Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Med Primatol ; 53(1): e12690, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38345331

ABSTRACT

BACKGROUND: Cervical cancer is an abnormal growth of cervical tissue epithelial cells due to persistent human papilloma virus (HPV) infection. Cynomolgus monkeys (Macaca fascicularis) can be naturally and spontaneously infected with M. fascicularis Papillomavirus Type 3 (MfPV3), a virus that is phylogenetically closely related to human oncogenic HPV (HPV-16 and HPV-34), and therefore a potentially beneficial for modeling HPV disease. This study aims to evaluate the expression of the integrin alpha 6 (ITGα6) receptor in cynomolgus monkeys spontaneously infected with MfPV3, which this receptor also found in human infected with HPV. METHODS: The study was done on archived Formalin-fixed Paraffin-Embedded (FFPE) samples of uterine and cervix tissue of cynomolgus monkeys. Immunohistochemistry was also performed to quantify the expression levels of ITGα6. RESULTS: The results showed 80% of the samples positive Cervical Intraepithelial Neoplasia (CIN) and increased expression of ITGα6 significantly in Positive-MfPV3 group than negative-MfPV3 group. CONCLUSIONS: This indicated the potential of cynomolgus monkeys as a spontaneous oncogenesis model of PV infection type.


Subject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Animals , Cervix Uteri/metabolism , Macaca fascicularis , Papillomavirus Infections/veterinary , Uterine Cervical Neoplasms/chemistry , Uterine Cervical Neoplasms/metabolism , Papillomaviridae , Integrins/analysis
2.
Int J Mol Sci ; 24(12)2023 Jun 09.
Article in English | MEDLINE | ID: mdl-37373123

ABSTRACT

Expression of human endogenous retrovirus type W (HERV-W) has been linked to cancer, making HERV-W antigens potential targets for therapeutic cancer vaccines. In a previous study, we effectively treated established tumours in mice by using adenoviral-vectored vaccines targeting the murine endogenous retrovirus envelope and group-specific antigen (Gag) of melanoma-associated retrovirus (MelARV) in combination with anti-PD-1. To break the immunological tolerance to MelARV, we mutated the immunosuppressive domain (ISD) of the MelARV envelope. However, reports on the immunogenicity of the HERV-W envelope, Syncytin-1, and its ISD are conflicting. To identify the most effective HERV-W cancer vaccine candidate, we evaluated the immunogenicity of vaccines encoding either the wild-type or mutated HERV-W envelope ISD in vitro and in vivo. Here, we show that the wild-type HERV-W vaccine generated higher activation of murine antigen-presenting cells and higher specific T-cell responses than the ISD-mutated counterpart. We also found that the wild-type HERV-W vaccine was sufficient to increase the probability of survival in mice subjected to HERV-W envelope-expressing tumours compared to a control vaccine. These findings provide the foundation for developing a therapeutic cancer vaccine targeting HERV-W-positive cancers in humans.


Subject(s)
Cancer Vaccines , Endogenous Retroviruses , Neoplasms , Humans , Animals , Mice , Endogenous Retroviruses/genetics , T-Lymphocytes , Immunosuppression Therapy
3.
Cell Mol Life Sci ; 78(21-22): 6963-6978, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34586443

ABSTRACT

The endogenous chemokines CCL19 and CCL21 signal via their common receptor CCR7. CCL21 is the main lymph node homing chemokine, but a weak chemo-attractant compared to CCL19. Here we show that the 41-amino acid positively charged peptide, released through C-terminal cleavage of CCL21, C21TP, boosts the immune cell recruiting activity of CCL21 by up to 25-fold and the signaling activity via CCR7 by ~ 100-fold. Such boosting is unprecedented. Despite the presence of multiple basic glycosaminoglycan (GAG) binding motifs, C21TP boosting of CCL21 signaling does not involve interference with GAG mediated cell-surface retention. Instead, boosting is directly dependent on O-glycosylations in the CCR7 N-terminus. As dictated by the two-step binding model, the initial chemokine binding involves interaction of the chemokine fold with the receptor N-terminus, followed by insertion of the chemokine N-terminus deep into the receptor binding pocket. Our data suggest that apart from a role in initial chemokine binding, the receptor N-terminus also partakes in a gating mechanism, which could give rise to a reduced ligand activity, presumably through affecting the ligand positioning. Based on experiments that support a direct interaction of C21TP with the glycosylated CCR7 N-terminus, we propose that electrostatic interactions between the positively charged peptide and sialylated O-glycans in CCR7 N-terminus may create a more accessible version of the receptor and thus guide chemokine docking to generate a more favorable chemokine-receptor interaction, giving rise to the peptide boosting effect.


Subject(s)
Chemokine CCL21/metabolism , Dendritic Cells/metabolism , Lymph Nodes/metabolism , Receptors, CCR7/metabolism , Receptors, Lymphocyte Homing/metabolism , Signal Transduction/physiology , Animals , CHO Cells , Cells, Cultured , Cricetulus , Glycosylation , Humans , Ligands , Peptides/metabolism , Protein Binding/physiology , Static Electricity
4.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163254

ABSTRACT

Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections that have become fixed in the human genome. While HERV genes are typically silenced in healthy somatic cells, there are numerous reports of HERV transcription and translation across a wide spectrum of cancers, while T and B cell responses against HERV proteins have been detected in cancer patients. This review systematically categorizes the published evidence on the expression of and adaptive immune response against specific HERVs in distinct cancer types. A systematic literature search was performed using Medical Search Headings (MeSH) in the PubMed/Medline database. Papers were included if they described the translational activity of HERVs. We present multiple tables that pair the protein expression of specific HERVs and cancer types with information on the quality of the evidence. We find that HERV-K is the most investigated HERV. HERV-W (syncytin-1) is the second-most investigated, while other HERVs have received less attention. From a therapeutic perspective, HERV-K and HERV-E are the only HERVs with experimental demonstration of effective targeted therapies, but unspecific approaches using antiviral and demethylating agents in combination with chemo- and immunotherapies have also been investigated.


Subject(s)
Antibody Formation/immunology , Endogenous Retroviruses/immunology , Neoplasms/immunology , Neoplasms/therapy , Retroviridae Infections/immunology , Viral Proteins/immunology , Animals , Host-Pathogen Interactions/immunology , Humans
5.
J Immunol ; 202(8): 2320-2331, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30833346

ABSTRACT

Adenoviral vectors can induce T and B cell immune responses to Ags encoded in the recombinant vector. The MHC class II invariant chain (Ii) has been used as an adjuvant to enhance T cell responses to tethered Ag encoded in adenoviral vectors. In this study, we modified the Ii adjuvant by insertion of a furin recognition site (Ii-fur) to obtain a secreted version of the Ii. To test the capacity of this adjuvant to enhance immune responses, we recombined vectors to encode Plasmodium falciparum virulence factors: two cysteine-rich interdomain regions (CIDR) α1 (IT4var19 and PFCLINvar30 var genes), expressed as a dimeric Ag. These domains are members of a highly polymorphic protein family involved in the vascular sequestration and immune evasion of parasites in malaria. The Ii-fur molecule directed secretion of both Ags in African green monkey cells and functioned as an adjuvant for MHC class I and II presentation in T cell hybridomas. In mice, the Ii-fur adjuvant induced a similar T cell response, as previously demonstrated with Ii, accelerated and enhanced the specific Ab response against both CIDR Ags, with an increased binding capacity to the cognate endothelial protein C receptor, and enhanced the breadth of the response toward different CIDRs. We also demonstrate that the endosomal sorting signal, secretion, and the C-terminal part of Ii were needed for the full adjuvant effect for Ab responses. We conclude that engineered secretion of Ii adjuvant-tethered Ags establishes a single adjuvant and delivery vehicle platform for potent T and B cell-dependent immunity.


Subject(s)
Adenoviridae , Antibodies, Protozoan/immunology , Antibody Formation , Histocompatibility Antigens Class II/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Vaccination , Animals , COS Cells , Chlorocebus aethiops , Female , Humans , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Plasmodium falciparum/genetics
6.
J Transl Med ; 17(1): 175, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31126293

ABSTRACT

BACKGROUND: In non-human primates (NHPs) and humans, partial protection from HIV/SIV infection or suppression of replication is achievable by Env-binding antibodies and Gag-specific CD8+ T-cells targeting protective epitopes. Unfortunately, such T-cell responses are frequently dominated by responses to non-protective, variable epitopes. In this study we attempt to combine three independent approaches, each developed to prevent immunodominance of non-protective epitopes. These approaches were (1) vaccines consisting exclusively of putatively protective p24 Gag highly conserved elements (CEs), (2) vaccines using solely subdominant antigens which were acutely protective in a recent NHP trial, and (3) virus-encoded virus-like particle vaccines (virus-like vaccines/VLVs) using heterologous Env and Gag sequences to enable selection of broadly cross-reactive responses and to avoid immunodominance of non-conserved sequences in prime-boost regimens as previously observed. METHODS: We vaccinated outbred CD1 mice with HIV-1 clade B Gag/Env encoded in an adenoviral prime and SIVmac239 Gag/Env in an MVA boost. We combined this completely heterologous immunization regimen and the homologous SIVmac239 Gag/Env immunization regimen with an additional prime encoding SIV CEs and accessory antigens Rev, Vif and Vpr (Ad-Ii-SIVCErvv). T-cell responses were analyzed by intracellular cytokine staining of splenocytes and antibody responses by trimer-specific ELISA, avidity and isotype-specific ELISA. RESULTS: Env dominance could be avoided successfully in the completely heterologous prime-boost regimen, but Env immunodominance reappeared when Ad-Ii-SIVCErvv was added to the prime. This regimen did however still induce more cross-reactive Gag-specific CD8+ T-cells and Env-specific antibodies. Including Ad-Ii-SIVCErvv in the homologous prime-boost not only elicited accessory antigen-specific CD8+ memory T-cells, but also significantly increased the ratio of Gag- to Env-specific CD8+ T-cells. The CD4+ T-cell response shifted away from structural antigens previously associated with infection-enhancement. CONCLUSION: The homologous Gag/Env prime-boost with Ad-Ii-SIVCErvv prime combined acutely protective CD8+ T-cell responses to subdominant antigens and Env-binding antibodies with chronically protective Gag-specific CD8+ T-cells in outbred mice. This vaccine regimen should be tested in an NHP efficacy trial.


Subject(s)
AIDS Vaccines/immunology , Antigens, Viral/immunology , HIV Infections/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , T-Lymphocytes/immunology , Adenoviridae/genetics , Animals , Antibody Formation/immunology , Female , Genetic Vectors/metabolism , HIV-1/immunology , Immunization, Secondary , Mice , Simian Immunodeficiency Virus/immunology
7.
Vet Res ; 49(1): 4, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29316978

ABSTRACT

Actinobacillus pleuropneumoniae (A. pleuropneumoniae) is a Gram-negative bacterium that represents the main cause of porcine pleuropneumonia in pigs, causing significant economic losses to the livestock industry worldwide. A. pleuropneumoniae, as the majority of Gram-negative bacteria, excrete vesicles from its outer membrane (OM), accordingly defined as outer membrane vesicles (OMVs). Thanks to their antigenic similarity to the OM, OMVs have emerged as a promising tool in vaccinology. In this study we describe the in vivo testing of several vaccine prototypes for the prevention of infection by all known A. pleuropneumoniae serotypes. Previously identified vaccine candidates, the recombinant proteins ApfA and VacJ, administered individually or in various combinations with the OMVs, were employed as vaccination strategies. Our data show that the addition of the OMVs in the vaccine formulations significantly increased the specific IgG titer against both ApfA and VacJ in the immunized animals, confirming the previously postulated potential of the OMVs as adjuvant. Unfortunately, the antibody response raised did not translate into an effective protection against A. pleuropneumoniae infection, as none of the immunized groups following challenge showed a significantly lower degree of lesions than the controls. Interestingly, quite the opposite was true, as the animals with the highest IgG titers were also the ones bearing the most extensive lesions in their lungs. These results shed new light on A. pleuropneumoniae pathogenicity, suggesting that antibody-mediated cytotoxicity from the host immune response may play a central role in the development of the lesions typically associated with A. pleuropneumoniae infections.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/immunology , Bacterial Vaccines/immunology , Pleuropneumonia/veterinary , Swine Diseases/prevention & control , Actinobacillus Infections/microbiology , Actinobacillus Infections/prevention & control , Actinobacillus pleuropneumoniae/genetics , Animals , Pleuropneumonia/microbiology , Pleuropneumonia/prevention & control , Recombinant Proteins/immunology , Serogroup , Swine , Swine Diseases/microbiology , Vaccination/veterinary
8.
Vet Res ; 48(1): 74, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29122004

ABSTRACT

Despite numerous actions to prevent disease, Actinobacillus pleuropneumoniae (A. pleuropneumoniae) remains a major cause of porcine pleuropneumonia, resulting in economic losses to the swine industry worldwide. In this paper, we describe the utilization of a reverse vaccinology approach for the selection and in vitro testing of serovar-independent A. pleuropneumoniae immunogens. Potential immunogens were identified in the complete genomes of three A. pleuropneumoniae strains belonging to different serovars using the following parameters: predicted outer-membrane subcellular localization; ≤ 1 trans-membrane helices; presence of a signal peptide in the protein sequence; presence in all known A. pleuropneumoniae genomes; homology with other well characterized factors with relevant data regarding immunogenicity/protective potential. Using this approach, we selected the proteins ApfA and VacJ to be expressed and further characterized, both in silico and in vitro. Additionally, we analysed outer membrane vesicles (OMVs) of A. pleuropneumoniae MIDG2331 as potential immunogens, and compared deletions in degS and nlpI for increasing yields of OMVs compared to the parental strain. Our results indicated that ApfA and VacJ are highly conserved proteins, naturally expressed during infection by all A. pleuropneumoniae serovars tested. Furthermore, OMVs, ApfA and VacJ were shown to possess a high immunogenic potential in vitro. These findings favour the immunogen selection protocol used, and suggest that OMVs, along with ApfA and VacJ, could represent effective immunogens for the prevention of A. pleuropneumoniae infections in a serovar-independent manner. This hypothesis is nonetheless predictive in nature, and in vivo testing in a relevant animal model will be necessary to verify its validity.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/immunology , Bacterial Vaccines/immunology , Pleuropneumonia/veterinary , Swine Diseases/prevention & control , Actinobacillus Infections/microbiology , Actinobacillus Infections/prevention & control , Animals , Pleuropneumonia/microbiology , Pleuropneumonia/prevention & control , Swine , Swine Diseases/microbiology , Vaccines, Synthetic/immunology
9.
J Transl Med ; 14(1): 343, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27998269

ABSTRACT

BACKGROUND: A major obstacle for the development of HIV vaccines is the virus' worldwide sequence diversity. Nevertheless, the presence of T cell epitopes within conserved regions of the virus' structural Gag protein and conserved structures in the envelope (env) sequence raises the possibility that cross-reactive responses may be induced by vaccination. In this study, the aim was to investigate the importance of antigenic match on immunodominance and breadth of obtainable T cell responses. METHODS: Outbred CD1 mice were immunized with either heterologous (SIVmac239 and HIV-1 clade B consensus) or homologous (SIVmac239) gag sequences using adenovirus (Ad5) and MVA vectors. Env (SIVmac239) was co-encoded in the vectors to study the induction of antibodies, which is a primary target of current HIV vaccine designs. All three vaccines were designed as virus-encoded virus-like particle vaccines. Antibody responses were analysed by ELISA, avidity ELISA, and neutralization assay. T cell responses were determined by intracellular cytokine staining of splenocytes. RESULTS: The homologous Env/Gag prime-boost regimen induced higher Env binding antibodies, and induced stronger and broader Gag specific CD8+ T cell responses than the homologous Env/heterologous Gag prime-boost regimen. Homologous Env/heterologous Gag immunization resulted in selective boosting of Env specific CD8+ T cell responses and consequently a paradoxical decreased recognition of variant sequences including conserved elements of p24 Gag. CONCLUSIONS: These results contrast with related studies using Env or Gag as the sole antigen and suggest that prime-boost immunizations based on homologous SIVmac239 Gag inserts is an efficient component of genetic VLP vaccines-both for induction of potent antibody responses and cross-reactive CD8+ T cell responses.


Subject(s)
Antibody Formation/immunology , Gene Products, env/immunology , Gene Products, gag/immunology , Genetic Vectors/metabolism , Immunization, Secondary , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Animals, Outbred Strains , Antibodies, Viral , CD8-Positive T-Lymphocytes/immunology , Female , Immunity, Humoral , Interferon-gamma/metabolism , Lymphocyte Count , Mice , Sequence Homology, Amino Acid , Vaccination
10.
Blood ; 124(9): 1450-9, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25037628

ABSTRACT

Although the accumulation of highly-differentiated and granzyme B (GrB)-expressing CD8(+)CD28(-) T cells has been associated with aging, the mechanism for their enrichment and contribution to immune function remains poorly understood. Here we report a novel B-cell subset expressing 4-1BBL, which increases with age in humans, rhesus macaques, and mice, and with immune reconstitution after chemotherapy and autologous progenitor cell transplantation. These cells (termed 4BL cells) induce GrB(+)CD8(+) T cells by presenting endogenous antigens and using the 4-1BBL/4-1BB axis. We found that the 4BL cells increase antitumor responses in old mice, which may explain in part the paradox of retarded tumor growth in the elderly. 4BL cell accumulation and its capacity to evoke the generation of GrB(+)CD8(+) T cells can be eliminated by inducing reconstitution of B cells in old mice, suggesting that the age-associated skewed cellular immune responses are reversible. We propose that 4BL cells and the 4-1BBL signaling pathway are useful targets for improved effectiveness of natural antitumor defenses and therapeutic immune manipulations in the elderly.


Subject(s)
4-1BB Ligand/metabolism , Aging/immunology , B-Lymphocyte Subsets/immunology , CD8-Positive T-Lymphocytes/immunology , Granzymes/metabolism , 4-1BB Ligand/deficiency , 4-1BB Ligand/genetics , Adult , Aged , Aged, 80 and over , Aging/metabolism , Aging/pathology , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/enzymology , Female , Humans , Immunity, Cellular , Immunity, Innate , Macaca mulatta , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
11.
Mol Ther ; 22(12): 2107-2117, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25023330

ABSTRACT

We have previously shown that for the majority of antigens, adenoviral vaccines expressing the target antigen fused to the MHC associated invariant chain (Ii) induce an accelerated, augmented, and prolonged transgene-specific CD8(+) T-cell response. Here we describe a new adenoviral vaccine vector approach where the target antigen fused to Ii is expressed from the adenoviral E1 region and IL-2 is expressed from the E3 region. Immunization of mice with this new vector construct resulted in an augmented primary effector CD8(+) T-cell response. Furthermore, in a melanoma model we observed significantly prolonged tumor control in vaccinated wild type (WT) mice. The improved tumor control required antigen-specific cells, since no tumor control was observed, unless the melanoma cells expressed the vaccine targeted antigen. We also tested our new vaccine in immunodeficient (CD80/86 deficient) mice. Following vaccination with the IL-2 expressing construct, these mice were able to raise a delayed but substantial CD8(+) T-cell response, and to control melanoma growth nearly as efficaciously as similarly vaccinated WT mice. Taken together, these results demonstrate that current vaccine vectors can be improved and even tailored to meet specific demands: in the context of therapeutic vaccination, the capacity to promote an augmented effector T-cell response.


Subject(s)
Antigens, Viral, Tumor/genetics , CD8-Positive T-Lymphocytes/metabolism , Genetic Vectors/administration & dosage , Interleukin-2/genetics , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Animals , Antigens, Viral, Tumor/immunology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Female , Interleukin-2/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Skin Neoplasms/immunology , Spleen/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology
12.
J Virol ; 87(11): 6283-95, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536658

ABSTRACT

It has been reported that adenovirus (Ad)-primed CD8 T cells may display a distinct and partially exhausted phenotype. Given the practical implications of this claim, we decided to analyze in detail the quality of Ad-primed CD8 T cells by directly comparing these cells to CD8 T cells induced through infection with lymphocytic choriomeningitis virus (LCMV). We found that localized immunization with intermediate doses of Ad vector induces a moderate number of functional CD8 T cells which qualitatively match those found in LCMV-infected mice. The numbers of these cells may be efficiently increased by additional adenoviral boosting, and, importantly, the generated secondary memory cells cannot be qualitatively differentiated from those induced by primary infection with replicating virus. Quantitatively, DNA priming prior to Ad vaccination led to even higher numbers of memory cells. In this case, the vaccination led to the generation of a population of memory cells characterized by relatively low CD27 expression and high CD127 and killer cell lectin-like receptor subfamily G member 1 (KLRG1) expression. These memory CD8 T cells were capable of proliferating in response to viral challenge and protecting against infection with live virus. Furthermore, viral challenge was followed by sustained expansion of the memory CD8 T-cell population, and the generated memory cells did not appear to have been driven toward exhaustive differentiation. Based on these findings, we suggest that adenovirus-based prime-boost regimens (including Ad serotype 5 [Ad5] and Ad5-like vectors) represent an effective means to induce a substantially expanded, long-lived population of high-quality transgene-specific memory CD8 T cells.


Subject(s)
Adenoviruses, Human/immunology , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/immunology , Glycoproteins/immunology , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Viral Proteins/immunology , Adenoviruses, Human/genetics , Animals , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , Female , Genetic Vectors/genetics , Glycoproteins/administration & dosage , Glycoproteins/genetics , Humans , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred C57BL , Vaccination , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
13.
J Immunol ; 186(7): 3997-4007, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21357263

ABSTRACT

The impact of prophylactic vaccination against acute and chronic infection in a Th-deficient host has not been adequately addressed because of difficulties in generating protective immunity in the absence of CD4(+) T cell help. In this study, we demonstrated that a broad CD8(+) T cell immune response could be elicited in MHC class II-deficient mice by vaccination with adenovirus encoding lymphocytic choriomeningitis virus (LCMV) glycoprotein tethered to MHC class II-associated invariant chain. Moreover, the response induced conferred significant cytolytic CD8(+) T cell-mediated protection against challenge with a high dose of the invasive clone 13 strain of LCMV. In contrast, vaccination with adenovirus encoding unlinked LCMV glycoprotein induced weak virus control in the absence of CD4(+) T cells, and mice may die of increased immunopathology associated with incomplete protection. Acute mortality was not observed in any vaccinated mice following infection with the less-invasive Traub strain. However, LCMV Traub infection caused accelerated late mortality in unvaccinated MHC class II-deficient mice; in this case, we observed a strong trend toward delayed mortality in vaccinated mice, irrespective of the nature of the vaccine. These results indicated that optimized vaccination may lead to efficient protection against acute viral infection, even in Th-deficient individuals, but that the duration of such immunity is limited. Nevertheless, for select immunodeficiencies in which CD4(+) T cell deficiency is incomplete or transient, these results are very encouraging.


Subject(s)
Histocompatibility Antigens Class II/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Helper-Inducer/immunology , Viral Vaccines/immunology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Chronic Disease , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Epitopes, T-Lymphocyte/genetics , Lymphocytic Choriomeningitis/immunology , Lymphopenia/immunology , Lymphopenia/pathology , Lymphopenia/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Helper-Inducer/virology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
14.
J Immunol ; 186(4): 2355-64, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21257961

ABSTRACT

Potent and broad cellular immune responses against the nonstructural (NS) proteins of hepatitis C virus (HCV) are associated with spontaneous viral clearance. In this study, we have improved the immunogenicity of an adenovirus (Ad)-based HCV vaccine by fusing NS3 from HCV (Strain J4; Genotype 1b) to the MHC class II chaperone protein invariant chain (Ii). We found that, after a single vaccination of C57BL/6 or BALB/c mice with Ad-IiNS3, the HCV NS3-specific CD8(+) T cell responses were significantly enhanced, accelerated, and prolonged compared with the vaccine encoding NS3 alone. The AdIiNS3 vaccination induced polyfunctional CD8(+) T cells characterized by coproduction of IFN-γ, TNF-α and IL-2, and this cell phenotype is associated with good viral control. The memory CD8(+) T cells also expressed high levels of CD27 and CD127, which are markers of long-term survival and maintenance of T cell memory. Functionally, the AdIiNS3-vaccinated mice had a significantly increased cytotoxic capacity compared with the AdNS3 group. The AdIiNS3-induced CD8(+) T cells protected mice from infection with recombinant vaccinia virus expressing HCV NS3 of heterologous 1b strains, and studies in knockout mice demonstrated that this protection was mediated primarily through IFN-γ production. On the basis of these promising results, we suggest that this vaccination technology should be evaluated further in the chimpanzee HCV challenge model.


Subject(s)
Adenoviridae/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/immunology , Hepacivirus/immunology , Histocompatibility Antigens Class II/immunology , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Adenoviridae/genetics , Animals , Antigens, Differentiation, B-Lymphocyte/administration & dosage , Antigens, Differentiation, B-Lymphocyte/genetics , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Survival/genetics , Cell Survival/immunology , Cytotoxicity Tests, Immunologic/methods , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors/administration & dosage , Hepacivirus/genetics , Hepatitis C/immunology , Hepatitis C/pathology , Hepatitis C/prevention & control , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Immunologic Memory/genetics , Immunophenotyping , Interleukin-7 Receptor alpha Subunit/biosynthesis , Interleukin-7 Receptor alpha Subunit/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Hepatitis Vaccines/administration & dosage , Viral Hepatitis Vaccines/genetics , Viral Nonstructural Proteins/administration & dosage , Viral Nonstructural Proteins/genetics
15.
Cancer Immunol Res ; 11(2): 261-275, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36534088

ABSTRACT

Human papillomavirus (HPV) infections are the main cause of cervical and oropharyngeal cancers. As prophylactic vaccines have no curative effect, an efficient therapy would be highly desired. Most therapeutic vaccine candidates target only a small subset of HPV regulatory proteins, namely, E6 and E7, and are therefore restricted in the breadth of their immune response. However, research has suggested E1 and E2 as promising targets to fight HPV+ cancer. Here, we report the design of adenoviral vectors efficiently expressing HPV16 E1 and E2 in addition to transformation-deficient E6 and E7. Vaccination elicited vigorous CD4+ and CD8+ T-cell responses against all encoded HPV16 proteins in outbred mice and against E1 and E7 in C57BL/6 mice. Therapeutic vaccination of C3 tumor-bearing mice led to significantly reduced tumor growth and enhanced survival for both small and established tumors. Tumor biopsies revealed increased numbers of tumor-infiltrating CD8+ T cells in treated mice. Cisplatin enhanced the effect of therapeutic vaccination, accompanied by enhanced infiltration of dendritic cells into the tumor. CD8+ T cells were identified as effector cells in T-cell depletion assays, seemingly under regulation by FoxP3+CD4+ regulatory T cells. Finally, therapeutic vaccination with Ad-Ii-E1E2E6E7 exhibited significantly enhanced survival compared with vaccination with two peptides each harboring a known E6/E7 epitope. We hypothesize that this difference could be due to the induction of additional T-cell responses against E1. These results support the use of this novel vaccine candidate targeting an extended set of antigens (Ad-Ii-E1E2E6E7), in combination with cisplatin, as an advanced strategy to combat HPV+ cancers.


Subject(s)
Cancer Vaccines , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Animals , Mice , Humans , Female , Cisplatin/pharmacology , Papillomavirus E7 Proteins/genetics , Mice, Inbred C57BL , CD8-Positive T-Lymphocytes , Adenoviridae/genetics
16.
Viruses ; 15(8)2023 08 03.
Article in English | MEDLINE | ID: mdl-37632028

ABSTRACT

Human endogenous retrovirus type W (HERV-W) is expressed in various cancers. We previously developed an adenovirus-vectored cancer vaccine targeting HERV-W by encoding an assembled HERV-W group-specific antigen sequence and the HERV-W envelope sequence Syncytin-1. Syncytin-1 is constitutively fusogenic and forms large multinucleated cell fusions when overexpressed. Consequently, immunising humans with a vaccine encoding Syncytin-1 can lead to the formation of extensive syncytia, which is undesirable and poses a potential safety issue. Here, we show experiments in cell lines that restoring an evolutionary lost cleavage site of the fusion inhibitory R-peptide of Syncytin-1 inhibit cell fusion. Interestingly, this modification of the HERV-W vaccine's fusogenicity increased the expression of the vaccine antigens in vitro. It also enhanced Syncytin-1-specific antibody responses and CD8+-mediated T-cell responses compared to the wildtype vaccine in vaccinated mice, with a notable enhancement in responses to subdominant T-cell epitopes but equal responses to dominant epitopes and similar rates of survival following a tumour challenge. The impairment of cell-cell fusion and the enhanced immunogenicity profile of this HERV-W vaccine strengthens the prospects of obtaining a meaningful immune response against HERV-W in patients with HERV-W-overexpressing cancers.


Subject(s)
Cancer Vaccines , Endogenous Retroviruses , Pregnancy Proteins , Humans , Animals , Mice , Amino Acids , Endogenous Retroviruses/genetics , Epitopes, T-Lymphocyte
17.
Viruses ; 15(4)2023 04 06.
Article in English | MEDLINE | ID: mdl-37112906

ABSTRACT

Endogenous retroviruses (ERVs) account for 8% of our genome, and, although they are usually silent in healthy tissues, they become reactivated and expressed in pathological conditions such as cancer. Several studies support a functional role of ERVs in tumour development and progression, specifically through their envelope (Env) protein, which contains a region described as an immunosuppressive domain (ISD). We have previously shown that targeting of the murine ERV (MelARV) Env using virus-like vaccine (VLV) technology, consisting of an adenoviral vector encoding virus-like particles (VLPs), induces protection against small tumours in mice. Here, we investigate the potency and efficacy of a novel MelARV VLV with a mutated ISD (ISDmut) that can modify the properties of the adenoviral vaccine-encoded Env protein. We show that the modification of the vaccine's ISD significantly enhanced T-cell immunogenicity in both prime and prime-boost vaccination regimens. The modified VLV in combination with an α-PD1 checkpoint inhibitor (CPI) exhibited excellent curative efficacy against large established colorectal CT26 tumours in mice. Furthermore, only ISDmut-vaccinated mice that survived CT26 challenge were additionally protected against rechallenge with a triple-negative breast cancer cell line (4T1), showing that our modified VLV provides cross-protection against different tumour types expressing ERV-derived antigens. We envision that translating these findings and technology into human ERVs (HERVs) could provide new treatment opportunities for cancer patients with unmet medical needs.


Subject(s)
Endogenous Retroviruses , Neoplasms , Viral Vaccines , Animals , Humans , Mice , Endogenous Retroviruses/genetics , Genetic Vectors/genetics , Neoplasms/prevention & control , Neoplasms/genetics , T-Lymphocytes , Viral Vaccines/genetics , Programmed Cell Death 1 Receptor/immunology
18.
Cancers (Basel) ; 15(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38136407

ABSTRACT

Persistent human papillomavirus (HPV) infection is responsible for practically all cervical and a high proportion of anogenital and oropharyngeal cancers. Therapeutic HPV vaccines in clinical development show great promise in improving outcomes for patients who mount an anti-HPV T-cell response; however, far from all patients elicit a sufficient immunological response. This demonstrates a translational gap between animal models and human patients. Here, we investigated the potential of a new assay consisting of co-culturing vaccine-transduced dendritic cells (DCs) with syngeneic, healthy, human peripheral blood mononuclear cells (PBMCs) to mimic a human in vivo immunization. This new promising human ex vivo PBMC assay was evaluated using an innovative therapeutic adenovirus (Adv)-based HPV vaccine encoding the E1, E2, E6, and E7 HPV16 genes. This new method allowed us to show that vaccine-transduced DCs yielded functional effector T cells and unveiled information on immunohierarchy, showing E1-specific T-cell immunodominance over time. We suggest that this assay can be a valuable translational tool to complement the known animal models, not only for HPV therapeutic vaccines, and supports the use of E1 as an immunotherapeutic target. Nevertheless, the findings reported here need to be validated in a larger number of donors and preferably in patient samples.

19.
J Biol Chem ; 286(52): 44632-45, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22027838

ABSTRACT

The glucose-dependent insulinotropic polypeptide receptor (GIPr) has been implicated in high fat diet-induced obesity and is proposed as an anti-obesity target despite an uncertainty regarding the mechanism of action. To independently investigate the contribution of the insulinotropic effects and the direct effects on adipose tissue, we generated transgenic mice with targeted expression of the human GIPr to white adipose tissue or beta-cells, respectively. These mice were then cross-bred with the GIPr knock-out strain. The central findings of the study are that mice with GIPr expression targeted to adipose tissue have a similar high fat diet -induced body weight gain as control mice, significantly greater than the weight gain in mice with a general ablation of the receptor. Surprisingly, this difference was due to an increase in total lean body mass rather than a gain in total fat mass that was similar between the groups. In contrast, glucose-dependent insulinotropic polypeptide-mediated insulin secretion does not seem to be important for regulation of body weight after high fat feeding. The study supports a role of the adipocyte GIPr in nutrient-dependent regulation of body weight and lean mass, but it does not support a direct and independent role for the adipocyte or beta-cell GIPr in promoting adipogenesis.


Subject(s)
Dietary Fats/adverse effects , Gastric Inhibitory Polypeptide/metabolism , Obesity/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Peptide/metabolism , Transgenes , Weight Gain , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Dietary Fats/pharmacology , Gastric Inhibitory Polypeptide/genetics , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Mice, Knockout , Obesity/genetics , Receptors, Gastrointestinal Hormone/genetics , Receptors, Peptide/genetics
20.
J Immunol ; 184(8): 4431-9, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20212099

ABSTRACT

Adenoviral vectors have been widely used for experimental gene therapy and vaccination, yet there is a surprising lack of knowledge connecting the route and dose of adenovirus administration to the induced transgene-specific immune response. We have recently demonstrated polyfunctional CD8(+) T cells and protective memory responses using adenoviral vectors, which seem to contrast with recent reports suggesting that an exhausted CD8(+) T cell phenotype is induced by inoculation with adenoviral vectors. Accordingly, we investigated the route and dose interrelationship for transgene-specific CD8(+) T cells using adenoviral vectors encoding beta-galactosidase applied either s.c. or i.v. Irrespective of the route of inoculation, most of the adenoviral inoculum was found to disseminate systemically as the dose was raised beyond 10(9) particles. The number of transgene-specific CD8(+) T cells correlated positively with dissemination, whereas the functional capacity of the generated T cells correlated inversely with vector dissemination. A comparison of the immune response to s.c. or i.v. administration at moderate doses revealed that inoculation by both routes induced a transient peak of IFN-gamma-producing CD8(+) T cells 2 to 3 wk postinfection, but following i.v. administration, these cells were only detected in the liver. Two to four months after systemic, but not peripheral, immunization, dysfunctional transgene-specific CD8(+) T cells impaired in both cytokine production and important in vivo effector functions, accumulated in the spleen. These findings indicate that the localization of the adenoviral inoculum and not the total Ag load determines the quality of the CD8(+) T cell response induced with adenoviral vaccines.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity Tests, Immunologic/methods , Genetic Vectors/immunology , Transgenes/immunology , Viral Load/immunology , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/prevention & control , Animals , CD8-Positive T-Lymphocytes/virology , Cytotoxicity Tests, Immunologic/trends , Dose-Response Relationship, Immunologic , Foot , Genetic Vectors/administration & dosage , Genetic Vectors/standards , Hindlimb , Immunologic Memory/genetics , Injections, Intravenous , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Vaccinia virus/enzymology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Load/standards , beta-Galactosidase/administration & dosage , beta-Galactosidase/genetics , beta-Galactosidase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL