Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Invest New Drugs ; 35(5): 556-565, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28516360

RESUMEN

Advances in the understanding of the molecular basis for acute myeloid leukemia (AML) have generated new potential targets for treatment. Fms-like tyrosine kinase 3 (FLT3) is one of the most frequently mutated genes in AML and mutations in this gene are associated with poor overall survival. AXL plays a role in the activation of FLT3 and has been implicated in the pathogenesis of AML. The studies reported here evaluated the ability of a novel FLT3/AXL inhibitor, gilteritinib, to block mutated FLT3 in cellular and animal models of AML. Initial kinase studies showed that gilteritinib, a type I tyrosine kinase inhibitor, was highly selective for both FLT3 and AXL while having weak activity against c-KIT. Gilteritinib demonstrated potent inhibitory activity against the internal tandem duplication (FLT3-ITD) and FLT3-D835Y point mutations in cellular assays using MV4-11 and MOLM-13 cells as well as Ba/F3 cells expressing mutated FLT3. Gilteritinib also inhibited FLT3-F691 mutations, although to a lesser degree, in these assays. Furthermore, gilteritinib decreased the phosphorylation levels of FLT3 and its downstream targets in both cellular and animal models. In vivo, gilteritinib was distributed at high levels in xenografted tumors after oral administration. The decreased FLT3 activity and high intratumor distribution of gilteritinib translated to tumor regression and improved survival in xenograft and intra-bone marrow transplantation models of FLT3-driven AML. No overt toxicity was seen in mouse models treated with gilteritinib. These results indicate that gilteritinib may be an important next-generation FLT3 inhibitor for use in the treatment of FLT3 mutation-positive AML.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Pirazinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Desnudos , Mutación/efectos de los fármacos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Tirosina Quinasa del Receptor Axl
2.
Eur J Immunol ; 41(1): 67-75, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21182078

RESUMEN

Galectins comprise a family of animal lectins that differ in their affinity for ß-galactosides. Galectin-9 (Gal-9) is a tandem-repeat-type galectin that was recently shown to function as a ligand for T-cell immunoglobin domain and mucin domain-3 (Tim-3) expressed on terminally differentiated CD4(+) Th1 cells. Gal-9 modulates immune reactions, including the induction of apoptosis in Th1 cells. In this study, we investigated the effects of Gal-9 in murine models of acute GVH disease (aGVHD). First, we demonstrated that recombinant human Gal-9 inhibit MLR in a dose-dependent manner, involving both Ca(2+) influx and apoptosis in T cells. Next, we revealed that recombinant human Gal-9 significantly inhibit the progression of aGVHD in murine BM transplantation models. In conclusion, Gal-9 ameliorates aGVHD, possibly by inducing T-cell apoptosis, suggesting that gal-9 may be an attractive candidate for the treatment of aGVHD.


Asunto(s)
Apoptosis , Galectinas/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Proteínas Recombinantes/uso terapéutico , Enfermedad Aguda , Animales , Canales de Calcio/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología
3.
Blood ; 116(12): 2089-95, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20548094

RESUMEN

Despite promising clinical results from imatinib mesylate and second-generation ABL tyrosine kinase inhibitors (TKIs) for most BCR-ABL(+) leukemia, BCR-ABL harboring the mutation of threonine 315 to isoleucine (BCR-ABL/T315I) is not targeted by any of these agents. We describe the in vitro and in vivo effects of AT9283 (1-cyclopropyl-3[5-morpholin-4yl methyl-1H-benzomidazol-2-yl]-urea), a potent inhibitor of several protein kinases, including Aurora A, Aurora B, Janus kinase 2 (JAK2), JAK3, and ABL on diverse imatinib-resistant BCR-ABL(+) cells. AT9283 showed potent antiproliferative activity on cells transformed by wild-type BCR-ABL and BCR-ABL/T315I. AT9283 inhibited proliferation in a panel of BaF3 and human BCR-ABL(+) cell lines both sensitive and resistant to imatinib because of a variety of mechanisms. In BCR-ABL(+) cells, we confirmed inhibition of substrates of both BCR-ABL (signal transducer and activator of transcription-5) and Aurora B (histone H3) at physiologically achievable concentrations. The in vivo effects of AT9283 were examined in several mouse models engrafted either subcutaneously or intravenously with BaF3/BCR-ABL, human BCR-ABL(+) cell lines, or primary patient samples expressing BCR-ABL/T315I or glutamic acid 255 to lysine, another imatinib-resistant mutation. These data together support further clinical investigation of AT9283 in patients with imatinib- and second-generation ABL TKI-resistant BCR-ABL(+) cells, including T315I.


Asunto(s)
Bencimidazoles/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Urea/análogos & derivados , Animales , Antineoplásicos , Benzamidas , Bencimidazoles/uso terapéutico , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/farmacología , Urea/farmacología , Urea/uso terapéutico
4.
Cancer Sci ; 102(3): 591-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21166958

RESUMEN

Treatment with Abl tyrosine kinase inhibitors (TKI) drastically improves the prognosis of chronic myelogenous leukemia (CML) patients. However, quiescent CML cells are insensitive to TKI and can lead to relapse of the disease. Thus, research is needed to elucidate the properties of these quiescent CML cells, including their microenvironment, in order to effectively target them. Hypoxia is known to be a common feature of solid tumors that contributes to therapeutic resistance. Leukemic cells are also able to survive and proliferate in severely hypoxic environments. The hypoxic conditions in the bone marrow (BM) allow leukemic cells that reside there to become insensitive to cell death stimuli. To target leukemic cells in hypoxic conditions, we focused on the hypoxia-selective cytotoxin, Rakicidin A. A previous report showed that Rakicidin A, a natural product produced by the Micromonospora strain, induced hypoxia-selective cytotoxicity in solid tumors. Here, we describe Rakicidin A-induced cell death in hypoxia-adapted (HA)-CML cells with stem cell-like characteristics. Interestingly, apoptosis was induced via caspase-dependent and -independent pathways. In addition, treatment with Rakicidin A in combination with the TKI, imatinib, resulted in synergistic cytotoxicity against HA-CML cells. In conclusion, Rakicidin A is a promising compound for targeting TKI-resistant quiescent CML stem cells in the hypoxic BM environment.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Hipoxia de la Célula , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Lipopéptidos/farmacología , Péptidos Cíclicos/farmacología , Adaptación Fisiológica , Caspasa 3/metabolismo , Resistencia a Antineoplásicos , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología
5.
Clin Cancer Res ; 15(8): 2731-8, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19351774

RESUMEN

PURPOSE: beta-catenin is the downstream effector of the Wnt signaling pathway, and it regulates cell proliferation. beta-catenin overexpression correlates positively with prognosis in several types of malignancies. We herein assessed its effects on growth of multiple myeloma cells using a xenograft model. EXPERIMENTAL DESIGN: We first investigated the expression of beta-catenin in multiple myeloma cell lines and multiple myeloma cells obtained from patients. Next, we investigated the growth inhibitory effects of beta-catenin small interfering RNA on the growth of multiple myeloma cells in vivo. Six-week-old male BALB/c nu/nu mice were inoculated s.c. in the right flank with 5 x 10(6) RPMI8226 cells, followed by s.c. injections of beta-catenin small interfering RNA, scramble small interfering RNA, or PBS/atelocollagen complex twice a week for a total of eight injections. RESULTS: Significantly higher levels of beta-catenin expression were observed in multiple myeloma cell lines and in samples from patients with multiple myeloma than those found in mononuclear cells obtained from healthy volunteers. In in vivo experiments, no inhibitory effects were observed following treatment with scramble small interfering RNA or PBS/atelocollagen complexes, whereas treatment with beta-catenin small interfering RNA/atelocollagen complex significantly inhibited growth of multiple myeloma tumors (P < 0.05). CONCLUSIONS: beta-catenin small interfering RNA treatment inhibited the growth of multiple myeloma tumors in a xenograft model. To our knowledge, this is the first report showing that the treatment with beta-catenin small interfering RNA produces an inhibitory effects on growth of hematologic malignancies in vivo. Because treatment with beta-catenin small interfering RNA inhibited growth of multiple myeloma cells, beta-catenin is the attractive novel target for treating multiple myeloma.


Asunto(s)
Mieloma Múltiple/metabolismo , ARN Interferente Pequeño/genética , beta Catenina/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mieloma Múltiple/patología , Trasplante Heterólogo/patología , beta Catenina/genética
6.
Mol Cancer Ther ; 7(9): 2904-12, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18790771

RESUMEN

Liver metastasis is one of the most important prognostic factors in lung cancer patients. However, current therapies are not sufficient. RNA interference provides us a powerful and promising approach for treating human diseases including cancers. Herein, we investigated the in vitro effects of PLK-1 small interfering RNA (siRNA) on human lung cancer cell lines and the in vivo usage of PLK-1 siRNA with atelocollagen as a drug delivery system in a murine liver metastasis model of lung cancer. PLK-1 was overexpressed in cell lines and in cancerous tissues from lung cancer patients. PLK-1 siRNA treatment inhibited growth and induced apoptosis in a concentration-dependent manner. To verify in vivo efficacy, we confirmed that atelocollagen was a useful drug delivery system in our model of implanted luciferase-labeled A549LUC cells by detecting reduced bioluminescence after an i.v. injection of luciferase GL3 siRNA/atelocollagen. PLK-1 siRNA/atelocollagen was also successfully transfected into cells and inhibited the progression of metastases. This study shows the efficacy of i.v. administration of PLK-1 siRNA/atelocollagen for liver metastases of lung cancer. We believe siRNA therapy will be a powerful and promising strategy against advanced lung cancer.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Colágeno/farmacología , Neoplasias Hepáticas/prevención & control , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , Anciano , Animales , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colágeno/administración & dosificación , Femenino , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/ultraestructura , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
7.
Br J Haematol ; 140(2): 181-90, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18028486

RESUMEN

The effect of ABT-737, a BH3-mimicking inhibitor for anti-apoptotic Bcl-2 and Bcl-X(L), but not Mcl-1, against Bcr-Abl-positive (Bcr-Abl(+)) leukaemic cells was examined. ABT-737 potently induced apoptosis in Bcr-Abl(+) chronic myeloid leukaemia (CML) cell lines and primary CML samples in vitro and prolonged the survival of mice xenografted with BV173 cells, a CML cell line. Higher expression of anti-apoptotic Bcl-2 proteins reduced cell killing by ABT-737 in each cell line, but there was no correlation between the sensitivities to ABT-737 and the specific expression patterns of Bcl-2 family proteins among cell lines. Thus, the cell killing effect of ABT-737 must be determined not only by the expression patterns of Bcl-2 family proteins but also by other mechanisms, such as high expression of Bcr-Abl, or a drug-efflux pump, in CML cells. ABT-737 augmented the cell killing effect of imatinib in Bcr-Abl(+) cells with diverse drug-resistance mechanisms unless leukaemic cells harboured imatinib-insensitive Abl kinase domain mutations, such as T315I. The combination of homoharringtonine that reduces Mcl-1 enhanced the killing by ABT-737 strongly in Bcr-Abl(+) cells even with T315I mutation. These results suggest that ABT-737 is a useful component of chemotherapies for CML with diverse drug-resistance mechanisms.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Resistencia a Antineoplásicos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Nitrofenoles/farmacología , Sulfonamidas/farmacología , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/uso terapéutico , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Harringtoninas/farmacología , Homoharringtonina , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Nitrofenoles/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Análisis de Supervivencia , Trasplante Heterólogo , Células Tumorales Cultivadas
8.
Leuk Res ; 32(9): 1462-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18328559

RESUMEN

JAK2V617F is a common mutation in chronic myeloproliferative diseases (CMPDs). We have developed a system utilizing JAK2V617F-specific guanine quenching probe (QP-system) to detect JAK2V617F. With QP-system, results can be obtained from 100 microl of blood within 90 min. We compared QP-system with direct sequencing using 42 CMPD patients' specimens. JAK2V617F was detected in 25 specimens by QP-system, while direct sequencing failed to detect JAK2V617F in 7 of those 25. The presence of JAK2V617F mutation in these 7 specimens was confirmed by allele-specific PCR. These findings indicate that QP-system is more sensitive and useful than direct sequencing for diagnoses of CMPDs.


Asunto(s)
Janus Quinasa 2/genética , Trastornos Mieloproliferativos/genética , Mutación Puntual , Adulto , Anciano , Anciano de 80 o más Años , Automatización , Enfermedad Crónica , Análisis Mutacional de ADN , Cartilla de ADN , Femenino , Humanos , Janus Quinasa 2/sangre , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/sangre , Reacción en Cadena de la Polimerasa
9.
Int J Hematol ; 87(5): 507-515, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18415656

RESUMEN

Since a variety of cell intrinsic and extrinsic molecular abnormalities cooperatively promote tumor formation in multiple myeloma (MM), therapeutic approaches that concomitantly target more than one molecule are increasingly attractive. We herein demonstrate the anti-myeloma effect of a cephalotaxus alkaloid, homoharringtonine (HHT), an inhibitor of protein synthesis, through the induction of apoptosis. HHT significantly reduced Mcl-1, a crucial protein involved in myeloma cell survival, in all three myeloma cell lines examined, whereas certain BH3-only proteins, such as Bim, Bik, and Puma, remained unchanged following HHT treatment, and their expression levels depended on the cell type. HHT also reduced the levels of c-FLIP(L/S), activated caspase-8, and induced active truncated-Bid. Thus, HHT-induced apoptosis appears to be mediated via both intrinsic and extrinsic apoptosis pathways, and the resultant imbalance between BH3-only proteins and Mcl-1 may be pivotal for apoptosis by HHT. In addition, HHT treatment resulted in reduced levels of beta-catenin and XIAP proteins, which also contribute to disease progression and resistance to chemotherapy in MM. In combination, HHT enhanced the effects of melphalan, bortezomib, and ABT-737. These results suggest that HHT could constitute an attractive option for MM treatment though its ability to simultaneously target multiple tumor-promoting molecules.


Asunto(s)
Antineoplásicos/farmacología , Harringtoninas/farmacología , Mieloma Múltiple/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteína Inhibidora de la Apoptosis Ligada a X/biosíntesis , beta Catenina/biosíntesis , Antineoplásicos/agonistas , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Harringtoninas/agonistas , Homoharringtonina , Humanos , Mieloma Múltiple/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Inhibidores de la Síntesis de la Proteína/agonistas
10.
Acta Haematol ; 120(3): 177-81, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19129688

RESUMEN

Overlapping of essential thrombocythemia (ET) and multiple myeloma (MM) has been extremely rare. Our report concerns a case with concomitant ET and MM, where JAK2V617F was present in non-myeloma peripheral blood leukocytes and bone marrow (BM) hematopoietic cells, but not in BM-derived CD138-positive myeloma cells. In contrast, double-color fluorescence in situ hybridization analysis showed that BM-derived CD138-positive myeloma cells possessed the gene translocation between the immunoglobulin heavy chain gene and the cyclin D1 gene, which was not involved in non-myeloma hematopoietic cells. This is the first case with concomitant ET and MM in which the 2 hematologic neoplasms were shown to have originated from separate malignant clones at hierarchically different differentiation levels resulting from independent acquisition of different molecular aberrations. Among the 10 reported cases, including ours, ET preceded MM in 8 cases, but MM never preceded ET. We suggest that MM clones may have greater proliferative potency compared with ET clones, and that the treatment modification from ET to MM did not seem to exacerbate ET in most reported cases, perhaps because of the suppression of the ET clone by both the cytotoxic effect of anti-myeloma therapy and the clonal repression by MM progression.


Asunto(s)
Ciclina D1/genética , Cadenas Pesadas de Inmunoglobulina/genética , Janus Quinasa 2/genética , Mieloma Múltiple/genética , Mutación Missense , Trombocitopenia/genética , Translocación Genética , Anciano , Sustitución de Aminoácidos , Ciclina D1/metabolismo , Femenino , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Cadenas Pesadas de Inmunoglobulina/metabolismo , Janus Quinasa 2/metabolismo , Leucocitos/metabolismo , Leucocitos/patología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Sindecano-1/metabolismo , Trombocitopenia/metabolismo , Trombocitopenia/patología
11.
Cancer Lett ; 312(1): 91-100, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21906872

RESUMEN

We investigated the effect of a novel Wnt/ß-catenin signaling inhibitor, AV65 on imatinib mesylate (IM)-sensitive and -resistant human chronic myeloid leukemia (CML) cells in vitro. AV65 inhibited the proliferation of various CML cell lines including T315I mutation-harboring cells. AV65 reduced the expression of ß-catenin in CML cells, resulting in the induction of apoptosis. Moreover, AV65 inhibited the proliferation of hypoxia-adapted primitive CML cells that overexpress ß-catenin. The combination of AV65 with IM had a synergistic inhibitory effect on the proliferation of CML cells. These findings suggest that AV65 could be a novel therapeutic agent for the treatment of CML.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Mutación , Piperazinas/farmacología , Pirimidinas/farmacología , Proteínas Wnt/antagonistas & inhibidores , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Benzamidas , Procesos de Crecimiento Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Fase G1/efectos de los fármacos , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Fase S/efectos de los fármacos , Proteínas Wnt/metabolismo , beta Catenina/biosíntesis , beta Catenina/metabolismo
12.
Cancer Lett ; 312(2): 228-34, 2011 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21890264

RESUMEN

ABL tyrosine kinase inhibitor (TKI), imatinib is used for BCR-ABL(+) leukemias. We developed an automatic method utilizing guanine-quenching probes (QP) to detect 17 kinds of mutations frequently observed in imatinib-resistance. Results were obtained from 100µL of whole blood within 90min by this method. Detected mutations were almost identical between QP method and direct sequencing. Furthermore, the mutation-biased PCR (MBP) was added to the QP method to increase sensitivity, resulting earlier detection of T315I mutation which was insensitive to any ABL TKIs. Thus, the QP and MBP-QP may become useful methods for the management of ABL TKI-treated patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Automatización , Mutación , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-abl/genética , Pirimidinas/uso terapéutico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Benzamidas , Niño , Preescolar , Resistencia a Antineoplásicos , Femenino , Humanos , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Adulto Joven
13.
Leuk Res ; 34(6): 793-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19781772

RESUMEN

Osteoclasts (OCs) are specialized cells for the resorption of bone matrix that have also been recently reported to be involved in the mobilization of hematopoietic progenitor cells. When Ba/F3 cells expressing wild-type bcr-abl were co-cultured with osteoblasts (OBs), OCs, and bone slices, their proliferation was significantly suppressed, and the Ki-67 negative population, which is believed to be in G(0) phase, was increased. The results of our in vitro experiments suggest that OCs could be involved in the maintenance of dormant leukemic cells in the bone marrow (BM) microenvironment through the release of soluble factors, one of which could be TGF-beta.


Asunto(s)
Proliferación Celular , Leucemia/patología , Leucemia/fisiopatología , Osteoclastos/fisiología , Animales , Animales Recién Nacidos , Huesos/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Ciclo Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Leucemia/genética , Leucemia/metabolismo , Ratones , Ratones Endogámicos BALB C , Osteoblastos/metabolismo , Osteoblastos/fisiología , Osteoclastos/metabolismo , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/patología , Factor de Crecimiento Transformador beta1/farmacología
14.
Cancer Lett ; 294(2): 245-53, 2010 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-20206440

RESUMEN

Although novel agents effective against malignant mesothelioma (MM) have been developed, the prognosis of patients with MM is still poor. We generated a DNA-chimeric siRNA against polo-like kinase-1 (PLK-1), which was more stable in human serum than the non-chimeric siRNA. The chimeric PLK-1 siRNA inhibited MM cell proliferation through the induction of apoptosis. Next, we investigated the effects of zoledronic acid (ZOL) on MM cells, and found that ZOL also induced apoptosis in MM cells. Furthermore, ZOL augmented the inhibitory effects of the PLK-1 siRNA. In conclusion, combining a PLK-1 siRNA with ZOL treatment is an attractive strategy against MM.


Asunto(s)
Proteínas de Ciclo Celular/genética , ADN/genética , Difosfonatos/farmacología , Imidazoles/farmacología , Mesotelioma/terapia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/biosíntesis , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Quimera/genética , Terapia Combinada , ADN/sangre , Terapia Genética/métodos , Humanos , Mesotelioma/enzimología , Mesotelioma/genética , Mesotelioma/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/biosíntesis , ARN Interferente Pequeño/sangre , ARN Interferente Pequeño/genética , Transfección , Ácido Zoledrónico , Quinasa Tipo Polo 1
15.
Expert Rev Anticancer Ther ; 8(9): 1387-98, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18759691

RESUMEN

Treatment of chronic myeloid leukemia (CML) has changed drastically with the emergence of the Abl tyrosine kinase inhibitor (TKI), imatinib mesylate. However, primary and secondary resistance have frequently been reported, particularly in patients with advanced-stage disease. Point mutations within the Abl kinase domain that interfere with imatinib binding are the most critical cause of imatinib resistance. In order to override this resistance, several second generation ATP-competitive Abl TKIs including dasatinib, nilotinib, bosutinib and INNO-406 have been developed. Despite promising clinical results from these novel Abl TKIs for most mutations, the frequently observed mutant T315I is not effectively targeted by any of these agents. Thus, identification of novel agents and the development of new strategies for the effective treatment of CML patients with the T315I mutation are important and challenging tasks. In this review, the current status of novel agents for CML treatment is overviewed as follows: pathogenesis and features of CML; imatinib and second-generation Abl TKIs; why Abl TKIs are not effective against T315I; and novel agents that may override the T315I mutation.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas de Fusión bcr-abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Benzamidas , Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Piperazinas/uso terapéutico , Mutación Puntual , Pirimidinas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA