1
|
Mkulo EM, Wang B, Amoah K, Huang Y, Cai J, Jin X, Wang Z. The current status and development forecasts of vaccines for aquaculture and its effects on bacterial and viral diseases. Microb Pathog 2024; 196:106971. [PMID: 39307198 DOI: 10.1016/j.micpath.2024.106971] [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: 04/09/2024] [Revised: 08/19/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
The aquaculture sector predicts protein-rich meals by 2040 and has experienced significant economic shifts since 2000. However, challenges emanating from disease control measures, brood stock improvement, feed advancements, hatchery technology, and water quality management due to environmental fluctuations have been taken as major causative agents for hindering the sector's growth. For the past years, aquatic disease prevention and control have principally depended on the use of various antibiotics, ecologically integrated control, other immunoprophylaxis mechanisms, and chemical drugs, but the long-term use of chemicals such as antibiotics not only escalates antibiotic-resistant bacteria and genes but also harms the fish and the environments, resulting in drug residues in aquatic products, severely obstructing the growth of the aquaculture sector. The field of science has opened new avenues in basic and applied research for creating and producing innovative and effective vaccines and the enhancement of current vaccines to protect against numerous infectious diseases. Recent advances in vaccines and vaccinology could lead to novel vaccine candidates that can tackle fish diseases, including parasitic organism agents, for which the current vaccinations are inadequate. In this review, we study and evaluate the growing aquaculture production by focusing on the current knowledge, recent progress, and prospects related to vaccinations and immunizations in the aquaculture industry and their effects on treating bacterial and viral diseases. The subject matter covers a variety of vaccines, such as conventional inactivated and attenuated vaccines as well as advanced vaccines, and examines their importance in real-world aquaculture scenarios. To encourage enhanced importation of vaccines for aquaculture sustainability and profitability and also help in dealing with challenges emanating from diseases, national and international scientific and policy initiatives need to be informed about the fundamental understanding of vaccines.
Collapse
Affiliation(s)
- Evodia Moses Mkulo
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524088, China
| | - Bei Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Ocean University, Zhanjiang, 524088, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 327005, China
| | - Kwaku Amoah
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Ocean University, Zhanjiang, 524088, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 327005, China.
| | - Yu Huang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Ocean University, Zhanjiang, 524088, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 327005, China
| | - Jia Cai
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Ocean University, Zhanjiang, 524088, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 327005, China
| | - Xiao Jin
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Ocean University, Zhanjiang, 524088, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 327005, China
| | - Zhongliang Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Ocean University, Zhanjiang, 524088, China; Agro-Tech Extension Center of Guangdong Province, Guangzhou, China.
| |
Collapse
|
2
|
Goetz MJ, Park KS, Joshi M, Gottlieb AP, Dowling DJ, Mitragotri S. An ionic liquid-based adjuvant for modulating cellular and humoral immune responses. J Control Release 2024; 376:632-645. [PMID: 39437967 DOI: 10.1016/j.jconrel.2024.10.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Vaccination is an important strategy for the prevention of infectious diseases worldwide. Adjuvants can be incorporated in vaccine formulations to enhance the resultant immune response and subsequently confer more robust protection upon natural infection. While adjuvants have exciting potential to improve vaccination, the landscape of materials employed in clinical adjuvants is small and its expansion is needed to facilitate vaccine development against current and future infectious diseases. This study introduces the first ionic liquid (IL) adjuvant comprised of choline and sorbic acid (ChoSorb) to produce an antigen-specific cellular as well as humoral immune response against multiple antigens. The abilities of ChoSorb as a vaccine adjuvant is evaluated and characterized through material analysis, innate immune responses, and adaptive responses to both a model and clinical grade antigen. With the robust immune responses generated by ChoSorb and the accompanying mechanistic insights, this study introduces ILs as a new class of adjuvant materials for future vaccine design.
Collapse
Affiliation(s)
- Morgan J Goetz
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA
| | - Kyung Soo Park
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA
| | - Maithili Joshi
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA
| | - Alexander P Gottlieb
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA.
| |
Collapse
|
3
|
Lan J, Feng D, He X, Zhang Q, Zhang R. Basic Properties and Development Status of Aluminum Adjuvants Used for Vaccines. Vaccines (Basel) 2024; 12:1187. [PMID: 39460352 PMCID: PMC11511158 DOI: 10.3390/vaccines12101187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Aluminum adjuvants, renowned for their safety and efficacy, act as excellent adsorbents and vaccine immunogen enhancers, significantly contributing to innate, endogenous, and humoral immunity. An ideal adjuvant not only boosts the immune response but also ensures optimal protective immunity. Aluminum adjuvants are the most widely used vaccine adjuvants and have played a crucial role in both the prevention of existing diseases and the development of new vaccines. With the increasing emergence of new vaccines, traditional immune adjuvants are continually being researched and upgraded. The future of vaccine development lies in the exploration and integration of novel adjuvant technologies that surpass the capabilities of traditional aluminum adjuvants. One promising direction is the incorporation of nanoparticles, which offer precise delivery and controlled release of antigens, thereby enhancing the overall immune response. CONCLUSIONS This review summarizes the types, mechanisms, manufacturers, patents, advantages, disadvantages, and future prospects of aluminum adjuvants. Although aluminum adjuvants have certain limitations, their contribution to enhancing vaccine immunity is significant and cannot be ignored. Future research should continue to explore their mechanisms of action and address potential adverse reactions to achieve improved vaccine efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Rong Zhang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 117004, China; (J.L.); (D.F.); (Q.Z.)
| |
Collapse
|
4
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2024; 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] [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
|
5
|
Oboge H, Riitho V, Nyamai M, Omondi GP, Lacasta A, Githaka N, Nene V, Aboge G, Thumbi SM. Safety and efficacy of toll-like receptor agonists as therapeutic agents and vaccine adjuvants for infectious diseases in animals: a systematic review. Front Vet Sci 2024; 11:1428713. [PMID: 39355141 PMCID: PMC11442433 DOI: 10.3389/fvets.2024.1428713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/20/2024] [Indexed: 10/03/2024] Open
Abstract
Introduction Strengthening global health security relies on adequate protection against infectious diseases through vaccination and treatment. Toll-like receptor (TLR) agonists exhibit properties that can enhance immune responses, making them potential therapeutic agents or vaccine adjuvants. Methods We conducted an extensive systematic review to assess the efficacy of TLR agonists as therapeutic agents or vaccine adjuvants for infectious diseases and their safety profile in animals, excluding rodents and cold-blooded animals. We collected qualitative and available quantitative data on the efficacy and safety outcomes of TLR agonists and employed descriptive analysis to summarize the outcomes. Results Among 653 screened studies, 51 met the inclusion criteria. In this review, 82% (42/51) of the studies used TLR agonists as adjuvants, while 18% (9/51) applied TLR agonist as therapeutic agents. The predominant TLR agonists utilized in animals against infectious diseases was CpG ODN, acting as a TLR9 agonist in mammals, and TLR21 agonists in chickens. In 90% (46/51) of the studies, TLR agonists were found effective in stimulating specific and robust humoral and cellular immune responses, thereby enhancing the efficacy of vaccines or therapeutics against infectious diseases in animals. Safety outcomes were assessed in 8% (4/51) of the studies, with one reporting adverse effects. Discussion Although TLR agonists are efficacious in enhancing immune responses and the protective efficacy of vaccines or therapeutic agents against infectious diseases in animals, a thorough evaluation of their safety is imperative to in-form future clinical applications in animal studies. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=323122.
Collapse
Affiliation(s)
- Harriet Oboge
- Department of Public Health Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - Victor Riitho
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - Mutono Nyamai
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - George P Omondi
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
- Department of Clinical Studies, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Anna Lacasta
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - Naftaly Githaka
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - Vishvanath Nene
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
| | - Gabriel Aboge
- Department of Public Health Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - S M Thumbi
- Centre for Epidemiological Modelling and Analysis, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
- Feed the Future Innovation Lab for Animal Health, Washington State University, Pullman, WA, United States
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
6
|
D’Onofrio V, Porrez S, Jacobs B, Alhatemi A, De Boever F, Waerlop G, Michels E, Vanni F, Manenti A, Leroux-Roels G, Platenburg PP, Hilgers L, Leroux-Roels I. Safety and Immunogenicity of a Carbohydrate Fatty Acid Monosulphate Ester Adjuvant Combined with a Low-Dose Quadrivalent Split-Virion Inactivated Influenza Vaccine: A Randomised, Observer-Blind, Active-Controlled, First-in-Human, Phase 1 Study. Vaccines (Basel) 2024; 12:1036. [PMID: 39340066 PMCID: PMC11435821 DOI: 10.3390/vaccines12091036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Seasonal influenza vaccine effectiveness is low. Carbohydrate fatty acid monosulphate ester (CMS), a new oil-in-water adjuvant, has proven potency in animal models with suggested capacity for dose-sparing. The objective was to evaluate safety and immunogenicity of CMS when added to a low-dose influenza vaccine (QIV) in humans. In a randomised, double-blind, active-controlled, first-in-human study, sixty participants (18-50 years) received either 0.5 mg CMS or 2 mg CMS with 1/5th dose QIV, or a full dose QIV without CMS. Adverse events (AE) were monitored until 7 days post-vaccination. Haemagglutinin inhibition (HI) titres in serum and CD4+ T cells in PBMCs were determined at day 0, 7, 28, and 180. Mean age was 37.6 (±10.1) years and 42/60 (70.0%) were female. Pain at injection site (42/60, 86.7%) and headache (34/60, 56.7%) were reported most and more frequently in the 2 mg CMS group. HI titres and the frequency of influenza specific CD4+ T cells were equal across strains for the three cohorts on all visits, increased until day 28 and decreased at day 180 to values higher than baseline. CMS was safe in humans. Humoral and cell-mediated immunogenicity was similar across vaccines, even with 1/5th antigen dose. CMS can have beneficial implications in low-resource settings or in a pandemic context.
Collapse
Affiliation(s)
- Valentino D’Onofrio
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Sharon Porrez
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Bart Jacobs
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Azhar Alhatemi
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Fien De Boever
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Gwenn Waerlop
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Els Michels
- Harmony Clinical Research BV, 9090 Melle, Belgium;
| | - Francesca Vanni
- VisMederi S.r.l., 53035 Monteriggioni, Italy; (F.V.); (A.M.)
| | | | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | | | - Luuk Hilgers
- LiteVax, 4061 BJ Ophemert, The Netherlands; (P.P.P.); (L.H.)
| | - Isabel Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| |
Collapse
|
7
|
Shi Q, Wang Q, Shen Y, Chen S, Gan S, Lin T, Song F, Ma Y. Escherichia coli LTB26 mutant enhances immune responses to rotavirus antigen VP8 in a mouse model. Mol Immunol 2024; 173:10-19. [PMID: 39004021 DOI: 10.1016/j.molimm.2024.07.001] [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: 06/10/2023] [Revised: 06/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Adjuvant is a major supplementary component of vaccines to boost adaptive immune responses. To select an efficient adjuvant from the heat-labile toxin B subunit (LTB) of E. coli, four LTB mutants (numbered LTB26, LTB34, LTB57, and LTB85) were generated by multi-amino acid random replacement. Mice have been intranasally vaccinated with human rotavirus VP8 admixed. Among the four mutants, enzyme-linked immunosorbent assay (ELISA) revealed that LTB26 had enhanced mucosal immune adjuvanticity compared to LTB, showing significantly enhanced immune responses in both serum IgG and mucosal sIgA levels. The 3D modeling analysis suggested that the enhanced immune adjuvanticity of LTB26 might be due to the change of the first LTB α-helix to a β-sheet. The molecular mechanism was studied using transcriptomic and flow cytometric (FCM) analysis. The transcriptomic data demonstrated that LTB26 enhanced immune response by enhancing B cell receptor (BCR) and major histocompatibility complex (MHC) II+-related pathways. Furthermore, LTB26 promoted Th1 and Th2-type immune responses which were confirmed by detecting IFN-γ and IL-4 expression levels. Immunohistochemical analysis demonstrated that LTB26 enhanced both Th1 and Th2 type immunity. Therefore, LTB26 was a potent mucosal immune adjuvant meeting the requirement for use in human clinics in the future.
Collapse
Affiliation(s)
- Qinlin Shi
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qiujuan Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yanxi Shen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijing Chen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijie Gan
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Tao Lin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Fangzhou Song
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yongping Ma
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China.
| |
Collapse
|
8
|
Chen Y, Wang Y, Li Z, Jiang H, Pan W, Liu M, Jiang W, Zhang X, Wang F. Preparation and immunological activity evaluation of an intranasal protein subunit vaccine against ancestral and mutant SARS-CoV-2 with curdlan sulfate/O-linked quaternized chitosan nanoparticles as carrier and adjuvant. Int J Biol Macromol 2024; 276:133733. [PMID: 39002905 DOI: 10.1016/j.ijbiomac.2024.133733] [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: 11/28/2023] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Chitosan and its derivatives are ideal nasal vaccine adjuvant to deliver antigens to immune cells. Previously, we successfully used a chitosan derivative, O-(2-Hydroxyl) propyl-3-trimethyl ammonium chitosan chloride (O-HTCC), and a β-glucan derivative, curdlan sulfate (CS), to prepare a nanoparticle adjuvant CS/O-HTCC which could deliver ovalbumin to antigen presenting cells (APCs) through nasal inhalation. In this article, we used SARS-CoV-2 spike receptor binding domain (S-RBD) as the antigen and CS/O-HTCC nanoparticles as the adjuvant to develop a nasal mucosal protein subunit vaccine, CS/S-RBD/O-HTCC. The humoral immunity, cell-mediated immunity and mucosal immunity induced by vaccines were evaluated. The results showed that CS/S-RBD/O-HTCC could induce desirable immunization with single or bivalent antigen through nasal inoculation, giving one booster vaccination with mutated S-RBD (beta) could bring about a broad cross reaction with ancestral and different mutated S-RBD, and vaccination of the BALB/c mice with CS/S-RBD/O-HTCC containing S-RBD mix antigens (ancestral and omicron) could induce the production of binding and neutralizing antibodies against both of the two antigens. Our results indicate that CS/O-HTCC is a promising nasal mucosal adjuvant to prepare protein subunit vaccine for both primary and booster immunization, and the adjuvant is suitable for loading more than one antigen for preparing multivalent vaccines.
Collapse
MESH Headings
- Chitosan/chemistry
- Animals
- Nanoparticles/chemistry
- beta-Glucans/chemistry
- beta-Glucans/immunology
- SARS-CoV-2/immunology
- Vaccines, Subunit/immunology
- Mice
- Administration, Intranasal
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Adjuvants, Immunologic/pharmacology
- Mice, Inbred BALB C
- COVID-19/prevention & control
- COVID-19/immunology
- Female
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/chemistry
- Antibodies, Viral/immunology
- Immunity, Mucosal/drug effects
- Mutation
- Antibodies, Neutralizing/immunology
- Drug Carriers/chemistry
- Adjuvants, Vaccine/chemistry
- Humans
Collapse
Affiliation(s)
- Yipan Chen
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Yan Wang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Zuyi Li
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Honglei Jiang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Wei Pan
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Minghui Liu
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Wenjie Jiang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China.
| | - Xinke Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Fengshan Wang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China; NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, National Glycoengineering Research Center, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
9
|
Mendoza-Morales LF, Fiorani F, Morán KD, Hecker YP, Cirone KM, Sánchez-López EF, Ramos-Duarte VA, Corigliano MG, Bilbao MG, Clemente M, Moore DP, Sander VA. Immunogenicity, safety and dual DIVA-like character of a recombinant candidate vaccine against neosporosis in cattle. Acta Trop 2024; 257:107293. [PMID: 38901525 DOI: 10.1016/j.actatropica.2024.107293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Neosporosis is the major infectious cause of abortion and reproductive losses in cattle worldwide; however, there are no available vaccines or drugs to control this disease. Recently, a dual (positive and negative) DIVA-like (Differentiation of Infected from Vaccinated Animals) vaccine was evaluated in a pregnant mouse model of neosporosis, showing promising immunogenic and protective results. The current report aimed to study the safety, the dose-dependent immunogenicity and the dual DIVA-like character of a recombinant subunit vaccine composed of the major surface antigen from Neospora caninum (rNcSAG1) and the carrier/adjuvant Heat shock protein 81.2 from Arabidopsis thaliana (rAtHsp81.2) in cattle. Healthy heifers were separated and assigned to experimental groups A-F and subcutaneously immunized with 2 doses of vaccine formulations 30 days apart as follows: A (n = 4): 50 μg rNcSAG1 + 150 μg rAtHsp81.2; B (n = 4): 200 μg rNcSAG1 + 600 μg rAtHsp81.2; C (n = 4): 500 μg rNcSAG1 + 1,500 μg rAtHsp81.2; D (n = 3): 150 μg rAtHsp81.2; E (n = 3):1,500 μg rAtHsp81.2, and F (n = 3) 2 ml of sterile PBS. The immunization of heifers with the different vaccine or adjuvant doses (groups A-E) was demonstrated to be safe and did not modify the mean value of the evaluated serum biomarkers of metabolic function (GOT/ASP, GPT/ALT, UREA, Glucose and total proteins). The kinetics and magnitude of the immune responses were dose-dependent. The higher dose of the vaccine formulation (group C) stimulated a broad and potent humoral and cellular immune response, characterized by an IgG1/IgG2 isotype profile and IFN-γ secretion. In addition, this was the first time that dual DIVA-like character of a vaccine against neosporosis was demonstrated, allowing us to differentiate vaccinated from infected heifers by two different DIVA compliant test approaches. These results encourage us to evaluate its protective efficacy in infected pregnant cattle in the future.
Collapse
Affiliation(s)
- Luisa Fernanda Mendoza-Morales
- Laboratorio de Biotecnologías en Bovinos y Ovinos, INTECH, CONICET-UNSAM, Intendente Marino Km 8,2; CC 164 (B7130IWA), Chascomús, Buenos Aires, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires, Argentina
| | - Franco Fiorani
- Facultad de Ciencias Agrarias, Universidad Nacional de Mar del Plata, Balcarce, Buenos Aires, Argentina; Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), Instituto Nacional de Tecnología Agropecuaria Estación Experimental Agropecuaria Balcarce (CONICET-INTA), Balcarce, Buenos Aires, Argentina
| | - Karen Daiana Morán
- Laboratorio de Reproducción, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), General Pico, La Pampa, Argentina; Facultad de Ciencias Veterinarias, Universidad Nacional de La Pampa, General Pico, La Pampa, Argentina
| | - Yanina Paola Hecker
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), Instituto Nacional de Tecnología Agropecuaria Estación Experimental Agropecuaria Balcarce (CONICET-INTA), Balcarce, Buenos Aires, Argentina
| | - Karina Mariela Cirone
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), Instituto Nacional de Tecnología Agropecuaria Estación Experimental Agropecuaria Balcarce (CONICET-INTA), Balcarce, Buenos Aires, Argentina
| | - Edwin Fernando Sánchez-López
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires, Argentina; Laboratorio de Molecular Farming y Vacunas, INTECH, CONICET-UNSAM, Chascomús, Argentina
| | - Victor Andrés Ramos-Duarte
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires, Argentina; Laboratorio de Molecular Farming y Vacunas, INTECH, CONICET-UNSAM, Chascomús, Argentina
| | - Mariana Georgina Corigliano
- Laboratorio de Biotecnologías en Bovinos y Ovinos, INTECH, CONICET-UNSAM, Intendente Marino Km 8,2; CC 164 (B7130IWA), Chascomús, Buenos Aires, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires, Argentina
| | - María Guillermina Bilbao
- Laboratorio de Reproducción, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), General Pico, La Pampa, Argentina; Facultad de Ciencias Veterinarias, Universidad Nacional de La Pampa, General Pico, La Pampa, Argentina
| | - Marina Clemente
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires, Argentina; Laboratorio de Molecular Farming y Vacunas, INTECH, CONICET-UNSAM, Chascomús, Argentina
| | - Dadín Prando Moore
- Facultad de Ciencias Agrarias, Universidad Nacional de Mar del Plata, Balcarce, Buenos Aires, Argentina; Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), Instituto Nacional de Tecnología Agropecuaria Estación Experimental Agropecuaria Balcarce (CONICET-INTA), Balcarce, Buenos Aires, Argentina
| | - Valeria Analía Sander
- Laboratorio de Biotecnologías en Bovinos y Ovinos, INTECH, CONICET-UNSAM, Intendente Marino Km 8,2; CC 164 (B7130IWA), Chascomús, Buenos Aires, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Honorato L, Bonilla JJA, Valdez AF, Frases S, Araújo GRDS, Sabino ALRDN, da Silva NM, Ribeiro L, Ferreira MDS, Kornetz J, Rodrigues ML, Cunningham I, Gow NAR, Gacser A, Guimarães AJ, Dutra FF, Nimrichter L. Toll-like receptor 4 (TLR4) is the major pattern recognition receptor triggering the protective effect of a Candida albicans extracellular vesicle-based vaccine prototype in murine systemic candidiasis. mSphere 2024; 9:e0046724. [PMID: 39037263 PMCID: PMC11351041 DOI: 10.1128/msphere.00467-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024] Open
Abstract
Systemic candidiasis remains a significant public health concern worldwide, with high mortality rates despite available antifungal drugs. Drug-resistant strains add to the urgency for alternative therapies. In this context, vaccination has reemerged as a prominent immune-based strategy. Extracellular vesicles (EVs), nanosized lipid bilayer particles, carry a diverse array of native fungal antigens, including proteins, nucleic acids, lipids, and glycans. Previous studies from our laboratory demonstrated that Candida albicans EVs triggered the innate immune response, activating bone marrow-derived dendritic cells (BMDCs) and potentially acting as a bridge between innate and adaptive immunity. Vaccination with C. albicans EVs induced the production of specific antibodies, modulated cytokine production, and provided protection in immunosuppressed mice infected with lethal C. albicans inoculum. To elucidate the mechanisms underlying EV-induced immune activation, our study investigated pathogen-associated molecular patterns (PAMPs) and pattern recognition receptors (PRRs) involved in EVs-phagocyte engagement. EVs from wild-type and mutant C. albicans strains with truncated mannoproteins were compared for their ability to stimulate BMDCs. Our findings revealed that EV decoration with O- and N-linked mannans and the presence of β-1,3-glucans and chitin oligomers may modulate the activation of specific PRRs, in particular Toll-like receptor 4 (TLR4) and dectin-1. The protective effect of vaccination with wild-type EVs was found to be dependent on TLR4. These results suggest that fungal EVs can be harnessed in vaccine formulations to selectively activate PRRs in phagocytes, offering potential avenues for combating or preventing candidiasis.IMPORTANCESystemic candidiasis is a serious global health concern with high mortality rates and growing drug resistance. Vaccination offers a promising solution. A unique approach involves using tiny lipid-coated particles called extracellular vesicles (EVs), which carry various fungal components. Previous studies found that Candida albicans EVs activate the immune response and may bridge the gap between innate and adaptive immunity. To understand this better, we investigated how these EVs activate immune cells. We demonstrated that specific components on EV surfaces, such as mannans and glucans, interact with receptors on immune cells, including Toll-like receptor 4 (TLR4) and dectin-1. Moreover, vaccinating with these EVs led to strong immune responses and full protection in mice infected with Candida. This work shows how harnessing fungal EVs might lead to effective vaccines against candidiasis.
Collapse
Affiliation(s)
- Leandro Honorato
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jhon J. Artunduaga Bonilla
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro F. Valdez
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susana Frases
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filhos (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
| | - Glauber Ribeiro de Sousa Araújo
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filhos (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Natalia Martins da Silva
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Ribeiro
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marina da Silva Ferreira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Julio Kornetz
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcio L. Rodrigues
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Carlos Chagas (ICC), Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
| | - Iain Cunningham
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Neil A. R. Gow
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Attila Gacser
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Allan J. Guimarães
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Fabianno F. Dutra
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Nimrichter
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rede Micologia, RJ, FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Zhong L, Zhang W, Liu H, Zhang X, Yang Z, Wen Z, Chen L, Chen H, Luo Y, Chen Y, Feng Q, Zeng MS, Zhao Q, Liu L, Krummenacher C, Zeng YX, Chen Y, Xu M, Zhang X. A cocktail nanovaccine targeting key entry glycoproteins elicits high neutralizing antibody levels against EBV infection. Nat Commun 2024; 15:5310. [PMID: 38906867 PMCID: PMC11192767 DOI: 10.1038/s41467-024-49546-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 06/10/2024] [Indexed: 06/23/2024] Open
Abstract
Epstein-Barr virus (EBV) infects more than 95% of adults worldwide and is closely associated with various malignancies. Considering the complex life cycle of EBV, developing vaccines targeting key entry glycoproteins to elicit robust and durable adaptive immune responses may provide better protection. EBV gHgL-, gB- and gp42-specific antibodies in healthy EBV carriers contributed to sera neutralizing abilities in vitro, indicating that they are potential antigen candidates. To enhance the immunogenicity of these antigens, we formulate three nanovaccines by co-delivering molecular adjuvants (CpG and MPLA) and antigens (gHgL, gB or gp42). These nanovaccines induce robust humoral and cellular responses through efficient activation of dendritic cells and germinal center response. Importantly, these nanovaccines generate high levels of neutralizing antibodies recognizing vulnerable sites of all three antigens. IgGs induced by a cocktail vaccine containing three nanovaccines confer superior protection from lethal EBV challenge in female humanized mice compared to IgG elicited by individual NP-gHgL, NP-gB and NP-gp42. Importantly, serum antibodies elicited by cocktail nanovaccine immunization confer durable protection against EBV-associated lymphoma. Overall, the cocktail nanovaccine shows robust immunogenicity and is a promising candidate for further clinical trials.
Collapse
Affiliation(s)
- Ling Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wanlin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Hong Liu
- Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Xinyu Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zeyu Yang
- Center for Functional Biomaterials, School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Zhenfu Wen
- Center for Functional Biomaterials, School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Ling Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Haolin Chen
- Center for Functional Biomaterials, School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Yanran Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yanhong Chen
- Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, 475004, China
| | - Qisheng Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China
| | - Lixin Liu
- Center for Functional Biomaterials, School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Claude Krummenacher
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ, USA.
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Yongming Chen
- College of Chemistry and Molecular Science, Henan University, Zhengzhou, 450046, China.
| | - Miao Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Xiao Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
12
|
Ahmed M, Kurungottu P, Swetha K, Atla S, Ashok N, Nagamalleswari E, Bonam SR, Sahu BD, Kurapati R. Role of NLRP3 inflammasome in nanoparticle adjuvant-mediated immune response. Biomater Sci 2024. [PMID: 38867716 DOI: 10.1039/d4bm00439f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is pivotal in orchestrating the immune response induced by nanoparticle adjuvants. Understanding the intricate mechanisms underlying the activation of NLRP3 inflammasome by these adjuvants is crucial for deciphering their immunomodulatory properties. This review explores the involvement of the NLRP3 inflammasome in mediating immune responses triggered by nanoparticle adjuvants. It delves into the signaling pathways and cellular mechanisms involved in NLRP3 activation, highlighting its significance in modulating the efficacy and safety of nanoparticle-based adjuvants. A comprehensive grasp of the interplay between NLRP3 inflammasome and nanoparticle adjuvants holds promise for optimizing vaccine design and advancing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Momitul Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Pavithra Kurungottu
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - K Swetha
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Sandeep Atla
- Texas A&M Drug Discovery Center, Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | - Nivethitha Ashok
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Easa Nagamalleswari
- MTCC and Gene Bank, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| |
Collapse
|
13
|
Zhang H, Heng X, Yang H, Rao Y, Yao L, Zhu Z, Chen G, Chen H. Metal-Free Atom Transfer Radical Polymerization to Prepare Recylable Micro-Adjuvants for Dendritic Cell Vaccine. Angew Chem Int Ed Engl 2024; 63:e202402853. [PMID: 38598262 DOI: 10.1002/anie.202402853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Abstract
In the development of dendritic cell (DC) vaccines, the maturation of DCs is a critical stage. Adjuvants play a pivotal role in the maturation of DCs, with a major concern being to ensure both efficacy and safety. This study introduces an innovative approach that combines high efficacy with safety through the synthesis of micro-adjuvants grafted with copolymers of 2-(methacrylamido) glucopyranose (MAG) and methacryloxyethyl trimethyl ammonium chloride (DMC). The utilization of metal-free surface-initiated atom transfer radical polymerization enables the production of safe and recyclable adjuvants. These micrometer-sized adjuvants surpass the optimal size range for cellular endocytosis, enabling the retrieval and reuse of them during the ex vivo maturation process, mitigating potential toxicity concerns associated with the endocytosis of non-metabolized nanoparticles. Additionally, the adjuvants exhibit a "micro-ligand-mediated maturation enhancement" effect for DC maturation. This effect is influenced by the shape of the particle, as evidenced by the distinct promotion effects of rod-like and spherical micro-adjuvants with comparable sizes. Furthermore, the porous structure of the adjuvants enables them to function as cargo-carrying "micro-shuttles", releasing antigens upon binding to DCs to facilitate efficient antigen delivery.
Collapse
Affiliation(s)
- Hengyuan Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xingyu Heng
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - He Yang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yu Rao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Lihua Yao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Zhichen Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Gaojian Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hong Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| |
Collapse
|
14
|
Saldanha L, Langel Ü, Vale N. A Physiologically Based Pharmacokinetic (PBPK) Study to Assess the Adjuvanticity of Three Peptides in an Oral Vaccine. Pharmaceutics 2024; 16:780. [PMID: 38931901 PMCID: PMC11207434 DOI: 10.3390/pharmaceutics16060780] [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: 04/26/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Following up on the first PBPK model for an oral vaccine built for alpha-tocopherol, three peptides are explored in this article to verify if they could support an oral vaccine formulation as adjuvants using the same PBPK modeling approach. A literature review was conducted to verify what peptides have been used as adjuvants in the last decades, and it was noticed that MDP derivatives have been used, with one of them even being commercially approved and used as an adjuvant when administered intravenously in oncology. The aim of this study was to build optimized models for three MDP peptides (MDP itself, MTP-PE, and murabutide) and to verify if they could act as adjuvants for an oral vaccine. Challenges faced by peptides in an oral delivery system are taken into consideration, and improvements to the formulations to achieve better results are described in a step-wise approach to reach the most-optimized model. Once simulations are performed, results are compared to determine what would be the best peptide to support as an oral adjuvant. According to our results, MTP-PE, the currently approved and commercialized peptide, could have potential to be incorporated into an oral formulation. It would be interesting to proceed with further in vivo experiments to determine the behavior of this peptide when administered orally with a proper formulation to overcome the challenges of oral delivery systems.
Collapse
Affiliation(s)
- Leonor Saldanha
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ülo Langel
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia;
- Department of Biochemistry and Biophysics, Stockholm University, 10691 Stockholm, Sweden
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
15
|
Qian G, Gao C, Zhang M, Chen Y, Xie L. A Review of Protein-Based COVID-19 Vaccines: From Monovalent to Multivalent Formulations. Vaccines (Basel) 2024; 12:579. [PMID: 38932308 PMCID: PMC11209593 DOI: 10.3390/vaccines12060579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
The emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulting in the COVID-19 pandemic, has profoundly impacted global healthcare systems and the trajectory of economic advancement. As nations grapple with the far-reaching consequences of this unprecedented health crisis, the administration of COVID-19 vaccines has proven to be a pivotal strategy in managing this crisis. Protein-based vaccines have garnered significant attention owing to their commendable safety profile and precise immune targeting advantages. Nonetheless, the unpredictable mutations and widespread transmission of SARS-CoV-2 have posed challenges for vaccine developers and governments worldwide. Monovalent and multivalent vaccines represent two strategies in COVID-19 vaccine development, with ongoing controversy surrounding their efficacy. This review concentrates on the development of protein-based COVID-19 vaccines, specifically addressing the transition from monovalent to multivalent formulations, and synthesizes data on vaccine manufacturers, antigen composition, pivotal clinical study findings, and other features that shape their distinct profiles and overall effectiveness. Our hypothesis is that multivalent vaccine strategies for COVID-19 could offer enhanced capability with broad-spectrum protection.
Collapse
Affiliation(s)
- Gui Qian
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (G.Q.); (C.G.); (M.Z.); (Y.C.)
| | - Cuige Gao
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (G.Q.); (C.G.); (M.Z.); (Y.C.)
| | - Miaomiao Zhang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (G.Q.); (C.G.); (M.Z.); (Y.C.)
| | - Yuanxin Chen
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (G.Q.); (C.G.); (M.Z.); (Y.C.)
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China; (G.Q.); (C.G.); (M.Z.); (Y.C.)
- Cell Culture Engineering Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100006, China
| |
Collapse
|
16
|
Saleemi MA, Zhang Y, Zhang G. Current Progress in the Science of Novel Adjuvant Nano-Vaccine-Induced Protective Immune Responses. Pathogens 2024; 13:441. [PMID: 38921739 PMCID: PMC11206999 DOI: 10.3390/pathogens13060441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Vaccinations are vital as they protect us from various illness-causing agents. Despite all the advancements in vaccine-related research, developing improved and safer vaccines against devastating infectious diseases including Ebola, tuberculosis and acquired immune deficiency syndrome (AIDS) remains a significant challenge. In addition, some of the current human vaccines can cause adverse reactions in some individuals, which limits their use for massive vaccination program. Therefore, it is necessary to design optimal vaccine candidates that can elicit appropriate immune responses but do not induce side effects. Subunit vaccines are relatively safe for the vaccination of humans, but they are unable to trigger an optimal protective immune response without an adjuvant. Although different types of adjuvants have been used for the formulation of vaccines to fight pathogens that have high antigenic diversity, due to the toxicity and safety issues associated with human-specific adjuvants, there are only a few adjuvants that have been approved for the formulation of human vaccines. Recently, nanoparticles (NPs) have gain specific attention and are commonly used as adjuvants for vaccine development as well as for drug delivery due to their excellent immune modulation properties. This review will focus on the current state of adjuvants in vaccine development, the mechanisms of human-compatible adjuvants and future research directions. We hope this review will provide valuable information to discovery novel adjuvants and drug delivery systems for developing novel vaccines and treatments.
Collapse
Affiliation(s)
| | | | - Guoquan Zhang
- Department of Molecular Microbiology and Immunology, College of Sciences, University of Texas at San Antonio, San Antonio, TX 78249, USA; (M.A.S.); (Y.Z.)
| |
Collapse
|
17
|
Meng X, Xu Y, Yang J, Meng S, Ding N, Sun T, Zong C. Strategic development of a self-adjuvanting SARS-CoV-2 RBD vaccine: From adjuvant screening to enhanced immunogenicity with a modified TLR7 agonist. Int Immunopharmacol 2024; 132:111909. [PMID: 38554446 DOI: 10.1016/j.intimp.2024.111909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/10/2024] [Accepted: 03/19/2024] [Indexed: 04/01/2024]
Abstract
Adjuvants enhance the body's immune response to a vaccine, often leading to better protection against diseases. Monophosphoryl lipid A analogues (MPLA, TLR4 agonists), α-galactosylceramide analogues (NKT cell agonists), and imidazoquinoline compounds (TLR7/8 agonists) are emerging novel adjuvants on market or under clinical trials. Despite significant interest in these adjuvants, a direct comparison of their adjuvant activities remains unexplored. We initially assessed the activities of various adjuvants from three distinct categories using the SARS-CoV-2 RBD trimer antigen. TLR4 and TLR7/8 agonists are discovered to elicit robust IgG2a/2b antibodies, which is crucial for eliciting antibody dependent cytotoxicity. While α-galactosylceramide analogs induced mainly IgG1 antibody. Then, because of the flexibility of the TLR7/8 agonist, we designed and synthesized a tri-component self-adjuvanting SARS-CoV-2 RBD vaccine, featuring a covalent TLR7 agonist and targeting mannoside. Animal studies indicated that this vaccine generated antigen-specific humoral immunity. Yet, its immunogenicity seems compromised, indicating the complexity of the vaccine.
Collapse
Affiliation(s)
- Xiongyan Meng
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Ying Xu
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Jing Yang
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Shuai Meng
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Ning Ding
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Tiantian Sun
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Chengli Zong
- School of Pharmaceutical Sciences, School of Marine Biology and Fisheries, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China.
| |
Collapse
|
18
|
Hiller J, Göen T, Drexler H, Berking C, Wagner N. Elevated aluminum excretion in patients by long-term subcutaneous immunotherapy - A cross-sectional case-control study. Int J Hyg Environ Health 2024; 258:114337. [PMID: 38461738 DOI: 10.1016/j.ijheh.2024.114337] [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: 10/31/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Aluminum (Al) adjuvants have been used in vaccines and subcutaneous immunotherapy (SCIT) for decades. Despite indisputable neurotoxic properties of Al, there is no clear evidence of a causal relationship between their use and any neurotoxic side effects. However, recent rat studies have shown an accumulation of Al from adjuvants in tissues, especially in bones. OBJECTIVES Since the human toxicokinetics of Al-adjuvants are poorly understood, this study aimed to evaluate whether up-dosed or long-term SCIT with Al-coupled extracts leads to increased Al load in humans. METHODS This observational cross-sectional case-control study explored Al excretion in hymenoptera venom allergy patients recruited in 2020 before initiation (n = 10) and during ongoing (n = 12) SCIT with Al-based preparations. Urine samples were collected before and 24 h after the SCIT injections and analyzed for aluminum content by using atomic absorption spectrometry. The cumulative administered Al dose was extracted from patient records. Patients receiving long-term immunotherapy were treated between 2.8 and 13.6 years (mean 7.1). Other potential sources of Al exposure were surveyed. RESULTS Patients who had received Al-coupled immunotherapy for several years showed significantly (p < 0.001) higher Al excretion than the controls at initiation of immunotherapy (mean 18.2 μg/gC vs. 7.9 μg/gC) and predominantly (73%) were above the 95th percentile of the general populations' exposure (>15 μg/gC), however, without reaching levels of toxicological concern (>50 μg/gC). Taking both groups together excreted Al levels correlated with the cumulative administered Al dose from SCIT (linear regression: Alurine = 8.258 + 0.133*Alcum; p = 0.001). DISCUSSION These results suggest a relevant iatrogenic contribution of long-term SCIT to human internal Al burden and potential accumulation. Considering the medical benefits of Al-adjuvants and SCIT a differentiated risk-benefit analysis is needed. For certain scenarios of potential toxicological concern in clinical practice biomonitoring might be advisable.
Collapse
Affiliation(s)
- Julia Hiller
- Institute and Outpatient Clinic of Occupational, Social, and Environmental Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestraße 9-11, 91054, Erlangen, Germany.
| | - Thomas Göen
- Institute and Outpatient Clinic of Occupational, Social, and Environmental Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestraße 9-11, 91054, Erlangen, Germany.
| | - Hans Drexler
- Institute and Outpatient Clinic of Occupational, Social, and Environmental Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestraße 9-11, 91054, Erlangen, Germany.
| | - Carola Berking
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054, Erlangen, Germany.
| | - Nicola Wagner
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054, Erlangen, Germany.
| |
Collapse
|
19
|
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
|
20
|
Nurdin A, Movieta Nency Y, Maddeppungeng M, Sekartini R, Mulia Sari R, Surachman F, Fitry Yani F, Raveinal, Anggrainy F, Hafiz A, Linosefa, Machmud R, Awaliyah Deza P, Rujiana V, Bella Rahimi M, Farhanah N, Gundi Pramudo S, Hapsari R, Tri Anantyo D, Mulyono, Mahati E, Maharani N, Darma S, Husni Esa Darussalam A, Shakinah S, Nasrum Massi M, Soedjatmiko. Immunogenicity and safety of SARS-CoV-2 recombinant protein subunit vaccine (IndoVac) adjuvanted with alum and CpG 1018 in Indonesian adults: A phase 3, randomized, active-controlled, multicenter trial. Vaccine 2024; 42:3009-3017. [PMID: 38575433 DOI: 10.1016/j.vaccine.2024.03.077] [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: 12/14/2023] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Bio Farma has developed a recombinant protein subunit vaccine (IndoVac) that is indicated for active immunization in population of all ages. This article reported the results of the phase 3 immunogenicity and safety study in Indonesian adults aged 18 years and above. METHODS We conducted a randomized, active-controlled, multicenter, prospective intervention study to evaluate the immunogenicity and safety of IndoVac in adults aged 18 years and above. Participants who were SARS-CoV-2 vaccine-naïve received two doses of either IndoVac or control (Covovax) with 28 days interval between doses and were followed up until 12 months after complete vaccination. RESULTS A total of 4050 participants were enrolled from June to August 2022 and received at least one dose of vaccine. The geometric mean ratio (GMR) of neutralizing antibody at 14 days after the second dose was 1.01 (95 % confidence interval (CI) 0.89-1.16), which met the WHO non-inferiority criteria for immunobridging (95 % CI lower bound > 0.67). The antibody levels were maintained through 12 months after the second dose. The incidence rate of adverse events (AEs) were 27.95 % in IndoVac group and 32.15 % in Covovax group with mostly mild intensity (27.70 %). The most reported solicited AEs were pain (14.69 %) followed by myalgia (7.48 %) and fatigue (6.77 %). Unsolicited AEs varied, with each of the incidence rate under 5 %. There were no serious AEs assessed as possibly, probably, or likely related to vaccine. CONCLUSIONS IndoVac in adults showed favourable safety profile and elicited non-inferior immune response to Covovax. (ClinicalTrials.gov: NCT05433285, Indonesian Clinical Research Registry: INA-R5752S9).
Collapse
Affiliation(s)
| | | | | | - Rini Sekartini
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | | | | | - Raveinal
- Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | | | - Al Hafiz
- Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | - Linosefa
- Faculty of Medicine, Universitas Andalas, Padang, Indonesia
| | | | | | | | | | - Nur Farhanah
- Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | | | | | | | - Mulyono
- Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Endang Mahati
- Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Nani Maharani
- Faculty of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Sidrah Darma
- Faculty of Medicine, Universitas Muslim Indonesia, Makassar, Indonesia
| | | | | | | | - Soedjatmiko
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
21
|
Arunachalam AB. Vaccines Induce Homeostatic Immunity, Generating Several Secondary Benefits. Vaccines (Basel) 2024; 12:396. [PMID: 38675778 PMCID: PMC11053716 DOI: 10.3390/vaccines12040396] [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: 02/27/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The optimal immune response eliminates invading pathogens, restoring immune equilibrium without inflicting undue harm to the host. However, when a cascade of immunological reactions is triggered, the immune response can sometimes go into overdrive, potentially leading to harmful long-term effects or even death. The immune system is triggered mostly by infections, allergens, or medical interventions such as vaccination. This review examines how these immune triggers differ and why certain infections may dysregulate immune homeostasis, leading to inflammatory or allergic pathology and exacerbation of pre-existing conditions. However, many vaccines generate an optimal immune response and protect against the consequences of pathogen-induced immunological aggressiveness, and from a small number of unrelated pathogens and autoimmune diseases. Here, we propose an "immuno-wave" model describing a vaccine-induced "Goldilocks immunity", which leaves fine imprints of both pro-inflammatory and anti-inflammatory milieus, derived from both the innate and the adaptive arms of the immune system, in the body. The resulting balanced, 'quiet alert' state of the immune system may provide a jump-start in the defense against pathogens and any associated pathological inflammatory or allergic responses, allowing vaccines to go above and beyond their call of duty. In closing, we recommend formally investigating and reaping many of the secondary benefits of vaccines with appropriate clinical studies.
Collapse
Affiliation(s)
- Arun B Arunachalam
- Analytical Sciences, R&D Sanofi Vaccines, 1 Discovery Dr., Swiftwater, PA 18370, USA
| |
Collapse
|
22
|
Su J, Harati Taji Z, Kosinska AD, Ates Oz E, Xie Z, Bielytskyi P, Shein M, Hagen P, Esmaeili S, Steiger K, Protzer U, Schütz AK. Introducing adjuvant-loaded particulate hepatitis B core antigen as an alternative therapeutic hepatitis B vaccine component. JHEP Rep 2024; 6:100997. [PMID: 38425450 PMCID: PMC10904195 DOI: 10.1016/j.jhepr.2023.100997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 11/27/2023] [Accepted: 12/19/2023] [Indexed: 03/02/2024] Open
Abstract
Background & Aims Particulate hepatitis B core antigen (HBcoreAg) is a potent immunogen used as a vaccine carrier platform. HBcoreAg produced in E. coli encapsidates random bacterial RNA (bRNA). Using the heterologous protein-prime, viral-vector-boost therapeutic hepatitis B vaccine TherVacB, we compared the properties of different HBcoreAg forms. We explored how the content of HBcoreAg modulates antigen stability, immunogenicity, and antiviral efficacy. Methods bRNA was removed from HBcoreAg by capsid disassembly, followed by reassembly in the absence or presence of specific nucleic acid-based adjuvants poly I:C or CpG. The morphology and structure of empty, bRNA-containing and adjuvant-loaded HBcoreAg were monitored by electron microscopy and nuclear magnetic resonance spectroscopy. Empty, bRNA-containing or adjuvant-loaded HBcoreAg were applied together with HBsAg and with or without nucleic acid-based external adjuvants within the TherVacB regimen in both wild-type and HBV-carrier mice. Results While HBcoreAg retained its structure upon bRNA removal, its stability and immunogenicity decreased significantly. Loading HBcoreAg with nucleic acid-based adjuvants re-established stability of the capsid-like antigen. Immunization with poly I:C- or CpG-loaded HBcoreAg induced high antibody titers against co-administered HBsAg. When applied within the TherVacB regimen, they activated vigorous HBcoreAg- and HBsAg-specific T-cell responses in wild-type and HBV-carrier mice, requiring a significantly lower dose of adjuvant compared to externally added adjuvant. Finally, immunization with adjuvant-loaded HBcoreAg mixed with HBsAg led to long-term control of persistent HBV replication in the HBV-carrier mice. Conclusion Adjuvant-loaded HBcoreAg retained capsid integrity and stability, was as immunogenic in vivo as externally adjuvanted HBcoreAg, requiring lower adjuvant levels, and supported immunity against co-administered, non-adjuvanted HBsAg. Thus, adjuvant-loaded HBcoreAg represents a promising novel platform for vaccine development. Impact and implications Hepatitis B core antigen (HBcoreAg) recapitulates the capsid of the HBV that hosts the viral genome. Produced recombinantly, it is not infectious but emerges as a potent immunogen in vaccine development. In this preclinical study, we show that loading HBcoreAg with defined nucleic-acid-based adjuvants on the one hand stabilizes the HBcoreAg with standardized capsid content and, on the other hand, efficiently promotes the immunity of HBcoreAg and a co-administered antigen, allowing for reduced adjuvant doses. Therefore, adjuvant-loaded HBcoreAg not only serves as an encouraging option for therapeutic hepatitis B vaccines, but could also act as an efficient adjuvant delivery system for other types of vaccine.
Collapse
Affiliation(s)
- Jinpeng Su
- Institute of Virology, Technical University of Munich / Helmholtz Munich, 81675, Munich, Germany
- German Center for Infection Research (DZIF), Munich partner site, Germany
| | - Zahra Harati Taji
- Ludwig Maximilians University of Munich, 81377, Munich, Germany
- Bavarian NMR Center, Technical University of Munich, 85748, Garching, Germany
- Institute of Structural Biology, Helmholtz Munich, 85764, Neuherberg, Germany
| | - Anna D. Kosinska
- Institute of Virology, Technical University of Munich / Helmholtz Munich, 81675, Munich, Germany
- German Center for Infection Research (DZIF), Munich partner site, Germany
| | - Edanur Ates Oz
- Institute of Virology, Technical University of Munich / Helmholtz Munich, 81675, Munich, Germany
| | - Zhe Xie
- Institute of Virology, Technical University of Munich / Helmholtz Munich, 81675, Munich, Germany
| | - Pavlo Bielytskyi
- Bavarian NMR Center, Technical University of Munich, 85748, Garching, Germany
- Institute of Structural Biology, Helmholtz Munich, 85764, Neuherberg, Germany
| | - Mikhail Shein
- Ludwig Maximilians University of Munich, 81377, Munich, Germany
- Bavarian NMR Center, Technical University of Munich, 85748, Garching, Germany
- Institute of Structural Biology, Helmholtz Munich, 85764, Neuherberg, Germany
| | - Philipp Hagen
- Institute of Virology, Technical University of Munich / Helmholtz Munich, 81675, Munich, Germany
| | - Shohreh Esmaeili
- Ludwig Maximilians University of Munich, 81377, Munich, Germany
- Bavarian NMR Center, Technical University of Munich, 85748, Garching, Germany
- Institute of Structural Biology, Helmholtz Munich, 85764, Neuherberg, Germany
| | - Katja Steiger
- Comparative Experimental Pathology, Institute of Pathology, School of Medicine and Health, Technical University Munich, 81675, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich / Helmholtz Munich, 81675, Munich, Germany
- German Center for Infection Research (DZIF), Munich partner site, Germany
| | - Anne K. Schütz
- Ludwig Maximilians University of Munich, 81377, Munich, Germany
- Bavarian NMR Center, Technical University of Munich, 85748, Garching, Germany
- Institute of Structural Biology, Helmholtz Munich, 85764, Neuherberg, Germany
| |
Collapse
|
23
|
Pullen RH, Sassano E, Agrawal P, Escobar J, Chehtane M, Schanen B, Drake DR, Luna E, Brennan RJ. A Predictive Model of Vaccine Reactogenicity Using Data from an In Vitro Human Innate Immunity Assay System. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:904-916. [PMID: 38276072 DOI: 10.4049/jimmunol.2300185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 01/02/2024] [Indexed: 01/27/2024]
Abstract
A primary concern in vaccine development is safety, particularly avoiding an excessive immune reaction in an otherwise healthy individual. An accurate prediction of vaccine reactogenicity using in vitro assays and computational models would facilitate screening and prioritization of novel candidates early in the vaccine development process. Using the modular in vitro immune construct model of human innate immunity, PBMCs from 40 healthy donors were treated with 10 different vaccines of varying reactogenicity profiles and then cell culture supernatants were analyzed via flow cytometry and a multichemokine/cytokine assay. Differential response profiles of innate activity and cell viability were observed in the system. In parallel, an extensive adverse event (AE) dataset for the vaccines was assembled from clinical trial data. A novel reactogenicity scoring framework accounting for the frequency and severity of local and systemic AEs was applied to the clinical data, and a machine learning approach was employed to predict the incidence of clinical AEs from the in vitro assay data. Biomarker analysis suggested that the relative levels of IL-1B, IL-6, IL-10, and CCL4 have higher predictive importance for AE risk. Predictive models were developed for local reactogenicity, systemic reactogenicity, and specific individual AEs. A forward-validation study was performed with a vaccine not used in model development, Trumenba (meningococcal group B vaccine). The clinically observed Trumenba local and systemic reactogenicity fell on the 26th and 93rd percentiles of the ranges predicted by the respective models. Models predicting specific AEs were less accurate. Our study presents a useful framework for the further development of vaccine reactogenicity predictive models.
Collapse
|
24
|
Song Y, Mehl F, Zeichner SL. Vaccine Strategies to Elicit Mucosal Immunity. Vaccines (Basel) 2024; 12:191. [PMID: 38400174 PMCID: PMC10892965 DOI: 10.3390/vaccines12020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccines are essential tools to prevent infection and control transmission of infectious diseases that threaten public health. Most infectious agents enter their hosts across mucosal surfaces, which make up key first lines of host defense against pathogens. Mucosal immune responses play critical roles in host immune defense to provide durable and better recall responses. Substantial attention has been focused on developing effective mucosal vaccines to elicit robust localized and systemic immune responses by administration via mucosal routes. Mucosal vaccines that elicit effective immune responses yield protection superior to parenterally delivered vaccines. Beyond their valuable immunogenicity, mucosal vaccines can be less expensive and easier to administer without a need for injection materials and more highly trained personnel. However, developing effective mucosal vaccines faces many challenges, and much effort has been directed at their development. In this article, we review the history of mucosal vaccine development and present an overview of mucosal compartment biology and the roles that mucosal immunity plays in defending against infection, knowledge that has helped inform mucosal vaccine development. We explore new progress in mucosal vaccine design and optimization and novel approaches created to improve the efficacy and safety of mucosal vaccines.
Collapse
Affiliation(s)
- Yufeng Song
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Frances Mehl
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Steven L. Zeichner
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
25
|
Kanuri SH, Sirrkay PJ. Adjuvants in COVID-19 vaccines: innocent bystanders or culpable abettors for stirring up COVID-heart syndrome. Ther Adv Vaccines Immunother 2024; 12:25151355241228439. [PMID: 38322819 PMCID: PMC10846003 DOI: 10.1177/25151355241228439] [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: 08/18/2023] [Accepted: 01/05/2024] [Indexed: 02/08/2024] Open
Abstract
COVID-19 infection is a multi-system clinical disorder that was associated with increased morbidity and mortality. Even though antiviral therapies such as Remdesvir offered modest efficacy in reducing the mortality and morbidity, they were not efficacious in reducing the risk of future infections. So, FDA approved COVID-19 vaccines which are widely administered in the general population worldwide. These COVID-19 vaccines offered a safety net against future infections and re-infections. Most of these vaccines contain inactivated virus or spike protein mRNA that are primarily responsible for inducing innate and adaptive immunity. These vaccines were also formulated to contain supplementary adjuvants that are beneficial in boosting the immune response. During the pandemic, clinicians all over the world witnessed an uprise in the incidence and prevalence of cardiovascular diseases (COVID-Heart Syndrome) in patients with and without cardiovascular risk factors. Clinical researchers were not certain about the underlying reason for the upsurge of cardiovascular disorders with some blaming them on COVID-19 infections while others blaming them on COVID-19 vaccines. Based on the literature review, we hypothesize that adjuvants included in the COVID-19 vaccines are the real culprits for causation of cardiovascular disorders. Operation of various pathological signaling events under the influence of these adjuvants including autoimmunity, bystander effect, direct toxicity, anti-phospholipid syndrome (APS), anaphylaxis, hypersensitivity, genetic susceptibility, epitope spreading, and anti-idiotypic antibodies were partially responsible for stirring up the onset of cardiovascular disorders. With these mechanisms in place, a minor contribution from COVID-19 virus itself cannot be ruled out. With that being said, we strongly advocate for careful selection of vaccine adjuvants included in COVID-19 vaccines so that future adverse cardiac disorders can be averted.
Collapse
Affiliation(s)
- Sri Harsha Kanuri
- Research Fellow, Stark Neurosciences Institute, Indiana University School of Medicine, 320 W 15 ST, Indianapolis, IN 46202, USA
| | | |
Collapse
|
26
|
Zhu S, Nie Z, Che Y, Shu J, Wu S, He Y, Wu Y, Qian H, Feng H, Zhang Q. The Chinese Hamster Ovary Cell-Based H9 HA Subunit Avian Influenza Vaccine Provides Complete Protection against the H9N2 Virus Challenge in Chickens. Viruses 2024; 16:163. [PMID: 38275973 PMCID: PMC10821000 DOI: 10.3390/v16010163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/08/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
(1) Background: Avian influenza has attracted widespread attention because of its severe effect on the poultry industry and potential threat to human health. The H9N2 subtype of avian influenza viruses was the most prevalent in chickens, and there are several commercial vaccines available for the prevention of the H9N2 subtype of avian influenza viruses. However, due to the prompt antigenic drift and antigenic shift of influenza viruses, outbreaks of H9N2 viruses still continuously occur, so surveillance and vaccine updates for H9N2 subtype avian influenza viruses are particularly important. (2) Methods: In this study, we constructed a stable Chinese hamster ovary cell line (CHO) to express the H9 hemagglutinin (HA) protein of the major prevalent H9N2 strain A/chicken/Daye/DY0602/2017 with genetic engineering technology, and then a subunit H9 avian influenza vaccine was prepared using the purified HA protein with a water-in-oil adjuvant. (3) Results: The results showed that the HI antibodies significantly increased after vaccination with the H9 subunit vaccine in specific-pathogen-free (SPF) chickens with a dose-dependent potency of the immunized HA protein, and the 50 μg or more per dose HA protein could provide complete protection against the H9N2 virus challenge. (4) Conclusions: These results indicate that the CHO expression system could be a platform used to develop the subunit vaccine against H9 influenza viruses in chickens.
Collapse
Affiliation(s)
- Shunfan Zhu
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (S.Z.); (Z.N.); (J.S.); (Y.H.)
| | - Zhenyu Nie
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (S.Z.); (Z.N.); (J.S.); (Y.H.)
| | - Ying Che
- Zhejiang Novo Biotech Co., Ltd., Shaoxing 312366, China; (Y.C.); (S.W.); (Y.W.); (H.Q.)
| | - Jianhong Shu
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (S.Z.); (Z.N.); (J.S.); (Y.H.)
| | - Sufang Wu
- Zhejiang Novo Biotech Co., Ltd., Shaoxing 312366, China; (Y.C.); (S.W.); (Y.W.); (H.Q.)
| | - Yulong He
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (S.Z.); (Z.N.); (J.S.); (Y.H.)
| | - Youqiang Wu
- Zhejiang Novo Biotech Co., Ltd., Shaoxing 312366, China; (Y.C.); (S.W.); (Y.W.); (H.Q.)
| | - Hong Qian
- Zhejiang Novo Biotech Co., Ltd., Shaoxing 312366, China; (Y.C.); (S.W.); (Y.W.); (H.Q.)
| | - Huapeng Feng
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (S.Z.); (Z.N.); (J.S.); (Y.H.)
| | - Qiang Zhang
- Zhejiang Novo Biotech Co., Ltd., Shaoxing 312366, China; (Y.C.); (S.W.); (Y.W.); (H.Q.)
| |
Collapse
|
27
|
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
|
28
|
Tomljenovic L, McHenry LB. A reactogenic "placebo" and the ethics of informed consent in Gardasil HPV vaccine clinical trials: A case study from Denmark. INTERNATIONAL JOURNAL OF RISK & SAFETY IN MEDICINE 2024; 35:159-180. [PMID: 38788092 PMCID: PMC11191454 DOI: 10.3233/jrs-230032] [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: 06/07/2023] [Accepted: 03/18/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Medical ethics guidelines require of clinical trial investigators and sponsors to inform prospective trial participants of all known and potential risks associated with investigational medical products, and to obtain their free informed consent. These guidelines also require that clinical research be so designed as to minimize harms and maximize benefits. OBJECTIVE To examine Merck's scientific rationale for using a reactogenic aluminum-containing "placebo" in Gardasil HPV vaccine pre-licensure clinical trials. METHODS We examined the informed consent form and the recruitment brochure for the FUTURE II Gardasil vaccine trial conducted in Denmark; and we interviewed several FUTURE II trial participants and their treating physicians. We also reviewed regulatory documentation related to Gardasil vaccine approval process and the guidelines on evaluation of adjuvants used in human vaccines. RESULTS It was found that the vaccine manufacturer Merck made several inaccurate statements to trial participants that compromised their right to informed consent. First, even though the study protocol listed safety testing as one of the study's primary objectives, the recruitment brochure emphasized that FUTURE II was not a safety study, and that the vaccine had already been proven safe. Second, the advertising material for the trial and the informed consent forms stated that the placebo was saline or an inactive substance, when, in fact, it contained Merck's proprietary highly reactogenic aluminum adjuvant which does not appear to have been properly evaluated for safety. Several trial participants experienced chronic disabling symptoms, including some randomized to the adjuvant "placebo" group. CONCLUSION In our view, the administration of a reactive placebo in Gardasil clinical trials was without any possible benefit, needlessly exposed study subjects to risks, and was therefore a violation of medical ethics. The routine use of aluminum adjuvants as "placebos" in vaccine clinical trials is inappropriate as it hinders the discovery of vaccine-related safety signals.
Collapse
Affiliation(s)
| | - Leemon B. McHenry
- Department of Philosophy, California State University, Northridge, CA, USA
| |
Collapse
|
29
|
Kara A, Coskun A, Temel F, Özelci P, Topal S, Ateş I. Analysis of participant-reported adverse events following the first dose of inactivated SARS-Cov-2 vaccine (TURKOVAC™) through telephone survey in Türkiye. Ann Med 2023; 55:1070-1079. [PMID: 36908270 PMCID: PMC10795555 DOI: 10.1080/07853890.2023.2183985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 02/17/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND/OBJECTIVE(S)/INTRODUCTION TURKOVAC™ is a whole-virion inactivated COVID-19 vaccine, which was developed and recently granted emergency use authorization (conditional marketing authorization) in Türkiye. The objective of this study is to assess the spectrum and the distribution of adverse events reported following the administration of the first 150,000 doses as primary and booster vaccine doses in 22 state hospitals of 17 provinces in Türkiye. PATIENTS/MATERIALS AND METHODS In this cohort study, a verbal survey was conducted via telephone calls between 10 January and 17 January 2022, utilizing a structured questionnaire algorithm on a sample group of 20,000 persons on the third- and seventh-days following vaccination. The algorithm consisted of two parts focusing on both systemic and local adverse effects. Other adverse events reported by the participants were also recorded. 6023 people and 5345 people agreed to participate in the telephone survey on the 3rd- and 7th- days of having received the first dose of the vaccine, respectively. RESULTS Thirty-six-point-six percent of the participants on the 3rd day and 22.5% of the participants on the 7th day reported any adverse event following the first dose of the vaccine. On both follow-up days, the most commonly reported (29.7% for Day 3 and 13.1% for Day 7) adverse events were on the injection site. Among the local adverse events, the most frequently reported one was the pain on the injection site (27.9% for Day 3 and 12.4% for Day 7), induration (4.8% for Day 3 and 2.7% for Day 7) and swelling (3.5% for Day 3 and 2.0% for Day 7). Fatigue/weakness (9.6% for Day 3 and 8.3% for Day 7) and headache (7.9% for Day 3 and 8.0% for Day 7) were the most frequent systemic adverse events. Younger age, vaccine dose, and female sex were associated with having any adverse event and pain (on the injection site). Female sex was associated with more swelling (on the injection site), induration (on the injection site), fever, and a higher impact on daily living. CONCLUSION(S) In this study, we conducted a rapid assessment of adverse events following the first dose of the TURKOVAC vaccine. The vaccine appears to have a good safety profile in the first 7 days following vaccination. Younger age, vaccine dose, and female sex are associated with any adverse event and pain (on the injection site). These results present valuable information for the community and may contribute to increasing vaccine confidence.KEY MESSAGESAs a whole-virion inactivated SARS-CoV-2 vaccine, the TURKOVAC™ vaccine, which has a favorable safety profile, can be an alternative to other COVID-19 vaccines including mRNA and viral vector vaccines.
Collapse
Affiliation(s)
- Ateş Kara
- Pediatric Infectious Disease Unit, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Aslihan Coskun
- Health Institutes of Türkiye, Turkish Vaccine Institute, Ankara, Turkey
| | - Fehminaz Temel
- Field Epidemiology Unit, General Directorate of Public Health, Department of Communicable Diseases and Early Warning, Ministry of Health, Ankara, Turkey
| | - Pervin Özelci
- Health Institutes of Türkiye, Turkish Vaccine Institute, Ankara, Turkey
| | - Selmur Topal
- Field Epidemiology Unit, General Directorate of Public Health, Department of Communicable Diseases and Early Warning, Ministry of Health, Ankara, Turkey
| | - Ihsan Ateş
- Ankara City Hospital, Internal Medicine Clinic, Ankara, Turkey
| |
Collapse
|
30
|
Hidalgo-Gajardo A, Gutiérrez N, Lamazares E, Espinoza F, Escobar-Riquelme F, Leiva MJ, Villavicencio C, Mena-Ulecia K, Montesino R, Altamirano C, Sánchez O, Rivas CI, Ruíz Á, Toledo JR. Co-Formulation of Recombinant Porcine IL-18 Enhances the Onset of Immune Response in a New Lawsonia intracellularis Vaccine. Vaccines (Basel) 2023; 11:1788. [PMID: 38140192 PMCID: PMC10747595 DOI: 10.3390/vaccines11121788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Pig is one of the most consumed meats worldwide. One of the main conditions for pig production is Porcine Enteropathy caused by Lawsonia intracellularis. Among the effects of this disease is chronic mild diarrhea, which affects the weight gain of pigs, generating economic losses. Vaccines available to prevent this condition do not have the desired effect, but this limitation can be overcome using adjuvants. Pro-inflammatory cytokines, such as interleukin 18 (IL-18), can improve an immune response, reducing the immune window of protection. In this study, recombinant porcine IL-18 was produced and expressed in Escherichia coli and Pichia pastoris. The protein's biological activity was assessed in vitro and in vivo, and we determined that the P. pastoris protein had better immunostimulatory activity. A vaccine candidate against L. intracellularis, formulated with and without IL-18, was used to determine the pigs' cellular and humoral immune responses. Animals injected with the candidate vaccine co-formulated with IL-18 showed a significant increase of Th1 immune response markers and an earlier increase of antibodies than those vaccinated without the cytokine. This suggests that IL-18 acts as an immunostimulant and vaccine adjuvant to boost the immune response against the antigens, reducing the therapeutic window of recombinant protein-based vaccines.
Collapse
Affiliation(s)
- Angela Hidalgo-Gajardo
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
- Centro de Desarrollo e Innovación Biovacuvet SpA, VIII Región, Concepción 4090838, Chile
| | - Nicolás Gutiérrez
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
- Centro de Desarrollo e Innovación Biovacuvet SpA, VIII Región, Concepción 4090838, Chile
| | - Emilio Lamazares
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
| | - Felipe Espinoza
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
- Centro de Desarrollo e Innovación Biovacuvet SpA, VIII Región, Concepción 4090838, Chile
| | - Fernanda Escobar-Riquelme
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
| | - María J. Leiva
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
| | - Carla Villavicencio
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
| | - Karel Mena-Ulecia
- Departamento de Ciencias Biológicas y Químicas, Facultad de Recursos Naturales, Universidad Católica de Temuco, IX Región, Temuco 4813302, Chile;
| | - Raquel Montesino
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
| | - Claudia Altamirano
- Laboratorio de Cultivos Celulares, Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, V Región, Valparaíso 2362803, Chile;
| | - Oliberto Sánchez
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
| | - Coralia I. Rivas
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
| | - Álvaro Ruíz
- Departamento de Patología y Medicina Preventiva, Facultad de Ciencias Veterinarias, Universidad de Concepción, XVI Región, Chillán 3812120, Chile;
| | - Jorge R. Toledo
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, VIII Región, Concepción 4070386, Chile; (A.H.-G.); (M.J.L.); (C.V.); (C.I.R.)
| |
Collapse
|
31
|
Moni SS, Abdelwahab SI, Jabeen A, Elmobark ME, Aqaili D, Ghoal G, Oraibi B, Farasani AM, Jerah AA, Alnajai MMA, Mohammad Alowayni AMH. Advancements in Vaccine Adjuvants: The Journey from Alum to Nano Formulations. Vaccines (Basel) 2023; 11:1704. [PMID: 38006036 PMCID: PMC10674458 DOI: 10.3390/vaccines11111704] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Vaccination is a groundbreaking approach in preventing and controlling infectious diseases. However, the effectiveness of vaccines can be greatly enhanced by the inclusion of adjuvants, which are substances that potentiate and modulate the immune response. This review is based on extensive searches in reputable databases such as Web of Science, PubMed, EMBASE, Scopus, and Google Scholar. The goal of this review is to provide a thorough analysis of the advances in the field of adjuvant research, to trace the evolution, and to understand the effects of the various adjuvants. Historically, alum was the pioneer in the field of adjuvants because it was the first to be approved for use in humans. It served as the foundation for subsequent research and innovation in the field. As science progressed, research shifted to identifying and exploiting the potential of newer adjuvants. One important area of interest is nano formulations. These advanced adjuvants have special properties that can be tailored to enhance the immune response to vaccines. The transition from traditional alum-based adjuvants to nano formulations is indicative of the dynamism and potential of vaccine research. Innovations in adjuvant research, particularly the development of nano formulations, are a promising step toward improving vaccine efficacy and safety. These advances have the potential to redefine the boundaries of vaccination and potentially expand the range of diseases that can be addressed with this approach. There is an optimistic view of the future in which improved vaccine formulations will contribute significantly to improving global health outcomes.
Collapse
Affiliation(s)
- Sivakumar S. Moni
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.J.)
| | | | - Aamena Jabeen
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.J.)
| | - Mohamed Eltaib Elmobark
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.J.)
| | - Duaa Aqaili
- Physiology Department, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia
| | - Gassem Ghoal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia
| | - Bassem Oraibi
- Medical Research Centre, Jazan University, Jazan 45142, Saudi Arabia (B.O.)
| | | | - Ahmed Ali Jerah
- College of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Mahdi Mohammed A. Alnajai
- General Directorate of Health Services and University Hospital, Jazan University, Jazan 45142, Saudi Arabia;
| | | |
Collapse
|
32
|
Mosadegh S, Abtahi H, Amani J, Karizi SZ, Salmanian AH. Protective immunization against Enterohemorrhagic Escherichia coli and Shigella dysenteriae Type 1 by chitosan nanoparticle loaded with recombinant chimeric antigens comprising EIT and STX1B-IpaD. Microb Pathog 2023; 184:106344. [PMID: 37704060 DOI: 10.1016/j.micpath.2023.106344] [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: 07/05/2023] [Revised: 09/09/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Increasing evidence demonstrated that Enterohemorrhagic Escherichia coli (EHEC) and Shigella dysenteriae type 1 (S. dysenteriae1) are considered pathogens, that are connected with diarrhea and are still the greatest cause of death in children under the age of five years, worldwide. EHEC and S. dysenteriae 1 infections can be prevented and managed using a vaccination strategy against pathogen attachment stages. In this study, the chitosan nanostructures were loaded with recombinant EIT and STX1B-IpaD polypeptides. The immunogenic properties of this nano-vaccine candidate were investigated. The EIT and STX1B-IpaD recombinant proteins were heterologous expressed, purified, and confirmed by western blotting. The chitosan nanoparticles, were used to encapsulate the purified proteins. The immunogenicity of recombinant nano vaccine candidate, was examined in three groups of BalB/c mice by injection, oral delivery, and combination of oral-injection. ELISA and antibody titer, evaluated the humoral immune response. Finally, all three mice groups were challenged by two pathogens to test the ability of the nano-vaccine candidate to protect against bacterial infection. The Sereny test in guinea pigs was used to confirm the neutralizing effect of immune sera in controlling S. dysenteriae 1, infections. SDS-PAGE and western blotting, confirmed the presence and specificity of 63 and 27 kDa recombinant EIT and STX1B-IpaD, respectively. The results show that the nanoparticles containing recombinant proteins could stimulate the systemic and mucosal immune systems by producing IgG and IgA, respectively. The challenge test showed that, the candidate nano-vaccine could protect the animal model from bacterial infection. The combination of multiple recombinant proteins, carrying several epitopes and natural nanoparticles could evocate remarkable humoral and mucosal responses and improve the protection properties of synthetic antigens. Furthermore, compared with other available antigen delivery methods, using oral delivery as immune priming and injection as a booster method, could act as combinatorial methods to achieve a higher level of immunity. This approach could present an appropriate vaccine candidate against both EHEC and S. dysenteriae 1.
Collapse
Affiliation(s)
- Shadi Mosadegh
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hamid Abtahi
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shohreh Zare Karizi
- Department of Biology, Varamin Pishva Branch, Islamic Azad University, Pishva, Varamin, Iran
| | - Ali Hatef Salmanian
- Department of Agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| |
Collapse
|
33
|
Si Y, Wang Y, Tian Q, Wang Q, Pollard JM, Srivastava PK, Esser-Kahn AP, Collier JH, Sperling AI, Chong AS. Lung cDC1 and cDC2 dendritic cells priming naive CD8 + T cells in situ prior to migration to draining lymph nodes. Cell Rep 2023; 42:113299. [PMID: 37864794 PMCID: PMC10676754 DOI: 10.1016/j.celrep.2023.113299] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/21/2023] [Accepted: 10/02/2023] [Indexed: 10/23/2023] Open
Abstract
The current paradigm indicates that naive T cells are primed in secondary lymphoid organs. Here, we present evidence that intranasal administration of peptide antigens appended to nanofibers primes naive CD8+ T cells in the lung independently and prior to priming in the draining mediastinal lymph node (MLN). Notably, comparable accumulation and transcriptomic responses of CD8+ T cells in lung and MLN are observed in both Batf3KO and wild-type (WT) mice, indicating that, while cDC1 dendritic cells (DCs) are the major subset for cross-presentation, cDC2 DCs alone are capable of cross-priming CD8+ T cells both in the lung and draining MLN. Transcription analyses reveal distinct transcriptional responses in lung cDC1 and cDC2 to intranasal nanofiber immunization. However, both DC subsets acquire shared transcriptional responses upon migration into the lymph node, thus uncovering a stepwise activation process of cDC1 and cDC2 toward their ability to cross-prime effector and functional memory CD8+ T cell responses.
Collapse
Affiliation(s)
- Youhui Si
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Department of Surgery, The University of Chicago, Chicago, IL 60637, USA.
| | - Yihan Wang
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiaomu Tian
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiang Wang
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Jared M Pollard
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Pramod K Srivastava
- Department of Immunology and Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Aaron P Esser-Kahn
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Anne I Sperling
- Department of Medicine, Pulmonary and Critical Care, University of Virginia, Charlottesville, VA 22908, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
34
|
Liu X, Liu Y, Yang X, Lu X, Xu XN, Zhang J, Chen R. Potentiating the Immune Responses of HBsAg-VLP Vaccine Using a Polyphosphoester-Based Cationic Polymer Adjuvant. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48871-48881. [PMID: 37816068 PMCID: PMC10614196 DOI: 10.1021/acsami.3c07491] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023]
Abstract
Virus-like particle (VLP)-based vaccines are required to be associated with a suitable adjuvant to potentiate their immune responses. Herein, we report a novel, biodegradable, and biocompatible polyphosphoester-based amphiphilic cationic polymer, poly(ethylene glycol)-b-poly(aminoethyl ethylene phosphate) (PEG-PAEEP), as a Hepatitis B surface antigen (HBsAg)-VLP vaccine adjuvant. The polymer adjuvant effectively bound with HBsAg-VLP through electrostatic interactions to form a stable vaccine nanoformulation with a net positive surface charge. The nanoformulations exhibited enhanced cellular uptake by macrophages. HBsAg-VLP/PEG-PAEEP induced a significantly higher HBsAg-specific IgG titer in mice than HBsAg-VLP alone after second immunization, indicative of the antigen-dose sparing advantage of PEG-PAEEP. Furthermore, the nanoformulations exhibited a favorable biocompatibility and in vivo tolerability. This work presents the PEG-PAEEP copolymer as a promising vaccine adjuvant and as a potentially effective alternative to aluminum adjuvants.
Collapse
Affiliation(s)
- Xuhan Liu
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
- Department
of Emergency Medicine, Shenzhen University
General Hospital, Shenzhen University, Shenzhen 518051, China
| | - Yifan Liu
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
| | - Xiaoyu Yang
- AIM
Honesty Biopharmaceutical Co., Ltd, Dalian 116620, China
| | - Xinyu Lu
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
| | - Xiao-Ning Xu
- Department
of Infectious Diseases, Imperial College
London, London W12 0NN, U.K.
| | - Jiancheng Zhang
- AIM
Honesty Biopharmaceutical Co., Ltd, Dalian 116620, China
| | - Rongjun Chen
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
| |
Collapse
|
35
|
Zahedipour F, Jamialahmadi K, Zamani P, Reza Jaafari M. Improving the efficacy of peptide vaccines in cancer immunotherapy. Int Immunopharmacol 2023; 123:110721. [PMID: 37543011 DOI: 10.1016/j.intimp.2023.110721] [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: 05/24/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Peptide vaccines have shown great potential in cancer immunotherapy by targeting tumor antigens and activating the patient's immune system to mount a specific response against cancer cells. However, the efficacy of peptide vaccines in inducing a sustained immune response and achieving clinical benefit remains a major challenge. In this review, we discuss the current status of peptide vaccines in cancer immunotherapy and strategies to improve their efficacy. We summarize the recent advancements in the development of peptide vaccines in pre-clinical and clinical settings, including the use of novel adjuvants, neoantigens, nano-delivery systems, and combination therapies. We also highlight the importance of personalized cancer vaccines, which consider the unique genetic and immunological profiles of individual patients. We also discuss the strategies to enhance the immunogenicity of peptide vaccines such as multivalent peptides, conjugated peptides, fusion proteins, and self-assembled peptides. Although, peptide vaccines alone are weak immunogens, combining peptide vaccines with other immunotherapeutic approaches and developing novel approaches such as personalized vaccines can be promising methods to significantly enhance their efficacy and improve the clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Troese MJ, Burlet E, Cunningham MW, Alvarez K, Bentley R, Thomas N, Carwell S, Morefield GL. Group A Streptococcus Vaccine Targeting the Erythrogenic Toxins SpeA and SpeB Is Safe and Immunogenic in Rabbits and Does Not Induce Antibodies Associated with Autoimmunity. Vaccines (Basel) 2023; 11:1504. [PMID: 37766180 PMCID: PMC10534881 DOI: 10.3390/vaccines11091504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Group A streptococcus (GAS) is a global pathogen associated with significant morbidity and mortality for which there is currently no licensed vaccine. Vaccine development has been slow, mostly due to safety concerns regarding streptococcal antigens associated with autoimmunity and related complications. For a GAS vaccine to be safe, it must be ensured that the antigens used in the vaccine do not elicit an antibody response that can cross-react with host tissues. In this study, we evaluated the safety of our GAS vaccine candidate called VaxiStrep in New Zealand White rabbits. VaxiStrep is a recombinant fusion protein comprised of streptococcal pyrogenic exotoxin A (SpeA) and exotoxin B (SpeB), also known as erythrogenic toxins, adsorbed to an aluminum adjuvant. The vaccine elicited a robust immune response against the two toxins in the rabbits without any adverse events or toxicity. No signs of autoimmune pathology were detected in the rabbits' brains, hearts, and kidneys via immunohistochemistry, and serum antibodies did not cross-react with cardiac or neuronal tissue proteins associated with rheumatic heart disease or Sydenham chorea (SC). This study further confirms that VaxiStrep does not elicit autoantibodies and is safe to be tested in a first-in-human trial.
Collapse
Affiliation(s)
| | | | - Madeleine W. Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kathy Alvarez
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Rebecca Bentley
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
37
|
Madge HYR, Alexander S, Azuar A, Zhang J, Koirala P, Burne TH, Toth I, Stephenson RJ. Synthetic Anti-Cocaine Nanoaccine Successfully Prevents Cocaine-Induced Hyperlocomotion. J Med Chem 2023; 66:12407-12419. [PMID: 37646732 DOI: 10.1021/acs.jmedchem.3c00889] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Cocaine is one of the most widely used and increasingly popular illicit psychoactive drugs. Unlike other commonly used substances of abuse, cocaine has no pharmacological therapies to treat addiction or aid in rehabilitation. Immunopharmacology has long been touted as a possible avenue to develop effective anticocaine therapies; however, lack of efficacy and designs which are not consistent with simple large-scale production have hindered vaccine translation. We have designed and synthesized a peptide-based anti-cocaine immunogen which we have shown is capable of inducing physiologically relevant immune responses in mice as part of a self-adjuvanting delivery system or in combination with the human-approved commercial adjuvant MF59. We have demonstrated that immunization with the reported vaccine elicits high titers of anti-cocaine IgG and prevents cocaine-induced hyperlocomotion in an in vivo murine model. This peptide-hapten immunogen along with self-adjuvanting liposomal-based delivery system provides a platform for the development of effective anti-drug vaccines.
Collapse
Affiliation(s)
- Harrison Y R Madge
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Suzy Alexander
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia
- Queensland Centre for Mental Health Research, Wacol, Queensland, 4076, Australia
| | - Armira Azuar
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Jiahui Zhang
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Prashamsa Koirala
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Thomas H Burne
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia
- Queensland Centre for Mental Health Research, Wacol, Queensland, 4076, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- School of Pharmacy, The University of Queensland, Brisbane 4072, Australia
| | - Rachel J Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| |
Collapse
|
38
|
Pérez-Baños A, Gleisner MA, Flores I, Pereda C, Navarrete M, Araya JP, Navarro G, Quezada-Monrás C, Tittarelli A, Salazar-Onfray F. Whole tumour cell-based vaccines: tuning the instruments to orchestrate an optimal antitumour immune response. Br J Cancer 2023; 129:572-585. [PMID: 37355722 PMCID: PMC10421921 DOI: 10.1038/s41416-023-02327-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023] Open
Abstract
Immunotherapy, particularly those based on immune checkpoint inhibitors (ICIs), has become a useful approach for many neoplastic diseases. Despite the improvements of ICIs in supporting tumour regression and prolonging survival, many patients do not respond or develop resistance to treatment. Thus, therapies that enhance antitumour immunity, such as anticancer vaccines, constitute a feasible and promising therapeutic strategy. Whole tumour cell (WTC) vaccines have been extensively tested in clinical studies as intact or genetically modified cells or tumour lysates, injected directly or loaded on DCs with distinct adjuvants. The essential requirements of WTC vaccines include the optimal delivery of a broad battery of tumour-associated antigens, the presence of tumour cell-derived molecular danger signals, and adequate adjuvants. These factors trigger an early and robust local innate inflammatory response that orchestrates an antigen-specific and proinflammatory adaptive antitumour response capable of controlling tumour growth by several mechanisms. In this review, the strengths and weaknesses of our own and others' experiences in studying WTC vaccines are revised to discuss the essential elements required to increase anticancer vaccine effectiveness.
Collapse
Affiliation(s)
- Amarilis Pérez-Baños
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Iván Flores
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cristián Pereda
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mariela Navarrete
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Juan Pablo Araya
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Giovanna Navarro
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, 5110566, Chile
| | - Claudia Quezada-Monrás
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, 5110566, Chile
| | - Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación (PIDi), Universidad Tecnológica Metropolitana (UTEM), Santiago, Chile.
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute and Section for Infectious Diseases, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
39
|
Shichinohe S, Watanabe T. Advances in Adjuvanted Influenza Vaccines. Vaccines (Basel) 2023; 11:1391. [PMID: 37631959 PMCID: PMC10459454 DOI: 10.3390/vaccines11081391] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/29/2023] [Accepted: 08/18/2023] [Indexed: 08/29/2023] Open
Abstract
The numerous influenza infections that occur every year present a major public health problem. Influenza vaccines are important for the prevention of the disease; however, their effectiveness against infection can be suboptimal. Particularly in the elderly, immune induction can be insufficient, and the vaccine efficacy against infection is usually lower than that in young adults. Vaccine efficacy can be improved by the addition of adjuvants, and an influenza vaccine with an oil-in-water adjuvant MF59, FLUAD, has been recently licensed in the United States and other countries for persons aged 65 years and older. Although the adverse effects of adjuvanted vaccines have been a concern, many adverse effects of currently approved adjuvanted influenza vaccines are mild and acceptable, given the overriding benefits of the vaccine. Since sufficient immunity can be induced with a small amount of vaccine antigen in the presence of an adjuvant, adjuvanted vaccines promote dose sparing and the prompt preparation of vaccines for pandemic influenza. Adjuvants not only enhance the immune response to antigens but can also be effective against antigenically different viruses. In this narrative review, we provide an overview of influenza vaccines, both past and present, before presenting a discussion of adjuvanted influenza vaccines and their future.
Collapse
Grants
- JP16H06429, JP16K21723, JP17H05809, JP16H06434, JP22H02521, JP22H02876 Japan Society for the Promotion of Science
- JP20jk0210021h0002, JP19fk0108113, JP223fa627002, JP22am0401030, JP23fk0108659, JP22gm1610010 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Shintaro Shichinohe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Tokiko Watanabe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease and Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
40
|
Ruzzi F, Semprini MS, Scalambra L, Palladini A, Angelicola S, Cappello C, Pittino OM, Nanni P, Lollini PL. Virus-like Particle (VLP) Vaccines for Cancer Immunotherapy. Int J Mol Sci 2023; 24:12963. [PMID: 37629147 PMCID: PMC10454695 DOI: 10.3390/ijms241612963] [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: 07/31/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer vaccines are increasingly being studied as a possible strategy to prevent and treat cancers. While several prophylactic vaccines for virus-caused cancers are approved and efficiently used worldwide, the development of therapeutic cancer vaccines needs to be further implemented. Virus-like particles (VLPs) are self-assembled protein structures that mimic native viruses or bacteriophages but lack the replicative material. VLP platforms are designed to display single or multiple antigens with a high-density pattern, which can trigger both cellular and humoral responses. The aim of this review is to provide a comprehensive overview of preventive VLP-based vaccines currently approved worldwide against HBV and HPV infections or under evaluation to prevent virus-caused cancers. Furthermore, preclinical and early clinical data on prophylactic and therapeutic VLP-based cancer vaccines were summarized with a focus on HER-2-positive breast cancer.
Collapse
Affiliation(s)
- Francesca Ruzzi
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Maria Sofia Semprini
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Laura Scalambra
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Arianna Palladini
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Stefania Angelicola
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Chiara Cappello
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Olga Maria Pittino
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Patrizia Nanni
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| | - Pier-Luigi Lollini
- Department of Medical and Surgical Sciences (DIMEC) and Alma Mater Institute on Healthy Planet, University of Bologna, 40126 Bologna, Italy; (F.R.); (M.S.S.); (L.S.); (S.A.); (C.C.); (O.M.P.); (P.N.)
| |
Collapse
|
41
|
Han S, Lee P, Choi HJ. Non-Invasive Vaccines: Challenges in Formulation and Vaccine Adjuvants. Pharmaceutics 2023; 15:2114. [PMID: 37631328 PMCID: PMC10458847 DOI: 10.3390/pharmaceutics15082114] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Given the limitations of conventional invasive vaccines, such as the requirement for a cold chain system and trained personnel, needle-based injuries, and limited immunogenicity, non-invasive vaccines have gained significant attention. Although numerous approaches for formulating and administrating non-invasive vaccines have emerged, each of them faces its own challenges associated with vaccine bioavailability, toxicity, and other issues. To overcome such limitations, researchers have created novel supplementary materials and delivery systems. The goal of this review article is to provide vaccine formulation researchers with the most up-to-date information on vaccine formulation and the immunological mechanisms available, to identify the technical challenges associated with the commercialization of non-invasive vaccines, and to guide future research and development efforts.
Collapse
Affiliation(s)
| | | | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada; (S.H.); (P.L.)
| |
Collapse
|
42
|
Desai DN, Mahal A, Varshney R, Obaidullah AJ, Gupta B, Mohanty P, Pattnaik P, Mohapatra NC, Mishra S, Kandi V, Rabaan AA, Mohapatra RK. Nanoadjuvants: Promising Bioinspired and Biomimetic Approaches in Vaccine Innovation. ACS OMEGA 2023; 8:27953-27968. [PMID: 37576639 PMCID: PMC10413842 DOI: 10.1021/acsomega.3c02030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023]
Abstract
Adjuvants are the important part of vaccine manufacturing as they elicit the vaccination effect and enhance the durability of the immune response through controlled release. In light of this, nanoadjuvants have shown unique broad spectrum advantages. As nanoparticles (NPs) based vaccines are fast-acting and better in terms of safety and usability parameters as compared to traditional vaccines, they have attracted the attention of researchers. A vaccine nanocarrier is another interesting and promising area for the development of next-generation vaccines for prophylaxis. This review looks at the various nanoadjuvants and their structure-function relationships. It compiles the state-of-art literature on numerous nanoadjuvants to help domain researchers orient their understanding and extend their endeavors in vaccines research and development.
Collapse
Affiliation(s)
- Dhruv N. Desai
- Department
of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ahmed Mahal
- Department
of Medical Biochemical Analysis, College of Health Technology, Cihan University−Erbil, Erbil, Kurdistan Region, Iraq
| | - Rajat Varshney
- Department
of Veterinary Microbiology, FVAS, Banaras
Hindu University, Mirzapur 231001, India
| | - Ahmad J. Obaidullah
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Bhawna Gupta
- School
of Biotechnology, KIIT Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | - Pratikhya Mohanty
- Bioenergy
Lab, BDTC, School of Biotechnology, KIIT
Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | | | | | - Snehasish Mishra
- Bioenergy
Lab, BDTC, School of Biotechnology, KIIT
Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | - Venkataramana Kandi
- Department
of Microbiology, Prathima Institute of Medical
Sciences, Karimnagar 505 417, Telangana, India
| | - Ali A. Rabaan
- Molecular
Diagnostic Laboratory, Johns Hopkins Aramco
Healthcare, Dhahran 31311, Saudi Arabia
- College
of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department
of Public Health and Nutrition, The University
of Haripur, Haripur 22610, Pakistan
| | - Ranjan K. Mohapatra
- Department
of Chemistry, Government College of Engineering, Keonjhar 758002, Odisha, India
| |
Collapse
|
43
|
Kamensek U, Cemazar M, Kranjc Brezar S, Jesenko T, Kos S, Znidar K, Markelc B, Modic Z, Komel T, Gorse T, Rebersek E, Jakopic H, Sersa G. What We Learned about the Feasibility of Gene Electrotransfer for Vaccination on a Model of COVID-19 Vaccine. Pharmaceutics 2023; 15:1981. [PMID: 37514166 PMCID: PMC10385748 DOI: 10.3390/pharmaceutics15071981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
DNA vaccination is one of the emerging approaches for a wide range of applications, including prophylactic vaccination against infectious diseases and therapeutic vaccination against cancer. The aim of this study was to evaluate the feasibility of our previously optimized protocols for gene electrotransfer (GET)-mediated delivery of plasmid DNA into skin and muscle tissues on a model of COVID-19 vaccine. Plasmids encoding the SARS-CoV-2 proteins spike (S) and nucleocapsid (N) were used as the antigen source, and a plasmid encoding interleukin 12 (IL-12) was used as an adjuvant. Vaccination was performed in the skin or muscle tissue of C57BL/6J mice on days 0 and 14 (boost). Two weeks after the boost, blood, spleen, and transfected tissues were collected to determine the expression of S, N, IL-12, serum interferon-γ, the induction of antigen-specific IgG antibodies, and cytotoxic T-cells. In accordance with prior in vitro experiments that indicated problems with proper expression of the S protein, vaccination with S did not induce S-specific antibodies, whereas significant induction of N-specific antibodies was detected after vaccination with N. Intramuscular vaccination outperformed skin vaccination and resulted in significant induction of humoral and cell-mediated immunity. Moreover, both boost and adjuvant were found to be redundant for the induction of an immune response. Overall, the study confirmed the feasibility of the GET for DNA vaccination and provided valuable insights into this approach.
Collapse
Affiliation(s)
- Urska Kamensek
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, SI-1000 Ljubljana, Slovenia
| | - Maja Cemazar
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310 Izola, Slovenia
| | | | - Tanja Jesenko
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia
| | - Spela Kos
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
| | - Katarina Znidar
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
| | - Bostjan Markelc
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena Pot 5, SI-1000 Ljubljana, Slovenia
| | - Ziva Modic
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia
| | - Tilen Komel
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia
| | - Tim Gorse
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, SI-1000 Ljubljana, Slovenia
| | - Eva Rebersek
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, SI-1000 Ljubljana, Slovenia
| | - Helena Jakopic
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, SI-1000 Ljubljana, Slovenia
| | - Gregor Sersa
- Institute of Oncology Ljubljana, Zaloska Cesta 2, SI-1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena Pot 5, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
44
|
Lin HH, Wang CY, Hsieh FJ, Liao FZ, Su YK, Pham MD, Lee CY, Chang HC, Hsu HH. Nanodiamonds-in-oil emulsions elicit potent immune responses for effective vaccination and therapeutics. Nanomedicine (Lond) 2023; 18:1045-1059. [PMID: 37610004 DOI: 10.2217/nnm-2023-0179] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
Background: The use of nanodiamonds (NDs) and fluorescent nanodiamonds (FNDs) as nonallergenic biocompatible additives in incomplete Freund's adjuvant (IFA) to elicit immune responses in vivo was investigated. Methods: C57BL/6 mice were immunized with chicken egg ovalbumin (OVA) in IFA and also OVA-conjugated NDs (or OVA-conjugated FNDs) in IFA to produce antibodies. OVA-expressing E.G7 lymphoma cells and OVA-negative EL4 cells were inoculated in mice to induce tumor formation. Results: The new formulation significantly enhanced immune responses and thus disease resistance. It exhibited specific therapeutic activities, effectively inhibiting the growth of E.G7 tumor cells in mice over 35 days. Conclusion: The high biocompatibility and multiple functionalities of NDs/FNDs render them applicable as active and trackable vaccine adjuvants and antitumor agents.
Collapse
Affiliation(s)
- Hsin-Hung Lin
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Chih-Yen Wang
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Feng-Jen Hsieh
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Fang-Zhen Liao
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Yu-Kai Su
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
| | - Minh Dinh Pham
- Institute of Biotechnology, Vietnam Academy of Science & Technology, Ha Noi 100000, Vietnam
| | - Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital & College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Huan-Cheng Chang
- Institute of Atomic & Molecular Sciences, Academia Sinica, Taipei 106, Taiwan
- Department of Chemical Engineering, National Taiwan University of Science & Technology, Taipei City 106, Taiwan
- Department of Chemistry, National Taiwan Normal University, Taipei City 106, Taiwan
| | - Hsao-Hsun Hsu
- Department of Surgery, National Taiwan University Hospital & College of Medicine, National Taiwan University, Taipei 100, Taiwan
- National Taiwan University Cancer Center, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
45
|
Kim JY, Rosenberger MG, Rutledge NS, Esser-Kahn AP. Next-Generation Adjuvants: Applying Engineering Methods to Create and Evaluate Novel Immunological Responses. Pharmaceutics 2023; 15:1687. [PMID: 37376133 PMCID: PMC10300703 DOI: 10.3390/pharmaceutics15061687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Adjuvants are a critical component of vaccines. Adjuvants typically target receptors that activate innate immune signaling pathways. Historically, adjuvant development has been laborious and slow, but has begun to accelerate over the past decade. Current adjuvant development consists of screening for an activating molecule, formulating lead molecules with an antigen, and testing this combination in an animal model. There are very few adjuvants approved for use in vaccines, however, as new candidates often fail due to poor clinical efficacy, intolerable side effects, or formulation limitations. Here, we consider new approaches using tools from engineering to improve next-generation adjuvant discovery and development. These approaches will create new immunological outcomes that will be evaluated with novel diagnostic tools. Potential improved immunological outcomes include reduced vaccine reactogenicity, tunable adaptive responses, and enhanced adjuvant delivery. Evaluations of these outcomes can leverage computational approaches to interpret "big data" obtained from experimentation. Applying engineering concepts and solutions will provide alternative perspectives, further accelerating the field of adjuvant discovery.
Collapse
Affiliation(s)
| | | | | | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA; (J.Y.K.); (M.G.R.); (N.S.R.)
| |
Collapse
|
46
|
Wu N, Chen Q, Zou Y, Miao C, Ma G, Wu J. Chitosan particle-emulsion complex adjuvants: The effect of particle distribution on the immune intensity and response type. Carbohydr Polym 2023; 309:120673. [PMID: 36906359 DOI: 10.1016/j.carbpol.2023.120673] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Particle-emulsion complex adjuvants as a new trend in the research of vaccine formulation, can improve the immune strength and balance the immune type. However, the location of the particle in the formulation is a key factor that has not been investigated extensively and its type of immunity. In order to investigate the effect of different combining modes of emulsion and particle on the immune response, three types of particle-emulsion complex adjuvant formulations were designed with the combination of chitosan nanoparticles (CNP) and an o/w emulsion with squalene as the oil phase. The complex adjuvants included the CNP-I group (particle inside the emulsion droplet), CNP-S group (particle on the surface of emulsion droplet) and CNP-O group (particle outside the emulsion droplet), respectively. The formulations with different particle locations behaved with different immunoprotective effects and immune-enhancing mechanisms. Compared with CNP-O, CNP-I and CNP-S significantly improve humoral and cellular immunity. CNP-O was more like two independent systems for immune enhancement. As a result, CNP-S triggered a Th1-type immune bias and CNP-I had more of a Th2-type of the immune response. These data highlight the key influence of the subtle difference of particle location in the droplets for immune response.
Collapse
Affiliation(s)
- Nan Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Qiuting Chen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Yongjuan Zou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Chunyu Miao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
47
|
Brai A, Poggialini F, Pasqualini C, Trivisani CI, Vagaggini C, Dreassi E. Progress towards Adjuvant Development: Focus on Antiviral Therapy. Int J Mol Sci 2023; 24:9225. [PMID: 37298177 PMCID: PMC10253057 DOI: 10.3390/ijms24119225] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
In recent decades, vaccines have been extraordinary resources to prevent pathogen diffusion and cancer. Even if they can be formed by a single antigen, the addition of one or more adjuvants represents the key to enhance the response of the immune signal to the antigen, thus accelerating and increasing the duration and the potency of the protective effect. Their use is of particular importance for vulnerable populations, such as the elderly or immunocompromised people. Despite their importance, only in the last forty years has the search for novel adjuvants increased, with the discovery of novel classes of immune potentiators and immunomodulators. Due to the complexity of the cascades involved in immune signal activation, their mechanism of action remains poorly understood, even if significant discovery has been recently made thanks to recombinant technology and metabolomics. This review focuses on the classes of adjuvants under research, recent mechanism of action studies, as well as nanodelivery systems and novel classes of adjuvants that can be chemically manipulated to create novel small molecule adjuvants.
Collapse
Affiliation(s)
- Annalaura Brai
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Federica Poggialini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Pasqualini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Immacolata Trivisani
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Chiara Vagaggini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Elena Dreassi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| |
Collapse
|
48
|
Van Rampelbergh J, Achenbach P, Leslie RD, Ali MA, Dayan C, Keymeulen B, Owen KR, Kindermans M, Parmentier F, Carlier V, Ahangarani RR, Gebruers E, Bovy N, Vanderelst L, Van Mechelen M, Vandepapelière P, Boitard C. First-in-human, double-blind, randomized phase 1b study of peptide immunotherapy IMCY-0098 in new-onset type 1 diabetes. BMC Med 2023; 21:190. [PMID: 37226224 DOI: 10.1186/s12916-023-02900-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a CD4+ T cell-driven autoimmune disease characterized by the destruction of insulin-producing pancreatic β-cells by CD8+ T cells. Achieving glycemic targets in T1D remains challenging in clinical practice; new treatments aim to halt autoimmunity and prolong β-cell survival. IMCY-0098 is a peptide derived from human proinsulin that contains a thiol-disulfide oxidoreductase motif at the N-terminus and was developed to halt disease progression by promoting the specific elimination of pathogenic T cells. METHODS This first-in-human, 24-week, double-blind phase 1b study evaluated the safety of three dosages of IMCY-0098 in adults diagnosed with T1D < 6 months before study start. Forty-one participants were randomized to receive four bi-weekly injections of placebo or increasing doses of IMCY-0098 (dose groups A/B/C received 50/150/450 μg for priming followed by three further administrations of 25/75/225 μg, respectively). Multiple T1D-related clinical parameters were also assessed to monitor disease progression and inform future development. Long-term follow-up to 48 weeks was also conducted in a subset of patients. RESULTS Treatment with IMCY-0098 was well tolerated with no systemic reactions; a total of 315 adverse events (AEs) were reported in 40 patients (97.6%) and were related to study treatment in 29 patients (68.3%). AEs were generally mild; no AE led to discontinuation of the study or death. No significant decline in C-peptide was noted from baseline to Week 24 for dose A, B, C, or placebo (mean change - 0.108, - 0.041, - 0.040, and - 0.012, respectively), suggesting no disease progression. CONCLUSIONS Promising safety profile and preliminary clinical response data support the design of a phase 2 study of IMCY-0098 in patients with recent-onset T1D. TRIAL REGISTRATION IMCY-T1D-001: ClinicalTrials.gov NCT03272269; EudraCT: 2016-003514-27; and IMCY-T1D-002: ClinicalTrials.gov NCT04190693; EudraCT: 2018-003728-35.
Collapse
Affiliation(s)
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | | | - Mohammad Alhadj Ali
- Diabetes Research Group, Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | - Colin Dayan
- Diabetes Research Group, Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | - Bart Keymeulen
- Member of Belgian Diabetes Registry, Academic Hospital and Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katharine R Owen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | | | | | - Vincent Carlier
- Imcyse S.A., Avenue Pré-Aily 14, Angleur, 4031, Liège, Belgium
| | | | | | - Nicolas Bovy
- Imcyse S.A., Avenue Pré-Aily 14, Angleur, 4031, Liège, Belgium
| | - Luc Vanderelst
- Imcyse S.A., Avenue Pré-Aily 14, Angleur, 4031, Liège, Belgium
| | | | | | - Christian Boitard
- Inserm U1016, Cochin Institute, Paris, France
- Medical Faculty, Université de Paris, Paris, France
| |
Collapse
|
49
|
Okumoto A, Nomura Y, Maki K, Ogawa T, Onodera H, Shikano M, Okabe N. Addressing practical issues in the smooth implementation of revised guidelines for non-clinical studies of vaccines for infectious disease prevention. Regul Toxicol Pharmacol 2023:105413. [PMID: 37230176 DOI: 10.1016/j.yrtph.2023.105413] [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: 12/14/2022] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Herein, we investigated possible practical issues for the smooth implementation of the revised Japanese Guidelines for Non-clinical Studies of Vaccines for the Prevention of Infectious Diseases, which were raised in response to public comments on the proposed guideline revision and a gap analysis of the World Health Organization and European Medicines Agency guidelines. We identified main issues such as the non-clinical safety studies of adjuvants and evaluation of local cumulative tolerance in toxicity studies. The revised Japanese Pharmaceuticals and Medical Devices Agency (PMDA)/Ministry of Health, Labour and Welfare (MHLW) guidelines require non-clinical safety studies for vaccines containing new adjuvants, but additional safety pharmacology studies or safety studies in two animal species may be required if non-clinical safety studies raise any concerns (i.e., systemic distribution). Adjuvant biodistribution studies may aid in understanding vaccine characteristics. The evaluation of local cumulative tolerance in non-clinical studies, which was the focus of the Japanese review, can be omitted by including a warning in the package insert to avoid injection to the same site. The study's findings will be reflected in a Q&A to be released by the Japanese MHLW. We hope that this study will contribute to the global and harmonized development of vaccines.
Collapse
Affiliation(s)
- Atsuko Okumoto
- Graduate School of Pharmaceutical Sciences, Tokyo University of Science, 162-8601, Tokyo, Japan; Pharmaceuticals and Medical Devices Agency, 100-0013, Tokyo, Japan.
| | - Yumiko Nomura
- Graduate School of Pharmaceutical Sciences, Tokyo University of Science, 162-8601, Tokyo, Japan; Ministry of Health, Labor, and Welfare, 100-8916, Tokyo, Japan
| | - Kazushige Maki
- Pharmaceuticals and Medical Devices Agency, 100-0013, Tokyo, Japan
| | - Takashi Ogawa
- Pharmaceuticals and Medical Devices Agency, 100-0013, Tokyo, Japan
| | - Hiroshi Onodera
- National Institute of Health Sciences, 210-9501, Kanagawa, Japan
| | - Mayumi Shikano
- Graduate School of Pharmaceutical Sciences, Tokyo University of Science, 162-8601, Tokyo, Japan; Faculty of Pharmaceutical Sciences, Tokyo University of Science, 162-8601, Tokyo, Japan
| | - Nobuhiko Okabe
- Kawasaki City Institute for Public Health, 210-0821, Kanagawa, Japan
| |
Collapse
|
50
|
Kassardjian A, Sun E, Sookhoo J, Muthuraman K, Boligan KF, Kucharska I, Rujas E, Jetha A, Branch DR, Babiuk S, Barber B, Julien JP. Modular adjuvant-free pan-HLA-DR-immunotargeting subunit vaccine against SARS-CoV-2 elicits broad sarbecovirus-neutralizing antibody responses. Cell Rep 2023; 42:112391. [PMID: 37053069 PMCID: PMC10067452 DOI: 10.1016/j.celrep.2023.112391] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/14/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Subunit vaccines typically require co-administration with an adjuvant to elicit protective immunity, adding development hurdles that can impede rapid pandemic responses. To circumvent the need for adjuvant in a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) subunit vaccine, we engineer a thermostable immunotargeting vaccine (ITV) that leverages the pan-HLA-DR monoclonal antibody 44H10 to deliver the viral spike protein receptor-binding domain (RBD) to antigen-presenting cells. X-ray crystallography shows that 44H10 binds to a conserved epitope on HLA-DR, providing the basis for its broad HLA-DR reactivity. Adjuvant-free ITV immunization in rabbits and ferrets induces robust anti-RBD antibody responses that neutralize SARS-CoV-2 variants of concern and protect recipients from SARS-CoV-2 challenge. We demonstrate that the modular nature of the ITV scaffold with respect to helper T cell epitopes and diverse RBD antigens facilitates broad sarbecovirus neutralization. Our findings support anti-HLA-DR immunotargeting as an effective means to induce strong antibody responses to subunit antigens without requiring an adjuvant.
Collapse
Affiliation(s)
- Audrey Kassardjian
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Eric Sun
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jamie Sookhoo
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3M4, Canada; Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Krithika Muthuraman
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Iga Kucharska
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Edurne Rujas
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; Pharmacokinetic, Nanotechnology and Gene Therapy Group, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, 01006 Vitoria, Spain
| | - Arif Jetha
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Donald R Branch
- Canadian Blood Services, Keenan Research Centre, Toronto, ON M5B 1W8, Canada; University of Toronto, Departments of Medicine and Laboratory Medicine and Pathobiology, Toronto, ON M5S 1A8, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3M4, Canada; Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Brian Barber
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|