1
|
Yang J, Hu X, Chen X, Li W, Yin Q, Xiong Y, An Y, Li H, Liu Z. A novel MF59 and CpG1018 adjuvant combination enhances the humoral and cellular immune responses against a truncated varicella-zoster viral glycoprotein E. Immunol Lett 2025; 275:107025. [PMID: 40239819 DOI: 10.1016/j.imlet.2025.107025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/18/2025]
Abstract
Vaccination is the only effective strategy for preventing herpes zoster (HZ), a disease caused by reactivation of the varicella-zoster virus (VZV). Cell-mediated immunity (CMI) plays a pivotal role in controlling VZV reactivation and is a critical factor in the efficacy of the HZ vaccine. This research introduced the preliminary utilization of truncated glycoprotein E (tgE) as the antigen in the formulation of an innovative recombinant HZ vaccine and explored the combination of tgE with several adjuvants to assess their effectiveness in eliciting robust humoral and CMI responses in C57BL/6 mice, followed by the immunogenicity validation of the optimal vaccine formulation in Sprague-Dawley (SD) rats and cynomolgus monkeys. The results demonstrated that the combination of tgE with MF59 and CpG1018, designated as tgE/MF59+CpG1018, elicited significantly stronger gE-specific humoral and cellular immune responses in C57BL/6 mice compared to any single adjuvant or other adjuvant combinations. The optimal dosages for MF59 and CpG1018 were determined to be 0.025 ml and 10 μg, respectively, for each 0.05 ml of the vaccine formulation. Notably, the increasing in the dosage of the adjuvant does not inherently correlate with a more pronounced immune response. Furthermore, the tgE/MF59+CpG1018 also elicited robust humoral and CMI responses in both SD rats and cynomolgus monkeys. These findings established the novel tgE/MF59+CpG1018 vaccine as a highly promising prophylactic candidate against HZ.
Collapse
Affiliation(s)
- Jing Yang
- Yither Biotech Co., Ltd., Pudong, Shanghai 200120, PR China
| | - Xue Hu
- Yither Biotech Co., Ltd., Pudong, Shanghai 200120, PR China
| | - Xiguang Chen
- Yither Biotech Co., Ltd., Pudong, Shanghai 200120, PR China
| | - Wanzhen Li
- Yither Biotech Co., Ltd., Pudong, Shanghai 200120, PR China
| | - Quanyi Yin
- Yither Biotech Co., Ltd., Pudong, Shanghai 200120, PR China
| | - Yelin Xiong
- Yither Biotech Co., Ltd., Pudong, Shanghai 200120, PR China
| | - Youcai An
- Ab&B Biotech Co., Ltd., Taizhou Jiangsu 225300, PR China
| | - Haiyan Li
- Yither Biotech Co., Ltd., Pudong, Shanghai 200120, PR China.
| | - Zhilei Liu
- Yither Biotech Co., Ltd., Pudong, Shanghai 200120, PR China.
| |
Collapse
|
2
|
Andrianov AK, Fuerst TR. Immunopotentiating and Delivery Systems for HCV Vaccines. Viruses 2021; 13:v13060981. [PMID: 34070543 PMCID: PMC8227888 DOI: 10.3390/v13060981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022] Open
Abstract
Development of preventive vaccines against hepatitis C virus (HCV) remains one of the main strategies in achieving global elimination of the disease. The effort is focused on the quest for vaccines capable of inducing protective cross-neutralizing humoral and cellular immune responses, which in turn dictate the need for rationally designed cross-genotype vaccine antigens and potent immunoadjuvants systems. This review provides an assessment of the current state of knowledge on immunopotentiating compounds and vaccine delivery systems capable of enhancing HCV antigen-specific immune responses, while focusing on the synergy and interplay of two modalities. Structural, physico-chemical, and biophysical features of these systems are discussed in conjunction with the analysis of their in vivo performance. Extreme genetic diversity of HCV-a well-known hurdle in the development of an HCV vaccine, may also present a challenge in a search for an effective immunoadjuvant, as the effort necessitates systematic and comparative screening of rationally designed antigenic constructs. The progress may be accelerated if the preference is given to well-defined molecular immunoadjuvants with greater formulation flexibility and adaptability, including those capable of spontaneous self-assembly behavior, while maintaining their robust immunopotentiating and delivery capabilities.
Collapse
Affiliation(s)
- Alexander K. Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA;
- Correspondence:
| | - Thomas R. Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA;
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
3
|
Protective cellular immune response against hepatitis C virus elicited by chimeric protein formulations in BALB/c mice. Arch Virol 2020; 165:593-607. [PMID: 32016547 PMCID: PMC7224087 DOI: 10.1007/s00705-019-04464-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023]
Abstract
The eradication of hepatitis C virus (HCV) infection is a public health priority. Despite the efficiency of treatment with direct-acting antivirals, the high cost of the therapy and the lack of accurate data about the HCV-infected population worldwide constitute important factors hampering this task. Hence, an affordable preventive vaccine is still necessary for reducing transmission and the future disease burden globally. In this work, chimeric proteins (EnvCNS3 and NS3EnvCo) encompassing conserved and immunogenic epitopes from the HCV core, E1, E2 and NS3 proteins were produced in Escherichia coli, and their immunogenicity was evaluated in BALB/c mice. The impact of recombinant HCV E2.680 protein and oligodeoxynucleotide 39M (ODN39M) on the immune response to chimeric proteins was also assessed. Immunization with chimeric proteins mixed with E2.680 enhanced the antibody and cellular response against HCV antigens and chimeric proteins. Interestingly, the combination of NS3EnvCo with E2.680 and ODN39M as adjuvant elicited a potent antibody response characterized by an increase in antibodies of the IgG2a subclass against E2.680, NS3 and chimeric proteins, suggesting the induction of a Th1-type response. Moreover, a cytotoxic T lymphocyte response and a broad response of IFN-γ-secreting cells against HCV antigens were induced with this formulation as well. This T cell response was able to protect vaccinated mice against challenge with a surrogate model based on HCV recombinant vaccinia virus. Overall, the vaccine candidate NS3EnvCo/E2.680/ODN39M might constitute an effective immunogen against HCV with potential for reducing the likelihood of viral persistence.
Collapse
|
4
|
Robinson C, Baehr C, Schmiel SE, Accetturo C, Mueller DL, Pravetoni M. Alum adjuvant is more effective than MF59 at prompting early germinal center formation in response to peptide-protein conjugates and enhancing efficacy of a vaccine against opioid use disorders. Hum Vaccin Immunother 2019; 15:909-917. [PMID: 30625019 DOI: 10.1080/21645515.2018.1558697] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Opioid use disorders (OUD) and fatal overdoses are a national emergency in the United States. Therapeutic vaccines offer a promising strategy to treat OUD and reduce the incidence of overdose. Immunization with opioid-based haptens conjugated to immunogenic carriers elicits opioid-specific antibodies that block opioid distribution to the brain and reduce opioid-induced behavior and toxicity in pre-clinical models. This study tested whether the efficacy of a lead oxycodone conjugate vaccine was improved by formulation in either aluminum hydroxide or the squalene-based oil-in-water emulsion MF59 adjuvant, which was recently FDA-approved for influenza vaccines in subjects 65+ years old. In adult BALB/c mice, alum formulation was more effective than MF59 at promoting the early expansion of hapten-specific B cells and the production of oxycodone-specific serum IgG antibodies, as well as blocking oxycodone distribution to the brain and oxycodone-induced motor activity. Alum was also more effective than MF59 at promoting early differentiation of peptide-specific MHCII-restricted CD4+ Tfh and GC-Tfh cells in adult C57Bl/6 mice immunized with a model peptide-protein conjugate. In contrast, alum and MF59 were equally effective in promoting hapten-specific B cells and peptide-specific MHCII-restricted CD4+ T cell differentiation in older C57Bl/6 mice. These data suggest that alum is a more effective adjuvant than MF59 for conjugate vaccines targeting synthetic small molecule haptens or peptide antigens in adult, but not aged, mice.
Collapse
Affiliation(s)
- Christine Robinson
- a Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation) , Minneapolis, MN , USA
| | - Carly Baehr
- a Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation) , Minneapolis, MN , USA.,b Department of Veterinary Population Medicine , University of Minnesota , Minneapolis, MN, , USA
| | | | - Claudia Accetturo
- d Department of Pharmaceutics , Universita degli Studi di Milano , Milan , Italy
| | - Daniel L Mueller
- e Center for Immunology , University of Minnesota , Minneapolis, MN , USA.,f Department of Medicine , University of Minnesota , Minneapolis, MN , USA
| | - Marco Pravetoni
- a Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation) , Minneapolis, MN , USA.,e Center for Immunology , University of Minnesota , Minneapolis, MN , USA.,f Department of Medicine , University of Minnesota , Minneapolis, MN , USA.,g Department of Pharmacology , University of Minnesota , Minneapolis, MN , USA
| |
Collapse
|
5
|
Agnolon V, Bruno C, Leuzzi R, Galletti B, D’Oro U, Pizza M, Seubert A, O’Hagan DT, Baudner BC. The potential of adjuvants to improve immune responses against TdaP vaccines: A preclinical evaluation of MF59 and monophosphoryl lipid A. Int J Pharm 2015; 492:169-76. [DOI: 10.1016/j.ijpharm.2015.06.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/15/2015] [Accepted: 06/18/2015] [Indexed: 01/11/2023]
|
6
|
Ilyinskii PO, Roy CJ, O'Neil CP, Browning EA, Pittet LA, Altreuter DH, Alexis F, Tonti E, Shi J, Basto PA, Iannacone M, Radovic-Moreno AF, Langer RS, Farokhzad OC, von Andrian UH, Johnston LPM, Kishimoto TK. Adjuvant-carrying synthetic vaccine particles augment the immune response to encapsulated antigen and exhibit strong local immune activation without inducing systemic cytokine release. Vaccine 2014; 32:2882-95. [PMID: 24593999 PMCID: PMC4059049 DOI: 10.1016/j.vaccine.2014.02.027] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Augmentation of immunogenicity can be achieved by particulate delivery of an antigen and by its co-administration with an adjuvant. However, many adjuvants initiate strong systemic inflammatory reactions in vivo, leading to potential adverse events and safety concerns. We have developed a synthetic vaccine particle (SVP) technology that enables co-encapsulation of antigen with potent adjuvants. We demonstrate that co-delivery of an antigen with a TLR7/8 or TLR9 agonist in synthetic polymer nanoparticles results in a strong augmentation of humoral and cellular immune responses with minimal systemic production of inflammatory cytokines. In contrast, antigen encapsulated into nanoparticles and admixed with free TLR7/8 agonist leads to lower immunogenicity and rapid induction of high levels of inflammatory cytokines in the serum (e.g., TNF-α and IL-6 levels are 50- to 200-fold higher upon injection of free resiquimod (R848) than of nanoparticle-encapsulated R848). Conversely, local immune stimulation as evidenced by cellular infiltration of draining lymph nodes and by intranodal cytokine production was more pronounced and persisted longer when SVP-encapsulated TLR agonists were used. The strong local immune activation achieved using a modular self-assembling nanoparticle platform markedly enhanced immunogenicity and was equally effective whether antigen and adjuvant were co-encapsulated in a single nanoparticle formulation or co-delivered in two separate nanoparticles. Moreover, particle encapsulation enabled the utilization of CpG oligonucleotides with the natural phosphodiester backbone, which are otherwise rapidly hydrolyzed by nucleases in vivo. The use of SVP may enable clinical use of potent TLR agonists as vaccine adjuvants for indications where cellular immunity or robust humoral responses are required.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Frank Alexis
- Laboratory of Nanomedicine and Biomaterials, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Tonti
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jinjun Shi
- Laboratory of Nanomedicine and Biomaterials, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Pamela A Basto
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Matteo Iannacone
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Aleksandar F Radovic-Moreno
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Robert S Langer
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Omid C Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
7
|
Llanes MS, Palacios NS, Piccione M, Ruiz MG, Layana C. [Molecular aspects of the antiviral response against hepatitis C virus implicated in vaccines development]. Enferm Infecc Microbiol Clin 2014; 33:273-80. [PMID: 24529681 DOI: 10.1016/j.eimc.2013.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/29/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
Hepatitis C is a contagious liver disease caused by hepacivirus of the Flaviviridae family. It has a RNA genome, a unique highly variable molecule. It encodes ten proteins which are necessary to infect cells and multiply. Replication occurs only in hepatocytes. Because of its wide genomic variability and the absence of symptoms, it is difficult to make an early diagnosis and successful treatment. In this review we analyze the molecular mechanism by which the virus infects the hepatocytes and causes the disease. We focused the analysis on different therapies, with the possibility of improving treatment with the use of new specific vaccines. We highlight the use of new therapies based on nucleic acids, mainly DNA vectors. In the near future, once this treatment is adequately evaluated in clinical trials, and the costs are calculated, it could be a very beneficial alternative to conventional methods.
Collapse
Affiliation(s)
- María Soledad Llanes
- Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Soledad Palacios
- Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de Buenos Aires, Buenos Aires, Argentina
| | - Magalí Piccione
- Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de Buenos Aires, Buenos Aires, Argentina
| | - María Guillermina Ruiz
- Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de Buenos Aires, Buenos Aires, Argentina
| | - Carla Layana
- Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de Buenos Aires, Buenos Aires, Argentina; Centro Regional de Estudios Genómicos, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Dey AK, Srivastava IK. Novel adjuvants and delivery systems for enhancing immune responses induced by immunogens. Expert Rev Vaccines 2014; 10:227-51. [DOI: 10.1586/erv.10.142] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
9
|
Fraser CK, Diener KR, Brown MP, Hayball JD. Improving vaccines by incorporating immunological coadjuvants. Expert Rev Vaccines 2014; 6:559-78. [PMID: 17669010 DOI: 10.1586/14760584.6.4.559] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
While vaccination continues to be the most successful interventionist health policy to date, infectious disease remains a significant cause of death worldwide. A primary reason that vaccination is not able to generate effective immunity is a lack of appropriate adjuvants capable of initiating the desired immune response. Adjuvant combinations can potentially overcome this problem; however, the possible permutations to consider, which include the route and kinetics of vaccination, as well as combinations of adjuvants, are practically limitless. This review aims to summarize the current understanding of adjuvants and related immunological processes and how this knowledge can and has been applied to the strategic selection of adjuvant combinations as components of vaccines against human infectious disease.
Collapse
Affiliation(s)
- Cara K Fraser
- Experimental Therapeutics Laboratory, Hanson Institute, and School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Australia.
| | | | | | | |
Collapse
|
10
|
Abstract
Hepatocellular carcinoma (HCC) is the fifth most prevalent cancer and the third leading cause of cancer-related death, and its incidence is increasing. The majority of HCC cases are associated with chronic viral hepatitis. With over 170 million individuals chronically infected with hepatitis C virus (HCV) worldwide, HCV is currently a serious global health concern, leading to chronic hepatitis, cirrhosis and HCC, thereby causing significant morbidity and mortality. With the incidence of HCV infection increasing, the problem of HCV-associated HCC is expected to worsen as well, with the majority of HCCs developing in the setting of cirrhosis. Thus, it is imperative to provide antiviral therapy to infected individuals prior to the development of established cirrhosis in order to reduce the risk of subsequent HCC. Indeed, the successful eradication of HCV is associated with clinical and histological improvement as well as a greatly reduced risk of subsequent HCC development. Even after the development of cirrhosis, successful viral clearance is still associated with reduced HCC risk. Current standard of care antiviral treatment consists of pegylated interferon-α and ribavirin, but viral clearance rates are suboptimal with this regimen, especially in difficult to treat cohorts. However, there is a myriad of different classes of HCV-specific direct-acting antiviral agents currently in development, which can be used in combination with one another or with standard of care treatment to improve HCV cure rates. Preventative and therapeutic vaccines against HCV remain an area of ongoing research with good progress towards developing an effective vaccine in the future.
Collapse
Affiliation(s)
- E J Lim
- Department of Gastroenterology, Austin Hospital, Heidelberg, Victoria, 3084, Australia,
| | | |
Collapse
|
11
|
Construction of an immunostimulatory plasmid, pUCpGs10, and research on its immune adjuvant effect. Mol Biotechnol 2013; 54:58-67. [PMID: 22544607 DOI: 10.1007/s12033-012-9544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
In order to overcome the instability of CpG ODN in vivo, sequence diversity, and individual differences, eleven CpG ODN fragments were meticulously selected and linked to form a Multi-CpG, which were repeatedly inserted into the cloning vector pUC19 for constructing the recombinant plasmid pUCpGs10 containing ten of Multi-CpG. Using the multi-genotype HCV E1 and multi-epitope complex HCV-T as immunogens, and plasmid pUCpGs10 as the immune adjuvant, Balb/c mice were immunized through nasal and subcutaneous immunization. Strong-specific humoral and cellular immune response were induced, which can obviously inhibit the growth of homograft expressing HCV antigen. The immune adjuvant effect of pUCpGs10 closely matched that of Freund's complete adjuvant. The plasmid pUCpGs10 can significantly improve IgA content in serum and different mucosal extract and systematical T-cell response via intranasal immunization. In conclusions, the newly constructed immunostimulatory plasmid pUCpGs10 is able to effectively activate the humoral and cellular immune activity, and possesses activation on mucosal immune response.
Collapse
|
12
|
Generation of a parvovirus B19 vaccine candidate. Vaccine 2013; 31:3872-8. [DOI: 10.1016/j.vaccine.2013.06.062] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/24/2013] [Accepted: 06/19/2013] [Indexed: 11/22/2022]
|
13
|
Bremer PT, Janda KD. Investigating the effects of a hydrolytically stable hapten and a Th1 adjuvant on heroin vaccine performance. J Med Chem 2012; 55:10776-80. [PMID: 23134263 DOI: 10.1021/jm301262z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We challenged the performance of our previous heroin vaccine with a similar vaccine containing a more hydrolytically stable hapten analogue and a Th1 adjuvant (CpG ODN). Our results indicate that the elements of our previous vaccine are essential for its anti-heroin potency, i.e., a chemically labile hapten and an exclusively Th2 humoral response elicited by alum. Such design elements are critical for producing next-generation heroin vaccines.
Collapse
Affiliation(s)
- Paul T Bremer
- Department of Chemistry, The Skaggs Institute for Chemical Biology, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | |
Collapse
|
14
|
Zhou Y, Zhang Y, Yao Z, Moorman JP, Jia Z. Dendritic cell-based immunity and vaccination against hepatitis C virus infection. Immunology 2012; 136:385-96. [PMID: 22486354 DOI: 10.1111/j.1365-2567.2012.03590.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hepatitis C virus (HCV) has chronically infected an estimated 170 million people worldwide. There are many impediments to the development of an effective vaccine for HCV infection. Dendritic cells (DC) remain the most important antigen-presenting cells for host immune responses, and are capable of either inducing productive immunity or maintaining the state of tolerance to self and non-self antigens. Researchers have recently explored the mechanisms by which DC function is regulated during HCV infection, leading to impaired antiviral T-cell responses and so to persistent viral infection. Recently, DC-based vaccines against HCV have been developed. This review summarizes the current understanding of DC function during HCV infection and explores the prospects of DC-based HCV vaccine. In particular, it describes the biology of DC, the phenotype of DC in HCV-infected patients, the effect of HCV on DC development and function, the studies on new DC-based vaccines against HCV infection, and strategies to improve the efficacy of DC-based vaccines.
Collapse
Affiliation(s)
- Yun Zhou
- Centre of Diagnosis and Treatment for Infectious Diseases of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | | | | | | | | |
Collapse
|
15
|
Masalova OV, Lesnova EI, Shingarova LN, Tunitskaya VL, Ulanova TI, Burkov AN, Kushch AA. The combined application of nucleotide and amino acid sequences of NS3 hepatitis C virus protein, DNA encoding granulocyte macrophage colony-stimulating factor, and inhibitor of regulatory T cells induces effective immune responce against Hepatitis C virus. Mol Biol 2012; 46:473-480. [DOI: 10.1134/s0026893312030077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 11/03/2011] [Indexed: 01/05/2025]
|
16
|
Roohvand F, Kossari N. Advances in hepatitis C virus vaccines, part two: advances in hepatitis C virus vaccine formulations and modalities. Expert Opin Ther Pat 2012; 22:391-415. [PMID: 22455502 DOI: 10.1517/13543776.2012.673589] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Developing a vaccine against HCV is an important medical and global priority. Unavailability and potential dangers associated with using attenuated HCV viral particles for vaccine preparation have resulted in the use of HCV genes and proteins formulated in novel vaccine modalities. AREAS COVERED In part one of this review, advances in basic knowledge for HCV vaccine design were provided. Herein, a detailed and correlated patents (searched by Espacenet) and literatures (searched by Pubmed) review on HCV vaccine formulations and modalities is provided, including: subunit, DNA, epitopic-peptide/polytopic, live vector- and whole yeast-based vaccines. Less-touched areas in vaccine studies such as mucosal, plant-based, and chimeric HBV/HCV vaccines are also discussed. Furthermore, results of preclinical/clinical studies on selected HCV vaccines as well as pros and cons of different strategies are reviewed. Finally, potential strategies for creation and/or improvement of HCV vaccine formulations are discussed. EXPERT OPINION Promising outcomes of a few HCV vaccine modalities in phase I/II clinical trials predict the accessibility of at least partially effective vaccines to inhibit or treat the chronic state of HCV infection (specially in combination with standard antiviral therapy). ChronVac-C (plasmid DNA), TG4040 (MVA-based), and GI-5005 (whole yeast-based) might be the most obvious HCV vaccine candidates to be approved in the near future.
Collapse
Affiliation(s)
- Farzin Roohvand
- Hepatitis & AIDS Department, Pasteur Institute of Iran, Tehran, Iran.
| | | |
Collapse
|
17
|
Gupte GM, Arankalle VA. Evaluation of the immunogenicity of liposome encapsulated HVR1 and NS3 regions of genotype 3 HCV, either singly or in combination. Virol J 2012; 9:74. [PMID: 22452828 PMCID: PMC3349533 DOI: 10.1186/1743-422x-9-74] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 03/27/2012] [Indexed: 01/15/2023] Open
Abstract
Background Hepatitis C virus displays a high rate of mutation and exists as a quasispecies in infected patients. In the absence of an effective universal vaccine, genotype-specific vaccine development represents an alternative. We have attempted to develop a genotype 3 based, liposome encapsulated HCV vaccine with hypervariable region-1 (HVR1) and non-structural region-3 (NS3) components. Results HCV RNA extracted from serum samples of 49 chronically infected patients was PCR amplified to obtain HVR1 region. These amplified products were cloned to obtain 20 clones per sample in order to identify the quasispecies pattern. The HVR1 consensus sequence, along with three variants was reverse transcribed to obtain peptides. The peptides were checked for immunoreactivity individually, as a pool or as a single peptide tetramer interspersed with four glycine residues. Anti-HCV positivity varied from 42.6% (tetramer) to 92.2% (variant-4) when 115 anti-HCV positive sera representing genotypes 1, 3, 4 and 6 were screened. All the 95 anti-HCV negatives were scored negative by all antigens. Mice were immunized with different liposome encapsulated or Al(OH)3 adjuvanted formulations of HVR1 variants and recombinant NS3 protein, and monitored for anti-HVR1 and anti-NS3 antibody titres, IgG isotypes and antigen specific cytokine levels. A balanced Th1/Th2 isotyping response with high antibody titres was observed in most of the liposome encapsulated antigen groups. The effect of liposomes and aluminium hydroxide on the expression of immune response genes was studied using Taqman Low Density Array. Both Th1 (IFN-gamma, Il18) and Th2 (Il4) genes were up regulated in the liposome encapsulated HVR1 variant pool-NS3 combination group. In-vitro binding of the virus to anti-HVR1 antibodies was demonstrated. Conclusion The optimum immunogen was identified to be combination of peptides of HVR1 consensus sequence and its variants along with pNS3 encapsulated in liposomes, which could generate both cellular and humoral immune responses in mice deserving further evaluation in a suitable cell culture system/non-human primate model.
Collapse
Affiliation(s)
- Gouri M Gupte
- Hepatitis Division, National Institute of Virology, Microbial Containment Complex, Sus Road, Pashan, Pune, India 411021
| | | |
Collapse
|
18
|
Chiang CLL, Kandalaft LE, Coukos G. Adjuvants for enhancing the immunogenicity of whole tumor cell vaccines. Int Rev Immunol 2011; 30:150-82. [PMID: 21557641 DOI: 10.3109/08830185.2011.572210] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Whole tumor cell lysates can serve as excellent multivalent vaccines for priming tumor-specific CD8(+) and CD4(+) T cells. Whole cell vaccines can be prepared with hypochlorous acid oxidation, UVB-irradiation and repeat cycles of freeze and thaw. One major obstacle to successful immunotherapy is breaking self-tolerance to tumor antigens. Clinically approved adjuvants, including Montanide™ ISA-51 and 720, and keyhole-limpet proteins can be used to enhance tumor cell immunogenicity by stimulating both humoral and cellular anti-tumor responses. Other potential adjuvants, such as Toll-like receptor agonists (e.g., CpG, MPLA and PolyI:C), and cytokines (e.g., granulocyte-macrophage colony stimulating factor), have also been investigated.
Collapse
Affiliation(s)
- Cheryl Lai-Lai Chiang
- Ovarian Cancer Research Center, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104-6142, USA
| | | | | |
Collapse
|
19
|
Torresi J, Johnson D, Wedemeyer H. Progress in the development of preventive and therapeutic vaccines for hepatitis C virus. J Hepatol 2011; 54:1273-85. [PMID: 21236312 DOI: 10.1016/j.jhep.2010.09.040] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 08/27/2010] [Accepted: 09/07/2010] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus (HCV) is a blood borne disease estimated to chronically infect 3% of the worlds' population causing significant morbidity and mortality. Current medical therapy is curative in approximately 50% of patients. While recent treatment advances of genotype 1 infection using directly acting antiviral agents (DAAs) are encouraging, there is still a need to develop vaccine strategies capable of preventing infection. Moreover, vaccines may also be used in future in combination with DAAs enabling interferon-free treatment regimens. Viral and host specific factors contribute to viral evasion and present important impediments to vaccine development. Both, innate and adaptive immune responses are of major importance for the control of HCV infection. However, HCV has evolved ways of evading the host's immune response in order to establish persistent infection. For example, HCV inhibits intracellular interferon signalling pathways, impairs the activation of dendritic cells, CD8(+) and CD4(+) T cell responses, induces a state of T-cell exhaustion and selects escape variants with mutations CD8(+) T cell epitopes. An effective vaccine will need to produce strong and broadly cross-reactive CD4(+), CD8(+) T cell and neutralising antibody (NAb) responses to be successful in preventing or clearing HCV. Vaccines in clinical trials now include recombinant proteins, synthetic peptides, virosome based vaccines, tarmogens, modified vaccinia Ankara based vaccines, and DNA based vaccines. Several preclinical vaccine strategies are also under development and include recombinant adenoviral vaccines, virus like particles, and synthetic peptide vaccines. This paper will review the vaccines strategies employed, their success to date and future directions of vaccine design.
Collapse
Affiliation(s)
- Joseph Torresi
- Austin Centre for Infection Research, Department of Infectious Diseases Austin Hospital, Heidelberg, Victoria 3084, Australia.
| | | | | |
Collapse
|
20
|
Kanduc D. HCV: Written in our DNA. SELF NONSELF 2011; 2:108-113. [PMID: 22299062 DOI: 10.4161/self.2.2.15795] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 04/08/2011] [Accepted: 04/11/2011] [Indexed: 11/19/2022]
Abstract
An inspection of the sequence similarity between the hepatitis C virus (HCV) polyprotein and human proteins revealed a high level of peptide sharing, with a limited number of motifs unique to the virus (i.e., with no counterpart in the human proteome). Using pentapeptide matching, only 214 motifs out of a total of 3,007 (7.11%) identified HCV as nonself compared to the Homo sapiens proteome. However, this virus-versus-human phenetic difference disappeared at the genetic level. Indeed, a BLAST analysis of pentadecameric oligodeoxynucleotide sequences corresponding to the 214 pentapeptides unique to HCV revealed that almost all of them are present in the human genome, located in the non-coding strand, introns, and/or pseudogenes, thus being, as such, untranslatable. The present data warn against using DNA-based vaccines to fight HCV infection and emphasize peptide uniqueness as the molecular basis for designing effective anti-HCV immunotherapeutic approaches.
Collapse
Affiliation(s)
- Darja Kanduc
- Department of Biochemistry and Molecular Biology; University of Bari; Bari, Italy
| |
Collapse
|
21
|
El Sahly H. MF59™ as a vaccine adjuvant: a review of safety and immunogenicity. Expert Rev Vaccines 2011; 9:1135-41. [PMID: 20923265 DOI: 10.1586/erv.10.111] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Approximately 70 years passed between the licensing of alum salts as vaccine adjuvants and that of MF59™ MF59, an oil-in-water emulsion, is currently licensed for use in the elderly as an adjuvant in seasonal influenza vaccines. Its mechanism of action is not fully understood, but enhancement of the interaction between the antigen and the dendritic cell seems to be involved. When used with seasonal influenza vaccines, an increase occurs in the hemagglutination inhibition antibody titers against some, but not all, seasonal vaccine influenza strains. The adjuvant effect is more pronounced when MF59 is combined with novel influenza antigens such as H9 and H5. The use of the adjuvant is associated with an increase in the frequency of local and systemic early post-vaccine adverse events (3-7 days), but no increase in adverse events was observed thereafter. Currently, MF59 is under evaluation as an adjuvant with other antigens such as pandemic influenza antigens and cytomegalovirus antigens.
Collapse
Affiliation(s)
- Hana El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
22
|
Krishnadas DK, Ahn JS, Han J, Kumar R, Agrawal B. Immunomodulation by hepatitis C virus-derived proteins: targeting human dendritic cells by multiple mechanisms. Int Immunol 2010; 22:491-502. [PMID: 20410260 DOI: 10.1093/intimm/dxq033] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) has the ability to persist in the majority of infected people. Strong, multispecific and sustained T-cell response is correlated with viral clearance. The mechanisms of chronicity by HCV are unclear. HCV could restrain the immune system and establish chronic infection by modulating dendritic cell (DC) function, T-cell function or both. DC dysfunction has been postulated to be either due to direct HCV infection or by the presence of HCV proteins. In this report, for the first time, we have examined whether soluble HCV proteins can impair DC function or directly inhibit T-cell responses in the cells obtained from healthy uninfected people. Our studies revealed that different HCV proteins used distinct mechanisms to down-regulate DC functions. Individual HCV proteins, Core, NS3, NS4, NS5 as well as fused Polyprotein (Core-NS3-NS4) were found to impair functions of both immature DCs and mature DCs by regulating the expression of co-stimulatory and antigen presentation molecules, strikingly reducing IL-12 secretion, inducing the expression of FasL to mediate apoptosis, interfering with allo-stimulatory capacity, inhibiting toll-like receptor signaling and inhibiting nuclear translocation of NFkappaB in DCs. Interestingly, HCV proteins did not directly inhibit T-cell proliferation. Our findings clearly demonstrate that HCV proteins impair T-cell responses indirectly by inhibiting DCs that could result in a sub-optimal cellular immune response allowing for persistent HCV infections. These studies delineate important mechanisms by which initial DC dysfunction can establish contributing to chronicity. Our data are in agreement with earlier observations that DCs are impaired in HCV infected people.
Collapse
Affiliation(s)
- Deepa K Krishnadas
- Department of Surgery, University of Alberta, Heritage Medical Research Centre, Edmonton, Canada
| | | | | | | | | |
Collapse
|
23
|
Masalova OV, Lesnova EI, Pichugin AV, Melnikova TM, Grabovetsky VV, Petrakova NV, Smirnova OA, Ivanov AV, Zaberezhny AD, Ataullakhanov RI, Isaguliants MG, Kushch AA. The successful immune response against hepatitis C nonstructural protein 5A (NS5A) requires heterologous DNA/protein immunization. Vaccine 2010; 28:1987-1996. [PMID: 20188254 DOI: 10.1016/j.vaccine.2009.10.097] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The aim of this study was to evaluate the immunogenicity of NS5A protein of human hepatitis C virus (HCV) when delivered as naked DNA (NS5A DNA), or recombinant protein (rNS5A). DBA/2J mice received NS5A DNA, rNS5A, or NS5A DNA/rNS5A in different prime-boost combinations with a peptidoglycan Immunomax((R)). The weakest response was induced after rNS5A prime and NS5A DNA boost; rNS5A alone induced an immune response with a strong Th2-component; and NS5A DNA alone, a relatively weak secretion of IL-2 and IFN-gamma. The most efficient was co-injection of NS5A DNA and rNS5A, which induced a significant increase in CD4(+) and CD8(+) T-cell counts, anti-NS5A antibodies, specific T-cell proliferation, and proinflammatory cytokine production in vitro against a broad spectrum of NS5A epitopes. Administration of the mixture of adjuvanted DNA and protein immunogens can be selected as the best regimen for further preclinical HCV-vaccine trials.
Collapse
Affiliation(s)
- Olga V Masalova
- D.I. Ivanovsky Institute of Virology, Russian Academy of Medical Sciences, Gamaleya str. 16, 123098 Moscow, Russian Federation.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Brunner R, Jensen-Jarolim E, Pali-Schöll I. The ABC of clinical and experimental adjuvants--a brief overview. Immunol Lett 2009; 128:29-35. [PMID: 19895847 DOI: 10.1016/j.imlet.2009.10.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2009] [Revised: 10/19/2009] [Accepted: 10/23/2009] [Indexed: 01/18/2023]
Abstract
Adjuvants are compounds that can increase and/or modulate the intrinsic immunogenicity of an antigen and elicit strong and long lasting immune responses. During the last 80 years many adjuvants have been used in experimental settings, but due to various shortcomings of most of them only aluminum compounds made it into regular clinical usage. However, during the last years promising candidates have arisen that may finally adjunct or displace aluminum substances as main adjuvant. This review summarizes information on adjuvants currently used in clinical as well as in experimental settings.
Collapse
Affiliation(s)
- Richard Brunner
- Department of Pathophysiology, Medical University of Vienna, Waehringer Guertel 18-20, AKH-EB 03.Q, Vienna, Austria
| | | | | |
Collapse
|
25
|
Koyama S, Coban C, Aoshi T, Horii T, Akira S, Ishii KJ. Innate immune control of nucleic acid-based vaccine immunogenicity. Expert Rev Vaccines 2009; 8:1099-1107. [PMID: 19627190 DOI: 10.1586/erv.09.57] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Optimal vaccine efficacy requires not only a protective antigen, but also a strong immune activator as an adjuvant. Most viral vaccines, such as influenza vaccines and nonviral genetic vaccines (e.g., DNA vaccines), contain nucleic acids, which appear to act as essential 'built-in' adjuvants. Specific receptors, including Toll-like receptors, retinoic acid-inducible protein-I-like receptors, and nucleotide-binding oligomerization domain-like receptors can detect specific nucleic acid patterns, depending on the immunized tissue, cell type and intracellular localization. The resulting immune activation is uniquely regulated by intra- and intercellular signaling pathways, which are indispensable for the ensuing vaccine immunogenicity, such as antigen-specific T- and B-cell responses. Thus, elucidation and manipulation of immune signaling and interactions by nucleic acid adjuvants are essential for maximizing the immunogenicity and safety of viral and DNA vaccines.
Collapse
Affiliation(s)
- Shohei Koyama
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | |
Collapse
|
26
|
De Gregorio E, D’Oro U, Wack A. Immunology of TLR-independent vaccine adjuvants. Curr Opin Immunol 2009; 21:339-45. [DOI: 10.1016/j.coi.2009.05.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Accepted: 05/04/2009] [Indexed: 12/13/2022]
|
27
|
O’Hagan DT, De Gregorio E. The path to a successful vaccine adjuvant – ‘The long and winding road’. Drug Discov Today 2009; 14:541-51. [DOI: 10.1016/j.drudis.2009.02.009] [Citation(s) in RCA: 200] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 02/19/2009] [Accepted: 02/23/2009] [Indexed: 11/28/2022]
|
28
|
Argentini C, Genovese D, Dettori S, Rapicetta M. HCV genetic variability: from quasispecies evolution to genotype classification. Future Microbiol 2009; 4:359-73. [PMID: 19327119 DOI: 10.2217/fmb.09.8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
HCV is a ssRNA virus belonging to the Flaviviruses and is found worldwide worldwide in humans. Following primary infection, persistent infection develops in more than 85% of cases, which in up to 30% of cases, may progress to liver disease, cirrhosis and hepatocellular carcinoma. The virus presents a high degree of genetic variability owing to the combination of a lack of proofreading by the RNA-dependent RNA polymerase and a high level of viral replication. This genetic variability allows the classification of genotypes, subtypes, isolates and quasispecies to which epidemiological and pathogenetic significance may be associated. The features and biological implications of HCV variability and of quasispecies dynamics in infection transmission, mechanisms of chronicity and resistance to antiviral therapy are discussed.
Collapse
Affiliation(s)
- Claudio Argentini
- Viral Hepatitis Unit, Department of Infectious, Parasitic & Immune-Mediated Disease, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy.
| | | | | | | |
Collapse
|
29
|
Caputo A, Castaldello A, Brocca-Cofano E, Voltan R, Bortolazzi F, Altavilla G, Sparnacci K, Laus M, Tondelli L, Gavioli R, Ensoli B. Induction of humoral and enhanced cellular immune responses by novel core-shell nanosphere- and microsphere-based vaccine formulations following systemic and mucosal administration. Vaccine 2009; 27:3605-15. [PMID: 19464541 DOI: 10.1016/j.vaccine.2009.03.047] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 03/12/2009] [Accepted: 03/17/2009] [Indexed: 10/20/2022]
Abstract
Anionic surfactant-free polymeric core-shell nanospheres and microspheres were previously described with an inner core constituted by poly(methylmethacrylate) (PMMA) and a highly hydrophilic outer shell composed of a hydrosoluble co-polymer (Eudragit L100-55). The outer shell is tightly linked to the core and bears carboxylic groups capable of adsorbing high amounts (antigen loading ability of up to 20%, w/w) of native basic proteins, mainly by electrostatic interactions, while preserving their activity. In the present study we have evaluated in mice the safety and immunogenicity of new vaccine formulations composed of these nano- and microspheres and the HIV-1 Tat protein. Vaccines were administered by different routes, including intramuscular, subcutaneous or intranasal and the results were compared to immunization with Tat alone or with Tat delivered with the alum adjuvant. The data demonstrate that the nano- and microspheres/Tat formulations are safe and induce robust and long-lasting cellular and humoral responses in mice after systemic and/or mucosal immunization. These delivery systems may have great potential for novel Tat protein-based vaccines against HIV-1 and hold promise for other protein-based vaccines.
Collapse
Affiliation(s)
- Antonella Caputo
- Department of Histology, University of Padova, Via A. Gabelli 63, 35122 Padova, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Itoh K, Yamada A, Mine T, Noguchi M. Recent advances in cancer vaccines: an overview. Jpn J Clin Oncol 2008; 39:73-80. [PMID: 19015149 DOI: 10.1093/jjco/hyn132] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The field of cancer vaccines is currently in an active state of clinical investigations. Human papilloma virus vaccine has been approved as a prophylactic cancer vaccine, while Oncophage (heat shock protein-peptide complex) was recently approved in Russia for a certain stage of kidney cancer, although to date none has been approved in Japan or the USA. We reviewed recent clinical trials of several different types of cancer vaccines, mainly by using PubMed from 2005 to 2008. There have been slow but substantial advances in peptide vaccines and dendritic cell-based vaccines with regard to both clinical responses and immunological markers. A personalized approach to boost immune responses, addition of chemotherapy to overcome robust cancers and changing of endpoints from tumor reduction to overall survival seem to be the three key elements for the development of therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Kyogo Itoh
- Department of Immunology and Immunotherapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka 830-0011, Japan.
| | | | | | | |
Collapse
|
31
|
De Gregorio E, Tritto E, Rappuoli R. Alum adjuvanticity: unraveling a century old mystery. Eur J Immunol 2008; 38:2068-71. [PMID: 18651701 DOI: 10.1002/eji.200838648] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The development of vaccine adjuvants for human use has been one of the slowest processes in the history of medicine. For almost one century, aluminium hydroxide (alum) has been the only vaccine adjuvant approved worldwide. Only in the past decade have two oil-in-water emulsions and one TLR agonist been approved by the European authorities as new vaccine adjuvants. Despite the fact that alum has been injected into billions of people, its mechanism of action is not fully understood. Recently, several reports have greatly increased our knowledge of the molecular and cellular events triggered by alum; however, the contribution of each of these processes to alum adjuvanticity is still unclear. A study published in this issue of the European Journal of Immunology, together with two recent publications, have demonstrated that the NOD-like receptor, pyrin domain containing 3 (Nlrp3)-inflammasome is the molecular target of alum immunostimulatory activity in vitro. Surprisingly, these three studies reported conflicting results on the requirement of the Nlrp3 inflammasome complex for alum adjuvant effects in vivo. This commentary attempts to resolve some of these discrepancies.
Collapse
Affiliation(s)
- Ennio De Gregorio
- Department of Immunology, Novartis Vaccines & Diagnostics, Siena, Italy.
| | | | | |
Collapse
|
32
|
Abstract
Oil-in-water emulsions are potent human adjuvants used for effective pandemic influenza vaccines; however, their mechanism of action is still unknown. By combining microarray and immunofluorescence analysis, we monitored the effects of the adjuvants MF59 oil-in-water emulsion, CpG, and alum in the mouse muscle. MF59 induced a time-dependent change in the expression of 891 genes, whereas CpG and alum regulated 387 and 312 genes, respectively. All adjuvants modulated a common set of 168 genes and promoted antigen-presenting cell recruitment. MF59 was the stronger inducer of cytokines, cytokine receptors, adhesion molecules involved in leukocyte migration, and antigen-presentation genes. In addition, MF59 triggered a more rapid influx of CD11b+ blood cells compared with other adjuvants. The early biomarkers selected by microarray, JunB and Ptx3, were used to identify skeletal muscle as a direct target of MF59. We propose that oil-in-water emulsions are the most efficient human vaccine adjuvants, because they induce an early and strong immunocompetent environment at the injection site by targeting muscle cells.
Collapse
|
33
|
Strickland GT, El-Kamary SS, Klenerman P, Nicosia A. Hepatitis C vaccine: supply and demand. THE LANCET. INFECTIOUS DISEASES 2008; 8:379-86. [DOI: 10.1016/s1473-3099(08)70126-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
34
|
Induction of broad CD4+ and CD8+ T-cell responses and cross-neutralizing antibodies against hepatitis C virus by vaccination with Th1-adjuvanted polypeptides followed by defective alphaviral particles expressing envelope glycoproteins gpE1 and gpE2 and nonstructural proteins 3, 4, and 5. J Virol 2008; 82:7492-503. [PMID: 18508900 DOI: 10.1128/jvi.02743-07] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Broad, multispecific CD4(+) and CD8(+) T-cell responses to the hepatitis C virus (HCV), as well as virus-cross-neutralizing antibodies, are associated with recovery from acute infection and may also be associated in chronic HCV patients with a favorable response to antiviral treatment. In order to recapitulate all of these responses in an ideal vaccine regimen, we have explored the use of recombinant HCV polypeptides combined with various Th1-type adjuvants and replication-defective alphaviral particles encoding HCV proteins in various prime/boost modalities in BALB/c mice. Defective chimeric alphaviral particles derived from the Sindbis and Venezuelan equine encephalitis viruses encoding either the HCV envelope glycoprotein gpE1/gpE2 heterodimer (E1E2) or nonstructural proteins 3, 4, and 5 (NS345) elicited strong CD8(+) T-cell responses but low CD4(+) T helper responses to these HCV gene products. In contrast, recombinant E1E2 glycoproteins adjuvanted with MF59 containing a CpG oligonucleotide elicited strong CD4(+) T helper responses but no CD8(+) T-cell responses. A recombinant NS345 polyprotein also stimulated strong CD4(+) T helper responses but no CD8(+) T-cell responses when adjuvanted with Iscomatrix containing CpG. Optimal elicitation of broad CD4(+) and CD8(+) T-cell responses to E1E2 and NS345 was obtained by first priming with Th1-adjuvanted proteins and then boosting with chimeric, defective alphaviruses expressing these HCV genes. In addition, this prime/boost regimen resulted in the induction of anti-E1E2 antibodies capable of cross-neutralizing heterologous HCV isolates in vitro. This vaccine formulation and regimen may therefore be optimal in humans for protection against this highly heterogeneous global pathogen.
Collapse
|
35
|
Higgins D, Marshall JD, Traquina P, Van Nest G, Livingston BD. Immunostimulatory DNA as a vaccine adjuvant. Expert Rev Vaccines 2007; 6:747-59. [PMID: 17931155 DOI: 10.1586/14760584.6.5.747] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Immunostimulatory DNA containing unmethylated CpG motifs is recognized by Toll-like receptor 9, resulting in the activation of innate immune responses that subsequently amplify the adaptive-immune response. Advances in the characterization of Toll-like receptor 9 signaling have identified immunostimulatory sequences (ISS) with distinct biological activities. Numerous animal models have demonstrated that synthetic ISS are effective adjuvants that enhance both humoral and cellular immune responses in diverse indications, ranging from infectious disease to cancer and allergy. An added benefit supporting the use of ISS as a vaccine adjuvant is that the specific activation of a pathway critical to the regulation of the immune response results in minimal toxicity. To date, clinical testing has largely affirmed the potency and safety of ISS-adjuvanted vaccines.
Collapse
Affiliation(s)
- Debbie Higgins
- Preclinical Research, Dynavax Technologies, 2929 Seventh Street, Suite 100, Berkeley, CA 94710, USA.
| | | | | | | | | |
Collapse
|
36
|
Abstract
Developing efficient and safe adjuvants for use in human vaccines remains both a challenge and a necessity. Past approaches have been largely empirical and generally used a single type of adjuvant, such as aluminium salts or emulsions. However, new vaccine targets often require the induction of well-defined cell-mediated responses in addition to antibodies, and thus new immunostimulants are required. Recent advances in basic immunology have elucidated how early innate immune signals can shape subsequent adaptive responses and this, coupled with improvements in biochemical techniques, has led to the design and development of more specific and focused adjuvants. In this Review, I discuss the research that has made it possible for vaccinologists to now be able to choose between a large panel of adjuvants, which potentially can act synergistically, and combine them in formulations that are specifically adapted to each target and to the relevant correlate(s) of protection.
Collapse
Affiliation(s)
- Bruno Guy
- Research Department, sanofi pasteur, Campus Merieux, 69280 Marcy l'Etoile, France.
| |
Collapse
|