Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Immunity ; 53(5): 1095-1107.e3, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33128877

RESUMEN

Developing effective strategies to prevent or treat coronavirus disease 2019 (COVID-19) requires understanding the natural immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We used an unbiased, genome-wide screening technology to determine the precise peptide sequences in SARS-CoV-2 that are recognized by the memory CD8+ T cells of COVID-19 patients. In total, we identified 3-8 epitopes for each of the 6 most prevalent human leukocyte antigen (HLA) types. These epitopes were broadly shared across patients and located in regions of the virus that are not subject to mutational variation. Notably, only 3 of the 29 shared epitopes were located in the spike protein, whereas most epitopes were located in ORF1ab or the nucleocapsid protein. We also found that CD8+ T cells generally do not cross-react with epitopes in the four seasonal coronaviruses that cause the common cold. Overall, these findings can inform development of next-generation vaccines that better recapitulate natural CD8+ T cell immunity to SARS-CoV-2.


Asunto(s)
Betacoronavirus/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Coronavirus/inmunología , Neumonía Viral/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adulto , Anciano , Betacoronavirus/aislamiento & purificación , COVID-19 , Convalecencia , Coronavirus/inmunología , Infecciones por Coronavirus/diagnóstico , Proteínas de la Nucleocápside de Coronavirus , Mapeo Epitopo , Epítopos de Linfocito T , Femenino , Humanos , Epítopos Inmunodominantes , Memoria Inmunológica , Masculino , Persona de Mediana Edad , Proteínas de la Nucleocápside/inmunología , Pandemias , Fosfoproteínas , Neumonía Viral/diagnóstico , Poliproteínas , SARS-CoV-2 , Proteínas Virales/inmunología , Adulto Joven
3.
Proc Natl Acad Sci U S A ; 116(15): 7533-7542, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30898885

RESUMEN

Activation of the Met receptor tyrosine kinase, either by its ligand, hepatocyte growth factor (HGF), or via ligand-independent mechanisms, such as MET amplification or receptor overexpression, has been implicated in driving tumor proliferation, metastasis, and resistance to therapy. Clinical development of Met-targeted antibodies has been challenging, however, as bivalent antibodies exhibit agonistic properties, whereas monovalent antibodies lack potency and the capacity to down-regulate Met. Through computational modeling, we found that the potency of a monovalent antibody targeting Met could be dramatically improved by introducing a second binding site that recognizes an unrelated, highly expressed antigen on the tumor cell surface. Guided by this prediction, we engineered MM-131, a bispecific antibody that is monovalent for both Met and epithelial cell adhesion molecule (EpCAM). MM-131 is a purely antagonistic antibody that blocks ligand-dependent and ligand-independent Met signaling by inhibiting HGF binding to Met and inducing receptor down-regulation. Together, these mechanisms lead to inhibition of proliferation in Met-driven cancer cells, inhibition of HGF-mediated cancer cell migration, and inhibition of tumor growth in HGF-dependent and -independent mouse xenograft models. Consistent with its design, MM-131 is more potent in EpCAM-high cells than in EpCAM-low cells, and its potency decreases when EpCAM levels are reduced by RNAi. Evaluation of Met, EpCAM, and HGF levels in human tumor samples reveals that EpCAM is expressed at high levels in a wide range of Met-positive tumor types, suggesting a broad opportunity for clinical development of MM-131.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/farmacología , Molécula de Adhesión Celular Epitelial/antagonistas & inhibidores , Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Molécula de Adhesión Celular Epitelial/metabolismo , Humanos , Ratones , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Nature ; 547(7664): 453-457, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28678785

RESUMEN

Plasticity of the cell state has been proposed to drive resistance to multiple classes of cancer therapies, thereby limiting their effectiveness. A high-mesenchymal cell state observed in human tumours and cancer cell lines has been associated with resistance to multiple treatment modalities across diverse cancer lineages, but the mechanistic underpinning for this state has remained incompletely understood. Here we molecularly characterize this therapy-resistant high-mesenchymal cell state in human cancer cell lines and organoids and show that it depends on a druggable lipid-peroxidase pathway that protects against ferroptosis, a non-apoptotic form of cell death induced by the build-up of toxic lipid peroxides. We show that this cell state is characterized by activity of enzymes that promote the synthesis of polyunsaturated lipids. These lipids are the substrates for lipid peroxidation by lipoxygenase enzymes. This lipid metabolism creates a dependency on pathways converging on the phospholipid glutathione peroxidase (GPX4), a selenocysteine-containing enzyme that dissipates lipid peroxides and thereby prevents the iron-mediated reactions of peroxides that induce ferroptotic cell death. Dependency on GPX4 was found to exist across diverse therapy-resistant states characterized by high expression of ZEB1, including epithelial-mesenchymal transition in epithelial-derived carcinomas, TGFß-mediated therapy-resistance in melanoma, treatment-induced neuroendocrine transdifferentiation in prostate cancer, and sarcomas, which are fixed in a mesenchymal state owing to their cells of origin. We identify vulnerability to ferroptic cell death induced by inhibition of a lipid peroxidase pathway as a feature of therapy-resistant cancer cells across diverse mesenchymal cell-state contexts.


Asunto(s)
Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Cadherinas/metabolismo , Muerte Celular , Línea Celular Tumoral , Linaje de la Célula , Transdiferenciación Celular , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal , Humanos , Hierro/metabolismo , Peróxidos Lipídicos/metabolismo , Masculino , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Melanoma/metabolismo , Melanoma/patología , Mesodermo/efectos de los fármacos , Mesodermo/enzimología , Mesodermo/metabolismo , Mesodermo/patología , Neoplasias/genética , Neoplasias/patología , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteómica , Proteínas Proto-Oncogénicas B-raf/genética , Reproducibilidad de los Resultados , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
5.
Mol Neuropsychiatry ; 2(2): 97-106, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27606323

RESUMEN

Many studies suggest the presence of aberrations in cellular metabolism in bipolar disorder. We studied the metabolome in bipolar disorder to gain insight into cellular pathways that may be dysregulated in bipolar disorder and to discover evidence of novel biomarkers. We measured polar and nonpolar metabolites in fibroblasts from subjects with bipolar I disorder and matched healthy control subjects, under normal conditions and with two physiologic perturbations: low-glucose media and exposure to the stress-mediating hormone dexamethasone. Metabolites that were significantly different between bipolar and control subjects showed distinct separation by principal components analysis methods. The most statistically significant findings were observed in the perturbation experiments. The metabolite with the lowest p value in both the low-glucose and dexamethasone experiments was α-aminoadipate, whose intracellular level was consistently lower in bipolar subjects. Our study implicates α-aminoadipate as a possible biomarker in bipolar disorder that manifests under cellular stress. This is an intriguing finding given the known role of α-aminoadipate in the modulation of kynurenic acid in the brain, especially as abnormal kynurenic acid levels have been implicated in bipolar disorder.

6.
ACS Chem Biol ; 11(2): 363-74, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26640968

RESUMEN

Modulation of histone deacetylase (HDAC) activity has been implicated as a potential therapeutic strategy for multiple diseases. However, it has been difficult to dissect the role of individual HDACs due to a lack of selective small-molecule inhibitors. Here, we report the synthesis of a series of highly potent and isoform-selective class I HDAC inhibitors, rationally designed by exploiting minimal structural changes to the clinically experienced HDAC inhibitor CI-994. We used this toolkit of isochemogenic or chemically matched inhibitors to probe the role of class I HDACs in ß-cell pathobiology and demonstrate for the first time that selective inhibition of an individual HDAC isoform retains beneficial biological activity and mitigates mechanism-based toxicities. The highly selective HDAC3 inhibitor BRD3308 suppressed pancreatic ß-cell apoptosis induced by inflammatory cytokines, as expected, or now glucolipotoxic stress, and increased functional insulin release. In addition, BRD3308 had no effect on human megakaryocyte differentiation, while inhibitors of HDAC1 and 2 were toxic. Our findings demonstrate that the selective inhibition of HDAC3 represents a potential path forward as a therapy to protect pancreatic ß-cells from inflammatory cytokines and nutrient overload in diabetes.


Asunto(s)
Citoprotección/efectos de los fármacos , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Diseño de Fármacos , Inhibidores de Histona Desacetilasas/farmacocinética , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Humanos , Células Secretoras de Insulina/citología , Datos de Secuencia Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratas
7.
Cell Rep ; 10(5): 755-770, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25660025

RESUMEN

Novel therapeutic approaches are urgently required for multiple myeloma (MM). We used a phenotypic screening approach using co-cultures of MM cells with bone marrow stromal cells to identify compounds that overcome stromal resistance. One such compound, BRD9876, displayed selectivity over normal hematopoietic progenitors and was discovered to be an unusual ATP non-competitive kinesin-5 (Eg5) inhibitor. A novel mutation caused resistance, suggesting a binding site distinct from known Eg5 inhibitors, and BRD9876 inhibited only microtubule-bound Eg5. Eg5 phosphorylation, which increases microtubule binding, uniquely enhanced BRD9876 activity. MM cells have greater phosphorylated Eg5 than hematopoietic cells, consistent with increased vulnerability specifically to BRD9876's mode of action. Thus, differences in Eg5-microtubule binding between malignant and normal blood cells may be exploited to treat multiple myeloma. Additional steps are required for further therapeutic development, but our results indicate that unbiased chemical biology approaches can identify therapeutic strategies unanticipated by prior knowledge of protein targets.

8.
ACS Chem Biol ; 10(3): 883-90, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25546293

RESUMEN

We examined the effects of isoform-specific histone deacetylase (HDAC) inhibitors on ß-catenin posttranslational modifications in neural progenitor cells (NPCs) derived from human induced pluripotent stem cells (iPSCs). ß-catenin is a multifunctional protein with important roles in the developing and adult central nervous system. Activation of the Wnt pathway results in stabilization and nuclear translocation of ß-catenin, resulting in activation of multiple target genes. In addition, ß-catenin forms a complex with cadherins at the plasma membrane as part of the adherens junctions. The N-terminus of ß-catenin has phosphorylation, ubiquitination, and acetylation sites that regulate its stability and signaling. In the absence of a Wnt signal, Ser33, Ser37, and Thr41 are constitutively phosphorylated by glycogen synthase kinase 3ß (GSK3ß). ß-Catenin phosphorylated at these sites is recognized by ß-transducin repeat-containing protein (ßTrCP), which results in ubiquitination and degradation by the ubiquitin-proteasome pathway. The N-terminal regulatory domain of ß-catenin also includes Ser45, a phosphorylation site for Casein Kinase 1α (CK1α) and Lys49, which is acetylated by the acetyltransferase p300/CBP-associated factor (PCAF). The relevance of Lys49 acetylation and Ser45 phosphorylation to the function of ß-catenin is an active area of investigation. We find that HDAC6 inhibitors increase Lys49 acetylation and Ser45 phosphorylation but do not affect Ser33, Ser37, and Thr41 phosphorylation. Lys49 acetylation results in decreased ubiquitination of ß-catenin in the presence of proteasome inhibition. While increased Lys49 acetylation does not affect total levels of ß-catenin, it results in increased membrane localization of ß-catenin.


Asunto(s)
Membrana Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Neuronas/efectos de los fármacos , Procesamiento Proteico-Postraduccional , beta Catenina/metabolismo , Acetilación , Sitios de Unión , Diferenciación Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Células Cultivadas , Expresión Génica , Inhibidores de Histona Desacetilasas/química , Histona Desacetilasas/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Lisina/química , Lisina/metabolismo , Modelos Moleculares , Neuronas/metabolismo , Neuronas/ultraestructura , Fosforilación , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transducción de Señal , Relación Estructura-Actividad , Ubiquitinación , beta Catenina/química , beta Catenina/genética
9.
Nat Chem Biol ; 9(12): 840-848, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24161946

RESUMEN

Efforts to develop more effective therapies for acute leukemia may benefit from high-throughput screening systems that reflect the complex physiology of the disease, including leukemia stem cells (LSCs) and supportive interactions with the bone marrow microenvironment. The therapeutic targeting of LSCs is challenging because LSCs are highly similar to normal hematopoietic stem and progenitor cells (HSPCs) and are protected by stromal cells in vivo. We screened 14,718 compounds in a leukemia-stroma co-culture system for inhibition of cobblestone formation, a cellular behavior associated with stem-cell function. Among those compounds that inhibited malignant cells but spared HSPCs was the cholesterol-lowering drug lovastatin. Lovastatin showed anti-LSC activity in vitro and in an in vivo bone marrow transplantation model. Mechanistic studies demonstrated that the effect was on target, via inhibition of HMG-CoA reductase. These results illustrate the power of merging physiologically relevant models with high-throughput screening.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Leucemia , Células Madre Neoplásicas/efectos de los fármacos , Línea Celular Tumoral , Células Madre Hematopoyéticas , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lovastatina/farmacología , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/fisiología
10.
Curr Opin Investig Drugs ; 11(12): 1409-23, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21154123

RESUMEN

More than 50 years after the introduction of the dihydrofolate reductase inhibitor, methotrexate, new antifolates have emerged and have been incorporated into the chemotherapeutic armamentarium. These include pralatrexate, with the same target as methotrexate, but with enhanced properties, and pemetrexed, with different enzyme targets and properties. Current synthetic efforts are focused on developing antifolates that are selectively delivered to cancer cells, but not to normal proliferating cells, exploiting the different properties of folate transporters. In another novel approach, drugs structurally and mechanistically unrelated to folates are linked to and use folic acid as a carrier to be endocytosed by folate receptors and then released to inhibit their cellular targets. This review describes the evolution and current status of antifolate pharmacology and prospects for the development of the next generation of folate analogs.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Antagonistas del Ácido Fólico/farmacología , Transportadores de Ácido Fólico/metabolismo , Ácido Fólico/análogos & derivados , Antimetabolitos Antineoplásicos/metabolismo , Portadores de Fármacos , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/metabolismo , Humanos , Péptidos/metabolismo
11.
Oncologist ; 12(7): 808-15, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17673612

RESUMEN

Pemetrexed is a novel antifolate recently approved for the treatment of pleural mesothelioma and non-small cell lung cancer. In clinical regimens, pemetrexed is administered in conjunction with folic acid to minimize toxicity. However, excessive folate supplementation may also diminish the activity of this agent. The current study demonstrates, in several human solid tumor cell lines, that when extracellular 5-formyltetrahydrofolate levels are increased in vitro, within the range of normal human blood levels, there is a substantial decrease in pemetrexed activity upon continuous exposure to the drug. This was accompanied by a comparable lower level of trimetrexate activity consistent with an expansion of tumor cell folate pools. Likewise, when cells were exposed to pemetrexed with a schedule that simulates in vivo pharmacokinetics, there was markedly less cell killing with higher extracellular folate levels. Data are provided to indicate that 5-formyltetrahydrofolate is an acceptable surrogate for 5-methyltetrahydrofolate, the major blood folate, for this type of in vitro study. These observations and other reports suggest that, in view of the rise in serum folate and fall in serum homocysteine that has accompanied folic acid supplementation of food in the U.S., the addition of folic acid to regimens with pemetrexed should be limited to the lowest recommended level that provides optimal protection from pemetrexed toxicity.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Antagonistas del Ácido Fólico/farmacocinética , Ácido Fólico/farmacología , Glutamatos/farmacocinética , Guanina/análogos & derivados , Tetrahidrofolatos/farmacocinética , Antimetabolitos Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Ácido Fólico/uso terapéutico , Antagonistas del Ácido Fólico/administración & dosificación , Glutamatos/administración & dosificación , Guanina/administración & dosificación , Guanina/farmacocinética , Humanos , Técnicas In Vitro , Neoplasias Pulmonares/tratamiento farmacológico , Mesotelioma/tratamiento farmacológico , Pemetrexed , Tetrahidrofolatos/administración & dosificación
12.
Mol Cancer Ther ; 6(2): 404-17, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17308042

RESUMEN

Pemetrexed is a new-generation antifolate, approved for the treatment of mesothelioma and non-small cell lung cancer, currently being evaluated for the treatment of a variety of other solid tumors. This review traces the history of antifolates that led to the development of pemetrexed and describes the unique properties of this agent that distinguish it from other antifolates. These include (a) its very rapid conversion to active polyglutamate derivatives in cells that build to high levels and are retained for long intervals to achieve prolonged and potent inhibition of its major target enzyme thymidylate synthase, (b) its high affinity for three folate transporters, and (c) its marked sensitivity to the level of physiologic folates in cells. The latter results in the unique and paradoxical finding that when transport mediated by the major folate transporter (the reduced folate carrier) is impaired, pemetrexed activity is preserved. This is due to concurrent contraction of competing cellular physiologic folates and utilization of a novel second transport carrier for which pemetrexed has high affinity, recently identified as the proton-coupled folate transporter (PCFT). Laboratory studies are reviewed that raise the possibility of new approaches to the use of folic acid supplementation in clinical regimens with pemetrexed.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Antagonistas del Ácido Fólico/farmacología , Glutamatos/farmacología , Guanina/análogos & derivados , Transporte Biológico , Ensayos Clínicos como Asunto , Ácido Fólico/metabolismo , Glutamatos/metabolismo , Guanina/farmacología , Humanos , Neoplasias/tratamiento farmacológico , Pemetrexed
13.
Cell ; 127(5): 917-28, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17129779

RESUMEN

Folates are essential nutrients that are required for one-carbon biosynthetic and epigenetic processes. While folates are absorbed in the acidic milieu of the upper small intestine, the underlying absorption mechanism has not been defined. We now report the identification of a human proton-coupled, high-affinity folate transporter that recapitulates properties of folate transport and absorption in intestine and in various cell types at low pH. We demonstrate that a loss-of-function mutation in this gene is the molecular basis for hereditary folate malabsorption in a family with this disease. This transporter was previously reported to be a lower-affinity, pH-independent heme carrier protein, HCP1. However, the current study establishes that a major function of this gene product is proton-coupled folate transport required for folate homeostasis in man, and we have thus amended the name to PCFT/HCP1.


Asunto(s)
Ácido Fólico/metabolismo , Mucosa Intestinal/metabolismo , Síndromes de Malabsorción/genética , Proteínas de Transporte de Membrana/metabolismo , Animales , Transporte Biológico , Células CACO-2 , Línea Celular Tumoral , Electrofisiología , Ácido Fólico/química , Regulación de la Expresión Génica , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Cinética , Proteínas de Transporte de Membrana/genética , Oocitos , Linaje , Transportador de Folato Acoplado a Protón , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especificidad por Sustrato , Xenopus
14.
Mol Cancer Ther ; 5(10): 2549-55, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17041099

RESUMEN

Pemetrexed is a new-generation antifolate inhibitor of thymidylate synthase (TS) and a weaker inhibitor of glycinamide ribonucleotide transformylase (GARFT) required for de novo purine synthesis. Methylthioadenosine phosphorylase (MTAP) salvages purines by releasing adenine from methylthioadenosine and is often deleted in mesothelioma. The current study addresses the effect of MTAP on pemetrexed activity using a highly potent transition state inhibitor of MTAP, MT-DADMe-Immucillin A (ImmA; K(i) = 86 pmol/L) in the MTAP(+) NCI-H28 and MTAP(-) NCI-H2052 mesothelioma cell lines. Based on selective nucleoside protection, TS was found to be the primary pemetrexed target in both cell lines with GARFT inhibition requiring 20- to 30-fold higher pemetrexed concentrations. ImmA had no effect on pemetrexed activity but, when thymidine was added, the pemetrexed IC(50) decreased by a factor of approximately 3 in MTAP(+) H28 cells with no effect in MTAP(-) H2052 cells. Conversely, the transfection of MTAP into H2052 cells increased the pemetrexed IC(50) by nearly 3-fold but only in the presence of thymidine; this was reversed by ImmA. An MTAP-specific short interfering RNA produced a 2-fold decrease in pemetrexed IC(50) in MTAP(+) HeLa cells in the presence of thymidine. These data indicate that suppression of constitutive MTAP has no effect on pemetrexed activity when the primary target is TS. There is a modest salutary effect when the pemetrexed target is GARFT alone.


Asunto(s)
Glutamatos/farmacología , Guanina/análogos & derivados , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Línea Celular Tumoral , Guanina/farmacología , Humanos , Pemetrexed , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Purina-Nucleósido Fosforilasa/biosíntesis , Purina-Nucleósido Fosforilasa/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Ensayo de Tumor de Célula Madre
15.
Mol Cancer Ther ; 5(2): 438-49, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16505119

RESUMEN

Pemetrexed, a new generation antifolate recently approved for the treatment of mesothelioma and non-small cell lung cancer, is an excellent substrate for the reduced folate carrier (RFC). To explore the carrier's effect on pemetrexed activity, RFC was inactivated in HCT-15 colon cancer cells by mutagenesis and PT632 selective pressure. A clone (PT1) was obtained with a glycine to arginine substitution at amino acid 401, resulting in the loss of RFC function. PT1 cells were resistant to PT632 (178-fold), methotrexate (4-fold), and ZD1694 (Tomudex, raltitrexed; 20-fold), but were 3-fold collaterally sensitive to pemetrexed when grown in 25 nmol/L of 5-formyltetrahydrofolate. PT1 cells transfected with wild-type RFC had antifolate sensitivities comparable to that of wild-type HCT-15 cells, indicating that the RFC mutation was the sole basis for resistance. Folate pools were contracted in PT1 cells by 32% or 60%, as measured by radiolabeling intracellular folates or by an enzyme binding assay, respectively. This was reflected in marked (6.5-fold) collateral sensitivity to trimetrexate. The initial uptake of pemetrexed in PT1 cells was markedly reduced ( approximately 85%) but intracellular pemetrexed levels increased to approximately 60% and approximately 70% to that of wild-type cells after 2 hours and 6 days, respectively. There was increased pemetrexed inhibition of glycinamide ribonucleotide transformylase and, to a lesser extent, thymidylate synthase in PT1 cells growing in 5-formyltetrahydrofolate based on nucleoside protection analyses. Hence, loss of RFC function leads to collateral sensitivity to pemetrexed in HCT-15 cells, likely due to cellular folate pool contraction resulting in partial preservation of pemetrexed polyglutamylation and increased target enzyme inhibition. micro


Asunto(s)
Antineoplásicos/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias del Colon/metabolismo , Resistencia a Antineoplásicos/genética , Glutamatos/metabolismo , Guanina/análogos & derivados , Receptores de Superficie Celular/metabolismo , Sustitución de Aminoácidos , Arginina/química , Arginina/genética , Proteínas Portadoras/genética , Línea Celular Tumoral , Neoplasias del Colon/genética , Receptores de Folato Anclados a GPI , Antagonistas del Ácido Fólico/farmacología , Glicina/química , Glicina/genética , Guanina/agonistas , Guanina/metabolismo , Humanos , Leucovorina/metabolismo , Metotrexato/metabolismo , Mutación , Pemetrexed , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Purinas/farmacología , Pirimidinas/farmacología , Quinazolinas/metabolismo , Receptores de Superficie Celular/genética , Tiofenos/metabolismo , Timidilato Sintasa/antagonistas & inhibidores
16.
Mol Cancer Ther ; 4(12): 1860-6, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16373701

RESUMEN

The p16(INK4A)/CDKN2A gene on chromosome 9p21 is a site of frequent allelic loss in human cancers, and in a subset of cases, homozygous deletions at this locus encompass the telomeric methylthioadenosine phosphorylase (MTAP) gene. The MTAP gene product is the principal enzyme involved in purine synthesis via the salvage pathway, such that MTAP-negative cancers are solely dependent on de novo purine synthesis mechanisms. Inhibitors of the de novo pathway can then be used to selectively blockade purine synthesis in cancer cells while causing minimal collateral damage to normal cells. In this study, we determine that 10 of 28 (35%) biliary tract cancers show complete lack of Mtap protein expression. In vitro analysis using a selective inhibitor of the de novo purine synthesis pathway, L-alanosine, shows robust growth inhibition in MTAP-negative biliary cancer cell lines CAK-1 and GBD-1 accompanied by striking depletion of intracellular ATP and failure to rescue this depletion via addition of exogenous methylthioadenosine, the principal substrate of the MTAP gene product; in contrast, no significant effects were observed in MTAP-expressing HuCCT1 and SNU308 cell lines. Colony formation studies confirmed that L-alanosine reduced both number and size of CAK-1 colonies in soft agar assays. Knockdown of Mtap protein by RNA interference in L-alanosine-resistant HuCCT1 cells conferred sensitivity to this agent, confirming that intracellular Mtap protein levels determine response to L-alanosine. Inhibitors of de novo purine synthesis can be a potential mechanism-based strategy for treatment of biliary tract cancers, one third of which show complete loss of MTAP function.


Asunto(s)
Neoplasias del Sistema Biliar/genética , Eliminación de Gen , Homocigoto , Purina-Nucleósido Fosforilasa/genética , Secuencia de Bases , Neoplasias del Sistema Biliar/enzimología , Neoplasias del Sistema Biliar/patología , Línea Celular Tumoral , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa
17.
Clin Cancer Res ; 11(3): 1294-301, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15709201

RESUMEN

Pemetrexed is a novel antifolate with polyglutamate derivatives that are potent inhibitors of thymidylate synthase (TS) and to a lesser extent glycinamide ribonucleotide formyltransferase (GARFT). Conditions that might modulate relative suppression of these sites were assessed by the pattern of hypoxanthine and thymidine protection. When grown with 25 nmol/L racemic 5-formyltetrahydrofolate, thymidine alone fully protected wild-type HeLa cells to at least 1 micromol/L pemetrexed, but protection of a reduced folate carrier (RFC)-null subline required both thymidine and hypoxanthine above a concentration of 30 nmol/L pemetrexed. As medium 5-formyltetrahydrofolate was decreased, protection by thymidine alone decreased, and was further diminished when HeLa cells were grown in dialyzed serum. There was little protection by thymidine of RFC-null HeLa cells under the latter conditions. Thymidine alone was not protective, and hypoxanthine alone produced only a small (2-fold) increase in IC(50), in a HeLa-derived line 8-fold resistant to pemetrexed due to a modest increase in TS. Finally, in MCF-7 breast cancer cells there was greater protection with thymidine alone than in HeLa cells when cells were grown in medium containing a low concentration of 5-formyltetrahydrofolate. These observations indicate that as intracellular folates decrease in HeLa cells, due to decreased extracellular reduced folate, or loss of RFC function, pemetrexed inhibition of GARFT increases. These data support the concept that the contribution to pemetrexed activity by inhibition of GARFT, particularly at low folate levels, is a contributing factor to drug activity but relative inhibition of TS and GARFT may vary among human tumors and cell lines.


Asunto(s)
Antagonistas del Ácido Fólico/farmacología , Ácido Fólico/metabolismo , Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/farmacología , Purinas/biosíntesis , Animales , Western Blotting , Bovinos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Medios de Cultivo/química , Medios de Cultivo/farmacología , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Hipoxantina/farmacología , Leucovorina/farmacología , Proteínas de Transporte de Membrana/fisiología , Pemetrexed , Proteína Portadora de Folato Reducido , Suero/química , Timidilato Sintasa/metabolismo , Timina/farmacología
18.
Clin Cancer Res ; 10(24): 8735-42, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15623659

RESUMEN

Prior studies from this laboratory documented the prevalence of methotrexate (MTX) transport activity with a low pH optimum in human solid tumor cell lines. In HeLa cells, this low pH activity has high affinity for pemetrexed [PMX (Alimta)] and is reduced folate carrier (RFC)-independent because it is not diminished in a RFC-null subline (R5). R5 cells also have residual transport activity, with high specificity for PMX, at neutral pH. In the current study, a R5 subline, R1, was selected under MTX selective pressure at a modest reduction in pH. There was markedly decreased MTX and PMX transport at both pH 5.5 and pH 7.4. When MTX was removed, there was a slow return of transport activity, and when MTX was added back, there was loss of transport at both pH values within 8 weeks. In R1 cells, there was a marked decrease in accumulation of PMX, MTX, and folic acid along with a decrease in growth inhibition by these and other antifolates that require a facilitative process to gain entry into cells. These data demonstrate that (i) RFC-independent transport in HeLa cells at low and neutral pH contributes to antifolate activity (in particular, to PMX activity) and can be diminished by antifolate selective pressure and (ii) the loss of these activities results in marked resistance to PMX, an agent for which there is little or no loss of activity when transport mediated by RFC is abolished. These observations suggest that transport activity in RFC-null HeLa R5 cells at neutral and low pH may reflect the same carrier-mediated process.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Antagonistas del Ácido Fólico/farmacología , Proteínas de Transporte de Membrana/farmacocinética , Transporte Biológico , Proteínas Portadoras/metabolismo , División Celular/efectos de los fármacos , Receptores de Folato Anclados a GPI , Ácido Fólico/metabolismo , Glutamatos/metabolismo , Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/metabolismo , Guanina/farmacología , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Proteínas de Transporte de Membrana/deficiencia , Metotrexato/metabolismo , Metotrexato/farmacología , Pemetrexed , Receptores de Superficie Celular/metabolismo , Proteína Portadora de Folato Reducido , Timidilato Sintasa/antagonistas & inhibidores , Factores de Tiempo
19.
Clin Cancer Res ; 10(23): 7986-93, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15585634

RESUMEN

Pemetrexed [PMX (Alimta)] is a new generation antifolate with activity in a variety of solid tumors. It is an excellent substrate for most folate transporters, notably the reduced folate carrier (RFC) and folate receptor (FR)-alpha. The role of FR-alpha in PMX pharmacological activity is uncertain. Whereas high-level expression may enhance the activity of this agent, it is not clear what role constitutive levels of this transporter contribute to PMX activity. In this study, constitutive levels of FR-alpha expression were abolished by small interfering RNA-induced silencing in HeLa cells and RFC-null HeLa R5 cells as confirmed by Northern blotting, immunohistochemistry, and cell surface binding. PMX growth inhibition was unchanged in HeLa and R5 cells in the absence of FR-alpha expression. Loss of FR-alpha expression did not decrease net accumulation of PMX in either wild-type or RFC-null HeLa cells. Likewise, folate pools in wild-type HeLa cells were not decreased by FR-alpha gene silencing and were negligibly affected in the RFC-null R5 subline grown with 5-formyltetrahydrofolate. FR-alpha surface binding in HeLa cells was shown to be greater than that in a variety of other human solid tumor cell lines. Hence, constitutively expressed FR-alpha in HeLa cells does not contribute to PMX activity in the presence or absence of RFC function. This is likely the case in many human solid tumor cell lines.


Asunto(s)
Proteínas Portadoras/genética , Antagonistas del Ácido Fólico/farmacología , Ácido Fólico/metabolismo , Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/farmacología , Neoplasias/enzimología , Interferencia de ARN , Receptores de Superficie Celular/genética , Apoptosis/efectos de los fármacos , Secuencia de Bases , Northern Blotting , Técnica del Anticuerpo Fluorescente , Receptores de Folato Anclados a GPI , Antagonistas del Ácido Fólico/metabolismo , Formiltetrahidrofolatos , Glutamatos/metabolismo , Guanina/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Neoplasias/genética , Pemetrexed , Receptores de Superficie Celular/deficiencia , Timidilato Sintasa/antagonistas & inhibidores , Células Tumorales Cultivadas
20.
Cancer Res ; 64(9): 3313-9, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15126375

RESUMEN

A methotrexate (MTX)-resistant HeLa subline (R5), developed in this laboratory, with impaired transport due to a genomic deletion of the reduced folate carrier (RFC) was only 2-fold resistant to pemetrexed (PMX), but 200- and 400-fold resistant to raltitrexed (ZD1694) and N(alpha)-(-4-amino-4-deoxypteroyl)-N(delta)-hemiphthaloyl-1-ornithine (PT523), respectively, compared with parental HeLa cells when grown with 2 microM folic acid. When folic acid was replaced with the more physiological 25 nM 5-formyltetrahydrofolate, R5 cells were 2-fold collaterally sensitive to PMX but still 40- and 200-fold resistant to ZD1694 and PT523, respectively. Sensitivity to PT523 and PMX could be completely restored, and sensitivity to ZD1694 nearly restored, by transfection of RFC cDNA into R5 cells, indicating that the defect in drug transport was the only, or major, factor in resistance. The preserved PMX activity in R5 cells could not be related to the very low expression of folate receptors. Rather, retained PMX activity in R5 cells was associated with residual transport by another process that exhibits good affinity for PMX (Kt = 12 microM) with much lower affinities for ZD1694, MTX, and PT523 (Kis of approximately 90, 100, and 250 microM, respectively). PMX transported by this route was rapidly converted to higher polyglutamates and, when grown with 25 nM 5-formyl-tetrahydrofolate, the rate of formation of these derivatives and their net accumulation in R5 cells was comparable to that of wild-type cells. These data suggest that selective preservation of PMX pharmacological activity in RFC-null R5 cells is due, in part, to partial preservation of transport by secondary process with a higher affinity for PMX than the other antifolates evaluated.


Asunto(s)
Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/farmacología , Proteínas de Transporte de Membrana/deficiencia , Ornitina/análogos & derivados , Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/farmacología , Transporte Biológico/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/fisiología , Resistencia a Antineoplásicos , Antagonistas del Ácido Fólico/farmacocinética , Antagonistas del Ácido Fólico/farmacología , Glutamatos/farmacocinética , Guanina/farmacocinética , Células HeLa , Humanos , Proteínas de Transporte de Membrana/metabolismo , Ornitina/farmacología , Pemetrexed , Ácido Poliglutámico/análogos & derivados , Ácido Poliglutámico/metabolismo , Pterinas/farmacología , Quinazolinas/farmacología , Proteína Portadora de Folato Reducido , Tiofenos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA