1
|
Zhang Y, Bourgine M, Wan Y, Song J, Li Z, Yu Y, Jiang W, Zhou M, Guo C, Santucci D, Liang X, Brechot C, Zhang W, Charneau P, Wu H, Qiu C. Therapeutic vaccination with lentiviral vector in HBV-persistent mice and two inactive HBsAg carriers. J Hepatol 2024; 80:31-40. [PMID: 37827470 DOI: 10.1016/j.jhep.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/09/2023] [Accepted: 09/06/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND & AIMS Immunotherapy for chronic hepatitis B virus (HBV) infection has not yet demonstrated sufficient efficacy. We developed a non-integrative lentiviral-vectored therapeutic vaccine for chronic hepatitis B and tested its antiviral effects in HBV-persistent mice and two inactive HBsAg carriers. METHODS Lentiviral vectors (LVs) encoding the core, preS1, or large HBsAg (LHBs) proteins of HBV were evaluated for immunogenicity in HBV-naïve mice and therapeutic efficacy in a murine model of chronic HBV infection. In addition, two inactive HBsAg carriers each received two doses of 5×107 transduction units (TU) or 1×108 TU of lentiviral-vectored LHBs (LV-LHBs), respectively. The endpoints were safety, LHBs-specific T-cell responses, and serum HBsAg levels during a 24-week follow-up. RESULTS In the mouse models, LV-LHBs was the most promising in eliciting robust antigen-specific T cells and in reducing the levels of serum HBsAg and viral load. By the end of the 34-week observation period, six out of ten (60%) HBV-persistent mice vaccinated with LV-LHBs achieved serum HBsAg loss and significant depletion of HBV-positive hepatocytes in the liver. In the two inactive HBsAg carriers, vaccination with LV-LHBs induced a considerable increase in the number of peripheral LHBs-specific T cells in one patient, and a weak but detectable response in the other, accompanied by a sustained reduction of HBsAg (-0.31 log10 IU/ml and -0.46 log10 IU/ml, respectively) from baseline to nadir. CONCLUSIONS A lentiviral-vectored therapeutic vaccine for chronic HBV infection demonstrated the potential to improve HBV-specific T-cell responses and deplete HBV-positive hepatocytes, leading to a sustained loss or reduction of serum HBsAg. IMPACT AND IMPLICATIONS Chronic HBV infection is characterized by an extremely low number and profound hypo-responsiveness of HBV-specific T cells. Therapeutic vaccines are designed to improve HBV-specific T-cell responses. We show that immunization with a lentiviral-vectored therapeutic HBV vaccine was able to expand HBV-specific T cells in vivo, leading to reductions of HBV-positive hepatocytes and serum HBsAg.
Collapse
Affiliation(s)
- Yumeng Zhang
- Department of Infectious Disease, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai, China; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, 200052, China
| | - Maryline Bourgine
- Institut Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| | - Yanmin Wan
- Department of Infectious Disease, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai, China; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, 200052, China
| | - Jieyu Song
- Department of Infectious Disease, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Yiqi Yu
- Department of Infectious Disease, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Mingzhe Zhou
- Department of Infectious Disease, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai, China
| | - Cuiyuan Guo
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Shanghai, China; Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan Province, Zhengzhou, China
| | | | | | - Christian Brechot
- TheraVectys S.A., Paris, France; University of South Florida, Tampa, USA.
| | - Wenhong Zhang
- Department of Infectious Disease, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai, China; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, 200052, China.
| | - Pierre Charneau
- Institut Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, F-75015 Paris, France.
| | - Hong Wu
- Changzhi People's Hospital, Changzhi, China.
| | - Chao Qiu
- Department of Infectious Disease, National Medical Center for Infectious Diseases and Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan University, Shanghai, China; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, 200052, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China..
| |
Collapse
|
2
|
Lopez J, Anna F, Authié P, Pawlik A, Ku MW, Blanc C, Souque P, Moncoq F, Noirat A, Hardy D, Sougakoff W, Brosch R, Guinet F, Charneau P, Majlessi L. A lentiviral vector encoding fusion of light invariant chain and mycobacterial antigens induces protective CD4 + T cell immunity. Cell Rep 2022; 40:111142. [PMID: 35905717 DOI: 10.1016/j.celrep.2022.111142] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 05/11/2022] [Accepted: 06/07/2022] [Indexed: 12/21/2022] Open
Abstract
Lentiviral vectors (LVs) are highly efficient at inducing CD8+ T cell responses. However, LV-encoded antigens are processed inside the cytosol of antigen-presenting cells, which does not directly communicate with the endosomal major histocompatibility complex class II (MHC-II) presentation pathway. LVs are thus poor at inducing CD4+ T cell response. To overcome this limitation, we devised a strategy whereby LV-encoded antigens are extended at their N-terminal end with the MHC-II-associated light invariant chain (li), which contains an endosome-targeting signal sequence. When evaluated with an LV-encoded polyantigen composed of CD4+ T cell targets from Mycobacterium tuberculosis, intranasal vaccination in mice triggers pulmonary polyfunctional CD4+ and CD8+ T cell responses. Adjuvantation of these LVs extends the mucosal immunity to Th17 and Tc17 responses. A systemic prime and an intranasal boost with one of these LV induces protection against M. tuberculosis. This strategy improves the protective power of LVs against infections and cancers, where CD4+ T cell immunity plays an important role.
Collapse
Affiliation(s)
- Jodie Lopez
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - François Anna
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Authié
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Alexandre Pawlik
- Institut Pasteur, Integrated Mycobacterial Pathogenomics Unit, CNRS UMR 3525, Université Paris Cité, 25 rue du Dr. Roux, 75015 Paris, France
| | - Min-Wen Ku
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Catherine Blanc
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Philippe Souque
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Fanny Moncoq
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Amandine Noirat
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - David Hardy
- Institut Pasteur, Histopathology Platform, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Wladimir Sougakoff
- Sorbonne Universités, UPMC Université Paris 06, CIMI-Paris, AP-HP, Hôpital Pitié-Salpêtrière, CNR-MyRMA, 75013 Paris, France
| | - Roland Brosch
- Institut Pasteur, Integrated Mycobacterial Pathogenomics Unit, CNRS UMR 3525, Université Paris Cité, 25 rue du Dr. Roux, 75015 Paris, France
| | - Françoise Guinet
- Institut Pasteur, Lymphocytes and Immunity Unit, Université Paris Cité, 25 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Charneau
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France
| | - Laleh Majlessi
- Institut Pasteur-TheraVectys Joint Lab, Université Paris Cité, 28 rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
3
|
Anna F, Lopez J, Moncoq F, Blanc C, Authié P, Noirat A, Fert I, Souque P, Nevo F, Pawlik A, Hardy D, Goyard S, Hudrisier D, Brosch R, Guinet F, Neyrolles O, Charneau P, Majlessi L. A lentiviral vector expressing a dendritic cell-targeting multimer induces mucosal anti-mycobacterial CD4 + T-cell immunity. Mucosal Immunol 2022; 15:1389-1404. [PMID: 36104497 PMCID: PMC9473479 DOI: 10.1038/s41385-022-00566-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 08/18/2022] [Accepted: 08/28/2022] [Indexed: 02/04/2023]
Abstract
Most viral vectors, including the potently immunogenic lentiviral vectors (LVs), only poorly direct antigens to the MHC-II endosomal pathway and elicit CD4+ T cells. We developed a new generation of LVs encoding antigen-bearing monomers of collectins substituted at their C-terminal domain with the CD40 ligand ectodomain to target and activate antigen-presenting cells. Host cells transduced with such optimized LVs secreted soluble collectin-antigen polymers with the potential to be endocytosed in vivo and reach the MHC-II pathway. In the murine tuberculosis model, such LVs induced efficient MHC-II antigenic presentation and triggered both CD8+ and CD4+ T cells at the systemic and mucosal levels. They also conferred a significant booster effect, consistent with the importance of CD4+ T cells for protection against Mycobacterium tuberculosis. Given the pivotal role of CD4+ T cells in orchestrating innate and adaptive immunity, this strategy could have a broad range of applications in the vaccinology field.
Collapse
Affiliation(s)
- François Anna
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Jodie Lopez
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Fanny Moncoq
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Catherine Blanc
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Pierre Authié
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Amandine Noirat
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Ingrid Fert
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Philippe Souque
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Fabien Nevo
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Alexandre Pawlik
- grid.428999.70000 0001 2353 6535Integrated Mycobacterial Pathogenomics Unit, CNRS UMR 3525, Institut Pasteur, Université Paris Cité, 25 rue du Dr. Roux, F-75015 Paris, France
| | - David Hardy
- grid.428999.70000 0001 2353 6535Histopathology Platform, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Sophie Goyard
- grid.428999.70000 0001 2353 6535Platform for Innovation and Development of Diagnostic Tests, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Denis Hudrisier
- grid.508721.9Institut de Pharmacologie et de Biologie Structurale, IPBS, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Roland Brosch
- grid.428999.70000 0001 2353 6535Integrated Mycobacterial Pathogenomics Unit, CNRS UMR 3525, Institut Pasteur, Université Paris Cité, 25 rue du Dr. Roux, F-75015 Paris, France
| | - Françoise Guinet
- grid.428999.70000 0001 2353 6535Lymphocytes and Immunity Unit, INSERM U1223, Institut Pasteur, Université Paris Cité, 25 rue du Dr. Roux, F-75015 Paris, France
| | - Olivier Neyrolles
- grid.508721.9Institut de Pharmacologie et de Biologie Structurale, IPBS, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Pierre Charneau
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| | - Laleh Majlessi
- grid.428999.70000 0001 2353 6535Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université Paris Cité, 28 rue du Dr. Roux, F-75015 Paris, France
| |
Collapse
|
4
|
Bona R, Michelini Z, Mazzei C, Gallinaro A, Canitano A, Borghi M, Vescio MF, Di Virgilio A, Pirillo MF, Klotman ME, Negri D, Cara A. Safety and efficiency modifications of SIV-based integrase-defective lentiviral vectors for immunization. Mol Ther Methods Clin Dev 2021; 23:263-275. [PMID: 34729374 PMCID: PMC8526422 DOI: 10.1016/j.omtm.2021.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/24/2021] [Indexed: 11/20/2022]
Abstract
Integrase-defective lentiviral vectors (IDLVs) represent an attractive platform for vaccine development as a result of the ability to induce persistent humoral- and cellular-mediated immune responses against the encoded transgene. Compared with the parental integrating vector, the main advantages for using IDLV are the reduced hazard of insertional mutagenesis and the decreased risk for vector mobilization by wild-type viruses. Here we report on the development and use in the mouse immunogenicity model of simian immunodeficiency virus (SIV)-based IDLV containing a long deletion in the U3 region and with the 3' polypurine tract (PPT) removed from the transfer vector for improving safety and/or efficacy. Results show that a safer extended deletion of U3 sequences did not modify integrase-mediated or -independent integration efficiency. Interestingly, 3' PPT deletion impaired integrase-mediated integration but did not reduce illegitimate, integrase-independent integration efficiency, contrary to what was previously reported in the HIV system. Importantly, although the extended deletion in the U3 did not affect expression or immunogenicity from IDLV, deletion of 3' PPT considerably reduced both expression and immunogenicity of IDLV.
Collapse
Affiliation(s)
- Roberta Bona
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Chiara Mazzei
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alessandra Gallinaro
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Maria Fenicia Vescio
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Antonio Di Virgilio
- Center for Animal Research and Welfare, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Maria Franca Pirillo
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Mary E. Klotman
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
5
|
Ku M, Authié P, Bourgine M, Anna F, Noirat A, Moncoq F, Vesin B, Nevo F, Lopez J, Souque P, Blanc C, Fert I, Chardenoux S, Lafosse L, Cussigh D, Hardy D, Nemirov K, Guinet F, Langa Vives F, Majlessi L, Charneau P. Brain cross-protection against SARS-CoV-2 variants by a lentiviral vaccine in new transgenic mice. EMBO Mol Med 2021; 13:e14459. [PMID: 34647691 PMCID: PMC8646827 DOI: 10.15252/emmm.202114459] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022] Open
Abstract
COVID-19 vaccines already in use or in clinical development may have reduced efficacy against emerging SARS-CoV-2 variants. In addition, although the neurotropism of SARS-CoV-2 is well established, the vaccine strategies currently developed have not taken into account protection of the central nervous system. Here, we generated a transgenic mouse strain expressing the human angiotensin-converting enzyme 2, and displaying unprecedented brain permissiveness to SARS-CoV-2 replication, in addition to high permissiveness levels in the lung. Using this stringent transgenic model, we demonstrated that a non-integrative lentiviral vector, encoding for the spike glycoprotein of the ancestral SARS-CoV-2, used in intramuscular prime and intranasal boost elicits sterilizing protection of lung and brain against both the ancestral virus, and the Gamma (P.1) variant of concern, which carries multiple vaccine escape mutations. Beyond induction of strong neutralizing antibodies, the mechanism underlying this broad protection spectrum involves a robust protective T-cell immunity, unaffected by the recent mutations accumulated in the emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Min‐Wen Ku
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Pierre Authié
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Maryline Bourgine
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - François Anna
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Amandine Noirat
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Fanny Moncoq
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Benjamin Vesin
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Fabien Nevo
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Jodie Lopez
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Philippe Souque
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Catherine Blanc
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Ingrid Fert
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Sébastien Chardenoux
- Plate‐Forme Centre d'Ingénierie Génétique Murine CIGMInstitut PasteurParisFrance
| | - llta Lafosse
- Plate‐Forme Centre d'Ingénierie Génétique Murine CIGMInstitut PasteurParisFrance
| | - Delphine Cussigh
- Plate‐Forme Centre d'Ingénierie Génétique Murine CIGMInstitut PasteurParisFrance
| | - David Hardy
- Experimental Neuropatholgy UnitInstitut PasteurParisFrance
| | - Kirill Nemirov
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Françoise Guinet
- Lymphocytes and Immunity UnitImmunology DepartmentInstitut PasteurParisFrance
| | - Francina Langa Vives
- Plate‐Forme Centre d'Ingénierie Génétique Murine CIGMInstitut PasteurParisFrance
| | - Laleh Majlessi
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| | - Pierre Charneau
- Virology DepartmentInstitut Pasteur‐TheraVectys Joint LabParisFrance
| |
Collapse
|
6
|
Ku MW, Bourgine M, Authié P, Lopez J, Nemirov K, Moncoq F, Noirat A, Vesin B, Nevo F, Blanc C, Souque P, Tabbal H, Simon E, Hardy D, Le Dudal M, Guinet F, Fiette L, Mouquet H, Anna F, Martin A, Escriou N, Majlessi L, Charneau P. Intranasal vaccination with a lentiviral vector protects against SARS-CoV-2 in preclinical animal models. Cell Host Microbe 2020; 29:236-249.e6. [PMID: 33357418 PMCID: PMC7738935 DOI: 10.1016/j.chom.2020.12.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/26/2020] [Accepted: 12/09/2020] [Indexed: 11/25/2022]
Abstract
To develop a vaccine candidate against coronavirus disease 2019 (COVID-19), we generated a lentiviral vector (LV) eliciting neutralizing antibodies against the Spike glycoprotein of SARS-CoV-2. Systemic vaccination by this vector in mice, in which the expression of the SARS-CoV-2 receptor hACE2 has been induced by transduction of respiratory tract cells by an adenoviral vector, confers only partial protection despite high levels of serum neutralizing activity. However, eliciting an immune response in the respiratory tract through an intranasal boost results in a >3 log10 decrease in the lung viral loads and reduces local inflammation. Moreover, both integrative and non-integrative LV platforms display strong vaccine efficacy and inhibit lung deleterious injury in golden hamsters, which are naturally permissive to SARS-CoV-2 replication and closely mirror human COVID-19 physiopathology. Our results provide evidence of marked prophylactic effects of LV-based vaccination against SARS-CoV-2 and designate intranasal immunization as a powerful approach against COVID-19.
Collapse
Affiliation(s)
- Min-Wen Ku
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Maryline Bourgine
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France; Molecular Virology and Vaccinology Unit, Virology Department, Institut Pasteur, Paris 75015, France
| | - Pierre Authié
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Jodie Lopez
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Kirill Nemirov
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Fanny Moncoq
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Amandine Noirat
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Benjamin Vesin
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Fabien Nevo
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Catherine Blanc
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Philippe Souque
- Molecular Virology and Vaccinology Unit, Virology Department, Institut Pasteur, Paris 75015, France
| | - Houda Tabbal
- Molecular Genetics of RNA Viruses Unit, Virology Department, Institut Pasteur, CNRS UMR3569, Université de Paris, Paris 75015, France
| | - Emeline Simon
- Molecular Genetics of RNA Viruses Unit, Virology Department, Institut Pasteur, CNRS UMR3569, Université de Paris, Paris 75015, France; Université de Paris, Paris 75006, France
| | - David Hardy
- Experimental Neuropathology Unit, Global Health Department, Institut Pasteur, Paris 75015, France
| | | | - Françoise Guinet
- Lymphocytes and Immunity Unit, Immunology Department, Institut Pasteur, Paris 75015, France
| | | | - Hugo Mouquet
- Laboratory of Humoral Immunology, Immunology Department, Institut Pasteur, INSERM U1222, Paris, France
| | - François Anna
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France
| | - Annette Martin
- Molecular Genetics of RNA Viruses Unit, Virology Department, Institut Pasteur, CNRS UMR3569, Université de Paris, Paris 75015, France
| | - Nicolas Escriou
- Innovation Lab, Vaccines, Virology Department, Institut Pasteur, Paris 75015, France
| | - Laleh Majlessi
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France.
| | - Pierre Charneau
- Institut Pasteur-TheraVectys Joint Lab, Virology Department, Institut Pasteur, Paris 75015, France; Molecular Virology and Vaccinology Unit, Virology Department, Institut Pasteur, Paris 75015, France.
| |
Collapse
|
7
|
Tasyurek HM, Altunbas HA, Balci MK, Griffith TS, Sanlioglu S. Therapeutic Potential of Lentivirus-Mediated Glucagon-Like Peptide-1 Gene Therapy for Diabetes. Hum Gene Ther 2018; 29:802-815. [PMID: 29409356 DOI: 10.1089/hum.2017.180] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Postprandial glucose-induced insulin secretion from the islets of Langerhans is facilitated by glucagon-like peptide-1 (GLP-1)-a metabolic hormone with insulinotropic properties. Among the variety of effects it mediates, GLP-1 induces delta cell secretion of somatostatin, inhibits alpha cell release of glucagon, reduces gastric emptying, and slows food intake. These events collectively contribute to weight loss over time. During type 2 diabetes (T2DM), however, the incretin response to glucose is reduced and accompanied by a moderate reduction in GLP-1 secretion. To compensate for the reduced incretin effect, a human immunodeficiency virus-based lentiviral vector was generated to deliver DNA encoding human GLP-1 (LentiGLP-1), and the anti-diabetic efficacy of LentiGLP-1 was tested in a high-fat diet/streptozotocin-induced model of T2DM. Therapeutic administration of LentiGLP-1 reduced blood glucose levels in obese diabetic Sprague Dawley rats, along with improving insulin sensitivity and glucose tolerance. Normoglycemia was correlated with increased blood GLP-1 and pancreatic beta cell regeneration in LentiGLP-1-treated rats. Plasma triglyceride levels were also normalized after LentiGLP-1 injection. Collectively, these data suggest the clinical potential of GLP-1 gene transfer therapy for the treatment of T2DM.
Collapse
Affiliation(s)
- Hale M Tasyurek
- 1 Human Gene and Cell Therapy Center of Akdeniz University Hospitals , Antalya, Turkey
| | - Hasan Ali Altunbas
- 2 Department of Internal Medicine, Division of Endocrinology and Metabolism, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Mustafa Kemal Balci
- 2 Department of Internal Medicine, Division of Endocrinology and Metabolism, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Thomas S Griffith
- 3 Department of Urology, University of Minnesota , School of Medicine, Minneapolis, Minnesota
| | - Salih Sanlioglu
- 1 Human Gene and Cell Therapy Center of Akdeniz University Hospitals , Antalya, Turkey
| |
Collapse
|
8
|
Norton TD, Miller EA. Corrigendum: Recent Advances in Lentiviral Vaccines for HIV-1 Infection. Front Immunol 2016; 7:354. [PMID: 27630642 PMCID: PMC5021684 DOI: 10.3389/fimmu.2016.00354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 08/31/2016] [Indexed: 11/13/2022] Open
Abstract
[This corrects the article on p. 243 in vol. 7, PMID: 27446074.].
Collapse
Affiliation(s)
- Thomas D Norton
- Department of Medicine, Division of Infectious Diseases, NYU School of Medicine , New York, NY , USA
| | - Elizabeth A Miller
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| |
Collapse
|
9
|
Dérian N, Bellier B, Pham HP, Tsitoura E, Kazazi D, Huret C, Mavromara P, Klatzmann D, Six A. Early Transcriptome Signatures from Immunized Mouse Dendritic Cells Predict Late Vaccine-Induced T-Cell Responses. PLoS Comput Biol 2016; 12:e1004801. [PMID: 26998760 PMCID: PMC4801398 DOI: 10.1371/journal.pcbi.1004801] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 02/08/2016] [Indexed: 01/19/2023] Open
Abstract
Systems biology offers promising approaches for identifying response-specific signatures to vaccination and assessing their predictive value. Here, we designed a modelling strategy aiming to predict the quality of late T-cell responses after vaccination from early transcriptome analysis of dendritic cells. Using standardized staining with tetramer, we first quantified antigen-specific T-cell expansion 5 to 10 days after vaccination with one of a set of 41 different vaccine vectors all expressing the same antigen. Hierarchical clustering of the responses defined sets of high and low T cell response inducers. We then compared these responses with the transcriptome of splenic dendritic cells obtained 6 hours after vaccination with the same vectors and produced a random forest model capable of predicting the quality of the later antigen-specific T-cell expansion. The model also successfully predicted vector classification as low or strong T-cell response inducers of a novel set of vaccine vectors, based on the early transcriptome results obtained from spleen dendritic cells, whole spleen and even peripheral blood mononuclear cells. Finally, our model developed with mouse datasets also accurately predicted vaccine efficacy from literature-mined human datasets. Vaccines are designed to elicit effective immune responses against antigens. The various vector platforms used in vaccine development are diverse and complex, rendering the selection of promising vaccines vector challenging. We have designed a modeling strategy that predicts the propensity of vaccine vectors to elicit strong late T-cell responses using transcriptome material obtained 6 hours after vaccination. Our model, designed with mouse datasets, also predicted vector efficacy from mined human data. Thus, molecular signatures obtained 6 hours after vaccination can predict vaccine efficacy at 2 weeks post vaccination, which should help in vaccine development.
Collapse
Affiliation(s)
- Nicolas Dérian
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 959, Immunology, Immunopathology, Immunotherapy, Paris, France
- AP-HP, Clinical Investigation Center in Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France
- INSERM, UMRS 959, "Immunology, Immunopathology, Immunotherapy", Paris, France
| | - Bertrand Bellier
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 959, Immunology, Immunopathology, Immunotherapy, Paris, France
- AP-HP, Clinical Investigation Center in Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France
- INSERM, UMRS 959, "Immunology, Immunopathology, Immunotherapy", Paris, France
| | - Hang Phuong Pham
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 959, Immunology, Immunopathology, Immunotherapy, Paris, France
- INSERM, UMRS 959, "Immunology, Immunopathology, Immunotherapy", Paris, France
| | - Eliza Tsitoura
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Athens, Greece
| | - Dorothea Kazazi
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Athens, Greece
| | - Christophe Huret
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 959, Immunology, Immunopathology, Immunotherapy, Paris, France
- INSERM, UMRS 959, "Immunology, Immunopathology, Immunotherapy", Paris, France
| | - Penelope Mavromara
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Athens, Greece
| | - David Klatzmann
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 959, Immunology, Immunopathology, Immunotherapy, Paris, France
- AP-HP, Clinical Investigation Center in Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France
- INSERM, UMRS 959, "Immunology, Immunopathology, Immunotherapy", Paris, France
- * E-mail: (DK); (AS)
| | - Adrien Six
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 959, Immunology, Immunopathology, Immunotherapy, Paris, France
- AP-HP, Clinical Investigation Center in Biotherapy, Hôpital Pitié-Salpêtrière, Paris, France
- INSERM, UMRS 959, "Immunology, Immunopathology, Immunotherapy", Paris, France
- * E-mail: (DK); (AS)
| |
Collapse
|
10
|
Soares HR, Castro R, Tomás HA, Rodrigues AF, Gomes-Alves P, Bellier B, Klatzmann D, Carrondo MJT, Alves PM, Coroadinha AS. Tetraspanins displayed in retrovirus-derived virus-like particles and their immunogenicity. Vaccine 2016; 34:1634-1641. [PMID: 26795367 DOI: 10.1016/j.vaccine.2015.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 10/22/2022]
Abstract
Virus-like particles (VLPs) are a particular subset of subunit vaccines which are currently explored as safer alternatives to live attenuated or inactivated vaccines. VLPs derived from retrovirus (retroVLPs) are commonly used as scaffolds for vaccine candidates due to their ability to incorporate heterologous envelope proteins. Pseudotyping retroVLPs is however not a selective process therefore, host cellular proteins such as tetraspanins are also included in the membrane. The contribution of these host-proteins to retrovirus immunogenicity remains unclear. In this work, human cells silenced and not silenced for tetraspanin CD81 were used to produce CD81(-) or CD81(+) retroVLPs. We first analyzed mice immune response against human CD81. Despite effective silencing of CD81 in retroVLP producing cells, both humoral and cellular immune responses showed persistent anti-CD81 immunogenicity, suggesting cross reactivity to related antigens. We thus compared the incorporation of related tetraspanins in retroVLPs and showed that decreased CD81 incorporation in CD81(-) retro-VLPs is compensated by an increased incorporation of CD9 and CD63 tetraspanins. These results highlight the dynamic nature of host-derived proteins incorporation in retroVLPs membrane, which should be considered when retrovirus-based biopharmaceuticals are produced in xenogeneic cells.
Collapse
Affiliation(s)
- H R Soares
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - R Castro
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - H A Tomás
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - A F Rodrigues
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - P Gomes-Alves
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - B Bellier
- Sorbonne Universités, UPMC Univ Paris 06, UMRS_959, I3, F-75013 Paris, France; INSERM, UMR_S 959, I3, F-75013 Paris, France
| | - D Klatzmann
- Sorbonne Universités, UPMC Univ Paris 06, UMRS_959, I3, F-75013 Paris, France; INSERM, UMR_S 959, I3, F-75013 Paris, France
| | - M J T Carrondo
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Faculdade de Ciências e Tecnologia/Universidade Nova de Lisboa, P-2825 Monte da Caparica, Portugal
| | - P M Alves
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal
| | - A S Coroadinha
- iBET - Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, António Xavier, Universidade Nova de Lisboa (ITQB-UNL), Av. da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
11
|
Abstract
Gene transfer vectors based on retroviridae are increasingly becoming a tool of choice for biomedical research and for the development of biotherapies in rare diseases or cancers. To meet the challenges of preclinical and clinical production, different steps of the production process of self-inactivating γ-retroviral (RVs) and lentiviral vectors (LVs) have been improved (e.g., transfection, media optimization, cell culture conditions). However, the increasing need for mass production of such vectors is still a challenge and could hamper their availability for therapeutic use. Recently, we observed that the use of a neutral pH during vector production is not optimal. The use of mildly acidic pH conditions (pH 6) can increase by two- to threefold the production of RVs and LVs pseudotyped with the vesicular stomatitis virus G (VSV-G) or gibbon ape leukemia virus (GALV) glycoproteins. Here, we describe the production protocol in mildly acidic pH conditions of GALVTR- and VSV-G-pseudotyped LVs using the transient transfection of HEK293T cells and the production protocol of GALV-pseudotyped RVs produced from a murine producer cell line. These protocols should help to achieve higher titers of vectors, thereby facilitating experimental research and therapeutic applications.
Collapse
Affiliation(s)
- Nathalie Holic
- Généthon, 91002, Evry, France.
- INSERM, UMR_S951, Généthon, 1bis, rue de l'Internationale-BP60, 91002, Evry, France.
- Université Evry Val d'Essonne, UMR_S951, 91002, Evry, France.
| | - David Fenard
- Généthon, 91002, Evry, France.
- INSERM, UMR_S951, Généthon, 1bis, rue de l'Internationale-BP60, 91002, Evry, France.
- Université Evry Val d'Essonne, UMR_S951, 91002, Evry, France.
| |
Collapse
|
12
|
de Wispelaere M, Ricklin M, Souque P, Frenkiel MP, Paulous S, Garcìa-Nicolàs O, Summerfield A, Charneau P, Desprès P. A Lentiviral Vector Expressing Japanese Encephalitis Virus-like Particles Elicits Broad Neutralizing Antibody Response in Pigs. PLoS Negl Trop Dis 2015; 9:e0004081. [PMID: 26437302 PMCID: PMC4593544 DOI: 10.1371/journal.pntd.0004081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/21/2015] [Indexed: 01/08/2023] Open
Abstract
Background Japanese encephalitis virus (JEV) is the major cause of viral encephalitis in Southeast Asia. Vaccination of domestic pigs has been suggested as a “one health” strategy to reduce viral disease transmission to humans. The efficiency of two lentiviral TRIP/JEV vectors expressing the JEV envelope prM and E glycoproteins at eliciting protective humoral response was assessed in a mouse model and piglets. Methodology/Principal Findings A gene encoding the envelope proteins prM and E from a genotype 3 JEV strain was inserted into a lentiviral TRIP vector. Two lentiviral vectors TRIP/JEV were generated, each expressing the prM signal peptide followed by the prM protein and the E glycoprotein, the latter being expressed either in its native form or lacking its two C-terminal transmembrane domains. In vitro transduction of cells with the TRIP/JEV vector expressing the native prM and E resulted in the efficient secretion of virus-like particles of Japanese encephalitis virus. Immunization of BALB/c mice with TRIP/JEV vectors resulted in the production of IgGs against Japanese encephalitis virus, and the injection of a second dose one month after the prime injection greatly boosted antibody titers. The TRIP/JEV vectors elicited neutralizing antibodies against JEV strains belonging to genotypes 1, 3, and 5. Immunization of piglets with two doses of the lentiviral vector expressing JEV virus-like particles led to high titers of anti-JEV antibodies, that had efficient neutralizing activity regardless of the JEV genotype tested. Conclusions/Significance Immunization of pigs with the lentiviral vector expressing JEV virus-like particles is particularly efficient to prime antigen-specific humoral immunity and trigger neutralizing antibody responses against JEV genotypes 1, 3, and 5. The titers of neutralizing antibodies elicited by the TRIP/JEV vector are sufficient to confer protection in domestic pigs against different genotypes of JEV and this could be of a great utility in endemic regions where more than one genotype is circulating. Japanese encephalitis virus is the etiologic agent of the most medically important viral encephalitis in South Asia with thousands of deaths per year. The virus is maintained in an enzootic cycle between Culex mosquitoes and amplifying vertebrate hosts, such as wild boars and pigs. Vaccination of domestic pigs has been suggested as a strategy to reduce viral disease transmission to humans, in line with the now-called “One Health” concept. Lentiviral gene transfer vectors represent a novel vaccination platform with an unprecedented ability to induce robust humoral immunity in various animal species. In our study, we demonstrated that immunization of pigs with a recombinant lentiviral vector expressing virus-like particles of Japanese encephalitis virus is particularly efficient at eliciting specific humoral immunity. The titers of neutralizing antibodies elicited by the lentiviral vector are sufficient to confer protection in domestic pigs against the different genotypes of Japanese encephalitis virus observed in Asia.
Collapse
Affiliation(s)
| | - Meret Ricklin
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Philippe Souque
- Virologie Moléculaire et Vaccinologie, Institut Pasteur, Paris, France
| | | | - Sylvie Paulous
- Interactions Moléculaires Flavivirus-Hôtes, Institut Pasteur, Paris, France
| | | | - Artur Summerfield
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Pierre Charneau
- Virologie Moléculaire et Vaccinologie, Institut Pasteur, Paris, France
- * E-mail: (PC); (PD)
| | - Philippe Desprès
- Interactions Moléculaires Flavivirus-Hôtes, Institut Pasteur, Paris, France
- * E-mail: (PC); (PD)
| |
Collapse
|
13
|
Kang W, Marasco WA, Tong HI, Byron MM, Wu C, Shi Y, Sun S, Sun Y, Lu Y. Anti-tat Hutat2:Fc mediated protection against tat-induced neurotoxicity and HIV-1 replication in human monocyte-derived macrophages. J Neuroinflammation 2014; 11:195. [PMID: 25416164 PMCID: PMC4256057 DOI: 10.1186/s12974-014-0195-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 11/05/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND HIV-1 Tat is essential for HIV replication and is also a well-known neurotoxic factor causing HIV-associated neurocognitive disorder (HAND). Currently, combined antiretroviral therapy targeting HIV reverse transcriptase or protease cannot prevent the production of early viral proteins, especially Tat, once HIV infection has been established. HIV-infected macrophages and glial cells in the brain still release Tat into the extracellular space where it can exert direct and indirect neurotoxicity. Therefore, stable production of anti-Tat antibodies in the brain would neutralize HIV-1 Tat and thus provide an effective approach to protect neurons. METHODS We constructed a humanized anti-Tat Hutat2:Fc fusion protein with the goal of antagonizing HIV-1 Tat and delivered the gene into cell lines and primary human monocyte-derived macrophages (hMDM) by an HIV-based lentiviral vector. The function of the anti-Tat Hutat2:Fc fusion protein and the potential side effects of lentiviral vector-mediated gene transfer were evaluated in vitro. RESULTS Our study demonstrated that HIV-1-based lentiviral vector-mediated gene transduction resulted in a high-level, stable expression of anti-HIV-1 Tat Hutat2:Fc in human neuronal and monocytic cell lines, as well as in primary hMDM. Hutat2:Fc was detectable in both cells and supernatants and continued to accumulate to high levels within the supernatant. Hutat2:Fc protected mouse cortical neurons against HIV-1 Tat86-induced neurotoxicity. In addition, both secreted Hutat2:Fc and transduced hMDM led to reducing HIV-1BaL viral replication in human macrophages. Moreover, lentiviral vector-based gene introduction did not result in any significant changes in cytomorphology and cell viability. Although the expression of IL8, STAT1, and IDO1 genes was up-regulated in transduced hMDM, such alternation in gene expression did not affect the neuroprotective effect of Hutat2:Fc. CONCLUSIONS Our study demonstrated that lentivirus-mediated gene transfer could efficiently deliver the Hutat2:Fc gene into primary hMDM and does not lead to any significant changes in hMDM immune-activation. The neuroprotective and HIV-1 suppressive effects produced by Hutat2:Fc were comparable to that of a full-length anti-Tat antibody. This study provides the foundation and insights for future research on the potential use of Hutat2:Fc as a novel gene therapy approach for HAND through utilizing monocytes/macrophages, which naturally cross the blood-brain barrier, for gene delivery.
Collapse
Affiliation(s)
- Wen Kang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi, 710038, China. .,Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Wayne A Marasco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 50 Brookline Avenue, Boston, MA, 02215, USA.
| | - Hsin-I Tong
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Mary Margaret Byron
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St., BSB, Suite 231, Honolulu, HI, 96813, USA.
| | - Chengxiang Wu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Yingli Shi
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Si Sun
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Yongtao Sun
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi, 710038, China.
| | - Yuanan Lu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| |
Collapse
|
14
|
Arrode-Brusés G, Moussa M, Baccard-Longere M, Villinger F, Chebloune Y. Long-term central and effector SHIV-specific memory T cell responses elicited after a single immunization with a novel lentivector DNA vaccine. PLoS One 2014; 9:e110883. [PMID: 25337803 PMCID: PMC4206452 DOI: 10.1371/journal.pone.0110883] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/18/2014] [Indexed: 12/13/2022] Open
Abstract
Prevention of HIV acquisition and replication requires long lasting and effective immunity. Given the state of HIV vaccine development, innovative vectors and immunization strategies are urgently needed to generate safe and efficacious HIV vaccines. Here, we developed a novel lentivirus-based DNA vector that does not integrate in the host genome and undergoes a single-cycle of replication. Viral proteins are constitutively expressed under the control of Tat-independent LTR promoter from goat lentivirus. We immunized six macaques once only with CAL-SHIV-IN- DNA using combined intramuscular and intradermal injections plus electroporation. Antigen-specific T cell responses were monitored for 47 weeks post-immunization (PI). PBMCs were assessed directly ex vivo or after 6 and 12 days of in vitro culture using antigenic and/or homeostatic proliferation. IFN-γ ELISPOT was used to measure immediate cytokine secretion from antigen specific effector cells and from memory precursors with high proliferative capacity (PHPC). The memory phenotype and functions (proliferation, cytokine expression, lytic content) of specific T cells were tested using multiparametric FACS-based assays. All immunized macaques developed lasting peripheral CD8+ and CD4+ T cell responses mainly against Gag and Nef antigens. During the primary expansion phase, immediate effector cells as well as increasing numbers of proliferating cells with limited effector functions were detected which expressed markers of effector (EM) and central (CM) memory phenotypes. These responses contracted but then reemerged later in absence of antigen boost. Strong PHPC responses comprising vaccine-specific CM and EM T cells that readily expanded and acquired immediate effector functions were detected at 40/47 weeks PI. Altogether, our study demonstrated that a single immunization with a replication-limited DNA vaccine elicited persistent vaccine-specific CM and EM CD8+ and CD4+ T cells with immediate and readily inducible effector functions, in the absence of ongoing antigen expression.
Collapse
Affiliation(s)
| | - Maha Moussa
- INRA, ANRS, Université Joseph Fourier, PAVAL Lab./Nanobio 2, UJF Grenoble, Grenoble, France
| | - Monique Baccard-Longere
- Institut de Biologie et Pathologie, Centre Hospitalo-Universitaire de Grenoble, Grenoble, France
| | - François Villinger
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Yahia Chebloune
- INRA, ANRS, Université Joseph Fourier, PAVAL Lab./Nanobio 2, UJF Grenoble, Grenoble, France
| |
Collapse
|
15
|
Macdonald DC, Hotblack A, Akbar S, Britton G, Collins MK, Rosenberg WC. 4-1BB ligand activates bystander dendritic cells to enhance immunization in trans. THE JOURNAL OF IMMUNOLOGY 2014; 193:5056-64. [PMID: 25305314 DOI: 10.4049/jimmunol.1301723] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Expression of the costimulatory receptor 4-1BB is induced by TCR recognition of Ag, whereas 4-1BB ligand (4-1BBL) is highly expressed on activated APC. 4-1BB signaling is particularly important for survival of activated and memory CD8(+) T cells. We wished to test whether coexpression of Ag and 4-1BBL by dendritic cells (DC) would be an effective vaccine strategy. Therefore, we constructed lentiviral vectors (LV) coexpressing 4-1BBL and influenza nucleoprotein (NP). Following s.c. immunization of mice, which targets DC, we found superior CD8(+) T cell responses against NP and protection from influenza when 4-1BBL was expressed. However, functionally superior CD8(+) T cell responses were obtained when two LV were coinjected: one expressing 4-1BBL and the other expressing NP. This surprising result suggested that 4-1BBL is more effective when expressed in trans, acting on adjacent DC. Therefore, we investigated the effect of LV expression of 4-1BBL in mouse DC cultures and observed induced maturation of bystander, untransduced cells. Maturation was blocked by anti-4-1BBL Ab, required cell-cell contact, and did not require the cytoplasmic signaling domain of 4-1BBL. Greater maturation of untransduced cells could be explained by LV expression of 4-1BBL, causing downregulation of 4-1BB. These data suggest that coexpression of 4-1BBL and Ag by vaccine vectors that target DC may not be an optimal strategy. However, 4-1BBL LV immunization activates significant numbers of bystander DC in the draining lymph nodes. Therefore, transactivation by 4-1BBL/4-1BB interaction following DC-DC contact may play a role in the immune response to infection or vaccination.
Collapse
Affiliation(s)
- Douglas C Macdonald
- Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - Alastair Hotblack
- Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - Saniath Akbar
- Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - Gary Britton
- Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - Mary K Collins
- Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom; National Institute for Biological Standards and Control, South Mimms, Potters Bar, Hertsfordshire EN6 3QG, United Kingdom; and
| | - William C Rosenberg
- Division of Medicine, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
16
|
Ciré S, Da Rocha S, Yao R, Fisson S, Buchholz CJ, Collins MK, Galy A. Immunization of mice with lentiviral vectors targeted to MHC class II+ cells is due to preferential transduction of dendritic cells in vivo. PLoS One 2014; 9:e101644. [PMID: 25058148 PMCID: PMC4109917 DOI: 10.1371/journal.pone.0101644] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/09/2014] [Indexed: 11/20/2022] Open
Abstract
Gene transfer vectors such as lentiviral vectors offer versatile possibilities to express transgenic antigens for vaccination purposes. However, viral vaccines leading to broad transduction and transgene expression in vivo, are undesirable. Therefore, strategies capable of directing gene transfer only to professional antigen-presenting cells would increase the specific activity and safety of genetic vaccines. A lentiviral vector pseudotype specific for murine major histocompatibilty complex class II (LV-MHCII) was recently developed and the present study aims to characterize the in vivo biodistribution profile and immunization potential of this vector in mice. Whereas the systemic administration of a vector pseudotyped with a ubiquitously-interacting envelope led to prominent detection of vector copies in the liver of animals, the injection of an equivalent amount of LV-MHCII resulted in a more specific biodistribution of vector and transgene. Copies of LV-MHCII were found only in secondary lymphoid organs, essentially in CD11c+ dendritic cells expressing the transgene whereas B cells were not efficiently targeted in vivo, contrary to expectations based on in vitro testing. Upon a single injection of LV-MHCII, naive mice mounted specific effector CD4 and CD8 T cell responses against the intracelllular transgene product with the generation of Th1 cytokines, development of in vivo cytotoxic activity and establishment of T cell immune memory. The targeting of dendritic cells by recombinant viral vaccines must therefore be assessed in vivo but this strategy is feasible, effective for immunization and cross-presentation and constitutes a potentially safe alternative to limit off-target gene expression in gene-based vaccination strategies with integrative vectors.
Collapse
Affiliation(s)
- Séverine Ciré
- Inserm, U 951, Molecular Immunology and Innovative Biotherapies, Genethon, Evry, France
- UMR_S951, University of Evry, Genethon, Evry, France
- Genethon, Evry, France
| | - Sylvie Da Rocha
- Inserm, U 951, Molecular Immunology and Innovative Biotherapies, Genethon, Evry, France
- UMR_S951, University of Evry, Genethon, Evry, France
- Genethon, Evry, France
| | - Roseline Yao
- Inserm, U 951, Molecular Immunology and Innovative Biotherapies, Genethon, Evry, France
- UMR_S951, University of Evry, Genethon, Evry, France
- Genethon, Evry, France
| | - Sylvain Fisson
- Inserm, U 951, Molecular Immunology and Innovative Biotherapies, Genethon, Evry, France
- UMR_S951, University of Evry, Genethon, Evry, France
- Genethon, Evry, France
| | | | - Mary K. Collins
- Infection and Immunity, University College London, London, United Kingdom
| | - Anne Galy
- Inserm, U 951, Molecular Immunology and Innovative Biotherapies, Genethon, Evry, France
- UMR_S951, University of Evry, Genethon, Evry, France
- Genethon, Evry, France
| |
Collapse
|
17
|
Garulli B, Di Mario G, Stillitano MG, Kawaoka Y, Castrucci MR. Exploring mucosal immunization with a recombinant influenza virus carrying an HIV-polyepitope in mice with pre-existing immunity to influenza. Vaccine 2014; 32:2501-6. [DOI: 10.1016/j.vaccine.2014.02.077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/07/2014] [Accepted: 02/25/2014] [Indexed: 12/12/2022]
|
18
|
Abstract
Glucagon-like peptide (GLP)-1 is an incretin hormone with several antidiabetic functions including stimulation of glucose-dependent insulin secretion, increase in insulin gene expression and beta-cell survival. Despite the initial technical difficulties and profound inefficiency of direct gene transfer into the pancreas that seriously restricted in vivo gene transfer experiments with GLP-1, recent exploitation of various routes of gene delivery and alternative means of gene transfer has permitted the detailed assessment of the therapeutic efficacy of GLP-1 in animal models of type 2 diabetes (T2DM). As a result, many clinical benefits of GLP-1 peptide/analogues observed in clinical trials involving induction of glucose tolerance, reduction of hyperglycaemia, suppression of appetite and food intake linked to weight loss have been replicated in animal models using gene therapy. Furthermore, GLP-1-centered gene therapy not only improved insulin sensitivity, but also reduced abdominal and/or hepatic fat associated with obesity-induced T2DM with drastic alterations in adipokine profiles in treated subjects. Thus, a comprehensive assessment of recent GLP-1-mediated gene therapy approaches with detailed analysis of current hurdles and resolutions, is discussed.
Collapse
|
19
|
Rodrigues A, Formas-Oliveira A, Bandeira V, Alves P, Hu W, Coroadinha A. Metabolic pathways recruited in the production of a recombinant enveloped virus: Mining targets for process and cell engineering. Metab Eng 2013; 20:131-45. [DOI: 10.1016/j.ymben.2013.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/22/2013] [Accepted: 10/03/2013] [Indexed: 11/27/2022]
|
20
|
Molino JVD, Viana Marques DDA, Júnior AP, Mazzola PG, Gatti MSV. Different types of aqueous two-phase systems for biomolecule and bioparticle extraction and purification. Biotechnol Prog 2013; 29:1343-53. [DOI: 10.1002/btpr.1792] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 06/28/2013] [Indexed: 12/19/2022]
Affiliation(s)
- João Vitor Dutra Molino
- Dept. of Biochemical and Pharmaceutical Technology; Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Block 16, Cidade Universitária; São Paulo 05508-000 Brazil
| | - Daniela de Araújo Viana Marques
- Dept. of Biochemical and Pharmaceutical Technology; Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Block 16, Cidade Universitária; São Paulo 05508-000 Brazil
| | - Adalberto Pessoa Júnior
- Dept. of Biochemical and Pharmaceutical Technology; Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Block 16, Cidade Universitária; São Paulo 05508-000 Brazil
| | - Priscila Gava Mazzola
- Dept. of Clinical Patology; Faculty of Medical Sciences; University of Campinas, Rua: Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz, Campinas; São Paulo 13083-887 Brazil
| | - Maria Silvia Viccari Gatti
- Genetics; Evolution and Bioagents Dept.; Biology Institute; University of Campinas, Rua: Monteiro Lobato, 255, Cidade Universitária “Zeferino Vaz,” Campinas; São Paulo 13083-862 Brazil
| |
Collapse
|
21
|
Situación actual de la infección pediátrica por virus de la inmunodeficiencia humana en España. An Pediatr (Barc) 2013; 79:133-5. [DOI: 10.1016/j.anpedi.2013.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 05/24/2013] [Indexed: 11/19/2022] Open
|
22
|
Suwanmanee T, Hu G, Gui T, Bartholomae CC, Kutschera I, von Kalle C, Schmidt M, Monahan PE, Kafri T. Integration-deficient lentiviral vectors expressing codon-optimized R338L human FIX restore normal hemostasis in Hemophilia B mice. Mol Ther 2013; 22:567-574. [PMID: 23941813 DOI: 10.1038/mt.2013.188] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 07/31/2013] [Indexed: 12/31/2022] Open
Abstract
Integration-deficient lentiviral vectors (IDLVs) have been shown to transduce a wide spectrum of target cells and organs in vitro and in vivo and to maintain long-term transgene expression in nondividing cells. However, epigenetic silencing of episomal vector genomes reduces IDLV transgene expression levels and renders these safe vectors less efficient. In this article, we describe for the first time a complete correction of factor IX (FIX) deficiency in hemophilia B mice by IDLVs carrying a novel, highly potent human FIX cDNA. A 50-fold increase in human FIX cDNA potency was achieved by combining two mechanistically independent yet synergistic strategies: (i) optimization of the human FIX cDNA codon usage to increase human FIX protein production per vector genome and (ii) generation of a highly catalytic mutant human FIX protein in which the arginine residue at position 338 was substituted with leucine. The enhanced human FIX activity was not associated with liver damage or with the formation of human FIX-directed inhibitory antibodies and rendered IDLV-treated FIX-knockout mice resistant to a challenging tail-clipping assay. A novel S1 nuclease-based B1-quantitative polymerase chain reaction assay showed low levels of IDLV integration in mouse liver. Overall, this study demonstrates that IDLVs carrying an improved human FIX cDNA safely and efficiently cure hemophilia B in a mouse model.
Collapse
Affiliation(s)
- Thipparat Suwanmanee
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Genlin Hu
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Tong Gui
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Cynthia C Bartholomae
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ina Kutschera
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul E Monahan
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Pediatrics, Division of Hematology/Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Tal Kafri
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA.
| |
Collapse
|
23
|
Gao J, Zhang C, Fu X, Yi Q, Tian F, Ning Q, Luo X. Effects of targeted suppression of glutaryl-CoA dehydrogenase by lentivirus-mediated shRNA and excessive intake of lysine on apoptosis in rat striatal neurons. PLoS One 2013; 8:e63084. [PMID: 23658800 PMCID: PMC3642093 DOI: 10.1371/journal.pone.0063084] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 03/29/2013] [Indexed: 12/31/2022] Open
Abstract
In glutaric aciduria type 1 (GA1), glutaryl-CoA dehydrogenase (GCDH) deficiency has been shown to be responsible for the accumulation of glutaric acid and striatal degeneration. However, the mechanisms by which GA1 induces striatal degeneration remain unclear. In this study, we aimed to establish a novel neuronal model of GA1 and to investigate the effects of GCDH deficiency and lysine-related metabolites on the viability of rat striatal neurons. Thus we constructed a lentiviral vector containing short hairpin RNA targeted against the GCDH gene expression (lentivirus-shRNA) in neurons. A virus containing a scrambled short hairpin RNA construct served as a control. Addition of lysine (5 mmol/L) was used to mimic hypermetabolism. Cell viability was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. Apoptosis was assessed using Hoechst33342 staining and Annexin V-PE/7-AAD staining. The mitochondrial membrane potential (MPP) was monitored using tetramethylrhodamine methyl ester. The expression levels of caspases 3, 8, and 9 were determined by Western blotting. We found that lentivirus-shRNA induced apoptosis and decreased MMP levels in neurons, and addition of 5 mmol/L lysine enhanced this effect markedly. Lentivirus-shRNA upregulated the protein levels of caspases 3 and 9 regardless of the presence of 5 mmol/L lysine. The expression level of caspase 8 was higher in neurons co-treated with lentivirus-shRNA and 5 mmol/L lysine than in control. Benzyloxy-carbonyl-Val-Ala-Asp(OMe)-fluoromethylketone, a pan-caspase inhibitor, blocked the apoptosis induced by lentivirus-shRNA and 5 mmol/L lysine to a great extent. These results indicate that the targeted suppression of GCDH by lentivirus-mediated shRNA and excessive intake of lysine may be a useful cell model of GA1. These also suggest that GA1-induced striatal degeneration is partially caspase-dependent.
Collapse
MESH Headings
- Amino Acid Metabolism, Inborn Errors/enzymology
- Amino Acid Metabolism, Inborn Errors/metabolism
- Amino Acid Metabolism, Inborn Errors/pathology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Base Sequence
- Biological Transport/genetics
- Brain Diseases, Metabolic/enzymology
- Brain Diseases, Metabolic/metabolism
- Brain Diseases, Metabolic/pathology
- Caspase Inhibitors/pharmacology
- Cell Survival/drug effects
- Cell Survival/genetics
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Gene Knockdown Techniques
- Glutaryl-CoA Dehydrogenase/deficiency
- Glutaryl-CoA Dehydrogenase/genetics
- Glutaryl-CoA Dehydrogenase/metabolism
- Lentivirus/genetics
- Lysine/metabolism
- Lysine/pharmacology
- Membrane Potential, Mitochondrial/drug effects
- Membrane Potential, Mitochondrial/genetics
- Neostriatum/cytology
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- RNA, Small Interfering/genetics
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Jinzhi Gao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cai Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Fu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Yi
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengyan Tian
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| |
Collapse
|
24
|
|
25
|
Coutant F, Sanchez David RY, Félix T, Boulay A, Caleechurn L, Souque P, Thouvenot C, Bourgouin C, Beignon AS, Charneau P. A nonintegrative lentiviral vector-based vaccine provides long-term sterile protection against malaria. PLoS One 2012; 7:e48644. [PMID: 23133649 PMCID: PMC3487763 DOI: 10.1371/journal.pone.0048644] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 09/27/2012] [Indexed: 01/06/2023] Open
Abstract
Trials testing the RTS,S candidate malaria vaccine and radiation-attenuated sporozoites (RAS) have shown that protective immunity against malaria can be induced and that an effective vaccine is not out of reach. However, longer-term protection and higher protection rates are required to eradicate malaria from the endemic regions. It implies that there is still a need to explore new vaccine strategies. Lentiviral vectors are very potent at inducing strong immunological memory. However their integrative status challenges their safety profile. Eliminating the integration step obviates the risk of insertional oncogenesis. Providing they confer sterile immunity, nonintegrative lentiviral vectors (NILV) hold promise as mass pediatric vaccine by meeting high safety standards. In this study, we have assessed the protective efficacy of NILV against malaria in a robust pre-clinical model. Mice were immunized with NILV encoding Plasmodium yoelii Circumsporozoite Protein (Py CSP) and challenged with sporozoites one month later. In two independent protective efficacy studies, 50% (37.5-62.5) of the animals were fully protected (p = 0.0072 and p = 0.0008 respectively when compared to naive mice). The remaining mice with detectable parasitized red blood cells exhibited a prolonged patency and reduced parasitemia. Moreover, protection was long-lasting with 42.8% sterile protection six months after the last immunization (p = 0.0042). Post-challenge CD8+ T cells to CSP, in contrast to anti-CSP antibodies, were associated with protection (r = -0.6615 and p = 0.0004 between the frequency of IFN-g secreting specific T cells in spleen and parasitemia). However, while NILV and RAS immunizations elicited comparable immunity to CSP, only RAS conferred 100% of sterile protection. Given that a better protection can be anticipated from a multi-antigen vaccine and an optimized vector design, NILV appear as a promising malaria vaccine.
Collapse
Affiliation(s)
- Frédéric Coutant
- Unité Virologie Moléculaire et Vaccinologie, Department of Virology, Institut Pasteur and CNRS URA3015, Institut Pasteur, Paris, France
| | - Raul Yusef Sanchez David
- Unité Virologie Moléculaire et Vaccinologie, Department of Virology, Institut Pasteur and CNRS URA3015, Institut Pasteur, Paris, France
| | - Tristan Félix
- Unité Virologie Moléculaire et Vaccinologie, Department of Virology, Institut Pasteur and CNRS URA3015, Institut Pasteur, Paris, France
| | - Aude Boulay
- Unité Virologie Moléculaire et Vaccinologie, Department of Virology, Institut Pasteur and CNRS URA3015, Institut Pasteur, Paris, France
| | - Laxmee Caleechurn
- Unité Virologie Moléculaire et Vaccinologie, Department of Virology, Institut Pasteur and CNRS URA3015, Institut Pasteur, Paris, France
| | - Philippe Souque
- Unité Virologie Moléculaire et Vaccinologie, Department of Virology, Institut Pasteur and CNRS URA3015, Institut Pasteur, Paris, France
| | - Catherine Thouvenot
- Centre de Production et d’Infection des Anophèles (CEPIA), Department of Parasitology and Mycology, Institut Pasteur, Paris, France
| | - Catherine Bourgouin
- Centre de Production et d’Infection des Anophèles (CEPIA), Department of Parasitology and Mycology, Institut Pasteur, Paris, France
| | - Anne-Sophie Beignon
- Unité Virologie Moléculaire et Vaccinologie, Department of Virology, Institut Pasteur and CNRS URA3015, Institut Pasteur, Paris, France
| | - Pierre Charneau
- Unité Virologie Moléculaire et Vaccinologie, Department of Virology, Institut Pasteur and CNRS URA3015, Institut Pasteur, Paris, France
| |
Collapse
|