Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 629
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 560(7716): 55-60, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30022166

RESUMEN

Acute lymphoblastic leukaemia (ALL) has a marked propensity to metastasize to the central nervous system (CNS). In contrast to brain metastases from solid tumours, metastases of ALL seldom involve the parenchyma but are isolated to the leptomeninges, which is an infrequent site for carcinomatous invasion. Although metastasis to the CNS occurs across all subtypes of ALL, a unifying mechanism for invasion has not yet been determined. Here we show that ALL cells in the circulation are unable to breach the blood-brain barrier in mice; instead, they migrate into the CNS along vessels that pass directly between vertebral or calvarial bone marrow and the subarachnoid space. The basement membrane of these bridging vessels is enriched in laminin, which is known to coordinate pathfinding of neuronal progenitor cells in the CNS. The laminin receptor α6 integrin is expressed in most cases of ALL. We found that α6 integrin-laminin interactions mediated the migration of ALL cells towards the cerebrospinal fluid in vitro. Mice with ALL xenografts were treated with either a PI3Kδ inhibitor, which decreased α6 integrin expression on ALL cells, or specific α6 integrin-neutralizing antibodies and showed significant reductions in ALL transit along bridging vessels, blast counts in the cerebrospinal fluid and CNS disease symptoms despite minimally decreased bone marrow disease burden. Our data suggest that α6 integrin expression, which is common in ALL, allows cells to use neural migratory pathways to invade the CNS.


Asunto(s)
Sistema Nervioso Central/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Animales , Anticuerpos Neutralizantes/inmunología , Membrana Basal/metabolismo , Barrera Hematoencefálica/metabolismo , Médula Ósea , Movimiento Celular , Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/metabolismo , Líquido Cefalorraquídeo/metabolismo , Circulación Cerebrovascular , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Progresión de la Enfermedad , Femenino , Xenoinjertos/inmunología , Xenoinjertos/patología , Integrina alfa6/inmunología , Integrina alfa6/metabolismo , Laminina/metabolismo , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Laminina/antagonistas & inhibidores , Receptores de Laminina/inmunología , Receptores de Laminina/metabolismo , Cráneo , Espacio Subaracnoideo
2.
Blood ; 137(7): 939-944, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32898857

RESUMEN

Blinatumomab is currently approved for use as a single agent in relapsed and refractory acute lymphoblastic leukemia (ALL). Cytotoxicity is mediated via signaling through the T-cell receptor (TCR). There is now much interest in combining blinatumomab with targeted therapies, particularly in Philadelphia chromosome-positive ALL (Ph+ ALL). However, some second- and third-generation ABL inhibitors also potently inhibit Src family kinases that are important in TCR signaling. We combined ABL inhibitors and dual Src/ABL inhibitors with blinatumomab in vitro from both healthy donor samples and primary samples from patients with Ph+ ALL. Blinatumomab alone led to both T-cell proliferation and elimination of target CD19+ cells and enhanced production of interferon-γ (IFN-γ). The addition of the ABL inhibitors imatinib or nilotinib to blinatumomab did not inhibit T-cell proliferation or IFN-γ production. However, the addition of dasatinib or ponatinib inhibited T-cell proliferation and IFN-γ production. Importantly, there was no loss of CD19+ cells treated with blinatumomab plus dasatinib or ponatinib in healthy samples or samples with a resistant ABL T315I mutation by dasatinib in combination with blinatumomab. These in vitro findings bring pause to the excitement of combination therapies, highlighting the importance of maintaining T-cell function with targeted therapies.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Activación de Linfocitos/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Linfocitos T/inmunología , Familia-src Quinasas/antagonistas & inhibidores , Linfocitos B , Dasatinib/farmacología , Humanos , Mesilato de Imatinib/farmacología , Imidazoles/farmacología , Ensayos de Liberación de Interferón gamma , Células Jurkat , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Mutación Missense , Proteínas de Neoplasias/fisiología , Fosforilación/efectos de los fármacos , Mutación Puntual , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-abl/genética , Piridazinas/farmacología , Pirimidinas/farmacología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/enzimología , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Familia-src Quinasas/fisiología
3.
Blood ; 133(21): 2263-2268, 2019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-30910786

RESUMEN

Mutations in the cytosolic 5' nucleotidase II (NT5C2) gene drive resistance to thiopurine chemotherapy in relapsed acute lymphoblastic leukemia (ALL). Mechanistically, NT5C2 mutant proteins have increased nucleotidase activity as a result of altered activating and autoregulatory switch-off mechanisms. Leukemias with NT5C2 mutations are chemoresistant to 6-mercaptopurine yet show impaired proliferation and self-renewal. Direct targeting of NT5C2 or inhibition of compensatory pathways active in NT5C2 mutant cells may antagonize the emergence of NT5C2 mutant clones driving resistance and relapse in ALL.


Asunto(s)
5'-Nucleotidasa , Proliferación Celular , Mercaptopurina/uso terapéutico , Mutación , Proteínas de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
4.
Int J Mol Sci ; 22(22)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34830095

RESUMEN

Compared to other ethnicities, Hispanic children incur the highest rates of leukemia, and most cases are diagnosed as Acute Lymphoblastic Leukemia (ALL). Despite improved treatment and survival for ALL, disproportionate health outcomes in Hispanics persist. Thus, it is essential to identify oncogenic mutations within this demographic to aid in the development of new strategies to diagnose and treat ALL. Using whole-exome sequencing, five single nucleotide polymorphisms within mitogen-activated protein kinase 3 (MAP2K3) were identified in an ALL cancer patient library from the U.S./Mexico border. MAP2K3 R26T and P11T are located near the substrate-binding site, while R65L and R67W localized to the kinase domain. Truncated-MAP2K3 mutant Q73* was also identified. Transfection in HEK293 cells showed that the quadruple-MEK3 mutant (4M-MEK3) impacted protein stability, inducing degradation and reducing expression. The expression of 4M-MEK3 could be rescued by cysteine/serine protease inhibition, and proteasomal degradation of truncated-MEK3 occurred in a ubiquitin-independent manner. MEK3 mutants displayed reduced auto-phosphorylation and enzymatic activity, as seen by decreases in p38 phosphorylation. Furthermore, uncoupling of the MEK3/p38 signaling pathway resulted in less suppressive activity on HEK293 cell viability. Thus, disruption of MEK3 activation may promote proliferative signals in ALL. These findings suggest that MEK3 represents a potential therapeutic target for treating ALL.


Asunto(s)
Proliferación Celular/genética , MAP Quinasa Quinasa 3 , Sistema de Señalización de MAP Quinasas/genética , Mutación , Proteínas de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras , Proteolisis , Células HEK293 , Células Hep G2 , Humanos , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
5.
Cancer Metastasis Rev ; 38(4): 829-837, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31802358

RESUMEN

Matrix metalloproteinases (MMPs) play a major role in extracellular matrix remodeling and are involved in tumor cell invasion. Cancers such as childhood leukemia are characterized by their capacity to infiltrate different organs. MMP production by leukemic cells may indicate a leukemic subtype or subpopulation with a more invasive phenotype. Therefore, clarifying the action mechanisms of MMPs as prognostic predictors or MMP targeting as a therapeutic strategy is necessary. MMP-targeting drugs have been developed for the treatment of hematological malignancies. In this review, we highlight current advances in understanding the molecular mechanisms and pathological characteristics of various MMPs, as well as recent therapeutic advances targeting MMPs in childhood leukemia. Several studies have been conducted on the therapeutic efficacy of MMP inhibitors in cancer, such as collagen peptidomimetics, nonpeptidomimetic inhibitors of MMP active sites, bisphosphonates, and tetracycline derivatives. Here, we conclude that more clinical trials are necessary to estimate the role of selective MMP inhibitors in the treatment and prevention of childhood leukemia.


Asunto(s)
Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/terapia , Metaloproteinasas de la Matriz/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Animales , Niño , Humanos
6.
Blood ; 131(13): 1464-1475, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29348129

RESUMEN

We investigated the role of copy number alterations to refine risk stratification in adult Philadelphia chromosome positive (Ph)+ acute lymphoblastic leukemia (ALL) treated with tyrosine kinase inhibitors (TKIs) and allogeneic stem cell transplantation (aSCT). Ninety-seven Ph+ ALL patients (median age 41 years; range 18-64 years) within the prospective multicenter German Multicenter ALL Study Group studies 06/99 (n = 8) and 07/2003 (n = 89) were analyzed. All patients received TKI and aSCT in first complete remission (CR1). Copy number analysis was performed with single nucleotide polymorphism arrays and validated by multiplex ligation-dependent probe amplification. The frequencies of recurrently deleted genes were: IKZF1, 76%; CDKN2A/2B, 45%; PAX5, 43%; BTG1, 18%; EBF1, 13%; ETV6, 5%; RB, 14%. In univariate analyses, the presence of CDKN2A/2B deletions had a negative impact on all endpoints: overall survival (P = .023), disease-free survival (P = .012), and remission duration (P = .036). The negative predictive value of CDKN2A/2B deletions was retained in multivariable analysis along with other factors such as timing of TKI therapy, intensity of conditioning, achieving remission after induction phase 1 and BTG1 deletions. We therefore conclude that acquired genomic CDKN2A/2B deletions identify a subgroup of Ph+ ALL patients, who have an inferior prognosis despite aSCT in CR1. Their poor outcome was attributable primarily to a high relapse rate after aSCT.


Asunto(s)
Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Eliminación de Gen , Trasplante de Células Madre Hematopoyéticas , Cromosoma Filadelfia , Leucemia-Linfoma Linfoblástico de Células Precursoras , Inhibidores de Proteínas Quinasas/administración & dosificación , Acondicionamiento Pretrasplante , Adolescente , Adulto , Aloinjertos , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Estudios Prospectivos , Tasa de Supervivencia
7.
Cancer Invest ; 38(8-9): 463-475, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32772580

RESUMEN

In the present study, we searched selective cytotoxicity and mitochondria mediated apoptosis of novel COX-2 inhibitor 2-(4-(Methylsulfonyl)phenyl)imidazo[1,2-a] pyridine-8-carboxylic acid on B-lymphocytes and their mitochondria isolated from normal subjects and acute lymphoblastic leukemia (ALL) patients' blood. Our results showed this compound can selectively induce cellular and mitochondrial toxicity on ALL B-lymphocytes and mitochondria without any toxic effects on normal B-lymphocytes and their mitochondria. Taken together, the results of this study suggest that cancerous mitochondria are a potential target for the ALL B-lymphocytes. Selective toxicity of COX-2 inhibitor in cancerous mitochondria could be an attractive therapeutic option for the effective clinical management of therapy-resistant ALL.


Asunto(s)
Linfocitos B/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Mitocondrias/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Linfocitos B/patología , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Niño , Preescolar , Citocromos c/metabolismo , Femenino , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/patología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/enzimología , Membranas Mitocondriales/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Especies Reactivas de Oxígeno/metabolismo , Succinato Deshidrogenasa/metabolismo
8.
Pediatr Blood Cancer ; 67(7): e28244, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32323890

RESUMEN

It is unclear if dosing intervals for Erwinase can be extended with intramuscular (i.m.) versus intravenous (i.v.) dosing. Children with acute lymphoblastic leukemia received Erwinase at 30 000-42 000 IU/m2 i.v. or i.m. I.m. Erwinase (n = 22) achieved activity above 0.1 IU/mL for longer than i.v. Erwinase (n = 33) (3.4 vs 2.9 days, P = 0.0007). With 30 000 IU/m2 Monday, Wednesday, Friday, more patients achieved adequate concentrations over the weekend with i.m. vs i.v. dosing (P = 5 × 10-36 ). A schedule with i.v. doses on Monday and Wednesday and i.m. doses on Friday of 30 000 IU/m2 maintained activity > 0.1 IU/mL over the weekend in 80% of patients.


Asunto(s)
Asparaginasa/administración & dosificación , Asparaginasa/sangre , Erwinia/enzimología , Inyecciones Intramusculares/métodos , Inyecciones Intravenosas/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Niño , Estudios de Seguimiento , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
9.
Pediatr Blood Cancer ; 67(2): e28044, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31625674

RESUMEN

We undertook this retrospective study to describe decisions made following asparaginase activity monitoring implementation at our center. Clinically apparent reactions (CARs) and asparaginase activity monitoring costs were described. Patients with acute lymphoblastic leukemia, aged <18 years who received asparaginase between April 2016 and September 2017, were included. Decisions made following receipt of asparaginase activity results were categorized as continuation, modification, premedication, or discontinuation. We included 129 patients (median age: 5.33 years) receiving 565 asparaginase doses. CARs were observed following 25 asparaginase doses (19/361 [5.3%] pegaspargase). A total of 224 asparaginase activity levels were ordered in 88 patients. Following receipt of 190 asparaginase activity results, asparaginase therapy was continued, modified, or premedicated in 188 (98.9%), 1 (0.005%), and 1 (0.005%) cases, respectively. Inadequate asparaginase activity was observed in three patients receiving Erwinia asparaginase. Asparaginase activity monitoring allowed patients with pegaspargase-associated CAR and adequate activity to continue therapy unchanged and was cost neutral.


Asunto(s)
Antineoplásicos/uso terapéutico , Asparaginasa/metabolismo , Biomarcadores de Tumor/análisis , Toma de Decisiones Clínicas , Polietilenglicoles/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Asparaginasa/uso terapéutico , Canadá/epidemiología , Niño , Preescolar , Hipersensibilidad a las Drogas/tratamiento farmacológico , Hipersensibilidad a las Drogas/epidemiología , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico , Estudios Retrospectivos
10.
Acta Haematol ; 143(3): 217-231, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31590170

RESUMEN

Treatment of chronic myeloid leukemia (CML) and Philadelphia chromosome-positive acute leukemia (Ph+ ALL) has been revolutionized with the advent of tyrosine kinase inhibitors (TKIs). Most patients with CML achieve long-term survival similar to individuals without CML due to treatment with TKIs not only in frontline but also in further lines of therapy. The third-generation TKI ponatinib has demonstrated efficacy in patients with refractory CML and Ph+ ALL. Ponatinib is currently the most potent TKI in this setting demonstrating activity against T315I mutant clones. However, ponatinib's safety data revealed a dose-dependent, increased risk of serious cardiovascular (CV) events. Guidance is needed to evaluate the benefit-risk profile of TKIs, such as ponatinib, and safety measures to prevent treatment-associated CV events. An expert panel of German hematologists and cardiologists summarize current evidence regarding ponatinib's efficacy and CV safety profile. We propose CV management strategies for patients who are candidates for ponatinib.


Asunto(s)
Antineoplásicos/uso terapéutico , Enfermedades Cardiovasculares/inducido químicamente , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Imidazoles/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridazinas/uso terapéutico , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/prevención & control , Ensayos Clínicos como Asunto , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Femenino , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/tratamiento farmacológico , Hiperlipidemias/complicaciones , Hiperlipidemias/tratamiento farmacológico , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Imidazoles/administración & dosificación , Imidazoles/efectos adversos , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Masculino , Persona de Mediana Edad , Cromosoma Filadelfia , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Supervivencia sin Progresión , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Piridazinas/administración & dosificación , Piridazinas/efectos adversos , Medición de Riesgo
11.
Nature ; 514(7523): 513-7, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25132549

RESUMEN

T-cell acute lymphoblastic leukaemia (T-ALL) is a haematological malignancy with a dismal overall prognosis, including a relapse rate of up to 25%, mainly because of the lack of non-cytotoxic targeted therapy options. Drugs that target the function of key epigenetic factors have been approved in the context of haematopoietic disorders, and mutations that affect chromatin modulators in a variety of leukaemias have recently been identified; however, 'epigenetic' drugs are not currently used for T-ALL treatment. Recently, we described that the polycomb repressive complex 2 (PRC2) has a tumour-suppressor role in T-ALL. Here we delineated the role of the histone 3 lysine 27 (H3K27) demethylases JMJD3 and UTX in T-ALL. We show that JMJD3 is essential for the initiation and maintenance of T-ALL, as it controls important oncogenic gene targets by modulating H3K27 methylation. By contrast, we found that UTX functions as a tumour suppressor and is frequently genetically inactivated in T-ALL. Moreover, we demonstrated that the small molecule inhibitor GSKJ4 (ref. 5) affects T-ALL growth, by targeting JMJD3 activity. These findings show that two proteins with a similar enzymatic function can have opposing roles in the context of the same disease, paving the way for treating haematopoietic malignancies with a new category of epigenetic inhibitors.


Asunto(s)
Histona Demetilasas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Animales , Benzazepinas/farmacología , Epigénesis Genética/efectos de los fármacos , Histona Demetilasas/genética , Histonas/química , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Lisina/metabolismo , Metilación/efectos de los fármacos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pirimidinas/farmacología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
J Cell Biochem ; 120(10): 17791-17810, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31264739

RESUMEN

Chemoresistance often causes treatment failure of B-cell acute lymphoblastic leukemia (B-ALL). However, the mechanism remains unclear at present. Herein, overexpression of heme oxygenase-1 (HO-1) was found in the bone marrow stromal cells (BMSCs) from B-ALL patients developing resistance to vincristine (VCR), a chemotherapeutic agent. Two B-ALL cell lines Super B15 and CCRF-SB were cocultured with BMSCs transfected with lentivirus to regulate the expression of HO-1. Silencing HO-1 expression in BMSCs increased the apoptotic rates of B-ALL cell lines induced by VCR, whereas upregulating HO-1 expression reduced the rate. Cell cycle can be arrested in the G2/M phase by VCR. In contrast, B-ALL cells were arrested in the G0/G1 phase due to HO-1 overexpression in BMSCs, which avoided damage from the G2/M phase. Vascular endothelial growth factor (VEGF) in BMSCs, as a key factor in the microenvironment-associated chemoresistance, was also positively coexpressed with HO-1. VEGF secretion was markedly increased in BMSCs with HO-1 upregulation but decreased in BMSCs with HO-1 silencing. B-ALL cell lines became resistant to VCR when cultured with VEGF recombinant protein, so VEGF secretion induced by HO-1 expression may promote the VCR resistance of B-ALL cells. As to the molecular mechanism, the PI3K/AKT pathway mediated regulation of VEGF by HO-1. In conclusion, this study clarifies a mechanism by which B-ALL is induced to resist VCR through HO-1 overexpression in BMSCs, and provides a novel strategy for overcoming VCR resistance in clinical practice.


Asunto(s)
Resistencia a Antineoplásicos , Hemo-Oxigenasa 1/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vincristina/farmacología , Adolescente , Adulto , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Niño , Preescolar , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Lentivirus/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Adulto Joven
13.
Am J Hematol ; 94(12): 1388-1395, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31595534

RESUMEN

Despite the advances in the management of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) with the introduction of tyrosine kinase inhibitors (TKIs), relapses remain challenging. We reviewed clinical data from adult patients with Ph + ALL who received frontline hyperCVAD chemotherapy with a TKI to determine their outcomes after first relapse. Patients with first morphological relapse after prior complete remission were evaluated for predictors of response and survival. For 57 of 233 (25%) patients, there was morphological relapse after a median of 15.9 months from first remission [range: 5.3-94]. The choice of salvage treatments was at the discretion of the treating physician. So, 43 (75%) patients received a TKI in combination with their salvage treatment. Second remission was achieved in 41 of 49 (84%) evaluable patients. Median relapse free survival (RFS) was 10.5 months [range, 0.2-81]. The 1-year and 2-year overall survival (OS) were 41% and 20% respectively. On multivariate analysis, only elevated LDH (units/L), the use of first-generation or no TKI at the time of first relapse and the achievement of a major molecular response (MMR) had a significant effect on OS (HR: 2.82, 95% CI:1.11-7.16, P = .029; HR = 2.39, 95% CI: 1.07,5.39, P = .034; HR = 0.39, 95% CI: 0.16-0.94, P = .03, respectively). Whereas, only achievement of MMR was significantly prognostic for RFS with a HR of 0.48 (95% CI: 0.23-0.98, P = .04). The OS and RFS were comparable between recipients and non-recipients of allogeneic hematopoietic stem cell transplantation (alloHSCT) at second remission, due to a higher non-relapse mortality (53%) seen in patients who underwent alloHSCT.


Asunto(s)
Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Terapia Molecular Dirigida , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Biespecíficos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Asparaginasa/uso terapéutico , Evolución Clonal , Ciclofosfamida/administración & dosificación , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Supervivencia sin Enfermedad , Doxorrubicina/administración & dosificación , Femenino , Proteínas de Fusión bcr-abl/genética , Genes abl , Humanos , Inotuzumab Ozogamicina/uso terapéutico , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mutación Missense , Cromosoma Filadelfia , Mutación Puntual , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Pronóstico , Modelos de Riesgos Proporcionales , Dominios Proteicos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Terapia Recuperativa , Vincristina/administración & dosificación , Adulto Joven
14.
Pediatr Blood Cancer ; 66(6): e27681, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30848065

RESUMEN

BACKGROUND/OBJECTIVES: Anthracyclines are used in induction therapy of pediatric acute lymphoblastic leukemia (ALL) and are known to generate oxidative stress; whether this translates into enhanced antileukemic activity or hemolytic effects in patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency is unknown. DESIGN/METHODS: Among 726 pediatric patients with newly diagnosed ALL treated at St. Jude Children's Research Hospital, 22 had deficient G6PD activity. We compared the prevalence of positive minimal residual disease (MRD) ≥1% at Day 15/Day 19 of induction or ≥0.01% at Day 42/Day 46 (end of induction) and the number of red blood cell (RBC) transfusions after daunorubicin in induction between patients with or without G6PD deficiency, adjusting for ALL risk group, treatment protocol, age, and gender. RESULTS: There was no difference in Day 15/19 (P = 1) or end of induction MRD (P = 0.76) nor in the number of RBC transfusions (P = 0.73); the lack of association with MRD was confirmed in a dataset of 1192 newly diagnosed male patients enrolled in a Children's Oncology Group trial (P = 0.78). CONCLUSION: We found no evidence that G6PD deficiency affects daunorubicin activity during induction treatment for ALL.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Daunorrubicina/uso terapéutico , Glucosafosfato Deshidrogenasa/metabolismo , Neoplasia Residual/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Niño , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Quimioterapia de Inducción , Masculino , Terapia Neoadyuvante , Neoplasia Residual/tratamiento farmacológico , Neoplasia Residual/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Pronóstico , Factores de Riesgo , Seguridad
15.
Pharmazie ; 74(11): 671-674, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31739835

RESUMEN

Gamma-Glutamyl hydrolase (GGH) plays an important role in the disposition of anti-folate analogs. Several studies noted the pharmacological relevance of rs3758149 C/T polymorphism located in the human GGH promoter. The present study aimed to investigate the role of rs3758149 C/T polymorphism and transcription factors in the regulation of GGH expression in human acute lymphoblastic leukemia (ALL) CEM/C1 cells. Compared with the rs3758149 T allele, the C allele showed significantly higher transcriptional activity in luciferase reporter assays, as well as a stronger binding affinity for the nuclear protein extracts in an electrophoretic mobility shift assay. Sp1 was identified as the target transcription factor that exhibited allele-specific binding to the location of rs3758149 C/T polymorphism in the chromatin immunoprecipitation assay. Overexpression of Sp1 led to enhanced GGH promoter activity and GGH mRNA expression in allele-specific manners. These findings suggested that Sp1 acted as a positive regulator of human GGH transcription through the rs3758149 polymorphism in CEM/C1 cells. This study contributed to the present understanding of the mechanisms underlying variable responses of ALL to anti-folates.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Factor de Transcripción Sp1/metabolismo , gamma-Glutamil Hidrolasa/genética , Alelos , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Regulación Neoplásica de la Expresión Génica , Humanos , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Regiones Promotoras Genéticas
16.
J Appl Microbiol ; 125(4): 1017-1029, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29896797

RESUMEN

AIMS: Matrix metalloproteinase-9 (MMP-9) and survivin are involved in several steps of carcinogenesis in acute lymphoblastic leukaemia (ALL). Yet, no MMP-9 and survivin-modulating drugs with low toxicity on normal cells but high efficacy against high MMP-9- and survivin-expressing leukaemia cells have been approved for clinical application in ALL. Prodigiosin, a secondary metabolite of Serratia marcescens, induces apoptosis in different kinds of cancer cells with low toxicity on normal cells. However, little is known about the effects of this compound on the high MMP-9- and survivin-expressing leukaemia cells. METHODS AND RESULTS: CCRF-CEM cells as a model for high MMP-9- and survivin-expressing ALL cells were treated with 100, 200 and 400 nmol l-1 prodigiosin after which cell number, proliferation rate, MMP-9 and survivin expression, caspase-3 activation and apoptosis were evaluated. After 24-, 48-, and 72-h treatments with 100, 200 and 400 nmol l-1 prodigiosin, proliferation rates were measured to be 92·3-76·7%, 82-63% and 63·7-46·6% respectively. Treatment with prodigiosin for 48 h decreased MMP-9 mRNA levels followed by decreases in secreted (S) and intracellular (I) MMP-9 protein levels by 20-22% and 69-72% for 100-400 nmol l-1 prodigiosin respectively. Prodigiosin decreased survivin protein levels from 40 to 26% followed by 3·7-5·6-fold increases in caspase-3 activation for the aforementioned prodigiosin concentration ranges. Treatment with 100-400 nmol l-1 prodigiosin increased the caspase-3/survivin, caspase-3/I-MMP-9 and caspase-3/S-MMP-9 ratios by 6-7·3-, 11·5-19·1- and 4·9-6·8-fold increases respectively. A dramatic increase in the number of apoptotic cells was also observed with increasing prodigiosin concentrations. CONCLUSION: The inhibitory effects of prodigiosin on MMP-9 and survivin expression, as well as its pro-apoptotic capacity, represent a novel therapeutic avenue against ALL cells. SIGNIFICANCE AND IMPACT OF THE STUDY: These findings provide an important and interesting basis to develop a new therapeutic compound with high potential against ALL cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/fisiopatología , Prodigiosina/farmacología , Serratia marcescens/química , Caspasa 3/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Metaloproteinasa 9 de la Matriz/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Prodigiosina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serratia marcescens/metabolismo , Survivin
17.
Pharmacogenomics J ; 17(3): 274-279, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27019981

RESUMEN

Asparaginase, which depletes asparagine and glutamine, activates amino-acid stress response. Oxidative stress mediated by excessive reactive oxygen species (ROS) causes enhanced mitochondrial permeabilization and subsequent cell apoptosis and is considered as a plausible mechanism for drug-induced hepatotoxicity, a common toxicity of asparaginase in adults with acute lymphoblastic leukemia (ALL). Studies investigating the pharmacogenetics of asparaginase in ALL are limited and focused on asparaginase-induced allergic reaction common in pediatric patients. Here, we sought to determine a potential association between the variant rs4880 in SOD2 gene, a key mitochondrial enzyme that protects cells against ROS, and hepatotoxicity during asparaginase-based therapy in 224 patients enrolled on CALGB-10102, a treatment trial for adults with ALL. We report that the CC genotype of rs4880 is associated with increased hepatotoxicity following asparaginase-based treatment. Thus, rs4880 likely contributes to asparaginase-induced hepatotoxicity, and functional studies investigating this single-nucleotide polymorphism (SNP) are needed to develop therapeutic approaches that mitigate this toxicity.


Asunto(s)
Antineoplásicos/efectos adversos , Asparaginasa/efectos adversos , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Variantes Farmacogenómicas , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Superóxido Dismutasa/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Pruebas de Farmacogenómica , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Medición de Riesgo , Factores de Riesgo , Resultado del Tratamiento , Adulto Joven
18.
Am J Hematol ; 92(1): 7-11, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27673440

RESUMEN

Phosphatidylinositol-3-kinase (PI3K) signaling plays a crucial role in oncogene-mediated tumor growth and proliferation. Buparlisib (BKM120) is an oral pan-class I PI3K inhibitor. This phase I study was conducted to determine the dose limiting toxicity (DLT) and maximum tolerated dose (MTD) of BKM120 in patients (pts) with relapsed/refractory acute leukemias. Fourteen pts (12 acute myeloid leukemia, 1 acute lymphoblastic leukemia, and 1 mixed phenotype leukemia) were enrolled. Twelve pts received BKM-120 80 mg/day and two 100 mg/day. The MTD was 80 mg/day. Of the 14 patients treated, the best response was stable disease in one patient that lasted 82 days. The median survival for all patients was 75 days (range 10-568). Three patients with a 3q26 chromosome abnormality had a significantly improved median survival of 360 days (range 278-568) as compared to a median survival of 57 days (range, 10-125) among the 11 other patients. The most frequent drug-related toxicities included confusion, mucositis, dysphagia, and fatigue. Western blot profiling revealed a decrease in p-pS6K/total pS6K in 5/7 (71%) available patient samples with a mean quantitative inhibition of 65% (range, 32-100%) and a decrease in p-FOXO3/total FOXO3 in 4/6 (67%) samples with a mean quantitative inhibition of 93% (range, 89-100%). BKM120 administered at 80 mg/day showed modest efficacy and was tolerable in advanced acute leukemias. Am. J. Hematol. 92:7-11, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Aminopiridinas , Antineoplásicos , Leucemia Mieloide Aguda/tratamiento farmacológico , Morfolinas , Inhibidores de las Quinasa Fosfoinosítidos-3 , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Aminopiridinas/administración & dosificación , Aminopiridinas/efectos adversos , Aminopiridinas/uso terapéutico , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Supervivencia sin Enfermedad , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/mortalidad , Dosis Máxima Tolerada , Persona de Mediana Edad , Morfolinas/administración & dosificación , Morfolinas/efectos adversos , Morfolinas/uso terapéutico , Fosfatidilinositol 3-Quinasas/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad
19.
Pediatr Blood Cancer ; 64(1): 32-38, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27555294

RESUMEN

BACKGROUND: l-Asparaginase is an important drug for treatment of childhood acute lymphoblastic leukemia (ALL), but is associated with serious toxicities, including pancreatitis and hypertriglyceridemia (HTG). Asparaginase-associated pancreatitis (AAP) is a common reason for stopping asparaginase treatment. The aim of this study was to explore if HTG or early elevations in pancreatic enzymes were associated with the subsequent development of AAP. METHOD: Children (1.0-17.9 years) diagnosed with ALL, treated with asparaginase for 30 weeks, according to the NOPHO ALL2008 protocol at the University Hospital Rigshospitalet, Copenhagen, Denmark, were eligible. Pancreatic enzymes, triglycerides, and cholesterol were measured regularly. RESULTS: Thirty-one patients were included. Seven patients were diagnosed with AAP. HTG was most evident when PEG-asparaginase and dexamethasone were administered concomitantly. Overall, there was no significant difference in triglyceride levels in patients who experienced AAP and patients who did not. An increase in triglyceride levels during concomitant dexamethasone therapy in delayed intensification was significantly associated with an increase in pancreas-specific amylase levels two weeks later (P = 0.005). CONCLUSIONS: AAP does not seem to be associated with HTG. Continuous monitoring of pancreas enzymes does not predict AAP.


Asunto(s)
Asparaginasa/efectos adversos , Biomarcadores de Tumor/metabolismo , Hipertrigliceridemia/epidemiología , alfa-Amilasas Pancreáticas/sangre , Pancreatitis/inducido químicamente , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Triglicéridos/metabolismo , Adolescente , Niño , Preescolar , Dinamarca/epidemiología , Femenino , Estudios de Seguimiento , Humanos , Hipertrigliceridemia/diagnóstico , Lactante , Masculino , Estadificación de Neoplasias , Pancreatitis/sangre , Pancreatitis/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico
20.
Nature ; 537(7619): S60-2, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27602742

Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Pruebas Genéticas , Farmacogenética/métodos , Farmacogenética/tendencias , Medicina de Precisión/métodos , Medicina de Precisión/tendencias , Antidepresivos/administración & dosificación , Antidepresivos/efectos adversos , Antidepresivos/metabolismo , Antidepresivos/uso terapéutico , Niño , Didesoxinucleósidos/efectos adversos , Hipersensibilidad a las Drogas/complicaciones , Hipersensibilidad a las Drogas/enzimología , Hipersensibilidad a las Drogas/genética , Medicina Basada en la Evidencia/tendencias , Pruebas Genéticas/estadística & datos numéricos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Antígenos HLA-B/genética , Humanos , Masculino , Mercaptopurina/administración & dosificación , Mercaptopurina/efectos adversos , Mercaptopurina/metabolismo , Mercaptopurina/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Primaquina/efectos adversos , Errores Innatos del Metabolismo de la Purina-Pirimidina/complicaciones , Errores Innatos del Metabolismo de la Purina-Pirimidina/enzimología , Errores Innatos del Metabolismo de la Purina-Pirimidina/genética , Succinilcolina/efectos adversos , Warfarina/administración & dosificación , Warfarina/efectos adversos , Warfarina/metabolismo , Warfarina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA