Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Exp Clin Cancer Res ; 41(1): 340, 2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-36482393

RESUMEN

BACKGROUND: Acute myeloid leukemia (AML) is an aggressive hematological cancer resulting from uncontrolled proliferation of differentiation-blocked myeloid cells. Seventy percent of AML patients are currently not cured with available treatments, highlighting the need of novel therapeutic strategies. A promising target in AML is the mammalian target of rapamycin complex 1 (mTORC1). Clinical inhibition of mTORC1 is limited by its reactivation through compensatory and regulatory feedback loops. Here, we explored a strategy to curtail these drawbacks through inhibition of an important effector of the mTORC1signaling pathway, the eukaryotic initiation factor 4A (eIF4A). METHODS: We tested the anti-leukemic effect of a potent and specific eIF4A inhibitor (eIF4Ai), CR-1-31-B, in combination with cytosine arabinoside (araC) or the BCL2 inhibitor venetoclax. We utilized the MOLM-14 human AML cell line to model chemoresistant disease both in vitro and in vivo. In eIF4Ai-treated cells, we assessed for changes in survival, apoptotic priming, de novo protein synthesis, targeted intracellular metabolite content, bioenergetic profile, mitochondrial reactive oxygen species (mtROS) and mitochondrial membrane potential (MMP). RESULTS: eIF4Ai exhibits anti-leukemia activity in vivo while sparing non-malignant myeloid cells. In vitro, eIF4Ai synergizes with two therapeutic agents in AML, araC and venetoclax. EIF4Ai reduces mitochondrial membrane potential (MMP) and the rate of ATP synthesis from mitochondrial respiration and glycolysis. Furthermore, eIF4i enhanced apoptotic priming while reducing the expression levels of the antiapoptotic factors BCL2, BCL-XL and MCL1. Concomitantly, eIF4Ai decreases intracellular levels of specific metabolic intermediates of the tricarboxylic acid cycle (TCA cycle) and glucose metabolism, while enhancing mtROS. In vitro redox stress contributes to eIF4Ai cytotoxicity, as treatment with a ROS scavenger partially rescued the viability of eIF4A inhibition. CONCLUSIONS: We discovered that chemoresistant MOLM-14 cells rely on eIF4A-dependent cap translation for survival in vitro and in vivo. EIF4A drives an intrinsic metabolic program sustaining bioenergetic and redox homeostasis and regulates the expression of anti-apoptotic proteins. Overall, our work suggests that eIF4A-dependent cap translation contributes to adaptive processes involved in resistance to relevant therapeutic agents in AML.


Asunto(s)
Antineoplásicos , Citarabina , Factor 4A Eucariótico de Iniciación , Leucemia Mieloide Aguda , Humanos , Citarabina/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2 , Línea Celular Tumoral , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Antineoplásicos/farmacología
2.
Sci Rep ; 11(1): 18516, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34531456

RESUMEN

Rocaglates are a class of eukaryotic translation initiation inhibitors that are being explored as chemotherapeutic agents. They function by targeting eukaryotic initiation factor (eIF) 4A, an RNA helicase critical for recruitment of the 40S ribosome (and associated factors) to mRNA templates. Rocaglates perturb eIF4A activity by imparting a gain-of-function activity to eIF4A and mediating clamping to RNA. To appreciate how rocaglates could best be enabled in the clinic, an understanding of resistance mechanisms is important, as this could inform on strategies to bypass such events as well as identify responsive tumor types. Here, we report on the results of a positive selection, ORFeome screen aimed at identifying cDNAs capable of conferring resistance to rocaglates. Two of the most potent modifiers of rocaglate response identified were the transcription factors FOXP3 and NR1I3, both of which have been implicated in ABCB1 regulation-the gene encoding P-glycoprotein (Pgp). Pgp has previously been implicated in conferring resistance to silvestrol, a naturally occurring rocaglate, and we show here that this extends to additional synthetic rocaglate derivatives. In addition, FOXP3 and NR1I3 impart a multi-drug resistant phenotype that is reversed upon inhibition of Pgp, suggesting a potential therapeutic combination strategy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Benzofuranos/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factores de Transcripción Forkhead/genética , Receptores Citoplasmáticos y Nucleares/genética , Línea Celular , Receptor de Androstano Constitutivo , Regulación de la Expresión Génica/efectos de los fármacos , Pruebas Genéticas , Humanos
3.
Bioorg Med Chem Lett ; 47: 128111, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34353608

RESUMEN

Flavaglines such as silvestrol (1) and rocaglamide (2) constitute an interesting class of natural products with promising anticancer activities. Their mode of action is based on inhibition of eukaryotic initiation factor 4A (eIF4A) dependent translation through formation of a stable ternary complex with eIF4A and mRNA, thus blocking ribosome scanning. Herein we describe initial SAR studies in a novel series of 1-aminomethyl substituted flavagline-inspired eIF4A inhibitors. We discovered that a variety of N-substitutions at the 1-aminomethyl group are tolerated, making this position pertinent for property and ADME profile tuning. The findings presented herein are relevant to future drug design efforts towards novel eIF4A inhibitors with drug-like properties.


Asunto(s)
Antineoplásicos/farmacología , Benzofuranos/farmacología , Productos Biológicos/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Triterpenos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Benzofuranos/síntesis química , Benzofuranos/química , Productos Biológicos/síntesis química , Productos Biológicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Factor 4A Eucariótico de Iniciación/metabolismo , Humanos , Estructura Molecular , Relación Estructura-Actividad , Triterpenos/síntesis química , Triterpenos/química
4.
Cell Mol Life Sci ; 78(17-18): 6337-6349, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34398253

RESUMEN

Signaling via the B-cell receptor (BCR) is a key driver and therapeutic target in chronic lymphocytic leukemia (CLL). BCR stimulation of CLL cells induces expression of eIF4A, an initiation factor important for translation of multiple oncoproteins, and reduces expression of PDCD4, a natural inhibitor of eIF4A, suggesting that eIF4A may be a critical nexus controlling protein expression downstream of the BCR in these cells. We, therefore, investigated the effect of eIF4A inhibitors (eIF4Ai) on BCR-induced responses. We demonstrated that eIF4Ai (silvestrol and rocaglamide A) reduced anti-IgM-induced global mRNA translation in CLL cells and also inhibited accumulation of MYC and MCL1, key drivers of proliferation and survival, respectively, without effects on upstream signaling responses (ERK1/2 and AKT phosphorylation). Analysis of normal naïve and non-switched memory B cells, likely counterparts of the two main subsets of CLL, demonstrated that basal RNA translation was higher in memory B cells, but was similarly increased and susceptible to eIF4Ai-mediated inhibition in both. We probed the fate of MYC mRNA in eIF4Ai-treated CLL cells and found that eIF4Ai caused a profound accumulation of MYC mRNA in anti-IgM treated cells. This was mediated by MYC mRNA stabilization and was not observed for MCL1 mRNA. Following drug wash-out, MYC mRNA levels declined but without substantial MYC protein accumulation, indicating that stabilized MYC mRNA remained blocked from translation. In conclusion, BCR-induced regulation of eIF4A may be a critical signal-dependent nexus for therapeutic attack in CLL and other B-cell malignancies, especially those dependent on MYC and/or MCL1.


Asunto(s)
Factor 4A Eucariótico de Iniciación/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Anticuerpos Antiidiotipos/farmacología , Benzofuranos/farmacología , Células Cultivadas , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Triterpenos/farmacología
5.
Leukemia ; 35(9): 2469-2481, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34127794

RESUMEN

Eukaryotic initiation factor 4A (eIF4A), the enzymatic core of the eIF4F complex essential for translation initiation, plays a key role in the oncogenic reprogramming of protein synthesis, and thus is a putative therapeutic target in cancer. As important component of its anticancer activity, inhibition of translation initiation can alleviate oncogenic activation of HSF1, a stress-inducible transcription factor that enables cancer cell growth and survival. Here, we show that primary acute myeloid leukemia (AML) cells exhibit the highest transcript levels of eIF4A1 compared to other cancer types. eIF4A inhibition by the potent and specific compound rohinitib (RHT) inactivated HSF1 in these cells, and exerted pronounced in vitro and in vivo anti-leukemia effects against progenitor and leukemia-initiating cells, especially those with FLT3-internal tandem duplication (ITD). In addition to its own anti-leukemic activity, genetic knockdown of HSF1 also sensitized FLT3-mutant AML cells to clinical FLT3 inhibitors, and this synergy was conserved in FLT3 double-mutant cells carrying both ITD and tyrosine kinase domain mutations. Consistently, the combination of RHT and FLT3 inhibitors was highly synergistic in primary FLT3-mutated AML cells. Our results provide a novel therapeutic rationale for co-targeting eIF4A and FLT3 to address the clinical challenge of treating FLT3-mutant AML.


Asunto(s)
Antineoplásicos/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factores de Transcripción del Choque Térmico/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Humanos , Leucemia Mieloide Aguda/patología , Terapia Molecular Dirigida
6.
Curr Protein Pept Sci ; 22(7): 559-566, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34042032

RESUMEN

Eukaryotic translation initiation factor 4A (eIF4A) is a highly conserved DEAD-box RNA helicase in eukaryotes with ATPase and RNA helicase activities. eIF4A plays an important role in capdependent translation at the initiation of mRNA translation, and carcinoma signal transduction pathways are focused on cap-dependent translation. eIF4A is highly expressed in a variety of cancers, and its high expression is associated with the degree of leukemia progression. Therefore, eIF4A, as a target for tumor therapy, has become a hot research topic. Many small-molecule inhibitors targeting eIF4A have been demonstrated in preclinical cancer model trials. The purpose of this review is to describe the function of eIF4A and the development of eIF4A targeting inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Factor 4A Eucariótico de Iniciación , Leucemia , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/metabolismo , Humanos , Leucemia/tratamiento farmacológico , Leucemia/metabolismo
7.
Cell Commun Signal ; 19(1): 54, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001163

RESUMEN

Many clinical trials are being conducted to clarify effective combinations of various drugs for immune checkpoint blockade (ICB) therapy. However, although extensive studies from multiple aspects have been conducted regarding treatments for pancreatic ductal adenocarcinoma (PDAC), there are still no effective ICB-based therapies or biomarkers for this cancer type. A series of our studies have identified that the small GTPase ARF6 and its downstream effector AMAP1 (also called ASAP1/DDEF1) are often overexpressed in different cancers, including PDAC, and closely correlate with poor patient survival. Mechanistically, the ARF6-AMAP1 pathway drives cancer cell invasion and immune evasion, via upregulating ß1-integrins and PD-L1, and downregulating E-cadherin, upon ARF6 activation by external ligands. Moreover, the ARF6-AMAP1 pathway enhances the fibrosis caused by PDAC, which is another barrier for ICB therapies. KRAS mutations are prevalent in PDACs. We have shown previously that oncogenic KRAS mutations are the major cause of the aberrant overexpression of ARF6 and AMAP1, in which KRAS signaling enhances eukaryotic initiation factor 4A (eIF4A)-dependent ARF6 mRNA translation and eIF4E-dependent AMAP1 mRNA translation. MYC overexpression is also a key pathway in driving cancer malignancy. MYC mRNA is also known to be under the control of eIF4A, and the eIF4A inhibitor silvestrol suppresses MYC and ARF6 expression. Using a KPC mouse model of human PDAC (LSL-Kras(G12D/+); LSL-Trp53(R172H/+)); Pdx-1-Cre), we here demonstrate that inhibition of the ARF6-AMAP1 pathway by shRNAs in cancer cells results in therapeutic synergy with an anti-PD-1 antibody in vivo; and furthermore, that silvestrol improves the efficacy of anti-PD-1 therapy, whereas silvestrol on its own promotes tumor growth in vivo. ARF6 and MYC are both essential for normal cell functions. We demonstrate that silvestrol substantially mitigates the overexpression of ARF6 and MYC in KRAS-mutated cells, whereas the suppression is moderate in KRAS-intact cells. We propose that targeting eIF4A, as well as mutant KRAS, provides novel methods to improve the efficacy of anti-PD-1 and associated ICB therapies against PDACs, in which ARF6 and AMAP1 overexpression, as well as KRAS mutations of cancer cells are biomarkers to identify patients with drug-susceptible disease. The same may be applicable to other cancers with KRAS mutations. Video abstract.


Asunto(s)
Factor 6 de Ribosilación del ADP/metabolismo , Antígeno B7-H1/inmunología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Inmunoterapia , Mutación/genética , Neoplasias Pancreáticas/terapia , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Factor 4A Eucariótico de Iniciación/metabolismo , Femenino , Humanos , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/inmunología
8.
Cancer Res ; 81(8): 2002-2014, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33632898

RESUMEN

Pancreatic adenocarcinoma (PDAC) epitomizes a deadly cancer driven by abnormal KRAS signaling. Here, we show that the eIF4A RNA helicase is required for translation of key KRAS signaling molecules and that pharmacological inhibition of eIF4A has single-agent activity against murine and human PDAC models at safe dose levels. EIF4A was uniquely required for the translation of mRNAs with long and highly structured 5' untranslated regions, including those with multiple G-quadruplex elements. Computational analyses identified these features in mRNAs encoding KRAS and key downstream molecules. Transcriptome-scale ribosome footprinting accurately identified eIF4A-dependent mRNAs in PDAC, including critical KRAS signaling molecules such as PI3K, RALA, RAC2, MET, MYC, and YAP1. These findings contrast with a recent study that relied on an older method, polysome fractionation, and implicated redox-related genes as eIF4A clients. Together, our findings highlight the power of ribosome footprinting in conjunction with deep RNA sequencing in accurately decoding translational control mechanisms and define the therapeutic mechanism of eIF4A inhibitors in PDAC. SIGNIFICANCE: These findings document the coordinate, eIF4A-dependent translation of RAS-related oncogenic signaling molecules and demonstrate therapeutic efficacy of eIF4A blockade in pancreatic adenocarcinoma.


Asunto(s)
Adenocarcinoma/metabolismo , Factor 4A Eucariótico de Iniciación/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Regiones no Traducidas 5' , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/tratamiento farmacológico , Animales , Línea Celular Tumoral , Cicloheximida/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , G-Cuádruplex , Genes ras/genética , Humanos , Ratones , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Oxidación-Reducción , Neoplasias Pancreáticas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Polirribosomas/metabolismo , Biosíntesis de Proteínas , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Helicasas , Análisis de Secuencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma , Triterpenos/farmacología , Proteínas Señalizadoras YAP , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP ral/genética , Proteínas de Unión al GTP ral/metabolismo , Proteína RCA2 de Unión a GTP
9.
Antiviral Res ; 186: 105012, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33422611

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of COVID-19, a severe respiratory disease with varying clinical presentations and outcomes, and responsible for a major pandemic that started in early 2020. With no vaccines or effective antiviral treatments available, the quest for novel therapeutic solutions remains an urgent priority. Rocaglates, a class of plant-derived cyclopenta[b]benzofurans, exhibit broad-spectrum antiviral activity against multiple RNA viruses including coronaviruses. Specifically, rocaglates inhibit eukaryotic initiation factor 4A (eIF4A)-dependent mRNA translation initiation, resulting in strongly reduced viral RNA translation. Here, we assessed the antiviral activity of the synthetic rocaglate CR-31-B (-) against SARS-CoV-2 using both in vitro and ex vivo cell culture models. In Vero E6 cells, CR-31-B (-) inhibited SARS-CoV-2 replication with an EC50 of ~1.8 nM. In primary human airway epithelial cells, CR-31-B (-) reduced viral titers to undetectable levels at a concentration of 100 nM. Reduced virus reproduction was accompanied by substantially reduced viral protein accumulation and replication/transcription complex formation. The data reveal a potent anti-SARS-CoV-2 activity by CR-31-B (-), corroborating previous results obtained for other coronaviruses and supporting the idea that rocaglates may be used in first-line antiviral intervention strategies against novel and emerging RNA virus outbreaks.


Asunto(s)
Antivirales/farmacología , Benzofuranos/farmacología , Ácidos Hidroxámicos/farmacología , SARS-CoV-2/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Benzofuranos/química , Bronquios/virología , Células Cultivadas , Chlorocebus aethiops , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Humanos , Ácidos Hidroxámicos/química , Mucosa Respiratoria/virología , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Células Vero , Carga Viral/efectos de los fármacos , Compartimentos de Replicación Viral/efectos de los fármacos
10.
Oncol Rep ; 45(1): 230-238, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33416145

RESUMEN

Induction of the apoptosis of tumor cells is a promising therapeutic approach for the treatment of cancer. Tumor necrosis factor­related apoptosis­inducing ligand (TRAIL) is a novel type of anticancer drug. However, gallbladder cancer cells (GBC) exhibit strong resistance to TRAIL. The aim of the present study was to assess the effect of rocaglate CR­1­31B (CR­31), an inhibitor of eukaryotic translation initiation factor 4A (eIF4A), on the sensitization of cells to TRAIL­induced apoptosis in TRAIL­resistant GBC. eIF4A was highly abundant in GBC tissues and cell lines (GBC­SD and SGC­996). GBC cells were treated using TRAIL and/or CR­31 and then apoptosis and TRAIL signaling were detected in vitro. CR­31 enhanced the sensitivity of TRAIL­resistant GBC cells, due to the CR­31­mediated eIF4A translational downregulation of c­FLIP and the subsequent activation of the caspase cascade. Furthermore, GBC­SD tumor xenografts models were established and the effects of CR­31 in vivo were assessed. CR­31 significantly reduced the growth and initiated the apoptosis of tumor cells, suggesting that CR­31 also increased sensitivity in vivo. Taken together, the results of the present study show that CR­31 treatment countered the resistance to TRAIL in GBC cells in vitro and in vivo. Therefore, eIF4A may serve as a novel therapeutic target and its combination with TRAIL­CR­31 as a therapy may serve as a novel strategy for GBC treatment.


Asunto(s)
Benzofuranos/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Ácidos Hidroxámicos/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzofuranos/uso terapéutico , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Línea Celular Tumoral , Supervivencia Celular , Regulación hacia Abajo/efectos de los fármacos , Factor 4A Eucariótico de Iniciación/genética , Factor 4A Eucariótico de Iniciación/metabolismo , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/patología , Técnicas de Silenciamiento del Gen , Humanos , Ácidos Hidroxámicos/uso terapéutico , Masculino , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Cancer Ther ; 20(1): 26-36, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33037136

RESUMEN

The PI3K/AKT/mTOR pathway is often activated in lymphoma through alterations in PI3K, PTEN, and B-cell receptor signaling, leading to dysregulation of eIF4A (through its regulators, eIF4B, eIF4G, and PDCD4) and the eIF4F complex. Activation of eIF4F has a direct role in tumorigenesis due to increased synthesis of oncogenes that are dependent on enhanced eIF4A RNA helicase activity for translation. eFT226, which inhibits translation of specific mRNAs by promoting eIF4A1 binding to 5'-untranslated regions (UTR) containing polypurine and/or G-quadruplex recognition motifs, shows potent antiproliferative activity and significant in vivo efficacy against a panel of diffuse large B-cell lymphoma (DLBCL), and Burkitt lymphoma models with ≤1 mg/kg/week intravenous administration. Evaluation of predictive markers of sensitivity or resistance has shown that activation of eIF4A, mediated by mTOR signaling, correlated with eFT226 sensitivity in in vivo xenograft models. Mutation of PTEN is associated with reduced apoptosis in vitro and diminished efficacy in vivo in response to eFT226. In models evaluated with PTEN loss, AKT was stimulated without a corresponding increase in mTOR activation. AKT activation leads to the degradation of PDCD4, which can alter eIF4F complex formation. The association of eFT226 activity with PTEN/PI3K/mTOR pathway regulation of mRNA translation provides a means to identify patient subsets during clinical development.


Asunto(s)
Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Linfoma de Células B/genética , Linfoma de Células B/patología , Oncogenes , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Factor 4A Eucariótico de Iniciación/metabolismo , Femenino , Humanos , Ratones Endogámicos NOD , Ratones SCID , Fosfohidrolasa PTEN/metabolismo , ARN Mensajero/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cell Chem Biol ; 28(4): 475-486.e8, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33296667

RESUMEN

The translation inhibitor rocaglamide A (RocA) has shown promising antitumor activity because it uniquely clamps eukaryotic initiation factor (eIF) 4A onto polypurine RNA for selective translational repression. As eIF4A has been speculated to be a unique target of RocA, alternative targets have not been investigated. Here, we reveal that DDX3 is another molecular target of RocA. Proximity-specific fluorescence labeling of an O-nitrobenzoxadiazole-conjugated derivative revealed that RocA binds to DDX3. RocA clamps the DDX3 protein onto polypurine RNA in an ATP-independent manner. Analysis of a de novo-assembled transcriptome from the plant Aglaia, a natural source of RocA, uncovered the amino acid critical for RocA binding. Moreover, ribosome profiling showed that because of the dominant-negative effect of RocA, high expression of eIF4A and DDX3 strengthens translational repression in cancer cells. This study indicates that sequence-selective clamping of DDX3 and eIF4A, and subsequent dominant-negative translational repression by RocA determine its tumor toxicity.


Asunto(s)
Benzofuranos/farmacología , ARN Helicasas DEAD-box/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Benzofuranos/química , Células Cultivadas , ARN Helicasas DEAD-box/metabolismo , Inhibidores Enzimáticos/química , Factor 4A Eucariótico de Iniciación/metabolismo , Femenino , Humanos , Masculino , Modelos Moleculares , Conformación Molecular
13.
Nucleic Acids Res ; 48(17): 9521-9537, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32766783

RESUMEN

Hippuristanol (Hipp) is a natural product that selectively inhibits protein synthesis by targeting eukaryotic initiation factor (eIF) 4A, a DEAD-box RNA helicase required for ribosome recruitment to mRNA templates. Hipp binds to the carboxyl-terminal domain of eIF4A, locks it in a closed conformation, and inhibits its RNA binding. The dependencies of mRNAs for eIF4A during initiation is contingent on the degree of secondary structure within their 5' leader region. Interest in targeting eIF4A therapeutically in cancer and viral-infected settings stems from the dependencies that certain cellular (e.g. pro-oncogenic, pro-survival) and viral mRNAs show towards eIF4A. Using a CRISPR/Cas9-based variomics screen, we identify functional EIF4A1 Hipp-resistant alleles, which in turn allowed us to link the translation-inhibitory and cytotoxic properties of Hipp to eIF4A1 target engagement. Genome-wide translational profiling in the absence or presence of Hipp were undertaken and our validation studies provided insight into the structure-activity relationships of eIF4A-dependent mRNAs. We find that mRNA 5' leader length, overall secondary structure and cytosine content are defining features of Hipp-dependent mRNAs.


Asunto(s)
Regiones no Traducidas 5' , Resistencia a Antineoplásicos/genética , Factor 4A Eucariótico de Iniciación/genética , Esteroles/farmacología , Sistemas CRISPR-Cas , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/metabolismo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Mutación , Ribosomas/genética , Ribosomas/metabolismo
14.
Org Lett ; 22(16): 6257-6261, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32806219

RESUMEN

Rocaglates, rocaglamides, and related flavagline natural products exert their remarkable anticancer activity through inhibition of eukaryotic initiation factor 4A (eIF4A) but generally display suboptimal drug-like properties. In our efforts to identify potent drug-like eIF4A inhibitors, we developed synthetic strategies for diastereoselectively functionalizing the C1 position of aza-rocaglamide scaffolds (cf. 14 and 18), which proceed via retention or inversion of configuration at C1 depending on the C2 substituent (cf. 15 and 21) and ultimately enabled the discovery of novel and potent eIF4A inhibitors such as 25.


Asunto(s)
Benzofuranos/química , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Sitios de Unión , Productos Biológicos , Factor 4A Eucariótico de Iniciación/metabolismo , Humanos , Estructura Molecular
15.
J Med Chem ; 63(11): 5879-5955, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32470302

RESUMEN

Dysregulation of protein translation is a key driver for the pathogenesis of many cancers. Eukaryotic initiation factor 4A (eIF4A), an ATP-dependent DEAD-box RNA helicase, is a critical component of the eIF4F complex, which regulates cap-dependent protein synthesis. The flavagline class of natural products (i.e., rocaglamide A) has been shown to inhibit protein synthesis by stabilizing a translation-incompetent complex for select messenger RNAs (mRNAs) with eIF4A. Despite showing promising anticancer phenotypes, the development of flavagline derivatives as therapeutic agents has been hampered because of poor drug-like properties as well as synthetic complexity. A focused effort was undertaken utilizing a ligand-based design strategy to identify a chemotype with optimized physicochemical properties. Also, detailed mechanistic studies were undertaken to further elucidate mRNA sequence selectivity, key regulated target genes, and the associated antitumor phenotype. This work led to the design of eFT226 (Zotatifin), a compound with excellent physicochemical properties and significant antitumor activity that supports clinical development.


Asunto(s)
Benzofuranos/química , Diseño de Fármacos , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Animales , Benzofuranos/farmacocinética , Benzofuranos/uso terapéutico , Sitios de Unión , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cristalografía por Rayos X , Factor 4A Eucariótico de Iniciación/genética , Factor 4A Eucariótico de Iniciación/metabolismo , Femenino , Semivida , Humanos , Ligandos , Ratones , Ratones Desnudos , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , ARN Mensajero/química , ARN Mensajero/metabolismo , Ratas , Relación Estructura-Actividad
16.
Biochem Cell Biol ; 98(4): 502-510, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32008367

RESUMEN

The natural product pateamineA (PatA) is a highly potent antiproliferative agent. PatA and the simplified analog desmethyl, desamino pateamineA (DMDAPatA) have exhibited cytotoxicity selective for rapidly proliferating cells, and have been shown to inhibit cap-dependent translation initiation through binding to eIF4A (eukaryotic initiation factor 4A) of the eIF4F complex. PatA and DMDAPatA are both known to stimulate the RNA-dependent ATPase, and ATP-dependent RNA helicase activities of eIF4A. The impact of other eIF4F components, eIF4E and eIF4G, on DMDAPatA action were investigated in vitro and in cultured mammalian cells. The perturbation of the eIF4A-eIF4G association was found to be eIF4E- and mRNA cap-dependent. An inhibitory effect on helicase activity of eIF4A was observed when it was part of a complex that mimicked the eIF4F complex. We propose a model of action for DMDAPatA (and by supposition PatA) where the cellular activity of the compound is dependent on an "active" eIF4F complex.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Compuestos Epoxi/química , Compuestos Epoxi/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Macrólidos/química , Macrólidos/farmacología , Neoplasias/tratamiento farmacológico , ARN Mensajero/metabolismo , Tiazoles/química , Tiazoles/farmacología , Adenosina Trifosfatasas/genética , Productos Biológicos , Línea Celular , Proliferación Celular , Humanos , Neoplasias/metabolismo , Neoplasias/patología , ARN Mensajero/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
RNA ; 26(5): 541-549, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32014999

RESUMEN

The PI3K/Akt/mTOR kinase pathway is extensively deregulated in human cancers. One critical node under regulation of this signaling axis is eukaryotic initiation factor (eIF) 4F, a complex involved in the control of translation initiation rates. eIF4F-dependent addictions arise during tumor initiation and maintenance due to increased eIF4F activity-generally in response to elevated PI3K/Akt/mTOR signaling flux. There is thus much interest in exploring eIF4F as a small molecule target for the development of new anticancer drugs. The DEAD-box RNA helicase, eIF4A, is an essential subunit of eIF4F, and several potent small molecules (rocaglates, hippuristanol, pateamine A) affecting its activity have been identified and shown to demonstrate anticancer activity in vitro and in vivo in preclinical models. Recently, a number of new small molecules have been reported as having the capacity to target and inhibit eIF4A. Here, we undertook a comparative analysis of their biological activity and specificity relative to the eIF4A inhibitor, hippuristanol.


Asunto(s)
Antineoplásicos/química , Factor 4A Eucariótico de Iniciación/química , Neoplasias/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/química , Esteroles/química , Antineoplásicos/farmacología , Benzofuranos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Compuestos Epoxi/química , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factor 4F Eucariótico de Iniciación/antagonistas & inhibidores , Factor 4F Eucariótico de Iniciación/química , Humanos , Macrólidos/química , Neoplasias/genética , Fosfatidilinositol 3-Quinasas/genética , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Esteroles/farmacología , Serina-Treonina Quinasas TOR/genética , Tiazoles/química
18.
Biochem Biophys Res Commun ; 523(3): 795-801, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-31954521

RESUMEN

The DEAD-box family of RNA helicases plays essential roles in both transcriptional and translational mRNA degradation; they unwind short double-stranded RNA by breaking the RNA-RNA interactions. Two DEAD-box RNA helicases, eukaryotic translation initiation factor 4A3 (eIF4A3) and DEAD-box helicase 3 (DDX3X), show high homology in the ATP-binding region and are considered key molecules for cancer progression. Several small molecules that target eIF4A3 and DDX3X have been reported to inhibit cancer cell growth; however, more potent compounds are required for cancer therapeutics, and there is a critical need for high-throughput assays to screen for RNA helicase inhibitors. In this study, we developed novel fluorescence resonance energy transfer-based high-throughput RNA helicase assays for eIF4A3 and DDX3X. Using these assays, we identified several eIF4A3 allosteric inhibitors whose inhibitory effect on eIF4A3 ATPase showed a strong correlation with inhibitory effect on helicase activity. From 102 compounds that exhibited eIF4A3 ATPase inhibition, we identified a selective DDX3X inhibitor, C1, which showed stronger inhibition of DDX3X than of eIF4A3. Small-molecule helicase inhibitors can be valuable for clarifying the molecular machinery of DEAD-box RNA helicases. The high-throughput quantitative assays established here should facilitate the evaluation of the helicase inhibitory activity of compounds.


Asunto(s)
ARN Helicasas DEAD-box/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , ARN Helicasas DEAD-box/metabolismo , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Pruebas de Enzimas/métodos , Factor 4A Eucariótico de Iniciación/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Bibliotecas de Moléculas Pequeñas/química
19.
Antiviral Res ; 175: 104706, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31931103

RESUMEN

Rocaglates, a class of natural compounds isolated from plants of the genus Aglaia, are potent inhibitors of translation initiation. They are proposed to form stacking interactions with polypurine sequences in the 5'-untranslated region (UTR) of selected mRNAs, thereby clamping the RNA substrate onto eIF4A and causing inhibition of the translation initiation complex. Since virus replication relies on the host translation machinery, it is not surprising that the rocaglate Silvestrol has broad-spectrum antiviral activity. Unfortunately, synthesis of Silvestrol is sophisticated and time-consuming, thus hampering the prospects for further antiviral drug development. Here, we present the less complex structured synthetic rocaglate CR-31-B (-) as a novel compound with potent broad-spectrum antiviral activity in primary cells and in an ex vivo bronchial epithelial cell system. CR-31-B (-) inhibited the replication of corona-, Zika-, Lassa-, Crimean Congo hemorrhagic fever viruses and, to a lesser extent, hepatitis E virus (HEV) at non-cytotoxic low nanomolar concentrations. Since HEV has a polypurine-free 5'-UTR that folds into a stable hairpin structure, we hypothesized that RNA clamping by Silvestrol and its derivatives may also occur in a polypurine-independent but structure-dependent manner. Interestingly, the HEV 5'-UTR conferred sensitivity towards Silvestrol but not to CR-31-B (-). However, if an exposed polypurine stretch was introduced into the HEV 5'-UTR, CR-31-B (-) became an active inhibitor comparable to Silvestrol. Moreover, thermodynamic destabilization of the HEV 5'-UTR led to reduced translational inhibition by Silvestrol, suggesting differences between rocaglates in their mode of action, most probably by engaging Silvestrol's additional dioxane moiety.


Asunto(s)
Antivirales/farmacología , Benzofuranos/farmacología , Triterpenos/farmacología , Replicación Viral/efectos de los fármacos , Virus/efectos de los fármacos , Células A549 , Animales , Antivirales/síntesis química , Benzofuranos/síntesis química , Bronquios/citología , Técnicas de Cultivo de Célula , Células Cultivadas , Células Epiteliales/virología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Hepatocitos/virología , Humanos , Ratones , Virus/clasificación
20.
Nat Prod Rep ; 37(5): 609-616, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31782447

RESUMEN

Covering: up to 2019Pharmacological targeting of eukaryotic mRNA translation initiation is a promising approach for cancer therapy, since several signaling pathways that are commonly deregulated during tumor progression converge on this process. The DEAD-box helicase, eukaryotic initiation factor (eIF) 4A, is essential for translation initiation and facilitates the loading of the 43S pre-initiation complex onto mRNAs. Hippuristanol, rocaglates, and pateamine A are natural products that each target eIF4A by interfering with the helicase's RNA-binding activity in distinct manners. They exert a selective change in gene expression that results in potent anti-tumorigenic activity in pre-clinical studies. This review will provide an update on the molecular mechanisms of action of these natural products.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Factor 4A Eucariótico de Iniciación/metabolismo , Animales , Productos Biológicos/química , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/metabolismo , Compuestos Epoxi/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Macrólidos/farmacología , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , Esteroles/farmacología , Tiazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...