1
|
Di Benedetto R, Massai L, Wright M, Mancini F, Cleveland M, Rossi O, Giannelli C, Berlanda Scorza F, Micoli F. Adjuvanted Modified Bacterial Antigens for Single-Dose Vaccines. Int J Mol Sci 2024; 25:11461. [PMID: 39519015 PMCID: PMC11546299 DOI: 10.3390/ijms252111461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Alum is the most used vaccine adjuvant, due to its safety, low cost and adjuvanticity to various antigens. However, the mechanism of action of alum is complex and not yet fully understood, and the immune responses elicited can be weak and antigen-dependent. While several antigens rapidly desorb from alum upon exposure to serum, phosphorylated proteins remain tightly bound through a ligand-exchange reaction with surface hydroxyls on alum. Here, bacterial proteins and glycoconjugates have been modified with phosphoserines, aiming at enhancing the binding to alum and prolonging their bioavailability. Tetanus toxoid protein and Salmonella Typhi fragmented Vi-CRM conjugate were used. Both antigens rapidly and completely desorbed from alum after incubation with serum, verified via a competitive ELISA assay, and set up to rapidly evaluate in vitro antigen desorption from alum. After antigen modification with phosphoserines, desorption from alum was slowed down, and modified antigens demonstrated more prolonged retention at the injection sites through in vivo optical imaging in mice. Both modified antigens elicited stronger immune responses in mice, after a single injection only, compared to unmodified antigens. A stronger binding to alum could result in potent single-dose vaccine candidates and opens the possibility to design novel carrier proteins for glycoconjugates and improved versions of bacterial recombinant proteins.
Collapse
Affiliation(s)
- Roberta Di Benedetto
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.D.B.); (L.M.); (F.M.); (O.R.); (C.G.); (F.B.S.)
| | - Luisa Massai
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.D.B.); (L.M.); (F.M.); (O.R.); (C.G.); (F.B.S.)
| | - Mark Wright
- GSK, Stevenage SG1 2NFX, Hertfordshire, UK; (M.W.); (M.C.)
| | - Francesca Mancini
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.D.B.); (L.M.); (F.M.); (O.R.); (C.G.); (F.B.S.)
| | | | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.D.B.); (L.M.); (F.M.); (O.R.); (C.G.); (F.B.S.)
| | - Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.D.B.); (L.M.); (F.M.); (O.R.); (C.G.); (F.B.S.)
| | - Francesco Berlanda Scorza
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.D.B.); (L.M.); (F.M.); (O.R.); (C.G.); (F.B.S.)
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), 53100 Siena, Italy; (R.D.B.); (L.M.); (F.M.); (O.R.); (C.G.); (F.B.S.)
| |
Collapse
|
2
|
Rungelrath V, Ahmed M, Hicks L, Miller SM, Ryter KT, Montgomery K, Ettenger G, Riffey A, Abdelwahab WM, Khader SA, Evans JT. Vaccination with Mincle agonist UM-1098 and mycobacterial antigens induces protective Th1 and Th17 responses. NPJ Vaccines 2024; 9:100. [PMID: 38844494 PMCID: PMC11156909 DOI: 10.1038/s41541-024-00897-x] [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: 01/31/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is one of the top infectious killers in the world. The only licensed vaccine against TB, Bacille Calmette-Guérin (BCG), provides variable protection against pulmonary TB, especially in adults. Hence, novel TB vaccine approaches are urgently needed. Both Th1 and Th17 responses are necessary for protection against TB, yet effective adjuvants and vaccine delivery systems for inducing robust Th1 and Th17 immunity are lacking. Herein we describe a synthetic Mincle agonist, UM-1098, and a silica nanoparticle delivery system that drives Th1/Th17 responses to Mtb antigens. Stimulation of human peripheral blood mononuclear cells (hPBMCs) with UM-1098 induced high levels of Th17 polarizing cytokines IL-6, IL-1β, IL-23 as well as IL-12p70, IL-4 and TNF-α in vitro. PBMCs from both C57BL/6 and BALB/c mice responded with a similar cytokine pattern in vitro and in vivo. Importantly, intramuscular (I.M.) vaccination with UM-1098-adjuvanted TB antigen M72 resulted in significantly higher antigen-specific IFN-γ and IL-17A levels in C57BL/6 wt mice than Mincle KO mice. Vaccination of C57BL/6 wt mice with immunodominant Mtb antigens ESAT6/Ag85B or M72 resulted in predominantly Th1 and Th17 responses and induced antigen-specific serum antibodies. Notably, in a virulent Mtb challenge model, vaccination with UM-1098 adjuvanted ESAT6/Ag85B or M72 significantly reduced lung bacterial burden when compared with unvaccinated mice and protection occurred in the absence of pulmonary inflammation. These data demonstrate that the synthetic Mincle agonist UM-1098 induces strong Th1 and Th17 immunity after vaccination with Mtb antigens and provides protection against Mtb infection in mice.
Collapse
Affiliation(s)
- Viktoria Rungelrath
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Mushtaq Ahmed
- Department of Microbiology, University of Chicago, 920 E. 58th St., Chicago, IL, 60637, USA
| | - Linda Hicks
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kendal T Ryter
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kyle Montgomery
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - George Ettenger
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Alexander Riffey
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Walid M Abdelwahab
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Shabaana Abdul Khader
- Department of Microbiology, University of Chicago, 920 E. 58th St., Chicago, IL, 60637, USA
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA.
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA.
| |
Collapse
|
3
|
Marin C, Ruiz Moreno FN, Sánchez Vallecillo MF, Pascual MM, Dho ND, Allemandi DA, Palma SD, Pistoresi-Palencia MC, Crespo MI, Gomez CG, Morón G, Maletto BA. Improved biodistribution and enhanced immune response of subunit vaccine using a nanostructure formed by self-assembly of ascorbyl palmitate. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 58:102749. [PMID: 38719107 DOI: 10.1016/j.nano.2024.102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/04/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
New adjuvant strategies are needed to improve protein-based subunit vaccine immunogenicity. We examined the potential to use nanostructure of 6-O-ascorbyl palmitate to formulate ovalbumin (OVA) protein and an oligodeoxynucleotide (CpG-ODN) (OCC). In mice immunized with a single dose, OCC elicited an OVA-specific immune response superior to OVA/CpG-ODN solution (OC). Rheological studies demonstrated OCC's self-assembling viscoelastic properties. Biodistribution studies indicated that OCC prolonged OVA and CpG-ODN retention at injection site and lymph nodes, reducing systemic spread. Flow-cytometry assays demonstrated that OCC promoted OVA and CpG-ODN co-uptake by Ly6ChiCD11bhiCD11c+ monocytes. OCC and OC induced early IFN-γ in lymph nodes, but OCC led to higher concentration. Conversely, mice immunized with OC showed higher serum IFN-γ concentration compared to those immunized with OCC. In mice immunized with OCC, NK1.1+ cells were the IFN-γ major producers, and IFN-γ was essential for OVA-specific IgG2c switching. These findings illustrate how this nanostructure improves vaccine's response.
Collapse
Affiliation(s)
- Constanza Marin
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Federico N Ruiz Moreno
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - María F Sánchez Vallecillo
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - María M Pascual
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Nicolas D Dho
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Daniel A Allemandi
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas; CONICET, UNITEFA, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Santiago D Palma
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas; CONICET, UNITEFA, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - María C Pistoresi-Palencia
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - María I Crespo
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Cesar G Gomez
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Orgánica, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina; CONICET, IPQA, Av. Vélez Sarsfield 1611, 5016 Córdoba, Argentina
| | - Gabriel Morón
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina
| | - Belkys A Maletto
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica; CONICET, CIBICI, Haya de la Torre y Medina Allende, X5000HUA Córdoba, Argentina.
| |
Collapse
|
4
|
Wang H, Wang S, Fang R, Li X, Xing J, Li Z, Song N. Enhancing TB Vaccine Efficacy: Current Progress on Vaccines, Adjuvants and Immunization Strategies. Vaccines (Basel) 2023; 12:38. [PMID: 38250851 PMCID: PMC10820143 DOI: 10.3390/vaccines12010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Tuberculosis (TB) remains a global infectious disease primarily transmitted via respiratory tract infection. Presently, vaccination stands as the primary method for TB prevention, predominantly reliant on the Bacillus Calmette-Guérin (BCG) vaccine. Although it is effective in preventing disseminated diseases in children, its impact on adults is limited. To broaden vaccine protection, efforts are underway to accelerate the development of new TB vaccines. However, challenges arise due to the limited immunogenicity and safety of these vaccines, necessitating adjuvants to bolster their ability to elicit a robust immune response for improved and safer immunization. These adjuvants function by augmenting cellular and humoral immunity against M. tuberculosis antigens via different delivery systems, ultimately enhancing vaccine efficacy. Therefore, this paper reviews and summarizes the current research progress on M. tuberculosis vaccines and their associated adjuvants, aiming to provide a valuable reference for the development of novel TB vaccines and the screening of adjuvants.
Collapse
Affiliation(s)
- Hui Wang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Shuxian Wang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Ren Fang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Xiaotian Li
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Jiayin Xing
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Zhaoli Li
- SAFE Pharmaceutical Technology Co., Ltd., Beijing 100000, China
| | - Ningning Song
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| |
Collapse
|
5
|
López-Serrano S, Mahmmod YS, Christensen D, Ebensen T, Guzmán CA, Rodríguez F, Segalés J, Aragón V. Immune responses following neonatal vaccination with conserved F4 fragment of VtaA proteins from virulent Glaesserella parasuis adjuvanted with CAF®01 or CDA. Vaccine X 2023; 14:100330. [PMID: 37361051 PMCID: PMC10285277 DOI: 10.1016/j.jvacx.2023.100330] [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: 03/08/2023] [Revised: 06/07/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Glaesserella parasuis is a Gram-negative bacterium that colonizes the upper airways of swine, capable of causing a systemic infection called Glässer's disease. This disease is more frequent in young post-weaning piglets. Current treatments against G. parasuis infection are based on the use of antimicrobials or inactivated vaccines, which promote limited cross-protection against different serovars. For this reason, there is an interest in developing novel subunit vaccines with the capacity to confer effective protection against different virulent strains. Herein, we characterize the immunogenicity and the potential benefits of neonatal immunization with two different vaccine formulations based on the F4 polypeptide, a conserved immunogenic protein fragment from the virulence-associated trimeric autotransporters of virulent G. parasuis strains. With this purpose, we immunized two groups of piglets with F4 combined with cationic adjuvant CAF®01 or cyclic dinucleotide CDA. Piglets immunized with a commercial bacterin and non-immunized animals served as control groups. The vaccinated piglets received two doses of vaccine, at 14 days old and 21 days later. The immune response induced against the F4 polypeptide varied depending on the adjuvant used. Piglets vaccinated with the F4+CDA vaccine developed specific anti-F4 IgGs, biased towards the induction of IgG1 responses, whereas no anti-F4 IgGs were de novo induced after immunization with the CAF®01 vaccine. Piglets immunized with both formulations displayed balanced memory T-cell responses, evidenced upon in vitro re-stimulation of peripheral blood mononuclear cells with F4. Interestingly, pigs immunized with F4+CAF®01 controlled more efficiently a natural nasal colonization by a virulent serovar 4 G. parasuis that spontaneously occurred during the experimental procedure. According to the results, the immunogenicity and the protection afforded by F4 depend on the adjuvant used. F4 may represent a candidate to consider for a Glässer's disease vaccine and could contribute to a better understanding of the mechanisms involved in protection against virulent G. parasuis colonization.
Collapse
Affiliation(s)
- Sergi López-Serrano
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Institut de Recerca i Tecnologia Agroalimentàries, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Catalonia, Spain
| | - Yasser S. Mahmmod
- Department of Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
- Section of Veterinary Sciences, Health Sciences Division, Al Ain Men’s College, Higher Colleges of Technology, Al Ain 17155, United Arab Emirates
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Fernando Rodríguez
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Institut de Recerca i Tecnologia Agroalimentàries, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Catalonia, Spain
| | - Joaquim Segalés
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Institut de Recerca i Tecnologia Agroalimentàries, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Departament de Sanitat i Anatomia animals. Facultat de Veterinària. Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
| | - Virginia Aragón
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Institut de Recerca i Tecnologia Agroalimentàries, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Catalonia, Spain
| |
Collapse
|
6
|
Thakur A, Wadhwa A, Lokras A, Müllertz OAO, Christensen D, Franzyk H, Foged C. Method of manufacturing CAF®09 liposomes affects immune responses induced by adjuvanted subunit proteins. Eur J Pharm Biopharm 2023; 189:84-97. [PMID: 37059402 DOI: 10.1016/j.ejpb.2023.04.005] [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: 12/22/2022] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
The ability to induce antigen-specific CD4+ and CD8+T-cell responses is one of the fundamental requirements when developing new efficacious vaccines against challenging infectious diseases and cancer. However, no adjuvants are currently approved for human subunit vaccines that induce T-cell immunity. Here, we incorporated a Toll-like receptor 4 agonist, i.e., the ionizable lipidoid L5N12, in the liposomal cationic adjuvant formulation 09 (CAF®09), and found that modified CAF®09 liposomes possess preserved adjuvant function as compared to unmodified CAF®09. CAF®09 consists of the cationic lipid dimethyldioctadecylammonium (DDA), monomycoloyl glycerol analogue 1 (MMG-1), and polyinosinic:polycytidylic acid [poly(I:C)]. By using the microfluidic mixing technology for liposome preparation, we gradually replaced DDA with L5N12, while keeping the molar ratios of MMG-1 and poly(I:C) constant. We found that this type of modification resulted in colloidally stable liposomes, which were significantly smaller and displayed reduced surface charge as compared to unmodified CAF®09, prepared by using the conventional thin film method. We showed that incorporation of L5N12 decreases the membrane rigidity of CAF®09 liposomes. Furthermore, vaccination with antigen adjuvanted with L5N12-modified CAF®09 or antigen adjuvanted with unmodified CAF®09, respectively, induced comparable antigen-specific serum antibody titers. We found that antigen adjuvanted with L5N12-modified CAF®09 induced antigen-specific effector and memory CD4+ and CD8+T-cell responses in the spleen comparable to those induced when unmodified CAF®09 was used as adjuvant. However, incorporating L5N12 did not have a synergistic immunopotentiating effect on the antibody and T-cell responses induced by CAF®09. Moreover, vaccination with antigen adjuvanted with unmodified CAF®09, which was manufactured by using microfluidic mixing, induced significantly lower antigen-specific CD4+ and CD8+T-cell responses than vaccination with antigen adjuvanted with unmodified CAF®09, which was prepared by using the thin film method. These results show that the method of manufacturing affects CAF®09 liposome adjuvanted antigen-specific immune responses, which should be taken into consideration when evaluating immunogenicity of subunit protein vaccines.
Collapse
Affiliation(s)
- Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| | - Abishek Wadhwa
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Abhijeet Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Olivia Amanda Oest Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, 2100 Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| |
Collapse
|
7
|
Verma SK, Mahajan P, Singh NK, Gupta A, Aggarwal R, Rappuoli R, Johri AK. New-age vaccine adjuvants, their development, and future perspective. Front Immunol 2023; 14:1043109. [PMID: 36911719 PMCID: PMC9998920 DOI: 10.3389/fimmu.2023.1043109] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/26/2023] [Indexed: 02/26/2023] Open
Abstract
In the present scenario, immunization is of utmost importance as it keeps us safe and protects us from infectious agents. Despite the great success in the field of vaccinology, there is a need to not only develop safe and ideal vaccines to fight deadly infections but also improve the quality of existing vaccines in terms of partial or inconsistent protection. Generally, subunit vaccines are known to be safe in nature, but they are mostly found to be incapable of generating the optimum immune response. Hence, there is a great possibility of improving the potential of a vaccine in formulation with novel adjuvants, which can effectively impart superior immunity. The vaccine(s) in formulation with novel adjuvants may also be helpful in fighting pathogens of high antigenic diversity. However, due to the limitations of safety and toxicity, very few human-compatible adjuvants have been approved. In this review, we mainly focus on the need for new and improved vaccines; the definition of and the need for adjuvants; the characteristics and mechanisms of human-compatible adjuvants; the current status of vaccine adjuvants, mucosal vaccine adjuvants, and adjuvants in clinical development; and future directions.
Collapse
Affiliation(s)
| | - Pooja Mahajan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI, United States
| | - Ankit Gupta
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | - Rupesh Aggarwal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
8
|
Müllertz OAO, Andersen P, Christensen D, Foged C, Thakur A. Pulmonary Administration of the Liposome-Based Adjuvant CAF01: Effect of Surface Charge on Mucosal Adjuvant Function. Mol Pharm 2023; 20:953-970. [PMID: 36583936 DOI: 10.1021/acs.molpharmaceut.2c00634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mucosal surfaces of the lungs represent a major site of entry for airborne pathogens, and pulmonary administration of vaccines is an attractive strategy to induce protective mucosal immunity in the airways. Recently, we demonstrated the potential of pulmonary vaccination with the tuberculosis subunit antigen H56 adjuvanted with the cationic liposomal adjuvant formulation CAF01, which consists of the cationic lipid dimethyldioctadecylammonium (DDA) bromide and the synthetic cord factor trehalose-6,6'-dibehenate. However, the cationic charge of DDA represents a major safety challenge. Hence, replacing DDA with a safer zwitterionic or anionic phospholipid is an attractive approach to improve vaccine safety, but the effect of liposomal surface charge on the induction of mucosal immunity after airway immunization is poorly understood. Here, we investigated the effect of surface charge by replacing the cationic DDA component of CAF01 with zwitterionic dipalmitoylphosphatidylcholine (DPPC) or anionic dipalmitoylphosphatidylglycerol (DPPG), and we show that charge modification enhances antigen-specific pulmonary T-cell responses against co-formulated H56. We systematically replaced DDA with either DPPC or DPPG and found that these modifications resulted in colloidally stable liposomes that have similar size and morphology to unmodified CAF01. DPPC- or DPPG-modified CAF01 displayed surface charge-dependent protein adsorption and induced slightly higher follicular helper T cells and germinal center B cells in the lung-draining lymph nodes than unmodified CAF01. In addition, modified CAF01 induced significantly higher levels of H56-specific Th17 cells and polyfunctional CD4+ T cells in the lungs, as compared to unmodified CAF01. However, the strong H56-specific humoral responses induced by CAF01 in the lungs and spleen were not influenced by surface charge. Hence, these results provide insights into the importance of surface charge for liposomal adjuvant function and can also guide the design of safe pulmonary subunit vaccines against other mucosal pathogens.
Collapse
Affiliation(s)
- Olivia Amanda Oest Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, Copenhagen S2300, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, Copenhagen S2300, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| |
Collapse
|
9
|
Nguyen NDNT, Guleed S, Olsen AW, Follmann F, Christensen JP, Dietrich J. Th1/Th17 T cell Tissue-Resident Immunity Increases Protection, But Is Not Required in a Vaccine Strategy Against Genital Infection With Chlamydia trachomatis. Front Immunol 2021; 12:790463. [PMID: 34925371 PMCID: PMC8674352 DOI: 10.3389/fimmu.2021.790463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
The requirement for vaccine-induced tissue-resident immunity for protection against one or repeated infections with Chlamydia trachomatis (C.t.) is still not fully resolved. In this study, our aim was to investigate to which degree tissue-resident Th1/Th17 T cells in the genital tract (GT) could add to the protection mediated by circulating immunity. Out of several mucosal vaccine strategies, a strategy termed SIM (for simultaneous intrauterine and parenteral immunization with CAF01 adjuvanted CTH522), was superior in generating genital tract tissue-resident Th1/Th17 T cell immunity. This led to a faster and stronger local CD4 T cell response post infection, consisting of multifunctional IFNγ/TNFα-producing Th1 T cells and IFNγ/TNFα/IL-17-producing Th17 T cells, and a faster recruitment of innate immune cells. Post infection, SIM animals showed an additional significant reduction in bacterial levels compared to mice having received only a parenteral vaccine. Nevertheless, the parenteral strategy reduced bacterial levels by 75%, and interestingly, post infection, these mice generated their own vaccine-derived genital tract tissue-resident memory Th1/Th17 T cells, which upon a subsequent infection showed as fast an activation in the genital tract, as observed in SIM mice. Furthermore, in contrast to after the first infection, both groups of mice now showed a similar infection-induced boost in local vaginal IgA and IgG titers. Thus, vaccine-induced resident immunity, generated pre-infection, led to an advantage in the response against the first infection, but not the second infection, suggesting that a parenteral vaccine strategy is a suitable vaccine strategy against infections with Chlamydia trachomatis.
Collapse
Affiliation(s)
| | - Safia Guleed
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Anja Weinreich Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | - Jes Dietrich
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
10
|
Wørzner K, Hvannastein J, Schmidt ST, Foged C, Rosenkrands I, Pedersen GK, Christensen D. Adsorption of protein antigen to the cationic liposome adjuvant CAF®01 is required for induction of Th1 and Th17 responses but not for antibody induction. Eur J Pharm Biopharm 2021; 165:293-305. [PMID: 34044110 PMCID: PMC8212872 DOI: 10.1016/j.ejpb.2021.05.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/20/2021] [Accepted: 05/19/2021] [Indexed: 11/24/2022]
Abstract
The degree of antigen adsorption to adjuvants in subunit vaccines may significantly influence the immune responses they induce upon vaccination. Commonly used approaches for studying how the level of adsorption affects the induction of antigen-specific immune responses include (i) using adjuvants with different abilities to adsorb antigens, and (ii) comparing different antigens selected based on their ability to adsorb to the adjuvant. A weakness of these approaches is that not only the antigen adsorption level is varied, but also other important functional factors such as adjuvant composition and/or the B/T cell epitopes, which may affect immunogenicity. Hence, we investigated how changing the adsorption capabilities of a single antigen to an adjuvant influenced the vaccine-induced immune responses. The model antigen lysozyme, which displays a positive net charge at physiological pH due to an isoelectric point (pI) of 11, was succinylated to different extents, resulting in a reduction of the pI value to 4.4–5.9, depending on the degree of succinylation. A pronounced inverse correlation was found between the pI value of the succinylated lysozyme analogues and the degree of adsorption to a cationic liposomal adjuvant consisting of dimethyldioctadecylammonium bromide (DDA) and trehalose dibehenate (TDB) (CAF®01). Furthermore, increased adsorption to this adjuvant correlated directly with the magnitude of lysozyme-specific Th1/Th17 immune responses induced by the vaccine in mice, while there was an inverse correlation with antibody induction. However, high lysozyme-specific antibody titers were induced with an increased antigen dose, even upon vaccination with a strongly adsorbed succinylated lysozyme analogue. Hence, these data illustrate that the degree of lysozyme adsorption to CAF®01 strongly affects the quality of the resulting immune responses.
Collapse
Affiliation(s)
- Katharina Wørzner
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Jóhanna Hvannastein
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Signe Tandrup Schmidt
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Ida Rosenkrands
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Gabriel Kristian Pedersen
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Dennis Christensen
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300 Copenhagen S, Denmark.
| |
Collapse
|
11
|
Abdelwahab WM, Riffey A, Buhl C, Johnson C, Ryter K, Evans JT, Burkhart DJ. Co-adsorption of synthetic Mincle agonists and antigen to silica nanoparticles for enhanced vaccine activity: A formulation approach to co-delivery. Int J Pharm 2020; 593:120119. [PMID: 33249249 DOI: 10.1016/j.ijpharm.2020.120119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/12/2020] [Accepted: 11/22/2020] [Indexed: 01/14/2023]
Abstract
To date there is no clinically approved adjuvant to drive a protective T-helper cell 17 (Th17) immune response against Mycobacterium tuberculosis (Mtb). Trehalose Dimycolate (TDM) is a glycolipid molecule found in the cell wall of Mtb and similar species. Our team has discovered novel synthetic TDM derivatives that target Mincle receptors and when presented on the surface of amine functionalized silica nanoparticles (A-SNPs) adopt the requisite supramolecular structure for Mincle receptor agonism. Here we describe the preparation and characterization methods for these critical silica nanoparticles (SNPs) co-loaded with Mincle agonists (MAs) and a model antigen. In this work, A-SNPs with a particle diameter of 246 ± 11 nm were prepared and examined for co-adsorption of two synthetic MAs along with ovalbumin (OVA). Due to the insolubility of the studied MAs in aqueous environment, aggregation of the MAs made separation of the adjuvant-loaded A-SNPs from the free-form MAs via centrifugation very challenging. To facilitate separation, we synthesized modified SNPs with comparable amine surface functionalization to the original A-SNPs, but with a superparamagnetic iron oxide core (M-A-SNPs), to allow for magnetic separation. We also substituted Alexa Fluor 488-labeled ovalbumin (AF 488 OVA) for the un-tagged OVA to improve the sensitivity of our quantitation method. A RP-HPLC method was developed to simultaneously determine the amount of adsorption of both the Mincle adjuvant and the model antigen to the A-SNPs. AF488 OVA demonstrated higher than 90% adsorption, with or without the co-adsorption of MAs. Likewise, MAs exhibited higher than 80% adsorption in the presence or absence of antigen. The developed formulations were tested in vitro using murine RAW cells and human peripheral blood mononuclear cells, exhibiting good cytokine induction in both cell lines. Results from these studies indicate that A-SNPs could be used as a customizable presentation platform to co-deliver antigens along with different MAs of varying structural features and biophysical properties.
Collapse
Affiliation(s)
- Walid M Abdelwahab
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Alexander Riffey
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Cassie Buhl
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Craig Johnson
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Kendal Ryter
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - David J Burkhart
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States.
| |
Collapse
|
12
|
Wang Y, Yu Z, Zhou Y, Zhu Y, Wang J, Fu J, Yuan Y, Chen S, Wang Y, Yu W, Gao P, Zhu W, Cheng Q, Cho SH, Kong W, Chen J. Different doses of ovalbumin exposure on dendritic cells determine their genetic/epigenetic regulation and T cell differentiation. Aging (Albany NY) 2020; 12:25432-25451. [PMID: 33234726 PMCID: PMC7803576 DOI: 10.18632/aging.104145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
It has been reported that allergen dosage can impact the differentiation of dendritic cells (DCs)-mediated T cells. However, the mechanisms of such dose-dependent differentiation are poorly understood. In this study, bone marrow-derived immature DCs stimulated with Ovalbumin (OVA) of different concentrations (0, 10, 100, 1000, 10000μg/ml, respectively). DCs were then co-cultured with naïve T cells. RNA-sequencing detection and DNA methylation of DCs were performed. We show that when DCs were stimulated with low-dose (10μg/ml), 77 genes were up-regulated and 87 genes down-regulated. Most activated genes were related to ribosome synthesis and ion channel inhibition. At the medium-dose (100μg/ml), 339 genes were up-regulated and 168 genes down-regulated. Most activated genes involved cytokine synthesis and regulation of immune responses. At high-dose (10000μg/ml), 2497 genes were up-regulated and 1156 genes down-regulated. TNF signaling pathway, NF-kappa B signaling pathway, antigen processing and presentation signaling pathway were mostly up-regulated. The related co-stimulators, co-inhibitory molecules, inhibitory cytokines, negative regulating enzymes were highly expressed. The monocarbate, coenzyme, fatty acid, glucolipid, starch, sucrose and other metabolism-related signaling pathways were down-regulated. The profiles of DNA methylation and RNA synthesis of DCs varied with different doses of OVA, which serves to induce T cells to differentiate in various directions.
Collapse
Affiliation(s)
- Ying Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zizhong Yu
- Department of Otorhinolaryngology, Taihe Hospital, Hubei University of Medicine, Wuhan, China
| | - Yue Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Zhu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinhui Wang
- Department of Otorhinolaryngology, First Hospital of Handan, Wuhan, China
| | - Junmei Fu
- Department of Otorhinolaryngology, First People’s Hospital of Jiangxia District, Wuhan, China
| | - Yang Yuan
- Department of Otorhinolaryngology, Wuhan General Hospital of the Chinese People’s Liberation Army, Wuhan, China
| | - Shan Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanjun Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenting Yu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Gao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanting Zhu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Cheng
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Seong H Cho
- Division of Allergy-Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Weijia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianjun Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Anderluzzi G, Schmidt ST, Cunliffe R, Woods S, Roberts CW, Veggi D, Ferlenghi I, O'Hagan DT, Baudner BC, Perrie Y. Rational design of adjuvants for subunit vaccines: The format of cationic adjuvants affects the induction of antigen-specific antibody responses. J Control Release 2020; 330:933-944. [PMID: 33152394 DOI: 10.1016/j.jconrel.2020.10.066] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/18/2022]
Abstract
A range of cationic delivery systems have been investigated as vaccine adjuvants, though few direct comparisons exist. To investigate the impact of the delivery platform, we prepared four cationic systems (emulsions, liposomes, polymeric nanoparticles and solid lipid nanoparticles) all containing equal concentrations of the cationic lipid dimethyldioctadecylammonium bromide in combination with the Neisseria adhesin A variant 3 subunit antigen. The formulations were physicochemically characterized and their ability to associate with cells and promote antigen processing (based on degradation of DQ-OVA, a substrate for proteases which upon hydrolysis is fluorescent) was compared in vitro and their vaccine efficacy (antigen-specific antibody responses and IFN-γ production) and biodistribution (antigen and adjuvant) were evaluated in vivo. Due to their cationic nature, all delivery systems gave high antigen loading (> 85%) with liposomes, lipid nanoparticles and emulsions being <200 nm, whilst polymeric nanoparticles were larger (~350 nm). In vitro, the particulate systems tended to promote cell uptake and antigen processing, whilst emulsions were less effective. Similarly, whilst the particulate delivery systems induced a depot (of both delivery system and antigen) at the injection site, the cationic emulsions did not. However, out of the systems tested the cationic emulsions induced the highest antibody responses. These results demonstrate that while cationic lipids can have strong adjuvant activity, their formulation platform influences their immunogenicity.
Collapse
Affiliation(s)
- Giulia Anderluzzi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK; GSK, Siena, Italy
| | - Signe Tandrup Schmidt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK; Department of Infectious Disease Immunology, Center for Vaccine Research, Statens Serum Institut, Artillerivej 5, Copenhagen S 2300, Denmark
| | - Robert Cunliffe
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK; GSK, Siena, Italy
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Craig W Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | | | | | | | | | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK.
| |
Collapse
|
14
|
Lofano G, Mallett CP, Bertholet S, O’Hagan DT. Technological approaches to streamline vaccination schedules, progressing towards single-dose vaccines. NPJ Vaccines 2020; 5:88. [PMID: 33024579 PMCID: PMC7501859 DOI: 10.1038/s41541-020-00238-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022] Open
Abstract
Vaccines represent the most successful medical intervention in history, with billions of lives saved. Although multiple doses of the same vaccine are typically required to reach an adequate level of protection, it would be advantageous to develop vaccines that induce protective immunity with fewer doses, ideally just one. Single-dose vaccines would be ideal to maximize vaccination coverage, help stakeholders to greatly reduce the costs associated with vaccination, and improve patient convenience. Here we describe past attempts to develop potent single dose vaccines and explore the reasons they failed. Then, we review key immunological mechanisms of the vaccine-specific immune responses, and how innovative technologies and approaches are guiding the preclinical and clinical development of potent single-dose vaccines. By modulating the spatio-temporal delivery of the vaccine components, by providing the appropriate stimuli to the innate immunity, and by designing better antigens, the new technologies and approaches leverage our current knowledge of the immune system and may synergize to enable the rational design of next-generation vaccination strategies. This review provides a rational perspective on the possible development of future single-dose vaccines.
Collapse
Affiliation(s)
- Giuseppe Lofano
- GSK, Slaoui Center for Vaccines Research, Rockville, MD 20850 USA
| | - Corey P. Mallett
- GSK, Slaoui Center for Vaccines Research, Rockville, MD 20850 USA
| | - Sylvie Bertholet
- GSK, Slaoui Center for Vaccines Research, Rockville, MD 20850 USA
| | - Derek T. O’Hagan
- GSK, Slaoui Center for Vaccines Research, Rockville, MD 20850 USA
| |
Collapse
|
15
|
Thakur A, Pinto FE, Hansen HS, Andersen P, Christensen D, Janfelt C, Foged C. Intrapulmonary (i.pulmon.) Pull Immunization With the Tuberculosis Subunit Vaccine Candidate H56/CAF01 After Intramuscular (i.m.) Priming Elicits a Distinct Innate Myeloid Response and Activation of Antigen-Presenting Cells Than i.m. or i.pulmon. Prime Immunization Alone. Front Immunol 2020; 11:803. [PMID: 32457748 PMCID: PMC7221191 DOI: 10.3389/fimmu.2020.00803] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/08/2020] [Indexed: 12/22/2022] Open
Abstract
Understanding the in vivo fate of vaccine antigens and adjuvants and their safety is crucial for the rational design of mucosal subunit vaccines. Prime and pull vaccination using the T helper 17-inducing adjuvant CAF01 administered parenterally and mucosally, respectively, has previously been suggested as a promising strategy to redirect immunity to mucosal tissues. Recently, we reported a promising tuberculosis (TB) vaccination strategy comprising of parenteral priming followed by intrapulmonary (i.pulmon.) mucosal pull immunization with the TB subunit vaccine candidate H56/CAF01, which resulted in the induction of lung-localized, H56-specific T cells and systemic as well as lung mucosal IgA responses. Here, we investigate the uptake of H56/CAF01 by mucosal and systemic innate myeloid cells, antigen-presenting cells (APCs), lung epithelial cells and endothelial cells in mice after parenteral prime combined with i.pulmon. pull immunization, and after parenteral or i.pulmon. prime immunization alone. We find that i.pulmon. pull immunization of mice with H56/CAF01, which are parenterally primed with H56/CAF01, substantially enhances vaccine uptake and presentation by pulmonary and splenic APCs, pulmonary endothelial cells and type I epithelial cells and induces stronger activation of dendritic cells in the lung-draining lymph nodes, compared with parenteral immunization alone, which suggests activation of both innate and memory responses. Using mass spectrometry imaging of lipid biomarkers, we further show that (i) airway mucosal immunization with H56/CAF01 neither induces apparent local tissue damage nor inflammation in the lungs, and (ii) the presence of CAF01 is accompanied by evidence of an altered phagocytic activity in alveolar macrophages, evident from co-localization of CAF01 with the biomarker bis(monoacylglycero)phosphate, which is expressed in the late endosomes and lysosomes of phagocytosing macrophages. Hence, our data demonstrate that innate myeloid responses differ after one and two immunizations, respectively, and the priming route and boosting route individually affect this outcome. These findings may have important implications for the design of mucosal vaccines intended for safe administration in the airways.
Collapse
Affiliation(s)
- Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Harald Severin Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Christian Janfelt
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Miyake Y, Yamasaki S. Immune Recognition of Pathogen-Derived Glycolipids Through Mincle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:31-56. [DOI: 10.1007/978-981-15-1580-4_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Schmidt ST, Pedersen GK, Christensen D. Rational Design and In Vivo Characterization of Vaccine Adjuvants. ILAR J 2019; 59:309-322. [PMID: 30624655 DOI: 10.1093/ilar/ily018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 09/05/2018] [Indexed: 12/14/2022] Open
Abstract
Many different adjuvants are currently being developed for subunit vaccines against a number of pathogens and diseases. Rational design is increasingly used to develop novel vaccine adjuvants, which requires extensive knowledge of, for example, the desired immune responses, target antigen-presenting cell subsets, their localization, and expression of relevant pattern-recognition receptors. The adjuvant mechanism of action and efficacy are usually evaluated in animal models, where mice are by far the most used. In this review, we present methods for assessing adjuvant efficacy and function in animal models: (1) whole-body biodistribution evaluated by using fluorescently and radioactively labeled vaccine components; (2) association and activation of immune cell subsets at the injection site, in the draining lymph node, and the spleen; (4) adaptive immune responses, such as cytotoxic T-lymphocytes, various T-helper cell subsets, and antibody responses, which may be quantitatively evaluated using ELISA, ELISPOT, and immunoplex assays and qualitatively evaluated using flow cytometric and single cell sequencing assays; and (5) effector responses, for example, antigen-specific cytotoxic potential of CD8+ T cells and antibody neutralization assays. While the vaccine-induced immune responses in mice often correlate with the responses induced in humans, there are instances where immune responses detected in mice are not translated to the human situation. We discuss some examples of correlation and discrepancy between mouse and human immune responses and how to understand them.
Collapse
Affiliation(s)
- Signe Tandrup Schmidt
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| | - Gabriel Kristian Pedersen
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| | - Dennis Christensen
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| |
Collapse
|
18
|
Roces CB, Khadke S, Christensen D, Perrie Y. Scale-Independent Microfluidic Production of Cationic Liposomal Adjuvants and Development of Enhanced Lymphatic Targeting Strategies. Mol Pharm 2019; 16:4372-4386. [DOI: 10.1021/acs.molpharmaceut.9b00730] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Carla B. Roces
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Swapnil Khadke
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, DK-2300 Copenhagen, Denmark
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| |
Collapse
|
19
|
Abstract
Introduction: Neonates are less responsive to vaccines than adults, making it harder to protect newborns against infection. Neonatal differences in antigen-presenting cell, B and T cell function, all likely contribute. A key question is whether novel adjuvants might be able to make neonatal vaccines more effective. Areas covered: This review addresses the issues of how to improve neonatal vaccines, which we have defined as vaccines given in the first 4 weeks of life in a human infant or the first week of life in a mouse. A search was performed using keywords including 'neonatal immunity', 'neonatal immunisation', 'vaccine' and 'adjuvant' of PubMed articles published between 1960 and 2018. Expert opinion: Sugar-like structures have recently been shown to prime the infant adaptive immune system to respond to vaccines, being potentially more effective than traditional adjuvants. Sugar-based compounds with beneficial adjuvant effects in neonatal vaccine models include delta inulin (Advax), curdlan, and trehalose 6,6'-dibehenate. Such compounds make interesting neonatal adjuvant candidates, either used alone or in combination with traditional innate immune adjuvants.
Collapse
Affiliation(s)
- Isaac G Sakala
- a Vaxine Pty Ltd , Adelaide , Australia.,b Department of Diabetes and Endocrinology, Flinders Medical Centre/Flinders University , Adelaide , Australia
| | - Katherine Marie Eichinger
- c Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh , Pittsburgh , PA , USA
| | - Nikolai Petrovsky
- a Vaxine Pty Ltd , Adelaide , Australia.,b Department of Diabetes and Endocrinology, Flinders Medical Centre/Flinders University , Adelaide , Australia
| |
Collapse
|
20
|
Smith AJ, Miller SM, Buhl C, Child R, Whitacre M, Schoener R, Ettenger G, Burkhart D, Ryter K, Evans JT. Species-Specific Structural Requirements of Alpha-Branched Trehalose Diester Mincle Agonists. Front Immunol 2019; 10:338. [PMID: 30873180 PMCID: PMC6403188 DOI: 10.3389/fimmu.2019.00338] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 02/08/2019] [Indexed: 12/31/2022] Open
Abstract
Despite the ever present need for an effective Mycobacterium tuberculosis (Mtb) vaccine, efforts for development have been largely unsuccessful. Correlates of immune protection against Mtb are not wholly defined, but Th1 and likely Th17 adaptive immune responses have been demonstrated to be necessary for vaccine-mediated protection. Unfortunately, no approved adjuvants are able to drive a Th17 response, though recent clinical trials with CAF01 have demonstrated proof of concept. Herein we present the discovery and characterization of a new class of potential Th17-inducing vaccine adjuvants, alpha-branched trehalose diester molecules (αTDE). Based off the Mtb immunostimulatory component trehalose dimycolate (TDM), we synthesized and evaluated the immunostimulatory capacity of a library of structural derivatives. We evaluated the structure activity relationship of the compounds in relation to chain length and engagement of the Mincle receptor, production of innate cytokines from human and murine cells, and a pro-Th17 cytokine profile from primary human peripheral blood mononuclear cells. Murine cells displayed more structural tolerance, engaging and responding to a wide array of compound chain lengths. Interestingly, human cells displayed a unique specificity for ester chains between 5 and 14 carbons for maximal immune stimulating activity. Evaluation of two distinct αTDEs, B16 and B42, in concert with a recombinant Mtb antigen demonstrated their ability to augment a Th17 immune response against a Mtb antigen in vivo. Collectively this data describes the species-specific structural requirements for maximal human activity of alpha-branched trehalose diester compounds and demonstrates their capacity to serve as potent Th17-inducing adjuvants.
Collapse
Affiliation(s)
- Alyson J Smith
- Center for Translational Medicine, Missoula, MT, United States.,Division of Biological Sciences, Missoula, MT, United States
| | - Shannon M Miller
- Center for Translational Medicine, Missoula, MT, United States.,Division of Biological Sciences, Missoula, MT, United States
| | - Cassandra Buhl
- Center for Translational Medicine, Missoula, MT, United States.,Division of Biological Sciences, Missoula, MT, United States
| | - Robert Child
- Center for Translational Medicine, Missoula, MT, United States.,Division of Biological Sciences, Missoula, MT, United States
| | - Margaret Whitacre
- Center for Translational Medicine, Missoula, MT, United States.,Division of Biological Sciences, Missoula, MT, United States
| | - Roman Schoener
- Center for Translational Medicine, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, Missoula, MT, United States
| | - George Ettenger
- Center for Translational Medicine, Missoula, MT, United States.,Department of Chemistry, University of Montana, Missoula, MT, United States
| | - David Burkhart
- Center for Translational Medicine, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, Missoula, MT, United States
| | - Kendal Ryter
- Center for Translational Medicine, Missoula, MT, United States.,Department of Chemistry, University of Montana, Missoula, MT, United States
| | - Jay T Evans
- Center for Translational Medicine, Missoula, MT, United States.,Division of Biological Sciences, Missoula, MT, United States
| |
Collapse
|
21
|
Thakur A, Rodríguez-Rodríguez C, Saatchi K, Rose F, Esposito T, Nosrati Z, Andersen P, Christensen D, Häfeli UO, Foged C. Dual-Isotope SPECT/CT Imaging of the Tuberculosis Subunit Vaccine H56/CAF01: Induction of Strong Systemic and Mucosal IgA and T-Cell Responses in Mice Upon Subcutaneous Prime and Intrapulmonary Boost Immunization. Front Immunol 2018; 9:2825. [PMID: 30555488 PMCID: PMC6284049 DOI: 10.3389/fimmu.2018.02825] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/15/2018] [Indexed: 12/20/2022] Open
Abstract
Pulmonary tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), remains a global pandemic, despite the widespread use of the parenteral live attenuated Bacillus Calmette–Guérin (BCG) vaccine during the past decades. Mucosal administration of next generation TB vaccines has great potential, but developing a safe and efficacious mucosal vaccine is challenging. Hence, understanding the in vivo biodistribution and pharmacokinetics of mucosal vaccines is essential for shaping the desired immune response and for optimal spatiotemporal targeting of the appropriate effector cells in the lungs. A subunit vaccine consisting of the fusion antigen H56 (Ag85B-ESAT-6-Rv2660) and the liposome-based cationic adjuvant formulation (CAF01) confers efficient protection in preclinical animal models. In this study, we devise a novel immunization strategy for the H56/CAF01 vaccine, which comply with the intrapulmonary (i.pulmon.) route of immunization. We also describe a novel dual-isotope (111In/67Ga) radiolabeling approach, which enables simultaneous non-invasive and longitudinal SPECT/CT imaging and quantification of H56 and CAF01 upon parenteral prime and/or i.pulmon. boost immunization. Our results demonstrate that the vaccine is distributed evenly in the lungs, and there are pronounced differences in the pharmacokinetics of H56 and CAF01. We provide convincing evidence that the H56/CAF01 vaccine is not only well-tolerated when administered to the respiratory tract, but it also induces strong lung mucosal and systemic IgA and polyfunctional Th1 and Th17 responses after parenteral prime and i.pulmon. boost immunization. The study furthermore evaluate the application of SPECT/CT imaging for the investigation of vaccine biodistribution after parenteral and i.pulmon. immunization of mice.
Collapse
Affiliation(s)
- Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada.,Department of Physics and Astronomy, The University of British Columbia, Vancouver, BC, Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Fabrice Rose
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tullio Esposito
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Zeynab Nosrati
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Urs O Häfeli
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Wilkinson A, Lattmann E, Roces CB, Pedersen GK, Christensen D, Perrie Y. Lipid conjugation of TLR7 agonist Resiquimod ensures co-delivery with the liposomal Cationic Adjuvant Formulation 01 (CAF01) but does not enhance immunopotentiation compared to non-conjugated Resiquimod+CAF01. J Control Release 2018; 291:1-10. [PMID: 30291987 DOI: 10.1016/j.jconrel.2018.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 09/30/2018] [Accepted: 10/01/2018] [Indexed: 01/12/2023]
Abstract
Pattern recognition receptors, including the Toll-like receptors (TLRs), are important in the induction and activation of two critical arms of the host defence to pathogens and microorganisms: the rapid innate immune response (as characterised by the production of Th1 promoting cytokines and type 1 interferons) and the adaptive immune response. Through this activation, ligands and agonists of TLRs can enhance immunotherapeutic efficacy. Resiquimod is a small (water-soluble) agonist of the endosome-located Toll-like receptors 7 and 8 (TLR7/8). However due to its molecular attributes it rapidly distributes throughout the body after injection. To circumvent this, these TLR agonists can be incorporated within delivery systems, such as liposomes, to promote the co-delivery of both antigen and agonists to antigen presenting cells. In this present study, resiquimod has been chemically conjugated to a lipid to form a lipid-TLR7/8 agonist conjugate which can be incorporated within immunogenic cationic liposomes composed of dimethyldioctadecylammonium bromide (DDA) and the immunostimulatory glycolipid trehalose 6,6' - dibehenate (TDB). This DDA:TDB-TLR7/8 formulation offers similar vesicle characteristics to DDA:TDB (size and charge) and offers high retention of both resiquimod and the electrostatically adsorbed TB subunit antigen Ag85B-ESAT6-Rv2660c (H56). Following immunisation through the intramuscular (i.m.) route, these cationic DDA:TDB-TLR7/8 liposomes form a vaccine depot at the injection site. However, immunisation studies have shown that this biodistribution does not translate into notably increased antibody nor Th1 responses at the spleen and draining popliteal lymph node compared to DDA:TDB liposomes. This work demonstrates that the conjugation of TLR7/8 agonists to cationic liposomes can promote co-delivery but the immune responses stimulated do not merit the added complexity considerations of the formulation.
Collapse
Affiliation(s)
| | - Eric Lattmann
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Carla B Roces
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St, Glasgow G4 0RE, UK
| | - Gabriel K Pedersen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St, Glasgow G4 0RE, UK.
| |
Collapse
|
23
|
|
24
|
Spray dried cubosomes with ovalbumin and Quil-A as a nanoparticulate dry powder vaccine formulation. Int J Pharm 2018; 550:35-44. [DOI: 10.1016/j.ijpharm.2018.08.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 01/30/2023]
|
25
|
Abstract
Adjuvants are included in vaccine formulations to enhance the immunogenicity and efficacy of vaccines. MF59® is an oil-in-water emulsion adjuvant and licensed for use in pandemic and seasonal influenza vaccines in many countries. MF59 is safe and well tolerated in humans. MF59-adjuvanted vaccination spares vaccine dose and enhances hemagglutination inhibiting antibodies against homologous and heterologous influenza virus strains. The mechanisms of MF59 involve rapid induction of chemokines, inflammatory cytokines, recruiting multiple immune cells, uric acid and benign apoptosis of certain innate immune cells. The adjuvant effects of MF59 on generating vaccine-specific isotype-switched IgG antibodies, effector CD8 T cells, and protective immunity were retained even in a CD4-deficient condition by inducing effective immune-competent microenvironment with various innate and antigen presenting cells in a mouse model. CD4-independent adjuvant effects of MF59 might contribute to improving the vaccine efficacy in children, the elderly, and immunocompromised patients as well as in healthy adults. Further studies will be needed to broaden the use of MF59 in various vaccine antigens and populations as well as lead to better understanding of the action mechanisms of MF59 adjuvant.
Collapse
Affiliation(s)
- Eun-Ju Ko
- a Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences , Georgia State University , Atlanta , GA , USA.,b Vaccine Branch, Center for Cancer Research, National Cancer Institute , National Institutes of Health , Bethesda , MD , USA
| | - Sang-Moo Kang
- a Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences , Georgia State University , Atlanta , GA , USA
| |
Collapse
|
26
|
Kononova TE, Urazova OI, Novitskii VV, Esimova IE, Churina EG. Subpopulation Structure of IFNγ-Producing T Lymphocytes in Patients with Pulmonary Tuberculosis. Bull Exp Biol Med 2018; 165:311-314. [PMID: 30003413 DOI: 10.1007/s10517-018-4157-z] [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: 07/12/2017] [Indexed: 11/29/2022]
Abstract
The study of subpopulation structure of IFNγ-producing T cells in patients with pulmonary tuberculosis revealed a decrease in the number of CD3+ IFNγ+ cells against the background of significantly increased IFNγ secretion in vitro irrespective of the clinical form of the disease and drug sensitivity of M. tuberculosis, most strongly expressed in case of the disseminated tuberculosis. In patients with infiltrative drug-sensitive and drug-resistant pulmonary tuberculosis, increased number of Th1/Th17 lymphocytes (CD4+ IFNγ+IL-17A+) and, conversely, decreased number of blood γδT cells was detected.
Collapse
Affiliation(s)
- T E Kononova
- Department of Pathophysiology, Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - O I Urazova
- Department of Pathophysiology, Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - V V Novitskii
- Department of Pathophysiology, Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - I E Esimova
- Department of Pathophysiology, Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - E G Churina
- Department of Pathophysiology, Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.,Laboratory of Translational and Cellular Biomedicine, National Research Tomsk State University, Tomsk, Russia
| |
Collapse
|
27
|
Bobbala S, Gibson B, Gamble AB, McDowell A, Hook S. Poloxamer 407-chitosan grafted thermoresponsive hydrogels achieve synchronous and sustained release of antigen and adjuvant from single-shot vaccines. Immunol Cell Biol 2018; 96:656-665. [PMID: 29499080 DOI: 10.1111/imcb.12031] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/24/2018] [Accepted: 02/26/2018] [Indexed: 01/22/2023]
Abstract
Sustained-release vaccine delivery systems may enhance the immunogenicity of subunit vaccines and reduce the need for multiple vaccinations. The aim of this study was to develop a thermoresponsive hydrogel using poloxamer 407-chitosan (CP) grafted copolymer as a delivery system for single-shot sustained-release vaccines. The CP copolymer was synthesized using 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide and N-hydroxysuccinimide chemistry. The CP copolymer was a free flowing solution at ambient temperature and transformed rapidly into a gel at body temperature. The hydrogels were loaded with vaccine antigen and adjuvants or the vaccine components were encapsulated in poly (lactic-co-glycolic acid) nanoparticles in order to ensure synchronous release. The CP hydrogels were stable for up to 18 days in vitro. Release of both nanoparticles and the individual components was complete, with release of the individual components being modulated by incorporation into nanoparticles. In vivo, a single dose of CP hydrogel vaccine induced strong, long lasting, cellular and humoral responses that could protect against the development of tumors in a murine melanoma model.
Collapse
Affiliation(s)
- Sharan Bobbala
- School of Pharmacy, University of Otago, Dunedin, New Zealand.,Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Blake Gibson
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Allan B Gamble
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Arlene McDowell
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
28
|
Vono M, Eberhardt CS, Mohr E, Auderset F, Christensen D, Schmolke M, Coler R, Meinke A, Andersen P, Lambert PH, Mastelic-Gavillet B, Siegrist CA. Overcoming the Neonatal Limitations of Inducing Germinal Centers through Liposome-Based Adjuvants Including C-Type Lectin Agonists Trehalose Dibehenate or Curdlan. Front Immunol 2018. [PMID: 29541075 PMCID: PMC5835515 DOI: 10.3389/fimmu.2018.00381] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Neonates and infants are more vulnerable to infections and show reduced responses to vaccination. Consequently, repeated immunizations are required to induce protection and early life vaccines against major pathogens such as influenza are yet unavailable. Formulating antigens with potent adjuvants, including immunostimulators and delivery systems, is a demonstrated approach to enhance vaccine efficacy. Yet, adjuvants effective in adults may not meet the specific requirements for activating the early life immune system. Here, we assessed the neonatal adjuvanticity of three novel adjuvants including TLR4 (glucopyranosyl lipid adjuvant-squalene emulsion), TLR9 (IC31®), and Mincle (CAF01) agonists, which all induce germinal centers (GCs) and potent antibody responses to influenza hemagglutinin (HA) in adult mice. In neonates, a single dose of HA formulated into each adjuvant induced T follicular helper (TFH) cells. However, only HA/CAF01 elicited significantly higher and sustained antibody responses, engaging neonatal B cells to differentiate into GCs already after a single dose. Although antibody titers remained lower than in adults, HA-specific responses induced by a single neonatal dose of HA/CAF01 were sufficient to confer protection against influenza viral challenge. Postulating that the neonatal adjuvanticity of CAF01 may result from the functionality of the C-type lectin receptor (CLR) Mincle in early life we asked whether other C-type lectin agonists would show a similar neonatal adjuvanticity. Replacing the Mincle agonist trehalose 6,6′-dibehenate by Curdlan, which binds to Dectin-1, enhanced antibody responses through the induction of similar levels of TFH, GCs and bone marrow high-affinity plasma cells. Thus, specific requirements of early life B cells may already be met after a single vaccine dose using CLR-activating agonists, identified here as promising B cell immunostimulators for early life vaccines when included into cationic liposomes.
Collapse
Affiliation(s)
- Maria Vono
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Christiane Sigrid Eberhardt
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland.,WHO Collaborative Center for Vaccine Immunology, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| | - Elodie Mohr
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Floriane Auderset
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Dennis Christensen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Mirco Schmolke
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Rhea Coler
- Infectious Disease Research Institute, Seattle, WA, United States
| | | | - Peter Andersen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Paul-Henri Lambert
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Beatris Mastelic-Gavillet
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland.,WHO Collaborative Center for Vaccine Immunology, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| |
Collapse
|
29
|
Razazan A, Behravan J, Arab A, Barati N, Arabi L, Gholizadeh Z, Hatamipour M, Reza Nikpoor A, Momtazi-Borojeni AA, Mosaffa F, Ghahremani MH, Jaafari MR. Conjugated nanoliposome with the HER2/neu-derived peptide GP2 as an effective vaccine against breast cancer in mice xenograft model. PLoS One 2017; 12:e0185099. [PMID: 29045460 PMCID: PMC5646774 DOI: 10.1371/journal.pone.0185099] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 09/06/2017] [Indexed: 01/07/2023] Open
Abstract
One of the challenging issues in vaccine development is peptide and adjuvant delivery into target cells. In this study, we developed a vaccine and therapeutic delivery system to increase cytotoxic T lymphocyte (CTL) response against a breast cancer model overexpressing HER2/neu. Gp2, a HER2/neu-derived peptide, was conjugated to Maleimide-mPEG2000-DSPE micelles and post inserted into liposomes composed of DMPC, DMPG phospholipids, and fusogenic lipid dioleoylphosphatidylethanolamine (DOPE) containing monophosphoryl lipid A (MPL) adjuvant (DMPC-DMPG-DOPE-MPL-Gp2). BALB/c mice were immunized with different formulations and the immune response was evaluated in vitro and in vivo. ELISpot and intracellular cytokine analysis by flow cytometry showed that the mice vaccinated with Lip-DOPE-MPL-GP2 incited the highest number of IFN-γ+ in CD8+ cells and CTL response. The immunization led to lower tumor sizes and longer survival time compared to the other groups of mice immunized and treated with the Lip-DOPE-MPL-GP2 formulation in both prophylactic and therapeutic experiments. These results showed that co-formulation of DOPE and MPL conjugated with GP2 peptide not only induces high antitumor immunity but also enhances therapeutic efficacy in TUBO mice model. Lip-DOPE-MPL-GP2 formulation could be a promising vaccine and a therapeutic delivery system against HER2 positive cancers and merits further investigation.
Collapse
Affiliation(s)
- Atefeh Razazan
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran Iran
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Behravan
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Arab
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nastaran Barati
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Gholizadeh
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdi Hatamipour
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Reza Nikpoor
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi-Borojeni
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohamad Hosein Ghahremani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
30
|
Kumar S, Kesharwani SS, Kuppast B, Bakkari MA, Tummala H. Pathogen-mimicking vaccine delivery system designed with a bioactive polymer (inulin acetate) for robust humoral and cellular immune responses. J Control Release 2017; 261:263-274. [PMID: 28669593 PMCID: PMC6487847 DOI: 10.1016/j.jconrel.2017.06.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/26/2017] [Indexed: 01/24/2023]
Abstract
New and improved vaccines are needed against challenging diseases such as malaria, tuberculosis, Ebola, influenza, AIDS, and cancer. The majority of existing vaccine adjuvants lack the ability to significantly stimulate the cellular immune response, which is required to prevent the aforementioned diseases. This study designed a novel particulate based pathogen-mimicking vaccine delivery system (PMVDS) to target antigen-presenting-cells (APCs) such as dendritic cells. The uniqueness of PMVDS is that the polymer used to prepare the delivery system, Inulin Acetate (InAc), activates the innate immune system. InAc was synthesized from the plant polysaccharide, inulin. PMVDS provided improved and persistent antigen delivery to APCs as an efficient vaccine delivery system, and simultaneously, activated Toll-Like Receptor-4 (TLR-4) on APCs to release chemokine's/cytokines as an immune-adjuvant. Through this dual mechanism, PMVDS robustly stimulated both the humoral (>32 times of IgG1 levels vs alum) and the cell-mediated immune responses against the encapsulated antigen (ovalbumin) in mice. More importantly, PMVDS stimulated both cytotoxic T cells and natural killer cells of cell-mediated immunity to provide tumor (B16-ova-Melanoma) protection in around 40% of vaccinated mice and significantly delayed tumor progression in rest of the mice. PMVDS is a unique bio-active vaccine delivery technology with broader applications for vaccines against cancer and several intracellular pathogens, where both humoral and cellular immune responses are desired.
Collapse
Affiliation(s)
- Sunny Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy & Allied Health Professions, South Dakota State University, SAV # 255, Box 2202C, Brookings, SD 57007, USA
| | - Siddharth S Kesharwani
- Department of Pharmaceutical Sciences, College of Pharmacy & Allied Health Professions, South Dakota State University, SAV # 255, Box 2202C, Brookings, SD 57007, USA
| | - Bhimanna Kuppast
- Department of Pharmaceutical Sciences, College of Pharmacy & Allied Health Professions, South Dakota State University, SAV # 255, Box 2202C, Brookings, SD 57007, USA; Chicago College of Pharmacy, 555, 31st Street, Downers Grove, IL 60515, USA
| | - Mohammed Ali Bakkari
- Department of Pharmaceutical Sciences, College of Pharmacy & Allied Health Professions, South Dakota State University, SAV # 255, Box 2202C, Brookings, SD 57007, USA
| | - Hemachand Tummala
- Department of Pharmaceutical Sciences, College of Pharmacy & Allied Health Professions, South Dakota State University, SAV # 255, Box 2202C, Brookings, SD 57007, USA.
| |
Collapse
|
31
|
Counoupas C, Pinto R, Nagalingam G, Britton WJ, Petrovsky N, Triccas JA. Delta inulin-based adjuvants promote the generation of polyfunctional CD4 + T cell responses and protection against Mycobacterium tuberculosis infection. Sci Rep 2017; 7:8582. [PMID: 28819247 PMCID: PMC5561132 DOI: 10.1038/s41598-017-09119-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/19/2017] [Indexed: 01/16/2023] Open
Abstract
There is an urgent need for the rational design of safe and effective vaccines to protect against chronic bacterial pathogens such as Mycobacterium tuberculosis. Advax™ is a novel adjuvant based on delta inulin microparticles that enhances immunity with a minimal inflammatory profile and has entered human trials to protect against viral pathogens. In this report we determined if Advax displays broad applicability against important human pathogens by assessing protective immunity against infection with M. tuberculosis. The fusion protein CysVac2, comprising the M. tuberculosis antigens Ag85B (Rv1886c) and CysD (Rv1285) formulated with Advax provided significant protection in the lungs of M. tuberculosis-infected mice. Protection was associated with the generation of CysVac2-specific multifunctional CD4+ T cells (IFN-γ+TNF+IL-2+). Addition to Advax of the TLR9 agonist, CpG oligonucleotide (AdvaxCpG), improved both the immunogenicity and protective efficacy of CysVac2. Immunisation with CysVac2/AdvaxCpG resulted in heightened release of the chemoattractants, CXCL1, CCL3, and TNF, and rapid influx of monocytes and neutrophils to the site of vaccination, with pronounced early priming of CysVac2-specific CD4+ T cells. As delta inulin adjuvants have shown an excellent safety and tolerability profile in humans, CysVac2/AdvaxCpG is a strong candidate for further preclinical evaluation for progression to human trials.
Collapse
Affiliation(s)
- Claudio Counoupas
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia
| | - Rachel Pinto
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia
| | - Gayathri Nagalingam
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia
| | - Warwick J Britton
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia
| | - Nikolai Petrovsky
- Department of Endocrinology, Flinders University, Adelaide, Australia
- Vaxine Pty Ltd, Flinders Medical Centre, Adelaide, Australia
| | - James A Triccas
- Department of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia.
- Mycobacterial Research Program, Centenary Institute, Sydney, Australia.
| |
Collapse
|
32
|
O'Hagan DT, Friedland LR, Hanon E, Didierlaurent AM. Towards an evidence based approach for the development of adjuvanted vaccines. Curr Opin Immunol 2017; 47:93-102. [PMID: 28755542 DOI: 10.1016/j.coi.2017.07.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/12/2017] [Indexed: 01/29/2023]
Abstract
In the last two decades, several vaccines formulated with a new generation of adjuvants have been licensed or approved to target diseases such as influenza, hepatitis B, cervical cancer, and malaria. These new generation adjuvants appear to work by delivering a localized activation signal to the innate immune system, which in turn promotes antigen-specific adaptive immunity. Advances in understanding of the innate immune system together with high-throughput discovery of synthetic immune potentiators are now expanding the portfolio of new generation adjuvants available for evaluation. Meanwhile, omics and systems biology are providing molecular benchmarks or signatures to assess vaccine safety and effectiveness. This accumulating knowledge and experience raises the prospect that the future selection of the right antigen/adjuvant combination can be more evidence based and can speed up the clinical development program for new adjuvanted vaccines.
Collapse
Affiliation(s)
- Derek T O'Hagan
- GSK Vaccines, 14200 Shady Grove Road, Rockville, MD, USA. derek.t.o'
| | | | - Emmanuel Hanon
- GSK Vaccines, Rue de l'Institut 89, 1330 Rixensart, Belgium
| | | |
Collapse
|
33
|
Schmidt ST, Neustrup MA, Harloff-Helleberg S, Korsholm KS, Rades T, Andersen P, Christensen D, Foged C. Systematic Investigation of the Role of Surfactant Composition and Choice of oil: Design of a Nanoemulsion-Based Adjuvant Inducing Concomitant Humoral and CD4 + T-Cell Responses. Pharm Res 2017; 34:1716-1727. [PMID: 28516400 DOI: 10.1007/s11095-017-2180-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 05/11/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Induction of cell-mediated immune (CMI) responses is crucial for vaccine-mediated protection against difficult vaccine targets, e.g., Chlamydia trachomatis (Ct). Adjuvants are included in subunit vaccines to potentiate immune responses, but many marketed adjuvants stimulate predominantly humoral immune responses. Therefore, there is an unmet medical need for new adjuvants, which potentiate humoral and CMI responses. The purpose was to design an oil-in-water nanoemulsion adjuvant containing a synthetic CMI-inducing mycobacterial monomycoloyl glycerol (MMG) analogue to concomitantly induce humoral and CMI responses. METHODS The influence of emulsion composition was analyzed using a systematic approach. Three factors were varied: i) saturation of the oil phase, ii) type and saturation of the applied surfactant mixture, and iii) surfactant mixture net charge. RESULTS The emulsions were colloidally stable with a droplet diameter of 150-250 nm, and the zeta-potential correlated closely with the net charge of the surfactant mixture. Only cationic emulsions containing the unsaturated surfactant mixture induced concomitant humoral and CMI responses upon immunization of mice with a Ct antigen, and the responses were enhanced when squalene was applied as the oil phase. In contrast, emulsions with neutral and net negative zeta-potentials did not induce CMI responses. The saturation degree of the oil phase did not influence the adjuvanticity. CONCLUSION Cationic, MMG analogue-containing nanoemulsions are potential adjuvants for vaccines against pathogens for which both humoral and CMI responses are needed.
Collapse
Affiliation(s)
- Signe Tandrup Schmidt
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark.,Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300, Copenhagen S, Denmark
| | - Malene Aaby Neustrup
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark
| | - Stine Harloff-Helleberg
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark
| | - Karen Smith Korsholm
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300, Copenhagen S, Denmark
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark
| | - Peter Andersen
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300, Copenhagen S, Denmark
| | - Dennis Christensen
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, 2300, Copenhagen S, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark.
| |
Collapse
|
34
|
Billeskov R, Wang Y, Solaymani-Mohammadi S, Frey B, Kulkarni S, Andersen P, Agger EM, Sui Y, Berzofsky JA. Low Antigen Dose in Adjuvant-Based Vaccination Selectively Induces CD4 T Cells with Enhanced Functional Avidity and Protective Efficacy. THE JOURNAL OF IMMUNOLOGY 2017; 198:3494-3506. [PMID: 28348274 DOI: 10.4049/jimmunol.1600965] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Abstract
T cells with high functional avidity can sense and respond to low levels of cognate Ag, a characteristic that is associated with more potent responses against tumors and many infections, including HIV. Although an important determinant of T cell efficacy, it has proven difficult to selectively induce T cells of high functional avidity through vaccination. Attempts to induce high-avidity T cells by low-dose in vivo vaccination failed because this strategy simply gave no response. Instead, selective induction of high-avidity T cells has required in vitro culturing of specific T cells with low Ag concentrations. In this study, we combined low vaccine Ag doses with a novel potent cationic liposomal adjuvant, cationic adjuvant formulation 09, consisting of dimethyldioctadecylammonium liposomes incorporating two immunomodulators (monomycolyl glycerol analog and polyinosinic-polycytidylic acid) that efficiently induces CD4 Th cells, as well as cross-primes CD8 CTL responses. We show that vaccination with low Ag dose selectively primes CD4 T cells of higher functional avidity, whereas CD8 T cell functional avidity was unrelated to vaccine dose in mice. Importantly, CD4 T cells of higher functional avidity induced by low-dose vaccinations showed higher cytokine release per cell and lower inhibitory receptor expression (PD-1, CTLA-4, and the apoptosis-inducing Fas death receptor) compared with their lower-avidity CD4 counterparts. Notably, increased functional CD4 T cell avidity improved antiviral efficacy of CD8 T cells. These data suggest that potent adjuvants, such as cationic adjuvant formulation 09, render low-dose vaccination a feasible and promising approach for generating high-avidity T cells through vaccination.
Collapse
Affiliation(s)
- Rolf Billeskov
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; .,Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen S, DK-2300, Denmark; and
| | - Yichuan Wang
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Corporation, Frederick, MD 21702
| | - Shahram Solaymani-Mohammadi
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Blake Frey
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Shweta Kulkarni
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen S, DK-2300, Denmark; and
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen S, DK-2300, Denmark; and
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
35
|
Olafsdottir TA, Lindqvist M, Nookaew I, Andersen P, Maertzdorf J, Persson J, Christensen D, Zhang Y, Anderson J, Khoomrung S, Sen P, Agger EM, Coler R, Carter D, Meinke A, Rappuoli R, Kaufmann SHE, Reed SG, Harandi AM. Comparative Systems Analyses Reveal Molecular Signatures of Clinically tested Vaccine Adjuvants. Sci Rep 2016; 6:39097. [PMID: 27958370 PMCID: PMC5153655 DOI: 10.1038/srep39097] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/17/2016] [Indexed: 01/22/2023] Open
Abstract
A better understanding of the mechanisms of action of human adjuvants could inform a rational development of next generation vaccines for human use. Here, we exploited a genome wide transcriptomics analysis combined with a systems biology approach to determine the molecular signatures induced by four clinically tested vaccine adjuvants, namely CAF01, IC31, GLA-SE and Alum in mice. We report signature molecules, pathways, gene modules and networks, which are shared by or otherwise exclusive to these clinical-grade adjuvants in whole blood and draining lymph nodes of mice. Intriguingly, co-expression analysis revealed blood gene modules highly enriched for molecules with documented roles in T follicular helper (TFH) and germinal center (GC) responses. We could show that all adjuvants enhanced, although with different magnitude and kinetics, TFH and GC B cell responses in draining lymph nodes. These results represent, to our knowledge, the first comparative systems analysis of clinically tested vaccine adjuvants that may provide new insights into the mechanisms of action of human adjuvants.
Collapse
Affiliation(s)
- Thorunn A Olafsdottir
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Madelene Lindqvist
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Intawat Nookaew
- Department of Biology and Biological Engineering, Chalmers, University of Technology, Gothenburg, Sweden.,Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Jeroen Maertzdorf
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Josefine Persson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Yuan Zhang
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jenna Anderson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sakda Khoomrung
- Department of Biology and Biological Engineering, Chalmers, University of Technology, Gothenburg, Sweden
| | - Partho Sen
- Department of Biology and Biological Engineering, Chalmers, University of Technology, Gothenburg, Sweden
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Rhea Coler
- Infectious Disease Research Institute, Seattle, Washington, USA
| | - Darrick Carter
- Infectious Disease Research Institute, Seattle, Washington, USA
| | - Andreas Meinke
- Valneva Austria GmbH, Campus Vienna Biocenter, Vienna, Austria
| | | | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, Washington, USA
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
36
|
van Haren SD, Dowling DJ, Foppen W, Christensen D, Andersen P, Reed SG, Hershberg RM, Baden LR, Levy O. Age-Specific Adjuvant Synergy: Dual TLR7/8 and Mincle Activation of Human Newborn Dendritic Cells Enables Th1 Polarization. THE JOURNAL OF IMMUNOLOGY 2016; 197:4413-4424. [PMID: 27793997 DOI: 10.4049/jimmunol.1600282] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 09/26/2016] [Indexed: 02/07/2023]
Abstract
Due to functionally distinct cell-mediated immunity, newborns and infants are highly susceptible to infection with intracellular pathogens. Indeed, neonatal Ag-presenting dendritic cells (DCs) demonstrate impaired Th1 responses to many candidate adjuvants, including most TLR agonists (TLRAs). Combination adjuvantation systems may provide enhanced immune activation but have typically been developed without regard to the age of the target population. We posited that distinct combinations of TLRAs and C-type lectin receptor agonists may enhance Th1 responses of newborn DCs. TLRA/C-type lectin receptor agonist combinations were screened for enhancement of TNF production by human newborn and adult monocyte-derived DCs cultured in 10% autologous plasma or in newborn cord, infant, adult, and elderly whole blood. Monocyte-derived DC activation was characterized by targeted gene expression analysis, caspase-1 and NF-κB studies, cytokine multiplex and naive autologous CD4+ T cell activation. Dual activation of newborn DCs via the C-type lectin receptor, macrophage-inducible C-type lectin (trehalose-6,6-dibehenate), and TLR7/8 (R848) greatly enhanced caspase-1 and NF-κB activation, Th1 polarizing cytokine production and autologous Th1 polarization. Combined activation via TLR4 (glycopyranosyl lipid adjuvant aqueous formulation) and Dectin-1 (β-glucan peptide) acted synergistically in newborns and adults, but to a lesser extent. The degree of synergy varied dramatically with age, and was the greatest in newborns and infants with less synergy in adults and elders. Overall, combination adjuvant systems demonstrate markedly different immune activation with age, with combined DC activation via Macrophage-inducible C-type lectin and TLR7/8 representing a novel approach to enhance the efficacy of early-life vaccines.
Collapse
Affiliation(s)
- Simon D van Haren
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA 02115.,Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115.,Harvard Medical School, Boston, MA 02115
| | - David J Dowling
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA 02115.,Harvard Medical School, Boston, MA 02115
| | - Willemina Foppen
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA 02115.,Harvard Medical School, Boston, MA 02115
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, DK-2300, Denmark
| | - Peter Andersen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115.,Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, DK-2300, Denmark
| | - Steven G Reed
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115.,Infectious Disease Research Institute, Seattle, WA 98102
| | | | - Lindsey R Baden
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115.,Division of Infectious Diseases, Brigham and Women's Hospital Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Ofer Levy
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA 02115; .,Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115.,Harvard Medical School, Boston, MA 02115
| |
Collapse
|
37
|
Liu Y, Chen S, Pan B, Guan Z, Yang Z, Duan L, Cai H. A subunit vaccine based on rH-NS induces protection against Mycobacterium tuberculosis infection by inducing the Th1 immune response and activating macrophages. Acta Biochim Biophys Sin (Shanghai) 2016; 48:909-922. [PMID: 27563010 DOI: 10.1093/abbs/gmw078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/07/2016] [Indexed: 11/12/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a Gram-positive pathogen which causes tuberculosis in both animals and humans. All tested rH-NS formulations induced a specific Th1 response, as indicated by increased production of interferon γ (IFN-γ) and interleukin 2 (IL-2) by lymphocytes in the spleen of mice which were immunized with rH-NS alone or with rH-NS and the adjuvant cyclic GMP-AMP (cGAMP). Serum from mice immunized with rH-NS with or without adjuvant also had higher levels of IL-12p40 and TNF-α, compared with those from control mice immunized with phosphate-buffered saline. Both vaccines increased protective efficacy in mice which were challenged with Mtb H37Rv, as measured by reduced relative CFU counts in the lungs. We found that rH-NS induced the production of TNF-α, IL-6, and IL-12p40, which relied on the activation of mitogen-activated protein kinases by stimulating the rapid phosphorylation of ERK1/2, p38, and JNK, and on the activation of transcription factor NF-κB in macrophages. Additionally, we also found that rH-NS could interact with TLR2 but not TLR4 in pull-down assays. The rH-NS-induced cytokine production from TLR2-silenced RAW264.7 cells was lower than that from BALB/c macrophages. Prolonged exposure (>24 h) of RAW264.7 cells to rH-NS resulted in a significant enhancement in IFN-γ-induced MHC II expression, which was not found in shTLR2-treated RAW264.7 cells. These results suggest that rH-NS is a TLR2 agonist which induces the production of cytokines by macrophages and up-regulates macrophage function.
Collapse
Affiliation(s)
- Yuan Liu
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
| | - Suting Chen
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing 101149, China
| | - Bowen Pan
- State Key Laboratory of Natural and Biomimetic Drugs, Peking Universtiy Health Science Center, Beijing 100191, China
| | - Zhu Guan
- State Key Laboratory of Natural and Biomimetic Drugs, Peking Universtiy Health Science Center, Beijing 100191, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking Universtiy Health Science Center, Beijing 100191, China
| | - Linfei Duan
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
| | - Hong Cai
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
38
|
Bobbala S, Hook S. Is There an Optimal Formulation and Delivery Strategy for Subunit Vaccines? Pharm Res 2016; 33:2078-97. [DOI: 10.1007/s11095-016-1979-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/21/2016] [Indexed: 12/16/2022]
|
39
|
Vaccination in early life: standing up to the challenges. Curr Opin Immunol 2016; 41:1-8. [PMID: 27104290 DOI: 10.1016/j.coi.2016.04.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/16/2016] [Accepted: 04/06/2016] [Indexed: 01/18/2023]
Abstract
The challenge for any vaccine design is to elicit protective humoral and/or cytotoxic immunity against life threatening pathogens while remaining innocuous. Neonatal vaccinology faces additional challenges linked to intrinsic peculiarities of the innate and adaptive neonatal immune system. These include anti-inflammatory rather than pro-inflammatory responses to innate signals, preferential Th2 differentiation limiting the induction of Th1 and cytotoxic responses, trends to immunoregulatory responses and weak plasma cell and germinal centre B cell responses. Recent progresses in our understanding of the molecular bases of these physiological peculiarities and of the mode of action of novel adjuvants open new opportunities to design vaccine formulations and immunization strategies better adapted to the early life period.
Collapse
|
40
|
Aboutorabian S, Hakimi J, Boudet F, Montano S, Dookie A, Roque C, Ausar SF, Rahman N, Brookes RH. A high ratio of IC31(®) adjuvant to antigen is necessary for H4 TB vaccine immunomodulation. Hum Vaccin Immunother 2016; 11:1449-55. [PMID: 25997147 PMCID: PMC4514381 DOI: 10.1080/21645515.2015.1023970] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
A tuberculosis (TB) vaccine consisting of a recombinant fusion protein (H4) and a novel TLR9 adjuvant (IC31) is in clinical development. To better understand the H4-IC31 ratio, we measured the binding capacity of IC31 for H4 protein and immunized mice with formulations that contained limiting to excess ratios of IC31 to H4. An immunomodulated H4-specific IFNγ response was only observed when IC31 was present in excess of H4. Since TLR expression is species-specific and the vaccine is intended to boost BCG-primed immunity, we questioned whether data in mice would translate to humans. To address this question, we used the fresh human Whole Blood (hWB) recovered from BCG-vaccinated subjects to screen H4-IC31 formulations. We found IC31 modulation in hWB to be quite distinct from the TLR4-Adjuvant. Unlike TLR4-Adjuvant, IC31 formulations did not induce the pro-inflammatory cytokine TNFα, but modulated a robust H4-specific IFNγ response after 12 d of culture. We then re-stimulated the fresh hWB of 5 BCG-primed subjects with formulations that had excess or limiting IC31 binding for H4 protein and again found that an immunomodulated H4-specific IFNγ response needed an excess of IC31. Finally, we monitored the zeta (ζ) potential of H4-IC31 formulations and found that the overall charge of H4-IC31 particles changes from negative to positive once IC31 is in greater than 9-fold excess. Using two diverse yet mutually supportive approaches, we confirm the need for an excess of IC31 adjuvant in H4 TB vaccine formulations and suggest surface potential may be an important factor.
Collapse
Key Words
- BCG, Bacillus Calmette-Guérin
- H4, TB, Antigen
- IC31 adjuvant modulation
- IC31®, Valneva Adjuvant consisting of KLK and ODN1a
- IFNγ, Interferon gamma
- KLK, Antimicrobial peptide H-KLKL5KLK-OH
- ODN1a, Oligodeoxynucleotide
- TB, Tuberculosis
- TLR, Toll-like Receptor
- TLR4, Toll-like Receptor 4
- TLR4-Adjuvant, TLR4A combined with an aluminum salt adjuvant
- TLR4A, TLR4 Agonist
- TLR9, Toll-like Receptor 9
- TNFα, Tumor Necrosis Factor alpha
- WBA
- functionality
- hWB, Human Whole Blood
- tuberculosis
- vaccine
Collapse
|
41
|
Liposome-Based Adjuvants for Subunit Vaccines: Formulation Strategies for Subunit Antigens and Immunostimulators. Pharmaceutics 2016; 8:pharmaceutics8010007. [PMID: 26978390 PMCID: PMC4810083 DOI: 10.3390/pharmaceutics8010007] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 11/25/2022] Open
Abstract
The development of subunit vaccines has become very attractive in recent years due to their superior safety profiles as compared to traditional vaccines based on live attenuated or whole inactivated pathogens, and there is an unmet medical need for improved vaccines and vaccines against pathogens for which no effective vaccines exist. The subunit vaccine technology exploits pathogen subunits as antigens, e.g., recombinant proteins or synthetic peptides, allowing for highly specific immune responses against the pathogens. However, such antigens are usually not sufficiently immunogenic to induce protective immunity, and they are often combined with adjuvants to ensure robust immune responses. Adjuvants are capable of enhancing and/or modulating immune responses by exposing antigens to antigen-presenting cells (APCs) concomitantly with conferring immune activation signals. Few adjuvant systems have been licensed for use in human vaccines, and they mainly stimulate humoral immunity. Thus, there is an unmet demand for the development of safe and efficient adjuvant systems that can also stimulate cell-mediated immunity (CMI). Adjuvants constitute a heterogeneous group of compounds, which can broadly be classified into delivery systems or immunostimulators. Liposomes are versatile delivery systems for antigens, and they can carefully be customized towards desired immune profiles by combining them with immunostimulators and optimizing their composition, physicochemical properties and antigen-loading mode. Immunostimulators represent highly diverse classes of molecules, e.g., lipids, nucleic acids, proteins and peptides, and they are ligands for pattern-recognition receptors (PRRs), which are differentially expressed on APC subsets. Different formulation strategies might thus be required for incorporation of immunostimulators and antigens, respectively, into liposomes, and the choice of immunostimulator should ideally be based on knowledge regarding the specific PRR expression profile of the target APCs. Here, we review state-of-the-art formulation approaches employed for the inclusion of immunostimulators and subunit antigens into liposome dispersion and their optimization towards robust vaccine formulations.
Collapse
|
42
|
Bobbala S, Tamboli V, McDowell A, Mitra AK, Hook S. Novel Injectable Pentablock Copolymer Based Thermoresponsive Hydrogels for Sustained Release Vaccines. AAPS JOURNAL 2015; 18:261-9. [PMID: 26589309 DOI: 10.1208/s12248-015-9843-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023]
Abstract
The need for multiple vaccinations to enhance the immunogenicity of subunit vaccines may be reduced by delivering the vaccine over an extended period of time. Here, we report two novel injectable pentablock copolymer based thermoresponsive hydrogels made of polyethyleneglycol-polycaprolactone-polylactide-polycaprolactone-polyethyleneglycol (PEG-PCL-PLA-PCL-PEG) with varying ratios of polycaprolactone (PCL) and polylactide (PLA), as single shot sustained release vaccines. Pentablock copolymer hydrogels were loaded with vaccine-encapsulated poly lactic-co-glycolic acid nanoparticles (PLGA-NP) or with the soluble vaccine components. Incorporation of PLGA-NP into the thermoresponsive hydrogels increased the complex viscosity of the gels, lowered the gelation temperature, and minimized the burst release of antigen and adjuvants. The two pentablock hydrogels stimulated both cellular and humoral responses. The addition of PLGA-NP to the hydrogels sustained immune responses for up to 49 days. The polymer with a higher ratio of PCL to PLA formed a more rigid gel, induced stronger immune responses, and stimulated effective anti-tumor responses in a prophylactic melanoma tumor model.
Collapse
Affiliation(s)
- Sharan Bobbala
- School of Pharmacy, University of Otago, Dunedin, 9054, New Zealand
| | - Viral Tamboli
- School of Pharmacy, UMKC, Kansas City, Missouri, USA
| | - Arlene McDowell
- School of Pharmacy, University of Otago, Dunedin, 9054, New Zealand
| | - Ashim K Mitra
- School of Pharmacy, UMKC, Kansas City, Missouri, USA
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, 9054, New Zealand.
| |
Collapse
|
43
|
Intranasal vaccination with killed Leishmania amazonensis promastigotes antigen (LaAg) associated with CAF01 adjuvant induces partial protection in BALB/c mice challenged with Leishmania (infantum) chagasi. Parasitology 2015; 142:1640-6. [DOI: 10.1017/s0031182015001250] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARYThe CAF01 adjuvant has previously been shown to be safe for human use and to be a potent adjuvant for several vaccine antigens. In the present work, we sought to optimize the Leishmania amazonensis antigens (LaAg) intranasal vaccine in an attempt to enhance the protective immune responses against Leishmania (infantum) chagasi by using the CAF01 association. LaAg/CAF01 vaccinated mice that were challenged 15 days after booster dose with L. (infantum) chagasi showed a significant reduction in their parasite burden in both the spleen and liver, which is associated with an increase in specific production of IFN-γ and nitrite, and a decrease in IL-4 production. In addition, LaAg/CAF01 intranasal delivery was able to increase lymphoproliferative immune responses after parasite antigen recall. These results suggest the feasibility of using the intranasal route for the delivery of crude antigens and of a human-compatible adjuvant against visceral leishmaniasis.
Collapse
|
44
|
Even MP, Bobbala S, Kooi KL, Hook S, Winter G, Engert J. Impact of implant composition of twin-screw extruded lipid implants on the release behavior. Int J Pharm 2015; 493:102-10. [DOI: 10.1016/j.ijpharm.2015.06.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 12/16/2022]
|
45
|
Long-term persistence of immunity induced by OVA-coupled gas-filled microbubble vaccination partially protects mice against infection by OVA-expressing Listeria. Biomaterials 2015; 57:153-60. [DOI: 10.1016/j.biomaterials.2015.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/02/2015] [Accepted: 04/03/2015] [Indexed: 02/06/2023]
|
46
|
Neeland MR, Elhay MJ, Powell DR, Rossello FJ, Meeusen ENT, de Veer MJ. Transcriptional profile in afferent lymph cells following vaccination with liposomes incorporating CpG. Immunology 2015; 144:518-529. [PMID: 25308816 DOI: 10.1111/imm.12401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/14/2014] [Accepted: 10/02/2014] [Indexed: 12/17/2022] Open
Abstract
Vaccine formulations incorporating innate immune stimulants are highly immunogenic; however, the biological signals that originate in the peripheral tissues at the site of injection and are transmitted to the local lymph node to induce immunity remain unclear. By directly cannulating the ovine afferent lymphatic vessels, we have previously shown that it takes 72 hr for mature antigen-loaded dendritic cells and monocytes to appear within afferent lymph following injection of a liposomal formulation containing the Toll-like receptor ligand CpG. In this present study, we characterize the global transcriptional signatures at this time-point in ovine afferent lymph cells as they migrate from the injection site into the lymphatics following vaccination with a liposome antigen formulation incorporating CpG. We show that at 72 hr post vaccination, liposomes alone induce no changes in gene expression and inflammatory profiles within afferent lymph; however, the incorporation of CpG drives interferon, antiviral and cytotoxic gene programmes. This study also measures the expression of key genes within individual cell types in afferent lymph. Antiviral gene signatures are most prominent in lymphocytes, which may play a significant and unexpected role in sustaining the immune response to vaccination at the site of injection. These findings provide a comprehensive analysis of the in vivo immunological pathways that connect the injection site with the local draining lymph node following vaccination.
Collapse
Affiliation(s)
- Melanie R Neeland
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Clayton, Vic., Australia
| | - Martin J Elhay
- Zoetis Research and Manufacturing Australia P/L, Parkville, Vic., Australia
| | - David R Powell
- Victorian Bioinformatics Consortium, Monash University, Clayton, Vic., Australia.,Victorian Life Sciences Computation Initiative, Life Sciences Computation Centre, Carlton, Vic., Australia
| | - Fernando J Rossello
- Victorian Bioinformatics Consortium, Monash University, Clayton, Vic., Australia.,Victorian Life Sciences Computation Initiative, Life Sciences Computation Centre, Carlton, Vic., Australia
| | - Els N T Meeusen
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Clayton, Vic., Australia.,Department of Microbiology, Monash University, Clayton, Vic., Australia
| | - Michael J de Veer
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Clayton, Vic., Australia
| |
Collapse
|
47
|
Kastner E, Schmidt ST, Wilkinson A, Christensen D, Perrie Y. The Application of Liposomes as Vaccine Adjuvants. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2015. [DOI: 10.1007/978-1-4939-1417-3_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
48
|
Modified thermoresponsive Poloxamer 407 and chitosan sol-gels as potential sustained-release vaccine delivery systems. Eur J Pharm Biopharm 2014; 89:74-81. [PMID: 25481034 DOI: 10.1016/j.ejpb.2014.11.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/19/2014] [Accepted: 11/26/2014] [Indexed: 01/06/2023]
Abstract
Thermoresponsive, particle-loaded, Poloxamer 407 (P407)-Pluronic-R® (25R4) or chitosan-methyl cellulose (MC) formulations were developed as single-dose, sustained release vaccines. The sol-gels, loaded either with a particulate vaccine (cubosomes) or soluble antigen (ovalbumin) and adjuvants (Quil A and monophosphoryl lipid A), were free-flowing liquids at room temperature and formed stable gels at physiological temperatures. Rheological results showed that both systems meet the criteria of being thermoresponsive gels. The P407-25R4 sol-gels did not significantly sustain the release of antigen in vivo while the chitosan-MC sol-gels sustained the release of antigen up to at least 14 days after administration. The chitosan-MC sol-gels stimulated both cellular and humoral responses. The inclusion of cubosomes in the sol-gels did not provide a definitive beneficial effect. Further analysis of the formulations with small-angle X-ray scattering (SAXS) revealed that while cubosomes were stable in chitosan-MC gels they were not stable in P407-25R4 formulations. The reason for the mixed response to cubosome-loaded vehicles requires more investigation, however it appears that the cubosomes did not facilitate synchronous vaccine release and may in fact retard release, reducing efficacy in some cases. From these results, chitosan-MC sol-gels show potential as sustained release vaccine delivery systems, as compared to the P407-25R4 system that had a limited ability to sustain antigen release.
Collapse
|
49
|
Schwendener RA. Liposomes as vaccine delivery systems: a review of the recent advances. THERAPEUTIC ADVANCES IN VACCINES 2014; 2:159-82. [PMID: 25364509 DOI: 10.1177/2051013614541440] [Citation(s) in RCA: 329] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Liposomes and liposome-derived nanovesicles such as archaeosomes and virosomes have become important carrier systems in vaccine development and the interest for liposome-based vaccines has markedly increased. A key advantage of liposomes, archaeosomes and virosomes in general, and liposome-based vaccine delivery systems in particular, is their versatility and plasticity. Liposome composition and preparation can be chosen to achieve desired features such as selection of lipid, charge, size, size distribution, entrapment and location of antigens or adjuvants. Depending on the chemical properties, water-soluble antigens (proteins, peptides, nucleic acids, carbohydrates, haptens) are entrapped within the aqueous inner space of liposomes, whereas lipophilic compounds (lipopeptides, antigens, adjuvants, linker molecules) are intercalated into the lipid bilayer and antigens or adjuvants can be attached to the liposome surface either by adsorption or stable chemical linking. Coformulations containing different types of antigens or adjuvants can be combined with the parameters mentioned to tailor liposomal vaccines for individual applications. Special emphasis is given in this review to cationic adjuvant liposome vaccine formulations. Examples of vaccines made with CAF01, an adjuvant composed of the synthetic immune-stimulating mycobacterial cordfactor glycolipid trehalose dibehenate as immunomodulator and the cationic membrane forming molecule dimethyl dioctadecylammonium are presented. Other vaccines such as cationic liposome-DNA complexes (CLDCs) and other adjuvants like muramyl dipeptide, monophosphoryl lipid A and listeriolysin O are mentioned as well. The field of liposomes and liposome-based vaccines is vast. Therefore, this review concentrates on recent and relevant studies emphasizing current reports dealing with the most studied antigens and adjuvants, and pertinent examples of vaccines. Studies on liposome-based veterinary vaccines and experimental therapeutic cancer vaccines are also summarized.
Collapse
Affiliation(s)
- Reto A Schwendener
- Institute of Molecular Cancer Research, Laboratory of Liposome Research, University of Zurich, Winterthurerstrasse 190, Zurich, 8057, Switzerland
| |
Collapse
|
50
|
van Dissel JT, Joosten SA, Hoff ST, Soonawala D, Prins C, Hokey DA, O'Dee DM, Graves A, Thierry-Carstensen B, Andreasen LV, Ruhwald M, de Visser AW, Agger EM, Ottenhoff THM, Kromann I, Andersen P. A novel liposomal adjuvant system, CAF01, promotes long-lived Mycobacterium tuberculosis-specific T-cell responses in human. Vaccine 2014; 32:7098-107. [PMID: 25454872 DOI: 10.1016/j.vaccine.2014.10.036] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 09/08/2014] [Accepted: 10/17/2014] [Indexed: 02/08/2023]
Abstract
Here, we report on a first-in-man trial where the tuberculosis (TB) vaccine Ag85B-ESAT-6 (H1) was adjuvanted with escalating doses of a novel liposome adjuvant CAF01. On their own, protein antigens cannot sufficiently induce immune responses in humans, and require the addition of an adjuvant system to ensure appropriate delivery and concomitant immune activation. To date no approved adjuvants are available for induction of cellular immunity, which seems essential for a number of vaccines, including vaccines against TB. We vaccinated four groups of human volunteers: a non-adjuvanted H1 group, followed by three groups with escalating doses of CAF01-adjuvanted H1 vaccine. All subjects were vaccinated at 0 and 8 weeks and followed up for 150 weeks. Vaccination did not cause local or systemic adverse effects besides transient soreness at the injection site. Two vaccinations elicited strong antigen-specific T-cell responses which persisted after 150 weeks follow-up, indicating the induction of a long-lasting memory response in the vaccine recipients. These results show that CAF01 is a safe and tolerable, Th1-inducing adjuvant for human TB vaccination trials and for vaccination studies in general where cellular immunity is required.
Collapse
Affiliation(s)
- Jaap T van Dissel
- Leiden University Medical Center (LUMC), Department of Infectious Diseases, Leiden, The Netherlands.
| | - Simone A Joosten
- Leiden University Medical Center (LUMC), Department of Infectious Diseases, Leiden, The Netherlands
| | - Søren T Hoff
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, Copenhagen 2300s, Denmark
| | - Darius Soonawala
- Leiden University Medical Center (LUMC), Department of Infectious Diseases, Leiden, The Netherlands
| | - Corine Prins
- Leiden University Medical Center (LUMC), Department of Infectious Diseases, Leiden, The Netherlands
| | | | | | | | - Birgit Thierry-Carstensen
- Statens Serum Institut, Department of Vaccine Development, Artillerivej 5, Copenhagen 2300s, Denmark
| | - Lars V Andreasen
- Statens Serum Institut, Department of Vaccine Development, Artillerivej 5, Copenhagen 2300s, Denmark
| | - Morten Ruhwald
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, Copenhagen 2300s, Denmark
| | - Adriëtte W de Visser
- Leiden University Medical Center (LUMC), Department of Infectious Diseases, Leiden, The Netherlands
| | - Else Marie Agger
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, Copenhagen 2300s, Denmark
| | - Tom H M Ottenhoff
- Leiden University Medical Center (LUMC), Department of Infectious Diseases, Leiden, The Netherlands
| | - Ingrid Kromann
- Statens Serum Institut, Department of Vaccine Development, Artillerivej 5, Copenhagen 2300s, Denmark
| | - Peter Andersen
- Statens Serum Institut, Department of Infectious Disease Immunology, Artillerivej 5, Copenhagen 2300s, Denmark.
| |
Collapse
|