Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 2(10): 100422, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34755134

RESUMEN

Provoked by sterile/nonsterile insults, prolonged monocyte mobilization and uncontrolled monocyte/macrophage activation can pose imminent or impending harm to the affected organs. Curiously, folate receptor beta (FRß), with subnanomolar affinity for the vitamin folic acid (FA), is upregulated during immune activation in hematopoietic cells of the myeloid lineage. This phenomenon has inspired a strong interest in exploring FRß-directed diagnostics/therapeutics. Previously, we have reported that FA-targeted aminopterin (AMT) therapy can modulate macrophage function and effectively treat animal models of inflammation. Our current investigation of a lead compound (EC2319) leads to discovery of a highly FR-specific mechanism of action independent of the root causes against inflammatory monocytes. We further show that EC2319 suppresses interleukin-6/interleukin-1ß release by FRß+ monocytes in a triple co-culture leukemic model of cytokine release syndrome with anti-CD19 chimeric antigen receptor T cells. Because of its chemical stability and metabolically activated linker, EC2319 demonstrates favorable pharmacokinetic characteristics and cross-species translatability to support future pre-clinical and clinical development.


Asunto(s)
Aminopterina/farmacología , Síndrome de Liberación de Citoquinas/prevención & control , Receptor 2 de Folato/genética , Antagonistas del Ácido Fólico/farmacología , Ácido Fólico/metabolismo , Macrófagos/efectos de los fármacos , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Células CHO , Cricetulus , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Femenino , Receptor 1 de Folato/antagonistas & inhibidores , Receptor 1 de Folato/genética , Receptor 1 de Folato/inmunología , Receptor 2 de Folato/antagonistas & inhibidores , Receptor 2 de Folato/inmunología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Modelos Biológicos , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/patología , Células RAW 264.7 , Ratas , Ratas Endogámicas Lew , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
2.
Int J Mol Sci ; 22(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070369

RESUMEN

Folate receptor beta (FRß) is a folate binding receptor expressed on myeloid lineage hematopoietic cells. FRß is commonly expressed at high levels on malignant blasts in patients with acute myeloid leukemia (AML), as well as on M2 polarized tumor-associated macrophages (TAMs) in the tumor microenvironment of many solid tumors. Therefore, FRß is a potential target for both direct and indirect cancer therapy. We demonstrate that FRß is expressed in both AML cell lines and patient-derived AML samples and that a high-affinity monoclonal antibody against FRß (m909) has the ability to cause dose- and expression-dependent ADCC against these cells in vitro. Importantly, we find that administration of m909 has a significant impact on tumor growth in a humanized mouse model of AML. Surprisingly, m909 functions in vivo with and without the infusion of human NK cells as mediators of ADCC, suggesting potential involvement of mouse macrophages as effector cells. We also found that TAMs from primary ovarian ascites samples expressed appreciable levels of FRß and that m909 has the ability to cause ADCC in these samples. These results indicate that the targeting of FRß using m909 has the potential to limit the outgrowth of AML in vitro and in vivo. Additionally, m909 causes cytotoxicity to TAMs in the tumor microenvironment of ovarian cancer warranting further investigation of m909 and its derivatives as therapeutic agents in patients with FRß-expressing cancers.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Receptor 2 de Folato , Inmunoterapia , Leucemia Mieloide Aguda , Proteínas de Neoplasias , Neoplasias Ováricas , Animales , Células CHO , Cricetulus , Femenino , Receptor 2 de Folato/antagonistas & inhibidores , Receptor 2 de Folato/inmunología , Células HL-60 , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Células THP-1 , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Biomed Pharmacother ; 131: 110711, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32919191

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic, fatal disease with high mortality and poor prognosis. It is characterized by a gradual decline in lung function, and there are currently no effective therapeutic methods. Folate is a water-soluble B vitamin that plays an important role in one-carbon transfer reactions, nucleic acid biosynthesis and methylation reactions. Studies have shown that folate may participate in the pathogenesis of IPF through ways of DNA repair, methylation, and reactive oxygen species. Macrophage activation is an important early cellular event in IPF and the inflammatory response that they trigger is a significant feature of IPF. Folate receptor-ß (FR-ß) is a cell surface glycosylphosphatidylinositol-anchored glycoprotein that can mediate the unidirectional transport of folate into cells. And it has been found in previous studies that FR-ß is usually overexpressed on activated macrophages, but the expression on resting macrophages was undetectable. Therefore, targeting FR-ß may have potential value for the early diagnosis and therapy of IPF. Our goal is to highlight the biological role of folate and FR-ß in IPF, and we hope to provide helpful insight for clinical treatment strategies.


Asunto(s)
Receptor 2 de Folato/fisiología , Ácido Fólico/fisiología , Fibrosis Pulmonar Idiopática/etiología , Macrófagos/fisiología , Polaridad Celular , Receptor 2 de Folato/antagonistas & inhibidores , Receptor 2 de Folato/química , Humanos , Fibrosis Pulmonar Idiopática/diagnóstico , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Receptor Toll-Like 4/fisiología
4.
Signal Transduct Target Ther ; 5(1): 6, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-32296026

RESUMEN

Tumor-associated macrophages (TAMs) facilitate cancer progression by promoting tumor invasion, angiogenesis, metastasis, inflammatory responses, and immunosuppression. Folate receptor ß (FRß) is overexpressed in TAMs. However, the clinical significance of FRß-positive macrophages in lung cancer remains poorly understood. In this study, we verified that FRß overexpression in lung cancer TAMs was associated with poor prognosis. We utilized a folate-modified lipoplex comprising a folate-modified liposome (F-PLP) delivering a BIM-S plasmid to target both lung cancer cells and FRß-positive macrophages in the tumor microenvironment. Transfection of LL/2 cells and MH-S cells with F-PLP/pBIM induced cell apoptosis. Injection of F-PLP/pBIM into LL/2 and A549 lung cancer models significantly depleted FRß-positive macrophages and reduced tumor growth. Treatment of tumor-bearing mice with F-PLP/pBIM significantly inhibited tumor growth in vivo by inducing tumor cell and macrophage apoptosis, reducing tumor proliferation, and inhibiting tumor angiogenesis. In addition, a preliminary safety evaluation demonstrated a good safety profile of F-PLP/pBIM as a gene therapy administered intravenously. This work describes a novel application of lipoplexes in lung cancer targeted therapy that influences the tumor microenvironment by targeting TAMs.


Asunto(s)
Receptor 2 de Folato/genética , Ácido Fólico/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Macrófagos Asociados a Tumores/efectos de los fármacos , Células A549 , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptor 2 de Folato/antagonistas & inhibidores , Ácido Fólico/química , Humanos , Liposomas/química , Liposomas/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Microambiente Tumoral/efectos de los fármacos
5.
Angew Chem Int Ed Engl ; 56(36): 10845-10849, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28686804

RESUMEN

Arsenic trioxide (ATO, As2 O3 ) is currently used to treat acute promyelocytic leukemia. However, expanding its use to include high-dose treatment of other cancers is severely hampered by serious side effects on healthy organs. To address these limitations, we loaded ATO onto folate (FA)-labeled human serum albumin (HSA) pretreated with glutathione (GSH) based on the low pH- and GSH-sensitive arsenic-sulfur bond, and we termed the resulting smart nanodrug as FA-HSA-ATO. FA-HSA-ATO could specifically recognize folate receptor-ß-positive (FRß+) chronic myeloid leukemia (CML) cells, resulting in more intracellular accumulation of ATO. Furthermore, the nanodrug could upregulate FRß expression in CML cancer cells and xenograft tumor model, facilitating even more recruitment and uptake of FRß-targeting drugs. In vitro and in vivo experiments indicate that the nanodrug significantly alleviates side effects and improves therapeutic efficacy of ATO on CML and xenograft tumor model.


Asunto(s)
Antineoplásicos/farmacología , Trióxido de Arsénico/farmacología , Receptor 2 de Folato/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Nanopartículas/química , Albúmina Sérica Humana/química , Animales , Antineoplásicos/química , Trióxido de Arsénico/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Receptor 2 de Folato/metabolismo , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Relación Estructura-Actividad
6.
Blood ; 125(22): 3466-76, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25887778

RESUMEN

T cells expressing a chimeric antigen receptor (CAR) can produce dramatic results in lymphocytic leukemia patients; however, therapeutic strategies for myeloid leukemia remain limited. Folate receptor ß (FRß) is a myeloid-lineage antigen expressed on 70% of acute myeloid leukemia (AML) patient samples. Here, we describe the development and evaluation of the first CARs specific for human FRß (m909) in vitro and in vivo. m909 CAR T cells exhibited selective activation and lytic function against engineered C30-FRß as well as endogenous FRß(+) AML cell lines in vitro. In mouse models of human AML, m909 CAR T cells mediated the regression of engrafted FRß(+) THP1 AML in vivo. In addition, we demonstrated that treatment of AML with all-trans retinoic acid (ATRA) enhanced FRß expression, resulting in improved immune recognition by m909 CAR T cells. Because many cell surface markers are shared between AML blasts and healthy hematopoietic stem and progenitor cells (HSCs), we evaluated FRß expression and recognition of HSCs by CAR T cells. m909 CAR T cells were not toxic against healthy human CD34(+) HSCs in vitro. Our results indicate that FRß is a promising target for CAR T-cell therapy of AML, which may be augmented by combination with ATRA.


Asunto(s)
Receptor 2 de Folato/antagonistas & inhibidores , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Animales , Células Cultivadas , Femenino , Receptor 2 de Folato/genética , Terapia Genética/métodos , Células HEK293 , Humanos , Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Terapia Molecular Dirigida , Proteínas Mutantes Quiméricas/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
7.
Arthritis Res Ther ; 14(3): R106, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22551402

RESUMEN

INTRODUCTION: We previously demonstrated that synovial sublining macrophages express folate receptor beta (FRß). The aim of this study was to evaluate the efficacy of intra-articular administration of a recombinant immunotoxin to FRß for treating rat antigen-induced arthritis. METHODS: A monoclonal antibody (mAb) to rat FRß was produced by immunizing mice with B300-19 cells (murine pre-B cells) transfected with the rat FRß gene. Recombinant immunotoxin was prepared by conjugating the Fv portion of the anti-rat FRß mAb heavy chain with a truncated Pseudomonas exotoxin A and the Fv portion of the anti-rat FRß mAb light chain. Antigen-induced arthritis was induced through intra-articular injection of methylated bovine serum albumin (mBSA) after two subcutaneous injections of mBSA and complete Freund's adjuvant. Immunotoxin was intra-articularly injected into the arthritis joint every other day for seven days after arthritis onset. Joint swelling was measured and histological scores of inflammation, synovial thickness, cartilage, and bone destruction were determined. Immunohistochemistry was performed to detect osteoclast and osteoclast precursor FRß-expressing macrophages and cathepsin K-positive cells on day 21. RESULTS: Intra-articular administration of the immunotoxin attenuated joint swelling (61% suppression; P < 0.01 compared to the control on day 21) and improved histological findings, particularly cartilage and bone destruction (scores of rats treated with control versus the immunotoxin: 2.2 versus 0.5; P < 0.01), by reducing the number of FRß-expressing macrophages and cathepsin K-positive cells. CONCLUSIONS: Intra-articular administration of an immunotoxin to FRß is effective for improving rat antigen-induced arthritis.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Receptor 2 de Folato/antagonistas & inhibidores , Receptor 2 de Folato/inmunología , Inmunotoxinas/administración & dosificación , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Artritis Reumatoide/patología , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Inmunotoxinas/inmunología , Inyecciones Intraarticulares , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratas , Ratas Endogámicas Lew
8.
Salud Publica Mex ; 52(5): 461-7, 2010.
Artículo en Español | MEDLINE | ID: mdl-21031253

RESUMEN

Fumonisins are mycotoxins that contaminate maize, disrupt the folate and sphingolipid metabolism, are associated with neural tube defects, and are considered by the International Agency for Research on Cancer (IARC) as possible human carcinogens. Since maize-based foods are significant components of the Mexican diet and there is a high prevalence of genetic susceptibility for folate deficiency among Mexicans, this essay presents international and national evidence of fumonisin exposure and the relevance that such exposure represents for Mexico.


Asunto(s)
Ácido Fólico/metabolismo , Contaminación de Alimentos , Fumonisinas/efectos adversos , Defectos del Tubo Neural/etiología , Adolescente , Adulto , Animales , Carcinógenos Ambientales/efectos adversos , Neoplasias del Sistema Digestivo/inducido químicamente , Neoplasias del Sistema Digestivo/epidemiología , Equidae , Femenino , Receptor 2 de Folato/antagonistas & inhibidores , Fumonisinas/química , Fumonisinas/farmacocinética , Fumonisinas/toxicidad , Homocistinuria/epidemiología , Homocistinuria/genética , Humanos , Necrosis Tubular Aguda/inducido químicamente , Leucoencefalopatías/inducido químicamente , Leucoencefalopatías/veterinaria , Masculino , Proteínas de Transporte de Membrana/metabolismo , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , México , Ratones , Espasticidad Muscular/epidemiología , Espasticidad Muscular/genética , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/epidemiología , Defectos del Tubo Neural/genética , Embarazo , Trastornos Psicóticos/epidemiología , Trastornos Psicóticos/genética , Ratas , Esfingolípidos/química , Esfingolípidos/metabolismo , Porcinos , Teratógenos/toxicidad , Adulto Joven , Zea mays/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...