1
|
Sallard E, Aydin M. Cutting-edge research frontiers in oral cavity vaccines for respiratory diseases: a roadmap for scientific advancement. Front Cell Infect Microbiol 2024; 14:1388222. [PMID: 38988815 PMCID: PMC11234472 DOI: 10.3389/fcimb.2024.1388222] [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: 02/19/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024] Open
Abstract
Intramuscular vaccines present limitations in eliciting robust mucosal immunity and preventing respiratory pathogens transmission. Sublingual vaccine administration offers promising advantages, including interconnected mucosal protection. Despite these advantages, only a few clinical trials have explored sublingual vaccines, underscoring the necessity of optimizing next-generation vaccine formulas. Critical research priorities include understanding vector behavior in the oral environment, understanding their interactions with mucosal immunity and developing formulations enabling sustained mucosal contact to facilitate efficient transduction. Consequently, tonsil organoids, as representative human mucosal models, could offer critical insights into sublingual immunization. Thus, a multi-disciplinary approach integrating pharmacological, immunological, and manufacturing considerations is pivotal for sublingual vaccines in targeting pathogen-aggravated prevalent respiratory diseases including asthma, COPD and lung cancer, as well as the antimicrobial resistance crisis.
Collapse
Affiliation(s)
- Erwan Sallard
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Malik Aydin
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Research, School of Life Sciences (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
- Institute for Medical Laboratory Diagnostics, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany
| |
Collapse
|
2
|
Gutiérrez RL, Riddle MS, Porter CK, Maciel M, Poole ST, Laird RM, Lane M, Turiansky GW, Jarell A, Savarino SJ. A First in Human Clinical Trial Assessing the Safety and Immunogenicity of Two Intradermally Delivered Enterotoxigenic Escherichia coli CFA/I Fimbrial Tip Adhesin Antigens with and without Heat-Labile Enterotoxin with Mutation LT(R192G). Microorganisms 2023; 11:2689. [PMID: 38004700 PMCID: PMC10672875 DOI: 10.3390/microorganisms11112689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
INTRODUCTION Enterotoxigenic E. coli (ETEC) is a leading cause of diarrhea in travelers as well as for children living in low- to middle-income countries. ETEC adhere to intestinal epithelium via colonization factors (CFs). CFA/I, a common CF, is composed of a polymeric stalk and a tip-localized minor adhesive subunit, CfaE. Vaccine delivery by the transcutaneous immunization of dscCfaE was safe but was poorly immunogenic in a phase 1 trial when administered to volunteers with LTR(192G) and mLT. To potentially enhance the immunogenicity of CfaE while still delivering via a cutaneous route, we evaluated the safety and immunogenicity of two CfaE constructs administered intradermally (ID) with or without mLT. METHODS CfaE was evaluated as a donor strand-complemented construct (dscCfaE) and as a chimeric construct (Chimera) in which dscCfaE replaces the A1 domain of the cholera toxin A subunit and assembles non-covalently with the pentamer of heat-labile toxin B (LTB). Subjects received three ID vaccinations three weeks apart with either dscCfaE (1, 5, and 25 µg) or Chimera (2.6 and 12.9 µg) with and without 0.1 µg of mLT. Subjects were monitored for local and systemic adverse events. Immunogenicity was evaluated by serum and antibody-secreting cell (ASC) responses. RESULTS The vaccine was well-tolerated with predominantly mild and moderate local vaccine site reactions characterized by erythema, induration and post-inflammatory hyperpigmentation. High rates of serologic and ASC responses were seen across study groups with the most robust responses observed in subjects receiving 25 µg of dscCfaE with 0.1 mcg of LT(R192G). CONCLUSION Both ETEC adhesin vaccine prototypes were safe and immunogenic when co-administered with mLT by the ID route. The observed immune responses induced with the high dose of dscCfaE and mLT warrant further assessment in a controlled human infection model.
Collapse
Affiliation(s)
- Ramiro L. Gutiérrez
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (S.T.P.)
| | - Mark S. Riddle
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (S.T.P.)
| | - Chad K. Porter
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (S.T.P.)
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Milton Maciel
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (S.T.P.)
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Steven T. Poole
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (S.T.P.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Renee M. Laird
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (S.T.P.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Michelle Lane
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (S.T.P.)
| | - George W. Turiansky
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Abel Jarell
- Walter Reed National Military Medical Center, Bethesda, MD 20814, USA
| | - Stephen J. Savarino
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (S.T.P.)
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
3
|
Svennerholm AM, Lundgren A. Developments in oral enterotoxigenic Escherichia coli vaccines. Curr Opin Immunol 2023; 84:102372. [PMID: 37523966 DOI: 10.1016/j.coi.2023.102372] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/11/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a leading cause of diarrhea in children in developing countries and in travelers. WHO has affirmed ETEC as a priority vaccine target, but there is no licensed ETEC vaccine available yet. We here describe recent, promising developments of different live, inactivated, and subunit ETEC candidate vaccines expressing or containing nontoxic enterotoxin and/or colonization factor antigens with a focus on oral vaccines. Many of the ETEC candidate vaccines have been tested in clinical trials for safety and immunogenicity and some of them also for protective efficacy in field trials or in challenge studies.
Collapse
Affiliation(s)
- Ann-Mari Svennerholm
- Dept. of Microbiology and Immunology, Inst. of Biomedicine, University of Gothenburg, Sweden.
| | - Anna Lundgren
- Dept. of Microbiology and Immunology, Inst. of Biomedicine, University of Gothenburg, Sweden
| |
Collapse
|
4
|
Prior JT, Limbert VM, Horowitz RM, D'Souza SJ, Bachnak L, Godwin MS, Bauer DL, Harrell JE, Morici LA, Taylor JJ, McLachlan JB. Establishment of isotype-switched, antigen-specific B cells in multiple mucosal tissues using non-mucosal immunization. NPJ Vaccines 2023; 8:80. [PMID: 37258506 PMCID: PMC10231862 DOI: 10.1038/s41541-023-00677-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Although most pathogens infect the human body via mucosal surfaces, very few injectable vaccines can specifically target immune cells to these tissues where their effector functions would be most desirable. We have previously shown that certain adjuvants can program vaccine-specific helper T cells to migrate to the gut, even when the vaccine is delivered non-mucosally. It is not known whether this is true for antigen-specific B cell responses. Here we show that a single intradermal vaccination with the adjuvant double mutant heat-labile toxin (dmLT) induces a robust endogenous, vaccine-specific, isotype-switched B cell response. When the vaccine was intradermally boosted, we detected non-circulating vaccine-specific B cell responses in the lamina propria of the large intestines, Peyer's patches, and lungs. When compared to the TLR9 ligand adjuvant CpG, only dmLT was able to drive the establishment of isotype-switched resident B cells in these mucosal tissues, even when the dmLT-adjuvanted vaccine was administered non-mucosally. Further, we found that the transcription factor Batf3 was important for the full germinal center reaction, isotype switching, and Peyer's patch migration of these B cells. Collectively, these data indicate that specific adjuvants can promote mucosal homing and the establishment of activated, antigen-specific B cells in mucosal tissues, even when these adjuvants are delivered by a non-mucosal route. These findings could fundamentally change the way future vaccines are formulated and delivered.
Collapse
Affiliation(s)
- John T Prior
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Vanessa M Limbert
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Rebecca M Horowitz
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Shaina J D'Souza
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Louay Bachnak
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Matthew S Godwin
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - David L Bauer
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jaikin E Harrell
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Lisa A Morici
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - James B McLachlan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
| |
Collapse
|
5
|
Mbani CJ, Nekoua MP, Moukassa D, Hober D. The Fight against Poliovirus Is Not Over. Microorganisms 2023; 11:1323. [PMID: 37317297 DOI: 10.3390/microorganisms11051323] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/13/2023] [Accepted: 05/14/2023] [Indexed: 06/16/2023] Open
Abstract
Poliovirus (PV), the virus that causes both acute poliomyelitis and post-polio syndrome, is classified within the Enterovirus C species, and there are three wild PV serotypes: WPV1, WPV2 and WPV3. The launch of the Global Polio Eradication Initiative (GPEI) in 1988 eradicated two of the three serotypes of WPV (WPV2 and WPV3). However, the endemic transmission of WPV1 persists in Afghanistan and Pakistan in 2022. There are cases of paralytic polio due to the loss of viral attenuation in the oral poliovirus vaccine (OPV), known as vaccine-derived poliovirus (VDPV). Between January 2021 and May 2023, a total of 2141 circulating VDPV (cVDPV) cases were reported in 36 countries worldwide. Because of this risk, inactivated poliovirus (IPV) is being used more widely, and attenuated PV2 has been removed from OPV formulations to obtain bivalent OPV (containing only types 1 and 3). In order to avoid the reversion of attenuated OPV strains, the new OPV, which is more stable due to genome-wide modifications, as well as sabin IPV and virus-like particle (VLP) vaccines, is being developed and offers promising solutions for eradicating WP1 and VDPV.
Collapse
Affiliation(s)
- Chaldam Jespère Mbani
- Laboratoire de Virologie URL3610, Université de Lille, CHU Lille, 59000 Lille, France
- Laboratoire de Biologie Cellulaire et Moléculaire, Faculté des Sciences et Technique, Université Marien Ngouabi, Brazzaville BP 69, Congo
| | | | - Donatien Moukassa
- Laboratoire de Biologie Cellulaire et Moléculaire, Faculté des Sciences et Technique, Université Marien Ngouabi, Brazzaville BP 69, Congo
| | - Didier Hober
- Laboratoire de Virologie URL3610, Université de Lille, CHU Lille, 59000 Lille, France
| |
Collapse
|
6
|
Stone AE, Rambaran S, Trinh IV, Estrada M, Jarand CW, Williams BS, Murrell AE, Huerter CM, Bai W, Palani S, Nakanishi Y, Laird RM, Poly FM, Reed WF, White JA, Norton EB. Route and antigen shape immunity to dmLT-adjuvanted vaccines to a greater extent than biochemical stress or formulation excipients. Vaccine 2023; 41:1589-1601. [PMID: 36732163 PMCID: PMC10308557 DOI: 10.1016/j.vaccine.2023.01.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 02/04/2023]
Abstract
A key aspect to vaccine efficacy is formulation stability. Biochemical evaluations provide information on optimal compositions or thermal stability but are routinely validated by ex vivo analysis and not efficacy in animal models. Here we assessed formulations identified to improve or reduce stability of the mucosal adjuvant dmLT being investigated in polio and enterotoxigenic E. coli (ETEC) clinical vaccines. We observed biochemical changes to dmLT protein with formulation or thermal stress, including aggregation or subunit dissociation or alternatively resistance against these changes with specific buffer compositions. However, upon injection or mucosal vaccination with ETEC fimbriae adhesin proteins or inactivated polio virus, experimental findings indicated immunization route and co-administered antigen impacted vaccine immunogenicity more so than dmLT formulation stability (or instability). These results indicate the importance of both biochemical and vaccine-derived immunity assessment in formulation optimization. In addition, these studies have implications for use of dmLT in clinical settings and for delivery in resource poor settings.
Collapse
Affiliation(s)
- Addison E Stone
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Saraswatie Rambaran
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ivy V Trinh
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Curtis W Jarand
- Department of Physics and Engineering Physics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Blake S Williams
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Amelie E Murrell
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Chelsea M Huerter
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - William Bai
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Surya Palani
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Renee M Laird
- Henry M. Jackson Foundation for Military Medicine, Bethesda, MD, USA; Enteric Diseases Department, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Frederic M Poly
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Wayne F Reed
- Department of Physics and Engineering Physics, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Elizabeth B Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
7
|
Estrada MR, Bzami A, Norton EB, White JA. Identifying a stable bulk dmLT adjuvant formulation at a clinically relevant concentration. Vaccine 2023; 41:1362-1367. [PMID: 36658044 PMCID: PMC9932622 DOI: 10.1016/j.vaccine.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/19/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023]
Abstract
Double mutant heat-labile toxin (dmLT) is a novel vaccine adjuvant under development with several different vaccine candidates. Studies using existing dmLT adjuvant stocks require significant dilution to achieve a clinically relevant dose. This dilution leads to wastage of the adjuvant. This manuscript describes a limited formulation study to improve the stability of bulk dmLT at a more clinically relevant concentration (20 µg/mL) with minimal changes to the existing bulk dmLT formulation. In vitro methods were used to evaluate dmLT stability after lyophilization and short-term accelerated stability studies. The addition of the excipient polysorbate 80 (PS80) at 0.05 % to the existing dmLT formulation was identified as the lead modification that provided improved stability of the lyophilized dmLT at 20 µg/mL through 4 weeks at 40 °C.
Collapse
Affiliation(s)
| | - Anan Bzami
- PATH, 2201 Westlake Ave, Seattle, WA 98122, United States
| | - Elizabeth B. Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, United States
| | - Jessica A. White
- PATH, 2201 Westlake Ave, Seattle, WA 98122, United States,Corresponding author.
| |
Collapse
|
8
|
Molina Estupiñan JL, Aradottir Pind AA, Foroutan Pajoohian P, Jonsdottir I, Bjarnarson SP. The adjuvants dmLT and mmCT enhance humoral immune responses to a pneumococcal conjugate vaccine after both parenteral or mucosal immunization of neonatal mice. Front Immunol 2023; 13:1078904. [PMID: 36741402 PMCID: PMC9896006 DOI: 10.3389/fimmu.2022.1078904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/12/2022] [Indexed: 01/21/2023] Open
Abstract
Immaturity of the neonatal immune system contributes to increased susceptibility to infectious diseases and poor vaccine responses. Therefore, better strategies for early life vaccination are needed. Adjuvants can enhance the magnitude and duration of immune responses. In this study we assessed the effects of the adjuvants dmLT and mmCT and different immunization routes, subcutaneous (s.c.) and intranasal (i.n.), on neonatal immune response to a pneumococcal conjugate vaccine Pn1-CRM197. Pn1-specific antibody (Ab) levels of neonatal mice immunized with Pn1-CRM197 alone were low. The adjuvants enhanced IgG Ab responses up to 8 weeks after immunization, more after s.c. than i.n. immunization. On the contrary, i.n. immunization with either adjuvant enhanced serum and salivary IgA levels more than s.c. immunization. In addition, both dmLT and mmCT enhanced germinal center formation and accordingly, dmLT and mmCT enhanced the induction and persistence of Pn1-specific IgG+ Ab-secreting cells (ASCs) in spleen and bone marrow (BM), irrespective of the immunization route. Furthermore, i.n. immunization enhanced Pn1-specific IgA+ ASCs in BM more than s.c. immunizatiofimmu.2022.1078904n. However, a higher i.n. dose of the Pn1-CRM197 was needed to achieve IgG response comparable to that elicited by s.c. immunization with either adjuvant. We conclude that dmLT and mmCT enhance both induction and persistence of the neonatal immune response to the vaccine Pn1-CRM197, following mucosal or parenteral immunization. This indicates that dmLT and mmCT are promising adjuvants for developing safe and effective early life vaccination strategies.
Collapse
Affiliation(s)
- Jenny Lorena Molina Estupiñan
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Audur Anna Aradottir Pind
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Poorya Foroutan Pajoohian
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Ingileif Jonsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Stefania P. Bjarnarson
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland,*Correspondence: Stefania P. Bjarnarson,
| |
Collapse
|
9
|
Anti-Cocaine IgA Rather Than IgG Mediates Vaccine Protection from Cocaine Use. Pharmaceutics 2022; 14:pharmaceutics14112368. [PMID: 36365186 PMCID: PMC9697488 DOI: 10.3390/pharmaceutics14112368] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
In developing a vaccine for fentanyl use disorder, we observed that IgA was the best correlate of vaccine-mediated protection from injected drug challenge, rather than IgG or binding affinity. Recent evidence shows that IgA secreting cells line the blood−brain barrier that capture pathogens and could prevent drug antigens from penetrating the brain. We assayed IgA and IgG antibodies from an anti-cocaine vaccine clinical trial and categorized each subject’s antibody levels using half-log cut-points for IgA: <1000, <5000, <10,000 and >10,000; and for IgG: <10,000 to >100,000. We compared these antibody groups on urine toxicology in 130 subjects at week 9 after 3 booster vaccinations. We also provided relevant data on benzoylecgonine (BE, cocaine metabolite) from this study’s placebo patients. BE urine levels were lowest for the highest IgA category; however, levels did not differ across IgG groups. Our findings linking IgA to protection from cocaine and fentanyl in mice, rats and humans are novel and suggest an increasingly recognized role of IgA in vaccine efficacy.
Collapse
|
10
|
Sahoo A, Jones AT, Cheedarla N, Gangadhara S, Roy V, Styles TM, Shiferaw A, Walter KL, Williams LD, Shen X, Ozorowski G, Lee WH, Burton S, Yi L, Song X, Qin ZS, Derdeyn CA, Ward AB, Clements JD, Varadarajan R, Tomaras GD, Kozlowski PA, Alter G, Amara RR. A clade C HIV-1 vaccine protects against heterologous SHIV infection by modulating IgG glycosylation and T helper response in macaques. Sci Immunol 2022; 7:eabl4102. [PMID: 35867800 PMCID: PMC9410801 DOI: 10.1126/sciimmunol.abl4102] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The rising global HIV-1 burden urgently requires vaccines capable of providing heterologous protection. Here, we developed a clade C HIV-1 vaccine consisting of priming with modified vaccinia Ankara (MVA) and boosting with cyclically permuted trimeric gp120 (CycP-gp120) protein, delivered either orally using a needle-free injector or through parenteral injection. We tested protective efficacy of the vaccine against intrarectal challenges with a pathogenic heterologous clade C SHIV infection in rhesus macaques. Both routes of vaccination induced a strong envelope-specific IgG in serum and rectal secretions directed against V1V2 scaffolds from a global panel of viruses with polyfunctional activities. Envelope-specific IgG showed lower fucosylation compared with total IgG at baseline, and most of the vaccine-induced proliferating blood CD4+ T cells did not express CCR5 and α4β7, markers associated with HIV target cells. After SHIV challenge, both routes of vaccination conferred significant and equivalent protection, with 40% of animals remaining uninfected at the end of six weekly repeated challenges with an estimated efficacy of 68% per exposure. Induction of envelope-specific IgG correlated positively with G1FB glycosylation, and G2S2F glycosylation correlated negatively with protection. Vaccine-induced TNF-α+ IFN-γ+ CD8+ T cells and TNF-α+ CD4+ T cells expressing low levels of CCR5 in the rectum at prechallenge were associated with decreased risk of SHIV acquisition. These results demonstrate that the clade C MVA/CycP-gp120 vaccine provides heterologous protection against a tier2 SHIV rectal challenge by inducing a polyfunctional antibody response with distinct Fc glycosylation profile, as well as cytotoxic CD8 T cell response and CCR5-negative T helper response in the rectum.
Collapse
Affiliation(s)
- Anusmita Sahoo
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Andrew T Jones
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Narayanaiah Cheedarla
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Sailaja Gangadhara
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Tiffany M Styles
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ayalnesh Shiferaw
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Korey L Walter
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - LaTonya D Williams
- Department of Surgery, Duke University Medical School, Duke University, Durham, NC 27710, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University Medical School, Duke University, Durham, NC 27710, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, San Diego, CA 92121, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, San Diego, CA 92121, USA
| | - Samantha Burton
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Lasanajak Yi
- Department of Biochemistry, Emory Glycomics and Molecular Interactions Core (EGMIC), School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Xuezheng Song
- Department of Biochemistry, Emory Glycomics and Molecular Interactions Core (EGMIC), School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Zhaohui S Qin
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Cynthia A Derdeyn
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, San Diego, CA 92121, USA
| | - John D Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 8638, USA
| | - Raghavan Varadarajan
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, Karnataka 560012, India.,Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560012, India
| | - Georgia D Tomaras
- Department of Surgery, Duke University Medical School, Duke University, Durham, NC 27710, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Rama Rao Amara
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
11
|
Garcia‐del Rio L, Diaz‐Rodriguez P, Pedersen GK, Christensen D, Landin M. Sublingual Boosting with a Novel Mucoadhesive Thermogelling Hydrogel Following Parenteral CAF01 Priming as a Strategy Against Chlamydia trachomatis. Adv Healthc Mater 2022; 11:e2102508. [PMID: 35124896 PMCID: PMC11468966 DOI: 10.1002/adhm.202102508] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/18/2022] [Indexed: 01/13/2023]
Abstract
Chlamydia trachomatis is the most prevalent sexually transmitted disease of bacterial origin. The high number of asymptomatic cases makes it difficult to stop the transmission, requiring vaccine development. Herein, a strategy is proposed to obtain local genital tract immunity against C. trachomatis through parenteral prime and sublingual boost. Subcutaneous administration of chlamydia CTH522 subunit vaccine loaded in the adjuvant CAF01 is combined with sublingual administration of CTH522 loaded in a novel thermosensitive and mucoadhesive hydrogel. Briefly, a ternary optimized hydrogel (OGEL) with desirable biological and physicochemical properties is obtained using artificial intelligence techniques. This formulation exhibits a high gel strength and a strong mucoadhesive, adhesive and cohesive nature. The thermosensitive properties of the hydrogel facilitate application under the tongue. Meanwhile the fast gelation at body temperature together with rapid antigen release should avoid CTH522 leakage by swallowing and increase the contact with sublingual tissue, thus promoting absorption. In vivo studies demonstrate that parenteral-sublingual prime-boost immunization, using CAF01 and OGEL as CTH522 vaccine carriers, shows a tendency to increase cellular (Th1/Th17) immune responses when compared to mucosal or parenteral vaccination alone. Furthermore, parenteral prime with CAF01/CTH522 followed by sublingual boosting with OGEL/CTH522 elicits a local IgA response in the genital tract.
Collapse
Affiliation(s)
- Lorena Garcia‐del Rio
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| | - Patricia Diaz‐Rodriguez
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| | - Gabriel Kristian Pedersen
- Department of Infectious Disease ImmunologyStatens Serum InstitutArtillerivej 5Copenhagen S2300Denmark
| | - Dennis Christensen
- Department of Infectious Disease ImmunologyStatens Serum InstitutArtillerivej 5Copenhagen S2300Denmark
| | - Mariana Landin
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| |
Collapse
|
12
|
Fratzke AP, van Schaik EJ, Samuel JE. Immunogenicity and Reactogenicity in Q Fever Vaccine Development. Front Immunol 2022; 13:886810. [PMID: 35693783 PMCID: PMC9177948 DOI: 10.3389/fimmu.2022.886810] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular bacterium which, in humans, causes the disease Q fever. Although Q fever is most often a mild, self-limiting respiratory disease, it can cause a range of severe syndromes including hepatitis, myocarditis, spontaneous abortion, chronic valvular endocarditis, and Q fever fatigue syndrome. This agent is endemic worldwide, except for New Zealand and Antarctica, transmitted via aerosols, persists in the environment for long periods, and is maintained through persistent infections in domestic livestock. Because of this, elimination of this bacterium is extremely challenging and vaccination is considered the best strategy for prevention of infection in humans. Many vaccines against C. burnetii have been developed, however, only a formalin-inactivated, whole cell vaccine derived from virulent C. burnetii is currently licensed for use in humans. Unfortunately, widespread use of this whole cell vaccine is impaired due to the severity of reactogenic responses associated with it. This reactogenicity continues to be a major barrier to access to preventative vaccines against C. burnetii and the pathogenesis of this remains only partially understood. This review provides an overview of past and current research on C. burnetii vaccines, our knowledge of immunogenicity and reactogenicity in C. burnetii vaccines, and future strategies to improve the safety of vaccines against C. burnetii.
Collapse
Affiliation(s)
- Alycia P. Fratzke
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Erin J. van Schaik
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
| | - James E. Samuel
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
13
|
Crothers JW, Ross Colgate E, Cowan KJ, Dickson DM, Walsh M, Carmolli M, Wright PF, Norton EB, Kirkpatrick BD. Intradermal fractional-dose inactivated polio vaccine (fIPV) adjuvanted with double mutant Enterotoxigenic Escherichia coli heat labile toxin (dmLT) is well-tolerated and augments a systemic immune response to all three poliovirus serotypes in a randomized placebo-controlled trial. Vaccine 2022; 40:2705-2713. [PMID: 35367069 PMCID: PMC9024222 DOI: 10.1016/j.vaccine.2022.03.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 11/19/2022]
Abstract
Eradication of poliomyelitis globally is constrained by fecal shedding of live polioviruses, both wild-type and vaccine-derived strains, into the environment. Although inactivated polio vaccines (IPV) effectively protect the recipient from clinical poliomyelitis, fecal shedding of live virus still occurs following infection with either wildtype or vaccine-derived strains of poliovirus. In the drive to eliminate the last cases of polio globally, improvements in both oral polio vaccines (OPV) (to prevent reversion to virulence) and injectable polio vaccines (to improve mucosal immunity and prevent viral shedding) are underway. The E. coli labile toxin with two or "double" attenuating mutations (dmLT) may boost immunologic responses to IPV, including at mucosal sites. We performed a double-blinded phase I controlled clinical trial to evaluate safety, tolerability, as well as systemic and mucosal immunogenicity of IPV adjuvanted with dmLT, given as a fractional (1/5th) dose intradermally (fIPV-dmLT). Twenty-nine volunteers with no past exposure to OPV were randomized to a single dose of fIPV-dmLT or fIPV alone. fIPV-dmLT was well tolerated, although three subjects had mild but persistent induration and hyperpigmentation at the injection site. A ≥ 4-fold rise in serotype-specific neutralizing antibody (SNA) titers to all three serotypes was seen in 84% of subjects receiving fIPV-dmLT vs. 50% of volunteers receiving IPV alone. SNA titers were higher in the dmLT-adjuvanted group, but only differences in serotype 1 were significant. Mucosal immune responses, as measured by polio serotype specific fecal IgA were minimal in both groups and differences were not seen. fIPV-dmLT may offer a benefit over IPV alone. Beyond NAB responses protecting the individual, studies demonstrating the ability of fIPV-dmLT to prevent viral shedding are necessary. Studies employing controlled human infection models, using monovalent OPV post-vaccine are ongoing. Studies specifically in children may also be necessary and additional biomarkers of mucosal immune responses in this population are needed. Clinicaltrials.gov Identifer: NCT03922061.
Collapse
Affiliation(s)
- Jessica W Crothers
- Department of Pathology and Laboratory Medicine, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA.
| | - Elizabeth Ross Colgate
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Kelly J Cowan
- Department of Pediatrics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Dorothy M Dickson
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - MaryClaire Walsh
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Marya Carmolli
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Elizabeth B Norton
- Department of Immunology and Microbiology, Tulane University, New Orleans, LA, USA
| | - Beth D Kirkpatrick
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| |
Collapse
|
14
|
Fleckenstein JM. Confronting challenges to enterotoxigenic Escherichia coli vaccine development. FRONTIERS IN TROPICAL DISEASES 2021; 2:709907. [PMID: 35937717 PMCID: PMC9355458 DOI: 10.3389/fitd.2021.709907] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023] Open
Abstract
The enterotoxigenic Escherichia coli (ETEC) are a diverse and genetically plastic pathologic variant (pathovar) of E. coli defined by their production of heat-labile (LT) and heat-stable (ST) enterotoxins. These pathogens, which came to recognition more than four decades ago in patients presenting with severe cholera-like diarrhea, are now known to cause hundreds of millions of cases of symptomatic infection annually. Children in low-middle income regions of the world lacking access to clean water and basic sanitation are disproportionately affected by ETEC. In addition to acute diarrheal morbidity, these pathogens remain a significant cause of mortality in children under the age of five years and have also been linked repeatedly to sequelae of childhood malnutrition and growth stunting. Vaccines that could prevent ETEC infections therefore remain a high priority. Despite several decades of effort, a licensed vaccine that protects against the breadth of these pathogens remains an aspirational goal, and the underlying genetic plasticity of E. coli has posed a fundamental challenge to development of a vaccine that can encompass the complete antigenic spectrum of ETEC. Nevertheless, novel strategies that include toxoids, a more complete understanding of ETEC molecular pathogenesis, structural details of target immunogens, and the discovery of more highly conserved antigens essential for virulence should accelerate progress and make a broadly protective vaccine feasible.
Collapse
Affiliation(s)
- James M. Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
- Medicine Service, Infectious Diseases, John Cochran Saint Louis Veterans Affairs Health Care System, Saint Louis, Missouri, USA
| |
Collapse
|
15
|
Lee T, Gutiérrez RL, Maciel M, Poole S, Testa KJ, Trop S, Duplessis C, Lane A, Riddle MS, Hamer M, Alcala A, Prouty M, Maier N, Erdem R, Louis Bourgeois A, Porter CK. Safety and immunogenicity of intramuscularly administered CS6 subunit vaccine with a modified heat-labile enterotoxin from enterotoxigenic Escherichia coli. Vaccine 2021; 39:5548-5556. [PMID: 34419306 PMCID: PMC8461560 DOI: 10.1016/j.vaccine.2021.08.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Enterotoxigenic Escherichia coli (ETEC) is a common cause of infectious diarrhoea and a leading cause of morbidity and mortality in children living in resource-limited settings. It is also the leading cause of travellers' diarrhoea among civilian and military travellers. Its dual importance in global public health and travel medicine highlights the need for an effective vaccine. ETEC express colonization factors (CFs) that mediate adherence to the small intestine. An epidemiologically prevalent CF is coli surface antigen 6 (CS6). We assessed the safety and immunogenicity of a CS6-targeted candidate vaccine, CssBA, co-administered intramuscularly with the double-mutant heat-labile enterotoxin, dmLT [LT(R192G/L211A)]. METHODS This was an open-label trial. Fifty subjects received three intramuscular injections (Days 1, 22 and 43) of CssBA alone (5 µg), dmLT alone (0.1 µg) or CssBA (5, 15, 45 µg) + dmLT (0.1 and 0.5 µg). Subjects were actively monitored for adverse events for 28 days following the third vaccination. Antibody responses (IgG and IgA) were characterized in the serum and from lymphocyte supernatants (ALS) to CS6 and the native ETEC heat labile enterotoxin, LT. RESULTS Across all dose cohorts, the vaccine was safe and well-tolerated with no vaccine-related severe or serious adverse events. Among vaccine-related adverse events, a majority (98%) were mild with 79% being short-lived vaccine site reactions. Robust antibody responses were induced in a dose-dependent manner with a clear dmLT adjuvant effect. Response rates in subjects receiving 45 µg CssBA and 0.5 µg dmLT ranged from 50 to 100% across assays. CONCLUSION This is the first study to demonstrate the safety and immunogenicity of CssBA and/or dmLT administered intramuscularly. Co-administration of the two components induced robust immune responses to CS6 and LT, paving the way for future studies to evaluate the efficacy of this vaccine target and development of a multivalent, subunit ETEC vaccine.
Collapse
Affiliation(s)
- Tida Lee
- Enteric Diseases Department, Naval Medical Research Center, United States
| | - Ramiro L Gutiérrez
- Enteric Diseases Department, Naval Medical Research Center, United States
| | - Milton Maciel
- Enteric Diseases Department, Naval Medical Research Center, United States; Henry M. Jackson Foundation, United States
| | - Steven Poole
- Enteric Diseases Department, Naval Medical Research Center, United States; Henry M. Jackson Foundation, United States
| | - Kayla J Testa
- Enteric Diseases Department, Naval Medical Research Center, United States; Henry M. Jackson Foundation, United States
| | - Stefanie Trop
- Enteric Diseases Department, Naval Medical Research Center, United States; Henry M. Jackson Foundation, United States
| | | | - Alison Lane
- Uniformed Services University, United States
| | | | - Melinda Hamer
- Walter Reed Army Institute of Research, United States; Uniformed Services University, United States
| | - Ashley Alcala
- Enteric Diseases Department, Naval Medical Research Center, United States; Henry M. Jackson Foundation, United States
| | - Michael Prouty
- Enteric Diseases Department, Naval Medical Research Center, United States
| | | | | | | | - Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, United States.
| |
Collapse
|
16
|
Abstract
Mucosal vaccines offer the potential to trigger robust protective immune responses at the predominant sites of pathogen infection. In principle, the induction of adaptive immunity at mucosal sites, involving secretory antibody responses and tissue-resident T cells, has the capacity to prevent an infection from becoming established in the first place, rather than only curtailing infection and protecting against the development of disease symptoms. Although numerous effective mucosal vaccines are in use, the major advances seen with injectable vaccines (including adjuvanted subunit antigens, RNA and DNA vaccines) have not yet been translated into licensed mucosal vaccines, which currently comprise solely live attenuated and inactivated whole-cell preparations. The identification of safe and effective mucosal adjuvants allied to innovative antigen discovery and delivery strategies is key to advancing mucosal vaccines. Significant progress has been made in resolving the mechanisms that regulate innate and adaptive mucosal immunity and in understanding the crosstalk between mucosal sites, and this provides valuable pointers to inform mucosal adjuvant design. In particular, increased knowledge on mucosal antigen-presenting cells, innate lymphoid cell populations and resident memory cells at mucosal sites highlights attractive targets for vaccine design. Exploiting these insights will allow new vaccine technologies to be leveraged to facilitate rational mucosal vaccine design for pathogens including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and for cancer.
Collapse
|
17
|
Stone AE, Scheuermann SE, Haile CN, Cuny GD, Velasquez ML, Linhuber JP, Duddupudi AL, Vigliaturo JR, Pravetoni M, Kosten TA, Kosten TR, Norton EB. Fentanyl conjugate vaccine by injected or mucosal delivery with dmLT or LTA1 adjuvants implicates IgA in protection from drug challenge. NPJ Vaccines 2021; 6:69. [PMID: 33986280 PMCID: PMC8119695 DOI: 10.1038/s41541-021-00329-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
Fentanyl is a major contributor to the devastating increase in overdose deaths from substance use disorders (SUD). A vaccine targeting fentanyl could be a powerful immunotherapeutic. Here, we evaluated adjuvant and delivery strategies for conjugate antigen vaccination with fentanyl-based haptens. We tested adjuvants derived from the heat-labile toxin of E. coli including dmLT and LTA1 by intramuscular, sublingual or intranasal delivery. Our results show anti-fentanyl serum antibodies and antibody secreting cells in the bone-marrow after vaccination with highest levels observed with an adjuvant (alum, dmLT, or LTA1). Vaccine adjuvanted with LTA1 or dmLT elicited the highest levels of anti-fentanyl antibodies, whereas alum achieved highest levels against the carrier protein. Vaccination with sublingual dmLT or intranasal LTA1 provided the most robust blockade of fentanyl-induced analgesia and CNS penetration correlating strongly to anti-FEN IgA. In conclusion, this study demonstrates dmLT or LTA1 adjuvant as well as mucosal delivery may be attractive strategies for improving the efficacy of vaccines against SUD.
Collapse
Affiliation(s)
- Addison E Stone
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sarah E Scheuermann
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Colin N Haile
- Department of Psychology, University of Houston, Houston, TX, USA.,Texas Institute of Measurement Evaluation and Statistics, University of Houston, Houston, TX, USA
| | - Gregory D Cuny
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Marcela Lopez Velasquez
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Joshua P Linhuber
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Anantha L Duddupudi
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Jennifer R Vigliaturo
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Marco Pravetoni
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA.,Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Therese A Kosten
- Department of Psychology, University of Houston, Houston, TX, USA.,Texas Institute of Measurement Evaluation and Statistics, University of Houston, Houston, TX, USA
| | - Thomas R Kosten
- Department of Psychology, University of Houston, Houston, TX, USA.,Texas Institute of Measurement Evaluation and Statistics, University of Houston, Houston, TX, USA
| | - Elizabeth B Norton
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
18
|
Sublingual vaccination and delivery systems. J Control Release 2021; 332:553-562. [DOI: 10.1016/j.jconrel.2021.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 12/22/2022]
|
19
|
Hanson SM, Singh S, Tabet A, Sastry KJ, Barry M, Wang C. Mucoadhesive wafers composed of binary polymer blends for sublingual delivery and preservation of protein vaccines. J Control Release 2021; 330:427-437. [DOI: 10.1016/j.jconrel.2020.12.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/02/2020] [Accepted: 12/17/2020] [Indexed: 01/31/2023]
|
20
|
Preclinical optimization of an enterotoxigenic Escherichia coli adjuvanted subunit vaccine using response surface design of experiments. NPJ Vaccines 2020; 5:83. [PMID: 32983577 PMCID: PMC7486917 DOI: 10.1038/s41541-020-00228-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
Abstract
Enterotoxigenic E. coli (ETEC) is a leading cause of moderate-to-severe diarrhoea. ETEC colonizes the intestine through fimbrial tip adhesin colonization factors and produces heat-stable and/or heat-labile (LT) toxins, stimulating fluid and electrolyte release leading to watery diarrhoea. We reported that a vaccine containing recombinant colonization factor antigen (CfaEB) targeting fimbrial tip adhesin of the colonization factor antigen I (CFA/I) and an attenuated LT toxoid (dmLT) elicited mucosal and systemic immune responses against both targets. Additionally, the toll-like receptor 4 ligand second-generation lipid adjuvant (TLR4-SLA) induced a potent mucosal response, dependent on adjuvant formulation. However, a combination of vaccine components at their respective individual optimal doses may not achieve the optimal immune profile. We studied a subunit ETEC vaccine prototype in mice using a response surface design of experiments (DoE), consisting of 64 vaccine dose-combinations of CfaEB, dmLT and SLA in four formulations (aqueous, aluminium oxyhydroxide, squalene-in-water stable nanoemulsion [SE] or liposomes containing the saponin Quillaja saponaria-21 [LSQ]). Nine readouts focusing on antibody functionality and plasma cell response were selected to profile the immune response of parenterally administered ETEC vaccine prototype. The data were integrated in a model to identify the optimal dosage of each vaccine component and best formulation. Compared to maximal doses used in mouse models (10 µg CfaEB, 1 µg dmLT and 5 µg SLA), a reduction in the vaccine components up to 37%, 60% and 88% for CfaEB, dmLT and SLA, respectively, maintained or even maximized immune responses, with SE and LSQ the best formulations. The DoE approach can help determine the best vaccine composition with a limited number of experiments and may accelerate development of multi-antigen/component ETEC vaccines.
Collapse
|
21
|
U-Omp19 from Brucella abortus increases dmLT immunogenicity and improves protection against Escherichia coli heat-labile toxin (LT) oral challenge. Vaccine 2020; 38:5027-5035. [PMID: 32536545 PMCID: PMC7327514 DOI: 10.1016/j.vaccine.2020.05.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 01/18/2023]
Abstract
Oral co-administration of dmLT with U-Omp19 increases dmLT immunogenicity. U-Omp19 oral co-delivery with dmLT induces anti-LT antibody responses. U-Omp19 co-administered with dmLT protects against oral challenge with LT. U-Omp19 can allow antigen dose sparing by oral route. U-Omp19 can be used as adjuvant in an oral vaccine formulation against ETEC.
Acute diarrhea disease caused by bacterial infections is a major global health problem. Enterotoxigenic Escherichia coli (ETEC) is one of the top causes of diarrhea-associated morbidity and mortality in young children and travelers to low-income countries. There are currently no licensed vaccines for ETEC. Induction of immunity at the site of entry of the bacteria is key to prevent infection. Current approaches to ETEC vaccines include a less toxic mutant form of E. coli heat-labile toxin (double-mutant heat-labile enterotoxin -dmLT-) with both antigenic and immunostimulatory properties. U-Omp19 is a protease inhibitor from Brucella spp. with immunostimulatory properties that has been used as oral adjuvant. In this work, we use U-Omp19 as adjuvant in an oral vaccine formulation against ETEC containing dmLT in outbred and inbred mice. To evaluate antigen dose sparing by U-Omp19 three different immunization protocols with three different doses of dmLT were evaluated. We demonstrated that U-Omp19 co-delivery increases anti-LT IgA in feces using a mid-dose of dmLT following a prime-boost protocol (after one or two boosts). Oral immunization with U-Omp19 induced protection against LT challenge when co-formulated with dmLT in CD-1 and BALB/c mice. Indeed, there was a significant increase in anti-LT IgG and IgA avidity after a single oral administration of dmLT plus U-Omp19 in comparison with dmLT delivered alone. Interestingly, sera from dmLT plus U-Omp19 vaccinated mice significantly neutralize LT effect on intestine inflammation in vivo compared with sera from the group immunized with dmLT alone. These results demonstrate the adjuvant capacity of U-Omp19 to increase dmLT immunogenicity by the oral route and support its use in an oral subunit vaccine formulation against ETEC.
Collapse
|
22
|
Xi M, Yao Q, Ge W, Chen Y, Cao B, Wang Z, Cui X, Sun Q. Effects of stachyose on intestinal microbiota and immunity in mice infected with enterotoxigenic Escherichia coli. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
23
|
Intradermal or Sublingual Delivery and Heat-Labile Enterotoxin Proteins Shape Immunologic Responses to a CFA/I Fimbria-Derived Subunit Antigen Vaccine against Enterotoxigenic Escherichia coli. Infect Immun 2019; 87:IAI.00460-19. [PMID: 31427449 DOI: 10.1128/iai.00460-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/08/2019] [Indexed: 01/06/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of infectious diarrhea in children, travelers, and deployed military personnel. As such, development of a vaccine would be advantageous for public health. One strategy is to use subunits of colonization factors combined with antigen/adjuvant toxoids as an ETEC vaccine. Here, we investigated the intradermal (i.d.) or sublingual (s.l.) delivery of CFA/I fimbrial antigens, including CfaEB and a CfaE-heat-labile toxin B subunit (LTB) chimera admixed with double mutant heat-labile toxin (LT) LT-R192G/L211A (dmLT). In addition, we compared dmLT with other LT proteins to better understand the generation of adjuvanted fimbrial and toxoid immunity as well as the influence on any local skin reactogenicity. We demonstrate that immunization with dmLT admixed with CfaEB induces robust serum and fecal antibody responses to CFA/I fimbriae and LT but that i.d. formulations are not optimal for s.l. delivery. Improved s.l. vaccination outcomes were observed when higher doses of dmLT (1 to 5 μg) were admixed with CfaEB or, even better, when a CfaE-LTB chimera antigen was used instead. Serum anti-CFA/I total antibodies, detected by enzyme-linked immunosorbent assay, were the best predictor of functional antibodies, based on the inhibition of red blood cell agglutination by ETEC. Immunization with other LT proteins or formulations with altered B-subunit binding during i.d. immunization (e.g., by addition of 5% lactose, LTA1, or LT-G33D) minimally altered the development of antibody responses and cytokine recall responses but reduced skin reactogenicity at the injection site. These results reveal how formulations and delivery parameters shape the adaptive immune responses to a toxoid and fimbria-derived subunit vaccine against ETEC.
Collapse
|