1
|
Zubair A, Al-Emam A, Ali M, Hussain SM, Elmagzoub RM. Targeting HIV-1 conserved regions: An immunoinformatic pathway to vaccine innovation for the Asia. PLoS One 2025; 20:e0317382. [PMID: 40117271 PMCID: PMC11927918 DOI: 10.1371/journal.pone.0317382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/26/2024] [Indexed: 03/23/2025] Open
Abstract
A combination of humoral and cell-mediated immune system stimulation is essential for developing an effective HIV vaccine. Traditional treatment options and the challenges posed by drug resistance necessitate the discovery of a viable vaccine candidate capable of eliciting a robust immunological response. This research aims to develop an HIV vaccine with a multi-epitope component using a unique immunoinformatics approach. A subunit vaccine comprising B-cell, helper T-cell, and cytotoxic T-cell epitopes, along with appropriate adjuvants and linkers, was employed to identify conserved regions in the Pol, Vpr, Gag, Tat, Env, Nef, and Vif proteins. The HIV subunit vaccine demonstrated the potential to activate both cell-mediated and humoral immune responses, indicating its immunogenicity. The application of homology modeling and refinement further enhanced the model's accuracy. Subsequently, the molecular docking procedure utilized the refined model structure to bind to the immunological receptor TLR-3 in lymphocyte cells. Following this, the potential interactions of the subunit vaccine with TLR-3 were investigated using molecular dynamics modeling. The vaccine's stability was improved through a meticulous disulfide engineering technique that involved inserting cysteine residues into highly flexible regions. Finally, in silico cloning was employed to validate the efficacy of translating and producing the vaccine in a microbiological setting. The vaccine shows promising results in terms of population coverage, reaching 82% of the global population, with extraordinary efficacy in Asia, covering up to 95% of the population. Our HIV vaccine candidate is highly stable and elicits a robust immune response against HIV-1.
Collapse
Affiliation(s)
- Akmal Zubair
- Department of Biotechnology, Quaid-i-Azam University Islamabad, Pakistan
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir , Saudi Arabia
| | - Muhammad Ali
- Department of Biotechnology, Quaid-i-Azam University Islamabad, Pakistan
| | - Syeda Maryam Hussain
- Department of Livestock Production and Management, Faculty of Veterinary and Animal Sciences PIR Mehr Ali Shah-Arid Agriculture University Rawalpindi, Shamsabad, Murree Road, Pakistan
| | - Ranya Mohammed Elmagzoub
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
2
|
Babutzka S, Gehrke M, Papadopoulou A, Diedrichs-Möhring M, Giannaki M, Hennis L, Föhr B, Kooyman C, Osterman A, Yannaki E, Wildner G, Ammer H, Michalakis S. A novel platform for engineered AAV-based vaccines. Mol Ther Methods Clin Dev 2025; 33:101418. [PMID: 40008090 PMCID: PMC11850754 DOI: 10.1016/j.omtm.2025.101418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025]
Abstract
Engineering of adeno-associated virus (AAV) capsids allowed for the development of gene therapy vectors with improved tropism and enhanced transduction efficiency. Capsid engineering can also be used to adapt the AAV technology for applications outside gene therapy. Here, we investigated modified AAV capsids as scaffolds for the presentation of large immunogenic antigens to elicit a strong and specific immune response against pathogens. Using SARS-CoV-2 as a model pathogen, we introduced ∼200 amino acids of the SARS-CoV-2 receptor-binding domain (RBD) into a surface-exposed variable loop region of AAV2 and AAV9, resulting in AAV2.RBD and AAV9.RBD capsids (AAV.RBDs). This engineering endowed AAV.RBDs with SARS-CoV-2-like properties, such as angiotensin-converting enzyme 2 receptor affinity. In line with this, AAV.RBDs were neutralized by sera from human donors vaccinated against SARS-CoV-2. When administered subcutaneously to rabbits, AAV.RBDs elicited a strong humoral response against SARS-CoV-2 RBD. Moreover, the AAV.RBDs were able to trigger RBD-specific cellular immune responses in peripheral human lymphocytes. In conclusion, this novel AAV-based next-generation vaccine platform allows for the presentation of large antigenic sequences to elicit strong and specific immune responses. This versatile vaccine technology could be explored in the context of diseases where conventional immunization approaches have been unsuccessful.
Collapse
Affiliation(s)
- Sabrina Babutzka
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Miranda Gehrke
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Anastasia Papadopoulou
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 570 10 Thessaloniki, Greece
| | | | - Maria Giannaki
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 570 10 Thessaloniki, Greece
| | - Lena Hennis
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Bastian Föhr
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Cale Kooyman
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Andreas Osterman
- Max Von Pettenkofer Institute and Gene Center, Virology, LMU Munich, 80336 Munich, Germany
| | - Evangelia Yannaki
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 570 10 Thessaloniki, Greece
- Department of Medicine, University of Washington, Seattle, WA 91895, USA
| | - Gerhild Wildner
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Hermann Ammer
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| |
Collapse
|
3
|
Wei L, Yu P, Wang H, Liu J. Adeno-associated viral vectors deliver gene vaccines. Eur J Med Chem 2025; 281:117010. [PMID: 39488197 DOI: 10.1016/j.ejmech.2024.117010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Adeno-associated viruses (AAVs) are leading platforms for in vivo delivery of gene therapies, with six licensed AAV-based therapeutics attributed to their non-pathogenic nature, low immunogenicity, and high efficiency. In the realm of gene-based vaccines, one of the most vital therapeutic areas, AAVs are also emerging as promising delivery tools. We scrutinized AAVs, focusing on their virological properties, as well as bioengineering and chemical modifications to demonstrate their significant potential in gene vaccine delivery, and detailing the preparation of AAV particles. Additionally, we summarized the use of AAV vectors in vaccines for both infectious and non-infectious diseases, such as influenza, COVID-19, Alzheimer's disease, and cancer. Furthermore, this review, along with the latest clinical trial updates, provides a comprehensive overview of studies on the potential of using AAV vectors for gene vaccine delivery. It aims to deepen our understanding of the challenges and limitations in nucleic acid delivery and pave the way for future clinical success.
Collapse
Affiliation(s)
- Lai Wei
- College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Peng Yu
- College of Biotechnology, Tianjin University of Science & Technology, 300457 Tianjin, China
| | - Haomeng Wang
- CanSino (Shanghai) Biological Research Co., Ltd, 201208, Shanghai, China.
| | - Jiang Liu
- Rosalind Franklin Institute, Harwell Campus, OX11 0QS, Oxford, United Kingdom; Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT, Oxford, United Kingdom.
| |
Collapse
|
4
|
Filipić B, Pantelić I, Nikolić I, Majhen D, Stojić-Vukanić Z, Savić S, Krajišnik D. Nanoparticle-Based Adjuvants and Delivery Systems for Modern Vaccines. Vaccines (Basel) 2023; 11:1172. [PMID: 37514991 PMCID: PMC10385383 DOI: 10.3390/vaccines11071172] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Ever since the development of the first vaccine, vaccination has had the great impact on global health, leading to the decrease in the burden of numerous infectious diseases. However, there is a constant need to improve existing vaccines and develop new vaccination strategies and vaccine platforms that induce a broader immune response compared to traditional vaccines. Modern vaccines tend to rely on certain nanotechnology platforms but are still expected to be readily available and easy for large-scale manufacturing and to induce a durable immune response. In this review, we present an overview of the most promising nanoadjuvants and nanoparticulate delivery systems and discuss their benefits from tehchnological and immunological standpoints as well as their objective drawbacks and possible side effects. The presented nano alums, silica and clay nanoparticles, nanoemulsions, adenoviral-vectored systems, adeno-associated viral vectors, vesicular stomatitis viral vectors, lentiviral vectors, virus-like particles (including bacteriophage-based ones) and virosomes indicate that vaccine developers can now choose different adjuvants and/or delivery systems as per the requirement, specific to combatting different infectious diseases.
Collapse
Affiliation(s)
- Brankica Filipić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ivana Pantelić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ines Nikolić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
- Section of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
| | - Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Snežana Savić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Danina Krajišnik
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| |
Collapse
|
5
|
Shi C, Tian L, Zheng W, Zhu Y, Sun P, Liu L, Liu W, Song Y, Xia X, Xue X, Zheng X. Recombinant adeno-associated virus serotype 9 AAV-RABVG expressing a Rabies Virus G protein confers long-lasting immune responses in mice and non-human primates. Emerg Microbes Infect 2022; 11:1439-1451. [PMID: 35579916 PMCID: PMC9154782 DOI: 10.1080/22221751.2022.2078226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Three or four intramuscular doses of the inactivated human rabies virus vaccines are needed for pre- or post-exposure prophylaxis in humans. This procedure has made a great contribution to prevent human rabies deaths, which bring huge economic burdens in developing countries. Herein, a recombinant adeno-associated virus serotype 9, AAV9-RABVG, harbouring a RABV G gene, was generated to serve as a single dose rabies vaccine candidate. The RABV G protein was stably expressed in the 293T cells infected with AAV9-RABVG. A single dose of 2 × 1011 v.p. of AAV9-RABVG induced robust and long-term positive seroconversions in BALB/c mice with a 100% survival from a lethal RABV challenge. In Cynomolgus Macaques vaccinated with a single dose of 1 × 1013 v.p. of AAV9-RABVG, the titres of rabies VNAs increased remarkably from 2 weeks after immunity, and maintained over 31.525 IU/ml at 52 weeks. More DCs were activated significantly for efficient antigen presentations of RABV G protein, and more B cells were activated to be responsible for antibody responses. Significantly more RABV G specific IFN-γ-secreting CD4+ and CD8+ T cells, and IL-4-secreting CD4+ T cells were activated, and significantly higher levels of IL-2, IFN-γ, IL-4, and IL-10 were secreted to aid immune responses. Overall, the AAV9-RABVG was a single dose rabies vaccine candidate with great promising by inducing robust, long-term humoral responses and both Th1 and Th2 cell-mediated immune responses in mice and non-human primates.
Collapse
Affiliation(s)
- Chenjuan Shi
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Li Tian
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Wenwen Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yelei Zhu
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.,Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, People's Republic of China
| | - Peilu Sun
- Institute of Materia Medical, Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Lele Liu
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Wenkai Liu
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yanyan Song
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Xianzhu Xia
- Institute of Military Veterinary Medicine, Academy of Military Medical Science, Changchun, People's Republic of China
| | - Xianghong Xue
- Divisions of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, People's Republic of China
| | - Xuexing Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
6
|
Intranasal application of adeno-associated viruses: a systematic review. Transl Res 2022; 248:87-110. [PMID: 35597541 DOI: 10.1016/j.trsl.2022.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/12/2022] [Accepted: 05/16/2022] [Indexed: 01/13/2023]
Abstract
Adeno-associated viruses (AAVs) represent some of the most commonly employed vectors for targeted gene delivery and their extensive study has resulted in the approval of multiple gene therapies to treat human diseases. The intranasal route of vector application in gene therapy offers several advantages over traditional ways of administration. In addition to targeting local tissue like the olfactory epithelium, it provides minimally invasive access to various organ systems, including the central nervous system and the respiratory tract. Through a systematic literature review, a total of 53 articles that investigated the intranasal application of AAVs were identified, included, and summarized in this manuscript. Within these studies, AAV-based gene therapy was mainly investigated for its application in various infectious, pulmonary, or neurologic and/or psychiatric diseases. This review gives a comprehensive overview of the current technological state of the art regarding the intranasal application of AAVs for gene transfer and discusses remaining hurdles, which still have to be resolved before this approach can effectively be implemented in the routine clinical setting.
Collapse
|
7
|
Travieso T, Li J, Mahesh S, Mello JDFRE, Blasi M. The use of viral vectors in vaccine development. NPJ Vaccines 2022; 7:75. [PMID: 35787629 PMCID: PMC9253346 DOI: 10.1038/s41541-022-00503-y] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/15/2022] [Indexed: 12/22/2022] Open
Abstract
Vaccines represent the single most cost-efficient and equitable way to combat and eradicate infectious diseases. While traditional licensed vaccines consist of either inactivated/attenuated versions of the entire pathogen or subunits of it, most novel experimental vaccines against emerging infectious diseases employ nucleic acids to produce the antigen of interest directly in vivo. These include DNA plasmid vaccines, mRNA vaccines, and recombinant viral vectors. The advantages of using nucleic acid vaccines include their ability to induce durable immune responses, high vaccine stability, and ease of large-scale manufacturing. In this review, we present an overview of pre-clinical and clinical data on recombinant viral vector vaccines and discuss the advantages and limitations of the different viral vector platforms.
Collapse
Affiliation(s)
- Tatianna Travieso
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Jenny Li
- Duke University, Durham, NC, USA
| | - Sneha Mahesh
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Juliana Da Fonzeca Redenze E Mello
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Maria Blasi
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA. .,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
8
|
Sarangi D, Pattanaik S. Nanoparticles in dentistry. ADVANCED NANOMATERIALS FOR POINT OF CARE DIAGNOSIS AND THERAPY 2022:335-358. [DOI: 10.1016/b978-0-323-85725-3.00008-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Shabani SH, Kardani K, Milani A, Bolhassani A. In Silico and in Vivo Analysis of HIV-1 Rev Regulatory Protein for Evaluation of a Multiepitope-based Vaccine Candidate. Immunol Invest 2021; 51:1-28. [PMID: 33416004 DOI: 10.1080/08820139.2020.1867163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In silico-designed multiepitope conserved regions of human immunodeficiency virus 1 (HIV-1) proteins would be a beneficial strategy for antigen design which induces effective anti-HIV-1 T-cell responses. The conserved multiple HLA-DR-binding epitopes of Rev protein were identified using IEDB MHC-I prediction tools and SYFPEITHI webserver to screen potential T-cell epitopes. We analyzed toxicity, allergenicity, immunogenicity, hemolytic activity, cross-reactivity, cell-penetrating peptide (CPP) potency, and molecular docking of the candidate epitopes using several immune-informatics tools. Afterward, we designed a novel multiepitope construct based on non-toxic and non-allergenic Rev, Nef, Gp160 and P24-derived cytotoxic T cell (CTL) and T-helper cell (HTL) epitopes. Next, the designed construct (Nef-Rev-Gp160-P24) was subjected to three B-cell epitope prediction webservers, ProtParam and Protein-Sol to obtain the physicochemical features. Then, the recombinant multiepitope DNA and polypeptide constructs were complexed with different CPPs for nanoparticle formation and pass them via the cell membranes. Finally, the immunogenicity of multiepitope constructs in a variety of modalities was evaluated in mice. The results demonstrated that groups immunized with heterologous DNA+ MPG or HR9 CPP prime/rNef-Rev-Gp160-P24 polypeptide + LDP-NLS CPP boost regimens could significantly produce higher levels of IFN-γ and Granzyme B, and lower amounts of IL-10 than other groups. Moreover, higher levels of IgG2a and IgG2b were observed in all heterologous prime-boost regimens than homologous DNA or polypeptide regimens. Altogether, the present findings indicated that the Nef-Rev-Gp160-P24 polypeptide meets the criteria to be potentially useful as a multiepitope-based vaccine candidate against HIV-1 infection.
Collapse
Affiliation(s)
- Samaneh H Shabani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
10
|
Kardani K, Hashemi A, Bolhassani A. Comparative analysis of two HIV-1 multiepitope polypeptides for stimulation of immune responses in BALB/c mice. Mol Immunol 2020; 119:106-122. [DOI: 10.1016/j.molimm.2020.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/20/2022]
|
11
|
Yusuf Y, Yoshii T, Iyori M, Yoshida K, Mizukami H, Fukumoto S, Yamamoto DS, Alam A, Emran TB, Amelia F, Islam A, Otsuka H, Takashima E, Tsuboi T, Yoshida S. Adeno-Associated Virus as an Effective Malaria Booster Vaccine Following Adenovirus Priming. Front Immunol 2019; 10:730. [PMID: 31024558 PMCID: PMC6460511 DOI: 10.3389/fimmu.2019.00730] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
An ideal malaria vaccine platform should potently induce protective immune responses and block parasite transmission from mosquito to human, and it should maintain these effects for an extended period. Here, we have focused on vaccine development based on adeno-associated virus serotype 1 (AAV1), a viral vector widely studied in the field of clinical gene therapy that is able to induce long-term transgene expression without causing toxicity in vivo. Our results show the potential utility of AAV1 vectors as an extremely potent booster vaccine to induce durable immunity when combined with an adenovirus-priming vaccine in a rodent malaria model. We generated a series of recombinant AAV1s and human adenovirus type 5 (AdHu5) expressing either Plasmodium falciparum circumsporozoite protein (PfCSP) or P25 (Pfs25) protein. Heterologous two-dose immunization with an AdHu5-prime and AAV1-boost (AdHu5-AAV1) elicited robust and durable PfCSP- or Pfs25-specific functional antibodies over 280 days. Regarding protective efficacy, AdHu5-AAV1 PfCSP achieved high sterile protection (up to 80% protection rate) against challenge with transgenic Plasmodium berghei sporozoites expressing PfCSP. When examining transmission-blocking (TB) efficacy, we found that immunization with AdHu5-AAV1 Pfs25 maintained TB activity in vivo against transgenic P. berghei expressing Pfs25 for 287 days (99% reduction in oocyst intensity, 85% reduction in oocyst prevalence). Our data indicate that AAV1-based malaria vaccines can confer potent and durable protection as well as TB efficacy when administered following an AdHu5 priming vaccine, supporting the further evaluation of this regimen in clinical trials as a next-generation malaria vaccine platform.
Collapse
Affiliation(s)
- Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
- Department of Parasitology, Faculty of Medicine, University of Hasanuddin, Makassar, Indonesia
| | - Tatsuya Yoshii
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Kunitaka Yoshida
- Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroaki Mizukami
- Division of Gene therapy, Jichi Medical University, Shimotsuke, Japan
| | - Shinya Fukumoto
- National Research Centre for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Daisuke S. Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | - Asrar Alam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Hiromu Otsuka
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
12
|
Zhu F, Wang Y, Xu Z, Qu H, Zhang H, Niu L, Xue H, Jing D, He H. Novel adeno‑associated virus‑based genetic vaccines encoding hepatitis C virus E2 glycoprotein elicit humoral immune responses in mice. Mol Med Rep 2018; 19:1016-1023. [PMID: 30569131 PMCID: PMC6323296 DOI: 10.3892/mmr.2018.9739] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/20/2018] [Indexed: 12/15/2022] Open
Abstract
Hepatitis C virus (HCV) infection remains a major public health issue despite the introduction of several direct-acting antiviral agents (DAAs), with some 185 million individuals infected with HCV worldwide. There is an urgent need for an effective prophylactic HCV vaccine. In the present study, we constructed genetic vaccines based on novel recombinant adeno-associated viral (rAAV) vectors (AAV2/8 or AAV2/rh32.33) that express the envelope glycoprotein E2 from the HCV genotype 1b. Expression of HCV E2 protein in 293 cells was confirmed by western blot analysis. rAAV2/8.HCV E2 vaccine or rAAV2/rh32.33.HCV E2 vaccine was intramuscularly injected into C57BL/6 mice. HCV E2-specific antigen was produced, and long-lasting specific antibody responses remained detectable XVI weeks following immunization. In addition, the rAAV2/rh32.33 vaccine induced higher antigen-specific antibody levels than the rAAV2/8 vaccine or AAV plasmid. Moreover, both AAV vaccines induced neutralizing antibodies against HCV genotypes 1a and 1b. Finally, it is worth mentioning that neutralizing antibody levels directed against AAV2/rh32.33 were lower than those against AAV2/8 in both mouse and human serum. These results demonstrate that AAV vectors, especially the AAVrh32.33, have particularly favorable immunogenicity for development into an effective HCV vaccine.
Collapse
Affiliation(s)
- Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Yibo Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Zhen Xu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Haiyang Qu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Hairong Zhang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Lingling Niu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Honglu Xue
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Dehuai Jing
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Heng He
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| |
Collapse
|
13
|
Dadar M, Chakraborty S, Dhama K, Prasad M, Khandia R, Hassan S, Munjal A, Tiwari R, Karthik K, Kumar D, Iqbal HMN, Chaicumpa W. Advances in Designing and Developing Vaccines, Drugs and Therapeutic Approaches to Counter Human Papilloma Virus. Front Immunol 2018; 9:2478. [PMID: 30483247 PMCID: PMC6240620 DOI: 10.3389/fimmu.2018.02478] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/08/2018] [Indexed: 02/05/2023] Open
Abstract
Human papillomavirus (HPV) is a viral infection with skin-to-skin based transmission mode. HPV annually caused over 500,000 cancer cases including cervical, anogenital and oropharyngeal cancer among others. HPV vaccination has become a public-health concern, worldwide, to prevent the cases of HPV infections including precancerous lesions, cervical cancers, and genital warts especially in adolescent female and male population by launching national programs with international alliances. Currently, available prophylactic and therapeutic vaccines are expensive to be used in developing countries for vaccination programs. The recent progress in immunotherapy, biotechnology, recombinant DNA technology and molecular biology along with alternative and complementary medicinal systems have paved novel ways and valuable opportunities to design and develop effective prophylactic and therapeutic vaccines, drugs and treatment approach to counter HPV effectively. Exploration and more researches on such advances could result in the gradual reduction in the incidences of HPV cases across the world. The present review presents a current global scenario and futuristic prospects of the advanced prophylactic and therapeutic approaches against HPV along with recent patents coverage of the progress and advances in drugs, vaccines and therapeutic regimens to effectively combat HPV infections and its cancerous conditions.
Collapse
Affiliation(s)
- Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, West Tripura, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Minakshi Prasad
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar, India
| | - Rekha Khandia
- Department of Genetics, Barkatullah University, Bhopal, India
| | - Sameer Hassan
- Department of Biomedical Informatics, National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Ashok Munjal
- Department of Genetics, Barkatullah University, Bhopal, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, U P Pt. Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan, Mathura, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Hafiz M. N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, Mexico
| | - Wanpen Chaicumpa
- Department of Parasitology, Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine SIriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
14
|
Paulk NK, Pekrun K, Charville GW, Maguire-Nguyen K, Wosczyna MN, Xu J, Zhang Y, Lisowski L, Yoo B, Vilches-Moure JG, Lee GK, Shrager JB, Rando TA, Kay MA. Bioengineered Viral Platform for Intramuscular Passive Vaccine Delivery to Human Skeletal Muscle. Mol Ther Methods Clin Dev 2018; 10:144-155. [PMID: 30101152 PMCID: PMC6077147 DOI: 10.1016/j.omtm.2018.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 01/08/2023]
Abstract
Skeletal muscle is ideal for passive vaccine administration as it is easily accessible by intramuscular injection. Recombinant adeno-associated virus (rAAV) vectors are in consideration for passive vaccination clinical trials for HIV and influenza. However, greater human skeletal muscle transduction is needed for therapeutic efficacy than is possible with existing serotypes. To bioengineer capsids with therapeutic levels of transduction, we utilized a directed evolution approach to screen libraries of shuffled AAV capsids in pools of surgically resected human skeletal muscle cells from five patients. Six rounds of evolution were performed in various muscle cell types, and evolved variants were validated against existing muscle-tropic serotypes rAAV1, 6, and 8. We found that evolved variants NP22 and NP66 had significantly increased primary human and rhesus skeletal muscle fiber transduction from surgical explants ex vivo and in various primary and immortalized myogenic lines in vitro. Importantly, we demonstrated reduced seroreactivity compared to existing serotypes against normal human serum from 50 adult donors. These capsids represent powerful tools for human skeletal muscle expression and secretion of antibodies from passive vaccines.
Collapse
Affiliation(s)
- Nicole K. Paulk
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Katja Pekrun
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Gregory W. Charville
- Glenn Center for Biology of Aging and Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Katie Maguire-Nguyen
- Glenn Center for Biology of Aging and Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Michael N. Wosczyna
- Glenn Center for Biology of Aging and Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Jianpeng Xu
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Yue Zhang
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Leszek Lisowski
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Bryan Yoo
- Glenn Center for Biology of Aging and Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Gordon K. Lee
- Department of Surgery, Division of Plastic & Reconstructive Surgery, Stanford University, Stanford, CA 94305, USA
| | - Joseph B. Shrager
- Department of Cardiothoracic Surgery, Division of Thoracic Surgery, Stanford University and VA Palo Alto Health Care System, Stanford, CA 94305, USA
| | - Thomas A. Rando
- Glenn Center for Biology of Aging and Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Neurology Service and Rehabilitation Research and Development Center of Excellence, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Mark A. Kay
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
15
|
Brady JM, Baltimore D, Balazs AB. Antibody gene transfer with adeno-associated viral vectors as a method for HIV prevention. Immunol Rev 2017; 275:324-333. [PMID: 28133808 DOI: 10.1111/imr.12478] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Broadly neutralizing antibodies (bNAbs) against human immunodeficiency virus (HIV) show great promise in HIV prevention as they are capable of potently neutralizing a considerable breadth of genetically diverse strains. Passive transfer of monoclonal bNAb proteins can confer protection in animal models of HIV infection at modest concentrations, inspiring efforts to develop an HIV vaccine capable of eliciting bNAb responses. However, these antibodies demonstrate high degrees of somatic mutation and other unique characteristics that may hinder the ability of conventional approaches to consistently and effectively produce bNAb analogs. As an alternative strategy, we and others have proposed vector-mediated gene transfer to generate long-term, systemic production of bNAbs in the absence of immunization. Herein, we review the use of adeno-associated virus (AAV) vectors for delivery of HIV bNAbs and antibody-like proteins and summarize both the advantages and disadvantages of this strategy as a method for HIV prevention.
Collapse
Affiliation(s)
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | |
Collapse
|
16
|
Gupta G, Glueck R, Patel PR. HPV vaccines: Global perspectives. Hum Vaccin Immunother 2017; 13:1-4. [PMID: 28362244 PMCID: PMC5489288 DOI: 10.1080/21645515.2017.1289301] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/27/2017] [Indexed: 01/07/2023] Open
Abstract
The discovery of HPV as the etiological factor for HPV-associated malignancies and disease has opened up several opportunities for prevention and therapy. Current commercially available HPV vaccines (Gardasil, Gardasil 9, and Cervarix) are prophylactic in nature and derived from adjuvanted L1-based virus-like particles of HPV. Globally, through several clinical trials, they were found to be very safe and efficacious. Certain limitations such as cost-effectiveness, low coverage against all HPV types and a 3-dose schedule make these vaccines difficult to use worldwide. Approaches to address these issues involve alternate expression systems using L1 or alternate antigen (L2) as well as optimizing doses and broadening protection to provide cheap and cross-protective vaccines. Additionally, promising preclinical immunogenicity results from our own studies using alternative hosts such as Pichia and an antigen delivery system-based measles vector have potential for development as next generation HPV prophylactic vaccines. Several other therapeutic approaches are also ongoing.
Collapse
Affiliation(s)
- Gaurav Gupta
- Vaccine Technology Centre, Zydus Biologics Compound, Changodar, Ahmedabad, Gujarat, India
| | - Reinhard Glueck
- Vaccine Technology Centre, Zydus Biologics Compound, Changodar, Ahmedabad, Gujarat, India
| | - Pankaj R. Patel
- Vaccine Technology Centre, Zydus Biologics Compound, Changodar, Ahmedabad, Gujarat, India
| |
Collapse
|
17
|
Santiago-Ortiz JL, Schaffer DV. Adeno-associated virus (AAV) vectors in cancer gene therapy. J Control Release 2016; 240:287-301. [PMID: 26796040 PMCID: PMC4940329 DOI: 10.1016/j.jconrel.2016.01.001] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/08/2015] [Accepted: 01/02/2016] [Indexed: 02/06/2023]
Abstract
Gene delivery vectors based on adeno-associated virus (AAV) have been utilized in a large number of gene therapy clinical trials, which have demonstrated their strong safety profile and increasingly their therapeutic efficacy for treating monogenic diseases. For cancer applications, AAV vectors have been harnessed for delivery of an extensive repertoire of transgenes to preclinical models and, more recently, clinical trials involving certain cancers. This review describes the applications of AAV vectors to cancer models and presents developments in vector engineering and payload design aimed at tailoring AAV vectors for transduction and treatment of cancer cells. We also discuss the current status of AAV clinical development in oncology and future directions for AAV in this field.
Collapse
Affiliation(s)
- Jorge L Santiago-Ortiz
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA; Department of Bioengineering, University of California, Berkeley, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
18
|
Rodríguez-Gascón A, Del Pozo-Rodríguez A, Isla A, Solinís MA. Vaginal gene therapy. Adv Drug Deliv Rev 2015; 92:71-83. [PMID: 26189799 DOI: 10.1016/j.addr.2015.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 06/09/2015] [Accepted: 07/09/2015] [Indexed: 02/01/2023]
Abstract
In the last years, vaginal gene therapy has gained increasing attention mainly for the treatment and control of sexually transmitted infections. DNA delivery has been also suggested to improve reproductive outcomes for women with deficiencies in the female reproductive tract. Although no product has reached clinical phase, preclinical investigations reveal the potential of the vaginal tract as an effective administration route for gene delivery. This review focuses on the main advantages and challenges of vaginal gene therapy, and on the most used nucleic acid delivery systems, including viral and non-viral vectors. Additionally, the advances in the application of vaginal gene therapy for the treatment and/or prevention of infectious diseases such as the human immunodeficiency virus (HIV), the human papillomavirus (HPV) or the herpes simplex virus (HSV) are presented.
Collapse
Affiliation(s)
- Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain.
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain
| | - Arantxazu Isla
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain
| | - María Angeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
19
|
Kianmehr Z, Ardestani SK, Soleimanjahi H, Farahmand B, Abdoli A, Khatami M, Akbari K, Fotouhi F. An effective DNA priming-protein boosting approach for the cervical cancer vaccination. Pathog Dis 2014; 73:1-8. [DOI: 10.1093/femspd/ftu012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
20
|
Abstract
Human papillomaviruses (HPV) are the causative agents of cervical cancer, the third most common cancer in women. The development of prophylactic HPV vaccines Gardasil® and Cervarix® targeting the major oncogenic HPV types is now the frontline of cervical cancer prevention. Both vaccines have been proven to be highly effective and safe although there are still open questions about their target population, cross-protection, and long-term efficacy. The main limitation for a worldwide implementation of Gardasil® and Cervarix® is their high cost. To develop more affordable vaccines research groups are concentrated in new formulations with different antigens including capsomeres, the minor capsid protein L2 and DNA. In this article we describe the vaccines' impact on HPV-associated disease, the main open questions about the marketed vaccines, and current efforts for the development of second-generation vaccines.
Collapse
|
21
|
Cho H, Lee HJ, Heo YK, Cho Y, Gwon YD, Kim MG, Park KH, Oh YK, Kim YB. Immunogenicity of a trivalent human papillomavirus L1 DNA-encapsidated, non-replicable baculovirus nanovaccine. PLoS One 2014; 9:e95961. [PMID: 24759938 PMCID: PMC3997520 DOI: 10.1371/journal.pone.0095961] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 03/31/2014] [Indexed: 12/04/2022] Open
Abstract
Previously, we developed a non-replicating recombinant baculovirus coated with human endogenous retrovirus envelope protein (AcHERV) for enhanced cellular delivery of human papillomavirus (HPV) 16L1 DNA. Here, we report the immunogenicity of an AcHERV-based multivalent HPV nanovaccine in which the L1 segments of HPV 16, 18, and 58 genes were inserted into a single baculovirus genome of AcHERV. To test whether gene expression levels were affected by the order of HPV L1 gene insertion, we compared the efficacy of bivalent AcHERV vaccines with the HPV 16L1 gene inserted ahead of the 18L1 gene (AcHERV-HP16/18L1) with that of AcHERV with the HPV 18L1 gene inserted ahead of the 16L1 gene (AcHERV-HP18/16L1). Regardless of the order, the bivalent AcHERV DNA vaccines retained the immunogenicity of monovalent AcHERV-HP16L1 and AcHERV-HP18L1 DNA vaccines. Moreover, the immunogenicity of bivalent AcHERV-HP16/18L1 was not significantly different from that of AcHERV-HP18/16L1. In challenge tests, both bivalent vaccines provided complete protection against HPV 16 and 18 pseudotype viruses. Extending these results, we found that a trivalent AcHERV nanovaccine encoding HPV 16L1, 18L1, and 58L1 genes (AcHERV-HP16/18/58L1) provided high levels of humoral and cellular immunogenicity against all three subtypes. Moreover, mice immunized with the trivalent AcHERV-based nanovaccine were protected from challenge with HPV 16, 18, and 58 pseudotype viruses. These results suggest that trivalent AcHERV-HPV16/18/58L1 could serve as a potential prophylactic baculoviral nanovaccine against concurrent infection with HPV 16, 18, and 58.
Collapse
Affiliation(s)
- Hansam Cho
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Hee-Jung Lee
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Yoon-Ki Heo
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Yeondong Cho
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Yong-Dae Gwon
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Mi-Gyeong Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Ki Hoon Park
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- * E-mail: (YKO) or (YBK)
| | - Young Bong Kim
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
- * E-mail: (YKO) or (YBK)
| |
Collapse
|
22
|
Nieto K, Salvetti A. AAV Vectors Vaccines Against Infectious Diseases. Front Immunol 2014; 5:5. [PMID: 24478774 PMCID: PMC3896988 DOI: 10.3389/fimmu.2014.00005] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 01/07/2014] [Indexed: 12/12/2022] Open
Abstract
Since their discovery as a tool for gene transfer, vectors derived from the adeno-associated virus (AAV) have been used for gene therapy applications and attracted scientist to this field for their exceptional properties of efficiency of in vivo gene transfer and the level and duration of transgene expression. For many years, AAVs have been considered as low immunogenic vectors due to their ability to induce long-term expression of non-self-proteins in contrast to what has been observed with other viral vectors, such as adenovirus, for which strong immune responses against the same transgene products were documented. The perceived low immunogenicity likely explains why the use of AAV vectors for vaccination was not seriously considered before the early 2000s. Indeed, while analyses conducted using a variety of transgenes and animal species slowly changed the vision of immunological properties of AAVs, an increasing number of studies were also performed in the field of vaccination. Even if the comparison with other modes of vaccination was not systemically performed, the analyses conducted so far in the field of active immunotherapy strongly suggest that AAVs possess some interesting features to be used as tools to produce an efficient and sustained antibody response. In addition, recent studies also highlighted the potential of AAVs for passive immunotherapy. This review summarizes the main studies conducted to evaluate the potential of AAV vectors for vaccination against infectious agents and discusses their advantages and drawbacks. Altogether, the variety of studies conducted in this field contributes to the understanding of the immunological properties of this versatile virus and to the definition of its possible future applications.
Collapse
Affiliation(s)
- Karen Nieto
- Tumor Immunology Program (D030), German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Anna Salvetti
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon , Lyon , France ; LabEx Ecofect, Université de Lyon , Lyon , France
| |
Collapse
|
23
|
Vesicular transport of progeny parvovirus particles through ER and Golgi regulates maturation and cytolysis. PLoS Pathog 2013; 9:e1003605. [PMID: 24068925 PMCID: PMC3777860 DOI: 10.1371/journal.ppat.1003605] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/24/2013] [Indexed: 01/30/2023] Open
Abstract
Progeny particles of non-enveloped lytic parvoviruses were previously shown to be actively transported to the cell periphery through vesicles in a gelsolin-dependent manner. This process involves rearrangement and destruction of actin filaments, while microtubules become protected throughout the infection. Here the focus is on the intracellular egress pathway, as well as its impact on the properties and release of progeny virions. By colocalization with cellular marker proteins and specific modulation of the pathways through over-expression of variant effector genes transduced by recombinant adeno-associated virus vectors, we show that progeny PV particles become engulfed into COPII-vesicles in the endoplasmic reticulum (ER) and are transported through the Golgi to the plasma membrane. Besides known factors like sar1, sec24, rab1, the ERM family proteins, radixin and moesin play (an) essential role(s) in the formation/loading and targeting of virus-containing COPII-vesicles. These proteins also contribute to the transport through ER and Golgi of the well described analogue of cellular proteins, the secreted Gaussia luciferase in absence of virus infection. It is therefore likely that radixin and moesin also serve for a more general function in cellular exocytosis. Finally, parvovirus egress via ER and Golgi appears to be necessary for virions to gain full infectivity through post-assembly modifications (e.g. phosphorylation). While not being absolutely required for cytolysis and progeny virus release, vesicular transport of parvoviruses through ER and Golgi significantly accelerates these processes pointing to a regulatory role of this transport pathway. Previously, it was thought that non-enveloped lytic parvoviruses were released through a lytic burst of cells at the end of infection. However, recent work demonstrated that these small non-enveloped single-stranded DNA viruses are actively transported through vesicles from the nucleus, the site of replication and assembly, to the cell periphery. The current investigation demonstrates that progeny particles become engulfed into COPII-vesicles in the endoplasmic reticulum (ER) and are transported through the Golgi to the plasma membrane (PM). ERM family proteins radixin and moesin appear to play an essential role in this cellular secretion pathway. While passing through ER and Golgi cisternae, PVs maturate through post-assembly modifications, which significantly increase the infectivity of progeny virions. Finally, the vesicular transport of parvoviral particles was shown to regulate virus-induced cytolysis, thereby accelerating the further release and spread of progeny virions. As rodent PVs are currently viewed as oncolytic agents for cancer virotherapy, it is important to further investigate the mechanism of PV egress — not only to improve the spreading of these agents through the tumor mass, but also to optimize the induction of an anti-tumor immune response upon virus — induced cytolysis.
Collapse
|
24
|
|
25
|
Ploquin A, Szécsi J, Mathieu C, Guillaume V, Barateau V, Ong KC, Wong KT, Cosset FL, Horvat B, Salvetti A. Protection against henipavirus infection by use of recombinant adeno-associated virus-vector vaccines. J Infect Dis 2012; 207:469-78. [PMID: 23175762 PMCID: PMC7107322 DOI: 10.1093/infdis/jis699] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Nipah virus (NiV) and Hendra virus (HeV) are closely related, recently emerged paramyxoviruses that are capable of causing considerable morbidity and mortality in several mammalian species, including humans. Henipavirus-specific vaccines are still commercially unavailable, and development of novel antiviral strategies to prevent lethal infections due to henipaviruses is highly desirable. Here we describe the development of adeno-associated virus (AAV) vaccines expressing the NiV G protein. Characterization of these vaccines in mice demonstrated that a single intramuscular AAV injection was sufficient to induce a potent and long-lasting antibody response. Translational studies in hamsters further demonstrated that all vaccinated animals were protected against lethal challenge with NiV. In addition, this vaccine induced a cross-protective immune response that was able to protect 50% of the animals against a challenge by HeV. This study presents a new efficient vaccination strategy against henipaviruses and opens novel perspectives on the use of AAV vectors as vaccines against emergent diseases.
Collapse
Affiliation(s)
- Aurélie Ploquin
- INSERM U758, 2Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Schiller JT, Castellsagué X, Garland SM. A review of clinical trials of human papillomavirus prophylactic vaccines. Vaccine 2012; 30 Suppl 5:F123-38. [PMID: 23199956 PMCID: PMC4636904 DOI: 10.1016/j.vaccine.2012.04.108] [Citation(s) in RCA: 558] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 04/18/2012] [Accepted: 04/20/2012] [Indexed: 02/06/2023]
Abstract
End of study analyses of the phase III trials of prophylactic human papillomavirus (HPV) virus-like particle (VLP) vaccines in young women are now largely completed. Two distinct vaccines were evaluated, Gardasil(®) (Merck & Co., Whitehouse Station, NJ USA) a quadrivalent vaccine containing VLPs of types 6, 11, 16 and 18 and Cervarix(®) (GlaxoSmithKline Biologicals, Rixensart, Belgium), a bivalent vaccine containing VLPs of types 16 and 18. Both vaccines exhibited excellent safety and immunogenicity profiles. The vaccines also demonstrated remarkably high and similar efficacy against the vaccine-targeted types for a range of cervical endpoints from persistent infection to cervical intraepithelial neoplasia grade 3 (CIN3) in women naïve to the corresponding type at the time of vaccination. However, protection from incident infection or disease from non-vaccine types was restricted, and the vaccines had no effect on prevalent infection or disease. Gardasil(®) also demonstrated strong protection against genital warts and vulvar/vaginal neoplasia associated with the vaccine types. In other trials, Gardasil(®) protected mid-adult women from incident infection and CIN caused by the vaccine types and protected men for incident infection, genital warts and anal intraepithelial neoplasia by the vaccine types. Cervarix(®) protected against vaccine-targeted anal infections in women in an end of study evaluation. For practical reasons, efficacy studies have not been conducted in the primary target populations of current vaccination programs, adolescent girls and boys. However, immunogenicity bridging studies demonstrating excellent safety and strong immune responses in adolescence, coupled with the documentation of durable antibody responses and protection in young adults, leads to an optimistic projection of the effectiveness of the vaccines in adolescent vaccination programs. Taken together, the excellent clinical trial results strongly support the potential of the vaccines as high value public health interventions and justify their widespread implementation to prevent anogenital HPV infections and their associated neoplasia. This article forms part of a special supplement entitled "Comprehensive Control of HPV Infections and Related Diseases" Vaccine Volume 30, Supplement 5, 2012.
Collapse
Affiliation(s)
- John T Schiller
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | | | |
Collapse
|
27
|
Nieto K, Weghofer M, Sehr P, Ritter M, Sedlmeier S, Karanam B, Seitz H, Müller M, Kellner M, Hörer M, Michaelis U, Roden RBS, Gissmann L, Kleinschmidt JA. Development of AAVLP(HPV16/31L2) particles as broadly protective HPV vaccine candidate. PLoS One 2012; 7:e39741. [PMID: 22761884 PMCID: PMC3384601 DOI: 10.1371/journal.pone.0039741] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 05/25/2012] [Indexed: 12/21/2022] Open
Abstract
The human papillomavirus (HPV) minor capsid protein L2 is a promising candidate for a broadly protective HPV vaccine yet the titers obtained in most experimental systems are rather low. Here we examine the potential of empty AAV2 particles (AAVLPs), assembled from VP3 alone, for display of L2 epitopes to enhance their immunogenicity. Insertion of a neutralizing epitope (amino acids 17–36) from L2 of HPV16 and HPV31 into VP3 at positions 587 and 453, respectively, permitted assembly into empty AAV particles (AAVLP(HPV16/31L2)). Intramuscularly vaccination of mice and rabbits with AAVLP(HPV16/31L2)s in montanide adjuvant, induced high titers of HPV16 L2 antibodies as measured by ELISA. Sera obtained from animals vaccinated with the AAVLP(HPV16/31L2)s neutralized infections with several HPV types in a pseudovirion infection assay. Lyophilized AAVLP(HPV16/31L2) particles retained their immunogenicity upon reconstitution. Interestingly, vaccination of animals that were pre-immunized with AAV2 - simulating the high prevalence of AAV2 antibodies in the population - even increased cross neutralization against HPV31, 45 and 58 types. Finally, passive transfer of rabbit antisera directed against AAVLP(HPV16/31L2)s protected naïve mice from vaginal challenge with HPV16 pseudovirions. In conclusion, AAVLP(HPV16/31L2) particles have the potential as a broadly protective vaccine candidate regardless of prior exposure to AAV.
Collapse
Affiliation(s)
- Karen Nieto
- Research Program Infection and Cancer, German Cancer Research Center, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Pilz IH, Di Pasquale G, Rzadzinska A, Leppla SH, Chiorini JA. Mutation in the platelet-derived growth factor receptor alpha inhibits adeno-associated virus type 5 transduction. Virology 2012; 428:58-63. [PMID: 22520943 DOI: 10.1016/j.virol.2012.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 11/21/2011] [Accepted: 03/05/2012] [Indexed: 01/01/2023]
Abstract
Due to its non-pathogenic lifecycle, little is known about the cellular determinants of infection by adeno-associated virus (AAV). To identify these critical cellular factors, we took advantage of the gene transfer abilities of AAV in combination with a forward genetic selection to identify proteins critical for transduction by this virus. AAV serotype 5 (AAV5) vectors encoding the furin gene were used to transduce furin-deficient cells followed by selection with furin-dependent toxins. A population of cells specifically resistant to AAV5 transduction was identified and sequence analysis suggested all had a single amino acid mutation in the leader sequence of the platelet-derived growth factor receptor alpha (PDGFRα) gene. Characterization of this mutation suggested it inhibited PDGFRα trafficking resulting in limited expression on the plasma membrane. Mutagenesis and transfection experiments confirmed the effect of this mutation on PDGFRα trafficking, and the AAV5 resistant phenotype could be rescued by transfection with wild type PDGFRα.
Collapse
Affiliation(s)
- Ingo H Pilz
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
29
|
Nieto K, Stahl-Hennig C, Leuchs B, Müller M, Gissmann L, Kleinschmidt JA. Intranasal vaccination with AAV5 and 9 vectors against human papillomavirus type 16 in rhesus macaques. Hum Gene Ther 2012; 23:733-41. [PMID: 22401308 DOI: 10.1089/hum.2011.202] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cervical cancer is the second most common cancer in women worldwide. Persistent high-risk human papillomavirus (HPV) infection has been identified as the causative event for the development of this type of cancer. Recombinant adeno-associated viruses (rAAVs) are currently being developed and evaluated as vaccine vector. In previous work, we demonstrated that rAAVs administered intranasally in mice induced high titers and long-lasting neutralizing antibodies against HPV type 16 (HPV16). To extend this approach to a more human-related species, we immunized rhesus macaques (Macaca mulatta) with AAVs expressing an HPV16 L1 protein using rAAV5 and 9 vectors in an intranasal prophylactic setting. An rAAV5-L1 vector followed by a boost with rAAV9-L1 induced higher titers of L1-specific serum antibodies than a single rAAV5-L1 immunization. L1-specific antibodies elicited by AAV9 vector neutralized HPV16 pseudovirions and persisted for at least 7 months post immunization. Interestingly, nasal application of rAAV9 was immunogenic even in the presence of high AAV9 antibody titers, allowing reimmunization with the same serotype without prevention of the transgene expression. Two of six animals did not respond to AAV-mediated intranasal vaccination, although they were not tolerant, as both developed antibodies after intramuscular vaccination with HPV16 virus-like particles. These data clearly show the efficacy of an intranasal immunization using rAAV9-L1 vectors without the need of an adjuvant. We conclude from our results that rAAV9 vector is a promising candidate for a noninvasive nasal vaccination strategy.
Collapse
Affiliation(s)
- Karen Nieto
- Research Program Infection and Cancer, German Cancer Research Center, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
30
|
Infante-Bourzac JF, Sifontes-Rodríguez S, Arencibia-Arrebola DF, Hernández-Salazar T, Fariñas-Medina M, Pérez O. Toxicological Assessment of the Cochleate Derived from Neisseria meningitidis Proteoliposome in Sprague Dawley Rats. NORTH AMERICAN JOURNAL OF MEDICAL SCIENCES 2012; 4:135-40. [PMID: 22454827 PMCID: PMC3309621 DOI: 10.4103/1947-2714.93888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The AFCo1 cochleate is a potential novel adjuvant derived from Neisseria meningitidis B proteoliposome. AIM The aim was to assessing the safety of AFCo1 by single and repeated doses in Sprague Dawley rats. MATERIALS AND METHODS Rats were grouped for treatment with AFCo1, placebo formulation or control. The first study was a single intranasal dose of 100 μl and monitoring body weight, water, and food intakes as well as clinical symptoms. Fourteen days later the rats were killed and anatomopathological studies were conducted. In a second study, four similar doses of the test substance were instilled every 5 days. Clinical observations were carried out as for the single dose study and a number of rats from each group were killed 3 and 14 days after the last dose in order to conduct hematological, hemochemical, and anatomopathological studies. RESULTS No variable showed differences of toxicological relevance; the histological changes found were mild and similarly frequently in the three groups. According to the irritability index calculated form histology of the nasal region, AFCo1 was also classified as nonirritating. CONCLUSION AFCo1 is potentially safe for human use by nasal route as evidenced by the absence of local and systemic signs of toxicity in Sprague Dawley rats.
Collapse
Affiliation(s)
| | - Sergio Sifontes-Rodríguez
- Department of Animal Model and Immunology, Vice Presidency of Research, Finlay Institute, Havana, Cuba
| | | | - Tamara Hernández-Salazar
- Department of Animal Model and Immunology, Vice Presidency of Research, Finlay Institute, Havana, Cuba
| | - Mildrey Fariñas-Medina
- Department of Animal Model and Immunology, Vice Presidency of Research, Finlay Institute, Havana, Cuba
| | - Oliver Pérez
- Department of Animal Model and Immunology, Vice Presidency of Research, Finlay Institute, Havana, Cuba
| |
Collapse
|
31
|
Gersch ED, Gissmann L, Garcea RL. New approaches to prophylactic human papillomavirus vaccines for cervical cancer prevention. Antivir Ther 2011; 17:425-34. [PMID: 22293302 DOI: 10.3851/imp1941] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2011] [Indexed: 12/12/2022]
Abstract
The currently licensed human papillomavirus (HPV) vaccines are safe and highly effective at preventing HPV infection for a select number of papillomavirus types, thus decreasing the incidence of precursors to cervical cancer. It is expected that vaccination will also ultimately reduce the incidence of this cancer. The licensed HPV vaccines are, however, type restricted and expensive, and also require refrigeration, multiple doses and intramuscular injection. Second-generation vaccines are currently being developed to address these shortcomings. New expression systems, viral and bacterial vectors for HPV L1 capsid protein delivery, and use of the HPV L2 capsid protein will hopefully aid in decreasing cost and increasing ease of use and breadth of protection. These second-generation vaccines could also allow affordable immunization of women in developing countries, where the incidence of cervical cancer is high.
Collapse
Affiliation(s)
- Elizabeth D Gersch
- Department of Molecular, Cellular & Developmental Biology, University of Colorado, Boulder, CO, USA
| | | | | |
Collapse
|
32
|
Adriouch S, Franck E, Drouot L, Bonneau C, Jolinon N, Salvetti A, Boyer O. Improved Immunological Tolerance Following Combination Therapy with CTLA-4/Ig and AAV-Mediated PD-L1/2 Muscle Gene Transfer. Front Microbiol 2011; 2:199. [PMID: 22046170 PMCID: PMC3202221 DOI: 10.3389/fmicb.2011.00199] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 09/06/2011] [Indexed: 12/12/2022] Open
Abstract
Initially thought as being non-immunogenic, recombinant AAVs have emerged as efficient vector candidates for treating monogenic diseases. It is now clear however that they induce potent immune responses against transgene products which can lead to destruction of transduced cells. Therefore, developing strategies to circumvent these immune responses and facilitate long-term expression of transgenic therapeutic proteins is a main challenge in gene therapy. We evaluated herein a strategy to inhibit the undesirable immune activation that follows muscle gene transfer by administration of CTLA-4/Ig to block the costimulatory signals required early during immune priming and by using gene transfer of PD-1 ligands to inhibit T cell functions at the tissue sites. We provide the proof of principle that this combination immunoregulatory therapy targeting two non-redundant checkpoints of the immune response, i.e., priming and effector functions, can improve persistence of transduced cells in experimental settings where cytotoxic T cells escape initial blockade. Therefore, CTLA-4/Ig plus PD-L1/2 combination therapy represents a candidate approach to circumvent the bottleneck of immune responses directed toward transgene products.
Collapse
Affiliation(s)
- Sahil Adriouch
- INSERM U905, Faculty of Medicine and Pharmacy Rouen, France
| | | | | | | | | | | | | |
Collapse
|
33
|
Stephen SL, Freestone K, Dunn S, Twigg MW, Homer-Vanniasinkam S, Walker JH, Wheatcroft SB, Ponnambalam S. Scavenger receptors and their potential as therapeutic targets in the treatment of cardiovascular disease. Int J Hypertens 2010; 2010:646929. [PMID: 20981357 PMCID: PMC2958427 DOI: 10.4061/2010/646929] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 07/07/2010] [Indexed: 12/12/2022] Open
Abstract
Scavenger receptors act as membrane-bound and soluble proteins that bind to macromolecular complexes and pathogens. This diverse supergroup of proteins mediates binding to modified lipoprotein particles which regulate the initiation and progression of atherosclerotic plaques. In vascular tissues, scavenger receptors are implicated in regulating intracellular signaling, lipid accumulation, foam cell development, and cellular apoptosis or necrosis linked to the pathophysiology of atherosclerosis. One approach is using gene therapy to modulate scavenger receptor function in atherosclerosis. Ectopic expression of membrane-bound scavenger receptors using viral vectors can modify lipid profiles and reduce the incidence of atherosclerosis. Alternatively, expression of soluble scavenger receptors can also block plaque initiation and progression. Inhibition of scavenger receptor expression using a combined gene therapy and RNA interference strategy also holds promise for long-term therapy. Here we review our current understanding of the gene delivery by viral vectors to cells and tissues in gene therapy strategies and its application to the modulation of scavenger receptor function in atherosclerosis.
Collapse
Affiliation(s)
- Sam L. Stephen
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Katie Freestone
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Sarah Dunn
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Michael W. Twigg
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
- Leeds Vascular Institute, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK
| | - Shervanthi Homer-Vanniasinkam
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
- Leeds Vascular Institute, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK
| | - John H. Walker
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Stephen B. Wheatcroft
- Academic Unit of Molecular and Vascular Medicine, Faculty of Medicine and Health, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - Sreenivasan Ponnambalam
- Endothelial Cell Biology Unit, Institute of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| |
Collapse
|
34
|
Nieto K, Kern A, Leuchs B, Gissmann L, Müller M, Kleinschmidt JA. Combined prophylactic and therapeutic intranasal vaccination against human papillomavirus type-16 using different adeno-associated virus serotype vectors. Antivir Ther 2010; 14:1125-37. [PMID: 20032542 DOI: 10.3851/imp1469] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Cervical cancer is the second most frequent cancer among woman worldwide and is considered to be caused by infection with high-risk papilloma viruses. Genetic immunization using recombinant adeno-associated virus (rAAV) vectors has shown great promise for vaccination against human papillomavirus (HPV) infections. METHODS rAAV5, -8 and -9 vectors expressing an HPV16 L1/E7 fusion gene were generated and applied intranasally for combined prophylactic and therapeutic vaccination of mice. RESULTS The rAAV5 and the rAAV9 vectors showed efficient induction of both humoral and cellular immune responses, whereas rAAV8 failed to immunize mice by the intranasal route. The L1-specific immune response evoked by expression of the L1/E7 fusion gene, however, was lower than that evoked by expression of the L1 antigen alone. This deficiency could be compensated by application of Escherichia coli heat-labile enterotoxin or monophsphoryl lipid as adjuvant upon vaccination with rAAV5-L1/E7. Coimmunization of rAAV9-L1/E7 with rAAV5-L1 or boosting of rAAV9-L1/E7 with rAAV5-L1 strongly increased L1-specific neutralizing antibody titres to levels above those achieved by vaccination with vectors expressing L1 alone. Both vectors elicited a vibrant cytotoxic T-lymphocyte response against L1 or E7. Nasal immunization with rAAV5 or rAAV9 was superior to vaccination with HPV16-L1 virus-like particles (VLPs) or HPV16-L1/E7 CVLPs with respect to humoral and cellular immune responses. Vaccination with the rAAV vectors led to a significant protection of animals against a challenge with different HPV tumour cell lines. CONCLUSIONS Our results show that rAAV5 and rAAV9 vectors are promising candidates for a non-invasive nasal vaccination strategy.
Collapse
Affiliation(s)
- Karen Nieto
- Infection and Cancer Research Program, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Nieto K, Gissmann L, Schädlich L. Human papillomavirus-specific immune therapy: failure and hope. Antivir Ther 2010; 15:951-7. [DOI: 10.3851/imp1665] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
36
|
Lee HJ, Park N, Cho HJ, Yoon JK, Van ND, Oh YK, Kim YB. Development of a novel viral DNA vaccine against human papillomavirus: AcHERV-HP16L1. Vaccine 2009; 28:1613-9. [PMID: 19961961 DOI: 10.1016/j.vaccine.2009.11.044] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 11/07/2009] [Accepted: 11/18/2009] [Indexed: 10/20/2022]
Abstract
In this study, we developed a novel DNA vaccine for HPV; a recombinant baculovirus bearing human endogenous retrovirus (HERV) envelope protein, which cannot replicate in mammals, was used as a nano-carrier for HPV-16L1 DNA vaccine (AcHERV-HP16L1). For in vivo test, mice were injected intramuscularly with 107 particles of the constructs, with two boosts at 2-week intervals. Compared with Gardasil (25 microL/dose), the AcHERV-HP16L1 immunized mice showed similar high levels of humoral immunity in IgG/IgA and in neutralization of HPV pseudovirions. Combined immunization (prime with AcHERV-HP16L1 and boost with Gardasil) induced slightly higher neutralizing activity. As compared to the group treated with Gardasil, the mice immunized with AcHERV-HP16L1 showed 450- and 490-fold increase in the IFN-gamma at 5 and 20 weeks after the first priming, respectively. The combined immunization conferred lower T cell immunity than AcHERV-HP16L1 treatment. The advantages of our novel AcHERV-HP16L1 vaccine over Gardasil include higher cellular immunogenicity, considerably lower production cost, and comparable safety. Therefore, we suggest that AcHERV-HP16L1 can be developed as an efficient prophylactic vaccine and therapeutic vaccine.
Collapse
Affiliation(s)
- Hee-Jung Lee
- Department of Animal Biotechnology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | | | | | | | | | | | | |
Collapse
|
37
|
A new genetic vaccine platform based on an adeno-associated virus isolated from a rhesus macaque. J Virol 2009; 83:12738-50. [PMID: 19812149 DOI: 10.1128/jvi.01441-09] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We created a hybrid adeno-associated virus (AAV) from two related rhesus macaque isolates, called AAVrh32.33, and evaluated it as a vaccine carrier for human immunodeficiency virus type 1 (HIV-1) and type A influenza virus antigens. The goal was to overcome the limitations of vaccines based on other AAVs, which generate dysfunctional T-cell responses and are inhibited by antibodies found in human sera. Injection of a Gag-expressing AAVrh32.33 vector into mice resulted in a high-quality CD8(+) T-cell response. The resulting Gag-specific T cells express multiple cytokines at high levels, including interleukin-2, with many having memory phenotypes; a subsequent boost with an adenovirus vector yielded a brisk expansion of Gag-specific T cells. A priming dose of AAVrh32.33 led to high levels of Gag antibodies, which exceed levels found after injection of adenovirus vectors. Importantly, passive transfer of pooled human immunoglobulin into mice does not interfere with the efficacy of AAVrh32.33 expressing nucleoproteins from influenza virus, as measured by protection to a lethal dose of influenza virus, which is consistent with the very low seroprevalence to this virus in humans. Studies of macaques with vectors expressing gp140 from HIV-1 (i.e., with AAVrh32.33 as the prime and simian adenovirus type 24 as the boost) demonstrated results similar to those for mice with high-level and high-quality CD8(+) T-cell responses to gp140 and high-titered neutralizing antibodies to homologous HIV-1. The biology of this novel AAV hybrid suggests that it should be a preferred genetic vaccine carrier, capable of generating robust T- and B-cell responses.
Collapse
|
38
|
Gissmann L. HPV Vaccines: Preclinical Development. Arch Med Res 2009; 40:466-70. [DOI: 10.1016/j.arcmed.2009.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 06/23/2009] [Indexed: 12/29/2022]
|
39
|
Abstract
Bacillus anthracis represents a formidable bioterrorism and biowarfare threat for which new vaccines are needed with improved safety and efficacy over current options. Toward this end, we created recombinant adeno-associated virus type 1 (rAAV1) vectors containing synthetic genes derived from the protective antigen (PA) or lethal factor (LF) of anthrax lethal toxin (LeTx) and tested them for immunogenicity and induction of toxin-neutralizing antibodies in rabbits. Codon-optimized segments encoding activated PA (PA63), or LF, were synthesized and cloned into optimized rAAV1 vectors containing a human cytomegalovirus (hCMV) promoter and synthetic optimized leader. Serum from rabbits immunized intramuscularly with rAAV1/PA (monovalent), rAAV1/LF (monovalent), rAAV1/PA + rAAV1/LF (bivalent), or rAAV1/enhanced green fluorescent protein (control) exhibited substantial PA- and LF-specific antibody responses at 4 weeks by both western blot (> 1:10,000 dilution) and enzyme-linked immunosorbent assay (ELISA) (mean end-point titer: 32,000-260,000), and contained anthrax LeTx-neutralizing activity in vitro, with peak titers approximating those of a rabbit hyperimmune antisera raised against soluble PA and LF. Compared to the monovalent groups (rAAV1/PA or rAAV1/LF), the bivalent group (rAAV1/PA + rAAV1/LF) exhibited marginally higher ELISA and neutralization activity with dual specificity for both PA and LF. The finding of robust neutralizing antibody responses after a single injection of these rAAV1-based vectors supports their further development as candidate anthrax vaccines.
Collapse
|
40
|
Abstract
High risk human papillomavirus (HPV) types 16 and 18 DNAs were initially identified in 1983-1984. Subsequently the DNA of several other high risk HPV types has been identified. HPV 16 is present in more than 50% of cervical cancer biopsies, and HPV 18 is close to 20%. Some geographic variations exist in the prevalence of HPV high risk types: e.g. HPV 45 is more frequently observed in equatorial Africa, whereas types 58 and 52 have been more often found in East Asia. Molecular as well as epidemiological studies demonstrate that high risk HPV are indeed the causative agents for cervical cancer, they are also involved in other anogenital cancers, and in 25-30% of oropharyngeal carcinomas. Some of the mechanistic aspects are discussed in this review.
Collapse
Affiliation(s)
- H zur Hausen
- Deutsches Krebsforschungszentrum Im Neuenheimer Feld 280, Heidelberg 69120, Germany.
| |
Collapse
|
41
|
Cesco-Gaspere M, Zentilin L, Giacca M, Burrone O. Boosting Anti-idiotype Immune Response with Recombinant AAV Enhances Tumour Protection Induced by Gene Gun Vaccination. Scand J Immunol 2008; 68:58-66. [DOI: 10.1111/j.1365-3083.2008.02119.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Du L, Zhao G, Lin Y, Chan C, He Y, Jiang S, Wu C, Jin DY, Yuen KY, Zhou Y, Zheng BJ. Priming with rAAV encoding RBD of SARS-CoV S protein and boosting with RBD-specific peptides for T cell epitopes elevated humoral and cellular immune responses against SARS-CoV infection. Vaccine 2008; 26:1644-51. [PMID: 18289745 PMCID: PMC2600875 DOI: 10.1016/j.vaccine.2008.01.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Revised: 12/30/2007] [Accepted: 01/14/2008] [Indexed: 12/17/2022]
Abstract
Development of vaccines against severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) is crucial in the prevention of SARS reemergence. The receptor-binding domain (RBD) of SARS-CoV spike (S) protein is an important target in developing safe and effective SARS vaccines. Our previous study has demonstrated that vaccination with adeno-associated virus encoding RBD (RBD-rAAV) induces high titer of neutralizing antibodies. In this study, we further assessed the immune responses and protective effect of the immunization with RBD-rAAV prime/RBD-specific T cell peptide boost. Compared with the RBD-rAAV prime/boost vaccination, RBD-rAAV prime/RBD-peptide (RBD-Pep) boost induced similar levels of Th1 and neutralizing antibody responses that protected the vaccinated mice from subsequent SARS-CoV challenge, but stronger Th2 and CTL responses. No significant immune responses and protective effects were detected in mice vaccinated with RBD-Pep or blank AAV alone. Since T cell epitopes are highly conserved and boosting with peptides may induce the production of effector memory T cells, which may be effective against viruses with mutations in the neutralizing epitopes, our results suggest that the vaccination protocol used may be ideal for providing effective, broad and long-term protection against SARS-CoV infection.
Collapse
Affiliation(s)
- Lanying Du
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Lindsley F. Kimball Research Institute, The New York Blood Center, New York, NY 10021, USA
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yongping Lin
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chris Chan
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuxian He
- Lindsley F. Kimball Research Institute, The New York Blood Center, New York, NY 10021, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, The New York Blood Center, New York, NY 10021, USA
| | - Changyou Wu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dong-Yan Jin
- Department of Biochemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Bo-Jian Zheng
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
43
|
Du L, Zhao G, Lin Y, Sui H, Chan C, Ma S, He Y, Jiang S, Wu C, Yuen KY, Jin DY, Zhou Y, Zheng BJ. Intranasal vaccination of recombinant adeno-associated virus encoding receptor-binding domain of severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein induces strong mucosal immune responses and provides long-term protection against SARS-CoV infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:948-56. [PMID: 18178835 PMCID: PMC2603051 DOI: 10.4049/jimmunol.180.2.948] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have previously reported that a subunit protein vaccine based on the receptor-binding domain (RBD) of severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein and a recombinant adeno-associated virus (rAAV)-based RBD (RBD-rAAV) vaccine could induce highly potent neutralizing Ab responses in immunized animals. In this study, systemic, mucosal, and cellular immune responses and long-term protective immunity induced by RBD-rAAV were further characterized in a BALB/c mouse model, with comparison of the i.m. and intranasal (i.n.) routes of administration. Our results demonstrated that: 1) the i.n. vaccination induced a systemic humoral immune response of comparable strength and shorter duration than the i.m. vaccination, but the local humoral immune response was much stronger; 2) the i.n. vaccination elicited stronger systemic and local specific cytotoxic T cell responses than the i.m. vaccination, as evidenced by higher prevalence of IL-2 and/or IFN-gamma-producing CD3+/CD8+ T cells in both lungs and spleen; 3) the i.n. vaccination induced similar protection as the i.m. vaccination against SARS-CoV challenge in mice; 4) higher titers of mucosal IgA and serum-neutralizing Ab were associated with lower viral load and less pulmonary pathological damage, while no Ab-mediated disease enhancement effect was observed; and 5) the vaccination could provide long-term protection against SARS-CoV infection. Taken together, our findings suggest that RBD-rAAV can be further developed into a vaccine candidate for prevention of SARS and that i.n. vaccination may be the preferred route of administration due to its ability to induce SARS-CoV-specific systemic and mucosal immune responses and its better safety profile.
Collapse
Affiliation(s)
- Lanying Du
- Department of Microbiology, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Van Vliet KM, Blouin V, Brument N, Agbandje-McKenna M, Snyder RO. The role of the adeno-associated virus capsid in gene transfer. Methods Mol Biol 2008; 437:51-91. [PMID: 18369962 PMCID: PMC7120696 DOI: 10.1007/978-1-59745-210-6_2] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adeno-associated virus (AAV) is one of the most promising viral gene transfer vectors that has been shown to effect long-term gene expression and disease correction with low toxicity in animal models, and is well tolerated in human clinical trials. The surface of the AAV capsid is an essential component that is involved in cell binding, internalization, and trafficking within the targeted cell. Prior to developing a gene therapy strategy that utilizes AAV, the serotype should be carefully considered since each capsid exhibits a unique tissue tropism and transduction efficiency. Several approaches have been undertaken in an effort to target AAV vectors to specific cell types, including utilizing natural serotypes that target a desired cellular receptor, producing pseudotyped vectors, and engineering chimeric and mosaic AAV capsids. These capsid modifications are being incorporated into vector production and purification methods that provide for the ability to scale-up the manufacturing process to support human clinical trials. Protocols for small-scale and large-scale production of AAV, as well as assays to characterize the final vector product, are presented here. The structures of AAV2, AAV4, and AAV5 have been solved by X-ray crystallography or cryo-electron microscopy (cryo-EM), and provide a basis for rational vector design in developing customized capsids for specific targeting of AAV vectors. The capsid of AAV has been shown to be remarkably stable, which is a desirable characteristic for a gene therapy vector; however, recently it has been shown that the AAV serotypes exhibit differential susceptibility to proteases. The capsid fragmentation pattern when exposed to various proteases, as well as the susceptibility of the serotypes to a series of proteases, provides a unique fingerprint for each serotype that can be used for capsid identity validation. In addition to serotype identification, protease susceptibility can also be utilized to study dynamic structural changes that must occur for the AAV capsid to perform its various functions during the virus life cycle. The use of proteases for structural studies in solution complements the crystal structural studies of the virus. A generic protocol based on proteolysis for AAV serotype identification is provided here.
Collapse
Affiliation(s)
- Kim M Van Vliet
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | | | | | | | | |
Collapse
|
46
|
Gasparić M, Rubio I, Thönes N, Gissmann L, Müller M. Prophylactic DNA immunization against multiple papillomavirus types. Vaccine 2007; 25:4540-53. [PMID: 17485151 DOI: 10.1016/j.vaccine.2007.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 02/13/2007] [Accepted: 04/02/2007] [Indexed: 10/23/2022]
Abstract
At least 15 different papillomavirus types are causatively associated with the development of tumors in humans. Since the middle of 2006 a protective, virus-like particle based vaccine against the tumor-related HPV types 16 and 18 is commercially available. We investigated the possibility of applying DNA vaccination to obtain protective antibody responses against multiple papillomavirus types. Our data indicate that low amounts of DNA were sufficient to induce neutralizing antibodies in mice although a DNA dose-dependency in respect to the L1-specific antibody titers was observed. Furthermore, we found that immune responses against different PV types could be induced by simultaneous DNA vaccination with a mixture of expression vectors encoding L1 proteins of different papillomavirus types. However, we observed that there was a strong interference when plasmids encoding different L1 genes were used together. HPV 16 responses were repressed by co-administration of HPV 11 and/or BPV 1 L1 expression constructs. Likewise, BPV 1 responses were repressed by co-administration of HPV 16 or HPV 11 L1 plasmids. This interference could be overcome by administration of the different constructs into different sites of the animals or by sequential immunization. Thus, our results suggest that the mode of repression was due to interference with L1 particle assembly and was not a consequence of immunodominance of certain L1 proteins.
Collapse
Affiliation(s)
- Maja Gasparić
- Deutsches Krebsforschungszentrum, Forschungsschwerpunkt Infektionen und Krebs, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
47
|
Manuri PR, Nehete B, Nehete PN, Reisenauer R, Wardell S, Courtney AN, Gambhira R, Lomada D, Chopra AK, Sastry KJ. Intranasal immunization with synthetic peptides corresponding to the E6 and E7 oncoproteins of human papillomavirus type 16 induces systemic and mucosal cellular immune responses and tumor protection. Vaccine 2007; 25:3302-10. [PMID: 17291642 PMCID: PMC2043498 DOI: 10.1016/j.vaccine.2007.01.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 12/20/2006] [Accepted: 01/04/2007] [Indexed: 11/20/2022]
Abstract
The E6 and E7 oncoproteins of the high-risk HPV type16 represent ideal targets for HPV vaccine development, they being consistently expressed in cervical cancer lesions. Since HPV-16 is primarily transmitted through genital mucosal route, mucosal immune responses constitute an essential feature for vaccination strategies against HPV-associated lesions. We present here evidence showing that mucosal immunization of mice by the intranasal route with a mixture of peptides E7(44-62) and E6(43-57) from the E7 and E6 oncoproteins of HPV-16, respectively, using a mutant cholera toxin adjuvant (CT-2*), primed strong antigen-specific cellular immune responses in systemic and mucosal tissues. Significant levels of IFN-gamma production by both CD4 and CD8 cells were observed along with CTL responses that were effective against both peptide-pulsed targets as well as syngeneic tumor cells (TC-1) expressing the cognate E6 and E7 proteins. Furthermore, mice immunized with the peptide mixture and CT-2* effectively resisted TC-1 tumor challenge. These results together with our earlier observations that T cell responses to these peptides correlate with recurrence-free survival in women after ablative treatment for HPV-associated cervical intraepithelial neoplasia, support the potential of these E6 and E7 peptides for inclusion in vaccine formulations.
Collapse
Affiliation(s)
- Pallavi R. Manuri
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| | - Bharti Nehete
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| | - Pramod N. Nehete
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| | - Rose Reisenauer
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| | - Seth Wardell
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| | - Amy N. Courtney
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| | - Ratish Gambhira
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| | - Dakshyani Lomada
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| | - Ashok K. Chopra
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555
| | - K. Jagannadha Sastry
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, Bastrop, TX 77030
| |
Collapse
|
48
|
Kuck D, Kern A, Kleinschmidt JA. Development of AAV serotype-specific ELISAs using novel monoclonal antibodies. J Virol Methods 2007; 140:17-24. [PMID: 17126418 DOI: 10.1016/j.jviromet.2006.10.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Revised: 09/28/2006] [Accepted: 10/19/2006] [Indexed: 01/30/2023]
Abstract
Adeno-associated viruses (AAV) have been developed and evaluated as recombinant vectors for gene therapy. More recently, due to the advantages they offer for gene transfer, several AAV serotypes have gained increasing interest. However, monoclonal antibodies for the characterization and quantitation of vectors derived from different serotypes are at present not available. Serotype-specific monoclonal antibodies (mAbs) against the capsids of the serotypes 1/6, 4 and 5 are described. These antibodies, designated as ADK1a and b, ADK4 or ADK5a and b, reacted specifically with the indicated serotype capsids in cell lysates. ADK 1a and b cross-reacted with its highly related AAV6 serotype, but not with the other serotypes tested. The new antibodies recognized exclusively assembled capsids and neither free nor denatured capsid proteins as shown by fractionation experiments. In immunofluorescence experiments, the mAbs stained only distinct intranuclear foci in cells expressing the capsid protein. The development of capture ELISAs for quantitation of AAV1 and 6, AAV4 or AAV5 capsids illustrates that these novel monoclonal antibodies provide valuable tools for characterization of vector stocks.
Collapse
Affiliation(s)
- Dirk Kuck
- German Cancer Research Center, Program Infection and Cancer, Heidelberg, Germany
| | | | | |
Collapse
|