Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Nature ; 603(7900): 328-334, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35197632

RESUMO

Effective antitumour immunity depends on the orchestration of potent T cell responses against malignancies1. Regression of human cancers has been induced by immune checkpoint inhibitors, T cell engagers or chimeric antigen receptor T cell therapies2-4. Although CD8 T cells function as key effectors of these responses, the role of CD4 T cells beyond their helper function has not been defined. Here we demonstrate that a trispecific antibody to HER2, CD3 and CD28 stimulates regression of breast cancers in a humanized mouse model through a mechanism involving CD4-dependent inhibition of tumour cell cycle progression. Although CD8 T cells directly mediated tumour lysis in vitro, CD4 T cells exerted antiproliferative effects by blocking cancer cell cycle progression at G1/S. Furthermore, when T cell subsets were adoptively transferred into a humanized breast cancer tumour mouse model, CD4 T cells alone inhibited HER2+ breast cancer growth in vivo. RNA microarray analysis revealed that CD4 T cells markedly decreased tumour cell cycle progression and proliferation, and also increased pro-inflammatory signalling pathways. Collectively, the trispecific antibody to HER2 induced T cell-dependent tumour regression through direct antitumour and indirect pro-inflammatory/immune effects driven by CD4 T cells.


Assuntos
Neoplasias da Mama , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Feminino , Humanos , Camundongos , Receptor ErbB-2/genética
3.
Sci Transl Med ; 13(610): eabc7804, 2021 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-34516826

RESUMO

Local immunotherapy ideally stimulates immune responses against tumors while avoiding toxicities associated with systemic administration. Current strategies for tumor-targeted, gene-based delivery, however, are limited by adverse effects such as off-targeting or antivector immunity. We investigated the intratumoral administration of saline-formulated messenger (m)RNA encoding four cytokines that were identified as mediators of tumor regression across different tumor models: interleukin-12 (IL-12) single chain, interferon-α (IFN-α), granulocyte-macrophage colony-stimulating factor, and IL-15 sushi. Effective antitumor activity of these cytokines relied on multiple immune cell populations and was accompanied by intratumoral IFN-γ induction, systemic antigen-specific T cell expansion, increased granzyme B+ T cell infiltration, and formation of immune memory. Antitumor activity extended beyond the treated lesions and inhibited growth of distant tumors and disseminated tumors. Combining the mRNAs with immunomodulatory antibodies enhanced antitumor responses in both injected and uninjected tumors, thus improving survival and tumor regression. Consequently, clinical testing of this cytokine-encoding mRNA mixture is now underway.


Assuntos
Citocinas , Neoplasias , Citocinas/genética , Humanos , Neoplasias/genética , Neoplasias/terapia , RNA Mensageiro
4.
Oncoimmunology ; 10(1): 1881268, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33796402

RESUMO

Immune checkpoint blockade elicits durable anti-cancer responses in the clinic, however a large proportion of patients do not benefit from treatment. Several mechanisms of innate and acquired resistance to checkpoint blockade have been defined and include mutations of MHC I and IFNγ signaling pathways. However, such mutations occur in a low frequency of patients and additional mechanisms have yet to be elucidated. In an effort to better understand acquired resistance to checkpoint blockade, we generated a mouse tumor model exhibiting in vivo resistance to anti-PD-1 antibody treatment. MC38 tumors acquired resistance to PD-1 blockade following serial in vivo passaging. Lack of sensitivity to PD-1 blockade was not attributed to dysregulation of PD-L1 or ß2M expression, as both were expressed at similar levels in parental and resistant cells. Similarly, IFNγ signaling and antigen processing and presentation pathways were functional in both parental and resistant cell lines. Unbiased gene expression analysis was used to further characterize potential resistance mechanisms. RNA-sequencing revealed substantial differences in global gene expression, with tumors resistant to anti-PD-1 displaying a marked reduction in expression of immune-related genes relative to parental MC38 tumors. Indeed, resistant tumors exhibited reduced immune infiltration across multiple cell types, including T and NK cells. Pathway analysis revealed activation of TGFß and Notch signaling in anti-PD-1 resistant tumors, and activation of these pathways was associated with poorer survival in human cancer patients. While pharmacological inhibition of TGFß and Notch in combination with PD-1 blockade decelerated tumor growth, a local mRNA-based immunotherapy potently induced regression of resistant tumors, resulting in complete tumor remission, and resensitized tumors to treatment with anti-PD-1. Overall, this study describes a novel anti-PD-1 resistant mouse tumor model and underscores the role of two well-defined signaling pathways in response to immune checkpoint blockade. Furthermore, our data highlights the potential of intratumoral mRNA therapy in overcoming acquired resistance to PD-1 blockade.


Assuntos
Imunoterapia , Neoplasias , Animais , Apresentação de Antígeno , Modelos Animais de Doenças , Humanos , Camundongos , RNA Mensageiro/genética
5.
Mol Cancer Ther ; 20(2): 250-262, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33310762

RESUMO

Primary treatment for estrogen receptor-positive (ER+) breast cancer is endocrine therapy. However, substantial evidence indicates a continued role for ER signaling in tumor progression. Selective estrogen receptor degraders (SERD), such as fulvestrant, induce effective ER signaling inhibition, although clinical studies with fulvestrant report insufficient blockade of ER signaling, possibly due to suboptimal pharmaceutical properties. Furthermore, activating mutations in the ER have emerged as a resistance mechanism to current endocrine therapies. New oral SERDs with improved drug properties are under clinical investigation, but the biological profile that could translate to improved therapeutic benefit remains unclear. Here, we describe the discovery of SAR439859, a novel, orally bioavailable SERD with potent antagonist and degradation activities against both wild-type and mutant Y537S ER. Driven by its fluoropropyl pyrrolidinyl side chain, SAR439859 has demonstrated broader and superior ER antagonist and degrader activities across a large panel of ER+ cells, compared with other SERDs characterized by a cinnamic acid side chain, including improved inhibition of ER signaling and tumor cell growth. Similarly, in vivo treatment with SAR439859 demonstrated significant tumor regression in ER+ breast cancer models, including MCF7-ESR1 wild-type and mutant-Y537S mouse tumors, and HCI013, a patient-derived tamoxifen-resistant xenograft tumor. These findings indicate that SAR439859 may provide therapeutic benefit to patients with ER+ breast cancer, including those who have resistance to endocrine therapy with both wild-type and mutant ER.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Receptores de Estrogênio/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos
6.
Adv Sci (Weinh) ; 7(24): 2002997, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33344141

RESUMO

Nonalcoholic steatohepatitis (NASH) is a progressive form of nonalcoholic fatty liver disease that can lead to irreversible liver cirrhosis and cancer. Early diagnosis of NASH is vital to detect disease before it becomes life-threatening, yet noninvasively differentiating NASH from simple steatosis is challenging. Herein, bifunctional probes have been developed that target the hepatocyte-specific asialoglycoprotein receptor (ASGPR), the expression of which decreases during NASH progression. The results show that the probes allow longitudinal, noninvasive monitoring of ASGPR levels by positron emission tomography in the newly developed rat model of NASH. The probes open new possibilities for research into early diagnosis of NASH and development of drugs to slow or reverse its progression.

7.
Mol Ther ; 28(2): 664-676, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31843448

RESUMO

Patients with α-dystroglycanopathies, a subgroup of rare congenital muscular dystrophies, present with a spectrum of clinical manifestations that includes muscular dystrophy as well as CNS and ocular abnormalities. Although patients with α-dystroglycanopathies are genetically heterogeneous, they share a common defect of aberrant post-translational glycosylation modification of the dystroglycan alpha-subunit, which renders it defective in binding to several extracellular ligands such as laminin-211 in skeletal muscles, agrin in neuromuscular junctions, neurexin in the CNS, and pikachurin in the eye, leading to various symptoms. The genetic heterogeneity associated with the development of α-dystroglycanopathies poses significant challenges to developing a generalized treatment to address the spectrum of genetic defects. Here, we propose the development of a bispecific antibody (biAb) that functions as a surrogate molecular linker to reconnect laminin-211 and the dystroglycan beta-subunit to ameliorate sarcolemmal fragility, a primary pathology in patients with α-dystroglycan-related muscular dystrophies. We show that the treatment of LARGEmyd-3J mice, an α-dystroglycanopathy model, with the biAb improved muscle function and protected muscles from exercise-induced damage. These results demonstrate the viability of a biAb that binds to laminin-211 and dystroglycan simultaneously as a potential treatment for patients with α-dystroglycanopathy.


Assuntos
Anticorpos Biespecíficos/farmacologia , Distroglicanas/metabolismo , Laminina/metabolismo , Síndrome de Walker-Warburg/metabolismo , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/metabolismo , Modelos Animais de Doenças , Distroglicanas/imunologia , Expressão Gênica , Humanos , Imuno-Histoquímica , Injeções Intramusculares , Laminina/genética , Laminina/imunologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Ligação Proteica/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas/genética , Sarcolema/efeitos dos fármacos , Sarcolema/metabolismo , Síndrome de Walker-Warburg/tratamento farmacológico , Síndrome de Walker-Warburg/etiologia
8.
PLoS One ; 14(12): e0226245, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31887144

RESUMO

Antibody therapies for Alzheimer's Disease (AD) hold promise but have been limited by the inability of these proteins to migrate efficiently across the blood brain barrier (BBB). Central nervous system (CNS) gene transfer by vectors like adeno-associated virus (AAV) overcome this barrier by allowing the bodies' own cells to produce the therapeutic protein, but previous studies using this method to target amyloid-ß have shown success only with truncated single chain antibodies (Abs) lacking an Fc domain. The Fc region mediates effector function and enhances antigen clearance from the brain by neonatal Fc receptor (FcRn)-mediated reverse transcytosis and is therefore desirable to include for such treatments. Here, we show that single chain Abs fused to an Fc domain retaining FcRn binding, but lacking Fc gamma receptor (FcγR) binding, termed a silent scFv-IgG, can be expressed and released into the CNS following gene transfer with AAV. While expression of canonical IgG in the brain led to signs of neurotoxicity, this modified Ab was efficiently secreted from neuronal cells and retained target specificity. Steady state levels in the brain exceeded peak levels obtained by intravenous injection of IgG. AAV-mediated expression of this scFv-IgG reduced cortical and hippocampal plaque load in a transgenic mouse model of progressive ß-amyloid plaque accumulation. These findings suggest that CNS gene delivery of a silent anti-Aß scFv-IgG was well-tolerated, durably expressed and functional in a relevant disease model, demonstrating the potential of this modality for the treatment of Alzheimer's disease.


Assuntos
Doença de Alzheimer/terapia , Sistema Nervoso Central/metabolismo , Vetores Genéticos/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Anticorpos de Cadeia Única/genética , Doença de Alzheimer/genética , Animais , Barreira Hematoencefálica , Linhagem Celular , Dependovirus/genética , Modelos Animais de Doenças , Progressão da Doença , Terapia Genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Camundongos Transgênicos , Domínios Proteicos , Receptores Fc/metabolismo , Receptores de IgG/metabolismo , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/metabolismo
9.
PLoS One ; 14(12): e0225835, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31794591

RESUMO

Systemic inhibition of dipeptidyl peptidase 4 (dpp4) represents an effective and established treatment option for type 2 diabetes (T2D). The current study investigated in mice if a liver selective knock-down of dpp4 by therapeutic siRNAs could be a novel, similarly effective treatment option for T2D. Furthermore, the potential effects on hepatic steatosis, inflammation and lipid metabolism were investigated after hepato-selective knock-down of dpp4. The knock-down efficiency and IC50 values of siRNAs targeting dpp4 were analyzed in PC3 cells. In two independent studies, either db/db mice or C57BL/6J mice were injected intravenously with a liposomal formulation of siRNAs targeting either dpp4 or a non-targeting control, followed by metabolically characterization. In comparator groups, additional cohorts of mice were treated with an oral dpp4 inhibitor. In both animal studies, we observed a robust knock-down (~75%) of hepatic dpp4 with a potent siRNA. Hepatic dpp4 knockdown did not significantly affect glucose metabolism or circulating incretin concentrations in both animal studies. However, in obese and diabetic db/db mice hepatic steatosis was reduced and hepatic mRNA expression of acaca, scd1, fasn and pparg was significantly lower after siRNA treatment. Systemic inhibition of the enzymatic dpp4 activity by an oral dpp4 inhibitor significantly improved glucose handling in db/db mice but did not affect hepatic endpoints. These data demonstrate that a targeted reduction of dpp4 expression in the liver may not be sufficient to improve whole-body glucose metabolism in obese and diabetic mice but may improve hepatic lipid metabolism.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , RNA Interferente Pequeno/metabolismo , Animais , Linhagem Celular Tumoral , Dipeptidil Peptidase 4/metabolismo , Feminino , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Hiperglicemia/metabolismo , Inflamação/genética , Inflamação/patologia , Fígado/patologia , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Especificidade de Órgãos
10.
Mol Ther ; 27(8): 1495-1506, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31208914

RESUMO

Neuronopathic glycosphingolipidoses are a sub-group of lysosomal storage disorders for which there are presently no effective therapies. Here, we evaluated the potential of substrate reduction therapy (SRT) using an inhibitor of glucosylceramide synthase (GCS) to decrease the synthesis of glucosylceramide (GL1) and related glycosphingolipids. The substrates that accumulate in Sandhoff disease (e.g., ganglioside GM2 and its nonacylated derivative, lyso-GM2) are distal to the drug target, GCS. Treatment of Sandhoff mice with a GCS inhibitor that has demonstrated CNS access (Genz-682452) reduced the accumulation of GL1 and GM2, as well as a variety of disease-associated substrates in the liver and brain. Concomitant with these effects was a significant decrease in the expression of CD68 and glycoprotein non-metastatic melanoma B protein (Gpnmb) in the brain, indicating a reduction in microgliosis in the treated mice. Moreover, using in vivo imaging, we showed that the monocytic biomarker translocator protein (TSPO), which was elevated in Sandhoff mice, was normalized following Genz-682452 treatment. These positive effects translated in turn into a delay (∼28 days) in loss of motor function and coordination, as measured by rotarod latency, and a significant increase in longevity (∼17.5%). Together, these results support the development of SRT for the treatment of gangliosidoses, particularly in patients with residual enzyme activity.


Assuntos
Carbamatos/farmacologia , Inibidores Enzimáticos/farmacologia , Glucosiltransferases/antagonistas & inibidores , Quinuclidinas/farmacologia , Doença de Sandhoff/enzimologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Ligantes , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Espectrometria de Massas , Camundongos , Camundongos Knockout , Imagem Molecular , Receptores de GABA/metabolismo , Doença de Sandhoff/diagnóstico , Doença de Sandhoff/genética , Doença de Sandhoff/terapia , Esfingolipídeos/metabolismo , Cadeia beta da beta-Hexosaminidase/genética , Cadeia beta da beta-Hexosaminidase/metabolismo
11.
Sci Rep ; 8(1): 469, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29323153

RESUMO

Achondroplasia, the most common form of dwarfism, affects more than a quarter million people worldwide and remains an unmet medical need. Achondroplasia is caused by mutations in the fibroblast growth factor receptor 3 (FGFR3) gene which results in over-activation of the receptor, interfering with normal skeletal development leading to disproportional short stature. Multiple mouse models have been generated to study achondroplasia. The characterization of these preclinical models has been primarily done with 2D measurements. In this study, we explored the transgenic model expressing mouse Fgfr3 containing the achondroplasia mutation G380R under the Col2 promoter (Ach). Survival and growth rate of the Ach mice were reduced compared to wild-type (WT) littermates. Axial skeletal defects and abnormalities of the sternebrae and vertebrae were observed in the Ach mice. Further evaluation of the Ach mouse model was performed by developing 3D parameters from micro-computed tomography (micro-CT) and magnetic resonance imaging (MRI). The 3-week-old mice showed greater differences between the Ach and WT groups compared to the 6-week-old mice for all parameters. Deeper understanding of skeletal abnormalities of this model will help guide future studies for evaluating novel and effective therapeutic approaches for the treatment of achondroplasia.


Assuntos
Acondroplasia/diagnóstico por imagem , Cifose/diagnóstico por imagem , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Coluna Vertebral/anormalidades , Acondroplasia/genética , Acondroplasia/mortalidade , Animais , Modelos Animais de Doenças , Humanos , Cifose/etiologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Transgênicos , Mutação , Coluna Vertebral/diagnóstico por imagem , Taxa de Sobrevida , Microtomografia por Raio-X
12.
Toxicol Pathol ; 45(7): 799-833, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29113559

RESUMO

The 2017 annual National Toxicology Program Satellite Symposium, entitled "Pathology Potpourri," was held in Montreal, Quebec, Canada at the Society of Toxicologic Pathology's 36th annual meeting. The goal of this symposium was to present and discuss challenging diagnostic pathology and/or nomenclature issues. This article presents summaries of the speakers' talks along with select images that were used by the audience for voting and discussion. Various lesions and other topics covered during the symposium included renal papillary degeneration in perinatally exposed animals, an atriocaval mesothelioma, an unusual presentation of an alveolar-bronchiolar carcinoma, a paraganglioma of the organ of Zuckerkandl (also called an extra-adrenal pheochromocytoma), the use of human muscle samples to illustrate the challenges of manual scoring of fluorescent staining, intertubular spermatocytic seminomas, medical device pathology assessment and discussion of the approval process, collagen-induced arthritis, incisor denticles, ameloblast degeneration and poorly mineralized enamel matrix, connective tissue paragangliomas, microcystin-LR toxicity, perivascular mast cells in the forebrain thalamus unrelated to treatment, and 2 cases that provided a review of the International Harmonization of Nomenclature and Diagnostic Criteria (INHAND) bone nomenclature and recommended application of the terminology in routine nonclinical toxicity studies.


Assuntos
Congressos como Assunto , Técnicas e Procedimentos Diagnósticos , Patologia , Sociedades Científicas , Toxicologia , Animais , Humanos , Processamento de Imagem Assistida por Computador , Quebeque
13.
J Gen Virol ; 98(4): 749-753, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28086071

RESUMO

Bovine adenovirus (AdV) type 3 (BAdV-3) E1 region shares functional homology with E1 of human AdV type C5. Sequence analysis of the BAdV-3 E1 region revealed the presence of a novel 155R ORF that is not observed in other AdVs, on the lower strand antiparallel to a portion of the E1B region. The 155R gene products in BAdV-3-infected cells were identified by Northern blot, reverse transcriptase PCR followed by sequencing and Western blot analysis using the155R-specific antibody. 155R seems to be a late protein and is present in purified BAdV-3 particles. Replication kinetics of BAdV mutants with either one (BAdV/155R/mt1) or two (BAdV/155R/mt2) stop codons in the 155R ORF were comparable to those of BAdV-3, indicating that 155R is not essential for virus replication in cell culture. These results suggest that 155R-deleted BAdV-3 vectors could be generated in a cell line that fully complements BAdV-3 E1 functions.


Assuntos
Adenoviridae/genética , Adenoviridae/fisiologia , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Replicação Viral , Animais , Northern Blotting , Western Blotting , Bovinos , Análise Mutacional de DNA , Perfilação da Expressão Gênica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
14.
Oncotarget ; 8(70): 114526-114539, 2017 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-29383099

RESUMO

Hepatocellular carcinoma (HCC) represents a serious public health challenge with few therapeutic options available to cancer patients.Wnt/ß-catenin pathway is thought to play a significant role in HCC pathogenesis. In this study, we confirmed high frequency of CTNNB1 (ß-catenin) mutations in two independent cohorts of HCC patients and demonstrated significant upregulation of ß-catenin protein in the overwhelming majority of HCC patient samples, patient-derived xenografts (PDX) and established cell lines. Using genetic tools validated for target specificity through phenotypic rescue experiments, we went on to investigate oncogenic dependency on ß-catenin in an extensive collection of human HCC cells lines. Our results demonstrate that dependency on ß-catenin generally tracks with its activation status. HCC cell lines that harbored activating mutations in CTNNB1 or displayed elevated levels of non-phosphorylated (active) ß-catenin were significantly more sensitive to ß-catenin siRNA treatment than cell lines with wild-type CTNNB1 and lower active ß-catenin. Finally, significant therapeutic benefit of ß-catenin knock-down was demonstrated in established HCC tumor xenografts using doxycycline-inducible shRNA system. ß-catenin downregulation and tumor growth inhibition was associated with reduction in AXIN2, direct transcriptional target of ß-catenin, and decreased cancer cell proliferation as measured by Ki67 staining. Taken together, our data highlight fundamental importance of aberrant ß-catenin signaling in the maintenance of oncogenic phenotype in HCC.

15.
Mol Med ; 21: 389-99, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25938659

RESUMO

Fabry disease, an X-linked glycosphingolipid storage disorder, is caused by the deficient activity of α-galactosidase A (α-Gal A). This results in the lysosomal accumulation in various cell types of its glycolipid substrates, including globotriaosylceramide (GL-3) and lysoglobotriaosylceramide (globotriaosyl lysosphingolipid, lyso-GL-3), leading to kidney, heart, and cerebrovascular disease. To complement and potentially augment the current standard of care, biweekly infusions of recombinant α-Gal A, the merits of substrate reduction therapy (SRT) by selectively inhibiting glucosylceramide synthase (GCS) were examined. Here, we report the development of a novel, orally available GCS inhibitor (Genz-682452) with pharmacological and safety profiles that have potential for treating Fabry disease. Treating Fabry mice with Genz-682452 resulted in reduced tissue levels of GL-3 and lyso-GL-3 and a delayed loss of the thermal nociceptive response. Greatest improvements were realized when the therapeutic intervention was administered to younger mice before they developed overt pathology. Importantly, as the pharmacologic profiles of α-Gal A and Genz-682452 are different, treating animals with both drugs conferred the greatest efficacy. For example, because Genz-682452, but not α-Gal A, can traverse the blood-brain barrier, levels of accumulated glycosphingolipids were reduced in the brain of Genz-682452-treated but not α-Gal A-treated mice. These results suggest that combining substrate reduction and enzyme replacement may confer both complementary and additive therapeutic benefits in Fabry disease.


Assuntos
Carbamatos/administração & dosagem , Doença de Fabry/tratamento farmacológico , Glucosiltransferases/metabolismo , Glicolipídeos/metabolismo , Quinuclidinas/administração & dosagem , Esfingolipídeos/metabolismo , Triexosilceramidas/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Modelos Animais de Doenças , Doença de Fabry/metabolismo , Doença de Fabry/patologia , Glucosiltransferases/antagonistas & inibidores , Humanos , Camundongos , alfa-Galactosidase/administração & dosagem , alfa-Galactosidase/metabolismo
16.
Am J Pathol ; 185(3): 651-65, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25553976

RESUMO

Fabry disease is an X-linked lysosomal storage disease caused by deficient activity of α-galactosidase A and the resultant systemic accumulation of globotrioasylceramide (GL-3) and related glycolipids. α-Galactosidase A gene knockout (Gla KO) mice have no α-galactosidase A activity and progressively accumulate GL-3 in tissues and fluids, similarly to FD patients. The nature and temporal effects of the progressive substrate accumulation on tissue histology in these mice have not previously been characterized. Here, we report the pathology of young to old (3 to 17 months old) Gla KO mice and compare these changes with those in strain-matched control animals. Gla KO mice accumulated GL-3 in various tissues and fluids with age. Lysosomal GL-3 inclusions increased with age in multiple cell types, including renal epithelial, intestinal, and vascular smooth muscle cells, and neurons in trigeminal and dorsal root ganglia, as detected by light and electron microscopy. However, unlike the case for male FD patients with the type 1 classic phenotype, GL-3 inclusions were not detected in vascular endothelial cells or cardiomyocytes. The histological changes in Gla KO mice better resemble the type 2 later-onset phenotype observed in patients with residual α-galactosidase A activity. GL-3 accumulation in the small intestine and sensory ganglia of Gla KO mice provides a model for study of enteropathy and neuropathy in Fabry disease.


Assuntos
Doença de Fabry/patologia , Intestinos/patologia , Rim/patologia , Músculo Liso Vascular/patologia , alfa-Galactosidase/genética , Animais , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patologia , Modelos Animais de Doenças , Progressão da Doença , Doença de Fabry/genética , Doença de Fabry/metabolismo , Feminino , Humanos , Mucosa Intestinal/metabolismo , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , alfa-Galactosidase/metabolismo
17.
J Inherit Metab Dis ; 37(6): 1013-22, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24850378

RESUMO

Fabry disease, a rare X-linked α-galactosidase A deficiency, causes progressive lysosomal accumulation of globotriaosylceramide (GL-3) in a variety of cell types. As the disease progresses, renal failure, left ventricular hypertrophy, and strokes may occur. Enzyme replacement therapy (ERT), with recombinant α-galactosidase A, is currently available for use to reduce GL-3 deposits. However, although it improves cardiac function and decreases left ventricular mass, GL-3 clearance upon ERT has been demonstrated in cardiac capillary endothelium but not in cardiomyocytes of patients. Relevant models are needed to understand the pathogenesis of cardiac disease and explore new therapeutic approaches. We generated induced pluripotent stem cells (iPSC) from Fabry patients and differentiated them into cardiomyocytes. In these cells, GL-3 accumulates in the lysosomes over time, resulting in phenotypic changes similar to those found in cardiac tissue from Fabry patients. Using this human in vitro model, we demonstrated that substrate reduction therapy via glucosylceramide synthase inhibition was able to prevent accumulation and to clear lysosomal GL-3 in cardiomyocytes. This new in vitro model recapitulates essential features of cardiomyocytes from patients with Fabry disease and therefore provides a useful and relevant tool for further investigations of new therapy.


Assuntos
Doença de Fabry/tratamento farmacológico , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Triexosilceramidas/metabolismo , alfa-Galactosidase/uso terapêutico , Adolescente , Células Cultivadas , Criança , Progressão da Doença , Terapia de Reposição de Enzimas , Humanos , Lisossomos/metabolismo , Masculino , Fenótipo
18.
J Gene Med ; 14(2): 77-89, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22228563

RESUMO

BACKGROUND: The receptor tyrosine kinase EphA2 is overexpressed in several types of cancers and is currently being pursued as a target for breast cancer therapeutics. The EphA2 ligand EphrinA1 induces EphA2 phosphorylation and intracellular internalization and degradation, thus inhibiting tumor progression. The hematopoietic growth factor, FMS-like tyrosine kinase 3 receptor ligand (Flt3L), promotes expansion and mobilization of functional dendritic cells. METHODS: We tested the EphrinA1-EphA2 interaction in MDA-MB-231 breast cancer cells focusing on the receptor-ligand-mediated apoptosis of breast cancer cells. To determine whether EphrinA1-EphA2 interaction-associated apoptosis and Flt3L-mediated immunotherapy would have an additive effect in inhibiting tumor growth, we used an immunocompetent mouse model of breast cancer to evaluate intratumoral (i.t.) inoculation strategies with human adenovirus (HAd) vectors expressing either EphrinA1 (HAd-EphrinA1-Fc), Flt3L (HAd-Flt3L) or a combination of EphrinA1-Fc + Flt3L (HAd-EphrinA1-Fc + HAd-Flt3L). RESULTS: In vitro analysis demonstrated that an EphrinA1-EphA2 interaction led to apoptosis-related changes in breast cancer cells. In vivo, three i.t. inoculations of HAd-EphrinA1-Fc showed potent inhibition of tumor growth. Furthermore, increased inhibition in tumor growth was observed with the combination of HAd-EphrinA1-Fc and HAd-Flt3L accompanied by the generation of an anti-tumor adaptive immune response. CONCLUSIONS: The results obtained in the present study, indicating the induction of apoptosis and inhibition of mammary tumor growth, show the potential therapeutic benefits of HAd-EphrinA1-Fc. In combination with HAd-Flt3L, this represents a promising strategy for effectively inducing mammary tumor regression by HAd vector-based therapy.


Assuntos
Apoptose/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Efrina-A1/metabolismo , Imunoterapia/métodos , Receptor EphA2/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Adenoviridae , Análise de Variância , Animais , Western Blotting , Linhagem Celular Tumoral , Células Dendríticas/metabolismo , ELISPOT , Feminino , Vetores Genéticos , Humanos , Imuno-Histoquímica , Camundongos , Fosforilação , Proteólise , Reação em Cadeia da Polimerase em Tempo Real
19.
Virus Res ; 163(1): 202-11, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21971215

RESUMO

The potential of a bovine adenovirus serotype 3 (BAd3)-based vector to bypass the human adenoviral serotype 5 (HAd5)-specific neutralizing immune response was evaluated in an immunocompetent mouse model of breast cancer. Initially we monitored vector biodistribution, genome persistence, transgene expression, and potential toxicity of HAd-GFP [HAd5 vector expressing green fluorescent protein (GFP)] or BAd-GFP (BAd3 vector expressing GFP) in FVB/n mice bearing tumors. A comparable biodistribution pattern for BAd-GFP and HAd-GFP was evident. In addition, following the development of vector-specific immune responses, animals were inoculated intratumorally (i.t.) with HAd-GFP or BAd-GFP. HAd-GFP immunity did not hamper the transduction and persistence of BAd-GFP into the tumors and other organs, and, similarly, BAd-GFP immunity did not hamper the transduction and persistence of HAd-GFP. Both BAd3 and HAd5 vectors showed relatively higher transgene expression in the presence of heterologous vector immunity. In contrast, the homologous vector immunity was associated with a rapid vector clearance and decline in transgene expression levels. Histopathological changes in BAd-GFP inoculated animals were generally mild with some acute but recoverable hepatic perturbations. Overall, the data suggest the importance of BAd3 vectors for sequential vector administration in overcoming the vector immunity for cancer gene therapy.


Assuntos
Adenoviridae/imunologia , Produtos Biológicos/administração & dosagem , Neoplasias da Mama/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Adenoviridae/genética , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Produtos Biológicos/imunologia , Modelos Animais de Doenças , Feminino , Expressão Gênica , Vetores Genéticos/imunologia , Humanos , Camundongos , Transdução Genética , Transgenes , Resultado do Tratamento
20.
Int J Oncol ; 40(2): 479-86, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22075622

RESUMO

Choriocarcinomas are a rare form of cancer that develops in the uterus from tissue which would normally become the placenta. Choriocarcinomas are a trophoblastic gestational disease and have been studied largely to investigate conditions related to pregnancy such as preeclampsia. Choriocarcinomas are highly angiogenic and produce high levels of placental growth factor (PlGF) to promote the development of blood vessels. Upregulation of PlGF expression also occurs during the development of other human malignancies such as breast cancer and melanoma. Both tumor specimens and plasma samples have higher levels of PlGF than normal tissues. Hence, PlGF has emerged as a valid target for anti-angiogenic therapy. The cell lines BeWo, JAR and JEG-3, derived from human choriocarcinomas, were investigated in vitro and in vivo for suitability as PlGF-dependent models. BeWo, JAR and JEG-3 cells were characterized in culture and were implanted into immunodeficient mice to generate subcutaneous tumors. The PlGF and VEGF angiogenic profiles of the choriocarcinoma cells and tumors were investigated by ELISA and by immunohistochemical methods. Double immunofluorescence methods were applied to choriocarcinoma xenograft sections to characterize the cellular components of the blood vessels. sFLT01, a fusion protein that neutralizes PlGF, was assessed in cell culture experiments and xenograft studies. The novel results presented here validate the importance of human choriocarcinoma cell lines and xenografts in further exploring the role of PlGF in tumor angiogenesis, for evaluating PlGF as an anti-angiogenic target, and for developing therapies that may provide clinical benefit.


Assuntos
Antineoplásicos/farmacologia , Coriocarcinoma/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Proteínas da Gravidez/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Neoplasias Uterinas/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Coriocarcinoma/irrigação sanguínea , Coriocarcinoma/patologia , Meios de Cultivo Condicionados/química , Feminino , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fator de Crescimento Placentário , Gravidez , Proteínas Recombinantes de Fusão/uso terapêutico , Carga Tumoral , Neoplasias Uterinas/irrigação sanguínea , Neoplasias Uterinas/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA