Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood Cells Mol Dis ; 87: 102522, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33260083

RESUMEN

Stathmin 1 (STMN1) is a cytosolic phosphoprotein that was discovered as a result of its high level of expression in leukemic cells. It plays an important role in the regulation of mitosis by promoting depolymerization of the microtubules that make up the mitotic spindle and, aging has been shown to impair STMN1 levels and change microtubule stability. We have previously demonstrated that a high level of STMN1 expression during early megakaryopoiesis is necessary for proliferation of megakaryocyte progenitors and that down-regulation of STMN1 expression during late megakaryopoiesis is important for megakaryocyte maturation and platelet production. In this report, we examined the effects of STMN1 deficiency on erythroid and megakaryocytic lineages in the mouse. Our studies show that STMN1 deficiency results in mild thrombocytopenia in young animals which converts into profound thrombocythemia as the mice age. STMN1 deficiency also lead to macrocytic changes in both erythrocytes and megakaryocytes that persisted throughout the life of STMN1 knock-out mice. Furthermore, STMN1 knock-out mice displayed a lower number of erythroid and megakaryocytic progenitor cells and had delayed recovery of their blood counts after chemotherapy. These studies show an important role for STMN1 in normal erythro-megakaryopoietic development and suggests potential implications for disorders affecting these hematopoietic lineages.


Asunto(s)
Anemia Macrocítica/genética , Células Precursoras Eritroides/patología , Megacariocitos/patología , Estatmina/genética , Trombocitosis/genética , Anemia Macrocítica/patología , Animales , Plaquetas/patología , Eritropoyesis , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombocitosis/patología
2.
Curr Oncol ; 29(7): 4779-4790, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35877239

RESUMEN

Background: Several studies suggest that patients with KRAS-mutant NSCLC fail to benefit from standard systemic therapies and do not respond to EGFR inhibitors. Most recently, KRAS 12c data suggest specific treatment for improving ORR and OS. There is a clear need for therapies specifically developed for these patients. Moreover, data that might be suggestive of a response to specific therapies, such as BRCA1, are needed, and two mutations that were studied in other malignancies show more response to PARP inhibitors. Molecular profiling has the potential to identify other potential targets that may provide better treatment and novel targeted therapy for KRAS-mutated NSCLC. Methods: We purified RNA from archived tissues of patients with stage I and II NSCLC with wild-type (wt) and mutant (mt) KRAS tumors; paired normal tissue adjacent to the tumor from 20 and 17 patients, respectively, and assessed, using real-time reverse transcriptase−polymerase chain reaction (RT-PCR), the expression of four genes involved in DNA synthesis and repair, including thymidylate synthase (TS), BRCA1, ECCR1, RAP80, and the proto-oncogene SRC. Additionally, we assessed the expression of PD-L1 in mt KRAS tumors with immunohistochemistry using an antibody against PD-L1. Results: Our results show that in mtKRAS tumors, the level of expression of ERCC1, TS, and SRC was significantly increased in comparison to paired normal lung tissue (p ≤ 0.04). The expression of BRCA1 and RAP80 was similar in both mt KRAS tumors and paired normal tissue. Furthermore, the expression of BRCA1, TS, and SRC was significantly increased in wt KRAS tumors relative to their expression in the normal lung tissue (p < 0.044). The expression of ERCC1 and RAP80 was similar in wt KRAS tumors and paired normal tissue. Interestingly, SRC expression in mtKRAS tumors was decreased in comparison to wt KRAS tumors. Notably, there was an expression of PD-L1 in the tumor and stromal cells in a few (5 out of 20) mtKRAS tumors. Our results suggest that a greater ERCC1 expression in mt KRAS tumors might increase platinum resistance in this group of patients, whereas the greater expression of BRCA1 in wt KRAS tumor might be suggestive of the sensitivity of taxanes. Our data also suggest that the combination of an SRC inhibitor with a TS inhibitor, such as pemetrexed, might improve the outcome of patients with NSCLC and in particular, patients with wt KRAS tumors. PD-L1 expression in tumors, and especially stromal cells, suggests a better outcome. Conclusion: mt KRAS NSCLC patients might benefit from a treatment strategy that targets KRAS in combination with therapeutic agents based on pharmacogenomic markers, such as SRC and BRCA1. mtKRAS tumors are likely to be platinum-, taxane-, and pemetrexed-resistant, as well as having a low level of PD-L1 expression; thus, they are less likely to receive single-agent immunotherapy, such as pembrolizumab, as the first-line therapy. wt KRAS tumors with BRCA1 positivity tend to be sensitive to taxane therapy and, potentially, platinum. Our results suggest the need to develop targeted therapies for KRAS-mutant NSCLC or combine the targeting of oncogenic KRAS in addition to other therapeutic agents specific to the molecular profile of the tumor.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Antígeno B7-H1 , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Pemetrexed/uso terapéutico , Platino (Metal)/metabolismo , Platino (Metal)/uso terapéutico , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
3.
Blood Cells Mol Dis ; 43(1): 58-62, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19346141

RESUMEN

Inducers of fetal hemoglobin (HbF) have shown considerable promise in the treatment of sickle cell disease (SCD). However, the same agents have shown less clinical activity in beta-thalassemia (beta-Thal). To understand the basis of these differences in clinical effectiveness, we compared the effects of butyrate and hemin on the expression of the different globin genes in progenitors-derived erythroid cells from patients with beta-Thal intermedia and SCD. Exposure to butyrate resulted in an augmentation of gamma-globin mRNA levels in both SCD and beta-Thal. Interestingly, butyrate exposure increased alpha-globin expression in beta-Thal, while alpha-globin mRNA levels decreased in SCD in response to butyrate. As a result, the favorable effects of the butyrate-induced increase in gamma-globin expression on alpha:beta-like globin mRNA imbalance in beta-Thal were reduced as a result of the associated increase in alpha-globin expression. Hemin had similar but less profound effects on all three globin genes in both categories of patients. Although the majority of patients with beta-Thal did not correct their globin imbalance in response to butyrate or hemin induction of HbF in a minority of patients resulted in marked reduction in globin imbalance. Thus, we believe that the poor clinical response in a majority of patients with beta-Thal to inducers of gamma-globin expression may be a reflection of unfavorable effects of these agents on the other globin genes.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Butiratos/uso terapéutico , Expresión Génica/efectos de los fármacos , Hemina/uso terapéutico , Hemoglobinas/genética , Talasemia beta/tratamiento farmacológico , Adulto , Butiratos/farmacología , Células Cultivadas , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Femenino , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Hemina/farmacología , Subunidades de Hemoglobina/genética , Subunidades de Hemoglobina/metabolismo , Hemoglobinas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , Adulto Joven
4.
Target Oncol ; 12(3): 323-332, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28357727

RESUMEN

BACKGROUND: The combination of everolimus and the imidazoquinoline derivative, BEZ235 (dactolisib), a dual PI3K/mTOR inhibitor, demonstrated synergy in a preclinical model. OBJECTIVE: To establish clinical feasibility, a phase Ib dose-escalation trial investigating safety and pharmacokinetics of this combination in patients with advanced tumors was performed. PATIENTS AND METHODS: BEZ235 was orally administered daily in escalating doses of 200, 400, and 800 mg along with everolimus at 2.5 mg daily in 28-day cycles. Nineteen patients were enrolled. Adverse events and tumor responses were evaluated using CTCAE v4.0 and RECIST 1.1, respectively. Pharmacokinetic analyses were performed. RESULTS: Common toxicities observed included fatigue, diarrhea, nausea, mucositis, and elevated liver enzymes. No confirmed responses were observed. BEZ235 pharmacokinetics exhibited dose-proportional increases in Cmax and AUC0-24 over the three doses, with high inter-individual variability. Non-compartmental and population pharmacokinetic-based simulations indicated significant increases in everolimus Cmax and AUC0-24 on day 28 and decreased clearance to 13.41 L/hr. CONCLUSIONS: The combination of BEZ235 and everolimus demonstrated limited efficacy and tolerance. BEZ235 systemic exposure increased in a dose-proportional manner while oral bioavailability was quite low, which may be related to gastrointestinal-specific toxicity. The changes in steady-state pharmacokinetics of everolimus with BEZ235 highlight potential drug-drug interactions when these two drugs are administered together. Clinicaltrials.gov: NCT01508104.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Everolimus/uso terapéutico , Imidazoles/uso terapéutico , Neoplasias/tratamiento farmacológico , Quinolinas/uso terapéutico , Administración Oral , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Everolimus/efectos adversos , Femenino , Humanos , Imidazoles/efectos adversos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Quinolinas/efectos adversos , Serina-Treonina Quinasas TOR/metabolismo , Síndrome de Lisis Tumoral/etiología
5.
Blood Rev ; 20(4): 227-34, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16513230

RESUMEN

Reactivation of fetal hemoglobin (HbF) expression is an important therapeutic option in patients with hemoglobin disorders. In sickle cell disease (SCD), an increase in HbF would interfere with the polymerization of sickle hemoglobin while in beta-thalassemia, an increase in gamma-globin chain synthesis would decrease non-alpha:alpha chain imbalance. Hydroxyurea, an inducer of HbF, is the only currently approved agent for the treatment of patients with moderate and/or severe SCD. However, about one third of patients with SCD do not respond to HU, and in beta-thalassemia, the clinical response is unimpressive. The last decade has seen a renewed interest in the use of inhibitors of DNA methylation in the treatment of patients with hemoglobin disorders. In this review, we discuss the role of DNA methylation in gamma-globin gene regulation, describe clinical trials with agents that hypomethylate DNA and speculate about the future role of DNA hypomethylation therapy in patients with SCD and beta-thalassemia.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Hemoglobinopatías/tratamiento farmacológico , Hemoglobina Fetal/biosíntesis , Hemoglobina Fetal/efectos de los fármacos , Hemoglobina Fetal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Talasemia/tratamiento farmacológico
6.
Ann N Y Acad Sci ; 1054: 228-37, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16339670

RESUMEN

The first studies of the pharmacological induction of fetal hemoglobin were conducted in patients with sickle cell disease and thalassemia. Although hydroxyurea was approved by the FDA for the treatment of sickle cell disease in 1996, no similar pharmacological agent(s) has been approved for the treatment of patients with thalassemic disorders. The small-scale studies of the induction of fetal hemoglobin in thalassemia have been generally disappointing. The aim of this report is to provide a critical analysis of the factors that may be responsible for our failure to develop an effective fetal hemoglobin induction therapy for patients with thalassemia. We also describe several areas for future investigation that may be critically important for the development of an effective therapy for thalassemia.


Asunto(s)
Azacitidina/uso terapéutico , Hemoglobina Fetal/biosíntesis , Expresión Génica/efectos de los fármacos , Globinas/biosíntesis , Hidroxiurea/uso terapéutico , Talasemia/tratamiento farmacológico , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Animales , Azacitidina/efectos adversos , Azacitidina/farmacología , Terapia Combinada , Aprobación de Drogas , Evaluación de Medicamentos , Evaluación Preclínica de Medicamentos , Eritropoyesis/efectos de los fármacos , Hemoglobina Fetal/genética , Regulación de la Expresión Génica , Globinas/genética , Humanos , Hidroxiurea/efectos adversos , Hidroxiurea/farmacología , Hierro/metabolismo , Células K562/efectos de los fármacos , Células K562/metabolismo , Modelos Animales , Papio , Talasemia/sangre , Talasemia/genética , Talasemia/terapia , Reacción a la Transfusión , Estados Unidos , United States Food and Drug Administration
8.
Hematol Oncol Clin North Am ; 24(6): 1131-44, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21075284

RESUMEN

Reactivation of fetal hemoglobin (HbF) expression is an important therapeutic option in adult patients with hemoglobin disorders. The understanding of the developmental regulation of γ-globin gene expression was followed by the identification of a number of chemical compounds that can reactivate HbF synthesis in vitro and in vivo in patients with hemoglobin disorders. These HbF inducers can be grouped in several classes based on their mechanisms of action. This article focuses on pharmacologic agents that were tested in humans and discusses current knowledge about the mechanisms by which they induce HbF.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Hemoglobina Fetal/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hemoglobinopatías/genética , Adulto , Azacitidina/análogos & derivados , Azacitidina/farmacología , Decitabina , Inhibidores Enzimáticos/farmacología , Hemoglobina Fetal/metabolismo , Hemoglobinopatías/tratamiento farmacológico , Hemoglobinopatías/metabolismo , Humanos , gamma-Globinas/genética , gamma-Globinas/metabolismo
9.
Blood Cells Mol Dis ; 40(2): 166-73, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18029204

RESUMEN

K562 erythroleukemia cells have been widely used as a model for the study of globin gene regulation. A number of agents have been shown to activate or suppress globin gene expression in these cells. However, the molecular effects of these agents on the epigenetic configuration of the alpha- and gamma-globin genes that encode HbF are not known. In this report, we investigated the relationship between globin expression and histone acetylation of the human alpha- and beta-globin clusters in the fetal erythroid environment of K562 cells. Our studies suggest that acetylation of histone H3 may be important in regulating developmental stage-specific expression of the different beta-like globin genes while acetylation of both histones H3 and H4 may be important for the regulation of tissue-specific expression of these genes. In contrast, acetylation of both histones H3 and H4 at the alpha-like globin promoters appears to be important for both developmental stage- and tissue-specific expression. Interestingly, butyrate-induced activation of alpha-globin gene expression in K562 cells is associated with significant increase in histone acetylation levels while TPA-induced inhibition is associated with decreased histone acetylation at its promoters. In contrast, changes in histone acetylation and DNA methylation do not appear to be important in the regulation of gamma-globin gene expression by the same agents. These data suggest that the butyrate-mediated induction of the fetal gamma-globin genes in K562 cells is not a direct result of its histone deacetylase inhibitor activity of butyrate on the chromatin of the gamma-globin promoters, while the induction of the alpha-globin genes could be a result of a direct effect of butyrate on chromatin at its promoters. This is another example of the important differences in the molecular mechanisms of regulation of the genes of the alpha- and beta-like globin clusters.


Asunto(s)
Epigénesis Genética , Globinas/genética , Histona Desacetilasas/metabolismo , Acetilación , Butiratos/farmacología , Metilación de ADN , Células Precursoras Eritroides/enzimología , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Globinas/metabolismo , Células HeLa , Hemina/farmacología , Histona Desacetilasas/genética , Histonas/genética , Histonas/metabolismo , Humanos , Células K562 , Familia de Multigenes , Regiones Promotoras Genéticas
10.
Blood ; 110(9): 3391-7, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17638855

RESUMEN

Butyrate is a prototype of histone deacetylase inhibitors that is believed to reactivate silent genes by inducing epigenetic modifications. Although butyrate was shown to induce fetal hemoglobin (HbF) production in patients with hemoglobin disorders, the mechanism of this induction has not been fully elucidated. Our studies of the epigenetic configuration of the beta-globin cluster suggest that DNA methylation and histone H3 acetylation are important for the regulation of developmental stage-specific expression of the beta-like globin genes, whereas acetylation of both histones H3 and H4 seem to be important for the regulation of tissue-specific expression. These studies suggest that DNA methylation may be important for the silencing of the beta-like globin genes in nonerythroid hematopoietic cells but may not be necessary for their silencing in nonhematopoietic cells. Furthermore, our studies demonstrate that butyrate exposure results in a true reversal of the normal developmental switch from gamma- to beta-globin expression. This is associated with increased histone acetylation and decreased DNA methylation of the gamma-globin genes, with opposite changes in the beta-globin gene. These studies provide strong support for the role of epigenetic modifications in the normal developmental and tissue-specific regulation of globin gene expression and in the butyrate-mediated pharmacologic induction of HbF production.


Asunto(s)
Butiratos/farmacología , Epigénesis Genética/fisiología , Hemoglobina Fetal/metabolismo , Globinas/genética , Acetilación , Células Cultivadas , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Metilación de ADN , Células HeLa , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Humanos , Células K562
11.
Artículo en Inglés | MEDLINE | ID: mdl-17124041

RESUMEN

Reactivation of fetal hemoglobin (HbF) expression is an important therapeutic option in patients with hemoglobin disorders. In sickle cell disease (SCD), an increase in HbF inhibits the polymerization of sickle hemoglobin and the resulting pathophysiology. Hydroxyurea, an inducer of HbF, has already been approved for the treatment of patients with moderate and/or severe SCD. Recent clinical trials with other pharmacological inducers of HbF, such as butyrate and decitabine, have shown considerable promise. In this chapter, we highlight the important clinical trials with pharmacological inducers of HbF, discuss their mechanisms of action and speculate about the future of this therapeutic approach in the treatment of patients with SCD.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Hemoglobina Fetal/genética , Azacitidina/análogos & derivados , Azacitidina/uso terapéutico , Butiratos/uso terapéutico , Decitabina , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidroxiurea/uso terapéutico
12.
Artículo en Inglés | MEDLINE | ID: mdl-14633775

RESUMEN

The outlook for patients with sickle cell disease has improved steadily during the last two decades. In spite of these improvements, curative therapies are currently available only to a small minority of patients. The main theme of this chapter is to describe new therapeutic options that are at different stages of development that might result in further improvements in the outlook for patients with these disorders. Dr. Joseph DeSimone and his colleagues had previously made the important observation that the hypomethylating agent 5-azacytidine can reverse the switch from adult to fetal hemoglobin in adult baboons. Although similar activity was demonstrated in patients with sickle cell disease and beta-thalassemia, concern about the toxicity of 5-azacytidine prevented its widespread use in these disorders. In Section I, Dr. DeSimone discusses the role of DNA methylation in globin gene regulation and describe recent clinical experience with decitabine (an analogue of 5-azacytidine) in patients with sickle cell disease. These encouraging studies demonstrate significant fetal hemoglobin inducing activity of decitabine in patients who fail to respond to hydroxyurea. In Section II, Dr. George Atweh continues the same theme by describing recent progress in the study of butyrate, another inducer of fetal hemoglobin, in patients with sickle cell disease and beta-thalassemia. The main focus of his section is on the use of a combination of butyrate and hydroxyurea to achieve higher levels of fetal hemoglobin that might be necessary for complete amelioration of the clinical manifestations of these disorders. Dr. Atweh also describes novel laboratory studies that shed new light on the mechanisms of fetal hemoglobin induction by butyrate. In Section III, Dr. Ronald Nagel discusses the different available transgenic sickle mice as experimental models for human sickle cell disease. These experimental models have already had a significant impact on our understanding of the pathophysiology of sickle cell disease. Dr. Nagel describes more recent studies in which transgenic sickle mice provide the first proof of principle that globin gene transfer into hematopoietic stem cells inhibits in vivo sickling and ameliorates the severity of the disease. Although stroke in adult patients with sickle cell disease is not as common as in children, adult hematologists, like their pediatric colleagues, need to make management decisions in adult patients with a stroke or a history of stroke. Dr. Robert Adams has led several large clinical studies that investigated the role of transfusions in the prevention of stroke in children with sickle cell disease. Much less is known, however, about the prevention of first or subsequent strokes in adult patients with sickle cell disease. In Section IV, Dr. Adams provides some general guidelines for the management of adult patients with stroke while carefully distinguishing between recommendations that are evidence-based and those that are anecdotal in nature.


Asunto(s)
Azacitidina/análogos & derivados , Hemoglobinopatías , Animales , Azacitidina/farmacología , Azacitidina/uso terapéutico , Butiratos/farmacología , Butiratos/uso terapéutico , Hemorragia Cerebral/etiología , Hemorragia Cerebral/prevención & control , Metilación de ADN/efectos de los fármacos , Decitabina , Modelos Animales de Enfermedad , Globinas/genética , Hemoglobinopatías/complicaciones , Hemoglobinopatías/tratamiento farmacológico , Hemoglobinopatías/genética , Humanos , Hidroxiurea/farmacología , Hidroxiurea/uso terapéutico , Ratones , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA