Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Mol Med ; 26(22): 5624-5633, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36300880

RESUMEN

Curcumin is a known epigenetic modifier that demonstrated antitumor effect in different types of cancer. The poor solubility and metabolic stability are major drawbacks that limit its development as an antitumor agent. Dimethoxycurcumin (DMC) is a more soluble and stable curcumin analog. In this study, we compared the effect of both drugs on a variety of histone posttranslational modifications and on the activity of histone lysine methyltransferase (HKMTs) and demethylase (HKDMTs) enzymes that target the H3K4, H3K9 and H3K27 epigenetic marks. Mass spectrometry was used to quantitate the changes in 95 histone posttranslational modifications induced by curcumin or DMC. The effect of both drugs on the enzymatic activity of HKMTs and HKDMs was measured using an antibody-based assay. Mass spectrometry analysis showed that curcumin and DMC modulated several histone modifications. Histone changes were not limited to lysine methylation and acetylation but included arginine and glutamine methylation. Only few histone modifications were similarly changed by both drugs. On the contrary, the effect of both drugs on the activity of HKMTs and HKDMs was very similar. Curcumin and DMC inhibited the HKMTs enzymes that target the H3K4, H3K9 and H3K27 marks and increased the activity of the HKDMs enzymes LSD1, JARID and JMJD2. In conclusion, we identified novel enzymatic targets for both curcumin and DMC that support their use and development as epigenetic modifiers in cancer treatment. The multiple targets modulated by both drugs could provide a therapeutic advantage by overcoming drug resistance development.


Asunto(s)
Curcumina , Leucemia , Humanos , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Curcumina/farmacología , Leucemia/tratamiento farmacológico
2.
Cancer Control ; 29: 10732748221074051, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35067084

RESUMEN

INTRODUCTION: The prototype DNA hypomethylating agents 5-azacytidine (5AC) and decitabine (DAC) are currently FDA-approved for treatment of blood and bone marrow disorders like myelodysplastic syndrome. 5AC and DAC are considered similar drugs and were shown to induce histone modifications that modulate gene expression. The aim of this study is to compare the effect of both drugs on histone acetylation and methylation at multiple histone amino acids residues. METHODS: Mass spectrometry was used to compare the effect of both drugs on 95 different histone posttranslational modifications (PTMs) in leukemia cells. ChIP-Seq analysis was used to compare the impact of both drugs on the genome-wide acetylation of the H3K9 mark using primary leukemia cells from six de-identified AML patients. RESULTS: Both DAC and 5AC induced histone PTMs in different histone isoforms like H1.4, H2A, H3, H3.1, and H4. Changes in both histone methylation and acetylation were observed with both drugs; however, there were distinct differences in the histone modifications induced by the two drugs. Since both drugs were shown to increase the activity of the HDAC SIRT6 previously, we tested the effect of 5AC on the acetylation of H3K9, the physiological substrate SIRT6, using ChIP-Seq analysis and compared it to the previously published DAC-induced changes. Significant H3K9 acetylation changes (P< .05) were detected at 925 genes after 5AC treatment vs only 182 genes after DAC treatment. Nevertheless, the gene set modified by 5AC was different from that modified by DAC with only ten similar genes modulated by both drugs. CONCLUSION: Despite similarity in chemical structure and DNA hypomethylating activity, 5AC and DAC induced widely different histone PTMs and considering them interchangeable should be carefully evaluated. The mechanism of these histone PTM changes is not clear and may involve modulation of the activity or the expression of the enzymes inducing histone PTMs.


Asunto(s)
Acetilación/efectos de los fármacos , Azacitidina/farmacología , Metilación de ADN/efectos de los fármacos , Decitabina/farmacología , Histonas/efectos de los fármacos , Línea Celular Tumoral , Humanos , Leucemia/tratamiento farmacológico , Procesamiento Proteico-Postraduccional/efectos de los fármacos
3.
Pharm Res ; 32(3): 863-75, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25186441

RESUMEN

PURPOSE: Curcumin is an ideal chemopreventive and antitumor agent characterized by poor bioavailability and low stability. The development of synthetic structural analogues like dimethoxycurcumin (DMC) could overcome these drawbacks. In this study we compared the cytotoxicity, metabolism and the epigenetic changes induced by both drugs in leukemia cells. METHODS: Apoptosis and cell cycle analysis were analyzed by flow cytometry. Real-time PCR was used for gene expression analysis. DNA methylation was analyzed by DNA pyrosequencing. The metabolic stability was determined using human pooled liver microsomes. Chromatin Immunoprecipitation was used to quantify histone methylation. RESULTS: Clinically relevant concentration of curcumin and DMC were not cytotoxic to leukemia cells and induced G2/M cell cycle arrest. DMC was more metabolically stable than curcumin. Curcumin and DMC were devoid of DNA hypomethylating activity. DMC induced the expression of promoter methylated genes without reversing DNA methylation and increased H3K36me3 mark near the promoter region of hypermethylated genes. CONCLUSION: DMC is a more stable analogue of curcumin that can induce epigenetic changes not induced by curcumin. DMC induced the expression of promoter methylated genes. The combination of DMC with DNA methyltransferase inhibitors could harness their combined induced epigenetic changes for optimal re-expression of epigenetically silenced genes.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Curcumina/análogos & derivados , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia/genética , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/metabolismo , Apoptosis/efectos de los fármacos , Biotransformación , Línea Celular Tumoral , Curcumina/química , Curcumina/metabolismo , Curcumina/farmacología , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Histonas/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/patología , Metilación , Microsomas Hepáticos/metabolismo , Regiones Promotoras Genéticas , Factores de Tiempo
4.
Epigenomes ; 7(3)2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37489400

RESUMEN

Azanucleosides, such as 5-azacytidine and decitabine, are DNA demethylating agents used in the treatment of acute myeloid leukemia and myelodysplastic syndromes. Researchers continue to explore their utility in the treatment of other hematologic and solid tumors. Based on the capacity of the compounds to inhibit DNA methyltransferase enzymes and the important role of DNA methylation in health and disease, it is essential to understand the molecular changes that azanucleosides induce and how these changes may improve treatment outcomes in subsets of patients. This review summarizes the molecular and therapeutic actions of azanucleosides and discusses recent clinical trials of these compounds as single agents or in combination therapy for the treatment of cancer and related conditions.

5.
Sr Care Pharm ; 37(8): 339-344, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35879841

RESUMEN

Objectives To discuss therapeutic management of Parkinson's disease psychosis (PDP) and the burdens associated with it. Parkinson's disease (PD) is an incurable, progressive, and devastating neurodegenerative disease. The prolonged use of dopamine agonists to improve the motor symptoms of PD may result in nonmotor complications such as psychosis. These complications are as devastating as the motor symptoms that accompany PD. PDP is associated with financial burden for patients and health care organizations. Data Sources PubMed search using the phrases Parkinson's disease, Parkinson's disease psychosis, clozapine, quetiapine, and "pimavanserin." Conclusion Pimavanserin is the only US Food and Drug Administration approved drug for the management of hallucinations and delusions associated with PD psychosis. The off-label use of clozapine and quetiapine has demonstrated clinical utility; however, there is still a need for the development of novel therapeutic molecules to avoid side effects associated with current therapeutic options and reduce the burdens of patients, caregivers, and health care institutions.


Asunto(s)
Clozapina , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Trastornos Psicóticos , Clozapina/uso terapéutico , Humanos , Enfermedades Neurodegenerativas/complicaciones , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/tratamiento farmacológico , Trastornos Psicóticos/tratamiento farmacológico , Trastornos Psicóticos/etiología , Fumarato de Quetiapina/uso terapéutico , Estados Unidos
6.
Pharmaceutics ; 14(3)2022 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-35335943

RESUMEN

BACKGROUND: With the Coronavirus becoming a new reality of our world, global efforts continue to seek answers to many questions regarding the spread, variants, vaccinations, and medications. Particularly, with the emergence of several strains (e.g., Delta, Omicron), vaccines will need further development to offer complete protection against the new variants. It is critical to identify antiviral treatments while the development of vaccines continues. In this regard, the repurposing of already FDA-approved drugs remains a major effort. In this paper, we investigate the hypothesis that a combination of FDA-approved drugs may be considered as a candidate for COVID-19 treatment if (1) there exists an evidence in the COVID-19 biomedical literature that suggests such a combination, and (2) there is match in the clinical trials space that validates this drug combination. METHODS: We present a computational framework that is designed for detecting drug combinations, using the following components (a) a Text-mining module: to extract drug names from the abstract section of the biomedical publications and the intervention/treatment sections of clinical trial records. (b) a network model constructed from the drug names and their associations, (c) a clique similarity algorithm to identify candidate drug treatments. RESULT AND CONCLUSIONS: Our framework has identified treatments in the form of two, three, or four drug combinations (e.g., hydroxychloroquine, doxycycline, and azithromycin). The identifications of the various treatment candidates provided sufficient evidence that supports the trustworthiness of our hypothesis.

7.
Blood ; 114(16): 3448-58, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19652201

RESUMEN

Increasing evidence shows aberrant hypermethylation of genes occurring in and potentially contributing to pathogenesis of myeloid malignancies. Several of these diseases, such as myelodysplastic syndromes (MDSs), are responsive to DNA methyltransferase inhibitors. To determine the extent of promoter hypermethylation in such tumors, we compared the distribution of DNA methylation of 14 000 promoters in MDS and secondary acute myeloid leukemia (AML) patients enrolled in a phase 1 trial of 5-azacytidine and the histone deacetylase inhibitor entinostat against de novo AML patients and normal CD34(+) bone marrow cells. The MDS and secondary AML patients displayed more extensive aberrant DNA methylation involving thousands of genes than did the normal CD34(+) bone marrow cells or de novo AML blasts. Aberrant methylation in MDS and secondary AML tended to affect particular chromosomal regions, occurred more frequently in Alu-poor genes, and included prominent involvement of genes involved in the WNT and MAPK signaling pathways. DNA methylation was also measured at days 15 and 29 after the first treatment cycle. DNA methylation was reversed at day 15 in a uniform manner throughout the genome, and this effect persisted through day 29, even without continuous administration of the study drugs. This trial was registered at www.clinicaltrials.gov as J0443.


Asunto(s)
Azacitidina/administración & dosificación , Metilación de ADN/efectos de los fármacos , ADN de Neoplasias/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Neoplasias Primarias Secundarias/tratamiento farmacológico , Neoplasias Primarias Secundarias/metabolismo , Antígenos CD34 , Células de la Médula Ósea/metabolismo , Femenino , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/metabolismo , Regiones Promotoras Genéticas , Factores de Tiempo , Proteínas Wnt/metabolismo
8.
Blood ; 114(13): 2764-73, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19546476

RESUMEN

Sequential administration of DNA methyltransferase (DNMT) inhibitors and histone deacetylase (HDAC) inhibitors has demonstrated clinical efficacy in patients with hematologic malignancies. However, the mechanism behind their clinical efficacy remains controversial. In this study, the methylation dynamics of 4 TSGs (p15(INK4B), CDH-1, DAPK-1, and SOCS-1) were studied in sequential bone marrow samples from 30 patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) who completed a minimum of 4 cycles of therapy with 5-azacytidine and entinostat. Reversal of promoter methylation after therapy was observed in both clinical responders and nonresponders across all genes. There was no association between clinical response and either baseline methylation or methylation reversal in the bone marrow or purified CD34(+) population, nor was there an association with change in gene expression. Transient global hypomethylation was observed in samples after treatment but was not associated with clinical response. Induction of histone H3/H4 acetylation and the DNA damage-associated variant histone gamma-H2AX was observed in peripheral blood samples across all dose cohorts. In conclusion, methylation reversal of candidate TSGs during cycle 1 of therapy was not predictive of clinical response to combination "epigenetic" therapy. This trial is registered with http://www.clinicaltrials.gov under NCT00101179.


Asunto(s)
Azacitidina/administración & dosificación , Benzamidas/administración & dosificación , Daño del ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/tratamiento farmacológico , Piridinas/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Análisis Citogenético , Daño del ADN/fisiología , Esquema de Medicación , Epigénesis Genética/fisiología , Femenino , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Tiempo
9.
Adv Cancer Res ; 152: 205-223, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34353438

RESUMEN

DNA methylation is an epigenetic modification that contributes to essential biological processes such as retrotransposon silencing, cell differentiation, genomic imprinting and X-chromosome inactivation. DNA methylation generates a stable epigenetic mark associated with silencing of gene expression. Aberrant DNA methylation is associated with the development of different tumor types. Reversing DNA methylation is a rational strategy to restore gene re-expression and induce cell differentiation in cancer. DNA hypomethylating agents is a class of drugs that demonstrated efficacy in different tumors. In this chapter, the classification of DNA hypomethylating agents, their pharmacodynamics and their potential drawbacks will be discussed.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/uso terapéutico , Azacitidina/uso terapéutico , Metilación de ADN , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Estudios Retrospectivos
10.
J Med Chem ; 63(24): 15906-15945, 2020 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-33314925

RESUMEN

HER2 kinase as a well-established target for breast cancer (BC) therapy is associated with aggressive clinical outcomes; thus, herein we present structural optimization for HER2-selective targeting. HER2 profiling of the developed derivatives demonstrated potent and selective inhibitions (IC50: 5.4-12 nM) compared to lapatinib (IC50: 95.5 nM). Favorably, 17d exhibited minimum off-target kinase activation. NCI-5-dose screening revealed broad-spectrum activities (GI50: 1.43-2.09 µM) and 17d had a remarkable selectivity toward BC. Our compounds revealed significant selective and potent antiproliferative activities (∼20-fold) against HER2+ (AU565, BT474) compared to HER2(-) cells. At 0.1 IC50, 15i, 17d, and 25b inhibited pERK1/2 and pAkt by immunoblotting. Furthermore, 17d demonstrated potent in vivo tumor regression against the BT474 xenograft model. Notably, a metastasis case was observed in the vehicle but not in the test mice groups. CD-1 mice metabolic stability assay revealed high stability and low intrinsic clearance of 17d (T1/2 > 145 min and CLint(mic) < 9.6 mL/min/kg).


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Diseño de Fármacos , Lapatinib/química , Terapia Molecular Dirigida , Quinazolinas/síntesis química , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas In Vitro , Ratones , Ratones Desnudos , Receptor ErbB-2/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Sci Rep ; 10(1): 10325, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32587297

RESUMEN

The FDA-approved DNA hypomethylating agents (DHAs) like 5-azacytidine (5AC) and decitabine (DAC) demonstrate efficacy in the treatment of hematologic malignancies. Despite previous reports that showed histone acetylation changes upon using these agents, the exact mechanism underpinning these changes is unknown. In this study, we investigated the relative potency of the nucleoside analogs and non-nucleoside analogs DHAs on DNA methylation reversal using DNA pyrosequencing. Additionally, we screened their effect on the enzymatic activity of the histone deacetylase sirtuin family (SIRT1, SIRT2, SIRT3, SIRT5 and SIRT6) using both recombinant enzymes and nuclear lysates from leukemia cells. The nucleoside analogs (DAC, 5AC and zebularine) were the most potent DHAs and increased the enzymatic activity of SIRT6 without showing any significant increase in other sirtuin isoforms. ChIP-Seq analysis of bone marrow cells derived from six acute myeloid leukemia (AML) patients and treated with the nucleoside analog DAC induced genome-wide acetylation changes in H3K9, the physiological substrate for SIRT6. Data pooling from the six patients showed significant acetylation changes in 187 gene loci at different chromosomal regions including promoters, coding exons, introns and distal intergenic regions. Signaling pathway analysis showed that H3K9 acetylation changes are linked to AML-relevant signaling pathways like EGF/EGFR and Wnt/Hedgehog/Notch. To our knowledge, this is the first report to identify the nucleoside analogs DHAs as activators of SIRT6. Our findings provide a rationale against the combination of the nucleoside analogs DHAs with SIRT6 inhibitors or chemotherapeutic agents in AML due to the role of SIRT6 in maintaining genome integrity and DNA repair.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Sirtuinas/metabolismo , Acetilación/efectos de los fármacos , Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Azacitidina/farmacología , Azacitidina/uso terapéutico , Médula Ósea/patología , Línea Celular Tumoral , Citidina/análogos & derivados , Citidina/farmacología , Citidina/uso terapéutico , Metilación de ADN/efectos de los fármacos , Decitabina/farmacología , Decitabina/uso terapéutico , Histonas/metabolismo , Humanos , Leucemia Mieloide Aguda/patología
12.
Sci Rep ; 10(1): 15386, 2020 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-32968125

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
Curr Med Chem ; 16(17): 2075-85, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19519382

RESUMEN

Although chemotherapy is considered the mainstay of cancer therapy, unfortunate side effects of chemotherapy create a continuous demand for developing other novel and specific targets for cancer therapy. Re-expression of epigenetically silenced tumor suppressor genes is a rational strategy for the treatment of human neoplasms. Epigenetic modifiers like DNA methyltransferase (DNMT) inhibitors and histone deacteylase (HDAC) inhibitors induce the re-expression of epigenetically silenced genes in vitro and in vivo. Moreover, they demonstrate safety and efficacy against neoplastic diseases in clinical trials. DNMT inhibitors like 5-azacytidine and 5-aza-2'-deoxycytidine are currently FDA approved for the treatment of myelodysplastic syndrome. Nonetheless, the mechanism of action behind their clinical efficacy remains unclear. Ongoing clinical trials are attempting to identify tumor suppressor genes that upon re-expression can induce remission and cure in patients. On the other hand, the pleiotropic biological effects of DNMT inhibitors and recent reports demonstrating lack of association between clinical response and methylation reversal of candidate tumor suppressor genes, suggest a complex mechanism behind their clinical efficacy that may involve a cytotoxic effect.


Asunto(s)
Antineoplásicos/uso terapéutico , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Ensayos Clínicos como Asunto , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/genética , Humanos , Estructura Molecular , Neoplasias/enzimología , Neoplasias/genética
14.
Mol Cancer ; 7: 60, 2008 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-18590557

RESUMEN

BACKGROUND: Drug resistance is a major concern in cancer therapy. Here, we investigate the clinical potential of the second mitochondria-derived activator of caspase (Smac/DIABLO) in enhancing the apoptosis-inducing potential of commonly used anticancer drugs (paclitaxel, doxorubicin, etoposide, tamoxifen), irradiation and TRAIL in breast carcinoma. METHODS: Breast cancer cells were overexpressed with Smac/DIABLO gene (full-length or Delta55 Smac/DIABLO) or treated with Smac/DIABLO peptide to enhance the apoptosis-inducing potential of chemotherapeutic drugs and irradiation, and sensitize TRAIL-resistant cells. Cell viability and apoptosis were measured by XTT assay and DAPI staining, respectively. Protein-protein interaction was determined by immunoprecipitation followed by the Western blot analysis. RESULTS: Overexpression of Smac/DIABLO gene (full-length or Delta55 Smac/DIABLO) or treatment with Smac/DIABLO peptide enhances apoptosis induced by paclitaxel, doxorubicin, etoposide, tamoxifen, and irradiation in breast cancer cells. Overexpression of Smac/DIABLO resulted in an increased interaction of Smac/DIABLO with IAPs, which correlated with an increase in caspase-3 activity and apoptosis. Furthermore, Smac/DIABLO sensitized TRAIL-resistant breast cancer cell lines to undergo apoptosis through caspase-3 activation. These data suggest that apoptotic events down-stream of mitochondria were intact in TRAIL-resistant cells since ectopic expression of Smac/DIABLO or pretreatment of cells with Smac/DIABLO peptide completely restored TRAIL sensitivity. CONCLUSION: The ability of Smac/DIABLO agonists to enhance the apoptosis-inducing potential of chemotherapeutic drugs and irradiation, and sensitize TRAIL-resistant tumor cells suggests that Smac/DIABLO may induce fundamental alterations in cell signaling pathways. Thus, Smac/DIABLO agonists can be used as promising new candidates for cancer treatment by potentiating cytotoxic therapies.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Mitocondriales/metabolismo , Fármacos Fotosensibilizantes/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Colágeno Tipo XI/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de la radiación , Activación Enzimática/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Mitocondriales/genética , Unión Proteica
16.
Clin Pharmacokinet ; 57(6): 687-703, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29188435

RESUMEN

Antibody-drug conjugates are an emerging class of biopharmaceuticals changing the landscape of targeted chemotherapy. These conjugates combine the target specificity of monoclonal antibodies with the anti-cancer activity of small-molecule therapeutics. Several antibody-drug conjugates have received approval for the treatment of various types of cancer including gemtuzumab ozogamicin (Mylotarg®), brentuximab vedotin (Adcetris®), trastuzumab emtansine (Kadcyla®), and inotuzumab ozogamicin, which recently received approval (Besponsa®). In addition to these approved therapies, there are many antibody-drug conjugates in the drug development pipeline and in clinical trials, although these fall outside the scope of this article. Understanding the pharmacokinetics and pharmacodynamics of antibody-drug conjugates and the development of pharmacokinetic/pharmacodynamic models is indispensable, albeit challenging as there are many parameters to incorporate including the disposition of the intact antibody-drug conjugate complex, the antibody, and the drug agents following their dissociation in the body. In this review, we discuss how antibody-drug conjugates progressed over time, the challenges in their development, and how our understanding of their pharmacokinetics/pharmacodynamics led to greater strides towards successful targeted therapy programs.


Asunto(s)
Inmunoconjugados , Modelos Biológicos , Ado-Trastuzumab Emtansina , Aminoglicósidos/farmacocinética , Aminoglicósidos/farmacología , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/farmacología , Gemtuzumab , Humanos , Inmunoconjugados/farmacocinética , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Inotuzumab Ozogamicina , Maitansina/análogos & derivados , Maitansina/farmacocinética , Maitansina/farmacología , Neoplasias/metabolismo , Trastuzumab/farmacocinética , Trastuzumab/farmacología
17.
Cancer Chemother Pharmacol ; 60(3): 313-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17187253

RESUMEN

PURPOSE: Flavopiridol is known to modulate the transcription of genes. We investigated the effect of flavopiridol pretreatment on TRAIL cytotoxicity and on the expression of FLIP(L) in different TRAIL-resistant cell lines, because FLIP expression is known to confer TRAIL-resistance. METHODS: Apoptosis was assessed by PI staining and protein expression by Western blotting. RT-PCR was used for mRNA quantitation. siRNA gene silencing was used to knock down FLIP(L). RESULTS: Flavopiridol pretreatment synergized TRAIL-induced apoptosis in human myeloma and breast cancer cells. Flavopiridol treatment repressed the transcription of FLIP(L) and downregulated its expression in both myeloma and breast cancer cells. Silencing of FLIP(L) gene by siRNA sensitized myeloma cells to TRAIL. Flavopiridol treatment downregulated the expression of the proapoptotic members of the Bcl-2 family proteins (Bak, Bax and PUMA-alpha). The expression of the antiapoptotic Bcl-2 members (Bcl-2 and Bcl-X(L)) was not altered by flavopiridol treatment in myeloma cells. CONCLUSION: Our data indicate that flavopiridol synergizes TRAIL cytotoxicity by downregulation of FLIP(L) and this synergistic effect is Bcl-2 family independent.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Flavonoides/toxicidad , Piperidinas/toxicidad , Ligando Inductor de Apoptosis Relacionado con TNF/toxicidad , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Colorantes , Regulación hacia Abajo , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen/efectos de los fármacos , Humanos , Mieloma Múltiple/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Mol Cancer Ther ; 5(2): 457-66, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16505121

RESUMEN

Multiple myeloma is an incurable disease for the majority of patients, therefore requiring new biological targeted therapies. In primary myeloma cells, IMP dehydrogenase (IMPDH) was shown to be consistently overexpressed. We therefore tested the IMPDH inhibitor mycophenolate mofetil (MMF) currently available as a clinical therapeutic agent for its antimyeloma activity in vitro. MMF depleted intracellular guanosine 5'-triphosphate (GTP) levels in myeloma cells. We showed apoptosis induction in myeloma cell lines and primary myeloma cells between 1 and 5 mumol/L MMF. MMF was also cytotoxic at this concentration in dexamethasone-resistant and Mcl-1-overexpressed myeloma cell lines shown by the tetrazolium salt XTT assay along with cell survival measured by a modified flow cytometric assay. Apoptosis was not inhibited by the presence of an antioxidant, suggesting that MMF-induced apoptosis is less likely to be associated with reactive oxygen species. However, apoptosis was abrogated by exogenously added guanosine, which activates an alternative pathway for GTP formation, implicating that this effect is directly mediated by IMPDH inhibition. MMF-induced G1-S phase cell cycle arrest and its apoptosis induction mechanism were associated with a caspase-dependent pathway as shown by alteration of mitochondrial membrane potential and cytochrome c release followed by activation of the caspases. MMF-induced apoptosis was also inhibited by a pan-caspase inhibitor Z-VAD-fmk. MMF-treated myeloma cells showed an up-regulation of Bak, which most likely together with Bax resulted in the release of cytochrome c. In summary, MMF attenuates G1-S phase cell cycle progression and activates the pathway of mitochondrial dysfunction, leading to cytochrome c release followed by activation of caspases.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Caspasas/metabolismo , IMP Deshidrogenasa/antagonistas & inhibidores , Mieloma Múltiple/metabolismo , Ácido Micofenólico/análogos & derivados , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Guanosina/farmacología , Humanos , Mieloma Múltiple/enzimología , Ácido Micofenólico/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Transcripción STAT3/metabolismo
19.
Cancer Chemother Pharmacol ; 58(4): 471-7, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16435155

RESUMEN

PURPOSE: In this study, we have investigated the effect of trichostatin A (TSA) pretreatment on the cytotoxicity of TRAIL in TRAIL-resistant myeloma cells. METHODS AND RESULTS: MM1S myeloma cells exhibited resistance to TRAIL-induced apoptosis even at high doses of TRAIL and was sensitive to low doses of TSA. Sequential treatment of myeloma cells with TSA followed by TRAIL enhanced TRAIL cytotoxicity. TSA induced the transcription of TRAIL death receptor DR5 without affecting the transcription of DR4 and the decoy receptors; DcR1 and DcR2. However, the surface expression of both DR4 and DR5 was not modulated by TSA treatment. TSA treatment repressed the transcription and downregulated the expression of the antiapoptotic Bcl-2 proteins; Bcl-2 and Bcl-XL. Surprisingly, the effect of TSA on the proapoptotic Bcl-2 proteins was mixed, the two isoforms of PUMA (alpha, beta), Noxa, Bax were downregulated, while Bim was upregulated. Although MM1S cells showed higher expression level of FLIP(S) than other TRAIL-sensitive myeloma cells, the enhancing effect of TSA was not accompanied by FLIP(S) downregulation. CONCLUSION: In conclusion, TSA sensitized TRAIL-resistant myeloma cells by downregulating the antiapoptotic Bcl-2 protein without altering FLIP(S) expression.


Asunto(s)
Antifúngicos/farmacología , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/farmacología , Apoptosis/efectos de los fármacos , Caspasas/efectos de los fármacos , Cartilla de ADN/genética , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Glicoproteínas de Membrana/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Caspasa 8 , Caspasas/metabolismo , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ligando Inductor de Apoptosis Relacionado con TNF , Células Tumorales Cultivadas
20.
Cancer Chemother Pharmacol ; 77(2): 259-67, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26666648

RESUMEN

PURPOSE: Vincamine, vinpocetine and eburnamonine are alkaloids known for their neuroprotective attributes, enhancement of cerebrovascular blood flow and antitumor effect of their derivatives. However, the relative metabolic stability of these alkaloids and their extrusion by the drug efflux transporters expressed at the blood-brain barrier (BBB) are not clear. In this study, we developed rapid and sensitive methods for the detection of these alkaloids and investigated their relative metabolic stability and their interaction with drug efflux transporters. METHODS: UPLC methods were developed to analyze metabolic in vitro samples. Intrinsic clearance was determined using rat liver microsomal enzymes. Drug-stimulated transporter activity was estimated by measuring inorganic phosphate released from ATP spectrophotometrically. RESULTS: The UPLC methods quantification level ranged from 0.02 to 0.025 µg/mL, indicating high sensitivity. The intrinsic clearance of eburnamonine was significantly less than both vincamine and vinpocetine. Different concentrations of the three drugs (4, 20 and 100 µM) induced minimal stimulation of the ATPase activity of the Bcrp and Pgp membrane transporters. CONCLUSIONS: The developed simple, sensitive and reliable UPLC analysis methods can be utilized in future in vitro and in vivo studies. The three alkaloids demonstrated minimal interaction with the drug efflux transporters Pgp and Bcrp, concordant with the ability of these alkaloids to cross the BBB. The relative metabolic stability of eburnamonine compared to the other alkaloids suggests the use of eburnamonine or its derivatives as lead compounds for the development of antitumor and nootropic agents that need to cross the BBB and produce their pharmacological effects in the CNS.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Microsomas Hepáticos/efectos de los fármacos , Alcaloides de la Vinca , Vincamina , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Transporte Biológico/efectos de los fármacos , Estabilidad de Medicamentos , Proteínas de Transporte de Membrana/metabolismo , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Reproducibilidad de los Resultados , Alcaloides de la Vinca/metabolismo , Alcaloides de la Vinca/farmacología , Vincamina/metabolismo , Vincamina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA