1
|
Chen X, Liao B, Ren T, Liao Z, Huang Z, Lin Y, Zhong S, Li J, Wen S, Li Y, Lin X, Du X, Yang Y, Guo J, Zhu X, Lin H, Liu R, Wang J. Adjuvant activity of cordycepin, a natural derivative of adenosine from Cordyceps militaris, on an inactivated rabies vaccine in an animal model. Heliyon 2024; 10:e24612. [PMID: 38293396 PMCID: PMC10826310 DOI: 10.1016/j.heliyon.2024.e24612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
Vaccination is the most feasible way of preventing rabies, an ancient zoonosis that remains a major public health concern globally. However, administration of inactivated rabies vaccination without adjuvants is always inefficient and necessitates four to five injections. In the current study, we explored the adjuvant characteristics of cordycepin, a major bioactive component of Cordyceps militaris, to boost immune responses against a commercially available rabies vaccine. We found that cordycepin could stimulate stronger phenotypic and functional maturation of dendritic cells (DCs). For animal experiments, mice were immunized 3 times with rabies vaccine in the presence or absence of cordycepin at 1-week interval. Analysis of T cell differentiation and serum antibody isotypes showed that humoral immunity was dominant with a Th2 biased immune response. These results were also supported by the raised ratio of follicular helper T cells (TFH) and germinal center B cells (GCB). Thus, titer of rabies virus neutralizing antibody (RVNAb) and rabies virus-specific memory B cells were both raised as a result. Furthermore, administration of cordycepin did not cause pathological phenomena or body weight loss. The findings indicate that cordycepin could be used as a promising adjuvant for rabies vaccines to get a higher range of protection without any side effects.
Collapse
Affiliation(s)
- Xin Chen
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518118, China
| | - Boyu Liao
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518118, China
| | - Tianci Ren
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Zhipeng Liao
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Zijie Huang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Yujuan Lin
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Shouhao Zhong
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Jiaying Li
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Shun Wen
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Yingyan Li
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Xiaohan Lin
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Xingchen Du
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Yuhui Yang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518118, China
| | - Jiubiao Guo
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Xiaohui Zhu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Haishu Lin
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518118, China
| | - Rui Liu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jingbo Wang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518118, China
| |
Collapse
|
2
|
Zhou X, Wang H, Zhang J, Guan Y, Zhang Y. Single-injection subunit vaccine for rabies prevention using lentinan as adjuvant. Int J Biol Macromol 2024; 254:128118. [PMID: 37977452 DOI: 10.1016/j.ijbiomac.2023.128118] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Current rabies vaccines require 5 doses to provide full protection from the deadly virus, which significantly reduce the compliance of recipients. To minimize the number of immunizations herein single injection vaccines were developed. First a single injection vaccine was designed using rabies virus glycoprotein (G protein) as antigen. A time-controlled release system which uses dynamic layer-by-layer films as erodible coating was employed to accomplish multiply pulsatile releases of G protein. The single-injection vaccine elicits potent humoral and cellular immune responses comparable to the corresponding multi-dose ordinary vaccines because of their similar release pattern of G protein. To further improve its performance, a second single injection vaccine, in which lentinan was added as adjuvant, was designed. This single-injection vaccine again elicits humoral and cellular immune responses comparable to the corresponding multi-dose ordinary vaccines because of their similar release pattern of antigen and adjuvant. In addition, the second single-injection vaccine elicits higher level immune response and provides higher efficiency on virus inhibition than the first one because lentinan can booster immune response.
Collapse
Affiliation(s)
- Xiaoyong Zhou
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Haozheng Wang
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jianchen Zhang
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Ying Guan
- Key Laboratory of Functional Polymer Materials, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Yongjun Zhang
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Pharmaceutical Sciences, Tiangong University, Tianjin 300387, China.
| |
Collapse
|
3
|
Wang Z, Yuan Y, Chen C, Zhang C, Huang F, Zhou M, Chen H, Fu ZF, Zhao L. Colloidal Manganese Salt Improves the Efficacy of Rabies Vaccines in Mice, Cats, and Dogs. J Virol 2021; 95:e0141421. [PMID: 34495701 PMCID: PMC8577392 DOI: 10.1128/jvi.01414-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
Rabies, caused by rabies virus (RABV), remains a serious threat to public health in most countries worldwide. At present, the administration of rabies vaccines has been the most effective strategy to control rabies. Herein, we evaluate the effect of colloidal manganese salt (Mn jelly [MnJ]) as an adjuvant of rabies vaccine in mice, cats, and dogs. The results showed that MnJ promoted type I interferon (IFN-I) and cytokine production in vitro and the maturation of dendritic cells (DCs) in vitro and in vivo. Besides, MnJ serving as an adjuvant for rabies vaccines could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, plasma cells (PCs), and RABV-specific antibody-secreting cells (ASCs), consequently improve the immunogenicity of rabies vaccines, and provide better protection against virulent RABV challenge. Similarly, MnJ enhanced the humoral immune response in cats and dogs as well. Collectively, our results suggest that MnJ can facilitate the maturation of DCs during rabies vaccination, which can be a promising adjuvant candidate for rabies vaccines. IMPORTANCE Extending the humoral immune response by using adjuvants is an important strategy for vaccine development. In this study, a novel adjuvant, MnJ, supplemented in rabies vaccines was evaluated in mice, cats, and dogs. Our results in the mouse model revealed that MnJ increased the numbers of mature DCs, Tfh cells, GC B cells, PCs, and RABV-specific ASCs, resulting in enhanced immunogenicity and protection rate of rabies vaccines. We further found that MnJ had the same stimulative effect in cats and dogs. Our study provides the first evidence that MnJ serving as a novel adjuvant of rabies vaccines can boost the immune response in both a mouse and pet model.
Collapse
Affiliation(s)
- Zongmei Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yueming Yuan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chen Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengguang Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fei Huang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
4
|
Premature neonatal gut microbial community patterns supporting an epithelial TLR-mediated pathway for necrotizing enterocolitis. BMC Microbiol 2021; 21:225. [PMID: 34362295 PMCID: PMC8343889 DOI: 10.1186/s12866-021-02285-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/21/2021] [Indexed: 01/04/2023] Open
Abstract
Background Necrotising enterocolitis (NEC) is a devastating bowel disease, primarily affecting premature infants, with a poorly understood aetiology. Prior studies have found associations in different cases with an overabundance of particular elements of the faecal microbiota (in particular Enterobacteriaceae or Clostridium perfringens), but there has been no explanation for the different results found in different cohorts. Immunological studies have indicated that stimulation of the TLR4 receptor is involved in development of NEC, with TLR4 signalling being antagonised by the activated TLR9 receptor. We speculated that differential stimulation of these two components of the signalling pathway by different microbiota might explain the dichotomous findings of microbiota-centered NEC studies. Here we used shotgun metagenomic sequencing and qPCR to characterise the faecal microbiota community of infants prior to NEC onset and in a set of matched controls. Bayesian regression was used to segregate cases from control samples using both microbial and clinical data. Results We found that the infants suffering from NEC fell into two groups based on their microbiota; one with low levels of CpG DNA in bacterial genomes and the other with high abundances of organisms expressing LPS. The identification of these characteristic communities was reproduced using an external metagenomic validation dataset. We propose that these two patterns represent the stimulation of a common pathway at extremes; the LPS-enriched microbiome suggesting overstimulation of TLR4, whilst a microbial community with low levels of CpG DNA suggests reduction of the counterbalance to TLR4 overstimulation. Conclusions The identified microbial community patterns support the concept of NEC resulting from TLR-mediated pathways. Identification of these signals suggests characteristics of the gastrointestinal microbial community to be avoided to prevent NEC. Potential pre- or pro-biotic treatments may be designed to optimise TLR signalling. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02285-0.
Collapse
|
5
|
Negm II, Ragab YM, Mohamed AF. Outer membrane proteins of Salmonella typhimurium as an adjuvant in rabies vaccine. Clin Exp Vaccine Res 2021; 10:132-140. [PMID: 34222125 PMCID: PMC8217580 DOI: 10.7774/cevr.2021.10.2.132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/26/2021] [Indexed: 11/15/2022] Open
Abstract
PURPOSE The objective of the present study was to evaluate the immune-enhancing potential of Salmonella typhimurium outer membrane protein (OMP) and alum as adjuvants towards inactivated Vero cells rabies vaccine (FRV/K2). MATERIALS AND METHODS Six groups of female Sprague Dawley albino rats (10/group) were used in the evaluation of immunogenicity and safety of vaccines and adjuvants. Total immunoglobulin G secreted interferon-gamma (IFN-γ), and the percentage of proliferated CD4+ and CD8+ T cells were measured. Biochemical analysis and histopathological examination were used to test safety profiles. RESULTS OMP adjuvanted rabies vaccine (FRV/K2+OMP) (OMP combined locally prepared vaccine) induced significantly higher neutralizing antibodies on day 21 post-vaccination relative to free (FRV/K2) vaccine and alum adsorbed vaccine (FRV/K2+alum) (alum adsorbed locally prepared vaccine). (FRV/K2+OMP) induced a significantly higher level of IFN-γ on day 14 post-vaccination. CD8+ T cells were significantly higher post-vaccination with reference (RV), free (FRV/K2), and (FRV/K2+OMP) than (FRV/K2+alum). On the contrary, CD4+ T cells were significantly elevated post-vaccination with (FRV/K2+alum) at p<0.05. Biochemical analysis and histopathological examination revealed that OMP could be used safely as an adjuvant for the development of more effective rabies vaccines. CONCLUSION Outer membrane proteins adjuvanted rabies vaccines would be beneficial to induce rapid neutralizing antibodies and essential cytokines.
Collapse
Affiliation(s)
- Iman Ibrahim Negm
- The Egyptian Holding Company for the Production of Vaccines, Sera and Drugs, Cairo, Egypt
| | - Yasser M. Ragab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aly Fahmy Mohamed
- International Center for Training and Advanced Researches (ICTAR-Egypt), Cairo, Egypt
| |
Collapse
|
6
|
Liu C, Huang P, Zhao D, Xia M, Zhong W, Jiang X, Tan M. Effects of rotavirus NSP4 protein on the immune response and protection of the S R69A-VP8* nanoparticle rotavirus vaccine. Vaccine 2021; 39:263-271. [PMID: 33309483 PMCID: PMC7822095 DOI: 10.1016/j.vaccine.2020.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/05/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022]
Abstract
Rotavirus causes severe diarrhea and dehydration in young children. Even with the implementation of the current live vaccines, rotavirus infections still cause significant mortality and morbidity, indicating a need for new rotavirus vaccines with improved efficacy. To this end, we have developed an SR69A-VP8*/S60-VP8* nanoparticle rotavirus vaccine candidate that will be delivered parenterally with Alum adjuvant. In this study, as parts of our further development of this nanoparticle vaccine, we evaluated 1) roles of rotavirus nonstructural protein 4 (NSP4) that is the rotavirus enterotoxin, a possible vaccine target, and an immune stimulator, and 2) effects of CpG adjuvant that is a toll-like receptor 9 (TLR9) ligand and agonist on the immune response and protection of our SR69A-VP8*/S60-VP8* nanoparticle vaccine. The resulted vaccine candidates were examined for their IgG responses in mice. In addition, the resulted mouse sera were assessed for i) blocking titers against interactions of rotavirus VP8* proteins with their glycan ligands, ii) neutralization titers against rotavirus replication in cell culture, and iii) passive protection against rotavirus challenge with diarrhea in suckling mice. Our data showed that the Alum adjuvant appeared to work better with the SR69A-VP8*/S60-VP8* nanoparticles than the CpG adjuvant, while an addition of the NSP4 antigen to the SR69A-VP8*/S60-VP8* vaccine may not help to further increase the immune response and protection of the resulted vaccine.
Collapse
Affiliation(s)
- Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China
| | - Pengwei Huang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Dandan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Weiming Zhong
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
7
|
Abramova EG, Nikiforov AK, Movsesyants AA, Zhulidov IM. Rabies and rabies immunobiological preparations: vaccinations Pasteur to the contemporary biotechnology. JOURNAL OF MICROBIOLOGY, EPIDEMIOLOGY AND IMMUNOBIOLOGY 2019. [DOI: 10.36233/0372-9311-2019-5-83-94] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The review provides information on topical issues of rabies spread in the world and the Russian Federation, the stages of development and directions of improvement of available preventive anti-rabies immunobiological preparation used in medical practice for active and passive immunization against rabies. The current level of biotechnology development with the use of molecular biology and genetic engineering methods opens up prospects for the design of new safe effective anti-rabies drugs using recombinant technologies. Expanding the range of immunobiological drugs against rabies and their introduction into health practice will contribute to the elimination of human mortality from rabies.
Collapse
Affiliation(s)
| | - A. K. Nikiforov
- Russian Research Institute for Plague Control «Microb»;
Vavilov Saratov State Agrarian University
| | | | | |
Collapse
|
8
|
Incorporating B cell activating factor (BAFF) into the membrane of rabies virus (RABV) particles improves the speed and magnitude of vaccine-induced antibody responses. PLoS Negl Trop Dis 2019; 13:e0007800. [PMID: 31725816 PMCID: PMC6855436 DOI: 10.1371/journal.pntd.0007800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/20/2019] [Indexed: 12/25/2022] Open
Abstract
B cell activating factor (BAFF) is a member of the tumor necrosis factor (TNF) superfamily of cytokines that links innate with adaptive immunity. BAFF signals through receptors on B cells, making it an attractive molecule to potentiate vaccine-induced B cell responses. We hypothesized that a rabies virus (RABV)-based vaccine displaying both antigen and BAFF on the surface of the same virus particle would target antigen-specific B cells for activation and improve RABV-specific antibody responses. To test this hypothesis, we constructed a recombinant RABV-based vector expressing virus membrane-anchored murine BAFF (RABV-ED51-mBAFF). BAFF was incorporated into the RABV particle and determined to be biologically functional, as demonstrated by increased B cell survival of primary murine B cells treated ex-vivo with RABV-ED51-mBAFF. B cell survival was inhibited by pre-treating RABV-ED51-mBAFF with an antibody that blocks BAFF functions. RABV-ED51-mBAFF also activated primary murine B cells ex-vivo more effectively than RABV as shown by significant upregulation of CD69, CD40, and MHCII on the surface of infected B cells. In-vivo, RABV-ED51-mBAFF induced significantly faster and higher virus neutralizing antibody (VNA) titers than RABV while not adversely affecting the longevity of the vaccine-induced antibody response. Since BAFF was incorporated into the virus particle and genome replication was not required for BAFF expression in-vivo, we hypothesized that RABV-ED51-mBAFF would be effective as an inactivated vaccine. Mice immunized with 250 ng/mouse of β-propriolactone-inactivated RABV-ED51-mBAFF showed faster and higher anti-RABV VNA titers compared to mice immunized with inactivated RABV. Together, this model stands as a potential foundation for exploring other virus membrane-anchored molecular adjuvants to make safer, more effective inactivated RABV-based vaccines.
Collapse
|
9
|
Fooks AR, Banyard AC, Ertl HCJ. New human rabies vaccines in the pipeline. Vaccine 2019; 37 Suppl 1:A140-A145. [PMID: 30153997 PMCID: PMC6863069 DOI: 10.1016/j.vaccine.2018.08.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/17/2018] [Accepted: 08/16/2018] [Indexed: 12/24/2022]
Abstract
Rabies remains endemic in more than 150 countries. In 99% of human cases, rabies virus is transmitted by dogs. The disease, which is nearly always fatal, is preventable by vaccines given either before and/or after exposure to a rabid animal. Numerous factors including the high cost of vaccines, the relative complexity of post-exposure vaccination protocols requiring multiple doses of vaccine, which in cases of severe exposure have to be combined with a rabies immune globulin, lack of access to health care, and insufficient surveillance contribute to the estimated 59,000 human deaths caused by rabies each year. New, less expensive and more immunogenic rabies vaccines are needed together with improved surveillance and dog rabies control to reduce the death toll of human rabies. Here, we discuss new rabies vaccines that are in clinical and pre-clinical testing and evaluate their potential to replace current vaccines.
Collapse
|
10
|
Comberlato A, Paloja K, Bastings MMC. Nucleic acids presenting polymer nanomaterials as vaccine adjuvants. J Mater Chem B 2019; 7:6321-6346. [PMID: 31460563 DOI: 10.1039/c9tb01222b] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most vaccines developed today include only the antigens that best stimulate the immune system rather than the entire virus or microbe, which makes vaccine production and use safer and easier, though they lack potency to induce acceptable immunity and long-term protection. The incorporation of additional immune stimulating components, named adjuvants, is required to generate a strong protective immune response. Nucleic acids (DNA and RNA) and their synthetic analogs are promising candidates as vaccine adjuvants activating Toll-like receptors (TLRs). Additionally, in the last few years several nanocarriers have emerged as platforms for targeted co-delivery of antigens and adjuvants. In this review, we focus on the recent developments in polymer nanomaterials presenting nucleic acids as vaccine adjuvants. We aim to compare the effectiveness of the various classes of polymers in immune modulating materials (nanoparticles, dendrimers, single-chain particles, nanogels, polymersomes and DNA-based architectures). In particular, we address the critical role of parameters such as size, shape, complexation and release of TLR ligands, cellular uptake, stability, toxicity and potential importance of spatial control in ligand presentation.
Collapse
Affiliation(s)
- Alice Comberlato
- IMX/IBI, EPFL, EPFL-STI-IMX-PBL MXC 340 Station 12, Lausanne, 1015, Switzerland.
| | - Kaltrina Paloja
- IMX/IBI, EPFL, EPFL-STI-IMX-PBL MXC 340 Station 12, Lausanne, 1015, Switzerland.
| | - Maartje M C Bastings
- IMX/IBI, EPFL, EPFL-STI-IMX-PBL MXC 340 Station 12, Lausanne, 1015, Switzerland.
| |
Collapse
|
11
|
|
12
|
Yu P, Yan J, Wu W, Tao X, Lu X, Liu S, Zhu W. A CpG oligodeoxynucleotide enhances the immune response to rabies vaccination in mice. Virol J 2018; 15:174. [PMID: 30424815 PMCID: PMC6234694 DOI: 10.1186/s12985-018-1089-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 11/01/2018] [Indexed: 11/27/2022] Open
Abstract
Background Rabies is a fatal disease that is preventable when post exposure prophylaxis (PEP) is administered in a timely fashion. CpG oligodeoxynucleotides (ODNs) can trigger cells that express Toll-like receptor 9, and their immunopotentiation activity in an inactivated aluminum-adjuvanted rabies vaccine for dogs has been identified using mouse and dog models. Methods A human diploid cell rabies vaccine (HDCV) of humans and a CpG ODNs with cross-immunostimulatory activity in humans and mice were used to evaluate the immunogenicity and protective efficacy of CpG ODN in a mouse model that simulates human PEP. Results HDCV combined with CpG ODN (HDCV–CpG) stimulated mice to produce rabies virus-specific neutralizing antibody (RVNA) earlier and increased the seroconversion rate. Compared with HDCV alone, either HDCV–1.25 μg CpG or HDCV–5 μg CpG increased the levels of RVNA. In particular, 5 μg CpG ODN per mouse significantly boosted the levels of RVNA compared with HDCV alone. IFN-γ producing splenocytes generated in the HDCV-5 μg CpG group were significantly increased compared to the group treated with HDCV alone. When the immunization regimen was reduced to three injections or the dose was reduced to half of the recommended HDCV combined with CpG ODN, the RVNA titers were still higher than those induced by HDCV alone. After viral challenge, 50% of mice immunized with a half-dose HDCV–CpG survived, while the survival rate of mice immunized with HDCV alone was 30%. Conclusions The immunopotentiation activity of CpG ODNs for a commercially available human rabies vaccine was first evaluated in a mouse model on the basis of the Essen regimen. Our results suggest that the CpG ODN used in this study is a potential adjuvant to rabies vaccines for human use. Electronic supplementary material The online version of this article (10.1186/s12985-018-1089-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pengcheng Yu
- Key Laboratory for Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jianghong Yan
- Key Laboratory for Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Weicheng Wu
- Key Laboratory for Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoyan Tao
- Key Laboratory for Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xuexin Lu
- Key Laboratory for Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuqing Liu
- Key Laboratory for Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wuyang Zhu
- Key Laboratory for Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
13
|
Sadati SF, Jamali A, Abdoli A, Abedi-Valugerdi M, Gholami S, Alipour S, Soleymani S, Kheiri MT, Atyabi F. Simultaneous formulation of influenza vaccine and chitosan nanoparticles within CpG oligodesoxi nucleotides leads to dose-sparing and protects against lethal challenge in the mouse model. Pathog Dis 2018; 76:5089974. [PMID: 30184220 DOI: 10.1093/femspd/fty070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/01/2018] [Indexed: 12/22/2022] Open
Abstract
Lack of efficient delivery systems for transporting antigenic molecules to the cytosol of antigen-presenting cells presents a major obstacle for antigen uptake by immune cells. To this end, influenza whole inactivated virus vaccines were formulated with chitosan nanoparticles and CpG oligonucleotide as a biodegradable delivery system and a Th1-specific adjuvant, respectively. Intradermal injections of a single high dose and low dose of formulated candidate vaccines were carried out. Thirty days after injection, cell proliferation assay (MTT), IFN-gamma and IL-4 ELISpot assays were conducted. Sera samples were collected 21 days after immunization to measure IgG1 and IgG2a levels. In addition, the mice challenged with mouse-adopted virus were monitored for weight loss. The results show a significant stimulation of both humoral and cellular immunities; also, weight gain and a decrease in mortality in the mice receiving both dosages of inactivated influenza virus vaccines with CpG and Chitosan coating were observed. Based on the results, it can be concluded that formulation of inactivated influenza virus with CpG and its delivery by chitosan as low-dose can return the same results as with high-dose balanced between cellular and humeral immune responses. This formulation could potentially lead to a significant saving in vaccine production.
Collapse
Affiliation(s)
- Seyed Farid Sadati
- Department of Medical Microbiology, Ondokuz Mayis University Medical School, Samsun, Turkey.,Amasya University Research Laboratory Center, Ipekkoy Campus, Amasya, Turkey
| | - Abbas Jamali
- Influenza Unit, Pasteur Institute of Iran, Tehran, Iran
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Shima Gholami
- Department of Pharmaceutical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Samira Alipour
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45195-1159, Iran
| | - Sepehr Soleymani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Shi W, Kou Y, Xiao J, Zhang L, Gao F, Kong W, Su W, Jiang C, Zhang Y. Comparison of immunogenicity, efficacy and transcriptome changes of inactivated rabies virus vaccine with different adjuvants. Vaccine 2018; 36:5020-5029. [DOI: 10.1016/j.vaccine.2018.07.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/29/2018] [Accepted: 07/03/2018] [Indexed: 12/12/2022]
|
15
|
Li Z, Ding W, Guo Q, Liu Z, Zhu Z, Song S, Li W, Liao G. Analysis of the dose-sparing effect of adjuvanted Sabin-inactivated poliovirus vaccine (sIPV). Hum Vaccin Immunother 2018; 14:1987-1994. [PMID: 29601259 PMCID: PMC6150041 DOI: 10.1080/21645515.2018.1454571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Sabin-based inactivated poliovirus vaccine(sIPV) is gradually replacing live-attenuated oral polio vaccine(OPV). Sabin-inactivated poliovirus vaccine(sIPV) has played a vital role in reducing economic burden of poliomyelitis and maintaining appropriate antibody levels in the population. However, due to its high cost and limited manufacturing capacity, sIPV cannot reach its full potential for global poliovirus eradication in developing countries. Therefore, to address this situation, we designed this study to evaluate the dose-sparing effects of AS03, CpG oligodeoxynucleotides (CpG-ODN) and polyinosinic:polycytidylic acid (PolyI:C) admixed with sIPV in rats. Our results showed that a combination of 1/4-dose sIPV adjuvanted with AS03 or AS03 with BW006 provides a seroconversion rate similar to that of full-dose sIPV without adjuvant and that, this rate is 5-fold higher than that of 1/4-dose sIPV without adjuvant after the first immunization. The combination of AS03 or AS03 with BW006 as an adjuvant effectively reduced sIPV dose by at least 4-fold and induced both humoral and cellular immune responses. Therefore, our study revealed that the combination of AS03 or AS03 with BW006 is a promising adjuvant for sIPV development.
Collapse
Affiliation(s)
- Zhuofan Li
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Wenting Ding
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Qi Guo
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Ze Liu
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Zhe Zhu
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Shaohui Song
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Weidong Li
- b The Department of Production Administration , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Guoyang Liao
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| |
Collapse
|
16
|
A novel rabies virus lipopeptide provides a better protection by improving the magnitude of DCs activation and T cell responses. Virus Res 2016; 221:66-73. [DOI: 10.1016/j.virusres.2016.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 03/20/2016] [Accepted: 05/09/2016] [Indexed: 11/22/2022]
|
17
|
Liu R, Wang J, Yang Y, Khan I, Zhu N. Rabies virus lipopeptide conjugated to a TLR7 agonist improves the magnitude and quality of the Th1-biased humoral immune response in mice. Virology 2016; 497:102-110. [PMID: 27449478 DOI: 10.1016/j.virol.2016.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/06/2016] [Accepted: 06/23/2016] [Indexed: 12/17/2022]
Abstract
In this study, we conjugated the rabies-derived lipopeptide CE536 to a TLR7 agonist, imiquimod, and evaluated its adjuvanticity. The synthetic construct (Lipo-I) targeted to TLR7, induced dendritic cell phenotypic maturation and production of both type I interferon and pro-inflammatory cytokines more efficiently than unconjugated TLR7 ligands or lipopeptide alone. The immunostimulatory effects of the conjugate were apparently the result of IκBα degradation and sustained p38 and JNK phosphorylation. The analysis of IgG isotypes and T cell differentiation showed that IgG2a dominant Th1-biased humoral and CD8(+) IFN-γ T cell responses were induced by Lipo-I. Lipo-I could facilitate the rabies vaccine to induce the production of an earlier and more vigorous rabies virus neutralizing antibody. In the post-exposure test, the Lipo-I adjuvanted vaccine provided a 73.3% survival rate, while the traditional vaccine bestowed only a 26.7% survival. Therefore, Lipo-I is a promising adjuvant for the development of more effective rabies vaccines.
Collapse
Affiliation(s)
- Rui Liu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Jingbo Wang
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yan Yang
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Inamullah Khan
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Naishuo Zhu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
18
|
DiStefano D, Antonello JM, Bett AJ, Medi MB, Casimiro DR, ter Meulen J. Immunogenicity of a reduced-dose whole killed rabies vaccine is significantly enhanced by ISCOMATRIX™ adjuvant, Merck amorphous aluminum hydroxylphosphate sulfate (MAA) or a synthetic TLR9 agonist in rhesus macaques. Vaccine 2013; 31:4888-93. [PMID: 23941913 DOI: 10.1016/j.vaccine.2013.07.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 07/09/2013] [Accepted: 07/16/2013] [Indexed: 12/25/2022]
Abstract
There is a need for novel rabies vaccines suitable for short course, pre- and post-exposure prophylactic regimens which require reduced doses of antigen to address the current worldwide supply issue. We evaluated in rhesus macaques the immunogenicity of a quarter-dose of a standard rabies vaccine formulated with Merck's amorphous aluminum hydroxylphosphate sulfate adjuvant, the saponin-based ISCOMATRIX™ adjuvant, or a synthetic TLR9 agonist. All adjuvants significantly increased the magnitude and durability of the humoral immune response as measured by rapid fluorescent focus inhibition test (RFFIT). Several three-dose vaccine regimens resulted in adequate neutralizing antibody of ≥ 0.5 IU/ml earlier than the critical day seven post the first dose. Rabies vaccine with ISCOMATRIX™ adjuvant given at days 0 and 3 resulted in neutralizing antibody titers which developed faster and were up to one log10 higher compared to WHO-recommended intramuscular and intradermal regimens and furthermore, passive administration of human rabies immunoglobulin did not interfere with immunogenicity of this reduced dose, short course vaccine regimen. Adjuvantation of whole-killed rabies vaccine for intramuscular injection may therefore be a viable alternative to intradermal application of non-adjuvanted vaccine for both pre- and post-exposure regimens.
Collapse
Affiliation(s)
- Daniel DiStefano
- Department of Vaccine Basic Research, Merck Research Laboratories, West Point, PA, USA
| | | | | | | | | | | |
Collapse
|
19
|
Hu X, Liu R, Zhu N. Enhancement of humoral and cellular immune responses by monophosphoryl lipid A (MPLA) as an adjuvant to the rabies vaccine in BALB/c mice. Immunobiology 2013; 218:1524-8. [PMID: 23816301 DOI: 10.1016/j.imbio.2013.05.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 05/16/2013] [Accepted: 05/22/2013] [Indexed: 11/30/2022]
Abstract
The development of effective vaccines against the rabies virus could prevent infection with this fatal virus. However, the current rabies vaccine fails to provide a full range of protection because of its limited ability to elicit a cellular immune response and the requirement for repeat vaccination. Monophosphoryl lipid A (MPLA) is well known as a potent adjuvant to enhance immune responses against virus infection. Here we investigated the efficacy of MPLA as an adjuvant to improve the humoral and cellular immune responses to the rabies vaccine in BALB/c mice. Supplementation of the rabies vaccine with MPLA significantly accelerated the production of specific antibodies by 10 days compared to the original vaccines. Furthermore, MPLA promoted the induction of stronger cellular immune responses by the rabies vaccine, including the production of IL-4, IFN-γ and the activation of CD4⁺/CD8⁺ T cells, than those elicited without MPLA. Collectively, our findings indicated that MPLA enhances humoral and cellular immunity and is a promising adjuvant for the development of more effective rabies vaccines.
Collapse
Affiliation(s)
- Xiaobo Hu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | | | | |
Collapse
|
20
|
Hou J, Liu Y, Liu Y, Shao Y. The MSHA strain of Pseudomonas aeruginosa activated TLR pathway and enhanced HIV-1 DNA vaccine immunoreactivity. PLoS One 2012; 7:e47724. [PMID: 23077664 PMCID: PMC3471878 DOI: 10.1371/journal.pone.0047724] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/14/2012] [Indexed: 12/27/2022] Open
Abstract
The mannose-sensitive hemagglutination pilus strain of Pseudomonas aeruginosa (PA-MSHA) has been shown to trigger naïve immune responses through the activation of monocytes, macrophages, natural killer cells (NK cells) and antigen presenting cells (APCs). Based on the hypothesis that PA-MSHA activates natural immunity through the Toll-like receptor (TLR) pathway, we scanned several critical TLR pathway molecules in mouse splenocytes using high-throughput real-time QRT-PCR and co-stimulatory molecule in bone marrow-derived dendritic cells (BMDCs) following in vitro stimulation by PA-MSHA. PA-MSHA enabled activation of the TLR pathway mediated by NF-κB and JNK signaling in splenocytes, and the co-stimulatory molecule CD86 was up-regulated in BMDCs. We then assessed the adjuvant effect of PA-MSHA for HIV-1 DNA vaccines. In comparison to DNA inoculation alone, co-inoculation with low dosage of PA-MSHA enhanced specific immunoreactivity against HIV-1 Env in both cellular and humoral responses, and promoted antibody avidity maturation. However, high doses of adjuvant resulted in an immunosuppressive effect; a two- or three-inoculation regimen yielded low antibody responses and the two-inoculation regimen exhibited only a slight cellular immunity response. To our knowledge, this is the first report demonstrating the utility of PA-MSHA as an adjuvant to a DNA vaccine. Further research is needed to investigate the exact mechanisms through which PA-MSHA achieves its adjuvant effects on innate immune responses, especially on dendritic cells.
Collapse
Affiliation(s)
- Jue Hou
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yong Liu
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ying Liu
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yiming Shao
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- * E-mail:
| |
Collapse
|
21
|
Zhou X, Wei H, Sun P, Wu X, Wan M, Zhang P, Guo S, Zhao T, Yu Y, Wang L. Recombinant hepatitis B virus surface antigen formulated with B-type CpG oligodeoxynucleotide induces therapeutic immunity against hepatitis B virus surface antigen-expressing liver cancer cells in mice. Cancer Biother Radiopharm 2012; 27:234-42. [PMID: 22537404 DOI: 10.1089/cbr.2011.1127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
To develop a therapeutic vaccine against hepatitis B virus surface antigen (HBsAg)-expressing liver cancer, we tried to prepare a vaccine by formulating recombinant HBsAg with BW006, a B type CpG oligodeoxynucleotide (ODN) with Th1-biasing activity, and examined its potency of inducing therapeutic immunity against HBsAg-expressing liver cancer cells in mice. When applied therapeutically, BW006 could assist HBsAg to induce vigorous immune responses capable of inhibiting the growth of HBsAg-expressing liver cancer cells and prolonging the survival of mice bearing HBsAg-expressing liver cancer cells. In vivo and in vitro experiments showed that the BW006-adjuvanted HBsAg enhanced the production of IgG2a antibodies, interferon-γ, and interleukin-12 and facilitated the generation of specific cytotoxic T lymphocyte that killed the HBsAg-expressing liver cancer cells. These results suggest that the BW006-adjuvanted HBsAg might be developed into a candidate tumor vaccine for the treatment of HBsAg-expressing liver cancer.
Collapse
Affiliation(s)
- Xiaojing Zhou
- Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shen Y, Feng Z, Lin C, Hou X, Wang X, Wang J, Yu Y, Wang L, Sun X. An oligodeoxynucleotide that induces differentiation of bone marrow mesenchymal stem cells to osteoblasts in vitro and reduces alveolar bone loss in rats with periodontitis. Int J Mol Sci 2012; 13:2877-2892. [PMID: 22489131 PMCID: PMC3317693 DOI: 10.3390/ijms13032877] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 02/19/2012] [Accepted: 02/24/2012] [Indexed: 01/01/2023] Open
Abstract
To investigate the effect of oligodeoxynucleotides (ODNs) on the differentiation of rat bone marrow mesenchymal stem cells (BMSCs) to osteoblasts, in order to find a candidate ODN with potential for the treatment of periodontitis, a series of ODNs were designed and selected to test their effect on the promotion of the differentiation of BMSCs to osteoblasts in vitro and on the repair of periodontal tissue in rats with periodontitis. It was found that MT01, one of the ODNs with the sequences of human mitochondrial DNA, stimulated the proliferation of BMSCs, the differentiation of BMSCs to osteoblasts and mRNA expression of bone-associated factors including Runx2, Osterix, OPG, RANKL and collagen I in vitro. In vivo study showed that MT01 prevented the loss of alveolar bone in the rats with periodontitis and induced the production of proteins of OPG and Osterix in the bone tissue. These results indicated that MT01 could induce differentiation of BMSCs to osteoblasts and inhibit the alveolar bone absorption in rats with periodontitis.
Collapse
Affiliation(s)
- Yuqin Shen
- Department of Periodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (Y.S.); (C.L.)
| | - Zhiyuan Feng
- Department of Orthodontics, People’s Hospital of Shanxi, 29 Shuangta Road, Taiyuan 030012, China; E-Mail:
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (X.H.); (J.W.)
| | - Chongtao Lin
- Department of Periodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (Y.S.); (C.L.)
| | - Xu Hou
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (X.H.); (J.W.)
| | - Xueju Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mail:
| | - Jing Wang
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (X.H.); (J.W.)
| | - Yongli Yu
- Department of Immunology, Medical College of Norman Bethune, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mail:
| | - Liying Wang
- Department of Molecular Biology, Medical College of Norman Bethune, Jilin University, 1500 Qinghua Road, Changchun 130021, China
| | - Xinhua Sun
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (X.H.); (J.W.)
| |
Collapse
|
23
|
Yan Y, Cao Z, Yang M, Li H, Wei H, Fu Y, Song D, Wang L, Yu Y. A CpG oligodeoxynucleotide potentiates the anti-tumor effect of HSP65-Her2 fusion protein against Her2 positive B16 melanoma in mice. Int Immunopharmacol 2012; 12:402-7. [PMID: 22222115 DOI: 10.1016/j.intimp.2011.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 12/04/2011] [Accepted: 12/19/2011] [Indexed: 12/25/2022]
Abstract
Although being promising tumor vaccine candidates in animal models, heat shock protein (HSP)-based tumor vaccines have not yet succeeded in the clinical trials, implying the necessity to be formulated with appropriate adjutants to enhance their immunogenicity. In this study, we investigated whether a B-class CpG ODN (BW006), a TLR9 agonist, could facilitate HSP65-Her2, a recombinant protein between mycobacterial HSP65 and Her2-derived peptide, to induce vigorous anti-tumor activity against Her2 positive tumors in mice both prophylactically and therapeutically. It was found that BW006 could enhance prophylactic and therapeutic effect of HSP65-Her2 with improved survival of the mice bearing Her2(+) B16 melanoma and HSP65-Her2 specific Th1 response.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Montaner AD, Nichilo AD, Rodriguez JM, Hernando-Insua A, Fló J, Lopez RA, Sierra V, Paolazzi C, Larghi O, Horn DL, Zorzopulos J, Elias F. IMT504: A New and Potent Adjuvant for Rabies Vaccines Permitting Significant Dose Sparing. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/wjv.2012.24025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
25
|
Liu L, Shen L, Liu X, Yu Y, Li Y, Wang L, He C, Sun J, Li B. A safety study of a B-class CpG ODN in Sprague-Dawley rats. J Appl Toxicol 2011; 32:60-71. [PMID: 21538408 DOI: 10.1002/jat.1683] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2010] [Revised: 02/10/2011] [Accepted: 02/26/2011] [Indexed: 12/29/2022]
Abstract
Oligodeoxynucleotides containing CpG motifs (CpG ODNs) are potent immune activators and are being tested as anti-tumor, antimicrobial agents and as adjuvants in vaccines. Little has been reported, however, about the systematic and comprehensive safety evaluation on repeated CpG ODN administration. To investigate the safety profile of a newly developed CpG ODN, CpG 684, we conducted a 28-day repeated dose toxicity study in rats, at dose levels of 5, 20 and 150 µg CpG 684 per rat. No abnormalities in clinical observations, growth, urinalysis and bone marrow cell counts were found in CpG 684 treated rats. CpG 684 was proved biologically active, capable of up-regulating the expressions of CD40 and CD86 molecules. The monocyte numbers were increased at the dose levels of 20 and 150 µg per rat. The spleen weights were increased in female rats at the dose level of 150 µg per rat. Microscopically, 5, 20 and 150 µg per rat CpG 684 caused local inflammatory cell infiltration and hyperplasia of fibrous tissue at injection sites; the treatment of 5 and 150 µg per rat CpG 684 induced enhanced inflammatory reaction in inguinal lymphoid tissue, and the dose of 150 µg per rat induced cell hyperplasia in white pulp of spleen and white pulp expansion. CpG 684 at 150 µg per rat led to decreases in peripheral lymphocyte, serum globulin, glucose, alkaline phosphatase and K+ levels in female rats, and induced the decrease in serum albumin and total protein in rats of both sexes. The data from this study will provide an important reference for developing CpG 684 as an adjuvant for vaccines of human use.
Collapse
Affiliation(s)
- Li Liu
- Department of Chinese Herbal Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Feng Z, Shen Y, Wang L, Cheng L, Wang J, Li Q, Shi W, Sun X. An oligodeoxynucleotide with promising modulation activity for the proliferation and activation of osteoblast. Int J Mol Sci 2011; 12:2543-55. [PMID: 21731457 PMCID: PMC3127133 DOI: 10.3390/ijms12042543] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 04/06/2011] [Accepted: 04/07/2011] [Indexed: 12/22/2022] Open
Abstract
The paper explored the regulatory role of oligodeoxynucleotides (ODNs) with specific sequences in the proliferation and activation of osteoblast, using human osteoblast-like cell line MG 63 as the model. Through the administration of ODNs to MG 63 cells at a concentration of 1.0 μg/mL, ODN MT01 with positive effects on proliferation and activation of osteoblast was selected from 11 different ODNs by methyl thiazolyl tetrazolium (MTT) assay and alkaline phosphatase (ALP) activity measurement. To get a deeper insight into the molecular mechanism, effects of ODN MT01 treatment on the expression level of Sp7, runx-2, collagen-I, osteoprotegerin (OPG) and RANK ligand (RANKL) were determined using quantitative real time PCR and Western blotting. Remarkably, the mRNA and protein expression levels of Sp7, runx-2, collagen-I and OPG were improved after ODN MT01 treatment. Meanwhile, the protein expression level of RANKL was dramatically decreased. These results suggested that ODN MT01 had a significant impact in facilitating osteogenic proliferation and activation, and provided a direct evidence for the notion that single strand ODN could regulate the balance of bone formation and resorption, and thus was of great potential in the rebuilding of alveolar bone.
Collapse
Affiliation(s)
- Zhiyuan Feng
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (Z.F.); (J.W.)
| | - Yuqin Shen
- Department of Periodontal, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mail:
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medicine, Jilin University, Changchun 130021, China; E-Mail:
| | - Lin Cheng
- Department of Oral Cavity, The Second Affiliated Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; E-Mail:
| | - Jing Wang
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (Z.F.); (J.W.)
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, Jilin University, 2699 Qianjin Road, Changchun 130021, China; E-Mail:
| | - Wei Shi
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, Jilin University, 2699 Qianjin Road, Changchun 130021, China; E-Mail:
| | - Xinhua Sun
- Department of Orthodontics, School of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130021, China; E-Mails: (Z.F.); (J.W.)
| |
Collapse
|
27
|
Abstract
Rabies remains a global public health threat that kills more than 55,000 people per year. Rabies disproportionately affects children and, therefore, is ranked the seventh most important infectious disease due to years lost. Prevention of human rabies is accomplished by controlling rabies in domestic and wild animals, including the use of vaccination programs. The usefulness of human rabies vaccines is hampered by high cost, complicated vaccination regimens and lack of compliance, especially in areas of Africa and Asia where human rabies infections are endemic. A single-dose vaccine would greatly benefit efforts to combat this global health threat. However, a single-dose vaccine based on current inactivated vaccines does not appear feasible and other approaches are needed. Technology has advanced since modern human rabies vaccines were developed over 40 years ago. In addition, our understanding of immunological principles that influence the outcome of vaccination has increased. This article describes the current status of inactivated rabies virus vaccines and recent developments arising from the use of reverse genetics technologies designed to develop replication-deficient or single-cycle live rabies virus-based vectors for use as a single-dose rabies vaccine for humans.
Collapse
Affiliation(s)
- James P McGettigan
- Department of Microbiology and Immunology, Jefferson Vaccine Center, Jefferson Medical College of Thomas Jefferson University, 1020 Locust Street, JAH 466, Philadelphia, PA 19107, USA.
| |
Collapse
|
28
|
de Vries IJM, Tel J, Benitez-Ribas D, Torensma R, Figdor CG. Prophylactic vaccines mimic synthetic CpG oligonucleotides in their ability to modulate immune responses. Mol Immunol 2011; 48:810-7. [PMID: 21257206 DOI: 10.1016/j.molimm.2010.12.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 12/18/2010] [Indexed: 11/29/2022]
Abstract
Synthetic oligonucleotide ligands that bind to toll-like receptors are known to modulate the immune response via the activation of antigen presenting cells, and were therefore proposed as a novel form of vaccine adjuvant. Clinical-grade they are, however, not readily available. Here, we show that commonly used prophylactic vaccines for infectious diseases like measles, mumps and tuberculosis exhibit the same immune modulating behavior as synthetic CpG oligonucleotides in terms of their ability to stimulate IFN-α production and plasmacytoid dendritic cell maturation. Featuring the additional advantages of low-cost and proven safety, these vaccines could therefore be attractive alternatives to CpG oligonucleotides as adjuvants for immunotherapy. This previously undiscovered characteristic of prophylactic vaccines also sheds new light on the mechanisms by which they operate and is extremely interesting for vaccine development. Moreover, the finding that prophylactic vaccines trigger TLRs like synthetic oligonucleotides opens the possibility to predict the immune response of new vaccines.
Collapse
Affiliation(s)
- I Jolanda M de Vries
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
29
|
|
30
|
Yang G, Wan M, Zhang Y, Sun L, Sun R, Hu D, Zhou X, Wang L, Wu X, Wang L, Yu Y. Inhibition of a C-rich oligodeoxynucleotide on activation of immune cells in vitro and enhancement of antibody response in mice. Immunology 2010; 131:501-12. [PMID: 20646074 DOI: 10.1111/j.1365-2567.2010.03322.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To explore the possibility that human mitochondrial genomic DNA-mimicking oligodeoxynucleotides could regulate the immune response, a series of mitochondrial DNA-based oligodeoxynucleotides (MTODNs) were designed and studied to determine their immunoregulatory effects on immune cells activated by toll-like receptor (TLR) stimulation. The results showed that a C-rich MTODN, designated MT01, was able to inhibit the proliferation of human peripheral blood mononuclear cells (PBMCs) induced by cytosine-phosphate-guanosine (CpG) oligodeoxynucleotides (ODNs) and the production of type I interferon (IFN) from human PBMCs stimulated by TLR agonists, including inactivated influenza virus, imiquimod, inactivated herpes simplex virus-1 (HSV-1) and CpG ODNs. In addition, MT01 inhibited the CpG ODN-enhanced antibody response and this inhibition could be related to the antagonism of TLR9-activation pathways in B cells. Notably, unlike the G-rich suppressive ODNs reported, MT01 is composed of ACCCCCTCT repeats. These data imply that MT01 represents a novel class of immunosuppressive ODNs that could be candidate biologicals with therapeutic use in TLR activation-associated diseases.
Collapse
Affiliation(s)
- Guang Yang
- Department of Molecular Biology, Medical College of Norman Bethune, Jilin University, Changchun, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ren J, Sun L, Yang L, Wang H, Wan M, Zhang P, Yu H, Guo Y, Yu Y, Wang L. A novel canine favored CpG oligodeoxynucleotide capable of enhancing the efficacy of an inactivated aluminum-adjuvanted rabies vaccine of dog use. Vaccine 2010; 28:2458-64. [PMID: 20067754 DOI: 10.1016/j.vaccine.2009.12.077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 12/23/2009] [Accepted: 12/25/2009] [Indexed: 10/20/2022]
Abstract
In order to develop novel canine CpG ODNs as adjuvant for rabies vaccine of dog use, a panel of CpG ODNs containing different CpG motifs was designed and screened for their ability to induce the proliferation of canine splenocytes. Three AACGTT motif-containing CpG ODNs, designated as YW07, YW08 and YW09, respectively, were outshined with stronger ability to activate canine immune cells. The CpG ODNs were tested for their adjuvant activity for rabies vaccine in mice and dogs. It was found that YW07 could facilitate the rabies vaccine to induce more vigorous and long-lasting specific antibody response in mice and dogs, respectively. These findings suggest that YW07, a canine favored CpG ODN, could be used as a novel adjuvant for developing more efficient rabies vaccine of dog use.
Collapse
Affiliation(s)
- Jiling Ren
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kaushik P, Singh DK, Kumar SV, Tiwari AK, Shukla G, Dayal S, Chaudhuri P. Protection of mice against Brucella abortus 544 challenge by vaccination with recombinant OMP28 adjuvanted with CpG oligonucleotides. Vet Res Commun 2009; 34:119-32. [PMID: 20013309 DOI: 10.1007/s11259-009-9337-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2009] [Indexed: 11/24/2022]
Abstract
Brucella abortus, a gram negative, facultative intracellular pathogen causes brucellosis in many animal species and humans. Although live, attenuated vaccines are available against this infection, they suffer from certain limitations. Therefore, the development of an effective subunit vaccine against brucellosis is an area of intense research. The outer membrane proteins (OMPs) of Brucella species have been extensively studied for its immunogenicity and protective ability. We have investigated the potential of CpG ODN to enhance the immunogenicity and protective efficacy of recombinant 28 kDa outer membrane protein (rOMP28) of Brucella melitensis. The study demonstrated vigorous immunoglobulin G (IgG) response of OMP28. The administration of rOMP28 with CpG caused increased cell mediated immune response in terms of induced IgG2a, T-cell proliferation and up-regulation of type I cytokine expression. In contrast, the free antigen suppressed the interferon gamma (type I cytokine) production on in-vitro stimulation of spleenocytes. The result indicates the role of OMP28 in the down regulation of IFN-gamma production. Moreover, the B. abortus S-19 vaccinated mice showed highest production of IL-4 and IFN-gamma. The protective ability of the antigen was evaluated by systemic bacterial clearance after challenging the mouse with B. abortus 544 pathogen. The level of protection was significant in rOMP28+CpG treated mice but was lower than the required level. The results of the present study indicate that rOMP28 could be an immunogen capable of inducing both humoral and cellular immune response. The humoral response was biased towards Th1 type when it was co-administered with CpG.
Collapse
Affiliation(s)
- Purushottam Kaushik
- Division of Veterinary Public Health, Bihar Veterinary College, Patna, Bihar, India.
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Rabies, the most fatal of all infectious diseases, remains a major public health problem in developing countries, claiming the lives of an estimated 55,000 people each year. Most fatal rabies cases, with more than half of them in children, result from dog bites and occur among low-income families in Southeast Asia and Africa. Safe and efficacious vaccines are available to prevent rabies. However, they have to be given repeatedly, three times for pre-exposure vaccination and four to five times for post-exposure prophylaxis (PEP). In cases of severe exposure, a regimen of vaccine combined with a rabies immunoglobulin (RIG) preparation is required. The high incidence of fatal rabies is linked to a lack of knowledge on the appropriate treatment of bite wounds, lack of access to costly PEP, and failure to follow up with repeat immunizations. New, more immunogenic but less costly rabies virus vaccines are needed to reduce the toll of rabies on human lives. A preventative vaccine used for the immunization of children, especially those in high incidence countries, would be expected to lower fatality rates. Such a vaccine would have to be inexpensive, safe, and provide sustained protection, preferably after a single dose. Novel regimens are also needed for PEP to reduce the need for the already scarce and costly RIG and to reduce the number of vaccine doses to one or two. In this review, the pipeline of new rabies vaccines that are in pre-clinical testing is provided and an opinion on those that might be best suited as potential replacements for the currently used vaccines is offered.
Collapse
Affiliation(s)
- Hildegund C. J. Ertl
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
34
|
CpG oligonucleotide as an adjuvant for the treatment of prostate cancer. Adv Drug Deliv Rev 2009; 61:268-74. [PMID: 19166887 DOI: 10.1016/j.addr.2008.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2008] [Indexed: 12/18/2022]
Abstract
The use of an adenovirus transduced to express a prostate cancer antigen (PSA) as a vaccine for the treatment of prostate cancer has been shown to be active in the destruction of antigen-expressing prostate tumor cells in a pre-clinical model, using Balb/C or PSA transgenic mice. The destruction of PSA-secreting mouse prostate tumors was observed in Ad/PSA immunized mice in a prophylaxis study with 70% of the mice surviving long term tumor free. This successful immunotherapy was not observed in therapeutic studies in which tumors were established before vaccination and the development of anti-PSA immune response was not as easily generated in PSA transgenic mice. Immunization of conventional and transgenic animals was enhanced by incorporating a collagen matrix into the immunizing injection. Therefore the need to strengthen anti-PSA and anti-prostate cancer immunity was an obvious next step in developing a successful prostate cancer immunotherapy. Because the use of immunostimulatory CpG motifs was shown to enhance immune responses to a wide variety of antigens, our studies incorporated CpG into the Ad/PSA vaccine experimental plans. The results of the subsequent studies demonstrated a dichotomy where Ad/PSA plus CpG enhanced the in vivo destruction of PSA-secreting tumors and the survival of experimental animals, but revealed that the number and in vitro activities of antigen specific CD8+ T cells was decreased as compared to the values observed when the vaccine alone was used for immunization. The dichotomous observations were confirmed using another antigen system, OVA also incorporated into a replication defective adenovirus. Despite the reduction in antigen-specific CD8+ cells after vaccine plus CpG immunization the enhanced destruction of sc and systemic tumors was shown to be mediated entirely by CD8+ T cells. Finally, the reduction of the CD8+ T cells was the result of an observed decrease in the proliferation of the antigen specific cell population.
Collapse
|
35
|
Therapeutic injection of C-class CpG ODN in draining lymph node area induces potent activation of immune cells and rejection of established breast cancer in mice. Clin Immunol 2009; 131:426-37. [PMID: 19233733 DOI: 10.1016/j.clim.2009.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 12/19/2008] [Accepted: 01/14/2009] [Indexed: 01/16/2023]
Abstract
In order to develop novel CpG ODNs for the treatment of breast cancer, we have designed a series of CpG ODNs and evaluated their anti-tumor activity in a breast cancer mouse model. Interestingly, a C-class CpG ODN, designated as YW002, showed a vigorous activity on the inhibition of tumor growth in mice and completely cured some of the tumor-bearing mice through injection at tumor draining lymph node (TDLN) area. The expansion of immune cells in the TDLN and tumor and the generation of tumor specific immune memory were found associated with YW002-induced anti-tumor activity in mice. These results indicate that C-class CpG ODN could be developed into a medicament in a monotherapeutic regimen for the treatment of breast cancer through injection at TDLN area in clinic.
Collapse
|