Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 24(6)2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36983050

ABSTRACT

Cancer testis antigens are ideal for tumor immunotherapy due to their testis-restricted expression. We previously showed that an immunotherapeutic vaccine targeting the germ cell-specific transcription factor BORIS (CTCFL) was highly effective in treating aggressive breast cancer in the 4T1 mouse model. Here, we further tested the therapeutic efficacy of BORIS in a rat 13762 breast cancer model. We generated a recombinant VEE-VRP (Venezuelan Equine Encephalitis-derived replicon particle) vector-expressing modified rat BORIS lacking a DNA-binding domain (VRP-mBORIS). Rats were inoculated with the 13762 cells, immunized with VRP-mBORIS 48 h later, and then, subsequently, boosted at 10-day intervals. The Kaplan-Meier method was used for survival analysis. Cured rats were re-challenged with the same 13762 cells. We demonstrated that BORIS was expressed in a small population of the 13762 cells, called cancer stem cells. Treatment of rats with VRP-BORIS suppressed tumor growth leading to its complete disappearance in up to 50% of the rats and significantly improved their survival. This improvement was associated with the induction of BORIS-specific cellular immune responses measured by T-helper cell proliferation and INFγ secretion. The re-challenging of cured rats with the same 13762 cells indicated that the immune response prevented tumor growth. Thus, a therapeutic vaccine against rat BORIS showed high efficacy in treating the rat 13762 carcinoma. These data suggest that targeting BORIS can lead to the elimination of mammary tumors and cure animals even though BORIS expression is detected only in cancer stem cells.


Subject(s)
Mammary Neoplasms, Animal , Vaccines , Animals , Male , Mice , Rats , DNA-Binding Proteins/metabolism , Immunotherapy/methods , Transcription Factors
2.
Int J Mol Sci ; 24(12)2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37372944

ABSTRACT

Post-translationally modified N-terminally truncated amyloid beta peptide with a cyclized form of glutamate at position 3 (pE3Aß) is a highly pathogenic molecule with increased neurotoxicity and propensity for aggregation. In the brains of Alzheimer's Disease (AD) cases, pE3Aß represents a major constituent of the amyloid plaque. The data show that pE3Aß formation is increased at early pre-symptomatic disease stages, while tau phosphorylation and aggregation mostly occur at later stages of the disease. This suggests that pE3Aß accumulation may be an early event in the disease pathogenesis and can be prophylactically targeted to prevent the onset of AD. The vaccine (AV-1986R/A) was generated by chemically conjugating the pE3Aß3-11 fragment to our universal immunogenic vaccine platform MultiTEP, then formulated in AdvaxCpG adjuvant. AV-1986R/A showed high immunogenicity and selectivity, with endpoint titers in the range of 105-106 against pE3Aß and 103-104 against the full-sized peptide in the 5XFAD AD mouse model. The vaccination showed efficient clearance of the pathology, including non-pyroglutamate-modified plaques, from the mice brains. AV-1986R/A is a novel promising candidate for the immunoprevention of AD. It is the first late preclinical candidate which selectively targets a pathology-specific form of amyloid with minimal immunoreactivity against the full-size peptide. Successful translation into clinic may offer a new avenue for the prevention of AD via vaccination of cognitively unimpaired individuals at risk of disease.


Subject(s)
Alzheimer Disease , Cancer Vaccines , Mice , Animals , Alzheimer Disease/prevention & control , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Pyrrolidonecarboxylic Acid , Immunotherapy , Plaque, Amyloid/pathology , Brain/metabolism , Mice, Transgenic , Disease Models, Animal
3.
Int J Mol Sci ; 23(11)2022 May 29.
Article in English | MEDLINE | ID: mdl-35682759

ABSTRACT

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are characterized by the aberrant accumulation of intracytoplasmic misfolded and aggregated α-synuclein (α-Syn), resulting in neurodegeneration associated with inflammation. The propagation of α-Syn aggregates from cell to cell is implicated in the spreading of pathological α-Syn in the brain and disease progression. We and others demonstrated that antibodies generated after active and passive vaccinations could inhibit the propagation of pathological α-Syn in the extracellular space and prevent/inhibit disease/s in the relevant animal models. We recently tested the immunogenicity and efficacy of four DNA vaccines on the basis of the universal MultiTEP platform technology in the DLB/PD mouse model. The antibodies generated by these vaccines efficiently reduced/inhibited the accumulation of pathological α-Syn in the different brain regions and improved the motor deficit of immunized female mice. The most immunogenic and preclinically effective vaccine, PV-1950D, targeting three B-cell epitopes of pathological α-Syn simultaneously, has been selected for future IND-enabling studies. However, to ensure therapeutically potent concentrations of α-Syn antibodies in the periphery of the vaccinated elderly, we developed a recombinant protein-based MultiTEP vaccine, PV-1950R/A, and tested its immunogenicity in young and aged D-line mice. Antibody responses induced by immunizations with the PV-1950R/A vaccine and its homologous DNA counterpart, PV-1950D, in a mouse model of PD/DLB have been compared.


Subject(s)
Lewy Body Disease , Parkinson Disease , Vaccines, DNA , Animals , Antibodies , Disease Models, Animal , Epitopes, B-Lymphocyte , Female , Lewy Body Disease/metabolism , Mice , Parkinson Disease/metabolism , Recombinant Proteins , alpha-Synuclein/metabolism
4.
Neurobiol Dis ; 139: 104823, 2020 06.
Article in English | MEDLINE | ID: mdl-32119976

ABSTRACT

The DNA vaccine, AV-1959D, targeting N-terminal epitope of Aß peptide, has been proven immunogenic in mice, rabbits, and non-human primates, while its therapeutic efficacy has been shown in mouse models of Alzheimer's disease (AD). Here we report for the first time on IND-enabling biodistribution and safety/toxicology studies of cGMP-grade AV-1959D vaccine in the Tg2576 mouse model of AD. We also tested acute neuropathology safety profiles of AV-1959D in another AD disease model, Tg-SwDI mice with established vascular and parenchymal Aß pathology in a pre-clinical translational study. Biodistribution studies two days after the injection demonstrated high copy numbers of AV-1959D plasmid after single immunization of Tg2576 mice at the injection sites but not in the tissues of distant organs. Plasmids persisted at the injection sites of some mice 60 days after vaccination. In Tg2576 mice with established amyloid pathology, we did not observe short- or long-term toxicities after multiple immunizations with three doses of AV-1959D. Assessment of the repeated dose acute safety of AV-1959D in cerebral amyloid angiopathy (CAA) prone Tg-SwDI mice did not reveal any immunotherapy-induced vasogenic edema detected by magnetic resonance imaging (MRI) or increased microhemorrhages. Multiple immunizations of Tg-SwDI mice with AV-1959D did not induce T and B cell infiltration, glial activation, vascular deposition of Aß, or neuronal degeneration (necrosis and apoptosis) greater than that in the control group determined by immunohistochemistry of brain tissues. Taken together, the safety data from two different mouse models of AD substantiate a favorable safety profile of the cGMP grade AV-1959D vaccine supporting its progression to first-in-human clinical trials.


Subject(s)
Alzheimer Vaccines/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic , Alzheimer Disease/immunology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Animals , Antibody Formation , Cerebral Amyloid Angiopathy/immunology , Clinical Trials as Topic , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Peptide Fragments/metabolism
5.
J Immunol ; 200(8): 2656-2669, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29500244

ABSTRACT

Dendritic cells (DCs) are well-known for their functions in orchestrating the innate and adaptive arms of immune defense. However, under certain conditions, DCs can exert tumoricidal activity. We have elucidated the mechanism of tumor suppression by TLR4-activated bone marrow-derived DCs (BMDCs) isolated from BALB/c mice. We identified that two distinct subsets of BMDCs (CD11b+CD11c+I-A/Eint and CD11b+CD11c+I-A/Ehigh) have different cytotoxic mechanisms of action. The cytotoxicity of the former subset is mediated through NO and reactive oxygen species and type I IFN (IFN-ß), whereas the latter subset acts only through IFN-ß. TLR4 agonists, LPS or pharmaceutical-grade ImmunoMax, activate CD11c+ BMDCs, which, in turn, directly kill 4T1 mouse breast cancer cells or inhibit their proliferation in an MHC-independent manner. These data define two populations of BMDCs with different mechanisms of direct cytotoxicity, as well as suggest that the I-A/Eint subset could be less susceptible to counteracting mechanisms in the tumor microenvironment and support investigation of similar subsets in human DCs.


Subject(s)
Bone Marrow/metabolism , Dendritic Cells/metabolism , Toll-Like Receptor 4/agonists , Animals , Bone Marrow Cells/metabolism , CD11c Antigen/metabolism , Cell Line, Tumor , Cells, Cultured , Female , Interferon-beta/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Microenvironment/physiology
6.
Mol Ther ; 25(1): 153-164, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28129111

ABSTRACT

Previously, we reported that Alzheimer's disease (AD) epitope vaccines (EVs) composed of N-terminal ß-amyloid (Aß42) B cell epitope fused with universal foreign T helper (Th) epitope(s) were immunogenic, potent, and safe in different amyloid precursor protein (APP) transgenic mice with early AD-like pathology. However, developing an effective therapeutic vaccine is much more challenging, especially when a self-antigen such as Aß42 is a target. Here, we directly compare the efficacy of anti-Aß42 antibodies in Tg2576 mice with low or high levels of AD-like pathology at the start of immunizations: 6-6.5 months for preventive vaccinations and 16-19 months for therapeutic vaccinations. EV in a preventive setting induced high levels of anti-Aß antibodies, significantly reducing pathologic forms of Aß in the brains of Tg2576 mice. When used therapeutically for immunesenescent Tg2576 mice, EV induced low levels of antibodies not sufficient for clearing of AD-like pathology. Separately, we demonstrated that EV was also not effective in 11-11.5-month-old Tg2576 mice with moderate AD-like pathology. However, we augmented the titers of anti-Aß antibodies in transgenic (Tg) mice of the same age possessing the pre-existing memory Th cells and detected a significant decrease in diffuse and core plaques in cortical regions compared to control animals along with improved novel object recognition performance.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Vaccines/immunology , Alzheimer Disease/prevention & control , Alzheimer Disease/therapy , Animals , Antibodies/immunology , Astrocytes/immunology , Astrocytes/metabolism , Brain/immunology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Epitopes/immunology , Immunization , Mice , Mice, Transgenic , Neuroglia/immunology , Neuroglia/metabolism , Peptide Fragments/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Vaccines/administration & dosage
7.
Alzheimers Dement ; 11(10): 1246-59, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26192465

ABSTRACT

Traditional vaccination against infectious diseases relies on generation of cellular and humoral immune responses that act to protect the host from overt disease even though they do not induce sterilizing immunity. More recently, attempts have been made with mixed success to generate therapeutic vaccines against a wide range of noninfectious diseases including neurodegenerative disorders. After the exciting first report of successful vaccine prevention of progression of an Alzheimer's disease (AD) animal model in 1999, various epitope-based vaccines targeting amyloid beta (Aß) have proceeded to human clinical trials, with varied results. More recently, AD vaccines based on tau protein have advanced into clinical testing too. This review seeks to put perspective to the mixed results obtained so far in clinical trials of AD vaccines and discusses the many pitfalls and misconceptions encountered on the path to a successful AD vaccine, including better standardization of immunologic efficacy measures of antibodies, immunogenicity of platform/carrier and adjuvants.


Subject(s)
Alzheimer Disease/prevention & control , Alzheimer Vaccines/therapeutic use , Alzheimer Disease/therapy , Animals , Antibody Formation , Clinical Trials as Topic , Humans , Vaccination , tau Proteins/immunology
8.
J Neurosci ; 33(15): 6245-56, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23575824

ABSTRACT

Alzheimer's disease (AD) is hallmarked by amyloid plaques, neurofibrillary tangles, and widespread cortical neuronal loss (Selkoe, 2001). The "amyloid cascade hypothesis" posits that cerebral amyloid sets neurotoxic events into motion that precipitate Alzheimer dementia (Hardy and Allsop, 1991). Yet, faithful recapitulation of all AD features in widely used transgenic (Tg) mice engineered to overproduce Aß peptides has been elusive. We have developed a Tg rat model (line TgF344-AD) expressing mutant human amyloid precursor protein (APPsw) and presenilin 1 (PS1ΔE9) genes, each independent causes of early-onset familial AD. TgF344-AD rats manifest age-dependent cerebral amyloidosis that precedes tauopathy, gliosis, apoptotic loss of neurons in the cerebral cortex and hippocampus, and cognitive disturbance. These results demonstrate progressive neurodegeneration of the Alzheimer type in these animals. The TgF344-AD rat fills a critical need for a next-generation animal model to enable basic and translational AD research.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cerebral Cortex/pathology , Cognition Disorders/pathology , Hippocampus/pathology , Nerve Degeneration/pathology , Plaque, Amyloid/pathology , Tauopathies/pathology , Age Factors , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal , Cerebral Amyloid Angiopathy , Cerebral Cortex/metabolism , Cognition Disorders/complications , Cognition Disorders/genetics , Cognition Disorders/metabolism , Disease Models, Animal , Female , Gliosis/genetics , Gliosis/pathology , Hippocampus/metabolism , Humans , Male , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Plaque, Amyloid/genetics , Presenilin-1/genetics , Rats , Rats, Inbred F344 , Rats, Transgenic , Tauopathies/metabolism , tau Proteins/metabolism
9.
J Neurosci ; 33(11): 4923-34, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23486963

ABSTRACT

The Alzheimer's disease (AD) process is understood to involve the accumulation of amyloid plaques and tau tangles in the brain. However, attempts at targeting the main culprits, neurotoxic Aß peptides, have thus far proven unsuccessful for improving cognitive function. Recent clinical trials with passively administrated anti-Aß antibodies failed to slow cognitive decline in mild to moderate AD patients, but suggest that an immunotherapeutic approach could be effective in patients with mild AD. Using an AD mouse model (Tg2576), we tested the immunogenicity (cellular and humoral immune responses) and efficacy (AD-like pathology) of clinical grade Lu AF20513 vaccine. We found that Lu AF20513 induces robust "non-self" T-cell responses and the production of anti-Aß antibodies that reduce AD-like pathology in the brains of Tg2576 mice without inducing microglial activation and enhancing astrocytosis or cerebral amyloid angiopathy. A single immunization with Lu AF20513 induced strong humoral immunity in mice with preexisting memory T-helper cells. In addition, Lu AF20513 induced strong humoral responses in guinea pigs and monkeys. These data support the translation of Lu AF20513 to the clinical setting with the aims of: (1) inducing therapeutically potent anti-Aß antibody responses in patients with mild AD, particularly if they have memory T-helper cells generated after immunizations with conventional tetanus toxoid vaccine, and (2) preventing pathological autoreactive T-cell responses.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/therapy , Amyloid beta-Peptides/chemistry , Epitopes, T-Lymphocyte/immunology , Peptide Fragments/chemistry , Vaccination/methods , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Antibodies, Anti-Idiotypic/blood , Antibodies, Anti-Idiotypic/pharmacology , Antibody Formation/immunology , Brain/metabolism , Brain/pathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Female , Guinea Pigs , Humans , Immunologic Memory/drug effects , Immunologic Memory/immunology , Macaca fascicularis , Male , Mice , Mice, Transgenic , Mutation/genetics , Neuroglia/drug effects , Neuroglia/immunology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Binding/immunology , Surface Plasmon Resonance , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccines/immunology
10.
J Transl Med ; 12: 322, 2014 Nov 29.
Article in English | MEDLINE | ID: mdl-25432242

ABSTRACT

BACKGROUND: Previously we demonstrated that the resection of primary 4T1 tumors only slightly prolongs mouse survival, but importantly, creates a "window of opportunity" with attenuated suppressor cell and increased activated T cell populations. This suggests that additional activation of the immune system by immunostimulatory agents during this period may enhance anti-tumor immunity and potentially eradicate micro-metastatic disease in this stringent model. We hypothesized that the immunostimulator Immunomax®, which is comprised of a plant-derived polysaccharide, is non-toxic in humans and stimulates immune defense during the infectious diseases treatment, may have also anti-tumor activity and be beneficial in the adjuvant setting when endogenous anti-tumor responses are present and during the "window of opportunity" in post-resection metastatic breast cancer model. Here we provide the initial report that Immunomax® demonstrates the capacity to eliminate micro-metastatic disease in the post-resection, 4T1 mouse model of breast cancer. METHODS: The efficacy of Immunomax® was evaluated by analyzing survival rate and the number of spontaneous clonogenic tumor cells in the lung homogenates of mice. The frequencies of activated NK, CD4(+) and CD8(+) cells as well as myeloid-derived suppressor cells and Treg cells were evaluated using flow cytometry. Highly purified mouse and human dendritic and NK cells were sorted and the effect of Immunomax® on activation status of these cells was assessed by flow cytometry. The property of Immunomax® as TLR-4 agonist was determined by NF-κB/SEAP reporter gene assay, WB, RT-PCR. RESULTS: Immunomax® injections significantly prolonged overall survival and cured 31% of mice. This immunostimulator activates DCs via the TLR-4, which in turn stimulates tumoricidal NK cells and in vitro, completely inhibits growth of 4T1 cells. Incubation of PBMC from healthy donors with Immunomax® activates NK cells via activation of plasmacytoid DC leading significantly higher efficacy in killing of human NK-target cells K562 compared with non-treated cells. CONCLUSION: This is the first demonstration that Immunomax® is a TLR-4 agonist and the first report of a documented role for this pharmaceutical grade immunostimulator in augmenting anti-tumor activity, suggesting that incorporation of Immunomax® into developing breast cancer therapeutic strategies may be beneficial and with less potential toxicity than checkpoint inhibitors.


Subject(s)
Mammary Neoplasms, Experimental/therapy , Neoplasm Metastasis , Plant Extracts/pharmacology , Toll-Like Receptor 4/drug effects , Animals , Female , Lymphocytes/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C
11.
Alzheimers Dement ; 10(3): 271-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24560029

ABSTRACT

BACKGROUND: As a prelude to clinical trials we have characterized B- and T-cell immune responses in macaques to AD vaccine candidates: AV-1955 and its slightly modified version, AV-1959 (with 3 additional promiscuous Th epitopes). METHODS: T- and B-cell epitope mapping was performed using the ELISPOT assay and competition ELISA, respectively. RESULTS: AV-1955 and AV-1959 did not stimulate potentially harmful autoreactive T cells, but instead activated a broad but individualized repertoire of Th cells specific to the MultiTEP platform in macaques. Although both vaccines induced robust anti-Aß antibody responses without producing antibodies specific to Th epitopes of MultiTEP platforms, analyses of cellular immune responses in macaques demonstrated that the addition of Th epitopes in the case of AV-1959 created a more potent, superior vaccine. CONCLUSION: AV-1959 is a promising vaccine candidate capable of producing therapeutically potent anti-amyloid antibody in a broader population of vaccinated subjects with high MHC class II gene polymorphisms.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/therapy , Alzheimer Vaccines/immunology , Alzheimer Vaccines/therapeutic use , Lymphocyte Activation , Amyloid beta-Peptides/immunology , Animals , Antibodies/immunology , Antibody Formation , B-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Epitope Mapping , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Female , Macaca mulatta , Male , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, DNA/therapeutic use
12.
Alzheimers Dement ; 10(3): 284-95, 2014 May.
Article in English | MEDLINE | ID: mdl-23916838

ABSTRACT

BACKGROUND: Clinical trials with passive and active Alzheimer's disease (AD) vaccines suggest that early interventions are needed for improvement of cognitive and/or functional performance in patients, providing impetus for the development of safe and immunologically potent active vaccines targeting amyloid ß (Aß). The AN-1792 trial has indicated that Aß-specific T cells may be unsafe for humans; therefore, other vaccines based on small Aß epitopes are undergoing preclinical and clinical testing. METHODS: Humoral and cellular immune responses elicited in response to a novel DNA epitope-based vaccine (AV-1955) delivered to rhesus macaques using the TriGrid electroporation device were evaluated. Functional activities of anti-Aß antibodies generated in response to vaccination were assessed in vitro. RESULTS: AV-1955 generates long-term, potent anti-Aß antibodies and cellular immune responses specific to foreign T-helper epitopes but not to self-Aß. CONCLUSIONS: This translational study demonstrates that a DNA-based epitope vaccine for AD could be appropriate for human clinical testing.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/therapy , Alzheimer Vaccines/therapeutic use , Lymphocyte Activation , T-Lymphocytes, Helper-Inducer/physiology , Amyloid beta-Peptides/immunology , Animals , Antibodies/blood , Antibodies/immunology , Cell Line, Tumor , Dose-Response Relationship, Immunologic , Epitopes, T-Lymphocyte , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Longitudinal Studies , Macaca mulatta , Male , Plaque, Amyloid/immunology , Random Allocation , Time Factors , Translational Research, Biomedical , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use
13.
Vaccines (Basel) ; 12(6)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38932388

ABSTRACT

The escalating global healthcare challenge posed by Alzheimer's Disease (AD) and compounded by the lack of effective treatments emphasizes the urgent need for innovative approaches to combat this devastating disease. Currently, passive and active immunotherapies remain the most promising strategy for AD. FDA-approved lecanemab significantly reduces Aß aggregates from the brains of early AD patients administered biweekly with this humanized monoclonal antibody. Although the clinical benefits noted in these trials have been modest, researchers have emphasized the importance of preventive immunotherapy. Importantly, data from immunotherapy studies have shown that antibody concentrations in the periphery of vaccinated people should be sufficient for targeting Aß in the CNS. To generate relatively high concentrations of antibodies in vaccinated people at risk of AD, we generated a universal vaccine platform, MultiTEP, and, based on it, developed a DNA vaccine, AV-1959D, targeting pathological Aß, completed IND enabling studies, and initiated a Phase I clinical trial with early AD volunteers. Our current pilot study combined our advanced MultiTEP technology with a novel mRNA approach to develop an mRNA vaccine encapsulated in lipid-based nanoparticles (LNPs), AV-1959LR. Here, we report our initial findings on the immunogenicity of 1959LR in mice and non-human primates, comparing it with the immunogenicity of its DNA counterpart, AV-1959D.

14.
Neurodegener Dis ; 10(1-4): 261-4, 2012.
Article in English | MEDLINE | ID: mdl-22301697

ABSTRACT

BACKGROUND: Induction of a humoral response against amyloid-ß peptide may be beneficial for Alzheimer's disease (AD) patients and may alleviate the onset and progression of AD. DNA-based vaccination provides a unique alternative method of immunization for treatment and prevention of AD. Currently, the two major delivery methods used for enhancing DNA uptake and immune responses to DNA vaccines in humans are electroporation (EP) and gene gun (GG). OBJECTIVE: The goal of this translational study was to evaluate the efficacy of an AD DNA epitope vaccine (DepVac) delivered intramuscularly by EP or intradermally by GG. METHODS: Humoral and cellular immune responses to immunization with DepVac were evaluated by ELISA and ELISPOT, respectively. Functional activity of the antibodies was also assessed. RESULTS: EP- and GG-mediated immunizations with DepVac induced similar anti-amyloid-ß (Aß) antibody and T cell responses. Anti-Aß antibodies bound to amyloid plaques in AD brain tissue and to toxic forms of Aß(42) peptide. CONCLUSION: Both delivery methods are effective at promoting potent antibodies specific for Aß.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/immunology , Antibodies/blood , Electroporation/methods , Vaccines, DNA/administration & dosage , Alzheimer Disease/blood , Alzheimer Disease/immunology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Animals , Biolistics/methods , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/immunology , Malaria Vaccines/immunology , Mice , Mice, Inbred C57BL , Peptide Fragments/immunology , Peptide Fragments/metabolism , Thymidine/metabolism , Time Factors , Tritium/metabolism
15.
NPJ Vaccines ; 7(1): 117, 2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36224191

ABSTRACT

Pathological forms of Tau protein are directly associated with neurodegeneration and correlate with Alzheimer's Disease (AD) symptoms, progression, and severity. Previously, using various mouse models of Tauopathies and AD, we have demonstrated the immunogenicity and efficacy of the MultiTEP-based adjuvanted vaccine targeting the phosphatase activating domain (PAD) of Tau, AV-1980R/A. Here, we analyzed its immunogenicity in non-human primates (NHP), the closest phylogenic relatives to humans with a similar immune system, to initiate the transition of this vaccine into clinical trials. We have demonstrated that AV-1980R/A is highly immunogenic in these NHPs, activating a broad but unique to each monkey repertoire of MultiTEP-specific T helper (Th) cells that, in turn, activate B cells specific to PAD. The resulting anti-PAD IgG antibodies recognize pathological Tau tangles and Tau-positive neuritis in AD case brain sections with no staining in control non-AD cases. These published data and efficacy results support the AV-1980R/A vaccine progression to first-in-human clinical trials.

16.
NPJ Vaccines ; 7(1): 1, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35013319

ABSTRACT

Accumulation of misfolded proteins such as amyloid-ß (Aß), tau, and α-synuclein (α-Syn) in the brain leads to synaptic dysfunction, neuronal damage, and the onset of relevant neurodegenerative disorder/s. Dementia with Lewy bodies (DLB) and Parkinson's disease (PD) are characterized by the aberrant accumulation of α-Syn intracytoplasmic Lewy body inclusions and dystrophic Lewy neurites resulting in neurodegeneration associated with inflammation. Cell to cell propagation of α-Syn aggregates is implicated in the progression of PD/DLB, and high concentrations of anti-α-Syn antibodies could inhibit/reduce the spreading of this pathological molecule in the brain. To ensure sufficient therapeutic concentrations of anti-α-Syn antibodies in the periphery and CNS, we developed four α-Syn DNA vaccines based on the universal MultiTEP platform technology designed especially for the elderly with immunosenescence. Here, we are reporting on the efficacy and immunogenicity of these vaccines targeting three B-cell epitopes of hα-Syn aa85-99 (PV-1947D), aa109-126 (PV-1948D), aa126-140 (PV-1949D) separately or simultaneously (PV-1950D) in a mouse model of synucleinopathies mimicking PD/DLB. All vaccines induced high titers of antibodies specific to hα-Syn that significantly reduced PD/DLB-like pathology in hα-Syn D line mice. The most significant reduction of the total and protein kinase resistant hα-Syn, as well as neurodegeneration, were observed in various brain regions of mice vaccinated with PV-1949D and PV-1950D in a sex-dependent manner. Based on these preclinical data, we selected the PV-1950D vaccine for future IND enabling preclinical studies and clinical development.

17.
J Transl Med ; 9: 127, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21806809

ABSTRACT

BACKGROUND: Numerous pre-clinical studies and clinical trials demonstrated that induction of antibodies to the ß-amyloid peptide of 42 residues (Aß42) elicits therapeutic effects in Alzheimer's disease (AD). However, an active vaccination strategy based on full length Aß42 is currently hampered by elicitation of T cell pathological autoreactivity. We attempt to improve vaccine efficacy by creating a novel chimeric flu vaccine expressing the small immunodominant B cell epitope of Aß42. We hypothesized that in elderly people with pre-existing memory Th cells specific to influenza this dual vaccine will simultaneously boost anti-influenza immunity and induce production of therapeutically active anti-Aß antibodies. METHODS: Plasmid-based reverse genetics system was used for the rescue of recombinant influenza virus containing immunodominant B cell epitopes of Aß42 (Aß1-7/10). RESULTS: Two chimeric flu viruses expressing either 7 or 10 aa of Aß42 (flu-Aß1-7 or flu-Aß1-10) were generated and tested in mice as conventional inactivated vaccines. We demonstrated that this dual vaccine induced therapeutically potent anti-Aß antibodies and anti-influenza antibodies in mice. CONCLUSION: We suggest that this strategy might be beneficial for treatment of AD patients as well as for prevention of development of AD pathology in pre-symptomatic individuals while concurrently boosting immunity against influenza.


Subject(s)
Alzheimer Disease/immunology , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Agglutination , Amyloid beta-Peptides/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , Antibody Specificity/immunology , Epitopes, B-Lymphocyte/immunology , Female , Humans , Immunity, Humoral/immunology , Immunization , Influenza, Human/virology , Kinetics , Mice , Mice, Inbred C57BL , Neutralization Tests , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
18.
J Transl Med ; 9: 213, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22168535

ABSTRACT

BORIS, or CTCFL, the so called Brother of the Regulator of Imprinted Sites because of the extensive homology in the central DNA binding region of the protein to the related regulator, CTCF, is expressed in early gametogenesis and in multiple cancers but not in differentiated somatic cells. Thus it is a member of the cancer testes antigen group (CTAs). Since BORIS and CTCF target common DNA binding sites, these proteins function on two levels, the first level is their regulation via the methylation context of the DNA target site and the second level is their distinct and different epigenetic associations due to differences in the non-homologous termini of the proteins. The regulation on both of these levels is extensive and complex and the sphere of influence of each of these proteins is associated with vastly different cellular signaling processes. On the level of gene expression, BORIS has three known promoters and multiple spliced mRNAs which adds another level of complexity to this intriguing regulator. BORIS expression is observed in the majority of cancer tissues and cell lines analyzed up to today. The expression profile and essential role of BORIS in cancer make this molecule very attractive target for cancer immunotherapy. This review summarizes what is known about BORIS regarding its expression, structure, and function and then presents some theoretical considerations with respect to its genome wide influence and its potential for use as a vaccine for cancer immunotherapy.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Genome, Human/genetics , CCCTC-Binding Factor , DNA-Binding Proteins/genetics , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Protein Binding , Repressor Proteins
19.
Cell Immunol ; 270(2): 188-97, 2011.
Article in English | MEDLINE | ID: mdl-21641588

ABSTRACT

Here, we analyze for the first time the immunological and therapeutic efficacy of a dendritic cell (DC) vaccine based on a cancer-testis antigen, Brother of regulator of imprinted sites (BORIS), an epigenetically acting tumor-promoting transcription factor. Vaccination of mice with DC loaded with truncated form of BORIS (DC/mBORIS) after 4T1 mammary tumor implantation induced strong anti-cancer immunity, inhibited tumor growth (18.75% of mice remained tumor-free), and dramatically lowered the number of spontaneous clonogenic metastases (50% of mice remained metastases-free). Higher numbers of immune effector CD4 and CD8 T cells infiltrated the tumors of vaccinated mice vs. control animals. Vaccination significantly decreased the number of myeloid-derived suppressor cells (MDSCs) infiltrating the tumor sites, but not MDSCs in the spleens of vaccinated animals. These data suggest that DC-based mBORIS vaccination strategies have significant anti-tumor activity in a therapeutic setting and will be more effective when combined with agents to attenuate tumor-associated immune suppression.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Mammary Neoplasms, Experimental/therapy , Animals , Female , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Testis/immunology , Vaccination/methods
20.
Sci Rep ; 9(1): 15455, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664089

ABSTRACT

Pathological tau correlates well with cognitive impairments in Alzheimer's disease (AD) patients and therefore represents a promising target for immunotherapy. Targeting an appropriate B cell epitope in pathological tau could in theory produce an effective reduction of pathology without disrupting the function of normal native tau. Recent data demonstrate that the N-terminal region of tau (aa 2-18), termed the "phosphatase activation domain (PAD)", is hidden within native Tau in a 'paperclip'-like conformation. Conversely, PAD is exposed in pathological tau and plays an essential role in the inhibition of fast axonal transport and tau polymerization. Thus, we hypothesized that anti-tau2-18 antibodies may safely and specifically reduce pathological tau and prevent further aggregation, which in turn would neutralize tau toxicity. Therefore, we evaluated the immunogenicity and therapeutic efficacy of our MultiTEP platform-based vaccine targeting tau2-18 formulated with AdvaxCpG adjuvant (AV-1980R/A) in PS19 tau transgenic mice. The AV-1980R/A induced extremely high antibody responses and the resulting sera recognized neurofibrillary tangles and plaque-associated dystrophic neurites in AD brain sections. In addition, under non-denaturing conditions AV-1980R/A sera preferentially recognized AD-associated tau. Importantly, vaccination also prevented age-related motor and cognitive deficits in PS19 mice and significantly reduced insoluble total and phosphorylated tau species. Taken together, these findings suggest that predominantly targeting misfolded tau with AV-1980R/A could represent an effective strategy for AD immunotherapy.


Subject(s)
Epitopes/immunology , Phosphoric Monoester Hydrolases/metabolism , Vaccines/immunology , tau Proteins/immunology , Animals , Antibody Formation , Immunotherapy , Mice , Neurofibrillary Tangles/immunology , Phosphorylation , tau Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL