1
|
Chippaux JP. Gaston Ramon's Big Four. Toxins (Basel) 2024; 16:33. [PMID: 38251249 PMCID: PMC10819242 DOI: 10.3390/toxins16010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
When immunology was still in its infancy, Gaston Ramon made several major contributions to humoral immunology [...].
Collapse
Affiliation(s)
- Jean-Philippe Chippaux
- MERIT, Institut de Recherche pour le Développement, Université Paris Cité, F-75006 Paris, France
| |
Collapse
|
2
|
Garcia-Vello P, Di Lorenzo F, Zucchetta D, Zamyatina A, De Castro C, Molinaro A. Lipopolysaccharide lipid A: A promising molecule for new immunity-based therapies and antibiotics. Pharmacol Ther 2022; 230:107970. [PMID: 34454000 DOI: 10.1016/j.pharmthera.2021.107970] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/24/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022]
Abstract
Lipopolysaccharides (LPS) are the main components of the external leaflet of the Gram-negative outer membrane and consist of three different moieties: lipid A, core oligosaccharide, and O-polysaccharide. The lipid A is a glucosamine disaccharide with different levels of acylation and phosphorylation, beside carrying, in certain cases, additional substituents on the sugar backbone. It is also the main immunostimulatory part of the LPS, as its recognition by the host immune system represents a fundamental event for detection of perilous microorganisms. Moreover, an uncontrolled immune response caused by a large amount of circulating LPS can lead to dramatic outcomes for human health, such as septic shock. The immunostimulant properties of an LPS incredibly vary depending on lipid A chemical structure, and for this reason, natural and synthetic variants of the lipid A are under study to develop new drugs that mimic or antagonise its natural effects. Here, we review past and recent findings on the lipid A as an antibiotic target and immune-therapeutic molecule, with a special attention on the crucial role of the chemical structure and its exploitation for conceiving novel strategies for treatment of several immune-related pathologies.
Collapse
Affiliation(s)
- Pilar Garcia-Vello
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy.
| | - Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy
| | - Daniele Zucchetta
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Cristina De Castro
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy.
| |
Collapse
|
3
|
Pan Y, Tang W, Fan W, Zhang J, Chen X. Development of nanotechnology-mediated precision radiotherapy for anti-metastasis and radioprotection. Chem Soc Rev 2022; 51:9759-9830. [DOI: 10.1039/d1cs01145f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radiotherapy (RT), including external beam RT and internal radiation therapy, uses high-energy ionizing radiation to kill tumor cells.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Wei Tang
- Departments of Pharmacy and Diagnostic Radiology, Nanomedicine Translational Research Program, Faculty of Science and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
4
|
Garcia-Vello P, Speciale I, Chiodo F, Molinaro A, De Castro C. Carbohydrate-based adjuvants. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 35-36:57-68. [PMID: 33388128 DOI: 10.1016/j.ddtec.2020.09.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/28/2020] [Accepted: 09/25/2020] [Indexed: 06/12/2023]
Abstract
Carbohydrate adjuvants are safe and biocompatible compounds usable as sustained delivery systems and stimulants of ongoing humoral and cellular immune responses, being especially suitable for the development of vaccines against intracellular pathogens where alum is useless. The development of new adjuvants is difficult and expensive, however, in the last two years, seven new carbohydrate-based adjuvants have been patented, also there are twelve ongoing clinical trials of vaccines that contain carbohydrate-based adjuvants, as well as numerous publications on their mechanism of action and safety. More research is necessary to improve the existent adjuvants and develop innovative ones.
Collapse
Affiliation(s)
- Pilar Garcia-Vello
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Naples (NA), Italy.
| | - Immacolata Speciale
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Naples (NA), Italy
| | - Fabrizio Chiodo
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Naples (NA), Italy
| | - Cristina De Castro
- Department of Agricultural Sciences, University of Naples Federico II, Via Università, 100, 80055 Portici (NA), Italy.
| |
Collapse
|
5
|
Lee JE, Kye YC, Park SM, Shim BS, Yoo S, Hwang E, Kim H, Kim SJ, Han SH, Park TS, Park BC, Yun CH. Bacillus subtilis spores as adjuvants against avian influenza H9N2 induce antigen-specific antibody and T cell responses in White Leghorn chickens. Vet Res 2020; 51:68. [PMID: 32448402 PMCID: PMC7245620 DOI: 10.1186/s13567-020-00788-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/16/2020] [Indexed: 01/06/2023] Open
Abstract
Low-pathogenicity avian influenza H9N2 remains an endemic disease worldwide despite continuous vaccination, indicating the need for an improved vaccine strategy. Bacillus subtilis (B. subtilis), a gram-positive and endospore-forming bacterium, is a non-pathogenic species that has been used in probiotic formulations for both animals and humans. The objective of the present study was to elucidate the effect of B. subtilis spores as adjuvants in chickens administered inactivated avian influenza virus H9N2. Herein, the adjuvanticity of B. subtilis spores in chickens was demonstrated by enhancement of H9N2 virus-specific IgG responses. B. subtilis spores enhanced the proportion of B cells and the innate cell population in splenocytes from chickens administered both inactivated H9N2 and B. subtilis spores (Spore + H9N2). Furthermore, the H9N2 and spore administration induced significantly increased expression of the pro-inflammatory cytokines IL-1β and IL-6 compared to that in the H9N2 only group. Additionally, total splenocytes from chickens immunized with inactivated H9N2 in the presence or absence of B. subtilis spores were re-stimulated with inactivated H9N2. The subsequent results showed that the extent of antigen-specific CD4+ and CD8+ T cell proliferation was higher in the Spore + H9N2 group than in the group administered only H9N2. Taken together, these data demonstrate that B. subtilis spores, as adjuvants, enhance not only H9N2 virus-specific IgG but also CD4+ and CD8+ T cell responses, with an increase in pro-inflammatory cytokine production. This approach to vaccination with inactivated H9N2 together with a B. subtilis spore adjuvant in chickens produces a significant effect on antigen-specific antibody and T cell responses against avian influenza virus.
Collapse
Affiliation(s)
- Ji Eun Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon-Chul Kye
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung-Moo Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea.,Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | | | - Sungsik Yoo
- Choong-Ang Vaccine Laboratory, Daejeon, Republic of Korea
| | - Eunmi Hwang
- Department of Biotechnology, Hoseo University, Asan, Chungcheongnam-do, Republic of Korea
| | - Hyungkuen Kim
- Department of Biotechnology, Hoseo University, Asan, Chungcheongnam-do, Republic of Korea
| | - Sung-Jo Kim
- Department of Biotechnology, Hoseo University, Asan, Chungcheongnam-do, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, DRI and BK21 Program, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Tae Sub Park
- Graduate School of International Agricultural Technology, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Republic of Korea
| | - Byung-Chul Park
- Graduate School of International Agricultural Technology, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Republic of Korea.
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea. .,Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea. .,Graduate School of International Agricultural Technology, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Republic of Korea.
| |
Collapse
|
6
|
Singh B, Maharjan S, Sindurakar P, Cho KH, Choi YJ, Cho CS. Needle-Free Immunization with Chitosan-Based Systems. Int J Mol Sci 2018; 19:E3639. [PMID: 30463211 PMCID: PMC6274840 DOI: 10.3390/ijms19113639] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/10/2018] [Accepted: 11/12/2018] [Indexed: 02/02/2023] Open
Abstract
Despite successful use, needle-based immunizations have several issues such as the risk of injuries and infections from the reuse of needles and syringes and the low patient compliance due to pain and fear of needles during immunization. In contrast, needle-free immunizations have several advantages including ease of administration, high level of patient compliance and the possibility of mass vaccination. Thus, there is an increasing interest on developing effective needle-free immunizations via cutaneous and mucosal approaches. Here, we discuss several methods of needle-free immunizations and provide insights into promising use of chitosan systems for successful immunization.
Collapse
Affiliation(s)
- Bijay Singh
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
- Research Institute for Bioscience and Biotechnology, Kathmandu 44600, Nepal.
| | - Sushila Maharjan
- Research Institute for Bioscience and Biotechnology, Kathmandu 44600, Nepal.
- Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| | - Princy Sindurakar
- Department of Biology, College of the Holy Cross, Worcester, MA 01610, USA.
| | - Ki-Hyun Cho
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
7
|
Hossain MK, Vartak A, Karmakar P, Sucheck SJ, Wall KA. Augmenting Vaccine Immunogenicity through the Use of Natural Human Anti-rhamnose Antibodies. ACS Chem Biol 2018; 13:2130-2142. [PMID: 29916701 PMCID: PMC6103300 DOI: 10.1021/acschembio.8b00312] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Utilizing
natural antibodies to augment vaccine immunogenicity
is a promising approach toward cancer immunotherapy. Anti-rhamnose
(anti-Rha) antibodies are some of the most common natural anti-carbohydrate
antibodies present in human serum. Therefore, rhamnose can be utilized
as a targeting moiety for a rhamnose-containing vaccine to prepare
an effective vaccine formulation. It was shown previously that anti-Rha
antibody generated in mice binds effectively with Rha-conjugated vaccine
and is picked up by antigen presenting cells (APCs) through stimulatory
Fc receptors. This leads to the effective uptake and processing of
antigen and eventually presentation by major histocompatibility complex
(MHC) molecules. In this article, we show that natural human anti-Rha
antibodies can also be used in a similar mechanism and immunogenicity
can be enhanced by targeting Rha-conjugated antigens. In doing so,
we have purified human anti-Rha antibodies from human serum using
a rhamnose affinity column. In vitro, human anti-Rha
antibodies are shown to enhance the uptake of a model antigen, Rha-ovalbumin
(Rha-Ova), by APCs. In vivo, they improved the priming
of CD4+ T cells to Rha-Ova in comparison to non-anti-Rha human antibodies.
Additionally, increased priming of both CD4+ and CD8+ T cells toward
the cancer antigen MUC1-Tn was observed in mice that received human
anti-Rha antibodies prior to vaccination with a rhamnose-modified
MUC1-Tn cancer vaccine. The vaccine conjugate contained Pam3CysSK4, a Toll-like receptor (TLR) agonist linked via copper-free cycloaddition chemistry to a 20-amino-acid
glycopeptide derived from the tumor marker MUC-1 containing the tumor-associated
carbohydrate antigen α-N-acetyl galactosamine
(GalNAc). The primed CD8+ T cells released IFN-γ and killed
tumor cells. Therefore, we have confirmed that human anti-Rha antibodies
can be effectively utilized as a targeting moiety for making an effective
vaccine.
Collapse
Affiliation(s)
- Md Kamal Hossain
- Dept. of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Abhishek Vartak
- Dept. of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Partha Karmakar
- Dept. of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Steven J. Sucheck
- Dept. of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Katherine A. Wall
- Dept. of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43606, United States
| |
Collapse
|
8
|
Vartak A, Sucheck SJ. Recent Advances in Subunit Vaccine Carriers. Vaccines (Basel) 2016; 4:vaccines4020012. [PMID: 27104575 PMCID: PMC4931629 DOI: 10.3390/vaccines4020012] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/28/2016] [Accepted: 04/01/2016] [Indexed: 12/11/2022] Open
Abstract
The lower immunogenicity of synthetic subunit antigens, compared to live attenuated vaccines, is being addressed with improved vaccine carriers. Recent reports indicate that the physio-chemical properties of these carriers can be altered to achieve optimal antigen presentation, endosomal escape, particle bio-distribution, and cellular trafficking. The carriers can be modified with various antigens and ligands for dendritic cells targeting. They can also be modified with adjuvants, either covalently or entrapped in the matrix, to improve cellular and humoral immune responses against the antigen. As a result, these multi-functional carrier systems are being explored for use in active immunotherapy against cancer and infectious diseases. Advancing technology, improved analytical methods, and use of computational methodology have also contributed to the development of subunit vaccine carriers. This review details recent breakthroughs in the design of nano-particulate vaccine carriers, including liposomes, polymeric nanoparticles, and inorganic nanoparticles.
Collapse
Affiliation(s)
- Abhishek Vartak
- Department of Chemistry and Biochemistry, The University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA.
| | - Steven J Sucheck
- Department of Chemistry and Biochemistry, The University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA.
| |
Collapse
|
9
|
Yang A, Jeang J, Cheng K, Cheng T, Yang B, Wu TC, Hung CF. Current state in the development of candidate therapeutic HPV vaccines. Expert Rev Vaccines 2016; 15:989-1007. [PMID: 26901118 DOI: 10.1586/14760584.2016.1157477] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The identification of human papillomavirus (HPV) as an etiological factor for HPV-associated malignancies creates the opportunity to control these cancers through vaccination. Currently, available preventive HPV vaccines have not yet demonstrated strong evidences for therapeutic effects against established HPV infections and lesions. Furthermore, HPV infections remain extremely common. Thus, there is urgent need for therapeutic vaccines to treat existing HPV infections and HPV-associated diseases. Therapeutic vaccines differ from preventive vaccines in that they are aimed at generating cell-mediated immunity rather than neutralizing antibodies. The HPV-encoded early proteins, especially oncoproteins E6 and E7, form ideal targets for therapeutic HPV vaccines since they are consistently expressed in HPV-associated malignancies and precancerous lesions, playing crucial roles in the generation and maintenance of HPV-associated disease. Our review will cover various therapeutic vaccines in development for the treatment of HPV-associated lesions and cancers. Furthermore, we review strategies to enhance vaccine efficacy and the latest clinical trials on therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Andrew Yang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Jessica Jeang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Kevin Cheng
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Ting Cheng
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Benjamin Yang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - T-C Wu
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA.,b Department of Obstetrics and Gynecology , Johns Hopkins University , Baltimore , MD , USA.,c Department of Molecular Microbiology and Immunology , Johns Hopkins University , Baltimore , MD , USA.,d Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| | - Chien-Fu Hung
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA.,d Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
10
|
Powles L, Xiang SD, Selomulya C, Plebanski M. The Use of Synthetic Carriers in Malaria Vaccine Design. Vaccines (Basel) 2015; 3:894-929. [PMID: 26529028 PMCID: PMC4693224 DOI: 10.3390/vaccines3040894] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/28/2015] [Accepted: 10/16/2015] [Indexed: 11/29/2022] Open
Abstract
Malaria vaccine research has been ongoing since the 1980s with limited success. However, recent improvements in our understanding of the immune responses required to combat each stage of infection will allow for intelligent design of both antigens and their associated delivery vaccine vehicles/vectors. Synthetic carriers (also known as vectors) are usually particulate and have multiple properties, which can be varied to control how an associated vaccine interacts with the host, and consequently how the immune response develops. This review comprehensively analyzes both historical and recent studies in which synthetic carriers are used to deliver malaria vaccines. Furthermore, the requirements for a synthetic carrier, such as size, charge, and surface chemistry are reviewed in order to understand the design of effective particle-based vaccines against malaria, as well as providing general insights. Synthetic carriers have the ability to alter and direct the immune response, and a better control of particle properties will facilitate improved vaccine design in the near future.
Collapse
Affiliation(s)
- Liam Powles
- Department of Chemical Engineering, Monash University, Clayton, VIC 3800, Australia.
| | - Sue D Xiang
- Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia.
- Therapeutics and Regenerative Medicine Division, The Monash Institute of Medical Engineering (MIME), Monash University, Clayton, VIC 3800, Australia.
| | - Cordelia Selomulya
- Department of Chemical Engineering, Monash University, Clayton, VIC 3800, Australia.
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia.
- Therapeutics and Regenerative Medicine Division, The Monash Institute of Medical Engineering (MIME), Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
11
|
Uchegbu IF, Schätzlein AG, Cheng WP, Lalatsa A. Vaccines. FUNDAMENTALS OF PHARMACEUTICAL NANOSCIENCE 2013. [PMCID: PMC7120629 DOI: 10.1007/978-1-4614-9164-4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vaccines continue to offer the key line of protection against a range of infectious diseases; however, the range of vaccines currently available is limited. One key consideration in the development of a vaccine is risk-versus-benefit, and in an environment of perceived low risk, the benefit of vaccination may not be recognised. To address this, there has been a move towards the use of subunit-based vaccines, which offer low side-effect profiles but are generally weakly immunogenic. This can be compensated for by the development of effective adjuvants. Nanotechnology offers key attributes in this field through the ability of nanoparticulates to incorporate and protect antigens from rapid degradation, combined with their potential to effectively deliver the antigens to appropriate cells within the immune system. These characteristics can be exploited in the development of new adjuvants. This chapter will outline the applications of nanosystems in vaccine formulations and consider the mechanisms of action behind a range of formulations.
Collapse
Affiliation(s)
- Ijeoma F. Uchegbu
- UCL School of Pharmacy, University College London, London, United Kingdom
| | | | | | - Aikaterini Lalatsa
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
12
|
Mori J, Vranac T, Smrekar B, Cernilec M, Serbec VČ, Horvat S, Ihan A, Benčina M, Jerala R. Chimeric flagellin as the self-adjuvanting antigen for the activation of immune response against Helicobacter pylori. Vaccine 2012; 30:5856-63. [PMID: 22819990 DOI: 10.1016/j.vaccine.2012.07.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/19/2012] [Accepted: 07/08/2012] [Indexed: 12/11/2022]
Abstract
Helicobacter pylori infection can cause gastritis, peptic ulcer and can lead to gastric cancer. Lengthy antibiotic therapy does not protect the host against reinfection. H. pylori evolved to evade the recognition of the immune response by modifying several of its components whose orthologous proteins from other bacteria activate the innate immune response. Flagella are essential for the H. pylori effective colonization of human duodenum and stomach. TLR5, a member of the Toll-like receptor family, recognizes flagellin of most bacteria, such as Escherichia coli, but does not recognize the flagellin FlaA of H. pylori. We restored the ability of FlaA for the recognition by TLR5 by engineering a chimeric flagellin, in which both terminal segments of H. pylori flagellin were replaced by the corresponding segments from TLR5-activating E. coli flagellin. Recombinant chimeric flagellin folded correctly and was able to activate TLR5. Significantly increased serum IgG and IgA antibody responses were determined in mice vaccinated with chimeric flagellin in comparison to mice vaccinated with a control protein (FlaA) or negative control. Antibody titers remained high even 8 months after the last immunization. Antibodies were able to bind native flagellin from H. pylori lysate. Vaccination with chimeric flagellin provided mice with significant protection against H. pylori. The approach of chimeric flagellin can therefore generate effective immunogens that enable activation of innate and adaptive immune response and can be used to construct efficient vaccines against H. pylori or other flagellated bacteria that evade TLR5 recognition.
Collapse
Affiliation(s)
- Jerneja Mori
- Department of Biotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Kumar HS, Singh PP, Qazi NA, Srinivas J, Malik F, Sidiq T, Gupta A, Khajuria A, Suri K, Satti N, Qazi G. Development of novel lipidated analogs of picroside as vaccine adjuvants: Acylated analogs of picroside-II elicit strong Th1 and Th2 response to ovalbumin in mice. Vaccine 2010; 28:8327-37. [DOI: 10.1016/j.vaccine.2010.07.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 06/05/2010] [Accepted: 07/21/2010] [Indexed: 11/16/2022]
|
14
|
Sundling C, Schön K, Mörner A, Forsell MNE, Wyatt RT, Thorstensson R, Hedestam GBK, Lycke NY. CTA1-DD adjuvant promotes strong immunity against human immunodeficiency virus type 1 envelope glycoproteins following mucosal immunization. J Gen Virol 2009; 89:2954-2964. [PMID: 19008380 DOI: 10.1099/vir.0.2008/005470-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Strategies to induce potent and broad antibody responses against the human immunodeficiency virus type 1 (HIV-1) envelope glycoproteins (Env) at both systemic and mucosal sites represent a central goal for HIV-1 vaccine development. Here, we show that the non-toxic CTA1-DD adjuvant promoted mucosal and systemic humoral and cell-mediated immune responses following intranasal (i.n.) immunizations with trimeric or monomeric forms of HIV-1 Env in mice and in non-human primates. Env-specific IgG subclasses in the serum of immunized mice reflected a balanced Th1/Th2 type of response. Strikingly, i.n. immunizations with Env and the CTA1-DD adjuvant induced substantial levels of mucosal anti-Env IgA in bronchial alveolar lavage and also detectable levels in vaginal secretions. By contrast, parenteral immunizations of Env formulated in Ribi did not stimulate mucosal IgA responses, while the two adjuvants induced a similar distribution of Env-specific IgG-subclasses in serum. A single parenteral boost with Env in Ribi adjuvant into mice previously primed i.n. with Env and CTA1-DD, augmented the serum anti-Env IgG levels to similar magnitudes as those observed after three intraperitoneal immunizations with Env in Ribi. The augmenting potency of CTA1-DD was similar to that of LTK63 or CpG oligodeoxynucleotides (ODN). However, in contrast to CpG ODN, the effect of CTA1-DD and LTK63 appeared to be independent of MyD88 and toll-like receptor signalling. This is the first demonstration that CTA1-DD augments specific immune responses also in non-human primates, suggesting that this adjuvant could be explored further as a clinically safe mucosal vaccine adjuvant for humoral and cell-mediated immunity against HIV-1 Env.
Collapse
Affiliation(s)
- Christopher Sundling
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden.,Swedish Institute for Infectious Disease Control, 171 82 Solna, Sweden
| | - Karin Schön
- Mucosal Immunobiology & Vaccine Center (MIVAC), Department of Microbiology & Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Mörner
- Swedish Institute for Infectious Disease Control, 171 82 Solna, Sweden
| | - Mattias N E Forsell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Richard T Wyatt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Gunilla B Karlsson Hedestam
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden.,Swedish Institute for Infectious Disease Control, 171 82 Solna, Sweden
| | - Nils Y Lycke
- Mucosal Immunobiology & Vaccine Center (MIVAC), Department of Microbiology & Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
15
|
Midha S, Bhatnagar R. Anthrax protective antigen administered by DNA vaccination to distinct subcellular locations potentiates humoral and cellular immune responses. Eur J Immunol 2009; 39:159-77. [PMID: 19130551 DOI: 10.1002/eji.200838058] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Based on the hypothesis that immune outcome can be influenced by the form of antigen administered and its ability to access various antigen-processing pathways, we targeted the 63 kDa fragment of protective antigen (PA) of Bacillus anthracis to various subcellular locations by DNA chimeras bearing a set of signal sequences. These targeting signals, namely, lysosome-associated membrane protein 1 (LAMP1), tissue plasminogen activator (TPA) and ubiquitin, encoded various forms of PA viz. lysosomal, secreted and cytosolic, respectively. Examination of IgG subclass distribution arising as a result of DNA vaccination indicated a higher IgG1:IgG2a ratio whenever the groups were immunized with chimeras bearing TPA, LAMP1 signals alone or when combined together. Importantly, high end-point titers of IgG antibodies were maintained until 24 wk. It was paralleled by high avidity toxin neutralizing antibodies (TNA) and effective cellular adaptive immunity in the systemic compartment. Anti-PA and TNA titers of approximately 10(5) and approximately 10(3), respectively, provided protection to approximately 90% of vaccinated animals in the group pTPA-PA63-LAMP1. A significant correlation was found between survival percentage and post-challenge anti-PA titers and TNA titers. Overall, immune kinetics pointed that differential processing through various compartments gave rise to qualitative differences in the immune response generated by various chimeras.
Collapse
Affiliation(s)
- Shuchi Midha
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | | |
Collapse
|
16
|
Rajput ZI, Hu SH, Xiao CW, Arijo AG. Adjuvant effects of saponins on animal immune responses. J Zhejiang Univ Sci B 2007; 8:153-61. [PMID: 17323426 PMCID: PMC1810383 DOI: 10.1631/jzus.2007.b0153] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2006] [Accepted: 05/26/2006] [Indexed: 11/11/2022]
Abstract
Vaccines require optimal adjuvants including immunopotentiator and delivery systems to offer long term protection from infectious diseases in animals and man. Initially it was believed that adjuvants are responsible for promoting strong and sustainable antibody responses. Now it has been shown that adjuvants influence the isotype and avidity of antibody and also affect the properties of cell-mediated immunity. Mostly oil emulsions, lipopolysaccharides, polymers, saponins, liposomes, cytokines, ISCOMs (immunostimulating complexes), Freund's complete adjuvant, Freund's incomplete adjuvant, alums, bacterial toxins etc., are common adjuvants under investigation. Saponin based adjuvants have the ability to stimulate the cell mediated immune system as well as to enhance antibody production and have the advantage that only a low dose is needed for adjuvant activity. In the present study the importance of adjuvants, their role and the effect of saponin in immune system is reviewed.
Collapse
Affiliation(s)
- Zahid Iqbal Rajput
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310029, China
| | - Song-hua Hu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310029, China
| | - Chen-wen Xiao
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310029, China
| | - Abdullah G. Arijo
- Department of Parasitology, Sindh Agriculture University, Tando Jam 70060, Pakistan
| |
Collapse
|
17
|
Kalkanidis M, Pietersz GA, Xiang SD, Mottram PL, Crimeen-Irwin B, Ardipradja K, Plebanski M. Methods for nano-particle based vaccine formulation and evaluation of their immunogenicity. Methods 2006; 40:20-9. [PMID: 16997710 DOI: 10.1016/j.ymeth.2006.05.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 05/12/2006] [Indexed: 11/19/2022] Open
Abstract
Nano- and microparticles have long been used for the delivery of drugs and are currently being evaluated as vaccine delivery systems. Particulates can elicit potent immune responses, either by direct immuno-stimulation of antigen presenting cells (APC) or/and by delivering antigen to specific cellular compartments and promoting antigen uptake by appropriate stimulatory cell types. Herein, we describe a detailed method for the preparation of a novel nanoparticle-based antigen delivery system which induces strong cellular and humoral immune responses in mice and sheep. This simple system is based on the use of 40 nanometer (nm) inert solid carrier beads to which antigen is covalently coupled before injection. Covalent conjugation of antigen to the nanobeads, assessment of conjugation efficiency, characterisation and measurement of in vivo immunogenicity by cytokine ELISPOT (to measure antigen-specific T-cell responses) and ELISA (to measure antibody titers), are described. Emphasis is placed on providing trouble-shooting advice to enable the reproducible production of soluble nano-size formulations that do not suffer from common problems such as aggregation, as well as understanding the causes and thus avoiding a range of prevalent technical problems that occur when using immune response detection assays, such as the cytokine ELISPOT assay and ELISA.
Collapse
Affiliation(s)
- Martha Kalkanidis
- Vaccine and Infectious Diseases Laboratory, Burnet Institute at Austin, Studley Road, Heidelberg, Vic. 3084, Australia
| | | | | | | | | | | | | |
Collapse
|
18
|
Lycke N. ADP-ribosylating bacterial enzymes for the targeted control of mucosal tolerance and immunity. Ann N Y Acad Sci 2005; 1029:193-208. [PMID: 15681758 DOI: 10.1196/annals.1309.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The questions of whether mucosal tolerance and IgA immunity are mutually exclusive or can coexist and whether they represent priming of the local immune system through the same or different activation pathways are addressed. Two strategies were attempted: the first using cholera toxin (CT) or the enzymatically inactive receptor-binding B subunit of CT (CTB), and the second using CTA1-DD or an enzymatically inactive mutant thereof, CTA1R7K-DD. The CTA1-DD adjuvant is a fusion protein composed of the ADP-ribosylating part of CT, CTA1, and DD, which is derived from Staphylococcus areus protein A and targets the molecule to B cells. Here, we provide compelling evidence that delivery of antigen in the absence of ADP ribosylation can promote tolerance, whereas ADP-ribosyltransferase activity induces IgA immunity and prevents tolerance. By linking antigen to the ADP-ribosylating enzymes we could show that CT, although potentially binding to all nucleated cells, in fact, bound preferentially to dendritic cells (DCs) in vivo. On the other hand, DD-bound antigen was distinctly targeted to B cells and probably also to follicular dendritic cells (FDCs) in vivo. Interestingly, the CT and CTA1-DD adjuvants gave equally enhancing effects on mucosal and systemic responses, but appeared to target different APCs in vivo. CT- or CTB-conjugated antigen accumulated in mucosal and systemic DCs. Whereas only CT promoted an active IgA response, CTB induced tolerance to the conjugated antigen. Following intravenous injection of CT-conjugated antigen, DCs in the marginal zone (MZ) of the spleen were selectively targeted. Interestingly, CTB delivered antigen to the same MZ DCs, but failed to induce maturation and upregulation of costimulatory molecules in these cells. Thus, ADP-ribosylation was necessary for a strong enhancing effect of immune responses following CT/CTB-dependent delivery of antigen to the MZ DCs. Moreover, using CTA1-DD, antigen was targeted to the B cell follicle and FDC in the spleen after intravenous injection. Only active CTA1-DD, but not the inactive mutant CTA1R7K-DD, provided enhancing effects on immune responses. By contrast, antigen delivered by the CTA1R7K-DD stimulated specific tolerance in adoptively transferred T cell receptor transgenic CD4(+) T cells. Whether targeting of B cells suffices for tolerance induction or requires participation of DCs remains to be investigated. With CT we found that enzyme-dependent modulation of DCs affects migration, maturation, and differentiation of DCs, which resulted in CD4(+) T cell help for IgA B cell development. On the contrary, antigen presentation in the absence of ADP-ribosylating enzyme, as seen with CTB or CTA1R7K-DD, appears to expand specific T cells to a similar extent as enzymatically active CT or CTA1-DD, but fails to recruit help for germinal center (GC) formation and the necessary expansion of activated B cells. Also, the CD41 T cells that are primed in a suboptimal, tolerogenic, fashion do not migrate to the B cell follicle to provide T cell help. Thus, ADP-ribosylating enzymes may be used to selectively control the induction of an active IgA response or promote the development of tolerance. In particular, on the targeted APC, modulation of the expression of costimulatory molecules, CD80, CD86, CD83, and B7RP-1, plays an important role in the effect of the ADP-ribosylating CTA1-based adjuvants on the development of tolerance or active IgA immunity. For example, the expression of CD86 in vivo was a prominent feature of the enzymatically active CT or CTA1-DD adjuvants. By contrast, CD80 expression appeared not to be important in CTA1-augmented APCs for an adjuvant function.
Collapse
Affiliation(s)
- Nils Lycke
- Department of Clinical Immunology, University of Göteborg, S413 46 Göteborg, Sweden.
| |
Collapse
|
19
|
Lycke N. From toxin to adjuvant: basic mechanisms for the control of mucosal IgA immunity and tolerance. Immunol Lett 2005; 97:193-8. [PMID: 15752558 DOI: 10.1016/j.imlet.2004.12.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2004] [Revised: 11/27/2004] [Accepted: 11/30/2004] [Indexed: 12/01/2022]
Abstract
We provide compelling evidence that delivery of Ag in the absence of ADP-ribosylation can promote tolerance, whereas ADP-ribosyltransferase activity induces IgA immunity and prevents tolerance. By linking Ag to the ADP-ribosylating enzyme, cholera toxin subunit A1 (CTA1), we could show that the combination of targeting to antigen-presenting cells (APC) and enzymatic activity is a highly effective means of controlling the induction of tolerance or immunity. Firstly, we demonstrated that cholera toxin (CT), although potentially binding to all nucleated cells, in fact, bound preferentially to dendritic cells (DC) in vivo. Following injection of CT-conjugated Ag, we found that DC in the marginal zone (MZ) of the spleen accumulated Ag, a process that was GM1-ganglioside receptor dependent. Contrary to CTB, which also delivered Ag to the MZ DC, CT matured and activated co-stimulatory functions in the targeted DC and greatly augmented immune responses to Ag. Secondly, when Ag was incorporated into the CTA1-DD fusion protein, which equals the CT in adjuvant function but lacks GM1-ganglioside-binding ability, we greatly augmented specific responses to Ag. The DD-bound Ag was distinctly targeted to B cells and probably also to follicular dendritic cells (FDC) in vivo. Thus, in both constructs Ag was targeted to APC and associated with an ADP-ribosylating enzyme, which resulted in greatly enhanced immunogenicity. When the enzymatic activity was absent, as in CT B-subunit (CTB) or in the inactive CTA1R7K-DD mutant, Ag largely failed to stimulate an active immune response. Rather, this type of Ag exposure resulted in Ag-specific tolerance, especially when mucosal delivery of Ag was attempted. Therefore, targeting to APC in the absence or presence of the CTA1-enzyme appears to be an effective means to control tolerance and active protective IgA immunity.
Collapse
Affiliation(s)
- Nils Lycke
- Department of Clinical Immunology, University of Göteborg, Guldhedsgatan 10A, S41346 Göteborg, Sweden.
| |
Collapse
|
20
|
Abstract
Mucosally active vaccine adjuvants which will prime a full range of local and systemic immune responses against defined antigenic epitopes are much needed. Cholera toxin (CT) and lipophilic immune stimulating complexes (ISCOMs) containing Quil A can both act as adjuvants for orally administered antigens, but through separate pathways, as evidenced by the dependence on IL-12 for the effect of ISCOMs, whereas CT is independent of this cytokine. Unfortunately the toxicity of CT and recent findings of accumulation of CT in the olfactory nerve and bulb after intranasal administration precludes the clinical use of CT. However, we have been successful in separating the adjuvant and toxic effects of CT, by constructing a gene fusion protein, CTA1-DD, that combines the enzymatically active CTA1-subunit with a B cell targeting moiety, D, derived from Staphylococcus aureus protein A. The present review gives a background to mucosal immunization and the use of -adjuvants in general, followed by a description of a strategy to rationally design a vaccine adjuvant vector that fulfils the criteria of targeting and immunomodulating innate immunity in order to boost a strong adaptive immune response. We have combined CTA1-DD and ISCOMs into a new highly promising vaccine adjuvant vector, CTA1-DD/ISCOMs. The combined vector is immunogenic when given by the subcutaneous, oral or nasal routes, inducing strong cell--mediated and humoral immune responses, including local mucosal IgA. It requires the ADP ribosylating property of the CTA1-enzyme and the effect of the combined vector greatly exceeded the effect of either ISCOMs or CT used alone. Antigens could be incorporated into or just admixed with the new vector. Thus, we have demonstrated that rationally designed vectors consisting of CTA1-DD and ISCOMS may provide a novel strategy for the generation of potent and safe mucosal vaccines.
Collapse
Affiliation(s)
- Nils Lycke
- Department of Clinical Immunology, University of Göteborg, S413 46 Göteborg, Sweden.
| |
Collapse
|
21
|
Mittenbühler K, v d Esche U, Heinevetter L, Bessler WG, Huber M. Lipopeptides: adjuvanticity in conventional and genetic immunization. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2003; 37:193-200. [PMID: 12832125 DOI: 10.1016/s0928-8244(03)00069-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Synthetic lipopeptides derived from the bacterial cell wall component lipoprotein activate B-lymphocytes and macrophages/monocytes in vitro. In vivo they constitute potent immunoadjuvants for a broad range of different antigens and species comparable or superior to Freund's adjuvant. Here, we demonstrate that P(3)CSK(4), representing a highly active lipopentapeptide derivative in vitro, significantly enhances and accelerates the humoral immune response to tetanus toxoid. P(3)CSK(4) could substitute for up to 90% of the antigen without any decrease in the specific IgG level, and the presence of the lipopeptide resulted in a prolonged production of specific IgG in time. Investigations using P(3)CSK(4) as an adjuvant in genetic immunization confirmed earlier data demonstrating that lipopeptides constitute adjuvants for low-immunogenic DNA constructs and/or for application routes resulting in weak immune responses. We monitored a lipopeptide-dependent shift from a Th1-type to Th2-type response, when DNA immunization was followed by i.p. administration of the lipopeptide adjuvant.
Collapse
Affiliation(s)
- K Mittenbühler
- Institut für Molekulare Medizin und Zellforschung, Universitätsklinikum Freiburg, AK Tumorimmunologie/Vakzine, Stefan-Meier-Str. 8, D-79104 Freiburg, Germany.
| | | | | | | | | |
Collapse
|
22
|
Pitt ML, Little SF, Ivins BE, Fellows P, Barth J, Hewetson J, Gibbs P, Dertzbaugh M, Friedlander AM. In vitro correlate of immunity in a rabbit model of inhalational anthrax. Vaccine 2001; 19:4768-73. [PMID: 11535328 DOI: 10.1016/s0264-410x(01)00234-1] [Citation(s) in RCA: 209] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A serological correlate of vaccine-induced immunity was identified in the rabbit model of inhalational anthrax. Animals were inoculated intramuscularly at 0 and 4 weeks with varying doses of Anthrax Vaccine Adsorbed (AVA) ranging from a human dose to a 1:256 dilution in phosphate-buffered saline (PBS). At 6 and 10 weeks, both the quantitative anti-protective antigen (PA) IgG ELISA and the toxin-neutralizing antibody (TNA) assays were used to measure antibody levels to PA. Rabbits were aerosol-challenged at 10 weeks with a lethal dose (84-133 LD(50)) of Bacillus anthracis spores. All the rabbits that received the undiluted and 1:4 dilution of vaccine survived, whereas those receiving the higher dilutions of vaccine (1:16, 1:64 and 1:256) had deaths in their groups. Results showed that antibody levels to PA at both 6 and 10 weeks were significant (P<0.0001) predictors of survival.
Collapse
Affiliation(s)
- M L Pitt
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702-5011, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Affiliation(s)
- S Sasaki
- Department of Bacteriology, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | | | | |
Collapse
|
24
|
McNicholl JM, Bond KB, Ruhadze ER, Olsen MR, Takayama K, Hunter RL. Enhancement of HIV type 1 vaccine immunogenicity by block copolymer adjuvants. I. Induction of high-titer, long-lasting, cross-reactive antibodies of broad isotype. AIDS Res Hum Retroviruses 1998; 14:1457-71. [PMID: 9824324 DOI: 10.1089/aid.1998.14.1457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Improvements in HIV-1 vaccines are urgently needed since many of the available vaccines are weak immunogens. We examined the ability of CRL1005, a novel nonionic block copolymer adjuvant, to improve the immunogenicity of multiple HIV-1 envelope vaccines: six gp120s and single and multiple V3 peptides (MAPs). Formulation of vaccine with adjuvant, as compared with alum or saline, enhanced antibody titer in mice up to 200-fold, with antibody half-lives of >200 days. For most vaccinations, an oil-in-water formulation induced the highest antibody titers; for some antigens, however, particularly single peptides, water-in-oil (w/o) was better. Antigen cross-reactivity was optimized by formulation in w/o, while addition of detoxified lipopolysaccharide enhanced levels of IgG2a and IgG2b. After more than 1 year of observation, no vaccine-related toxicity was observed and emulsified antigen in encapsulated depots was found at immunization sites of w/o-immunized animals. No other adjuvant has been reported to induce such long-lasting antibodies, and the ability of CRL1005 to greatly amplify and qualitatively modify antibody responses suggests that it may be useful in developing improved HIV vaccines for humans.
Collapse
Affiliation(s)
- J M McNicholl
- Immunology Branch, DASLTR, NCID, CDC, Atlanta, Georgia 30333, USA
| | | | | | | | | | | |
Collapse
|
25
|
Lambros MP, Schafer F, Blackstock R, Murphy JW. Liposomes, a potential immunoadjuvant and carrier for a cryptococcal vaccine. J Pharm Sci 1998; 87:1144-8. [PMID: 9724568 DOI: 10.1021/js9704184] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mice immunized with a cryptococcal culture filtrate antigen (CneF) emulsified in complete Freund's adjuvant (CFA) develop an anticryptococcal cell-mediated immune response (CMI). CMI is detected by delayed-type hypersensitivity (DTH) reactions and by enhanced clearance of Cryptococcus neoformans from infected tissues. The objective of this research was to evaluate anticryptococcal DTH reactivity and clearance of cryptococci from groups of mice immunized with CneF encapsulated into liposomes (CneF-liposome) and compare the results to results from mice immunized with CneF-CFA. CBA/J mice were injected subcutaneously with vaccines or control formulations (saline-liposome or saline-CFA). Six days later the mice were footpad tested to assess their DTH response to CneF or the animals were challenged intravenously with 10(5) viable C. neoformans to determine clearance of infection. Clearance was evaluated 7 days later by enumeration of cryptococcal colony forming units (CFU) in lungs, spleens, livers, and brains of the infected mice. The CneF-liposome formulation induced a positive anticryptococcal DTH response and elicited increased clearance of C. neoformans from tissues as compared to mice treated with saline-liposome. Even though the CneF-liposome preparation did not induce as strong of a DTH response or as much protection as did CneF-CFA, our results indicate that liposomes are promising carriers for immunization with cryptococcal antigen and that such immunization can provide some protection to subsequent infection with C. neoformans.
Collapse
Affiliation(s)
- M P Lambros
- College of Pharmacy and Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73190, USA
| | | | | | | |
Collapse
|
26
|
Hamajima K, Sasaki S, Fukushima J, Kaneko T, Xin KQ, Kudoh I, Okuda K. Intranasal administration of HIV-DNA vaccine formulated with a polymer, carboxymethylcellulose, augments mucosal antibody production and cell-mediated immune response. CLINICAL IMMUNOLOGY AND IMMUNOPATHOLOGY 1998; 88:205-10. [PMID: 9714699 DOI: 10.1006/clin.1998.4566] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that intramuscular (i.m.) immunization of DNA vaccine encoding human immunodeficiency virus type 1 (HIV-1)IIIB env and rev genes alone or in combination with appropriate adjuvant induces substantial and enhanced immune response against HIV-1. In the present study, we examined whether a polymer, low-viscosity carboxymethylcellulose sodium salt (CMCS-L), has an adjuvant effect on immune response induced by DNA vaccination. BALB/c mice were immunized with HIV-DNA vaccine formulated with CMCS-L via the intranasal (i.n.) and i.m. routes. The combination with the polymer elicited higher levels of antigen-specific serum IgG and fecal IgA antibodies than DNA vaccine alone. For cell-mediated immunity, HIV-specific delayed-type hypersensitivity response and cytotoxic T lymphocyte activity were measured by the footpad-swelling test and the 51Cr-release assay, respectively. Both were enhanced by the combination with CMCS-L via i.n. and i.m. inoculation. Cytokine analysis in culture media of bulk splenocytes harvested from immunized animals showed higher levels of IL-4 production in i.n. -immunized mice compared with i.m.-immunized mice. Nevertheless, the increased IFN-gamma production resulting from the combination with CMCS-L was observed only in i.n.-immunized mice. These data indicate that i.n. immunization of HIV-DNA vaccine formulated with CMCS-L enhances HIV-specific mucosal antibody (Ab) and systemic Ab and cell-mediated immune response.
Collapse
MESH Headings
- AIDS Vaccines/immunology
- AIDS Vaccines/pharmacology
- Adjuvants, Immunologic/pharmacology
- Administration, Intranasal
- Animals
- Antibodies/blood
- Carboxymethylcellulose Sodium/pharmacology
- Cells, Cultured
- Female
- HIV/genetics
- HIV/immunology
- Hypersensitivity, Delayed
- Immunity, Cellular/drug effects
- Immunity, Mucosal/drug effects
- Immunoglobulin G/classification
- Injections, Intramuscular
- Interferon-gamma/biosynthesis
- Interleukin-4/biosynthesis
- Mice
- Mice, Inbred BALB C
- Polymers/pharmacology
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- Titrimetry
- Vaccines, DNA/immunology
- Vaccines, DNA/pharmacology
Collapse
Affiliation(s)
- K Hamajima
- Department of Bacteriology, Yokohama City University School of Medicine, Yokohama, 236, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Sasaki S, Sumino K, Hamajima K, Fukushima J, Ishii N, Kawamoto S, Mohri H, Kensil CR, Okuda K. Induction of systemic and mucosal immune responses to human immunodeficiency virus type 1 by a DNA vaccine formulated with QS-21 saponin adjuvant via intramuscular and intranasal routes. J Virol 1998; 72:4931-9. [PMID: 9573261 PMCID: PMC110054 DOI: 10.1128/jvi.72.6.4931-4939.1998] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/1997] [Accepted: 02/20/1998] [Indexed: 02/07/2023] Open
Abstract
Induction of mucosal and cell-mediated immunity is critical for development of an effective vaccine against human immunodeficiency virus (HIV). We compared intramuscular and intranasal immunizations with a DNA vaccine encoding env of HIV-1 and evaluated the QS-21 saponin adjuvant for augmentation of the systemic and mucosal immune responses to HIV-1 in a murine model. Vaccination via the two routes elicited comparable systemic immune responses, and QS-21 consistently enhanced antigen-specific serum immunoglobulin G2a (IgG2a) production, delayed-type hypersensitivity reaction, and cytolytic activity of splenocytes. Intestinal secretory IgA production and cytolytic activity of the mesenteric lymph node cells are preferentially elicited by intranasal immunization, and QS-21 augmented these activities as well. This adjuvant augmented production of interleukin-2 (IL-2) and gamma interferon (IFN-gamma) associated with decrease in IL-4 synthesis by antigen-restimulated splenocytes. The serum immunoglobulin subtype profile showed a dominant IgG2a response and less strong IgG1 and IgE production in a QS-21 dose-dependent manner. As expected, enhancements of humoral and cell-mediated immune responses by QS-21 were abrogated by treatment with anti-IL-2 and anti-IFN-gamma monoclonal antibodies. These results suggest that the intranasal route of DNA immunization is more efficient than the intramuscular route in inducing mucosal immunity mediated by sIgA and mesenteric lymphocytes. Furthermore, QS-21 is able to act as a mucosal adjuvant in DNA vaccination and demonstrates its immunomodulatory property via stimulation of the Th1 subset. This study emphasizes the importance of the route of immunization and the use of an adjuvant for effective DNA vaccination against HIV-1.
Collapse
Affiliation(s)
- S Sasaki
- Department of Bacteriology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Despite the early success demonstrated with the hepatitis B vaccine, no other recombinant engineered vaccine has been approved for use in humans. It is unlikely that a recombinant vaccine will be developed to replace an existing licensed human vaccine with a proven record of safety and efficacy. This is due to the economic reality of making vaccines for human use. Genetically engineered subunit vaccines are more costly to manufacture than conventional vaccines, since the antigen must be purified to a higher standard than was demanded of older, conventional vaccines. Each vaccine must also be subjected to extensive testing and review by the FDA, as it would be considered a new product. This is costly to a company in terms of both time and money and is unnecessary if a licensed product is already on the market. Although recombinant subunit vaccines hold great promise, they do present some potential limitations. In addition to being less reactogenic, recombinant subunit vaccines have a tendency to be less immunogenic than their conventional counterparts. This can be attributed to these vaccines being held to a higher degree of purity than was traditionally done for an earlier generation of licensed subunit vaccines. Ironically, the contaminants often found in conventional subunit vaccines may have aided in the inflammatory process, which is essential for initiating a vigorous immune response. This potential problem may be overcome by employing one of the many new types of adjuvants that are becoming available for use in humans. Recombinant subunit vaccines may also suffer from being too well-defined, because they are composed of a single antigen. In contrast, conventional vaccines contain trace amounts of other antigens that may aid in conferring an immunity to infectious agents that is more solid than could be provided by a monovalent vaccine. This problem can be minimized, where necessary, by creating recombinant vaccines that are composed of multiple antigens from the same pathogen. These issues are less of a concern with a live attenuated vaccine, since these vaccines are less costly, require fewer steps to manufacture, and elicit long-lived immunity after only a single dose. Unfortunately, live vaccines carry a higher risk of vaccine-induced complications in recipients that make their use in highly developed, litiginous countries unlikely. In lesser developed countries, where the prevalence of disease and the need for effective vaccines outweighs the risk associated with their administration, live vaccines may play an important role in human health. This review has attempted to make the reader aware of some of the current approaches and issues that are associated with the development of these vaccines. Genetically engineered vaccines hold great promise for the future, but the potential of these vaccines to improve human and animal health has yet to be fully realized.
Collapse
Affiliation(s)
- M T Dertzbaugh
- Toxinology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702-5011, USA.
| |
Collapse
|
29
|
Sasaki S, Hamajima K, Fukushima J, Ihata A, Ishii N, Gorai I, Hirahara F, Mohri H, Okuda K. Comparison of intranasal and intramuscular immunization against human immunodeficiency virus type 1 with a DNA-monophosphoryl lipid A adjuvant vaccine. Infect Immun 1998; 66:823-6. [PMID: 9453648 PMCID: PMC107976 DOI: 10.1128/iai.66.2.823-826.1998] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We compared immune responses to intranasal and intramuscular DNA vaccinations against human immunodeficiency virus type 1 with monophosphoryl lipid A (MPL) used as an adjuvant. Both routes of vaccination resulted in similar levels of cell-mediated immunity, but the intestinal secretory immunoglobulin A response was higher following intranasal immunization than after intramuscular immunization. MPL demonstrated its adjuvanticity in vaccination by both routes.
Collapse
Affiliation(s)
- S Sasaki
- Department of Bacteriology, Yokohama City University School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kurth BE, Bryant D, Naaby-Hansen S, Reddi PP, Weston C, Foley P, Bhattacharya R, Flickinger CJ, Herr JC. Immunological response in the primate oviduct to a defined recombinant sperm immunogen. J Reprod Immunol 1997; 35:135-50. [PMID: 9421797 DOI: 10.1016/s0165-0378(97)00053-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Assessment of immune responses in the oviduct is of importance in understanding reproductive tract responses to infections, vaccination against reproductive tract pathogens, or contraceptive immunogens. This review discusses a technique that permits repeated sampling of oviductal fluid from the same monkey at intervals spanning up to several years, and the analysis of antigen-specific immunoglobulins in the fluid. This technique is important to immunocontraceptive development because previous studies in primates have lacked information on oviductal immune responses and contraceptive efficacy may not correlate well with serum antibody titers. Thus, a reliable method of sampling oviductal fluid before and after immunization with a defined antigen is required to determine the quantity and type of local immune responses necessary to achieve contraceptive effects. Implantation of access ports proved useful for repeatedly aspirating oviductal fluid in vivo from cynomolgus monkeys that was free from artifactual contaminants and with no observable changes in the behavior or health of the animals. Subsequent assays of relative and absolute concentrations of antibodies in oviductal fluid and serum demonstrated the presence of IgA and IgG specific for the recombinant sperm immunogen SP-10 in fluid collected from the periovulatory oviduct of primates after intramuscular inoculations. The antibodies evoked by the recombinant sperm vaccinogen recognized the endogenous antigen target on both human and macaque sperm, lending support for the possibility of developing a contraceptive immunogen that prevents fertilization.
Collapse
Affiliation(s)
- B E Kurth
- Center for Recombinant Gamete Contraceptive Vaccinogens, Department of Cell Biology, University of Virginia, Charlottesville 22908, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Goldenthal KL, Burns DL, McVittie LD, Lewis BP, Williams JC. Overview--combination vaccines and simultaneous administration. Past, present, and future. Ann N Y Acad Sci 1995; 754:xi-xv. [PMID: 7625640 DOI: 10.1111/j.1749-6632.1995.tb44430.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- K L Goldenthal
- Division of Vaccines and Related Products Applications, United States Food and Drug Administration, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
32
|
Key Recent Literature. Viral Immunol 1995. [DOI: 10.1089/vim.1995.8.199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|