1
|
Calvo-Pinilla E, Moreno S, Barreiro-Piñeiro N, Sánchez-Puig JM, Blasco R, Martínez-Costas J, Brun A, Lorenzo G. Prime-Boost Vaccination Based on Nanospheres and MVA Encoding the Nucleoprotein of Crimean-Congo Hemorrhagic Fever Virus Elicits Broad Immune Responses. Vaccines (Basel) 2025; 13:291. [PMID: 40266214 PMCID: PMC11946443 DOI: 10.3390/vaccines13030291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: Crimean-Congo hemorrhagic fever virus (CCHFV) is an emerging, widely distributed zoonotic tick-borne pathogen. The virus causes severe disease in humans, and numerous wild and domestic animals act as reservoirs of it. Unfortunately, there are no effective therapies or safe vaccines commercialized nowadays for this particular virus. As CCHF (Crimean-Congo hemorrhagic fever) is a serious threat to public health, there is an urgent need to investigate the development of safe and effective vaccination strategies further. Methods: In this work, we have employed two immunization platforms based on protein nanoparticles and a modified vaccinia Ankara (MVA) viral vector using the nucleoprotein (NP) as the target antigen. The humoral and cellular immune responses were characterized by ELISA, ICS, and cytokine measurement. Results: This work shows that a single dose of the vaccine candidates was not as immunogenic as the heterologous vaccination using nanoparticles and MVA. A prime with NP nanoparticles (NS-NP) and a boost with MVA-expressing NP were capable of triggering significant levels of humoral and cellular immune responses against CCHFV in mice. Conclusions: Our study shows that the NS-NP/MVA-NP vaccination strategy effectively elicits a robust humoral and cellular immune response in a mouse model, emphasizing its potential as a protective approach against CCHFV lineages.
Collapse
Affiliation(s)
- Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal, INIA, CSIC, Valdeolmos, 28130 Madrid, Spain; (S.M.); (A.B.)
| | - Sandra Moreno
- Centro de Investigación en Sanidad Animal, INIA, CSIC, Valdeolmos, 28130 Madrid, Spain; (S.M.); (A.B.)
| | - Natalia Barreiro-Piñeiro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Bioquímica e Bioloxía Molecular, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain; (N.B.-P.); (J.M.-C.)
| | - Juana M. Sánchez-Puig
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. de la Coruña km 7.5, 28040 Madrid, Spain; (J.M.S.-P.); (R.B.)
| | - Rafael Blasco
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Ctra. de la Coruña km 7.5, 28040 Madrid, Spain; (J.M.S.-P.); (R.B.)
| | - José Martínez-Costas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Bioquímica e Bioloxía Molecular, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain; (N.B.-P.); (J.M.-C.)
| | - Alejandro Brun
- Centro de Investigación en Sanidad Animal, INIA, CSIC, Valdeolmos, 28130 Madrid, Spain; (S.M.); (A.B.)
| | - Gema Lorenzo
- Centro de Investigación en Sanidad Animal, INIA, CSIC, Valdeolmos, 28130 Madrid, Spain; (S.M.); (A.B.)
| |
Collapse
|
2
|
Sun L, Liu C, Peng J. Specific Immune Responses and Oncolytic Effects Induced by EBV LMP2A-Armed Modified Ankara-Vaccinia Virus Vectored Vaccines in Nasopharyngeal Cancer. Pharmaceutics 2025; 17:52. [PMID: 39861700 PMCID: PMC11768126 DOI: 10.3390/pharmaceutics17010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The Epstein-Barr virus (EBV) is intricately linked to a range of human malignancies, with EBV latent membrane protein 2A (LMP2A) emerging as a potential target antigen for immunotherapeutic strategies in the treatment of nasopharyngeal carcinoma (NPC). METHODS The modified vaccinia virus Ankara (MVA) is universally used in vector vaccine research because of its excellent safety profile and highly efficient recombinant gene expression. Here, we constructed a novel MVA-LMP2A recombinant virus and investigated its specific immune response induction and oncolytic effect. RESULTS An immunization dose of 2 × 107 PFU induced the highest specific immune response, which was no longer increased by boost injections after four doses. Three weeks post-final immunization, the specific immune response reached its peak. The MVA-LMP2A vaccine-induced LMP2A-specific cytotoxic T lymphocytes (CTLs), which exhibited substantial efficacy against target cells and effectively inhibited tumor growth. CONCLUSIONS Thus, the MVA-LMP2A recombinant virus effectively induces strong LMP2A-specific cellular and humoral immune responses and anti-tumor activity. This work provides a promising therapeutic strategy for developing NPC candidate vaccines, as well as a reference for the treatment of EBV LMP2-associated malignancies.
Collapse
Affiliation(s)
- Liying Sun
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China;
| | - Chao Liu
- State Key Laboratory of Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Junping Peng
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China;
| |
Collapse
|
3
|
Link EK, Tscherne A, Sutter G, Smith ER, Gurwith M, Chen RT, Volz A. A Brighton collaboration standardized template with key considerations for a benefit/risk assessment for a viral vector vaccine based on a non-replicating modified vaccinia virus Ankara viral vector. Vaccine 2025; 43:126521. [PMID: 39612556 DOI: 10.1016/j.vaccine.2024.126521] [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: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
The Brighton Collaboration Benefit-Risk Assessment of VAccines by TechnolOgy (BRAVATO) was formed to evaluate the safety and other key features of new platform technology vaccines. This manuscript provides an overview of Modified Vaccinia virus Ankara (MVA)-vectored vaccines and reviews molecular and biological key features of this platform. In particular, this review aims to provide fundamental information about the promising candidate vaccine MVA-MERS-S which has been evaluated successfully in different preclinical animal models and has undergone clinical testing including a phase Ib study involving more than 170 participants. Infectious diseases continue to be a major cause of human death worldwide. In this context, emerging zoonotic infectious diseases pose a particular challenge for public health systems. In the last two decades, three different respiratory coronaviruses, including the Middle East respiratory syndrome Coronavirus (MERS-CoV) have emerged. For many years, safe and efficacious vaccines have been a major tool to combat infectious diseases. Here, we report on a promising candidate vaccine (MVA-MERS-S) against MERS-CoV based on MVA. Upon application, MVA-MERS-S has been well tolerated and immunogenic, inducing both, cellular and humoral immune responses in different animal models and humans. We demonstrate that the MVA vector platform, with the example of MVA-MERS-S, is a viable and effective tool for producing safe, immunogenic, and efficient vaccines against emerging infectious diseases.
Collapse
Affiliation(s)
- Ellen K Link
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Alina Tscherne
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Gerd Sutter
- Ludwig-Maximilians-University Munich (LMU Munich), Department of Veterinary Sciences, Division of Virology, Oberschleißheim, Germany
| | - Emily R Smith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA.
| | - Marc Gurwith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Robert T Chen
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Asisa Volz
- University of Veterinary Medicine Hannover, Institute of Virology, Hannover, Germany
| |
Collapse
|
4
|
Jiménez-Cabello L, Utrilla-Trigo S, Rodríguez-Sabando K, Carra-Valenzuela A, Illescas-Amo M, Calvo-Pinilla E, Ortego J. Vaccine candidates based on MVA viral vectors expressing VP2 or VP7 confer full protection against Epizootic hemorrhagic disease virus in IFNAR(-/-) mice. J Virol 2024; 98:e0168724. [PMID: 39508577 DOI: 10.1128/jvi.01687-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 11/15/2024] Open
Abstract
Epizootic hemorrhagic disease (EHD), caused by Epizootic hemorrhagic disease virus (EHDV), is an emerging and severe livestock disease. Recent incursion and distribution of EHDV in Europe have outlined the need for vaccine research against this viral disease. In this work, we report modified vaccinia virus Ankara (MVA)-vectored vaccines designed to express protein VP2 of EHDV-8 or protein VP7 of EHDV-2. Prime boost immunization of adult IFNAR(-/-) mice with the MVA-VP2 vaccine candidate induced high titers of EHDV-8-specific neutralizing antibodies (NAbs) and conferred full protection against homologous lethal challenge with EHDV-8. However, no heterologous protection was observed after lethal challenge with EHDV-6. In contrast, the MVA-VP7 vaccine candidate elicited strong cytotoxic CD8+ T-cell responses against VP7 and conferred complete protection against lethal challenge with either EHDV-8 or EHDV-6 in IFNAR(-/-) mice in the absence of NAbs, being the first multiserotype vaccine candidate against EHDV. Moreover, we expressed recombinant proteins VP2 and VP7 of EHDV in the baculovirus expression system, which were used to analyze the potential DIVA (differentiating infected from vaccinated animals) character of these vaccine candidates.IMPORTANCEEmergence and re-emergence of arthropod-borne viruses are major concerns for both human and animal health. The most recent example is the fast expansion of EHDV-8 through Europe. Besides, EHDV-8 relates with a high prevalence of pathologic cases in cattle populations. No vaccine is currently available in Europe, and vaccine research against this arboviral disease is negligible. In this work, we present novel DIVA vaccine candidates against EHDV, and most importantly, we identified the protein VP7 of EHDV as an antigen capable of inducing multiserotype protection, one of the major challenges in vaccine research against orbiviruses.
Collapse
Affiliation(s)
- Luis Jiménez-Cabello
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Sergio Utrilla-Trigo
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Karen Rodríguez-Sabando
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Alejandro Carra-Valenzuela
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Miguel Illescas-Amo
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| |
Collapse
|
5
|
Metz C, Haug V, Müller M, Amann R. Pharmacokinetic and Environmental Risk Assessment of Prime-2-CoV, a Non-Replicating Orf Virus-Based Vaccine against SARS-CoV-2. Vaccines (Basel) 2024; 12:492. [PMID: 38793743 PMCID: PMC11126055 DOI: 10.3390/vaccines12050492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Viral vector vaccines represent a substantial advancement in immunization technology, offering numerous benefits over traditional vaccine modalities. The Orf virus (ORFV) strain D1701-VrV is a particularly promising candidate for vaccine development due to its distinctive attributes, such as a good safety profile, the ability to elicit both humoral and cellular immunity, and its favorable genetic and thermal stability. Despite ORFV's theoretical safety advantages, such as its narrow host range and limited systemic spread post-inoculation, a critical gap persists between these theoretical benefits and the empirical evidence regarding its in vivo safety profile. This discrepancy underscores the need for comprehensive preclinical validations to bridge this knowledge gap, especially considering ORFV's use in humans. Our research introduces Prime-2-CoV, an innovative ORFV-based vaccine candidate against COVID-19, designed to elicit a robust immune response by expressing SARS-CoV-2 Nucleocapsid and Spike proteins. Currently under clinical trials, Prime-2-CoV marks the inaugural application of ORFV in human subjects. Addressing the aforementioned safety concerns, our extensive preclinical evaluation, including an environmental risk assessment (ERA) and detailed pharmacokinetic studies in rats and immunocompromised NOG mice, demonstrates Prime-2-CoV's favorable pharmacokinetic profile, negligible environmental impact, and minimal ERA risks. These findings not only affirm the vaccine's safety and efficacy but also pioneer the use of ORFV-based therapeutics, highlighting its potential for wider therapeutic applications.
Collapse
Affiliation(s)
- Carina Metz
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
- Institute for Tropical Medicine, Travel Medicine, and Human Parasitology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Verena Haug
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
- Institute for Tropical Medicine, Travel Medicine, and Human Parasitology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Melanie Müller
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
| | - Ralf Amann
- Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany; (C.M.); (V.H.); (M.M.)
| |
Collapse
|
6
|
Hernandez JM. Biosafety Considerations for Viral Vector Gene Therapy: An Explanation and Guide for the Average Everyday-Hero Pharmacist. J Pharm Pract 2023; 36:1532-1539. [PMID: 35583499 DOI: 10.1177/08971900221104250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Purpose: An overview of the multi-faceted biosafety points that must be taken into consideration by pharmacists and pharmacies in order to provide viral vector gene therapy to their practice site. Summary: As science and medicine evolves, pharmacists and other healthcare workers are continually faced with unique challenges in the workplace. They are expected to be informed and proficient on new therapies and standards of practice, and be able to apply this knowledge appropriately for their patients. One such advancement that seems to be picking up speed in recent years is gene therapy, which is often achieved with the assistance of a viral vector. As these viral vector doses move closer to mainstream medicine, a host of issues and concerns for the pharmacists, nurses, and caregivers that are involved in the process begin to rise to the surface, often rooted in the critical concern: "How do we dispense, utilize, and administer these doses safely?" Unfortunately, there is no singular, concise source of information for addressing biosafety with viral vector products, and guidance must be gathered from a variety of resources in order to mesh together a reasonable working process. Conclusion: While this may seem to be a daunting task, facilities that already meet USP 797 and USP 800 guidelines are well on their way to being ready to provide viral vector doses. By incorporating additional steps and reviewing biosafety specific resources, these sites can easily adapt to provide these new and novel therapies for their patient population.
Collapse
|
7
|
Froechlich G, Finizio A, Napolano A, Amiranda S, De Chiara A, Pagano P, Mallardo M, Leoni G, Zambrano N, Sasso E. The common H232 STING allele shows impaired activities in DNA sensing, susceptibility to viral infection, and in monocyte cell function, while the HAQ variant possesses wild-type properties. Sci Rep 2023; 13:19541. [PMID: 37945588 PMCID: PMC10636114 DOI: 10.1038/s41598-023-46830-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
Different innate immune pathways converge to Stimulator of interferon genes (STING) and trigger type I interferon responses after recognition of abnormal nucleic acids in the cells. This non-redundant function renders STING a major player in immunosurveillance, and an emerging target for cancer and infectious diseases therapeutics. Beyond somatic mutations that often occur in cancer, the human gene encoding STING protein, TMEM173 (STING1), holds great genetic heterogeneity; R232, HAQ (R71H-G230A-R293Q) and H232 are the most common alleles. Although some of these alleles are likely to be hypomorphic, their function is still debated, due to the available functional assessments, which have been performed in biased biological systems. Here, by using genetic background-matched models, we report on the functional evaluation of R232, HAQ and H232 variants on STING function, and on how these genotypes affect the susceptibility to clinically relevant viruses, thus supporting a potential contributing cause to differences in inter-individual responses to infections. Our findings also demonstrate a novel toll-like receptor-independent role of STING in modulating monocytic cell function and differentiation into macrophages. We further supported the interplay of STING1 variants and human biology by demonstrating how monocytes bearing the H232 allele were impaired in M1/M2 differentiation, interferon response and antigen presentation. Finally, we assessed the response to PD-1 inhibitor in a small cohort of melanoma patients stratified according to STING genotype. Given the contribution of the STING protein in sensing DNA viruses, bacterial pathogens and misplaced cancer DNA, these data may support the development of novel therapeutic options for infectious diseases and cancer.
Collapse
Affiliation(s)
- Guendalina Froechlich
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Arianna Finizio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Alessandra Napolano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Sara Amiranda
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Arianna De Chiara
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Pasqualina Pagano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Massimo Mallardo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy
| | | | - Nicola Zambrano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy.
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Via Gaetano Salvatore 486, 80145, Naples, Italy.
| | - Emanuele Sasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini 5, 80131, Napoli, NA, Italy.
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Via Gaetano Salvatore 486, 80145, Naples, Italy.
- ImGen-T Srl, Viale del Parco Carelli, Napoli, NA, Italy.
| |
Collapse
|
8
|
Nave L, Margalit I, Tau N, Cohen I, Yelin D, Lienert F, Yahav D. Immunogenicity and Safety of Modified Vaccinia Ankara (MVA) Vaccine-A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Vaccines (Basel) 2023; 11:1410. [PMID: 37766090 PMCID: PMC10536351 DOI: 10.3390/vaccines11091410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Prevention of mpox has become an important public health interest. We aimed to evaluate the safety and immunogenicity of the Modified Vaccinia Ankara (MVA) vaccine. We conducted a systematic review and meta-analysis of randomized-controlled trials (RCTs) comparing MVA versus no intervention, placebo, or another vaccine. Outcomes included safety and immunogenicity outcomes. We also performed a systematic review of RCTs evaluating various MVA regimens. Fifteen publications were included in the quantitative meta-analysis. All but one (ACAM2000) compared MVA with placebo. We found that cardiovascular adverse events following two MVA doses were significantly more common compared to placebo (relative risk [RR] 4.07, 95% confidence interval [CI] 1.10-15.10), though serious adverse events (SAEs) were not significantly different. Following a single MVA dose, no difference was demonstrated in any adverse event outcomes. Seroconversion rates were significantly higher compared with placebo after a single or two doses. None of the RCTs evaluated clinical effectiveness in preventing mpox. This meta-analysis provides reassuring results concerning the immunogenicity and safety of MVA. Further studies are needed to confirm the immunogenicity of a single dose and its clinical effectiveness. A single vaccine dose may be considered according to vaccine availability, with preference for two doses.
Collapse
Affiliation(s)
- Lior Nave
- Internal Medicine E, Sheba Medical Center, Ramat-Gan 52621, Israel; (L.N.)
| | - Ili Margalit
- Faculty of Medicine, Tel Aviv University, Ramat-Aviv, Tel Aviv 69978, Israel; (I.M.)
- Infectious Diseases Unit, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Noam Tau
- Faculty of Medicine, Tel Aviv University, Ramat-Aviv, Tel Aviv 69978, Israel; (I.M.)
- Department of Diagnostic Imaging, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Ido Cohen
- Internal Medicine E, Sheba Medical Center, Ramat-Gan 52621, Israel; (L.N.)
| | - Dana Yelin
- Faculty of Medicine, Tel Aviv University, Ramat-Aviv, Tel Aviv 69978, Israel; (I.M.)
- Infectious Diseases Unit, Sheba Medical Center, Ramat-Gan 52621, Israel
| | | | - Dafna Yahav
- Faculty of Medicine, Tel Aviv University, Ramat-Aviv, Tel Aviv 69978, Israel; (I.M.)
- Infectious Diseases Unit, Sheba Medical Center, Ramat-Gan 52621, Israel
| |
Collapse
|
9
|
Saunders JE, Gilbride C, Dowall S, Morris S, Ulaszewska M, Spencer AJ, Rayner E, Graham VA, Kennedy E, Thomas K, Hewson R, Gilbert SC, Belij-Rammerstorfer S, Lambe T. Adenoviral vectored vaccination protects against Crimean-Congo Haemorrhagic Fever disease in a lethal challenge model. EBioMedicine 2023; 90:104523. [PMID: 36933409 PMCID: PMC10025009 DOI: 10.1016/j.ebiom.2023.104523] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND The tick-borne bunyavirus, Crimean-Congo Haemorrhagic Fever virus (CCHFV), can cause severe febrile illness in humans and has a wide geographic range that continues to expand due to tick migration. Currently, there are no licensed vaccines against CCHFV for widespread usage. METHODS In this study, we describe the preclinical assessment of a chimpanzee adenoviral vectored vaccine (ChAdOx2 CCHF) which encodes the glycoprotein precursor (GPC) from CCHFV. FINDINGS We demonstrate here that vaccination with ChAdOx2 CCHF induces both a humoral and cellular immune response in mice and 100% protection in a lethal CCHF challenge model. Delivery of the adenoviral vaccine in a heterologous vaccine regimen with a Modified Vaccinia Ankara vaccine (MVA CCHF) induces the highest levels of CCHFV-specific cell-mediated and antibody responses in mice. Histopathological examination and viral load analysis of the tissues of ChAdOx2 CCHF immunised mice reveals an absence of both microscopic changes and viral antigen associated with CCHF infection, further demonstrating protection against disease. INTERPRETATION There is the continued need for an effective vaccine against CCHFV to protect humans from lethal haemorrhagic disease. Our findings support further development of the ChAd platform expressing the CCHFV GPC to seek an effective vaccine against CCHFV. FUNDING This research was supported by funding from the Biotechnology and Biological Sciences Research Council (UKRI-BBSRC) [BB/R019991/1 and BB/T008784/1].
Collapse
Affiliation(s)
- Jack E Saunders
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Ciaran Gilbride
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Stuart Dowall
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Susan Morris
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marta Ulaszewska
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandra J Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Emma Rayner
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Victoria A Graham
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Emma Kennedy
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Kelly Thomas
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Roger Hewson
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Sarah C Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sandra Belij-Rammerstorfer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Smith ES, Balch LA, Scrivens M, Shi S, Wang W, Harvey CD, Cornelison AA, Gil-Moore M, Kirk RA, Mueller LL, Hall RL, Howell AP, Reilly CA, Mayer JM, Murante FG, Viggiani KA, Gersz EM, Bussler H, Keefe MR, Evans EE, Paris MJ, Zauderer M. Use of poxvirus display to select antibodies specific for complex membrane antigens. MAbs 2023; 15:2249947. [PMID: 37635331 PMCID: PMC10464538 DOI: 10.1080/19420862.2023.2249947] [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: 02/15/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023] Open
Abstract
Antibody discovery against complex antigens is limited by the availability of a reproducible pure source of concentrated properly folded antigen. We have developed a technology to enable direct incorporation of membrane proteins such as GPCRs and into the membrane of poxvirus. The protein of interest is correctly folded and expressed in the cell-derived viral membrane and does not require any detergents or refolding before downstream use. The poxvirus is selective in which proteins are incorporated into the viral membrane, making the antigen poxvirus an antigenically cleaner target for in vitro panning. Antigen-expressing virus can be readily purified at scale and used for antibody selection using any in vitro display platform.
Collapse
Affiliation(s)
| | | | | | | | - Wei Wang
- Research, Vaccinex, Inc, Rochester, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Safety and immunogenicity of orally administered poxvirus vectored constructs in the white-footed mouse ( Peromyscus leucopus). Vaccine X 2022; 13:100259. [PMID: 36654838 PMCID: PMC9841169 DOI: 10.1016/j.jvacx.2022.100259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 11/15/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022] Open
Abstract
Globally, zoonotic spillover is becoming more frequent and represents a growing public health concern. Reservoir-targeted vaccination offers an intriguing alternative to traditional vaccine practices by establishing protection in wild populations that maintain the natural pathogen cycle. As an important pathogen reservoir, Peromyscus leucopus Rafinesque or the white-footed mouse has been the target of several experimental vaccines. However, strategies are limited by the method of administration, need for repeated dosing, or safety of constructs in the field. To address these concerns, we evaluated two highly attenuated poxviruses, raccoonpox virus (RCN) and modified vaccinia Ankara (MVA) virus as potential oral vaccine vectors in white-footed mice. Following oral administration, P. leucopus showed no adverse signs. A single oral dose elicited robust immune responses in mice to the foreign influenza hemagglutinin protein expressed by poxvirus vaccine vectors. Serum hemagglutinin inhibition antibody titers were detected by day 7 post immunization and persisted until study termination (77 days post immunization). This study establishes the safety and immunogenicity of recombinant MVA and RCN poxviruses in P. leucopus and demonstrates the suitability of these vectors as part of a reservoir-targeted vaccine strategy for white-footed mice.
Collapse
|
12
|
Mattar R, Neto ARB, Luz AG, Hatanaka A, Zaconeta A, Guazzelli CAF, Traina E, Baptista FS, Osanan G, Duarte G, Ramos JGL, Oppermann ML, Francisco RPV, Cardoso SMLDQ, Quintana SM, Sun SY, Borges VTM. Expert Recommendations on Monkeypox (MPX) in Pregnancy, Postpartum and Lactating Women. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2022; 44:1122-1125. [PMID: 36580939 PMCID: PMC9800143 DOI: 10.1055/s-0042-1759635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Rosiane Mattar
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Antonio Rodrigues Braga Neto
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Universidade Federal Fluminense, Niterói, RJ, Brazil
| | | | - Alan Hatanaka
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | | | - Evelyn Traina
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Gabriel Osanan
- Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Geraldo Duarte
- Faculdade de Medicina, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | - Sue Yazaki Sun
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
13
|
Coupet CA, Dubois C, Evlachev A, Kehrer N, Baldazza M, Hofman S, Vierboom M, Martin P, Inchauspe G. Intravenous injection of a novel viral immunotherapy encoding human interleukin-7 in nonhuman primates is safe and increases absolute lymphocyte count. Hum Vaccin Immunother 2022; 18:2133914. [PMID: 36315906 PMCID: PMC9746448 DOI: 10.1080/21645515.2022.2133914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Persistence of an immunosuppression, affecting both the innate and adaptive arms of the immune system, plays a role in sepsis patients' morbidity and late mortality pointing to the need for broad and effective immune interventions. MVA-hIL-7-Fc is a non-replicative recombinant Modified Vaccinia virus Ankara encoding the human interleukin-7 fused to human IgG2 Fc fragment. We have shown in murine sepsis models the capacity of this new virotherapy to stimulate both arms of the immune system and increase survival. Herein, an exploratory study in nonhuman primates was performed following a single intravenous injection of the MVA-hIL-7-Fc used at the clinical dose to assess its safety and biological activities. Four cynomolgus macaques were followed for 3 weeks post-injection (p.i), without observed acute adverse reactions. Circulating hIL-7-Fc was detected during the first 3-5 days p.i with a detection peaking at 12 h p.i. IL-7 receptor engagement and downstream signal transduction were detected in T cells demonstrating functionality of the expressed IL-7. Expansion of blood lymphocytes, mainly CD4 and CD8 naïve and central memory T cells, was observed on day 7 p.i. together with a transient increase of Ki67 expression on T lymphocytes. In addition, we observed an increase in circulating B and NK cells as well as monocytes were albeit with different kinetics and levels. This study indicates that a vectorized IL-7-Fc, injected by intravenous route at a relevant clinical dose in a large animal model, is active without adverse reactions supporting the clinical development of this novel virotherapy for treatment of sepsis patients.
Collapse
Affiliation(s)
| | | | | | - Nadine Kehrer
- Infectious Diseases Department, Transgene SA, Lyon, France
| | - Marie Baldazza
- Infectious Diseases Department, Transgene SA, Lyon, France
| | - Sam Hofman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Michel Vierboom
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Perrine Martin
- Infectious Diseases Department, Transgene SA, Lyon, France
| | - Geneviève Inchauspe
- Infectious Diseases Department, Transgene SA, Lyon, France,CONTACT Geneviève Inchauspe Infectious Diseases department, Transgene SA, 317 Avenue Jean Jaures, Lyon69007, France
| |
Collapse
|
14
|
Advances in Next-Generation Coronavirus Vaccines in Response to Future Virus Evolution. Vaccines (Basel) 2022; 10:vaccines10122035. [PMID: 36560445 PMCID: PMC9785936 DOI: 10.3390/vaccines10122035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread to more than 230 countries and territories worldwide since its outbreak in late 2019. In less than three years, infection by SARS-CoV-2 has resulted in over 600 million cases of COVID-19 and over 6.4 million deaths. Vaccines have been developed with unimaginable speed, and 11 have already been approved by the World Health Organization and given Emergency Use Listing. The administration of several first-generation SARS-CoV-2 vaccines has successfully decelerated the spread of COVID-19 but not stopped it completely. In the ongoing fight against viruses, genetic mutations frequently occur in the viral genome, resulting in a decrease in vaccine-induced antibody neutralization and widespread breakthrough infection. Facing the evolution and uncertainty of SARS-CoV-2 in the future, and the possibility of the spillover of other coronaviruses to humans, the need for vaccines with a broad spectrum of antiviral variants against multiple coronaviruses is recognized. It is imperative to develop a universal coronavirus or pan-coronavirus vaccine or drug to combat the ongoing COVID-19 pandemic as well as to prevent the next coronavirus pandemic. In this review, in addition to summarizing the protective effect of approved vaccines, we systematically summarize current work on the development of vaccines aimed at suppressing multiple SARS-CoV-2 variants of concern as well as multiple coronaviruses.
Collapse
|
15
|
Sacherl J, Kosinska AD, Kemter K, Kächele M, Laumen SC, Kerth HA, Öz EA, Wolff LS, Su J, Essbauer S, Sutter G, Scholz M, Singethan K, Altrichter J, Protzer U. Efficient stabilization of therapeutic hepatitis B vaccine components by amino-acid formulation maintains its potential to break immune tolerance. JHEP Rep 2022; 5:100603. [PMID: 36714793 PMCID: PMC9880034 DOI: 10.1016/j.jhepr.2022.100603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 09/05/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
Background & Aims Induction of potent, HBV-specific immune responses is crucial to control and finally cure HBV. The therapeutic hepatitis B vaccine TherVacB combines protein priming with a Modified Vaccinia virus Ankara (MVA)-vector boost to break immune tolerance in chronic HBV infection. Particulate protein and vector vaccine components, however, require a constant cooling chain for storage and transport, posing logistic and financial challenges to vaccine applications. We aimed to identify an optimal formulation to maintain stability and immunogenicity of the protein and vector components of the vaccine using a systematic approach. Methods We used stabilizing amino acid (SAA)-based formulations to stabilize HBsAg and HBV core particles (HBcAg), and the MVA-vector. We then investigated the effect of lyophilization and short- and long-term high-temperature storage on their integrity. Immunogenicity and safety of the formulated vaccine was validated in HBV-naïve and adeno-associated virus (AAV)-HBV-infected mice. Results In vitro analysis proved the vaccine's stability against thermal stress during lyophilization and the long-term stability of SAA-formulated HBsAg, HBcAg and MVA during thermal stress at 40 °C for 3 months and at 25 °C for 12 months. Vaccination of HBV-naïve and AAV-HBV-infected mice demonstrated that the stabilized vaccine was well tolerated and able to brake immune tolerance established in AAV-HBV mice as efficiently as vaccine components constantly stored at 4 °C/-80 °C. Even after long-term exposure to elevated temperatures, stabilized TherVacB induced high titre HBV-specific antibodies and strong CD8+ T-cell responses, resulting in anti-HBs seroconversion and strong suppression of the virus in HBV-replicating mice. Conclusion SAA-formulation resulted in highly functional and thermostable HBsAg, HBcAg and MVA vaccine components. This will facilitate global vaccine application without the need for cooling chains and is important for the development of prophylactic as well as therapeutic vaccines supporting vaccination campaigns worldwide. Impact and implications Therapeutic vaccination is a promising therapeutic option for chronic hepatitis B that may enable its cure. However, its application requires functional cooling chains during transport and storage that can hardly be guaranteed in many countries with high demand. In this study, the authors developed thermostable vaccine components that are well tolerated and that induce immune responses and control the virus in preclinical mouse models, even after long-term exposure to high surrounding temperatures. This will lower costs and ease application of a therapeutic vaccine and thus be beneficial for the many people affected by hepatitis B around the world.
Collapse
Key Words
- AAV, adeno-associated virus
- ALT, alanine aminotransferase
- CHB, chronic hepatitis B
- CTC, controlled temperature chain
- Ctrl, control
- DLS, dynamic light scattering
- HBcAg
- HBcAg, hepatitis B core antigen
- HBeAg, hepatitis B e antigen
- HBsAg
- HBsAg, hepatitis B surface antigen
- Heat-stable vaccine
- ICS, intracellular cytokine staining
- IFNα, interferon alpha
- MVA
- MVA, Modified Vaccinia virus Ankara
- NAGE, native agarose gel electrophoresis
- RH, relative humidity
- RT, room temperature
- SAA, stabilizing amino acids
- SEC-HPLC, size exclusion-high performance liquid chromatography
- SPS®
- TCID50, median tissue culture infection dose
- TherVacBCtrl, non-lyophilized
- WHO, World Health Organization
- anti-HBc, hepatitis B core antibodies
- anti-HBs, hepatitis B surface antibodies
- cccDNA, covalently closed circular DNA
- formulation
- hepatitis B virus
- heterologous prime/boost vaccination
- lyophilization
- non-stressed, non-stabilized TherVacB
- stabilization
- stabilizing amino acid-based formulation
- stabilizing excipients
Collapse
Affiliation(s)
- Julia Sacherl
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | - Anna D. Kosinska
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | | | - Martin Kächele
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | - Sabine C. Laumen
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | - Hélène A. Kerth
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Edanur Ates Öz
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | - Lisa S. Wolff
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | - Jinpeng Su
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
| | | | - Gerd Sutter
- Institute of Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | | | - Katrin Singethan
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
- Bundeswehr Institute of Microbiology, Munich, Germany
| | | | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
- Corresponding author. Address: Institute of Virology, Trogerstr. 30, 81675 Munich, Germany; Tel.: +49-89-4140-6821, fax: +49-89-4140-6823.
| |
Collapse
|
16
|
Orlova OV, Glazkova DV, Bogoslovskaya EV, Shipulin GA, Yudin SM. Development of Modified Vaccinia Virus Ankara-Based Vaccines: Advantages and Applications. Vaccines (Basel) 2022; 10:vaccines10091516. [PMID: 36146594 PMCID: PMC9503770 DOI: 10.3390/vaccines10091516] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) is a promising viral vector for vaccine development. MVA is well studied and has been widely used for vaccination against smallpox in Germany. This review describes the history of the origin of the virus and its properties as a vaccine, including a high safety profile. In recent years, MVA has found its place as a vector for the creation of vaccines against various diseases. To date, a large number of vaccine candidates based on the MVA vector have already been developed, many of which have been tested in preclinical and clinical studies. We discuss data on the immunogenicity and efficacy of some of these vaccines.
Collapse
|
17
|
Khalil A, Samara A, O'Brien P, Morris E, Draycott T, Lees C, Ladhani S. Monkeypox vaccines in pregnancy: lessons must be learned from COVID-19. Lancet Glob Health 2022; 10:e1230-e1231. [PMID: 35772413 PMCID: PMC9236565 DOI: 10.1016/s2214-109x(22)00284-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Asma Khalil
- Fetal Medicine Unit, St George's Hospital, St George's University of London, London, UK; Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK.
| | - Athina Samara
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Pat O'Brien
- The Royal College of Obstetricians and Gynaecologists, London, UK; Department of Women's Health, University College London Hospitals NHS Foundation Trust, London, UK
| | - Edward Morris
- The Royal College of Obstetricians and Gynaecologists, London, UK; Department of Women's Health, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, Norfolk, UK
| | - Tim Draycott
- The Royal College of Obstetricians and Gynaecologists, London, UK; North Bristol NHS Trust Department of Women's Health, Westbury on Trym, UK
| | - Christoph Lees
- Department of Women's Health, Imperial College, London, UK
| | - Shamez Ladhani
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute of Infection and Immunity, St George's University of London, London, UK; Immunisation and Countermeasures Division, Public Health England, London, UK; British Paediatric Surveillance Unit, Royal College of Paediatrics and Child Health, London, UK
| |
Collapse
|
18
|
Lélu K, Dubois C, Evlachev A, Crausaz M, Baldazza M, Kehrer N, Brandely R, Schlesinger Y, Silvestre N, Marchand JB, Bastien B, Leung-Theung-Long S, Unsinger J, Martin P, Inchauspé G. Viral Delivery of IL-7 Is a Potent Immunotherapy Stimulating Innate and Adaptive Immunity and Confers Survival in Sepsis Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:99-117. [PMID: 35667841 DOI: 10.4049/jimmunol.2101145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/25/2022] [Indexed: 12/29/2022]
Abstract
Persistence of an immunosuppressive state plays a role in septic patient morbidity and late mortality. Both innate and adaptive pathways are impaired, pointing toward the need for immune interventions targeting both arms of the immune system. We developed a virotherapy using the nonpropagative modified vaccinia virus Ankara (MVA), which harbors the intrinsic capacity to stimulate innate immunity, to deliver IL-7, a potent activator of adaptive immunity. The rMVA-human IL-7 (hIL-7)-Fc encoding the hIL-7 fused to the human IgG2-Fc was engineered and shown to express a dimeric, glycosylated, and biologically active cytokine. Following a single i.v. injection in naive mice, the MVA-hIL-7-Fc increased the number of total and activated B, T, and NK cells but also myeloid subpopulations (Ly6Chigh, Ly6Cint, and Ly6Cneg cells) in both lung and spleen. It triggered differentiation of T cells in central memory, effector memory, and acute effector phenotypes and enhanced polyfunctionality of T cells, notably the number of IFN-γ-producing cells. The MVA vector contributed significantly to immune cell activation, particularly of NK cells. The MVA-hIL-7-Fc conferred a significant survival advantage in the cecal ligation and puncture (CLP) and Candida albicans sepsis models. It significantly increased cell numbers and activation in both spleen and lung of CLP mice. Comparatively, in naive and CLP mice, the rhIL-7-Fc soluble counterpart overall induced less vigorous, shorter lasting, and narrower immune activities than did the MVA-hIL-7-Fc and favored TNF-α-producing cells. The MVA-hIL-7-Fc represents a novel class of immunotherapeutic with clinical potential for treatment of septic patients.
Collapse
Affiliation(s)
- Karine Lélu
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Clarisse Dubois
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Alexei Evlachev
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Morgane Crausaz
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Marie Baldazza
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Nadine Kehrer
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Renée Brandely
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | - Yasmin Schlesinger
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | - Nathalie Silvestre
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | | | - Bérangère Bastien
- Department of Medical Affairs, Transgene SA, Illkirch-Graffenstraden, France
| | | | - Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO.,Department of Medicine, Washington University School of Medicine, St. Louis, MO; and.,Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Perrine Martin
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | | |
Collapse
|
19
|
Chakraborty E, Sarkar D. Emerging Therapies for Hepatocellular Carcinoma (HCC). Cancers (Basel) 2022; 14:2798. [PMID: 35681776 PMCID: PMC9179883 DOI: 10.3390/cancers14112798] [Citation(s) in RCA: 185] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 01/30/2023] Open
Abstract
Hepatocellular carcinoma (HCC) arises from hepatocytes and accounts for 90% of primary liver cancer. According to Global Cancer Incidence, Mortality and Prevalence (GLOBOCAN) 2020, globally HCC is the sixth most common cancer and the third most common cause of cancer-related deaths. Reasons for HCC prognosis remaining dismal are that HCC is asymptomatic in its early stages, leading to late diagnosis, and it is markedly resistant to conventional chemo- and radiotherapy. Liver transplantation is the treatment of choice in early stages, while surgical resection, radiofrequency ablation (RFA) and trans arterial chemoembolization (TACE) are Food and Drug Administration (FDA)-approved treatments for advanced HCC. Additional first line therapy for advanced HCC includes broad-spectrum tyrosine kinase inhibitors (TKIs), such as sorafenib and lenvatinib, as well as a combination of immunotherapy and anti-angiogenesis therapy, namely atezolizumab and bevacizumab. However, these strategies provide nominal extension in the survival curve, cause broad spectrum toxic side effects, and patients eventually develop therapy resistance. Some common mutations in HCC, such as in telomerase reverse transcriptase (TERT), catenin beta 1 (CTNNB1) and tumor protein p53 (TP53) genes, are still considered to be undruggable. In this context, identification of appropriate gene targets and specific gene delivery approaches create the potential of gene- and immune-based therapies for the safe and effective treatment of HCC. This review elaborates on the current status of HCC treatment by focusing on potential gene targets and advanced techniques, such as oncolytic viral vectors, nanoparticles, chimeric antigen receptor (CAR)-T cells, immunotherapy, and clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9), and describes future prospects in HCC treatment.
Collapse
Affiliation(s)
- Eesha Chakraborty
- C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Massey Cancer Center, VCU Institute of Molecular Medicine (VIMM), Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
20
|
Measles-based Zika vaccine induces long-term immunity and requires NS1 antibodies to protect the female reproductive tract. NPJ Vaccines 2022; 7:43. [PMID: 35440656 PMCID: PMC9018676 DOI: 10.1038/s41541-022-00464-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Zika virus (ZIKV) can cause devastating effects in the unborn fetus of pregnant women. To develop a candidate vaccine that can protect human fetuses, we generated a panel of live measles vaccine (MV) vectors expressing ZIKV-E and -NS1. Our MV-based ZIKV-E vaccine, MV-E2, protected mice from the non-lethal Zika Asian strain (PRVABC59) and the lethal African strain (MR766) challenge. Despite 100% survival of the MV-E2 mice, however, complete viral clearance was not achieved in the brain and reproductive tract of the lethally challenged mice. We then tested MV-based vaccines that expressed E and NS1 together or separately in two different vaccines. We observed complete clearance of ZIKV from the female reproductive tract and complete fetal protection in the lethal African challenge model in animals that received the dual antigen vaccines. Additionally, MV-E2 and MV-NS1, when administered together, induced durable plasma cell responses. Our findings suggest that NS1 antibodies are required to enhance the protection of ZIKV-E antibodies in the female reproductive tract.
Collapse
|
21
|
Pursell T, Spencer Clinton JL, Tan J, Peng R, Ling PD. Modified vaccinia Ankara expressing EEHV1A glycoprotein B elicits humoral and cell-mediated immune responses in mice. PLoS One 2022; 17:e0265424. [PMID: 35312707 PMCID: PMC8936464 DOI: 10.1371/journal.pone.0265424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
Elephant endotheliotropic herpesvirus (EEHV) can cause lethal hemorrhagic disease (EEHV-HD) in Asian elephants and is the largest cause of death in captive juvenile Asian elephants in North America and Europe. EEHV-HD also has been documented in captive and wild elephants in their natural range countries. A safe and effective vaccine to prevent lethal EEHV infection would significantly improve conservation efforts for this endangered species. Recent studies from our laboratory suggest that EEHV morbidity and mortality are often associated with primary infection. Therefore, we aim to generate a vaccine, particularly for EEHV1 naïve animals, with the goal of preventing lethal EEHV-HD. To address this goal, we generated a Modified Vaccinia Ankara (MVA) recombinant virus expressing a truncated form of glycoprotein B (gBΔfur731) from EEHV1A, the strain associated with the majority of lethal EEHV cases. Vaccination of CD-1 mice with this recombinant virus induced robust antibody and polyfunctional T cell responses significantly above mice inoculated with wild-type MVA. Although the vaccine-induced T cell response was mainly observed in CD8+ T cell populations, the CD4+ T cell response was also polyfunctional. No adverse responses to vaccination were observed. Overall, our data demonstrates that MVA-gBΔfur731 stimulates robust humoral and cell-mediated responses, supporting its potential translation for use in elephants.
Collapse
Affiliation(s)
- Taylor Pursell
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jennifer L. Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jie Tan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rongsheng Peng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Paul D. Ling
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
22
|
Lee MH, Kim BJ. COVID-19 vaccine development based on recombinant viral and bacterial vector systems: combinatorial effect of adaptive and trained immunity. J Microbiol 2022; 60:321-334. [PMID: 35157221 PMCID: PMC8853094 DOI: 10.1007/s12275-022-1621-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/11/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 virus (SARS-CoV-2) infection, which causes coronavirus disease 2019 (COVID-19), has led to many cases and deaths worldwide. Therefore, a number of vaccine candidates have been developed to control the COVID-19 pandemic. Of these, to date, 21 vaccines have received emergency approval for human use in at least one country. However, the recent global emergence of SARS-CoV-2 variants has compromised the efficacy of the currently available vaccines. To protect against these variants, the use of vaccines that modulate T cell-mediated immune responses or innate immune cell memory function, termed trained immunity, is needed. The major advantage of a vaccine that uses bacteria or viral systems for the delivery of COVID-19 antigens is the ability to induce both T cell-mediated and humoral immune responses. In addition, such vaccine systems can also exert off-target effects via the vector itself, mediated partly through trained immunity; compared to other vaccine platforms, suggesting that this approach can provide better protection against even vaccine escape variants. This review presents the current status of the development of COVID-19 vaccines based on recombinant viral and bacterial delivery systems. We also discuss the current status of the use of licensed live vaccines for other infections, including BCG, oral polio and MMR vaccines, to prevent COVID-19 infections.
Collapse
Affiliation(s)
- Mi-Hyun Lee
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Seoul National University Medical Research Center (SNUMRC), Seoul, 03080, Republic of Korea.
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
23
|
Damerval M, Fagnoni-Legat C, Louvrier A, Fischer S, Limat S, Clairet AL, Nerich V, Madelaine I, Kroemer M. ATMP Environmental Exposure Assessment in European Healthcare Settings: A Systematic Review of the Literature. Front Med (Lausanne) 2021; 8:713047. [PMID: 34926483 PMCID: PMC8671638 DOI: 10.3389/fmed.2021.713047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
Since 2007, a new class of biologic products for human use called "advanced therapy medicinal products (ATMP)" have been legally integrated in the European Medical Agency. They consist of recombinant nucleic acid, engineered cells, cells, or tissues. In the United States, ATMP fall under the regulatory framework of biological products and the term "cell and gene therapy product" is used in the legislative and regulatory documents. Potential clinical applications are broad, particularly, in the field of cancer, inherited genetic disease, and regenerative medicine. Indeed, the benefit conferred by CD19 chimeric antigen receptor T cells led to the first engineered cell therapy products to be approved by the Food and Drug Administration (FDA) in 2017. Gene therapy products to treat orphan diseases are also extensively developed with many clinical trials ongoing in the world. Nevertheless, the use of these therapeutic products is complex and requires careful considerations in the terms of regulatory and hospital setting requirements, such as storage, handling, administration, and disposal which justify the implementation of a secured medication circuit. Through this systematic review of the literature, the authors wanted to compile data on the assessment of environmental exposure related to the use of ATMP in healthcare setting to secure their medication circuit. A literature search was conducted on PubMed and Web of Science, and 32 publications dealing with environmental exposure assessment and ATMP were selected. In addition, marketed ATMPs were identified and data regarding the environmental concerns were extracted from product information sections from European Public Assessment Reports (EPAR). The environmental contamination assessments were mainly addressed in the reviews rather than in original articles related to the use of ATMP. Most of the product information sections from EPAR suggested precautions rather than requirements when dealing with environmental consideration following ATMP handling. Nevertheless, these precautions usually remain elusive especially concerning waste disposal and the detection of biological material on the work surfaces, and mainly relate to the genetically modified organisms (GMO) over non-GMO cellular products. Pharmaceutical oversight and adherence to the good preparation practices and good clinical practices are essential to ensure the safe use in term of environmental concern of these new therapeutic products in healthcare setting.
Collapse
Affiliation(s)
- Margaux Damerval
- Department of Pharmacy, University Hospital of Besançon, Besançon, France
| | | | - Aurélien Louvrier
- Host-Graft Interactions Lab – Tumor - Cell and Tissue engineering (UMR 1098 INSERM/UFC/EFS), University of Franche-Comté, Besançon, France
- Department of Oral and Maxillofacial Surgery, University Hospital of Besançon, Besançon, France
| | - Sarah Fischer
- Department of Pharmacy, University Hospital of Besançon, Besançon, France
| | - Samuel Limat
- Department of Pharmacy, University Hospital of Besançon, Besançon, France
- Department of Oral and Maxillofacial Surgery, University Hospital of Besançon, Besançon, France
| | - Anne-Laure Clairet
- Department of Pharmacy, University Hospital of Besançon, Besançon, France
| | - Virginie Nerich
- Department of Pharmacy, University Hospital of Besançon, Besançon, France
- Department of Oral and Maxillofacial Surgery, University Hospital of Besançon, Besançon, France
| | | | - Marie Kroemer
- Department of Pharmacy, University Hospital of Besançon, Besançon, France
- Department of Oral and Maxillofacial Surgery, University Hospital of Besançon, Besançon, France
| |
Collapse
|
24
|
Kulkarni R, Chen WC, Lee Y, Kao CF, Hu SL, Ma HH, Jan JT, Liao CC, Liang JJ, Ko HY, Sun CP, Lin YS, Wang YC, Wei SC, Lin YL, Ma C, Chao YC, Chou YC, Chang W. Vaccinia virus-based vaccines confer protective immunity against SARS-CoV-2 virus in Syrian hamsters. PLoS One 2021; 16:e0257191. [PMID: 34499677 PMCID: PMC8428573 DOI: 10.1371/journal.pone.0257191] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
COVID-19 in humans is caused by Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) that belongs to the beta family of coronaviruses. SARS-CoV-2 causes severe respiratory illness in 10-15% of infected individuals and mortality in 2-3%. Vaccines are urgently needed to prevent infection and to contain viral spread. Although several mRNA- and adenovirus-based vaccines are highly effective, their dependence on the "cold chain" transportation makes global vaccination a difficult task. In this context, a stable lyophilized vaccine may present certain advantages. Accordingly, establishing additional vaccine platforms remains vital to tackle SARS-CoV-2 and any future variants that may arise. Vaccinia virus (VACV) has been used to eradicate smallpox disease, and several attenuated viral strains with enhanced safety for human applications have been developed. We have generated two candidate SARS-CoV-2 vaccines based on two vaccinia viral strains, MVA and v-NY, that express full-length SARS-CoV-2 spike protein. Whereas MVA is growth-restricted in mammalian cells, the v-NY strain is replication-competent. We demonstrate that both candidate recombinant vaccines induce high titers of neutralizing antibodies in C57BL/6 mice vaccinated according to prime-boost regimens. Furthermore, our vaccination regimens generated TH1-biased immune responses in mice. Most importantly, prime-boost vaccination of a Syrian hamster infection model with MVA-S and v-NY-S protected the hamsters against SARS-CoV-2 infection, supporting that these two vaccines are promising candidates for future development. Finally, our vaccination regimens generated neutralizing antibodies that partially cross-neutralized SARS-CoV-2 variants of concern.
Collapse
Affiliation(s)
- Rakesh Kulkarni
- Molecular and Cell Biology, Taiwan International Graduate Program, National Defense Medical Center, Academia Sinica and Graduate Institute of Life Science, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Wen-Ching Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ying Lee
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chi-Fei Kao
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Shiu-Lok Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington, United States of America
| | - Hsiu-Hua Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chun-Che Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jian-Jong Liang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Ying Ko
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Cheng-Pu Sun
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yin-Shoiou Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Chiuan Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Academi Sinica SPF Animal Facility, Academia Sinica, Taipei, Taiwan
| | - Sung-Chan Wei
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Che Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chan Chao
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, Taiwan
| | - Wen Chang
- Molecular and Cell Biology, Taiwan International Graduate Program, National Defense Medical Center, Academia Sinica and Graduate Institute of Life Science, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
25
|
Volkmann A, Williamson AL, Weidenthaler H, Meyer TPH, Robertson JS, Excler JL, Condit RC, Evans E, Smith ER, Kim D, Chen RT. The Brighton Collaboration standardized template for collection of key information for risk/benefit assessment of a Modified Vaccinia Ankara (MVA) vaccine platform. Vaccine 2021; 39:3067-3080. [PMID: 33077299 PMCID: PMC7568176 DOI: 10.1016/j.vaccine.2020.08.050] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/25/2022]
Abstract
The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety and characteristics of live, recombinant viral vector vaccines. The Modified Vaccinia Ankara (MVA) vector system is being explored as a platform for development of multiple vaccines. This paper reviews the molecular and biological features specifically of the MVA-BN vector system, followed by a template with details on the safety and characteristics of an MVA-BN based vaccine against Zaire ebolavirus and other filovirus strains. The MVA-BN-Filo vaccine is based on a live, highly attenuated poxviral vector incapable of replicating in human cells and encodes glycoproteins of Ebola virus Zaire, Sudan virus and Marburg virus and the nucleoprotein of the Thai Forest virus. This vaccine has been approved in the European Union in July 2020 as part of a heterologous Ebola vaccination regimen. The MVA-BN vector is attenuated following over 500 serial passages in eggs, showing restricted host tropism and incompetence to replicate in human cells. MVA has six major deletions and other mutations of genes outside these deletions, which all contribute to the replication deficiency in human and other mammalian cells. Attenuation of MVA-BN was demonstrated by safe administration in immunocompromised mice and non-human primates. In multiple clinical trials with the MVA-BN backbone, more than 7800 participants have been vaccinated, demonstrating a safety profile consistent with other licensed, modern vaccines. MVA-BN has been approved as smallpox vaccine in Europe and Canada in 2013, and as smallpox and monkeypox vaccine in the US in 2019. No signal for inflammatory cardiac disorders was identified throughout the MVA-BN development program. This is in sharp contrast to the older, replicating vaccinia smallpox vaccines, which have a known risk for myocarditis and/or pericarditis in up to 1 in 200 vaccinees. MVA-BN-Filo as part of a heterologous Ebola vaccination regimen (Ad26.ZEBOV/MVA-BN-Filo) has undergone clinical testing including Phase III in West Africa and is currently in use in large scale vaccination studies in Central African countries. This paper provides a comprehensive picture of the MVA-BN vector, which has reached regulatory approvals, both as MVA-BN backbone for smallpox/monkeypox, as well as for the MVA-BN-Filo construct as part of an Ebola vaccination regimen, and therefore aims to provide solutions to prevent disease from high-consequence human pathogens.
Collapse
Affiliation(s)
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine at the University of Cape Town, South Africa
| | | | | | | | | | - Richard C Condit
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Eric Evans
- Brighton Collaboration, a Program of the Task Force for Global Health, Decatur, GA, USA
| | - Emily R Smith
- Brighton Collaboration, a Program of the Task Force for Global Health, Decatur, GA, USA.
| | - Denny Kim
- Janssen Pharmaceuticals, Titusville, NJ, USA
| | - Robert T Chen
- Brighton Collaboration, a Program of the Task Force for Global Health, Decatur, GA, USA
| |
Collapse
|
26
|
Baldo A, Leunda A, Willemarck N, Pauwels K. Environmental Risk Assessment of Recombinant Viral Vector Vaccines against SARS-Cov-2. Vaccines (Basel) 2021; 9:453. [PMID: 34063733 PMCID: PMC8147846 DOI: 10.3390/vaccines9050453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/23/2021] [Accepted: 05/01/2021] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the coronavirus disease 2019 (COVID-19) pandemic. Over the past months, considerable efforts have been put into developing effective and safe drugs and vaccines against SARS-CoV-2. Various platforms are being used for the development of COVID-19 vaccine candidates: recombinant viral vectors, protein-based vaccines, nucleic acid-based vaccines, and inactivated/attenuated virus. Recombinant viral vector vaccine candidates represent a significant part of those vaccine candidates in clinical development, with two already authorised for use in the European Union and one currently under rolling review by the European Medicines Agency (EMA). Since recombinant viral vector vaccine candidates are considered as genetically modified organisms (GMOs), their regulatory oversight includes besides an assessment of their quality, safety and efficacy, also an environmental risk assessment (ERA). The present article highlights the main characteristics of recombinant viral vector vaccine (candidates) against SARS-CoV-2 in the pipeline and discusses their features from an environmental risk point of view.
Collapse
Affiliation(s)
- Aline Baldo
- Sciensano, Service Biosafety and Biotechnology, Rue Juliette Wytsmanstraat 14, B-1050 Brussels, Belgium; (A.L.); (N.W.); (K.P.)
| | | | | | | |
Collapse
|
27
|
Hazlewood JE, Dumenil T, Le TT, Slonchak A, Kazakoff SH, Patch AM, Gray LA, Howley PM, Liu L, Hayball JD, Yan K, Rawle DJ, Prow NA, Suhrbier A. Injection site vaccinology of a recombinant vaccinia-based vector reveals diverse innate immune signatures. PLoS Pathog 2021; 17:e1009215. [PMID: 33439897 PMCID: PMC7837487 DOI: 10.1371/journal.ppat.1009215] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/26/2021] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Poxvirus systems have been extensively used as vaccine vectors. Herein a RNA-Seq analysis of intramuscular injection sites provided detailed insights into host innate immune responses, as well as expression of vector and recombinant immunogen genes, after vaccination with a new multiplication defective, vaccinia-based vector, Sementis Copenhagen Vector. Chikungunya and Zika virus immunogen mRNA and protein expression was associated with necrosing skeletal muscle cells surrounded by mixed cellular infiltrates. The multiple adjuvant signatures at 12 hours post-vaccination were dominated by TLR3, 4 and 9, STING, MAVS, PKR and the inflammasome. Th1 cytokine signatures were dominated by IFNγ, TNF and IL1β, and chemokine signatures by CCL5 and CXCL12. Multiple signatures associated with dendritic cell stimulation were evident. By day seven, vaccine transcripts were absent, and cell death, neutrophil, macrophage and inflammation annotations had abated. No compelling arthritis signatures were identified. Such injection site vaccinology approaches should inform refinements in poxvirus-based vector design. Poxvirus vector systems have been widely developed for vaccine applications. Despite considerable progress, so far only one recombinant poxvirus vectored vaccine has to date been licensed for human use, with ongoing efforts seeking to enhance immunogenicity whilst minimizing reactogenicity. The latter two characteristics are often determined by early post-vaccination events at the injection site. We therefore undertook an injection site vaccinology approach to analyzing gene expression at the vaccination site after intramuscular inoculation with a recombinant, multiplication defective, vaccinia-based vaccine. This provided detailed insights into inter alia expression of vector-encoded immunoregulatory genes, as well as host innate and adaptive immune responses. We propose that such injection site vaccinology can inform rational vaccine vector design, and we discuss how the information and approach elucidated herein might be used to improve immunogenicity and limit reactogenicity of poxvirus-based vaccine vector systems.
Collapse
Affiliation(s)
- Jessamine E. Hazlewood
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Troy Dumenil
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Thuy T. Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andrii Slonchak
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - Stephen H. Kazakoff
- Clinical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Ann-Marie Patch
- Clinical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Lesley-Ann Gray
- Australian Genome Research Facility Ltd., Melbourne, Australia
| | | | - Liang Liu
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - John D. Hayball
- Sementis Ltd., Hackney, Australia
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Daniel J. Rawle
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Natalie A. Prow
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Australian Infectious Disease Research Centre, Brisbane, Australia
- * E-mail:
| |
Collapse
|
28
|
Koban R, Lam T, Schwarz F, Kloke L, Bürge S, Ellerbrok H, Neumann M. Simplified Bioprinting-Based 3D Cell Culture Infection Models for Virus Detection. Viruses 2020; 12:E1298. [PMID: 33198291 PMCID: PMC7698278 DOI: 10.3390/v12111298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Studies of virus-host interactions in vitro may be hindered by biological characteristics of conventional monolayer cell cultures that differ from in vivo infection. Three-dimensional (3D) cell cultures show more in vivo-like characteristics and may represent a promising alternative for characterisation of infections. In this study, we established easy-to-handle cell culture platforms based on bioprinted 3D matrices for virus detection and characterisation. Different cell types were cultivated on these matrices and characterised for tissue-like growth characteristics regarding cell morphology and polarisation. Cells developed an in vivo-like morphology and long-term cultivation was possible on the matrices. Cell cultures were infected with viruses which differed in host range, tissue tropism, cytopathogenicity, and genomic organisation and virus morphology. Infections were characterised on molecular and imaging level. The transparent matrix substance allowed easy optical monitoring of cells and infection even via live-cell microscopy. In conclusion, we established an enhanced, standardised, easy-to-handle bioprinted 3D-cell culture system. The infection models are suitable for sensitive monitoring and characterisation of virus-host interactions and replication of different viruses under physiologically relevant conditions. Individual cell culture models can further be combined to a multicellular array. This generates a potent diagnostic tool for propagation and characterisation of viruses from diagnostic samples.
Collapse
Affiliation(s)
- Robert Koban
- Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Seestr. 10, 13353 Berlin, Germany; (R.K.); (F.S.); (M.N.)
| | - Tobias Lam
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355 Berlin, Germany; (T.L.); (L.K.)
| | - Franziska Schwarz
- Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Seestr. 10, 13353 Berlin, Germany; (R.K.); (F.S.); (M.N.)
| | - Lutz Kloke
- Cellbricks GmbH, Gustav-Meyer-Allee 25, 13355 Berlin, Germany; (T.L.); (L.K.)
| | - Silvio Bürge
- Advanced Light and Electron Microscopy, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Seestr. 10, 13353 Berlin, Germany;
| | - Heinz Ellerbrok
- Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Seestr. 10, 13353 Berlin, Germany; (R.K.); (F.S.); (M.N.)
| | - Markus Neumann
- Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Seestr. 10, 13353 Berlin, Germany; (R.K.); (F.S.); (M.N.)
| |
Collapse
|
29
|
Lothert K, Pagallies F, Feger T, Amann R, Wolff MW. Selection of chromatographic methods for the purification of cell culture-derived Orf virus for its application as a vaccine or viral vector. J Biotechnol 2020; 323:62-72. [PMID: 32763261 PMCID: PMC7403136 DOI: 10.1016/j.jbiotec.2020.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/23/2020] [Accepted: 07/31/2020] [Indexed: 12/02/2022]
Abstract
Estimation of the isoelectric point and size of Vero cell-derived Orf virus. Limited dynamic binding capacity of tested Orf virus to sulfated cellulose. Purification of Orf virus by steric exclusion chromatography lead to 84 % recovery. Hydrophobic interaction chromatography suitable for Orf virus purification. Promising unit operations for a scalable DSP to produce Orf virus viral vectors.
In recent years, the Orf virus has become a promising tool for protective recombinant vaccines and oncolytic therapy. However, suitable methods for an Orf virus production, including up- and downstream, are very limited. The presented study focuses on downstream processing, describing the evaluation of different chromatographic unit operations. In this context, ion exchange-, pseudo-affinity- and steric exclusion chromatography were employed for the purification of the cell culture-derived Orf virus, aiming at a maximum in virus recovery and contaminant depletion. The most promising chromatographic methods for capturing the virus particles were the steric exclusion- or salt-tolerant anion exchange membrane chromatography, recovering 84 % and 86 % of the infectious virus. Combining the steric exclusion chromatography with a subsequent Capto™ Core 700 resin or hydrophobic interaction membrane chromatography as a secondary chromatographic step, overall virus recoveries of up to 76 % were achieved. Furthermore, a complete cellular protein removal and a host cell DNA depletion of up to 82 % was possible for the steric exclusion membranes and the Capto™ Core 700 combination. The study reveals a range of possible unit operations suited for the chromatographic purification of the cell culture-derived Orf virus, depending on the intended application, i.e. a human or veterinary use, and the required purity.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany
| | - Felix Pagallies
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Thomas Feger
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany.
| |
Collapse
|
30
|
A vital gene for modified vaccinia virus Ankara replication in human cells. Proc Natl Acad Sci U S A 2020; 117:6289-6291. [PMID: 32179684 PMCID: PMC7104218 DOI: 10.1073/pnas.2001335117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
31
|
del Moral-Sánchez I, Sliepen K. Strategies for inducing effective neutralizing antibody responses against HIV-1. Expert Rev Vaccines 2019; 18:1127-1143. [PMID: 31791150 PMCID: PMC6961309 DOI: 10.1080/14760584.2019.1690458] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: Despite intensive research efforts, there is still no effective prophylactic vaccine available against HIV-1. Currently, substantial efforts are devoted to the development of vaccines aimed at inducing broadly neutralizing antibodies (bNAbs), which are capable of neutralizing most HIV-1 strains. All bNAbs target the HIV-1 envelope glycoprotein (Env), but Env immunizations usually only induce neutralizing antibodies (NAbs) against the sequence-matched virus and not against other strains.Areas covered: We describe the different strategies that have been explored to improve the breadth and potency of anti-HIV-1 NAb responses. The discussed strategies include the application of engineered Env immunogens, optimization of (bNAb) epitopes, different cocktail and sequential vaccination strategies, nanoparticles and nucleic acid-based vaccines.Expert opinion: A combination of the strategies described in this review and future approaches are probably needed to develop an effective HIV-1 vaccine that can induce broad, potent and long-lasting NAb responses.
Collapse
Affiliation(s)
- Iván del Moral-Sánchez
- Department of Medical Microbiology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Kwinten Sliepen
- Department of Medical Microbiology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands,CONTACT Kwinten Sliepen Department of Medical Microbiology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Salvato MS, Domi A, Guzmán-Cardozo C, Medina-Moreno S, Zapata JC, Hsu H, McCurley N, Basu R, Hauser M, Hellerstein M, Guirakhoo F. A Single Dose of Modified Vaccinia Ankara Expressing Lassa Virus-like Particles Protects Mice from Lethal Intra-cerebral Virus Challenge. Pathogens 2019; 8:E133. [PMID: 31466243 PMCID: PMC6789566 DOI: 10.3390/pathogens8030133] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/23/2019] [Accepted: 08/25/2019] [Indexed: 12/13/2022] Open
Abstract
Lassa fever surpasses Ebola, Marburg, and all other hemorrhagic fevers except Dengue in its public health impact. Caused by Lassa virus (LASV), the disease is a scourge on populations in endemic areas of West Africa, where reported incidence is higher. Here, we report construction, characterization, and preclinical efficacy of a novel recombinant vaccine candidate GEO-LM01. Constructed in the Modified Vaccinia Ankara (MVA) vector, GEO-LM01 expresses the glycoprotein precursor (GPC) and zinc-binding matrix protein (Z) from the prototype Josiah strain lineage IV. When expressed together, GP and Z form Virus-Like Particles (VLPs) in cell culture. Immunogenicity and efficacy of GEO-LM01 was tested in a mouse challenge model. A single intramuscular dose of GEO-LM01 protected 100% of CBA/J mice challenged with a lethal dose of ML29, a Mopeia/Lassa reassortant virus, delivered directly into the brain. In contrast, all control animals died within one week. The vaccine induced low levels of antibodies but Lassa-specific CD4+ and CD8+ T cell responses. This is the first report showing that a single dose of a replication-deficient MVA vector can confer full protection against a lethal challenge with ML29 virus.
Collapse
Affiliation(s)
- Maria S Salvato
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | | | | | | | - Juan Carlos Zapata
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | - Haoting Hsu
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | - Nathanael McCurley
- Office of Technology Licensing and Commercialization, Georgia State University, Atlanta, GA 30303, USA
| | - Rahul Basu
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | | | | | |
Collapse
|
33
|
Arabipour I, Amani J, Mirhosseini SA, Salimian J. The study of genes and signal transduction pathways involved in mustard lung injury: A gene therapy approach. Gene 2019; 714:143968. [PMID: 31323308 DOI: 10.1016/j.gene.2019.143968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 07/06/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Sulfur mustard (SM) is a destructive and harmful chemical agent for the eyes, skin and lungs that causes short-term and long-term lesions and was widely used in Iraq war against Iran (1980-1988). SM causes DNA damages, oxidative stress, and Inflammation. Considering the similarities between SM and COPD (Chronic Obstructive Pulmonary Disease) pathogens and limited available treatments, a novel therapeutic approach is not developed. Gene therapy is a novel therapeutic approach that uses genetic engineering science in treatment of most diseases including chronic obstructive pulmonary disease. In this review, attempts to presenting a comprehensive study of mustard lung and introducing the genes therapy involved in chronic obstructive pulmonary disease and emphasizing the pathways and genes involved in the pathology and pathogenesis of sulfur Mustard. It seems that, given the high potential of gene therapy and the fact that this experimental technique is a candidate for the treatment of pulmonary diseases, further study of genes, vectors and gene transfer systems can draw a very positive perspective of gene therapy in near future.
Collapse
Affiliation(s)
- Iman Arabipour
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Langenmayer MC, Lülf-Averhoff AT, Adam-Neumair S, Fux R, Sutter G, Volz A. Distribution and absence of generalized lesions in mice following single dose intramuscular inoculation of the vaccine candidate MVA-MERS-S. Biologicals 2018; 54:58-62. [PMID: 29759890 PMCID: PMC7128986 DOI: 10.1016/j.biologicals.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 03/14/2018] [Accepted: 05/04/2018] [Indexed: 11/29/2022] Open
Abstract
Modified Vaccinia Virus Ankara (MVA) is a highly attenuated and replication-deficient virus serving as vaccine against infectious diseases. Here, we assessed the in vivo distribution of a recombinant MVA candidate vaccine against the Middle Eastern Respiratory Syndrome (MVA-MERS-S) in mice. Intramuscularly inoculated mice were necropsied at different time points and examined by histology, immunohistochemistry and real-time PCR. We detected inflammation and myonecrosis at the parenteral site and hyperplasia of the draining lymph nodes. MVA-MERS-S did not result in detectable lesions in tissues peripheral to the parenteral site and draining lymph nodes. Real-time PCR analysis of >240 tissue samples detected MVA-DNA predominantly at the injection site and in the draining lymph nodes, and suggested continuous clearance of the candidate vaccine during the observation period. Levels of parenteral site inflammation and hyperplasia of draining lymph nodes were considered in line with immunological responses to vaccine inoculation.
Collapse
Affiliation(s)
- Martin C Langenmayer
- Institute for Infectious Diseases and Zoonoses, LMU Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Germany
| | - Anna-Theresa Lülf-Averhoff
- Institute for Infectious Diseases and Zoonoses, LMU Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Germany
| | | | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, LMU Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Germany
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, LMU Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Germany.
| | - Asisa Volz
- Institute for Infectious Diseases and Zoonoses, LMU Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Germany
| |
Collapse
|
35
|
Altenburg AF, van Trierum SE, de Bruin E, de Meulder D, van de Sandt CE, van der Klis FRM, Fouchier RAM, Koopmans MPG, Rimmelzwaan GF, de Vries RD. Effects of pre-existing orthopoxvirus-specific immunity on the performance of Modified Vaccinia virus Ankara-based influenza vaccines. Sci Rep 2018; 8:6474. [PMID: 29692427 PMCID: PMC5915537 DOI: 10.1038/s41598-018-24820-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/10/2018] [Indexed: 11/26/2022] Open
Abstract
The replication-deficient orthopoxvirus modified vaccinia virus Ankara (MVA) is a promising vaccine vector against various pathogens and has an excellent safety record. However, pre-existing vector-specific immunity is frequently suggested to be a drawback of MVA-based vaccines. To address this issue, mice were vaccinated with MVA-based influenza vaccines in the presence or absence of orthopoxvirus-specific immunity. Importantly, protective efficacy of an MVA-based influenza vaccine against a homologous challenge was not impaired in the presence of orthopoxvirus-specific pre-existing immunity. Nonetheless, orthopoxvirus-specific pre-existing immunity reduced the induction of antigen-specific antibodies under specific conditions and completely prevented induction of antigen-specific T cell responses by rMVA-based vaccination. Notably, antibodies induced by vaccinia virus vaccination, both in mice and humans, were not capable of neutralizing MVA. Thus, when using rMVA-based vaccines it is important to consider the main correlate of protection induced by the vaccine, the vaccine dose and the orthopoxvirus immune status of vaccine recipients.
Collapse
Affiliation(s)
- Arwen F Altenburg
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Stella E van Trierum
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Erwin de Bruin
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Dennis de Meulder
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Carolien E van de Sandt
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Fiona R M van der Klis
- Centre for Infectious Disease Control (Cib), National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Guus F Rimmelzwaan
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine, Hannover, Germany
| | - Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
36
|
Cooper CJ, Koonjan S, Nilsson AS. Enhancing Whole Phage Therapy and Their Derived Antimicrobial Enzymes through Complex Formulation. Pharmaceuticals (Basel) 2018; 11:ph11020034. [PMID: 29671806 PMCID: PMC6027540 DOI: 10.3390/ph11020034] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/11/2018] [Accepted: 04/17/2018] [Indexed: 12/11/2022] Open
Abstract
The resurgence of research into phage biology and therapy is, in part, due to the increasing need for novel agents to treat multidrug-resistant infections. Despite a long clinical history in Eastern Europe and initial success within the food industry, commercialized phage products have yet to enter other sectors. This relative lack of success is, in part, due to the inherent biological limitations of whole phages. These include (but are not limited to) reaching target sites at sufficiently high concentrations to establish an infection which produces enough progeny phages to reduce the bacterial population in a clinically meaningful manner and the limited host range of some phages. Conversely, parallels can be drawn between antimicrobial enzymes derived from phages and conventional antibiotics. In the current article the biological limitations of whole phage-based therapeutics and their derived antimicrobial enzymes will be discussed. In addition, the ability of more complex formulations to address these issues, in the context of medical and non-medical applications, will also be included.
Collapse
Affiliation(s)
- Callum J Cooper
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden.
| | - Shazeeda Koonjan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden.
| | - Anders S Nilsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden.
| |
Collapse
|
37
|
Swindle S. Biosafety in Handling Gene Transfer Vectors. ACTA ACUST UNITED AC 2018; 96:12.1.1-12.1.17. [DOI: 10.1002/cphg.54] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Scott Swindle
- Occupational Medicine and Research Safety, The University of Alabama at Birmingham Birmingham Alabama
| |
Collapse
|
38
|
Rey-Jurado E, Tapia F, Muñoz-Durango N, Lay MK, Carreño LJ, Riedel CA, Bueno SM, Genzel Y, Kalergis AM. Assessing the Importance of Domestic Vaccine Manufacturing Centers: An Overview of Immunization Programs, Vaccine Manufacture, and Distribution. Front Immunol 2018; 9:26. [PMID: 29403503 PMCID: PMC5778105 DOI: 10.3389/fimmu.2018.00026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 01/04/2018] [Indexed: 12/03/2022] Open
Abstract
Vaccines have significantly reduced the detrimental effects of numerous human infectious diseases worldwide, helped to reduce drastically child mortality rates and even achieved eradication of major pathogens, such as smallpox. These achievements have been possible due to a dedicated effort for vaccine research and development, as well as an effective transfer of these vaccines to public health care systems globally. Either public or private institutions have committed to developing and manufacturing vaccines for local or international population supply. However, current vaccine manufacturers worldwide might not be able to guarantee sufficient vaccine supplies for all nations when epidemics or pandemics events could take place. Currently, different countries produce their own vaccine supplies under Good Manufacturing Practices, which include the USA, Canada, China, India, some nations in Europe and South America, such as Germany, the Netherlands, Italy, France, Argentina, and Brazil, respectively. Here, we discuss some of the vaccine programs and manufacturing capacities, comparing the current models of vaccine management between industrialized and developing countries. Because local vaccine production undoubtedly provides significant benefits for the respective population, the manufacture capacity of these prophylactic products should be included in every country as a matter of national safety.
Collapse
Affiliation(s)
- Emma Rey-Jurado
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Tapia
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Natalia Muñoz-Durango
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
39
|
Okeke MI, Okoli AS, Diaz D, Offor C, Oludotun TG, Tryland M, Bøhn T, Moens U. Hazard Characterization of Modified Vaccinia Virus Ankara Vector: What Are the Knowledge Gaps? Viruses 2017; 9:v9110318. [PMID: 29109380 PMCID: PMC5707525 DOI: 10.3390/v9110318] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/21/2017] [Accepted: 10/26/2017] [Indexed: 12/17/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) is the vector of choice for human and veterinary applications due to its strong safety profile and immunogenicity in vivo. The use of MVA and MVA-vectored vaccines against human and animal diseases must comply with regulatory requirements as they pertain to environmental risk assessment, particularly the characterization of potential adverse effects to humans, animals and the environment. MVA and recombinant MVA are widely believed to pose low or negligible risk to ecosystem health. However, key aspects of MVA biology require further research in order to provide data needed to evaluate the potential risks that may occur due to the use of MVA and MVA-vectored vaccines. The purpose of this paper is to identify knowledge gaps in the biology of MVA and recombinant MVA that are of relevance to its hazard characterization and discuss ongoing and future experiments aimed at providing data necessary to fill in the knowledge gaps. In addition, we presented arguments for the inclusion of uncertainty analysis and experimental investigation of verifiable worst-case scenarios in the environmental risk assessment of MVA and recombinant MVA. These will contribute to improved risk assessment of MVA and recombinant MVA vaccines.
Collapse
Affiliation(s)
- Malachy I Okeke
- Genome Editing Research Group, GenØk-Center for Biosafety, Siva Innovation Center, N-9294 Tromso, Norway.
| | - Arinze S Okoli
- Genome Editing Research Group, GenØk-Center for Biosafety, Siva Innovation Center, N-9294 Tromso, Norway.
| | - Diana Diaz
- Molecular Inflammation Research Group, Institute of Medical Biology, University i Tromsø (UiT)-The Arctic University of Norway, N-9037 Tromso, Norway.
| | - Collins Offor
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences Piaristengasse 1, A-3500 Krems, Austria.
| | - Taiwo G Oludotun
- Department of Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences Piaristengasse 1, A-3500 Krems, Austria.
| | - Morten Tryland
- Genome Editing Research Group, GenØk-Center for Biosafety, Siva Innovation Center, N-9294 Tromso, Norway.
- Artic Infection Biology, Department of Artic and Marine Biology, UIT-The Artic University of Norway, N-9037 Tromso, Norway.
| | - Thomas Bøhn
- Genome Editing Research Group, GenØk-Center for Biosafety, Siva Innovation Center, N-9294 Tromso, Norway.
| | - Ugo Moens
- Molecular Inflammation Research Group, Institute of Medical Biology, University i Tromsø (UiT)-The Arctic University of Norway, N-9037 Tromso, Norway.
| |
Collapse
|
40
|
Exploiting 2A peptides to elicit potent neutralizing antibodies by a multi-subunit herpesvirus glycoprotein complex. J Virol Methods 2017; 251:30-37. [PMID: 28989096 DOI: 10.1016/j.jviromet.2017.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/22/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Neutralizing antibodies (NAb) interfering with glycoprotein complex-mediated virus entry into host cells are thought to contribute to the protection against herpesvirus infection. However, using herpesvirus glycoprotein complexes as vaccine antigens can be complicated by the necessity of expressing multiple subunits simultaneously to allow efficient complex assembly and formation of conformational NAb epitopes. By using a novel bacterial artificial chromosome (BAC) clone of the clinically deployable Modified Vaccinia Ankara (MVA) vector and exploiting ribosomal skipping mediated by 2A peptides, MVA vectors were generated that expressed self-processing subunits of the human cytomegalovirus (HCMV) pentamer complex (PC) composed of gH, gL, UL128, UL130, and UL131A. These MVA vectors expressed 2A-linked HCMV PC subunits that were efficiently cleaved and transported to the cell surface as protein complexes forming conformational neutralizing epitopes. In addition, vaccination of mice by only two immunizations with these MVA vectors resulted in potent HCMV NAb responses that remained stable over a period of at least six months. This method of eliciting NAb by 2A-linked, self-processing HCMV PC subunits could contribute to develop a HCMV vaccine candidate and may serve as a template to facilitate the development of subunit vaccine strategies against other herpesviruses.
Collapse
|
41
|
Perez EM, Foley J, Tison T, Silva R, Ogembo JG. Novel Epstein-Barr virus-like particles incorporating gH/gL-EBNA1 or gB-LMP2 induce high neutralizing antibody titers and EBV-specific T-cell responses in immunized mice. Oncotarget 2017; 8:19255-19273. [PMID: 27926486 PMCID: PMC5386682 DOI: 10.18632/oncotarget.13770] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/07/2016] [Indexed: 01/02/2023] Open
Abstract
Previous Epstein-Barr virus (EBV) prophylactic vaccines based on the major surface glycoprotein gp350/220 as an immunogen have failed to block viral infection in humans, suggesting a need to target other viral envelope glycoproteins. In this study, we reasoned that incorporating gH/gL or gB, critical glycoproteins for viral fusion and entry, on the surface of a virus-like particle (VLP) would be more immunogenic than gp350/220 for generating effective neutralizing antibodies to prevent viral infection of both epithelial and B cell lines. To boost the humoral response and trigger cell-mediated immunity, EBV nuclear antigen 1 (EBNA1) and latent membrane protein 2 (LMP2), intracellular latency proteins expressed in all EBV-infected cells, were also included as critical components of the polyvalent EBV VLP. gH/gL-EBNA1 and gB-LMP2 VLPs were efficiently produced in Chinese hamster ovary cells, an FDA-approved vehicle for mass-production of biologics. Immunization with gH/gL-EBNA1 and gB-LMP2 VLPs without adjuvant generated both high neutralizing antibody titers in vitro and EBV-specific T-cell responses in BALB/c mice. These data demonstrate that EBV glycoprotein(s)-based VLPs have excellent immunogenicity, and represent a potentially safe vaccine that will be invaluable not only in preventing EBV infection, but importantly, in preventing and treating the 200,000 cases of EBV-associated cancers that occur globally every year.
Collapse
Affiliation(s)
- Elizabeth M Perez
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Joslyn Foley
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Timelia Tison
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Rute Silva
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Javier Gordon Ogembo
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA, USA
| |
Collapse
|
42
|
Tapia F, Jordan I, Genzel Y, Reichl U. Efficient and stable production of Modified Vaccinia Ankara virus in two-stage semi-continuous and in continuous stirred tank cultivation systems. PLoS One 2017; 12:e0182553. [PMID: 28837572 PMCID: PMC5570375 DOI: 10.1371/journal.pone.0182553] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 07/19/2017] [Indexed: 11/18/2022] Open
Abstract
One important aim in cell culture-based viral vaccine and vector production is the implementation of continuous processes. Such a development has the potential to reduce costs of vaccine manufacturing as volumetric productivity is increased and the manufacturing footprint is reduced. In this work, continuous production of Modified Vaccinia Ankara (MVA) virus was investigated. First, a semi-continuous two-stage cultivation system consisting of two shaker flasks in series was established as a small-scale approach. Cultures of the avian AGE1.CR.pIX cell line were expanded in the first shaker, and MVA virus was propagated and harvested in the second shaker over a period of 8-15 days. A total of nine small-scale cultivations were performed to investigate the impact of process parameters on virus yields. Harvest volumes of 0.7-1 L with maximum TCID50 titers of up to 1.0×109 virions/mL were obtained. Genetic analysis of control experiments using a recombinant MVA virus containing green-fluorescent-protein suggested that the virus was stable over at least 16 d of cultivation. In addition, a decrease or fluctuation of infectious units that may indicate an excessive accumulation of defective interfering particles was not observed. The process was automated in a two-stage continuous system comprising two connected 1 L stirred tank bioreactors. Stable MVA virus titers, and a total production volume of 7.1 L with an average TCID50 titer of 9×107 virions/mL was achieved. Because titers were at the lower range of the shake flask cultivations potential for further process optimization at large scale will be discussed. Overall, MVA virus was efficiently produced in continuous and semi-continuous cultivations making two-stage stirred tank bioreactor systems a promising platform for industrial production of MVA-derived recombinant vaccines and viral vectors.
Collapse
Affiliation(s)
- Felipe Tapia
- International Max Planck Research School for Advanced Methods in Process and Systems Engineering, Magdeburg, Germany
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | | | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- Bioprocess Engineering, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
43
|
Modified Vaccinia Virus Ankara Preferentially Targets Antigen Presenting Cells In Vitro, Ex Vivo and In Vivo. Sci Rep 2017; 7:8580. [PMID: 28819261 PMCID: PMC5561217 DOI: 10.1038/s41598-017-08719-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/13/2017] [Indexed: 11/09/2022] Open
Abstract
Modified Vaccinia virus Ankara (MVA) is a promising vaccine vector with an excellent safety profile. However, despite extensive pre-clinical and clinical testing, surprisingly little is known about the cellular tropism of MVA, especially in relevant animal species. Here, we performed in vitro, ex vivo and in vivo experiments with recombinant MVA expressing green fluorescent protein (rMVA-GFP). In both human peripheral blood mononuclear cells and mouse lung explants, rMVA-GFP predominantly infected antigen presenting cells. Subsequent in vivo experiments performed in mice, ferrets and non-human primates indicated that preferential targeting of dendritic cells and alveolar macrophages was observed after respiratory administration, although subtle differences were observed between the respective animal species. Following intramuscular injection, rMVA-GFP was detected in interdigitating cells between myocytes, but also in myocytes themselves. These data are important in advancing our understanding of the basis for the immunogenicity of MVA-based vaccines and aid rational vaccine design and delivery strategies.
Collapse
|
44
|
Altenburg AF, Magnusson SE, Bosman F, Stertman L, de Vries RD, Rimmelzwaan GF. Protein and modified vaccinia virus Ankara-based influenza virus nucleoprotein vaccines are differentially immunogenic in BALB/c mice. Clin Exp Immunol 2017; 190:19-28. [PMID: 28665497 DOI: 10.1111/cei.13004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2017] [Indexed: 02/06/2023] Open
Abstract
Because of the high variability of seasonal influenza viruses and the eminent threat of influenza viruses with pandemic potential, there is great interest in the development of vaccines that induce broadly protective immunity. Most probably, broadly protective influenza vaccines are based on conserved proteins, such as nucleoprotein (NP). NP is a vaccine target of interest as it has been shown to induce cross-reactive antibody and T cell responses. Here we tested and compared various NP-based vaccine preparations for their capacity to induce humoral and cellular immune responses to influenza virus NP. The immunogenicity of protein-based vaccine preparations with Matrix-M™ adjuvant as well as recombinant viral vaccine vector modified Vaccinia virus Ankara (MVA) expressing the influenza virus NP gene, with or without modifications that aim at optimization of CD8+ T cell responses, was addressed in BALB/c mice. Addition of Matrix-M™ adjuvant to NP wild-type protein-based vaccines significantly improved T cell responses. Furthermore, recombinant MVA expressing the influenza virus NP induced strong antibody and CD8+ T cell responses, which could not be improved further by modifications of NP to increase antigen processing and presentation.
Collapse
Affiliation(s)
- A F Altenburg
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, the Netherlands
| | | | - F Bosman
- AmatsiQ-Biologicals, Ghent, Belgium
| | | | - R D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - G F Rimmelzwaan
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
45
|
Wingerath J, Ostroumov D, Woller N, Manns MP, Pinschewer DD, Orlinger K, Berka U, Kühnel F, Wirth TC. Recombinant LCMV Vectors Induce Protective Immunity following Homologous and Heterologous Vaccinations. Mol Ther 2017; 25:2533-2545. [PMID: 28803862 DOI: 10.1016/j.ymthe.2017.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/11/2017] [Accepted: 07/16/2017] [Indexed: 01/15/2023] Open
Abstract
Successful vaccination against cancer and infectious diseases relies on the induction of adaptive immune responses that induce high-titer antibodies or potent cytoxic T cell responses. In contrast to humoral vaccines, the amplification of cellular immune responses is often hampered by anti-vector immunity that either pre-exists or develops after repeated homologous vaccination. Replication-defective lymphocytic choriomeningitis virus (LCMV) vectors represent a novel generation of vaccination vectors that induce potent immune responses while escaping recognition by neutralizing antibodies. Here, we characterize the CD8 T cell immune response induced by replication-defective recombinant LCMV (rLCMV) vectors with regard to expansion kinetics, trafficking, phenotype, and function and we perform head-to-head comparisons of the novel rLCMV vectors with established vectors derived from adenovirus, vaccinia virus, or Listeria monocytogenes. Our results demonstrate that replication-deficient rLCMV vectors are safe and ideally suited for both homologous and heterologous vaccination regimens to achieve optimal amplification of CD8 T cell immune responses in vivo.
Collapse
Affiliation(s)
- Jessica Wingerath
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Dmitrij Ostroumov
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Norman Woller
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Daniel D Pinschewer
- Division of Experimental Virology, Department of Biomedicine, University of Basel, 4051 Basel, Switzerland
| | - Klaus Orlinger
- Hookipa Biotech AG, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Ursula Berka
- Hookipa Biotech AG, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Thomas C Wirth
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
46
|
Olbert M, Römer-Oberdörfer A, Herden C, Malberg S, Runge S, Staeheli P, Rubbenstroth D. Viral vector vaccines expressing nucleoprotein and phosphoprotein genes of avian bornaviruses ameliorate homologous challenge infections in cockatiels and common canaries. Sci Rep 2016; 6:36840. [PMID: 27830736 PMCID: PMC5103271 DOI: 10.1038/srep36840] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/17/2016] [Indexed: 12/05/2022] Open
Abstract
Avian bornaviruses are causative agents of proventricular dilatation disease (PDD), an often fatal disease of parrots and related species (order Psittaciformes) which is widely distributed in captive psittacine populations and may affect endangered species. Here, we established a vaccination strategy employing two different well described viral vectors, namely recombinant Newcastle disease virus (NDV) and modified vaccinia virus Ankara (MVA) that were engineered to express the phosphoprotein and nucleoprotein genes of two avian bornaviruses, parrot bornavirus 4 (PaBV-4) and canary bornavirus 2 (CnBV-2). When combined in a heterologous prime/boost vaccination regime, NDV and MVA vaccine viruses established self-limiting infections and induced a bornavirus-specific humoral immune response in cockatiels (Nymphicus hollandicus) and common canaries (Serinus canaria forma domestica). After challenge infection with a homologous bornavirus, shedding of bornavirus RNA and viral loads in tissue samples were significantly reduced in immunized birds, indicating that vaccination markedly delayed the course of infection. However, cockatiels still developed signs of PDD if the vaccine failed to prevent viral persistence. Our work demonstrates that avian bornavirus infections can be repressed by vaccine-induced immunity. It represents a first crucial step towards a protective vaccination strategy to combat PDD in psittacine birds.
Collapse
Affiliation(s)
- Marita Olbert
- Institute for Virology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 11, D-79104 Freiburg, Germany
| | - Angela Römer-Oberdörfer
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Südufer 10, D-17493 Greifswald – Insel Riems, Germany
| | - Christiane Herden
- Institute for Veterinary Pathology, University Justus Liebig Gießen, Frankfurter Str. 96, D-35392 Gießen, Germany
| | - Sara Malberg
- Institute for Veterinary Pathology, University Justus Liebig Gießen, Frankfurter Str. 96, D-35392 Gießen, Germany
| | - Solveig Runge
- Institute for Virology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 11, D-79104 Freiburg, Germany
| | - Peter Staeheli
- Institute for Virology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 11, D-79104 Freiburg, Germany
| | - Dennis Rubbenstroth
- Institute for Virology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 11, D-79104 Freiburg, Germany
| |
Collapse
|
47
|
A MVA construct expressing a secretable form of the Dengue virus 3 envelope protein protects immunized mice from dengue-induced encephalitis. Vaccine 2016; 34:6120-6122. [PMID: 27817963 DOI: 10.1016/j.vaccine.2016.10.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/13/2016] [Accepted: 10/19/2016] [Indexed: 11/22/2022]
Abstract
Dengue is no longer restricted to tropical developing countries, but is now a major global public health problem. Despite the recent license approval of the CYD-TDV vaccine in some countries, efforts to develop a more efficient vaccine against Dengue virus (DENV) continue. Herein, we evaluate the immunogenicity and level of protection of two potential vaccines against DENV based on recombinant modified vaccinia virus Ankara (rMVA). The vaccine addressing the Envelope protein from DENV serotype 3 to the endoplasmic reticulum elicited neutralizing antibodies titers which correlate with protection, and also confers protection upon challenge in a mouse model. Our results support the development of a tetravalent dengue vaccine with the further construction of rMVAs expressing proteins from the other DENV serotypes.
Collapse
|
48
|
Vector-based genetically modified vaccines: Exploiting Jenner's legacy. Vaccine 2016; 34:6436-6448. [PMID: 28029542 PMCID: PMC7115478 DOI: 10.1016/j.vaccine.2016.06.059] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/02/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022]
Abstract
The global vaccine market is diverse while facing a plethora of novel developments. Genetic modification (GM) techniques facilitate the design of ’smarter’ vaccines. For many of the major infectious diseases of humans, like AIDS and malaria, but also for most human neoplastic disorders, still no vaccines are available. It may be speculated that novel GM technologies will significantly contribute to their development. While a promising number of studies is conducted on GM vaccines and GM vaccine technologies, the contribution of GM technology to newly introduced vaccines on the market is disappointingly limited. In this study, the field of vector-based GM vaccines is explored. Data on currently available, actually applied, and newly developed vectors is retrieved from various sources, synthesised and analysed, in order to provide an overview on the use of vector-based technology in the field of GM vaccine development. While still there are only two vector-based vaccines on the human vaccine market, there is ample activity in the fields of patenting, preclinical research, and different stages of clinical research. Results of this study revealed that vector-based vaccines comprise a significant part of all GM vaccines in the pipeline. This study further highlights that poxviruses and adenoviruses are among the most prominent vectors in GM vaccine development. After the approval of the first vectored human vaccine, based on a flavivirus vector, vaccine vector technology, especially based on poxviruses and adenoviruses, holds great promise for future vaccine development. It may lead to cheaper methods for the production of safe vaccines against diseases for which no or less perfect vaccines exist today, thus catering for an unmet medical need. After the introduction of Jenner’s vaccinia virus as the first vaccine more than two centuries ago, which eventually led to the recent eradication of smallpox, this and other viruses may now be the basis for constructing vectors that may help us control other major scourges of mankind.
Collapse
|
49
|
Léon A, David AL, Madeline B, Guianvarc'h L, Dureau E, Champion-Arnaud P, Hebben M, Huss T, Chatrenet B, Schwamborn K. The EB66® cell line as a valuable cell substrate for MVA-based vaccines production. Vaccine 2016; 34:5878-5885. [PMID: 27997338 DOI: 10.1016/j.vaccine.2016.10.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/15/2016] [Accepted: 10/17/2016] [Indexed: 11/17/2022]
Abstract
The selection of a cell substrate is a critical step for the development and manufacturing of a viral vaccine candidate. Several parameters such as cell susceptibility and permissiveness to the viral pathogens but also performance in terms of viral antigens quality and production yields are important considerations when identifying the ideal match between a viral vaccine and cell substrate. The modified vaccinia virus Ankara (MVA) is a replication-deficient viral vector that holds great promise as a vaccine platform, however only limited cell substrates have been tested or are available for industrialization. Here we evaluate the duck embryo-derived EB66® cell line as potential cell substrate for MVA production. To this end, we used two recombinant MVA constructs and demonstrated that EB66® cells are propagating the tested MVA viruses very efficiently, while preserving viral attenuation and transgene expression for up to 20 serial passages. Furthermore we developed upstream and downstream processes that enable industrialization of the virus production. In conclusion, we showed that EB66® cells can be used as potent cell substrate for MVA-based vaccines and represent therefore an attractive alternative for vaccine production.
Collapse
Affiliation(s)
- Arnaud Léon
- Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France.
| | | | - Brice Madeline
- Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France
| | | | - Elodie Dureau
- Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France
| | | | - Matthias Hebben
- Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France
| | - Thierry Huss
- Transgene SA, 400 boulevard Gonthier d'Andernach - Parc d'Innovation - CS80166, 67405 Illkirch Graffenstaden cedex, France
| | | | | |
Collapse
|
50
|
Fernández-Escobar M, Baldanta S, Reyburn H, Guerra S. Use of functional genomics to understand replication deficient poxvirus-host interactions. Virus Res 2016; 216:1-15. [PMID: 26519757 DOI: 10.1016/j.virusres.2015.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 10/22/2022]
Abstract
High-throughput genomics technologies are currently being used to study a wide variety of viral infections, providing insight into which cellular genes and pathways are regulated after infection, and how these changes are related, or not, to efficient elimination of the pathogen. This article will focus on how gene expression studies of infections with non-replicative poxviruses currently used as vaccine vectors provide a global perspective of the molecular events associated with the viral infection in human cells. These high-throughput genomics approaches have the potential to lead to the identification of specific new properties of the viral vector or novel cellular targets that may aid in the development of more effective pox-derived vaccines and antivirals.
Collapse
Affiliation(s)
- Mercedes Fernández-Escobar
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma, E-28029 Madrid, Spain
| | - Sara Baldanta
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma, E-28029 Madrid, Spain
| | - Hugh Reyburn
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Campus Universidad Autónoma, E-28049 Madrid, Spain
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma, E-28029 Madrid, Spain.
| |
Collapse
|