Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(23): e2407437121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38814864

ABSTRACT

The accessory protease transmembrane protease serine 2 (TMPRSS2) enhances severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uptake into ACE2-expressing cells, although how increased entry impacts downstream viral and host processes remains unclear. To investigate this in more detail, we performed infection assays in engineered cells promoting ACE2-mediated entry with and without TMPRSS2 coexpression. Electron microscopy and inhibitor experiments indicated TMPRSS2-mediated cell entry was associated with increased virion internalization into endosomes, and partially dependent upon clathrin-mediated endocytosis. TMPRSS2 increased panvariant uptake efficiency and enhanced early rates of virus replication, transcription, and secretion, with variant-specific profiles observed. On the host side, transcriptional profiling confirmed the magnitude of infection-induced antiviral and proinflammatory responses were linked to uptake efficiency, with TMPRSS2-assisted entry boosting early antiviral responses. In addition, TMPRSS2-enhanced infections increased rates of cytopathology, apoptosis, and necrosis and modulated virus secretion kinetics in a variant-specific manner. On the virus side, convergent signatures of cell-uptake-dependent innate immune induction were recorded in viral genomes, manifesting as switches in dominant coupled Nsp3 residues whose frequencies were correlated to the magnitude of the cellular response to infection. Experimentally, we demonstrated that selected Nsp3 mutations conferred enhanced interferon antagonism. More broadly, we show that TMPRSS2 orthologues from evolutionarily diverse mammals facilitate panvariant enhancement of cell uptake. In summary, our study uncovers previously unreported associations, linking cell entry efficiency to innate immune activation kinetics, cell death rates, virus secretion dynamics, and convergent selection of viral mutations. These data expand our understanding of TMPRSS2's role in the SARS-CoV-2 life cycle and confirm its broader significance in zoonotic reservoirs and animal models.


Subject(s)
COVID-19 , Immunity, Innate , SARS-CoV-2 , Serine Endopeptidases , Virus Internalization , SARS-CoV-2/immunology , SARS-CoV-2/physiology , SARS-CoV-2/metabolism , Humans , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , COVID-19/virology , COVID-19/immunology , COVID-19/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Virus Replication , Animals , Endocytosis , HEK293 Cells , Chlorocebus aethiops , Cytology
2.
Hum Gene Ther ; 34(17-18): 896-904, 2023 09.
Article in English | MEDLINE | ID: mdl-37639360

ABSTRACT

The development of prophylatic or therapeutic medicines for infectious diseases is one of the priorities for health organizations worldwide. Innovative solutions are required to achieve effective, safe, and accessible treatments for most if not all infectious diseases, particularly those that are chronic in nature or that emerge unexpectedly over time. Genetic technologies offer versatile possibilities to design therapies against pathogens. Recent developments such as mRNA vaccines, CRISPR gene editing, and immunotherapies provide unprecedented hope to achieve significant results in the field of infectious diseases. This review will focus on advances in this domain, showcasing the cross-fertilization with other fields (e.g., oncology), and addressing some of the logistical and economic concerns important to consider when making these advances accessible to diverse populations around the world.


Subject(s)
Communicable Diseases , Humans , Communicable Diseases/genetics , Communicable Diseases/therapy , Genetic Therapy , Vaccination , Cloning, Molecular , Clustered Regularly Interspaced Short Palindromic Repeats
3.
Nat Microbiol ; 8(5): 860-874, 2023 05.
Article in English | MEDLINE | ID: mdl-37012419

ABSTRACT

Vaccines play a critical role in combating the COVID-19 pandemic. Future control of the pandemic requires improved vaccines with high efficacy against newly emerging SARS-CoV-2 variants and the ability to reduce virus transmission. Here we compare immune responses and preclinical efficacy of the mRNA vaccine BNT162b2, the adenovirus-vectored spike vaccine Ad2-spike and the live-attenuated virus vaccine candidate sCPD9 in Syrian hamsters, using both homogeneous and heterologous vaccination regimens. Comparative vaccine efficacy was assessed by employing readouts from virus titrations to single-cell RNA sequencing. Our results show that sCPD9 vaccination elicited the most robust immunity, including rapid viral clearance, reduced tissue damage, fast differentiation of pre-plasmablasts, strong systemic and mucosal humoral responses, and rapid recall of memory T cells from lung tissue after challenge with heterologous SARS-CoV-2. Overall, our results demonstrate that live-attenuated vaccines offer advantages over currently available COVID-19 vaccines.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Cricetinae , Humans , Vaccines, Attenuated , COVID-19/prevention & control , COVID-19 Vaccines , BNT162 Vaccine , Pandemics , Mesocricetus
4.
NPJ Vaccines ; 8(1): 46, 2023 Mar 24.
Article in English | MEDLINE | ID: mdl-36964176

ABSTRACT

A novel Influenza A virus (subtype H7N9) emerged in spring 2013 and caused considerable mortality in zoonotically infected patients. To be prepared for potential pandemics, broadly effective and safe vaccines are crucial. Recombinant measles virus (MeV) encoding antigens of foreign pathogens constitutes a promising vector platform to generate novel vaccines. To characterize the efficacy of H7N9 antigens in a prototypic vaccine platform technology, we generated MeVs encoding either neuraminidase (N9) or hemagglutinin (H7). Moraten vaccine strain-derived vaccine candidates were rescued; they replicated with efficiency comparable to that of the measles vaccine, robustly expressed H7 and N9, and were genetically stable over 10 passages. Immunization of MeV-susceptible mice triggered the production of antibodies against H7 and N9, including hemagglutination-inhibiting and neutralizing antibodies induced by MVvac2-H7(P) and neuraminidase-inhibiting antibodies by MVvac2-N9(P). Vaccinated mice also developed long-lasting H7- and N9-specific T cells. Both MVvac2-H7(P) and MVvac2-N9(P)-vaccinated mice were protected from lethal H7N9 challenge.

5.
STAR Protoc ; 4(1): 101957, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36542521

ABSTRACT

In infectious disease research, single-cell RNA sequencing allows dissection of host-pathogen interactions. As a prerequisite, we provide a protocol to transform solid and complex organs such as lungs into representative diverse, viable single-cell suspensions. Our protocol describes performance of vascular perfusion, pneumonectomy, enzymatic digestion, and mechanical dissociation of lung tissue, as well as red blood cell lysis and counting of isolated cells. A challenge remains, however, to further increase the proportion of pulmonary endothelial cells without compromising on viability. For complete details on the use and execution of this protocol, please refer to Nouailles et al. (2021),1 Wyler et al. (2022),2 and Ebenig et al. (2022).3.


Subject(s)
Endothelial Cells , Single-Cell Gene Expression Analysis , Cricetinae , Animals , Mice , Cell Death , Dissection , Lung
6.
NPJ Vaccines ; 7(1): 119, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36243743

ABSTRACT

Live-attenuated measles virus (MeV) has been extraordinarily effective in preventing measles infections and their often deadly sequelae, accompanied by remarkable safety and stability since their first licensing in 1963. The advent of recombinant DNA technologies, combined with systems to generate infectious negative-strand RNA viruses on the basis of viral genomes encoded on plasmid DNA in the 1990s, paved the way to generate recombinant, vaccine strain-derived MeVs. These live-attenuated vaccine constructs can encode and express additional foreign antigens during transient virus replication following immunization. Effective humoral and cellular immune responses are induced not only against the MeV vector, but also against the foreign antigen cargo in immunized individuals, which can protect against the associated pathogen. This review aims to present an overview of the versatility of this vaccine vector as platform technology to target various diseases, as well as current research and developmental stages, with one vaccine candidate ready to enter phase III clinical trials to gain marketing authorization, MV-CHIK.

7.
Cell Rep ; 40(7): 111214, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35952673

ABSTRACT

Vaccine-associated enhanced respiratory disease (VAERD) is a severe complication for some respiratory infections. To investigate the potential for VAERD induction in coronavirus disease 2019 (COVID-19), we evaluate two vaccine leads utilizing a severe hamster infection model: a T helper type 1 (TH1)-biased measles vaccine-derived candidate and a TH2-biased alum-adjuvanted, non-stabilized spike protein. The measles virus (MeV)-derived vaccine protects the animals, but the protein lead induces VAERD, which can be alleviated by dexamethasone treatment. Bulk transcriptomic analysis reveals that our protein vaccine prepares enhanced host gene dysregulation in the lung, exclusively up-regulating mRNAs encoding the eosinophil attractant CCL-11, TH2-driving interleukin (IL)-19, or TH2 cytokines IL-4, IL-5, and IL-13. Single-cell RNA sequencing (scRNA-seq) identifies lung macrophages or lymphoid cells as sources, respectively. Our findings imply that VAERD is caused by the concerted action of hyperstimulated macrophages and TH2 cytokine-secreting lymphoid cells and potentially links VAERD to antibody-dependent enhancement (ADE). In summary, we identify the cytokine drivers and cellular contributors that mediate VAERD after TH2-biased vaccination.


Subject(s)
COVID-19 , Vaccines , Animals , Antibodies, Viral , Cricetinae , Cytokines/metabolism , Immunization , Lung/pathology , Mice , Mice, Inbred BALB C , Th1 Cells , Th2 Cells , Vaccination
8.
J Gen Virol ; 103(11)2022 11.
Article in English | MEDLINE | ID: mdl-36748683

ABSTRACT

The generation of recombinant measles virus (MeV) from manipulated genomes on plasmid DNA is quite a complex and inefficient process. As a member of the order Mononegavirales its single-stranded ssRNA genome in negative sense orientation is not infectious, but requires co-availability of the viral RNA-dependent RNA polymerase L, the polymerase co-factor phosphoprotein P, and the nucleocapsid protein N in defined relative amounts to establish infectious centres in transfected cell cultures that release replication-competent recombinant MeV particles. For this so-called rescue, different rescue systems were developed that rely on at least four different components. In this work, we establish a functional MeV rescue system just being composed of two components: the plasmid encoding the (modified) viral genome, and a one-helper-plasmid bundling all helper functions. In contrast to a rescue-system for Newcastle Disease Virus, another paramyxovirus, co-expression of all helper proteins by the same promoter failed. Instead, adaptation of the strength of the respective promoters to drive each helper gene´s expression to the relative expression found in MeV-infected cells or other rescue systems, which indeed adjusted respective mRNA and protein expression, yielded success, albeit not yet to the same efficacy as the four-component system. Thereby, our study paves the way for the development of easier and, after further optimization, more efficient rescue systems to generate recombinant MeV for e.g. the application as a vaccine platform or oncolytic virus, for example.


Subject(s)
Measles virus , Virus Replication , Animals , Measles virus/genetics , Transfection , Plasmids/genetics , Virus Replication/genetics , RNA, Viral/genetics , Genome, Viral
9.
Viruses ; 13(8)2021 07 21.
Article in English | MEDLINE | ID: mdl-34452286

ABSTRACT

Virotherapy research involves the development, exploration, and application of oncolytic viruses that combine direct killing of cancer cells by viral infection, replication, and spread (oncolysis) with indirect killing by induction of anti-tumor immune responses. Oncolytic viruses can also be engineered to genetically deliver therapeutic proteins for direct or indirect cancer cell killing. In this review-as part of the special edition on "State-of-the-Art Viral Vector Gene Therapy in Germany"-the German community of virotherapists provides an overview of their recent research activities that cover endeavors from screening and engineering viruses as oncolytic cancer therapeutics to their clinical translation in investigator-initiated and sponsored multi-center trials. Preclinical research explores multiple viral platforms, including new isolates, serotypes, or fitness mutants, and pursues unique approaches to engineer them towards increased safety, shielded or targeted delivery, selective or enhanced replication, improved immune activation, delivery of therapeutic proteins or RNA, and redirecting antiviral immunity for cancer cell killing. Moreover, several oncolytic virus-based combination therapies are under investigation. Clinical trials in Germany explore the safety and potency of virotherapeutics based on parvo-, vaccinia, herpes, measles, reo-, adeno-, vesicular stomatitis, and coxsackie viruses, including viruses encoding therapeutic proteins or combinations with immune checkpoint inhibitors. These research advances represent exciting vantage points for future endeavors of the German virotherapy community collectively aimed at the implementation of effective virotherapeutics in clinical oncology.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Clinical Trials as Topic , Genetic Engineering , Germany , Humans , Oncolytic Viruses/genetics
10.
J Gen Virol ; 102(4)2021 04.
Article in English | MEDLINE | ID: mdl-33830908

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection has caused a pandemic with tens of millions of cases and more than a million deaths. The infection causes COVID-19, a disease of the respiratory system of divergent severity. No treatment exists. Epigallocatechin-3-gallate (EGCG), the major component of green tea, has several beneficial properties, including antiviral activities. Therefore, we examined whether EGCG has antiviral activity against SARS-CoV-2. EGCG blocked not only the entry of SARS-CoV-2, but also MERS- and SARS-CoV pseudotyped lentiviral vectors and inhibited virus infections in vitro. Mechanistically, inhibition of the SARS-CoV-2 spike-receptor interaction was observed. Thus, EGCG might be suitable for use as a lead structure to develop more effective anti-COVID-19 drugs.


Subject(s)
Antiviral Agents/pharmacology , Catechin/analogs & derivatives , SARS-CoV-2/drug effects , Tea/chemistry , Animals , Betacoronavirus/drug effects , Betacoronavirus/physiology , COVID-19/prevention & control , COVID-19/virology , Catechin/pharmacology , Cell Survival/drug effects , Chlorocebus aethiops , HEK293 Cells , Humans , Lentivirus/drug effects , Lentivirus/genetics , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/genetics , Vero Cells , Virus Attachment/drug effects , Virus Replication/drug effects
11.
Proc Natl Acad Sci U S A ; 117(51): 32657-32666, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33257540

ABSTRACT

The COVID-19 pandemic is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and has spread worldwide, with millions of cases and more than 1 million deaths to date. The gravity of the situation mandates accelerated efforts to identify safe and effective vaccines. Here, we generated measles virus (MeV)-based vaccine candidates expressing the SARS-CoV-2 spike glycoprotein (S). Insertion of the full-length S protein gene in two different MeV genomic positions resulted in modulated S protein expression. The variant with lower S protein expression levels was genetically stable and induced high levels of effective Th1-biased antibody and T cell responses in mice after two immunizations. In addition to neutralizing IgG antibody responses in a protective range, multifunctional CD8+ and CD4+ T cell responses with S protein-specific killing activity were detected. Upon challenge using a mouse-adapted SARS-CoV-2, virus loads in vaccinated mice were significantly lower, while vaccinated Syrian hamsters revealed protection in a harsh challenge setup using an early-passage human patient isolate. These results are highly encouraging and support further development of MeV-based COVID-19 vaccines.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/prevention & control , Measles virus/immunology , SARS-CoV-2/immunology , Th1 Cells/immunology , Animals , Antibodies, Viral/immunology , COVID-19/epidemiology , COVID-19/immunology , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/genetics , Humans , Measles Vaccine/genetics , Measles Vaccine/immunology , Measles virus/genetics , Mice , Pandemics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , T-Lymphocytes/immunology
12.
Science ; 370(6513): 203-208, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32817270

ABSTRACT

The spike protein (S) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is required for cell entry and is the primary focus for vaccine development. In this study, we combined cryo-electron tomography, subtomogram averaging, and molecular dynamics simulations to structurally analyze S in situ. Compared with the recombinant S, the viral S was more heavily glycosylated and occurred mostly in the closed prefusion conformation. We show that the stalk domain of S contains three hinges, giving the head unexpected orientational freedom. We propose that the hinges allow S to scan the host cell surface, shielded from antibodies by an extensive glycan coat. The structure of native S contributes to our understanding of SARS-CoV-2 infection and potentially to the development of safe vaccines.


Subject(s)
Betacoronavirus/chemistry , Molecular Dynamics Simulation , Spike Glycoprotein, Coronavirus/chemistry , Cryoelectron Microscopy , Electron Microscope Tomography , Glycosylation , Humans , Protein Domains , Protein Multimerization , SARS-CoV-2
13.
Curr Opin Virol ; 41: 85-97, 2020 04.
Article in English | MEDLINE | ID: mdl-32861945

ABSTRACT

Over the last years, the development of viruses to treat cancer patients has re-gained considerable attention. A genetically modified herpesvirus, Talimogene laherparepvec, has already been authorized for the treatment of melanoma patients. Also recombinant measles virus (MeV) is developed as an oncolytic virus. Because of its high genetic flexibility, a number of different MeV strains have been the basis for the generation of targeted, armed, or shielded viruses that are highly specific for a given tumor target, more effective, or protected against serum neutralization. Such MeV have been extensively tested in vitro and in vivo, whereby remarkable oncolytic potency is accompanied by safety also in non-human primates. Therefore, MeV has been introduced into 19 different clinical trials and has reached phase II against two different tumor entities, multiple myeloma and ovarian carcinoma. Remarkably, one patient with advanced stage myeloma experienced long-term remission after treatment, visualizing the potency of this approach.


Subject(s)
Measles virus/physiology , Melanoma/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Animals , Humans , Measles virus/genetics , Melanoma/virology , Oncolytic Viruses/genetics
14.
Methods Mol Biol ; 2058: 51-75, 2020.
Article in English | MEDLINE | ID: mdl-31486031

ABSTRACT

This chapter describes the development of recombinant oncolytic measles viruses (MeV) that selectively enter and destroy tumor cells. The envelope of MeV is a favorable targeting substrate because receptor attachment and membrane fusion functions are separated on two proteins: the hemagglutinin (H) that binds receptors, and the fusion (F) protein that fuses the viral envelope with the cell membrane. The cell entry process, which depends on receptor recognition and occurs at the plasma membrane at neutral pH, results in the delivery of encapsidated genomes to the cytoplasm, where they replicate. Towards improving cancer specificity of oncolytic MeV, two types of cell entry targeting have been achieved. First, entry has been redirected through cancer-specific cell surface proteins. This was done by displaying specificity domains on H while also ablating binding to its natural receptors. Second, activation of the F protein was made dependent on secreted cancer proteases, while also interfering with F cleavage/activation by a ubiquitous intracellular protease. This chapter describes how entry-targeted MeV are produced: In short, gene cassettes with modified H or F coding regions are generated, and then introduced into the viral genome available on plasmid DNA. Such full-length genome plasmids are transfected in cell lines expressing, stably or transiently, the three viral proteins necessary for genome replication. Infectious centers form among these "rescue" cells, which allow isolation of clonal recombinant viruses. These are amplified, characterized in vitro, and then evaluated for their oncolytic activity in appropriate preclinical animal models.


Subject(s)
Genetic Engineering , Genetic Vectors/genetics , Measles virus/genetics , Oncolytic Viruses/genetics , Animals , Chlorocebus aethiops , Cloning, Molecular , Gene Expression , Gene Order , Genetic Engineering/methods , HEK293 Cells , Humans , Mice , Plasmids/genetics , Receptors, Virus/metabolism , Vero Cells , Virus Attachment , Virus Internalization
15.
Mol Ther Oncolytics ; 15: 186-200, 2019 Dec 20.
Article in English | MEDLINE | ID: mdl-31788553

ABSTRACT

Measles virus (MeV) is naturally cytolytic by extensive cell-to-cell fusion. Vaccine-derived MeV is toxic for cancer cells and is clinically tested as oncolytic virus. To combine the potential of MeV with enhanced safety, different targeting strategies have been described. We generated a receptor-targeted MeV by using receptor-blind viral attachment protein genetically fused to designed ankyrin repeat protein (DARPin) binding domains specific for the epidermal growth factor receptor (EGFR). To reduce on-target toxicity for EGFR+ healthy cells, we used an engineered viral fusion protein activatable by tumor-associated matrix metalloproteases (MMPs) for additional protease targeting. The dual-targeted virus replicated exclusively on EGFR+/MMP+ tumor cells but was safe on healthy EGFR+ target cells, primary human keratinocytes. Nevertheless, glioblastoma and other tumor cells were efficiently killed by all targeted viruses, although replication and oncolysis were slower for protease-targeted MeV. In vivo, efficacy of EGFR-targeted MeV was virtually unimpaired, whereas also dual-targeted MeV showed significant intra-tumoral spread and efficacy and could be armed with a prodrug convertase. The use of DARPin-domains resulted in potent EGFR-targeted MeV and for the first time effective dual retargeting of an oncolytic virus, further enhancing tumor selectivity. Together with powerful cell-toxic genes, the application as highly tumor-specific platform is promising.

16.
Mol Ther Oncolytics ; 12: 147-161, 2019 Mar 29.
Article in English | MEDLINE | ID: mdl-30775418

ABSTRACT

Glioblastoma is an aggressive primary brain tumor with bad prognosis. On the other hand, oncolytic measles virus (MeV) therapy is an experimental glioma treatment strategy with clinical safety and first evidence of anti-tumoral efficacy. Therefore, we investigated the combination of MeV with conventional therapies by cytotoxic survival assays in long-term glioma cell lines LN229, LNZ308, and glioma stem-like GS8 cells, as well as the basal viral infectivity in primary glioblastoma cultures T81/16, T1094/17, and T708/16. We employed Chou-Talalay analysis to identify the synergistic treatment sequence chemotherapy, virotherapy, and finally radiotherapy (CT-VT-RT). RNA sequencing and immunopeptidome analyses were used to delineate treatment-induced molecular and immunological profiles. CT-VT-RT displayed synergistic anti-glioma activity and initiated a type 1 interferon response, along with canonical Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling, and downstream interferon-stimulated genes were induced, resulting in apoptotic cascades. Furthermore, antigen presentation along with immunostimulatory chemokines was increased in CT-VT-RT-treated glioma cells, indicating a treatment-induced pro-inflammatory phenotype. We identified novel treatment-induced viral and tumor-associated peptides through HLA ligandome analysis. Our data delineate an actionable treatment-induced molecular and immunological signature of CT-VT-RT, and they could be exploited for the design of novel tailored treatment strategies involving virotherapy and immunotherapy.

17.
J Virol ; 93(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30429338

ABSTRACT

The impact of the Zika virus (ZIKV) epidemic highlights the need for vaccines that reduce or prevent infection and reliably prevent teratogenic complications. The live-attenuated measles virus (MV) vaccine strains are a promising vaccine platform, since they induce robust humoral and cellular immune responses against additional antigens and have an excellent safety record. To explore its potential to protect against ZIKV, we compared a recombinant Schwarz strain MV that encodes ZIKV prM and soluble E proteins (MV-Zika-sE) with a prototypic alum-adjuvanted whole inactivated ZIKV particle vaccine. Analysis of MV-Zika-sE-infected cells confirmed antigen expression, and the virus replicated with vaccine strain characteristics. Immunized IFNAR-/--CD46Ge mice developed E protein-specific and neutralizing antibodies, and ZIKV E-specific cellular immune responses were observed by gamma interferon (IFN-γ) enzyme-linked immunospot (ELISpot) and in vitro T cell proliferation assays. To analyze protective efficacy, vaccinated female mice were challenged with ZIKV after allogeneic mating. In MV-Zika-sE-vaccinated mice, weight gain was similar to that in uninfected mice, while no plasma viremia was detectable in the majority of the animals. In contrast, infected control animals gained less weight and experienced about 100-fold higher viremia over at least 3 days. Moreover, vaccination with MV-Zika-sE reduced the ZIKV load in different organs and the placentas and prevented infection of the fetus. Consequently, no fetal growth retardation, anemia, or death due to ZIKV infection was seen in MV-Zika-sE-vaccinated dams. In contrast, the inactivated ZIKV vaccine had little to no effect in our studies. Therefore, the MV-derived ZIKV vaccine is a promising candidate for further preclinical and clinical development.IMPORTANCE Zika virus (ZIKV) is a mosquito-borne flavivirus that causes a variety of neurological complications, including congenital birth defects. Despite the urgent need, no ZIKV vaccine has yet been licensed. Recombinant vaccine strain-derived measles viruses (MV) constitute a promising vector platform to induce immunity against foreign pathogens by expressing antigens from additional transcription units while at the same time possessing a remarkable safety profile. This concept has already been validated against different pathogens, including at least 3 other flaviviruses, and our data show that vaccination with MV expressing soluble ZIKV E protein significantly diminishes infection and prevents fetal loss or damage in an allogeneic mouse pregnancy model. It can thus be regarded as a promising emergency vaccine candidate with the potential for inclusion in routine vaccination settings in areas of endemicity to prevent teratogenic effects of circulating ZIKV during pregnancy, comparable to standard rubella virus vaccination.


Subject(s)
Disease Models, Animal , Measles Vaccine/administration & dosage , Measles virus/immunology , Viral Envelope Proteins/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Antibodies, Viral/blood , Female , Genome, Viral , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Measles Vaccine/immunology , Membrane Cofactor Protein/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pregnancy , Receptor, Interferon alpha-beta/physiology , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Zika Virus/genetics , Zika Virus Infection/immunology , Zika Virus Infection/virology
18.
Mol Immunol ; 101: 19-28, 2018 09.
Article in English | MEDLINE | ID: mdl-29852456

ABSTRACT

The development of novel vaccination strategies is a persistent challenge to provide effective prophylactic treatments to encounter viral infections. In general, the physical conjugation of selected vaccine components, e.g. antigen and adjuvant, has been shown to enhance the immunogenicity and hence, can increase effectiveness of the vaccine. In our proof-of-concept study, we generated non-infectious, replication deficient Murine Leukemia Virus (MLV)-derived virus-like particles (VLPs) that physically link antigen and adjuvant in a modular fashion by co-displaying them on their surface. For this purpose, we selected the immunodominant peptides of the model antigen ovalbumin (OVA) and the cytokine granulocyte macrophage-colony stimulating factor (GM-CSF) as non-classical adjuvant. Our results show that murine GM-CSF displayed on MLV-VLPs mediates expansion and proliferation of CD11b+ cells within murine bone marrow and total spleen cells. Moreover, we show increased immunogenicity of modular VLPs co-displaying OVA peptides and GM-CSF by their elevated capacity to induce OVA-specific T cell-activation and -proliferation within OT-I and OT-II splenocyte cultures. These enhanced effects were not achieved by using an equimolar mixture of VLPs displaying either OVA or GM-CSF. Taken together, OVA and GM-CSF co-displaying MLV-VLPs are able to target and expand antigen presenting cells which in turn results in enhanced antigen-specific T cell activation and proliferation in vitro. These data suggest MLV-VLPs to be an attractive platform to flexibly combine antigen and adjuvant for novel modular vaccination approaches.


Subject(s)
Antigen-Presenting Cells/metabolism , CD11b Antigen/metabolism , Epitopes/immunology , Leukemia Virus, Murine/metabolism , Ovalbumin/metabolism , Peptides/metabolism , T-Lymphocytes/immunology , Virion/metabolism , Animals , Cell Proliferation , Granulocyte-Macrophage Colony-Stimulating Factor , Lymphocyte Activation/immunology , Mice, Inbred C57BL
19.
Virology ; 521: 99-107, 2018 08.
Article in English | MEDLINE | ID: mdl-29902727

ABSTRACT

Cases of Middle East respiratory syndrome coronavirus (MERS-CoV) continue to occur, making it one of the WHO´s targets for accelerated vaccine development. One vaccine candidate is based on live-attenuated measles virus (MV) vaccine encoding the MERS-CoV spike glycoprotein (MERS-S). MVvac2-MERS-S(H) induces robust humoral and cellular immunity against MERS-S mediating protection. Here, the induction and nature of immunity after vaccination with MVvac2-MERS-S(H) or novel MVvac2-MERS-N were further characterized. We focused on the necessity for vector replication and the nature of induced T cells, since functional CD8+ T cells contribute importantly to clearance of MERS-CoV. While no immunity against MERS-CoV or MV was detected in MV-susceptible mice after immunization with UV-inactivated virus, replication-competent MVvac2-MERS-S(H) triggered robust neutralizing antibody titers also in adult mice. Furthermore, a significant fraction of MERS CoV-specific CD8+ T cells and MV-specific CD4+ T cells simultaneously expressing IFN-γ and TNF-α were induced, revealing that MVvac2-MERS-S(H) induces multifunctional cellular immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Coronavirus Infections/prevention & control , Middle East Respiratory Syndrome Coronavirus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Coronavirus Infections/immunology , Disease Models, Animal , Immunity, Cellular , Measles virus , Mice, Inbred BALB C , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Attenuated/immunology , Viral Vaccines/genetics
20.
Sci Rep ; 7(1): 16892, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203786

ABSTRACT

Recombinant vaccine strain-derived measles virus (MV) is clinically tested both as vaccine platform to protect against other pathogens and as oncolytic virus for tumor treatment. To investigate the potential synergism in anti-tumoral efficacy of oncolytic and vaccine properties, we chose Ovalbumin and an ideal tumor antigen, claudin-6, for pre-clinical proof of concept. To enhance immunogenicity, both antigens were presented by retroviral virus-like particle produced in situ during MV-infection. All recombinant MV revealed normal growths, genetic stability, and proper expression and presentation of both antigens. Potent antigen-specific humoral and cellular immunity were found in immunized MV-susceptible IFNAR-/--CD46Ge mice. These immune responses significantly inhibited metastasis formation or increased therapeutic efficacy compared to control MV in respective novel in vivo tumor models using syngeneic B16-hCD46/mCLDN6 murine melanoma cells. These data indicate the potential of MV to trigger selected tumor antigen-specific immune responses on top of direct tumor lysis for enhanced efficacy.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/immunology , Measles virus/genetics , Melanoma, Experimental/therapy , Vaccines, Virus-Like Particle/immunology , Animals , Antigen Presentation , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Autoantibodies/blood , Autoantibodies/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Chlorocebus aethiops , Claudins/genetics , Claudins/immunology , Claudins/metabolism , Immunity, Cellular , Immunity, Humoral , Interferon-gamma/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/immunology , Mice , Mice, Transgenic , Oncolytic Virotherapy , Ovalbumin/genetics , Ovalbumin/immunology , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/therapeutic use , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...