Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Angew Chem Int Ed Engl ; 60(1): 321-330, 2021 01 04.
Article in English | MEDLINE | ID: mdl-32886840

ABSTRACT

Matching of symmetry at interfaces is a fundamental obstacle in molecular assembly. Virus-like particles (VLPs) are important vaccine platforms against pathogenic threats, including Covid-19. However, symmetry mismatch can prohibit vaccine nanoassembly. We established an approach for coupling VLPs to diverse antigen symmetries. SpyCatcher003 enabled efficient VLP conjugation and extreme thermal resilience. Many people had pre-existing antibodies to SpyTag:SpyCatcher but less to the 003 variants. We coupled the computer-designed VLP not only to monomers (SARS-CoV-2) but also to cyclic dimers (Newcastle disease, Lyme disease), trimers (influenza hemagglutinins), and tetramers (influenza neuraminidases). Even an antigen with dihedral symmetry could be displayed. For the global challenge of influenza, SpyTag-mediated display of trimer and tetramer antigens strongly induced neutralizing antibodies. SpyCatcher003 conjugation enables nanodisplay of diverse symmetries towards generation of potent vaccines.


Subject(s)
COVID-19 Vaccines/chemistry , Nanostructures/chemistry , Vaccines, Virus-Like Particle/chemistry , Antibodies, Neutralizing/analysis , Antibodies, Viral , Antigens, Viral/chemistry , Antigens, Viral/immunology , Freezing , Humans , Models, Molecular
2.
Angew Chem Weinheim Bergstr Ger ; 133(1): 325-334, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-38504824

ABSTRACT

Matching of symmetry at interfaces is a fundamental obstacle in molecular assembly. Virus-like particles (VLPs) are important vaccine platforms against pathogenic threats, including Covid-19. However, symmetry mismatch can prohibit vaccine nanoassembly. We established an approach for coupling VLPs to diverse antigen symmetries. SpyCatcher003 enabled efficient VLP conjugation and extreme thermal resilience. Many people had pre-existing antibodies to SpyTag:SpyCatcher but less to the 003 variants. We coupled the computer-designed VLP not only to monomers (SARS-CoV-2) but also to cyclic dimers (Newcastle disease, Lyme disease), trimers (influenza hemagglutinins), and tetramers (influenza neuraminidases). Even an antigen with dihedral symmetry could be displayed. For the global challenge of influenza, SpyTag-mediated display of trimer and tetramer antigens strongly induced neutralizing antibodies. SpyCatcher003 conjugation enables nanodisplay of diverse symmetries towards generation of potent vaccines.

3.
Vaccine ; 37(22): 2952-2959, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31006497

ABSTRACT

CD8+ T cells are known to control infections, but their role in preventing latent infection from establishing has not been thoroughly investigated. We hypothesized that a potent CD8+ T cell response patrolling the mucosal viral entry points could kill the first infected cells and thereby abrogate the infection before latency is established. To investigate this, replication deficient adenovirus serotype 5 vectors encoding murine γ-herpesvirus-68 CD8+ T cell epitopes linkedto the T cell adjuvant Invariant chain, were developed. We show that intranasal vaccination of mice reduces the risk of establishment of latent infection from multiple intranasal ID50 challenges with murine γ-herpesvirus-68 by 81% per exposure at 14 days post vaccination. Protection waned over time, but immune responses were extended by heterologous prime-boost vaccination applied simultaneously intramuscularly and intranasally, and animals vaccinated 66 days prior to challenge showed a strong trend of long-term protection. Our data provides evidence that CD8+ T cells are able to protect against establishment of latent infection. Although the protective efficacy is difficult to maintain over time, this proof-of-concept study suggests a role for a CD8+ T cell arm in future vaccine strategies against latent human viral infections caused by pathogens such as HIV and multiple herpes virus.


Subject(s)
Adenoviridae/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae/pathogenicity , Animals , Female , Flow Cytometry , Herpesviridae/immunology , Mice , Mice, Inbred C57BL
4.
Sci Rep ; 9(1): 4625, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30874593

ABSTRACT

For many infectious diseases there is still no vaccine, even though potential protective antigens have been identified. Suitable platforms and conjugation routes are urgently needed to convert the promise of such antigens into broadly protective and scalable vaccines. Here we apply a newly established peptide-peptide ligation approach, SnoopLigase, for specific and irreversible coupling of antigens onto an oligomerization platform. SnoopLigase was engineered from a Streptococcus pneumoniae adhesin and enables isopeptide bond formation between two peptide tags: DogTag and SnoopTagJr. We expressed in bacteria DogTag linked to the self-assembling coiled-coil nanoparticle IMX313. This platform was stable over months at 37 °C when lyophilized, remaining reactive even after boiling. IMX-DogTag was efficiently coupled to two blood-stage malarial proteins (from PfEMP1 or CyRPA), with SnoopTagJr fused at the N- or C-terminus. We also showed SnoopLigase-mediated coupling of a telomerase peptide relevant to cancer immunotherapy. SnoopLigase-mediated nanoassembly enhanced the antibody response to both malaria antigens in a prime-boost model. Including or depleting SnoopLigase from the conjugate had little effect on the antibody response to the malarial antigens. SnoopLigase decoration represents a promising and accessible strategy for modular plug-and-display vaccine assembly, as well as providing opportunities for robust nanoconstruction in synthetic biology.


Subject(s)
Adhesins, Bacterial/immunology , Peptides/immunology , Vaccines/biosynthesis , Adhesins, Bacterial/metabolism , Antigens, Bacterial/immunology , Antigens, Protozoan/immunology , Immunoconjugates/metabolism , Immunoconjugates/pharmacology , Nanoparticles/chemistry , Peptides/chemistry , Streptococcus pneumoniae/metabolism , Vaccination
5.
ACS Nano ; 12(9): 8855-8866, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30028591

ABSTRACT

Nanoscale organization is crucial to stimulating an immune response. Using self-assembling proteins as multimerization platforms provides a safe and immunogenic system to vaccinate against otherwise weakly immunogenic antigens. Such multimerization platforms are generally based on icosahedral viruses and have led to vaccines given to millions of people. It is unclear whether synthetic protein nanoassemblies would show similar potency. Here we take the computationally designed porous dodecahedral i301 60-mer and rationally engineer this particle, giving a mutated i301 (mi3) with improved particle uniformity and stability. To simplify the conjugation of this nanoparticle, we employ a SpyCatcher fusion of mi3, such that an antigen of interest linked to the SpyTag peptide can spontaneously couple through isopeptide bond formation (Plug-and-Display). SpyCatcher-mi3 expressed solubly to high yields in Escherichia coli, giving more than 10-fold greater yield than a comparable phage-derived icosahedral nanoparticle, SpyCatcher-AP205. SpyCatcher-mi3 nanoparticles showed high stability to temperature, freeze-thaw, lyophilization, and storage over time. We demonstrate approximately 95% efficiency coupling to different transmission-blocking and blood-stage malaria antigens. Plasmodium falciparum CyRPA was conjugated to SpyCatcher-mi3 nanoparticles and elicited a high avidity antibody response, comparable to phage-derived virus-like particles despite their higher valency and RNA cargo. The simple production, precise derivatization, and exceptional ruggedness of this nanoscaffold should facilitate broad application for nanobiotechnology and vaccine development.


Subject(s)
Nanoparticles/chemistry , Peptides/chemistry , Plasmodium falciparum/chemistry , Nanotechnology , Particle Size , Peptides/immunology , Plasmodium falciparum/immunology , Porosity , Surface Properties , Vaccination
6.
Vaccines (Basel) ; 6(1)2018 Feb 11.
Article in English | MEDLINE | ID: mdl-29439476

ABSTRACT

Protection against chronic infections has necessitated the development of ever-more potent vaccination tools. HIV seems to be the most challenging foe, with a remarkable, poorly immunogenic and fragile surface glycoprotein and the ability to overpower the cell immune system. Virus-like-particle (VLP) vaccines have emerged as potent inducers of antibody and helper T cell responses, while replication-deficient viral vectors have yielded potent cytotoxic T cell responses. Here, we review the emerging concept of merging these two technologies into virus-like-vaccines (VLVs) for the targeting of HIV. Such vaccines are immunologically perceived as viruses, as they infect cells and produce VLPs in situ, but they only resemble viruses, as the replication defective vectors and VLPs cannot propagate an infection. The inherent safety of such a platform, despite robust particle production, is a distinct advantage over live-attenuated vaccines that must balance safety and immunogenicity. Previous studies have delivered VLVs encoded in modified Vaccinia Ankara vectors and we have developed the concept into a single-reading adenovirus-based technology capable of eliciting robust CD8⁺ and CD4⁺ T cells responses and trimer binding antibody responses. Such vaccines offer the potential to display the naturally produced immunogen directly and induce an integrated humoral and cellular immune response.

7.
J Immunother ; 40(2): 51-61, 2017.
Article in English | MEDLINE | ID: mdl-28166180

ABSTRACT

Currently available prophylactic vaccines have no therapeutic efficacy for preexisting human papillomavirus (HPVs) infections, do not target all oncogenic HPVs and are insufficient to eliminate the burden of HPV induced cancer. We aim to develop an alternative HPV vaccine which is broadly effective and capable of clearing preexisting infection. In an initial attempt to develop a broadly reactive therapeutic vaccine, we designed a putative papillomavirus (PV) ancestor antigen (circulating sequence derived antigenic sequences E1E2-CDSE1E2) based on the conserved E1 and E2 protein sequences from existing oncogenic HPV strains. This antigen was found to be as related to circulating oncogenic Macaca fascicularis papillomaviruses (MfPVs) as to oncogenic HPVs. The CDSE1E2 antigen was fused to a T-cell adjuvant and encoded in chimpanzee 3 and 63 adenoviral vectors. We first showed that the combination of these 2 vaccines induced long-lasting potent CDSE1E2 specific T cell responses in outbred mice. This prime-boost regimen was then tested in female macaques naturally infected with MfPVs. All immunized animals (16/16) responded to the vaccine antigen but with reduced cross-reactivity against existing PVs. Preexisting MfPV infections did not prime vaccine inducible immune responses. Importantly, immunized oncogenic MfPV type 3 (MfPV3) infected animals that responded toward MfPV3 were able to diminish cervical MfPV3 DNA content. Although insufficient breadth was achieved, our results suggest that a relevant level of E1E2 specific T cell immunity is achievable and might be sufficient for the elimination of PV infection. Importantly, naturally infected macaques, offer a relevant model for testing vaccines aimed at eliminating mucosal PV infections.


Subject(s)
Cervix Uteri/immunology , Oncogenic Viruses/immunology , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Papillomavirus Vaccines/immunology , T-Lymphocytes/immunology , Uterine Cervical Neoplasms/prevention & control , Animals , Animals, Outbred Strains , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Viral/genetics , Cells, Cultured , Cervix Uteri/virology , DNA, Viral/analysis , Disease Models, Animal , Female , Genetic Engineering , Histocompatibility Antigens Class II/genetics , Humans , Immunity, Cellular , Immunization, Secondary , Macaca fascicularis , Mice , Oncogenic Viruses/genetics , Pan troglodytes , Papillomaviridae/genetics , Papillomavirus Infections/complications , Recombinant Fusion Proteins/genetics , T-Lymphocytes/virology , Uterine Cervical Neoplasms/etiology , Vaccines, DNA , Viral Proteins/genetics
8.
Vaccine ; 35(8): 1140-1147, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28131394

ABSTRACT

The malaria parasite Plasmodium falciparum presents antigens on the infected erythrocyte surface that bind human receptors expressed on the vascular endothelium. The VAR2CSA mediated binding to a distinct chondroitin sulphate A (CSA) is a crucial step in the pathophysiology of placental malaria and the CSA binding region of VAR2CSA has been identified as a promising vaccine target against placental malaria. Here we designed adenovirus encoded virus-like particles (VLP) by co-encoding Simian Immunodeficiency Virus (SIV) gag and VAR2CSA. The VAR2CSA antigen was fused to the transmembrane (TM) and cytoplasmic tail (CT) domains of either the envelope protein of mouse mammary tumour virus (MMTV) or the hemagglutinin (HA) of influenza A. For a non-VLP incorporation control, a third design was made where VAR2CSA was expressed without TM-CT domains. In the primary immunogenicity study in Balb/c mice, VAR2CSA fused to HA TM-CT was significantly superior in inducing ID1-ID2a specific antibodies after the first immunization. A sequential study was performed to include a comparison to the soluble VAR2CSA protein vaccine, which has entered a phase I clinical trial (NCT02647489). The results revealed the induction of higher antibody responses and increased inhibition of parasite binding to CSA using either VAR2CSA HA TM-CT or VAR2CSA MMTV TM-CT as priming vaccines for protein double-boost immunizations, compared to protein prime-double boost regimen. Analysis of pooled serum samples on peptide arrays revealed a unique targeting of several epitopes in mice that had been primed with VAR2CSA HA TM-CT. Consequently, modification of VLP anchors is an important point of optimization in virus-encoded retroviral VLP-based vaccines, and adenovirus VLPs boosted by recombinant proteins offer hope of increasing the levels of protective VAR2CSA specific antibodies.


Subject(s)
Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Vaccines, Virus-Like Particle/administration & dosage , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Female , Gene Products, gag/chemistry , Gene Products, gag/genetics , Gene Products, gag/immunology , Genetic Vectors/chemistry , Genetic Vectors/immunology , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunization , Malaria Vaccines/biosynthesis , Malaria Vaccines/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mammary Tumor Virus, Mouse , Mice , Mice, Inbred BALB C , Placenta/chemistry , Placenta/immunology , Placenta/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/drug effects , Pregnancy , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Simian Immunodeficiency Virus , Vaccines, Virus-Like Particle/biosynthesis , Vaccines, Virus-Like Particle/genetics
9.
Vaccine ; 34(44): 5344-5351, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27633665

ABSTRACT

The low number of envelope (Env) spikes presented on native HIV-1 particles is a major impediment for HIV-1 prophylactic vaccine development. We designed virus-like particle encoding adenoviral vectors utilizing SIVmac239 Gag as an anchor for full length and truncated HIV-1 M consensus Env. Truncated Env overexpressed VRC01 and 17b binding antigen on the surface of transduced cells while the full length Env vaccine presented more and similar amounts of antigen binding to the trimer conformation sensitive antibodies PGT151 and PGT145, respectively. The adenoviral vectors were used to prime Balb/c mice followed by sequential boosting with chimpanzee type 63, and chimpanzee type 3 adenoviral vectors encoding SIVmac239 Gag and full length consensus Env. Both vaccine regimens induced increasing titers of binding antibody responses after each immunization, and significant differences in immune responses between the two groups were observed after the final immunization. Full length Env priming skewed antibody responses towards gp41, while truncated Env priming induced responses primarily targeting gp120 containing and derived antigens. Importantly, no differences in neutralizing antibody responses were found between the different priming regimens as both induced high titered tier 1 neutralizing antibodies, but no tier 2 antibodies, possibly reflecting the similar presentation of trimer specific antibody epitopes. The described vaccine regimens provide insight into the effects of the HIV-1 Env cytoplasmic tail on epitope presentation and subsequent immune responses, which is relevant for the interpretation of current clinical trials that are using truncated Env as an immunogen. The regimens described here provide similar neutralization titers, and thus are useful for investigating the importance of specificity in non-neutralizing antibody mediated protection against viral challenge.


Subject(s)
AIDS Vaccines/immunology , Adenoviridae/genetics , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Vaccines, Virus-Like Particle/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Adenoviridae/immunology , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Epitopes/immunology , Genes, gag , Genetic Vectors , HIV Antibodies/immunology , Immunity, Humoral , Immunogenicity, Vaccine , Mice, Inbred BALB C , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology
10.
Vaccine ; 34(18): 2147-56, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27004934

ABSTRACT

Previous studies have shown promising results when using an agonistic anti-4-1BB antibody treatment against established tumors. While this is promising, this type of treatment can induce severe side effects. Therefore, we decided to incorporate the membrane form of 4-1BB ligand (4-1BBL) in a replicative deficient adenovirus vaccine expressing the invariant chain (Ii) adjuvant fused to a tumor associated antigen (TAA). The Ii adjuvant increases and prolongs TAA specific CD8+ T cells as previously shown and local expression of 4-1BBL was chosen to avoid the toxicity associated with systemic antibody administration. Furthermore, adenovirus encoded 4-1BBL expression has previously been successfully used to enhance responses toward Plasmodium falciparum and Influenza A antigens. We showed that the incorporation of 4-1BBL in the adenovirus vector led to surface expression of 4-1BBL on antigen presenting cells, but it did not enhance T cell responses in mice towards the Ii linked antigen. In tumor-bearing mice, our vaccine was found to decrease the frequency of TAA specific CD8+ T cells, but this difference did not alter the therapeutic efficacy. In order to reconcile our findings with the previous reports of increased anti-cancer efficacy using systemically delivered 4-1BB agonists, we incorporated a secreted version of 4-1BBL (Fc-4-1BBL) in our vaccine and co-expressed it with the Ii linked to TAA. In tumor bearing mice, this vaccine initially delayed tumor growth and slightly increased survival compared to the vaccine expressing the membrane form of 4-1BBL. Accordingly, secreted 4-1BBL co-encoded with the Ii linked antigen may offer a simplification compared to administration of drug and vaccine separately.


Subject(s)
4-1BB Ligand/immunology , Adenoviridae , Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Melanoma, Experimental/therapy , Animals , Antigen-Presenting Cells/immunology , B7-2 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , COS Cells , Female , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Recombinant Proteins/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...