1
|
He G, Long H, He J, Zhu C. The Immunomodulatory Effects and Applications of Probiotic Lactiplantibacillus plantarum in Vaccine Development. Probiotics Antimicrob Proteins 2024; 16:2229-2250. [PMID: 39101975 DOI: 10.1007/s12602-024-10338-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/06/2024]
Abstract
Lactiplantibacillus plantarum (previously known as Lactobacillus plantarum) is a lactic acid bacterium that exists in various niches. L. plantarum is a food-grade microorganism that is commonly considered a safe and beneficial microorganism. It is widely used in food fermentation, agricultural enhancement, and environmental protection. L. plantarum is also part of the normal flora that can regulate the intestinal microflora and promote intestinal health. Some strains of L. plantarum are powerful probiotics that induce and modulate the innate and adaptive immune responses. Due to its outstanding immunoregulatory capacities, an increasing number of studies have examined the use of probiotic L. plantarum strains as natural immune adjuvants or alternative live vaccine carriers. The present review summarizes the main immunomodulatory characteristics of L. plantarum and discusses the preliminary immunological effects of L. plantarum as a vaccine adjuvant and delivery carrier. Different methods for improving the immune capacities of recombinant vector vaccines are also discussed.
Collapse
Affiliation(s)
- Guiting He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Huanbing Long
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Jiarong He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China.
| |
Collapse
|
2
|
Sharma A, Bhatia D. Programmable bionanomaterials for revolutionizing cancer immunotherapy. Biomater Sci 2024; 12:5415-5432. [PMID: 39291418 DOI: 10.1039/d4bm00815d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Cancer immunotherapy involves a cutting-edge method that utilizes the immune system to detect and eliminate cancer cells. It has shown substantial effectiveness in treating different types of cancer. As a result, its growing importance is due to its distinct benefits and potential for sustained recovery. However, the general deployment of this treatment is hindered by ongoing issues in maintaining minimal toxicity, high specificity, and prolonged effectiveness. Nanotechnology offers promising solutions to these challenges due to its notable attributes, including expansive precise surface areas, accurate ability to deliver drugs and controlled surface chemistry. This review explores the current advancements in the application of nanomaterials in cancer immunotherapy, focusing on three primary areas: monoclonal antibodies, therapeutic cancer vaccines, and adoptive cell treatment. In adoptive cell therapy, nanomaterials enhance the expansion and targeting capabilities of immune cells, such as T cells, thereby improving their ability to locate and destroy cancer cells. For therapeutic cancer vaccines, nanoparticles serve as delivery vehicles that protect antigens from degradation and enhance their uptake by antigen-presenting cells, boosting the immune response against cancer. Monoclonal antibodies benefit from nanotechnology through improved delivery mechanisms and reduced off-target effects, which increase their specificity and effectiveness. By highlighting the intersection of nanotechnology and immunotherapy, we aim to underscore the transformative potential of nanomaterials in enhancing the effectiveness and safety of cancer immunotherapies. Nanoparticles' ability to deliver drugs and biomolecules precisely to tumor sites reduces systemic toxicity and enhances therapeutic outcomes.
Collapse
Affiliation(s)
- Ayushi Sharma
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura, Uttar Pradesh-281406, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj 382355, Gandhinagar, India
| |
Collapse
|
3
|
Immunogenicity Associated with Aesthetic Botulinumtoxin A: A Survey of Asia-Pacific Physicians' Experiences and Recommendations. Plast Reconstr Surg Glob Open 2022; 10:e4217. [PMID: 35450268 PMCID: PMC9015201 DOI: 10.1097/gox.0000000000004217] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/31/2022] [Indexed: 12/27/2022]
Abstract
Background: Most botulinum toxin A (BoNT/A) products contain unnecessary bacterial components that increase the risk of developing neutralizing antibodies (nAbs). Reports of secondary nonresponse and treatment failures (STF) due to nAbs have accompanied a surge in new BoNT/A products. Methods: To formulate recommendations on managing toxin resistance, we reviewed the evidence on BoNT/A-associated immunogenicity and evaluated Asian physicians' current BoNT/A practices, knowledge, and real-world experiences, as provided by survey outcomes conducted with 128 Asian experts (regular botulinum toxin injectors). Results: Most doctors believe STF occurs, some patients exhibit partial symptoms, and impurities (eg, complexing proteins) in BoNT/A preparations risk STF. Bioassays that distinguish non-nAbs from nAbs that hinder toxin function remain unavailable to most doctors, though most would perform testing if given the option. Doctors in the Asia-Pacific region have differing strategies for managing STF, depending on the availability of alternatives or tests. They recommended switching to a highly-purified formulation free of complexing proteins and other impurities to lower the risk of immunogenicity, or offering treatment holidays of 2 -2.5 years. They suggested restarting treatment with the same highly purified formulation, especially for repeated treatments, large-dose injections, and younger patients who will accumulate higher lifetime doses, so as to minimize immunogenic risks and preserve long-term treatment outcomes. Importantly, doctors should always initiate patients on pure formulations rather than switching to these only after resistance develops. Conclusion: Choosing highly purified BoNT/A products at treatment initiation enhances long-term efficacy and patient satisfaction while minimizing the risk of immune activation and nAb formation.
Collapse
|
4
|
Mechanistic Insights from the Review and Evaluation of Ayurvedic Herbal Medicines for the Prevention and Management of COVID-19 Patients. J Herb Med 2022; 32:100554. [PMID: 35251909 PMCID: PMC8885324 DOI: 10.1016/j.hermed.2022.100554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 12/31/2022]
Abstract
Introduction The need for specific therapeutics against infectious diseases is made very important at this moment by the COVID-19 pandemic caused by SARS-COV-2. Vaccines containing live attenuated or heat-inactivated pathogens elicit robust immune responses, but their safety is sometimes not assured. Subunit vaccines consisting of the most potent antigenic protein or carbohydrates of the pathogen are safer but often induce a weak immune response. Traditional Ayurveda medicines have a long history of safety and may act as immuno-modulators or vaccine adjuvants. They can reduce the amount of vaccine booster doses required to elicit an immune response against any pathogen. The main objective of this review is a mechanistic evaluation of the antiviral potential of Ayurveda herbal compositions for their ability to increase cytokine expression and enhance NK cell activity, activate CD4/ CD8 + T cells, and increase the formation of IL-2 and IFNγ against SARS-CoV-2 infection. Methods Various peer-reviewed publications, books, monographs, and reputed search engines were reviewed in depth. Information available from the Ayurvedic Pharmacopoeia and in recent in silico analyses were compared in order to understand the mechanism of action of herbal components against SARS-CoV-2. Results It was found in various molecular docking and molecular dynamics studies that many bioactive natural components of Ayurvedic medicines could prevent viral entry or multiplication within a human host. Conclusion Ayurvedic herbal medicines can be used either independently as therapeutics or as a complement to the modern-day recombinant vaccines with immediate effect. Ayurveda-based adjuvant therapy can also efficiently manage the secondary symptoms of COVID 19 patients.
Collapse
|
5
|
Mekonnen D, Mengist HM, Jin T. SARS-CoV-2 subunit vaccine adjuvants and their signaling pathways. Expert Rev Vaccines 2022; 21:69-81. [PMID: 34633259 PMCID: PMC8567292 DOI: 10.1080/14760584.2021.1991794] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/07/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Vaccines are the agreed upon weapon against the COVID-19 pandemic. This review discusses about COVID-19 subunit vaccines adjuvants and their signaling pathways, which could provide a glimpse into the selection of appropriate adjuvants for prospective vaccine development studies. AREAS COVERED In the introduction, a brief background about the SARS-CoV-2 pandemic, the vaccine development race and classes of vaccine adjuvants were provided. . The antigen, trial stage, and types of adjuvants were extracted from the included articles and thun assimilated. Finally, the pattern recognition receptors (PRRs), their classes, cognate adjuvants, and potential signaling pathways were comprehended. EXPERT OPINION Adjuvants are unsung heroes of subunit vaccines. The in silico studies are very vital in avoiding several costly trial errors and save much work times. The majority of the (pre)clinical studies are promising. It is encouraging that most of the selected adjuvants are novel. Much emphasis must be paid to the optimal paring of antigen-adjuvant-PRRs for obtaining the desired vaccine effect. A good subunit vaccine/adjuvant is one that has high efficacy, safety, dose sparing, and rapid seroconversion rate and broad spectrum of immune response. In the years to come, COVID-19 adjuvanted subunit vaccines are expected to have superior utility than any other vaccines for various reasons.
Collapse
Affiliation(s)
- Daniel Mekonnen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hylemariam Mihiretie Mengist
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, Cas Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, Cas Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
6
|
Mohammadian Haftcheshmeh S, Zamani P, Mashreghi M, Nikpoor AR, Tavakkol-Afshari J, Jaafari MR. Immunoliposomes bearing lymphocyte activation gene 3 fusion protein and P5 peptide: A novel vaccine for breast cancer. Biotechnol Prog 2020; 37:e3095. [PMID: 33118322 DOI: 10.1002/btpr.3095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/14/2020] [Accepted: 10/26/2020] [Indexed: 01/31/2023]
Abstract
LAG3-Ig as an immune adjuvant has elicited potent anti-tumor immune responses in several preclinical and clinical studies, but the full potential immunostimulatory of LAG3-Ig has yet to be achieved. We hypothesized that by anchoring LAG3-Ig to the surface of liposomes, the adjuvant activity of LAG3-Ig could be improved. We also investigated the immunotherapy by co-delivery of liposome-coupled LAG3-Ig and P5 tumor antigen in mice model of TUBO breast cancer. We prepared and characterized novel PEGylated liposomes bearing surface conjugated LAG3-Ig and P5. Consistent with our hypothesis, liposomes-conjugated LAG3-Ig via multivalent binding to MHC class II molecules exerted immunostimulatory of LAG3-Ig and markedly induced maturation of dendritic cells more efficiently than free LAG3-Ig. LAG3-Ig-P5-immunoliposomes effectively elicited protective anti-tumor responses more than locally injected soluble LAG3-Ig + P5. The higher percentage of CD4+ and CD8+ T cells in the spleen and more rapid and pronounced infiltration of these effector cells into the site of the tumor were seen following immunoliposome therapy. Finally, anti-tumor immunity induced by LAG3-Ig-P5-immunoliposomes translated into the more tumor regression and prolonged survival of treated mice, compared to soluble immunotherapy. Taken together, our findings suggest that LAG3-Ig-P5-immunoliposomes can be considered as a valuable candidate for developing a liposome-based therapeutic cancer vaccine in treating HER2/ neu+ breast cancer patients.
Collapse
Affiliation(s)
- Saeed Mohammadian Haftcheshmeh
- Department of Medical Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Reza Nikpoor
- Department of Immunology, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Sefidi-Heris Y, Jahangiri A, Mokhtarzadeh A, Shahbazi MA, Khalili S, Baradaran B, Mosafer J, Baghbanzadeh A, Hejazi M, Hashemzaei M, Hamblin MR, Santos HA. Recent progress in the design of DNA vaccines against tuberculosis. Drug Discov Today 2020; 25:S1359-6446(20)30345-7. [PMID: 32927065 DOI: 10.1016/j.drudis.2020.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022]
Abstract
Current tuberculosis (TB) vaccines have some disadvantages and many efforts have been undertaken to produce effective TB vaccines. As a result of their advantages, DNA vaccines are promising future vaccine candidates. This review focuses on the design and delivery of novel DNA-based vaccines against TB.
Collapse
Affiliation(s)
- Youssof Sefidi-Heris
- Department of Biology, College of Sciences, Shiraz University, 7146713565, Shiraz, Iran
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, 193955487, Tehran, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, 5166614731, Tabriz, Iran.
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran.
| | - Saeed Khalili
- Department of Biology Sciences, Faculty of Sciences, Shahid Rajaee Teacher Training University, 1678815811, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, 5166614731, Tabriz, Iran
| | - Jafar Mosafer
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, 9516915169, Torbat Heydariyeh, Iran; Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, 9196773117, Mashhad, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, 5166614731, Tabriz, Iran
| | - Maryam Hejazi
- Immunology Research Center, Tabriz University of Medical Sciences, 5166614731, Tabriz, Iran
| | - Mahmoud Hashemzaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, 9861615881, Zabol, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki FI-00014, Finland.
| |
Collapse
|
8
|
MPL nano-liposomal vaccine containing P5 HER2/neu-derived peptide pulsed PADRE as an effective vaccine in a mice TUBO model of breast cancer. J Control Release 2019; 303:223-236. [DOI: 10.1016/j.jconrel.2019.04.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/11/2019] [Accepted: 04/13/2019] [Indexed: 11/21/2022]
|
9
|
Non-human papillomaviruses for gene delivery in vitro and in vivo. PLoS One 2018; 13:e0198996. [PMID: 29912929 PMCID: PMC6005490 DOI: 10.1371/journal.pone.0198996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/18/2018] [Indexed: 12/17/2022] Open
Abstract
Papillomavirus capsids are known to have the ability to package DNA plasmids and deliver them both in vitro and in vivo. Of all known papillomavirus types, human papillomaviruses (HPVs) are by far the most intensely studied. Although HPVs work well as gene transfer vectors, their use is limited as most individuals are exposed to this virus either through a HPV vaccination or natural infection. To circumvent these constraints, we produced pseudovirions (PsVs) of ten non-human papillomavirus types and tested their transduction efficiencies in vitro. PsVs based on Macaca fascicularis papillomavirus-11 and Puma concolor papillomavirus-1 were further tested in vivo. Intramuscular transduction by PsVs led to months-long expression of a reporter plasmid, indicating that PsVs have potential as gene delivery vectors.
Collapse
|
10
|
Zamani P, Momtazi‐Borojeni AA, Nik ME, Oskuee RK, Sahebkar A. Nanoliposomes as the adjuvant delivery systems in cancer immunotherapy. J Cell Physiol 2018; 233:5189-5199. [DOI: 10.1002/jcp.26361] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 11/29/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Parvin Zamani
- BuAli Research InstituteStudent Research CommitteeDepartment of Medical BiotechnologyNanotechnology Research CenterSchool of MedicineMashhad University of Medical SciencesMashhadIran
| | - Amir Abbas Momtazi‐Borojeni
- BuAli Research InstituteStudent Research CommitteeDepartment of Medical BiotechnologyNanotechnology Research CenterSchool of MedicineMashhad University of Medical SciencesMashhadIran
| | - Maryam Ebrahimi Nik
- Student Research CommitteeFaculty of PharmacyDepartment of NanotechnologyMashhad University of Medical SciencesMashhadIran
| | - Reza Kazemi Oskuee
- Targeted Drug Delivery Research CenterMashhad University of Medical SciencesMashhadIran
| | - Amirhossein Sahebkar
- Biotechnology Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
11
|
Reddy Bonam S, Naidu Gorantla J, Thangarasu AK, Lankalapalli RS, Sampath Kumar HM. Polyhydroxy-N-alkyl-2-pyrrolidinones as a new class of glycolipid analogues with immune modulation potential. J Carbohydr Chem 2018. [DOI: 10.1080/07328303.2017.1413193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Srinivasa Reddy Bonam
- Vaccine Immunology Laboratory, Natural Products Chemistry Division, Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- CNRS UPR 3572, Laboratory of Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, University of Strasbourg, Strasbourg, France
| | - Jaggaiah Naidu Gorantla
- Organic Chemistry Section, Chemical Sciences and Technology Division, Academy of Scientific and Innovative Research (AcSIR), CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, Kerala, India
| | - Arun Kumar Thangarasu
- Organic Chemistry Section, Chemical Sciences and Technology Division, Academy of Scientific and Innovative Research (AcSIR), CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, Kerala, India
| | - Ravi Shankar Lankalapalli
- Organic Chemistry Section, Chemical Sciences and Technology Division, Academy of Scientific and Innovative Research (AcSIR), CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, Kerala, India
| | - Halmuthur Mahabalarao Sampath Kumar
- Vaccine Immunology Laboratory, Natural Products Chemistry Division, Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| |
Collapse
|
12
|
Jung H, Kim D, Kang YY, Kim H, Lee JB, Mok H. CpG incorporated DNA microparticles for elevated immune stimulation for antigen presenting cells. RSC Adv 2018; 8:6608-6615. [PMID: 35540407 PMCID: PMC9078369 DOI: 10.1039/c7ra13293j] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/22/2019] [Accepted: 01/30/2018] [Indexed: 11/21/2022] Open
Abstract
As emerging evidence supports the immune stimulating capability of the CpG oligodeoxynucleotides (ODN), CpG-based adjuvants have been widely used. For efficient induction of immune responses, current issues affecting the use of nucleic acid-based adjuvants, e.g. stability in physiological conditions, delivery to immune cells, and successful release within the phagolysosome, should be addressed. Here, we present CpG-based DNA microparticles (DNA-MPs) fabricated by complementary rolling circle amplification (cRCA) as adjuvants for enhancing immune response and production of selective antibody production. Using cRCA method, the sizes of CpG-based DNA-MPs were finely controlled (0.5 and 1 μm) with superior and provided mismatched single stranded form of CpG ODN region for specific cleavage site by DNase II within the phagolysosome. Fabricated CpG-based 1 μm DNA-MPs (DNA-MP-1.0) were successfully internalized into primary macrophages and macrophage cell line (RAW264.7 cells), and elicited superior cytokine production e.g. TNF-α and IL-6, compared to conventional CpG ODNs. After in vivo administration of DNA-MP-1.0 with model antigen ovalbumin (OVA), significantly elevated OVA-specific antibody production was observed. With this in mind, DNA-MP-1.0 could serve as a novel type of adjuvant for the activation of macrophages and the following production of selective antibodies without any noticeable toxicity in vitro and in vivo. As emerging evidence supports the immune stimulating capability of the CpG oligodeoxynucleotides (ODN), CpG-based adjuvants have been widely used.![]()
Collapse
Affiliation(s)
- Heejung Jung
- Department of Bioscience and Biotechnology
- Konkuk University
- Seoul 05029
- Republic of Korea
| | - Dajeong Kim
- Department of Chemical Engineering
- University of Seoul
- Seoul 02504
- Republic of Korea
| | - Yoon Young Kang
- Department of Bioscience and Biotechnology
- Konkuk University
- Seoul 05029
- Republic of Korea
| | - Hyejin Kim
- Department of Chemical Engineering
- University of Seoul
- Seoul 02504
- Republic of Korea
| | - Jong Bum Lee
- Department of Chemical Engineering
- University of Seoul
- Seoul 02504
- Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology
- Konkuk University
- Seoul 05029
- Republic of Korea
| |
Collapse
|
13
|
Nano-sized Soluplus® polymeric micelles enhance the induction of tetanus toxin neutralising antibody response following transcutaneous immunisation with tetanus toxoid. Vaccine 2017; 35:2489-2495. [DOI: 10.1016/j.vaccine.2017.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 02/14/2017] [Accepted: 03/06/2017] [Indexed: 11/27/2022]
|
14
|
Bigaeva E, van Doorn E, Liu H, Hak E. Meta-Analysis on Randomized Controlled Trials of Vaccines with QS-21 or ISCOMATRIX Adjuvant: Safety and Tolerability. PLoS One 2016; 11:e0154757. [PMID: 27149269 PMCID: PMC4858302 DOI: 10.1371/journal.pone.0154757] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/19/2016] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES QS-21 shows in vitro hemolytic effect and causes side effects in vivo. New saponin adjuvant formulations with better toxicity profiles are needed. This study aims to evaluate the safety and tolerability of QS-21 and the improved saponin adjuvants (ISCOM, ISCOMATRIX and Matrix-M™) from vaccine trials. METHODS A systematic literature search was conducted from MEDLINE, EMBASE, Cochrane library and Clinicaltrials.gov. We selected for the meta-analysis randomized controlled trials (RCTs) of vaccines adjuvanted with QS-21, ISCOM, ISCOMATRIX or Matrix-M™, which included a placebo control group and reported safety outcomes. Pooled risk ratios (RRs) and their 95% confidence intervals (CIs) were calculated using a random-effects model. Jadad scale was used to assess the study quality. RESULTS Nine RCTs were eligible for the meta-analysis: six trials on QS-21-adjuvanted vaccines and three trials on ISCOMATRIX-adjuvanted, with 907 patients in total. There were no studies on ISCOM or Matrix-M™ adjuvanted vaccines matching the inclusion criteria. Meta-analysis identified an increased risk for diarrhea in patients receiving QS21-adjuvanted vaccines (RR 2.55, 95% CI 1.04-6.24). No increase in the incidence of the reported systemic AEs was observed for ISCOMATRIX-adjuvanted vaccines. QS-21- and ISCOMATRIX-adjuvanted vaccines caused a significantly higher incidence of injection site pain (RR 4.11, 95% CI 1.10-15.35 and RR 2.55, 95% CI 1.41-4.59, respectively). ISCOMATRIX-adjuvanted vaccines also increased the incidence of injection site swelling (RR 3.43, 95% CI 1.08-10.97). CONCLUSIONS Our findings suggest that vaccines adjuvanted with either QS-21 or ISCOMATRIX posed no specific safety concern. Furthermore, our results indicate that the use of ISCOMATRIX enables a better systemic tolerability profile when compared to the use of QS-21. However, no better local tolerance was observed for ISCOMATRIX-adjuvanted vaccines in immunized non-healthy subjects. This meta-analysis is limited by the relatively small number of individuals recruited in the included trials, especially in the control groups.
Collapse
Affiliation(s)
- Emilia Bigaeva
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Eva van Doorn
- Department of Pharmacy, Unit of PharmacoEpidemiology & PharmacoEconomics (PE2), University of Groningen, Groningen, The Netherlands
| | - Heng Liu
- Department of Pharmacy, Unit of PharmacoEpidemiology & PharmacoEconomics (PE2), University of Groningen, Groningen, The Netherlands
| | - Eelko Hak
- Department of Pharmacy, Unit of PharmacoEpidemiology & PharmacoEconomics (PE2), University of Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
Immunogenicity of multi-epitope-based vaccine candidates administered with the adjuvant Gp96 against rabies. Virol Sin 2016; 31:168-75. [PMID: 27068655 DOI: 10.1007/s12250-016-3734-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/16/2016] [Indexed: 10/22/2022] Open
Abstract
Rabies, a zoonotic disease, causes > 55,000 human deaths globally and results in at least 500 million dollars in losses every year. The currently available rabies vaccines are mainly inactivated and attenuated vaccines, which have been linked with clinical diseases in animals. Thus, a rabies vaccine with high safety and efficacy is urgently needed. Peptide vaccines are known for their low cost, simple production procedures and high safety. Therefore, in this study, we examined the efficacy of multi-epitope-based vaccine candidates against rabies virus. The ability of various peptides to induce epitope-specific responses was examined, and the two peptides that possessed the highest antigenicity and conservation, i.e., AR16 and hPAB, were coated with adjuvant canine-Gp96 and used to prepare vaccines. The peptides were prepared as an emulsion of oil in water (O/W) to create three batches of bivalent vaccine products. The vaccine candidates possessed high safety. Virus neutralizing antibodies were detected on the day 14 after the first immunization in mice and beagles, reaching 5-6 IU/mL in mice and 7-9 IU/mL in beagles by day 28. The protective efficacy of the vaccine candidates was about 70%-80% in mice challenged by a virulent strain of rabies virus. Thus, a novel multi-epitope-based rabies vaccine with Gp96 as an adjuvant was developed and validated in mice and dogs. Our results suggest that synthetic peptides hold promise for the development of novel vaccines against rabies.
Collapse
|
16
|
Liposome-Based Adjuvants for Subunit Vaccines: Formulation Strategies for Subunit Antigens and Immunostimulators. Pharmaceutics 2016; 8:pharmaceutics8010007. [PMID: 26978390 PMCID: PMC4810083 DOI: 10.3390/pharmaceutics8010007] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 11/25/2022] Open
Abstract
The development of subunit vaccines has become very attractive in recent years due to their superior safety profiles as compared to traditional vaccines based on live attenuated or whole inactivated pathogens, and there is an unmet medical need for improved vaccines and vaccines against pathogens for which no effective vaccines exist. The subunit vaccine technology exploits pathogen subunits as antigens, e.g., recombinant proteins or synthetic peptides, allowing for highly specific immune responses against the pathogens. However, such antigens are usually not sufficiently immunogenic to induce protective immunity, and they are often combined with adjuvants to ensure robust immune responses. Adjuvants are capable of enhancing and/or modulating immune responses by exposing antigens to antigen-presenting cells (APCs) concomitantly with conferring immune activation signals. Few adjuvant systems have been licensed for use in human vaccines, and they mainly stimulate humoral immunity. Thus, there is an unmet demand for the development of safe and efficient adjuvant systems that can also stimulate cell-mediated immunity (CMI). Adjuvants constitute a heterogeneous group of compounds, which can broadly be classified into delivery systems or immunostimulators. Liposomes are versatile delivery systems for antigens, and they can carefully be customized towards desired immune profiles by combining them with immunostimulators and optimizing their composition, physicochemical properties and antigen-loading mode. Immunostimulators represent highly diverse classes of molecules, e.g., lipids, nucleic acids, proteins and peptides, and they are ligands for pattern-recognition receptors (PRRs), which are differentially expressed on APC subsets. Different formulation strategies might thus be required for incorporation of immunostimulators and antigens, respectively, into liposomes, and the choice of immunostimulator should ideally be based on knowledge regarding the specific PRR expression profile of the target APCs. Here, we review state-of-the-art formulation approaches employed for the inclusion of immunostimulators and subunit antigens into liposome dispersion and their optimization towards robust vaccine formulations.
Collapse
|
17
|
Chen L, Zhang J, Sun H. Immunological adjuvant effect of the peptide fraction from the larvae of Musca domestica. Altern Ther Health Med 2015; 15:427. [PMID: 26630909 PMCID: PMC4668601 DOI: 10.1186/s12906-015-0951-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 11/26/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND The larvae of Musca domestica (Diptera: Muscidae) have been used traditionally for malnutritional stagnation, decubital necrosis, osteomyelitis, ecthyma and lip scald and also to treat coma and gastric cancer in the traditional Chinese medicine. Its immunomodulatory effects in naïve mice in relation to the traditional uses were also reported. However, the immunological adjuvant potentials of this insect have not yet been studied. METHODS The peptide fraction from the larvae of Musca domestica L. (MDPF) was evaluated for its adjuvant potentials on the immune responses to ovalbumin (OVA) and avian influenza vaccine (rL-H5) by determining antigen-specific antibody titers, splenocyte proliferation, activity of natural killer (NK) cell, the secretion of cytokines from splenocytes in the immunized mice. RESULTS MDPF significantly enhanced not only the concanavalin A (Con A)-, lipopolysaccharide (LPS)- and antigen-stimulated splenocyte proliferation, but serum antigen-specific IgG, IgG1, IgG2a, and IgG2b antibody titers in the mice immunized with OVA and rL-H5. MDPF also remarkably promoted the killing activities of NK cells in splenocytes from the mice immunized with rL-H5. Furthermore, MDPF significantly promoted the production of Th1 (IL-2 and IFN-γ) and Th2 (IL-10) cytokines from splenocytes in the immunized mice. CONCLUSIONS The results indicated that MDPF had a potential to increase both cellular and humoral immune responses and elicit a balanced Th1/Th2 response, and that MDPF may be a safe and efficacious vaccine adjuvant candidate.
Collapse
|
18
|
Galliher-Beckley A, Pappan LK, Madera R, Burakova Y, Waters A, Nickles M, Li X, Nietfeld J, Schlup JR, Zhong Q, McVey S, Dritz SS, Shi J. Characterization of a novel oil-in-water emulsion adjuvant for swine influenza virus and Mycoplasma hyopneumoniae vaccines. Vaccine 2015; 33:2903-8. [PMID: 25936722 DOI: 10.1016/j.vaccine.2015.04.065] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/14/2015] [Accepted: 04/16/2015] [Indexed: 11/16/2022]
Abstract
Vaccines consisting of subunit or inactivated bacteria/virus and potent adjuvants are widely used to control and prevent infectious diseases. Because inactivated and subunit antigens are often less antigenic than live microbes, a growing need exists for the development of new and improved vaccine adjuvants that can elicit rapid and long-lasting immunity. Here we describe the development and characterization of a novel oil-in-water emulsion, OW-14. OW-14 contains low-cost plant-based emulsifiers and was added to antigen at a ratio of 1:3 with simple hand mixing. OW-14 was stable for prolonged periods of time at temperatures ranging from 4 to 40°C and could be sterilized by autoclaving. Our results showed that OW-14 adjuvanted inactivated swine influenza viruses (SIV; H3N2 and H1N1) and Mycoplasma hyopneumoniae (M. hyo) vaccines could be safely administered to piglets in two doses, three weeks apart. Injection sites were monitored and no adverse reactions were observed. Vaccinated pigs developed high and prolonged antibody titers to both SIV and M. hyo. Interestingly, antibody titers were either comparable or greater than those produced by commercially available FluSure (SIV) or RespiSure (M. hyo) vaccines. We also found that OW-14 can induce high antibody responses in pigs that were vaccinated with a decreased antigen dose. This study provides direct evidence that we have developed an easy-to-use and low-cost emulsion that can act as a powerful adjuvant in two common types of swine vaccines.
Collapse
Affiliation(s)
- A Galliher-Beckley
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - L K Pappan
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Rachel Madera
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Y Burakova
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; Department of Chemical Engineering, College of Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - A Waters
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - M Nickles
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - X Li
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - J Nietfeld
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - J R Schlup
- Department of Chemical Engineering, College of Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Q Zhong
- Department of Food Science and Technology, University of Tennessee, Knoxville, TN 37996, USA
| | - S McVey
- United States Department of Agriculture, Agricultural Research Service, Arthropod Borne Animal Disease Research Unit, Manhattan, KS 66502, USA
| | - S S Dritz
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - J Shi
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
19
|
Seth A, Ritchie FK, Wibowo N, Lua LHL, Middelberg APJ. Non-carrier nanoparticles adjuvant modular protein vaccine in a particle-dependent manner. PLoS One 2015; 10:e0117203. [PMID: 25756283 PMCID: PMC4355484 DOI: 10.1371/journal.pone.0117203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/20/2014] [Indexed: 11/18/2022] Open
Abstract
Nanoparticles are increasingly used to adjuvant vaccine formulations due to their biocompatibility, ease of manufacture and the opportunity to tailor their size, shape, and physicochemical properties. The efficacy of similarly-sized silica (Si-OH), poly (D,L-lactic-co-glycolic acid) (PLGA) and poly caprolactone (PCL) nanoparticles (nps) to adjuvant recombinant capsomere presenting antigenic M2e modular peptide from Influenza A virus (CapM2e) was investigated in vivo. Formulation of CapM2e with Si-OH or PLGA nps significantly boosted the immunogenicity of modular capsomeres, even though CapM2e was not actively attached to the nanoparticles prior to injection (i.e., formulation was by simple mixing). In contrast, PCL nps showed no significant adjuvant effect using this simple-mixing approach. The immune response induced by CapM2e alone or formulated with nps was antibody-biased with very high antigen-specific antibody titer and less than 20 cells per million splenocytes secreting interferon gamma. Modification of silica nanoparticle surface properties through amine functionalization and pegylation did not lead to significant changes in immune response. This study confirms that simple mixing-based formulation can lead to effective adjuvanting of antigenic protein, though with antibody titer dependent on nanoparticle physicochemical properties.
Collapse
Affiliation(s)
- Arjun Seth
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD, Australia
| | - Fiona K Ritchie
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD, Australia
| | - Nani Wibowo
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD, Australia
| | - Linda H L Lua
- The University of Queensland, Protein Expression Facility, St Lucia, QLD, Australia
| | - Anton P J Middelberg
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD, Australia
| |
Collapse
|
20
|
Onishi M, Ozasa K, Kobiyama K, Ohata K, Kitano M, Taniguchi K, Homma T, Kobayashi M, Sato A, Katakai Y, Yasutomi Y, Wijaya E, Igarashi Y, Nakatsu N, Ise W, Inoue T, Yamada H, Vandenbon A, Standley DM, Kurosaki T, Coban C, Aoshi T, Kuroda E, Ishii KJ. Hydroxypropyl-β-cyclodextrin spikes local inflammation that induces Th2 cell and T follicular helper cell responses to the coadministered antigen. THE JOURNAL OF IMMUNOLOGY 2015; 194:2673-82. [PMID: 25681338 DOI: 10.4049/jimmunol.1402027] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cyclodextrins are commonly used as a safe excipient to enhance the solubility and bioavailability of hydrophobic pharmaceutical agents. Their efficacies and mechanisms as drug-delivery systems have been investigated for decades, but their immunological properties have not been examined. In this study, we reprofiled hydroxypropyl-β-cyclodextrin (HP-β-CD) as a vaccine adjuvant and found that it acts as a potent and unique adjuvant. HP-β-CD triggered the innate immune response at the injection site, was trapped by MARCO(+) macrophages, increased Ag uptake by dendritic cells, and facilitated the generation of T follicular helper cells in the draining lymph nodes. It significantly enhanced Ag-specific Th2 and IgG Ab responses as potently as did the conventional adjuvant, aluminum salt (alum), whereas its ability to induce Ag-specific IgE was less than that of alum. At the injection site, HP-β-CD induced the temporary release of host dsDNA, a damage-associated molecular pattern. DNase-treated mice, MyD88-deficient mice, and TBK1-deficient mice showed significantly reduced Ab responses after immunization with this adjuvant. Finally, we demonstrated that HP-β-CD-adjuvanted influenza hemagglutinin split vaccine protected against a lethal challenge with a clinically isolated pandemic H1N1 influenza virus, and the adjuvant effect of HP-β-CD was demonstrated in cynomolgus macaques. Our results suggest that HP-β-CD acts as a potent MyD88- and TBK1-dependent T follicular helper cell adjuvant and is readily applicable to various vaccines.
Collapse
Affiliation(s)
- Motoyasu Onishi
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan; Infectious Diseases, Medicinal Research Laboratories, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Koji Ozasa
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Kouji Kobiyama
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Keiichi Ohata
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Mitsutaka Kitano
- Infectious Diseases, Medicinal Research Laboratories, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Keiichi Taniguchi
- Infectious Diseases, Medicinal Research Laboratories, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Tomoyuki Homma
- Infectious Diseases, Medicinal Research Laboratories, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Masanori Kobayashi
- Infectious Diseases, Medicinal Research Laboratories, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Akihiko Sato
- Infectious Diseases, Medicinal Research Laboratories, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Yuko Katakai
- Corporation for the Production and Research of Laboratory Primates, Tsukuba, Ibaraki 305-0843, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Tsukuba, Ibaraki 305-0843, Japan
| | - Edward Wijaya
- System Immunology Laboratory, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Yoshinobu Igarashi
- Toxicogenomics Project, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Noriyuki Nakatsu
- Toxicogenomics Project, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Wataru Ise
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Hiroshi Yamada
- Toxicogenomics Project, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
| | - Alexis Vandenbon
- System Immunology Laboratory, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Daron M Standley
- System Immunology Laboratory, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan; Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Kanagawa 230-0045, Japan; and
| | - Cevayir Coban
- Laboratory of Malaria Immunology, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Taiki Aoshi
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Etsushi Kuroda
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center, World Premier Institute, Osaka University, Osaka 565-0871, Japan;
| |
Collapse
|
21
|
Manrique A, Adams E, Barouch DH, Fast P, Graham BS, Kim JH, Kublin JG, McCluskey M, Pantaleo G, Robinson HL, Russell N, Snow W, Johnston MI. The immune space: a concept and template for rationalizing vaccine development. AIDS Res Hum Retroviruses 2014; 30:1017-22. [PMID: 24857015 PMCID: PMC4208609 DOI: 10.1089/aid.2014.0040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Empirical testing of candidate vaccines has led to the successful development of a number of lifesaving vaccines. The advent of new tools to manipulate antigens and new methods and vectors for vaccine delivery has led to a veritable explosion of potential vaccine designs. As a result, selection of candidate vaccines suitable for large-scale efficacy testing has become more challenging. This is especially true for diseases such as dengue, HIV, and tuberculosis where there is no validated animal model or correlate of immune protection. Establishing guidelines for the selection of vaccine candidates for advanced testing has become a necessity. A number of factors could be considered in making these decisions, including, for example, safety in animal and human studies, immune profile, protection in animal studies, production processes with product quality and stability, availability of resources, and estimated cost of goods. The "immune space template" proposed here provides a standardized approach by which the quality, level, and durability of immune responses elicited in early human trials by a candidate vaccine can be described. The immune response profile will demonstrate if and how the candidate is unique relative to other candidates, especially those that have preceded it into efficacy testing and, thus, what new information concerning potential immune correlates could be learned from an efficacy trial. A thorough characterization of immune responses should also provide insight into a developer's rationale for the vaccine's proposed mechanism of action. HIV vaccine researchers plan to include this general approach in up-selecting candidates for the next large efficacy trial. This "immune space" approach may also be applicable to other vaccine development endeavors where correlates of vaccine-induced immune protection remain unknown.
Collapse
Affiliation(s)
| | - Elizabeth Adams
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Pat Fast
- International AIDS Vaccine Initiative, New York, New York
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Jerome H. Kim
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - James G. Kublin
- HIV Vaccine Trials Network and the Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | | | - Nina Russell
- The Bill & Melinda Gates Foundation, Seattle, Washington
| | - William Snow
- Global HIV Vaccine Enterprise, New York, New York
| | | |
Collapse
|
22
|
Sun H, He S, Shi M. Adjuvant-active fraction from Albizia julibrissin saponins improves immune responses by inducing cytokine and chemokine at the site of injection. Int Immunopharmacol 2014; 22:346-55. [DOI: 10.1016/j.intimp.2014.07.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/28/2014] [Accepted: 07/16/2014] [Indexed: 11/26/2022]
|
23
|
Uraki R, Das SC, Hatta M, Kiso M, Iwatsuki-Horimoto K, Ozawa M, Coban C, Ishii KJ, Kawaoka Y. Hemozoin as a novel adjuvant for inactivated whole virion influenza vaccine. Vaccine 2014; 32:5295-300. [PMID: 25108216 DOI: 10.1016/j.vaccine.2014.07.079] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 05/27/2014] [Accepted: 07/22/2014] [Indexed: 11/27/2022]
Abstract
Because vaccination is an effective means to protect humans from influenza viruses, extensive efforts have been made to develop not only new vaccines, but also for new adjuvants to enhance the efficacy of existing inactivated vaccines. Here, we examined the adjuvanticity of synthetic hemozoin, a synthetic version of the malarial by-product hemozoin, on the vaccine efficacy of inactivated whole influenza viruses in a mouse model. We found that mice immunized twice with hemozoin-adjuvanted inactivated A/California/04/2009 (H1N1pdm09) or A/Vietnam/1203/2004 (H5N1) virus elicited higher virus-specific antibody responses than did mice immunized with non-adjuvanted counterparts. Furthermore, mice immunized with hemozoin-adjuvanted inactivated viruses were better protected from lethal challenge with influenza viruses than were mice immunized with non-adjuvanted inactivated vaccines. Our results show that hemozoin improves the immunogenicity of inactivated influenza viruses, and is thus a promising adjuvant for inactivated whole virion influenza vaccines.
Collapse
Affiliation(s)
- Ryuta Uraki
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Subash C Das
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Masato Hatta
- Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Maki Kiso
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Kiyoko Iwatsuki-Horimoto
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Makoto Ozawa
- Laboratory of Animal Hygiene, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan; Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan
| | - Cevayir Coban
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, Osaka, Japan; Laboratory of Vaccine Science, IFReC, Osaka University, Osaka, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; Influenza Research Institute, Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53711, USA; ERATO Infection-Induced Host Responses Project (JST), Saitama 332-0012, Japan; Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
24
|
Kumru OS, Joshi SB, Smith DE, Middaugh CR, Prusik T, Volkin DB. Vaccine instability in the cold chain: mechanisms, analysis and formulation strategies. Biologicals 2014; 42:237-59. [PMID: 24996452 DOI: 10.1016/j.biologicals.2014.05.007] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 05/12/2014] [Accepted: 05/27/2014] [Indexed: 12/15/2022] Open
Abstract
Instability of vaccines often emerges as a key challenge during clinical development (lab to clinic) as well as commercial distribution (factory to patient). To yield stable, efficacious vaccine dosage forms for human use, successful formulation strategies must address a combination of interrelated topics including stabilization of antigens, selection of appropriate adjuvants, and development of stability-indicating analytical methods. This review covers key concepts in understanding the causes and mechanisms of vaccine instability including (1) the complex and delicate nature of antigen structures (e.g., viruses, proteins, carbohydrates, protein-carbohydrate conjugates, etc.), (2) use of adjuvants to further enhance immune responses, (3) development of physicochemical and biological assays to assess vaccine integrity and potency, and (4) stabilization strategies to protect vaccine antigens and adjuvants (and their interactions) during storage. Despite these challenges, vaccines can usually be sufficiently stabilized for use as medicines through a combination of formulation approaches combined with maintenance of an efficient cold chain (manufacturing, distribution, storage and administration). Several illustrative case studies are described regarding mechanisms of vaccine instability along with formulation approaches for stabilization within the vaccine cold chain. These include live, attenuated (measles, polio) and inactivated (influenza, polio) viral vaccines as well as recombinant protein (hepatitis B) vaccines.
Collapse
Affiliation(s)
- Ozan S Kumru
- Macromolecule and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Sangeeta B Joshi
- Macromolecule and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Dawn E Smith
- Temptime Corporation, Morris Plains, NJ 07950, USA
| | - C Russell Middaugh
- Macromolecule and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Ted Prusik
- Temptime Corporation, Morris Plains, NJ 07950, USA
| | - David B Volkin
- Macromolecule and Vaccine Stabilization Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA.
| |
Collapse
|
25
|
Wibowo N, Chuan YP, Seth A, Cordoba Y, Lua LHL, Middelberg APJ. Co-administration of non-carrier nanoparticles boosts antigen immune response without requiring protein conjugation. Vaccine 2014; 32:3664-9. [PMID: 24793947 DOI: 10.1016/j.vaccine.2014.04.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 04/11/2014] [Accepted: 04/16/2014] [Indexed: 11/28/2022]
Abstract
Nanotechnology promises a revolution in medicine including through new vaccine approaches. The use of nanoparticles in vaccination has, to date, focused on attaching antigen directly to or within nanoparticle structures to enhance antigen uptake by immune cells. Here we question whether antigen incorporation with the nanoparticle is actually necessary to boost vaccine effectiveness. We show that the immunogenicity of a sub-unit protein antigen was significantly boosted by formulation with silica nanoparticles even without specific conjugation of antigen to the nanoparticle. We further show that this effect was observed only for virus-sized nanoparticles (50 nm) but not for larger (1,000 nm) particles, demonstrating a pronounced effect of nanoparticle size. This non-attachment approach has potential to radically simplify the development and application of nanoparticle-based formulations, leading to safer and simpler nanoparticle applications in vaccine development.
Collapse
Affiliation(s)
- Nani Wibowo
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD 4072, Australia
| | - Yap P Chuan
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD 4072, Australia
| | - Arjun Seth
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD 4072, Australia
| | - Yoann Cordoba
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD 4072, Australia
| | - Linda H L Lua
- The University of Queensland, Protein Expression Facility, St. Lucia, QLD 4072, Australia
| | - Anton P J Middelberg
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St. Lucia, QLD 4072, Australia.
| |
Collapse
|
26
|
Levast B, Awate S, Babiuk L, Mutwiri G, Gerdts V, van Drunen Littel-van den Hurk S. Vaccine Potentiation by Combination Adjuvants. Vaccines (Basel) 2014; 2:297-322. [PMID: 26344621 PMCID: PMC4494260 DOI: 10.3390/vaccines2020297] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/22/2014] [Accepted: 03/28/2014] [Indexed: 01/02/2023] Open
Abstract
Adjuvants are crucial components of vaccines. They significantly improve vaccine efficacy by modulating, enhancing, or extending the immune response and at the same time reducing the amount of antigen needed. In contrast to previously licensed adjuvants, current successful adjuvant formulations often consist of several molecules, that when combined, act synergistically by activating a variety of immune mechanisms. These "combination adjuvants" are already registered with several vaccines, both in humans and animals, and novel combination adjuvants are in the pipeline. With improved knowledge of the type of immune responses needed to successfully induce disease protection by vaccination, combination adjuvants are particularly suited to not only enhance, but also direct the immune responses desired to be either Th1-, Th2- or Th17-biased. Indeed, in view of the variety of disease and population targets for vaccine development, a panel of adjuvants will be needed to address different disease targets and populations. Here, we will review well-known and new combination adjuvants already licensed or currently in development-including ISCOMs, liposomes, Adjuvant Systems Montanides, and triple adjuvant combinations-and summarize their performance in preclinical and clinical trials. Several of these combination adjuvants are promising having promoted improved and balanced immune responses.
Collapse
Affiliation(s)
- Benoît Levast
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
| | - Sunita Awate
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
| | - Lorne Babiuk
- University Hall, University of Alberta, Edmonton, AB T6G 2J9, Canada.
| | - George Mutwiri
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
- School of Public Health, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada.
| | - Volker Gerdts
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
- Veterinary Microbiology, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada.
| | - Sylvia van Drunen Littel-van den Hurk
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
- Microbiology and Immunology, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
27
|
Sasaki H, Ishikawa H, Kojima K, Itoh M, Matsumoto T, Itoh T, Hosomi O, Kawamoto E. Intranasal immunization with a non-adjuvanted adhesive protein descended from Pasteurella pneumotropica and its preventive efficacy against opportunistic infection in mice. Vaccine 2013; 31:5729-35. [DOI: 10.1016/j.vaccine.2013.09.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 08/25/2013] [Accepted: 09/19/2013] [Indexed: 12/14/2022]
|
28
|
García A, De Sanctis JB. An overview of adjuvant formulations and delivery systems. APMIS 2013; 122:257-67. [PMID: 23919674 DOI: 10.1111/apm.12143] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 05/30/2013] [Indexed: 12/19/2022]
Abstract
Adjuvants may promote immune responses: by recruiting professional antigen-presenting cells (APCs) to the vaccination site, increasing the delivery of antigens to APCs, or by activating APCs to produce cytokines and by triggering T cell responses. Aluminium salts have been effective at promoting protective humoral immunity; however, they are not effective in generating cell-mediated immunity. A number of different approaches have been developed to potentiate immune response and they have been partially successful. Research has been conducted into vaccine delivery systems (VDS); enhancing cross-presentation by targeting antigens to (APCs). Antigen discovery has increased over the past decade, and consequently, it has accelerated vaccine development demanding a new generation of VDS that combines different types of adjuvants into specific formulations with greater activity. The new approaches offer a wide spectrum of opportunities in vaccine research with direct applications in the near future.
Collapse
Affiliation(s)
- Alexis García
- Instituto de Inmunologia, Facultad de Medicina, Universidad Central de Venezuela, Apartado, Venezuela
| | | |
Collapse
|
29
|
Influenza vaccine: Development of a novel intranasal and subcutaneous recombinant adjuvant. Vaccine 2013; 31:4009-16. [DOI: 10.1016/j.vaccine.2013.05.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 04/29/2013] [Accepted: 05/10/2013] [Indexed: 12/15/2022]
|
30
|
Vargas-Inchaustegui DA, Robert-Guroff M. Fc receptor-mediated immune responses: new tools but increased complexity in HIV prevention. Curr HIV Res 2013; 11:407-20. [PMID: 24191937 PMCID: PMC6288814 DOI: 10.2174/1570162x113116660063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/04/2013] [Accepted: 06/06/2013] [Indexed: 12/21/2022]
Abstract
The modest success of the RV144 HIV vaccine trial in Thailand and the ensuing suggestion that a Fc-receptormediated antibody activity might have played a role in the protection observed have intensified investigations on Fcrelated immune responses. HIV neutralizing antibodies have been and continue to be the focal point of research into humoral immune protection. However, recent knowledge that their protective efficacy can be augmented by Fc-FcR interactions has increased the complexity of identifying immune correlates of protection. If anything, continued studies of both humoral and cellular immune mechanisms point to the lack of a single protective anti-HIV immune response. Here we focus on humoral immunity, analyzing the role played by Fc receptor-related responses and discussing how new knowledge of their interactions requires further investigation, but may also spur novel vaccination approaches. We initially address classical Fc-receptor mediated anti-viral mechanisms including antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cell mediated viral inhibition (ADCVI), and antibody-dependent cellular phagocytosis (ADCP), as well as the effector cells that mediate these functions. Next, we summarize key aspects of FcR-Fc interactions that are important for potential control of HIV/SIV such as FcR polymorphisms and post-transcriptional modifications. Finally we discuss less commonly studied non-mechanistic anti-HIV immune functions: antibody avidity and envelopespecific B cell memory. Overall, a spectrum of immune responses, reflecting the immune system's redundancy, will likely be needed to prevent HIV infection and/or disease progression. Aside from elicitation of critical immune mechanisms, a successful vaccine will need to induce mature B cell responses and long-lasting immune memory.
Collapse
Affiliation(s)
- Diego A Vargas-Inchaustegui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, 41 Medlars Drive, Building 41, Room D804, Bethesda, MD 20192-5065, USA.
| | | |
Collapse
|
31
|
Matyas GR, Mayorov AV, Rice KC, Jacobson AE, Cheng K, Iyer MR, Li F, Beck Z, Janda KD, Alving CR. Liposomes containing monophosphoryl lipid A: a potent adjuvant system for inducing antibodies to heroin hapten analogs. Vaccine 2013; 31:2804-10. [PMID: 23624097 PMCID: PMC4120113 DOI: 10.1016/j.vaccine.2013.04.027] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Revised: 03/08/2013] [Accepted: 04/09/2013] [Indexed: 12/21/2022]
Abstract
In order to create an effective immunization approach for a potential vaccine to heroin, liposomes containing monophosphoryl lipid A [L(MPLA)] were tested as an adjuvant system to induce antibodies to heroin hapten analogs. Four synthetic haptens and two immunization strategies were employed. In the first strategy, a hydrophobic 23 amino acid immunogenic peptide derived from the membrane proximal external region of gp41 from HIV-1 envelope protein was embedded as a carrier in the outer surface of L(MPLA), to which was conjugated a 15 amino acid universal T cell epitope and a terminal heroin hapten analog. In the second strategy, tetanus toxoid (TT) carrier protein was decorated with haptens by conjugation, and the hapten-conjugated protein was mixed with L(MPLA). After immunization of mice, each of the immunization strategies was effective for induction of IgG anti-hapten antibodies. The first immunization strategy induced a mean end-point IgG titer against one of two haptens tested of approximately 12,800; however, no detectable antibodies were induced against the liposome-associated HIV-1 carrier peptide. In the second immunization strategy, depending on the hapten used for decorating the TT, end-point IgG titers ranged from 100,000 to 6,500,000. In this strategy, in which hapten was conjugated to the TT, end-point IgG titers of 400,000 to the TT carrier were observed with each conjugate. However, upon mixing unconjugated TT with L(MPLA), anti-TT titers of 6,500,000 were observed. We conclude that L(MPLA) serves as a potent adjuvant for inducing antibodies to candidate heroin haptens. However, antibodies to the carrier peptide or protein were partly or completed inhibited by the presence of conjugated hapten.
Collapse
Affiliation(s)
- Gary R. Matyas
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Alexander V. Mayorov
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA
| | - Kenner C. Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Arthur E. Jacobson
- Chemical Biology Research Branch, National Institute on Drug Abuse, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Kejun Cheng
- Chemical Biology Research Branch, National Institute on Drug Abuse, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Malliga R. Iyer
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Fuying Li
- Chemical Biology Research Branch, National Institute on Drug Abuse, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Zoltan Beck
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA
| | - Kim D. Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology and Worm Institute of Research and Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Carl R. Alving
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| |
Collapse
|
32
|
Fernandes da Costa SP, Mot D, Bokori-Brown M, Savva CG, Basak AK, Van Immerseel F, Titball RW. Protection against avian necrotic enteritis after immunisation with NetB genetic or formaldehyde toxoids. Vaccine 2013; 31:4003-8. [PMID: 23727000 PMCID: PMC3763374 DOI: 10.1016/j.vaccine.2013.05.063] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 05/02/2013] [Accepted: 05/14/2013] [Indexed: 11/25/2022]
Abstract
NetB (necrotic enteritis toxin B) is a recently identified β-pore-forming toxin produced by Clostridium perfringens. This toxin has been shown to play a major role in avian necrotic enteritis. In recent years, a dramatic increase in necrotic enteritis has been observed, especially in countries where the use of antimicrobial growth promoters in animal feedstuffs has been banned. The aim of this work was to determine whether immunisation with a NetB toxoid would provide protection against necrotic enteritis. The immunisation of poultry with a formaldehyde NetB toxoid or with a NetB genetic toxoid (W262A) resulted in the induction of antibody responses against NetB and provided partial protection against disease.
Collapse
Affiliation(s)
- Sérgio P Fernandes da Costa
- College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
33
|
Singh M. Strategies for the Nonclinical Safety Assessment of Vaccines. NOVEL IMMUNE POTENTIATORS AND DELIVERY TECHNOLOGIES FOR NEXT GENERATION VACCINES 2013. [PMCID: PMC7120100 DOI: 10.1007/978-1-4614-5380-2_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Over the past century, vaccines have made a large impact on public health. Prophylactic vaccines prevent disability and disease, saving millions of dollars in potential health-care spending. Since prophylactic vaccines are administered to healthy individuals, including infants and children, it is important to demonstrate the safety of vaccines preclinically prior to testing the vaccine in clinical studies. A benefit-to-risk profile is considered for each individual vaccine and depends on many factors including preclinical and clinical toxicities that are observed, frequency of administration and intended target population. For prophylactic vaccines, in particular, the concerns about potential risks often outweigh the perception of benefit [1]. Therefore, over the past decade, there has been an increased focus on nonclinical safety assessment of vaccines, including toxicity testing.
Collapse
Affiliation(s)
- Manmohan Singh
- Novartis Vaccines Research, Cambridge, 02139 Massachusetts USA
| |
Collapse
|
34
|
Alving CR, Rao M, Steers NJ, Matyas GR, Mayorov AV. Liposomes containing lipid A: an effective, safe, generic adjuvant system for synthetic vaccines. Expert Rev Vaccines 2012; 11:733-44. [PMID: 22873129 DOI: 10.1586/erv.12.35] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liposomes containing monophosphoryl lipid A (MPLA) have previously exhibited considerable potency and safety in human trials with a variety of candidate vaccines, including vaccines to malaria, HIV-1 and several different types of cancer. The long history of research and development of MPLA and liposomal MPLA as vaccine adjuvants reveals that there are numerous opportunities for creation and development of generic (nonproprietary) adjuvant system formulations with these materials that are not only highly potent and safe, but also readily available as native materials or as synthetic compounds. They are easily manufactured as potentially inexpensive and easy to use adjuvant systems and might be effective even with synthetic peptides as antigens.
Collapse
Affiliation(s)
- Carl R Alving
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | | | | | | | | |
Collapse
|
35
|
Giddam AK, Zaman M, Skwarczynski M, Toth I. Liposome-based delivery system for vaccine candidates: constructing an effective formulation. Nanomedicine (Lond) 2012; 7:1877-93. [DOI: 10.2217/nnm.12.157] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The discovery of liposomes in 1965 by Bangham and coworkers changed the prospects of drug delivery systems. Since then, the application of liposomes as vaccine delivery systems has been studied extensively. Liposomal vaccine delivery systems are made up of nano- or micro-sized vesicles consisting of phospholipid bilayers, in which the bioactive molecule is encapsulated/entrapped, adsorbed or surface coupled. In general, liposomes are not immunogenic on their own; thus, liposomes combined with immunostimulating ligands (adjuvants) or various other formulations have been used as vaccine delivery systems. A thorough understanding of formulation parameters allows the design of effective liposomal vaccine delivery systems. This article provides an overview of various factors that influence liposomal immunogenicity. In particular, the effects of vesicle size, surface charge, bilayer composition, lamellarity, pegylation and targeting of liposomes are described.
Collapse
Affiliation(s)
- Ashwini Kumar Giddam
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD 4072, Australia
| | - Mehfuz Zaman
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD 4072, Australia
| | - Mariusz Skwarczynski
- The University of Queensland, School of Chemistry & Molecular Biosciences, St Lucia, QLD 4072, Australia
| | - Istvan Toth
- The University of Queensland, School of Pharmacy, St Lucia, QLD 4072, Australia
| |
Collapse
|
36
|
Intranasal vaccination with murabutide enhances humoral and mucosal immune responses to a virus-like particle vaccine. PLoS One 2012; 7:e41529. [PMID: 22855691 PMCID: PMC3405106 DOI: 10.1371/journal.pone.0041529] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 06/27/2012] [Indexed: 01/12/2023] Open
Abstract
Murabutide (MB) is a synthetic immunomodulator recognized by the nucleotide-binding oligomerization domain-containing protein 2 (NOD2) receptor on mammalian cells. MB has previously been approved for testing in multiple human clinical trials to determine its value as an antiviral therapeutic, and as an adjuvant for injected vaccines. We have found a new use for this immunomodulator; it functions as a mucosal adjuvant that enhances immunogenicity of virus-like particles (VLP) administered intranasally. MB enhanced Norwalk virus (NV) VLP-specific IgG systemically and IgA production at distal mucosal sites following intranasal (IN) vaccination. A dose escalation study identified 100 µg as the optimal MB dosage in mice, based on the magnitude of VLP-specific IgG, IgG1, IgG2a and IgA production in serum and VLP-specific IgA production at distal mucosal sites. IN vaccination using VLP with MB was compared to IN delivery VLP with cholera toxin (CT) or gardiquimod (GARD) and to parenteral VLP delivery with alum; the MB groups were equivalent to CT and GARD and superior to alum in inducing mucosal immune responses and stimulated equivalent systemic VLP-specific antibodies. These data support the further testing of MB as a potent mucosal adjuvant for inducing robust and durable antibody responses to non-replicating subunit vaccines.
Collapse
|
37
|
Okamura M, Matsumoto W, Seike F, Tanaka Y, Teratani C, Tozuka M, Kashimoto T, Takehara K, Nakamura M, Yoshikawa Y. Efficacy of Soluble Recombinant FliC Protein from Salmonella enterica Serovar Enteritidis as a Potential Vaccine Candidate Against Homologous Challenge in Chickens. Avian Dis 2012; 56:354-8. [DOI: 10.1637/9986-111011-reg.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
38
|
Boraschi D, Costantino L, Italiani P. Interaction of nanoparticles with immunocompetent cells: nanosafety considerations. Nanomedicine (Lond) 2012; 7:121-31. [PMID: 22191781 DOI: 10.2217/nnm.11.169] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Intentional delivery of high doses of nanoparticle-based drugs or diagnostic/imaging systems sets a new scenario in safety evaluation, compared with the environmentally borne unintentional exposure to engineered nanoparticles. Intravenously administered nanomedicines immediately interact with blood components such as serum proteins, changing their own characteristics and consequently the features of their interaction with cells and tissues. Of major importance is the interaction of nanomedicines with the immune system, which is essential in the recognition and elimination of foreign dangerous agents. Nanomedicines need to avoid immune recognition in order to reach their therapeutic target and display their effect, should not trigger defensive mechanisms that can damage the body tissues (e.g., complement activation or inflammation) and should not interfere with immunocompetent cells in order to avoid promoting immune-related diseases. This review will briefly cover these issues, and propose some knowledge-based approaches for future 'safe-by-design' nanomedicines.
Collapse
Affiliation(s)
- Diana Boraschi
- Institute of Biomedical Technologies, National Research Council, Pisa, Italy.
| | | | | |
Collapse
|
39
|
Lai RPJ, Seaman MS, Tonks P, Wegmann F, Seilly DJ, Frost SDW, LaBranche CC, Montefiori DC, Dey AK, Srivastava IK, Sattentau Q, Barnett SW, Heeney JL. Mixed adjuvant formulations reveal a new combination that elicit antibody response comparable to Freund's adjuvants. PLoS One 2012; 7:e35083. [PMID: 22509385 PMCID: PMC3324409 DOI: 10.1371/journal.pone.0035083] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/08/2012] [Indexed: 12/04/2022] Open
Abstract
Adjuvant formulations capable of inducing high titer and high affinity antibody responses would provide a major advance in the development of vaccines to viral infections such as HIV-1. Although oil-in-water emulsions, such as Freund's adjuvant (FCA/FIA), are known to be potent, their toxicity and reactogenicity make them unacceptable for human use. Here, we explored different adjuvants and compared their ability to elicit antibody responses to FCA/FIA. Recombinant soluble trimeric HIV-1 gp140 antigen was formulated in different adjuvants, including FCA/FIA, Carbopol-971P, Carbopol-974P and the licensed adjuvant MF59, or combinations of MF59 and Carbopol. The antigen-adjuvant formulation was administered in a prime-boost regimen into rabbits, and elicitation of antigen binding and neutralizing antibodies (nAbs) was evaluated. When used individually, only FCA/FIA elicited significantly higher titer of nAbs than the control group (gp140 in PBS (p<0.05)). Sequential prime-boost immunizations with different adjuvants did not offer improvements over the use of FCA/FIA or MF59. Remarkably however, the concurrent use of the combination of Carbopol-971P and MF59 induced potent adjuvant activity with significantly higher titer nAbs than FCA/FIA (p<0.05). This combination was not associated with any obvious local or systemic adverse effects. Antibody competition indicated that the majority of the neutralizing activities were directed to the CD4 binding site (CD4bs). Increased antibody titers to the gp41 membrane proximal external region (MPER) and gp120 V3 were detected when the more potent adjuvants were used. These data reveal that the combination of Carbopol-971P and MF59 is unusually potent for eliciting nAbs to a variety of HIV-1 nAb epitopes.
Collapse
Affiliation(s)
- Rachel P. J. Lai
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Michael S. Seaman
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Paul Tonks
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Frank Wegmann
- The Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - David J. Seilly
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Simon D. W. Frost
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Celia C. LaBranche
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David C. Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Antu K. Dey
- Novartis Vaccines and Diagnostics Inc., Massachusetts, United States of America
| | | | - Quentin Sattentau
- The Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Susan W. Barnett
- Novartis Vaccines and Diagnostics Inc., Massachusetts, United States of America
| | - Jonathan L. Heeney
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Dey AK, Burke B, Sun Y, Hartog K, Heeney JL, Montefiori D, Srivastava IK, Barnett SW. Use of a polyanionic carbomer, Carbopol971P, in combination with MF59, improves antibody responses to HIV-1 envelope glycoprotein. Vaccine 2012; 30:2749-59. [PMID: 22366638 DOI: 10.1016/j.vaccine.2012.02.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Revised: 02/09/2012] [Accepted: 02/11/2012] [Indexed: 12/20/2022]
Abstract
Identification of optimal antigen(s) and adjuvant combination(s) to elicit potent, protective, and long-lasting immunity has been a major challenge for the development of effective vaccines against chronic viral pathogens, such as HIV-1, for which there are not yet any licensed vaccines. Here we describe the use of a novel adjuvant approach employing Carbopol 971P(®) NF (hereafter referred to as Carbopol971P), a cross-linked polyanionic carbomer, in combination with the Novartis proprietary oil-in-water adjuvant, MF59, as a potentially safe and effective adjuvant to augment humoral immune responses to the HIV-1 envelope glycoprotein (Env). Intramuscular immunization of small animals with recombinant Env glycoprotein (gp140) formulated in Carbopol971P plus MF59 gave significantly higher titers of binding and virus neutralizing antibodies as compared to immunization using gp140 with either MF59 or Carbopol971P alone. In addition, the antibodies generated were of higher avidity. Importantly, the use of Carbopol971P plus MF59 did not cause any serious adverse reactions or any obvious health problems in animals upon intramuscular administration. Hence, the Carbopol971P plus MF59 adjuvant formulation may provide a benefit for future vaccine applications.
Collapse
Affiliation(s)
- Antu K Dey
- Novartis Vaccines & Diagnostics, 45 Sidney Street, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Ahmed FK, Clark BE, Burton DR, Pantophlet R. An engineered mutant of HIV-1 gp120 formulated with adjuvant Quil A promotes elicitation of antibody responses overlapping the CD4-binding site. Vaccine 2012; 30:922-30. [PMID: 22142583 PMCID: PMC3733221 DOI: 10.1016/j.vaccine.2011.11.089] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/20/2011] [Accepted: 11/22/2011] [Indexed: 01/08/2023]
Abstract
A major priority in HIV vaccine research is the development of an immunogen to elicit broadly neutralizing antibodies (NAbs). Monoclonal antibody (mAb) b12 is one of now several broadly neutralizing mAbs that bind epitopes overlapping the CD4-binding site (CD4bs) on HIV-1 gp120 and that serve as templates to engineer effective immunogens. We are exploring a strategy whereby extra glycans are incorporated onto gp120 to occlude the epitopes of non-neutralizing mAbs while maintaining exposure of the b12 site. Immunizing with these so-called hyperglycosylated gp120s is hypothesized to preferentially elicit b12-like NAbs. Here, the effects of two adjuvants, monophosphoryl lipid A (MPL) and Quil A, on eliciting b12-like responses when formulated with a new hyperglycosylated mutant, ΔN2mCHO(Q105N), is presented. Sera from ΔN2mCHO(Q105N)_MPL immunized animals bound the homologous antigen ΔN2mCHO(Q105N) with greater preference than sera from ΔN2mCHO(Q105N)_QuilA immunized animals, demonstrating the modulation of antibody fine specificity by these two adjuvants. We also found that sera from ΔN2mCHO(Q105N)_QuilA immunized animals bound best to a resurfaced HIV gp120 core protein on which non-CD4bs epitopes are substituted with non-HIV residues, suggesting that these sera contain a relatively larger fraction of CD4bs-specific antibodies. Consistent with these data, inhibition assays revealed epitope overlap with the binding sites of the CD4bs-specific antibodies b12, b13 and VRC03. Unexpectedly, these sera did not exhibit significant neutralizing activity against a set of HIV-1 primary strains. Our results show that although formulating mutant ΔN2mCHO(Q105N) with Quil A promotes the elicitation of CD4bs-directed antibodies relative to wild-type gp120, tweaking of the immunization regimen is needed to yield robust, CD4bs-focused NAbs.
Collapse
Affiliation(s)
- Fatima K. Ahmed
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A1S6, Canada
| | - Brenda E. Clark
- Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A1S6, Canada
| | - Dennis R. Burton
- Department of Immunology and Microbial Science, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Boston, MA 02129, USA
| | - Ralph Pantophlet
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A1S6, Canada
- Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A1S6, Canada
| |
Collapse
|
42
|
Greenwood B, Salisbury D, Hill AVS. Vaccines and global health. Philos Trans R Soc Lond B Biol Sci 2011; 366:2733-42. [PMID: 21893534 DOI: 10.1098/rstb.2011.0076] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Vaccines have made a major contribution to global health in recent decades but they could do much more. In November 2011, a Royal Society discussion meeting, 'New vaccines for global health', was held in London to discuss the past contribution of vaccines to global health and to consider what more could be expected in the future. Papers presented at the meeting reviewed recent successes in the deployment of vaccines against major infections of childhood and the challenges faced in developing vaccines against some of the world's remaining major infectious diseases such as human immunodeficiency virus (HIV), malaria and tuberculosis. The important contribution that development of more effective veterinary vaccines could make to global health was also addressed. Some of the social and financial challenges to the development and deployment of new vaccines were reviewed. The latter issues were also discussed at a subsequent satellite meeting, 'Accelerating vaccine development', held at the Kavli Royal Society International Centre. Delegates at this meeting considered challenges to the more rapid development and deployment of both human and veterinary vaccines and how these might be addressed. Papers based on presentations at the discussion meeting and a summary of the main conclusions of the satellite meeting are included in this issue of Philosophical Transactions of the Royal Society B.
Collapse
Affiliation(s)
- Brian Greenwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| | | | | |
Collapse
|
43
|
Abstract
The implementation of vaccination as an empiric strategy to protect against infectious diseases was introduced even before the advent of hygiene and antimicrobials in the medical practice. Nevertheless, it was not until a few decades ago that we really started understanding the underlying mechanisms of protection triggered by vaccination. Vaccines were initially based on attenuated or inactivated organisms. Subunit vaccines were then introduced as more refined formulations, exhibiting improved safety profiles. However, purified antigens tend to be poorly immunogenic and often require the use of adjuvants to achieve adequate stimulation of the immune system. Vaccination strategies, such as mucosal administration, also require potent adjuvants to improve performance. In the 1990s, immunologists found that pathogens could be sensed as ‘danger signals’ by receptors recognizing conserved motifs. Although our knowledge is still limited, tremendous advances were made in the understanding of host defence mechanisms regulated by these evolutionary conserved receptors, and the molecular structures which are recognized by them. This opened a new era in adjuvant development. Some of the latest players arrived to this field are the cyclic di‐nucleotides, which are ubiquitous prokaryotic intracellular signalling molecules. This review is focused on their potential for the development of vaccines and immunotherapies.
Collapse
Affiliation(s)
- Rimma Libanova
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany.
| | | | | |
Collapse
|