1
|
Declercq J, Gerlo S, Van Nevel S, De Ruyck N, Holtappels G, Delesie L, Tobback E, Lammens I, Gerebtsov N, Sedeyn K, Saelens X, Lambrecht BN, Gevaert P, Vandekerckhove L, Vanhee S. Repeated COVID-19 mRNA-based vaccination contributes to SARS-CoV-2 neutralizing antibody responses in the mucosa. Sci Transl Med 2024; 16:eadn2364. [PMID: 39441904 DOI: 10.1126/scitranslmed.adn2364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
To prevent infection by respiratory viruses and consequently limit virus circulation, vaccines need to promote mucosal immunity. The extent to which the currently used messenger RNA (mRNA)-based COVID-19 vaccines induce mucosal immunity remains poorly characterized. We evaluated mucosal neutralizing antibody responses in a cohort of 183 individuals. Participants were sampled at several time points after primary adenovirus vector-based or mRNA-based COVID-19 vaccination and after mRNA-based booster vaccinations. Our findings revealed that repeated vaccination with mRNA boosters promoted severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing antibodies in nasal secretions. Nasal and serum neutralizing antibody titers of both IgG and IgA isotypes correlated to one another. We investigated the source of these mucosal antibodies in a mouse model wherein mice received repeated mRNA vaccines for SARS-CoV-2. These experiments indicated that neutralizing antibody-producing cells reside in the spleen and bone marrow, whereas no proof of tissue homing to the respiratory mucosa was observed, despite the detection of mucosal antibodies. Serum transfer experiments confirmed that circulating antibodies were able to migrate to the respiratory mucosa. Collectively, these results demonstrate that, especially upon repeated vaccination, the currently used COVID-19 mRNA vaccines can elicit mucosal neutralizing antibodies and that vaccination might also stimulate mucosal immunity induced by previous SARS-CoV-2 infection. Moreover, migration of circulating antibodies to the respiratory mucosa might be a main mechanism. These findings advance our understanding of mRNA vaccine-induced immunity and have implications for the design of vaccine strategies to combat respiratory infections.
Collapse
Affiliation(s)
- Jozefien Declercq
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Sarah Gerlo
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Sharon Van Nevel
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Natalie De Ruyck
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Gabriele Holtappels
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Liesbeth Delesie
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Els Tobback
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Inés Lammens
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Nikita Gerebtsov
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Koen Sedeyn
- VIB Center for Medical Biotechnology, 9000 Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
| | - Xavier Saelens
- VIB Center for Medical Biotechnology, 9000 Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus Medical Center, 3015 GD Rotterdam, Netherlands
| | - Philippe Gevaert
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Linos Vandekerckhove
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Stijn Vanhee
- Laboratory of Mucosal Immunology, VIB Center for Inflammation Research, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
- Upper Airways Research Laboratory, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
2
|
Tian T, Zhu Y, Shi J, Shang K, Yin Z, Shi H, He Y, Ding J, Zhang F. The development of a human Brucella mucosal vaccine: What should be considered? Life Sci 2024; 355:122986. [PMID: 39151885 DOI: 10.1016/j.lfs.2024.122986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Brucellosis is a chronic infectious disease that is zoonotic in nature. Brucella can infect humans through interactions with livestock, primarily via the digestive tract, respiratory tract, and oral cavity. This bacterium has the potential to be utilized as a biological weapon and is classified as a Category B pathogen by the Centers for Disease Control and Prevention. Currently, there is no approved vaccine for humans against Brucella, highlighting an urgent need for the development of a vaccine to mitigate the risks posed by this pathogen. Brucella primarily infects its host by adhering to and penetrating mucosal surfaces. Mucosal immunity plays a vital role in preventing local infections, clearing microorganisms from mucosal surfaces, and inhibiting the spread of pathogens. As mucosal vaccine strategies continue to evolve, the development of a safe and effective mucosal vaccine against Brucella appears promising.This paper reviews the immune mechanism of mucosal vaccines, the infection mechanism of Brucella, successful Brucella mucosal vaccines in animals, and mucosal adjuvants. Additionally, it elucidates targeting and optimization strategies for mucosal vaccines to facilitate the development of human vaccines against Brucella.
Collapse
Affiliation(s)
- Tingting Tian
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Yuejie Zhu
- Reproductive Fertility Assistance Center, First Afffliated Hospital of Xinjiang Medical University, China
| | - Juan Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Kaiyu Shang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Zhengwei Yin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Huidong Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Yueyue He
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Jianbing Ding
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China
| | - Fengbo Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated hospital of Xinjiang Medical University, China; Department of Clinical laboratory, The First Affiliated hospital of Xinjiang Medical University, China.
| |
Collapse
|
3
|
Shi Q, Wang Q, Shen Y, Chen S, Gan S, Lin T, Song F, Ma Y. Escherichia coli LTB26 mutant enhances immune responses to rotavirus antigen VP8 in a mouse model. Mol Immunol 2024; 173:10-19. [PMID: 39004021 DOI: 10.1016/j.molimm.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 06/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Adjuvant is a major supplementary component of vaccines to boost adaptive immune responses. To select an efficient adjuvant from the heat-labile toxin B subunit (LTB) of E. coli, four LTB mutants (numbered LTB26, LTB34, LTB57, and LTB85) were generated by multi-amino acid random replacement. Mice have been intranasally vaccinated with human rotavirus VP8 admixed. Among the four mutants, enzyme-linked immunosorbent assay (ELISA) revealed that LTB26 had enhanced mucosal immune adjuvanticity compared to LTB, showing significantly enhanced immune responses in both serum IgG and mucosal sIgA levels. The 3D modeling analysis suggested that the enhanced immune adjuvanticity of LTB26 might be due to the change of the first LTB α-helix to a β-sheet. The molecular mechanism was studied using transcriptomic and flow cytometric (FCM) analysis. The transcriptomic data demonstrated that LTB26 enhanced immune response by enhancing B cell receptor (BCR) and major histocompatibility complex (MHC) II+-related pathways. Furthermore, LTB26 promoted Th1 and Th2-type immune responses which were confirmed by detecting IFN-γ and IL-4 expression levels. Immunohistochemical analysis demonstrated that LTB26 enhanced both Th1 and Th2 type immunity. Therefore, LTB26 was a potent mucosal immune adjuvant meeting the requirement for use in human clinics in the future.
Collapse
Affiliation(s)
- Qinlin Shi
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qiujuan Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yanxi Shen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijing Chen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijie Gan
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Tao Lin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Fangzhou Song
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yongping Ma
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China.
| |
Collapse
|
4
|
Berndsen ZT, Akhtar M, Thapa M, Vickers TJ, Schmitz A, Torres JL, Baboo S, Kumar P, Khatoon N, Sheikh A, Hamrick M, Diedrich JK, Martinez-Bartolome S, Garrett PT, Yates JR, Turner JS, Laird RM, Poly F, Porter CK, Copps J, Ellebedy AH, Ward AB, Fleckenstein JM. Repeat modules and N-linked glycans define structure and antigenicity of a critical enterotoxigenic E. coli adhesin. PLoS Pathog 2024; 20:e1012241. [PMID: 39283948 PMCID: PMC11463764 DOI: 10.1371/journal.ppat.1012241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/09/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of cases of infectious diarrhea annually, predominantly in children from low-middle income regions. Notably, in children, as well as volunteers challenged with ETEC, diarrheal severity is significantly increased in blood group A (bgA) individuals. EtpA, is a secreted glycoprotein adhesin that functions as a blood group A lectin to promote critical interactions between ETEC and blood group A glycans on intestinal epithelia for effective bacterial adhesion and toxin delivery. EtpA is highly immunogenic resulting in robust antibody responses following natural infection and experimental challenge of volunteers with ETEC. To understand how EtpA directs ETEC-blood group A interactions and stimulates adaptive immunity, we mutated EtpA, mapped its glycosylation by mass-spectrometry (MS), isolated polyclonal (pAbs) and monoclonal antibodies (mAbs) from vaccinated mice and ETEC-infected volunteers, and determined structures of antibody-EtpA complexes by cryo-electron microscopy. Both bgA and mAbs that inhibited EtpA-bgA interactions and ETEC adhesion, bound to the C-terminal repeat domain highlighting this region as crucial for ETEC pathogen-host interaction. MS analysis uncovered extensive and heterogeneous N-linked glycosylation of EtpA and cryo-EM structures revealed that mAbs directly engage these unique glycan containing epitopes. Finally, electron microscopy-based polyclonal epitope mapping revealed antibodies targeting numerous distinct epitopes on N and C-terminal domains, suggesting that EtpA vaccination generates responses against neutralizing and decoy regions of the molecule. Collectively, we anticipate that these data will inform our general understanding of pathogen-host glycan interactions and adaptive immunity relevant to rational vaccine subunit design.
Collapse
Affiliation(s)
- Zachary T. Berndsen
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Marjahan Akhtar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Mahima Thapa
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Tim J. Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Aaron Schmitz
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Jonathan L. Torres
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Sabyasachi Baboo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Pardeep Kumar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Nazia Khatoon
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Melissa Hamrick
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Jolene K. Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Salvador Martinez-Bartolome
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Patrick T. Garrett
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - John R. Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jackson S. Turner
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Renee M. Laird
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Frédéric Poly
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Chad K. Porter
- Translational and Clinical Research Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, Missouri, United States of America
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - James M. Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
- Medicine Service, Infectious Diseases, Veterans Affairs Health Care System, Saint Louis, Missouri, United States of America
| |
Collapse
|
5
|
Zhang B, Chen S, Yin X, McBride CD, Gertie JA, Yurieva M, Bielecka AA, Hoffmann B, Travis Hinson J, Grassmann J, Xu L, Siniscalco ER, Soldatenko A, Hoyt L, Joseph J, Norton EB, Uthaman G, Palm NW, Liu E, Eisenbarth SC, Williams A. Metabolic fitness of IgA + plasma cells in the gut requires DOCK8. Mucosal Immunol 2024; 17:431-449. [PMID: 38159726 PMCID: PMC11571232 DOI: 10.1016/j.mucimm.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/16/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Dedicator of cytokinesis 8 (DOCK8) mutations lead to a primary immunodeficiency associated with recurrent gastrointestinal infections and poor antibody responses but, paradoxically, heightened IgE to food antigens, suggesting that DOCK8 is central to immune homeostasis in the gut. Using Dock8-deficient mice, we found that DOCK8 was necessary for mucosal IgA production to multiple T cell-dependent antigens, including peanut and cholera toxin. Yet DOCK8 was not necessary in T cells for this phenotype. Instead, B cell-intrinsic DOCK8 was required for maintenance of antigen-specific IgA-secreting plasma cells (PCs) in the gut lamina propria. Unexpectedly, DOCK8 was not required for early B cell activation, migration, or IgA class switching. An unbiased interactome screen revealed novel protein partners involved in metabolism and apoptosis. Dock8-deficient IgA+ B cells had impaired cellular respiration and failed to engage glycolysis appropriately. These results demonstrate that maintenance of the IgA+ PC compartment requires DOCK8 and suggest that gut IgA+ PCs have unique metabolic requirements for long-term survival in the lamina propria.
Collapse
Affiliation(s)
- Biyan Zhang
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Shuting Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiangyun Yin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Caleb D McBride
- The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jake A Gertie
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marina Yurieva
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Agata A Bielecka
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Microbial Immunoregulation, Helmholtz Center for Infection Research, 38124 Braunschweig, Germany
| | - Brian Hoffmann
- Mass Spectrometry and Protein Chemistry, The Jackson Laboratory for Genomic Medicine, Bar Harbor, ME 04609, USA
| | - J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Cardiology center, Department of Medicine, UConn Health, Farmington, CT, USA
| | - Jessica Grassmann
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Lan Xu
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Emily R Siniscalco
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Arielle Soldatenko
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Laura Hoyt
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Julie Joseph
- Department of Laboratory Medicine, USA; Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Elizabeth B Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Gowthaman Uthaman
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Elise Liu
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Adam Williams
- The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
6
|
Wellford SA, Moseman EA. Olfactory immunology: the missing piece in airway and CNS defence. Nat Rev Immunol 2024; 24:381-398. [PMID: 38097777 PMCID: PMC11560121 DOI: 10.1038/s41577-023-00972-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2023] [Indexed: 12/23/2023]
Abstract
The olfactory mucosa is a component of the nasal airway that mediates the sense of smell. Recent studies point to an important role for the olfactory mucosa as a barrier to both respiratory pathogens and to neuroinvasive pathogens that hijack the olfactory nerve and invade the CNS. In particular, the COVID-19 pandemic has demonstrated that the olfactory mucosa is an integral part of a heterogeneous nasal mucosal barrier critical to upper airway immunity. However, our insufficient knowledge of olfactory mucosal immunity hinders attempts to protect this tissue from infection and other diseases. This Review summarizes the state of olfactory immunology by highlighting the unique immunologically relevant anatomy of the olfactory mucosa, describing what is known of olfactory immune cells, and considering the impact of common infectious diseases and inflammatory disorders at this site. We will offer our perspective on the future of the field and the many unresolved questions pertaining to olfactory immunity.
Collapse
Affiliation(s)
- Sebastian A Wellford
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - E Ashley Moseman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
7
|
Berndsen ZT, Akhtar M, Thapa M, Vickers T, Schmitz A, Torres JL, Baboo S, Kumar P, Khatoom N, Sheikh A, Hamrick M, Diedrich JK, Martinez-Bartolome S, Garrett PT, Yates JR, Turner JS, Laird RM, Poly F, Porter CK, Copps J, Ellebedy AH, Ward AB, Fleckenstein JM. Repeat modules and N-linked glycans define structure and antigenicity of a critical enterotoxigenic E. coli adhesin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593125. [PMID: 38766097 PMCID: PMC11100705 DOI: 10.1101/2024.05.08.593125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of cases of infectious diarrhea annually, predominantly in children from low-middle income regions. Notably, in children, as well as human volunteers challenged with ETEC, diarrheal severity is significantly increased severity in blood group A (bgA) individuals. EtpA, is a secreted glycoprotein adhesin that functions as a blood group A lectin to promote critical interactions between ETEC and blood group A glycans on intestinal epithelia for effective bacterial adhesion and toxin delivery. EtpA is highly immunogenic resulting in robust antibody responses following natural infection and experimental challenge of human volunteers with ETEC. To understand how EtpA directs ETEC-blood group A interactions and stimulates adaptive immunity, we mutated EtpA, mapped its glycosylation by mass-spectrometry (MS), isolated polyclonal (pAbs) and monoclonal antibodies (mAbs) from vaccinated mice and ETEC-infected human volunteers, and determined structures of antibody-EtpA complexes by cryo-electron microscopy. Both bgA and mAbs that inhibited EtpA-bgA interactions and ETEC adhesion, bound to the C-terminal repeat domain highlighting this region as crucial for ETEC pathogen-host interaction. MS analysis uncovered extensive and heterogeneous N-linked glycosylation of EtpA and cryo-EM structures revealed that mAbs directly engage these unique glycan containing epitopes. Finally, electron microscopy-based polyclonal epitope mapping revealed antibodies targeting numerous distinct epitopes on N and C-terminal domains, suggesting that EtpA vaccination generates responses against neutralizing and decoy regions of the molecule. Collectively, we anticipate that these data will inform our general understanding of pathogen-host glycan interactions and adaptive immunity relevant to rational vaccine subunit design.
Collapse
Affiliation(s)
- Zachary T Berndsen
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Marjahan Akhtar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Mahima Thapa
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Tim Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Aaron Schmitz
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Jonathan L Torres
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Sabyasachi Baboo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Pardeep Kumar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Nazia Khatoom
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Melissa Hamrick
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Jolene K Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Patrick T Garrett
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Renee M Laird
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
| | - Frédéric Poly
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
| | - Chad K Porter
- Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - 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, Veterans Affairs Health Care System, Saint Louis Missouri, USA
| |
Collapse
|
8
|
Abstract
Substance use disorders (SUD) present a worldwide challenge with few effective therapies except for the relative efficacy of opioid pharmacotherapies, despite limited treatment access. However, the proliferation of illicit fentanyl use initiated a dramatic and cascading epidemic of lethal overdoses. This rise in fentanyl overdoses regenerated an interest in vaccine immunotherapy, which, despite an optimistic start in animal models over the past 50 years, yielded disappointing results in human clinical trials of vaccines against nicotine, stimulants (cocaine and methamphetamine), and opioids. After a brief review of clinical and selected preclinical vaccine studies, the "lessons learned" from the previous vaccine clinical trials are summarized, and then the newest challenge of a vaccine against fentanyl and its analogs is explored. Animal studies have made significant advances in vaccine technology for SUD treatment over the past 50 years, and the resulting anti-fentanyl vaccines show remarkable promise for ending this epidemic of fentanyl deaths.
Collapse
Affiliation(s)
- Thomas R Kosten
- Waggoner Professor of Psychiatry, Pharmacology, Neuroscience, Immunology, Baylor College of Medicine, Houston
| |
Collapse
|
9
|
Liu Y, Lam DMK, Luan M, Zheng W, Ai H. Recent development of oral vaccines (Review). Exp Ther Med 2024; 27:223. [PMID: 38590568 PMCID: PMC11000446 DOI: 10.3892/etm.2024.12511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/08/2024] [Indexed: 04/10/2024] Open
Abstract
Oral immunization can elicit an effective immune response and immune tolerance to specific antigens. When compared with the traditional injection route, delivering antigens via the gastrointestinal mucosa offers superior immune effects and compliance, as well as simplicity and convenience, making it a more optimal route for immunization. At present, various oral vaccine delivery systems exist. Certain modified bacteria, such as Salmonella, Escherichia coli and particularly Lactobacillus, are considered promising carriers for oral vaccines. These carriers can significantly enhance immunization efficiency by actively replicating in the intestinal tract following oral administration. The present review provided a discussion of the main mechanisms of oral immunity and the research progress made in the field of oral vaccines. Additionally, it introduced the advantages and disadvantages of the currently more commonly administered injectable COVID-19 vaccines, alongside the latest advancements in this area. Furthermore, recent developments in oral vaccines are summarized, and their potential benefits and side effects are discussed.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Follicular Development and Reproductive Health in Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | | | - Mei Luan
- Department of Geriatric Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Wenfu Zheng
- Chinese Academy of Sciences Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hao Ai
- Key Laboratory of Follicular Development and Reproductive Health in Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
10
|
Norton E. Is there a role for vaccines in combatting the opioid epidemic? VACCINE INSIGHTS 2024; 3:83-89. [PMID: 39156761 PMCID: PMC11329228 DOI: 10.18609/vac.2024.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Fentanyl is at the center of the opioid crisis in the USA, causing an increasing number of overdoses and deaths. Casey Nevins, Assistant Editor, Vaccine Insights, speaks with Elizabeth Norton, Associate Professor, Tulane School of Medicine, about her work in developing a mucosal vaccination tailored to protect the brain from the effects of fentanyl.
Collapse
|
11
|
Sutter RW, Eisenhawer M, Molodecky NA, Verma H, Okayasu H. Inactivated Poliovirus Vaccine: Recent Developments and the Tortuous Path to Global Acceptance. Pathogens 2024; 13:224. [PMID: 38535567 PMCID: PMC10974833 DOI: 10.3390/pathogens13030224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/21/2024] Open
Abstract
Inactivated poliovirus vaccine (IPV), available since 1955, became the first vaccine to be used to protect against poliomyelitis. While the immunogenicity of IPV to prevent paralytic poliomyelitis continues to be irrefutable, its requirement for strong containment (due to large quantities of live virus used in the manufacturing process), perceived lack of ability to induce intestinal mucosal immunity, high cost and increased complexity to administer compared to oral polio vaccine (OPV), have limited its use in the global efforts to eradicate poliomyelitis. In order to harvest the full potential of IPV, a program of work has been carried out by the Global Polio Eradication Initiative (GPEI) over the past two decades that has focused on: (1) increasing the scientific knowledge base of IPV; (2) translating new insights and evidence into programmatic action; (3) expanding the IPV manufacturing infrastructure for global demand; and (4) continuing to pursue an ambitious research program to develop more immunogenic and safer-to-produce vaccines. While the knowledge base of IPV continues to expand, further research and product development are necessary to ensure that the program priorities are met (e.g., non-infectious production through virus-like particles, non-transmissible vaccine inducing humoral and intestinal mucosal immunity and new methods for house-to-house administration through micro-needle patches and jet injectors), the discussions have largely moved from whether to how to use this vaccine most effectively. In this review, we summarize recent developments on expanding the science base of IPV and provide insight into policy development and the expansion of IPV manufacturing and production, and finally we provide an update on the current priorities.
Collapse
Affiliation(s)
| | - Martin Eisenhawer
- Polio Eradication Department, World Health Organization, 1211 Geneva, Switzerland; (M.E.); (H.V.)
| | - Natalia A. Molodecky
- Polio Surge Capacity Support Program, The Task Force for Global Health, Inc., Decatur, GE 30030, USA;
| | - Harish Verma
- Polio Eradication Department, World Health Organization, 1211 Geneva, Switzerland; (M.E.); (H.V.)
| | - Hiromasa Okayasu
- Division of Healthy Environments and Population, Regional Office for the Western Pacific, World Health Organization, Manila 1000, Philippines
| |
Collapse
|
12
|
Desalegn G, Tamilselvi CS, Lemme-Dumit JM, Heine SJ, Dunn D, Ndungo E, Kapoor N, Oaks EV, Fairman J, Pasetti MF. Shigella virulence protein VirG is a broadly protective antigen and vaccine candidate. NPJ Vaccines 2024; 9:2. [PMID: 38167387 PMCID: PMC10761965 DOI: 10.1038/s41541-023-00797-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Diarrhea caused by Shigella has been associated with high morbidity and mortality in young children worldwide. There are no licensed vaccines, and those clinically advanced have restricted coverage as they elicit serotype-specific immunity while disease is caused by multiple circulating serotypes. Our group had previously reported a close association between serum antibodies to the Shigella virulence factor VirG (or IcsA) and clinical protection in infected individuals. VirG is highly conserved among Shigella strains and appealing as a broad-spectrum vaccine candidate. In this study, we investigated the immunogenicity and protective capacity of VirG as a subunit vaccine in mice. The surface-exposed alpha (α) domain of VirG (VirGα) was produced as a recombinant protein. This region has almost identical immune reactivity to full-length VirG. Administered intramuscularly with alum, VirGα elicited robust immune responses and high protective efficacy against S. flexneri 2a and S. sonnei. Almost complete protection was afforded by VirGα given intranasally with the E. coli double mutant heat-labile toxin (dmLT). VirGα-specific antibodies recognized VirG expressed on live Shigella, and blocked Shigella adhesion and invasion to human colonic cells. These results show for the first time that VirGα is a promising cross-protective vaccine candidate to prevent Shigella infection.
Collapse
Affiliation(s)
- Girmay Desalegn
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Chitradevi S Tamilselvi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Jose M Lemme-Dumit
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Shannon J Heine
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Dylan Dunn
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Esther Ndungo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Neeraj Kapoor
- Vaxcyte, Inc., 825 Industrial Road, San Carlos, CA, 94070, USA
| | - Edwin V Oaks
- Patuxent Research and Consulting Group, 3106 Arrowhead Farm Rd, Gambrills, MD, 21054, USA
| | - Jeff Fairman
- Vaxcyte, Inc., 825 Industrial Road, San Carlos, CA, 94070, USA
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685W. Baltimore Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
13
|
Ma J, Hermans L, Dierick M, Van der Weken H, Cox E, Devriendt B. Enterotoxigenic Escherichia coli heat labile enterotoxin affects neutrophil effector functions via cAMP/PKA/ERK signaling. Gut Microbes 2024; 16:2399215. [PMID: 39284098 PMCID: PMC11407407 DOI: 10.1080/19490976.2024.2399215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are a major cause of diarrheal illness in humans and animals, induced by enterotoxins produced by these pathogens. Despite the crucial role of neutrophils in combatting bacterial infections, our understanding of how enterotoxins impact neutrophil function is limited. To address this knowledge gap, we used heat-labile enterotoxin (LT) and heat-stable enterotoxin a (STa) to investigate their impact on the effector functions of neutrophils. Our study reveals that pSTa does not exert any discernible effect on the function of neutrophils. In contrast, LT altered the migration and phagocytosis of neutrophils and induced the production of inflammatory factors via activation of cAMP/PKA and ERK1/2 signaling. LT also attenuated the release of neutrophil extracellular traps by neutrophils via the PKA signaling pathway. Our findings provide novel insights into the impact of LT on neutrophil function, shedding light on the underlying mechanisms that govern its immunoregulatory effects. This might help ETEC in subverting the immune system and establishing infection.
Collapse
Affiliation(s)
- Jinglin Ma
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Leen Hermans
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Matthias Dierick
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hans Van der Weken
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eric Cox
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bert Devriendt
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
14
|
Hossain MJ, Svennerholm AM, Carlin N, D’Alessandro U, Wierzba TF. A Perspective on the Strategy for Advancing ETVAX ®, An Anti-ETEC Diarrheal Disease Vaccine, into a Field Efficacy Trial in Gambian Children: Rationale, Challenges, Lessons Learned, and Future Directions. Microorganisms 2023; 12:90. [PMID: 38257916 PMCID: PMC10819518 DOI: 10.3390/microorganisms12010090] [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: 11/15/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
For the first time in over 20 years, an Enterotoxigenic Escherichia coli (ETEC) vaccine candidate, ETVAX®, has advanced into a phase 2b field efficacy trial for children 6-18 months of age in a low-income country. ETVAX® is an inactivated whole cell vaccine that has gone through a series of clinical trials to provide a rationale for the design elements of the Phase 2b trial. This trial is now underway in The Gambia and will be a precursor to an upcoming pivotal phase 3 trial. To reach this point, numerous findings were brought together to define factors such as safe and immunogenic doses for children, and the possible benefit of a mucosal adjuvant, double mutant labile toxin (dmLT). Considering the promising but still underexplored potential of inactivated whole cells in oral vaccination, we present a perspective compiling key observations from past ETVAX® trials that informed The Gambian trial design. This report will update the trial's status and explore future directions for ETEC vaccine trials. Our aim is to provide not only an update on the most advanced ETEC vaccine candidate but also to offer insights beneficial for the development of other much-needed oral whole-cell vaccines against enteric and other pathogens.
Collapse
Affiliation(s)
- M. Jahangir Hossain
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul P.O. Box 273, The Gambia
| | - Ann-Mari Svennerholm
- Department of Microbiology and Immunology, Gothenburg University Research Institute (GUVAX), Gothenburg University, 40530 Gothenburg, Sweden
| | - Nils Carlin
- Scandinavian Biopharma, Industrivägen 1, 17148 Solna, Sweden
| | - Umberto D’Alessandro
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul P.O. Box 273, The Gambia
| | - Thomas F. Wierzba
- Section on Infectious Diseases, Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| |
Collapse
|
15
|
Zhou S, Yu KOA, Mabrouk MT, Jahagirdar D, Huang WC, Guerra JA, He X, Ortega J, Poole ST, Hall ER, Gomez-Duarte OG, Maciel M, Lovell JF. Antibody induction in mice by liposome-displayed recombinant enterotoxigenic Escherichia coli (ETEC) colonization antigens. Biomed J 2023; 46:100588. [PMID: 36925108 PMCID: PMC10711177 DOI: 10.1016/j.bj.2023.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
BACKGROUND Enterotoxigenic Escherichia coli (ETEC) strains cause infectious diarrhea and colonize host intestine epithelia via surface-expressed colonization factors. Colonization factor antigen I (CFA/I), a prevalent ETEC colonization factor, is a vaccine target since antibodies directed to this fimbria can block ETEC adherence and prevent diarrhea. METHODS Two recombinant antigens derived from CFA/I were investigated with a vaccine adjuvant system that displays soluble antigens on the surface of immunogenic liposomes. The first antigen, CfaEB, is a chimeric fusion protein comprising the minor (CfaE) and major (CfaB) subunits of CFA/I. The second, CfaEad, is the adhesin domain of CfaE. RESULTS Owing to their His-tag, recombinant CfaEB and CfaEad, spontaneously bound upon admixture with nanoliposomes containing cobalt-porphyrin phospholipid (CoPoP), as well as a synthetic monophosphoryl lipid A (PHAD) adjuvant. Intramuscular immunization of mice with sub-microgram doses CfaEB or CfaEad admixed with CoPoP/PHAD liposomes elicited serum IgG and intestinal IgA antibodies. The smaller CfaEad antigen benefitted more from liposome display. Serum and intestine antibodies from mice immunized with liposome-displayed CfaEB or CfaEad recognized native CFA/I fimbria as evidenced by immunofluorescence and hemagglutination inhibition assays using the CFA/I-expressing H10407 ETEC strain. CONCLUSION These data show that colonization factor-derived recombinant ETEC antigens exhibit immunogenicity when delivered in immunogenic particle-based formulations.
Collapse
Affiliation(s)
- Shiqi Zhou
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, USA
| | - Karl O A Yu
- Division of Pediatrics Infectious Diseases, Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - Moustafa T Mabrouk
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, USA
| | | | - Wei-Chiao Huang
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, USA
| | - Julio A Guerra
- Division of Pediatrics Infectious Diseases, Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - Xuedan He
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, USA
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Steven T Poole
- Naval Medical Research Center, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Eric R Hall
- Naval Medical Research Center, Silver Spring, MD, USA
| | - Oscar G Gomez-Duarte
- Division of Pediatrics Infectious Diseases, Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - Milton Maciel
- Naval Medical Research Center, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA; Department of Microbiology and Immunology, Uniformed Services University Health System, Bethesda, MD, USA.
| | - Jonathan F Lovell
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
16
|
Crothers JW, Norton EB. Recent advances in enterotoxin vaccine adjuvants. Curr Opin Immunol 2023; 85:102398. [PMID: 37976963 PMCID: PMC11258862 DOI: 10.1016/j.coi.2023.102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/21/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
Enterotoxin adjuvants have been researched for their ability to promote immunity to co-delivered antigens. Outside of cholera vaccines, however, these proteins have yet to be included in any currently licensed vaccines. They include molecules derived from the bacterial toxins of Vibrio cholerae, cholera toxin, or Escherichia coli, heat-labile toxin, such as detoxified mutants or subunits. This class of adjuvants is distinguished by their delivery possibilities, which include parenteral injection, skin applications, or direct mucosal administration by oral, sublingual, or nasal routes. In addition, inclusion of an enterotoxin adjuvant is associated with development of multifaceted cellular and humoral immune responses to vaccination. Here, we review exciting progress in the past few years in clinical trials for safety and efficacy, preclinical vaccines studies, and new mechanistic insights for enterotoxin adjuvants. This includes recent reports of their use in vaccines targeting microbial infections (bacterial, viral, parasitic) or substance abuse drugs.
Collapse
Affiliation(s)
- Jessica W Crothers
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | | |
Collapse
|
17
|
Hsieh HC, Chen CC, Chou PH, Liu WC, Wu SC. Induction of neutralizing antibodies and mucosal IgA through intranasal immunization with the receptor binding domain of SARS-CoV-2 spike protein fused with the type IIb E. coli heat-labile enterotoxin A subunit. Antiviral Res 2023; 220:105752. [PMID: 37949318 DOI: 10.1016/j.antiviral.2023.105752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
The outbreak of SARS-CoV-2 infections had led to the COVID-19 pandemic which has a significant impact on global public health and the economy. The spike (S) protein of SARS-CoV-2 contains the receptor binding domain (RBD) which binds to human angiotensin-converting enzyme 2 receptor. Numerous RBD-based vaccines have been developed and recently focused on the induction of neutralizing antibodies against the immune evasive Omicron BQ.1.1 and XBB.1.5 subvariants. In this preclinical study, we reported the use of a direct fusion of the type IIb Escherichia coli heat-labile enterotoxin A subunit with SARS CoV-2 RBD protein (RBD-LTA) as an intranasal vaccine candidate. The results showed that intranasal immunization with the RBD-LTA fusion protein in BALB/c mice elicited potent neutralizing antibodies against the Wuhan-Hu-1 and several SARS-CoV-2 variants as well as the production of IgA antibodies in bronchoalveolar lavage fluids (BALFs). Furthermore, the heterologous RBD representing the same strains used in the bivalent mRNA vaccine were used as a second-dose RBD-LTA/RBD protein booster after bivalent mRNA vaccination. The results showed that the neutralizing antibody titers elicited by the intranasal bivalent RBD-LTA/RBD protein booster were similar to the intramuscular bivalent mRNA booster, but the RBD-specific IgA titers in sera and BALFs significantly increased. Overall, this preclinical study suggests that the RBD-LTA fusion protein could be a promising candidate as a mucosal booster COVID-19 vaccine.
Collapse
Affiliation(s)
- He-Chin Hsieh
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Chung-Chu Chen
- Department of Internal Medicine, MacKay Memorial Hospital, Hsinchu, 30071, Taiwan; Teaching Center of Natural Science, Minghsin University of Science and Technology, Hsinchu, 30401, Taiwan.
| | - Pin-Han Chou
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Wen-Chun Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11529, Taiwan.
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan; Adimmune Corporation, Taichung, 42723, Taiwan.
| |
Collapse
|
18
|
Kantele A, Riekkinen M, Jokiranta TS, Pakkanen SH, Pietilä JP, Patjas A, Eriksson M, Khawaja T, Klemets P, Marttinen K, Siikamäki H, Lundgren A, Holmgren J, Lissmats A, Carlin N, Svennerholm AM. Safety and immunogenicity of ETVAX®, an oral inactivated vaccine against enterotoxigenic Escherichia coli diarrhoea: a double-blinded, randomized, placebo-controlled trial amongst Finnish travellers to Benin, West Africa. J Travel Med 2023; 30:taad045. [PMID: 37099803 PMCID: PMC10658657 DOI: 10.1093/jtm/taad045] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/31/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND No licensed human vaccines are available against enterotoxigenic Escherichia coli (ETEC), a major diarrhoeal pathogen affecting children in low- and middle-income countries and foreign travellers alike. ETVAX®, a multivalent oral whole-cell vaccine containing four inactivated ETEC strains and the heat-labile enterotoxin B subunit (LTB), has proved promising in Phase 1 and Phase 1/ 2 studies. METHODS We conducted a Phase 2b double-blinded, randomized, placebo-controlled trial amongst Finnish travellers to Benin, West Africa. This report presents study design and safety and immunogenicity data. Volunteers aged 18-65 years were randomized 1:1 to receive ETVAX® or placebo. They visited Benin for 12 days, provided stool and blood samples and completed adverse event (AE) forms. IgA and IgG antibodies to LTB and O78 lipopolysaccharide (LPS) were measured by electrochemiluminescence. RESULTS The AEs did not differ significantly between vaccine (n = 374) and placebo (n = 375) recipients. Of the solicited AEs, loose stools/diarrhoea (26.7/25.9%) and stomach ache (23.0/20.0%) were reported most commonly. Of all possibly/probably vaccine-related AEs, the most frequent were gastrointestinal symptoms (54.0/48.8%) and nervous system disorders (20.3/25.1%). Serious AEs were recorded for 4.3/5.6%, all unlikely to be vaccine related. Amongst the ETVAX® recipients, LTB-specific IgA antibodies increased 22-fold. For the 370/372 vaccine/placebo recipients, the frequency of ≥2-fold increases against LTB was 81/2.4%, and against O78 LPS 69/2.7%. The majority of ETVAX® recipients (93%) responded to either LTB or O78. CONCLUSIONS This Phase 2b trial is the largest on ETVAX® undertaken amongst travellers to date. ETVAX® showed an excellent safety profile and proved strongly immunogenic, which encourages the further development of this vaccine.
Collapse
Affiliation(s)
- Anu Kantele
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
- Travel Clinic, Aava Medical Center, Helsinki, Finland
| | - Marianna Riekkinen
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
- Travel Clinic, Aava Medical Center, Helsinki, Finland
| | - T Sakari Jokiranta
- United Medix Laboratories/Synlab Finland Ltd, Helsinki, Finland
- Medicum, Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Mobidiag Ltd, Espoo, Finland
| | - Sari H Pakkanen
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
| | - Jukka-Pekka Pietilä
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
- Travel Clinic, Aava Medical Center, Helsinki, Finland
| | - Anu Patjas
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
- Travel Clinic, Aava Medical Center, Helsinki, Finland
| | - Mari Eriksson
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
| | - Tamim Khawaja
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
| | - Peter Klemets
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
| | - Kati Marttinen
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
| | - Heli Siikamäki
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
| | - Anna Lundgren
- Gothenburg University Vaccine Research Institute, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Holmgren
- Gothenburg University Vaccine Research Institute, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Ann-Mari Svennerholm
- Gothenburg University Vaccine Research Institute, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
19
|
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
|
20
|
Doss-Gollin S, Thomas S, Brook B, Abedi K, Lebas C, Auderset F, Lugo-Rodriguez Y, Sanchez-Schmitz G, Dowling DJ, Levy O, van Haren SD. Human in vitro modeling of adjuvant formulations demonstrates enhancement of immune responses to SARS-CoV-2 antigen. NPJ Vaccines 2023; 8:163. [PMID: 37884538 PMCID: PMC10603059 DOI: 10.1038/s41541-023-00759-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Adjuvants can enhance vaccine immunogenicity, but their mechanism of action is often incompletely understood, hampering rapid applicability for pandemic vaccines. Herein, we characterized the cellular and molecular activity of adjuvant formulations available for pre-clinical evaluation, including several developed for global open access. We applied four complementary human in vitro platforms to assess individual and combined adjuvants in unformulated, oil-in-water, and liposomal delivery platforms. Liposomal co-formulation of MPLA and QS-21 was most potent in promoting dendritic cell maturation, selective production of Th1-polarizing cytokines, and activation of SARS-CoV-2 Spike-specific CD4+ and CD8+ T cells in a co-culture assay. Select formulations also significantly enhanced Spike antigen-specific humoral immunity in vivo. This study confirms the utility of the cumulative use of human in vitro tools to predict adjuvanticity potential. Thus, human in vitro modeling may advance public health by accelerating the development of affordable and scalable adjuvants for vaccines tailored to vulnerable populations.
Collapse
Affiliation(s)
- Simon Doss-Gollin
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Sanya Thomas
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Byron Brook
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Kimia Abedi
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Célia Lebas
- Vaccine Formulation Institute, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Floriane Auderset
- Vaccine Formulation Institute, 1228 Plan-les-Ouates, Geneva, Switzerland
| | | | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - David J Dowling
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Simon D van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
21
|
Dotiwala F, Upadhyay AK. Next Generation Mucosal Vaccine Strategy for Respiratory Pathogens. Vaccines (Basel) 2023; 11:1585. [PMID: 37896988 PMCID: PMC10611113 DOI: 10.3390/vaccines11101585] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Inducing humoral and cytotoxic mucosal immunity at the sites of pathogen entry has the potential to prevent the infection from getting established. This is different from systemic vaccination, which protects against the development of systemic symptoms. The field of mucosal vaccination has seen fewer technological advances compared to nucleic acid and subunit vaccine advances for injectable vaccine platforms. The advent of the next-generation adenoviral vectors has given a boost to mucosal vaccine research. Basic research into the mechanisms regulating innate and adaptive mucosal immunity and the discovery of effective and safe mucosal vaccine adjuvants will continue to improve mucosal vaccine design. The results from clinical trials of inhaled COVID-19 vaccines demonstrate their ability to induce the proliferation of cytotoxic T cells and the production of secreted IgA and IgG antibodies locally, unlike intramuscular vaccinations. However, these mucosal vaccines induce systemic immune responses at par with systemic vaccinations. This review summarizes the function of the respiratory mucosa-associated lymphoid tissue and the advantages that the adenoviral vectors provide as inhaled vaccine platforms.
Collapse
Affiliation(s)
- Farokh Dotiwala
- Ocugen Inc., 11 Great Valley Parkway, Malvern, PA 19355, USA
| | | |
Collapse
|
22
|
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
|
23
|
Akhtar M, Basher SR, Nizam NN, Hossain L, Bhuiyan TR, Qadri F, Lundgren A. T helper cell responses in adult diarrheal patients following natural infection with enterotoxigenic Escherichia coli are primarily of the Th17 type. Front Immunol 2023; 14:1220130. [PMID: 37809062 PMCID: PMC10552643 DOI: 10.3389/fimmu.2023.1220130] [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: 05/10/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
Background Infection with enterotoxigenic Escherichia coli (ETEC) gives rise to IgA antibodies against both the heat labile toxin (LT) and colonization factors (CFs), which are considered to synergistically protect against ETEC diarrhea. Since the development of ETEC-specific long lived plasma cells and memory B cells is likely to be dependent on T helper (Th) cells, we investigated if natural ETEC diarrhea elicits ETEC-specific Th cells and their relation to IgA responses. Methods Th cell subsets were analyzed in adult Bangladeshi patients hospitalized due to ETEC diarrhea by flow cytometric analysis of peripheral blood mononuclear cells (PBMCs) isolated from blood collected day 2, 7, 30 and 90 after hospitalization as well as in healthy controls. The LT- and CF-specific Th responses were determined by analysis of IL-17A and IFN-γ in antigen stimulated PBMC cultures using ELISA. ETEC-specific IgA secreted by circulating antibody secreting cells (plasmablasts) were analyzed by using the antibodies in lymphocyte supernatants (ALS) ELISA-based method and plasma IgA was also measured by ELISA. Results ETEC patients mounted significant ALS and plasma IgA responses against LTB and CFs on day 7 after hospitalization. ETEC patients had significantly elevated proportions of memory Th cells with a Th17 phenotype (CCR6+CXCR3-) in blood compared to controls, while frequencies of Th1 (CCR6-CXCR3+) or Th2 (CCR6-CXCR3-) cells were not increased. Antigen stimulation of PBMCs revealed IL-17A responses to LT, most clearly observed after stimulation with double mutant heat labile toxin (dmLT), but also with LT B subunit (LTB), and to CS6 in samples from patients with LT+ or CS6+ ETEC bacteria. Some individuals also mounted IFN-γ responses to dmLT and LTB. Levels of LTB specific IgA antibodies in ALS, but not plasma samples correlated with both IL-17A (r=0.5, p=0.02) and IFN-γ (r=0.6, p=0.01) responses to dmLT. Conclusions Our results show that ETEC diarrhea induces T cell responses, which are predominantly of the Th17 type. The correlations between IL-17A and IFN-g and intestine-derived plasmablast responses support that Th responses may contribute to the development of protective IgA responses against ETEC infection. These observations provide important insights into T cell responses that need to be considered in the evaluation of advanced ETEC vaccine candidates.
Collapse
Affiliation(s)
- Marjahan Akhtar
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Salima Raiyan Basher
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Nuder Nower Nizam
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Lazina Hossain
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Taufiqur Rahman Bhuiyan
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Anna Lundgren
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
24
|
Lu T, Das S, Howlader DR, Jain A, Hu G, Dietz ZK, Zheng Q, Ratnakaram SSK, Whittier SK, Varisco DJ, Ernst RK, Picking WD, Picking WL. Impact of the TLR4 agonist BECC438 on a novel vaccine formulation against Shigella spp. Front Immunol 2023; 14:1194912. [PMID: 37744341 PMCID: PMC10512073 DOI: 10.3389/fimmu.2023.1194912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Shigellosis (bacillary dysentery) is a severe gastrointestinal infection with a global incidence of 90 million cases annually. Despite the severity of this disease, there is currently no licensed vaccine against shigellosis. Shigella's primary virulence factor is its type III secretion system (T3SS), which is a specialized nanomachine used to manipulate host cells. A fusion of T3SS injectisome needle tip protein IpaD and translocator protein IpaB, termed DBF, when admixed with the mucosal adjuvant double-mutant labile toxin (dmLT) from enterotoxigenic E. coli was protective using a murine pulmonary model. To facilitate the production of this platform, a recombinant protein that consisted of LTA-1, the active moiety of dmLT, and DBF were genetically fused, resulting in L-DBF, which showed improved protection against Shigella challenge. To extrapolate this protection from mice to humans, we modified the formulation to provide for a multivalent presentation with the addition of an adjuvant approved for use in human vaccines. Here, we show that L-DBF formulated (admix) with a newly developed TLR4 agonist called BECC438 (a detoxified lipid A analog identified as Bacterial Enzymatic Combinatorial Chemistry candidate #438), formulated as an oil-in-water emulsion, has a very high protective efficacy at low antigen doses against lethal Shigella challenge in our mouse model. Optimal protection was observed when this formulation was introduced at a mucosal site (intranasally). When the formulation was then evaluated for the immune response it elicits, protection appeared to correlate with high IFN-γ and IL-17 secretion from mucosal site lymphocytes.
Collapse
Affiliation(s)
- Ti Lu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Sayan Das
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - Debaki R. Howlader
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Akshay Jain
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Gang Hu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Zackary K. Dietz
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Qi Zheng
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | | | - Sean K. Whittier
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - David J. Varisco
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - William D. Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Wendy L. Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
25
|
Westcott MM, Blevins M, Wierzba TF, Morse AE, White KR, Sanders LA, Sanders JW. The Immunogenicity and Properties of a Whole-Cell ETEC Vaccine Inactivated with Psoralen and UVA Light in Comparison to Formalin. Microorganisms 2023; 11:2040. [PMID: 37630600 PMCID: PMC10458022 DOI: 10.3390/microorganisms11082040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Inactivated whole-cell vaccines present a full repertoire of antigens to the immune system. Formalin treatment, a standard method for microbial inactivation, can modify or destroy protein antigenic epitopes. We tested the hypothesis that photochemical inactivation with psoralen and UVA light (PUVA), which targets nucleic acid, would improve the immunogenicity of an Enterotoxigenic E. coli (ETEC) vaccine relative to a formalin-inactivated counterpart. Exposure of ETEC H10407 to PUVA using the psoralen drug 4'-Aminomethyltrioxsalen hydrochloride (AMT) yielded replication-incompetent bacteria that retained their metabolic activity. CFA/I-mediated mannose-resistant hemagglutination (MRHA) was equivalent for PUVA-inactivated and live ETEC, but was severely reduced for formalin-ETEC, indicating that PUVA preserved fimbrial protein functional integrity. The immunogenicity of PUVA-ETEC and formalin-ETEC was compared in mice ± double mutant heat-labile enterotoxin (dmLT) adjuvant. Two weeks after an intramuscular prime/boost, serum anti-ETEC IgG titers were similar for the two vaccines and were increased by dmLT. However, the IgG responses raised against several conserved ETEC proteins were greater after vaccination with PUVA-ETEC. In addition, PUVA-ETEC generated IgG specific for heat-labile toxin (LT) in the absence of dmLT, which was not a property of formalin-ETEC. These data are consistent with PUVA preserving ETEC protein antigens in their native-like form and justify the further testing of PUVA as a vaccine platform for ETEC using murine challenge models.
Collapse
Affiliation(s)
- Marlena M. Westcott
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 575 Patterson Ave, Winston Salem, NC 27101, USA; (A.E.M.); (K.R.W.)
| | - Maria Blevins
- Infectious Diseases Section, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC 27157, USA; (M.B.); (T.F.W.); (L.A.S.); (J.W.S.)
| | - Thomas F. Wierzba
- Infectious Diseases Section, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC 27157, USA; (M.B.); (T.F.W.); (L.A.S.); (J.W.S.)
| | - Alexis E. Morse
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 575 Patterson Ave, Winston Salem, NC 27101, USA; (A.E.M.); (K.R.W.)
| | - Kinnede R. White
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 575 Patterson Ave, Winston Salem, NC 27101, USA; (A.E.M.); (K.R.W.)
| | - Leigh Ann Sanders
- Infectious Diseases Section, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC 27157, USA; (M.B.); (T.F.W.); (L.A.S.); (J.W.S.)
| | - John W. Sanders
- Infectious Diseases Section, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC 27157, USA; (M.B.); (T.F.W.); (L.A.S.); (J.W.S.)
| |
Collapse
|
26
|
Han S, Lee P, Choi HJ. Non-Invasive Vaccines: Challenges in Formulation and Vaccine Adjuvants. Pharmaceutics 2023; 15:2114. [PMID: 37631328 PMCID: PMC10458847 DOI: 10.3390/pharmaceutics15082114] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Given the limitations of conventional invasive vaccines, such as the requirement for a cold chain system and trained personnel, needle-based injuries, and limited immunogenicity, non-invasive vaccines have gained significant attention. Although numerous approaches for formulating and administrating non-invasive vaccines have emerged, each of them faces its own challenges associated with vaccine bioavailability, toxicity, and other issues. To overcome such limitations, researchers have created novel supplementary materials and delivery systems. The goal of this review article is to provide vaccine formulation researchers with the most up-to-date information on vaccine formulation and the immunological mechanisms available, to identify the technical challenges associated with the commercialization of non-invasive vaccines, and to guide future research and development efforts.
Collapse
Affiliation(s)
| | | | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada; (S.H.); (P.L.)
| |
Collapse
|
27
|
Das S, Howlader DR, Lu T, Whittier SK, Hu G, Sharma S, Dietz ZK, Ratnakaram SSK, Varisco DJ, Ernst RK, Picking WD, Picking WL. Immunogenicity and protective efficacy of nanoparticle formulations of L-SseB against Salmonella infection. Front Immunol 2023; 14:1208848. [PMID: 37457702 PMCID: PMC10347375 DOI: 10.3389/fimmu.2023.1208848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Salmonella enterica, a Gram-negative pathogen, has over 2500 serovars that infect a wide range of hosts. In humans, S. enterica causes typhoid or gastroenteritis and is a major public health concern. In this study, SseB (the tip protein of the Salmonella pathogenicity island 2 type III secretion system) was fused with the LTA1 subunit of labile-toxin from enterotoxigenic E. coli to make the self-adjuvanting antigen L-SseB. Two unique nanoparticle formulations were developed to allow multimeric presentation of L-SseB. Mice were vaccinated with these formulations and protective efficacy determined via challenging the mice with S. enterica serovars. The polysaccharide (chitosan) formulation was found to elicit better protection when compared to the squalene nanoemulsion. When the polysaccharide formulation was used to vaccinate rabbits, protection from S. enterica challenge was elicited. In summary, L-SseB in a particulate polysaccharide formulation appears to be an attractive candidate vaccine capable of broad protection against S. enterica.
Collapse
Affiliation(s)
- Sayan Das
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Debaki R. Howlader
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Ti Lu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Sean K. Whittier
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Gang Hu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Simran Sharma
- Department of Veterinary Pathobiology and Bond Life Science Center, University of Missouri, Columbia, MO, United States
| | - Zackary K. Dietz
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Siva S. K. Ratnakaram
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - David J. Varisco
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, United States
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, United States
| | - William D. Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Wendy L. Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
28
|
Park JY, Cho SH. Production of monoclonal antibody of heat-labile toxin A subunit to identify enterotoxigenic Escherichia coli by epitope mapping using synthetic peptides. Front Immunol 2023; 14:1152910. [PMID: 37275900 PMCID: PMC10232981 DOI: 10.3389/fimmu.2023.1152910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/05/2023] [Indexed: 06/07/2023] Open
Abstract
Background Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrhea through two enterotoxins, a heat-labile toxin and a heat-stable toxin. These toxins alter the cellular signaling pathways, ultimately triggering an increase in chloride secretion and watery diarrhea. Objective For the development of an ETEC vaccine, we attempted to construct a peptide-specific monoclonal antibody library against heat-labile enterotoxin A subunit (LT-A) by epitope mapping using synthetic peptides. Methods Sera produced by five mice immunized with recombinant LT-A protein were examined for specific recognition with synthetic 15-mer and 34-mer peptides of LT-A proteins using enzyme-linked immunosorbent assay. The analysis revealed that the synthetic peptides number 8, 16, 24, 33, 36, 38, and 39 reacted with an anti-LT-A polyclonal antibody. For the possible prediction of LT-A epitopes, each full-length protein sequence was subjected to BCPreds analysis and three-dimensional protein structure analysis. The data showed that three peptides (synthetic peptide numbers: 33, 36, and 38-39) have identical antigenic specificities with LT-A protein, suggesting the usefulness of these linear peptide epitopes. Results Based on these peptides, we produced monoclonal antibodies to improve the specificity of LT-A detection. Monoclonal antibodies produced from two peptides (numbers 33 and 36) showed affinity for an LT-A recombinant antigen. Moreover, peptide epitope prediction analysis showed that the sites of the three peptides were identical to those exhibiting actual antigenicity. Also, it was confirmed that the amino acid sequence that actually showed antigenicity was included in the peptide predicted only by ETEC-LT-A-33. Also, the specificity of the antibody for ETEC-LT-A-33 was validated using bacterial cells, and the neutralizing effect of the antibody was determined by assessing cytokine release in infected HCT-8 cells. Conclusion The monoclonal antibodies produced in this study are useful toolsfor vaccine production against ETEC and can be used to identify peptide antigencandidates.
Collapse
Affiliation(s)
- Jun-Young Park
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seung-Hak Cho
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
| |
Collapse
|
29
|
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
|
30
|
Bauer DL, Bachnak L, Limbert VM, Horowitz RM, Baudier RL, D'Souza SJ, Immethun VE, Kurtz JR, Grant SB, McLachlan JB. The Adjuvant Combination of dmLT and Monophosphoryl Lipid A Activates the Canonical, Nonpyroptotic NLRP3 Inflammasome in Dendritic Cells and Significantly Interacts to Expand Antigen-Specific CD4 T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1519-1530. [PMID: 37023458 PMCID: PMC10159919 DOI: 10.4049/jimmunol.2200221] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023]
Abstract
Adjuvants are often essential additions to vaccines that enhance the activation of innate immune cells, leading to more potent and protective T and B cell responses. Only a few vaccine adjuvants are currently used in approved vaccine formulations in the United States. Combinations of one or more adjuvants have the potential to increase the efficacy of existing and next-generation vaccines. In this study, we investigated how the nontoxic double mutant Escherichia coli heat-labile toxin R192G/L211A (dmLT), when combined with the TLR4 agonist monophosphoryl lipid A (MPL-A), impacted innate and adaptive immune responses to vaccination in mice. We found that the combination of dmLT and MPL-A induced an expansion of Ag-specific, multifaceted Th1/2/17 CD4 T cells higher than that explained by adding responses to either adjuvant alone. Furthermore, we observed more robust activation of primary mouse bone marrow-derived dendritic cells in the combination adjuvant-treated group via engagement of the canonical NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome complex. This was marked by a multiplicative increase in the secretion of active IL-1β that was independent of classical gasdermin D-mediated pyroptosis. Moreover, the combination adjuvant increased the production of the secondary messengers cAMP and PGE2 in dendritic cells. These results demonstrate how certain adjuvant combinations could be used to potentiate better vaccine responses to combat a variety of pathogens.
Collapse
Affiliation(s)
- David L Bauer
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| | - Louay Bachnak
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| | - Vanessa M Limbert
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| | - Rebecca M Horowitz
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| | - Robin L Baudier
- Department of Epidemiology, Tulane School of Public Health and Tropical Medicine, New Orleans, LA
| | - Shaina J D'Souza
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| | - Victoria E Immethun
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| | - Jonathan R Kurtz
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| | - Samuel B Grant
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| | - James B McLachlan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
31
|
Mubanga C, Simuyandi M, Mwape K, Chibesa K, Chisenga C, Chilyabanyama ON, Randall A, Liang X, Glashoff RH, Chilengi R. Use of an ETEC Proteome Microarray to Evaluate Cross-Reactivity of ETVAX ® Vaccine-Induced IgG Antibodies in Zambian Children. Vaccines (Basel) 2023; 11:vaccines11050939. [PMID: 37243042 DOI: 10.3390/vaccines11050939] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/02/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Developing a broadly protective vaccine covering most ETEC variants has been elusive. The most clinically advanced candidate yet is an oral inactivated ETEC vaccine (ETVAX®). We report on the use of a proteome microarray for the assessment of cross-reactivity of anti-ETVAX® IgG antibodies against over 4000 ETEC antigens and proteins. We evaluated 40 (pre-and post-vaccination) plasma samples from 20 Zambian children aged 10-23 months that participated in a phase 1 trial investigating the safety, tolerability, and immunogenicity of ETVAX® adjuvanted with dmLT. Pre-vaccination samples revealed high IgG responses to a variety of ETEC proteins including classical ETEC antigens (CFs and LT) and non-classical antigens. Post-vaccination reactivity to CFA/I, CS3, CS6, and LTB was stronger than baseline among the vaccinated compared to the placebo group. Interestingly, we noted significantly high post-vaccination responses to three non-vaccine ETEC proteins: CS4, CS14, and PCF071 (p = 0.043, p = 0.028, and p = 0.00039, respectively), suggestive of cross-reactive responses to CFA/I. However, similar responses were observed in the placebo group, indicating the need for larger studies. We conclude that the ETEC microarray is a useful tool for investigating antibody responses to numerous antigens, especially because it may not be practicable to include all antigens in a single vaccine.
Collapse
Affiliation(s)
- Cynthia Mubanga
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
- Division of Medical Microbiology, Department of Pathology, Stellenbosch University & National Health Laboratory Service, Tygerberg Hospital Francie van Zijl Drive, P.O. Box 241, Cape Town 8000, South Africa
| | - Michelo Simuyandi
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
| | - Kapambwe Mwape
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
- Water and Health Research Center, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Kennedy Chibesa
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
- Division of Virology, School of Pathology, Faculty of Health Sciences, University of the Free State, 205 Nelson Mandela, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Caroline Chisenga
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
| | | | - Arlo Randall
- Antigen Discovery Inc., 1 Technology Dr., Suite E309, Irvine, CA 92618, USA
| | - Xiaowu Liang
- Antigen Discovery Inc., 1 Technology Dr., Suite E309, Irvine, CA 92618, USA
| | - Richard H Glashoff
- Division of Medical Microbiology, Department of Pathology, Stellenbosch University & National Health Laboratory Service, Tygerberg Hospital Francie van Zijl Drive, P.O. Box 241, Cape Town 8000, South Africa
| | - Roma Chilengi
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
| |
Collapse
|
32
|
So KK, Le NMT, Nguyen NL, Kim DH. Improving expression and assembly of difficult-to-express heterologous proteins in Saccharomyces cerevisiae by culturing at a sub-physiological temperature. Microb Cell Fact 2023; 22:55. [PMID: 36959657 PMCID: PMC10035479 DOI: 10.1186/s12934-023-02065-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/19/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Escherichia coli heat labile toxin B subunit (LTB) is one of the most popular oral vaccine adjuvants and intestine adsorption enhancers. It is often expressed as a fusion partner with target antigens to enhance their immunogenicity as well as gut absorbability. However, high expression levels of a fusion protein are critical to the outcome of immunization experiments and the success of subsequent vaccine development efforts. In order to improve the expression and functional assembly of LTB-fusion proteins using Saccharomyces cerevisiae, we compared their expression under culture conditions at a sub-physiological temperature 20 °C with their expression under a standard 30 °C. RESULTS The assembled expression of LTB-EDIII2 (LTB fused to the envelope domain III (EDIII) of Dengue virus serotype 2), which was expressed at the level of 20 µg/L in our previous study, was higher when the expression temperature was 20 °C as opposed to 30 °C. We also tested whether the expression and functional assembly of a difficult-to-express LTB fusion protein could be increased. The assembled expression of the difficult-to-express LTB-VP1 fusion protein (LTB fused to VP1 antigen of Foot-and-Mouth Disease Virus) dramatically increased, although the total amount of expressed protein was still lower than that of LTB-EDIII2. Slight but significant increase in the expression of well-known reporter protein eGFP, which has previously been shown to be increased by cultivation at 20 °C, was also observed in our expression system. As no significant changes in corresponding transcripts levels and cell growth were observed between 20 °C and 30 °C, we infer that translation and post-translational assembly are responsible for these enhancements. CONCLUSIONS The effects of lowering the expression temperature from 30 °C to 20 °C on protein expression and folding levels in S. cerevisiae, using several proteins as models, are reported. When heterologous proteins are expressed at 20 °C, a greater amount of (specially, more assembled) functional proteins accumulated than at 30 °C. Although further studies are required to understand the molecular mechanisms, our results suggest that lowering the expression temperature is a convenient strategy for improving the expression of relatively complexly structured and difficult-to-express proteins in S. cerevisiae.
Collapse
Affiliation(s)
- Kum-Kang So
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Jeonbuk National University, Jeonju, Jeollabuk-Do, 54896, Republic of Korea
| | - Ngoc My Tieu Le
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Jeollabuk-Do, 54896, Republic of Korea
| | - Ngoc-Luong Nguyen
- Department of Biology, College of Sciences, Hue University, Hue, 530000, Vietnam.
| | - Dae-Hyuk Kim
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Jeonbuk National University, Jeonju, Jeollabuk-Do, 54896, Republic of Korea.
- Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju, Jeollabuk-Do, 54896, Republic of Korea.
| |
Collapse
|
33
|
Walker RI, Bourgeois AL. Oral inactivated whole cell vaccine for mucosal immunization: ETVAX case study. Front Immunol 2023; 14:1125102. [PMID: 36936951 PMCID: PMC10018008 DOI: 10.3389/fimmu.2023.1125102] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/03/2023] [Indexed: 03/06/2023] Open
Abstract
Oral immunization is an effective strategy for inducing protective immunity against mucosal enteric pathogens. Although live-attenuated as well as subunit approaches have been explored for vaccination against enteric pathogens, inactivated whole bacterial cells may also be effective in introducing protective immunity. Successfully accomplishing this goal with inactivated whole bacterial cells will require that a complex antigenic repertoire be presented in controlled immunogenic amounts, in a safe and relatively simple and self-contained delivery format. The benefit from immunization with whole cell vaccines can be further enhanced through genetic engineering to over-express selected antigens and also by the use of mucosal adjuvants to direct a more robust immunologic response. These steps are being taken for the development of ETVAX, the most clinically advanced vaccine candidate against the major enteric pathogen, enterotoxigenic Escherichia coli (ETEC) with significant positive impact.
Collapse
Affiliation(s)
- Richard I. Walker
- Center for Vaccine Innovation and Access, PATH, Washington, DC, United States
| | | |
Collapse
|
34
|
Peletta A, Lemoine C, Courant T, Collin N, Borchard G. Meeting vaccine formulation challenges in an emergency setting: Towards the development of accessible vaccines. Pharmacol Res 2023; 189:106699. [PMID: 36796463 DOI: 10.1016/j.phrs.2023.106699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Vaccination is considered one of the most successful strategies to prevent infectious diseases. In the event of a pandemic or epidemic, the rapid development and distribution of the vaccine to the population is essential to reduce mortality, morbidity and transmission. As seen during the COVID-19 pandemic, the production and distribution of vaccines has been challenging, in particular for resource-constrained settings, essentially slowing down the process of achieving global coverage. Pricing, storage, transportation and delivery requirements of several vaccines developed in high-income countries resulted in limited access for low-and-middle income countries (LMICs). The capacity to manufacture vaccines locally would greatly improve global vaccine access. In particular, for the development of classical subunit vaccines, the access to vaccine adjuvants is a pre-requisite for more equitable access to vaccines. Vaccine adjuvants are agents required to augment or potentiate, and possibly target the specific immune response to such type of vaccine antigens. Openly accessible or locally produced vaccine adjuvants may allow for faster immunization of the global population. For local research and development of adjuvanted vaccines to expand, knowledge on vaccine formulation is of paramount importance. In this review, we aim to discuss the optimal characteristics of a vaccine developed in an emergency setting by focusing on the importance of vaccine formulation, appropriate use of adjuvants and how this may help overcome barriers for vaccine development and production in LMICs, achieve improved vaccine regimens, delivery and storage requirements.
Collapse
Affiliation(s)
- Allegra Peletta
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| | - Céline Lemoine
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Thomas Courant
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| |
Collapse
|
35
|
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
|
36
|
Verma SK, Mahajan P, Singh NK, Gupta A, Aggarwal R, Rappuoli R, Johri AK. New-age vaccine adjuvants, their development, and future perspective. Front Immunol 2023; 14:1043109. [PMID: 36911719 PMCID: PMC9998920 DOI: 10.3389/fimmu.2023.1043109] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/26/2023] [Indexed: 02/26/2023] Open
Abstract
In the present scenario, immunization is of utmost importance as it keeps us safe and protects us from infectious agents. Despite the great success in the field of vaccinology, there is a need to not only develop safe and ideal vaccines to fight deadly infections but also improve the quality of existing vaccines in terms of partial or inconsistent protection. Generally, subunit vaccines are known to be safe in nature, but they are mostly found to be incapable of generating the optimum immune response. Hence, there is a great possibility of improving the potential of a vaccine in formulation with novel adjuvants, which can effectively impart superior immunity. The vaccine(s) in formulation with novel adjuvants may also be helpful in fighting pathogens of high antigenic diversity. However, due to the limitations of safety and toxicity, very few human-compatible adjuvants have been approved. In this review, we mainly focus on the need for new and improved vaccines; the definition of and the need for adjuvants; the characteristics and mechanisms of human-compatible adjuvants; the current status of vaccine adjuvants, mucosal vaccine adjuvants, and adjuvants in clinical development; and future directions.
Collapse
Affiliation(s)
| | - Pooja Mahajan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI, United States
| | - Ankit Gupta
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | - Rupesh Aggarwal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
37
|
Recent Advances in the Development of Adenovirus-Vectored Vaccines for Parasitic Infections. Pharmaceuticals (Basel) 2023; 16:ph16030334. [PMID: 36986434 PMCID: PMC10058461 DOI: 10.3390/ph16030334] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Vaccines against parasites have lagged centuries behind those against viral and bacterial infections, despite the devastating morbidity and widespread effects of parasitic diseases across the globe. One of the greatest hurdles to parasite vaccine development has been the lack of vaccine strategies able to elicit the complex and multifaceted immune responses needed to abrogate parasitic persistence. Viral vectors, especially adenovirus (AdV) vectors, have emerged as a potential solution for complex disease targets, including HIV, tuberculosis, and parasitic diseases, to name a few. AdVs are highly immunogenic and are uniquely able to drive CD8+ T cell responses, which are known to be correlates of immunity in infections with most protozoan and some helminthic parasites. This review presents recent developments in AdV-vectored vaccines targeting five major human parasitic diseases: malaria, Chagas disease, schistosomiasis, leishmaniasis, and toxoplasmosis. Many AdV-vectored vaccines have been developed for these diseases, utilizing a wide variety of vectors, antigens, and modes of delivery. AdV-vectored vaccines are a promising approach for the historically challenging target of human parasitic diseases.
Collapse
|
38
|
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
|
39
|
García-Silva I, Govea-Alonso DO, Rosales-Mendoza S. Current status of mucosal vaccines against SARS-CoV2: a hope for protective immunity. Expert Opin Biol Ther 2023; 23:207-222. [PMID: 36594264 DOI: 10.1080/14712598.2022.2156284] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION The current vaccines used to fight against COVID-19 are effective, however the induction of protective immunity is a pending goal required to prevent viral transmission, prevent the generation of new variants, and ultimately eradicate SARS-CoV-2. Mucosal immunization stands as a promising approach to achieve protective immunity against SARS-CoV-2; therefore, it is imperative to innovate the current vaccines by developing mucosal candidates, focusing not only on their ability to prevent severe COVID-19 but to neutralize the virus before invasion of the respiratory system and other mucosal compartments. AREAS COVERED This review covers the current advances on the development of anti-COVID-19 mucosal vaccines. Biomedical literature, including PubMed and clinicaltrials.gov website, was analyzed to identify the state of the art for this field. The achievements in preclinical and clinical evaluations are presented and critically analyzed. EXPERT OPINION There is a significant advance on the development of mucosal vaccines against SARSCoV-2, which is a promise to increase the efficacy of immunization against this pathogen. Both preclinical and clinical evaluation for several candidates have been performed. The challenges in this road (e.g. low immunogenicity, a reduced number of adjuvants available, and inaccurate dosage) are identified and also critical perspectives for the field are provided.
Collapse
Affiliation(s)
- Ileana García-Silva
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| | - Dania O Govea-Alonso
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| | - Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, San Luis Potosí, México.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, 78210, San Luis Potosí, México
| |
Collapse
|
40
|
Xing Y, Clark JR, Chang JD, Chirman DM, Green S, Zulk JJ, Jelinski J, Patras KA, Maresso AW. Broad protective vaccination against systemic Escherichia coli with autotransporter antigens. PLoS Pathog 2023; 19:e1011082. [PMID: 36800400 PMCID: PMC9937491 DOI: 10.1371/journal.ppat.1011082] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 12/26/2022] [Indexed: 02/18/2023] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is the leading cause of adult life-threatening sepsis and urinary tract infections (UTI). The emergence and spread of multidrug-resistant (MDR) ExPEC strains result in a considerable amount of treatment failure and hospitalization costs, and contribute to the spread of drug resistance amongst the human microbiome. Thus, an effective vaccine against ExPEC would reduce morbidity and mortality and possibly decrease carriage in healthy or diseased populations. A comparative genomic analysis demonstrated a gene encoding an invasin-like protein, termed sinH, annotated as an autotransporter protein, shows high prevalence in various invasive ExPEC phylogroups, especially those associated with systemic bacteremia and UTI. Here, we evaluated the protective efficacy and immunogenicity of a recombinant SinH-based vaccine consisting of either domain-3 or domains-1,2, and 3 of the putative extracellular region of surface-localized SinH. Immunization of a murine host with SinH-based antigens elicited significant protection against various strains of the pandemic ExPEC sequence type 131 (ST131) as well as multiple sequence types in two distinct models of infection (colonization and bacteremia). SinH immunization also provided significant protection against ExPEC colonization in the bladder in an acute UTI model. Immunized cohorts produced significantly higher levels of vaccine-specific serum IgG and urinary IgG and IgA, findings consistent with mucosal protection. Collectively, these results demonstrate that autotransporter antigens such as SinH may constitute promising ExPEC phylogroup-specific and sequence-type effective vaccine targets that reduce E. coli colonization and virulence.
Collapse
Affiliation(s)
- Yikun Xing
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - James D. Chang
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Dylan M. Chirman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sabrina Green
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jacob J. Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Joseph Jelinski
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
41
|
Pokharel P, Dhakal S, Dozois CM. The Diversity of Escherichia coli Pathotypes and Vaccination Strategies against This Versatile Bacterial Pathogen. Microorganisms 2023; 11:344. [PMID: 36838308 PMCID: PMC9965155 DOI: 10.3390/microorganisms11020344] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Escherichia coli (E. coli) is a gram-negative bacillus and resident of the normal intestinal microbiota. However, some E. coli strains can cause diseases in humans, other mammals and birds ranging from intestinal infections, for example, diarrhea and dysentery, to extraintestinal infections, such as urinary tract infections, respiratory tract infections, meningitis, and sepsis. In terms of morbidity and mortality, pathogenic E. coli has a great impact on public health, with an economic cost of several billion dollars annually worldwide. Antibiotics are not usually used as first-line treatment for diarrheal illness caused by E. coli and in the case of bloody diarrhea, antibiotics are avoided due to the increased risk of hemolytic uremic syndrome. On the other hand, extraintestinal infections are treated with various antibiotics depending on the site of infection and susceptibility testing. Several alarming papers concerning the rising antibiotic resistance rates in E. coli strains have been published. The silent pandemic of multidrug-resistant bacteria including pathogenic E. coli that have become more difficult to treat favored prophylactic approaches such as E. coli vaccines. This review provides an overview of the pathogenesis of different pathotypes of E. coli, the virulence factors involved and updates on the major aspects of vaccine development against different E. coli pathotypes.
Collapse
Affiliation(s)
- Pravil Pokharel
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Sabin Dhakal
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Charles M. Dozois
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
- Pasteur Network, Laval, QC H7V 1B7, Canada
| |
Collapse
|
42
|
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
|
43
|
Singleton KL, Joffe A, Leitner WW. Review: Current trends, challenges, and success stories in adjuvant research. Front Immunol 2023; 14:1105655. [PMID: 36742311 PMCID: PMC9892189 DOI: 10.3389/fimmu.2023.1105655] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Vaccine adjuvant research is being fueled and driven by progress in the field of innate immunity that has significantly advanced in the past two decades with the discovery of countless innate immune receptors and innate immune pathways. Receptors for pathogen-associated molecules (PAMPs) or host-derived, danger-associated molecules (DAMPs), as well as molecules in the signaling pathways used by such receptors, are a rich source of potential targets for agonists that enable the tuning of innate immune responses in an unprecedented manner. Targeted modulation of immune responses is achieved not only through the choice of immunostimulator - or select combinations of adjuvants - but also through formulation and systematic modifications of the chemical structure of immunostimulatory molecules. The use of medium and high-throughput screening methods for finding immunostimulators has further accelerated the identification of promising novel adjuvants. However, despite the progress that has been made in finding new adjuvants through systematic screening campaigns, the process is far from perfect. A major bottleneck that significantly slows the process of turning confirmed or putative innate immune receptor agonists into vaccine adjuvants continues to be the lack of defined in vitro correlates of in vivo adjuvanticity. This brief review discusses recent developments, exciting trends, and notable successes in the adjuvant research field, albeit acknowledging challenges and areas for improvement.
Collapse
|
44
|
Tsai CJY, Loh JMS, Fujihashi K, Kiyono H. Mucosal vaccination: onward and upward. Expert Rev Vaccines 2023; 22:885-899. [PMID: 37817433 DOI: 10.1080/14760584.2023.2268724] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION The unique mucosal immune system allows the generation of robust protective immune responses at the front line of pathogen encounters. The needle-free delivery route and cold chain-free logistic requirements also provide additional advantages in ease and economy. However, the development of mucosal vaccines faces several challenges, and only a handful of mucosal vaccines are currently licensed. These vaccines are all in the form of live attenuated or inactivated whole organisms, whereas no subunit-based mucosal vaccine is available. AREAS COVERED The selection of antigen, delivery vehicle, route and adjuvants for mucosal vaccination are highly important. This is particularly crucial for subunit vaccines, as they often fail to elicit strong immune responses. Emerging research is providing new insights into the biological and immunological uniqueness of mucosal tissues. However, many aspects of the mucosal immunology still await to be investigated. EXPERT OPINION This article provides an overview of the current understanding of mucosal vaccination and discusses the remaining knowledge gaps. We emphasize that because of the potential benefits mucosal vaccines can bring from the biomedical, social and economic standpoints, the unmet goal to achieve mucosal vaccine success is worth the effort.
Collapse
Affiliation(s)
- Catherine J Y Tsai
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
| | - Jacelyn M S Loh
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hiroshi Kiyono
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
- CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
- Future Medicine Education and Research Organization, Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan
| |
Collapse
|
45
|
Conti BJ, Santiago KB, Cardoso EO, Conte FL, Golim MA, Cruz MT, Sforcin JM. Effect of propolis on Th2 and Th17 cells: interplay with EtxB- and LPS-treated dendritic cells. Braz J Med Biol Res 2023; 56:e12659. [PMID: 37075347 PMCID: PMC10125804 DOI: 10.1590/1414-431x2023e12659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/07/2023] [Indexed: 04/21/2023] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells that drive the differentiation of T CD4+ cells into different profiles according to the nature of the antigen or immunomodulator. Propolis is a resinous product made by bees that has numerous pharmacological properties, including an immunomodulatory action. To assess whether propolis can modulate the activation of CD4+ T cells by stimulating DCs with heat-labile enterotoxin B subunit (EtxB) or lipopolysaccharide (LPS), we aimed to elucidate the mechanisms affected by propolis in the differential activation of T lymphocytes. Cell viability, lymphocyte proliferation, gene expression (GATA-3 and RORc), and cytokine production (interleukin (IL)-4 and IL-17A) were analyzed. Propolis, EtxB, and LPS induced a higher lymphoproliferation compared with the control. Propolis induced GATA-3 expression and, in combination with EtxB, maintained the baseline levels. Propolis alone or in combination with LPS inhibited RORc expression. EtxB alone and in combination with propolis increased IL-4 production. Propolis in combination with LPS prevented LPS-induced IL-17A production. These results opened perspectives for the study of biological events that may be favored by propolis by promoting Th2 activation or helping in the treatment of inflammatory conditions mediated by Th17 cells.
Collapse
Affiliation(s)
- B J Conti
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - K B Santiago
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - E O Cardoso
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - F L Conte
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - M A Golim
- Hemocentro de Botucatu, Faculdade de Medicina, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | - M T Cruz
- Faculty of Pharmacy, Center for Neurosciences and Cellular Biology, University of Coimbra, Coimbra, Portugal
| | - J M Sforcin
- Departamento de Ciências Químicas e Biológicas, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| |
Collapse
|
46
|
Characterization of Systemic and Mucosal Humoral Immune Responses to an Adjuvanted Intranasal SARS-CoV-2 Protein Subunit Vaccine Candidate in Mice. Vaccines (Basel) 2022; 11:vaccines11010030. [PMID: 36679875 PMCID: PMC9865305 DOI: 10.3390/vaccines11010030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Continuous viral evolution of SARS-CoV-2 has resulted in variants capable of immune evasion, vaccine breakthrough infections and increased transmissibility. New vaccines that invoke mucosal immunity may provide a solution to reducing virus transmission. Here, we evaluated the immunogenicity of intranasally administered subunit protein vaccines composed of a stabilized SARS-CoV-2 spike trimer or the receptor binding domain (RBD) adjuvanted with either cholera toxin (CT) or an archaeal lipid mucosal adjuvant (AMVAD). We show robust induction of immunoglobulin (Ig) G and IgA responses in plasma, nasal wash and bronchoalveolar lavage in mice only when adjuvant is used in the vaccine formulation. While the AMVAD adjuvant was more effective at inducing systemic antibodies against the RBD antigen than CT, CT was generally more effective at inducing overall higher IgA and IgG titers against the spike antigen in both systemic and mucosal compartments. Furthermore, vaccination with adjuvanted spike led to superior mucosal IgA responses than with the RBD antigen and produced broadly targeting neutralizing plasma antibodies against ancestral, Delta and Omicron variants in vitro; whereas adjuvanted RBD elicited a narrower antibody response with neutralizing activity only against ancestral and Delta variants. Our study demonstrates that intranasal administration of an adjuvanted protein subunit vaccine in immunologically naïve mice induced both systemic and mucosal neutralizing antibody responses that were most effective at neutralizing SARS-CoV-2 variants when the trimeric spike was used as an antigen compared to RBD.
Collapse
|
47
|
Gong X, Gao Y, Shu J, Zhang C, Zhao K. Chitosan-Based Nanomaterial as Immune Adjuvant and Delivery Carrier for Vaccines. Vaccines (Basel) 2022; 10:1906. [PMID: 36423002 PMCID: PMC9696061 DOI: 10.3390/vaccines10111906] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 08/26/2023] Open
Abstract
With the support of modern biotechnology, vaccine technology continues to iterate. The safety and efficacy of vaccines are some of the most important areas of development in the field. As a natural substance, chitosan is widely used in numerous fields-such as immune stimulation, drug delivery, wound healing, and antibacterial procedures-due to its good biocompatibility, low toxicity, biodegradability, and adhesion. Chitosan-based nanoparticles (NPs) have attracted extensive attention with respect to vaccine adjuvants and delivery systems due to their excellent properties, which can effectively enhance immune responses. Here, we list the classifications and mechanisms of action of vaccine adjuvants. At the same time, the preparation methods of chitosan, its NPs, and their mechanism of action in the delivery system are introduced. The extensive applications of chitosan and its NPs in protein vaccines and nucleic acid vaccines are also introduced. This paper reviewed the latest research progress of chitosan-based NPs in vaccine adjuvant and drug delivery systems.
Collapse
Affiliation(s)
- Xiaochen Gong
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
- School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Yuan Gao
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
| | - Jianhong Shu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Hom-Sun Biotechnology Co., Ltd., Shaoxing 312366, China
| | - Chunjing Zhang
- School of Medical Technology, Qiqihar Medical University, Qiqihar 161006, China
| | - Kai Zhao
- Institute of Nanobiomaterials and Immunology, School of Pharmaceutical Sciences & School of Life Science, Taizhou University, Taizhou 318000, China
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Hom-Sun Biotechnology Co., Ltd., Shaoxing 312366, China
| |
Collapse
|
48
|
Wellford SA, Moseman AP, Dao K, Wright KE, Chen A, Plevin JE, Liao TC, Mehta N, Moseman EA. Mucosal plasma cells are required to protect the upper airway and brain from infection. Immunity 2022; 55:2118-2134.e6. [PMID: 36137543 PMCID: PMC9649878 DOI: 10.1016/j.immuni.2022.08.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/25/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
While blood antibodies mediate protective immunity in most organs, whether they protect nasal surfaces in the upper airway is unclear. Using multiple viral infection models in mice, we found that blood-borne antibodies could not defend the olfactory epithelium. Despite high serum antibody titers, pathogens infected nasal turbinates, and neurotropic microbes invaded the brain. Using passive antibody transfers and parabiosis, we identified a restrictive blood-endothelial barrier that excluded circulating antibodies from the olfactory mucosa. Plasma cell depletions demonstrated that plasma cells must reside within olfactory tissue to achieve sterilizing immunity. Antibody blockade and genetically deficient models revealed that this local immunity required CD4+ T cells and CXCR3. Many vaccine adjuvants failed to generate olfactory plasma cells, but mucosal immunizations established humoral protection of the olfactory surface. Our identification of a blood-olfactory barrier and the requirement for tissue-derived antibody has implications for vaccinology, respiratory and CNS pathogen transmission, and B cell fate decisions.
Collapse
Affiliation(s)
| | - Annie Park Moseman
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Kianna Dao
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Katherine E Wright
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Allison Chen
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Jona E Plevin
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Tzu-Chieh Liao
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Naren Mehta
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - E Ashley Moseman
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
49
|
Monge C, Ayad C, Paris AL, Rovera R, Colomb E, Verrier B. Mucosal Adjuvants Delivered by a Mucoadhesive Patch for Sublingual Administration of Subunit Vaccines. Int J Mol Sci 2022; 23:13440. [PMID: 36362224 PMCID: PMC9655718 DOI: 10.3390/ijms232113440] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 11/19/2023] Open
Abstract
Among mucosal administration routes for vaccines, the sublingual route has been proven capable of inducing a potent systemic and mucosal immune response. However, the absence of a simple and compliant delivery system and the lack of robust mucosal adjuvants impede the development of sublingual vaccines. Here, we describe a mucoadhesive patch made of a layer-by-layer assembly of polysaccharides, chitosan, and hyaluronic acid. The mucoadhesive patch was covered by adjuvanted nanoparticles carrying viral proteins. We showed that the nanoparticles effectively cross the outer layers of the sublingual mucosa to reach the epithelium. Furthermore, the encapsulated adjuvants, 3M-052 and mifamurtide, targeting toll-like receptor (TLR) 7/8 and nucleotide-binding oligomerization domain-2 (NOD2), respectively, remain fully active after encapsulation into nanoparticles and exhibit a cytokine/chemokine signature similar to the mucosal gold-standard adjuvant, the cholera toxin. However, the particulate adjuvants induced more moderate levels of proinflammatory interleukin (IL)-6 and keratinocyte chemoattractant (KC), suggesting a controlled activation of the innate immune response.
Collapse
Affiliation(s)
- Claire Monge
- UMR 5305: Laboratoire de Biologie Tissulaire et d’Ingénierie Thérapeutique, Institut de Biologie et Chimie des Protéines, CNRS/Université Claude Bernard Lyon 1, 7 Passage du Vercors, 69007 Lyon, France
| | | | | | | | | | | |
Collapse
|
50
|
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
|