1
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [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: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Tammas I, Bitchava K, Gelasakis AI. Advances in Vaccine Adjuvants for Teleost Fish: Implications for Aquatic Welfare and the Potential of Nanoparticle-Based Formulations. Vaccines (Basel) 2024; 12:1347. [PMID: 39772009 PMCID: PMC11679523 DOI: 10.3390/vaccines12121347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Vaccine adjuvants are crucial for reinforcing the immunogenicity of vaccines. Therefore, they are widely used in the aquaculture sector as vaccine components, facilitating the efficient prevention of infectious diseases and promoting sustainable teleost fish growth. Despite their benefits, there has been a growing concern about the potential adverse effects of vaccine adjuvants in teleost fish, connoting a valid impact on their overall health and welfare. Among the adjuvants used in aquaculture vaccinology, nanoparticle-based formulations have given rise to a promising new alternative to traditional options, such as oil-based emulsions and aluminum compounds, offering the benefit of minimizing relevant side effects. The aim of this paper was to review the current status of the adjuvants used in aquaculture, provide a description and an evaluation of their mode of action and side effects, and explore the potential use of nanoparticle formulations as adjuvants to improve the efficacy of aquaculture vaccines. By demonstrating and assessing the equilibrium between teleost fish welfare and immunological efficacy, this review presents a collective perspective that will assist in establishing a framework for the utilization of effective species-specific practices around adjuvant use in aquaculture, while also addressing the challenges of welfare-friendly immunization.
Collapse
Affiliation(s)
- Iosif Tammas
- Laboratory of Applied Hydrobiology, Department of Animal Science, Agricultural University of Athens, 11855 Athens, Greece;
| | - Konstantina Bitchava
- Laboratory of Applied Hydrobiology, Department of Animal Science, Agricultural University of Athens, 11855 Athens, Greece;
| | - Athanasios I. Gelasakis
- Laboratory of Anatomy & Physiology of Farm Animals, Agricultural University of Athens, 11855 Athens, Greece
| |
Collapse
|
3
|
Cui Y, Ho M, Hu Y, Shi Y. Vaccine adjuvants: current status, research and development, licensing, and future opportunities. J Mater Chem B 2024; 12:4118-4137. [PMID: 38591323 PMCID: PMC11180427 DOI: 10.1039/d3tb02861e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Vaccines represent one of the most significant inventions in human history and have revolutionized global health. Generally, a vaccine functions by triggering the innate immune response and stimulating antigen-presenting cells, leading to a defensive adaptive immune response against a specific pathogen's antigen. As a key element, adjuvants are chemical materials often employed as additives to increase a vaccine's efficacy and immunogenicity. For over 90 years, adjuvants have been essential components in many human vaccines, improving their efficacy by enhancing, modulating, and prolonging the immune response. Here, we provide a timely and comprehensive review of the historical development and the current status of adjuvants, covering their classification, mechanisms of action, and roles in different vaccines. Additionally, we perform systematic analysis of the current licensing processes and highlights notable examples from clinical trials involving vaccine adjuvants. Looking ahead, we anticipate future trends in the field, including the development of new adjuvant formulations, the creation of innovative adjuvants, and their integration into the broader scope of systems vaccinology and vaccine delivery. The article posits that a deeper understanding of biochemistry, materials science, and vaccine immunology is crucial for advancing vaccine technology. Such advancements are expected to lead to the future development of more effective vaccines, capable of combating emerging infectious diseases and enhancing public health.
Collapse
Affiliation(s)
- Ying Cui
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Megan Ho
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Yongjie Hu
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
4
|
Oladejo M, Tijani AO, Puri A, Chablani L. Adjuvants in cutaneous vaccination: A comprehensive analysis. J Control Release 2024; 369:475-492. [PMID: 38569943 DOI: 10.1016/j.jconrel.2024.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Skin is the body's largest organ and serves as a protective barrier from physical, thermal, and mechanical environmental challenges. Alongside, the skin hosts key immune system players, such as the professional antigen-presenting cells (APCs) like the Langerhans cells in the epidermis and circulating macrophages in the blood. Further, the literature supports that the APCs can be activated by antigen or vaccine delivery via multiple routes of administration through the skin. Once activated, the stimulated APCs drain to the associated lymph nodes and gain access to the lymphatic system. This further allows the APCs to engage with the adaptive immune system and activate cellular and humoral immune responses. Thus, vaccine delivery via skin offers advantages such as reliable antigen delivery, superior immunogenicity, and convenient delivery. Several preclinical and clinical studies have demonstrated the significance of vaccine delivery using various routes of administration via skin. However, such vaccines often employ adjuvant/(s), along with the antigen of interest. Adjuvants augment the immune response to a vaccine antigen and improve the therapeutic efficacy. Due to these reasons, adjuvants have been successfully used with infectious disease vaccines, cancer immunotherapy, and immune-mediated diseases. To capture these developments, this review will summarize preclinical and clinical study results of vaccine delivery via skin in the presence of adjuvants. A focused discussion regarding the FDA-approved adjuvants will address the experiences of using such adjuvant-containing vaccines. In addition, the challenges and regulatory concerns with these adjuvants will be discussed. Finally, the review will share the prospects of adjuvant-containing vaccines delivered via skin.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA.
| | - Lipika Chablani
- Wegmans School of Pharmacy, St. John Fisher University, 3690 East Ave, Rochester, NY 14618, USA.
| |
Collapse
|
5
|
Gurunathan S, Thangaraj P, Wang L, Cao Q, Kim JH. Nanovaccines: An effective therapeutic approach for cancer therapy. Biomed Pharmacother 2024; 170:115992. [PMID: 38070247 DOI: 10.1016/j.biopha.2023.115992] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Cancer vaccines hold considerable promise for the immunotherapy of solid tumors. Nanomedicine offers several strategies for enhancing vaccine effectiveness. In particular, molecular or (sub) cellular vaccines can be delivered to the target lymphoid tissues and cells by nanocarriers and nanoplatforms to increase the potency and durability of antitumor immunity and minimize negative side effects. Nanovaccines use nanoparticles (NPs) as carriers and/or adjuvants, offering the advantages of optimal nanoscale size, high stability, ample antigen loading, high immunogenicity, tunable antigen presentation, increased retention in lymph nodes, and immunity promotion. To induce antitumor immunity, cancer vaccines rely on tumor antigens, which are administered in the form of entire cells, peptides, nucleic acids, extracellular vesicles (EVs), or cell membrane-encapsulated NPs. Ideal cancer vaccines stimulate both humoral and cellular immunity while overcoming tumor-induced immune suppression. Herein, we review the key properties of nanovaccines for cancer immunotherapy and highlight the recent advances in their development based on the structure and composition of various (including synthetic and semi (biogenic) nanocarriers. Moreover, we discuss tumor cell-derived vaccines (including those based on whole-tumor-cell components, EVs, cell membrane-encapsulated NPs, and hybrid membrane-coated NPs), nanovaccine action mechanisms, and the challenges of immunocancer therapy and their translation to clinical applications.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India.
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India
| | - Lin Wang
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Qilong Cao
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
6
|
Ekwemalor K, Asiamah E, Adjei-Fremah S, Eluka-Okoludoh E, Mulakala B, Osei B, Worku M. Diverse pathogen-associated molecular patterns affect transcription of genes in the toll-like receptor signaling pathway in goat blood. Anim Biotechnol 2023; 34:3729-3738. [PMID: 37226739 DOI: 10.1080/10495398.2023.2214189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS), peptidoglycan (PGN), Polyinosinic-polycytidylic acid (poly I:C), and CpG Oligodeoxynucleotides (ODN) are recognized by Toll-like receptors (TLR). This study aimed to investigate the effect of diverse PAMPs on the transcription of TLR signaling pathway genes in goat blood. Whole blood was collected from 3 female BoerXSpanish goats and treated with the following PAMPs: 10 µg/ml LPS, PGN, CpG ODN (2216), CpG ODN (2006), and 12.5 µg/ml Poly I:C. Blood-treated PBS served as a control. The expression of 84 genes in the human TLR signaling pathway RT2 PCR Array (Qiagen) was evaluated using real-time PCR. Treatment with PBS affected the expression of 74 genes, Poly I:C affected the expression of 40 genes, t ODN 2006 affected the expression of 50 genes, ODN 2216 affected the expression of 52 genes, LPS affected the expression of 49 genes, while PGN affected the expression of 49 genes. Our results show that PAMPs modulated and increased the expression of genes in the TLR signaling pathway. These results highlight important insights into how the host responds to different pathogens and may help design adjuvants for therapeutics and vaccines that target different.
Collapse
Affiliation(s)
- Kingsley Ekwemalor
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Emmanuel Asiamah
- Department of Agriculture, University of Arkansas at Pine Bluff, Pine Bluff, AR, USA
| | - Sarah Adjei-Fremah
- Department of Biological Sciences, Winston-Salem State University, Winston-Salem, NC, USA
| | - Eboghoye Eluka-Okoludoh
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Bharath Mulakala
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Bertha Osei
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Mulumebet Worku
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| |
Collapse
|
7
|
Zhu J, Liu J, Yan C, Wang D, Pan W. Trained immunity: a cutting edge approach for designing novel vaccines against parasitic diseases? Front Immunol 2023; 14:1252554. [PMID: 37868995 PMCID: PMC10587610 DOI: 10.3389/fimmu.2023.1252554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
The preventive situation of parasitosis, a global public health burden especially for developing countries, is not looking that good. Similar to other infections, vaccines would be the best choice for preventing and controlling parasitic infection. However, ideal antigenic molecules for vaccine development have not been identified so far, resulting from the complicated life history and enormous genomes of the parasites. Furthermore, the suppression or down-regulation of anti-infectious immunity mediated by the parasites or their derived molecules can compromise the effect of parasitic vaccines. Comparing the early immune profiles of several parasites in the permissive and non-permissive hosts, a robust innate immune response is proposed to be a critical event to eliminate the parasites. Therefore, enhancing innate immunity may be essential for designing novel and effective parasitic vaccines. The newly emerging trained immunity (also termed innate immune memory) has been increasingly recognized to provide a novel perspective for vaccine development targeting innate immunity. This article reviews the current status of parasitic vaccines and anti-infectious immunity, as well as the conception, characteristics, and mechanisms of trained immunity and its research progress in Parasitology, highlighting the possible consideration of trained immunity in designing novel vaccines against parasitic diseases.
Collapse
Affiliation(s)
- Jinhang Zhu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiaxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dahui Wang
- Liangshan College (Li Shui) China, Lishui University, Lishui, Zhejiang, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
8
|
da Silva JGL, Gonçalves AAM, Oliveira LT, Garcia GM, Batista MA, de Mendonça LZ, Viana KF, Sant’Ana RDCO, Melo Júnior OADO, Silveira-Lemos D, Dutra WO, Martins-Filho OA, Galdino AS, de Moura SAL, Mosqueira VCF, Giunchetti RC. Polymeric Delivery Systems as a Potential Vaccine against Visceral Leishmaniasis: Formulation Development and Immunogenicity. Vaccines (Basel) 2023; 11:1309. [PMID: 37631877 PMCID: PMC10459565 DOI: 10.3390/vaccines11081309] [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: 04/26/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/27/2023] Open
Abstract
Recent studies suggest that the association of antigens in microparticles increases the anti-Leishmania vaccine immunogenicity. This study aims to investigate the in situ effect of the adjuvant performance consisting of chitosan-coated poly(D,L-lactic) acid submicrometric particles (SMP) and analyze the inflammatory profile and toxicity. Two formulations were selected, SMP1, containing poly(D,L-lactide) (PLA) 1% wt/v and chitosan 1% wt/v; and SMP2, containing PLA 5% wt/v and chitosan 5% wt/v. After a single dose of the unloaded SMP1 or SMP2 in mice, the SMPs promoted cell recruitment without tissue damage. In addition, besides the myeloperoxidase (MPO) activity having demonstrated similar results among the analyzed groups, a progressive reduction in the levels of N-acetyl-β-D-glucosaminidase (NAG) until 72 h was observed for SMPs. While IL-6 levels were similar among all the analyzed groups along the kinetics, only the SMPs groups had detectable levels of TNF-α. Additionally, the Leishmania braziliensis antigen was encapsulated in SMPs (SMP1Ag and SMP2Ag), and mice were vaccinated with three doses. The immunogenicity analysis by flow cytometry demonstrated a reduction in NK (CD3-CD49+) cells in all the SMPs groups, in addition to impairment in the T cells subsets (CD3+CD4+) and CD3+CD8+) and B cells (CD19+) of the SMP2 group. The resulting data demonstrate that the chitosan-coated SMP formulations stimulate the early events of an innate immune response, suggesting their ability to increase the immunogenicity of co-administered Leishmania antigens.
Collapse
Affiliation(s)
- João Guilherme Lino da Silva
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
- Nucleus for Research in Biological Sciences (NUPEB), Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil
| | - Ana Alice Maia Gonçalves
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
| | - Liliam Teixeira Oliveira
- Laboratory of Pharmaceutics and Nanotechnology (LDGNano), School of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.T.O.); (G.M.G.); (V.C.F.M.)
| | - Giani Martins Garcia
- Laboratory of Pharmaceutics and Nanotechnology (LDGNano), School of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.T.O.); (G.M.G.); (V.C.F.M.)
| | - Maurício Azevedo Batista
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
| | - Ludmila Zanandreis de Mendonça
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
| | - Kelvinson Fernandes Viana
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
| | - Rita de Cássia Oliveira Sant’Ana
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
| | - Otoni Alves de Oliveira Melo Júnior
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
| | - Denise Silveira-Lemos
- Integrated Research Group on Biomarkers, René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte 30190-009, Brazil;
| | - Walderez Ornelas Dutra
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
- National Institute of Science and Technology in Tropical Diseases, INCT-DT, Salvador 40110-060, Brazil
| | - Olindo Assis Martins-Filho
- Laboratory of Diagnosis and Monitoring Biomarkers, René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte 30190-009, Brazil;
| | - Alexsandro Sobreira Galdino
- Laboratory of Microorganism Biotechnology, Federal University of São João Del-Rei (UFSJ), Midwest Campus, Divinópolis 35501-296, Brazil;
| | - Sandra Aparecida Lima de Moura
- Laboratory of Biomaterials and Experimental Pathology, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35402-136, Brazil;
| | - Vanessa Carla Furtado Mosqueira
- Laboratory of Pharmaceutics and Nanotechnology (LDGNano), School of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (L.T.O.); (G.M.G.); (V.C.F.M.)
| | - Rodolfo Cordeiro Giunchetti
- Laboratory of Biology of Cell Interactions, Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (J.G.L.d.S.); (A.A.M.G.); (M.A.B.); (L.Z.d.M.); (K.F.V.); (R.d.C.O.S.); (O.A.d.O.M.J.); (W.O.D.)
- National Institute of Science and Technology in Tropical Diseases, INCT-DT, Salvador 40110-060, Brazil
| |
Collapse
|
9
|
Thanvi R, Nada S, Dissanayake R, Vartak A, Sebilleau CO, Alom NE, Prestwich EG, Wall KA, Sucheck SJ. Synthesis and Evaluation of a Self-Adjuvanting Pseudomonal Vaccine Based on Major Outer Membrane Porin OprF Epitopes Formulated with Low-Toxicity QS-21-Containing Liposomes. Bioconjug Chem 2023; 34:893-910. [PMID: 37092892 PMCID: PMC10723056 DOI: 10.1021/acs.bioconjchem.3c00103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Pseudomonas aeruginosa (PA) is a Gram-negative pathogen that the World Health Organization has ranked as a priority 1 (critical) threat. One potential prophylactic approach to preventing or reducing the incidence of PA would be development of a long sought-after vaccine. Both antibody and CD4+ T-cell responses have been noted as playing key roles in protection against infection. In these studies, we have designed a prototype vaccine consisting of several known linear B-cell epitopes derived from an outer membrane porin F (OprF). The resulting thiol-containing protein was conjugated to a version of the lipopeptide-based Toll-like receptor agonist Pam3CysSK4Mal (10) containing a maleimide moiety and formulated into dipalmitoylphosphatidylcholine (DPPC)/cholesterol (Chol) liposomes. Mice immunized with the resulting vaccine generated antibodies that bound PA14 (serotype O10) in vitro and induced opsonization in the presence of rabbit complement and murine macrophage RAW264.7 cells. The liposome was optimized to contain 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1,2-dimyristoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (DMPG), Chol, Pam3CysSK4-OprF (12) and the Quillaja saponaria-derived saponin adjuvant QS-21. The resulting vaccine formulation produced significantly higher antibody titers, increased the IgG2a antibody isotype, and increased the number of IgG-producing B-cells as well as splenic primed T-cells. In summary, the liposomal vaccine platform was found highly useful for the generation of a robust and balanced TH1/TH2 response.
Collapse
Affiliation(s)
- Radhika Thanvi
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Shadia Nada
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43614, United States
| | - Ravindika Dissanayake
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43614, United States
| | - Abhishek Vartak
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Chloé Olayinka Sebilleau
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Nur-E Alom
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| | - Erin G Prestwich
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43614, United States
| | - Katherine A Wall
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43614, United States
| | - Steven J Sucheck
- Department of Chemistry and Biochemistry, University of Toledo, 2801 West Bancroft Street, Toledo, Ohio 43606, United States
| |
Collapse
|
10
|
Naletova I, Tomasello B, Attanasio F, Pleshkan VV. Prospects for the Use of Metal-Based Nanoparticles as Adjuvants for Local Cancer Immunotherapy. Pharmaceutics 2023; 15:1346. [PMID: 37242588 PMCID: PMC10222518 DOI: 10.3390/pharmaceutics15051346] [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: 02/28/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Immunotherapy is among the most effective approaches for treating cancer. One of the key aspects for successful immunotherapy is to achieve a strong and stable antitumor immune response. Modern immune checkpoint therapy demonstrates that cancer can be defeated. However, it also points out the weaknesses of immunotherapy, as not all tumors respond to therapy and the co-administration of different immunomodulators may be severely limited due to their systemic toxicity. Nevertheless, there is an established way through which to increase the immunogenicity of immunotherapy-by the use of adjuvants. These enhance the immune response without inducing such severe adverse effects. One of the most well-known and studied adjuvant strategies to improve immunotherapy efficacy is the use of metal-based compounds, in more modern implementation-metal-based nanoparticles (MNPs), which are exogenous agents that act as danger signals. Adding innate immune activation to the main action of an immunomodulator makes it capable of eliciting a robust anti-cancer immune response. The use of an adjuvant has the peculiarity of a local administration of the drug, which positively affects its safety. In this review, we will consider the use of MNPs as low-toxicity adjuvants for cancer immunotherapy, which could provide an abscopal effect when administered locally.
Collapse
Affiliation(s)
- Irina Naletova
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, V.le Andrea Doria 6, 95125 Catania, Italy
| | - Francesco Attanasio
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Victor V. Pleshkan
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| |
Collapse
|
11
|
The Use of an Adjuvant System Improves Innate and Adaptive Immune Response When Associated with a Leishmania ( Viannia) braziliensis Antigen in a Vaccine Candidate against L. ( Leishmania) infantum Infection. Vaccines (Basel) 2023; 11:vaccines11020395. [PMID: 36851272 PMCID: PMC9962147 DOI: 10.3390/vaccines11020395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND The adjuvants' optimal dose and the administration route can directly influence the epitope recognition patterns and profiles of innate response. We aimed to establish the effect and the optimal dose of adjuvant systems for proposing a vaccine candidate to be employed with Leishmania (Viannia) braziliensis. METHODS We evaluated the adjuvants saponin (SAP), monophosphoryl lipid A (MPL) and resiquimod (R-848) isolated and combined as adjuvant systems in a lower dose corresponding to 25%, 33%, and 50% of each adjuvant total dose. Male outbred BALB/c mice were divided into 13 groups, SAP, MPL, and R-848 isolated, and the adjuvant systems SAP plus MPL (SM), SAP plus R-848 (SR), and MPL plus R-848 (MR). RESULTS SM50 increased levels of all chemokines analyzed and TNF production, while it presented an increased inflammatory cell infiltrate in the skin with macrophage recruitment. Thus, we proposed a vaccine candidate employing L. (V.) braziliensis antigen associated with the SM adjuvant system against experimental L. (Leishmania) infantum challenge. We observed a significant increase in the frequency of cells expressing the central and effector memory CD4+ T cells phenotype in immunized mice with the LBSM50. In the liver, there was a decreased parasite load when mice received LBSM50. CONCLUSIONS When combined with L. (V.) braziliensis antigen, SM50 increases TNF and IFN-γ, which generates central and effector memory CD4+ T cells. Therefore, using an adjuvant system can promote an effective innate immune response with the potential to compose future vaccines.
Collapse
|
12
|
Shi T, Sun M, Lu C, Meng F. Self-assembled nanoparticles: A new platform for revolutionizing therapeutic cancer vaccines. Front Immunol 2023; 14:1125253. [PMID: 36895553 PMCID: PMC9988954 DOI: 10.3389/fimmu.2023.1125253] [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: 12/16/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Cancer vaccines have had some success in the past decade. Based on in-depth analysis of tumor antigen genomics, many therapeutic vaccines have already entered clinical trials for multiple cancers, including melanoma, lung cancer, and head and neck squamous cell carcinoma, which have demonstrated impressive tumor immunogenicity and antitumor activity. Recently, vaccines based on self-assembled nanoparticles are being actively developed as cancer treatment, and their feasibility has been confirmed in both mice and humans. In this review, we summarize recent therapeutic cancer vaccines based on self-assembled nanoparticles. We describe the basic ingredients for self-assembled nanoparticles, and how they enhance vaccine immunogenicity. We also discuss the novel design method for self-assembled nanoparticles that pose as a promising delivery platform for cancer vaccines, and the potential in combination with multiple therapeutic approaches.
Collapse
Affiliation(s)
- Tianyu Shi
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Mengna Sun
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Changchang Lu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
13
|
Kwak HW, Hong SH, Park HJ, Park HJ, Bang YJ, Kim JY, Lee YS, Bae SH, Yoon H, Nam JH. Adjuvant effect of IRES-based single-stranded RNA on melanoma immunotherapy. BMC Cancer 2022; 22:1041. [PMID: 36199130 PMCID: PMC9533600 DOI: 10.1186/s12885-022-10140-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Background Adjuvant therapies such as radiation therapy, chemotherapy, and immunotherapy are usually given after cancer surgery to improve the survival of cancer patients. However, despite advances in several adjuvant therapies, they are still limited in the prevention of recurrences. Methods We evaluated the immunological effects of RNA-based adjuvants in a murine melanoma model. Single-stranded RNA (ssRNA) were constructed based on the cricket paralysis virus (CrPV) internal ribosome entry site (IRES). Populations of immune cells in bone marrow cells and lymph node cells following immunization with CrPVIRES-ssRNA were determined using flow cytometry. Activated cytokine levels were measured using ELISA and ELISpot. The tumor protection efficacy of CrPVIRES-ssRNA was analyzed based on any reduction in tumor size or weight, and overall survival. Results CrPVIRES-ssRNA treatment stimulated antigen-presenting cells in the drain lymph nodes associated with activated antigen-specific dendritic cells. Next, we evaluated the expression of CD40, CD86, and XCR1, showing that immunization with CrPVIRES-ssRNA enhanced antigen presentation by CD8a+ conventional dendritic cell 1 (cDC1), as well as activated antigen-specific CD8 T cells. In addition, CrPVIRES-ssRNA treatment markedly increased the frequency of antigen-specific CD8 T cells and interferon-gamma (IFN-γ) producing cells, which promoted immune responses and reduced tumor burden in melanoma-bearing mice. Conclusions This study provides evidence that the CrPVIRES-ssRNA adjuvant has potential for use in therapeutic cancer vaccines. Moreover, CrPVIRES-ssRNA possesses protective effects on various cancer cell models. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10140-2.
Collapse
Affiliation(s)
- Hye Won Kwak
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea
| | - So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Hyeong-Jun Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea
| | - Yoo-Jin Bang
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea
| | - Jae-Yong Kim
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea
| | - Yu-Sun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Seo-Hyeon Bae
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea.,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Hyunho Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea. .,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43-1 Yeokgok-dong, Wonmi-gu, Bucheon, 14662, Republic of Korea. .,BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea. .,, SML biopharm, Gyeonggi-do, Gwangmyeong, Republic of Korea.
| |
Collapse
|
14
|
Liao Z, Huang J, Lo PC, Lovell JF, Jin H, Yang K. Self-adjuvanting cancer nanovaccines. J Nanobiotechnology 2022; 20:345. [PMID: 35883176 PMCID: PMC9316869 DOI: 10.1186/s12951-022-01545-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/04/2022] [Indexed: 11/12/2022] Open
Abstract
Nanovaccines, a new generation of vaccines that use nanoparticles as carriers and/or adjuvants, have been widely used in the prevention and treatment of various diseases, including cancer. Nanovaccines have sparked considerable interest in cancer therapy due to a variety of advantages, including improved access to lymph nodes (LN), optimal packing and presentation of antigens, and induction of a persistent anti-tumor immune response. As a delivery system for cancer vaccines, various types of nanoparticles have been designed to facilitate the delivery of antigens and adjuvants to lymphoid organs and antigen-presenting cells (APCs). Particularly, some types of nanoparticles are able to confer an immune-enhancing capability and can themselves be utilized for adjuvant-like effect for vaccines, suggesting a direction for a better use of nanomaterials and the optimization of cancer vaccines. However, this role of nanoparticles in vaccines has not been well studied. To further elucidate the role of self-adjuvanting nanovaccines in cancer therapy, we review the mechanisms of antitumor vaccine adjuvants with respect to nanovaccines with self-adjuvanting properties, including enhancing cross-presentation, targeting signaling pathways, biomimicking of the natural invasion process of pathogens, and further unknown mechanisms. We surveyed self-adjuvanting cancer nanovaccines in clinical research and discussed their advantages and challenges. In this review, we classified self-adjuvanting cancer nanovaccines according to the underlying immunomodulatory mechanism, which may provide mechanistic insights into the design of nanovaccines in the future.
Collapse
Affiliation(s)
- Zhiyun Liao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
15
|
Autoimmune/autoinflammatory syndrome induced by adjuvants: a focus on silicone. Clin Rheumatol 2022; 41:3275-3283. [PMID: 35798923 DOI: 10.1007/s10067-022-06274-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/30/2022] [Accepted: 06/29/2022] [Indexed: 11/03/2022]
Abstract
Autoimmune/autoinflammatory syndrome induced by adjuvants (ASIA) was first described in 2011 to cover disorders characterized by dysregulation of the immune system after exposure to an adjuvant. In the present review, the authors focus on silicone-induced ASIA. In the last two decades, there has been worldwide increase in the use of silicone breast implant (SBI) as an aesthetic procedure, raising concerns for possible effects on the immune system, especially in people who already have previous immune dysregulation. The authors did a critical review of the most important articles referring to silicone-induced ASIA, including most recent studies regarding physiopathologic mechanism. Despite large-scale epidemiological studies conducted to assess the association between SBI and autoimmune/rheumatic disorders, the results remain inconclusive, and the debate over the safety of SBIs remains heated. The explantation of silicone breast has been indicated for silicone-induced ASIA with improvement of unspecific symptoms in the majority of patients; however, the outcome seems different in patients with definitive autoimmune rheumatic disease (AIRD). There is no prospective study evaluating the risk of flares after SBI in patients who already have an AIRD. Therefore, based on the literature, we cannot contraindicate the procedure; however, we need to advertise about the risk of ASIA to the patients with AIRD. Long-term safety and implant-related outcomes should be discussed with these patients, considering each case individually, assessing genetic and environmental factors, and determining if the autoimmune disease is in remission or not, for shared decision among patient and the physician.
Collapse
|
16
|
Ali Dahhas M, Alsenaidy MA. Role of site-directed mutagenesis and adjuvants in the stability and potency of anthrax protective antigen. Saudi Pharm J 2022; 30:595-604. [PMID: 35693445 PMCID: PMC9177452 DOI: 10.1016/j.jsps.2022.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/21/2022] [Indexed: 11/03/2022] Open
Abstract
Anthrax is a zoonotic infection caused by the gram-positive, aerobic, spore-forming bacterium Bacillus anthracis. Depending on the origin of the infection, serious health problems or mortality is possible. The virulence of B. anthracis is reliant on three pathogenic factors, which are secreted upon infection: protective antigen (PA), lethal factor (LF), and edema factor (EF). Systemic illness results from LF and EF entering cells through the formation of a complex with the heptameric form of PA, bound to the membrane of infected cells through its receptor. The currently available anthrax vaccines have multiple drawbacks, and recombinant PA is considered a promising second-generation vaccine candidate. However, the inherent chemical instability of PA through Asn deamidation at multiple sites prevents its use after long-term storage owing to loss of potency. Moreover, there is a distinct possibility of B. anthracis being used as a bioweapon; thus, the developed vaccine should remain efficacious and stable over the long-term. Second-generation anthrax vaccines with appropriate adjuvant formulations for enhanced immunogenicity and safety are desired. In this article, using protein engineering approaches, we have reviewed the stabilization of anthrax vaccine candidates that are currently licensed or under preclinical and clinical trials. We have also proposed a formulation to enhance recombinant PA vaccine potency via adjuvant formulation.
Collapse
|
17
|
Shah SM, Alsaab HO, Rawas-Qalaji MM, Uddin MN. A Review on Current COVID-19 Vaccines and Evaluation of Particulate Vaccine Delivery Systems. Vaccines (Basel) 2021; 9:vaccines9101086. [PMID: 34696194 PMCID: PMC8540464 DOI: 10.3390/vaccines9101086] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/21/2022] Open
Abstract
First detected in Wuhan, China, a highly contagious coronavirus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), also known as COVID-19, spread globally in December of 2019. As of 19 September 2021, approximately 4.5 million people have died globally, and 215 million active cases have been reported. To date, six vaccines have been developed and approved for human use. However, current production and supply capabilities are unable to meet global demands to immunize the entire world population. Only a few countries have been able to successfully vaccinate many of their residents. Therefore, an alternative vaccine that can be prepared in an easy and cost-effective manner is urgently needed. A vaccine that could be prepared in this manner, as well as can be preserved and transported at room temperature, would be of great benefit to public health. It is possible to develop such an alternative vaccine by using nano- or microparticle platforms. These platforms address most of the existing vaccine limitations as they are stable at room temperature, are inexpensive to produce and distribute, can be administered orally, and do not require cold chain storage for transportation or preservation. Particulate vaccines can be administered as either oral solutions or in sublingual or buccal film dosage forms. Besides improved patient compliance, the major advantage of oral, sublingual, and buccal routes of administration is that they can elicit mucosal immunity. Mucosal immunity, along with systemic immunity, can be a strong defense against SARS-CoV-2 as the virus enters the system through inhalation or saliva. This review discusses the possibility to produce a particulate COVID vaccine by using nano- or microparticles as platforms for oral administration or in sublingual or buccal film dosage forms in order to accelerate global vaccination.
Collapse
Affiliation(s)
- Sarthak M. Shah
- College of Pharmacy, Mercer University, Atlanta, GA 31207, USA;
| | - Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Mutasem M. Rawas-Qalaji
- College of Pharmacy, University of Sharjah, Sharjah 26666, United Arab Emirates;
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 26666, United Arab Emirates
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 27272, USA
| | - Mohammad N. Uddin
- College of Pharmacy, Mercer University, Atlanta, GA 31207, USA;
- Correspondence: ; Tel.: +1-678-547-6224
| |
Collapse
|
18
|
Bacterial Outer Membrane Vesicles as a Versatile Tool in Vaccine Research and the Fight against Antimicrobial Resistance. mBio 2021; 12:e0170721. [PMID: 34372691 PMCID: PMC8406158 DOI: 10.1128/mbio.01707-21] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gram-negative bacteria include a number of pathogens that cause disease in humans and animals. Although antibiotics are still effective in treating a considerable range of infections caused by Gram-negative bacteria, the alarming increase of antimicrobial resistance (AMR) induced by excessive use of antibiotics has raised global concerns. Therefore, alternative strategies must be developed to prevent and treat bacterial infections and prevent the advent of a postantibiotic era. Vaccines, one of the greatest achievements in the history of medical science, hold extraordinary potential to prevent bacterial infections and thereby reduce the need for antibiotics. Novel bacterial vaccines are urgently needed, however, and outer membrane vesicles (OMVs), naturally produced by Gram-negative bacteria, represent a promising and versatile tool that can be employed as adjuvants, antigens, and delivery platforms in the development of vaccines against Gram-negative bacteria. Here, we provide an overview of the many roles OMVs can play in vaccine development and the mechanisms behind these applications. Methods to improve OMV yields and a comparison of different strategies for OMV isolation aiming at cost-effective production of OMV-based vaccines are also reviewed.
Collapse
|
19
|
Bachour Y, Kadouch JA, Niessen FB. The Aetiopathogenesis of Late Inflammatory Reactions (LIRs) After Soft Tissue Filler Use: A Systematic Review of the Literature. Aesthetic Plast Surg 2021; 45:1748-1759. [PMID: 33913021 PMCID: PMC8316155 DOI: 10.1007/s00266-021-02306-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 04/11/2021] [Indexed: 11/03/2022]
Abstract
Background Late inflammatory reactions (LIRs) are the most challenging complications after filler use. The immune system plays a prominent role in its etiology, albeit to an unknown extent. Bacterial contamination in situ has been hypothesized to be causative for LIRs. How this relates to the immunological processes involved is unknown. This article aims to provide an overview of immunological and bacterial factors involved in development of LIRs. Methods We undertook a systematic literature review focused on immunological factors and microbiota in relation to LIRs after filler use. This systematic review was performed in accordance with the PRISMA guidelines. PubMed, EMBASE and the Cochrane databases were searched from inception up to August 2019. Included studies were assessed for the following variables: subject characteristics, number of patients, primary indication for filler injection, implant type/amount and injection site, type of complication, follow-up or injection duration, study methods, type of antibiotics or medical therapies and outcomes related to microbiota and immunological factors. Results Data on immunological factors and bacterial contamination were retrieved from 21 included studies. Notably, the presence of histocytes, giant cells and Staphylococcus epidermidis within biopsies were often associated with LIRs. Conclusion This review provides a clear overview of the immunological factors associated with LIRs and provides a hypothetical immunological model for development of the disease. Furthermore, an overview of bacterial contamination and associations with LIRs has been provided. Follow-up research may result in clinical recommendations to prevent LIRs. Level of Evidence III This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors-www.springer.com/00266.. Supplementary Information The online version contains supplementary material available at 10.1007/s00266-021-02306-3.
Collapse
|
20
|
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
|
21
|
Kardani K, Sadat SM, Kardani M, Bolhassani A. The next generation of HCV vaccines: a focus on novel adjuvant development. Expert Rev Vaccines 2021; 20:839-855. [PMID: 34114513 DOI: 10.1080/14760584.2021.1941895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Considerable efforts have been made to treat and prevent acute and chronic infections caused by the hepatitis C virus (HCV). Current treatments are unable to protect people from reinfection. Hence, there is a need for development of both preventive and therapeutic HCV vaccines. Many vaccine candidates are in development to fight against HCV, but their efficacy has so far proven limited partly due to low immunogenicity. AREAS COVERED We explore development of novel and powerful adjuvants to achieve an effective HCV vaccine. The basis for developing strong adjuvants is to understand the innate immunity pathway, which subsequently stimulates humoral and cellular immune responses. We have also investigated immunogenicity of developed adjuvants that have been used in recent studies available in online databases such as PubMed, PMC, ScienceDirect, Google Scholar, etc. EXPERT OPINION Adjuvants are used as a part of vaccine formulation to boost vaccine immunogenicity and antigen delivery. Several FDA-approved adjuvants are used in licensed human vaccines. Unfortunately, no adjuvant has yet been proven to boost HCV immune responses to the extent needed for an effective vaccine. One of the promising approaches for developing an effective adjuvant is the combination of various adjuvants to trigger several innate immune responses, leading to activation of adaptive immunity.[Figure: see text].
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Mona Kardani
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
22
|
Decates T, Kadouch J, Velthuis P, Rustemeyer T. Immediate nor Delayed Type Hypersensitivity Plays a Role in Late Inflammatory Reactions After Hyaluronic Acid Filler Injections. Clin Cosmet Investig Dermatol 2021; 14:581-589. [PMID: 34103958 PMCID: PMC8178514 DOI: 10.2147/ccid.s312198] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/12/2021] [Indexed: 12/02/2022]
Abstract
Purpose The exact etiology of late inflammatory reactions (LIRs) to hyaluronic acid (HA) fillers is currently unknown. Some argue that these result from a hypersensitivity reaction, although evidence to support this is very scarce. Most reports on such reactions are not substantiated by positive skin tests. The purpose of our study was to determine whether immediate or delayed type hypersensitivity reaction follows hyaluronic acid (HA) filler injections. Patients and Methods Twelve patients were referred for general allergic screening (patch tests), as well as specific intradermal testing (injection of 0.1cc boluses) on the medial upper arm with a selection of several currently available hyaluronic acid (HA) fillers on the market. A positive allergic reaction was defined as erythema, firmness or swelling. Results During the 4 month follow-up, no reactions to any of the tested HA fillers were reported. No correlation was found between results from the general allergic screening and a history with LIRs to HA fillers. Conclusion The results suggest that neither type I nor type IV hypersensitivity plays a role in late inflammatory reactions (LIRs) to hyaluronic acid (HA) fillers. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/ldw2cZOftOw
Collapse
Affiliation(s)
- Tom Decates
- Department of Dermatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jonathan Kadouch
- Department of Dermatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Peter Velthuis
- Department of Dermatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Thomas Rustemeyer
- Department of Dermatology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Sánchez-Pavón E, López-Monteon A, Hernández-Romero D, de la Soledad Lagunes-Castro M, Zanatta-García DY, Ramos-Ligonio A. Design and Synthesis of IMR-23, an Oxime Derived from Nitroimidazole as an Immunomodulatory Molecule. Curr Drug Deliv 2021; 17:324-332. [PMID: 32056525 DOI: 10.2174/1567201817666200214110442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 09/26/2019] [Accepted: 02/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Adjuvants have been obtained empirically by trial and error experiments and today, there is a tendency to the rational design of adjuvants candidates, which will increasingly achieve effective and safe products. The aim of this work was to design and evaluate the compound IMR-23 derived from nitroimidazole as an immunomodulatory molecule. MATERIALS AND METHODS The IMR-23 molecule was obtained by a condensation reaction, cytotoxicity was tested by the sulforhodamine B assay. Adjuvanticity was evaluated in vivo and in vitro in J774A.1 cells and in the mouse model, respectively. RESULTS IMR-23 that did not show cytotoxicity on HeLa, Vero cells and macrophages J774A.1, was able to induce the production of molecules involved in the inflammatory process, such as cytokines and chemokines determined by ELISA, to induce the production of antibodies and to generate antigenspecific cells to ovalbumin and against the antigen GST-L1b. CONCLUSION These results open the possibility of further studies to obtain a proper balance of immunogenicity- toxicity in the use of IMR-23 as an adjuvant molecule.
Collapse
Affiliation(s)
- Esmeralda Sánchez-Pavón
- LADISER Quimica Organica, Facultad de Ciencias Quimicas, Universidad Veracruzana, Orizaba, Veracruz, Mexico
| | - Aracely López-Monteon
- LADISER Inmunologia y Biologia Molecular, Facultad de Ciencias Quimicas, Universidad Veracruzana, Orizaba, Veracruz, Mexico.,Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Veracruz, México
| | - Delia Hernández-Romero
- LADISER Quimica Organica, Facultad de Ciencias Quimicas, Universidad Veracruzana, Orizaba, Veracruz, Mexico
| | | | | | - Angel Ramos-Ligonio
- LADISER Inmunologia y Biologia Molecular, Facultad de Ciencias Quimicas, Universidad Veracruzana, Orizaba, Veracruz, Mexico.,Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Veracruz, México
| |
Collapse
|
24
|
Schijns V, Majhen D, van der Ley P, Thakur A, Summerfield A, Berisio R, Nativi C, Fernández-Tejada A, Alvarez-Dominguez C, Gizurarson S, Zamyatina A, Molinaro A, Rosano C, Jakopin Ž, Gursel I, McClean S. Rational Vaccine Design in Times of Emerging Diseases: The Critical Choices of Immunological Correlates of Protection, Vaccine Antigen and Immunomodulation. Pharmaceutics 2021; 13:501. [PMID: 33917629 PMCID: PMC8067490 DOI: 10.3390/pharmaceutics13040501] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 01/21/2023] Open
Abstract
Vaccines are the most effective medical intervention due to their continual success in preventing infections and improving mortality worldwide. Early vaccines were developed empirically however, rational design of vaccines can allow us to optimise their efficacy, by tailoring the immune response. Establishing the immune correlates of protection greatly informs the rational design of vaccines. This facilitates the selection of the best vaccine antigens and the most appropriate vaccine adjuvant to generate optimal memory immune T cell and B cell responses. This review outlines the range of vaccine types that are currently authorised and those under development. We outline the optimal immunological correlates of protection that can be targeted. Finally we review approaches to rational antigen selection and rational vaccine adjuvant design. Harnessing current knowledge on protective immune responses in combination with critical vaccine components is imperative to the prevention of future life-threatening diseases.
Collapse
Affiliation(s)
- Virgil Schijns
- Intravacc, Institute for Translational Vaccinology (Intravacc), Utrecht Science Park, 3721 MA Bilthoven, The Netherlands;
- Epitopoietic Research Corporation (ERC), 5374 RE Schaijk, The Netherlands
| | - Dragomira Majhen
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Instiute, HR-10000 Zagreb, Croatia;
| | - Peter van der Ley
- Intravacc, Institute for Translational Vaccinology (Intravacc), Utrecht Science Park, 3721 MA Bilthoven, The Netherlands;
| | - Aneesh Thakur
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Artur Summerfield
- Institute of Virology and Immunology, 3147 Mittelhausern, Switzerland;
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, National Research Council, I-80134 Naples, Italy;
| | - Cristina Nativi
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy;
| | - Alberto Fernández-Tejada
- Chemical Immunology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Biscay Science and Technology Park, 48160 Derio-Bilbao, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Carmen Alvarez-Dominguez
- Facultativo en plantilla (Research Faculty), Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Sveinbjörn Gizurarson
- Faculty of Pharmaceutical Sciences, University of Iceland, 107 Reykjavik, Iceland;
- Department of Pharmacy, College of Medicine, University of Malawi, Blantyre 3, Malawi
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria;
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Santangelo, I-80126 Napoli, Italy;
- Department of Chemistry, School of Science, Osaka University, 1-1 Osaka University Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Camillo Rosano
- Proteomics and Mass Spectrometry Unit, IRCCS Policlinico San Martino, 16132 Genova-1, Italy;
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubiljana, Slovenia;
| | - Ihsan Gursel
- Molecular Biology and Genetics Department, Science Faculty, Bilkent University, Bilkent, 06800 Ankara, Turkey;
| | - Siobhán McClean
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
25
|
Gupta D, Gangwar A, Jyoti K, Sainaga Jyothi VG, Sodhi RK, Mehra NK, Singh SB, Madan J. Self healing hydrogels: A new paradigm immunoadjuvant for delivering peptide vaccine. Colloids Surf B Biointerfaces 2020; 194:111171. [DOI: 10.1016/j.colsurfb.2020.111171] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/24/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022]
|
26
|
Gupta N, Regar H, Verma VK, Prusty D, Mishra A, Prajapati VK. Receptor-ligand based molecular interaction to discover adjuvant for immune cell TLRs to develop next-generation vaccine. Int J Biol Macromol 2020; 152:535-545. [PMID: 32112848 DOI: 10.1016/j.ijbiomac.2020.02.297] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/11/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022]
Abstract
Human immune cell toll-like receptors (TLRs) provide a novel chance for the development of the vaccine adjuvant engaging TLR signaling. A library of peptides was developed and peptides structure was generated through homology modeling and refinement. Further, these peptides were subjected to receptor-ligand interaction study against human immune cell TLRs using Schrödinger-suite software. Here, we identified the most potent ligands for each human immune cell receptor and identified it as a potent adjuvant. This work portrays the ability of binding of different known protein adjuvants with human TLRs 1--10. The significance of the study deals with the identification of adjuvant (ligand) for human TLRs individually which assist in the development of the optimal highly immunogenic vaccine.
Collapse
Affiliation(s)
- Nidhi Gupta
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, 305817 Ajmer, Rajasthan, India
| | - Hansa Regar
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, 305817 Ajmer, Rajasthan, India
| | - Vijay Kumar Verma
- Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, 305817 Ajmer, Rajasthan, India
| | - Dhaneswar Prusty
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, 305817 Ajmer, Rajasthan, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan 342011, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, 305817 Ajmer, Rajasthan, India.
| |
Collapse
|
27
|
Effect on cellular recruitment and the innate immune response by combining saponin, monophosphoryl lipid-A and Incomplete Freund’s Adjuvant with Leishmania (Viannia) braziliensis antigens for a vaccine formulation. Vaccine 2019; 37:7269-7279. [DOI: 10.1016/j.vaccine.2019.09.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 01/24/2023]
|
28
|
Zhou Y, Banday AH, Hruby VJ, Cai M. Development of N-Acetylated Dipalmitoyl- S-Glyceryl Cysteine Analogs as Efficient TLR2/TLR6 Agonists. Molecules 2019; 24:molecules24193512. [PMID: 31569697 PMCID: PMC6803979 DOI: 10.3390/molecules24193512] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/19/2019] [Accepted: 09/25/2019] [Indexed: 12/30/2022] Open
Abstract
Cancer vaccine is a promising immunotherapeutic approach to train the immune system with vaccines to recognize and eliminate tumors. Adjuvants are compounds that are necessary in cancer vaccines to mimic an infection process and amplify immune responses. The Toll-like receptor 2 and 6 (TLR2/TLR6) agonist dipalmitoyl-S-glyceryl cysteine (Pam2Cys) was demonstrated as an ideal candidate for synthetic vaccine adjuvants. However, the synthesis of Pam2Cys requires expensive N-protected cysteine as a key reactant, which greatly limits its application as a synthetic vaccine adjuvant in large-scaled studies. Here, we report the development of N-acetylated Pam2Cys analogs as TLR2/TLR6 agonists. Instead of N-protected cysteine, the synthesis utilizes N-acetylcysteine to bring down the synthetic costs. The N-acetylated Pam2Cys analogs were demonstrated to activate TLR2/TLR6 in vitro. Moreover, molecular docking studies were performed to provide insights into the molecular mechanism of how N-acetylated Pam2Cys analogs bind to TLR2/TLR6. Together, these results suggest N-acetylated Pam2Cys analogs as inexpensive and promising synthetic vaccine adjuvants to accelerate the development of cancer vaccines in the future.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ 85721, USA.
| | - Abid H Banday
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ 85721, USA.
| | - Victor J Hruby
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ 85721, USA.
| | - Minying Cai
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
29
|
Nascimento LFMD, Moura LDD, Lima RT, Cruz MDSPE. Novos adjuvantes vacinais: importante ferramenta para imunoterapia da leishmaniose visceral. HU REVISTA 2019. [DOI: 10.34019/1982-8047.2018.v44.14123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Atualmente, muitas das vacinas em desenvolvimento são aquelas compostas de proteínas antigênicas individuais de parasitas ou uma combinação de vários antígenos individuais que são produzidos como produtos recombinantes obtidos por técnicas de biologia molecular. Dentre elas a Leish-111f e sua variação Leish-110f tem ganhado destaque na proteção contra a LV e LC e alcançaram estudos de fase II em seres humanos. A eficácia de uma vacina é otimizada pela adição de adjuvantes imunológicos. No entanto, embora os adjuvantes tenham sido usados por mais de um século, até o momento, apenas alguns adjuvantes são aprovados para o uso em humanos, a maioria destinada a melhorar a eficácia da vacina e a produção de anticorpos protetores específicos do antígeno. Os mecanismos de ação dos adjuvantes imunológicos são diversos, dependendo da sua natureza química e molecular sendo capazes de ativar células imunes especificas que conduzem a respostas imunes inatas e adaptativas melhoradas. Embora o mecanismo de ação molecular detalhado de muitos adjuvantes ainda seja desconhecido, a descoberta de receptores Toll-like (TLRs) forneceu informações críticas sobre o efeito imunoestimulador de numerosos componentes bacterianos que envolvem interação com receptores TLRs, mostrando que estes ligantes melhoram tanto a qualidade como a quantidade de respostas imunes adaptativas do hospedeiro quando utilizadas em formulações de vacinais direcionadas para doenças. O potencial desses adjuvantes de TLR em melhorar o design e os resultados de várias vacinas está em constante evolução, à medida que novos agonistas são descobertos e testados em modelos experimentais e estudos clínicos de vacinação. Nesta revisão, é apresentado um resumo do progresso recente no desenvolvimento de proteínas recombinantes de segunda geração e adjuvantes de TLR, sendo o foco principal nos TLR4 e suas melhorias.
Collapse
|
30
|
Construction and Immunogenicity of Novel Chimeric Virus-Like Particles Bearing Antigens of Infectious Bronchitis Virus and Newcastle Disease Virus. Viruses 2019; 11:v11030254. [PMID: 30871190 PMCID: PMC6465995 DOI: 10.3390/v11030254] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/22/2022] Open
Abstract
Infectious bronchitis virus (IBV) and Newcastle disease virus (NDV) are two poultry pathogens seriously affecting the poultry industry. Here, IBV S1 and the ectodomain of NDV F proteins were separately linked with the trans-membrane and carboxy-terminal domain of IBV S protein (STMCT), composing rS and rF; thus, a novel chimeric infectious bronchitis-Newcastle disease (IB-ND) virus-like particles (VLPs) vaccine containing the rS, rF, and IBV M protein was constructed. Under the transmission electron microscope (TEM), VLPs possessing similar morphology to natural IBV were observed. To evaluate the immunogenicity of chimeric IB-ND VLPs, specific pathogen-free (SPF) chickens were immunized with three increasing doses (50, 75, and 100 μg protein of VLPs). Results of ELISAs detecting IBV and NDV specific antibodies and IL-4 and IFN-γ T cell cytokines indicated that vaccination with chimeric IB-ND VLPs could efficiently induce humoral and cellular immune responses. In the challenge study, chimeric IB-ND VLPs (100 μg protein) provided 100% protection against IBV or NDV virulent challenge from death, and viral RNA levels in tissues and swabs were greatly reduced. Collectively, chimeric IB-ND VLPs are highly immunogenic and could provide complete protection from an IBV or NDV virulent challenge. Chimeric IB-ND VLPs are an appealing vaccine candidate and a promising vaccine platform bearing multivalent antigens.
Collapse
|
31
|
Magiri R, Lai K, Huang Y, Mutwiri G, Wilson HL. Innate immune response profiles in pigs injected with vaccine adjuvants polydi(sodium carboxylatoethylphenoxy)phosphazene (PCEP) and Emulsigen. Vet Immunol Immunopathol 2019; 209:7-16. [PMID: 30885308 DOI: 10.1016/j.vetimm.2019.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/23/2022]
Abstract
Vaccines are formulated with adjuvants to enhance or direct antigen-specific immune responses against pathogens. However, the mechanisms of action (MOA) of adjuvants are not well understood and are under-investigated in large animal species. We have previously reported that injection of mice induced innate immune responses as indicated by increased cell recruitment and cytokine production at the site of injection with polyphosphazene (PCEP) adjuvant. In the present study, we evaluated whether PCEP induced similar innate immune responses in pigs. Piglets were injected with either PCEP or Emulsigen intradermally (I.D.) and the local cellular infiltration and cytokine production were evaluated at the site of injection and the draining lymph nodes. PCEP induced infiltration of macrophages, T and B cells, leucocytes and necrotic debris at the site of injection as well as PCEP-induced leucocyte infiltration in the draining lymph nodes. Emulsigen induced diffuse infiltration of leucocytes, macrophages, and lymphocytes at the site of injection as well as at the draining lymph nodes. PCEP induced significant production of interleukin IL-1β, and IL-13 at the site of injection and IL-1β, and IL-6 at the draining lymph nodes. Emulsigen promoted the production of IL-1β, IL-6, and IL-12 at the site of injection but not in the draining lymph nodes. No cytokines were detected in blood after injection of either adjuvant. Together, our data indicate that in pigs, the adjuvants PCEP and Emulsigen stimulate early innate immune responses at the injection site by creating an immunocompetent environment that may contribute to increased immunogenicity of the co-administered antigens.
Collapse
Affiliation(s)
- Royford Magiri
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada; Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK, Canada
| | - Ken Lai
- Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK, Canada
| | - Yanyun Huang
- Prairie Diagnostic Services, 52 Campus Drive, Saskatoon, SK, Canada
| | - George Mutwiri
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada; Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK, Canada
| | - Heather L Wilson
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada; Vaccine & Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
32
|
Deng Z, Geng Y, Wang K, Yu Z, Yang PO, Yang Z, He C, Huang C, Yin L, He M, Tang L, Lai W. Adjuvant effects of interleukin-2 co-expression with VP60 in an oral vaccine delivered by attenuated Salmonella typhimurium against rabbit hemorrhagic disease. Vet Microbiol 2019; 230:49-55. [PMID: 30827404 DOI: 10.1016/j.vetmic.2019.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 01/06/2019] [Accepted: 01/07/2019] [Indexed: 01/15/2023]
Abstract
Rabbit hemorrhagic disease (RHD) is a highly contagious infection that has caused significant damage to the rabbit industry since 1984. Inactivated vaccines, the currently used prevention measures, are effective in controlling RHD. However, these vaccines are derived from the livers of infected rabbits, which constitutes a major concern in terms of animal welfare and safety. Administration of DNA vaccines in collaboration with appropriate adjuvants, in particular, cytokines, to strengthen the immune response presents a novel optimization strategy to generate more efficient vaccines. In this study, the adjuvant effect of interleukin (IL)-2 co-expression with the VP60 gene in a DNA vaccine was evaluated. In total, four groups of 60 RHD virus (RHDV)-free rabbits (30 days old) were orally or subcutaneously administered recombinant SL7207-pVAX1-IL2-VP60, SL7207-pVAX1-VP60, SL7207-pVAX1 bacteria or the commercial inactive vaccine, and the induced immunity evaluated by challenge with the RHDV(Y8504/China) strain on day 56. The Recombinant SL7207-pVAX1-IL2-VP60 induced a higher level of antibodies than the vaccine SL7207-pVAX1-VP60 and inactivated vaccines to a significant extent. The concentrations of interleukin (IL)-4 were markedly higher than those in groups immunized with the naked or inactive vaccine alone. Furthermore, the fusion gene vaccine provided higher protection (93.33%) after virus challenge relative to immunization with the single gene (SL7207-pVAX1-VP60). The collective results indicate that recombinant SL7207-pVAX1-IL-2-VP60 bacteria exert enhanced protective effects against RHDV and therefore present a strong candidate as a potential vaccine. Moreover, IL-2 enhanced both humoral and cellular responses, highlighting the utility of rabbit IL-2 as an effective adjuvant.
Collapse
Affiliation(s)
- Zhaobin Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yi Geng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Kaiyu Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Zehui Yu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Ping Ou Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Zexiao Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Changliang He
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Lizi Yin
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Min He
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Li Tang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Weimin Lai
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| |
Collapse
|
33
|
Malik A, Gupta M, Gupta V, Gogoi H, Bhatnagar R. Novel application of trimethyl chitosan as an adjuvant in vaccine delivery. Int J Nanomedicine 2018; 13:7959-7970. [PMID: 30538470 PMCID: PMC6260144 DOI: 10.2147/ijn.s165876] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The application of natural carbohydrate polysaccharides for antigen delivery and its adjuvanation potential has garnered interest in the scientific community in the recent years. These biomaterials are considered favorable candidates for adjuvant development due to their desirable properties like enormous bioavailability, non-toxicity, biodegradability, stability, affordability, and immunostimulating ability. Chitosan is the one such extensively studied natural polymer which has been appreciated for its excellent applications in pharmaceuticals. Trimethyl chitosan (TMC), a derivative of chitosan, possesses these properties. In addition it has the properties of high aqueous solubility, high charge density, mucoadhesive, permeation enhancing (ability to cross tight junction), and stability over a range of ionic conditions which makes the spectrum of its applicability much broader. It has also been seen to perform analogously to alum, complete Freund’s adjuvant, incomplete Freund’s adjuvant, and cyclic guanosine monophosphate adjuvanation, which justifies its role as a potent adjuvant. Although many review articles detailing the applications of chitosan in vaccine delivery are available, a comprehensive review of the applications of TMC as an adjuvant is not available to date. This article provides a comprehensive overview of structural and chemical properties of TMC which affect its adjuvant characteristics; the efficacy of various delivery routes for TMC antigen combination; and the recent advances in the elucidation of its mechanism of action.
Collapse
Affiliation(s)
- Anshu Malik
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Manish Gupta
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Vatika Gupta
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Himanshu Gogoi
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| |
Collapse
|
34
|
Tandon A, Pathak M, Harioudh MK, Ahmad S, Sayeed M, Afshan T, Siddiqi MI, Mitra K, Bhattacharya SM, Ghosh JK. A TLR4-derived non-cytotoxic, self-assembling peptide functions as a vaccine adjuvant in mice. J Biol Chem 2018; 293:19874-19885. [PMID: 30385503 DOI: 10.1074/jbc.ra118.002768] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 09/01/2018] [Indexed: 12/18/2022] Open
Abstract
Vaccination is devised/formulated to stimulate specific and prolonged immune responses for long-term protection against infection or disease. A vaccine component, namely adjuvant, enhances antigen recognition by the host immune system and thereby stimulates its cellular and adaptive responses. Especially synthetic Toll-like receptor (TLR) agonists having self-assembling properties are considered as good candidates for adjuvant development. Here, a human TLR4-derived 20-residue peptide (TR-433), present in the dimerization interface of the TLR4-myeloid differentiation protein-2 (MD2) complex, displayed self-assembly and adopted a nanostructure. Both in vitro studies and in vivo experiments in mice indicated that TR-433 is nontoxic. TR-433 induced pro-inflammatory responses in THP-1 monocytes and HEK293T cells that were transiently transfected with TLR4/CD14/MD2 and also in BALB/c mice. In light of the self-assembly and pro-inflammatory properties of TR-433, we immunized with a mixture of TR-433 and either ovalbumin or filarial antigen trehalose-6-phosphate phosphatase (TPP). A significant amount of IgG titers was produced, suggesting adjuvanting capability of TR-433 that was comparable with that of Freund's complete adjuvant (FCA) and appreciably higher than that of alum. We found that TR-433 preferentially activates type 1 helper T cell (Th1) response rather than type 2 helper T cell (Th2) response. To our knowledge, this is the first report on the identification of a short TLR4-derived peptide that possesses both self-assembling and pro-inflammatory properties and has significant efficacy as an adjuvant, capable of activating cellular responses in mice. These results indicate that TR-433 possesses significant potential for development as a new adjuvant in therapeutic application.
Collapse
Affiliation(s)
| | | | | | | | - Mohd Sayeed
- From the Molecular and Structural Biology Division
| | | | - M I Siddiqi
- From the Molecular and Structural Biology Division
| | - Kalyan Mitra
- Electron Microscopy Unit, SAIF Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road Lucknow-226 031, India
| | | | | |
Collapse
|
35
|
Kaurav M, Madan J, Sudheesh MS, Pandey RS. Combined adjuvant-delivery system for new generation vaccine antigens: alliance has its own advantage. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S818-S831. [DOI: 10.1080/21691401.2018.1513941] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Monika Kaurav
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Jitender Madan
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, India
| | - M. S. Sudheesh
- Faculty of Pharmacy, VNS Group of Institutions, Nathu Barkheda, Bhopal, India
| | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
36
|
Yan S, Gu W, Zhang B, Rolfe BE, Xu ZP. High adjuvant activity of layered double hydroxide nanoparticles and nanosheets in anti-tumour vaccine formulations. Dalton Trans 2018; 47:2956-2964. [PMID: 29168855 DOI: 10.1039/c7dt03725b] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Effective adjuvants in anti-tumour vaccine formulations are very important in the development of new-generation vaccines. In this study, two layered double hydroxide (LDH) nanomaterial forms, i.e. nanoparticles (NPs) and nanosheets (NSs), were synthesised and examined as adjuvants to provoke the immune responses for anti-tumour purpose. Immunogen ovalbumin (OVA) delivered by both nanomaterials induced much stronger humoral and cell-medicated immune responses, together with an immune stimulant (TLR9 ligand CpG), as evidenced by higher levels of IgG1, IgG2a and interferon-γ. By comparison, LDH NSs showed higher activity to promote specific antibody responses than LDH NPs but with a similar cell-mediated immune response. The mice immunised with OVA-CpG vaccines formulated with both nanomaterials showed stronger inhibition of the inoculated tumour growth and had a longer survival. Altogether, these data indicate that LDH NPs and NSs can be used as potential nanoadjuvants for efficient protein-based anti-tumour vaccines.
Collapse
Affiliation(s)
- Shiyu Yan
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | | | |
Collapse
|
37
|
Li X, Wang X, Ito A. Tailoring inorganic nanoadjuvants towards next-generation vaccines. Chem Soc Rev 2018; 47:4954-4980. [PMID: 29911725 DOI: 10.1039/c8cs00028j] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vaccines, one of the most effective and powerful public health measures, have saved countless lives over the past century and still have a tremendous global impact. As an indispensable component of modern vaccines, adjuvants play a critical role in strengthening and/or shaping a specific immune response against infectious diseases as well as malignancies. The application of nanotechnology provides the possibility of precisely tailoring the building blocks of nanoadjuvants towards modern vaccines with the desired immune response. The last decade has witnessed great academic progress in inorganic nanomaterials for vaccine adjuvants in terms of nanometer-scale synthesis, structure control, and functionalization design. Inorganic adjuvants generally facilitate the delivery of antigens, allowing them to be released in a sustained manner, enhance immunogenicity, deliver antigens efficiently to specific targets, and induce a specific immune response. In particular, the recent discovery of the intrinsic immunomodulatory function of inorganic nanomaterials further allows us to shape the immune response towards the desired type and increase the efficacy of vaccines. In this article, we comprehensively review state-of-the-art research on the use of inorganic nanomaterials as vaccine adjuvants. Attention is focused on the physicochemical properties of versatile inorganic nanoadjuvants, such as composition, size, morphology, shape, hydrophobicity, and surface charge, to effectively stimulate cellular immunity, considering that the clinically used alum adjuvants can only induce strong humoral immunity. In addition, the efforts made to date to expand the application of inorganic nanoadjuvants in cancer vaccines are summarized. Finally, we discuss the future prospects and our outlook on tailoring inorganic nanoadjuvants towards next-generation vaccines.
Collapse
Affiliation(s)
- Xia Li
- Health Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.
| | | | | |
Collapse
|
38
|
Zhang C, Zhou J, Cai K, Zhang W, Liao C, Wang C. Gene cloning, expression and immune adjuvant properties of the recombinant fusion peptide Tα1-BLP on avian influenza inactivate virus vaccine. Microb Pathog 2018; 120:147-154. [DOI: 10.1016/j.micpath.2018.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 10/17/2022]
|
39
|
Fatima MT, Ahmad E, Hoque M. Effective antigen delivery via dual entrapment in erythrocytes and autologous plasma beads. J Drug Target 2017; 26:162-171. [DOI: 10.1080/1061186x.2017.1350859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | - Ejaj Ahmad
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Mehboob Hoque
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
40
|
Guo M, Tang X, Sheng X, Xing J, Zhan W. The Immune Adjuvant Effects of Flounder (Paralichthys olivaceus) Interleukin-6 on E. tarda Subunit Vaccine OmpV. Int J Mol Sci 2017; 18:ijms18071445. [PMID: 28678171 PMCID: PMC5535936 DOI: 10.3390/ijms18071445] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/22/2017] [Accepted: 07/01/2017] [Indexed: 12/20/2022] Open
Abstract
Interleukin-6 (IL-6) as a pleiotropic cytokine was widely used as an effective adjuvant for vaccines in mammals. In this study, the immune adjuvant effects of two forms of flounder (Paralichthys olivaceus) IL-6, including recombinant IL-6 (rIL-6) and pcDNA3.1-IL-6 (pcIL-6), were evaluated and comparatively analyzed on E. tarda subunit vaccine recombinant outer membrane protein V (rOmpV). The results showed that the relative percent survivals of flounder vaccinated with rOmpV plus rIL-6 or pcIL-6 were significantly higher than that in the two control groups, rOmpV plus recombinant 6× histidine-tag (rHis) or empty expression vector pcDNA3.1 (pcN3). The levels of specific serum antibodies and surface membrane immunoglobulin-positive (sIg+) lymphocytes in peripheral blood, spleen, and head kidney in the two adjuvant groups were also much higher than that in the two control groups. Compared with the two control groups, higher upregulated expressions of major histocompatibility complex class Iα (MHCIα), cluster of differentiation 8α (CD8α), MHCIIα, CD4-1, interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) were detected in flounder vaccinated with rOmpV plus rIL-6 or pcIL-6 after challenge. In addition, the rOmpV plus rIL-6 could induce significant higher levels of specific serum antibodies, sIg+ lymphocytes and four genes expressions than rOmpV plus pcIL-6. These results demonstrated that both rIL-6 and pcIL-6 used as adjuvants could enhance the immune response and evoke immune protections against E. tarda infection, which has a significant value in controlling diseases using vaccines in flounder.
Collapse
Affiliation(s)
- Ming Guo
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Aoshanwei Town, Jimo, Qingdao 266071, China.
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Aoshanwei Town, Jimo, Qingdao 266071, China.
| |
Collapse
|
41
|
Abstract
Vaccines have helped considerably in eliminating some life-threatening infectious diseases in past two hundred years. Recently, human medicine has focused on vaccination against some of the world's most common infectious diseases (AIDS, malaria, tuberculosis, etc.), and vaccination is also gaining popularity in the treatment of cancer or autoimmune diseases. The major limitation of current vaccines lies in their poor ability to generate a sufficient level of protective antibodies and T cell responses against diseases such as HIV, malaria, tuberculosis and cancers. Among the promising vaccination systems that could improve the potency of weakly immunogenic vaccines belong macromolecular carriers (water soluble polymers, polymer particels, micelles, gels etc.) conjugated with antigens and immunistumulatory molecules. The size, architecture, and the composition of the high molecular-weight carrier can significantly improve the vaccine efficiency. This review includes the most recently developed (bio)polymer-based vaccines reported in the literature.
Collapse
Affiliation(s)
- G MuŽíková
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | | |
Collapse
|
42
|
Tissera MS, Cowley D, Bogdanovic-Sakran N, Hutton ML, Lyras D, Kirkwood CD, Buttery JP. Options for improving effectiveness of rotavirus vaccines in developing countries. Hum Vaccin Immunother 2017; 13:921-927. [PMID: 27835052 PMCID: PMC5404363 DOI: 10.1080/21645515.2016.1252493] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/09/2016] [Accepted: 10/19/2016] [Indexed: 02/08/2023] Open
Abstract
Rotavirus gastroenteritis is a leading global cause of mortality and morbidity in young children due to diarrhea and dehydration. Over 85% of deaths occur in developing countries. In industrialised countries, 2 live oral rotavirus vaccines licensed in 2006 quickly demonstrated high effectiveness, dramatically reducing severe rotavirus gastroenteritis admissions in many settings by more than 90%. In contrast, the same vaccines reduced severe rotavirus gastroenteritis by only 30-60% in developing countries, but have been proven life-saving. Bridging this "efficacy gap" offers the possibility to save many more lives of children under the age of 5. The reduced efficacy of rotavirus vaccines in developing settings may be related to differences in transmission dynamics, as well as host luminal, mucosal and immune factors. This review will examine strategies currently under study to target the issue of reduced efficacy and effectiveness of oral rotavirus vaccines in developing settings.
Collapse
Affiliation(s)
- Marion S. Tissera
- Department of Paediatrics, Monash University, Melbourne, Australia; Enteric Virus Group, Murdoch Childrens Research Institute, Melbourne, Australia
| | - Daniel Cowley
- Enteric Virus Group, Murdoch Childrens Research Institute, Melbourne, Australia
| | | | | | - Dena Lyras
- Department of Microbiology, Monash University, Melbourne, Australia
| | - Carl D. Kirkwood
- Enteric Virus Group, Murdoch Childrens Research Institute, Melbourne, Australia; Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - Jim P. Buttery
- Department of Paediatrics & The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Melbourne, Australia; Infection and Immunity, Monash Children's Hospital, Monash Health, Melbourne, Australia; SAEFVIC, Murdoch Childrens Research Institute, Melbourne, Australia
| |
Collapse
|
43
|
Barinova K, Khomyakova E, Kuravsky M, Schmalhausen E, Muronetz V. Denaturing action of adjuvant affects specificity of polyclonal antibodies. Biochem Biophys Res Commun 2017; 482:1265-1270. [DOI: 10.1016/j.bbrc.2016.12.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/03/2016] [Indexed: 01/18/2023]
|
44
|
Liu Y, Chen S, Pan B, Guan Z, Yang Z, Duan L, Cai H. A subunit vaccine based on rH-NS induces protection against Mycobacterium tuberculosis infection by inducing the Th1 immune response and activating macrophages. Acta Biochim Biophys Sin (Shanghai) 2016; 48:909-922. [PMID: 27563010 DOI: 10.1093/abbs/gmw078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/07/2016] [Indexed: 11/12/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a Gram-positive pathogen which causes tuberculosis in both animals and humans. All tested rH-NS formulations induced a specific Th1 response, as indicated by increased production of interferon γ (IFN-γ) and interleukin 2 (IL-2) by lymphocytes in the spleen of mice which were immunized with rH-NS alone or with rH-NS and the adjuvant cyclic GMP-AMP (cGAMP). Serum from mice immunized with rH-NS with or without adjuvant also had higher levels of IL-12p40 and TNF-α, compared with those from control mice immunized with phosphate-buffered saline. Both vaccines increased protective efficacy in mice which were challenged with Mtb H37Rv, as measured by reduced relative CFU counts in the lungs. We found that rH-NS induced the production of TNF-α, IL-6, and IL-12p40, which relied on the activation of mitogen-activated protein kinases by stimulating the rapid phosphorylation of ERK1/2, p38, and JNK, and on the activation of transcription factor NF-κB in macrophages. Additionally, we also found that rH-NS could interact with TLR2 but not TLR4 in pull-down assays. The rH-NS-induced cytokine production from TLR2-silenced RAW264.7 cells was lower than that from BALB/c macrophages. Prolonged exposure (>24 h) of RAW264.7 cells to rH-NS resulted in a significant enhancement in IFN-γ-induced MHC II expression, which was not found in shTLR2-treated RAW264.7 cells. These results suggest that rH-NS is a TLR2 agonist which induces the production of cytokines by macrophages and up-regulates macrophage function.
Collapse
Affiliation(s)
- Yuan Liu
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
| | - Suting Chen
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing 101149, China
| | - Bowen Pan
- State Key Laboratory of Natural and Biomimetic Drugs, Peking Universtiy Health Science Center, Beijing 100191, China
| | - Zhu Guan
- State Key Laboratory of Natural and Biomimetic Drugs, Peking Universtiy Health Science Center, Beijing 100191, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking Universtiy Health Science Center, Beijing 100191, China
| | - Linfei Duan
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hong Cai
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
45
|
Kotla S, Sanghratna Vishanath B, H.J. D, K. G, V.V.S. S, Reddy G. DNA vaccine (P1-2A-3C-pCDNA) co-administered with Bovine IL-18 gives protective immune response against Foot and Mouth Disease in cattle. Vet Microbiol 2016; 193:106-15. [DOI: 10.1016/j.vetmic.2016.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 07/02/2016] [Accepted: 07/06/2016] [Indexed: 10/21/2022]
|
46
|
Delivery of an inactivated avian influenza virus vaccine adjuvanted with poly(D,L-lactic-co-glycolic acid) encapsulated CpG ODN induces protective immune responses in chickens. Vaccine 2016; 34:4807-13. [PMID: 27543454 DOI: 10.1016/j.vaccine.2016.08.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/28/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022]
Abstract
In poultry, systemic administration of commercial vaccines consisting of inactivated avian influenza virus (AIV) requires the simultaneous delivery of an adjuvant (water-in-oil emulsion). These vaccines are often limited in their ability to induce quantitatively better local (mucosal) antibody responses capable of curtailing virus shedding. Therefore, more efficacious adjuvants with the ability to provide enhanced immunogenicity and protective anti-AIV immunity in chickens are needed. While the Toll-like receptor (TLR) 21 agonist, CpG oligodeoxynucleotides (ODNs) has been recognized as a potential vaccine adjuvant in chickens, poly(D,L-lactic-co-glycolic acid) (PLGA) nanoparticles, successfully tested as vaccine delivery systems in other species, have not been extensively explored. The present study, therefore, assessed both systemic and mucosal antibody-mediated responses following intramuscular vaccination (administered at 7 and 21days post-hatch) of chickens with PLGA encapsulated H9N2 AIV plus encapsulated CpG ODN 2007 (CpG 2007), and nonencapsulated AIV plus PLGA encapsulated CpG 2007 vaccine formulations. Virus challenge was performed at 2weeks post-secondary vaccination using the oculo-nasal route. Our results showed that chickens vaccinated with the nonencapsulated AIV vaccine plus PLGA encapsulated CpG 2007 developed significantly higher systemic IgY and local (mucosal) IgY antibodies as well as haemagglutination inhibition antibody titres compared to PLGA encapsulated AIV plus encapsulated CpG 2007 vaccinated chickens. Furthermore, chickens that received CpG 2007 as an adjuvant in the vaccine formulation had antibodies exhibiting higher avidity indicating that the TLR21-mediated pathway may enhance antibody affinity maturation qualitatively. Collectively, our data indicate that vaccination of chickens with nonencapsulated AIV plus PLGA encapsulated CpG 2007 results in qualitatively and quantitatively augmented antibody responses leading to a reduction in virus shedding compared to the encapsulated AIV plus PLGA encapsulated CpG 2007 formulation.
Collapse
|
47
|
Huang FJ, Ma YL, Tang RY, Gong WC, Li J, Chen CX, Yin L, Chen XP. Interleukin-4- and NACHT, LRR and PYD domains-containing protein 3-independent mechanisms of alum enhanced T helper type 2 responses on basophils. Immunology 2016; 149:238-51. [PMID: 27315109 DOI: 10.1111/imm.12636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 06/11/2016] [Accepted: 06/12/2016] [Indexed: 12/12/2022] Open
Abstract
Aluminium hydroxide (alum), the most widely used adjuvant in human and animal vaccines, has long been known to promote T helper type 2 (Th2) responses and Th2-associated humoral responses, but the mechanisms have remained poorly understood. In this study, we explored whether alum is able to directly modulate antigen-presenting cells to enhance their potency for Th2 polarization. We found that alum treatment of dendritic cells failed to show any Th2-promoting activities. In contrast, alum was able to enhance the capacity of basophils to induce Th2 cells. When basophils from interleukin-4 (IL-4) knockout mice were examined, the intrinsic Th2-promoting activities by basophils were largely abrogated, but the alum-enhanced Th2-promoting activities on basophils were still detectable. More importantly, Th2-promoting adjuvant activities by alum found in IL-4 knockout mice were also largely reduced when basophils were depleted by antibody administration. Therefore, basophils can mediate Th2-promoting activities by alum both in vitro and in vivo through IL-4-independent mechanisms. Further studies revealed that secreted soluble molecules from alum-treated basophils were able to confer the Th2-promoting activities, and neutralization of thymic stromal lymphopoietin or IL-25 attenuated the IL-4-independent development of Th2 cells elicited by alum-treated basophils. Finally, alum was able to activate NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome in murine basophils in the same way as alum in professional antigen-presenting cells, but NLRP3 was not required for Th2-promoting activities on basophils by alum in vitro. These results demonstrated that alum can enhance the capacities of basophils to polarize Th2 cells via IL-4- and NLRP3-independent pathways.
Collapse
Affiliation(s)
- Feng-Juan Huang
- Department of Immunology and Pathogen Biology, TongJi University School of Medicine, Shanghai, China
| | - Yi-Lei Ma
- Department of Immunology and Pathogen Biology, TongJi University School of Medicine, Shanghai, China
| | - Ruo-Yu Tang
- Department of Immunology and Pathogen Biology, TongJi University School of Medicine, Shanghai, China
| | - Wen-Ci Gong
- Department of Immunology and Pathogen Biology, TongJi University School of Medicine, Shanghai, China
| | - Jun Li
- Department of Immunology and Pathogen Biology, TongJi University School of Medicine, Shanghai, China
| | - Chun-Xia Chen
- Department of Immunology and Pathogen Biology, TongJi University School of Medicine, Shanghai, China
| | - Lan Yin
- Department of Immunology and Pathogen Biology, TongJi University School of Medicine, Shanghai, China
| | - Xiao-Ping Chen
- Department of Immunology and Pathogen Biology, TongJi University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Adjuvants: Classification, Modus Operandi, and Licensing. J Immunol Res 2016; 2016:1459394. [PMID: 27274998 PMCID: PMC4870346 DOI: 10.1155/2016/1459394] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/02/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023] Open
Abstract
Vaccination is one of the most efficient strategies for the prevention of infectious diseases. Although safer, subunit vaccines are poorly immunogenic and for this reason the use of adjuvants is strongly recommended. Since their discovery in the beginning of the 20th century, adjuvants have been used to improve immune responses that ultimately lead to protection against disease. The choice of the adjuvant is of utmost importance as it can stimulate protective immunity. Their mechanisms of action have now been revealed. Our increasing understanding of the immune system, and of correlates of protection, is helping in the development of new vaccine formulations for global infections. Nevertheless, few adjuvants are licensed for human vaccines and several formulations are now being evaluated in clinical trials. In this review, we briefly describe the most well known adjuvants used in experimental and clinical settings based on their main mechanisms of action and also highlight the requirements for licensing new vaccine formulations.
Collapse
|
49
|
Chen W, Zhang B, Mahony T, Gu W, Rolfe B, Xu ZP. Efficient and Durable Vaccine against Intimin β of Diarrheagenic E. Coli Induced by Clay Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:1627-39. [PMID: 27000499 DOI: 10.1002/smll.201503359] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/09/2015] [Indexed: 05/21/2023]
Abstract
Improved strategies are urgently required to control infections with enterohemorrhagic Escherichia coli and enteropathogenic E. coli, two dominant zoonotic enteric pathogens responsible for a wide spectrum of illnesses as well as deaths of human being, with tremendous financial cost worldwide. The present study investigates the capacity of two clay nanoparticles (NPs) with opposite surface charges, namely synthetic layered double hydroxide (LDH) and hectorite (HEC) NPs as adjuvants to promote strong immune responses against the infections. Here both LDH and HEC NPs are showed to be able to carry an appreciable amount of Intimin β (1.1 and 4.4 mg per mg clay nanomaterials, respectively) and significantly facilitate antigen uptake by antigen-presenting cells. Remarkably, these clay NPs induce strong antibody and cell-mediated immune responses, which are much higher than that by the potent adjuvant, QuilA. Furthermore, these strong immune responses are well maintained for at least four months in the mouse model, during which there are no changes in histopathology of the animal organs. Collectively these data demonstrate the suitability of LDH and HEC NPs as useful adjuvants in new-generation vaccine formulations to control various infectious diseases.
Collapse
Affiliation(s)
- Weiyu Chen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Bing Zhang
- Vaccine Delivery, Animal Science, Agri-Science Queensland Department of Agriculture, Fisheries and Forestry, St Lucia, QLD, 4067, Australia
| | - Timothy Mahony
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Barbara Rolfe
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| |
Collapse
|
50
|
Kimber I, Carrillo JC. Oral exposure to mineral oils: Is there an association with immune perturbation and autoimmunity? Toxicology 2016; 344-346:19-25. [DOI: 10.1016/j.tox.2016.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/19/2016] [Accepted: 01/22/2016] [Indexed: 12/18/2022]
|