1
|
Lee MJ, Shin S, Kim HW, Ko MK, Park SH, Kim SM, Park JH. Oral Administration of Zinc Sulfate with Intramuscular Foot-and-Mouth Disease Vaccine Enhances Mucosal and Systemic Immunity. Vaccines (Basel) 2024; 12:1268. [PMID: 39591171 PMCID: PMC11598382 DOI: 10.3390/vaccines12111268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Foot-and-mouth disease (FMD) remains a significant global threat to livestock farming. Current commercial FMD vaccines present several challenges, including the risk of infection and adverse injection site reactions due to oil-based adjuvants. The complex immune environment of the gut-associated lymphoid tissue has the potential to induce broad and diverse immune responses. Therefore, we aimed to explore the potential of zinc sulfate as an oral adjuvant to enhance intestinal mucosal immunity and complement the effects of intramuscular (IM) FMD vaccination. Methods: We conducted serological analyses on mice and pigs, measuring secretory IgA (sIgA) levels and evaluating the expression of mucosal immunity-related genes in pigs. These assessments were used to investigate the systemic and mucosal immune responses induced by oral zinc sulfate administration in combination with an IM FMD vaccine. Results: This combination strategy significantly increased structural protein antibody titers and virus neutralization titers in experimental animals (mice) and target animals (pigs) across early, mid-, and long-term periods. Additionally, this approach enhanced the expression of key cytokines associated with mucosal immunity and increased sIgA levels, which are critical markers of mucosal immunity. Conclusions: Oral zinc sulfate administration may synergize with inactivated FMD vaccines, leading to sustained and enhanced long-term immune responses. This novel strategy could reduce the frequency of required vaccinations or allow for a lower antigen dose in vaccines, effectively stimulating the mucosal immune system and boosting systemic immunity. This approach has the potential to improve the overall efficacy of commercial FMD vaccines.
Collapse
|
2
|
Hillman T. The application of plant-exosome-like nanovesicles as improved drug delivery systems for cancer vaccines. Discov Oncol 2024; 15:136. [PMID: 38683256 PMCID: PMC11058161 DOI: 10.1007/s12672-024-00974-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 04/08/2024] [Indexed: 05/01/2024] Open
Abstract
The use of cancer immunotherapeutics is currently increasing. Cancer vaccines, as a form of immunotherapy, are gaining much attention in the medical community since specific tumor-antigens can activate immune cells to induce an anti-tumor immune response. However, the delivery of cancer vaccines presents many issues for research scientists when designing cancer treatments and requires further investigation. Nanoparticles, synthetic liposomes, bacterial vectors, viral particles, and mammalian exosomes have delivered cancer vaccines. In contrast, the use of many of these nanotechnologies produces many issues of cytotoxicity, immunogenicity, and rapid clearance by the mononuclear phagocyte system (MPS). Plant-exosome-like nanovesicles (PELNVs) can provide solutions for many of these challenges because they are innocuous and nonimmunogenic when delivering nanomedicines. Hence, this review will describe the potential use of PELNVs to deliver cancer vaccines. In this review, different approaches of cancer vaccine delivery will be detailed, the mechanism of oral vaccination for delivering cancer vaccines will be described, and the review will discuss the use of PELNVs as improved drug delivery systems for cancer vaccines via oral administration while also addressing the subsequent challenges for advancing their usage into the clinical setting.
Collapse
|
3
|
Pomatto MAC, Gai C, Negro F, Massari L, Deregibus MC, De Rosa FG, Camussi G. Oral Delivery of mRNA Vaccine by Plant-Derived Extracellular Vesicle Carriers. Cells 2023; 12:1826. [PMID: 37508491 PMCID: PMC10378442 DOI: 10.3390/cells12141826] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
mRNA-based vaccines were effective in contrasting SARS-CoV-2 infection. However, they presented several limitations of storage and supply chain, and their parenteral administration elicited a limited mucosal IgA immune response. Extracellular vesicles (EVs) have been recognized as a mechanism of cell-to-cell communication well-preserved in all life kingdoms, including plants. Their membrane confers protection from enzyme degradation to encapsulated nucleic acids favoring their transfer between cells. In the present study, EVs derived from the juice of an edible plant (Citrus sinensis) (oEVs) were investigated as carriers of an orally administered mRNA vaccine coding for the S1 protein subunit of SARS-CoV-2 with gastro-resistant oral capsule formulation. The mRNA loaded into oEVs was protected and was stable at room temperature for one year after lyophilization and encapsulation. Rats immunized via gavage administration developed a humoral immune response with the production of specific IgM, IgG, and IgA, which represent the first mucosal barrier in the adaptive immune response. The vaccination also triggered the generation of blocking antibodies and specific lymphocyte activation. In conclusion, the formulation of lyophilized mRNA-containing oEVs represents an efficient delivery strategy for oral vaccines due to their stability at room temperature, optimal mucosal absorption, and the ability to trigger an immune response.
Collapse
Affiliation(s)
- Margherita A C Pomatto
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Chiara Gai
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | | | | | - Maria Chiara Deregibus
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Francesco Giuseppe De Rosa
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Giovanni Camussi
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| |
Collapse
|
4
|
Rathore APS, St John AL. Promises and challenges of mucosal COVID-19 vaccines. Vaccine 2023; 41:4042-4049. [PMID: 37045682 PMCID: PMC10083204 DOI: 10.1016/j.vaccine.2023.04.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Coronavirus disease-2019 (COVID-19) is an ongoing pandemic caused by the newly emerged virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Currently, COVID-19 vaccines are given intramuscularly and they have been shown to evoke systemic immune responses that are highly efficacious towards preventing severe disease and death. However, vaccine-induced immunity wanes within a short time, and booster doses are currently recommended. Furthermore, current vaccine formulations do not adequately restrict virus infection at the mucosal sites, such as in the nasopharyngeal tract and, therefore, have limited capacity to block virus transmission. With these challenges in mind, several mucosal vaccines are currently being developed with the aim of inducing long-lasting protective immune responses at the mucosal sites where SARS-COV-2 infection begins. Past successes in mucosal vaccinations underscore the potential of these developmental stage SARS-CoV-2 vaccines to reduce disease burden, if not eliminate it altogether. Here, we discuss immune responses that are triggered at the mucosal sites and recent advances in our understanding of mucosal responses induced by SARS-CoV-2 infection and current COVID-19 vaccines. We also highlight several mucosal SARS-COV-2 vaccine formulations that are currently being developed or tested for human use and discuss potential challenges to mucosal vaccination.
Collapse
Affiliation(s)
- Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27705, USA
| | - Ashley L St John
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27705, USA; Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857 Singapore, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; SingHealth Duke-NUS Global Health Institute, Singapore.
| |
Collapse
|
5
|
Pomatto MAC, Gai C, Negro F, Massari L, Deregibus MC, Grange C, De Rosa FG, Camussi G. Plant-Derived Extracellular Vesicles as a Delivery Platform for RNA-Based Vaccine: Feasibility Study of an Oral and Intranasal SARS-CoV-2 Vaccine. Pharmaceutics 2023; 15:pharmaceutics15030974. [PMID: 36986835 PMCID: PMC10058531 DOI: 10.3390/pharmaceutics15030974] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023] Open
Abstract
Plant-derived extracellular vesicles (EVs) may represent a platform for the delivery of RNA-based vaccines, exploiting their natural membrane envelope to protect and deliver nucleic acids. Here, EVs extracted from orange (Citrus sinensis) juice (oEVs) were investigated as carriers for oral and intranasal SARS-CoV-2 mRNA vaccine. oEVs were efficiently loaded with different mRNA molecules (coding N, subunit 1 and full S proteins) and the mRNA was protected from degrading stress (including RNase and simulated gastric fluid), delivered to target cells and translated into protein. APC cells stimulated with oEVs loaded with mRNAs induced T lymphocyte activation in vitro. The immunization of mice with oEVs loaded with S1 mRNA via different routes of administration including intramuscular, oral and intranasal stimulated a humoral immune response with production of specific IgM and IgG blocking antibodies and a T cell immune response, as suggested by IFN-γ production by spleen lymphocytes stimulated with S peptide. Oral and intranasal administration also triggered the production of specific IgA, the mucosal barrier in the adaptive immune response. In conclusion, plant-derived EVs represent a useful platform for mRNA-based vaccines administered not only parentally but also orally and intranasally.
Collapse
Affiliation(s)
- Margherita A. C. Pomatto
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
- Correspondence: (M.A.C.P.); (G.C.)
| | - Chiara Gai
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | | | | | - Maria Chiara Deregibus
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Cristina Grange
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Francesco Giuseppe De Rosa
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Giovanni Camussi
- EvoBiotech s.r.l., 10122 Turin, Italy
- Department of Medical Science, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, 10126 Turin, Italy
- Correspondence: (M.A.C.P.); (G.C.)
| |
Collapse
|
6
|
Hughes SM, Levy CN, Katz R, Lokken EM, Anahtar MN, Hall MB, Bradley F, Castle PE, Cortez V, Doncel GF, Fichorova R, Fidel PL, Fowke KR, Francis SC, Ghosh M, Hwang LY, Jais M, Jespers V, Joag V, Kaul R, Kyongo J, Lahey T, Li H, Makinde J, McKinnon LR, Moscicki AB, Novak RM, Patel MV, Sriprasert I, Thurman AR, Yegorov S, Mugo NR, Roxby AC, Micks E, Hladik F. Changes in concentrations of cervicovaginal immune mediators across the menstrual cycle: a systematic review and meta-analysis of individual patient data. BMC Med 2022; 20:353. [PMID: 36195867 PMCID: PMC9533580 DOI: 10.1186/s12916-022-02532-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 08/16/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hormonal changes during the menstrual cycle play a key role in shaping immunity in the cervicovaginal tract. Cervicovaginal fluid contains cytokines, chemokines, immunoglobulins, and other immune mediators. Many studies have shown that the concentrations of these immune mediators change throughout the menstrual cycle, but the studies have often shown inconsistent results. Our understanding of immunological correlates of the menstrual cycle remains limited and could be improved by meta-analysis of the available evidence. METHODS We performed a systematic review and meta-analysis of cervicovaginal immune mediator concentrations throughout the menstrual cycle using individual participant data. Study eligibility included strict definitions of the cycle phase (by progesterone or days since the last menstrual period) and no use of hormonal contraception or intrauterine devices. We performed random-effects meta-analyses using inverse-variance pooling to estimate concentration differences between the follicular and luteal phases. In addition, we performed a new laboratory study, measuring select immune mediators in cervicovaginal lavage samples. RESULTS We screened 1570 abstracts and identified 71 eligible studies. We analyzed data from 31 studies, encompassing 39,589 concentration measurements of 77 immune mediators made on 2112 samples from 871 participants. Meta-analyses were performed on 53 immune mediators. Antibodies, CC-type chemokines, MMPs, IL-6, IL-16, IL-1RA, G-CSF, GNLY, and ICAM1 were lower in the luteal phase than the follicular phase. Only IL-1α, HBD-2, and HBD-3 were elevated in the luteal phase. There was minimal change between the phases for CXCL8, 9, and 10, interferons, TNF, SLPI, elafin, lysozyme, lactoferrin, and interleukins 1β, 2, 10, 12, 13, and 17A. The GRADE strength of evidence was moderate to high for all immune mediators listed here. CONCLUSIONS Despite the variability of cervicovaginal immune mediator measurements, our meta-analyses show clear and consistent changes during the menstrual cycle. Many immune mediators were lower in the luteal phase, including chemokines, antibodies, matrix metalloproteinases, and several interleukins. Only interleukin-1α and beta-defensins were higher in the luteal phase. These cyclical differences may have consequences for immunity, susceptibility to infection, and fertility. Our study emphasizes the need to control for the effect of the menstrual cycle on immune mediators in future studies.
Collapse
Affiliation(s)
- Sean M Hughes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Claire N Levy
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Erica M Lokken
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Melis N Anahtar
- Ragon Institute of MIT and Harvard, Massachusetts General Hospital, Boston, MA, USA
| | | | - Frideborg Bradley
- Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Philip E Castle
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Valerie Cortez
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - Raina Fichorova
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Paul L Fidel
- Louisiana State University Health, New Orleans, LA, USA
| | - Keith R Fowke
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Suzanna C Francis
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Mimi Ghosh
- Department of Epidemiology, The George Washington University, Washington, DC, USA
| | - Loris Y Hwang
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mariel Jais
- Office of Laboratory Safety, The George Washington University, Washington, DC, USA
| | | | - Vineet Joag
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Rupert Kaul
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jordan Kyongo
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Timothy Lahey
- University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Huiying Li
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Julia Makinde
- IAVI Human Immunology Laboratory, Imperial College, London, England, UK
- IAVI, New York, NY, USA
| | - Lyle R McKinnon
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi, Kenya
| | - Anna-Barbara Moscicki
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Mickey V Patel
- Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Intira Sriprasert
- Department of OB/GYN, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Sergey Yegorov
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Nelly Rwamba Mugo
- Department of Global Health, University of Washington, Seattle, WA, USA
- Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Alison C Roxby
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA
| | - Elizabeth Micks
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA.
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
- Vaccine and Infectious Disease Division, Fred Hutch, Seattle, WA, USA.
| |
Collapse
|
7
|
Bo Y, Wang H. Materials‐based vaccines for infectious diseases. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1824. [PMID: 35708013 PMCID: PMC9541041 DOI: 10.1002/wnan.1824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/16/2022] [Indexed: 12/03/2022]
Abstract
Infectious diseases that result from pathogen infection are among the leading causes of human death, with pathogens such as human immunodeficiency virus, malaria, influenza, and ongoing SARS‐COV‐2 viruses constantly threatening the global population. While the mechanisms behind various infectious diseases are not entirely clear and thus retard the development of effective therapeutics, vaccines have served as a universal approach to containing infectious diseases. However, conventional vaccines that solely consist of antigens or simply mix antigens and adjuvants have failed to control various highly infective or deadly pathogens. Biomaterials‐based vaccines have provided a promising solution due to their ability to synergize the function of antigens and adjuvants, troubleshoot delivery issues, home and manipulate immune cells in situ. In this review, we will summarize different types of materials‐based vaccines for generating cellular and humoral responses against pathogens and discuss the design criteria for amplifying the efficacy of materials‐based vaccines against infectious diseases. This article is categorized under:Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease
Collapse
Affiliation(s)
- Yang Bo
- Department of Materials Science and Engineering University of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Hua Wang
- Department of Materials Science and Engineering University of Illinois at Urbana‐Champaign Urbana Illinois USA
- Cancer Center at Illinois (CCIL) Urbana Illinois USA
- Department of Bioengineering University of Illinois at Urbana‐Champaign Urbana Illinois USA
- Carle College of Medicine University of Illinois at Urbana‐Champaign Urbana Illinois USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign Urbana Illinois USA
| |
Collapse
|
8
|
From Concept to Clinical Product: A Brief History of the Novel Shigella Invaplex Vaccine’s Refinement and Evolution. Vaccines (Basel) 2022; 10:vaccines10040548. [PMID: 35455297 PMCID: PMC9025769 DOI: 10.3390/vaccines10040548] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/12/2022] [Accepted: 03/23/2022] [Indexed: 01/13/2023] Open
Abstract
The Shigella invasin complex or Invaplex vaccine is a unique subunit approach to generate a protective immune response. Invaplex is a large, macromolecular complex consisting of the major Shigella antigens: lipopolysaccharide (LPS) and the invasion plasmid antigen (Ipa) proteins B and C. Over the past several decades, the vaccine has progressed from initial observations through pre-clinical studies to cGMP manufacture and clinical evaluations. The Invaplex product maintains unique biological properties associated with the invasiveness of virulent shigellae and also presents both serotype-specific epitopes, as well as highly conserved invasin protein epitopes, to the immunized host. The vaccine product has evolved from a native product isolated from wild-type shigellae (native Invaplex) to a more defined vaccine produced from purified LPS and recombinant IpaB and IpaC (artificial Invaplex). Each successive “generation” of the vaccine is derived from earlier versions, resulting in improved immunogenicity, homogeneity and effectiveness. The current vaccine, detoxified artificial Invaplex (InvaplexAR-Detox), was developed for parenteral administration by incorporating LPS with under-acylated lipid A. InvaplexAR-Detox has demonstrated an excellent safety and immunogenicity profile in initial clinical studies and is advancing toward evaluations in the target populations of children and travelers to endemic countries.
Collapse
|
9
|
Feng F, Wen Z, Chen J, Yuan Y, Wang C, Sun C. Strategies to Develop a Mucosa-Targeting Vaccine against Emerging Infectious Diseases. Viruses 2022; 14:v14030520. [PMID: 35336927 PMCID: PMC8952777 DOI: 10.3390/v14030520] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
Numerous pathogenic microbes, including viruses, bacteria, and fungi, usually infect the host through the mucosal surfaces of the respiratory tract, gastrointestinal tract, and reproductive tract. The mucosa is well known to provide the first line of host defense against pathogen entry by physical, chemical, biological, and immunological barriers, and therefore, mucosa-targeting vaccination is emerging as a promising strategy for conferring superior protection. However, there are still many challenges to be solved to develop an effective mucosal vaccine, such as poor adhesion to the mucosal surface, insufficient uptake to break through the mucus, and the difficulty in avoiding strong degradation through the gastrointestinal tract. Recently, increasing efforts to overcome these issues have been made, and we herein summarize the latest findings on these strategies to develop mucosa-targeting vaccines, including a novel needle-free mucosa-targeting route, the development of mucosa-targeting vectors, the administration of mucosal adjuvants, encapsulating vaccines into nanoparticle formulations, and antigen design to conjugate with mucosa-targeting ligands. Our work will highlight the importance of further developing mucosal vaccine technology to combat the frequent outbreaks of infectious diseases.
Collapse
Affiliation(s)
- Fengling Feng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Ziyu Wen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Jiaoshan Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yue Yuan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Congcong Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Caijun Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (F.F.); (Z.W.); (J.C.); (Y.Y.); (C.W.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- Correspondence:
| |
Collapse
|
10
|
Jiang L, Driedonks TA, Jong WS, Dhakal S, Bart van den Berg van Saparoea H, Sitaras I, Zhou R, Caputo C, Littlefield K, Lowman M, Chen M, Lima G, Gololobova O, Smith B, Mahairaki V, Riley Richardson M, Mulka KR, Lane AP, Klein SL, Pekosz A, Brayton C, Mankowski JL, Luirink J, Villano JS, Witwer KW. A bacterial extracellular vesicle-based intranasal vaccine against SARS-CoV-2 protects against disease and elicits neutralizing antibodies to wild-type and Delta variants. J Extracell Vesicles 2022; 11:e12192. [PMID: 35289114 PMCID: PMC8920961 DOI: 10.1002/jev2.12192] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Several vaccines have been introduced to combat the coronavirus infectious disease-2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Current SARS-CoV-2 vaccines include mRNA-containing lipid nanoparticles or adenoviral vectors that encode the SARS-CoV-2 Spike (S) protein of SARS-CoV-2, inactivated virus, or protein subunits. Despite growing success in worldwide vaccination efforts, additional capabilities may be needed in the future to address issues such as stability and storage requirements, need for vaccine boosters, desirability of different routes of administration, and emergence of SARS-CoV-2 variants such as the Delta variant. Here, we present a novel, well-characterized SARS-CoV-2 vaccine candidate based on extracellular vesicles (EVs) of Salmonella typhimurium that are decorated with the mammalian cell culture-derived Spike receptor-binding domain (RBD). RBD-conjugated outer membrane vesicles (RBD-OMVs) were used to immunize the golden Syrian hamster (Mesocricetus auratus) model of COVID-19. Intranasal immunization resulted in high titres of blood anti-RBD IgG as well as detectable mucosal responses. Neutralizing antibody activity against wild-type and Delta variants was evident in all vaccinated subjects. Upon challenge with live virus, hamsters immunized with RBD-OMV, but not animals immunized with unconjugated OMVs or a vehicle control, avoided body mass loss, had lower virus titres in bronchoalveolar lavage fluid, and experienced less severe lung pathology. Our results emphasize the value and versatility of OMV-based vaccine approaches.
Collapse
Affiliation(s)
- Linglei Jiang
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Tom A.P. Driedonks
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | | | - Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | | | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Ruifeng Zhou
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Christopher Caputo
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Kirsten Littlefield
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Maggie Lowman
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mengfei Chen
- Department of Otolaryngology‐Head and Neck SurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Gabriela Lima
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Barbara Smith
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Vasiliki Mahairaki
- Department of Genetic Medicine and The Richman Family Precision Medicine Centre of Excellence in Alzheimer's Disease Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - M. Riley Richardson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Kathleen R. Mulka
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Andrew P. Lane
- Department of Otolaryngology‐Head and Neck SurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyThe Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Cory Brayton
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Joseph L. Mankowski
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Neurology and NeurosurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Joen Luirink
- Abera Bioscience ABUppsalaSweden
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS)Vrije UniversiteitAmsterdamThe Netherlands
| | - Jason S. Villano
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Genetic Medicine and The Richman Family Precision Medicine Centre of Excellence in Alzheimer's Disease Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Neurology and NeurosurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
11
|
Jiang L, Driedonks TA, Jong WS, Dhakal S, van den Berg van Saparoea HB, Sitaras I, Zhou R, Caputo C, Littlefield K, Lowman M, Chen M, Lima G, Gololobova O, Smith B, Mahairaki V, Richardson MR, Mulka KR, Lane AP, Klein SL, Pekosz A, Brayton CF, Mankowski JL, Luirink J, Villano JS, Witwer KW. A bacterial extracellular vesicle-based intranasal vaccine against SARS-CoV-2 protects against disease and elicits neutralizing antibodies to wild-type and Delta variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.06.28.450181. [PMID: 35132418 PMCID: PMC8820665 DOI: 10.1101/2021.06.28.450181] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Several vaccines have been introduced to combat the coronavirus infectious disease-2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Current SARS-CoV-2 vaccines include mRNA-containing lipid nanoparticles or adenoviral vectors that encode the SARS-CoV-2 Spike (S) protein of SARS-CoV-2, inactivated virus, or protein subunits. Despite growing success in worldwide vaccination efforts, additional capabilities may be needed in the future to address issues such as stability and storage requirements, need for vaccine boosters, desirability of different routes of administration, and emergence of SARS-CoV-2 variants such as the Delta variant. Here, we present a novel, well-characterized SARS-CoV-2 vaccine candidate based on extracellular vesicles (EVs) of Salmonella typhimurium that are decorated with the mammalian cell culture-derived Spike receptor-binding domain (RBD). RBD-conjugated outer membrane vesicles (RBD-OMVs) were used to immunize the golden Syrian hamster ( Mesocricetus auratus ) model of COVID-19. Intranasal immunization resulted in high titers of blood anti-RBD IgG as well as detectable mucosal responses. Neutralizing antibody activity against wild-type and Delta variants was evident in all vaccinated subjects. Upon challenge with live virus, hamsters immunized with RBD-OMV, but not animals immunized with unconjugated OMVs or a vehicle control, avoided body mass loss, had lower virus titers in bronchoalveolar lavage fluid, and experienced less severe lung pathology. Our results emphasize the value and versatility of OMV-based vaccine approaches.
Collapse
Affiliation(s)
- Linglei Jiang
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tom A.P. Driedonks
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ruifeng Zhou
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christopher Caputo
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kirsten Littlefield
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Maggie Lowman
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mengfei Chen
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gabriela Lima
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Olesia Gololobova
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara Smith
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vasiliki Mahairaki
- Department of Genetic Medicine and The Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - M. Riley Richardson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen R. Mulka
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew P. Lane
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Cory F. Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph L. Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joen Luirink
- Abera Bioscience AB, Uppsala, Sweden
- Department of Molecular Microbiology, Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Vrije Universiteit, Amsterdam, The Netherlands
- These authors are designated as co-corresponding authors. ; ;
| | - Jason S. Villano
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- These authors are designated as co-corresponding authors. ; ;
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine and The Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- These authors are designated as co-corresponding authors. ; ;
| |
Collapse
|
12
|
Sobia P, Archary D. Preventive HIV Vaccines-Leveraging on Lessons from the Past to Pave the Way Forward. Vaccines (Basel) 2021; 9:vaccines9091001. [PMID: 34579238 PMCID: PMC8472969 DOI: 10.3390/vaccines9091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/05/2022] Open
Abstract
Almost four decades on, since the 1980’s, with hundreds of HIV vaccine candidates tested in both non-human primates and humans, and several HIV vaccines trials later, an efficacious HIV vaccine continues to evade us. The enormous worldwide genetic diversity of HIV, combined with HIV’s inherent recombination and high mutation rates, has hampered the development of an effective vaccine. Despite the advent of antiretrovirals as pre-exposure prophylaxis and preventative treatment, which have shown to be effective, HIV infections continue to proliferate, highlighting the great need for a vaccine. Here, we provide a brief history for the HIV vaccine field, with the most recent disappointments and advancements. We also provide an update on current passive immunity trials, testing proof of the concept of the most clinically advanced broadly neutralizing monoclonal antibodies for HIV prevention. Finally, we include mucosal immunity, the importance of vaccine-elicited immune responses and the challenges thereof in the most vulnerable environment–the female genital tract and the rectal surfaces of the gastrointestinal tract for heterosexual and men who have sex with men transmissions, respectively.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: ; Tel.: +27-(0)-31-655-0540
| |
Collapse
|
13
|
Rehman S, Ravinayagam V, Nahvi I, Aldossary H, Al-Shammari M, Amiri MSA, Kishore U, Al-Suhaimi EA. Immunity, Sex Hormones, and Environmental Factors as Determinants of COVID-19 Disparity in Women. Front Immunol 2021; 12:680845. [PMID: 34484179 PMCID: PMC8416472 DOI: 10.3389/fimmu.2021.680845] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/29/2021] [Indexed: 01/16/2023] Open
Abstract
The current coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome virus 2 (SARS-CoV-2), has resulted in a major global pandemic, causing extreme morbidity and mortality. Few studies appear to suggest a significant impact of gender in morbidity and mortality, where men are reported at a higher risk than women. The infectivity, transmissibility, and varying degree of disease manifestation (mild, modest, and severe) in population studies reinforce the importance of a number of genetic and epigenetic factors, in the context of immune response and gender. The present review dwells on several contributing factors such as a stronger innate immune response, estrogen, angiotensin-converting enzyme 2 gene, and microbiota, which impart greater resistance to the SARS-CoV-2 infection and disease progression in women. In addition, the underlying importance of associated microbiota and certain environmental factors in gender-based disparity pertaining to the mortality and morbidity due to COVID-19 in women has also been addressed.
Collapse
Affiliation(s)
- Suriya Rehman
- Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Vijaya Ravinayagam
- Deanship of Scientific Research and Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Insha Nahvi
- Department of Basic Sciences, Preparatory Year Deanship, King Faisal University, Al Hofuf, Saudi Arabia
| | - Hanan Aldossary
- Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Maha Al-Shammari
- Department of Public Health, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mai Saad Al Amiri
- Department of Obstetrics and Gynecology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Ebtesam A. Al-Suhaimi
- Biology Department, College of Science and Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
14
|
Cho CS, Hwang SK, Gu MJ, Kim CG, Kim SK, Ju DB, Yun CH, Kim HJ. Mucosal Vaccine Delivery Using Mucoadhesive Polymer Particulate Systems. Tissue Eng Regen Med 2021; 18:693-712. [PMID: 34304387 PMCID: PMC8310561 DOI: 10.1007/s13770-021-00373-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/23/2022] Open
Abstract
Vaccination has been recently attracted as one of the most successful medical treatments of the prevalence of many infectious diseases. Mucosal vaccination has been interested in many researchers because mucosal immune responses play part in the first line of defense against pathogens. However, mucosal vaccination should find out an efficient antigen delivery system because the antigen should be protected from degradation and clearance, it should be targeted to mucosal sites, and it should stimulate mucosal and systemic immunity. Accordingly, mucoadhesive polymeric particles among the polymeric particles have gained much attention because they can protect the antigen from degradation, prolong the residence time of the antigen at the target site, and control the release of the loaded vaccine, and results in induction of mucosal and systemic immune responses. In this review, we discuss advances in the development of several kinds of mucoadhesive polymeric particles for mucosal vaccine delivery.
Collapse
Affiliation(s)
- Chong-Su Cho
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| | - Soo-Kyung Hwang
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea ,grid.31501.360000 0004 0470 5905Lab. of Adhesion & Bio-Composites, Department of Agriculture, Forestry and Bioresources, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| | - Min-Jeong Gu
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| | - Cheol-Gyun Kim
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| | - Seo-Kyung Kim
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| | - Do-Bin Ju
- grid.31501.360000 0004 0470 5905Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea. .,Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, Seoul, Republic of Korea. .,Center for Food and Bioconvergence, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Hyun-Joong Kim
- Lab. of Adhesion & Bio-Composites, Department of Agriculture, Forestry and Bioresources, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
15
|
Effects of Hormone Therapy and Flavonoids Capable on Reversal of Menopausal Immune Senescence. Nutrients 2021; 13:nu13072363. [PMID: 34371873 PMCID: PMC8308838 DOI: 10.3390/nu13072363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/19/2021] [Accepted: 07/07/2021] [Indexed: 01/15/2023] Open
Abstract
Menopause, probably the most important natural change in a woman’s life and a major component of female senescence, is characterized, inter alia, by cessation of ovarian estrogen and progesterone production, resulting in a gradual deterioration of the female immune system. Hormone replacement therapy (HRT) is used in postmenopausal women to relieve some of the peri- and postmenopausal symptoms, while there is also evidence that the therapy may additionally partially reverse menopausal immune senescence. Flavonoids, and especially isoflavones, are widely used for the treatment of menopausal symptoms, although it is not at present clear whether they can reverse or alleviate other menopausal changes. HRT reverses the menopausal CD4/CD8 ratio and also limits the general peri- and postmenopausal inflammatory state. Moreover, the increased levels of interleukins (IL)-1β, IL-6, and IL-8, as well as of tumor necrosis factor-α (TNF-α) are decreased after the initiation of HRT. However, some reports show no effect of HRT on IL-4, IL-10, and IL-12. It is thus evident that the molecular pathways connecting HRT and female immune senescence need to be clarified. Interestingly, recent studies have suggested that the anti-inflammatory properties of isoflavones possibly interact with inflammatory cytokines when applied in menopause treatments, thereby potentially reversing immune senescence. This narrative review presents the latest data on the effect of menopausal therapies, including administration of flavonoid-rich products, on age-associated immune senescence reversal with the aim of revealing possible directions for future research and treatment development.
Collapse
|
16
|
Camp JV, Wilson RL, Singletary M, Blanchard JL, Aldovini A, Kaminski RW, Oaks EV, Kozlowski PA. Invaplex functions as an intranasal adjuvant for subunit and DNA vaccines co-delivered in the nasal cavity of nonhuman primates. Vaccine X 2021; 8:100105. [PMID: 34258576 PMCID: PMC8255935 DOI: 10.1016/j.jvacx.2021.100105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/04/2021] [Accepted: 06/11/2021] [Indexed: 01/15/2023] Open
Abstract
Development of intranasal vaccines for HIV-1 and other mucosal pathogens has been hampered by the lack of adjuvants that can be given safely to humans. We have found that an intranasal Shigella vaccine (Invaplex) which is well tolerated in humans can also function as an adjuvant for intranasal protein and DNA vaccines in mice. To determine whether Invaplex could potentially adjuvant similar vaccines in humans, we simultaneously administered a simian immunodeficiency virus (SIV) envelope (Env) protein and DNA encoding simian-human immunodeficiency virus (SHIV) with or without Invaplex in the nasal cavity of female rhesus macaques. Animals were intranasally boosted with adenoviral vectors expressing SIV env or gag,pol to evaluate memory responses. Anti-SIV antibodies in sera and nasal, genital tract and rectal secretions were quantitated by ELISA. Intracellular cytokine staining was used to measure Th1-type T cells in blood. Macaques given DNA/protein immunizations with 0.5 mg Invaplex developed greater serum IgG, nasal IgA and cervicovaginal IgA responses to SIV Env and SHIV Gag,Pol proteins when compared to non-adjuvanted controls. Rectal IgA responses to Env were only briefly elevated and not observed to Gag,Pol. Invaplex increased frequencies of IFNγ-producing CD4 and CD8 T cells to the Env protein, but not T cell responses induced by the DNA. Ad-SIV boosting increased Env-specific polyfunctional T cells and Env- and Gag,Pol-specific antibodies in serum and all secretions. The data suggest that Invaplex could be highly effective as an adjuvant for intranasal protein vaccines in humans, especially those intended to prevent infections in the genital or respiratory tract.
Collapse
Key Words
- Ad, adenovirus
- CVS, cervicovaginal secretions
- Env, envelope
- HIV/AIDS
- ICS, intracellular cytokine staining
- IM, intramuscular
- IN, intranasal
- IgA
- Mucosal adjuvant
- NHP, nonhuman primates
- NS, nasal secretions
- RS, rectal secretions
- Reproductive
- Respiratory tract
- S-IgA, secretory IgA
- Th, T helper
Collapse
Affiliation(s)
- Jeremy V Camp
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Robert L Wilson
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Morgan Singletary
- Tulane National Primate Research Center, Division of Veterinary Medicine, Covington, LA 70433, USA
| | - James L Blanchard
- Tulane National Primate Research Center, Division of Veterinary Medicine, Covington, LA 70433, USA
| | - Anna Aldovini
- Departments of Medicine and Pediatrics, Children's Hospital and Harvard, Boston, MA 02115, USA
| | - Robert W Kaminski
- Department of Subunit Enteric Vaccines and Immunology, Division of Bacterial and Rickettsial Diseases, The Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Edwin V Oaks
- Department of Subunit Enteric Vaccines and Immunology, Division of Bacterial and Rickettsial Diseases, The Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
17
|
Velarde de la Cruz E, Wang L, Bose D, Gangadhara S, Wilson RL, Amara RR, Kozlowski PA, Aldovini A. Oral Vaccination Approaches for Anti-SHIV Immunity. Front Immunol 2021; 12:702705. [PMID: 34234789 PMCID: PMC8256843 DOI: 10.3389/fimmu.2021.702705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/04/2021] [Indexed: 11/16/2022] Open
Abstract
We modified a Sabin Oral Poliovirus Vaccine (OPV) vector to permit secretion of the antigens of interest with the goal of improving anti-HIV Env humoral responses in a SHIV mucosal immunization composed of DNA and recombinant OPVs. We evaluated stimulation of systemic and mucosal cell-mediated and humoral immunity in Rhesus macaques by two regimens, both involving a prime with a SHIVBG505 DNA construct producing non-infectious particles formulated in lipid nanoparticles, administered in the oral cavity, and two different viral vector boostings, administered in the oral cavity and intestinally. Group 1 was boosted with rMVA-SHIVBG505, expressing SIV Gag/Pol and HIVBG505 Env. Group 2 was boosted with a SHIVBG505-OPV vaccine including a non-secreting SIVmac239CA-p6-OPV, expressing Gag CA, NC and p6 proteins, and a HIVBG505C1-V2-OPV, secreting the C1-V2 fragment of HIV EnvBG505, recognized by the broadly neutralizing antibody PG16. A time course analysis of anti-SHIV Gag and Env CD4+ and CD8+ T-cell responses in PBMC and in lymph node, rectal, and vaginal MNC was carried out. Both regimens stimulated significant cell-mediated responses in all compartments, with SHIVBG505-OPV immunization stimulating more significant levels of responses than rMVA- SHIVBG505. Boolean analysis of these responses revealed predominantly monofunctional responses with multifunctional responses also present in all tissues. Stimulation of antibody responses was disappointing in both groups with negative anti-SHIV IgG in plasma, and IgA in salivary, rectal and vaginal secretions being restricted to a few animals. After repeated rectal challenge with SHIVBG505, two Group 1 animals remained uninfected at challenge termination. No significant differences were observed in post-infection viral loads between groups. After the acute phase decline, CD4+ T cell percentages returned to normal levels in vaccinated as well as control animals. However, when compared to controls, vaccinate groups had more significant preservation of PBMC and rectal MNC Th17/Treg ratios, considered the strongest surrogate marker of progression to AIDS. We conclude that the vaccine platforms used in this study are insufficient to stimulate significant humoral immunity at the tested doses and schedule but sufficient to stimulate significant mucosal and systemic cell-mediated immunity, impacting the preservation of key Th17 CD4+ T cells in blood and rectal mucosa.
Collapse
Affiliation(s)
- Erandi Velarde de la Cruz
- Department of Medicine, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Lingyun Wang
- Department of Medicine, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Deepanwita Bose
- Department of Medicine, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Sailaja Gangadhara
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, United States
| | - Robert L. Wilson
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Rama R. Amara
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, United States
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Anna Aldovini
- Department of Medicine, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Logerot S, Figueiredo-Morgado S, Charmeteau-de-Muylder B, Sandouk A, Drillet-Dangeard AS, Bomsel M, Bourgault-Villada I, Couëdel-Courteille A, Cheynier R, Rancez M. IL-7-Adjuvanted Vaginal Vaccine Elicits Strong Mucosal Immune Responses in Non-Human Primates. Front Immunol 2021; 12:614115. [PMID: 33717097 PMCID: PMC7947860 DOI: 10.3389/fimmu.2021.614115] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022] Open
Abstract
Mucosal immune responses are crucial in protecting against pathogens entering through mucosal surfaces. However, due to poor T-cell responsiveness upon mucosal antigenic stimulation, mucosal immunity remains difficult to obtain through vaccines and requires appropriate adjuvants. We previously demonstrated that administered systemically to healthy macaques or locally expressed in the intestinal mucosa of acutely SIV-infected macaques, interleukin-7 (IL-7) triggers chemokine expression and immune cell homing into mucosae, suggesting its important role in the development of mucosal immune responses. We therefore examined whether local delivery of recombinant glycosylated simian IL-7 (rs-IL-7gly) to the vaginal mucosa of rhesus macaques could prepare the lower female genital tract (FGT) for subsequent immunization and act as an efficient mucosal adjuvant. First, we showed that local administration of rs-IL-7gly triggers vaginal overexpression of chemokines and infiltration of mDCs, macrophages, NKs, B- and T-cells in the lamina propria while MamuLa-DR+ APCs accumulated in the epithelium. Subsequent mucosal anti-DT immunization in macaques resulted in a faster, stronger, and more persistent mucosal antibody response compared to DT-immunization alone. Indeed, we detected robust productions of DT-specific IgAs and IgGs in their vaginal secretions and identified cells secreting DT-specific IgAs in their vaginal mucosa and IgGs in draining lymph nodes. Finally, the expression of chemokines involved in the organization of tertiary lymphoid structures (TLS) was only increased in the vaginal mucosa of IL-7-adjuvanted immunized macaques. Interestingly, TLSs developed around PNAd+ high endothelial venules in their lower FGT sampled 2 weeks after the last immunization. Non-traumatic vaginal administration of rs-IL-7gly prepares the mucosa to respond to subsequent local immunization and allows the development of a strong mucosal immune response in macaques, through the chemokine-dependent recruitment of immune cells, the activation of mDCs and the formation of TLSs. The localization of DT-specific IgA+ plasma cells in the upper vaginal mucosa argues for their contribution to the production of specific immunoglobulins in the vaginal secretions. Our results highlight the potential of IL-7 as a potent mucosal adjuvant to stimulate the FGT immune system and elicit vaginal antibody responses to local immunization, which is the most promising way to confer protection against many sexually transmitted diseases.
Collapse
Affiliation(s)
- Sandrine Logerot
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Suzanne Figueiredo-Morgado
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Bénédicte Charmeteau-de-Muylder
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Abdelkader Sandouk
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Anne-Sophie Drillet-Dangeard
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Morgane Bomsel
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Isabelle Bourgault-Villada
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Anne Couëdel-Courteille
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Rémi Cheynier
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| | - Magali Rancez
- Laboratory of Dendritic Cells, B Lymphocytes and Cytokines in their Microenvironment During Viral Infections and Cancer, Department of Infection, Immunity and Inflammation, Université de Paris, INSERM, CNRS, Institut Cochin, Paris, France
| |
Collapse
|
19
|
Chaudhary O, Wang L, Bose D, Narayan V, Yeh MT, Carville A, Clements JD, Andino R, Kozlowski PA, Aldovini A. Comparative Evaluation of Prophylactic SIV Vaccination Modalities Administered to the Oral Cavity. AIDS Res Hum Retroviruses 2020; 36:984-997. [PMID: 32962398 PMCID: PMC7703093 DOI: 10.1089/aid.2020.0157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Attempts to develop a protective human immunodeficiency virus (HIV) vaccine have had limited success, especially in terms of inducing protective antibodies capable of neutralizing different viral strains. As HIV transmission occurs mainly via mucosal surfaces, HIV replicates significantly in the gastrointestinal tract, and the oral route of vaccination is a very convenient one to implement worldwide, we explored three SIV vaccine modalities administered orally and composed of simian immunodeficiency virus (SIV) DNA priming with different boosting immunogens, with the goal of evaluating whether they could provide lasting humoral and cellular responses, including at mucosal surfaces that are sites of HIV entry. Twenty-four Cynomolgus macaques (CyM) were primed with replication-incompetent SIV DNA provirus and divided into three groups for the following booster vaccinations, all administered in the oral cavity: Group 1 with recombinant SIV gp140 and Escherichia coli heat-labile toxin adjuvant dmLT, Group 2 with recombinant SIV-Oral Poliovirus (SIV-OPV), and Group 3 with recombinant SIV-modified vaccinia ankara (SIV-MVA). Cell-mediated responses were measured using blood, lymph node, rectal and vaginal mononuclear cells. Significant levels of systemic and mucosal T-cell responses against Gag and Env were observed in all groups. Some SIV-specific plasma IgG, rectal and salivary IgA antibodies were generated, mainly in animals that received SIV DNA + SIV-MVA, but no vaginal IgA was detected. Susceptibility to infection after SIVmac251 challenge was similar in vaccinated and nonvaccinated animals, but acute infection viremia levels were lower in the group that received SIV DNA + SIV-MVA. Nonvaccinated CyM maintained central memory and total CD4+ T-cell levels in the normal range during the 5 months of postinfection follow-up as did the vaccinated animals, precluding evaluation of vaccine impact on disease progression. We conclude that the oral cavity vaccination tested in these regimens can stimulate cell-mediated immunity systemically and mucosally, but humoral response stimulation was limited with the doses and the vaccine platforms used.
Collapse
Affiliation(s)
- Omkar Chaudhary
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Lingyun Wang
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Deepanwita Bose
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Vivek Narayan
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Ming Te Yeh
- Department of Microbiology and Immunology, UCSF, San Francisco, California, USA
| | | | - John D. Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Raul Andino
- Department of Microbiology and Immunology, UCSF, San Francisco, California, USA
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Anna Aldovini
- Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Gogoi H, Mansouri S, Jin L. The Age of Cyclic Dinucleotide Vaccine Adjuvants. Vaccines (Basel) 2020; 8:E453. [PMID: 32823563 PMCID: PMC7563944 DOI: 10.3390/vaccines8030453] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
As prophylactic vaccine adjuvants for infectious diseases, cyclic dinucleotides (CDNs) induce safe, potent, long-lasting humoral and cellular memory responses in the systemic and mucosal compartments. As therapeutic cancer vaccine adjuvants, CDNs induce potent anti-tumor immunity, including cytotoxic T cells and NK cells activation that achieve durable regression in multiple mouse models of tumors. Clinical trials are ongoing to fulfill the promise of CDNs (ClinicalTrials.gov: NCT02675439, NCT03010176, NCT03172936, and NCT03937141). However, in October 2018, the first clinical data with Merck's CDN MK-1454 showed zero activity as a monotherapy in patients with solid tumors or lymphomas (NCT03010176). Lately, the clinical trial from Aduro's CDN ADU-S100 monotherapy was also disappointing (NCT03172936). The emerging hurdle in CDN vaccine development calls for a timely re-evaluation of our understanding on CDN vaccine adjuvants. Here, we review the status of CDN vaccine adjuvant research, including their superior adjuvant activities, in vivo mode of action, and confounding factors that affect their efficacy in humans. Lastly, we discuss the strategies to overcome the hurdle and advance promising CDN adjuvants in humans.
Collapse
Affiliation(s)
| | | | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (H.G.); (S.M.)
| |
Collapse
|
21
|
Zhang E, Wang J, Li Y, Huang L, Wang Y, Yang Q. Comparison of oral and nasal immunization with inactivated porcine epidemic diarrhea virus on intestinal immunity in piglets. Exp Ther Med 2020; 20:1596-1606. [PMID: 32742391 PMCID: PMC7388329 DOI: 10.3892/etm.2020.8828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/20/2020] [Indexed: 12/11/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) has proven to be a major problem for the porcine industry worldwide. Conventional injectable vaccines induce effective systemic immune responses but are less effective in preventing PEDV at mucosal invasion sites, including the nasal or oral mucosa. Additionally, antigens delivered orally are easily degraded. Nasal immunization induces intestinal mucosal immune responses, which can aid in blocking viral invasion, and requires fewer antigen inoculation doses. Therefore, nasal immunizations are considered to be a potential approach to overcome viral infections. In the present study, nasal immunization of piglets was performed using inactivated PEDV combined with Bacillus subtilis as an immunopotentiator and the efficacy of nasal immunization was assessed. The results demonstrated that compared with oral immunization, piglets from the nasal immunization group exhibited higher levels of neutralizing antibodies (P<0.01) in the intestine, PEDV-specific immunoglobulin (Ig)G (P<0.01) in serum and PEDV-specific secretory IgA (SIgA) in saliva (P<0.01) and nasal secretions (P<0.01). An increased number of intestinal CD3+ T cells, IgA-secreting cells and intraepithelial lymphocytes (P<0.05) were also observed. Furthermore, the protein expression levels of interleukin-6 and interferon-γ, relative to the control PEDV infection, were also significantly elevated (P<0.05). The results of the present study indicate that nasal immunization is more effective at inducing the intestinal mucosal immune response, and provide new insights into a novel vaccination strategy that may be used to decrease the incidence of PEDV infections.
Collapse
Affiliation(s)
- En Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Jialu Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yuchen Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Lulu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yongheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| |
Collapse
|
22
|
Hepatitis B Vaccination Induces Mucosal Antibody Responses in the Female Genital Tract, Indicating Potential Mechanisms of Protection Against Infection. Sex Transm Dis 2020; 46:e53-e56. [PMID: 30444798 DOI: 10.1097/olq.0000000000000949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Vaccines against hepatitis B virus confer effective protection. Enzyme-linked immunosorbent assay was developed to test for specific antibodies in female genital tract secretions. Anti-hepatitis B IgG and IgA were detected in the cervicovaginal secretions of women after hepatitis B vaccination, indicating a potential genital tract role for neutralizing antibodies against sexually transmitted hepatitis B virus.
Collapse
|
23
|
Forsyth VS, Himpsl SD, Smith SN, Sarkissian CA, Mike LA, Stocki JA, Sintsova A, Alteri CJ, Mobley HLT. Optimization of an Experimental Vaccine To Prevent Escherichia coli Urinary Tract Infection. mBio 2020; 11:e00555-20. [PMID: 32345645 PMCID: PMC7188996 DOI: 10.1128/mbio.00555-20] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Urinary tract infections (UTI) affect half of all women at least once during their lifetime. The rise in the numbers of extended-spectrum beta-lactamase-producing strains and the potential for carbapenem resistance within uropathogenic Escherichia coli (UPEC), the most common causative agent of UTI, create an urgent need for vaccine development. Intranasal immunization of mice with UPEC outer membrane iron receptors FyuA, Hma, IreA, and IutA, conjugated to cholera toxin, provides protection in the bladder or kidneys under conditions of challenge with UPEC strain CFT073 or strain 536. On the basis of these data, we sought to optimize the vaccination route (intramuscular, intranasal, or subcutaneous) in combination with adjuvants suitable for human use, including aluminum hydroxide gel (alum), monophosphoryl lipid A (MPLA), unmethylated CpG synthetic oligodeoxynucleotides (CpG), polyinosinic:polycytidylic acid (polyIC), and mutated heat-labile E. coli enterotoxin (dmLT). Mice intranasally vaccinated with dmLT-IutA and dmLT-Hma displayed significant reductions in bladder colonization (86-fold and 32-fold, respectively), with 40% to 42% of mice having no detectable CFU. Intranasal vaccination of mice with CpG-IutA and polyIC-IutA significantly reduced kidney colonization (131-fold) and urine CFU (22-fold), respectively. dmLT generated the most consistently robust antibody response in intranasally immunized mice, while MPLA and alum produced greater concentrations of antigen-specific serum IgG with intramuscular immunization. On the basis of these results, we conclude that intranasal administration of Hma or IutA formulated with dmLT adjuvant provides the greatest protection from UPEC UTI. This report advances our progress toward a vaccine against uncomplicated UTI, which will significantly improve the quality of life for women burdened by recurrent UTI and enable better antibiotic stewardship.IMPORTANCE Urinary tract infections (UTI) are among the most common bacterial infection in humans, affecting half of all women at least once during their lifetimes. The rise in antibiotic resistance and health care costs emphasizes the need to develop a vaccine against the most common UTI pathogen, Escherichia coli Vaccinating mice intranasally with a detoxified heat-labile enterotoxin and two surface-exposed receptors, Hma or IutA, significantly reduced bacterial burden in the bladder. This work highlights progress in the development of a UTI vaccine formulated with adjuvants suitable for human use and antigens that encode outer membrane iron receptors required for infection in the iron-limited urinary tract.
Collapse
Affiliation(s)
- Valerie S Forsyth
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Stephanie D Himpsl
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sara N Smith
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Christina A Sarkissian
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Laura A Mike
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jolie A Stocki
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Anna Sintsova
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Christopher J Alteri
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Harry L T Mobley
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Wallis J, Shenton DP, Carlisle RC. Novel approaches for the design, delivery and administration of vaccine technologies. Clin Exp Immunol 2019; 196:189-204. [PMID: 30963549 PMCID: PMC6468175 DOI: 10.1111/cei.13287] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2019] [Indexed: 12/20/2022] Open
Abstract
It is easy to argue that vaccine development represents humankind's most important and successful endeavour, such is the impact that vaccination has had on human morbidity and mortality over the last 200 years. During this time the original method of Jenner and Pasteur, i.e. that of injecting live-attenuated or inactivated pathogens, has been developed and supplemented with a wide range of alternative approaches which are now in clinical use or under development. These next-generation technologies have been designed to produce a vaccine that has the effectiveness of the original live-attenuated and inactivated vaccines, but without the associated risks and limitations. Indeed, the method of development has undoubtedly moved away from Pasteur's three Is paradigm (isolate, inactivate, inject) towards an approach of rational design, made possible by improved knowledge of the pathogen-host interaction and the mechanisms of the immune system. These novel vaccines have explored methods for targeted delivery of antigenic material, as well as for the control of release profiles, so that dosing regimens can be matched to the time-lines of immune system stimulation and the realities of health-care delivery in dispersed populations. The methods by which vaccines are administered are also the subject of intense research in the hope that needle and syringe dosing, with all its associated issues regarding risk of injury, cross-infection and patient compliance, can be replaced. This review provides a detailed overview of new vaccine vectors as well as information pertaining to the novel delivery platforms under development.
Collapse
Affiliation(s)
- J. Wallis
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| | - D. P. Shenton
- Defence Science and Technology LaboratoryPorton DownUK
| | - R. C. Carlisle
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| |
Collapse
|
25
|
Kozlowski PA, Aldovini A. Mucosal Vaccine Approaches for Prevention of HIV and SIV Transmission. CURRENT IMMUNOLOGY REVIEWS 2019; 15:102-122. [PMID: 31452652 PMCID: PMC6709706 DOI: 10.2174/1573395514666180605092054] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/19/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
Optimal protective immunity to HIV will likely require that plasma cells, memory B cells and memory T cells be stationed in mucosal tissues at portals of viral entry. Mucosal vaccine administration is more effective than parenteral vaccine delivery for this purpose. The challenge has been to achieve efficient vaccine uptake at mucosal surfaces, and to identify safe and effective adjuvants, especially for mucosally administered HIV envelope protein immunogens. Here, we discuss strategies used to deliver potential HIV vaccine candidates in the intestine, respiratory tract, and male and female genital tract of humans and nonhuman primates. We also review mucosal adjuvants, including Toll-like receptor agonists, which may adjuvant both mucosal humoral and cellular immune responses to HIV protein immunogens.
Collapse
Affiliation(s)
- Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Anna Aldovini
- Department of Medicine, and Harvard Medical School, Boston Children’s Hospital, Department of Pediatrics, Boston MA, 02115, USA
| |
Collapse
|
26
|
Kim SJ, Seon SH, Luong TT, Ghosh P, Pyo S, Rhee DK. Immunization with attenuated non-transformable pneumococcal pep27 and comD mutant provides serotype-independent protection against pneumococcal infection. Vaccine 2018; 37:90-98. [PMID: 30467061 DOI: 10.1016/j.vaccine.2018.11.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/05/2018] [Accepted: 11/12/2018] [Indexed: 10/27/2022]
Abstract
Streptococcus pneumoniae is a well-known pathogenic bacterium with a high mortality rate. Currently, a 23-valent pneumococcal polysaccharide vaccine (PPV23) and protein-conjugate vaccines (PCVs) are available on the market. However, both of these vaccines have limitations; specifically, PPV23 produces weak antibody responses in children younger than 2 years and PCVs only partially protect against secondary infection. Previously, we showed serotype-nonspecific protection by Δpep27 vaccine, but the reversion of Δpep27 to the wild type serotype during immunization cannot be excluded. To ensure the safety of the Δpep27 vaccine, comD, an important protein that activates competence, was inactivated, and the transformability of the double mutant (Δpep27ΔcomD) was determined. The transformation ability of this double mutant was successfully abolished. Δpep27ΔcomD immunization significantly increased the survival time after heterologous challenge(s), and diminished colonization levels independent of serotype, including a non-typeable strain (NCC1). Moreover, the double mutant was found to be highly safe in both normal and immunocompromised mice. In conclusion, this pneumococcal Δpep27ΔcomD vaccine appears to be a highly feasible and safe vaccine to prevent various types of pneumococcal infections.
Collapse
Affiliation(s)
- Se-Jin Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Seung Han Seon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Truc Thanh Luong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Prachetash Ghosh
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Suhkneung Pyo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
27
|
Lopez E, Shattock RJ, Kent SJ, Chung AW. The Multifaceted Nature of Immunoglobulin A and Its Complex Role in HIV. AIDS Res Hum Retroviruses 2018; 34:727-738. [PMID: 30056749 DOI: 10.1089/aid.2018.0099] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IgA is the most abundant immunoglobulin in mucosal secretions, and understanding the role of IgA in both protection from HIV acquisition and modulation of HIV disease progression is a field of considerable controversy and renewed research interest. Analysis of the RV144 clinical trial associated plasma HIV envelope-specific monomeric IgA from vaccines with reduced vaccine efficacy. The RV144 trial, however, only assessed for plasma IgA, which was not further subclassed, and the role of mucosal IgA was not addressed as mucosal samples were not collected. On the other hand, several studies have detected envelope-specific IgA in mucosal secretions of highly exposed persistently seronegative cohorts, while recent macaque simian-HIV passive immunization studies have suggested a potentially protective role for mucosal IgA. It is well established that total IgA in serum appears to correlate with HIV disease progression. In contrast, a selective deficit of anti-HIV IgA responses in HIV infection is apparent, with a number of recent studies beginning to elucidate the mechanisms behind these dysfunctional IgA responses. In this review, we highlight the dichotomy that exists in the literature as to whether anti-HIV IgA is protective or harmful to the host. Herein, we emphasize the importance of distinguishing between monomeric, multimeric, and isoforms of IgA and review what is known about the complex and diverse interactions of various molecular forms of IgA with HIV in both the systemic circulation and mucosal compartments.
Collapse
Affiliation(s)
- Ester Lopez
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Robin J. Shattock
- Mucosal Infection and Immunity Group, Department of Medicine, Imperial College London, London, United Kingdom
| | - Stephen J. Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
- Infectious Diseases Department, Melbourne Sexual Health Centre, Alfred Health, Central Clinical School, Monash University, Melbourne, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Melbourne, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| |
Collapse
|
28
|
Palkola NV, Pakkanen SH, Heikinheimo O, Kantele JM, Kantele A. Circulating pathogen-specific plasmablasts in female patients with upper genital tract infection. J Reprod Immunol 2018. [PMID: 29525428 DOI: 10.1016/j.jri.2018.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mucosal antibodies constitute the first line of adaptive immune defence against invaders in the female genital tract (FGT), yet the sequence of events leading to their production is surprisingly poorly characterized. We explored the induction of pathogen-specific antibody-secreting cells (ASC) as a response to an acute infection in the upper FGT. We recruited 12 patients undergoing surgery due to an upper FGT infection (7/12 blood culture positive, 5/12 negative) and six healthy controls. Pathogens were sampled during surgery and PBMC collected in the acute phase of the disease (days 7-10). We searched by ELISPOT circulating pathogen-specific ASC and explored their frequency, immunoglobulin isotype distribution, and expressions of homing receptors (α4β7, L-selectin, and CLA). All patients had circulating ASC specific to the infective bacteria; the geometric mean was 434 (95%CI 155-1234) ASC (IgA + IgG + IgM)/106 PBMC. IgA ASC predominated in 7/12, IgG ASC in 3/12, and IgM ASC in 2/12 cases. Of all the pathogen-specific ASC, 60% expressed α4β7, 67% L-selectin, and 9% CLA. This study is the first to show induction of pathogen-specific ASC in the peripheral blood in bacterial infection in the human FGT. Our findings reveal that such FGT-originating pathogen-specific ASC are predominated by IgA ASC and exhibit a homing receptor profile resembling that of ASC in acute urinary tract infection. The data thus suggest a characteristic profile shared by the urogenital tract.
Collapse
Affiliation(s)
- Nina V Palkola
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Sari H Pakkanen
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jussi M Kantele
- Occupational Health and Environmental Medicine, University of Turku, Turku, Finland
| | - Anu Kantele
- Inflammation Center, Clinic of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Unit of Infectious Diseases, Department of Medicine/Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
29
|
Parenterally Administered Norovirus GII.4 Virus-Like Particle Vaccine Formulated with Aluminum Hydroxide or Monophosphoryl Lipid A Adjuvants Induces Systemic but Not Mucosal Immune Responses in Mice. J Immunol Res 2018; 2018:3487095. [PMID: 29682589 PMCID: PMC5851174 DOI: 10.1155/2018/3487095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/16/2018] [Indexed: 01/06/2023] Open
Abstract
Norovirus (NoV) is a main cause of acute gastroenteritis across all ages worldwide. NoV vaccine candidates currently in clinical trials are based on noninfectious highly immunogenic virus-like particles (VLPs) delivered intramuscularly (IM). Since NoV is an enteric pathogen, it is likely that mucosal immunity has a significant role in protection from infection in the intestine. Due to the fact that IM delivery of NoV VLPs does not generate mucosal immunity, we investigated whether NoV genotype GII.4 VLPs coadministered with aluminum hydroxide (Al(OH)3) or monophosphoryl lipid A (MPLA) would induce mucosal antibodies in mice. Systemic as well as mucosal IgG and IgA antibodies in serum and intestinal and nasal secretions were measured. As expected, strong serum IgG, IgG1, and IgG2a antibodies as well as a dose sparing effect were induced by both Al(OH)3 and MPLA, but no mucosal IgA antibodies were detected. In contrast, IN immunization with GII.4 VLPs without an adjuvant induced systemic as well as mucosal IgA antibody response. These results indicate that mucosal delivery of NoV VLPs is needed for induction of mucosal responses.
Collapse
|
30
|
Albritton HL, Kozlowski PA, Lillis RA, McGowin CL, Siren JD, Taylor SN, Ibana JA, Buckner LR, Shen L, Quayle AJ. A novel whole-bacterial enzyme linked-immunosorbant assay to quantify Chlamydia trachomatis specific antibodies reveals distinct differences between systemic and genital compartments. PLoS One 2017; 12:e0183101. [PMID: 28797112 PMCID: PMC5552291 DOI: 10.1371/journal.pone.0183101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/29/2017] [Indexed: 11/19/2022] Open
Abstract
Chlamydia trachomatis (CT) is the leading sexually transmitted bacterial infection. The continued global burden of CT infection strongly predicates the need for a vaccine to supplement current chlamydial control programs. The correlates of protection against CT are currently unknown, but they must be carefully defined to guide vaccine design. The localized nature of chlamydial infection in columnar epithelial cells of the genital tract necessitates investigation of immunity at the site of infection. The purpose of this study was to develop a sensitive whole bacterial enzyme-linked immunosorbent assay (ELISA) to quantify and compare CT-specific IgG and IgA in sera and genital secretions from CT-infected women. To achieve this, elementary bodies (EBs) from two of the most common genital serovars (D and E) were attached to poly-L-lysine-coated microtiter plates with glutaraldehyde. EB attachment and integrity were verified by the presence of outer membrane antigens and the absence of bacterial cytoplasmic antigens. EB-specific IgG and IgA standards were developed by pooling sera with high titers of CT-specific antibodies from infected women. Serum, endocervical and vaginal secretions, and endocervical cytobrush specimens from CT-infected women were used to quantify CT-specific IgG and IgA which were then normalized to total IgG and IgA, respectively. Analyses of paired serum and genital samples revealed significantly higher proportions of EB-specific antibodies in genital secretions compared to sera. Cervical and vaginal secretions and cytobrush specimens had similar proportions of EB-specific antibodies, suggesting any one of these genital sampling techniques could be used to quantify CT-specific antibodies when appropriate normalization methodologies are implemented. Overall, these results illustrate the need to investigate genital tract CT antibody responses, and our assay provides a useful quantitative tool to assess natural immunity in defined clinical groups and CT vaccine trials.
Collapse
Affiliation(s)
- Hannah L. Albritton
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Rebecca A. Lillis
- Department of Medicine, Division of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Chris L. McGowin
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Julia D. Siren
- Department of Medicine, Division of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Stephanie N. Taylor
- Department of Medicine, Division of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Joyce A. Ibana
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
- Institute of Biology, University of the Philippines Diliman, Quezon City, National Capital Region, Philippines
| | - Lyndsey R. Buckner
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Li Shen
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | - Alison J. Quayle
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
- * E-mail:
| |
Collapse
|
31
|
Kim GL, Seon SH, Rhee DK. Pneumonia and Streptococcus pneumoniae vaccine. Arch Pharm Res 2017; 40:885-893. [PMID: 28735461 PMCID: PMC7090487 DOI: 10.1007/s12272-017-0933-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/17/2017] [Indexed: 12/19/2022]
Abstract
Pneumonia is an inflammatory disease of the lung, responsible for high morbidity and mortality worldwide. It is caused by bacteria, viruses, fungi, or other microorganisms. Streptococcus pneumoniae, a gram-positive bacterium with over 90 serotypes, is the most common causative agent. Moreover, comorbid factors including heart failure, renal disease, and pulmonary disease could increase the risk of pneumococcal pneumonia. Since the advent of the pneumococcal vaccine in the 1980s, the incidence of pneumonia has decreased significantly. However, current vaccines confer only limited protection against serotypes included in the vaccine. Thus, to overcome this limitation, new types of pneumococcal vaccines have been sought and under clinical trials. In this review, we discuss pneumonia and summarize the various types of pneumococcal vaccines in progress.
Collapse
Affiliation(s)
- Gyu-Lee Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, South Korea
| | - Seung-Han Seon
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, South Korea
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, South Korea.
| |
Collapse
|
32
|
Hanna GJ, Sridharan V, Margalit DN, La Follette SK, Chau NG, Rabinowits G, Lorch JH, Haddad RI, Tishler RB, Anderson KS, Schoenfeld JD. Salivary and serum HPV antibody levels before and after definitive treatment in patients with oropharyngeal squamous cell carcinoma. Cancer Biomark 2017; 19:129-136. [DOI: 10.3233/cbm-160071] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Glenn J. Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Danielle N. Margalit
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham & Women's Hospital, Boston, MA, USA
| | - Stephanie K. La Follette
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham & Women's Hospital, Boston, MA, USA
| | - Nicole G. Chau
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Jochen H. Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Robert I. Haddad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Roy B. Tishler
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham & Women's Hospital, Boston, MA, USA
| | - Karen S. Anderson
- School of Life Sciences, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Jonathan D. Schoenfeld
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham & Women's Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Park J, Ramanathan R, Pham L, Woodrow KA. Chitosan enhances nanoparticle delivery from the reproductive tract to target draining lymphoid organs. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2015-2025. [PMID: 28435136 DOI: 10.1016/j.nano.2017.04.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/29/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022]
Abstract
To prime adaptive immune responses from the female reproductive tract (FRT), particulate antigens must be transported to draining lymph nodes (dLNs) since there are no local organized lymphoid structures equivalent to those found in the respiratory or gastrointestinal tracts. However, little is known about how to safely and effectively navigate successive barriers to transport such as crossing the epithelium and gaining access to migratory cells and lymphatic drainage that provide entry into dLNs. Here, we demonstrate that intravaginal pre-treatment with chitosan significantly facilitates translocation of nanoparticles (NPs) across the multilayered vaginal epithelium to target dLNs. In addition, chitosan pre-treatment was found to enhance NP associations with immunogenic antigen presenting cells in the vaginal submucosa. These observations indicate that chitosan may have great potential as an adjuvant for both local and systemic protective immunity against viral infections in the FRT.
Collapse
Affiliation(s)
- Jaehyung Park
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Renuka Ramanathan
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Linhchi Pham
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
34
|
McKay PF, Mann JFS, Pattani A, Kett V, Aldon Y, King D, Malcolm RK, Shattock RJ. Intravaginal immunisation using a novel antigen-releasing ring device elicits robust vaccine antigen-specific systemic and mucosal humoral immune responses. J Control Release 2017; 249:74-83. [PMID: 28115243 PMCID: PMC5333785 DOI: 10.1016/j.jconrel.2017.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/23/2016] [Accepted: 01/10/2017] [Indexed: 01/28/2023]
Abstract
The generation of effective levels of antigen-specific immunity at the mucosal sites of pathogen entry is a key goal for vaccinologists. We explored topical vaginal application as an approach to initiate local antigen-specific immunity, enhance previously existing systemic immunity or re-target responses to the mucosae. To deliver a protein vaccine formulation to the vaginal mucosal surface, we used a novel vaginal ring device comprising a silicone elastomer body into which three freeze-dried, rod-shaped, hydroxypropylmethylcellulose inserts were incorporated. Each rod contained recombinant HIV-1 CN54gp140 protein (167μg)±R848 (167μg) adjuvant. The inserts were loaded into cavities within each ring such that only the ends of the inserts were initially exposed. Sheep received a prime-boost vaccination regime comprising intramuscular injection of 100μg CN54gp140+200μg R848 followed by three successive ring applications of one week duration and separated by one month intervals. Other sheep received only the ring devices without intramuscular priming. Serum and vaginal mucosal fluids were sampled every two weeks and analysed by CN54gp140 ELISA and antigen-specific B cells were measured by flow cytometry at necropsy. Vaccine antigen-specific serum antibody responses were detected in both the intramuscularly-primed and vaginal mucosally-primed groups. Those animals that received only vaginal vaccinations had identical IgG but superior IgA responses. Analysis revealed that all animals exhibited mucosal antigen-specific IgG and IgA with the IgA responses 30-fold greater than systemic levels. Importantly, very high numbers of antigen-specific B cells were detected in local genital draining lymph nodes. We have elicited local genital antigen-specific immune responses after topical application of an adjuvanted antigen formulation within a novel vaginal ring vaccine release device. This regimen and delivery method elicited high levels of antigen-specific mucosal IgA and large numbers of local antigen-reactive B cells, both likely essential for effective mucosal protection.
Collapse
Affiliation(s)
- Paul F McKay
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK.
| | - Jamie F S Mann
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK
| | - Aditya Pattani
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Vicky Kett
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Yoann Aldon
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK
| | - Deborah King
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK
| | - R Karl Malcolm
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Robin J Shattock
- Imperial College London, Department of Medicine, Division of Infectious Diseases, Section of Virology, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
35
|
Mucosal Vaccine Development Based on Liposome Technology. J Immunol Res 2016; 2016:5482087. [PMID: 28127567 PMCID: PMC5227169 DOI: 10.1155/2016/5482087] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/27/2016] [Indexed: 12/01/2022] Open
Abstract
Immune protection against infectious diseases is most effective if located at the portal of entry of the pathogen. Hence, there is an increasing demand for vaccine formulations that can induce strong protective immunity following oral, respiratory, or genital tract administration. At present, only few mucosal vaccines are found on the market, but recent technological advancements and a better understanding of the principles that govern priming of mucosal immune responses have contributed to a more optimistic view on the future of mucosal vaccines. Compared to live attenuated vaccines, subcomponent vaccines, most often protein-based, are considered safer, more stable, and less complicated to manufacture, but they require the addition of nontoxic and clinically safe adjuvants to be effective. In addition, another limiting factor is the large antigen dose that usually is required for mucosal vaccines. Therefore, the combination of mucosal adjuvants with the recent progress in nanoparticle technology provides an attractive solution to these problems. In particular, the liposome technology is ideal for combining protein antigen and adjuvant into an effective mucosal vaccine. Here, we describe and discuss recent progress in nanoparticle formulations using various types of liposomes that convey strong promise for the successful development of the next generation of mucosal vaccines.
Collapse
|
36
|
Hajishengallis G, Arce S, Gockel CM, Connell TD, Russell MW. Immunomodulation with Enterotoxins for the Generation of Secretory Immunity or Tolerance: Applications for Oral Infections. J Dent Res 2016; 84:1104-16. [PMID: 16304439 DOI: 10.1177/154405910508401205] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The heat-labile enterotoxins, such as cholera toxin (CT), and the labile toxins types I and II (LT-I and LT-II) of Escherichia coli have been extensively studied for their immunomodulatory properties, which result in the enhancement of immune responses. Despite superficial similarity in structure, in which a toxic A subunit is coupled to a pentameric binding B subunit, different toxins have different immunological properties. Administration of appropriate antigens admixed with or coupled to these toxins by oral, intranasal, or other routes in experimental animals induces mucosal IgA and circulating IgG antibodies that have protective potential against a variety of enteric, respiratory, or genital infections. These include the generation of salivary antibodies that may protect against colonization with mutans streptococci and the development of dental caries. However, exploitation of these adjuvants for human use requires an understanding of their mode of action and the separation of their desirable immunomodulatory properties from their toxicity. Recent findings have revealed that adjuvant action is not critically dependent upon the enzymic activity of the A subunits, and that the isolated B subunits may exert different effects on cells of the immune system than do the intact toxins. Interaction of the toxins with immunocompetent cells is not exclusively dependent upon their conventional ganglioside receptors. Immunomodulatory effects have been observed on dendritic cells, macrophages, CD4+ and CD8+ T-cells, and B-cells. Numerous factors—including the precise form of the toxin adjuvant, properties of the antigen, whether and how they are coupled, route of administration, and species of animal model—affect the outcome, whether this is enhanced humoral and cellular immunity, or specific induced tolerance toward the antigen.
Collapse
Affiliation(s)
- G Hajishengallis
- Department of Microbiology, Immunology, and Parasitology, and Center of Excellence in Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | | | | | | | | |
Collapse
|
37
|
Kim GL, Choi SY, Seon SH, Lee S, Park SS, Song JY, Briles DE, Rhee DK. Pneumococcal pep27 mutant immunization stimulates cytokine secretion and confers long-term immunity with a wide range of protection, including against non-typeable strains. Vaccine 2016; 34:6481-6492. [DOI: 10.1016/j.vaccine.2016.10.071] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 10/30/2016] [Accepted: 10/31/2016] [Indexed: 12/18/2022]
|
38
|
Nakayama M, Ozaki H, Itoh Y, Soda K, Ishigaki H, Okamatsu M, Sakoda Y, Park CH, Tsuchiya H, Kida H, Ogasawara K. Vaccination against H9N2 avian influenza virus reduces bronchus-associated lymphoid tissue formation in cynomolgus macaques after intranasal virus challenge infection. Pathol Int 2016; 66:678-686. [DOI: 10.1111/pin.12472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/09/2016] [Accepted: 10/07/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Misako Nakayama
- Department of Pathology; Shiga University of Medical Science; Otsu Shiga 520-2192 Japan
| | - Hiroichi Ozaki
- Joint Department of Veterinary Medicine, Faculty of Agriculture; Tottori University; Tottori 680-8553 Japan
| | - Yasushi Itoh
- Department of Pathology; Shiga University of Medical Science; Otsu Shiga 520-2192 Japan
| | - Kosuke Soda
- Joint Department of Veterinary Medicine, Faculty of Agriculture; Tottori University; Tottori 680-8553 Japan
| | - Hirohito Ishigaki
- Department of Pathology; Shiga University of Medical Science; Otsu Shiga 520-2192 Japan
| | - Masatoshi Okamatsu
- Laboratory of Microbiology, Graduate School of Veterinary Medicine; Hokkaido University; Sapporo 060-0818 Japan
| | - Yoshihiro Sakoda
- Laboratory of Microbiology, Graduate School of Veterinary Medicine; Hokkaido University; Sapporo 060-0818 Japan
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE); Hokkaido University; Sapporo Hokkaido 060-0815 Japan
| | - Chun-Ho Park
- School of Veterinary Medicine; Kitasato University; Towada Aomori 034-8628 Japan
| | - Hideaki Tsuchiya
- Research Center for Animal Life Science; Shiga University of Medical Science; Otsu Shiga 520-2192 Japan
| | - Hiroshi Kida
- Laboratory of Microbiology, Graduate School of Veterinary Medicine; Hokkaido University; Sapporo 060-0818 Japan
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE); Hokkaido University; Sapporo Hokkaido 060-0815 Japan
- Research Center for Zoonosis Control; Hokkaido University; Sapporo Hokkaido 001-0020 Japan
| | - Kazumasa Ogasawara
- Department of Pathology; Shiga University of Medical Science; Otsu Shiga 520-2192 Japan
- Research Center for Animal Life Science; Shiga University of Medical Science; Otsu Shiga 520-2192 Japan
| |
Collapse
|
39
|
Cosgrove CA, Lacey CJ, Cope AV, Bartolf A, Morris G, Yan C, Baden S, Cole T, Carter D, Brodnicki E, Shen X, Joseph S, DeRosa SC, Peng L, Yu X, Ferrari G, Seaman M, Montefiori DC, Frahm N, Tomaras GD, Stöhr W, McCormack S, Shattock RJ. Comparative Immunogenicity of HIV-1 gp140 Vaccine Delivered by Parenteral, and Mucosal Routes in Female Volunteers; MUCOVAC2, A Randomized Two Centre Study. PLoS One 2016; 11:e0152038. [PMID: 27159166 PMCID: PMC4861263 DOI: 10.1371/journal.pone.0152038] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/07/2016] [Indexed: 12/16/2022] Open
Abstract
Background Defining optimal routes for induction of mucosal immunity represents an important research priority for the HIV-1 vaccine field. In particular, it remains unclear whether mucosal routes of immunization can improve mucosal immune responses. Methods In this randomized two center phase I clinical trial we evaluated the systemic and mucosal immune response to a candidate HIV-1 Clade C CN54gp140 envelope glycoprotein vaccine administered by intramuscular (IM), intranasal (IN) and intravaginal (IVAG) routes of administration in HIV negative female volunteers. IM immunizations were co-administered with Glucopyranosyl Lipid Adjuvant (GLA), IN immunizations with 0.5% chitosan and IVAG immunizations were administered in an aqueous gel. Results Three IM immunizations of CN54 gp140 at either 20 or 100 μg elicited significantly greater systemic and mucosal antibodies than either IN or IVAG immunizations. Following additional intramuscular boosting we observed an anamnestic antibody response in nasally primed subjects. Modest neutralizing responses were detected against closely matched tier 1 clade C virus in the IM groups. Interestingly, the strongest CD4 T-cell responses were detected after IN and not IM immunization. Conclusions These data show that parenteral immunization elicits systemic and mucosal antibodies in women. Interestingly IN immunization was an effective prime for IM boost, while IVAG administration had no detectable impact on systemic or mucosal responses despite IM priming. Clinical Trials Registration EudraCT 2010-019103-27 and the UK Clinical Research Network (UKCRN) Number 11679
Collapse
Affiliation(s)
| | - Charles J. Lacey
- Hull York Medical School & Centre for Immunology and Infection, University of York, York, United Kingdom
| | - Alethea V. Cope
- Mucosal Infection & Immunity Group, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Angela Bartolf
- Centre for Infection, St George’s, University of London, London, United Kingdom
| | - Georgina Morris
- Hull York Medical School & Centre for Immunology and Infection, University of York, York, United Kingdom
| | - Celine Yan
- Mucosal Infection & Immunity Group, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Susan Baden
- Centre for Infection, St George’s, University of London, London, United Kingdom
| | - Tom Cole
- Mucosal Infection & Immunity Group, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Darrick Carter
- Infectious Disease Research Institute (IDRI), Seattle, WA, United States of America
| | - Elizabeth Brodnicki
- Medical Research Council, Clinical Trials Unit at UCL, University College London, London, United Kingdom
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States of America
| | - Sarah Joseph
- Medical Research Council, Clinical Trials Unit at UCL, University College London, London, United Kingdom
| | - Stephen C. DeRosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lili Peng
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Xuesong Yu
- Statistical Center for HIV/AIDS Research and Prevention, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States of America
- Department of Surgery, Duke University Medical Center, Durham, NC, United States of America
| | - Mike Seaman
- CAVD Neutralizing Antibody Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - David C. Montefiori
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States of America
- Department of Surgery, Duke University Medical Center, Durham, NC, United States of America
| | - Nicole Frahm
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States of America
- Department of Surgery, Duke University Medical Center, Durham, NC, United States of America
| | - Wolfgang Stöhr
- Medical Research Council, Clinical Trials Unit at UCL, University College London, London, United Kingdom
| | - Sheena McCormack
- Medical Research Council, Clinical Trials Unit at UCL, University College London, London, United Kingdom
| | - Robin J. Shattock
- Mucosal Infection & Immunity Group, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Combined HIV-1 Envelope Systemic and Mucosal Immunization of Lactating Rhesus Monkeys Induces a Robust Immunoglobulin A Isotype B Cell Response in Breast Milk. J Virol 2016; 90:4951-4965. [PMID: 26937027 DOI: 10.1128/jvi.00335-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/24/2016] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Maternal vaccination to induce anti-HIV immune factors in breast milk is a potential intervention to prevent postnatal HIV-1 mother-to-child transmission (MTCT). We previously demonstrated that immunization of lactating rhesus monkeys with a modified vaccinia Ankara (MVA) prime/intramuscular (i.m.) protein boost regimen induced functional IgG responses in milk, while MVA prime/intranasal (i.n.) boost induced robust milk Env-specific IgA responses. Yet, recent studies have suggested that prevention of postnatal MTCT may require both Env-specific IgA and functional IgG responses in milk. Thus, to investigate whether both responses could be elicited by a combined systemic/mucosal immunization strategy, animals previously immunized with the MVA prime/i.n. boost regimen received an i.n./i.m. combined C.1086 gp120 boost. Remarkably, high-magnitude Env-specific IgA responses were observed in milk, surpassing those in plasma. Furthermore, 29% of vaccine-elicited Env-specific B cells isolated from breast milk were IgA isotype, in stark contrast to the overwhelming predominance of IgG isotype Env-specific B cells in breast milk of chronically HIV-infected women. A clonal relationship was identified between Env-specific blood and breast milk B cells, suggesting trafficking of that cell population between the two compartments. Furthermore, IgA and IgG monoclonal antibodies isolated from Env-specific breast milk B cells demonstrated diverse Env epitope specificities and multiple effector functions, including tier 1 neutralization, antibody-dependent cellular cytotoxicity (ADCC), infected cell binding, and inhibition of viral attachment to epithelial cells. Thus, maternal i.n./i.m. combined immunization is a novel strategy to enhance protective Env-specific IgA in milk, which is subsequently transferred to the infant via breastfeeding. IMPORTANCE Efforts to increase the availability of antiretroviral therapy to pregnant and breastfeeding women in resource-limited areas have proven remarkably successful at reducing HIV vertical transmission rates. However, more than 200,000 children are infected annually due to failures in therapy implementation, monitoring, and adherence, nearly half by postnatal HIV exposure via maternal breast milk. Intriguingly, in the absence of antiretroviral therapy, only 10% of breastfed infants born to HIV-infected mothers acquire the virus, suggesting the existence of naturally protective immune factors in milk. Enhancement of these protective immune factors through maternal vaccination will be a critical strategy to reduce the global pediatric AIDS epidemic. We have previously demonstrated that a high magnitude of HIV Env-specific IgA in milk correlates with reduced risk of infant HIV acquisition. In this study, we describe a novel HIV vaccine regimen that induces potent IgA responses in milk and therefore could potentially protect against breast milk HIV MTCT.
Collapse
|
41
|
Engelmann F, Rivera A, Park B, Messerle-Forbes M, Jensen JT, Messaoudi I. Impact of Estrogen Therapy on Lymphocyte Homeostasis and the Response to Seasonal Influenza Vaccine in Post-Menopausal Women. PLoS One 2016; 11:e0149045. [PMID: 26859566 PMCID: PMC4747494 DOI: 10.1371/journal.pone.0149045] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/25/2016] [Indexed: 01/01/2023] Open
Abstract
It is widely recognized that changes in levels of ovarian steroids modulate severity of autoimmune disease and immune function in young adult women. These observations suggest that the loss of ovarian steroids associated with menopause could affect the age-related decline in immune function, known as immune senescence. Therefore, in this study, we determined the impact of menopause and estrogen therapy (ET) on lymphocyte subset frequency as well as the immune response to seasonal influenza vaccine in three different groups: 1) young adult women (regular menstrual cycles, not on hormonal contraception); 2) post-menopausal (at least 2 years) women who are not receiving any form of hormone therapy (HT) and 3) post-menopausal hysterectomized women receiving ET. Although the numbers of circulating CD4 and CD20 B cells were reduced in the post-menopausal group receiving ET, we also detected a better preservation of naïve B cells, decreased CD4 T cell inflammatory cytokine production, and slightly lower circulating levels of the pro-inflammatory cytokine IL-6. Following vaccination, young adult women generated more robust antibody and T cell responses than both post-menopausal groups. Despite similar vaccine responses between the two post-menopausal groups, we observed a direct correlation between plasma 17β estradiol (E2) levels and fold increase in IgG titers within the ET group. These findings suggest that ET affects immune homeostasis and that higher plasma E2 levels may enhance humoral responses in post-menopausal women.
Collapse
Affiliation(s)
- Flora Engelmann
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
| | - Andrea Rivera
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
| | - Byung Park
- Division of Biostatistics, Department of Public Health and Preventive Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Marci Messerle-Forbes
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Jeffrey T. Jensen
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon, United States of America
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Ilhem Messaoudi
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- * E-mail:
| |
Collapse
|
42
|
Ramanathan R, Woodrow K. Engineering immunity in the mucosal niche against sexually transmitted infections. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:107-22. [PMID: 26153141 PMCID: PMC6467227 DOI: 10.1002/wnan.1359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 05/04/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022]
Abstract
The mucosal surfaces of the genital tract are the site of entry to over 30 different bacterial, parasitic, and viral pathogens that are the cause of sexually transmitted infections (STIs) including HIV. Women and adolescent girls are more severely impacted by STIs than men due in part to a greater biological susceptibility for acquiring infections and differences in disease sequelae. While it is widely accepted that preventative vaccines against the most commonly transmitted STIs would have a major impact on decreasing the global health burden of STIs for women worldwide, several challenges preclude their development. The female genital tract is a complex niche of microflora, hormonal influences, and immune tissues and cells that result in a mucosal immune system that is distinct from other mucosal sites and from our systemic immune system. An appreciation of these differences and their effect on shaping mucosal immunity to sexually transmitted pathogens is an important determinant for the design of effective STI vaccines. Here we describe the anatomy and mucosal immune system of the female reproductive tract, and discuss bioengineering strategies to design mucosal vaccines that overcome delivery challenges and coordinate the presentation kinetics and compartmentalization of antigens and adjuvants to relevant mucosal immune cell subsets. In particular, we describe recent progress in understanding the role of specific mucosal dendritic cell subsets in facilitating immune responses to pathogenic microbes in the genital mucosa. We also discuss the development of pathogen-mimicking materials that may be useful for engineering protective immunity in this mucosal niche.
Collapse
Affiliation(s)
- Renuka Ramanathan
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Kim Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
43
|
Lin CY, Lin SJ, Yang YC, Wang DY, Cheng HF, Yeh MK. Biodegradable polymeric microsphere-based vaccines and their applications in infectious diseases. Hum Vaccin Immunother 2015; 11:650-6. [PMID: 25839217 PMCID: PMC4514183 DOI: 10.1080/21645515.2015.1009345] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Vaccination, which provides effective, safe infectious disease protection, is among the most important recent public health and immunological achievements. However, infectious disease remains the leading cause of death in developing countries because several vaccines require repeated administrations and children are often incompletely immunized. Microsphere-based systems, providing controlled release delivery, can obviate the need for repeat immunizations. Here, we review the function of sustained and pulsatile release of biodegradable polymeric microspheres in parenteral and mucosal single-dose vaccine administration. We also review the active-targeting function of polymeric particles. With their shield and co-delivery functions, polymeric particles are applied to develop single-dose and mucosally administered vaccines as well as to improve subunit vaccines. Because polymeric particles are easily surface-modified, they have been recently used in vaccine development for cancers and many infectious diseases without effective vaccines (e.g., human immunodeficiency virus infection). These polymeric particle functions yield important vaccine carriers and multiple benefits.
Collapse
Key Words
- APC,antigen-presenting cell
- DC, dendritic cell
- DEN-1–DEN-4, dengue virus serotypes 1–4
- DT or TD, diphtheria + tetanus vaccine
- DT, diphtheria toxoid
- DTP, diphtheria + tetanus + pertussis vaccine
- NS1, nonstructural protein 1
- PEG, poly (ethylene glycol)
- PLA, poly (lactide)
- PLGA, Poly (lactic-co-glycolic acid)
- TT, tetanus-toxoid
- VC, Vibrio cholera
- WHO, World Health Organization
- biodegradable
- immunization
- infectious diseases
- polymeric microspheres
- vaccines
Collapse
Affiliation(s)
- Chi-Ying Lin
- a Food and Drug Administration ; Ministry of Health and Welfare ; Taiwan (R.O.C.)
| | | | | | | | | | | |
Collapse
|
44
|
Pavot V, Climent N, Rochereau N, Garcia F, Genin C, Tiraby G, Vernejoul F, Perouzel E, Lioux T, Verrier B, Paul S. Directing vaccine immune responses to mucosa by nanosized particulate carriers encapsulating NOD ligands. Biomaterials 2015; 75:327-339. [PMID: 26539801 DOI: 10.1016/j.biomaterials.2015.10.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/09/2015] [Accepted: 10/14/2015] [Indexed: 12/30/2022]
Abstract
Mucosal surfaces are a major portal of entry for many pathogens that are the cause of infectious diseases. Therefore, effective vaccines that induce a protective immune response at these sites are much needed. However, despite early success with the live attenuated oral polio vaccine over 50 years ago, only a few new mucosal vaccines have been subsequently licensed. Development of new adjuvants, comprising antigen delivery platforms and immunostimulatory molecules, are critical for the successful development of new mucosal vaccines. Among them, biodegradable nanoparticle delivery systems are promising and NOD-like receptors are considered as potential new targets for immunostimulatory molecules. In this work, different NOD1 and NOD2 ligands were encapsulated in polylactic acid (PLA) nanoparticles, coated with HIV-1 gag p24 antigen. We showed that these new formulations are able to induce proliferation of HIV-specific T cells from HIV(+) individuals as well as autophagy. In vivo, these formulations highly enhanced p24-specific systemic and mucosal immune responses in mice not only after mucosal administration but also after immunization via the parenteral route. Our results provide a rational approach for combining nanosized particulate carriers and encapsulated NOD receptor ligands as potent synergistic tools for induction of specific mucosal immunity.
Collapse
Affiliation(s)
- Vincent Pavot
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thrérapeutique, IBCP, Université Lyon 1, CNRS, UMR 5305, Lyon, France
| | - Nuria Climent
- Hospital Clinic-IDIBAPS, HIVACAT, University of Barcelona, 08036 Barcelona, Spain
| | - Nicolas Rochereau
- Groupe Immunité des Muqueuses et Agents Pathogènes - INSERM CIC1408 Vaccinologie, Faculté de Médecine de Saint-Etienne, France
| | - Felipe Garcia
- Hospital Clinic-IDIBAPS, HIVACAT, University of Barcelona, 08036 Barcelona, Spain
| | - Christian Genin
- Groupe Immunité des Muqueuses et Agents Pathogènes - INSERM CIC1408 Vaccinologie, Faculté de Médecine de Saint-Etienne, France
| | | | | | | | | | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thrérapeutique, IBCP, Université Lyon 1, CNRS, UMR 5305, Lyon, France
| | - Stéphane Paul
- Groupe Immunité des Muqueuses et Agents Pathogènes - INSERM CIC1408 Vaccinologie, Faculté de Médecine de Saint-Etienne, France.
| |
Collapse
|
45
|
Lorenz TK, Demas GE, Heiman JR. Interaction of menstrual cycle phase and sexual activity predicts mucosal and systemic humoral immunity in healthy women. Physiol Behav 2015; 152:92-8. [PMID: 26394125 DOI: 10.1016/j.physbeh.2015.09.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/14/2015] [Accepted: 09/18/2015] [Indexed: 12/23/2022]
Abstract
Several studies have documented shifts in humoral immune parameters (e.g., immunoglobulins) across the menstrual cycle in healthy women. It is thought that these shifts may reflect dynamic balancing between reproduction and pathogen defense, as certain aspects of humoral immunity may disrupt conception and may be temporarily downregulated at ovulation. If so, one could expect maximal cycle-related shifts of humoral immunity in individuals invested in reproduction - that is, women who are currently sexually active - and less pronounced shifts in women who are not reproductively active (i.e., abstinent). We investigated the interaction of sexual activity, menstrual cycle phase, and humoral immunity in a sample of 32 healthy premenopausal women (15 sexually active, 17 abstinent). Participants provided saliva samples during their menses, follicular phase, ovulation (as indicated by urine test for LH surge), and luteal phase, from which IgA was assayed. Participants also provided blood samples at menses and ovulation, from which IgG was assayed. Sexually active participants provided records of their frequency of sexual activity as well as condom use. At ovulation, sexually active women had higher IgG than abstinent women (d=0.77), with women reporting regular condom use showing larger effects (d=0.63) than women reporting no condom use (d=0.11). Frequency of sexual activity predicted changes in IgA (Cohen's f(2)=0.25), with women reporting high frequency of sexual activity showing a decrease in IgA at ovulation, while women reporting low frequency or no sexual activity showing an increase in IgA at ovulation. Taken together, these findings support the hypothesis that shifts in humoral immunity across the menstrual cycle are associated with reproductive effort, and could contribute to the mechanisms by which women's physiology navigates tradeoffs between reproduction and immunity.
Collapse
Affiliation(s)
- Tierney K Lorenz
- Center for Integrative Study for Animal Behavior, Indiana University, Bloomington, 409 N Park Ave, Bloomington, IN, United States; The Kinsey Institute, Indiana University, Bloomington, 1165 E 3rd St., Bloomington, IN 47405, United States.
| | - Gregory E Demas
- Center for Integrative Study for Animal Behavior, Indiana University, Bloomington, 409 N Park Ave, Bloomington, IN, United States; Department of Biology, Indiana University, Bloomington, 1001 E 3rd St, Bloomington, IN, United States; Department of Psychological and Brain Sciences, Indiana University, Bloomington, 1101 E 10th St., Bloomington, IN, United States.
| | - Julia R Heiman
- Center for Integrative Study for Animal Behavior, Indiana University, Bloomington, 409 N Park Ave, Bloomington, IN, United States; The Kinsey Institute, Indiana University, Bloomington, 1165 E 3rd St., Bloomington, IN 47405, United States; Department of Psychological and Brain Sciences, Indiana University, Bloomington, 1101 E 10th St., Bloomington, IN, United States.
| |
Collapse
|
46
|
Thiam F, Charpilienne A, Poncet D, Kohli E, Basset C. B subunits of cholera toxin and thermolabile enterotoxin of Escherichia coli have similar adjuvant effect as whole molecules on rotavirus 2/6-VLP specific antibody responses and induce a Th17-like response after intrarectal immunization. Microb Pathog 2015; 89:27-34. [PMID: 26318874 DOI: 10.1016/j.micpath.2015.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/28/2015] [Accepted: 08/24/2015] [Indexed: 01/04/2023]
Abstract
The purpose of this study was to evaluate the adjuvant effect of the B subunits of cholera toxin (CT) and the thermolabile enterotoxin of Escherichia coli (LT) by the intrarectal route of immunization and compare them to the whole molecules CT and LT-R192G, a non toxic mutant of LT, using 2/6-VLP as an antigen, in mice. All molecules induced similar antigen specific antibody titers in serum and feces, whereas different T cell profiles were observed. CTB and LTB, conversely to CT and LT-R192G, did not induce detectable production of IL-2 by antigen specific T cells. Moreover, CTB, conversely to LT-R192G, CT and LTB, did not induce antigen specific CD4+CD25+Foxp3- and Foxp3+ T cells, thus showing different effects between the B subunits themselves. However, all molecules induced an antigen specific Th17 response. In conclusion, B subunits are potent adjuvants on B cell responses by the intrarectal route. Although their impact on T cell responses are different, all molecules induce a 2/6-VLP-specific Th17 T cell response that may play a major role in helping B cell responses and thus in adjuvanticity and protection.
Collapse
Affiliation(s)
- Fatou Thiam
- Centre de Recherche Inserm UMR866 'Lipides, Nutrition, Cancer', Université de Bourgogne Franche-Comté, Dijon, France
| | - Annie Charpilienne
- Virologie Moléculaire et Structurale, UMR CNRS 2472 INRA 1157, Gif/Yvette, France
| | - Didier Poncet
- Virologie Moléculaire et Structurale, UMR CNRS 2472 INRA 1157, Gif/Yvette, France
| | - Evelyne Kohli
- Centre de Recherche Inserm UMR866 'Lipides, Nutrition, Cancer', Université de Bourgogne Franche-Comté, Dijon, France
| | - Christelle Basset
- Centre de Recherche Inserm UMR866 'Lipides, Nutrition, Cancer', Université de Bourgogne Franche-Comté, Dijon, France.
| |
Collapse
|
47
|
Giefing-Kröll C, Berger P, Lepperdinger G, Grubeck-Loebenstein B. How sex and age affect immune responses, susceptibility to infections, and response to vaccination. Aging Cell 2015; 14:309-21. [PMID: 25720438 PMCID: PMC4406660 DOI: 10.1111/acel.12326] [Citation(s) in RCA: 505] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2014] [Indexed: 12/13/2022] Open
Abstract
Do men die young and sick, or do women live long and healthy? By trying to explain the sexual dimorphism in life expectancy, both biological and environmental aspects are presently being addressed. Besides age-related changes, both the immune and the endocrine system exhibit significant sex-specific differences. This review deals with the aging immune system and its interplay with sex steroid hormones. Together, they impact on the etiopathology of many infectious diseases, which are still the major causes of morbidity and mortality in people at old age. Among men, susceptibilities toward many infectious diseases and the corresponding mortality rates are higher. Responses to various types of vaccination are often higher among women thereby also mounting stronger humoral responses. Women appear immune-privileged. The major sex steroid hormones exhibit opposing effects on cells of both the adaptive and the innate immune system: estradiol being mainly enhancing, testosterone by and large suppressive. However, levels of sex hormones change with age. At menopause transition, dropping estradiol potentially enhances immunosenescence effects posing postmenopausal women at additional, yet specific risks. Conclusively during aging, interventions, which distinctively consider the changing level of individual hormones, shall provide potent options in maintaining optimal immune functions.
Collapse
Affiliation(s)
- Carmen Giefing-Kröll
- Institute for Biomedical Aging Research of Innsbruck University; Innsbruck Austria
| | - Peter Berger
- Institute for Biomedical Aging Research of Innsbruck University; Innsbruck Austria
| | - Günter Lepperdinger
- Institute for Biomedical Aging Research of Innsbruck University; Innsbruck Austria
| | | |
Collapse
|
48
|
Rodriguez Garcia M, Patel MV, Shen Z, Fahey JV, Biswas N, Mestecky J, Wira CR. Mucosal Immunity in the Human Female Reproductive Tract. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00108-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
49
|
Gupta PN. Mucosal Vaccine Delivery and M Cell Targeting. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2015. [DOI: 10.1007/978-3-319-11355-5_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
50
|
|