Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
BMC Cancer ; 19(1): 1251, 2019 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-31881855

RESUMO

BACKGROUND: Acute Myeloid Leukemia (AML) is a malignancy of myeloid precursor cells that arise from genomic alterations in the expression of key growth regulatory genes causing cells to assume an undifferentiated state and continue to proliferate. Recent efforts have focused on developing therapies that target specific protein products of aberrantly expressed genes. However, many of the identified proteins are difficult to target and thought to be "undrugable" because of structural challenges, protein overexpression, or mutations that confer resistance to therapy. A novel technology that circumvents some of these issues is the use of small molecules that stabilize secondary DNA structures present in the promoters of many potential oncogenes and modulate their transcription. METHODS: This study characterizes the in vitro activity of the G-quadruplex-stabilizing small molecule GQC-05 in AML cells. The effect of GQC-05 on three AML cell lines was analyzed using viability and apoptosis assays. GQC-05 has been shown to down-regulate MYC through G-quadruplex stabilization in Burkitt's lymphoma cell lines. MYC expression was evaluated through qPCR and immunoblotting in the three AML cell lines following the treatment of GQC-05. In order to identify other therapeutic agents that potentiate the activity of GQC-05, combination drug screening was performed. The drug combinations were validated using in vitro cytotoxicity assays and compared to other commonly used chemotherapeutic agents. RESULTS: GQC-05 treatment of KG-1a, CMK and TF-1 cells decreased cell viability and resulted in increased DNA damage and apoptosis. Additionally, treatment of KG-1a, CMK and TF-1 with GQC-05 resulted in decreased expression of MYC mRNA and protein, with a more pronounced effect in KG-1a cells. Combination drug screening identified the Bcl-2/Bcl-XL inhibitor Navitoclax as a compound that potentiated GQC-05 activity. Co-treatment with GQC-05 and Navitoclax showed a synergistic decrease in cell viability of AML cells as determined by Chou-Talalay analysis, and induced more DNA damage, apoptosis, and rapid cytotoxicity. The cytotoxicity induced by GQC-05 and Navitoclax was more potent than that of Navitoclax combined with either cytarabine or doxorubicin. CONCLUSION: These results suggest that the G-quadruplex stabilizing small molecule GQC-05 induces down regulated MYC expression and DNA damage in AML cells. Treatment with both GQC-05 with a Bcl-2/Bcl-XL inhibitor Navitoclax results in increased cytotoxic activity, which is more pronounced than Navitoclax or GQC-05 alone, and more significant than Navitoclax in combination with cytarabine and doxorubicin that are currently being used clinically.


Assuntos
Compostos de Anilina/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Elipticinas/farmacologia , Quadruplex G/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Sulfonamidas/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Dano ao DNA , Elipticinas/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Proto-Oncogênicas c-myc/genética , Resultado do Tratamento
2.
Pediatr Blood Cancer ; 65(9): e27237, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29768711

RESUMO

Patients with Langerhans cell histiocytosis (LCH) harbor BRAF V600E and activating mutations of MAP2K1/MEK1 in 50% and 25% of cases, respectively. We evaluated a patient with treatment-refractory LCH for mutations in the RAS-RAF-MEK-ERK pathway and identified a novel mutation in the MAP2K1 gene resulting in a p.L98_K104 > Q deletion and predicted to be auto-activating. During treatment with the MEK inhibitor trametinib, the patient's disease showed significant progression. In vitro characterization of the MAP2K1 p.L98_K104 > Q deletion confirmed its effect on cellular activation of the ERK pathway and drug resistance.


Assuntos
Resistência a Medicamentos/genética , Histiocitose de Células de Langerhans/tratamento farmacológico , MAP Quinase Quinase 1/genética , Sistema de Sinalização das MAP Quinases/genética , Inibidores de Proteínas Quinases/uso terapêutico , Piridonas/uso terapêutico , Pirimidinonas/uso terapêutico , Adolescente , Corticosteroides/uso terapêutico , Butadienos/farmacologia , Terapia Combinada , Citarabina/uso terapêutico , Progressão da Doença , Quimioterapia Combinada , Ativação Enzimática/genética , Éxons/genética , Células HEK293 , Transplante de Células-Tronco Hematopoéticas , Histiocitose de Células de Langerhans/genética , Histiocitose de Células de Langerhans/terapia , Humanos , Masculino , Terapia de Alvo Molecular , Mutação , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Pirazóis/uso terapêutico , Piridonas/farmacologia , Pirimidinonas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Tiofenos/uso terapêutico , Vincristina/uso terapêutico
3.
Pediatr Blood Cancer ; 64(5)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27804235

RESUMO

BACKGROUND: Langerhans cell histiocytosis (LCH) is a clonal neoplasm characterized by widely varied clinical presentations, including multisystem involvement and systemic inflammatory symptoms. The AKT pathway is relevant to survival and proliferation of dendritic cells, and is also often upregulated in hematopoietic malignancies. A clinical response in an adult patient with LCH participating in the first-in-human trial of afuresertib prompted this prospective trial. PROCEDURE: The population in the current study included treatment-naïve (n = 7) and recurrent/refractory patients with LCH (n = 10), who received oral afuresertib (125 mg). The majority of patients were treated for > 24 weeks, with four patients receiving treatment for > 48 weeks. RESULTS: Pharmacokinetic analysis showed similar exposures in previously reported patients with other hematologic malignancies. Primary drug-related toxicities included Grade 1/2 nausea, diarrhea, dyspepsia, and vomiting. Grade 3 toxicities included fatigue, diarrhea, and pain (one of each). Another severe adverse event involved soft tissue necrosis. The overall response rate in evaluable subjects was 33% in treatment-naïve patients and 28% in patients with recurrent/refractory disease, which did not meet the predefined Bayesian criteria for efficacy. CONCLUSION: Afuresertib has clinical activity in some patients with newly diagnosed and advanced LCH.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Hematológicas/tratamento farmacológico , Histiocitose de Células de Langerhans/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Pirazóis/uso terapêutico , Tiofenos/uso terapêutico , Adulto , Idoso , Antineoplásicos/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Inibidores de Proteínas Quinases/efeitos adversos , Proteínas Proto-Oncogênicas B-raf/genética , Pirazóis/efeitos adversos , Pirazóis/farmacocinética , Tiofenos/efeitos adversos , Tiofenos/farmacocinética , Resultado do Tratamento , Estados Unidos
4.
Pediatr Blood Cancer ; 63(8): 1332-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27100618

RESUMO

Side effects of tyrosine kinase inhibitor (TKI) treatment vary in children and adults with chronic myelogenous leukemia (CML). As children have a much longer life expectancy than adults, TKI therapy may continue for decades and with long-term consequences that differ from adults. Children may develop endocrinopathies related to "off-target" effects of TKIs, such as delayed growth, changes in bone metabolism, thyroid abnormalities, and effects on puberty and fertility. These endocrinopathies present additional challenges for pediatric patients with CML. This review critically evaluates the literature on long-term endocrine side effects of TKIs in the pediatric CML population and provides suggested recommendations.


Assuntos
Doenças do Sistema Endócrino/induzido quimicamente , Sistema Endócrino/efeitos dos fármacos , Sistema Endócrino/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/uso terapêutico , Glândulas Suprarrenais/efeitos dos fármacos , Compostos de Anilina/efeitos adversos , Compostos de Anilina/uso terapêutico , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Remodelação Óssea/efeitos dos fármacos , Criança , Dasatinibe/efeitos adversos , Dasatinibe/uso terapêutico , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Transtornos do Metabolismo de Glucose/induzido quimicamente , Humanos , Mesilato de Imatinib/efeitos adversos , Mesilato de Imatinib/uso terapêutico , Imidazóis/efeitos adversos , Imidazóis/uso terapêutico , Nitrilas/efeitos adversos , Nitrilas/uso terapêutico , Piridazinas/efeitos adversos , Piridazinas/uso terapêutico , Pirimidinas/efeitos adversos , Pirimidinas/uso terapêutico , Quinolinas/efeitos adversos , Quinolinas/uso terapêutico , Glândula Tireoide/efeitos dos fármacos
5.
Pediatr Blood Cancer ; 63(2): 358-60, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26452062

RESUMO

Sinus histiocytosis with massive lymphadenopathy (SHML), or Rosai-Dorfman disease (RDD), is a non-neoplastic, lymphoproliferative disorder that usually resolves spontaneously or with minimal conventional chemotherapy. Rarely, SHML can be associated with autoimmune findings. Such cases are often treatment resistant and have high rates of morbidity and mortality. We present a case of a patient with long-standing autoimmunity in the context of SHML, dependent on standard-treatment until he was transitioned to novel monotherapy with sirolimus. Sirolimus treatment resulted in a complete remission, now sustained after discontinuation of all treatments for over 23 months, with no observable long-term sequelae.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Histiocitose Sinusal/tratamento farmacológico , Imunossupressores/uso terapêutico , Sirolimo/uso terapêutico , Doenças Autoimunes/complicações , Pré-Escolar , Histiocitose Sinusal/complicações , Humanos , Masculino
6.
Pediatr Blood Cancer ; 63(10): 1704-12, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27314817

RESUMO

Langerhans cell histiocytosis (LCH) remains a poorly understood disorder with heterogeneous clinical presentations characterized by focal or disseminated lesions that contain excessive CD1a+ langerin+ cells with dendritic cell features known as "LCH cells." Two of the major questions investigated over the past century have been (i) the origin of LCH cells and (ii) whether LCH is primarily an immune dysregulatory disorder or a neoplasm. Current opinion is that LCH cells are likely to arise from hematopoietic precursor cells, although the stage of derailment and site of transformation remain unclear and may vary in patients with different extent of disease. Over the years, evidence has provided the view that LCH is a neoplasm. The demonstration of clonality of LCH cells, insufficient evidence alone for neoplasia, is now bolstered by finding driver somatic mutations in BRAF in up to 55% of patients with LCH, and activation of the RAS-RAF-MEK-ERK (where MEK and ERK are mitogen-activated protein kinase and extracellular signal-regulated kinase, respectively) pathway in nearly 100% of patients with LCH. Herein, we review the evidence that recurrent genetic abnormalities characterized by activating oncogenic mutations should satisfy prerequisites for LCH to be called a neoplasm. As a consequence, recurrent episodes of LCH should be considered relapsed disease rather than disease reactivation. Mapping the complete genetic landscape of this intriguing disease will provide additional support for the conclusion that LCH is a neoplasm and is likely to provide more potential opportunities for molecularly targeted therapies.


Assuntos
Histiocitose de Células de Langerhans/genética , Neoplasias/genética , Evolução Clonal , Histiocitose de Células de Langerhans/tratamento farmacológico , Humanos , Sistema de Sinalização das MAP Quinases , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Recidiva
7.
Blood ; 122(14): 2487-90, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23943650

RESUMO

Classical 5q- syndrome is an acquired macrocytic anemia of the elderly. Similar to Diamond Blackfan anemia (DBA), an inherited red cell aplasia, the bone marrow is characterized by a paucity of erythroid precursors. RPS14 deletions in combination with other deletions in the region have been implicated as causative of the 5q- syndrome phenotype. We asked whether smaller, less easily detectable deletions could account for a syndrome with a modified phenotype. We employed single-nucleotide polymorphism array genotyping to identify small deletions in patients diagnosed with DBA and other anemias lacking molecular diagnoses. Diminutive mosaic deletions involving RPS14 were identified in a 5-year-old patient with nonclassical DBA and in a 17-year-old patient with myelodysplastic syndrome. Patients with nonclassical DBA and other hypoproliferative anemias may have somatically acquired 5q deletions with RPS14 haploinsufficiency not identified by fluorescence in situ hybridization or cytogenetic testing, thus refining the spectrum of disorders with 5q- deletions.


Assuntos
Anemia de Diamond-Blackfan/genética , Anemia Macrocítica/genética , Análise Citogenética/métodos , Proteínas Ribossômicas/genética , Adolescente , Anemia de Diamond-Blackfan/diagnóstico , Anemia Macrocítica/diagnóstico , Anemia Macrocítica/tratamento farmacológico , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Feminino , Genótipo , Humanos , Fatores Imunológicos/uso terapêutico , Lenalidomida , Fenótipo , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase em Tempo Real , Talidomida/análogos & derivados , Talidomida/uso terapêutico
8.
Pediatr Blood Cancer ; 62(10): 1761-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25929242

RESUMO

BACKGROUND: Despite the frequent use and radiation exposure of computed tomography (CT) scans, there is little information on patterns of CT use and their utility in the management of pediatric patients with fever and neutropenia (FN). We examined the contribution of either the commonly employed pan-CT (multiple anatomical locations) or targeted CT (single location) scanning to identify possible infectious etiologies in this challenging clinical scenario. Procedure Pediatric patients with an underlying malignancy admitted for fever (temperature ≥ 38.3 °C) and an absolute neutrophil count <500 cells/µL from 2003-2009 were included. Risk factors associated with utilization, results, and effects on clinical management of CT scans were identified. Results Charts for 635 admissions for FN from 263 patients were reviewed. Overall, 139 (22%) admissions (93 individuals) had at least one scan. Of 188 scans, 103 (55%) were pan-scans. Changes in management were most strongly associated with the identification of evidence consistent with infection (OR = 12.64, 95% CI: 5.05-31.60, P < 0.001). Seventy-eight (41%) of all CT scans led to a change in clinical management, most commonly relating to use of antibiotic (N = 41, 53%) or antifungal/antiviral medications (N = 33, 42%). The odds of a change in clinical management did not differ for those receiving a pan-scan compared to those receiving a targeted scan (OR = 1.23; 95% CI, 0.61-2.46; P = 0.57). Conclusions When CT is clinically indicated, it is important for clinicians to strongly consider utilizing a targeted scan to reduce radiation exposure to patients as well as to decrease costs without compromising care.


Assuntos
Febre/diagnóstico por imagem , Neutropenia/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Tomografia Computadorizada por Raios X/estatística & dados numéricos , Adolescente , Criança , Pré-Escolar , Feminino , Febre/etiologia , Humanos , Masculino , Oncologia/métodos , Neoplasias/complicações , Neutropenia/etiologia , Pediatria , Estudos Retrospectivos , Tomografia Computadorizada por Raios X/efeitos adversos , Adulto Jovem
9.
Pediatr Blood Cancer ; 62(4): 629-36, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25704135

RESUMO

BACKGROUND: Juvenile myelomonocytic leukemia (JMML) is not durably responsive to chemotherapy, and approximately 50% of patients relapse after hematopoietic stem cell transplant (HSCT). Here we report the activity and acute toxicity of the farnesyl transferase inhibitor tipifarnib, the response rate to 13-cis retinoic acid (CRA) in combination with cytoreductive chemotherapy, and survival following HSCT in children with JMML. PROCEDURE: Eighty-five patients with newly diagnosed JMML were enrolled on AAML0122 between 2001 and 2006. Forty-seven consented to receive tipifarnib in a phase II window before proceeding to a phase III trial of CRA in combination with fludarabine and cytarabine followed by HSCT and maintenance CRA. Thirty-eight patients enrolled only in the phase III trial. RESULTS: Overall response rate was 51% after tipifarnib and 68% after fludarabine/cytarabine/CRA. Tipifarnib did not increase pre-transplant toxicities. Forty-six percent of the 44 patients who received protocol compliant HSCT relapsed. Five-year overall survival was 55 ± 11% and event-free survival was 41 ± 11%, with no significant difference between patients who did or did not receive tipifarnib. CONCLUSIONS: Administration of tipifarnib in the window setting followed by HSCT in patients with newly diagnosed JMML was safe and yielded a 51% initial response rate as a single agent, but failed to reduce relapse rates or improve long-term overall survival.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Farnesil-Difosfato Farnesiltransferase/antagonistas & inibidores , Leucemia Mielomonocítica Juvenil/tratamento farmacológico , Quinolonas/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Citarabina/administração & dosagem , Intervalo Livre de Doença , Inibidores Enzimáticos/administração & dosagem , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Lactente , Isotretinoína/administração & dosagem , Leucemia Mielomonocítica Juvenil/enzimologia , Leucemia Mielomonocítica Juvenil/mortalidade , Leucemia Mielomonocítica Juvenil/patologia , Masculino , Pessoa de Meia-Idade , Taxa de Sobrevida , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados
10.
Am J Hematol ; 89(10): 985-91, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25042156

RESUMO

Diamond Blackfan anemia (DBA), a syndrome primarily characterized by anemia and physical abnormalities, is one among a group of related inherited bone marrow failure syndromes (IBMFS) which share overlapping clinical features. Heterozygous mutations or single-copy deletions have been identified in 12 ribosomal protein genes in approximately 60% of DBA cases, with the genetic etiology unexplained in most remaining patients. Unlike many IBMFS, for which functional screening assays complement clinical and genetic findings, suspected DBA in the absence of typical alterations of the known genes must frequently be diagnosed after exclusion of other IBMFS. We report here a novel deletion in a child that presented such a diagnostic challenge and prompted development of a novel functional assay that can assist in the diagnosis of a significant fraction of patients with DBA. The ribosomal proteins affected in DBA are required for pre-rRNA processing, a process which can be interrogated to monitor steps in the maturation of 40S and 60S ribosomal subunits. In contrast to prior methods used to assess pre-rRNA processing, the assay reported here, based on capillary electrophoresis measurement of the maturation of rRNA in pre-60S ribosomal subunits, would be readily amenable to use in diagnostic laboratories. In addition to utility as a diagnostic tool, we applied this technique to gene discovery in DBA, resulting in the identification of RPL31 as a novel DBA gene.


Assuntos
Precursores de RNA , Processamento Pós-Transcricional do RNA/genética , RNA Ribossômico , Proteínas Ribossômicas , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Feminino , Humanos , Lactente , Células K562 , Precursores de RNA/genética , Precursores de RNA/metabolismo , RNA Ribossômico/metabolismo , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Subunidades Ribossômicas Maiores de Eucariotos/genética , Subunidades Ribossômicas Maiores de Eucariotos/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/genética , Subunidades Ribossômicas Menores de Eucariotos/metabolismo
11.
Pediatr Blood Cancer ; 61(8): 1403-10, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24648275

RESUMO

BACKGROUND: Little is known about childhood ALL in the Middle East. This study was undertaken by MECCA as initial efforts in collaborative data collection to provide clinical and demographic information on children with ALL in the Middle East. PROCEDURE: Clinical and laboratory data for patients with ALL between January 2008 and April 2012 were prospectively collected from institutions in 14 Middle East countries and entered into a custom-built-database during induction phase. All laboratory studies including cytogenetics were done at local institutions. RESULTS: The 1,171 voluntarily enrolled patients had a mean age of 6.1 ± 3.9 years and 59.2% were boys. T-ALL represented 14.8% and 84.2% had B-precursor ALL. At diagnosis, 5.6% had CNS disease. The distribution of common genetic abnormalities reflected a similar percentage of hyperdiploidy (25.6%), but a lower percentage of ETV6-RUNX1 translocation (14.7%) compared to large series reported from Western populations. By clinical criteria, 47.1% were low/standard risk, 16.9% were intermediate risk, and 36% were high risk. Most patients received all their care at the same unit (96.9%). Patients had excellent induction response to chemotherapy with an overall complete remission rate of 96%. Induction toxicities were acceptable. CONCLUSIONS: This first collaborative study has established a process for prospective data collection and future multinational collaborative research in the Middle East. Despite the limitations of an incomplete population-based study, it provides the first comprehensive baseline data on clinical characteristics, laboratory evaluation, induction outcome, and toxicity. Further work is planned to uncover possible biologic differences of ALL in the region and to improve diagnosis and management.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Translocação Genética , Adolescente , Criança , Pré-Escolar , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Intervalo Livre de Doença , Feminino , Humanos , Lactente , Masculino , Oriente Médio/epidemiologia , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Taxa de Sobrevida
12.
Nat Med ; 13(9): 1060-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17694067

RESUMO

Transformation and cancer growth are regulated by the coordinate actions of oncogenes and tumor suppressors. Here, we show that the novel E3 ubiquitin ligase HACE1 is frequently downregulated in human tumors and maps to a region of chromosome 6q21 implicated in multiple human cancers. Genetic inactivation of HACE1 in mice results in the development of spontaneous, late-onset cancer. A second hit from either environmental triggers or genetic heterozygosity of another tumor suppressor, p53, markedly increased tumor incidence in a Hace1-deficient background. Re-expression of HACE1 in human tumor cells directly abrogates in vitro and in vivo tumor growth, whereas downregulation of HACE1 via siRNA allows non-tumorigenic human cells to form tumors in vivo. Mechanistically, the tumor-suppressor function of HACE1 is dependent on its E3 ligase activity and HACE1 controls adhesion-dependent growth and cell cycle progression during cell stress through degradation of cyclin D1. Thus, HACE1 is a candidate chromosome 6q21 tumor-suppressor gene involved in multiple cancers.


Assuntos
Cromossomos Humanos Par 6 , Genes Supressores de Tumor , Neoplasias Renais/genética , Ubiquitina-Proteína Ligases/genética , Tumor de Wilms/genética , Animais , Repetição de Anquirina/genética , Mapeamento Cromossômico , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Mutantes , RNA Mensageiro/genética
13.
Nat Rev Cancer ; 5(6): 488-94, 2005 06.
Artigo em Inglês | MEDLINE | ID: mdl-15928676

RESUMO

Histiocytoses are a group of rare diseases that involve histiocytes (literally tissue cells (Greek), but in reality tissue-resident macrophages and dendritic cells), which are derived from bone-marrow stem cells. Histiocytoses pose problems similar to those of other rare diseases of childhood. Individual physicians see few cases, disease material is hard to collect and families suffer from lack of information and understanding. In this article, we describe how a series of 'think tank' meetings, the Nikolas Symposia, which have concentrated on Langerhans cell histiocytosis, have furthered our understanding of this enigmatic disease.


Assuntos
Histiocitose de Células de Langerhans/fisiopatologia , Diferenciação Celular , Criança , Células Dendríticas , Diagnóstico Diferencial , Histiocitose de Células de Langerhans/complicações , Humanos , Macrófagos , Monócitos , Prognóstico
14.
Am J Hum Genet ; 86(2): 222-8, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20116044

RESUMO

Diamond-Blackfan anemia (DBA), an inherited bone marrow failure syndrome characterized by anemia that usually presents before the first birthday or in early childhood, is associated with birth defects and an increased risk of cancer. Although anemia is the most prominent feature of DBA, the disease is also characterized by growth retardation and congenital malformations, in particular craniofacial, upper limb, heart, and urinary system defects that are present in approximately 30%-50% of patients. DBA has been associated with mutations in seven ribosomal protein (RP) genes, RPS19, RPS24, RPS17, RPL35A, RPL5, RPL11, and RPS7, in about 43% of patients. To continue our large-scale screen of RP genes in a DBA population, we sequenced 35 ribosomal protein genes, RPL15, RPL24, RPL29, RPL32, RPL34, RPL9, RPL37, RPS14, RPS23, RPL10A, RPS10, RPS12, RPS18, RPL30, RPS20, RPL12, RPL7A, RPS6, RPL27A, RPLP2, RPS25, RPS3, RPL41, RPL6, RPLP0, RPS26, RPL21, RPL36AL, RPS29, RPL4, RPLP1, RPL13, RPS15A, RPS2, and RPL38, in our DBA patient cohort of 117 probands. We identified three distinct mutations of RPS10 in five probands and nine distinct mutations of RPS26 in 12 probands. Pre-rRNA analysis in lymphoblastoid cells from patients bearing mutations in RPS10 and RPS26 showed elevated levels of 18S-E pre-rRNA. This accumulation is consistent with the phenotype observed in HeLa cells after knockdown of RPS10 or RPS26 expression with siRNAs, which indicates that mutations in the RPS10 and RPS26 genes in DBA patients affect the function of the proteins in rRNA processing.


Assuntos
Anemia de Diamond-Blackfan/genética , Mutação/genética , Proteínas Ribossômicas/genética , Sequência de Bases , Humanos , Processamento Pós-Transcricional do RNA
15.
Blood ; 118(26): 6752-9; quiz 6996, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-21849481

RESUMO

Transient myeloproliferative disorder (TMD), restricted to newborns with trisomy 21, is a megakaryocytic leukemia that although lethal in some is distinguished by its spontaneous resolution. Later development of acute myeloid leukemia (AML) occurs in some. Prospective enrollment (n = 135) elucidated the natural history in Down syndrome (DS) patients diagnosed with TMD via the use of uniform monitoring and intervention guidelines. Prevalent at diagnosis were leukocytosis, peripheral blast exceeding marrow blast percentage, and hepatomegaly. Among those with life-threatening symptoms, most (n = 29/38; 76%) received intervention therapy until symptoms abated and then were monitored similarly. Organomegaly with cardiopulmonary compromise most frequently led to intervention (43%). Death occurred in 21% but only 10% were attributable to TMD (intervention vs observation patients: 13/14 vs 1/15 because of TMD). Among those solely observed, peripheral blasts and all other TMD symptoms cleared at a median of 36 and 49 days from diagnosis, respectively. On the basis of the diagnostic clinical findings of hepatomegaly with or without life-threatening symptoms, 3 groups were identified with differing survival: low risk with neither finding (38%), intermediate risk with hepatomegaly alone (40%), and high risk with both (21%; overall survival: 92% ± 8%, 77% ± 12%, and 51% ± 19%, respectively; P ≤ .001). Among all, AML subsequently occurred in 16% at a median of 441 days (range, 118-1085 days). The trial is registered at http://www.clinicaltrials.gov as NCT00003593.


Assuntos
Citarabina/uso terapêutico , Síndrome de Down/complicações , Leucemia Megacarioblástica Aguda/tratamento farmacológico , Transtornos Mieloproliferativos/tratamento farmacológico , Doença Aguda , Antimetabólitos Antineoplásicos/uso terapêutico , Progressão da Doença , Feminino , Seguimentos , Humanos , Lactente , Recém-Nascido , Estimativa de Kaplan-Meier , Leucemia Megacarioblástica Aguda/complicações , Leucemia Megacarioblástica Aguda/diagnóstico , Leucemia Mieloide/diagnóstico , Masculino , Transtornos Mieloproliferativos/complicações , Transtornos Mieloproliferativos/diagnóstico , Prognóstico , Estudos Prospectivos , Fatores de Tempo , Resultado do Tratamento
16.
Blood ; 118(26): 6943-51, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-22045982

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital BM failure syndrome characterized by hypoproliferative anemia, associated physical abnormalities, and a predisposition to cancer. Perturbations of the ribosome appear to be critically important in DBA; alterations in 9 different ribosomal protein genes have been identified in multiple unrelated families, along with rarer abnormalities of additional ribosomal proteins. However, at present, only 50% to 60% of patients have an identifiable genetic lesion by ribosomal protein gene sequencing. Using genome-wide single-nucleotide polymorphism array to evaluate for regions of recurrent copy variation, we identified deletions at known DBA-related ribosomal protein gene loci in 17% (9 of 51) of patients without an identifiable mutation, including RPS19, RPS17, RPS26, and RPL35A. No recurrent regions of copy variation at novel loci were identified. Because RPS17 is a duplicated gene with 4 copies in a diploid genome, we demonstrate haploinsufficient RPS17 expression and a small subunit ribosomal RNA processing abnormality in patients harboring RPS17 deletions. Finally, we report the novel identification of variable mosaic loss involving known DBA gene regions in 3 patients from 2 kindreds. These data suggest that ribosomal protein gene deletion is more common than previously suspected and should be considered a component of the initial genetic evaluation in cases of suspected DBA.


Assuntos
Anemia de Diamond-Blackfan/genética , Deleção de Genes , Proteínas Ribossômicas/genética , Northern Blotting , Pré-Escolar , Feminino , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla/métodos , Técnicas de Genotipagem/métodos , Haploinsuficiência , Humanos , Lactente , Recém-Nascido , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Pediatr Blood Cancer ; 60(2): 230-6, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23024067

RESUMO

BACKGROUND: Platinum agents have been used for a variety of cancers, including pivotal use in pediatric tumors for many years. Oxaliplatin, a third generation platinum, has a different side effect profile and may provide improved activity in pediatric cancers. PROCEDURE: Patients 21 years or younger with progressive or refractory malignant solid tumors, including tumors of the central nervous system were enrolled on this multi-center open label, non-randomized Phase 1 dose escalation study. The study used a standard 3 + 3 dose escalation design with 2 dose levels (85 and 100 mg/m(2) ) with an expansion cohort of 15 additional patients at the recommended dose. Patients received oxaliplatin at the assigned dose level and 5-fluorouracil (5-FU) bolus 400 mg/m(2) followed by a 46-hour 5-FU infusion of 2,400 mg/m(2) every 14 days. The leucovorin dose was fixed at 400 mg/m(2) for all cohorts. RESULTS: Thirty-one evaluable patients were enrolled, 8 at 85 mg/m(2) and 23 at 100 mg/m(2) for a total of 121 courses. The median age was 12 years (range 2-19 years). The main toxicities were hematologic, primarily neutrophils and platelets. The most common non-hematologic toxicities were gastrointestinal. Stable disease was noted in 11 patients (54% of evaluable patients) and 1 confirmed partial response in a patient with osteosarcoma. CONCLUSIONS: The maximum planned dose of oxaliplatin at 100 mg/m(2) per dose in combination with 5-FU and leucovorin was safe and well tolerated and in this patient population. This combination demonstrated modest activity in patients with refractory or relapsed solid tumor and warrants further study. Pediatr Blood Cancer 2013;60:230-236. © 2012 Wiley Periodicals, Inc.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Criança , Pré-Escolar , Relação Dose-Resposta a Droga , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/efeitos adversos , Fluoruracila/farmacocinética , Fluoruracila/uso terapêutico , Humanos , Leucovorina/administração & dosagem , Leucovorina/efeitos adversos , Leucovorina/farmacocinética , Masculino , Dose Máxima Tolerável , Compostos Organoplatínicos/administração & dosagem , Compostos Organoplatínicos/efeitos adversos , Compostos Organoplatínicos/farmacocinética , Compostos Organoplatínicos/uso terapêutico , Oxaliplatina
18.
Cancer ; 118(19): 4806-14, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22392565

RESUMO

BACKGROUND: Children who are treated for myeloid leukemia associated with Down syndrome (DS) experience superior survival compared with children who have myeloid leukemia without DS. To maintain excellent outcomes while avoiding toxicity, the Children's Oncology Group (COG) conducted the phase 3 trial COG A2971, the first trial solely designed to provide uniform treatment of myeloid leukemia in North American children with DS. A2971 eliminated 2 induction drugs and 3 months of maintenance therapy from the standard-timing regimen of dexamethasone, cytarabine, 6-thioguanine, etoposide, and rubidomycin/daunomycin (DCTER) used in the previous study (Children's Cancer Group [CCG] 2891). METHODS: COG A2971 was a multi-institutional, nonrandomized, clinical trial that enrolled 132 patients who had DS with either acute myeloid leukemia (n = 91) or myelodysplastic syndrome (n = 41). RESULTS: The median follow-up was 4.8 years (range, 0.8-8.6 years), the median age at diagnosis was 1.7 years (range, 0.3-13.6 years), and the median white blood cell count was 6200/µL (range, 900-164,900/µL). The remission rate (92.7% ± 6%) was similar to that reported in the CCG 2891 study (91.3% ± 5%; P = .679). The 5-year event free survival (EFS) rate was 79% ± 7% (vs 77% ± 7% in CCG 2891; P = .589), the disease-free survival (DFS) rate was 89% ± 6% (vs 85% ± 6% in CCG 2891; P = .337), and the overall survival rate was 84% ± 6% (vs 79% ± 7% in CCG 2891; P = .302). Induction day-14 bone marrow response trended toward a more favorable outcome (EFS: P = .12). Age >4 years was an adverse risk factor (5-year EFS rate: 33% ± 38% for children aged >4 years [median, 8.5 years; n = 6] vs 81% ± 7% for children ages 0-4 years [median, 1.7 years; n = 126]; P = .001). CONCLUSIONS: The COG A2971 trial reduced the chemotherapy dose and maintained survival to that achieved by the CCG 2891 trial in children who had myeloid leukemia associated with DS.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Síndrome de Down/complicações , Leucemia Mieloide/tratamento farmacológico , Adolescente , Transplante de Medula Óssea , Criança , Pré-Escolar , Citarabina/administração & dosagem , Daunorrubicina/administração & dosagem , Dexametasona/administração & dosagem , Intervalo Livre de Doença , Esquema de Medicação , Etoposídeo/administração & dosagem , Feminino , Humanos , Lactente , Estimativa de Kaplan-Meier , Leucemia Mieloide/complicações , Leucemia Mieloide/cirurgia , Masculino , Tioguanina/administração & dosagem , Resultado do Tratamento
19.
Cancer ; 118(3): 761-9, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21766293

RESUMO

BACKGROUND: The development of antigen-targeted therapies may provide additional options to improve outcomes in children with acute myeloid leukemia (AML). The Children's Oncology Group AAML03P1 trial sought to determine the safety of adding 2 doses of gemtuzumab ozogamicin, a humanized anti-CD33 antibody-targeted agent, to intensive chemotherapy during remission induction and postremission intensification for children with de novo AML. METHODS: AAML03P1 enrolled 350 children with previously untreated AML. Patients with a matched family donor received 3 courses of chemotherapy followed by hematopoietic stem cell transplantation; those without a matched family donor received 5 courses of chemotherapy. Gemtuzumab ozogamicin 3 mg/m(2)/dose was administered on Day 6 of Course 1 and Day 7 of Course 4. RESULTS: Toxicities observed in all courses of therapy were typical of AML chemotherapy regimens, with infection being most common. Patients achieved a complete remission rate of 83% after 1 course and 87% after 2 courses. The mortality rate was 1.5% after the first gemtuzumab ozogamicin-containing induction course and 2.6% after 2 induction courses. The 3-year event-free survival and overall survival rates were 53 ± 6% and 66 ± 5%, respectively. CONCLUSIONS: This trial determined that it is safe and feasible to include gemtuzumab ozogamicin in combination with intensive chemotherapy. The survival rates compare favorably with the recently published results of clinical trials worldwide.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/mortalidade , Adolescente , Adulto , Aminoglicosídeos/administração & dosagem , Amsacrina/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Criança , Pré-Escolar , Citarabina/administração & dosagem , Daunorrubicina/administração & dosagem , Etoposídeo/administração & dosagem , Feminino , Gemtuzumab , Humanos , Lactente , Recém-Nascido , Masculino , Estadiamento de Neoplasias , Projetos Piloto , Prognóstico , Taxa de Sobrevida , Adulto Jovem
20.
Br J Haematol ; 159(3): 299-310, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22934889

RESUMO

CD1A is a cell surface protein expressed on Langerhans cells and cortical thymocytes that could potentially be used as an immunotherapeutic target in Langerhans Cell Histiocytosis (LCH), the cortical subtype of T-cell acute lymphocytic leukaemia (T-ALL) and other CD1A-positive tumours. The monoclonal antibody (mAb) CR2113 was selected from a panel of six fully human mAbs isolated from a semi-synthetic phage display library, based on specificity and avidity against cells expressing CD1 antigen variants. CR2113 recognized CD1A in T-ALL cell lines and patient samples. Confocal microscopy revealed that the CR2113-CD1A complex was internalized at 37°C. Furthermore, while CR2113 induced moderate complement-dependent cytotoxicity (CDC), potent antibody-dependent cell cytotoxicity (ADCC) activity was observed against CD1A expressing cell lines as well as T-ALL cell lines and T-ALL patient samples. In vivo experiments showed that CR2113 as a naked antibody has modest but specific anti-tumour activity against CD1A-expressing tumours. CR2113 is a high-affinity human anti-CD1A mAb with significant ADCC activity. These properties make CR2113 a candidate for clinical diagnostic imaging and therapeutic targeting of LCH as well as potential use in other clinical applications.


Assuntos
Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD1/imunologia , Técnicas de Visualização da Superfície Celular , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/isolamento & purificação , Afinidade de Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Antígenos CD1/metabolismo , Linhagem Celular Tumoral , Humanos , Imunoglobulina G/imunologia , Cinética , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Biblioteca de Peptídeos , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA