1
|
Tscherne A, Kalodimou G, Kupke A, Rohde C, Freudenstein A, Jany S, Kumar S, Sutter G, Krähling V, Becker S, Volz A. Rapid Development of Modified Vaccinia Virus Ankara (MVA)-Based Vaccine Candidates Against Marburg Virus Suitable for Clinical Use in Humans. Vaccines (Basel) 2024; 12:1316. [PMID: 39771978 PMCID: PMC11680136 DOI: 10.3390/vaccines12121316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Marburg virus (MARV) is the etiological agent of Marburg Virus Disease (MVD), a rare but severe hemorrhagic fever disease with high case fatality rates in humans. Smaller outbreaks have frequently been reported in countries in Africa over the last few years, and confirmed human cases outside Africa are, so far, exclusively imported by returning travelers. Over the previous years, MARV has also spread to non-endemic African countries, demonstrating its potential to cause epidemics. Although MARV-specific vaccines are evaluated in preclinical and clinical research, none have been approved for human use. Modified Vaccinia virus Ankara (MVA), a well-established viral vector used to generate vaccines against emerging pathogens, can deliver multiple antigens and has a remarkable clinical safety and immunogenicity record, further supporting its evaluation as a vaccine against MARV. The rapid availability of safe and effective MVA-MARV vaccine candidates would expand the possibilities of multi-factored intervention strategies in endemic countries. METHODS We have used an optimized methodology to rapidly generate and characterize recombinant MVA candidate vaccines that meet the quality requirements to proceed to human clinical trials. As a proof-of-concept for the optimized methodology, we generated two recombinant MVAs that deliver either the MARV glycoprotein (MVA-MARV-GP) or the MARV nucleoprotein (MVA-MARV-NP). RESULTS Infections of human cell cultures with recombinant MVA-MARV-GP and MVA-MARV-NP confirmed the efficient synthesis of MARV-GP and MARV-NP proteins in mammalian cells, which are non-permissive for MVA replication. Prime-boost immunizations in C57BL/6J mice readily induced circulating serum antibodies binding to recombinant MARV-GP and MARV-NP proteins. Moreover, the MVA-MARV-candidate vaccines elicited MARV-specific T-cell responses in C57BL/6J mice. CONCLUSIONS We confirmed the suitability of our two backbone viruses MVA-mCherry and MVA-GFP in a proof-of-concept study to rapidly generate candidate vaccines against MARV. However, further studies are warranted to characterize the protective efficacy of these recombinant MVA-MARV vaccines in other preclinical models and to evaluate them as vaccine candidates in humans.
Collapse
Affiliation(s)
- Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany
| | - Georgia Kalodimou
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany
| | - Alexandra Kupke
- Institute of Virology, Philipps University Marburg, 35037 Marburg, Germany; (A.K.); (C.R.); (S.B.)
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, 35037 Marburg, Germany
| | - Cornelius Rohde
- Institute of Virology, Philipps University Marburg, 35037 Marburg, Germany; (A.K.); (C.R.); (S.B.)
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, 35037 Marburg, Germany
| | - Astrid Freudenstein
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
| | - Sylvia Jany
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
| | - Satendra Kumar
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximilians University (LMU Munich), 85764 Oberschleißheim, Germany; (A.T.); (G.K.); (A.F.); (S.J.); (S.K.); (G.S.)
- German Center for Infection Research, Partner Site Munich, 85764 Oberschleißheim, Germany
| | - Verena Krähling
- Institute of Virology, Philipps University Marburg, 35037 Marburg, Germany; (A.K.); (C.R.); (S.B.)
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, 35037 Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, 35037 Marburg, Germany; (A.K.); (C.R.); (S.B.)
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, 35037 Marburg, Germany
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- German Center for Infection Research, Partner Site Hannover-Braunschweig, 30559 Hannover, Germany
| |
Collapse
|
2
|
D'Alise AM, Leoni G, Cotugno G, Siani L, Vitale R, Ruzza V, Garzia I, Antonucci L, Micarelli E, Venafra V, Gogov S, Capone A, Runswick S, Martin‐Liberal J, Calvo E, Moreno V, Symeonides SN, Scarselli E, Bechter O. Phase I Trial of Viral Vector-Based Personalized Vaccination Elicits Robust Neoantigen-Specific Antitumor T-Cell Responses. Clin Cancer Res 2024; 30:2412-2423. [PMID: 38506710 PMCID: PMC11145154 DOI: 10.1158/1078-0432.ccr-23-3940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/13/2024] [Accepted: 03/18/2024] [Indexed: 03/21/2024]
Abstract
PURPOSE Personalized vaccines targeting multiple neoantigens (nAgs) are a promising strategy for eliciting a diversified antitumor T-cell response to overcome tumor heterogeneity. NOUS-PEV is a vector-based personalized vaccine, expressing 60 nAgs and consists of priming with a nonhuman Great Ape Adenoviral vector (GAd20) followed by boosts with Modified Vaccinia Ankara. Here, we report data of a phase Ib trial of NOUS-PEV in combination with pembrolizumab in treatment-naïve patients with metastatic melanoma (NCT04990479). PATIENTS AND METHODS The feasibility of this approach was demonstrated by producing, releasing, and administering to 6 patients 11 of 12 vaccines within 8 weeks from biopsy collection to GAd20 administration. RESULTS The regimen was safe, with no treatment-related serious adverse events observed and mild vaccine-related reactions. Vaccine immunogenicity was demonstrated in all evaluable patients receiving the prime/boost regimen, with detection of robust neoantigen-specific immune responses to multiple neoantigens comprising both CD4 and CD8 T cells. Expansion and diversification of vaccine-induced T-cell receptor (TCR) clonotypes was observed in the posttreatment biopsies of patients with clinical response, providing evidence of tumor infiltration by vaccine-induced neoantigen-specific T cells. CONCLUSIONS These findings indicate the ability of NOUS-PEV to amplify and broaden the repertoire of tumor-reactive T cells to empower a diverse, potent, and durable antitumor immune response. Finally, a gene signature indicative of the reduced presence of activated T cells together with very poor expression of the antigen-processing machinery genes has been identified in pretreatment biopsies as a potential biomarker of resistance to the treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Veronica Venafra
- Department of Biology, University of Rome “Tor Vergata,” Rome, Italy
| | | | | | | | | | - Emiliano Calvo
- START Madrid‐CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Victor Moreno
- START Madrid‐FJD, Hospital Fundacion Jimenez Díaz, Madrid, Spain
| | - Stefan N. Symeonides
- Edinburgh Experimental Cancer Medicine Centre, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Oliver Bechter
- Leuven Cancer Institute, Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Dawson LM, Alshawabkeh M, Schröer K, Arakrak F, Ehrhardt A, Zhang W. Role of homologous recombination/recombineering on human adenovirus genome engineering: Not the only but the most competent solution. ENGINEERING MICROBIOLOGY 2024; 4:100140. [PMID: 39628785 PMCID: PMC11611009 DOI: 10.1016/j.engmic.2024.100140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 12/06/2024]
Abstract
Adenoviruses typically cause mild illnesses, but severe diseases may occur primarily in immunodeficient individuals, particularly children. Recently, adenoviruses have garnered significant interest as a versatile tool in gene therapy, tumor treatment, and vaccine vector development. Over the past two decades, the advent of recombineering, a method based on homologous recombination, has notably enhanced the utility of adenoviral vectors in therapeutic applications. This review summarizes recent advancements in the use of human adenoviral vectors in medicine and discusses the pivotal role of recombineering in the development of these vectors. Additionally, it highlights the current achievements and potential future impact of therapeutic adenoviral vectors.
Collapse
Affiliation(s)
| | | | | | - Fatima Arakrak
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Stockumer Str. 10 58453 Witten, Germany
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Stockumer Str. 10 58453 Witten, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Stockumer Str. 10 58453 Witten, Germany
| |
Collapse
|
4
|
Boselli D, Panigada M, Di Terlizzi S, Romanò M, Canonico E, Villa C, Minici C, van Anken E, Soprana E, Siccardi AG. Time- and cost-effective production of untagged recombinant MVA by flow virometry and direct virus sorting. J Transl Med 2023; 21:495. [PMID: 37482614 PMCID: PMC10364397 DOI: 10.1186/s12967-023-04353-7] [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: 06/17/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Recombinant MVAs (rMVAs) are widely used both in basic and clinical research. Our previously developed Red-to-Green Gene Swapping Method (RGGSM), a cytometry-based Cell-Sorting protocol, revolves around the transient expression of a green fluorescent cytoplasmic marker, to subsequently obtain purified untagged rMVA upon loss of that marker by site-specific recombination. The standard RGSSM is quite costly in terms of bench work, reagents, and Sorting Facility fees. Although faster than other methods to obtain recombinant MVAs, the standard RGSSM still is time-consuming, taking at least 25 days to yield the final product. METHODS The direct sorting of fluorescent virions is made amenable by the marker HAG, a flu hemagglutinin/EGFP fusion protein, integrated into the external envelope of extracellular enveloped virions (EEVs). Fluorescent EEVs-containing supernatants of infected cultures are used instead of purified virus. Direct Virus-Sorting was performed on BD FACSAria Fusion cell sorter equipped with 4 lasers and a 100-mm nozzle, with 20 psi pressure and a minimal flow rate, validated using Megamix beads. RESULTS Upon infection of cells with recombinant EEVs, at the first sorting step virions that contain HAG are harvested and cloned, while the second sorting step yields EEVs that have lost HAG, allowing to clone untagged rMVA. Because only virion-containing supernatants are used, no virus purification steps and fewer sortings are necessary. Therefore, the final untagged rMVA product can be obtained in a mere 8 days. CONCLUSIONS Altogether, we report that the original RGSSM has been markedly improved in terms of time- and cost efficiency by substituting Cell-Sorting with direct Virus-Sorting from the supernatants of infected cells. The improved virometry-based RGGSM may find wide applicability, considering that rMVAs hold great promise to serve as personalized vaccines for therapeutic intervention against cancer and various types of infectious diseases.
Collapse
Affiliation(s)
| | - Maddalena Panigada
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Monica Romanò
- FRACTAL - San Raffaele Scientific Institute, Milan, Italy
| | | | - Chiara Villa
- FRACTAL - San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Minici
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Elisa Soprana
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio G Siccardi
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
5
|
Seclì L, Infante L, Nocchi L, De Lucia M, Cotugno G, Leoni G, Micarelli E, Garzia I, Avalle L, Sdruscia G, Troise F, Allocca S, Romano G, Scarselli E, D'Alise AM. Vector Aided Microenvironment programming (VAMP): reprogramming the TME with MVA virus expressing IL-12 for effective antitumor activity. J Immunother Cancer 2023; 11:jitc-2023-006718. [PMID: 37117006 PMCID: PMC10151998 DOI: 10.1136/jitc-2023-006718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Tumor microenvironment (TME) represents a critical hurdle in cancer immunotherapy, given its ability to suppress antitumor immunity. Several efforts are made to overcome this hostile TME with the development of new therapeutic strategies modifying TME to boost antitumor immunity. Among these, cytokine-based approaches have been pursued for their known immunomodulatory effects on different cell populations within the TME. IL-12 is a potent pro-inflammatory cytokine that demonstrates striking immune activation and tumor control but causes severe adverse effects when systemically administered. Thus, local administration is considered a potential strategy to achieve high cytokine concentrations at the tumor site while sparing systemic adverse effects. METHODS Modified Vaccinia Ankara (MVA) vector is a potent inducer of pro-inflammatory response. Here, we cloned IL-12 into the genome of MVA for intratumoral immunotherapy, combining the immunomodulatory properties of both the vector and the cargo. The antitumor activity of MVA-IL-12 and its effect on TME reprogramming were investigated in preclinical tumor models. RNA sequencing (RNA-Seq) analysis was performed to assess changes in the TME in treated and distal tumors and the effect on the intratumoral T-cell receptor repertoire. RESULTS Intratumoral injection of MVA-IL-12 resulted in strong antitumor activity with the complete remission of established tumors in multiple murine models, including those resistant to checkpoint inhibitors. The therapeutic activity of MVA-IL-12 was associated with very low levels of circulating cytokine. Effective TME reprogramming was demonstrated on treatment, with the reduction of immunosuppressive M2 macrophages while increasing pro-inflammatory M1, and recruitment of dendritic cells. TME switch from immunosuppressive into immunostimulatory environment allowed for CD8 T cells priming and expansion leading to tumor attack. CONCLUSIONS Intratumoral administration of MVA-IL-12 turns immunologically 'cold' tumors 'hot' and overcomes resistance to programmed cell death protein-1 blockade.
Collapse
Affiliation(s)
| | - Luigia Infante
- NousCom, Rome, Italy
- University of Rome Tor Vergata, Roma, Lazio, Italy
| | | | | | | | | | | | | | - Lidia Avalle
- Department of Molecular Biotechnology and Health Science, University of Turin, Torino, Piemonte, Italy
| | | | | | | | | | | | | |
Collapse
|
6
|
Natalini A, Simonetti S, Favaretto G, Lucantonio L, Peruzzi G, Muñoz-Ruiz M, Kelly G, Contino AM, Sbrocchi R, Battella S, Capone S, Folgori A, Nicosia A, Santoni A, Hayday AC, Di Rosa F. Improved memory CD8 T cell response to delayed vaccine boost is associated with a distinct molecular signature. Front Immunol 2023; 14:1043631. [PMID: 36865556 PMCID: PMC9973452 DOI: 10.3389/fimmu.2023.1043631] [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: 09/13/2022] [Accepted: 01/09/2023] [Indexed: 02/16/2023] Open
Abstract
Effective secondary response to antigen is a hallmark of immunological memory. However, the extent of memory CD8 T cell response to secondary boost varies at different times after a primary response. Considering the central role of memory CD8 T cells in long-lived protection against viral infections and tumors, a better understanding of the molecular mechanisms underlying the changing responsiveness of these cells to antigenic challenge would be beneficial. We examined here primed CD8 T cell response to boost in a BALB/c mouse model of intramuscular vaccination by priming with HIV-1 gag-encoding Chimpanzee adenovector, and boosting with HIV-1 gag-encoding Modified Vaccinia virus Ankara. We found that boost was more effective at day(d)100 than at d30 post-prime, as evaluated at d45 post-boost by multi-lymphoid organ assessment of gag-specific CD8 T cell frequency, CD62L-expression (as a guide to memory status) and in vivo killing. RNA-sequencing of splenic gag-primed CD8 T cells at d100 revealed a quiescent, but highly responsive signature, that trended toward a central memory (CD62L+) phenotype. Interestingly, gag-specific CD8 T cell frequency selectively diminished in the blood at d100, relative to the spleen, lymph nodes and bone marrow. These results open the possibility to modify prime/boost intervals to achieve an improved memory CD8 T cell secondary response.
Collapse
Affiliation(s)
- Ambra Natalini
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
| | - Sonia Simonetti
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
| | - Gabriele Favaretto
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
| | - Lorenzo Lucantonio
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy.,Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Miguel Muñoz-Ruiz
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Gavin Kelly
- Bioinformatic and Biostatistics Science and Technology Platform, The Francis Crick Institute, London, United Kingdom
| | | | | | | | | | | | - Alfredo Nicosia
- CEINGE, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Adrian C Hayday
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom.,Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom.,National Institute for Health Research (NIHR), Biomedical Research Center (BRC), Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Francesca Di Rosa
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
| |
Collapse
|
7
|
Zhao C, Gao J, Wang Y, Ji L, Qin H, Hu W, Yang Y. A Novel Rabies Vaccine Based on a Recombinant Bovine Herpes Virus Type 1 Expressing Rabies Virus Glycoprotein. Front Microbiol 2022; 13:931043. [PMID: 35755997 PMCID: PMC9213812 DOI: 10.3389/fmicb.2022.931043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 11/24/2022] Open
Abstract
Rabies is a highly prevalent zoonotic disease and a public health threat worldwide. Currently licensed rabies vaccines are effective but less is known which would protect cattle. This study describes the construction of a novel recombinant bovine herpes virus type I (BHV-1) expressing rabies virus glycoprotein (RABV G) instead of its gE glycoprotein (gE) by CRISPR-Cas9 and homologous recombination technology (BHV-1-ΔgE-G). Insertion of the RABV G gene is stable after 20 rounds of in vitro passaging and the recombinant virus replicates to high titers in MDBK cells. The RABV G expresses in the recombinant virus-infected cells and on the virion surface of BHV-1-ΔgE-G. One single immunization with BHV-1-ΔgE-G-activated dendritic cells (DCs) and B cells furthermore induced a protective immune response in mice against severe lethal challenge infection. A protective level of RABV-specific virus-neutralizing antibody (VNA) was detected in intramuscular immunized mice and cattle without any clinical symptoms. This research demonstrated that the BHV-1 vector-based RABV vaccine is a potential candidate for cattle.
Collapse
Affiliation(s)
- Caiquan Zhao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China.,School of Biological Science and Technology, Baotou Teachers' College, Baotou, China
| | - Jie Gao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yongzhi Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Lina Ji
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Hui Qin
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Wei Hu
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yang Yang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
8
|
Reguzova A, Fischer N, Müller M, Salomon F, Jaenisch T, Amann R. A Novel Orf Virus D1701-VrV-Based Dengue Virus (DENV) Vaccine Candidate Expressing HLA-Specific T Cell Epitopes: A Proof-of-Concept Study. Biomedicines 2021; 9:biomedicines9121862. [PMID: 34944678 PMCID: PMC8698572 DOI: 10.3390/biomedicines9121862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/02/2022] Open
Abstract
Although dengue virus (DENV) affects almost half of the world’s population there are neither preventive treatments nor any long-lasting and protective vaccines available at this time. The complexity of the protective immune response to DENV is still not fully understood. The most advanced vaccine candidates focus specifically on humoral immune responses and the production of virus-neutralizing antibodies. However, results from several recent studies have revealed the protective role of T cells in the immune response to DENV. Hence, in this study, we generated a novel and potent DENV vaccine candidate based on an Orf virus (ORFV, genus Parapoxvirus) vector platform engineered to encode five highly conserved or cross-reactive DENV human leukocyte antigen (HLA)-A*02- or HLA-B*07-restricted epitopes as minigenes (ORFV-DENV). We showed that ORFV-DENV facilitates the in vitro priming of CD8+ T cells from healthy blood donors based on responses to each of the encoded immunogenic peptides. Moreover, we demonstrated that peripheral blood mononuclear cells isolated from clinically confirmed DENV-positive donors stimulated with ORFV-DENV generate cytotoxic T cell responses to at least three of the expressed DENV peptides. Finally, we showed that ORFV-DENV could activate CD8+ T cells isolated from donors who had recovered from Zika virus (ZIKV) infection. ZIKV belongs to the same virus family (Flaviviridae) and has epitope sequences that are homologous to those of DENV. We found that highly conserved HLA-B*07-restricted ZIKV and DENV epitopes induced functional CD8+ T cell responses in PBMCs isolated from confirmed ZIKV-positive donors. In summary, this proof-of-concept study characterizes a promising new ORFV D1701-VrV-based DENV vaccine candidate that induces broad and functional epitope-specific CD8+ T cell responses.
Collapse
Affiliation(s)
- Alena Reguzova
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (A.R.); (M.M.); (F.S.)
| | - Nico Fischer
- Department of Infectious Diseases, Heidelberg Institute of Global Health (HIGH) & Tropical Medicine, Heidelberg University Hospital, 69120 Heidelberg, Germany; (N.F.); (T.J.)
| | - Melanie Müller
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (A.R.); (M.M.); (F.S.)
| | - Ferdinand Salomon
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (A.R.); (M.M.); (F.S.)
| | - Thomas Jaenisch
- Department of Infectious Diseases, Heidelberg Institute of Global Health (HIGH) & Tropical Medicine, Heidelberg University Hospital, 69120 Heidelberg, Germany; (N.F.); (T.J.)
| | - Ralf Amann
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany; (A.R.); (M.M.); (F.S.)
- Correspondence: ; Tel.: +49-707-1298-7614
| |
Collapse
|
9
|
Leoni G, D'Alise AM, Cotugno G, Langone F, Garzia I, De Lucia M, Fichera I, Vitale R, Bignone V, Tucci FG, Mori F, Leuzzi A, Di Matteo E, Troise F, Abbate A, Merone R, Ruzza V, Diodoro MG, Yadav M, Gordon-Alonso M, Vanhaver C, Panigada M, Soprana E, Siccardi A, Folgori A, Colloca S, van der Bruggen P, Nicosia A, Lahm A, Catanese MT, Scarselli E. A Genetic Vaccine Encoding Shared Cancer Neoantigens to Treat Tumors with Microsatellite Instability. Cancer Res 2020; 80:3972-3982. [PMID: 32690723 DOI: 10.1158/0008-5472.can-20-1072] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/01/2020] [Accepted: 07/15/2020] [Indexed: 11/16/2022]
Abstract
Tumors with microsatellite instability (MSI) are caused by a defective DNA mismatch repair system that leads to the accumulation of mutations within microsatellite regions. Indels in microsatellites of coding genes can result in the synthesis of frameshift peptides (FSP). FSPs are tumor-specific neoantigens shared across patients with MSI. In this study, we developed a neoantigen-based vaccine for the treatment of MSI tumors. Genetic sequences from 320 MSI tumor biopsies and matched healthy tissues in The Cancer Genome Atlas database were analyzed to select shared FSPs. Two hundred nine FSPs were selected and cloned into nonhuman Great Ape Adenoviral and Modified Vaccinia Ankara vectors to generate a viral-vectored vaccine, referred to as Nous-209. Sequencing tumor biopsies of 20 independent patients with MSI colorectal cancer revealed that a median number of 31 FSPs out of the 209 encoded by the vaccine was detected both in DNA and mRNA extracted from each tumor biopsy. A relevant number of peptides encoded by the vaccine were predicted to bind patient HLA haplotypes. Vaccine immunogenicity was demonstrated in mice with potent and broad induction of FSP-specific CD8 and CD4 T-cell responses. Moreover, a vaccine-encoded FSP was processed in vitro by human antigen-presenting cells and was subsequently able to activate human CD8 T cells. Nous-209 is an "off-the-shelf" cancer vaccine encoding many neoantigens shared across sporadic and hereditary MSI tumors. These results indicate that Nous-209 can induce the optimal breadth of immune responses that might achieve clinical benefit to treat and prevent MSI tumors. SIGNIFICANCE: These findings demonstrate the feasibility of an "off-the-shelf" vaccine for treatment and prevention of tumors harboring frameshift mutations and neoantigenic peptides as a result of microsatellite instability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Cristophe Vanhaver
- de Duve Institute and the Université catholique de Louvain, Brussels, Belgium
| | - Maddalena Panigada
- Molecular Immunology Unit, San Raffaele Research Institute, Milan, Italy
| | - Elisa Soprana
- Molecular Immunology Unit, San Raffaele Research Institute, Milan, Italy
| | - Antonio Siccardi
- Molecular Immunology Unit, San Raffaele Research Institute, Milan, Italy
| | | | | | | | - Alfredo Nicosia
- Nouscom AG, Bäumleingasse, Basel, Switzerland.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.,CEINGE, Via Comunale Margherita, Naples, Italy
| | | | | | | |
Collapse
|
10
|
EPPIC (Efficient Purification by Parental Inducer Constraint) Platform for Rapid Generation of Recombinant Vaccinia Viruses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:731-738. [PMID: 32346550 PMCID: PMC7177189 DOI: 10.1016/j.omtm.2020.03.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
Abstract
Vaccinia virus (VACV) was successfully used as a vaccine in the smallpox eradication campaign. Since then, it has been widely used in the development of vaccine and therapeutic vectors. However, methods of generating and purifying recombinant VACVs (rVACVs) are often time-consuming, cumbersome, and in some cases require specialized cell lines or equipment. Here, we describe a novel EPPIC (Efficient Purification by Parental Inducer Constraint) platform for the rapid generation of rVACVs using a replication-inducible VACV (vIND) as a parental virus for homologous recombination. Purification of the rVACV from the parental vIND is achieved by two serial passages in the absence of inducer (i.e., parental inducer "constraint") in standard laboratory cell lines, without the need for specialized equipment, within 1 week. We determined the optimal conditions for homologous recombination and serial purification and generated a suite of vIND parental viruses to facilitate customization of the platform. Importantly, the EPPIC platform can be adapted to rapidly generate replication-deficient and replication-competent rVACVs expressing vaccine or therapeutic antigens, with or without screening markers, by simple modifications to a DNA shuttle vector, thus allowing the rapid development, updating, and refinement of personalized or custom vaccines and therapeutic vectors in a matter of days.
Collapse
|
11
|
Genomic Characterization of Orf Virus Strain D1701-V ( Parapoxvirus) and Development of Novel Sites for Multiple Transgene Expression. Viruses 2019; 11:v11020127. [PMID: 30704093 PMCID: PMC6409557 DOI: 10.3390/v11020127] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 02/06/2023] Open
Abstract
The Orf virus (ORFV; Parapoxvirus) strain D1701 with an attenuated phenotype and excellent immunogenic capacity is successfully used for the generation of recombinant vaccines against different viral infections. Adaption for growth in Vero cells was accompanied by additional major genomic changes resulting in ORFV strain variant D1701-V. In this study, restriction enzyme mapping, blot hybridization and DNA sequencing of the deleted region s (A, AT and D) in comparison to the predecessor strain D1701-B revealed the loss of 7 open reading frames (ORF008, ORF101, ORF102, ORF114, ORF115, ORF116, ORF117). The suitability of deletion site D for expression of foreign genes is demonstrated using novel synthetic early promoter eP1 and eP2. Comparison of promoter strength showed that the original vegf-e promoter Pv as well as promoter eP2 display an up to 11-fold stronger expression than promoter eP1, irrespective of the insertion site. Successful integration and expression of the fluorescent marker genes is demonstrated by gene- and insertion-site specific PCR assays, fluorescence microscopy and flow cytometry. For the first time ORFV recombinants are generated simultaneously expressing transgenes in two different insertion loci. That allows production of polyvalent vaccines containing several antigens against one or different pathogens in a single vectored ORFV vaccine.
Collapse
|
12
|
Simonetti S, Natalini A, Folgori A, Capone S, Nicosia A, Santoni A, Di Rosa F. Antigen-specific CD8 T cells in cell cycle circulate in the blood after vaccination. Scand J Immunol 2019; 89:e12735. [PMID: 30488973 PMCID: PMC6850756 DOI: 10.1111/sji.12735] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/20/2022]
Abstract
Although clonal expansion is a hallmark of adaptive immunity, the location(s) where antigen‐responding T cells enter cell cycle and complete it have been poorly explored. This lack of knowledge stems partially from the limited experimental approaches available. By using Ki67 plus DNA staining and a novel strategy for flow cytometry analysis, we distinguished antigen‐specific CD8 T cells in G0, in G1 and in S‐G2/M phases of cell cycle after intramuscular vaccination of BALB/c mice with antigen‐expressing viral vectors. Antigen‐specific cells in S‐G2/M were present at early times after vaccination in lymph nodes (LNs), spleen and, surprisingly, also in the blood, which is an unexpected site for cycling of normal non‐leukaemic cells. Most proliferating cells had high scatter profile and were undetected by current criteria of analysis, which under‐estimated up to 6 times antigen‐specific cell frequency in LNs. Our discovery of cycling antigen‐specific CD8 T cells in the blood opens promising translational perspectives.
Collapse
Affiliation(s)
- Sonia Simonetti
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy.,Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy
| | - Ambra Natalini
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy.,Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy
| | | | | | - Alfredo Nicosia
- Keires AG, Basel, Switzerland.,CEINGE - Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Angela Santoni
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy.,Department of Molecular Medicine, University of Rome "Sapienza", Rome, Italy.,Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Francesca Di Rosa
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
| |
Collapse
|
13
|
Conrad SJ, Liu J. Poxviruses as Gene Therapy Vectors: Generating Poxviral Vectors Expressing Therapeutic Transgenes. Methods Mol Biol 2019; 1937:189-209. [PMID: 30706397 DOI: 10.1007/978-1-4939-9065-8_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Treatments with poxvirus vectors can have long-lasting immunological impact in the host, and thus they have been extensively studied to treat diseases and for vaccine development. More importantly, the oncolytic properties of poxviruses have led to their development as cancer therapeutics. Two poxviruses, vaccinia virus (VACV) and myxoma virus (MYXV), have been extensively studied as virotherapeutics with promising results. Vaccinia virus vectors have advanced to the clinic and have been tested as oncolytic therapeutics for several cancer types with successes in phase I/II clinical trials. In addition to oncolytic applications, MYXV has been explored for additional applications including immunotherapeutics, purging of cancer progenitor cells, and treatments for graft-versus-host diseases. These novel therapeutic applications have encouraged its advancement into clinical trials. To meet the demands of different treatment needs, VACV and MYXV can be genetically engineered to express therapeutic transgenes. The engineering process used in poxvirus vectors can be very different from that of other DNA virus vectors (e.g., the herpesviruses). This chapter is intended to serve as a guide to those wishing to engineer poxvirus vectors for therapeutic transgene expression and to produce viral preparations for preclinical studies.
Collapse
Affiliation(s)
- Steven J Conrad
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA
| | - Jia Liu
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA. .,The Center for Microbial Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
14
|
Barbieri A, Panigada M, Soprana E, Di Mario G, Gubinelli F, Bernasconi V, Recagni M, Donatelli I, Castrucci MR, Siccardi AG. Strategies to obtain multiple recombinant modified vaccinia Ankara vectors. Applications to influenza vaccines. J Virol Methods 2017; 251:7-14. [PMID: 28987424 DOI: 10.1016/j.jviromet.2017.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 10/02/2017] [Accepted: 10/02/2017] [Indexed: 11/27/2022]
Abstract
As a vaccination vector, MVA has been widely investigated both in animal models and humans. The construction of recombinant MVA (rMVA) relies on homologous recombination between an acceptor virus and a donor plasmid in infected/transfected permissive cells. Our construction strategy "Red-to-Green gene swapping" - based on the exchange of two fluorescent markers within the flanking regions of MVA deletion ΔIII, coupled to fluorescence activated cell sorting - is here extended to a second insertion site, within the flanking regions of MVA deletion ΔVI. Exploiting this strategy, both double and triple rMVA were constructed, expressing as transgenes the influenza A proteins HA, NP, M1, and PB1. Upon validation of the harbored transgenes co-expression, double and triple recombinants rMVA(ΔIII)-NP-P2A-M1 and rMVA(ΔIII)-NP-P2A-M1-(ΔVI)-PB1 were assayed for in vivo immunogenicity and protection against lethal challenge. In vivo responses were identical to those obtained with the reported combinations of single recombinants, supporting the feasibility and reliability of the present improvement and the extension of Red-to-Green gene swapping to insertion sites other than ΔIII.
Collapse
Affiliation(s)
- Andrea Barbieri
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli, 32, 20129, Milan, Italy
| | - Maddalena Panigada
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Elisa Soprana
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Giuseppina Di Mario
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Francesco Gubinelli
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Valentina Bernasconi
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Marta Recagni
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Isabella Donatelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Maria R Castrucci
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Antonio G Siccardi
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy.
| |
Collapse
|
15
|
Guo ZS, Liu Z, Sathaiah M, Wang J, Ravindranathan R, Kim E, Huang S, Kenniston TW, Bell JC, Zeh HJ, Butterfield LH, Gambotto A, Bartlett DL. Rapid Generation of Multiple Loci-Engineered Marker-free Poxvirus and Characterization of a Clinical-Grade Oncolytic Vaccinia Virus. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:112-122. [PMID: 29085848 PMCID: PMC5651493 DOI: 10.1016/j.omtm.2017.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/27/2017] [Indexed: 12/13/2022]
Abstract
Recombinant poxviruses, utilized as vaccine vectors and oncolytic viruses, often require manipulation at multiple genetic loci in the viral genome. It is essential for viral vectors to possess no adventitious mutations and no (antibiotic) selection marker in the final product for human patients in order to comply with the guidance from the regulatory agencies. Rintoul et al. have previously developed a selectable and excisable marker (SEM) system for the rapid generation of recombinant vaccinia virus. In the current study, we describe an improved methodology for rapid creation and selection of recombinant poxviruses with multiple genetic manipulations solely based on expression of a fluorescent protein and with no requirement for drug selection that can lead to cellular stress and the risk of adventitious mutations throughout the viral genome. Using this improved procedure combined with the SEM system, we have constructed multiple marker-free oncolytic poxviruses expressing different cytokines and other therapeutic genes. The high fidelity of inserted DNA sequences validates the utility of this improved procedure for generation of therapeutic viruses for human patients. We have created an oncolytic poxvirus expressing human chemokine CCL5, designated as vvDD-A34R-hCCL5, with manipulations at two genetic loci in a single virus. Finally, we have produced and purified this virus in clinical grade for its use in a phase I clinical trial and presented data on initial in vitro characterization of the virus.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Corresponding author: Zong Sheng Guo, PhD, UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Zuqiang Liu
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Magesh Sathaiah
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiahu Wang
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Roshni Ravindranathan
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eun Kim
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shaohua Huang
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Thomas W. Kenniston
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John C. Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Herbert J. Zeh
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lisa H. Butterfield
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Departments of Medicine and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrea Gambotto
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L. Bartlett
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Di Mario G, Soprana E, Gubinelli F, Panigada M, Facchini M, Fabiani C, Garulli B, Basileo M, Cassone A, Siccardi A, Donatelli I, Castrucci MR. Immunogenicity of modified vaccinia virus Ankara expressing the hemagglutinin stalk domain of pandemic (H1N1) 2009 influenza virus. Pathog Glob Health 2017; 111:69-75. [PMID: 28081672 PMCID: PMC5375617 DOI: 10.1080/20477724.2016.1275464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND Vaccination offers protection against influenza, although current vaccines need to be reformulated each year. The development of a broadly protective influenza vaccine would guarantee the induction of heterosubtypic immunity also against emerging influenza viruses of a novel subtype. Vaccine candidates based on the stalk region of the hemagglutinin (HA) have the potential to induce broad and persistent protection against diverse influenza A viruses. METHODS Modified vaccinia virus Ankara (MVA) expressing a headless HA (hlHA) of A/California/4/09 (CA/09) virus was used as a vaccine to immunize C57BL/6 mice. Specific antibody and cell-mediated immune responses were determined, and challenge experiments were performed by infecting vaccinated mice with CA/09 virus. RESULTS Immunization of mice with CA/09-derived hlHA, vectored by MVA, was able to elicit influenza-specific broad cross-reactive antibodies and cell-mediated immune responses, but failed to induce neutralizing antibodies and did not protect mice against virus challenge. CONCLUSION Although highly immunogenic, our vaccine was unable to induce a protective immunity against influenza. A misfolded and unstable conformation of the hlHA molecule may have affected its capacity of inducing neutralizing antiviral, conformational antibodies. Design of stable hlHA-based immunogens and their delivery by recombinant MVA-based vectors has the potential of improving this promising approach for a universal influenza vaccine.
Collapse
Affiliation(s)
- Giuseppina Di Mario
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| | - Elisa Soprana
- b Molecular Immunology Unit , San Raffaele Research Institute , Milan , Italy
| | - Francesco Gubinelli
- b Molecular Immunology Unit , San Raffaele Research Institute , Milan , Italy
| | - Maddalena Panigada
- b Molecular Immunology Unit , San Raffaele Research Institute , Milan , Italy
| | - Marzia Facchini
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| | - Concetta Fabiani
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| | - Bruno Garulli
- c Department of Biology and Biotechnology "Charles Darwin" , Sapienza University of Rome , Rome , Italy
| | - Michela Basileo
- d Polo d'Innovazione della Genomica, Genetica e Biologia , University of Perugia , Perugia , Italy
| | - Antonio Cassone
- d Polo d'Innovazione della Genomica, Genetica e Biologia , University of Perugia , Perugia , Italy
| | - Antonio Siccardi
- b Molecular Immunology Unit , San Raffaele Research Institute , Milan , Italy
| | - Isabella Donatelli
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| | - Maria R Castrucci
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| |
Collapse
|
17
|
Di Mario G, Sciaraffia E, Facchini M, Gubinelli F, Soprana E, Panigada M, Bernasconi V, Garulli B, Siccardi A, Donatelli I, Castrucci MR. Protective immunity against influenza in HLA-A2 transgenic mice by modified vaccinia virus Ankara vectored vaccines containing internal influenza proteins. Pathog Glob Health 2017; 111:76-82. [PMID: 28079473 PMCID: PMC5375616 DOI: 10.1080/20477724.2016.1275465] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The emergence of novel strains of influenza A viruses with hemagglutinins (HAs) that are antigenically distinct from those circulating in humans, and thus have pandemic potential, pose concerns and call for the development of more broadly protective influenza vaccines. In the present study, modified vaccinia virus Ankara (MVA) encoding internal influenza antigens were evaluated for their immunogenicity and ability to protect HLA-A2.1 transgenic (AAD) mice from infection with influenza viruses. METHODS MVAs expressing NP (MVA-NP), M1 (MVA-M1) or polymerase PB1 (MVA-PB1) of A/California/4/09 (CA/09) virus were generated and used to immunize AAD mice. Antibodies and CD8+T cell responses were assessed by ELISA and ELISPOT, respectively, and challenge experiments were performed by infecting vaccinated mice with CA/09 virus. RESULTS CD8+T cells specific to immunodominant and subdominant epitopes on the internal influenza proteins were elicited by MVA-based vectors in AAD mice, whereas influenza-specific antibodies were detected only in MVA-NP-immunized mice. Both M1- and NP-based MVA vaccines, regardless of whether they were applied individually or in combination, conferred protection against lethal influenza virus challenge. CONCLUSION Our data further emphasize the promising potential of MVA vector expressing internal antigens toward the development of a universal influenza vaccine.
Collapse
Affiliation(s)
- Giuseppina Di Mario
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| | - Ester Sciaraffia
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| | - Marzia Facchini
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| | - Francesco Gubinelli
- b Molecular Immunology Unit , San Raffaele Research Institute , Milan , Italy
| | - Elisa Soprana
- b Molecular Immunology Unit , San Raffaele Research Institute , Milan , Italy
| | - Maddalena Panigada
- b Molecular Immunology Unit , San Raffaele Research Institute , Milan , Italy
| | | | - Bruno Garulli
- c Department of Biology and Biotechnology "Charles Darwin" , Sapienza University of Rome , Rome , Italy
| | - Antonio Siccardi
- b Molecular Immunology Unit , San Raffaele Research Institute , Milan , Italy
| | - Isabella Donatelli
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| | - Maria R Castrucci
- a Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| |
Collapse
|
18
|
Hoffmann PR, Panigada M, Soprana E, Terry F, Bandar IS, Napolitano A, Rose AH, Hoffmann FW, Ndhlovu LC, Belcaid M, Moise L, De Groot AS, Carbone M, Gaudino G, Matsui T, Siccardi A, Bertino P. Preclinical development of HIvax: Human survivin highly immunogenic vaccines. Hum Vaccin Immunother 2016; 11:1585-95. [PMID: 26042612 DOI: 10.1080/21645515.2015.1050572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Our previous work involved the development of a recombinant fowlpox virus encoding survivin (FP-surv) vaccine that was evaluated for efficacy in mesothelioma mouse models. Results showed that FP-surv vaccination generated significant immune responses, which led to delayed tumor growth and improved animal survival. We have extended those previous findings in the current study, which involves the pre-clinical development of an optimized version of FP-surv designed for human immunization (HIvax). Survivin-derived peptides for the most common haplotypes in the human population were identified and their immunogenicity confirmed in co-culture experiments using dendritic cells and T cells isolated from healthy donors. Peptides confirmed to induce CD8(+) and CD4(+) T cells activation in humans were then included in 2 transgenes optimized for presentation of processed peptides on MHC-I (HIvax1) and MHC-II (HIvax2). Fowlpox vectors expressing the HIvax transgenes were then generated and their efficacy was evaluated with subsequent co-culture experiments to measure interferon-γ and granzyme B secretion. In these experiments, both antigen specific CD4(+) and CD8(+) T cells were activated by HIvax vaccines with resultant cytotoxic activity against survivin-overexpressing mesothelioma cancer cells. These results provide a rationale for clinical testing of HIvax1 and HIvax2 vaccines in patients with survivin-expressing cancers.
Collapse
Affiliation(s)
- Peter R Hoffmann
- a Department of Cell and Molecular Biology; John A. Burns School of Medicine ; University of Hawai'i ; Honolulu , HI , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Pierantoni A, Esposito ML, Ammendola V, Napolitano F, Grazioli F, Abbate A, del Sorbo M, Siani L, D’Alise AM, Taglioni A, Perretta G, Siccardi A, Soprana E, Panigada M, Thom M, Scarselli E, Folgori A, Colloca S, Taylor G, Cortese R, Nicosia A, Capone S, Vitelli A. Mucosal delivery of a vectored RSV vaccine is safe and elicits protective immunity in rodents and nonhuman primates. Mol Ther Methods Clin Dev 2015; 2:15018. [PMID: 26015988 PMCID: PMC4441047 DOI: 10.1038/mtm.2015.18] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 03/20/2015] [Indexed: 01/27/2023]
Abstract
Respiratory Syncytial Virus (RSV) is a leading cause of severe respiratory disease in infants and the elderly. No vaccine is presently available to address this major unmet medical need. We generated a new genetic vaccine based on chimpanzee Adenovirus (PanAd3-RSV) and Modified Vaccinia Ankara RSV (MVA-RSV) encoding the F, N, and M2-1 proteins of RSV, for the induction of neutralizing antibodies and broad cellular immunity. Because RSV infection is restricted to the respiratory tract, we compared intranasal (IN) and intramuscular (M) administration for safety, immunogenicity, and efficacy in different species. A single IN or IM vaccination completely protected BALB/c mice and cotton rats against RSV replication in the lungs. However, only IN administration could prevent infection in the upper respiratory tract. IM vaccination with MVA-RSV also protected cotton rats from lower respiratory tract infection in the absence of detectable neutralizing antibodies. Heterologous prime boost with PanAd3-RSV and MVA-RSV elicited high neutralizing antibody titers and broad T-cell responses in nonhuman primates. In addition, animals primed in the nose developed mucosal IgA against the F protein. In conclusion, we have shown that our vectored RSV vaccine induces potent cellular and humoral responses in a primate model, providing strong support for clinical testing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Alessandra Taglioni
- Cellular Biology and Neurobiology Institute (IBCN) National Research Council of Italy, Rome, Italy
| | - Gemma Perretta
- Cellular Biology and Neurobiology Institute (IBCN) National Research Council of Italy, Rome, Italy
| | | | | | | | | | | | | | | | | | - Riccardo Cortese
- ReiThera Srl, Rome, Italy (former Okairos Srl)
- Keires AG, Basel, Switzerland
| | - Alfredo Nicosia
- ReiThera Srl, Rome, Italy (former Okairos Srl)
- CEINGE, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | | | | |
Collapse
|
20
|
Stanley DA, Honko AN, Asiedu C, Trefry JC, Lau-Kilby AW, Johnson JC, Hensley L, Ammendola V, Abbate A, Grazioli F, Foulds KE, Cheng C, Wang L, Donaldson MM, Colloca S, Folgori A, Roederer M, Nabel GJ, Mascola J, Nicosia A, Cortese R, Koup RA, Sullivan NJ. Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge. Nat Med 2014; 20:1126-9. [DOI: 10.1038/nm.3702] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/02/2014] [Indexed: 12/13/2022]
|
21
|
Capone S, Naddeo M, D'Alise AM, Abbate A, Grazioli F, Del Gaudio A, Del Sorbo M, Esposito ML, Ammendola V, Perretta G, Taglioni A, Colloca S, Nicosia A, Cortese R, Folgori A. Fusion of HCV nonstructural antigen to MHC class II-associated invariant chain enhances T-cell responses induced by vectored vaccines in nonhuman primates. Mol Ther 2014; 22:1039-47. [PMID: 24476798 DOI: 10.1038/mt.2014.15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 01/23/2014] [Indexed: 02/06/2023] Open
Abstract
Despite viral vectors being potent inducers of antigen-specific T cells, strategies to further improve their immunogenicity are actively pursued. Of the numerous approaches investigated, fusion of the encoded antigen to major histocompatibility complex class II-associated invariant chain (Ii) has been reported to enhance CD8(+) T-cell responses. We have previously shown that adenovirus vaccine encoding nonstructural (NS) hepatitis C virus (HCV) proteins induces potent T-cell responses in humans. However, even higher T-cell responses might be required to achieve efficacy against different HCV genotypes or therapeutic effect in chronically infected HCV patients. In this study, we assessed fusion of the HCV NS antigen to murine and human Ii expressed by the chimpanzee adenovirus vector ChAd3 or recombinant modified vaccinia Ankara in mice and nonhuman primates (NHPs). A dramatic increase was observed in outbred mice in which vaccination with ChAd3 expressing the fusion antigen resulted in a 10-fold increase in interferon-γ(+) CD8(+) T cells. In NHPs, CD8(+) T-cell responses were enhanced and accelerated with vectors encoding the Ii-fused antigen. These data show for the first time that the enhancement induced by vector vaccines encoding li-fused antigen was not species specific and can be translated from mice to NHPs, opening the way for testing in humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Gemma Perretta
- Cellular Biology and Neurobiology Institute (IBCN) National Research Council of Italy, Rome, Italy
| | - Alessandra Taglioni
- Cellular Biology and Neurobiology Institute (IBCN) National Research Council of Italy, Rome, Italy
| | | | - Alfredo Nicosia
- 1] Okairos, Rome, Italy [2] CEINGE, Naples, Italy [3] Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Riccardo Cortese
- 1] Okairos, Rome, Italy [2] Okairos AG, c/o OBC Suisse AG, Basel, Switzerland
| | | |
Collapse
|
22
|
Castrucci MR, Facchini M, Di Mario G, Garulli B, Sciaraffia E, Meola M, Fabiani C, De Marco MA, Cordioli P, Siccardi A, Kawaoka Y, Donatelli I. Modified vaccinia virus Ankara expressing the hemagglutinin of pandemic (H1N1) 2009 virus induces cross-protective immunity against Eurasian 'avian-like' H1N1 swine viruses in mice. Influenza Other Respir Viruses 2013; 8:367-75. [PMID: 24373385 PMCID: PMC4181486 DOI: 10.1111/irv.12221] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2013] [Indexed: 11/27/2022] Open
Abstract
Objectives To examine cross-reactivity between hemagglutinin (HA) derived from A/California/7/09 (CA/09) virus and that derived from representative Eurasian “avian-like” (EA) H1N1 swine viruses isolated in Italy between 1999 and 2008 during virological surveillance in pigs. Design Modified vaccinia virus Ankara (MVA) expressing the HA gene of CA/09 virus (MVA-HA-CA/09) was used as a vaccine to investigate cross-protective immunity against H1N1 swine viruses in mice. Sample Two classical swine H1N1 (CS) viruses and four representative EA-like H1N1 swine viruses previously isolated during outbreaks of respiratory disease in pigs on farms in Northern Italy were used in this study. Setting Female C57BL/6 mice were vaccinated with MVA/HA/CA/09 and then challenged intranasally with H1N1 swine viruses. Main outcome measures Cross-reactive antibody responses were determined by hemagglutination- inhibition (HI) and virus microneutralizing (MN) assays of sera from MVA-vaccinated mice. The extent of protective immunity against infection with H1N1 swine viruses was determined by measuring lung viral load on days 2 and 4 post-challenge. Results and Conclusions Systemic immunization of mice with CA/09-derived HA, vectored by MVA, elicited cross-protective immunity against recent EA-like swine viruses. This immune protection was related to the levels of cross-reactive HI antibodies in the sera of the immunized mice and was dependent on the similarity of the antigenic site Sa of H1 HAs. Our findings suggest that the herd immunity elicited in humans by the pandemic (H1N1) 2009 virus could limit the transmission of recent EA-like swine HA genes into the influenza A virus gene pool in humans.
Collapse
Affiliation(s)
- Maria R Castrucci
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Nigro E, Siccardi A, Vangelista L. Role and Redirection of IgE against Cancer. Antibodies (Basel) 2013; 2:371-391. [DOI: 10.3390/antib2020371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
IgE is a highly elusive antibody class, yet a tremendously powerful elicitor of immune reactions. Despite huge efforts spent on the characterization and understanding of the IgE system many questions remain either unanswered or only marginally addressed. One above all relates to the role of IgE. A common doubt is based on whether IgE mode of action should only be relegated to anti-parasite immunity and allergic manifestations. In search for a hidden role of IgE, reports from several laboratories are described herein in which a natural IgE link to cancer or the experimental redirection of IgE against cancer have been investigated. Epidemiological and investigational studies are trying to elucidate a possible direct intervention of endogenous IgE against cancer, raising thus far no definitive evidence. Conversely, experimental approaches implementing several strategies and engineered IgE formats built up a series of convincing results indicating that cancer might be tackled by the effector functions of this immunoglobulin class. Because of its peculiar immune features, IgE may present a superior anti-tumor performance as compared to IgG. However, extreme care should be taken on how IgE-based anti-tumor approaches should be devised. Overall, IgE appears as a promising resource, likely destined to enrich the anti-cancer arsenal.
Collapse
Affiliation(s)
- Elisa Nigro
- Molecular Immunology Group, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, via Olgettina 58, 20132 Milan, Italy
| | - Antonio Siccardi
- Molecular Immunology Group, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, via Olgettina 58, 20132 Milan, Italy
| | - Luca Vangelista
- Protein Engineering and Therapeutics Group, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, via Olgettina 58, 20132 Milan, Italy
| |
Collapse
|
24
|
Bertino P, Panigada M, Soprana E, Bianchi V, Bertilaccio S, Sanvito F, Rose AH, Yang H, Gaudino G, Hoffmann PR, Siccardi A, Carbone M. Fowlpox-based survivin vaccination for malignant mesothelioma therapy. Int J Cancer 2013; 133:612-23. [PMID: 23335100 DOI: 10.1002/ijc.28048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 01/08/2013] [Indexed: 12/12/2022]
Abstract
Survivin protein is an attractive candidate for cancer immunotherapy since it is abundantly expressed in most common human cancers and mostly absent in normal adult tissues. Malignant mesothelioma (MM) is a deadly cancer associated with asbestos or erionite exposure for which no successful therapies are currently available. In this study, we evaluated the therapeutic efficacy of a novel survivin-based vaccine by subcutaneous or intraperitoneum injection of BALB/c mice with murine fiber-induced MM tumor cells followed by vaccination with recombinant Fowlpox virus replicons encoding survivin. Vaccination generated significant immune responses in both models, leading to delayed tumor growth and improved animal survival. Flow cytometry and immunofluorescence analyses of tumors from vaccinated mice showed CD8(+) T-cell infiltration, and real-time PCR demonstrated increased mRNA and protein levels of immunostimulatory cytokines. Analyses of survivin peptide-pulsed spleen and lymph node cells from vaccinated mice using ELISPOT and intracellular cytokine staining confirmed antigen-specific, interferon-γ-producing CD8(+) T-cell responses. In addition pentamer-based flow cytometry showed that vaccination generated survivin-specific CD8(+) T cells. Importantly, vaccination did not affect fertility or induce autoimmune abnormalities in mice. Our results demonstrate that vaccination with recombinant Fowlpox expressing survivin improves T-cell responses against aggressive MM tumors and may form the basis for promising clinical applications.
Collapse
Affiliation(s)
- Pietro Bertino
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI 96813, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Modified vaccinia virus Ankara (MVA) has become a widely used vector for vaccine and laboratory purposes. Despite significant advances in recombinant MVA technology, the isolation of recombinant viruses remains a tedious and difficult process. This chapter describes the use of an efficient and easy-to-use selection system adapted for MVA. The system is based on the requirement of the viral gene F13L for efficient virus spread in cell culture, which results in a severe block in virus transmission when F13L gene is deleted (Blasco R, Moss B. J Virol 65:5910-5920, 1991; Blasco R, Moss B. J Virol 66:4170-4179, 1992). The insertion of foreign genes in the MVA genome is accomplished by recombination of a transfected plasmid carrying the foreign genes and the F13L with the genome of an F13L knockout virus. Subsequently, selection of virus recombinants is carried out by serial passage and/or plaque purification of viruses that have recovered the F13L gene.
Collapse
|
26
|
Nigro EA, Soprana E, Brini AT, Ambrosi A, Yenagi VA, Dombrowicz D, Siccardi AG, Vangelista L. An Antitumor Cellular Vaccine Based on a Mini-Membrane IgE. THE JOURNAL OF IMMUNOLOGY 2011; 188:103-10. [DOI: 10.4049/jimmunol.1101842] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
27
|
Soprana E, Panigada M, Knauf M, Radaelli A, Vigevani L, Palini A, Villa C, Malnati M, Cassina G, Kurth R, Norley S, Siccardi AG. Joint production of prime/boost pairs of Fowlpox Virus and Modified Vaccinia Ankara recombinants carrying the same transgene. J Virol Methods 2011; 174:22-8. [PMID: 21419167 DOI: 10.1016/j.jviromet.2011.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 03/03/2011] [Accepted: 03/09/2011] [Indexed: 10/18/2022]
Abstract
Pairs of recombinant MVA (Modified Vaccinia Ankara) and FPV (Fowlpox Virus) expressing the same transgene are reasonable candidates for prime/boost regimens, because cross-reacting immune responses between the two vectors, both non-replicative in mammalian hosts, are very limited. The acceptor virus FPD-Red, a derivative of FPV, carrying a red fluorescent protein gene flanked by the homology regions of MVA deletion III, was constructed. The same MVA Transfer Plasmid Green, designed to insert transgenes into the MVA deletion III locus, can therefore be used to transfer transgenes into both acceptor viruses MVA-Red and FPD-Red with the described recently Red-to-Green gene swapping method. Cells infected by either recombinant virus can be sorted differentially by a simple and reliable FACS-based purification protocol. The procedure is carried out in primary chick embryo fibroblasts grown in serum-free media and was applied to the production of three rMVA/rFPV pairs expressing the H5N1 avian influenza antigens M1, M2 and NP. The viral genes were human codon-optimized and expressed at high levels in both chick and mammalian cells. Both single-step and multiple-step growth analyses showed no significant differences in growth due to the transgenes in either rMVA or rFPV derivatives.
Collapse
|
28
|
Cottingham MG, Gilbert SC. Rapid generation of markerless recombinant MVA vaccines by en passant recombineering of a self-excising bacterial artificial chromosome. J Virol Methods 2010; 168:233-6. [DOI: 10.1016/j.jviromet.2010.04.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 03/26/2010] [Accepted: 04/19/2010] [Indexed: 10/19/2022]
|