Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 43(2): 575-88, 2015.
Article in English | MEDLINE | ID: mdl-25096615

ABSTRACT

Amyloid-ß (Aß) immunotherapy for Alzheimer's disease (AD) has good preclinical support from transgenic mouse models and clinical data suggesting that a long-term treatment effect is possible. Soluble Aß protofibrils have been shown to exhibit neurotoxicity in vitro and in vivo, and constitute an attractive target for immunotherapy. Here, we demonstrate that the humanized antibody BAN2401 and its murine version mAb158 exhibit a strong binding preference for Aß protofibrils over Aß monomers. Further, we confirm the presence of the target by showing that both antibodies efficiently immunoprecipitate soluble Aß aggregates in human AD brain extracts. mAb158 reached the brain and reduced the brain protofibril levels by 42% in an exposure-dependent manner both after long-term and short-term treatment in tg-ArcSwe mice. Notably, a 53% reduction of protofibrils/oligomers in cerebrospinal fluid (CSF) that correlated with reduced brain protofibril levels was observed after long-term treatment, suggesting that CSF protofibrils/oligomers could be used as a potential biomarker. No change in native monomeric Aß42 could be observed in brain TBS extracts after mAb158-treatment in tg-ArcSwe mice. By confirming the specific ability of mAb158 to selectively bind and reduce soluble Aß protofibrils, with minimal binding to Aß monomers, we provide further support in favor of its position as an attractive new candidate for AD immunotherapy. BAN2401 has undergone full phase 1 development, and available data indicate a favorable safety profile in AD patients.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/therapeutic use , Brain/metabolism , Immunologic Factors/therapeutic use , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Antibodies, Monoclonal/pharmacology , Brain/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , Immunologic Factors/pharmacology , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid , Presenilin-1/genetics , Protein Binding/drug effects , Protein Binding/genetics
2.
J Alzheimers Dis ; 37(1): 29-40, 2013.
Article in English | MEDLINE | ID: mdl-23780660

ABSTRACT

Evidence suggests that amyloid-ß (Aß) protofibrils/oligomers are pathogenic agents in Alzheimer's disease (AD). Unfortunately, techniques enabling quantitative estimates of these species in patients or patient samples are still rather limited. Here we describe the in vitro and ex vivo characteristics of a new antibody-based radioactive ligand, [125I]mAb158, which binds to Aß protofibrils with high affinity. [125I]mAb158 was specifically taken up in brain of transgenic mice expressing amyloid-ß protein precursor (AßPP) as shown ex vivo. This was in contrast to [125I]mAb-Ly128 which does not bind to Aß. The uptake of intraperitoneally-administered [125I]mAb158 into the brain was age- and time-dependent, and saturable in AßPP transgenic mice with modest Aß deposition. Brain uptake was also found in young AßPP transgenic mice that were devoid of Aß deposits, suggesting that [125I]mAb158 targets soluble Aß protofibrils. The radioligand was diffusely located in the parenchyma, sometimes around senile plaques and only occasionally colocalized with cerebral amyloid angiopathy. A refined iodine-124-labeled version of mAb158 with much improved blood-brain barrier passage and a shorter plasma half-life might be useful for PET imaging of Aß protofibrils.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid/metabolism , Antibodies, Monoclonal/metabolism , Brain/metabolism , Iodine Radioisotopes/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid/immunology , Amyloid beta-Protein Precursor/immunology , Animals , Brain/immunology , Brain/pathology , Humans , Mice , Mice, Transgenic , Protein Binding/immunology
3.
PLoS One ; 8(6): e65964, 2013.
Article in English | MEDLINE | ID: mdl-23785460

ABSTRACT

Ligands of pattern recognition receptors (PRRs) including Toll-like receptors (TLRs) stimulate innate and adaptive immune responses and are considered as potent adjuvants. Combinations of ligands might act in synergy to induce stronger and broader immune responses compared to stand-alone ligands. Alphaviruses stimulate endosomal TLRs 3, 7 and 8 as well as the cytoplasmic PRR MDA-5, resulting in induction of a strong type I interferon (IFN) response. Bacterial flagellin stimulates TLR5 and when delivered intracellularly the cytosolic PRR NLRC4, leading to secretion of proinflammatory cytokines. Both alphaviruses and flagellin have independently been shown to act as adjuvants for antigen-specific antibody responses. Here, we hypothesized that alphavirus and flagellin would act in synergy when combined. We therefore cloned the Salmonella Typhimurium flagellin (FliC) gene into an alphavirus replicon and assessed its adjuvant activity on the antibody response against co-administered antigen. In mice immunized with recombinant alphavirus, antibody responses were greatly enhanced compared to soluble FliC or control alphavirus. Both IgG1 and IgG2a/c responses were increased, indicating an enhancement of both Th1 and Th2 type responses. The adjuvant activity of FliC-expressing alphavirus was diminished but not abolished in the absence of TLR5 or type I IFN signaling, suggesting the contribution of several signaling pathways and some synergistic and redundant activity of its components. Thus, we have created a recombinant adjuvant that stimulates multiple signaling pathways of innate immunity resulting in a strong and broad antibody response.


Subject(s)
Alphavirus/genetics , Alphavirus/immunology , Flagellin/genetics , Flagellin/immunology , Replicon , Adjuvants, Immunologic , Alphavirus/metabolism , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cell Line , Cricetinae , Gene Expression , Immunoglobulin G/immunology , Interferon Type I/metabolism , Mice , Mice, Knockout , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Signal Transduction , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism
4.
PLoS One ; 6(8): e23828, 2011.
Article in English | MEDLINE | ID: mdl-21858228

ABSTRACT

Virus-like particles (VLPs) consist of capsid proteins from viruses and have been shown to be usable as carriers of protein and peptide antigens for immune therapy. In this study, we have produced and assayed murine polyomavirus (MPyV) VLPs carrying the entire human Prostate Specific Antigen (PSA) (PSA-MPyVLPs) for their potential use for immune therapy in a mouse model system. BALB/c mice immunized with PSA-MPyVLPs were only marginally protected against outgrowth of a PSA-expressing tumor. To improve protection, PSA-MPyVLPs were co-injected with adjuvant CpG, either alone or loaded onto murine dendritic cells (DCs). Immunization with PSA-MPyVLPs loaded onto DCs in the presence of CpG was shown to efficiently protect mice from tumor outgrowth. In addition, cellular and humoral immune responses after immunization were examined. PSA-specific CD4(+) and CD8(+) cells were demonstrated, but no PSA-specific IgG antibodies. Vaccination with DCs loaded with PSA-MPyVLPs induced an eight-fold lower titre of anti-VLP antibodies than vaccination with PSA-MPyVLPs alone. In conclusion, immunization of BALB/c mice with PSA-MPyVLPs, loaded onto DCs and co-injected with CpG, induces an efficient PSA-specific tumor protective immune response, including both CD4(+) and CD8(+) cells with a low induction of anti-VLP antibodies.


Subject(s)
Cancer Vaccines/immunology , Neoplasms, Experimental/immunology , Polyomavirus/immunology , Prostate-Specific Antigen/immunology , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/transplantation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunotherapy/methods , Interferon-gamma/immunology , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Prostate-Specific Antigen/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Treatment Outcome , Tumor Burden/immunology , Vaccines, Virus-Like Particle/administration & dosage
5.
PLoS One ; 5(7): e11580, 2010 Jul 19.
Article in English | MEDLINE | ID: mdl-20657846

ABSTRACT

BACKGROUND: Immunization with murine pneumotropic virus virus-like particles carrying Her2/neu (Her2MPtVLPs) prevents tumour outgrowth in mice when given prophylactically, and therapeutically if combined with the adjuvant CpG. We investigated which components of the immune system are involved in tumour rejection, and whether long-term immunological memory can be obtained. METHODOLOGY AND RESULTS: During the effector phase in BALB/c mice, only depletion of CD4+ and CD8+ in combination, with or without NK cells, completely abrogated tumour protection. Depletion of single CD4+, CD8+ or NK cell populations only had minor effects. During the immunization/induction phase, combined depletion of CD4+ and CD8+ cells abolished protection, while depletion of each individual subset had no or negligible effect. When tumour rejection was studied in knock-out mice with a C57Bl/6 background, protection was lost in CD4-/-CD8-/- and CD4-/-, but not in CD8-/- mice. In contrast, when normal C57Bl/6 mice were depleted of different cell types, protection was lost irrespective of whether only CD4+, only CD8+, or CD4+ and CD8+ cells in combination were eradicated. No anti-Her2/neu antibodies were detected but a Her2/neu-specific IFNgamma response was seen. Studies of long-term memory showed that BALB/c mice could be protected against tumour development when immunized together with CpG as long as ten weeks before challenge. CONCLUSION: Her2MPtVLP immunization is efficient in stimulating several compartments of the immune system, and induces an efficient immune response including long-term memory. In addition, when depleting mice of isolated cellular compartments, tumour protection is not as efficiently abolished as when depleting several immune compartments together.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Receptor, ErbB-2/immunology , Virion/metabolism , Animals , Cancer Vaccines/genetics , Cell Line , Cell Line, Tumor , Humans , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptor, ErbB-2/genetics , Virion/genetics
6.
Int J Cancer ; 124(1): 150-6, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18839427

ABSTRACT

Virus-like particles (VLPs) have increasingly attracted attention as DNA-free and safe antigen carriers in tumor immunotherapy, requiring only minute amounts of antigens. Previously, we have immunized with murine polyomavirus (MPyV) VLPs carrying human Her2/neu and prevented the outgrowth of a human Her2/neu expressing tumor in a transplantable tumor model as well as outgrowth of spontaneous rat Her2/neu carcinomas in BALB-neuT mice. Here, we examine if prophylactic and therapeutic protection could be obtained with murine pneumotropic virus (MPtV) VLPs, and study the cross-reactivity between human and rat Her2/neu. VLPs from MPyV and MPtV carrying human or rat Her2/neu were tested in two transplantable tumor models against a human Her2/neu positive (D2F2/E2) and a rat Her2/neu positive tumor cell line (TUBO). Rat Her2/neu-VLPs were also tested in BALB-neuT mice. Her2/neu-MPtVLPs were as efficient as prophylactic vaccines against D2F2/E2 and TUBO as those from MPyV. Homologous Her2/neu was better than heterologous, i.e. human Her2/neu-VLPs were better than rat Her2/neu-VLPs against D2F2/E2 and vice versa. Moreover, therapeutic immunization with human Her2/neu-VLPs together with CpG given up to 6 days after challenge protected against D2F2/E2. In BALB-neuT mice, rat Her2/neu-VLPs were less efficient than human Her2/neu-VLPs used in our previous study, implying that protection seen in that study was partly due to the use of human rather than rat Her2/neu. In conclusion, Her2/neu-MPtVLPs are effective both as prophylactic and therapeutic tumor vaccines. Homologous Her2/neu-VLPs are superior to heterologous in transplantable tumor models, while the opposite is true in BALB-neuT mice.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy/methods , Neoplasms/immunology , Polyomavirus/metabolism , Receptor, ErbB-2/immunology , Animals , DNA/metabolism , Female , Humans , Kinetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Transplantation , Neoplasms/metabolism , Neoplasms/therapy , Polyomavirus/genetics , Rats
7.
Cancer Immunol Immunother ; 56(9): 1335-44, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17268793

ABSTRACT

One immunization with murine polyomavirus (MPyV) VP1 virus-like particles containing a fusion protein between MPyV VP2 and the extra cellular and transmembrane domain of Her2 (Her2(1-683)PyVLPs) efficiently protects BALB/c mice from outgrowth of the Her2 expressing tumor D2F2/E2. To possibly enhance the anti-Her2 immune response and abrogate the induced anti-VLP antibody response, immunization with murine dendritic cells (DCs) loaded with Her2(1-683)PyVLPs was performed. Mice were immunized once or more with 5 or 50 microg Her2(1-683)PyVLPs alone or loaded on DCs, and challenged 14 days after the last immunization with a lethal dose of Her2-positive D2F2/E2 cells. Mice were protected from tumor outgrowth, when immunized only once with 5 or 50 mug Her2(1-683)PyVLPs loaded on DCs, or 50 mug of Her2(1-683)PyVLPs alone, whereas immunization once or more with 5 mug of Her2(1-683)PyVLPs alone only protected half of the mice. Immunization with recombinant Her2 protein alone, or loaded on DCs, did not induce tumor immunity. Using both immunization strategies, Her2-specific T cell immunity was demonstrated, while Her2-specific antibodies were not detected. Loading VLPs on DCs reduced anti-VLP antibodies sixfold, but did not influence the efficiency of subsequent immunizations. Notably, DC maturation by Her2(1-683)PyVLPs in vitro was not demonstrated although the IL-12 production was significantly increased. In conclusion, loading of VLPs on DCs can enhance specific VLP immunization considerably.


Subject(s)
Cancer Vaccines/therapeutic use , Capsid Proteins/therapeutic use , Dendritic Cells/immunology , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/therapeutic use , Animals , Cancer Vaccines/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Line , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , Humans , Interleukin-12/metabolism , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Peptide Fragments/immunology , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology
9.
Anticancer Res ; 25(4): 2601-8, 2005.
Article in English | MEDLINE | ID: mdl-16080500

ABSTRACT

This review describes the use of murine polyomavirus "virus-like" particles (MPyV-VLPs), free from viral genes, as vectors for gene and immune therapy and as vaccines. For large-scale MPyV-VLP manufacture, VP1 is produced in a baculovirus insect cell system, E. coli or in yeast. MPyV-VLPs bind eukaryotic DNA and introduce this DNA into various cell types in vitro and in vivo. In normal and T-cell-deficient mice, this results in the production of anti-MPyV-VLP (and MPyV) antibodies. Furthermore, repeated MPyV-VLP vaccination has been shown to prevent primary MPyV infection in normal and T-cell-deficient mice, and the outgrowth of some MPyV-induced tumours in normal mice. Moreover, when inoculated with gene constructs encoding for HIV p24, MPyV-VLPs augment the antibody response to p24. In addition, MPyV-VLPs, containing fusion proteins between the VP2 or VP3 capsid protein and selected antigens, can be used as vaccines. Notably, one vaccination with MPyV-VLPs, containing a fusion protein between VP2 and the extracellular and transmembrane parts of the HER-2/neu oncogene, immunizes against outgrowth of a HER-2/neu-expressing tumour in Balb/c mice and also against the development of mammary carcinomas in BALB-neuT transgenic mice. Finally, a second polyoma VLP-vector based on murine pneumotropic virus (MPtV-VLP), which does not cross-react serologically with MPyV-VLP (and MPyV), has been developed and can be used to conduct prime boost gene and immune therapy and vaccination. In summary, MPyV-VLPs are useful vectors for gene therapy, immune therapy and as vaccines and, in combination with MPyV-VLPs, MPtV-VLPs are potentially useful as prime-boost vectors.


Subject(s)
Cancer Vaccines/genetics , Genetic Therapy/methods , Immunotherapy/methods , Polyomavirus/genetics , Viral Vaccines/genetics , Animals , Genetic Vectors/genetics , Humans , Mice
10.
Cancer Res ; 65(13): 5953-7, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15994974

ABSTRACT

Murine polyomavirus (MPyV) VP1 virus-like particles (VLPs), containing a fusion protein between MPyV VP2 and the extracellular and transmembrane domain of HER-2/neu (Her2), Her2(1-683)PyVLPs, were tested for their ability to vaccinate against Her2-expressing tumors in two different in vivo models. Protection was assessed both against a lethal challenge with a BALB/c mammary carcinoma transfected with human Her2 (D2F2/E2) and against the outgrowth of autochthonous mammary carcinomas in BALB-neuT mice, transgenic for the activated rat Her2 oncogene. A single injection of Her2(1-683)PyVLPs before tumor inoculation induced a complete rejection of D2F2/E2 tumor cells in BALB/c mice. Similarly, a single injection of Her2(1-683)PyVLPs at 6 weeks of age protected BALB-neuT mice with atypical hyperplasia from a later outgrowth of mammary carcinomas, whereas all controls developed palpable tumors in all mammary glands. VLPs containing only VP1 and VP2 did not induce protection. The protection elicited by Her2(1-683)PyVLPs vaccination was most likely due to a cellular immune response because a Her2-specific response was shown in an ELISPOT assay, whereas antibodies against Her2 were not detected in any of the two models. The results show the feasibility of using MPyV-VLPs carrying Her2 fusion proteins as safe and efficient vaccines against Her2-expressing tumors.


Subject(s)
Cancer Vaccines/immunology , Capsid Proteins/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Receptor, ErbB-2/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Cancer Vaccines/genetics , Cancer Vaccines/pharmacology , Capsid Proteins/genetics , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Polyomavirus/genetics , Polyomavirus/immunology , Rats , Receptor, ErbB-2/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Vaccination/methods
11.
In Vivo ; 19(2): 323-6, 2005.
Article in English | MEDLINE | ID: mdl-15796193

ABSTRACT

The ability of murine polyomavirus (MPyV)-VP1 virus-like particles (MPyV-VLPs) to immunize against MPyV tumour outgrowth was investigated. Non-immunized and mice immunized three times were challenged with MPyV or non-MPyV tumours and followed for tumour outgrowth. MPyV-VLP immunization abrogated outgrowth of some, but not all, tested MPyV tumours and delayed the outgrowth of a non-MPyV tumour to some extent. However, when mice were irradiated prior to tumour challenge to avoid an unspecific immune response, protection was MPyV-specific. In conclusion, VLP immunization for prevention of viral infection could also contribute to immune-protection against some tumours induced by the corresponding virus.


Subject(s)
Capsid Proteins/immunology , Neoplasms, Experimental/prevention & control , Polyomavirus Infections/prevention & control , Polyomavirus/immunology , Tumor Virus Infections/prevention & control , Animals , Antigens, Neoplasm/immunology , Capsid Proteins/genetics , Cell Line, Tumor , Glutathione Transferase/genetics , Immunization , Mice , Mice, Inbred CBA , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Polyomavirus Infections/immunology , Polyomavirus Infections/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/pathology , Virus Replication
12.
J Med Virol ; 70(2): 293-300, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12696121

ABSTRACT

The ability to vaccinate against polyomavirus infection in a T-cell deficient as well as a normal immune context was studied using polyomavirus major capsid protein (VP1) pseudocapsids (VP1-ps) or a glutathione-S-transferase-VP1 (GST-VP1) fusion protein. VP1-ps (1 or 10 microg) were administered subcutaneously, alone or together with Freund's complete and incomplete adjuvant, to CD4(-/-)8(-/-) T-cell deficient or normal C57Bl/6 mice on four occasions. Alternatively, CD4(-/-)8(-/-) and normal mice were inoculated with either GST-VP1 or Py-VP1-ps (5 microg). Following immunisation, antibody titres were tested by ELISA to VP1-ps or GST-VP1 or by haemagglutination inhibition (HAI). Mice were then infected with polyomavirus. Three weeks post-infection, the mice were killed and examined for the presence of polyomavirus DNA by PCR. Viral DNA was not detected in CD4(-/-)8(-/-) mice immunised with either VP1-ps alone or in combination with Freund's complete and incomplete adjuvant, or in any of the normal mice immunised with VP1-ps or GST-VP1. However, viral DNA was detected in 2/5 of the CD4(-/-)8(-/-) mice immunised with GST-VP1 and in non-immunised controls. Greater antibody titres were observed to VP1-ps than to GST-VP1 in CD4(-/-)8(-/-) mice after VP1-ps compared to GST-VP1 immunisation and antibody responses were better in normal than in immune-deficient mice. Only immunisation with VP1-ps resulted in haemagglutination inhibition. Complete protection against polyomavirus infection in the T-cell deficient context was obtained with VP1-ps, but not with GST-VP1, immunisation using the present vaccination protocol.


Subject(s)
Capsid Proteins/immunology , Immunologic Deficiency Syndromes/immunology , Polyomavirus Infections/prevention & control , T-Lymphocytes/immunology , Tumor Virus Infections/prevention & control , Viral Vaccines/administration & dosage , Animals , Antibodies, Viral/blood , Capsid , Capsid Proteins/genetics , Capsid Proteins/metabolism , Disease Models, Animal , Glutathione Transferase/genetics , Glutathione Transferase/immunology , Glutathione Transferase/metabolism , Guinea Pigs , Hemagglutination , Humans , Immunization , Mice , Mice, Inbred C57BL , Polyomavirus/immunology , Polyomavirus Infections/virology , Recombinant Fusion Proteins/immunology , Tumor Virus Infections/virology , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...