Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Int J Mol Sci ; 24(12)2023 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-37372944

RESUMO

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.


Assuntos
Doença de Alzheimer , Vacinas Anticâncer , Camundongos , Animais , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Ácido Pirrolidonocarboxílico , Imunoterapia , Placa Amiloide/patologia , Encéfalo/metabolismo , Camundongos Transgênicos , Modelos Animais de Doenças
2.
Int J Mol Sci ; 24(6)2023 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-36983050

RESUMO

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.


Assuntos
Neoplasias Mamárias Animais , Vacinas , Animais , Masculino , Camundongos , Ratos , Proteínas de Ligação a DNA/metabolismo , Imunoterapia/métodos , Fatores de Transcrição
3.
J Immunol ; 200(8): 2656-2669, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29500244

RESUMO

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.


Assuntos
Medula Óssea/metabolismo , Células Dendríticas/metabolismo , Receptor 4 Toll-Like/agonistas , Animais , Células da Medula Óssea/metabolismo , Antígeno CD11c/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microambiente Tumoral/fisiologia
4.
Neurobiol Aging ; 59: 156-170, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28870518

RESUMO

We have previously demonstrated that anti-beta amyloid DNA vaccine (AV-1959D) based on our proprietary MultiTEP platform technology is extremely immunogenic in mice, rabbits, and monkeys. Importantly, MultiTEP platform enables development of vaccines targeting pathological molecules involved in various neurodegenerative disorders. Taking advantage of the universality of MultiTEP platform, we developed DNA vaccines targeting 3 B-cell epitopes (amino acids [aa]85-99, aa109-126, and aa126-140) of human alpha-synuclein (hα-Syn) separately or all 3 epitopes simultaneously. All 4 DNA vaccines (1) generate high titers of anti-hα-Syn antibodies and (2) induce robust MultiTEP-specific T-helper cell responses without activation of potentially detrimental autoreactive anti-hα-Syn T-helper cells. Generated antibodies recognize misfolded hα-Syn produced by neuroblastoma cells, hα-Syn in the brain tissues of transgenic mouse strains and in the brain tissues of dementia with Lewy body cases. Based on these results, the most promising vaccine targeting 3 B-cell epitopes of hα-Syn simultaneously (PV-1950D) has been chosen for ongoing preclinical assessment in mouse models of hα-Syn with the aim to translate it to the human clinical trials.


Assuntos
Epitopos de Linfócito B/imunologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/terapia , Vacinas de DNA/imunologia , alfa-Sinucleína/imunologia , Animais , Anticorpos , Feminino , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doenças Neurodegenerativas/genética , Linfócitos T Auxiliares-Indutores/imunologia , Vacinas de DNA/uso terapêutico
5.
Vaccine ; 35(16): 2015-2024, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28320590

RESUMO

BACKGROUND: By the time clinical symptoms of Alzheimer's disease (AD) manifest in patients there is already substantial tau pathology in the brain. Recent evidence also suggests that tau pathology can become self-propagating, further accelerating disease progression. Over the last decade several groups have tested the efficacy of protein-based anti-tau immunotherapeutics in various animal models of tauopathy. Here we report on the immunological and therapeutic potency of the first anti-tau DNA vaccine based on the MultiTEP platform, AV-1980D, in THY-Tau22 transgenic mice. METHODS: Starting at 3months of age, mice were immunized intramuscularly with AV-1980D vaccine targeting a tau B cell epitope spanning aa2-18 followed by electroporation (EP). Humoral and cellular immune responses in vaccinated animals were analyzed by ELISA and ELISpot, respectively. Neuropathological changes in the brains of experimental and control mice were then analyzed by biochemical (WB and ELISA) and immunohistochemical (IHC) methods at 9months of age. RESULTS: EP-mediated AV-1980D vaccinations of THY-Tau22 mice induced activation of Th cells specific to the MultiTEP vaccine platform and triggered robust humoral immunity response specific to tau. Importantly, no activation of potentially harmful autoreactive Th cell responses specific to endogenous tau species was detected. The maximum titers of anti-tau antibodies were reached after two immunizations and remained slightly lower, but steady during five subsequent monthly immunizations. Vaccinations with AV-1980D followed by EP significantly reduced total tau and pS199 and AT180 phosphorylated tau levels in the brains extracts of vaccinated mice, but produced on subtle non-significant effects on other phosphorylated tau species. CONCLUSIONS: These data demonstrate that MultiTEP-based DNA epitope vaccination targeting the N-terminus of tau is highly immunogenic and therapeutically potent in the THY-Tau22 mouse model of tauopathy and indicate that EP-mediated DNA immunization is an attractive alternative to protein-based adjuvanted vaccines for inducing high concentrations of anti-tau antibodies.


Assuntos
Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/terapia , Epitopos/imunologia , Vacinas de DNA/imunologia , Proteínas tau/imunologia , Imunidade Adaptativa , Doença de Alzheimer/patologia , Animais , Anticorpos/sangue , Western Blotting , Encéfalo/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , ELISPOT , Epitopos/genética , Feminino , Humanos , Imuno-Histoquímica , Injeções Intramusculares , Linfócitos/imunologia , Camundongos Transgênicos , Resultado do Tratamento , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Proteínas tau/genética
6.
J Transl Med ; 12: 322, 2014 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-25432242

RESUMO

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.


Assuntos
Neoplasias Mamárias Experimentais/terapia , Metástase Neoplásica , Extratos Vegetais/farmacologia , Receptor 4 Toll-Like/efeitos dos fármacos , Animais , Feminino , Linfócitos/imunologia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C
7.
Clin Exp Metastasis ; 31(2): 185-98, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24096737

RESUMO

It is believed that primary tumor resection modulates host-tumor immune interaction, but this has not been characterized in a stringent breast cancer tumor model. This report, using the 4T1 murine mammary tumor model, characterizes for the first time the dynamic longitudinal changes in immunosuppressive and effector components of the immune system after resection of an established orthotopic primary tumor with a defined natural history of developing lung metastases. More specifically, we analyzed changes of absolute numbers and frequencies of MDSC, regulatory T cells (Treg), as well as activated CD4 and CD8 positive T cells in spleens and, in some studies, lungs of 4T1 tumor-bearing mice and mice after primary tumor resection. Importantly, using mathematical analyses we established that primary resection of an orthotopic tumor had created a "window of opportunity" with decreased tumor-associated immune suppression that existed for approximately 10 days. Although tumor resection did slightly prolong survival, it did not affect the ultimate development of metastatic disease since animals with resected tumors or intact primary tumors eventually died by day 47 and 43, respectively. This window of opportunity likely occurs in humans providing a rationale and parameters for integration and testing of immunotherapeutic strategies in this critical "window of opportunity" to combat the development of metastatic disease.


Assuntos
Imunoterapia , Neoplasias Mamárias Experimentais/cirurgia , Neoplasias Mamárias Experimentais/terapia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias Mamárias Experimentais/imunologia , Camundongos
8.
Alzheimers Dement ; 10(3): 284-95, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-23916838

RESUMO

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.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/terapia , Vacinas contra Alzheimer/uso terapêutico , Ativação Linfocitária , Linfócitos T Auxiliares-Indutores/fisiologia , Peptídeos beta-Amiloides/imunologia , Animais , Anticorpos/sangue , Anticorpos/imunologia , Linhagem Celular Tumoral , Relação Dose-Resposta Imunológica , Epitopos de Linfócito T , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Estudos Longitudinais , Macaca mulatta , Masculino , Placa Amiloide/imunologia , Distribuição Aleatória , Fatores de Tempo , Pesquisa Translacional Biomédica , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico
9.
J Neurosci ; 33(15): 6245-56, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23575824

RESUMO

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.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/patologia , Transtornos Cognitivos/patologia , Hipocampo/patologia , Degeneração Neural/patologia , Placa Amiloide/patologia , Tauopatias/patologia , Fatores Etários , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Comportamento Animal , Angiopatia Amiloide Cerebral , Córtex Cerebral/metabolismo , Transtornos Cognitivos/complicações , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Feminino , Gliose/genética , Gliose/patologia , Hipocampo/metabolismo , Humanos , Masculino , Degeneração Neural/genética , Degeneração Neural/metabolismo , Placa Amiloide/genética , Presenilina-1/genética , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos , Tauopatias/metabolismo , Proteínas tau/metabolismo
10.
J Transl Med ; 9: 213, 2011 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-22168535

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Genoma Humano/genética , Fator de Ligação a CCCTC , Proteínas de Ligação a DNA/genética , Humanos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/terapia , Ligação Proteica , Proteínas Repressoras
11.
Cell Immunol ; 270(2): 188-97, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21641588

RESUMO

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.


Assuntos
Antígenos de Neoplasias/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Proteínas de Ligação a DNA/administração & dosagem , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/imunologia , Neoplasias Mamárias Experimentais/terapia , Animais , Feminino , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Masculino , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Testículo/imunologia , Vacinação/métodos
12.
Biochem Biophys Res Commun ; 370(1): 109-12, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18355444

RESUMO

Brother of the regulator of imprinted sites (BORIS) is an epigenetically acting transcription factor which represses the tumor inhibitor functions of the tumor suppressor protein CTCF. BORIS expression has not been documented in adult females, making it an exciting molecular target for drug development in breast cancer. Previously, we demonstrated that vaccination of mice with zing-finger (ZF)-deleted non-functional BORIS results in regression of breast cancer and generation of potent anti-tumor immune responses. RNAi induction can be used as an alternative approach for selective tumor cell killing. Short interfering RNA (siRNA) molecules targeting BORIS were generated and their efficacy was tested in MDA-MB-231 breast cancer and non-malignant epithelial cell lines. Treatment with BORIS-specific siRNA, but not control siRNA led to a concentration-dependent reduction in BORIS expression and proportional apoptotic death of the cancer but not control cells. To our knowledge this is first report demonstrating a critical role of BORIS in maintaining tumor cell viability.


Assuntos
Apoptose , Neoplasias da Mama/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , RNA Interferente Pequeno/farmacologia , Apoptose/genética , Sequência de Bases , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Proteínas de Ligação a DNA/genética , Feminino , Humanos , Dados de Sequência Molecular , RNA Interferente Pequeno/genética , Transfecção
13.
J Immunol ; 178(1): 566-73, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17182597

RESUMO

Brother of the regulator of imprinted sites (BORIS) was previously described as a transcription factor for epigenetic reprogramming the expression of which is strictly confined to germ cells of adult testes but is aberrantly activated in the vast majority of neoplastic cells. Considering the critical role of BORIS in cancerogenesis and the fact that its expression pattern may preclude thymic tolerance, we generated DNA- and protein-based mouse BORIS antitumor vaccines using a non-DNA-binding version of the BORIS molecule. Clinical use of BORIS as a vaccine Ag would require that certain safety concerns be met. Specifically, administration of the functional BORIS protein would hypothetically pose a risk of BORIS accelerating the progression of cancer. To alleviate such safety concerns, we have developed vaccines based on the BORIS molecule lacking the DNA-binding zinc fingers domain. To enhance anti-BORIS cellular immune responses, we used a standard molecular adjuvant approach. It consisted of plasmids encoding murine IL-12 and IL-18 for a DNA-based vaccine and conventional Th1 type adjuvant, Quil A, for a protein-based vaccine. Both DNA- and protein-based vaccines induced Ag-specific CD4(+) T cell proliferation with Th1 and Th2 cytokine profiles, respectively. Protein-based, but not DNA-based, BORIS vaccine induced a significant level of Ab production in immunized animals. Importantly, potent anticancer CD8(+)-cytotoxic lymphocytes were generated after immunization with the DNA-based, but not protein-based, BORIS vaccine. These cytolytic responses were observed across a wide range of different mouse cancers including mammary adenocarcinoma, glioma, leukemia, and mastocytoma.


Assuntos
Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacologia , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/farmacologia , Neoplasias/imunologia , Células Th1/efeitos dos fármacos , Adjuvantes Imunológicos/administração & dosagem , Animais , Formação de Anticorpos , Antígenos de Neoplasias/administração & dosagem , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos CD4/análise , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Proteínas de Ligação a DNA/genética , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunização , Interleucina-12/genética , Interleucina-18/genética , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/patologia , Plasmídeos/genética , Deleção de Sequência , Testículo/imunologia , Células Th1/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia
14.
DNA Cell Biol ; 25(10): 571-80, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17132088

RESUMO

In Alzheimer's disease (AD) the accumulation of pathological forms of the beta-amyloid (Abeta) peptide are believed to be causal factors in the neurodegeneration that results in the loss of cognitive function in patients. Anti-Abeta antibodies have been shown to reduce Abeta levels in transgenic mouse models of AD and in AN-1792 clinical trial on AD patients; however, the clinical trial was halted when some patients developed meningoencephalitis. Theories on the cause of the adverse events include proinflammatory "primed patients," a Th1-inducing adjuvant, and Abeta autoreactive T cells. New immunotherapy approaches are being developed to eliminate these putative risk factors. Mannan, which is recognized by pattern recognition receptors of the innate immune system, can be utilized as a molecular adjuvant to promote a Th2-mediated immune response to conjugated B cell epitopes. The N-terminus of Abeta was conjugated to mannan, and used to immunize mice with low concentrations of immunoconjugate, without a conventional adjuvant. Mannan induced a significant and highly polarized toward Th2 phenotype anti-Abeta antibody response not only in BALB/c, but also in B6SJL F1 mice. New preclinical trials in AD mouse models may help to develop novel immunogen-adjuvant configurations with the potential to avoid the adverse immune response that occurred in the first clinical trial.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/química , Imunoterapia/métodos , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/farmacologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Epitopos de Linfócito B/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peptídeos/química , Estrutura Terciária de Proteína , Fatores de Risco
15.
J Cell Biochem ; 98(5): 1037-43, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16741971

RESUMO

Cancer testis (CT) antigens are promising candidates for tumor vaccines due to their immunogenicity and tissue-restricted expression. Recently, we identified a novel cancer testis gene, BORIS, whose expression is restricted to male testis after puberty and is strictly absent in non-malignant female tissue. BORIS encodes a DNA-binding protein that shares 11 zing finger (ZF) with transcription factor CTCF and differs at the N- and C-termini. CTCF has been implicated in epigenetic regulation of imprinting, X chromosome inactivation, repression, and activation of cancer testis antigens. BORIS expression has been documented in cancers of diverse histological origin, including, but not limited to breast, prostate, ovary, gastric, liver, endometrial, glia, colon, and esophagus. Interestingly, BORIS induces demethylation and subsequent expression of many cancer-testis genes, including MAGE-A1 and NY-ESO-1, indicating that it is expressed very early in malignancy and might be an attractive candidate for immunotherapy. In this study we tested BORIS as a vaccine in a very aggressive, highly metastatic, and poorly immunogenic murine model of mammary carcinoma. Immunizations with a DNA encoding the mutant form of murine BORIS antigen (pmBORIS lacking DNA-binding function) significantly prolonged survival, and inhibited tumor growth in BALB/c mice inoculated with 4T1 cells. Priming with pmBORIS mixed with molecular adjuvant and boosting with adenoviral vector expressing mBORIS was generally more effective, suggesting that the vaccination protocol could be further optimized. This is the first report demonstrating the feasibility of vaccination with a cancer associated epigenetic regulator for the induction of tumor inhibition.


Assuntos
Adjuvantes Imunológicos , Antígeno B7-1/imunologia , Vacinas Anticâncer/imunologia , Proteínas de Ligação a DNA/imunologia , DNA/genética , DNA/imunologia , Vacinação , Animais , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Cricetinae , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Vetores Genéticos/genética , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Taxa de Sobrevida
16.
J Transl Med ; 4: 22, 2006 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-16725035

RESUMO

Historically cancer vaccines have yielded suboptimal clinical results. We have developed a novel strategy for eliciting antitumor immunity based upon homology between neoplastic tissue and the developing placenta. Placenta formation shares several key processes with neoplasia, namely: angiogenesis, activation of matrix metalloproteases, and active suppression of immune function. Immune responses against xenoantigens are well known to break self-tolerance. Utilizing xenogeneic placental protein extracts as a vaccine, we have successfully induced anti-tumor immunity against B16 melanoma in C57/BL6 mice, whereas control xenogeneic extracts and B16 tumor extracts where ineffective, or actually promoted tumor growth, respectively. Furthermore, dendritic cells were able to prime tumor immunity when pulsed with the placental xenoantigens. While vaccination-induced tumor regression was abolished in mice depleted of CD4 T cells, both CD4 and CD8 cells were needed to adoptively transfer immunity to naïve mice. Supporting the role of CD8 cells in controlling tumor growth are findings that only freshly isolated CD8 cells from immunized mice were capable of inducing tumor cell caspases-3 activation ex vivo. These data suggest feasibility of using xenogeneic placental preparations as a multivalent vaccine potently targeting not just tumor antigens, but processes that are essential for tumor maintenance of malignant potential.

17.
Clin Exp Metastasis ; 20(6): 489-98, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14598882

RESUMO

Human epithelial mucin (MUC1) is expressed by many carcinomas, including breast cancer cells. This breast cancer-associated antigen has been widely used for immunotherapy, despite the fact that cellular immune responses to MUC1 are impaired in breast cancer patients and MUC1 transgenic animals. Previously, we found that immunogenicity to MUC1 was also impaired in BALB/c mice injected with a mammary tumor cell line (410.4) expressing human MUC1. We suggested that one reason for its weak immunogenicity was the lack of expression of B7 molecules by 410.4 cells. Recognition of antigenic epitopes in conjunction with MHCI/II by the T-cell receptor without co-stimulation by B7/CD28 association resulted in T-cell anergy. Therefore, we attempted to enhance protective anti-MUC1-specific immunity in mice using B7 co-stimulatory molecules as a component of the MUC1 vaccine. We also compared cell-based with DNA-based vaccination strategies. One group of mice was vaccinated with an irradiated, 410.4 syngeneic mammary tumor cell line co-expressing human MUC1 and CD80 or CD86 co-stimulatory molecules, and a second group of mice was vaccinated with plasmids encoding MUC1 and CD80 or CD86. These mice along with appropriate controls were challenged with mammary tumor cell line 4T1, which expresses MUC1. There were significant inhibition on rates of tumor growth and survival in mice vaccinated with irradiated 410.4/MUC1 cells co-expressing either CD80 or CD86 molecules, compared to non-vaccinated animals. In addition, there were also significant delays in the appearance of measurable tumors and their growth in mice vaccinated by gene-gun immunization with plasmids encoding MUC1 and CD80 or CD86.


Assuntos
Antígeno B7-1/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Neoplasias Mamárias Animais/prevenção & controle , Mucina-1/uso terapêutico , Vacinas Sintéticas/uso terapêutico , Animais , Divisão Celular , Feminino , Humanos , Neoplasias Mamárias Animais/mortalidade , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Taxa de Sobrevida , Fatores de Tempo
18.
J Immunol ; 171(8): 4311-9, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14530356

RESUMO

There is great interest in understanding the role of costimulatory molecules in immune activation. In both the influenza and HIV DNA immunization models, several groups have reported that coimmunization of mice with plasmids encoding immunogen and CD86, but not CD80, effectively boosts Ag-specific T cell activation. This difference in immune priming provided an opportunity to examine the functional importance of different regions of the B.7 molecules in immune activation. To examine this issue, we developed a series of chimeric CD80 and CD86 constructs as well as deletion mutants, and examined their immune activating potential in the DNA vaccine model. We demonstrate that the lack of an Ig constant-like region in the CD80 molecule is critically important to the enhanced immune activation observed. CD80 C-domain deletion mutants induce a highly inflammatory Ag-specific cellular response when administered as part of a plasmid vaccine. The data suggest that the constant-like domains, likely through intermolecular interactions, are critically important for immune regulation during costimulation and that engineered CD80/86 molecules represent more potent costimulatory molecules and may improve vaccine adjuvant efficacy.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/fisiologia , Antígenos CD/fisiologia , Antígeno B7-1/fisiologia , Regiões Constantes de Imunoglobulina/fisiologia , Glicoproteínas de Membrana/fisiologia , Modelos Imunológicos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/antagonistas & inibidores , Adjuvantes Imunológicos/genética , Animais , Antígenos CD/administração & dosagem , Antígenos CD/biossíntese , Antígenos CD/genética , Antígeno B7-1/administração & dosagem , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Antígeno B7-2 , Movimento Celular/genética , Movimento Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação para Baixo/genética , Regulação para Baixo/imunologia , HIV-1/genética , HIV-1/imunologia , Humanos , Regiões Constantes de Imunoglobulina/administração & dosagem , Regiões Constantes de Imunoglobulina/genética , Região Variável de Imunoglobulina/administração & dosagem , Região Variável de Imunoglobulina/genética , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Glicoproteínas de Membrana/administração & dosagem , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Plasmídeos , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Deleção de Sequência/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Vírus 40 dos Símios/genética , Vírus 40 dos Símios/imunologia , Transfecção , Regulação para Cima/genética , Regulação para Cima/imunologia , Vacinas de DNA/genética
19.
Int Immunol ; 15(4): 505-14, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12663680

RESUMO

The role of adjuvant on the T(h)1 and T(h)2 immune responses to Abeta-immunotherapy (Abeta(42 )peptide) was examined in wild-type mice. Fine epitope analysis with overlapping oligomers of the Abeta(42) sequence identified the 1-15 region as a dominant B cell epitope. The 6-20 peptide was recognized only weakly by antisera from mice administrated with Abeta(42) peptide formulated in complete Freund's adjuvant (CFA), alum or TiterMax Gold (TMG). However, mice immunized with Abeta(42) mixed with QS21 induced a significant antibody response to the 6-20 peptide. The only T cell epitope found was within the 6-28 sequence of Abeta(42). QS21 and CFA induced the strongest humoral response to Abeta, alum was intermediate, and TMG the weakest adjuvant. Analysis of antibody isotypes specific for Abeta indicates that alum induces primarily T(h)2-type immune response, whereas TMG, CFA and QS21 shift the immune responses toward a T(h)1 phenotype. Stimulation of splenocytes from Abeta-immunized mice with Abeta(40) peptide induced strikingly different cytokine expression profiles. QS21 and CFA induced significant IFN-gamma, IL-4 and tumor necrosis factor-alpha expression, whereas alum induced primarily IL-4 production. As T(h)1-type immune responses have been implicated in many autoimmune disorders, whereas T(h)2-type responses have been shown to inhibit autoimmune disease, the choice of adjuvant may be critical for the design of a safe and effective immunotherapy for Alzheimer's disease.


Assuntos
Adjuvantes Imunológicos/farmacologia , Peptídeos beta-Amiloides/imunologia , Fragmentos de Peptídeos/imunologia , Células Th1/imunologia , Células Th2/imunologia , Compostos de Alúmen/farmacologia , Animais , Formação de Anticorpos , Linfócitos B/imunologia , Citocinas/biossíntese , Mapeamento de Epitopos , Feminino , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Saponinas/farmacologia , Linfócitos T/imunologia
20.
DNA Cell Biol ; 21(3): 137-49, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12015893

RESUMO

Effective activation of T cells requires engagement of two separate T-cell receptors. The antigen-specific T-cell receptor (TCR) binds foreign peptide antigen-MHC complexes, and the CD28 receptor binds to the B7 (CD80/CD86) costimulatory molecules expressed on the surface of antigen-presenting cells (APC). The simultaneous triggering of these T-cell surface receptors with their specific ligands results in an activation of this cell. In contrast, CTLA-4 (CD152) is a distinct T-cell receptor that, upon binding to B7 molecules, sends an inhibitory signal to T cell activation. Many in vitro and in vivo studies demonstrated that both CD80 and CD86 ligands have an identical role in the activation of T cells. Recently, functions of B7 costimulatory molecules in vivo have been investigated in B7-1 and/or B7-2 knockout mice, and the authors concluded that CD86 could be more important for initiating T-cell responses, while CD80 could be more significant for maintaining these immune responses. In this study, we directly compared the role of CD80 and CD86 in initiating and maintaining proliferation of resting CD4(+) T cells in an in vitro mode system that allowed to provide the first signal-to-effector cells through the use of suboptimal doses of PHA and the second costimulatory signal through cells expressing CD80 or CD86, but not any other costimulatory molecules. Using this experimental system we demonstrate that the CD80 and CD86 molecules can substitute for each other in the initial activation of resting CD4(+) T cells and in the maintenance of their proliferative response.


Assuntos
Antígenos CD/fisiologia , Antígeno B7-1/fisiologia , Linfócitos T CD4-Positivos/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/fisiologia , Fito-Hemaglutininas/farmacologia , Antígeno B7-2 , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Cinética , Fito-Hemaglutininas/administração & dosagem , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA