Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Protein Expr Purif ; 210: 106295, 2023 10.
Article in English | MEDLINE | ID: mdl-37201590

ABSTRACT

The human cell line HEK293 is one of the preferred choices for manufacturing therapeutic proteins and viral vectors for human applications. Despite its increased use, it is still considered in disadvantage in production aspects compared to cell lines such as the CHO cell line. We provide here a simple workflow for the rapid generation of stably transfected HEK293 cells expressing an engineered variant of the SARS-CoV-2 Receptor Binding Domain (RBD) carrying a coupling domain for linkage to VLPs through a bacterial transpeptidase-sortase (SrtA). To generate stable suspension cells expressing the RBD-SrtA, a single two plasmids transfection was performed, with hygromycin selection. The suspension HEK293 were grown in adherent conditions, with 20% FBS supplementation. These transfection conditions increased cell survival, allowing the selection of stable cell pools, which was otherwise not possible with standard procedures in suspension. Six pools were isolated, expanded and successfully re-adapted to suspension with a gradual increase of serum-free media and agitation. The complete process lasted four weeks. Stable expression with viability over 98% was verified for over two months in culture, with cell passages every 4-5 days. With process intensification, RBD-SrtA yields reached 6.4 µg/mL and 13.4 µg/mL in fed-batch and perfusion-like cultures, respectively. RBD-SrtA was further produced in fed-batch stirred tank 1L-bioreactors, reaching 10-fold higher yields than perfusion flasks. The trimeric antigen displayed the conformational structure and functionality expected. This work provides a series of steps for stable cell pool development using suspension HEK293 cells aimed at the scalable production of recombinant proteins.


Subject(s)
COVID-19 , Humans , HEK293 Cells , SARS-CoV-2 , Bioreactors , Recombinant Proteins/genetics
2.
Nanomedicine ; 44: 102584, 2022 08.
Article in English | MEDLINE | ID: mdl-35850421

ABSTRACT

A vaccine candidate to SARS-CoV-2 was constructed by coupling the viral receptor binding domain (RBD) to the surface of the papaya mosaic virus (PapMV) nanoparticle (nano) to generate the RBD-PapMV vaccine. Immunization of mice with the coupled RBD-PapMV vaccine enhanced the antibody titers and the T-cell mediated immune response directed to the RBD antigen as compared to immunization with the non-coupled vaccine formulation (RBD + PapMV nano). Anti-RBD antibodies, generated in vaccinated animals, neutralized SARS-CoV-2 infection in vitro against the ancestral, Delta and the Omicron variants. At last, immunization of mice susceptible to the infection by SARS-CoV-2 (K18-hACE2 transgenic mice) with the RBD-PapMV vaccine induced protection to the ancestral SARS-CoV-2 infectious challenge. The induction of the broad neutralization against SARS-CoV-2 variants induced by the RBD-PapMV vaccine demonstrate the potential of the PapMV vaccine platform in the development of efficient vaccines against viral respiratory infections.


Subject(s)
COVID-19 , Nanoparticles , Animals , Antibodies, Neutralizing , Antibodies, Viral , Broadly Neutralizing Antibodies , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Mice , Mice, Inbred BALB C , Potexvirus , SARS-CoV-2
3.
PLoS One ; 16(12): e0261987, 2021.
Article in English | MEDLINE | ID: mdl-34972158

ABSTRACT

Nanoparticles made of the coat protein of papaya mosaic virus (PapMV) and a single-strand RNA were previously shown to be an efficient antigen presentation system for the trigger of cellular immunity. Engineering of PapMV nano with a cytotoxic T lymphocyte epitope was previously shown activating specific T lymphocytes through a proteasome-independent major histocompatibility complex class I (MHC-I) cross-presentation. In this study, we provide new insights into the mechanism of the MHC-I cross-presentation mediated by PapMV nanoparticles. We demonstrate that PapMV nanoparticles do not require the transporter associated with antigen presentation (TAP), but rather depend on lysosome acidification and cathepsin S protease activity for presentation of the T cell epitope. We have also linked the induction of autophagy with this vacuolar MHC-I cross-presentation process. Interestingly, autophagy is induced in antigen-presenting cells after PapMV nanoparticles exposure and inhibition of autophagy reduce MHC-I cross-presentation. This study demonstrates that autophagy is associated with TAP- and proteasome-independent MHC-I cross-presentation. A deeper understanding of the autophagy-dependent MHC-I cross-presentation will be useful in designing vaccination platforms that aim to trigger an efficient cytotoxic T lymphocyte response.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , Autophagy , Cross-Priming/immunology , Histocompatibility Antigens Class I/immunology , Cathepsins/chemistry , Chloroquine/chemistry , Epitopes/chemistry , Epitopes, T-Lymphocyte/chemistry , Humans , Lysosomes/chemistry , Microscopy, Confocal , Nanoparticles/chemistry , Potexvirus , Protein Engineering , RNA/chemistry
4.
Vaccines (Basel) ; 9(1)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33435570

ABSTRACT

BACKGROUND: The papaya mosaic virus (PapMV) vaccine platform is a rod-shaped nanoparticle made of the recombinant PapMV coat protein (CP) self-assembled around a noncoding single-stranded RNA (ssRNA) template. The PapMV nanoparticle induces innate immunity through stimulation of the Toll-like receptors (TLR) 7 and 8. The display of the vaccine antigen at the surface of the nanoparticle, associated with the co-stimulation signal via TLR7/8, ensures a strong stimulation of the immune response, which is ideal for the development of candidate vaccines. In this study, we assess the impact of where the peptide antigen is fused, whether at the surface or at the extremities of the nanoparticles, on the immune response directed to that antigen. METHODS: Two different peptides from influenza A virus were used as model antigens. The conserved M2e peptide, derived from the matrix protein 2 was chosen as the B-cell epitope, and a peptide derived from the nucleocapsid was chosen as the cytotoxic T lymphocytes (CTL) epitope. These peptides were coupled at two different positions on the PapMV CP, the N- (PapMV-N) or the C-terminus (PapMV-C), using the transpeptidase activity of Sortase A (SrtA). The immune responses, both humoral and CD8+ T-cell-mediated, directed to the peptide antigens in the two different fusion contexts were analyzed and compared. The impact of coupling density at the surface of the nanoparticle was also investigated. CONCLUSIONS: The results demonstrate that coupling of the peptide antigens at the N-terminus (PapMV-N) of the PapMV CP led to an enhanced immune response to the coupled peptide antigens as compared to coupling to the C-terminus. The difference between the two vaccine platforms is linked to the enhanced capacity of the PapMV-N vaccine platform to stimulate TLR7/8. We also demonstrated that the strength of the immune response increases with the density of coupling at the surface of the nanoparticles.

5.
Vaccines (Basel) ; 8(4)2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33158147

ABSTRACT

Vaccine design strategies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are focused on the Spike protein or its subunits as the main antigen target of neutralizing antibodies. In this work, we propose rapid production methods of an extended segment of the Spike Receptor Binding Domain (RBD) in HEK293SF cells cultured in suspension, in serum-free media, as a major component of a COVID-19 subunit vaccine under development. The expression of RBD, engineered with a sortase-recognition motif for protein-based carrier coupling, was achieved at high yields by plasmid transient transfection or human type-5-adenoviral infection of the cells, in a period of only two and three weeks, respectively. Both production methods were evaluated in 3L-controlled bioreactors with upstream and downstream bioprocess improvements, resulting in a product recovery with over 95% purity. Adenoviral infection led to over 100 µg/mL of RBD in culture supernatants, which was around 7-fold higher than levels obtained in transfected cultures. The monosaccharide and sialic acid content was similar in the RBD protein from the two production approaches. It also exhibited a proper conformational structure as recognized by monoclonal antibodies directed against key native Spike epitopes. Efficient direct binding to ACE2 was also demonstrated at similar levels in RBD obtained from both methods and from different production lots. Overall, we provide bioprocess-related data for the rapid, scalable manufacturing of low cost RBD based vaccines against SARS-CoV-2, with the added value of making a functional antigen available to support further research on uncovering mechanisms of virus binding and entry as well as screening for potential COVID-19 therapeutics.

6.
Vaccines (Basel) ; 8(3)2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32698532

ABSTRACT

Inactivated influenza vaccines efficacy is variable and often poor. We conducted a phase 1 trial (NCT02188810), to assess the safety and immunogenicity of a novel nanoparticle Toll-like receptor 7/8 agonist adjuvant (Papaya Mosaic Virus) at different dose levels combined with trivalent influenza vaccine in healthy persons 18-50 years of age. Hemagglutination-inhibition assays, antibody to Influenza A virus nucleoprotein and peripheral blood mononuclear cells for measurement of interferon-gamma ELISPOT response to influenza antigens, Granzyme B and IFNγ:IL-10 ratio were measured. The most common adverse events were transient mild to severe injection site pain and no safety signals were observed. A dose-related adjuvant effect was observed. Geometric mean hemagglutination-inhibition titers increased at day 28 in most groups and waned over time, but fold-antibody responses were poor in all groups. Cell mediated immunity results were consistent with humoral responses. The Papaya Mosaic Virus adjuvant in doses of 30 to 240 µg combined with reduced influenza antigen content was safe with no signals up to 3 years after vaccination. A dose-related adjuvant effect was observed and immunogenicity results suggest that efficacy study should be conducted in influenza antigen-naïve participants.

7.
Vaccines (Basel) ; 7(2)2019 Jun 12.
Article in English | MEDLINE | ID: mdl-31212772

ABSTRACT

Background: Flexuous rod-shape nanoparticles-made of the coat protein of papaya mosaic virus (PapMV)-provide a promising vaccine platform for the presentation of viral antigens to immune cells. The PapMV nanoparticles can be combined with viral antigens or covalently linked to them. The coupling to PapMV was shown to improve the immune response triggered against peptide antigens (<39 amino acids) but it remains to be tested if large proteins can be coupled to this platform and if the coupling will lead to an immune response improvement. Methods: Two full-length recombinant viral proteins, the influenza nucleoprotein (NP) and the simian immunodeficiency virus group-specific protein antigen (GAG) were coupled to PapMV nanoparticles using sortase A. Mice were immunized with the nanoparticles coupled to the antigens and the immune response directed to the antigens were analyzed by ELISA and ELISPOT. Results: We showed the feasibility of coupling two different full-length proteins (GAG and NP) to the nanoparticle. We also showed that the coupling to PapMV nanoparticles improved significantly the humoral and the cytotoxic T lymphocyte (CTL) immune response to the antigens. Conclusion: This proof of concept demonstrates the versatility and the efficacy of the PapMV vaccine platform in the design of vaccines against viral diseases.

8.
Nanomedicine ; 14(8): 2563-2574, 2018 11.
Article in English | MEDLINE | ID: mdl-30193813

ABSTRACT

Influenza virus infections are a significant public threat and the best approach to prevent them is through vaccination. Because of the perpetual changes of circulating influenza strains, the efficacy of influenza vaccines rarely exceeds 50%. To improve the protection efficacy, we have designed a novel vaccine formulation that shows a broad range of protection. The formulation is made of the matrix protein 2 (M2e) and the nucleoprotein (NP) antigens. The multimerization of NP into nanoparticles improved significantly the immune response to NP. The combination of the NP nanoparticles with the PapMV-M2e nanoparticles enhances significantly the immune response directed to NP revealing the adjuvant property of the PapMV platform. The vaccine formulation combining these two types of nanoparticles protects mice from infectious challenges by two different influenza strains (H1N1 and H3N2) and is a promising influenza A vaccine capable to elicit a broad protection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Nanoparticles/administration & dosage , Orthomyxoviridae Infections/prevention & control , Potexvirus/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Orthomyxoviridae Infections/immunology
9.
Nanomedicine ; 14(7): 2317-2327, 2018 10.
Article in English | MEDLINE | ID: mdl-29128662

ABSTRACT

Rod-shaped virus-like nanoparticles (VLNP) made of papaya mosaic virus (PapMV) coat proteins (CP) self-assembled around a single stranded RNA (ssRNA) were showed to be a TLR7 agonist. Their utilization as an immune modulator in cancer immunotherapy was shown to be promising. To establish a clinical relevance in human for PapMV VLNP, we showed that stimulation of human peripheral blood mononuclear cells (PBMC) with VLNP induces the secretion of interferon alpha (IFNα) and other pro-inflammatory cytokines and chemokines. Plasmacytoid dendritic cells (pDCs) were activated and secreted IFN-α upon VLNP exposure. Monocyte-derived dendritic cells upregulate maturation markers and produce IL-6 in response to PapMV VLNP stimulation, which suggests the activation of TLR8. Finally, when co-cultured with NK cells, PapMV induced pDCs promoted the NK cytolytic activity against cancer cells. These data obtained with primary human immune cells further strengthen the clinical relevance of PapMV VLNPs as a cancer immunotherapy agent.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Leukocytes, Mononuclear/immunology , Nanoparticles/administration & dosage , Potexvirus/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Dendritic Cells/metabolism , Humans , Interferon-alpha/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Nanoparticles/chemistry , Potexvirus/chemistry
10.
J Nanobiotechnology ; 15(1): 54, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28720097

ABSTRACT

BACKGROUND: Flexuous rod-shaped nanoparticles made of the coat protein (CP) of papaya mosaic virus (PapMV) have been shown to trigger innate immunity through engagement of toll-like receptor 7 (TLR7). PapMV nanoparticles can also serve as a vaccine platform as they can increase the immune response to fused peptide antigens. Although this approach shows great potential, fusion of antigens directly to the CP open reading frame (ORF) is challenging because the fused peptides can alter the structure of the CP and its capacity to self assemble into nanoparticles-a property essential for triggering an efficient immune response to the peptide. This represents a serious limitation to the utility of this approach as fusion of small peptides only is tolerated. RESULTS: We have developed a novel approach in which peptides are fused directly to pre-formed PapMV nanoparticles. This approach is based on the use of a bacterial transpeptidase (sortase A; SrtA) that can attach the peptide directly to the nanoparticle. An engineered PapMV CP harbouring the SrtA recognition motif allows efficient coupling. To refine our engineering, and to predict the efficacy of coupling with SrtA, we modeled the PapMV structure based on the known structure of PapMV CP and on recent reports revealing the structure of two closely related potexviruses: pepino mosaic virus (PepMV) and bamboo mosaic virus (BaMV). We show that SrtA can allow the attachment of long peptides [Influenza M2e peptide (26 amino acids) and the HIV-1 T20 peptide (39 amino acids)] to PapMV nanoparticles. Consistent with our PapMV structural model, we show that around 30% of PapMV CP subunits in each nanoparticle can be fused to the peptide antigen. As predicted, engineered nanoparticles were capable of inducing a strong antibody response to the fused antigen. Finally, in a challenge study with influenza virus, we show that mice vaccinated with PapMV-M2e are protected from infection. CONCLUSIONS: This technology will allow the development of vaccines harbouring long peptides containing several B and/or T cell epitopes that can contribute to a broad and robust protection from infection. The design can be fast, versatile and can be adapted to the development of vaccines for many infectious diseases as well as cancer vaccines.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Capsid Proteins/chemistry , Cysteine Endopeptidases/chemistry , HIV Envelope Protein gp41/chemistry , Influenza Vaccines/chemistry , Nanoparticles , Peptide Fragments/chemistry , Potexvirus/immunology , Viral Matrix Proteins/chemistry , Animals , Capsid Proteins/immunology , Enfuvirtide , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Female , HIV Envelope Protein gp41/immunology , HIV-1/drug effects , Influenza Vaccines/immunology , Mice, Inbred BALB C , Models, Molecular , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Peptide Fragments/immunology , Potexvirus/chemistry , Surface Properties , Toll-Like Receptor 7/chemistry , Toll-Like Receptor 7/immunology , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology , Viral Matrix Proteins/immunology
11.
Front Immunol ; 8: 1885, 2017.
Article in English | MEDLINE | ID: mdl-29354118

ABSTRACT

Although vaccination has been an effective way of preventing infections ever since the eighteenth century, the generation of therapeutic vaccines and immunotherapies is still a work in progress. A number of challenges impede the development of these therapeutic approaches such as safety issues related to the administration of whole pathogens whether attenuated or inactivated. One safe alternative to classical vaccination methods gaining recognition is the use of nanoparticles, whether synthetic or naturally derived. We have recently demonstrated that the papaya mosaic virus (PapMV)-like nanoparticle can be used as a prophylactic vaccine against various viral and bacterial infections through the induction of protective humoral and cellular immune responses. Moreover, PapMV is also very efficient when used as an immune adjuvant in an immunotherapeutic setting at slowing down the growth of aggressive mouse melanoma tumors in a type I interferon (IFN-I)-dependent manner. In the present study, we were interested in exploiting the capacity of PapMV of inducing robust IFN-I production as treatment for the chronic viral infection model lymphocytic choriomeningitis virus (LCMV) clone 13 (Cl13). Treatment of LCMV Cl13-infected mice with two systemic administrations of PapMV was ineffective, as shown by the lack of changes in viral titers and immune response to LCMV following treatment. Moreover, IFN-α production following PapMV administration was almost completely abolished in LCMV-infected mice. To better isolate the mechanisms at play, we determined the influence of a pretreatment with PapMV on secondary PapMV administration, therefore eliminating potential variables emanating from the infection. Pretreatment with PapMV led to the same outcome as an LCMV infection in that IFN-α production following secondary PapMV immunization was abrogated for up to 50 days while immune activation was also dramatically impaired. We showed that two distinct and overlapping mechanisms were responsible for this outcome. While short-term inhibition was partially the result of interleukin-1 receptor-associated kinase 1 degradation, a crucial component of the toll-like receptor 7 signaling pathway, long-term inhibition was mainly due to interference by PapMV-specific antibodies. Thus, we identified a possible pitfall in the use of virus-like particles for the systemic treatment of chronic viral infections and discuss mitigating alternatives to circumvent these potential problems.

12.
J Immunol ; 198(1): 292-299, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27864474

ABSTRACT

The increasing use of plant viruses for the development of new vaccines and immunotherapy approaches poses questions regarding the mechanism by which the mammalian immune system recognizes these viruses. For example, although natural Abs (NA) and complement are key components of the innate immune system involved in the opsonization, phagocytosis, and destruction of microorganisms infecting mammals, their implication in plant virus recognition and immunogenicity is not well defined. In this study, we address the involvement of NA and the complement system in the activation of innate immunity through engagement of TLR7 with papaya mosaic virus (PapMV)-like nanoparticles. We demonstrate that NA, although binding to PapMV, are not involved in its recognition by the immune system. On the other hand, C3 strongly binds to PapMV nanoparticles and its depletion significantly reduces PapMV's interaction with immune cells. Unexpectedly, however, we observed increased immune cell activation following administration of PapMV to complement-depleted mice. TLR7 activation by PapMV in the absence of C3 induced higher IFN-α production, resulting in superior immune cell activation and increased immunotherapeutic properties. In conclusion, in this study we established the involvement of the complement system in the recognition and the phagocytosis of PapMV nanoparticles and identified an unsuspected role for C3 in regulating the production of IFN-α following TLR7 activation.


Subject(s)
Complement C3/immunology , Dendritic Cells/immunology , Interferon-gamma/biosynthesis , Membrane Glycoproteins/immunology , Mosaic Viruses/immunology , Toll-Like Receptor 7/immunology , Animals , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles , Phagocytosis/immunology , Polymerase Chain Reaction , Toll-Like Receptor 7/metabolism
13.
J Nanobiotechnology ; 14(1): 43, 2016 Jun 10.
Article in English | MEDLINE | ID: mdl-27282291

ABSTRACT

BACKGROUND: The addition of an adjuvant to a vaccine is a promising approach to increasing strength and immunogenicity towards antigens. Despite the fact that adjuvants have been used in vaccines for decades, their mechanisms of action and their influence on the kinetics of the immune response are still not very well understood. The use of papaya mosaic virus (PapMV) nanoparticles-a novel TLR7 agonist-was recently shown to improve and broaden the immune response directed to trivalent inactivated flu vaccine (TIV) in mice and ferrets. RESULTS: We investigated the capacity of PapMV nanoparticles to increase the speed of the immune response toward TIV. PapMV nanoparticles induced a faster and stronger humoral response to TIV that was measured as early as 5 days post-immunization. The addition of PapMV nanoparticles was shown to speed up the differentiation of B-cells into early plasma cells, and increased the growth of germinal centers in a CD4+ dependent manner. TIV vaccination with PapMV nanoparticles as an adjuvant protected mice against a lethal infection as early as 10 days post-immunization. CONCLUSION: In conclusion, PapMV nanoparticles are able to accelerate a broad humoral response to TIV. This property is of the utmost importance in the field of vaccination, especially in the case of pandemics, where populations need to be protected as soon as possible after vaccination.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antibody Formation , Influenza Vaccines/therapeutic use , Mosaic Viruses/immunology , Nanoparticles/therapeutic use , Orthomyxoviridae Infections/prevention & control , Vaccines, Inactivated/therapeutic use , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Carica/virology , Female , Immunization , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Mosaic Viruses/chemistry , Nanoparticles/chemistry , Nanoparticles/virology , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Vaccines, Inactivated/immunology
14.
Nano Lett ; 16(3): 1826-32, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26891174

ABSTRACT

The recent development of novel immunotherapies is revolutionizing cancer treatment. These include, for example, immune checkpoint blockade, immunomodulation, or therapeutic vaccination. Although effective on their own, combining multiple approaches will most likely be required in order to achieve the maximal therapeutic benefit. In this regard, the papaya mosaic virus nanoparticle (PapMV) has shown tremendous potential as (i) an immunostimulatory molecule, (ii) an adjuvant, and (iii) a vaccine platform through its intrinsic capacity to activate the innate immune response in an IFN-α-dependent manner. Here, we demonstrate that intratumor administration of PapMV significantly slows down melanoma progression and prolongs survival. This correlates with enhanced chemokine and pro-inflammatory-cytokine production in the tumor and increased immune-cell infiltration. Proportions of total and tumor-specific CD8(+) T cells dramatically increase following PapMV treatment whereas those of myeloid-derived suppressor cells (MDSC) concomitantly decrease. Moreover, systemic PapMV administration prevents metastatic tumor-implantation in the lungs. Importantly, PapMV also synergistically improves the therapeutic benefit of dendritic cell (DC)-based vaccination and PD-1 blockade by potentiating antitumor immune responses. This study illustrates the immunostimulatory potential of a plant virus-derived nanoparticle for cancer therapy either alone or in conjunction with other promising immunotherapies in clinical development.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Melanoma/prevention & control , Mosaic Viruses/immunology , Nanoparticles , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Carica/virology , Cell Line, Tumor , Cytokines/immunology , Female , Immunotherapy , Melanoma/immunology , Melanoma/pathology , Mice, Inbred C57BL , Mosaic Viruses/chemistry , Nanoparticles/chemistry
15.
Vaccine ; 33(51): 7245-7253, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26549362

ABSTRACT

The emergence of highly virulent influenza strains and the risks of pandemics as well as the limited efficiency of the current seasonal vaccines are important public health concerns. There is a major need for new influenza vaccines that would be broadly cross-protective. The ectodomain of matrix protein 2 (M2e) is highly conserved amongst different influenza strains and could be used as a broad spectrum antigen. To overcome its low immunogenicity we have fused a short peptide epitope derived from the human consensus sequence of M2e (amino acids 6-14, EVETPIRNE) to the N-terminus of papaya mosaic virus coat protein. The fusion harboring coat proteins were assembled around a single stranded RNA into virus-like particles (PapMV-sM2e). The resulting PapMV-sM2e rod-shaped particle was stable and indistinguishable from regular PapMV particles. A single intramuscular immunization with PapMV-sM2e was sufficient to mount appreciable levels of CD4 dependent M2e specific total IgG and IgG2a antibody in mice sera. PapMV-sM2e proved to be self-adjuvanting since the addition of PapMV as an exogenous adjuvant did not result in significantly improved antibody titers. In addition, we confirmed the adjuvant property of PapMV-sM2e using the trivalent inactivated flu vaccine as antigen and demonstrated that the newly engineered nanoparticles areas efficacious as an adjuvant than the original PapMV nanoparticles. Upon infection with a sub-lethal dose of influenza, PapMV-sM2e vaccinated animals were completely protected from virus induced morbidity and mortality. Mice immunized with decreasing amounts of PapMV-sM2e and challenged with a more stringent dose of influenza virus displayed dose-dependent levels of protection. Seventy percent of the mice immunized once with the highest dose of PapMV-sM2e survived the challenged. The survival of the mice correlated mainly with the levels of anti-M2e IgG2a antibodies obtained before the infection. These results demonstrate that PapMV-sM2e can be an important component of a broadly cross-reactive influenza vaccine.


Subject(s)
Drug Carriers , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Potexvirus/genetics , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/blood , Capsid Proteins/genetics , Disease Models, Animal , Dose-Response Relationship, Immunologic , Immunoglobulin G/blood , Influenza Vaccines/genetics , Injections, Intramuscular , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survival Analysis , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Virus-Like Particle/genetics , Viral Matrix Proteins/genetics
16.
Vaccines (Basel) ; 3(3): 620-37, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26350598

ABSTRACT

Vaccines are considered one of the greatest medical achievements in the battle against infectious diseases. However, the intractability of various diseases such as hepatitis C, HIV/AIDS, malaria, tuberculosis, and cancer poses persistent hurdles given that traditional vaccine-development methods have proven to be ineffective; as such, these challenges have driven the emergence of novel vaccine design approaches. In this regard, much effort has been put into the development of new safe adjuvants and vaccine platforms. Of particular interest, the utilization of plant virus-like nanoparticles and recombinant plant viruses has gained increasing significance as an effective tool in the development of novel vaccines against infectious diseases and cancer. The present review summarizes recent advances in the use of plant viruses as nanoparticle-based vaccines and adjuvants and their mechanism of action. Harnessing plant-virus immunogenic properties will enable the design of novel, safe, and efficacious prophylactic and therapeutic vaccines against disease.

17.
J Nanobiotechnology ; 12: 19, 2014 May 03.
Article in English | MEDLINE | ID: mdl-24885884

ABSTRACT

BACKGROUND: Trivalent inactivated flu vaccines (TIV) are currently the best means to prevent influenza infections. However, the protection provided by TIV is partial (about 50%) and it is needed to improve the efficacy of protection. Since the respiratory tract is the main site of influenza replications, a vaccine that triggers mucosal immunity in this region can potentially improve protection against this disease. Recently, PapMV nanoparticles used as an adjuvant in a formulation with TIV administered by the subcutaneous route have shown improving the immune response directed to the TIV and protection against an influenza challenge. FINDINGS: In the present study, we showed that intranasal instillation with a formulation containing TIV and PapMV nanoparticles significantly increase the amount of IgG, IgG2a and IgA in lungs of vaccinated mice as compared to mice that received TIV only. Instillation with the adjuvanted formulation leads to a more robust protection against an influenza infection with a strain that is lethal to mice vaccinated with the TIV. CONCLUSIONS: We demonstrate for the first time that PapMV nanoparticles are an effective and potent mucosal adjuvant for vaccination.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunity, Mucosal , Influenza Vaccines/immunology , Mosaic Viruses/immunology , Nanoparticles/administration & dosage , Orthomyxoviridae Infections/prevention & control , Vaccines, Inactivated/immunology , Adjuvants, Immunologic/chemistry , Animals , Immunoglobulin A/analysis , Immunoglobulin A/immunology , Immunoglobulin G/analysis , Immunoglobulin G/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Lung/immunology , Mice , Mice, Inbred BALB C , Mosaic Viruses/chemistry , Nanoparticles/chemistry , Orthomyxoviridae Infections/immunology , Vaccines, Inactivated/administration & dosage
18.
PLoS One ; 9(6): e99211, 2014.
Article in English | MEDLINE | ID: mdl-24911872

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is found in multiple malignancies and exerts immunosuppressive effects that are central in protecting tumors from host T lymphocyte rejection. IDO is an enzyme involved in the catabolism of tryptophan resulting in inhibition of T lymphocyte function. While inhibition of IDO enzymatic activity results in tumor rejection, it is still unknown how we can directly target IDO expression within tumors using drugs. We have chosen to interfere with IDO expression by targeting the key-signaling event signal transducer and activator of transcription 1 (STAT1). We evaluated the efficacy of fludarabine, previously described to inhibit STAT1 phosphorylation. Interestingly, fludarabine was efficient in suppressing protein expression and consequently IDO activity in two different cell lines derived from breast cancer and melanoma when IDO was activated with interferon-gamma (IFN-γ) or supernatants prepared from activated T lymphocytes. However, fludarabine had no inhibitory effect on STAT1 phosphorylation. Other IFN-γ-responsive genes were only marginally inhibited by fludarabine. The level of IDO transcript was unaffected by this inhibitor, suggesting the involvement of post-transcriptional control. Strikingly, we have found that the inhibition of proteasome partially protected IDO from fludarabine-induced degradation, indicating that fludarabine induces IDO degradation through a proteasome-dependent pathway. Currently used in the clinic to treat some malignancies, fludarabine has the potential for use in the treatment of human tumors through induction of IDO degradation and consequently, for the promotion of T cell-mediated anti-tumor response.


Subject(s)
Antineoplastic Agents/toxicity , Down-Regulation/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Proteasome Endopeptidase Complex/metabolism , Vidarabine/analogs & derivatives , B7-H1 Antigen/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/pharmacology , Phosphorylation/drug effects , Protein Stability , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Vidarabine/toxicity
19.
Curr Top Microbiol Immunol ; 375: 47-59, 2014.
Article in English | MEDLINE | ID: mdl-22021043

ABSTRACT

The 'easiest' vaccines, base on production of neutralizing antibodies, have been made. With the emergence of chronic diseases, vaccine developers have understood the importance to trigger an efficient cellular mediated immune response (CTL response) to respond to this medical need. Several options are currently in development and the utilization of plant virus as vaccine platform for the trigger of a CTL response is considered as an interesting avenue. The highly ordered structures of plant viruses are good triggers of the innate immune system, which in turn, is used to initiate an immune response to a vaccine target. It is likely that plant viruses will play an important role in the development of the vaccine of the futures even if there is still several challenges to face.


Subject(s)
Plant Viruses/genetics , Vaccines, Synthetic/immunology , Artificial Gene Fusion , Capsid Proteins/genetics , Epitopes, T-Lymphocyte/immunology , Humans , T-Lymphocytes, Cytotoxic/immunology , Vaccination , Vaccines, Synthetic/genetics
20.
J Immunol ; 192(3): 1071-8, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24376264

ABSTRACT

Developing new adjuvants and vaccination strategies is of paramount importance to successfully fight against many life-threatening infectious diseases and cancer. Very few adjuvants are currently authorized for human use, and these mainly stimulate a humoral response. However, specific Abs are not sufficient to confer protection against persisting infections or cancer. Therefore, development of adjuvants and immunomodulators able to enhance cell-mediated immune responses represents a major medical need. We recently showed that papaya mosaic virus nanoparticles (PapMV), self-assembled from the coat protein of a plant virus and a noncoding ssRNA molecule, are highly immunogenic in mice. PapMV can be used either as a vaccine delivery platform, through fusion of various epitopes to the coat protein or as adjuvant to enhance humoral immune responses against coadministered Ags or vaccines. However, the mechanisms that confer these immunomodulatory properties to PapMV and its ability to enhance T cell vaccines remain unknown. Using immunization studies in mice, we demonstrate in this paper that PapMV represents a novel TLR7 agonist with strong immunostimulatory properties. More importantly, pretreatment with PapMV significantly improves effector and memory CD8(+) T cell responses generated through dendritic cell vaccination increasing protection against a Listeria monocytogenes challenge.


Subject(s)
Adjuvants, Immunologic , CD8-Positive T-Lymphocytes/immunology , Listeria monocytogenes/immunology , Listeriosis/prevention & control , Membrane Glycoproteins/agonists , T-Lymphocyte Subsets/immunology , Toll-Like Receptor 7/agonists , Tymovirus/immunology , Vaccination , Adaptive Immunity , Animals , Dendritic Cells/immunology , Drug Evaluation, Preclinical , Female , Immunoglobulin G/biosynthesis , Immunologic Memory , Interferon Type I/immunology , Listeriosis/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Nanoparticles , Ovalbumin/immunology , RNA, Viral/immunology , Receptor, Interferon alpha-beta/deficiency , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/immunology , Tymovirus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...