1
|
Feng L, Zhang J, Ma C, Li K, Zhai J, Cai S, Yin J. Application prospect of polysaccharide in the development of vaccine adjuvants. Int J Biol Macromol 2025; 297:139845. [PMID: 39824409 DOI: 10.1016/j.ijbiomac.2025.139845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/26/2024] [Accepted: 01/12/2025] [Indexed: 01/20/2025]
Abstract
Vaccination is an effective strategy for preventing infectious diseases. Subunit vaccines offer more precise targeting and safer protection compared with traditional inactivated virus vaccines. However, due to their poor immunogenicity, subunit vaccines necessitate the use of adjuvants to stimulate the immune system. Adjuvants have long been incorporated into vaccines to enhance the body's immune response, allowing for reduced dosage and lower production costs. Despite the development of numerous vaccine adjuvants, few exhibit the necessary potency and low toxicity for clinical use, often due to limited efficacy or adverse side effects. This underscores the urgent need for novel human vaccine adjuvants that are safe, effective, and cost-efficient. Recent studies have identified certain natural polysaccharides as promising human vaccine adjuvants due to their immunostimulatory properties, low toxicity, and high safety profiles, which enhance both humoral and cellular immunity. These natural polysaccharides are primarily derived from traditional Chinese medicine (TCM) plants, bacteria, and yeast. This review comprehensively analyzes several promising polysaccharide adjuvants, discussing their clinical applications, market potential, and immunoregulatory activities. In summary, the future prospects of polysaccharides provide valuable insights for the application and development of vaccine adjuvants.
Collapse
Affiliation(s)
- Lei Feng
- Department of Pharmacy, the First Hospital of China Medical University, Shenyang 110001, China; School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Jiarui Zhang
- Department of Intensive Care Medicine, the First Hospital of China Medical University, Shenyang 110001, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, the First Hospital of China Medical University, Shenyang 110001, China
| | - Kai Li
- Department of Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Jianxiu Zhai
- Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Shuang Cai
- Department of Pharmacy, the First Hospital of China Medical University, Shenyang 110001, China; School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Jun Yin
- Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
2
|
Production and Evaluation of a Novel Multi-Epitope Bivalent Vaccine Against Echinococcus multilocaularis Metacestode. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10421-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
3
|
Woods N, Niwasabutra K, Acevedo R, Igoli J, Altwaijry N, Tusiimire J, Gray A, Watson D, Ferro V. Natural Vaccine Adjuvants and Immunopotentiators Derived From Plants, Fungi, Marine Organisms, and Insects. IMMUNOPOTENTIATORS IN MODERN VACCINES 2017. [PMCID: PMC7148613 DOI: 10.1016/b978-0-12-804019-5.00011-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Immunopotentiators derived from different natural sources are under investigation with varying success. This chapter gives an overview of developments from plants, fungi, marine organisms, and insects. Plant-derived immune stimulators consist of a diverse range of small molecules or large polysaccharides. Notable examples that have been assessed in both preclinical and clinical trials include saponins, tomatine, and inulin. Similarly, fungi produce a range of potential candidate molecules, with β-glucans showing the most promise. Other complex molecules that have established adjuvant activity include α-galactosylceramide (originally obtained from a marine sponge), chitosan (commonly produced from chitin from shrimps), and peptides (found in bee venom). Some organisms, for example, endophytic fungi and bees, produce immunostimulants using compounds obtained from plants. The main challenges facing this type of research and tools being developed to overcome them are examined.
Collapse
Affiliation(s)
- N. Woods
- University of Strathclyde, Glasgow, Scotland
| | | | | | - J. Igoli
- University of Strathclyde, Glasgow, Scotland,University of Agriculture, Makurdi, Benue State, Nigeria
| | | | | | - A.I. Gray
- University of Strathclyde, Glasgow, Scotland
| | - D.G. Watson
- University of Strathclyde, Glasgow, Scotland
| | - V.A. Ferro
- University of Strathclyde, Glasgow, Scotland
| |
Collapse
|
4
|
Aboul-Ata AAE, Vitti A, Nuzzaci M, El-Attar AK, Piazzolla G, Tortorella C, Harandi AM, Olson O, Wright SA, Piazzolla P. Plant-based vaccines: novel and low-cost possible route for Mediterranean innovative vaccination strategies. Adv Virus Res 2014; 89:1-37. [PMID: 24751193 DOI: 10.1016/b978-0-12-800172-1.00001-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A plant bioreactor has enormous capability as a system that supports many biological activities, that is, production of plant bodies, virus-like particles (VLPs), and vaccines. Foreign gene expression is an efficient mechanism for getting protein vaccines against different human viral and nonviral diseases. Plants make it easy to deal with safe, inexpensive, and provide trouble-free storage. The broad spectrum of safe gene promoters is being used to avoid risk assessments. Engineered virus-based vectors have no side effect. The process can be manipulated as follows: (a) retrieve and select gene encoding, use an antigenic protein from GenBank and/or from a viral-genome sequence, (b) design and construct hybrid-virus vectors (viral vector with a gene of interest) eventually flanked by plant-specific genetic regulatory elements for constitutive expression for obtaining chimeric virus, (c) gene transformation and/or transfection, for transient expression, into a plant-host model, that is, tobacco, to get protocols processed positively, and then moving into edible host plants, (d) confirmation of protein expression by bioassay, PCR-associated tests (RT-PCR), Northern and Western blotting analysis, and serological assay (ELISA), (e) expression for adjuvant recombinant protein seeking better antigenicity, (f) extraction and purification of expressed protein for identification and dosing, (g) antigenicity capability evaluated using parental or oral delivery in animal models (mice and/or rabbit immunization), and (h) growing of construct-treated edible crops in protective green houses. Some successful cases of heterologous gene-expressed protein, as edible vaccine, are being discussed, that is, hepatitis C virus (HCV). R9 mimotope, also named hypervariable region 1 (HVR1), was derived from the HVR1 of HCV. It was used as a potential neutralizing epitope of HCV. The mimotope was expressed using cucumber mosaic virus coat protein (CP), alfalfa mosaic virus CP P3/RNA3, and tobacco mosaic virus (TMV) CP-tobacco mild green mosaic virus (TMGMV) CP as expression vectors into tobacco plants. Expressed recombinant protein has not only been confirmed as a therapeutic but also as a diagnostic tool. Herpes simplex virus 2 (HSV-2), HSV-2 gD, and HSV-2 VP16 subunits were transfected into tobacco plants, using TMV CP-TMGMV CP expression vectors.
Collapse
Affiliation(s)
- Aboul-Ata E Aboul-Ata
- Molecular Biology Laboratory II, Plant Virus and Phytoplasma Research Department, Plant Pathology Research Institute, ARC, Giza, Egypt.
| | - Antonella Vitti
- School of Agricultural, Forestry, Food and Environmental Sciences, University of Basilicata, Potenza, Italy
| | - Maria Nuzzaci
- School of Agricultural, Forestry, Food and Environmental Sciences, University of Basilicata, Potenza, Italy
| | - Ahmad K El-Attar
- Molecular Biology Laboratory II, Plant Virus and Phytoplasma Research Department, Plant Pathology Research Institute, ARC, Giza, Egypt
| | - Giuseppina Piazzolla
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Allergology and Immunology, University of Bari, Bari, Italy
| | - Cosimo Tortorella
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Allergology and Immunology, University of Bari, Bari, Italy
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Olof Olson
- Department of Pure and Applied Biochemistry, Lund University, Lund, Sweden
| | - Sandra A Wright
- Department of Electronics, Mathematics and Natural Sciences, University of Gävle, Gävle, Sweden
| | - Pasquale Piazzolla
- School of Agricultural, Forestry, Food and Environmental Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|
5
|
Delayed but effective induction of mucosal memory immune responses against genital HSV-2 in the absence of secondary lymphoid organs. Mucosal Immunol 2013; 6:56-68. [PMID: 22718264 DOI: 10.1038/mi.2012.48] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To examine whether local immunization in the absence of secondary lymphoid organs (SLOs) could establish effective antiviral memory responses in the female genital tract, we examined immunity in the vaginal tracts of LTα-/- mice, LTα-/- SPL (splenectomized), and control C57BL/6 (WT) mice. All three groups of mice were immunized intravaginally (IVAG) with attenuated thymidine kinase-negative (TK(-)) Herpes simplex virus type 2 (HSV-2) and challenged 4-6 weeks later with wild-type (WT) HSV-2. Both groups of LTα-/- mice exhibited delayed viral clearance and prolonged genital pathology after immunization. Following IVAG WT HSV-2 challenge, LTα-/- and LTα-/- SPL mice had significantly lower levels of HSV-2-specific IgG and IgA in the vaginal secretions. Although the frequency of B and T cells in the vaginal mucosa was comparable or higher in both groups of LTα-/- mice, lower frequency of HSV-2-specific interferon-γ (IFNγ)-producing CD3+ T cells was seen after immunization and after challenge, compared with WT group. Despite this, immunized mice in all three groups showed complete sterile protection against IVAG WT HSV-2 challenge. These results show that even in the absence of SLOs, IVAG immunization generates effector memory immune responses at genital mucosa that can provide antiviral protection against subsequent viral exposures. This will inform new strategies to design mucosal vaccines against sexually transmitted infections.
Collapse
|
6
|
Roth K, Ferreira VH, Kaushic C. HSV-2 vaccine: current state and insights into development of a vaccine that targets genital mucosal protection. Microb Pathog 2012; 58:45-54. [PMID: 23159485 DOI: 10.1016/j.micpath.2012.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 11/05/2012] [Accepted: 11/06/2012] [Indexed: 11/19/2022]
Abstract
HSV-2 is one of the most prevalent sexually transmitted infections that result in significant morbidity and financial burden on health systems around the world. Recurrent and asymptomatic re-activation accompanied by viral shedding is common among sero-positive individuals, leading to relatively high efficiency of transmission. Prophylactic HSV-2 vaccines are the best and cheapest option to address the problems associated with HSV-2 infections globally. However, despite persistent efforts, the search for an efficacious vaccine for HSV-2 remains elusive. In this review, the current state of HSV-2 vaccines and the outcome of past human trials are examined. Furthermore, we discuss the evidence and strategies from experimental mouse models that have been successful in inducing protective immunity in the genital tract against HSV-2, following immunization. Future vaccination strategies that focus on induction of robust mucosal immunity in the genital tract may hold the key for a successful vaccine against HSV-2.
Collapse
Affiliation(s)
- Kristy Roth
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote Center for Learning and Discovery, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
7
|
Hou J, Liu Y, Liu Y, Shao Y. The MSHA strain of Pseudomonas aeruginosa activated TLR pathway and enhanced HIV-1 DNA vaccine immunoreactivity. PLoS One 2012; 7:e47724. [PMID: 23077664 PMCID: PMC3471878 DOI: 10.1371/journal.pone.0047724] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/14/2012] [Indexed: 12/27/2022] Open
Abstract
The mannose-sensitive hemagglutination pilus strain of Pseudomonas aeruginosa (PA-MSHA) has been shown to trigger naïve immune responses through the activation of monocytes, macrophages, natural killer cells (NK cells) and antigen presenting cells (APCs). Based on the hypothesis that PA-MSHA activates natural immunity through the Toll-like receptor (TLR) pathway, we scanned several critical TLR pathway molecules in mouse splenocytes using high-throughput real-time QRT-PCR and co-stimulatory molecule in bone marrow-derived dendritic cells (BMDCs) following in vitro stimulation by PA-MSHA. PA-MSHA enabled activation of the TLR pathway mediated by NF-κB and JNK signaling in splenocytes, and the co-stimulatory molecule CD86 was up-regulated in BMDCs. We then assessed the adjuvant effect of PA-MSHA for HIV-1 DNA vaccines. In comparison to DNA inoculation alone, co-inoculation with low dosage of PA-MSHA enhanced specific immunoreactivity against HIV-1 Env in both cellular and humoral responses, and promoted antibody avidity maturation. However, high doses of adjuvant resulted in an immunosuppressive effect; a two- or three-inoculation regimen yielded low antibody responses and the two-inoculation regimen exhibited only a slight cellular immunity response. To our knowledge, this is the first report demonstrating the utility of PA-MSHA as an adjuvant to a DNA vaccine. Further research is needed to investigate the exact mechanisms through which PA-MSHA achieves its adjuvant effects on innate immune responses, especially on dendritic cells.
Collapse
Affiliation(s)
- Jue Hou
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yong Liu
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ying Liu
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yiming Shao
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- * E-mail:
| |
Collapse
|
8
|
Wang SH, Thompson AL, Hickey AJ, Staats HF. Dry powder vaccines for mucosal administration: critical factors in manufacture and delivery. Curr Top Microbiol Immunol 2011; 354:121-56. [PMID: 21822816 DOI: 10.1007/82_2011_167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dry powder vaccine formulations have proved effective for induction of systemic and mucosal immune responses. Here we review the use of dry vaccines for immunization in the respiratory tract. We discuss techniques for powder formulation, manufacture, characterization and delivery in addition to methods used for evaluation of stability and safety. We review the immunogenicity and protective efficacy of dry powder vaccines as compared to liquid vaccines delivered by mucosal or parenteral routes. Included is information on mucosal adjuvants and mucoadhesives that can be used to enhance nasal or pulmonary dry vaccines. Mucosal immunization with dry powder vaccines offers the potential to provide a needle-free and cold chain-independent vaccination strategy for the induction of protective immunity against either systemic or mucosal pathogens.
Collapse
Affiliation(s)
- Sheena H Wang
- Division of Molecular Pharmaceutics, School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
9
|
Kaushic C. The role of the local microenvironment in regulating susceptibility and immune responses to sexually transmitted viruses in the female genital tract. J Reprod Immunol 2009; 83:168-72. [PMID: 19857903 DOI: 10.1016/j.jri.2009.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 07/22/2009] [Accepted: 08/01/2009] [Indexed: 12/28/2022]
Abstract
Sexually transmitted viruses cause chronic infections that have serious long-term health consequences. Based on the evidence from clinical and epidemiological studies, women carry a disproportionately higher burden of sexually transmitted diseases. The reasons for this are not well understood and possibly relate to a variety of social, behavioral and economic factors. In addition to these factors there are biological reasons that contribute to the higher prevalence in women. In this context it is critical to focus on and understand the local microenvironment of the female genital tract, since the majority of viral infections in women occur by heterosexual transmission. The genital tract is also the target site for initiation and maintenance of protective immune responses that could prevent or eliminate viral infections. The epithelial cells of the genital tract provide the first line of defense against viral entry. The interactions between each sexually transmitted virus and the genital epithelium are distinct and determine the outcome of exposure. They are also influenced by a number of factors in the local genital milieu. Among these factors are the female sex hormones that regulate both the susceptibility as well as immune responses to viral infections in the genital tract. Better understanding of the interactions of viruses with the local environment in the female genital tract will lead to development of novel methods to prevent sexually transmitted infections as well as to enhance innate and adaptive immunity.
Collapse
Affiliation(s)
- Charu Kaushic
- Department of Pathology and Molecular Medicine, Center for Gene Therapeutics, Michael G. DeGroote Center for Learning and Discovery Room 4014, McMaster University, Ontario, Canada.
| |
Collapse
|
10
|
Babiuk S, Babiuk LA, van Drunen Littel-van den Hurk S. Editorial: DNA Vaccination: A Simple Concept with Challenges Regarding Implementation. Int Rev Immunol 2009; 25:51-81. [PMID: 16818365 DOI: 10.1080/08830180600743008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
11
|
Dasgupta G, Chentoufi AA, Nesburn AB, Wechsler SL, BenMohamed L. New concepts in herpes simplex virus vaccine development: notes from the battlefield. Expert Rev Vaccines 2009; 8:1023-35. [PMID: 19627185 PMCID: PMC2760836 DOI: 10.1586/erv.09.60] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The recent discovery that T cells recognize different sets of herpes simplex virus type 1 and type 2 epitopes from seropositive symptomatic and asymptomatic individuals might lead to a fundamental immunologic advance in vaccine development against herpes infection and diseases. The newly introduced needle-free mucosal (i.e., topical ocular and intravaginal) lipopeptide vaccines provide a novel strategy that might target ocular and genital herpes and possibly provide 'heterologous protection' from HIV-1. Indeed, mucosal self-adjuvanting lipopeptide vaccines are easy to manufacture, simple to characterize, extremely pure, cost-effective, highly immunogenic and safe. In this review, we bring together recent published and unpublished data that illuminates the status of epitope-based herpes vaccine development and present an overview of our recent approach to an 'asymptomatic epitope'-based lipopeptide vaccine.
Collapse
Affiliation(s)
- Gargi Dasgupta
- The Gavin S Herbert Eye Institute, Cellular and Molecular Immunology Laboratory, Department of Ophthalmology, University of California, Irvine, College of Medicine, Irvine, CA 92697-4375, USA, Tel.: +1 714 456 6465, Fax: +1 714 456 5073,
| | - Aziz A Chentoufi
- The Gavin S Herbert Eye Institute, Cellular and Molecular Immunology Laboratory, Department of Ophthalmology, University of California, Irvine, College of Medicine, Irvine, CA 92697-4375, USA, Tel.: +1 714 456 6465, Fax: +1 714 456 5073,
| | - Anthony B Nesburn
- The Gavin S Herbert Eye Institute, Cellular and Molecular Immunology Laboratory, Department of Ophthalmology, University of California, Irvine, College of Medicine, Irvine, CA 92697-4375, USA, Tel.: +1 714 456 6465, Fax: +1 714 456 5073,
| | - Steven L Wechsler
- The Gavin S Herbert Eye Institute, Cellular and Molecular Immunology Laboratory, Department of Ophthalmology, and Department of Microbiology and Molecular Genetics, University of California, Irvine, College of Medicine, Irvine, CA 92697-4375, USA, Tel.: +1 714 456 6465, Fax: +1 714 456 5073,
| | - Lbachir BenMohamed
- The Gavin S Herbert Eye Institute, Cellular and Molecular Immunology Laboratory, University of California Irvine, College of Medicine, Building 55, Room 202, Orange, CA 92868, USA and Center for Immunology, University of California, Irvine, Irvine, CA 92697-1450, USA, Tel.: +1 714 456 7371, Fax: +1 714 456 5073,
| |
Collapse
|
12
|
McGowen AL, Hale LP, Shelburne CP, Abraham SN, Staats HF. The mast cell activator compound 48/80 is safe and effective when used as an adjuvant for intradermal immunization with Bacillus anthracis protective antigen. Vaccine 2009; 27:3544-52. [PMID: 19464533 PMCID: PMC2743390 DOI: 10.1016/j.vaccine.2009.03.069] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 03/20/2009] [Accepted: 03/24/2009] [Indexed: 11/18/2022]
Abstract
We evaluated the safety and efficacy of the mast cell activator compound 48/80 (C48/80) when used as an adjuvant delivered intradermally (ID) with recombinant anthrax protective antigen (rPA) in comparison with two well-known adjuvants. Mice were vaccinated in the ear pinnae with rPA or rPA+C48/80, CpG oligodeoxynucleotides (CpG), or cholera toxin (CT). All adjuvants induced similar increases in serum anti-rPA IgG and lethal toxin neutralizing antibodies. C48/80 induced a balanced cytokine production (Th1/Th2/Th17) by antigen-restimulated splenocytes, minimal injection site inflammation, and no antigen-specific IgE. Histological analysis demonstrated that vaccination with C48/80 reduced the number of resident mast cells and induced an injection site neutrophil influx within 24h. Our data demonstrate that C48/80 is a safe and effective adjuvant, when used by the intradermal route, to induce protective antibody and balanced Th1/Th2/Th17 responses.
Collapse
Affiliation(s)
- Afton L. McGowen
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Laura P. Hale
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
- The Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | - Soman N. Abraham
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Molecular Genetics & Microbiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Herman F. Staats
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
- The Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
13
|
Campo JD, Zayas C, Romeu B, Acevedo R, González E, Bracho G, Cuello M, Cabrera O, Balboa J, Lastre M. Mucosal immunization using proteoliposome and cochleate structures from Neisseria meningitidis serogroup B induce mucosal and systemic responses. Methods 2009; 49:301-8. [PMID: 19410000 DOI: 10.1016/j.ymeth.2009.03.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 02/27/2009] [Accepted: 03/09/2009] [Indexed: 11/25/2022] Open
Abstract
Most pathogens either invade the body or establish infection in mucosal tissues and represent an enormous challenge for vaccine development by the absence of good mucosal adjuvants. A proteoliposome-derived adjuvant from Neisseria meningitidis serogroup B (AFPL1, Adjuvant Finlay Proteoliposome 1) and its derived cochleate form (Co, AFCo1) contain multiple pathogen-associated molecular patterns as immunopotentiators, and can also serve as delivery systems to elicit a Th1-type immune response. The present studies demonstrate the ability of AFPL1and AFCo1 to induce mucosal and systemic immune responses by different mucosal immunizations routes and significant adjuvant activity for antibody responses of both structures: a microparticle and a nanoparticle with a heterologous antigen. Therefore, we used female mice immunized by intragastric, intravaginal, intranasal or intramuscular routes with both structures alone or incorporated with ovalbumin (OVA). High levels of specific IgG antibody were detected in all sera and in vaginal washes, but specific IgA antibody in external secretions was only detected in mucosally immunized mice. Furthermore, antigen specific IgG1 and IgG2a isotypes were all induced. AFPL1 and AFCo1 are capable of inducing IFN-gamma responses, and chemokine secretions, like MIP-1alpha and MIP-1beta. However, AFCo1 is a better alternative to induce immune responses at mucosal level. Even when we use a heterologous antigen, the AFCo1 response was better than with AFPL1 in inducing mucosal and systemic immune responses. These results support the use of AFCo1 as a potent Th1 inducing adjuvant particularly suitable for mucosal immunization.
Collapse
Affiliation(s)
- Judith Del Campo
- Immunology Department, Finlay Institute, PO Box 16017, Havana, Cuba.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Zhang X, Chentoufi AA, Dasgupta G, Nesburn AB, Wu M, Zhu X, Carpenter D, Wechsler SL, You S, BenMohamed L. A genital tract peptide epitope vaccine targeting TLR-2 efficiently induces local and systemic CD8+ T cells and protects against herpes simplex virus type 2 challenge. Mucosal Immunol 2009; 2:129-143. [PMID: 19129756 PMCID: PMC4509510 DOI: 10.1038/mi.2008.81] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The next generation of needle-free mucosal vaccines is being rationally designed according to rules that govern the way in which the epitopes are recognized by and stimulate the genital mucosal immune system. We hypothesized that synthetic peptide epitopes extended with an agonist of Toll-like receptor 2 (TLR-2), that are abundantly expressed by dendritic and epithelial cells of the vaginal mucosa, would lead to induction of protective immunity against genital herpes. To test this hypothesis, we intravaginally (IVAG) immunized wild-type B6, TLR-2 (TLR2(-/-)) or myeloid differentiation factor 88 deficient (MyD88(-/-)) mice with a herpes simplex virus type 2 (HSV-2) CD8+ T-cell peptide epitope extended by a palmitic acid moiety (a TLR-2 agonist). IVAG delivery of the lipopeptide generated HSV-2-specific memory CD8+ cytotoxic T cells both locally in the genital tract draining lymph nodes and systemically in the spleen. Moreover, lipopeptide-immunized TLR2(-/-) and MyD88(-/-) mice developed significantly less HSV-specific CD8+ T-cell response, earlier death, faster disease progression, and higher vaginal HSV-2 titers compared to lipopeptide-immunized wild-type B6 mice. IVAG immunization with self-adjuvanting lipid-tailed peptides appears to be a novel mucosal vaccine approach, which has attractive practical and immunological features.
Collapse
Affiliation(s)
- X Zhang
- Laboratory of Cellular and Molecular Immunology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA
| | - AA Chentoufi
- Laboratory of Cellular and Molecular Immunology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA
| | - G Dasgupta
- Laboratory of Cellular and Molecular Immunology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA
| | - AB Nesburn
- Laboratory of Cellular and Molecular Immunology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA
| | - M Wu
- Laboratory of Cellular and Molecular Immunology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA
| | - X Zhu
- Laboratory of Cellular and Molecular Immunology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA
| | - D Carpenter
- Laboratory of Cellular and Molecular Immunology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA
| | - SL Wechsler
- Laboratory of Virology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA,Department of Microbiology and Molecular Genetics, University of California Irvine, School of Medicine, Irvine, CA, USA,The Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - S You
- INSERM U580, University Paris Descartes, Paris, France
| | - L BenMohamed
- Laboratory of Cellular and Molecular Immunology, The Gavin S. Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, USA,Center for Immunology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
15
|
Wilson KD, Raney SG, Sekirov L, Chikh G, deJong SD, Cullis PR, Tam YK. Effects of intravenous and subcutaneous administration on the pharmacokinetics, biodistribution, cellular uptake and immunostimulatory activity of CpG ODN encapsulated in liposomal nanoparticles. Int Immunopharmacol 2007; 7:1064-75. [PMID: 17570323 DOI: 10.1016/j.intimp.2007.04.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 04/02/2007] [Accepted: 04/04/2007] [Indexed: 11/23/2022]
Abstract
We have previously demonstrated that the immune response to an unmethylated cytidine-guanosine (CpG)-containing oligonucleotide (ODN) is greatly enhanced when encapsulated in a lipid nanoparticle (LN-CpG ODN). In this study, the pharmacokinetics, biodistribution and cellular uptake of LN-CpG ODN following intravenous (i.v.) and subcutaneous (s.c.) administration was characterized and correlated with immunostimulatory activity. It is shown that, despite dramatic differences in tissue distribution profiles and considerable differences in uptake by CD11c-positive, CD11b-positive, Mac-3-positive and CD45R/B220-positive cells following i.v. and s.c. administration, the resultant immune response is very similar with respect to levels of cellular activation (DX5, Mac-3, CD11b, CD45/B220, CD4, CD8 and CD11c) and cytolytic activity of immune cells [natural killer (NK) cells and monocytes/macrophages] in the spleen and blood compartments. Some differences in response kinetics and antibody-dependent cellular cytotoxicity (ADCC) activity were noted in the peripheral blood NK cell population. Analyses of particle biodistribution and cell types involved in uptake leads to the conclusion that the inherent ability of antigen-presenting cells (APCs) to sequester LN-CpG ODN results in efficient uptake of the particle, even when present at very low concentrations, leading to similar responses following i.v. and s.c. administration. These results contrast with the behavior of free CpG ODN, for which distinctly different immune responses are observed following i.v. or s.c. administration.
Collapse
Affiliation(s)
- Kaley D Wilson
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|