le
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Dong-Ha Lee
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jaehan Jung
- Department of Materials Science and Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea.
| | - Eun-Ju Ko
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Republic of Korea; Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
21
|
Fan J, Jin S, Gilmartin L, Toth I, Hussein WM, Stephenson RJ. Advances in Infectious Disease Vaccine Adjuvants. Vaccines (Basel) 2022; 10:1120. [PMID: 35891284 PMCID: PMC9316175 DOI: 10.3390/vaccines10071120] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Vaccines are one of the most significant medical interventions in the fight against infectious diseases. Since their discovery by Edward Jenner in 1796, vaccines have reduced the worldwide transmission to eradication levels of infectious diseases, including smallpox, diphtheria, hepatitis, malaria, and influenza. However, the complexity of developing safe and effective vaccines remains a barrier for combating many more infectious diseases. Immune stimulants (or adjuvants) are an indispensable factor in vaccine development, especially for inactivated and subunit-based vaccines due to their decreased immunogenicity compared to whole pathogen vaccines. Adjuvants are widely diverse in structure; however, their overall function in vaccine constructs is the same: to enhance and/or prolong an immunological response. The potential for adverse effects as a result of adjuvant use, though, must be acknowledged and carefully managed. Understanding the specific mechanisms of adjuvant efficacy and safety is a key prerequisite for adjuvant use in vaccination. Therefore, rigorous pre-clinical and clinical research into adjuvant development is essential. Overall, the incorporation of adjuvants allows for greater opportunities in advancing vaccine development and the importance of immune stimulants drives the emergence of novel and more effective adjuvants. This article highlights recent advances in vaccine adjuvant development and provides detailed data from pre-clinical and clinical studies specific to infectious diseases. Future perspectives into vaccine adjuvant development are also highlighted.
Collapse
Affiliation(s)
- Jingyi Fan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Shengbin Jin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Lachlan Gilmartin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Rachel J. Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| |
Collapse
|
22
|
Singh Y, Kumar A, Saxena A, Bhatt P, Singh SP, Kumar A, Mrigesh M, Saxena MK. Assessment of free radicals and reactive oxygen species milieu in nanoparticles adjuvanted outer membrane proteins vaccine against Salmonella typhi. Arch Microbiol 2022; 204:479. [PMID: 35831726 DOI: 10.1007/s00203-022-03096-1] [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: 01/28/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/26/2022]
Abstract
In this study, calcium phosphate nanoparticles-based (STCNV) and montanide oil adjuvant vaccine (STOAV) containing outer membrane proteins (Omps) of S. Typhi were evaluated for inducing oxidative stress indicators [reduced glutathione (GSH), lipid peroxidation (LPO), catalase, superoxide dismutase (SOD), and total protein] in the tissues of mice after vaccination. The GSH levels though slightly high in the liver, kidney, and lungs of STCNV group were not significantly different from STOAV and the control group (STC). There was no significant difference in LPO levels in any group for any tissue. The significantly lower activities of catalase were observed in the kidney and lungs of the STCNV group as compared to STOAV and STC group, while in the liver, STCNV group revealed lower catalase activity in comparison to the control group. No significant difference in the SOD activities between the two vaccinated groups was observed. The total protein contents in all the organs showed no significant difference in the vaccinated and the control group. The vaccines may induce long-term inflammatory response and consequently damage vital organs; this study revealed no long-term oxidative stress in all the three vital organs, suggesting that these vaccines may not cause oxidative damages in the vital organs of mice.
Collapse
Affiliation(s)
- Yashpal Singh
- Department of Molecular Biology and Genetic Engineering, College of Basic Sciences and Humanities, G.B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| | - Amit Kumar
- Division of Animal Biotechnology, College of Biotechnology, Sardar Vallabh Bhai Patel University of Agriculture and Technology, Meerut, Uttar Pradesh, 250110, India
| | - Anjani Saxena
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary and Animal Sciences, G.B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| | - Pankaj Bhatt
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - S P Singh
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary and Animal Sciences, G.B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| | - Avadhesh Kumar
- Department of Veterinary and Animal Husbandry Extension Education, College of Veterinary and Animal Sciences, G.B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| | - Meena Mrigesh
- Department of Veterinary Anatomy, College of Veterinary and Animal Sciences, G.B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| | - Mumtesh Kumar Saxena
- Department of Animal Genetics and Breeding, College of Veterinary and Animal Sciences, G.B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India.
| |
Collapse
|
23
|
Singh Y, Saxena A, Singh SP, Verma MK, Kumar A, Kumar A, Mrigesh M, Saxena MK. Calcium phosphate adjuvanted nanoparticles of outer membrane proteins of Salmonella Typhi as a candidate for vaccine development against Typhoid fever. J Med Microbiol 2022; 71. [PMID: 35476604 DOI: 10.1099/jmm.0.001529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. The conventional adjuvants used in vaccines have limitations like induction of an imbalanced Th1 and Th2 immune response. To overcome this limitation, novel adjuvants and newer forms of existing adjuvants like calcium phosphate nanoparticles are being tested.Hypothesis/Gap Statement. Calcium phosphate adjuvanted outer membrane proteins vaccine may work as an efficient, safe and cost effective vaccine against Salmonella Typhi.Aim. Our goals were to evaluate the potential of calcium phosphate nanoparticles as an adjuvant using outer membrane proteins (Omps) of Salmonella Typhi as antigens for immune response, with montanide (commercially available adjuvant) as control, and its toxicity in rats.Methodology. Calcium phosphate adjuvanted outer membrane proteins nanoparticles were synthesized and characterized. The efficacy of vaccine formulation in mice and toxicity assay were carried out in rats.Results. The calcium phosphate nanoparticles varying in size between 20-50 nm had entrapment efficiency of 41.5% and loading capacity of 54%. The calcium phosphate nanoparticle-Omps vaccine formulation (nanoparticle-Omps) induced a strong humoral immune response, which was significantly higher than the control group for the entire period of study. In the montanide-Omps group the initial very high immune response declined steeply and then remained steady. The immune response induced by nanoparticle-Omps did not change appreciably. The cell mediated immune response as measured by lymphocyte proliferation assay and delayed type hypersensitivity test showed a higher response (P<0.01) for the nanoparticles-Omps group as compared to montanide-Omps group. The bacterial clearance assay also showed higher clearance in the nanoparticles-Omps group as compared to montanide-Omps group (approx 1.4%). The toxicity analysis in rats showed no difference in the values of toxicity biomarkers and blood chemistry parameters, revealing vaccine formulation was non-toxic in rats.Conclusion. Calcium phosphate nanoparticles as adjuvant in vaccines is safe, have good encapsulation and loading capacity and induce a strong cell mediated, humoral and protective immune response.
Collapse
Affiliation(s)
- Yashpal Singh
- Department of Molecular Biology & Genetic Engineering, College of Basic Sciences & Humanities, Pantnagar, Uttarakhand, India
| | - Anjani Saxena
- Department of Veterinary Pharmacology & Toxicology, College of Veterinary & Animal Sciences, Pantnagar, Uttarakhand, India
| | - S P Singh
- Department of Veterinary Pharmacology & Toxicology, College of Veterinary & Animal Sciences, Pantnagar, Uttarakhand, India
| | - Manish Kumar Verma
- Department of Veterinary Pharmacology & Toxicology, College of Veterinary & Animal Sciences, Pantnagar, Uttarakhand, India
| | - Arun Kumar
- Department of Veterinary Surgery and Radiology, College of Veterinary & Animal Sciences, Pantnagar, Uttarakhand, India
| | - Avadhesh Kumar
- Department of Veterinary & Animal Husbandry Extension Education, College of Veterinary & Animal Sciences, Pantnagar, Uttarakhand, India
| | - Meena Mrigesh
- Department of Veterinary Anatomy, College of Veterinary & Animal Sciences, Pantnagar, Uttarakhand, India
| | - Mumtesh Kumar Saxena
- Department of Animal Genetics & Breeding, College of Veterinary & Animal Sciences G.B. Pant University of Agriculture & Technology, Pantnagar, Uttarakhand, India
| |
Collapse
|
24
|
van Rijt S, de Groot K, Leeuwenburgh SCG. Calcium phosphate and silicate-based nanoparticles: history and emerging trends. Tissue Eng Part A 2022; 28:461-477. [PMID: 35107351 DOI: 10.1089/ten.tea.2021.0218] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bulk calcium phosphates and silicate-based bioglasses have been extensively studied since the early 1970s due to their unique capacity to bind to host bone, which led to their clinical translation and commercialization in the 1980s. Since the mid-1990s, researchers have synthesized nanoscale calcium phosphate and silicate-based particles of increased specific surface area, chemical reactivity and solubility which offer specific advantages as compared to their bulk counterparts. This review provides a critical perspective on the history and emerging trends of these two classes of ceramic nanoparticles. Their synthesis and functional properties in terms of particle composition, size, shape, charge, dispersion, and toxicity are discussed as a function of relevant processing parameters. Specifically, emerging trends such as the influence of ion doping and mesoporosity on the biological and pharmaceutical performance of these nanoparticles are reviewed in more detail. Finally, a broad comparative overview is provided on the physicochemical properties and applicability of calcium phosphate and silicate-based nanoparticles within the fields of i) local delivery of therapeutic agents, ii) functionalization of biomaterial scaffolds or implant coatings, and iii) bio-imaging applications.
Collapse
Affiliation(s)
- Sabine van Rijt
- Maastricht University, 5211, MERLN Institute-Instructive Biomaterial Engineering, Maastricht, Limburg, Netherlands;
| | - Klaas de Groot
- Vrije Universiteit Amsterdam, 1190, Academic Center for Dentistry Amsterdam (ACTA)-Department of Oral Implantology and Prosthetic Dentistry, Amsterdam, Noord-Holland, Netherlands;
| | - Sander C G Leeuwenburgh
- Radboudumc, 6034, Dept. of Dentistry-Regenerative Biomaterials, Nijmegen, Gelderland, Netherlands;
| |
Collapse
|
25
|
Koirala P, Bashiri S, Toth I, Skwarczynski M. Current Prospects in Peptide-Based Subunit Nanovaccines. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2412:309-338. [PMID: 34918253 DOI: 10.1007/978-1-0716-1892-9_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Vaccination renders protection against pathogens via stimulation of the body's natural immune responses. Classical vaccines that utilize whole organisms or proteins have several disadvantages, such as induction of undesired immune responses, poor stability, and manufacturing difficulties. The use of minimal immunogenic pathogen components as vaccine antigens, i.e., peptides, can greatly reduce these shortcomings. However, subunit antigens require a specific delivery system and immune adjuvant to increase their efficacy. Recently, nanotechnology has been extensively utilized to address this issue. Nanotechnology-based formulation of peptide vaccines can boost immunogenicity and efficiently induce cellular and humoral immune responses. This chapter outlines the recent developments and advances of nano-sized delivery platforms for peptide antigens, including nanoparticles composed of polymers, peptides, lipids, and inorganic materials.
Collapse
Affiliation(s)
- Prashamsa Koirala
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Sahra Bashiri
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia. .,Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia. .,School of Pharmacy, The University of Queensland, St Lucia, QLD, Australia.
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
26
|
Sun HC, Huang J, Fu Y, Hao LL, Liu X, Shi TY. Enhancing Immune Responses to a DNA Vaccine Encoding Toxoplasma gondii GRA7 Using Calcium Phosphate Nanoparticles as an Adjuvant. Front Cell Infect Microbiol 2022; 11:787635. [PMID: 34976863 PMCID: PMC8716823 DOI: 10.3389/fcimb.2021.787635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
Toxoplasma gondii infects almost all warm-blooded animals, including humans. DNA vaccines are an effective strategy against T. gondii infection, but these vaccines have often been poorly immunogenic due to the poor distribution of plasmids or degradation by lysosomes. It is necessary to evaluate the antigen delivery system for optimal vaccination strategy. Nanoparticles (NPs) have been shown to modulate and enhance the cellular humoral immune response. Here, we studied the immunological properties of calcium phosphate nanoparticles (CaPNs) as nanoadjuvants to enhance the protective effect of T. gondii dense granule protein (GRA7). BALB/c mice were injected three times and then challenged with T. gondii RH strain tachyzoites. Mice vaccinated with GRA7-pEGFP-C2+nano-adjuvant (CaPNs) showed a strong cellular immune response, as monitored by elevated levels of anti-T. gondii-specific immunoglobulin G (IgG), a higher IgG2a-to-IgG1 ratio, elevated interleukin (IL)-12 and interferon (IFN)-γ production, and low IL-4 levels. We found that a significantly higher level of splenocyte proliferation was induced by GRA7-pEGFP-C2+nano-adjuvant (CaPNs) immunization, and a significantly prolonged survival time and decreased parasite burden were observed in vaccine-immunized mice. These data indicated that CaPN-based immunization with T. gondii GRA7 is a promising approach to improve vaccination.
Collapse
Affiliation(s)
- Hong-Chao Sun
- Department of Animal Parasitology, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Jing Huang
- Department of Animal Epidemic Surveillance, Zhejiang Provincial Animal Disease Prevention and Control Center, Hangzhou, China
| | - Yuan Fu
- Department of Animal Parasitology, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Li-Li Hao
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Xin Liu
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Tuan-Yuan Shi
- Department of Animal Parasitology, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, China
| |
Collapse
|
27
|
Zhang L, Liang Z, Chen C, Yang X, Fu D, Bao H, Li M, Shi S, Yu G, Zhang Y, Zhang C, Zhang W, Xue C, Sun B. Engineered Hydroxyapatite Nanoadjuvants with Controlled Shape and Aspect Ratios Reveal Their Immunomodulatory Potentials. ACS APPLIED MATERIALS & INTERFACES 2021; 13:59662-59672. [PMID: 34894655 DOI: 10.1021/acsami.1c17804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hydroxyapatite (HAP) has been formulated as adjuvants in vaccines for human use. However, the optimal properties required for HAP nanoparticles to elicit adjuvanticity and the underlying immunopotentiation mechanisms have not been fully elucidated. Herein, a library of HAP nanorods and nanospheres was synthesized to explore the effect of the particle shape and aspect ratio on the immune responses in vitro and adjuvanticity in vivo. It was demonstrated that long aspect ratio HAP nanorods induced a higher degree of cell membrane depolarization and subsequent uptake, and the internalized particles elicited cathepsin B release and mitochondrial reactive oxygen species generation, which further led to pro-inflammatory responses. Furthermore, the physicochemical property-dependent immunostimulation capacities were correlated with their humoral responses in a murine hepatitis B surface antigen immunization model, with long aspect ratio HAP nanorods inducing higher antigen-specific antibody productions. Importantly, HAP nanorods significantly up-regulated the IFN-γ secretion and CD107α expression on CD8+ T cells in immunized mice. Further mechanistic studies demonstrated that HAP nanorods with defined properties exerted immunomodulatory effects by enhanced antigen persistence and immune cell recruitments. Our study provides a rational design strategy for engineered nanomaterial-based vaccine adjuvants.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Zhihui Liang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Chen Chen
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Xuecheng Yang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Duo Fu
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Hang Bao
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Min Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Shuting Shi
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Ge Yu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Yixuan Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Caiqiao Zhang
- NCPC Genetech Biotechnology Co., Ltd., Shijiazhuang 050035, P. R. China
| | - Weiting Zhang
- NCPC Genetech Biotechnology Co., Ltd., Shijiazhuang 050035, P. R. China
| | - Changying Xue
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| |
Collapse
|
28
|
A Self-Biomineralized Novel Adenovirus Vectored COVID-19 Vaccine for Boosting Immunization of Mice. Virol Sin 2021; 36:1113-1123. [PMID: 34581961 PMCID: PMC8476980 DOI: 10.1007/s12250-021-00434-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/28/2021] [Indexed: 12/28/2022] Open
Abstract
SARS-CoV-2 has caused more than 3.8 million deaths worldwide, and several types of COVID-19 vaccines are urgently approved for use, including adenovirus vectored vaccines. However, the thermal instability and pre-existing immunity have limited its wide applications. To circumvent these obstacles, we constructed a self-biomineralized adenovirus vectored COVID-19 vaccine (Sad23L-nCoV-S-CaP) by generating a calcium phosphate mineral exterior (CaP) based on Sad23L vector carrying the full-length gene of SARS-CoV-2 spike protein (S) under physiological condition. This Sad23L-nCoV-S-CaP vaccine was examined for its characteristics of structure, thermostability, immunogenicity and avoiding the problem of preexisting immunity. In thermostability test, Sad23L-nCoV-S-CaP could be stored at 4 °C for over 45 days, 26 °C for more than 8 days and 37 °C for approximately 2 days. Furthermore, Sad23L-nCoV-S-CaP induced higher level of S-specific antibody and T cell responses, and was not affected by the pre-existing anti-Sad23L immunity, suggesting it could be used as boosting immunization on Sad23L-nCoV-S priming vaccination. The boosting with Sad23L-nCoV-S-CaP vaccine induced high titers of 105.01 anti-S1, 104.77 anti-S2 binding antibody, 103.04 pseudovirus neutralizing antibody (IC50), and robust T-cell response of IFN-γ (1466.16 SFCs/106 cells) to S peptides, respectively. In summary, the self-biomineralization of the COVID-19 vaccine Sad23L-nCoV-S-CaP improved vaccine efficacy, which could be used in prime-boost regimen for prevention of SARS-CoV-2 infection in humans.
Collapse
|
29
|
Guerrero Manriquez GG, Tuero I. Adjuvants: friends in vaccine formulations against infectious diseases. Hum Vaccin Immunother 2021; 17:3539-3550. [PMID: 34288795 DOI: 10.1080/21645515.2021.1934354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Infectious diseases represent a major cause of deaths worldwide. No vaccine or effective treatment exists nowadays, especially against intracellular pathogens. The increase in multiple drug and superbug antibiotic resistance strains, excessive medication, or misuse of drugs has prompted the search for other safe and effective alternatives. Consistent with this, adjuvants (Latin word "adjuvare": "help or aid") co-administered (Exo) in vaccines have emerged as a promising alternative to initiate and boost an innate, downstream signal that led to adaptative immune response. Nowadays, a promising model of strong immunogens and adjuvants at mucosal sites are the microbial bacterial toxins. Other adjuvants that are also used and might successfully replace aluminum salts in combination with nanotechnology are CpG-ODN, poly IC, type I IFNs, mRNA platforms. Therefore, in the present review, we focused to revisit the old to the new adjuvants compounds, the properties that make them friends in vaccine formulations against infectious diseases.
Collapse
Affiliation(s)
| | - I Tuero
- Faculty of Science and Phylosophy, Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
30
|
Min Y, Xu W, Xiao Y, Xiao J, Shu Z, Li S, Zhang J, Liu Y, Yin Y, Zhang X, Meng J. Biomineralization improves the stability of a Streptococcus pneumoniae protein vaccine at high temperatures. Nanomedicine (Lond) 2021; 16:1747-1761. [PMID: 34264093 DOI: 10.2217/nnm-2021-0023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Protein vaccines have been the focus of research for vaccine development due to their safety record and facile production. Improving the stability of proteins is of great significance to the application of protein vaccines. Materials & methods: Based on the proteins pneumolysin and DnaJ of Streptococcus pneumoniae, biomineralization was carried out to prepare protein nanoparticles, and their thermal stability was tested both in vivo and in vitro. Results: Mineralized nanoparticles were formed successfully and these calcium phosphate-encapsulated proteins were resistant to proteinase K degradation and were thermally stable at high temperatures. The mineralized proteins retained the immunoreactivity of the original proteins. Conclusion: Mineralization technology is an effective means to stabilize protein vaccines, presenting a safe and economical method for vaccine administration.
Collapse
Affiliation(s)
- Yajun Min
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China.,Department of Obstetrics & Gynecology, Assisted Reproductive Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Wenchun Xu
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Yunju Xiao
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Jiangming Xiao
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Zhaoche Shu
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Sijie Li
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Jinghui Zhang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Yusi Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Yibing Yin
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Xuemei Zhang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Jiangping Meng
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
31
|
Zhang X, Zhang Z, Xia N, Zhao Q. Carbohydrate-containing nanoparticles as vaccine adjuvants. Expert Rev Vaccines 2021; 20:797-810. [PMID: 34101528 DOI: 10.1080/14760584.2021.1939688] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Adjuvants are essential to vaccines for immunopotentiation in the elicitation of protective immunity. However, classical and widely used aluminum-based adjuvants have limited capacity to induce cellular response. There are increasing needs for appropriate adjuvants with improved profiles for vaccine development toward emerging pathogens. Carbohydrate-containing nanoparticles (NPs) with immunomodulatory activity and particulate nanocarriers for effective antigen presentation are capable of eliciting a more balanced humoral and cellular immune response.Areas covered: We reviewed several carbohydrates with immunomodulatory properties. They include chitosan, β-glucan, mannan, and saponins, which have been used in vaccine formulations. The mode of action, the preparation methods, characterization of these carbohydrate-containing NPs and the corresponding vaccines are presented.Expert opinion: Several carbohydrate-containing NPs have entered the clinical stage or have been used in licensed vaccines for human use. Saponin-containing NPs are being evaluated in a vaccine against SARS-CoV-2, the pathogen causing the on-going worldwide pandemic. Vaccines with carbohydrate-containing NPs are in different stages of development, from preclinical studies to late-stage clinical trials. A better understanding of the mode of action for carbohydrate-containing NPs as vaccine carriers and as immunostimulators will likely contribute to the design and development of new generation vaccines against cancer and infectious diseases.
Collapse
Affiliation(s)
- Xinyuan Zhang
- 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, Fujian, PR China
| | - Zhigang Zhang
- 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, Fujian, PR China
| | - Ningshao Xia
- 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, Fujian, PR China.,School of Life Sciences, Xiamen University, Xiamen, Fujian, PR China.,The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, Fujian, PR China
| | - Qinjian Zhao
- 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, Fujian, PR China
| |
Collapse
|
32
|
Walkowski W, Bassett J, Bhalla M, Pfeifer BA, Ghanem ENB. Intranasal Vaccine Delivery Technology for Respiratory Tract Disease Application with a Special Emphasis on Pneumococcal Disease. Vaccines (Basel) 2021; 9:vaccines9060589. [PMID: 34199398 PMCID: PMC8230341 DOI: 10.3390/vaccines9060589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/17/2021] [Accepted: 05/22/2021] [Indexed: 12/17/2022] Open
Abstract
This mini-review will cover recent trends in intranasal (IN) vaccine delivery as it relates to applications for respiratory tract diseases. The logic and rationale for IN vaccine delivery will be compared to methods and applications accompanying this particular administration route. In addition, we will focus extended discussion on the potential role of IN vaccination in the context of respiratory tract diseases, with a special emphasis on pneumococcal disease. Here, elements of this disease, including its prevalence and impact upon the elderly population, will be viewed from the standpoint of improving health outcomes through vaccine design and delivery technology and how IN administration can play a role in such efforts.
Collapse
Affiliation(s)
- William Walkowski
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (W.W.); (J.B.); (B.A.P.)
| | - Justin Bassett
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (W.W.); (J.B.); (B.A.P.)
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA;
| | - Blaine A. Pfeifer
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA; (W.W.); (J.B.); (B.A.P.)
| | - Elsa N. Bou Ghanem
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, NY 14203, USA;
- Correspondence:
| |
Collapse
|
33
|
Abstract
Calcium phosphate nanoparticles have a high biocompatibility and biodegradability due to their chemical similarity to human hard tissue, for example, bone and teeth. They can be used as efficient carriers for different kinds of biomolecules such as nucleic acids, proteins, peptides, antibodies, or drugs, which alone are not able to enter cells where their biological effect is required. They can be loaded with cargo molecules by incorporating them, unlike solid nanoparticles, and also by surface functionalization. This offers protection, for example, against nucleases, and the possibility for cell targeting. If such nanoparticles are functionalized with fluorescing dyes, they can be applied for imaging in vitro and in vivo. Synthesis, functionalization and cell uptake mechanisms of calcium phosphate nanoparticles are discussed together with applications in transfection, gene silencing, imaging, immunization, and bone substitution. Biodistribution data of calcium phosphate nanoparticles in vivo are reviewed.
Collapse
Affiliation(s)
- Viktoriya Sokolova
- Inorganic chemistryUniversity of Duisburg-EssenUniversitaetsstr. 5–745117EssenGermany
| | - Matthias Epple
- Inorganic chemistryUniversity of Duisburg-EssenUniversitaetsstr. 5–745117EssenGermany
| |
Collapse
|
34
|
Guo Z, Kubiatowicz LJ, Fang RH, Zhang L. Nanotoxoids: Biomimetic Nanoparticle Vaccines against Infections. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Luke J. Kubiatowicz
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| |
Collapse
|
35
|
Mao L, Chen Z, Wang Y, Chen C. Design and application of nanoparticles as vaccine adjuvants against human corona virus infection. J Inorg Biochem 2021; 219:111454. [PMID: 33878530 PMCID: PMC8007196 DOI: 10.1016/j.jinorgbio.2021.111454] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/08/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022]
Abstract
In recent years, some viruses have caused a grave crisis to global public health, especially the human coronavirus. A truly effective vaccine is therefore urgently needed. Vaccines should generally have two features: delivering antigens and modulating immunity. Adjuvants have an unshakable position in the battle against the virus. In addition to the perennial use of aluminium adjuvant, nanoparticles have become the developing adjuvant candidates due to their unique properties. Here we introduce several typical nanoparticles and their antivirus vaccine adjuvant applications. Finally, for the combating of the coronavirus, we propose several design points, hoping to provide ideas for the development of personalized vaccines and adjuvants and accelerate the clinical application of adjuvants.
Collapse
Affiliation(s)
- Lichun Mao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ziwei Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yaling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, PR China; GBA National Institute for Nanotechnology Innovation, Guangdong 510700, PR China.
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; GBA National Institute for Nanotechnology Innovation, Guangdong 510700, PR China; Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100021, PR China.
| |
Collapse
|
36
|
An Overview of Nanocarrier-Based Adjuvants for Vaccine Delivery. Pharmaceutics 2021; 13:pharmaceutics13040455. [PMID: 33801614 PMCID: PMC8066039 DOI: 10.3390/pharmaceutics13040455] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022] Open
Abstract
The development of vaccines is one of the most significant medical accomplishments which has helped to eradicate a large number of diseases. It has undergone an evolutionary process from live attenuated pathogen vaccine to killed whole organisms or inactivated toxins (toxoids), each of them having its own advantages and disadvantages. The crucial parameters in vaccination are the generation of memory response and protection against infection, while an important aspect is the effective delivery of antigen in an intelligent manner to evoke a robust immune response. In this regard, nanotechnology is greatly contributing to developing efficient vaccine adjuvants and delivery systems. These can protect the encapsulated antigen from the host’s in-vivo environment and releasing it in a sustained manner to induce a long-lasting immunostimulatory effect. In view of this, the present review article summarizes nanoscale-based adjuvants and delivery vehicles such as viral vectors, virus-like particles and virosomes; non-viral vectors namely nanoemulsions, lipid nanocarriers, biodegradable and non-degradable nanoparticles, calcium phosphate nanoparticles, colloidally stable nanoparticles, proteosomes; and pattern recognition receptors covering c-type lectin receptors and toll-like receptors.
Collapse
|
37
|
Dodangeh S, Fasihi-Ramandi M, Daryani A, Valadan R, Asgarian-Omran H, Hosseininejad Z, Nayeri Chegeni T, Pagheh AS, Javidnia J, Sarvi S. Protective efficacy by a novel multi-epitope vaccine, including MIC3, ROP8, and SAG1, against acute Toxoplasma gondii infection in BALB/c mice. Microb Pathog 2021; 153:104764. [PMID: 33548480 DOI: 10.1016/j.micpath.2021.104764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/21/2020] [Accepted: 01/24/2021] [Indexed: 10/22/2022]
Abstract
Toxoplasma gondii is an intracellular apicomplexan parasite, which can cause a serious infectious disease in pregnant women and immunocompromised individuals. Therefore, the development of a polyvalent vaccine consisting of all stages of the parasite life cycle using the epitopes from tachyzoites, bradyzoites, and sporozoites is likely to be required for complete protective immunity. In this study, we designed protein vaccine candidate based on the prediction of specific epitopes (i.e., B cell and T cell) from three Toxoplasma gondii antigens. The MRS protein (MIC3: 30-180, ROP8: 85-185, and SAG1: 85-235) was expressed in Escherichia coli, and purification was performed using a HisTrap HP column and then we evaluated immunogenicity and protective property in BALB/c mice. Seventy-two mice were randomly divided into six groups, including three vaccinations (i.e., MRS, MRS-Freund, and MRS-Calcium Phosphate Nanoparticles (MRS-CaPNs)) and three control (i.e., Phosphate-buffered saline, Freund, and CaPNs) groups. All groups were immunized three times via subcutaneous injection within three-week intervals. In the vaccination groups, the BALB/c mice were injected with 20 μg of MRS protein for the first time and 10 μg of MRS for the next two times. Antibodies, cytokines, and splenocytes proliferation in the immunized mice were assayed using the enzyme-linked immunosorbent assay. Protective efficacy was analyzed by challenging the immunized mice with T. gondii of RH strain. Antibody, cytokine, and lymphocyte proliferation assays showed that the mice immunized with MRS induced stronger humoral and T helper type 1 cell-mediated immune responses, compared to the control mice. However, co-immunization with adjuvants (i.e., Freund and CaNPs) resulted in impaired immune responses. Effective protection against the parasite achieved an increase in survival time in the immunized mice, especially in the MRS-CaNPs group. The obtained results of the present study demonstrated that multi-epitope protein vaccination, MRS, is a potential strategy against toxoplasmosis infection. In addition, the vaccine co-delivered with CaPNs could provide an important key for vaccine candidate to control T. gondii infection.
Collapse
Affiliation(s)
- Samira Dodangeh
- Department of Medical Parasitology and Mycology, Children Growth Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Valadan
- Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Hosseininejad
- Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Tooran Nayeri Chegeni
- Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdol Sattar Pagheh
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Javad Javidnia
- Department of Medical Mycology, Invasive Fungi Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
38
|
Zhu W, Dong C, Wei L, Wang BZ. Promising Adjuvants and Platforms for Influenza Vaccine Development. Pharmaceutics 2021; 13:pharmaceutics13010068. [PMID: 33430259 PMCID: PMC7825707 DOI: 10.3390/pharmaceutics13010068] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023] Open
Abstract
Influenza is one of the major threats to public health. Current influenza vaccines cannot provide effective protection against drifted or shifted influenza strains. Researchers have considered two important strategies to develop novel influenza vaccines with improved immunogenicity and broader protective efficacy. One is applying fewer variable viral antigens, such as the haemagglutinin stalk domain. The other is including adjuvants in vaccine formulations. Adjuvants are promising and helpful boosters to promote more rapid and stronger immune responses with a dose-sparing effect. However, few adjuvants are currently licensed for human influenza vaccines, although many potential candidates are in different trials. While many advantages have been observed using adjuvants in influenza vaccine formulations, an improved understanding of the mechanisms underlying viral infection and vaccination-induced immune responses will help to develop new adjuvant candidates. In this review, we summarize the works related to adjuvants in influenza vaccine research that have been used in our studies and other laboratories. The review will provide perspectives for the utilization of adjuvants in developing next-generation and universal influenza vaccines.
Collapse
|
39
|
Inflammasome-Mediated Immunogenicity of Clinical and Experimental Vaccine Adjuvants. Vaccines (Basel) 2020; 8:vaccines8030554. [PMID: 32971761 PMCID: PMC7565252 DOI: 10.3390/vaccines8030554] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
In modern vaccines, adjuvants can be sophisticated immunological tools to promote robust and long-lasting protection against prevalent diseases. However, there is an urgent need to improve immunogenicity of vaccines in order to protect mankind from life-threatening diseases such as AIDS, malaria or, most recently, COVID-19. Therefore, it is important to understand the cellular and molecular mechanisms of action of vaccine adjuvants, which generally trigger the innate immune system to enhance signal transition to adaptive immunity, resulting in pathogen-specific protection. Thus, improved understanding of vaccine adjuvant mechanisms may aid in the design of “intelligent” vaccines to provide robust protection from pathogens. Various commonly used clinical adjuvants, such as aluminium salts, saponins or emulsions, have been identified as activators of inflammasomes - multiprotein signalling platforms that drive activation of inflammatory caspases, resulting in secretion of pro-inflammatory cytokines of the IL-1 family. Importantly, these cytokines affect the cellular and humoral arms of adaptive immunity, which indicates that inflammasomes represent a valuable target of vaccine adjuvants. In this review, we highlight the impact of different inflammasomes on vaccine adjuvant-induced immune responses regarding their mechanisms and immunogenicity. In this context, we focus on clinically relevant adjuvants that have been shown to activate the NLRP3 inflammasome and also present various experimental adjuvants that activate the NLRP3-, NLRC4-, AIM2-, pyrin-, or non-canonical inflammasomes and could have the potential to improve future vaccines. Together, we provide a comprehensive overview on vaccine adjuvants that are known, or suggested, to promote immunogenicity through inflammasome-mediated signalling.
Collapse
|
40
|
Cao P, Han FY, Grøndahl L, Xu ZP, Li L. Enhanced Oral Vaccine Efficacy of Polysaccharide-Coated Calcium Phosphate Nanoparticles. ACS OMEGA 2020; 5:18185-18197. [PMID: 32743193 PMCID: PMC7392379 DOI: 10.1021/acsomega.0c01792] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/06/2020] [Indexed: 05/21/2023]
Abstract
Oral administration of vaccines has been limited due to low immune response compared to parenteral administration. Antigen degradation in the acidic gastrointestinal environment (GI), mucus barriers, and inefficient cellular uptake by immune cells are the major challenges for oral vaccine delivery. To solve these issues, the current study investigates calcium phosphate nanoparticles (CaP NPs) coated with polysaccharides as nanocarriers for oral protein antigen delivery. In this design, the CaP NP core had an optimized antigen encapsulation capacity of 90 mg (BSA-FITC)/g (CaP NPs). The polysaccharides chitosan and alginate were coated onto the CaP NPs to protect the antigens against acidic degradation in the GI environment and enhance the immune response in the small intestine. The antigen release profiles showed that alginate-chitosan-coated CaP NPs prevented antigen release in a simulated gastric fluid (pH 1.2), followed by sustained release in simulated intestinal (pH 6.8) and colonic (pH 7.4) fluids. Cellular uptake and macrophage stimulation data revealed that the chitosan coating enhanced antigen uptake by intestine epithelia cells (Caco-2) and macrophages and improved surface expression of costimulatory molecules on macrophages. In vivo test further demonstrated that oral administration of alginate-chitosan-coated CaP@OVA NPs significantly enhanced the mucosal IgA and serum IgG antibody responses as compared to naked OVA, indicating that the CaP-Chi-Alg nanoparticle can potentially be used as a promising oral vaccine delivery system.
Collapse
Affiliation(s)
- Pei Cao
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Felicity Y. Han
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School
of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Lisbeth Grøndahl
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School
of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhi Ping Xu
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Li Li
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
41
|
Li M, Qin M, Song G, Deng H, Wang D, Wang X, Dai W, He B, Zhang H, Zhang Q. A biomimetic antitumor nanovaccine based on biocompatible calcium pyrophosphate and tumor cell membrane antigens. Asian J Pharm Sci 2020; 16:97-109. [PMID: 33613733 PMCID: PMC7878462 DOI: 10.1016/j.ajps.2020.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/12/2020] [Accepted: 06/28/2020] [Indexed: 12/18/2022] Open
Abstract
Currently, the cancer immunotherapy has made great progress while antitumor vaccine attracts substantial attention. Still, the selection of adjuvants as well as antigens are always the most crucial issues for better vaccination. In this study, we proposed a biomimetic antitumor nanovaccine based on biocompatible nanocarriers and tumor cell membrane antigens. Briefly, endogenous calcium pyrophosphate nanogranules with possible immune potentiating effect are designed and engineered, both as delivery vehicles and adjuvants. Then, these nanocarriers are coated with lipids and B16-OVA tumor cell membranes, so the biomembrane proteins can serve as tumor-specific antigens. It was found that calcium pyrophosphate nanogranules themselves were compatible and possessed adjuvant effect, while membrane proteins including tumor associated antigen were transferred onto the nanocarriers. It was demonstrated that such a biomimetic nanovaccine could be well endocytosed by dendritic cells, promote their maturation and antigen-presentation, facilitate lymph retention, and trigger obvious immune response. It was confirmed that the biomimetic vaccine could induce strong T-cell response, exhibit excellent tumor therapy and prophylactic effects, and simultaneously possess nice biocompatibility. In general, the present investigation might provide insights for the further design and application of antitumor vaccines.
Collapse
Affiliation(s)
- Minghui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Mengmeng Qin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ge Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hailiang Deng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Dakuan Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
42
|
Sadeghi Z, Fasihi-Ramandi M, Bouzari S. Nanoparticle-Based Vaccines for Brucellosis: Calcium Phosphate Nanoparticles-Adsorbed Antigens Induce Cross Protective Response in Mice. Int J Nanomedicine 2020; 15:3877-3886. [PMID: 32581535 PMCID: PMC7269176 DOI: 10.2147/ijn.s249942] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Introduction Vaccine formulation with appropriate adjuvants is an attractive approach to develop protective immunity against pathogens. Calcium phosphate nanoparticles (CaPNs) are considered as ideal adjuvants and delivery systems because of their great potential for enhancing immune responses. In the current study, we have designed nanoparticle-based vaccine candidates to induce immune responses and protection against B. melitensis and B. abortus. Materials and Methods For this purpose, we used three Brucella antigens (FliC, 7α-HSDH, BhuA) and two multi-epitopes (poly B and poly T) absorbed by CaPNs. The efficacy of each formulation was evaluated by measuring humoral, cellular and protective responses in immunized mice. Results The CaPNs showed an average size of about 90 nm with spherical shape and smooth surface. The CaPNs-adsorbed proteins displayed significant increase in cellular and humoral immune responses compared to the control groups. In addition, our results showed increased ratio of specific IgG2a (associated with Th1) to specific IgG1 (associated with Th2). Also, immunized mice with different vaccine candidate formulations were protected against B. melitensis 16M and B. abortus 544, and showed same levels of protection as commercial vaccines (B. melitensis Rev.1 and B. abortus RB51) except for BhuA-CaPNs. Discussion Our data support the hypothesis that these antigens absorbed with CaPNs could be effective vaccine candidates against B. melitensis and B. abortus.
Collapse
Affiliation(s)
- Zohre Sadeghi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
43
|
Alvaro-Lozano M, Akdis CA, Akdis M, Alviani C, Angier E, Arasi S, Arzt-Gradwohl L, Barber D, Bazire R, Cavkaytar O, Comberiati P, Dramburg S, Durham SR, Eifan AO, Forchert L, Halken S, Kirtland M, Kucuksezer UC, Layhadi JA, Matricardi PM, Muraro A, Ozdemir C, Pajno GB, Pfaar O, Potapova E, Riggioni C, Roberts G, Rodríguez Del Río P, Shamji MH, Sturm GJ, Vazquez-Ortiz M. EAACI Allergen Immunotherapy User's Guide. Pediatr Allergy Immunol 2020; 31 Suppl 25:1-101. [PMID: 32436290 PMCID: PMC7317851 DOI: 10.1111/pai.13189] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allergen immunotherapy is a cornerstone in the treatment of allergic children. The clinical efficiency relies on a well-defined immunologic mechanism promoting regulatory T cells and downplaying the immune response induced by allergens. Clinical indications have been well documented for respiratory allergy in the presence of rhinitis and/or allergic asthma, to pollens and dust mites. Patients who have had an anaphylactic reaction to hymenoptera venom are also good candidates for allergen immunotherapy. Administration of allergen is currently mostly either by subcutaneous injections or by sublingual administration. Both methods have been extensively studied and have pros and cons. Specifically in children, the choice of the method of administration according to the patient's profile is important. Although allergen immunotherapy is widely used, there is a need for improvement. More particularly, biomarkers for prediction of the success of the treatments are needed. The strength and efficiency of the immune response may also be boosted by the use of better adjuvants. Finally, novel formulations might be more efficient and might improve the patient's adherence to the treatment. This user's guide reviews current knowledge and aims to provide clinical guidance to healthcare professionals taking care of children undergoing allergen immunotherapy.
Collapse
Affiliation(s)
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cherry Alviani
- The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Newport, Isle of Wight, UK.,Clinical and Experimental Sciences and Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Elisabeth Angier
- Primary Care and Population Sciences, University of Southampton, Southampton, UK
| | - Stefania Arasi
- Pediatric Allergology Unit, Department of Pediatric Medicine, Bambino Gesù Children's research Hospital (IRCCS), Rome, Italy
| | - Lisa Arzt-Gradwohl
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| | - Domingo Barber
- School of Medicine, Institute for Applied Molecular Medicine (IMMA), Universidad CEU San Pablo, Madrid, Spain.,RETIC ARADYAL RD16/0006/0015, Instituto de Salud Carlos III, Madrid, Spain
| | - Raphaëlle Bazire
- Allergy Department, Hospital Infantil Niño Jesús, ARADyAL RD16/0006/0026, Madrid, Spain
| | - Ozlem Cavkaytar
- Department of Paediatric Allergy and Immunology, Faculty of Medicine, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Istanbul, Turkey
| | - Pasquale Comberiati
- Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Stephanie Dramburg
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Medical University, Berlin, Germany
| | - Stephen R Durham
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology, Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK.,the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Aarif O Eifan
- Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London and Royal Brompton Hospitals NHS Foundation Trust, London, UK
| | - Leandra Forchert
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Medical University, Berlin, Germany
| | - Susanne Halken
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Max Kirtland
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, UK
| | - Umut C Kucuksezer
- Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul University, Istanbul, Turkey
| | - Janice A Layhadi
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology, Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK.,the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.,Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, UK
| | - Paolo Maria Matricardi
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Medical University, Berlin, Germany
| | - Antonella Muraro
- The Referral Centre for Food Allergy Diagnosis and Treatment Veneto Region, Department of Women and Child Health, University of Padua, Padua, Italy
| | - Cevdet Ozdemir
- Institute of Child Health, Department of Pediatric Basic Sciences, Istanbul University, Istanbul, Turkey.,Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Turkey
| | | | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Ekaterina Potapova
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Medical University, Berlin, Germany
| | - Carmen Riggioni
- Pediatric Allergy and Clinical Immunology Service, Institut de Reserca Sant Joan de Deú, Barcelona, Spain
| | - Graham Roberts
- The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Newport, Isle of Wight, UK.,NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Paediatric Allergy and Respiratory Medicine (MP803), Clinical & Experimental Sciences & Human Development in Health Academic Units University of Southampton Faculty of Medicine & University Hospital Southampton, Southampton, UK
| | | | - Mohamed H Shamji
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology, Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK.,the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Gunter J Sturm
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| | | |
Collapse
|
44
|
Jensen‐Jarolim E, Bachmann MF, Bonini S, Jacobsen L, Jutel M, Klimek L, Mahler V, Mösges R, Moingeon P, O´Hehir RE, Palomares O, Pfaar O, Renz H, Rhyner C, Roth‐Walter F, Rudenko M, Savolainen J, Schmidt‐Weber CB, Traidl‐Hoffmann C, Kündig T. State-of-the-art in marketed adjuvants and formulations in Allergen Immunotherapy: A position paper of the European Academy of Allergy and Clinical Immunology (EAACI). Allergy 2020; 75:746-760. [PMID: 31774179 DOI: 10.1111/all.14134] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/07/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023]
Abstract
Since the introduction of allergen immunotherapy (AIT) over 100 years ago, focus has been on standardization of allergen extracts, with reliable molecular composition of allergens receiving the highest attention. While adjuvants play a major role in European AIT, they have been less well studied. In this Position Paper, we summarize current unmet needs of adjuvants in AIT citing current evidence. Four adjuvants are used in products marketed in Europe: aluminium hydroxide (Al(OH)3 ) is the most frequently used adjuvant, with microcrystalline tyrosine (MCT), monophosphoryl lipid A (MPLA) and calcium phosphate (CaP) used less frequently. Recent studies on humans, and using mouse models, have characterized in part the mechanisms of action of adjuvants on pre-existing immune responses. AIT differs from prophylactic vaccines that provoke immunity to infectious agents, as in allergy the patient is presensitized to the antigen. The intended mode of action of adjuvants is to simultaneously enhance the immunogenicity of the allergen, while precipitating the allergen at the injection site to reduce the risk of anaphylaxis. Contrasting immune effects are seen with different adjuvants. Aluminium hydroxide initially boosts Th2 responses, while the other adjuvants utilized in AIT redirect the Th2 immune response towards Th1 immunity. After varying lengths of time, each of the adjuvants supports tolerance. Further studies of the mechanisms of action of adjuvants may advise shorter treatment periods than the current three-to-five-year regimens, enhancing patient adherence. Improved lead compounds from the adjuvant pipeline are under development and are explored for their capacity to fill this unmet need.
Collapse
Affiliation(s)
- Erika Jensen‐Jarolim
- Institute of Pathophysiology & Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
- The Interuniversity Messerli Research Institute University of Veterinary Medicine Vienna Medical University of Vienna University of Vienna Vienna Austria
| | - Martin F. Bachmann
- Institute of Immunology Inselspital University of Berne Bern Switzerland
| | - Sergio Bonini
- Institute of Translational Pharmacology Italian National Research Council Rome Italy
| | - Lars Jacobsen
- ALC, Allergy Learning & Consulting Copenhagen Denmark
| | - Marek Jutel
- Department of Clinical Immunology Wroclaw Medical University Wrocław Poland
- ALL‐MED Medical Research Institute Wroclaw Poland
| | - Ludger Klimek
- Center of Rhinology and Allergology Wiesbaden Germany
| | - Vera Mahler
- Division of Allergology Paul‐Ehrlich‐Institut Federal Institute for Vaccines and Biomedicines Langen Germany
| | - Ralph Mösges
- CRI‐Clinical Research International Ltd Hamburg Germany
- Institute of Medical Statistics and Bioinformatics University of Cologne Cologne Germany
| | - Philippe Moingeon
- Center for Therapeutic Innovation – Immuno‐Inflammatory Disease Servier Suresnes France
| | - Robyn E. O´Hehir
- Department of Respiratory Medicine, Allergy and Clinical Immunology (Research) Central Clinical School Monash University and Alfred Hospital Melbourne Vic. Australia
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology Chemistry School Complutense University of Madrid Madrid Spain
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital MarburgPhilipps‐Universität Marburg Marburg Germany
| | - Harald Renz
- Institute of Laboratory Medicine Universities of Giessen and Marburg Lung Center (UGMLC) German Center for Lung Research (DZL) Philipps Universität Marburg Marburg Germany
| | - Claudio Rhyner
- SIAF – Swiss Institute of Allergy and Asthma Research Davos Switzerland
| | - Franziska Roth‐Walter
- The Interuniversity Messerli Research Institute University of Veterinary Medicine Vienna Medical University of Vienna University of Vienna Vienna Austria
| | | | - Johannes Savolainen
- Department of Pulmonary Diseases and Clinical Allergology University of Turku and Turku University Hospital Turku Finland
| | - Carsten B. Schmidt‐Weber
- Center of Allergy and Environment (ZAUM) German Center of Lung Research (DZL) and Helmholtz I&I Initiative Technical University, and Helmholtz Center Munich Munich Germany
| | - Claudia Traidl‐Hoffmann
- Institute of Environmental Medicine (IEM) Technical University Munich and Helmholtz Center Munich Munich Germany
| | - Thomas Kündig
- Department of Dermatology University Hospital Zurich Zurich Switzerland
| |
Collapse
|
45
|
Lung P, Yang J, Li Q. Nanoparticle formulated vaccines: opportunities and challenges. NANOSCALE 2020; 12:5746-5763. [PMID: 32124894 DOI: 10.1039/c9nr08958f] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Vaccines harness the inherent properties of the immune system to prevent diseases or treat existing ones. Continuous efforts have been devoted to both gaining a mechanistic understanding of how the immune system operates and designing vaccines with high efficacies and effectiveness. Advancements in nanotechnology in recent years have generated unique opportunities to meet the daunting challenges associated with immunology and vaccine development. Firstly, nanoparticle formulated systems provide ideal model systems for studying the operation of the immune system, making it possible to systematically identify key factors and understand their roles in specific immune responses. Also, the versatile compositions/architectures of nanoparticle systems enable new strategies/novel platforms for developing vaccines with high efficacies and effectiveness. In this review, we discuss the advantages of nanoparticles and the challenges faced during vaccine development, through the framework of the immunological mechanisms of vaccination, with the aim of bridging the gap between immunology and materials science, which are both involved in vaccine design. The knowledge obtained provides general guidelines for future vaccine development.
Collapse
Affiliation(s)
- Pingsai Lung
- Department of Physics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | | | | |
Collapse
|
46
|
Chi Q, Yang Z, Xu K, Wang C, Liang H. DNA Nanostructure as an Efficient Drug Delivery Platform for Immunotherapy. Front Pharmacol 2020; 10:1585. [PMID: 32063844 PMCID: PMC6997790 DOI: 10.3389/fphar.2019.01585] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/06/2019] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy has received increasing attention due to its low potential side effects and high specificity. For instance, cancer immunotherapy has achieved great success. CpG is a well-known and commonly used immunotherapeutic and vaccine adjuvant, but it has the disadvantage of being unstable and low in efficacy and needs to be transported through an effective nanocarrier. With perfect structural programmability, permeability, and biocompatibility, DNA nanostructures are one of the most promising candidates to deliver immune components to realize immunotherapy. However, the instability and low capability of the payload of ordinary DNA assemblies limit the relevant applications. Consequently, DNA nanostructure with a firm structure, high drug payloads is highly desirable. In the paper, the latest progress of biostable, high-payload DNA nanoassemblies of various structures, including cage-like DNA nanostructure, DNA particles, DNA polypods, and DNA hydrogel, are reviewed. Cage-like DNA structures hold drug molecules firmly inside the structure and leave a large space within the cavity. These DNA nanostructures use their unique structure to carry abundant CpG, and their biocompatibility and size advantages to enter immune cells to achieve immunotherapy for various diseases. Part of the DNA nanostructures can also achieve more effective treatment in conjunction with other functional components such as aPD1, RNA, TLR ligands.
Collapse
Affiliation(s)
- Qingjia Chi
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
- Hubei Key Laboratory of Theory and Application of Advanced Materials Mechanics, Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, China
| | - Zichang Yang
- Hubei Key Laboratory of Theory and Application of Advanced Materials Mechanics, Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, China
| | - Kang Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunli Wang
- “111” Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
47
|
Pagheh AS, Sarvi S, Gholami S, Asgarian-Omran H, Valadan R, Hassannia H, Ahmadpour E, Fasihi-Ramandie M, Dodangeh S, Hosseni-khah Z, Daryani A. Protective efficacy induced by DNA prime and recombinant protein boost vaccination with Toxoplasma gondii GRA14 in mice. Microb Pathog 2019; 134:103601. [DOI: 10.1016/j.micpath.2019.103601] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022]
|
48
|
Wallis J, Shenton DP, Carlisle RC. Novel approaches for the design, delivery and administration of vaccine technologies. Clin Exp Immunol 2019; 196:189-204. [PMID: 30963549 PMCID: PMC6468175 DOI: 10.1111/cei.13287] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2019] [Indexed: 12/20/2022] Open
Abstract
It is easy to argue that vaccine development represents humankind's most important and successful endeavour, such is the impact that vaccination has had on human morbidity and mortality over the last 200 years. During this time the original method of Jenner and Pasteur, i.e. that of injecting live-attenuated or inactivated pathogens, has been developed and supplemented with a wide range of alternative approaches which are now in clinical use or under development. These next-generation technologies have been designed to produce a vaccine that has the effectiveness of the original live-attenuated and inactivated vaccines, but without the associated risks and limitations. Indeed, the method of development has undoubtedly moved away from Pasteur's three Is paradigm (isolate, inactivate, inject) towards an approach of rational design, made possible by improved knowledge of the pathogen-host interaction and the mechanisms of the immune system. These novel vaccines have explored methods for targeted delivery of antigenic material, as well as for the control of release profiles, so that dosing regimens can be matched to the time-lines of immune system stimulation and the realities of health-care delivery in dispersed populations. The methods by which vaccines are administered are also the subject of intense research in the hope that needle and syringe dosing, with all its associated issues regarding risk of injury, cross-infection and patient compliance, can be replaced. This review provides a detailed overview of new vaccine vectors as well as information pertaining to the novel delivery platforms under development.
Collapse
Affiliation(s)
- J. Wallis
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| | - D. P. Shenton
- Defence Science and Technology LaboratoryPorton DownUK
| | - R. C. Carlisle
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| |
Collapse
|
49
|
A comparison between adjuvant and delivering functions of calcium phosphate, aluminum hydroxide and chitosan nanoparticles, using a model protein of Brucella melitensis Omp31. Immunol Lett 2019; 207:28-35. [DOI: 10.1016/j.imlet.2019.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 12/16/2022]
|
50
|
Galindo-Villegas J, García-Alcazar A, Meseguer J, Mulero V. Aluminum adjuvant potentiates gilthead seabream immune responses but induces toxicity in splenic melanomacrophage centers. FISH & SHELLFISH IMMUNOLOGY 2019; 85:31-43. [PMID: 29510253 DOI: 10.1016/j.fsi.2018.02.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/24/2018] [Accepted: 02/27/2018] [Indexed: 06/08/2023]
Abstract
A key goal of a successful vaccine formulation is the strong induction of persistent protective immune responses without producing side-effects. Adjuvants have been proved to be successful in several species at inducing increased immune responses against poorly immunogenic antigens. Fish are not the exception and promising results of adjuvanted vaccine formulations in many species are needed. In this study, over a period of 300 days, we characterized the apparent damage and immune response in gilthead seabream immunized by intraperitoneal injection with the model antigen keyhole limpet hemocyanin (KLH) alone or formulated with Montanide ISA water-in-oil (761 or 763), or Imject™ aluminum hydroxide (aluminium), as adjuvants. Throughout the trial, external tissue damage was examined visually, but no change was observed. Internally, severe adhesions, increased fat tissue, and hepatomegaly were recorded, but, without impairing animal health. At 120 days post priming (dpp), histopathological evaluations of head-kidney, spleen and liver revealed the presence of altered melanomacrophage centers (MMC) in HK and spleen, but not in liver. Surprisingly, in all aluminium treated fish, classical stains unmasked a toxic effect on splenic-MMC, unequivocally characterized by a strong cell depletion. Furthermore, at 170 dpp transmission electron microscopy confirmed this data. Paradoxically, at the same time powerful immune responses were recorded in most vaccinated groups, including the aluminium treatment. Whatever the case, despite the observed adhesions and MMC depletion, fish physiology was not affected, and most side-effects were resolved after 300 dpp. Therefore, our data support adjuvant inclusion, but strongly suggest that use of aluminium must be further explored in detail before it might benefit the rational design of new vaccination strategies in aquaculture.
Collapse
Affiliation(s)
- Jorge Galindo-Villegas
- Department of Cell Biology and Histology, Faculty of Biology, Institute of Biomedical Research of Murcia-Arrixaca, Campus Universitario de Espinardo, University of Murcia, 30100 Murcia, Spain.
| | | | - José Meseguer
- Department of Cell Biology and Histology, Faculty of Biology, Institute of Biomedical Research of Murcia-Arrixaca, Campus Universitario de Espinardo, University of Murcia, 30100 Murcia, Spain
| | - Victoriano Mulero
- Department of Cell Biology and Histology, Faculty of Biology, Institute of Biomedical Research of Murcia-Arrixaca, Campus Universitario de Espinardo, University of Murcia, 30100 Murcia, Spain.
| |
Collapse
|