1
|
Ferguson MR, Delgado KN, McBride S, Orbe IC, La Vake CJ, Caimano MJ, Mendez Q, Moraes TF, Schryvers AB, Moody MA, Radolf JD, Weiner MP, Hawley KL. Use of Epivolve phage display to generate a monoclonal antibody with opsonic activity directed against a subdominant epitope on extracellular loop 4 of Treponema pallidum BamA (TP0326). Front Immunol 2023; 14:1222267. [PMID: 37675118 PMCID: PMC10478084 DOI: 10.3389/fimmu.2023.1222267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/19/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction Syphilis, a sexually transmitted infection caused by the spirochete Treponema pallidum (Tp), is resurging globally. Tp's repertoire of outer membrane proteins (OMPs) includes BamA (β-barrel assembly machinery subunit A/TP0326), a bipartite protein consisting of a 16-stranded β-barrel with nine extracellular loops (ECLs) and five periplasmic POTRA (polypeptide transport-associated) domains. BamA ECL4 antisera promotes internalization of Tp by rabbit peritoneal macrophages. Methods Three overlapping BamA ECL4 peptides and a two-stage, phage display strategy, termed "Epivolve" (for epitope evolution) were employed to generate single-chain variable fragments (scFvs). Additionally, antisera generated by immunizing mice and rabbits with BamA ECL4 displayed by a Pyrococcus furiosus thioredoxin scaffold (PfTrxBamA/ECL4). MAbs and antisera reactivities were evaluated by immunoblotting and ELISA. A comparison of murine and rabbit opsonophagocytosis assays was conducted to evaluate the functional ability of the Abs (e.g., opsonization) and validate the mouse assay. Sera from Tp-infected mice (MSS) and rabbits (IRS) were evaluated for ECL4-specific Abs using PfTrxBamA/ECL4 and overlapping ECL4 peptides in immunoblotting and ELISA assays. Results Each of the five mAbs demonstrated reactivity by immunoblotting and ELISA to nanogram amounts of PfTrxBamA/ECL4. One mAb, containing a unique amino acid sequence in both the light and heavy chains, showed activity in the murine opsonophagocytosis assay. Mice and rabbits hyperimmunized with PfTrxBamA/ECL4 produced opsonic antisera that strongly recognized the ECL presented in a heterologous scaffold and overlapping ECL4 peptides, including S2. In contrast, Abs generated during Tp infection of mice and rabbits poorly recognized the peptides, indicating that S2 contains a subdominant epitope. Discussion Epivolve produced mAbs target subdominant opsonic epitopes in BamA ECL4, a top syphilis vaccine candidate. The murine opsonophagocytosis assay can serve as an alternative model to investigate the opsonic potential of vaccinogens. Detailed characterization of BamA ECL4-specific Abs provided a means to dissect Ab responses elicited by Tp infection.
Collapse
Affiliation(s)
- Mary R. Ferguson
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | | | | | - Isabel C. Orbe
- Department of Pediatrics, UConn Health, Farmington, CT, United States
| | - Carson J. La Vake
- Department of Pediatrics, UConn Health, Farmington, CT, United States
| | - Melissa J. Caimano
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, United States
| | - Qiana Mendez
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | - Trevor F. Moraes
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Anthony B. Schryvers
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - M. Anthony Moody
- Duke Human Vaccine Institute, Durham, NC, United States
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, United States
| | - Justin D. Radolf
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, United States
- Department of Immunology, UConn Health, Farmington, CT, United States
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
| | - Michael P. Weiner
- Department of Molecular Sciences, Abbratech, Branford, CT, United States
| | - Kelly L. Hawley
- Department of Medicine, UConn Health, Farmington, CT, United States
- Department of Pediatrics, UConn Health, Farmington, CT, United States
- Department of Immunology, UConn Health, Farmington, CT, United States
- Division of Infectious Diseases and Immunology, Connecticut Children’s, Hartford, CT, United States
| |
Collapse
|
2
|
Hendy DA, Johnson-Weaver BT, Batty CJ, Bachelder EM, Abraham SN, Staats HF, Ainslie KM. Delivery of small molecule mast cell activators for West Nile Virus vaccination using acetalated dextran microparticles. Int J Pharm 2023; 634:122658. [PMID: 36731641 PMCID: PMC9975031 DOI: 10.1016/j.ijpharm.2023.122658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023]
Abstract
Recently, there has been increasing interest in the activation of mast cells to promote vaccine efficacy. Several mast cell activating (MCA) compounds have been reported such as M7 and Compound 48/80 (C48/80). While these MCAs have been proven to be efficacious vaccine adjuvants, their translatability is limited by batch-to-batch variability, challenging large-scale manufacturing, and poor in vivo stability for the M7 peptide. Due to this, high throughput screening was performed to identify small molecule MCAs. Several potent MCAs were identified via this screening, but the in vivo translatability of the compounds was limited due to their poor aqueous solubility. To enhance the delivery of these MCAs we encapsulated them in acetalated dextran (Ace-DEX) microparticles (MPs). We have previously utilized Ace-DEX MPs for vaccine delivery due to their passive targeting to phagocytic cells, acid sensitivity, and tunable degradation. Four different MCA loaded MPs were combined with West Nile Virus Envelope III protein (EDIII) and their vaccine adjuvant activities were compared in vivo. MPs containing the small molecule MCA ST101036 produced the highest anti-EDIII IgG titers of all the MCAs tested. Further, ST101036 MPs produced higher titers than ST101036 formulated with PEG as a cosolvent which highlights the benefit of Ace-DEX MPs over a conventional formulation technique. Finally, in a mouse model of West Nile Virus infection ST101036 MPs produced similar survival to soluble M7 (80-90%). Overall, these data show that ST101036 MPs produce a robust antibody response against EDIII and survival emphasizing the benefits of using Ace-DEX as a delivery platform for the poorly soluble ST101036.
Collapse
Affiliation(s)
- Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | | | - Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | | | - Herman F Staats
- Department of Pathology, Duke University, USA; Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Ontiveros-Padilla L, Batty CJ, Hendy DA, Pena ES, Roque JA, Stiepel RT, Carlock MA, Simpson SR, Ross TM, Abraham SN, Staats HF, Bachelder EM, Ainslie KM. Development of a broadly active influenza intranasal vaccine adjuvanted with self-assembled particles composed of mastoparan-7 and CpG. Front Immunol 2023; 14:1103765. [PMID: 37033992 PMCID: PMC10081679 DOI: 10.3389/fimmu.2023.1103765] [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: 11/20/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Currently licensed vaccine adjuvants offer limited mucosal immunity, which is needed to better combat respiratory infections such as influenza. Mast cells (MCs) are emerging as a target for a new class of mucosal vaccine adjuvants. Here, we developed and characterized a nanoparticulate adjuvant composed of an MC activator [mastoparan-7 (M7)] and a TLR ligand (CpG). This novel nanoparticle (NP) adjuvant was co-formulated with a computationally optimized broadly reactive antigen (COBRA) for hemagglutinin (HA), which is broadly reactive against influenza strains. M7 was combined at different ratios with CpG and tested for in vitro immune responses and cytotoxicity. We observed significantly higher cytokine production in dendritic cells and MCs with the lowest cytotoxicity at a charge-neutralizing ratio of nitrogen/phosphate = 1 for M7 and CpG. This combination formed spherical NPs approximately 200 nm in diameter with self-assembling capacity. Mice were vaccinated intranasally with COBRA HA and M7-CpG NPs in a prime-boost-boost schedule. Vaccinated mice had significantly higher antigen-specific antibody responses (IgG and IgA) in serum and mucosa compared with controls. Splenocytes from vaccinated mice had significantly increased cytokine production upon antigen recall and the presence of central and effector memory T cells in draining lymph nodes. Finally, co-immunization with NPs and COBRA HA induced influenza H3N2-specific HA inhibition antibody titers across multiple strains and partially protected mice from a challenge against an H3N2 virus. These results illustrate that the M7-CpG NP adjuvant combination can induce a protective immune response with a broadly reactive influenza antigen via mucosal vaccination.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Erik S. Pena
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
| | - John A. Roque
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rebeca T. Stiepel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael A. Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Sean R. Simpson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ted M. Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Soman N. Abraham
- Departments of Pathology, Molecular Genetics and Microbiology and Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Herman F. Staats
- Department of Pathology, School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccines Institute, School of Medicine, Duke University, Durham, NC, United States
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Kristy M. Ainslie,
| |
Collapse
|
4
|
Inglefield J, Catania J, Harris A, Hickey T, Ma Z, Minang J, Baranji K, Spangler T, Look J, Ruiz C, Lu H, Alleva D, Reece JJ, Lacy MJ. Use of protective antigen of Bacillus anthracis as a model recombinant antigen to evaluate toll-like receptors 2, 3, 4, 7 and 9 agonists in mice using established functional antibody assays, antigen-specific antibody assays and cellular assays. Vaccine 2022; 40:5544-5555. [PMID: 35773119 DOI: 10.1016/j.vaccine.2022.06.017] [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: 05/31/2021] [Revised: 05/04/2022] [Accepted: 06/05/2022] [Indexed: 11/16/2022]
Abstract
Toll-like receptor (TLR) agonists can act as immune stimulants alone or as part of alum or oil formulations. Humoral and cellular immune responses were utilized to assess quantitative and qualitative immune response enhancement by TLR agonists using recombinant protective antigen (rPA) of B. anthracis as a model antigen. To rPA, combined with aluminum hydroxide (Alhydrogel; Al(OH)3) or squalene (AddaVax™), was added one of 7 TLR agonists: TLR2 agonist Pam3CysSK4 (PamS), TLR3 agonist double stranded polyinosinic:polycytidylic acid (PolyIC), TLR4 agonists Monophosphoryl lipid A (MPLA) or glucopyranosyl lipid A (GLA), TLR7-8 agonists 3M-052 or Resiquimod (Resiq), or TLR9 agonist CPG 7909 (CPG). CD-1 or BALB/c mice received two intraperitoneal or intramuscular immunizations 14 days apart, followed by serum or spleen sampling 14 days later. All TLR agonists except PamS induced high levels of B. anthracis lethal toxin-neutralizing antibodies and immunoglobulin G (IgG) anti-PA. Some responses were >100-fold higher than those without a TLR agonist, and IP delivery (0.5 mL) induced higher TLR-mediated antibody response increases compared to IM delivery (0.05 mL). TLR7-8 and TLR9 agonists induced profound shifts of IgG anti-PA response to IgG2a or IgG2b. Compared to the 14-day immunization schedule, use of a shortened immunization schedule of only 7 days between prime and boost found that TLR9 agonist CPG in a squalene formulation maintained higher interferon-γ-positive cells than TLR4 agonist GLA. Variability in antibody responses was lower in BALB/c mice than CD-1 mice but antibody responses were higher in CD-1 mice. Lower serum 50% effective concentration (EC50) values were found for rPA-agonist formulations and squalene formulations compared to Al(OH)3 formulations. Lower EC50 values also were associated with low frequency detection of linear peptide epitopes. In summary, TLR agonists elicited cellular immune responses and markedly boosted humoral responses.
Collapse
Affiliation(s)
- Jon Inglefield
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Jason Catania
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Andrea Harris
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Thomas Hickey
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Zhidong Ma
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Jacob Minang
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Katalin Baranji
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Tarl Spangler
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Jee Look
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Christian Ruiz
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Hang Lu
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - David Alleva
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Joshua J Reece
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA
| | - Michael J Lacy
- Emergent BioSolutions Inc., 300 Professional Drive, Gaithersburg, MD 20879, USA.
| |
Collapse
|
5
|
Johnson-Weaver BT, Choi HW, Yang H, Granek JA, Chan C, Abraham SN, Staats HF. Nasal Immunization With Small Molecule Mast Cell Activators Enhance Immunity to Co-Administered Subunit Immunogens. Front Immunol 2021; 12:730346. [PMID: 34566991 PMCID: PMC8461742 DOI: 10.3389/fimmu.2021.730346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/23/2021] [Indexed: 01/02/2023] Open
Abstract
Mast cell activators are a novel class of mucosal vaccine adjuvants. The polymeric compound, Compound 48/80 (C48/80), and cationic peptide, Mastoparan 7 (M7) are mast cell activators that provide adjuvant activity when administered by the nasal route. However, small molecule mast cell activators may be a more cost-efficient adjuvant alternative that is easily synthesized with high purity compared to M7 or C48/80. To identify novel mast cell activating compounds that could be evaluated for mucosal vaccine adjuvant activity, we employed high-throughput screening to assess over 55,000 small molecules for mast cell degranulation activity. Fifteen mast cell activating compounds were down-selected to five compounds based on in vitro immune activation activities including cytokine production and cellular cytotoxicity, synthesis feasibility, and selection for functional diversity. These small molecule mast cell activators were evaluated for in vivo adjuvant activity and induction of protective immunity against West Nile Virus infection in BALB/c mice when combined with West Nile Virus envelope domain III (EDIII) protein in a nasal vaccine. We found that three of the five mast cell activators, ST101036, ST048871, and R529877, evoked high levels of EDIII-specific antibody and conferred comparable levels of protection against WNV challenge. The level of protection provided by these small molecule mast cell activators was comparable to the protection evoked by M7 (67%) but markedly higher than the levels seen with mice immunized with EDIII alone (no adjuvant 33%). Thus, novel small molecule mast cell activators identified by high throughput screening are as efficacious as previously described mast cell activators when used as nasal vaccine adjuvants and represent next-generation mast cell activators for evaluation in mucosal vaccine studies.
Collapse
Affiliation(s)
| | - Hae Woong Choi
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
| | - Hang Yang
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Josh A. Granek
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Cliburn Chan
- Biostatistics and Bioinformatics Department, School of Medicine, Duke University, Durham, NC, United States
| | - Soman N. Abraham
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
- Department of Immunology, School of Medicine, Duke University, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - Herman F. Staats
- Pathology Department, School of Medicine, Duke University, Durham, NC, United States
- Department of Immunology, School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| |
Collapse
|
6
|
Fries CN, Chen JL, Dennis ML, Votaw NL, Eudailey J, Watts BE, Hainline KM, Cain DW, Barfield R, Chan C, Moody MA, Haynes BF, Saunders KO, Permar SR, Fouda GG, Collier JH. HIV envelope antigen valency on peptide nanofibers modulates antibody magnitude and binding breadth. Sci Rep 2021; 11:14494. [PMID: 34262096 PMCID: PMC8280189 DOI: 10.1038/s41598-021-93702-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/23/2021] [Indexed: 01/02/2023] Open
Abstract
A major challenge in developing an effective vaccine against HIV-1 is the genetic diversity of its viral envelope. Because of the broad range of sequences exhibited by HIV-1 strains, protective antibodies must be able to bind and neutralize a widely mutated viral envelope protein. No vaccine has yet been designed which induces broadly neutralizing or protective immune responses against HIV in humans. Nanomaterial-based vaccines have shown the ability to generate antibody and cellular immune responses of increased breadth and neutralization potency. Thus, we have developed supramolecular nanofiber-based immunogens bearing the HIV gp120 envelope glycoprotein. These immunogens generated antibody responses that had increased magnitude and binding breadth compared to soluble gp120. By varying gp120 density on nanofibers, we determined that increased antigen valency was associated with increased antibody magnitude and germinal center responses. This study presents a proof-of-concept for a nanofiber vaccine platform generating broad, high binding antibody responses against the HIV-1 envelope glycoprotein.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Maria L Dennis
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Nicole L Votaw
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Joshua Eudailey
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Brian E Watts
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Derek W Cain
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kevin O Saunders
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, New York-Presbyterian/Weill Cornell Medicine, New York, NY, 10065, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
7
|
St. John AL, Choi HW, Walker QD, Blough B, Kuhn CM, Abraham SN, Staats HF. Novel mucosal adjuvant, mastoparan-7, improves cocaine vaccine efficacy. NPJ Vaccines 2020; 5:12. [PMID: 32047657 PMCID: PMC7002721 DOI: 10.1038/s41541-020-0161-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/14/2020] [Indexed: 12/29/2022] Open
Abstract
Cocaine is one of the most potent and addictive psychostimulants known and there are no available pharmacotherapies to treat cocaine addiction. Here we describe a novel cocaine vaccine employing the mucosal adjuvant and mast cell-activating oligopeptide, mastoparan-7 (M7), to achieve optimal IgA antibody responses in mucosal secretions and effective induction of humoral immunity using a short immunization protocol. This formulation, using a hapten-carrier system to deliver cocaine as antigen, also reduced cocaine penetration of the blood brain barrier and protected mice from its psychoactive effects by reducing cocaine-induced locomotion. Surprisingly, the magnitude of cocaine-specific antibody titers induced by each adjuvant was not the major determinant of functional protection from cocaine challenge. A side-by-side comparison of the two haptens, cocaine and its analog GNC demonstrated that cocaine haptenation resulted in superior functional protection when used in combination with the novel mucosal adjuvant, M7. These results provide a new potential strategy for combatting cocaine addiction through mucosal vaccination.
Collapse
Affiliation(s)
- Ashley L. St. John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, 169857 Singapore
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228 Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore, 168753 Singapore
| | - Hae Woong Choi
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Present Address: Korea University, Division of Life Sciences, 108 Hana-Science Building, 145 Anam-ro, Seongbuk-gu, Seoul, South Korea
| | - Q. David Walker
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Bruce Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27709 USA
| | - Cynthia M. Kuhn
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Soman N. Abraham
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, 169857 Singapore
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Immunology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710 USA
| | - Herman F. Staats
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710 USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
8
|
Optimized Mucosal Modified Vaccinia Virus Ankara Prime/Soluble gp120 Boost HIV Vaccination Regimen Induces Antibody Responses Similar to Those of an Intramuscular Regimen. J Virol 2019; 93:JVI.00475-19. [PMID: 31068425 DOI: 10.1128/jvi.00475-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/01/2019] [Indexed: 12/29/2022] Open
Abstract
The benefits of mucosal vaccines over injected vaccines are difficult to ascertain, since mucosally administered vaccines often induce serum antibody responses of lower magnitude than those induced by injected vaccines. This study aimed to determine if mucosal vaccination using a modified vaccinia virus Ankara expressing human immunodeficiency virus type 1 (HIV-1) gp120 (MVAgp120) prime and a HIV-1 gp120 protein boost could be optimized to induce serum antibody responses similar to those induced by an intramuscularly (i.m.) administered MVAgp120 prime/gp120 boost to allow comparison of an i.m. immunization regimen to a mucosal vaccination regimen for the ability to protect against a low-dose rectal simian-human immunodeficiency virus (SHIV) challenge. A 3-fold higher antigen dose was required for intranasal (i.n.) immunization with gp120 to induce serum anti-gp120 IgG responses not significantly different than those induced by i.m. immunization. gp120 fused to the adenovirus type 2 fiber binding domain (gp120-Ad2F), a mucosal targeting ligand, exhibited enhanced i.n. immunogenicity compared to gp120. MVAgp120 was more immunogenic after i.n. delivery than after gastric or rectal delivery. Using these optimized vaccines, an i.n. MVAgp120 prime/combined i.m. (gp120) and i.n. (gp120-Ad2F) boost regimen (i.n./i.m.-plus-i.n.) induced serum anti-gp120 antibody titers similar to those induced by the intramuscular prime/boost regimen (i.m./i.m.) in rabbits and nonhuman primates. Despite the induction of similar systemic anti-HIV-1 antibody responses, neither the i.m./i.m. nor the i.n./i.m.-plus-i.n. regimen protected against a repeated low-dose rectal SHIV challenge. These results demonstrate that immunization regimens utilizing the i.n. route are able to induce serum antigen-specific antibody responses similar to those induced by systemic immunization.IMPORTANCE Mucosal vaccination is proposed as a method of immunization able to induce protection against mucosal pathogens that is superior to protection provided by parenteral immunization. However, mucosal vaccination often induces serum antigen-specific immune responses of lower magnitude than those induced by parenteral immunization, making the comparison of mucosal and parenteral immunization difficult. We identified vaccine parameters that allowed an immunization regimen consisting of an i.n. prime followed by boosters administered by both i.n. and i.m. routes to induce serum antibody responses similar to those induced by i.m. prime/boost vaccination. Additional studies are needed to determine the potential benefit of mucosal immunization for HIV-1 and other mucosally transmitted pathogens.
Collapse
|
9
|
Choi HW, Chan C, Shterev ID, Lynch HE, Robinette TJ, Johnson-Weaver BT, Shi J, Sempowski GD, Kim SY, Dickson JK, Gooden DM, Abraham SN, Staats HF. Identification of Novel Mast Cell Activators Using Cell-Based High-Throughput Screening. SLAS DISCOVERY 2019; 24:628-640. [PMID: 30917061 DOI: 10.1177/2472555219834699] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mast cells (MCs) are known to regulate innate and adaptive immunity. MC activators have recently been described as safe and effective vaccine adjuvants. Many currently known MC activators are inadequate for in vivo applications, however, and research on identifying novel MC activators is limited. In this study, we identified novel MC activators by using high-throughput screening (HTS) assays using approximately 55,000 small molecules. Data sets obtained by the primary HTS assays were statistically evaluated using quality control rules and the B-score calculation, and compounds with B-scores of >3.0 were chosen as mast cell activators (hits). These hits were re-evaluated with secondary and tertiary HTS assays, followed by further statistical analysis. From these hits, we selected 15 compounds that caused degranulation in murine and human MCs, with potential for flexible chemical modification for further study. Among these 15 compounds, ST101036, ST029248, and ST026567 exhibited higher degranulation potency than other hit compounds in both human and mouse MCs. In addition, the 15 compounds identified promote de novo synthesis of cytokines and induce the release of eicosanoids from human and mouse MCs. HTS enabled us to identify small-molecule MC activators with unique properties that may be useful as vaccine adjuvants.
Collapse
Affiliation(s)
- Hae Woong Choi
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Cliburn Chan
- 2 Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Ivo D Shterev
- 3 Duke Regional Biocontainment Laboratory, Duke University School of Medicine, Durham, NC, USA.,4 Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Heather E Lynch
- 3 Duke Regional Biocontainment Laboratory, Duke University School of Medicine, Durham, NC, USA.,4 Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,5 Departments of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Taylor J Robinette
- 5 Departments of Medicine, Duke University School of Medicine, Durham, NC, USA
| | | | - Jianling Shi
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Gregory D Sempowski
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA.,3 Duke Regional Biocontainment Laboratory, Duke University School of Medicine, Durham, NC, USA.,4 Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,5 Departments of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - So Young Kim
- 6 Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | | | - David M Gooden
- 8 Department of Chemistry, Duke University, Durham, NC, USA
| | - Soman N Abraham
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA.,9 Department of Molecular Genetics & Microbiology, Duke University Medical Center, Durham, NC, USA.,10 Department of Immunology, Duke University Medical Center, Durham, NC, USA.,11 Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore
| | - Herman F Staats
- 1 Department of Pathology, Duke University School of Medicine, Durham, NC, USA.,4 Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.,10 Department of Immunology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
10
|
Kozlowski PA, Aldovini A. Mucosal Vaccine Approaches for Prevention of HIV and SIV Transmission. CURRENT IMMUNOLOGY REVIEWS 2019; 15:102-122. [PMID: 31452652 PMCID: PMC6709706 DOI: 10.2174/1573395514666180605092054] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/19/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
Optimal protective immunity to HIV will likely require that plasma cells, memory B cells and memory T cells be stationed in mucosal tissues at portals of viral entry. Mucosal vaccine administration is more effective than parenteral vaccine delivery for this purpose. The challenge has been to achieve efficient vaccine uptake at mucosal surfaces, and to identify safe and effective adjuvants, especially for mucosally administered HIV envelope protein immunogens. Here, we discuss strategies used to deliver potential HIV vaccine candidates in the intestine, respiratory tract, and male and female genital tract of humans and nonhuman primates. We also review mucosal adjuvants, including Toll-like receptor agonists, which may adjuvant both mucosal humoral and cellular immune responses to HIV protein immunogens.
Collapse
Affiliation(s)
- Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Anna Aldovini
- Department of Medicine, and Harvard Medical School, Boston Children’s Hospital, Department of Pediatrics, Boston MA, 02115, USA
| |
Collapse
|
11
|
Hiradate Y, Sasaki E, Momose H, Asanuma H, Furuhata K, Takai M, Aoshi T, Yamada H, Ishii KJ, Tanemura K, Mizukami T, Hamaguchi I. Development of screening method for intranasal influenza vaccine and adjuvant safety in preclinical study. Biologicals 2018; 55:43-52. [DOI: 10.1016/j.biologicals.2018.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 05/29/2018] [Accepted: 07/05/2018] [Indexed: 11/26/2022] Open
|
12
|
Schubert N, Lisenko K, Auerbach C, Weitzmann A, Ghouse SM, Muhandes L, Haase C, Häring T, Schulze L, Voehringer D, Gunzer F, Müller W, Feyerabend TB, Rodewald HR, Dudeck A, Roers A. Unimpaired Responses to Vaccination With Protein Antigen Plus Adjuvant in Mice With Kit-Independent Mast Cell Deficiency. Front Immunol 2018; 9:1870. [PMID: 30210490 PMCID: PMC6123530 DOI: 10.3389/fimmu.2018.01870] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/30/2018] [Indexed: 12/23/2022] Open
Abstract
Innate inflammatory responses are crucial for induction and regulation of T cell and antibody responses. Mast cell (MC)-deficient Kit mutant mice showed impaired adaptive immunity, suggesting that MCs provide essential adjuvant activities, and pharmacological MC activation was proposed as a new adjuvant principle. However, the Kit mutations result in complex alterations of the immune system in addition to MC deficiency. We revisited the role of MCs in vaccination responses using Mcpt5-Cre R26DTA/DTA and Cpa3Cre/+ mice that lack connective tissue MCs or all MCs, respectively, but feature an otherwise normal immune system. These animals showed no impairment of T and B cell responses to intradermal vaccination with protein antigen plus complete Freund’s adjuvant. Moreover, we demonstrate that the adjuvant effects of the MC secretagogue c48/80 in intradermal or mucosal immunization are independent of the presence of MCs. We hence find no evidence for a regulation by MCs of adaptive immune responses to protein antigens. The finding that immunological MC functions differ from those suggested by experiments in Kit mutants, emphasizes the importance of rigorous tests in Kit-independent MC-deficiency models.
Collapse
Affiliation(s)
- Nadja Schubert
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Katharina Lisenko
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Christian Auerbach
- Medical Faculty Carl Gustav Carus, Institute of Medical Microbiology and Hygiene, University of Technology Dresden, Dresden, Germany
| | - Anke Weitzmann
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Shanawaz Mohammed Ghouse
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Lina Muhandes
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Christa Haase
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Tobias Häring
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - Livia Schulze
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Florian Gunzer
- Medical Faculty Carl Gustav Carus, Institute of Medical Microbiology and Hygiene, University of Technology Dresden, Dresden, Germany
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Anne Dudeck
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany.,Medical Faculty, Institute for Molecular and Clinical Immunology, Otto von Guericke University, Magdeburg, Germany
| | - Axel Roers
- Medical Faculty Carl Gustav Carus, Institute for Immunology, University of Technology Dresden, Dresden, Germany
| |
Collapse
|
13
|
Li B, Yuan H, Chen L, Sun H, Hu J, Wei S, Zhao Z, Zou Q, Wu C. The influence of adjuvant on UreB protection against Helicobacter pylori through the diversity of CD4+ T-cell epitope repertoire. Oncotarget 2017; 8:68138-68152. [PMID: 28978104 PMCID: PMC5620244 DOI: 10.18632/oncotarget.19248] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023] Open
Abstract
Adjuvants are widely used to enhance the effects of vaccines against pathogen infections. Interestingly, different adjuvants and vaccination routes usually induce dissimilar immune responses, and can even have completely opposite effects. The mechanism remains unclear. In this study, urease B subunit (UreB), an antigen of Helicobacter pylori (H. pylori) that can induce protective immune responses, was used as a model to vaccinate mice. We investigated the effects of different adjuvants and routes on consequent T cell epitope-specific targeting and protection against H. pylori infection. Comparison of the protective effects of UreB, administered either subcutaneously (sc) or intranasally (in), with the adjuvants AddaVax (sc), Complete Freund’s adjuvant (CFA; sc), or CpG oligonucleotide (CpG; sc or in), indicated that only CFA (sc) and CpG (in) were protective. Protective vaccines induced T cells targeting epitopes that differed from that targeted by control vaccination. Subsequent peptide vaccination demonstrated that only two of the identified epitopes were protective: UreB373–385 and UreB317–329. Overall, we found that both adjuvant and vaccination route affected the T cell response repertoire to antigen epitopes. The data obtained in this study contribute to improved characterization of the relationship between adjuvants, routes of vaccination, and epitope-specific T cell response repertoires.
Collapse
Affiliation(s)
- Bin Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Hanmei Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Li Chen
- Department of Blood Transfusion, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Heqiang Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Jian Hu
- Department of Intensive Care Unit, Chengdu Military General Hospital, Chengdu, PR China.,Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Shanshan Wei
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Chao Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
14
|
Burt T, Noveck RJ, MacLeod DB, Layton AT, Rowland M, Lappin G. Intra-Target Microdosing (ITM): A Novel Drug Development Approach Aimed at Enabling Safer and Earlier Translation of Biological Insights Into Human Testing. Clin Transl Sci 2017; 10:337-350. [PMID: 28419765 PMCID: PMC5593170 DOI: 10.1111/cts.12464] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/01/2017] [Indexed: 12/17/2022] Open
Affiliation(s)
- T Burt
- Burt Consultancy, LLC, Durham, North Carolina, USA
| | - R J Noveck
- Medical Director, Duke Clinical Research Unit, Durham, North Carolina, USA
| | - D B MacLeod
- Department of Anesthesiology, Duke University, Durham, North Carolina, USA
| | - A T Layton
- Robert R. and Katherine B. Penn Professor of Mathematics Arts and Sciences Council Chair Professor of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - M Rowland
- School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Manchester, UK
| | - G Lappin
- Reader in Pharmaceutical Science, Lincoln School of Pharmacy, University of Lincoln, Lincoln, Lincolnshire, UK
| |
Collapse
|
15
|
Gallovic MD, Schully KL, Bell MG, Elberson MA, Palmer JR, Darko CA, Bachelder EM, Wyslouzil BE, Keane-Myers AM, Ainslie KM. Acetalated Dextran Microparticulate Vaccine Formulated via Coaxial Electrospray Preserves Toxin Neutralization and Enhances Murine Survival Following Inhalational Bacillus Anthracis Exposure. Adv Healthc Mater 2016; 5:2617-2627. [PMID: 27594343 DOI: 10.1002/adhm.201600642] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 07/20/2016] [Indexed: 12/30/2022]
Abstract
Subunit formulations are regarded as the safest type of vaccine, but they often contain a protein-based antigen that can result in significant challenges, such as preserving antigenicity during formulation and administration. Many studies have demonstrated that encapsulation of protein antigens in polymeric microparticles (MPs) via emulsion techniques results in total IgG antibody titers comparable to alum formulations, however, the antibodies themselves are non-neutralizing. To address this issue, a coaxial electrohydrodynamic spraying (electrospray) technique is used to formulate a microparticulate-based subunit anthrax vaccine under conditions that minimize recombinant protective antigen (rPA) exposure to harsh solvents and high shear stress. rPA and the adjuvant resiquimod are encapsulated either in separate or the same acetalated dextran MPs. Using a murine model, the electrospray formulations lead to higher IgG2a subtype titers as well as comparable total IgG antibody titers and toxin neutralization relative to the FDA-approved vaccine (BioThrax). BioThrax provides no protection against a lethal inhalational challenge of the highly virulent Ames Bacillus anthracis anthrax strain, whereas 50% of the mice vaccinated with separately encapsulated electrospray MPs survive. Overall, this study demonstrates the potential use of electrospray for encapsulating protein antigens in polymeric MPs.
Collapse
Affiliation(s)
- Matthew D. Gallovic
- Department of Chemical and Biomolecular Engineering; College of Engineering; The Ohio State University; Columbus OH 43210 USA
- Division of Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina; Chapel Hill NC 27599 USA
| | - Kevin L. Schully
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Matthew G. Bell
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Margaret A. Elberson
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - John R. Palmer
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Christian A. Darko
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Eric M. Bachelder
- Division of Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina; Chapel Hill NC 27599 USA
| | - Barbara E. Wyslouzil
- Department of Chemical and Biomolecular Engineering; College of Engineering; The Ohio State University; Columbus OH 43210 USA
- Department of Chemistry and Biochemistry; College of Arts and Sciences; The Ohio State University; Columbus OH 43210 USA
| | - Andrea M. Keane-Myers
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Kristy M. Ainslie
- Division of Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina; Chapel Hill NC 27599 USA
| |
Collapse
|
16
|
Veazey RS, Siddiqui A, Klein K, Buffa V, Fischetti L, Doyle-Meyers L, King DF, Tregoning JS, Shattock RJ. Evaluation of mucosal adjuvants and immunization routes for the induction of systemic and mucosal humoral immune responses in macaques. Hum Vaccin Immunother 2016; 11:2913-22. [PMID: 26697975 DOI: 10.1080/21645515.2015.1070998] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Delivering vaccine antigens to mucosal surfaces is potentially very attractive, especially as protection from mucosal infections may be mediated by local immune responses. However, to date mucosal immunization has had limited successes, with issues of both safety and poor immunogenicity. One approach to improve immunogenicity is to develop adjuvants that are effective and safe at mucosal surfaces. Differences in immune responses between mice and men have overstated the value of some experimental adjuvants which have subsequently performed poorly in the clinic. Due to their closer similarity, non-human primates can provide a more accurate picture of adjuvant performance. In this study we immunised rhesus macaques (Macaca mulatta) using a unique matrix experimental design that maximised the number of adjuvants screened while reducing the animal usage. Macaques were immunised by the intranasal, sublingual and intrarectal routes with the model protein antigens keyhole limpet haemocyanin (KLH), β-galactosidase (β-Gal) and ovalbumin (OVA) in combination with the experimental adjuvants Poly(I:C), Pam3CSK4, chitosan, Thymic Stromal Lymphopoietin (TSLP), MPLA and R848 (Resiquimod). Of the routes used, only intranasal immunization with KLH and R848 induced a detectable antibody response. When compared to intramuscular immunization, intranasal administration gave slightly lower levels of antigen specific antibody in the plasma, but enhanced local responses. Following intranasal delivery of R848, we observed a mildly inflammatory response, but no difference to the control. From this we conclude that R848 is able to boost antibody responses to mucosally delivered antigen, without causing excess local inflammation.
Collapse
Affiliation(s)
- Ronald S Veazey
- a Tulane National Primate Research Center; Tulane University School of Medicine ; Covington , LA USA
| | - Asna Siddiqui
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| | - Katja Klein
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK.,c Present affiliation: University of Western Ontario ; Ontario , Canada
| | - Viviana Buffa
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| | - Lucia Fischetti
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| | - Lara Doyle-Meyers
- a Tulane National Primate Research Center; Tulane University School of Medicine ; Covington , LA USA
| | - Deborah F King
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK.,d Present affiliation: IAVI Human Immunology Lab; Chelsea and Westminster; Imperial College London ; London , UK
| | - John S Tregoning
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| | - Robin J Shattock
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| |
Collapse
|
17
|
Effect of particulate adjuvant on the anthrax protective antigen dose required for effective nasal vaccination. Vaccine 2015; 33:3609-13. [DOI: 10.1016/j.vaccine.2015.06.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/07/2015] [Accepted: 06/05/2015] [Indexed: 11/21/2022]
|
18
|
Bento D, Staats HF, Gonçalves T, Borges O. Development of a novel adjuvanted nasal vaccine: C48/80 associated with chitosan nanoparticles as a path to enhance mucosal immunity. Eur J Pharm Biopharm 2015; 93:149-64. [PMID: 25818119 DOI: 10.1016/j.ejpb.2015.03.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/19/2015] [Accepted: 03/20/2015] [Indexed: 11/15/2022]
Abstract
In a time in which mucosal vaccines development has been delayed by the lack of safe and effective mucosal adjuvants, the combination of adjuvants has started to be explored as a strategy to obtain potent vaccine formulations. This study describes a novel adjuvant combination as an effective approach for a nasal vaccine - the association of the mast cell activator compound 48/80 with chitosan based nanoparticles. It was hypothesized that mucoadhesive nanoparticles would promote the cellular uptake and prolong the antigen residence time on nasal cavity. Simultaneously, mast cell activation would promote a local microenvironment favorable to the development of an immune response. To test this hypothesis, two different C48/80 loaded nanoparticles (NPs) were prepared: Chitosan-C48/80 NP (Chi-C48/80 NP) and Chitosan/Alginate-C48/80 NP (Chi/Alg-C48/80 NP). The potential as a vaccine adjuvant of the two delivery systems was evaluated and directly compared. Both formulations had a mean size near 500nm and a positive charge; however, Chi-C48/80 NP was a more effective adjuvant delivery system when compared with Chi/Alg-C48/80 NP or C48/80 alone. Chi-C48/80 NP activated mast cells at a greater extent, were better internalized by antigen presenting cells than Chi/Alg-C48/80 NP and successfully enhanced the nasal residence time of a model antigen. Superiority of Chi-C48/80 NP as adjuvant was also observed in vivo. Therefore, nasal immunization of mice with Bacillus anthracis protective antigen (PA) adsorbed on Chi-C48/80 NP elicited high levels of serum anti-PA neutralizing antibodies and a more balanced Th1/Th2 profile than C48/80 in solution or Chi/Alg-C48/80 NP. The incorporation of C48/80 within Chi NP also promoted a mucosal immunity greater than all the other adjuvanted groups tested, showing that the combination of a mast cell activator and chitosan NP could be a promising strategy for nasal immunization.
Collapse
Affiliation(s)
- D Bento
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - H F Staats
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - T Gonçalves
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute of Microbiology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - O Borges
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
19
|
Zeng L, Liu Y, Wang H, Liao P, Song Z, Gao S, Wu Y, Zhang X, Yin Y, Xu W. Compound 48/80 acts as a potent mucosal adjuvant for vaccination against Streptococcus pneumoniae infection in young mice. Vaccine 2015; 33:1008-16. [DOI: 10.1016/j.vaccine.2015.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 12/25/2014] [Accepted: 01/06/2015] [Indexed: 10/24/2022]
|
20
|
|
21
|
Mucosal immunization with the live attenuated vaccine SPY1 induces humoral and Th2-Th17-regulatory T cell cellular immunity and protects against pneumococcal infection. Infect Immun 2014; 83:90-100. [PMID: 25312946 DOI: 10.1128/iai.02334-14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mucosal immunization with attenuated vaccine can protect against pneumococcal invasion infection, but the mechanism was unknown. Our study found that mucosal delivery with the live attenuated SPY1 vaccine strain can confer T cell- and B cell-dependent protection against pneumococcal colonization and invasive infection; yet it is still unclear which cell subsets contribute to the protection, and their roles in pneumococcal colonization and invasion remain elusive. Adoptive transfer of anti-SPY1 antibody conferred protection to naive μMT mice, and immune T cells were indispensable to protection examined in nude mice. A critical role of interleukin 17A (IL-17A) in colonization was demonstrated in mice lacking IL-17A, and a vaccine-specific Th2 immune subset was necessary for systemic protection. Of note, we found that SPY1 could stimulate an immunoregulatory response and that SPY1-elicited regulatory T cells participated in protection against colonization and lethal infection. The data presented here aid our understanding of how live attenuated strains are able to function as effective vaccines and may contribute to a more comprehensive evaluation of live vaccines and other mucosal vaccines.
Collapse
|