Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Clin Transl Sci ; 16(9): 1569-1581, 2023 09.
Article in English | MEDLINE | ID: mdl-37408165

ABSTRACT

Consensus of regulatory decisions on the same Marketing Authorization Application (MAA) are critical for stakeholders. In this context, regulatory decision patterns from the Swissmedic (SMC), the US Food and Drug Administration (FDA), and the European Medicines Agency (EMA) were analyzed for hemato-oncology products (OP) and non-oncology products (NOP). We compared 336 SMC regulatory decisions between 2009 and 2018 on new active substances with the EMA and the FDA for OP (n = 77) and NOP (n = 259) regarding approval rates, consensus, and divergent decisions. For OP MAA, we analyzed the underlying reasons for divergent decisions; for consensus decisions, the similarity and strictness of labeling. For OP, the approval rate for the SMC was 88.4%, the EMA 91.3%, and the FDA 95.7%. For NOP, the SMC had an approval rate of 86.2%, the EMA of 93.8%, and the FDA of 88.8%. The consensus decision rate among agencies was 88.4% for OP and 84.4% for NOP. The main clinical driver for divergent decisions for OP was nonrandomized trial design and low patient numbers. Comparing the approved indication wordings, the highest similarity was between the SMC and the EMA, and lowest for the FDA and the EMA. Investigating label strictness, the FDA numerically had the highest but not-statistically significant number of strict labels. The approval rate stratified by disease area (OP and NOP) differed among the SMC, the EMA, and the FDA. High concordance in regulatory decisions was observed between agencies for OP as well as NOP. Reasons for divergent decisions regarding OP were mainly due to scientific uncertainties. Comparing strictness of indications, numerical but no statistically significant differences were observed between agencies.


Subject(s)
Drug Approval , United States , Humans , United States Food and Drug Administration , Uncertainty , Europe
2.
Arthritis Rheumatol ; 66(9): 2601-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24909430

ABSTRACT

OBJECTIVE: Follicular regulatory T (Tfr) cells act as the regulatory counterpart of follicular helper T (Tfh) cells to suppress germinal center (GC) B cell differentiation. We recently showed that interleukin-21 (IL-21) promoted Tfh cell differentiation in autoimmune BXD2 mice that develop spontaneous GCs. This study was undertaken to determine the modulatory effects of IL-21 on Tfr cells and the Tfr cell to Tfh cell balance in BXD2 mice. METHODS: The percentage and phenotype of Tfr cells were determined in BXD2 and BXD2-IL21(-/-) mice. The effects of IL-21 on Tfr cells and the Tfr cell:Tfh cell ratio were evaluated. Sorted Tfr cells from BXD2-IL21(-/-) mice were cocultured with Tfh cells and B cells, or transferred into BXD2 mice to determine their function. RESULTS: The percentages and numbers of GC B cells and Tfh cells were significantly reduced, but the percentage of Tfr cells was 2-fold higher in BXD2-IL21(-/-) mice than in wild-type BXD2 mice. Administration of AdIL-21 to BXD2-IL21(-/-) mice decreased the percentages and numbers of Tfr cells and the Tfr cell:Tfh cell ratio but increased the number of GC B cells in the spleen. Recombinant murine IL-21 suppressed FoxP3 and significantly reduced Tgfb1, Il2, and Gitr but enhanced Il21, Il6, Pd1, Cxcr5, and Icos expression in Tfr cells. IL-21 also counteracted Tfr cell-mediated inhibition of antibody secretion in the Tfh cell-B cell coculture system. Transfer of Tfr cells into young BXD2 mice reduced GC size and decreased the numbers of autoantibody-producing B cells. CONCLUSION: Our findings indicate that high levels of IL-21 selectively enhance Tfh cell differentiation but inhibit Tfr cell commitment and the suppressive function of Tfr cells on Tfh cells and B cells, suggesting that IL-21 skews the balance from Tfr cells to Tfh cells to promote autoreactive GC reactions in BXD2 mice.


Subject(s)
Cell Differentiation/immunology , Germinal Center/metabolism , Interleukins/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Autoantibodies/immunology , Germinal Center/cytology , Germinal Center/immunology , Interleukins/genetics , Mice , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
3.
PLoS One ; 8(12): e82528, 2013.
Article in English | MEDLINE | ID: mdl-24349306

ABSTRACT

Interleukins (IL) are cytokines with stimulatory and modulatory functions in the immune system. In this study, we have chosen interleukins which are involved in the enhancement of TH2 responses and B cell functions to analyze their potential to improve a prophylactic adenovirus-based anti-retroviral vaccine with regard to antibody and virus-specific CD4(+) T cell responses. Mice were vaccinated with an adenoviral vector which encodes and displays the Friend Virus (FV) surface envelope protein gp70 (Ad.pIXgp70) in combination with adenoviral vectors encoding the interleukins IL4, IL5, IL6, IL7 or IL23. Co-application of Ad.pIXgp70 with Ad.IL5, Ad.IL6 or Ad.IL23 resulted in improved protection with high control over FV-induced splenomegaly and reduced viral loads. Mice co-immunized with adenoviral vectors encoding IL5 or IL23 showed increased neutralizing antibody responses while mice co-immunized with Ad.IL6 or Ad.IL23 showed improved FV-specific CD4(+) T cell responses compared to mice immunized with Ad.pIXgp70 alone. We show that the co-application of adenoviral vectors encoding specific interleukins is suitable to improve the vaccination efficacy of an anti-retroviral vaccine. Improved protection correlated with improved CD4(+) T cell responses and especially with higher neutralizing antibody titers. The co-application of selected interleukin-encoding adenoviral vectors is a valuable tool for vaccination with regard to enhancement of antibody mediated immunity.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Interleukins/genetics , Retroviridae Infections/genetics , Retroviridae Infections/immunology , Retroviridae/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Female , Friend murine leukemia virus/immunology , Genetic Vectors/administration & dosage , Humans , Mice , Retroviridae Infections/pathology , Retroviridae Infections/prevention & control , Splenomegaly , T-Lymphocyte Subsets/immunology , Vaccination , Viral Vaccines/administration & dosage , Virus Replication
4.
PLoS Pathog ; 9(6): e1003391, 2013.
Article in English | MEDLINE | ID: mdl-23785279

ABSTRACT

A potent therapeutic T-cell vaccine may be an alternative treatment of chronic hepatitis B virus (HBV) infection. Previously, we developed a DNA prime-adenovirus (AdV) boost vaccination protocol that could elicit strong and specific CD8+ T-cell responses to woodchuck hepatitis virus (WHV) core antigen (WHcAg) in mice. In the present study, we first examined whether this new prime-boost immunization could induce WHcAg-specific T-cell responses and effectively control WHV replication in the WHV-transgenic mouse model. Secondly, we evaluated the therapeutic effect of this new vaccination strategy in chronically WHV-infected woodchucks in combination with a potent antiviral treatment. Immunization of WHV-transgenic mice by DNA prime-AdV boost regimen elicited potent and functional WHcAg-specific CD8+ T-cell response that consequently resulted in the reduction of the WHV load below the detection limit in more than 70% of animals. The combination therapy of entecavir (ETV) treatment and DNA prime-AdV boost immunization in chronic WHV carriers resulted in WHsAg- and WHcAg-specific CD4+ and CD8+ T-cell responses, which were not detectable in ETV-only treated controls. Woodchucks receiving the combination therapy showed a prolonged suppression of WHV replication and lower WHsAg levels compared to controls. Moreover, two of four immunized carriers remained WHV negative after the end of ETV treatment and developed anti-WHs antibodies. These results demonstrate that the combined antiviral and vaccination approach efficiently elicited sustained immunological control of chronic hepadnaviral infection in woodchucks and may be a new promising therapeutic strategy in patients.


Subject(s)
Adenoviridae , Hepatitis B Vaccines/pharmacology , Hepatitis B, Chronic/prevention & control , Immunity, Cellular/drug effects , Immunization, Secondary , Vaccines, DNA/microbiology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Hepatitis B Vaccines/genetics , Hepatitis B Vaccines/immunology , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/immunology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Marmota , Mice , Mice, Transgenic , Vaccines, DNA/genetics
5.
J Virol ; 87(13): 7708-16, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23637419

ABSTRACT

Hepatitis D virus (HDV) superinfection of hepatitis B virus (HBV) carriers causes severe liver disease and a high rate of chronicity. Therefore, a vaccine protecting HBV carriers from HDV superinfection is needed. To protect from HDV infection an induction of virus-specific T cells is required, as antibodies to the two proteins of HDV, p24 and p27, do not neutralize the HBV-derived envelope of HDV. In mice, HDV-specific CD8(+) and CD4(+) T cell responses were induced by a DNA vaccine expressing HDV p27. In subsequent experiments, seven naive woodchucks were immunized with a DNA prime and adenoviral boost regimen prior to simultaneous woodchuck hepatitis virus (WHV) and HDV infection. Five of seven HDV-immunized woodchucks were protected against HDV infection, while acute self-limiting WHV infection occurred as expected. The two animals with the breakthrough had a shorter HDV viremia than the unvaccinated controls. The DNA prime and adenoviral vector boost vaccination protected woodchucks against HDV infection in the setting of simultaneous infection with WHV and HDV. In future experiments, the efficacy of this protocol to protect from HDV infection in the setting of HDV superinfection will need to be proven.


Subject(s)
Hepatitis B Virus, Woodchuck/immunology , Hepatitis B/complications , Hepatitis D/prevention & control , Hepatitis Delta Virus/immunology , Superinfection/prevention & control , T-Lymphocytes/immunology , Adenoviridae , Animals , Antibodies, Viral/blood , Aspartate Aminotransferases/metabolism , Blotting, Western , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors/immunology , Hepatitis D/complications , Immunization, Secondary , Marmota , Mice , Plasmids/genetics , Superinfection/virology , Viral Vaccines/genetics , Viral Vaccines/immunology
6.
Virol J ; 10: 108, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23560981

ABSTRACT

BACKGROUND: Inert nanoparticles are attracting attention as carriers for protein-based vaccines. Here we evaluate the immunogenicity of the model antigen ovalbumin delivered on polystyrene particles and directly compare particulate delivery with adenovirus-based immunization. FINDINGS: Mice were vaccinated with soluble ovalbumin, ovalbumin-coated polystyrene particles of different sizes, or an adenovirus-based expression-display vector that encodes and displays a pIX-ovalbumin fusion protein. Antibody responses were clearly higher when ovalbumin was administered on polystyrene particles compared to soluble protein administration, regardless of the particle size. Compared to adenovirus-based immunization, antibody levels were lower if an equivalent amount of protein was delivered, and no cellular immune response was detectable. CONCLUSIONS: We demonstrate in a side-by-side comparison that inert nanoparticles allow for the reduction of the administered antigen amount compared to immunization with soluble protein and induce strongly enhanced antibody responses, but responses are lower compared to adenovirus-based immunization.


Subject(s)
Adenoviridae/chemistry , Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Nanoparticles/chemistry , Vaccination/methods , Vaccines/administration & dosage , Vaccines/immunology , Animals , Antigens/administration & dosage , Antigens/immunology , Drug Carriers/chemistry , Mice , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Ovalbumin/immunology , Polystyrenes
7.
J Virol ; 86(17): 9297-310, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22718818

ABSTRACT

Induction of hepatitis B virus (HBV)-specific cytotoxic T cells by therapeutic immunization may be a strategy to treat chronic hepatitis B. In the HBV animal model, woodchucks, the application of DNA vaccine expressing woodchuck hepatitis virus (WHV) core antigen (WHcAg) in combination with antivirals led to the prolonged control of viral replication. However, it became clear that the use of more potent vaccines is required to overcome WHV persistence. Therefore, we asked whether stronger and more functional T-cell responses could be achieved using the modified vaccines and an optimized prime-boost vaccination regimen. We developed a new DNA plasmid (pCGWHc) and recombinant adenoviruses (AdVs) showing high expression levels of WHcAg. Mice vaccinated with the improved plasmid pCGWHc elicited a stronger WHcAg-specific CD8(+) T-cell response than with the previously used vaccines. Using multicolor flow cytometry and an in vivo cytotoxicity assay, we showed that immunization in a DNA prime-AdV boost regimen resulted in an even more vigorous and functional T-cell response than immunization with the new plasmid alone. Immunization of naïve woodchucks with pCGWHc plasmid or AdVs induced a significant WHcAg-specific degranulation response prior to the challenge, this response had not been previously detected. Consistently, this response led to a rapid control of infection after the challenge. Our results demonstrate that high antigen expression levels and the DNA prime-AdV boost immunization improved the T-cell response in mice and induced significant T-cell responses in woodchucks. Therefore, this new vaccination strategy may be a candidate for a therapeutic vaccine against chronic HBV infection.


Subject(s)
Adenoviridae/immunology , Hepatitis B Core Antigens/immunology , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Hepatitis B/immunology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Adenoviridae/genetics , Animals , Cells, Cultured , Disease Models, Animal , Female , Hepatitis B/virology , Hepatitis B Core Antigens/administration & dosage , Hepatitis B Core Antigens/genetics , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/genetics , Hepatitis B virus/genetics , Humans , Immunization, Secondary , Marmota , Mice , Mice, Inbred C57BL , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
8.
J Virol ; 86(3): 1706-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22090142

ABSTRACT

Processing and presentation of vaccine antigens by professional antigen-presenting cells (APCs) is of great importance for the efficient induction of protective immunity. We analyzed whether the efficacy of an adenovirus-based retroviral vaccine can be enhanced by coadministration of adenovirus-encoded chemokines that attract and stimulate APCs. In the Friend retrovirus (FV) mouse model we coexpressed CCL3, CCL20, CCL21, or CXCL14 from adenoviral vectors, together with FV Gag and Env antigens, and then analyzed immune responses and protection from pathogenic FV infection. Although most tested chemokines did not improve protection against FV challenge, mice that received adenoviral vectors encoding CCL3 together with FV antigens showed significantly better control over viral loads and FV-induced disease than mice immunized with the viral antigens only. Improved protection correlated with enhanced virus-specific CD4+ T cell responses and higher neutralizing antibody titers. To apply these results to an HIV vaccine, mice were immunized with adenoviral vectors encoding the HIV antigens Env and Gag-Pol and coadministered vectors encoding CCL3. Again, this combination vaccine induced higher virus-specific antibody titers and CD4+ T cell responses than did the HIV antigens alone. These results indicate that coexpression of the chemokine CCL3 by adenovirus-based vectors may be a promising tool to improve antiretroviral vaccination strategies.


Subject(s)
Adenoviridae/immunology , Chemokine CCL3/administration & dosage , Retroviridae Infections/prevention & control , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Cell Line , Humans , Mice
9.
Retrovirology ; 8: 75, 2011 Sep 26.
Article in English | MEDLINE | ID: mdl-21943056

ABSTRACT

BACKGROUND: Type I interferons (IFNs) exhibit direct antiviral effects, but also distinct immunomodulatory properties. In this study, we analyzed type I IFN subtypes for their effect on prophylactic adenovirus-based anti-retroviral vaccination of mice against Friend retrovirus (FV) or HIV. RESULTS: Mice were vaccinated with adenoviral vectors encoding FV Env and Gag proteins alone or in combination with vectors encoding IFNα1, IFNα2, IFNα4, IFNα5, IFNα6, IFNα9 or IFNß. Only the co-administration of adenoviral vectors encoding IFNα2, IFNα4, IFNα6 and IFNα9 resulted in strongly improved immune protection of vaccinated mice from subsequent FV challenge infection with high control over FV-induced splenomegaly and reduced viral loads. The level of protection correlated with augmented virus-specific CD4(+) T cell responses and enhanced antibody titers. Similar results were obtained when mice were vaccinated against HIV with adenoviral vectors encoding HIV Env and Gag-Pol in combination with various type I IFN encoding vectors. Here mainly CD4(+) T cell responses were enhanced by IFNα subtypes. CONCLUSIONS: Our results indicate that certain IFNα subtypes have the potential to improve the protective effect of adenovirus-based vaccines against retroviruses. This correlated with augmented virus-specific CD4(+) T cell and antibody responses. Thus, co-expression of select type I IFNs may be a valuable tool for the development of anti-retroviral vaccines.


Subject(s)
Antigens, Viral/immunology , HIV Infections/prevention & control , Interferon Type I/immunology , Retroviridae Infections/prevention & control , Viral Vaccines/immunology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antibodies, Viral/immunology , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , Cell Line , Female , Friend murine leukemia virus/genetics , Friend murine leukemia virus/immunology , Friend murine leukemia virus/physiology , Genetic Vectors/genetics , Genetic Vectors/metabolism , HIV/genetics , HIV/immunology , HIV/physiology , HIV Infections/immunology , HIV Infections/virology , Humans , Interferon Type I/administration & dosage , Interferon Type I/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Retroviridae Infections/immunology , Retroviridae Infections/virology , Viral Load , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
10.
J Virol ; 84(4): 1967-76, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20007267

ABSTRACT

We present a new type of adenoviral vector that both encodes and displays a vaccine antigen on the capsid, thus combining in itself gene-based and protein vaccination; this vector resulted in an improved vaccination outcome in the Friend virus (FV) model. For presentation of the envelope protein gp70 of Friend murine leukemia virus on the adenoviral capsid, gp70 was fused to the adenovirus capsid protein IX. When compared to vaccination with conventional FV Env- and Gag-encoding adenoviral vectors, vaccination with the adenoviral vector that encodes and displays pIX-gp70 combined with an FV Gag-encoding vector resulted in significantly improved protection against systemic FV challenge infection, with highly controlled viral loads in plasma and spleen. This improved protection correlated with improved neutralizing antibody titers and stronger CD4(+) T-cell responses. Using a vector that displays gp70 without encoding it, we found that while the antigen display on the capsid alone was sufficient to induce high levels of binding antibodies, in vivo expression was necessary for the induction of neutralizing antibodies. This new type of adenovirus-based vaccine could be a valuable tool for vaccination.


Subject(s)
Adenoviridae/genetics , Adenoviridae/immunology , Antigens, Viral/genetics , Friend murine leukemia virus/genetics , Friend murine leukemia virus/immunology , Vaccination , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Female , Genetic Vectors , Humans , Leukemia, Experimental/immunology , Leukemia, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Tumor Virus Infections/immunology , Tumor Virus Infections/prevention & control , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
11.
Virology ; 395(2): 243-54, 2009 Dec 20.
Article in English | MEDLINE | ID: mdl-19854461

ABSTRACT

Major strategies to increase oncolytic adenovirus efficacy, as required for clinical applications, are enhancing oncolysis by acceleration of virus release/spread and "arming" by insertion of therapeutic genes. We investigated whether these strategies can be effectively combined as it has been speculated that the arming approach rather benefits from delayed cell lysis and extended time for protein synthesis. We report that deleting adenoviral E1B19K results in an apoptosis-dependent early viral release and thus enhanced oncolysis in several tumor cells, but inhibits virus replication in others. In the former case, apoptosis induction and early cell lysis did not interfere with late transgene expression. Thus, transgene expression was dramatically increased over time due to better virus spread. In A549 cells, transgene expression by the E1B19K(-) virus at 5 days post-infection was higher than for the E1B19K(+) virus at 10 days. These properties may be of critical importance in patients, where time for virus spread is limited.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenoviridae/physiology , Carrier Proteins/metabolism , Gene Expression Regulation, Viral/physiology , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Carrier Proteins/genetics , Gene Deletion , Organisms, Genetically Modified , Viral Proteins/genetics
12.
Cancer Res ; 69(12): 5115-25, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19491256

ABSTRACT

We studied the susceptibility of primary ovarian cancer cells to oncolytic adenoviruses. Using gene expression profiling of cancer cells either resistant or susceptible to viral oncolysis, we discovered that the epithelial phenotype of ovarian cancer represents a barrier to infection by commonly used oncolytic adenoviruses targeted to coxsackie-adenovirus receptor or CD46. Specifically, we found that these adenovirus receptors were trapped in tight junctions and not accessible for virus binding. Accessibility to viral receptors was critically linked to depolarization and the loss of tight and adherens junctions, both hallmarks of epithelial-to-mesenchymal transition (EMT). We showed that specific, thus far little-explored adenovirus serotypes (Ad3, Ad7, Ad11, and Ad14) that use receptor(s) other than coxsackie-adenovirus receptor and CD46 were able to trigger EMT in epithelial ovarian cancer cells and cause efficient oncolysis. Our studies on ovarian cancer cultures and xenografts also revealed several interesting cancer cell biology features. Tumors in situ as well as tumor xenografts in mice mostly contained epithelial cells and cells that were in a hybrid stage where they expressed both epithelial and mesenchymal markers (epithelial/mesenchymal cells). These epithelial/mesenchymal cells are the only xenograft-derived cells that can be cultured and with passaging undergo EMT and differentiate into mesenchymal cells. Our study provides a venue for improved virotherapy of cancer as well as new insights into cancer cell biology.


Subject(s)
Adenoviridae/physiology , Epithelial Cells/pathology , Oncolytic Virotherapy , Ovarian Neoplasms/pathology , Adenoviridae/genetics , Animals , Cell Line, Tumor , Female , Genetic Vectors , Humans , Mice , Mice, SCID , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/virology , Phenotype , Receptors, Virus/metabolism , Tight Junctions/metabolism
13.
Vaccine ; 26(29-30): 3662-72, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18538453

ABSTRACT

Exosomes have been proposed as candidates for therapeutic immunization. The present study demonstrates that incorporation of the G protein of vesicular stomatitis virus (VSV-G) into exosome-like vesicles (ELVs) enhances their uptake and induces the maturation of dendritic cells. Targeting of VSV-G and ovalbumin as a model antigen to the same ELVs increased the cross-presentation of ovalbumin via an endosomal acidification mechanism. Immunization of mice with VSV-G and ovalbumin containing ELVs led to an increased IgG2a antibody response, expansion of antigen-specific CD8 T cells, strong in vivo CTL responses, and protection from challenge with ovalbumin expressing tumor cells. Thus, incorporation of VSV-G and targeting of antigens to ELVs are attractive strategies to improve exosomal vaccines.


Subject(s)
Dendritic Cells/immunology , Membrane Glycoproteins/immunology , Secretory Vesicles/immunology , Vaccines/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies/blood , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Endosomes/metabolism , Humans , Mice , Mice, Inbred C57BL , Neoplasms/prevention & control , Ovalbumin/immunology , Protein Transport
14.
World J Gastroenterol ; 14(12): 1842-50, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18350621

ABSTRACT

AIM: To promote the development of improved tumor vaccination strategies relying on the intratumoral expression of viral fusogenic membrane proteins, we elucidated whether the size of syncytia or the way tumor cells die has an effect on the therapeutic outcome. METHODS: In two syngeneic subcutaneous murine colon cancer models we assessed the anti-neoplastic effect on vector-treated and contralateral untreated tumors. RESULTS: Intratumoral injection of a replication-defective adenovirus encoding respiratory syncytial virus fusion protein (RSV-F) alone (Ad.RSV-F) or together with the attachment glycoprotein RSV-G (Ad.RSV-F/G) led to a significant growth reduction of the vector-treated and contralateral untreated tumors. The treatment response was associated with a strong tumor-specific CTL response and significantly improved survival with medians of 46 d and 44 d, respectively. Intratumoral injection of Ad.RSV-G or a soluble RSV-F encoding adenovirus (Ad.RSV-F(sol)) had no significant anti-neoplastic effect. The median survival of these treatment groups and of Ad.Null-treated control animals was about 30 d. CONCLUSION: Although in vitro transduction of colon cancer cell lines with Ad.RSV-F/G resulted in about 8-fold larger syncytia than with Ad.RSV-F, the in vivo outcome was not significantly different. Transduction of murine colon cancer cell lines with Ad.RSV-F or Ad.RSV-F/G caused apoptotic cell death, in contrast to transduction with Ad.RSV-G or Ad.RSV-F(sol), suggesting an importance of the mode of cell death. Overall, these findings provide insight into improved tumor vaccination strategies relying on the intratumoral expression of viral fusogenic membrane proteins.


Subject(s)
Apoptosis/physiology , Giant Cells , Respiratory Syncytial Viruses/metabolism , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antineoplastic Agents/metabolism , Cell Fusion , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/metabolism , Giant Cells/cytology , Giant Cells/physiology , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics
15.
Vaccine ; 26(5): 716-26, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18160188

ABSTRACT

We evaluated the suitability of the Friend Virus (FV) model for the development of improved adenovirus vectors for anti-retroviral vaccination using two types of adenovirus vectors, encoding F-MuLV Env and Gag, which differed only in their fiber genes (Ad5 and Ad5F35). Genetically FV-resistant C57BL/6 mice and highly susceptible CB6F1 hybrid mice were vaccinated by either homologous or heterologous prime-boost regimen. After FV challenge, viral loads in the spleens of C57BL/6 mice were reduced approximately 250-fold and were below the detection threshold in >50% of the mice. Vaccination outcome was critically influenced by the route of vector administration. In CB6F1 mice, vaccination resulted in reduced viremia, delayed onset of splenomegaly, and induction of FV-specific T cells as assessed by tetramer staining. Heterologous prime-boost vaccination resulted in significantly higher neutralizing antibody titers, translating into improved immune protection, in contrast to coexpression of cytokines. Our results suggest that the FV model can provide insight into the development of improved adenovirus vectors for HIV-1 vaccination.


Subject(s)
AIDS Vaccines/administration & dosage , Adenoviridae/genetics , Friend murine leukemia virus/genetics , Friend murine leukemia virus/immunology , Genetic Vectors , Retroviridae Infections/prevention & control , Tumor Virus Infections/prevention & control , Vaccination , AIDS Vaccines/immunology , Adenoviridae/immunology , Animals , Antibodies, Viral/blood , Cytokines/metabolism , Disease Models, Animal , Female , Friend murine leukemia virus/isolation & purification , Genes, env , Genes, gag , Humans , Immunization Schedule , Injections, Intradermal , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Neutralization Tests , Reassortant Viruses , Retroviridae Infections/immunology , Retroviridae Infections/virology , Species Specificity , Spleen/virology , T-Lymphocytes/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
16.
J Med Virol ; 80(2): 294-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18098135

ABSTRACT

Disseminated adenovirus (HAdV) infections are serious complications in allogenic stem cell transplant (SCT) recipients. Quantitative HAdV DNA detection in blood samples demonstrated the association of high virus loads with disease and improved early diagnosis. However, the pathogenesis of disseminated HAdV disease, for example sources of HAdV DNA shedding in the blood stream and association of HAdV replication sites with disease manifestations, remained obscure. In this report, 24 bioptic and autoptic organ and tissue samples of an adult SCT recipient suffering from disseminated infection were quantitatively analyzed for HAdV DNA. Results indicate subsequent virus replication in the colon, bone marrow and liver as origin of HAdV DNAemia, which increased from 1.4 x 10(4) copies/ml to a peak of 2 x 10(9) copies/ml over a period of 84 days in spite of antiviral therapy. Symptoms as diarrhoea, bone marrow failure and hepatic failure were clearly linked to high HAdV DNA concentrations in affected organs. For example, the HAdV DNA level was 2.2 x 10(3) copies/cell in a colon biopsy when the patient suffered from diarrhoea whereas only 1.1 x 10(1) copies/cell were detected when symptoms had improved. Focal HAdV infection of the liver as demonstrated by laser microdissection was followed by fulminant virus replication with 1.3 x 10(5) copies of HAdV DNA/cell causing terminal hepatic failure. In conclusion, pathogenesis of disseminated HAdV disease was associated with virus replication in affected organs and not immune mediated as suggested recently by a fatal case of gene therapy with a non-replication competent HAdV-C5 vector.


Subject(s)
Adenovirus Infections, Human/complications , Adenovirus Infections, Human/virology , Adenoviruses, Human/isolation & purification , Viral Load , Viremia/virology , Adenoviruses, Human/genetics , Adult , Bone Marrow/virology , Colon/virology , DNA, Viral/genetics , Diarrhea/virology , Humans , Liver/virology , Male , Multiple Organ Failure/virology , Stem Cell Transplantation/adverse effects , Transplantation, Homologous/adverse effects
17.
J Invest Dermatol ; 128(4): 988-98, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17960177

ABSTRACT

Advanced melanoma is associated with poor prognosis warranting the development of new therapeutics, such as oncolytic adenoviruses for immunovirotherapy. Since this approach critically depends on efficient transduction of targeted tumor cells, we screened a panel of 22 different adenovirus types for their internalization efficiency in melanoma cells. We demonstrated that the virions of Ad35, Ad38, and Ad3 have significantly higher internalization efficiency in melanoma cells than Ad5, so far the only adenovirus type used in clinical trials for melanoma. Therefore, we developed a conditionally replication-competent Ad5-based vector with the Ad35 fiber shaft and knob domains (Ad5/35) and compared its therapeutic efficacy with the homologous vector carrying the native Ad5 fiber. To further enhance virotherapy, we combined the oncolytic adenovirus vectors with intratumoral expression of measles virus fusogenic membrane glycoproteins H and F (MV-H/F) and dacarbazine chemotherapy. In a human melanoma xenograft model, established from a short-term culture of primary melanoma cells, we demonstrated that the Ad5/35-based therapy had a significantly greater anti-neoplastic effect than the homologous Ad5-based therapy. Furthermore, the combination of virotherapy, intratumoral expression of MV-H/F, and chemotherapy was clearly superior to single- or double-agent therapy. In conclusion, Ad35-based vectors are promising for the treatment of melanoma.


Subject(s)
Adenoviruses, Human , Genetic Vectors , Melanoma/therapy , Oncolytic Virotherapy , Skin Neoplasms/therapy , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Combined Modality Therapy , Dacarbazine/therapeutic use , Genetic Vectors/genetics , Humans , Melanoma/drug therapy , Membrane Cofactor Protein/analysis , Mice , Mice, Nude , Recoverin/analysis , Skin Neoplasms/drug therapy , Viral Fusion Proteins/genetics , Viral Proteins/genetics , Virus Internalization , Virus Replication , Xenograft Model Antitumor Assays
18.
Int J Mol Med ; 20(5): 673-81, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17912460

ABSTRACT

We assessed whether intratumoral expression of the fusogenic membrane protein of vesicular stomatitis virus (VSV-G), encoded by a replication-defective adenovirus vector (Ad.VSV-G), alone or in combination with local coexpression of cytokines induces tumor-specific immune responses in a syngeneic murine colon cancer model. We confirmed in vitro by dye colocalization that transduction of murine cells with Ad.VSV-G induces cell-cell fusion. In a bilateral syngeneic subcutaneous colon cancer model in C57BL/6 and BALB/c mice, we demonstrated that intratumoral injection of Ad.VSV-G leads to a significant growth reduction of the directly vector-treated tumor, but also of the contralateral not directly vector-treated tumor. When compared to monotherapy, the anti-neoplastic efficacy was significantly enhanced when intratumoral Ad.VSV-G administration was combined with adenovirus vectors encoding IL-2, IL-12, IL-18, IL-21, or GM-CSF. The anti-tumor effects of the first three cytokines in combination with VSV-G expression were somewhat greater than those of the latter two. However, the differences did not reach statistical significance. The combination therapy resulted also in a significantly enhanced survival when compared to monotherapy. In addition, we demonstrated that intratumoral expression of VSV-G in combination with the tested cytokines induced a strong tumor-specific cytotoxic T lymphocyte (CTL) response and infiltration of tumors with macrophages. The effects of the combination therapy were clearly greater than those of the monotherapy. Our experimental data indicate that intratumoral expression of VSV-G, particularly in combination with cytokines, is a promising novel tool for the development of in situ tumor vaccination approaches.


Subject(s)
Adenoviridae/metabolism , Cytokines/metabolism , Genetic Vectors/metabolism , Immunotherapy , Membrane Glycoproteins/metabolism , Neoplasms/immunology , Neoplasms/therapy , Viral Envelope Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Cancer Vaccines , Cell Fusion , Cell Line, Tumor , Cell Movement/drug effects , Colon/drug effects , Colon/pathology , Female , Giant Cells/drug effects , Giant Cells/pathology , Humans , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Splenomegaly/pathology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
19.
Virology ; 369(1): 131-42, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-17727912

ABSTRACT

Oncolytic adenoviruses as a treatment for cancer have demonstrated limited clinical activity. Contributing to this may be the relevance of preclinical animal models used to study these agents. Syngeneic mouse tumor models are generally non-permissive for adenoviral replication, whereas human tumor xenograft models exhibit attenuated immune responses to the vector. The cotton rat (Sigmodon hispidus) is susceptible to human adenovirus infection, permissive for viral replication and exhibits similar inflammatory pathology to humans with adenovirus replicating in the lungs, respiratory passages and cornea. We evaluated three transplantable tumorigenic cotton rat cell lines, CCRT, LCRT and VCRT as models for the study of oncolytic adenoviruses. All three cells lines were readily infected with adenovirus type-5-based vectors and exhibited high levels of transgene expression. The cell lines supported viral replication demonstrated by the induction of cytopathogenic effect (CPE) in tissue culture, increase in virus particle numbers and assembly of virions seen on transmission electron microscopy. In vivo, LCRT and VCRT tumors demonstrated delayed growth after injection with replicating adenovirus. No in vivo antitumor activity was seen in CCRT tumors despite in vitro oncolysis. Adenovirus was also rapidly cleared from the CCRT tumors compared to LCRT and VCRT tumors. The effect observed with the different cotton rat tumor cell lines mimics the variable results of human clinical trials highlighting the potential relevance of this model for assessing the activity and toxicity of oncolytic adenoviruses.


Subject(s)
Adenoviridae/growth & development , Disease Models, Animal , Neoplasms/therapy , Neoplasms/virology , Oncolytic Virotherapy/methods , Animals , Cell Line, Tumor , Cytopathogenic Effect, Viral , Microscopy, Electron, Transmission , Rats , Sigmodontinae , Transplantation, Isogeneic , Viral Plaque Assay , Virion/ultrastructure
20.
Mol Cancer Ther ; 6(7): 1942-50, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620425

ABSTRACT

Although cancers can naturally elicit immune responses, immune ignorance is a common observation preventing immune-mediated elimination of tumor cells. We assessed whether intratumoral expression of respiratory syncytial virus fusion (RSV-F) protein, encoded by a replication-defective adenovirus vector (Ad.RSV-F), alone or in combination with local coexpression of cytokines can induce tumor-specific immune responses in a syngeneic murine colon cancer model. We confirmed in vitro by dye colocalization that transduction of murine cells with Ad.RSV-F induces cell-cell fusion. In vivo, we showed in a bilateral syngeneic s.c. colon cancer model in C57BL/6 and BALB/c mice that intratumoral injection of Ad.RSV-F leads to a significant volume reduction not only of the directly vector-treated tumor but also of the contralateral not directly vector-treated tumor. The intratumoral administration of Ad.RSV-F in combination with adenovirus vectors encoding interleukin (IL)-2, IL-12, IL-18, IL-21, or granulocyte macrophage colony-stimulating factor significantly enhanced the antitumor effect on the directly vector-treated tumor and also on the contralateral tumor. The antineoplastic efficacy of this combined treatment was significantly higher than that of the individual treatment components and was associated with the induction of a tumor-specific CTL response and increased infiltration of the tumors by natural killer cells and macrophages. Intratumoral coexpression of RSV-F and IL-21 resulted in the highest tumor growth inhibition and improved survival. Our experimental data indicate that intratumoral expression of RSV-F in combination with cytokines is a promising novel tool for the development of in situ tumor vaccination approaches.


Subject(s)
Adenoviridae/genetics , Cancer Vaccines/immunology , Colorectal Neoplasms/prevention & control , Cytokines/genetics , Respiratory Syncytial Viruses , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Animals , Antineoplastic Agents/pharmacology , Cancer Vaccines/genetics , Cell Fusion , Colorectal Neoplasms/immunology , Cytokines/immunology , Female , Genetic Vectors/genetics , HT29 Cells , Humans , Killer Cells, Natural/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Splenomegaly/pathology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...