Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Br J Cancer ; 122(10): 1425-1427, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32161367

RESUMEN

The recognition that new cancer drugs can be truly tumour-agnostic based on mechanism-of-action is important for paediatric cancers, where access to novel targeted therapies developed for adult indications has sometimes been problematic. The recently approved drug larotrectinib is an excellent case study of the development of a tumour-agnostic drug relevant to children.


Asunto(s)
Neoplasias/tratamiento farmacológico , Proteínas de Fusión Oncogénica/efectos de los fármacos , Pediatría , Adulto , Antineoplásicos/uso terapéutico , Niño , Aprobación de Drogas , Humanos , Neoplasias/epidemiología , Neoplasias/genética , Preparaciones Farmacéuticas , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico
2.
Br J Cancer ; 120(2): 165-171, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30420614

RESUMEN

BACKGROUND: Next-generation sequencing has identified actionable genetic aberrations in intrahepatic cholangiocarcinomas (iCCA), including the fibroblast growth factor receptor 2 (FGFR2) fusions. Derazantinib (ARQ 087), an orally bioavailable, multi-kinase inhibitor with potent pan-FGFR activity, has shown preliminary therapeutic activity against FGFR2 fusion-positive iCCA. METHODS: This multicentre, phase 1/2, open-label study enrolled adult patients with unresectable iCCA with FGFR2 fusion, who progressed, were intolerant or not eligible to first-line chemotherapy (NCT01752920). Subjects received derazantinib in continuous daily doses. Tumour response was assessed according to RECIST 1.1 every 8 weeks. RESULTS: Twenty-nine patients (18 women/11 men; median age, 58.7 years), 2 treatment-naive and 27 who progressed after at least one prior systemic therapy, were enrolled. Overall response rate was 20.7%, disease control rate was 82.8%. Estimated median progression-free survival was 5.7 months (95% CI: 4.04-9.2 months). Treatment-related adverse events (AE) were observed in 27 patients (93.1%, all grades), including asthenia/fatigue (69.0%), eye toxicity (41.4%), and hyperphosphatemia (75.9%). Grade ≥ 3 AEs occurred in 8 patients (27.6%). CONCLUSION: Derazantinib demonstrated encouraging anti-tumour activity and a manageable safety profile in patients with advanced, unresectable iCCA with FGFR2 fusion who progressed after chemotherapy. A pivotal trial of derazantinib in iCCA is ongoing (NCT03230318).


Asunto(s)
Alcaloides/administración & dosificación , Colangiocarcinoma/tratamiento farmacológico , Proteínas de Fusión Oncogénica/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Adulto , Anciano , Anciano de 80 o más Años , Alcaloides/efectos adversos , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Ensayos Clínicos como Asunto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Proteínas de Fusión Oncogénica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos
3.
BMC Cancer ; 17(1): 107, 2017 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-28166781

RESUMEN

BACKGROUND: Desmoplastic small round cell tumor (DSRCT) is a rare and highly aggressive disease, that can be described as a member of the family of small round blue cell tumors. The molecular diagnostic marker is the t(11;22)(p13;q12) translocation, which creates an aberrant transcription factor, EWS-WT1, that underlies the oncogenesis of DSRCT. Current treatments are not very effective so new active drugs are needed. Trabectedin, now used as a single agent for the treatment of soft tissue sarcoma, was reported to be active in some pre-treated DSRCT patients. Using JN-DSRCT-1, a cell line derived from DSRCT expressing the EWS-WT1 fusion protein, we investigated the ability of trabectedin to modify the function of the chimeric protein, as in other sarcomas expressing fusion proteins. After detailed characterization of the EWS-WT1 transcripts structure, we investigated the mode of action of trabectedin, looking at the expression and function of the oncogenic chimera. METHODS: We characterized JN-DSRCT-1 cells using cellular approaches (FISH, Clonogenicity assay) and molecular approaches (Sanger sequencing, ChIP, GEP). RESULTS: JN-DSRCT-1 cells were sensitive to trabectedin at nanomolar concentrations. The cell line expresses different variants of EWS-WT1, some already identified in patients. EWS-WT1 mRNA expression was affected by trabectedin and chimeric protein binding on its target gene promoters was reduced. Expression profiling indicated that trabectedin affects the expression of genes involved in cell proliferation and apoptosis. CONCLUSIONS: The JN-DSRCT-1 cell line, in vitro, is sensitive to trabectedin: after drug exposure, EWS-WT1 chimera expression decreases as well as binding on its target promoters. Probably the heterogeneity of chimera transcripts is an obstacle to precisely defining the molecular mode of action of drugs, calling for further cellular models of DSRCT, possibly growing in vivo too, to mimic the biological complexity of this disease.


Asunto(s)
Tumor Desmoplásico de Células Pequeñas Redondas/tratamiento farmacológico , Dioxoles/farmacología , Regulación Neoplásica de la Expresión Génica , Proteínas de Fusión Oncogénica/efectos de los fármacos , Tetrahidroisoquinolinas/farmacología , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Tumor Desmoplásico de Células Pequeñas Redondas/metabolismo , Tumor Desmoplásico de Células Pequeñas Redondas/fisiopatología , Dioxoles/uso terapéutico , Humanos , Proteínas de Fusión Oncogénica/genética , Proteína EWS de Unión a ARN , Tetrahidroisoquinolinas/uso terapéutico , Trabectedina , Proteínas WT1
4.
Mol Cancer ; 15(1): 47, 2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27296891

RESUMEN

BACKGROUND: Fusion proteins have unique oncogenic properties and their identification can be useful either as diagnostic or therapeutic targets. Next generation sequencing data have previously shown a fusion gene formed between Rad51C and ATXN7 genes in the MCF7 breast cancer cell line. However, the existence of this fusion gene in colorectal patient tumor tissues is largely still unknown. METHODS: We evaluated for the presence of Rad51C-ATXN7 fusion gene in colorectal tumors and cells by RT-PCR, PCR, Topo TA cloning, Real time PCR, immunoprecipitation and immunoblotting techniques. RESULTS: We identified two forms of fusion mRNAs between Rad51C and ATXN7 in the colorectal tumors, including a Variant 1 (fusion transcript between Rad51C exons 1-7 and ATXN7 exons 6-13), and a Variant 2 (between Rad51C exons 1-6 and ATXN7 exons 6-13). In silico analysis showed that the Variant 1 produces a truncated protein, whereas the Variant 2 was predicted to produce a fusion protein with molecular weight of 110 KDa. Immunoprecipitation and Western blot analysis further showed a 110 KDa protein in colorectal tumors. 5-Azacytidine treatment of LS-174 T cells caused a 3.51-fold increase in expression of the fusion gene (Variant 2) as compared to no treatment controls evaluated by real time PCR. CONCLUSION: In conclusion we found a fusion gene between DNA repair gene Rad51C and neuro-cerebral ataxia Ataxin-7 gene in colorectal tumors. The in-frame fusion transcript of Variant 2 results in a fusion protein with molecular weight of 110 KDa. In addition, we found that expression of fusion gene is associated with functional impairment of Fanconi Anemia (FA) DNA repair pathway in colorectal tumors. The expression of Rad51C-ATXN7 in tumors warrants further investigation, as it suggests the potential of the fusion gene in treatment and predictive value in colorectal cancers.


Asunto(s)
Ataxina-7/genética , Clonación Molecular/métodos , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/genética , Proteínas de Fusión Oncogénica/genética , Ataxina-7/metabolismo , Azacitidina/farmacología , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Simulación por Computador , Metilación de ADN/efectos de los fármacos , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Variación Genética , Humanos , Peso Molecular , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/metabolismo
5.
Recent Results Cancer Res ; 201: 197-205, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24756793

RESUMEN

Crizotinib is an ATP-competitive small-molecule inhibitor of the receptor tyrosine kinases (RTK) c-Met, anaplastic lymphoma kinase (ALK), and ROS1. There is convincing clinical evidence for the effectiveness in non-small-cell lung cancer (NSCLC) harboring EML4-ALK rearrangements resulting in constitutional activation of the ALK-RTK. The drug is approved for this entity, which represents no more than 3-5% of all NSCLC. However, in this population, impressive response rates are generated. The same seems to be true for ROS-1 rearrangements; however, these only occur in approximately 1% of all NSCLC. The role in c-Met altered cancers needs to be determined. Toxicities include visual impairment, nausea, peripheral edema, QT-prolongation, and liver enzyme elevation. Also, the occurrence of renal cysts is reported. Fluorescence in situ hybridization (FISH) detecting the ALK rearrangement has to be performed on tumor tissue to predict crizotinib efficacy. The role of immunohistochemistry in this setting needs to be determined. It has high concordance with FISH results when strongly positive or completely negative. The high efficacy of crizotinib in ALK- and ROS-positive lung cancer as new molecular targets beside the epidermal growth factor receptor (EGFR) underscores the importance of molecular typing in NSCLC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Crizotinib , Humanos , Neoplasias Pulmonares/genética , Proteínas de Fusión Oncogénica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico
6.
J Pharmacol Sci ; 126(3): 179-85, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25319615

RESUMEN

Acute promyelocytic leukemia (APL) is characterized by the occurrence of translocations between chromosomes 15 and 17, resulting in generation of a fusion protein of promyelocytic leukemia (PML) and retinoid A receptor (RAR) α. APL cells are unable to differentiate into mature granulocytes since PML-RARα functions as a strong transcriptional repressor for a gene involved in granulocyte differentiation. All-trans retinoic acid (ATRA) is the first agent that has been developed to target specific disease-causing molecules, i.e., ATRA suppresses abnormal functions of oncogenic proteins. Moreover, ATRA facilitates the differentiation of APL cells toward mature granulocytes by changing epigenetic modifiers from corepressor complexes to co-activator complexes on target genes after binding to the ligand-binding domain at the RARα moiety of the PML-RARα oncoprotein. On the other hand, arsenic trioxide (ATO), another promising agent used to treat APL, directly binds to the PML moiety of the PML-RARα protein, causing oxidation and multimerization. ATO enhances the conjugation of small ubiquitin-like modifiers to PML-RARα, followed by ubiquitination and degradation, relieving the genes associated with granulocytic differentiation from suppressive restraint by the oncoprotein. Recent clinical studies have demonstrated that combination therapy with both ATRA and ATO is useful to achieve remission.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Arsenicales/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Óxidos/uso terapéutico , Tretinoina/uso terapéutico , Animales , Trióxido de Arsénico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Sinergismo Farmacológico , Epigénesis Genética/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Resultado del Tratamiento
7.
Gene ; 917: 148443, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38582263

RESUMEN

Acute promyelocytic leukemia (APL) is a type of acute myeloid leukemia (AML) with a high mortality rate, and the production of PML-RARα fusion protein is the cause of its pathogenesis. Our group has synthesized a novel compound, 4-amino-2-trifluoromethyl-phenyl retinate (ATPR), by structural modification of All-trans retinoic acid (ATRA), which has strong cell differentiation-inducing effects and inhibits the expression of PML-RARα. In this study, acute promyelocytic leukemia NB4 cells before and after ATPR induction were analyzed by whole transcriptome microarray, and the expression of lncRNA CONCR was found to be significantly downregulated. The role of CONCR in ATPR-induced cell differentiation and cycle arrest was explored through overexpression and silencing of CONCR. And then the database was used to predict that CONCR may bind to DEAD/H-Box Helicase 11 (DDX11) protein to further explore the role of CONCR binding to DDX11. The results showed that ATPR could reduce the expression of CONCR, and overexpression of CONCR could reverse the ATPR-induced cell differentiation and cycle blocking effect, and conversely silencing of CONCR could promote this effect. RNA immunoprecipitation (RIP) experiments showed that CONCR could bind to DDX11, the protein expression levels of DDX11 and PML-RARα were elevated after overexpression of CONCR. These results suggest that ATPR can regulate the expression of DDX11 through CONCR to affect the expression of PML-RARα fusion protein, which in turn induces the differentiation and maturation of APL cells.


Asunto(s)
Puntos de Control del Ciclo Celular , Diferenciación Celular , ARN Helicasas DEAD-box , Leucemia Promielocítica Aguda , Proteínas de Fusión Oncogénica , ARN Largo no Codificante , Transducción de Señal , Humanos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , ARN Helicasas DEAD-box/efectos de los fármacos , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Regulación Leucémica de la Expresión Génica , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , ARN Largo no Codificante/efectos de los fármacos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Tretinoina/farmacología
8.
Postepy Hig Med Dosw (Online) ; 67: 1083-9, 2013 Nov 25.
Artículo en Polaco | MEDLINE | ID: mdl-24379249

RESUMEN

During last decades acute promyelocytic leukemia, once considered the deadly disease, has evolved to the most treatable of all subtypes of acute myeloid leukemias. The intense clinical and basic research has led to a rational approach to treatment in which the use of the differentiating agent all-trans-retinoic acid has proven to be effective first-line therapy. Arsenic trioxide, used for relapsed disease, further improved the survival rate of patients. The classical model presented the therapeutic success as a result of over-coming of the differentiation block characteristic of neoplastic cells. However, the resent in vivo and ex vivo studies, seem to show that the induction of differentiation process is not required to cure acute promyelocytic leukemia. Rather than inducing differentiation, targeting clonogenic leukemia initiating cells or destroying PML-RARa fusion protein may represent a more effective therapeutic goal in this type of leukemia.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Tretinoina/uso terapéutico , Trióxido de Arsénico , Arsenicales/uso terapéutico , Humanos , Leucemia Promielocítica Aguda/mortalidad , Proteínas de Fusión Oncogénica/efectos de los fármacos , Óxidos/uso terapéutico , Prevención Secundaria , Tasa de Supervivencia
11.
Nat Med ; 9(11): 1413-7, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14566333

RESUMEN

Despite improved molecular characterization of malignancies and development of targeted therapies, acute leukemia is not curable and few patients survive more than 10 years after diagnosis. Recently, combinations of different therapeutic strategies (based on mechanisms of apoptosis, differentiation and cytotoxicity) have significantly increased survival. To further improve outcome, we studied the potential efficacy of boosting the patient's immune response using specific immunotherapy. In an animal model of acute promyelocytic leukemia, we developed a DNA-based vaccine by fusing the human promyelocytic leukemia-retinoic acid receptor-alpha (PML-RARA) oncogene to tetanus fragment C (FrC) sequences. We show for the first time that a DNA vaccine specifically targeted to an oncoprotein can have a pronounced effect on survival, both alone and when combined with all-trans retinoic acid (ATRA). The survival advantage is concomitant with time-dependent antibody production and an increase in interferon-gamma (IFN-gamma). We also show that ATRA therapy on its own triggers an immune response in this model. When DNA vaccination and conventional ATRA therapy are combined, they induce protective immune responses against leukemia progression in mice and may provide a new approach to improve clinical outcome in human leukemia.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/prevención & control , Proteínas de Neoplasias/inmunología , Proteínas de Fusión Oncogénica/inmunología , Vacunas de ADN/inmunología , Animales , Antineoplásicos/farmacología , Vacunas contra el Cáncer/farmacología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Tretinoina/farmacología , Vacunas de ADN/farmacología
12.
Nat Med ; 5(6): 669-76, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10371506

RESUMEN

Blockage in myeloid differentiation characterizes acute myeloid leukemia (AML); the stage of the blockage defines distinct AML subtypes (AML1/2 to AML5). Differentiation therapy in AML has recently raised interest because the survival of AML3 patients has been greatly improved using the differentiating agent retinoic acid. However, this molecule is ineffective in other AML subtypes. The CD44 surface antigen, on leukemic blasts from most AML patients, is involved in myeloid differentiation. Here, we report that ligation of CD44 with specific anti-CD44 monoclonal antibodies or with hyaluronan, its natural ligand, can reverse myeloid differentiation blockage in AML1/2 to AML5 subtypes. The differentiation of AML blasts was evidenced by the ability to produce oxidative bursts, the expression of lineage antigens and cytological modifications, all specific to normal differentiated myeloid cells. These results indicate new possibilities for the development of CD44-targeted differentiation therapy in the AML1/2 to AML5 subtypes.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Enfermedad Aguda , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Médula Ósea/metabolismo , Médula Ósea/patología , Relación Dosis-Respuesta a Droga , Factor Estimulante de Colonias de Granulocitos/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/genética , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Granulocitos/patología , Humanos , Receptores de Hialuranos/efectos de los fármacos , Receptores de Hialuranos/inmunología , Ácido Hialurónico/química , Ácido Hialurónico/metabolismo , Ácido Hialurónico/farmacología , Leucemia Mieloide/tratamiento farmacológico , Antígeno Lewis X/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Factor Estimulante de Colonias de Macrófagos/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/genética , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/metabolismo , ARN Mensajero/análisis , Estallido Respiratorio , Tretinoina/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/inmunología , Células Tumorales Cultivadas/metabolismo
13.
Theranostics ; 11(17): 8172-8184, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373735

RESUMEN

Chromosome translocations involving mixed lineage leukemia (MLL) gene cause acute leukemia with a poor prognosis. MLL is frequently fused with transcription cofactors AF4 (~35%), AF9 (25%) or its paralog ENL (10%). The AHD domain of AF9/ENL binds to AF4, its paralog AFF4, or histone-H3 lysine-79 (H3K79) methyltransferase DOT1L. Formation of AF9/ENL/AF4/AFF4-containing super elongation complexes (SEC) and the catalytic activity of DOT1L are essential for MLL-rearranged leukemia. Protein-protein interactions (PPI) between AF9/ENL and DOT1L/AF4/AFF4 are therefore a potential drug target. Methods: Compound screening followed by medicinal chemistry was used to find inhibitors of such PPIs, which were examined for their biological activities against MLL-rearranged leukemia and other cancer cells. Results: Compound-1 was identified to be a novel small-molecule inhibitor of the AF9/ENL-DOT1L/AF4/AFF4 interaction with IC50s of 0.9-3.5 µM. Pharmacological inhibition of the PPIs significantly reduced SEC and DOT1L-mediated H3K79 methylation in the leukemia cells. Gene profiling shows compound-1 significantly suppressed the gene signatures related to onco-MLL, DOT1L, HoxA9 and Myc. It selectively inhibited proliferation of onco-MLL- or Myc-driven cancer cells and induced cell differentiation and apoptosis. Compound-1 exhibited strong antitumor activity in a mouse model of MLL-rearranged leukemia. Conclusions: The AF9/ENL-DOT1L/AF4/AFF4 interactions are validated to be an anticancer target and compound-1 is a useful in vivo probe for biological studies as well as a pharmacological lead for further drug development.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Mieloide Aguda , Proteínas de Fusión Oncogénica , Animales , Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ratones , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Oncogenes/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/efectos de los fármacos , Factores de Elongación Transcripcional/genética
15.
Semin Oncol ; 36(2 Suppl 1): S36-41, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19393834

RESUMEN

Selective kinase inhibitors have emerged as an important class of cancer therapeutics. The clinical success of drugs such as imatinib, erlotinib, and lapatinib, together with findings demonstrating the important relationship between specific tumor genotypes and clinical response to these agents, also has brought to the forefront the concept of "personalized cancer medicine." The potential broader significance of this relationship has been further highlighted in preclinical studies using tumor-derived cell lines as a model system that can faithfully recapitulate the association of specific genotypes with drug sensitivity, suggesting the utility of cancer cell lines to identify novel candidate biomarkers for predicting clinically responsive patient subsets for newly developed anticancer agents. The case of the anaplastic lymphoma kinase (ALK) nicely exemplifies this, and cell line profiling has revealed that ALK mutations present in a subset of anaplastic large cell lymphomas (ALCLs), non-small cell lung cancers (NSCLCs), and neuroblastomas appear to sensitize cancer cells to treatment with selective ALK kinase inhibitors. Such findings suggest that genotype-based stratification of cancer patients for treatment with selective kinase inhibitors, even across multiple diseases of distinct tissue origin, may be essential for maximizing their clinical benefit.


Asunto(s)
Neoplasias/tratamiento farmacológico , Proteínas de Fusión Oncogénica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinasa de Linfoma Anaplásico , Células Cultivadas , Diseño de Fármacos , Humanos , Neoplasias/enzimología , Neoplasias/fisiopatología , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas Receptoras
16.
Medicine (Baltimore) ; 98(13): e14998, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30921216

RESUMEN

RATIONALE: Acute myeloid leukemia (AML) is a malignantly clonal and highly heterogeneous disease. Although the treatment of AML has brought promising outcomes for younger patients, prognosis of the elderly remains dismal. Innovative regimens are increasingly necessary to be investigated. PATIENT CONCERNS: We present an 86-year-old AML patient with fever, cough, and sputum production. DIAGNOSES: A diagnosis of AML with maturation (AML-M2) and AML1/ETO was made. INTERVENTIONS: The patient was treated with a regimen of Homoharringtonine coupled with arsenic trioxide. OUTCOMES: The AML-M2 patient with AML1/ETO achieved incomplete remission, but showed few toxic side effects and improved survival. Besides, we analyzed the dynamic counts of complete blood cells during the treatment. The count of white blood cell had a positive correlation with the percentage of blast cells (r = 0.65), both of which had a negative correlation with the percentage of segmented neutrophils (r = -0.63, -0.89). LESSONS: Homoharringtonine and arsenic trioxide may induce both the apoptosis and differentiation of leukemic cells in AML-M2 with AML1/ETO.


Asunto(s)
Antineoplásicos/administración & dosificación , Trióxido de Arsénico/administración & dosificación , Subunidad alfa 2 del Factor de Unión al Sitio Principal/efectos de los fármacos , Homoharringtonina/administración & dosificación , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteína 1 Compañera de Translocación de RUNX1/efectos de los fármacos , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Quimioterapia Combinada , Humanos , Masculino
17.
Int Arch Allergy Immunol ; 146 Suppl 1: 7-10, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18504399

RESUMEN

BACKGROUND: Acetylation and deacetylation of proteins occur in cells in response to various stimuli, and are reversibly catalyzed by histone acetyltransferase and histone deacetylase (HDAC), respectively. EoL-1 cells have an FIP1L1-PDGFRA fusion gene that causes transformation of eosinophilic precursor cells into leukemia cells. The HDAC inhibitors apicidin and n-butyrate suppress the proliferation of EoL-1 cells and induce differentiation into eosinophils by a decrease in the protein level of FIP1L1-PDGFRalpha without affecting the mRNA level for FIP1L1-PDGFRA. In this study, we analyzed the mechanism by which the protein level of FIP1L1-PDGFRalpha is decreased by apicidin and n-butyrate. METHODS: EoL-1 cells were incubated in the presence of the HDAC inhibitors apicidin, trichostatin A or n-butyrate. The protein levels of FIP1L1-PDGFRalpha and phosphorylated eIF-2alpha were determined by Western blotting. Actinomycin D and cycloheximide were used to block RNA synthesis and protein synthesis, respectively, in the chasing experiment of the amount of FIP1L1-PDGFRalpha protein. RESULTS: When apicidin- and n-butyrate-treated EoL-1 cells were incubated in the presence of actinomycin D, the decrease in the protein level of FIP1L1-PDGFRalpha was significantly enhanced when compared with controls. In contrast, the protein levels were not changed by cycloheximide among these groups. Apicidin and n-butyrate induced the continuous phosphorylation of eIF-2alpha for up to 8 days. CONCLUSIONS: The decrease in the level of FIP1L1-PDGFRalpha protein by continuous inhibition of HDAC may be due to the decrease in the translation rate of FIP1L1-PDGFRA.


Asunto(s)
Butiratos/farmacología , Inhibidores Enzimáticos/farmacología , Eosinófilos/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Inhibidores de Histona Desacetilasas , Síndrome Hipereosinofílico/tratamiento farmacológico , Proteínas de Fusión Oncogénica/análisis , Péptidos Cíclicos/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis , Factores de Escisión y Poliadenilación de ARNm/análisis , Acetilación/efectos de los fármacos , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Eosinófilos/metabolismo , Factor 2 Eucariótico de Iniciación/análisis , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Histona Desacetilasas/metabolismo , Humanos , Síndrome Hipereosinofílico/metabolismo , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Escisión y Poliadenilación de ARNm/efectos de los fármacos , Factores de Escisión y Poliadenilación de ARNm/metabolismo
18.
Rev Med Interne ; 29(1): 65-7, 2008 Jan.
Artículo en Francés | MEDLINE | ID: mdl-18036706

RESUMEN

Hypereosinophilic syndromes (HES) are a heterogeneous group of disorders characterized by marked peripheral blood and tissue eosinophilia resulting in organ damage. Recent advances in molecular biology have led to the identification of a FIP1L1-PDGFRA fusion gene as a recurrent abnormality in some patients with HES. This fusion gene results from a cryptic 4q12 interstitial deletion involving an 800 kb region. Recent reports indicate that this subtype of HES is imatinib responsive with rapid and complete haematological remissions. Here we report two patients successfully treated with imatinib.


Asunto(s)
Síndrome Hipereosinofílico/tratamiento farmacológico , Proteínas de Fusión Oncogénica/efectos de los fármacos , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Factores de Escisión y Poliadenilación de ARNm/efectos de los fármacos , Adulto , Benzamidas , Humanos , Síndrome Hipereosinofílico/genética , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
19.
Cancer Cell ; 33(2): 202-216.e6, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29358035

RESUMEN

Many cancer types are driven by oncogenic transcription factors that have been difficult to drug. Transcriptional inhibitors, however, may offer inroads into targeting these cancers. Through chemical genomics screening, we identified that Ewing sarcoma is a disease with preferential sensitivity to THZ1, a covalent small-molecule CDK7/12/13 inhibitor. The selective CDK12/13 inhibitor, THZ531, impairs DNA damage repair in an EWS/FLI-dependent manner, supporting a synthetic lethal relationship between response to THZ1/THZ531 and EWS/FLI expression. The combination of these molecules with PARP inhibitors showed striking synergy in cell viability and DNA damage assays in vitro and in multiple models of Ewing sarcoma, including a PDX, in vivo without hematopoietic toxicity.


Asunto(s)
Quinasas Ciclina-Dependientes/efectos de los fármacos , Fenilendiaminas/farmacología , Proteína Proto-Oncogénica c-fli-1/genética , Pirimidinas/farmacología , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/efectos de los fármacos , Proteína EWS de Unión a ARN/efectos de los fármacos , Mutaciones Letales Sintéticas/efectos de los fármacos , Mutaciones Letales Sintéticas/genética
20.
Exp Hematol ; 67: 1-9, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30075295

RESUMEN

Acute lymphoblastic leukemia (ALL) is an aggressive hematologic malignancy characterized by suboptimal outcomes in the adult age group. Recently, a new subtype called Philadelphia (Ph)-like ALL has been described. This subgroup is characterized by high cytokine receptor and tyrosine kinase signaling expression, resulting in kinase activation through stimulation of two main pathways, the ABL and JAK/STAT pathways. The diagnostic method or approach for Ph-like ALL is still not standardized and efforts are ongoing to identify an easy and applicable diagnostic method. Accurate and standard testing approaches are much needed and this will facilitate better understanding of this subgroup, including better estimation of the prevalence and incidence in different age groups and the clinical outcomes of such new entity. Here, we review the currently available diagnostic tools, activated pathways, and different therapeutic approaches used to target this subgroup.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Adolescente , Adulto , Distribución por Edad , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Niño , Preescolar , Ensayos Clínicos como Asunto , Manejo de la Enfermedad , Drogas en Investigación/uso terapéutico , Activación Enzimática , Femenino , Predicción , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/clasificación , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/genética , Proteínas Quinasas/fisiología , Receptores de Citocinas/efectos de los fármacos , Receptores de Citocinas/genética , Receptores de Citocinas/fisiología , Distribución por Sexo , Transducción de Señal/efectos de los fármacos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA