Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Genome Res ; 22(2): 407-19, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21613409

RESUMO

Most of the studies characterizing DNA methylation patterns have been restricted to particular genomic loci in a limited number of human samples and pathological conditions. Herein, we present a compromise between an extremely comprehensive study of a human sample population with an intermediate level of resolution of CpGs at the genomic level. We obtained a DNA methylation fingerprint of 1628 human samples in which we interrogated 1505 CpG sites. The DNA methylation patterns revealed show this epigenetic mark to be critical in tissue-type definition and stemness, particularly around transcription start sites that are not within a CpG island. For disease, the generated DNA methylation fingerprints show that, during tumorigenesis, human cancer cells underwent a progressive gain of promoter CpG-island hypermethylation and a loss of CpG methylation in non-CpG-island promoters. Although transformed cells are those in which DNA methylation disruption is more obvious, we observed that other common human diseases, such as neurological and autoimmune disorders, had their own distinct DNA methylation profiles. Most importantly, we provide proof of principle that the DNA methylation fingerprints obtained might be useful for translational purposes by showing that we are able to identify the tumor type origin of cancers of unknown primary origin (CUPs). Thus, the DNA methylation patterns identified across the largest spectrum of samples, tissues, and diseases reported to date constitute a baseline for developing higher-resolution DNA methylation maps and provide important clues concerning the contribution of CpG methylation to tissue identity and its changes in the most prevalent human diseases.


Assuntos
Metilação de DNA , Linhagem Celular , Transformação Celular Neoplásica/genética , Análise por Conglomerados , Ilhas de CpG , Epigenômica/métodos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Neoplasias/genética
2.
BMC Cancer ; 14: 33, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24443788

RESUMO

BACKGROUND: Expression of the cold shock protein Y-box protein 1 (YB-1) is associated with deleterious outcome in various malignant diseases. Our group recently showed that the detection of an 18 kDa YB-1 fragment (YB-1/p18) in human plasma identifies patients with malignant diseases. We now tested the prevalence, clinical, and diagnostic value of YB-1/p18 detection in common tumors. METHODS: A newly established monoclonal YB-1 antibody was used to detect YB-1/p18 by immunoblotting in plasma samples from 151 unselected tumor patients, alongside established tumor markers and various diagnostic measures, during evaluation for a cancerous disease and in follow-up studies after therapeutic interventions. RESULTS: Circulating YB-1/p18 was detected in 78% of patients having a tumor disease. YB-1/p18 positivity was highly prevalent in all examined malignancies, including lung cancer (32/37; 87%), breast cancer (7/10; 70%), cancer of unknown primary (CUP; 5/5, 100%) or hematological malignancies (42/62; 68%). Positivity for YB-1/p18 was independent of other routine laboratory parameters, tumor stage, or histology. In comparison to 13 established tumor markers (cancer antigens 15-3, 19-9, 72-4, and 125; carcinoembryonic antigen; cytokeratin fragments 21-1; neuron-specific enolase; alpha-fetoprotein; beta-2-microglobulin; squamous cell carcinoma antigen; thymidine kinase; tissue polypeptide antigen; pro-gastrin-releasing peptide), YB-1/p18 detection within serum samples was the most sensitive general parameter identifying malignant disorders. YB-1/p18 concentrations altered during therapeutic interventions, but did not predict prognosis. CONCLUSIONS: Plasma YB-1/p18 detection has a high specific prevalence in malignancies, thereby providing a novel tool for cancer screening independent of the tumor origin.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias/sangue , Fragmentos de Peptídeos/sangue , Proteína 1 de Ligação a Y-Box/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais , Biomarcadores Tumorais/imunologia , Western Blotting , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Fragmentos de Peptídeos/imunologia , Valor Preditivo dos Testes , Prognóstico , Fatores de Tempo , Proteína 1 de Ligação a Y-Box/imunologia , Adulto Jovem
3.
Int J Cancer ; 131(2): E138-42, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21918973

RESUMO

Aberrant DNA methylation and concomitant transcriptional silencing of death-associated protein kinase 1 (DAPK1) have been demonstrated to be key pathogenic events in chronic lymphocytic leukemia (CLL). In acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), however, the presence of elevated DNA methylation levels has been a matter of continued controversy. Several studies demonstrated highly variable frequencies of DAPK1 promoter methylation by the use of methylation-specific PCR (MSP). By quantitative high-resolution assessment, we demonstrate that aberrant DNA methylation is an extremely rare event in this region. We observed elevated levels just in one out of 246 (0.4%) AML patients, all 42 MDS patients were unmethylated. In conclusion, we present a refined DAPK1 methylation analysis in a large representative patient cohort of AML and MDS patients proofing almost complete absence of elevated DNA methylation. Our results highlight the importance of quantitative measurements for translational research questions on primary patient specimens, particularly.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicas/genética , Idoso , Idoso de 80 Anos ou mais , Proteínas Reguladoras de Apoptose/metabolismo , Células da Medula Óssea , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Estudos de Coortes , Metilação de DNA , Proteínas Quinases Associadas com Morte Celular , Feminino , Humanos , Leucemia Mieloide Aguda/patologia , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/patologia , Regiões Promotoras Genéticas , Análise de Sequência de DNA
4.
Haematologica ; 97(3): 393-401, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22058219

RESUMO

BACKGROUND: The treatment of acute myeloid leukemia of older, medically non-fit patients still poses a highly unmet clinical need, and only few large, prospective studies have been performed in this setting. Given the established activity of hypomethylating agents such as 5-aza-2'-deoxycytidine (decitabine) in myelodysplastic syndromes and acute myeloid leukemia with 20-30% bone marrow blasts, we investigated whether this drug is also active in patients with more than 30% blasts. DESIGN AND METHODS: To evaluate the efficacy and toxicity of decitabine in patients over 60 years old with untreated acute myeloid leukemia ineligible for induction chemotherapy, 227 patients (median age, 72 years), many with comorbidities, adverse cytogenetics and/or preceding myelodysplastic syndrome were treated with this hypomethylating agent. During the initial decitabine treatment (135 mg/m(2) total dose infused intravenously over 72 hours every 6 weeks), a median of two cycles was administered (range, 1-4). All-trans retinoic acid was administered to 100 patients during course 2. Fifty-two patients who completed four cycles of treatment subsequently received a median of five maintenance courses (range, 1-19) with a lower dose of decitabine (20 mg/m(2)) infused over 1 hour on 3 consecutive days every 4-6 weeks. RESULTS: The complete and partial remission rate was 26%, 95% CI (20%, 32%), and an antileukemic effect was noted in 26% of patients. Response rates did not differ between patients with or without adverse cytogenetics; patients with monosomal karyotypes also responded. The median overall survival from the start of decitabine treatment was 5.5 months (range, 0-57.5+) and the 1-year survival rate was 28%, 95%CI (22%,34%). Toxicities were predominantly hematologic. CONCLUSIONS: Decitabine is well tolerated by older, medically non-fit patients with acute myeloid leukemia; myelosuppression is the major toxicity. The response rate and overall survival were not adversely influenced by poor-risk cytogenetics or myelodysplastic syndrome. Because of these encouraging results, randomized studies evaluating single-agent decitabine versus conventional treatment are warranted. The study is registered with the German Clinical Trials Registry, number DRKS00000069.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Azacitidina/análogos & derivados , Leucemia Mieloide Aguda/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/efeitos adversos , Azacitidina/administração & dosagem , Azacitidina/efeitos adversos , Azacitidina/uso terapêutico , Comorbidade , Decitabina , Feminino , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/epidemiologia , Masculino , Pessoa de Meia-Idade , Prevalência , Resultado do Tratamento
5.
Pain Ther ; 11(1): 73-91, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34792789

RESUMO

INTRODUCTION: Peripheral neuropathic pain (PNP) is difficult to treat. Several oral drugs are recommended as first-line treatments. Nevertheless, many patients cannot obtain sufficient pain relief or do not tolerate systemically active treatments. Topical treatments, with a lower risk of systemic side effects such as lidocaine 700 mg medicated plaster, are also recommended in treatment guidelines. This analysis compares the benefit-risk balance of topical 700 mg lidocaine medicated plaster with the benefit-risk balance of oral pregabalin administration for the treatment of PNP following current recommendations on benefit-risk assessment (BRA) methodology. METHODS: The Benefit-Risk Action Team (BRAT) framework was used as structured approach. Selection of key benefits and risks was supported by a patient survey. Published randomized controlled clinical trials were the main source to identify data related to key benefits and risks. The outcome of randomized clinical trials was compared with real-world evidence (RWE) data for consistency. RESULTS: Identified key benefits were pain reduction and improvement in quality of life. Key risks identified were application site reactions, dizziness, confusion, weight gain, peripheral edema, and blurred vision. Overall, there was similarity in key benefits between the comparators; however, a clear advantage regarding key risks in favor of lidocaine 700 mg medicated plaster was observed. This observation was consistent across data from a direct comparison trial, randomized placebo-controlled trials, as well as data from RWE studies. The low number of randomized controlled trials for lidocaine 700 mg medicated plaster was the main limitation. CONCLUSION: Guided by the opinion of patients regarding key benefits and risks deemed important for treatments of peripheral neuropathic pain, our analysis showed that lidocaine 700 mg medicated plaster has a more favorable benefit-risk balance compared to pregabalin (300 and 600 mg daily).

6.
Br J Haematol ; 155(1): 65-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21790528

RESUMO

Methylation in the promoter region of many genes is involved in regulating gene expression patterns. Using the Illumina GoldenGate© methylation assay, we examined the methylation status of 1505 CpG-sites from 807 genes in 32 samples from patients with acute myeloid leukaemia (AML) at diagnosis, nine at relapse and 15 normal controls and performed additional pyrosequencing and semiquantitative methylation specific polymerase chain reaction (MSP) of the GNMT promoter in 113 diagnostic AML samples. We found a gain of overall methylation in AML samples with a further increase at relapse. Regional hypermethylation as assessed by array analysis could be confirmed by both MSP and pyrosequencing. Additionally, large-scale methylation analysis identified interesting candidate genes. Cluster analysis indicated that cytogenetic subgroups seemed to be characterized by additional distinct epigenetic modifications and that basic DNA methylation patterns remain at relapse. Therefore, promoter hypermethylation is a frequent event in AML and is accentuated at relapse. Array-based methylation analysis determined distinct methylation profiles for non-malignant controls and AML samples with specific chromosomal aberrations and can identify target genes for further evaluation.


Assuntos
Metilação de DNA , DNA de Neoplasias/genética , Leucemia Mieloide Aguda/genética , Estudos de Casos e Controles , Aberrações Cromossômicas , Análise por Conglomerados , Ilhas de CpG/genética , Impressões Digitais de DNA/métodos , Feminino , Predisposição Genética para Doença , Glicina N-Metiltransferase/genética , Humanos , Leucemia Mieloide Aguda/patologia , Masculino , Proteínas de Neoplasias/genética , Reação em Cadeia da Polimerase/métodos , Prognóstico , Regiões Promotoras Genéticas/genética , Recidiva
7.
Am J Hematol ; 85(7): 477-81, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20575043

RESUMO

In older patients suffering from acute myelogenous leukemia (AML), aggressive chemotherapy is accompanied with high treatment-related morbidity and mortality. Gemtuzumab ozogamicin (GO), a humanized monoclonal anti-CD33 antibody, represents a well tolerated treatment option, but optimal treatment schedules are still unknown. Additionally, Suppressor of cytokine signaling 3 (SOCS3) inhibits the CD33-induced block on cytokine-induced proliferation. Consequently, a variable response of AML cells to anti-CD33-targeted therapy may be caused by modulation of SOCS3 expression. Twenty-four patients with refractory or relapsed CD33-positive AML received GO as a single agent before or after conventional chemotherapy. The methylation status of the SOCS3 CpG island was assessed by methylation-specific polymerase chain reaction. Response (RR) and overall survival (OS) were significantly higher in 16 patients receiving chemotherapy before GO (RR 81%, OS 14.8 months) compared to three patients who received GO single agent therapy (RR 33%, OS 7.2 months) or 16 with GO before chemotherapy (RR 0% OS 2.2 months, P = 0.01 for RR and P < 0.001 for OS). Methylation of the SOCS3 CpG island was found in 8/24 patients. There was a trend towards a higher RR and longer OS in patients with SOCS3 hypermethylation (RR 86%, OS 25.1 months) compared to unmethylated SOCS3 (RR 56%, OS 10.3 months, P = 0.09). Administration of GO a few days after chemotherapy seems to provide better response and survival compared to administration of GO directly before chemotherapy. The potential role of SOCS3 hypermethylation as a biomarker should be further investigated in patients undergoing GO containing therapies.


Assuntos
Aminoglicosídeos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Metilação de DNA , Leucemia Mieloide Aguda/tratamento farmacológico , Terapia de Salvação/métodos , Proteínas Supressoras da Sinalização de Citocina/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Aminoglicosídeos/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Ilhas de CpG , Esquema de Medicação , Gemtuzumab , Humanos , Leucemia Mieloide Aguda/mortalidade , Pessoa de Meia-Idade , Estudos Retrospectivos , Proteína 3 Supressora da Sinalização de Citocinas , Taxa de Sobrevida , Resultado do Tratamento
8.
Breast Cancer Res ; 11(3): R34, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19527488

RESUMO

INTRODUCTION: Endothelin (EDN) signalling plays a crucial role in cell differentiation, proliferation and migration processes. There is compelling evidence that altered EDN signalling is involved in carcinogenesis by modulating cell survival and promoting invasiveness. To date, most reports have focused on the oncogenic potential of EDN1 and EDN2, both of which are overexpressed in various tumour entities. Here, we aimed at a first comprehensive analysis on EDN3 expression and its implication in human breast cancer. METHODS: EDN3 mRNA expression was assessed by Northern blotting in normal human tissues (n = 9) as well as in matched pairs of normal and tumourous tissues from breast specimens (n = 50). EDN3 mRNA expression in breast cancer was further validated by real-time polymerase chain reaction (PCR) (n = 77). A tissue microarray was used to study EDN3 protein expression in breast carcinoma (n = 150) and normal breast epithelium (n = 44). EDN3 promoter methylation was analysed by methylation-specific PCR in breast cell lines (n = 6) before and after demethylating treatment, normal breast tissues (n = 17) and primary breast carcinomas (n = 128). EDN3 expression and methylation data were statistically correlated with clinical patient characteristics and patient outcome. RESULTS: Loss of EDN3 mRNA expression in breast cancer, as initially detected by array-based expression profiling, could be confirmed by Northern blot analysis (> 2-fold loss in 96%) and real-time PCR (> 2-fold loss in 78%). Attenuated EDN3 expression in breast carcinoma was also evident at the protein level (45%) in association with adverse patient outcome in univariate (P = 0.022) and multivariate (hazard ratio 2.0; P = 0.025) analyses. Hypermethylation of the EDN3 promoter could be identified as the predominant mechanism leading to gene silencing. Reversion of the epigenetic lock by 5-aza-2'-deoxycytidine and trichostatin A resulted in EDN3 mRNA re-expression in vitro. Furthermore, EDN3 promoter hypermethylation was detected in 70% of primary breast carcinomas with significant association to loss of EDN3 mRNA expression (P = 0.005), whilst normal matched breast tissues revealed no EDN3 promoter methylation. CONCLUSIONS: EDN3 is a frequent target of epigenetic inactivation in human breast cancer, potentially contributing to imbalanced EDN signalling commonly found in this disease. The clinical implication supports the view that EDN3, in contrast to EDN1 and EDN2, may act as natural tumour suppressor in the human mammary gland.


Assuntos
Neoplasias da Mama/metabolismo , Endotelina-3/biossíntese , Endotelina-3/genética , Epigênese Genética , Neoplasias da Mama/genética , Diferenciação Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Endotelina-1/biossíntese , Endotelina-2/biossíntese , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais
9.
BMC Cancer ; 9: 173, 2009 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-19500415

RESUMO

BACKGROUND: Inhibitor of differentiation 4 (Id4), a member of the Id gene family is also a dominant negative regulator of basic helix loop helix (bHLH) transcription factors. Some of the functions of Id4 appear to be unique as compared to its other family members Id1, Id2 and Id3. Loss of Id4 gene expression in many cancers in association with promoter hypermethylation has led to the proposal that Id4 may act as a tumor suppressor. In this study we provide functional evidence that Id4 indeed acts as a tumor suppressor and is part of a cancer associated epigenetic re-programming. METHODS: Data mining was used to demonstrate Id4 expression in prostate cancer. Methylation specific polymerase chain reaction (MSP) analysis was performed to understand molecular mechanisms associated with Id4 expression in prostate cancer cell lines. The effect of ectopic Id4 expression in DU145 cells was determined by cell cycle analysis (3H thymidine incorporation and FACS), expression of androgen receptor, p53 and cyclin dependent kinase inhibitors p27 and p21 by a combination of RT-PCR, real time-PCR, western blot and immuno-cytochemical analysis. RESULTS: Id4 expression was down-regulated in prostate cancer. Id4 expression was also down-regulated in prostate cancer line DU145 due to promoter hyper-methylation. Ectopic Id4 expression in DU145 prostate cancer cell line led to increased apoptosis and decreased cell proliferation due in part by an S-phase arrest. In addition to S-phase arrest, ectopic Id4 expression in PC3 cells also resulted in prolonged G2/M phase. At the molecular level these changes were associated with increased androgen receptor (AR), p21, p27 and p53 expression in DU145 cells. CONCLUSION: The results suggest that Id4 acts directly as a tumor suppressor by influencing a hierarchy of cellular processes at multiple levels that leads to a decreased cell proliferation and change in morphology that is possibly mediated through induction of previously silenced tumor suppressors.


Assuntos
Genes Supressores de Tumor , Proteínas Inibidoras de Diferenciação/biossíntese , Neoplasias da Próstata/metabolismo , Apoptose/genética , Ciclo Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Metilação de DNA , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Inibidoras de Diferenciação/genética , Masculino , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores Androgênicos/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
10.
Acta Haematol ; 122(4): 216-20, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19887778

RESUMO

Infiltration of the liver with consecutive severe dysfunction is rarely seen in adult acute lymphoblastic leukemia (ALL). We describe a case of a 32-year-old woman with severe icterus, thrombocytopenia, neutropenia and hepatosplenomegaly. ALL was diagnosed. One day after admission, she developed hepatic encephalopathy with ammonia concentrations in plasma >100 micromol/l. Hepatic infiltration was presumed and chemotherapy was initiated immediately which led to resolution of hepatic encephalopathy and complete hematological remission. Clinicians should be aware of unusual presentations of ALL and difficulties for the application of chemotherapy in patients with liver failure.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Encefalopatia Hepática/tratamento farmacológico , Encefalopatia Hepática/etiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicações , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Adulto , Alanina Transaminase/sangue , Amônia/sangue , Aspartato Aminotransferases/sangue , Bilirrubina/sangue , Ciclofosfamida/administração & dosagem , Dexametasona/administração & dosagem , Feminino , Encefalopatia Hepática/sangue , Encefalopatia Hepática/diagnóstico , Humanos , Idarubicina/administração & dosagem , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Vincristina/administração & dosagem
11.
Onkologie ; 32(7): 400-4, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19556817

RESUMO

BACKGROUND: Chemotherapeutic options are often limited for patients with hepatic dysfunction induced by advanced metastases. The toxicity and efficacy of cytotoxic drugs are often unpredictable due to altered drug activation or inactivation and excretion. Therefore, numerous chemotherapeutic agents should not be administered to patients with liver failure. PATIENTS AND METHODS: We retrospectively analysed 14 patients with solid tumours presenting with liver dysfunction induced by metastasis who were treated with cisplatin single-agent chemotherapy. RESULTS: Tolerance of therapy was acceptable with 1 grade I and 1 grade II renal toxicity. 6 patients experienced grade III haematological toxicity. Partial remission of the disease was observed in 6 cases, and 6 patients could receive combination chemotherapy after improvement of liver function. The median overall survival of the patient cohort was 4.8 months. CONCLUSION: Cisplatin monotherapy may thus be considered as a treatment option for patients with liver dysfunction induced by different solid tumours.


Assuntos
Cisplatino/administração & dosagem , Falência Hepática/etiologia , Falência Hepática/prevenção & controle , Neoplasias Hepáticas , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/administração & dosagem , Feminino , Humanos , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
12.
BMC Cancer ; 8: 154, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18513385

RESUMO

BACKGROUND: Inhibitor of DNA binding/Inhibitor of differentiation 4 (ID4) is a critical factor for cell proliferation and differentiation in normal vertebrate development. ID4 has regulative functions for differentiation and growth of the developing brain. The role of ID1, ID2 and ID3 are expected to be oncogenic due to their overexpression in pancreatic cancer and colorectal adenocarcinomas, respectively. Aside from these findings, loss of ID3 expression was demonstrated in ovarian cancer. The aim of the present study was to reveal the factual role of ID4 in carcinogenesis in more detail, since its role for the pathogenesis of human breast cancer has been discussed controversially, assigning both oncogenic and tumour suppressive functions. METHODS: ID4 promoter methylation, ID4 mRNA expression and ID4 protein expression were analysed in primary human breast cancer specimens using methylation-specific PCR (MSP) (n=170), semiquantitative realtime RT-PCR (n=46) and immunhistochemistry (n=3), respectively. In order to demonstrate a functional association of ID4 promoter methylation with its gene silencing, we performed DNA demethylation analysis with four human breast cell lines using MSP and semiquantitative realtime RT-PCR. In addition, we performed correlations of ID4 promoter methylation with ID4 mRNA and ID4 protein expression in matched samples of breast tumour and corresponding normal tissue. We carried out statistical analyses in order to find correlations between ID4 promoter methylation and clinicopathological parameters. RESULTS: Frequent ID4 promoter methylation was observed in primary breast cancer samples (69%, 117/170). We found a tight correlation (P<0.0001) between ID4 promoter methylation and loss of ID4 expression in primary breast cancer 3 specimens. Demethylating treatment with breast cancer cell lines was associated with clear ID4 mRNA re-expression. Tumours with ID4 promoter methylation showed distinct loss of ID4 expression on both transcription and protein level. Interestingly, ID4 promoter methylation was a factor for unfavourable recurrence-free survival (P=0.036) and increased risk for lymph node metastasis (P=0.030). CONCLUSION: ID4 is indeed a novel tumour suppressor gene in normal human breast tissue and is epigenetically silenced during cancer development, indicating increased risk for tumour relapse. Thus, ID4 methylation status could serve as a prognostic biomarker in human breast cancer.


Assuntos
Neoplasias da Mama/genética , Metilação de DNA , Proteínas Inibidoras de Diferenciação/genética , Recidiva Local de Neoplasia/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Genes Supressores de Tumor , Humanos , Proteínas Inibidoras de Diferenciação/biossíntese , Proteínas Inibidoras de Diferenciação/deficiência , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
13.
Cancer Res ; 66(12): 6361-9, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778214

RESUMO

Optimal reexpression of most genes silenced through promoter methylation requires the sequential application of DNA methyltransferase inhibitors followed by histone deacetylase inhibitors in tumor cell cultures. Patients with myelodysplastic syndrome or acute myeloid leukemia (AML) were treated with the methyltransferase inhibitor 5-azacitidine (aza-CR) followed by the histone deacetylase inhibitor sodium phenylbutyrate. Major responses associated with cytogenetic complete response developed in patients receiving prolonged dosing schedules of aza-CR. Bisulfite sequencing of the p15 promoter in marrow DNA during the first cycle of treatment showed heterogeneous allelic demethylation in three responding patients, suggesting ongoing demethylation within the tumor clone, but no demethylation in two nonresponders. Six of six responding patients with pretreatment methylation of p15 or CDH-1 promoters reversed methylation during the first cycle of therapy (methylation-specific PCR), whereas none of six nonresponders showed any demethylation. Gene demethylation correlated with the area under the aza-CR plasma concentration-time curve. Administration of both drugs was associated with induction of acetylation of histones H3 and H4. This study provides the first demonstration that molecular mechanisms responsible for responses to DNA methyltransferase/histone deacetylase inhibitor combinations may include reversal of aberrant epigenetic gene silencing. The promising percentage of major hematologic responses justifies the testing of such combinations in prospective randomized trials.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Inibidores de Histona Desacetilases , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/enzimologia , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/enzimologia , Acetilação/efeitos dos fármacos , Doença Aguda , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Azacitidina/administração & dosagem , Azacitidina/efeitos adversos , Azacitidina/farmacocinética , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estudos de Viabilidade , Feminino , Histona Desacetilases/genética , Histonas/metabolismo , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Fenilbutiratos/administração & dosagem , Fenilbutiratos/efeitos adversos , Fenilbutiratos/farmacocinética , Regiões Promotoras Genéticas , Resultado do Tratamento
15.
Eur J Haematol ; 79(6): 546-9, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17903214

RESUMO

In the past decade, rituximab in combination with polychemotherapy has become the standard approach in most patients with advanced CD20-positive B-cell lymphoma. In mantle cell lymphoma (MCL), follicular lymphoma and diffuse large B-cell lymphoma, rituximab has been used as monotherapy and in combination with various chemotherapy regimens in different treatment situations. Routinely, rituximab is given intravenously, but other routes of administration have also been described. Here, we report a 64-yr-old woman who was previously treated with three different chemotherapy regimens for stage IV MCL. No sustained clinical and radiological response could be achieved. The patient's general status declined and she developed massive ascites as the dominant clinical problem. Local, intraperitoneal administration of rituximab was initiated as an experimental treatment approach. After 11 doses of rituximab, the general status of the patient improved significantly, ascites resolved completely and computed tomography (CT) scans demonstrated a partial remission of intra-abdominal lymph nodes and splenomegaly. Furthermore, we observed a regression of mediastinal lymph nodes, pleural effusion and centrocytes in peripheral blood as well as improvement of anaemia. The response to the experimental treatment has maintained for more than 6 months. In summary, we observed a sustained local and systemic response to intraperitoneal administration of rituximab in a patient with heavily pretreated advanced MCL.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Ascite/tratamento farmacológico , Infusões Parenterais/métodos , Linfoma de Célula do Manto/tratamento farmacológico , Anemia/tratamento farmacológico , Anticorpos Monoclonais Murinos , Antineoplásicos/administração & dosagem , Feminino , Humanos , Linfonodos/patologia , Metástase Linfática , Pessoa de Meia-Idade , Rituximab , Tomografia Computadorizada por Raios X/métodos , Resultado do Tratamento
16.
Oncogene ; 24(30): 4799-805, 2005 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-15870703

RESUMO

The tissue inhibitor of metalloproteinases-2 (TIMP-2) is known to antagonize matrix metalloproteinase activity and to suppress tumor growth, angiogenesis, invasion and metastasis. We analysed the methylation status of the CpG island in the TIMP-2 promoter region by methylation-specific polymerase chain reaction (MSP) in hematopoietic cell lines. TIMP-2 promoter hypermethylation in the lymphoma cell line Raji and the leukemia cell line KG1a was associated with transcriptional repression. Treatment with the demethylating agent 5-aza-2'-deoxycytidine resulted in TIMP-2 upregulation in both cell lines. TIMP-2 was expressed in the cell lines HL60, U266 and XG1, which carry an unmethylated promoter region. MSP analysis of primary patient samples revealed aberrant methylation of TIMP-2 in 33/90 (36.7%) cases of non-Hodgkin's lymphoma (NHL), but not in normal peripheral blood lymphocytes as well as in nonmalignant bone marrow and lymph nodes. The frequency of TIMP-2 methylation was slightly higher in aggressive NHL subtypes compared to those with an indolent subtype (38.6 versus 33.3%). In contrast, TIMP-2 was not hypermethylated in any of the 40 cases of acute myelogenous leukemia examined. We conclude that promoter hypermethylation of TIMP-2 is a novel epigenetic event in the pathogenesis of lymphoid malignancies and may contribute to a more aggressive NHL phenotype.


Assuntos
Metilação de DNA , Linfoma/genética , Linfoma/metabolismo , Regiões Promotoras Genéticas/genética , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Hematopoese , Humanos , Imuno-Histoquímica , Linfoma/classificação , Inibidor Tecidual de Metaloproteinase-2/análise , Inibidor Tecidual de Metaloproteinase-2/imunologia
17.
Blood Rev ; 20(1): 1-13, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16426940

RESUMO

The term epigenetics defines a heritable alteration in gene expression without an accompanying change in primary DNA sequence. Two major mechanisms that foster epigenetic changes are DNA methylation at cytosine bases within a CpG dinucleotide and post-translational histone modifications. Disruption of the balanced epigenetic network may have significant impact on chromatin structure and transcriptional activity. DNA methylation patterns are profoundly deranged in human cancer and comprise genome-wide losses as well as regional gains in DNA methylation. Hypermethylation of CpG islands within gene promoter regions in collaboration with deacetylation and other modifications of key histone amino acids is associated with transcriptional inactivation and represents, in addition to genetic aberrations, an important mechanism of gene silencing in the pathogenesis of human cancer. These epigenetic events act as alternatives to mutations and deletions to disrupt tumor suppressor gene function. A large number of genes involving fundamental cellular pathways may be affected by aberrant CpG island methylation in association with transcriptional silencing in virtually all tumor types. Altered DNA methylation patterns may serve as biomarkers for cancer detection, assessment of prognosis, and prediction of response to therapy. Furthermore, clinical trials using epigenetically targeted therapies have yielded promising results in hematopoietic malignancies. The ongoing exploration of basic events involved in altered gene transcription patterns and continued clinical investigative studies are helping to develop novel strategies for the diagnosis, prevention, and treatment of human cancer.


Assuntos
Epigênese Genética , Neoplasias Hematológicas/genética , Ilhas de CpG/genética , Metilação de DNA , Terapia Genética , Neoplasias Hematológicas/terapia , Histonas/fisiologia , Humanos , Processamento de Proteína Pós-Traducional
18.
Cancer Res ; 62(20): 5902-5, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12384555

RESUMO

The effects of retinol (vitamin A) depend on its transport and binding to nuclear receptors. The cellular retinol-binding protein 1 (CRBP1) and the retinoic acid receptor beta2 (RARbeta2) are key components of this process. Loss of CRBP1 expression occurs in breast tumors, but the mechanism is not known. We examined whether CpG island hypermethylation of CRBP1 was responsible for its inactivation in cancer cell lines (n = 36) and primary tumors (n = 553) and its relation to RARbeta2 methylation. Hypermethylation of CRBP1 was common in tumors and cancer cell lines, with the highest frequency in lymphoma and gastrointestinal carcinomas. Hypermethylation correlated with loss of CRBP1 mRNA, and in vitro treatment with the demethylating agent 5-aza-2'-deoxycytidine reactivated CRBP1 expression. CRBP1 methylation appeared in premalignant lesions and frequently occurred with RARbeta2 hypermethylation in the same tumors. Finally, we observed that a higher dietary retinol intake was associated with reduced frequencies of methylation of both genes. Epigenetic disruption of CRBP1 is a common event in human cancer that may have important implications for cancer prevention and treatment using retinoids.


Assuntos
Metilação de DNA , Inativação Gênica , Neoplasias/genética , Proteínas de Ligação ao Retinol/genética , Ilhas de CpG , Dieta , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/metabolismo , Regiões Promotoras Genéticas , Receptores do Ácido Retinoico/genética , Proteínas de Ligação ao Retinol/biossíntese , Proteínas Celulares de Ligação ao Retinol , Células Tumorais Cultivadas , Vitamina A/farmacologia
19.
Ann Hematol ; 84 Suppl 1: 39-46, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16231140

RESUMO

Hypermethylation of CpG islands near gene promoter regions is associated with transcriptional inactivation and represents an important mechanism of gene silencing in carcinogenesis. Such epigenetic phenomena can act alongside DNA mutations and deletions to disrupt tumor-suppressor gene function. The methylation status of the promoter-associated CpG islands from 11 well-characterized cancer-related genes was analyzed by methylation-specific polymerase chain reaction in 60 adult patients with acute myelogenous leukemia (AML) at diagnosis. The frequency of aberrant methylation among the patient samples was 45.0% (27/60) for suppressor of cytokine signaling-1, 31.7% (19/60) for p15, 20.0% (12/60) for retinoic acid receptor beta2, 13.3% (8/60) for p73 and E-cadherin, 5.0% (3/60) for O(6)-methylguanine DNA methyltransferase, 3.3% (2/60) for death-associated protein kinase 1 and hMLH1, 1.7% (1/60) for p16, and 0% (0/60) for the tissue inhibitor of matrix metalloproteinases-3 and Ras association domain family 1A. Aberrant DNA methylation was found in AML of all French-American-British subtypes and throughout all cytogenetic risk groups. There appeared to be a trend towards a higher methylation frequency in AML patients with an unfavorable karyotype, but this difference was not statistically significant. Our data indicate that hypermethylation of multiple genes involving fundamental cellular pathways is a common event in AML, which varies greatly in frequency among the genes examined. The accumulation of epigenetic events affecting genes which are involved in regulating cell cycle inhibition, cell adhesion, growth factor signaling, and apoptosis may contribute to the malignant AML phenotype. The growing knowledge of the role of epigenetics in the aberrant silencing of cancer-related genes provides a rationale and molecular basis for targeted therapeutic approaches with demethylating agents in AML.


Assuntos
Ilhas de CpG/genética , Metilação de DNA , Epigênese Genética , Genes Neoplásicos/genética , Leucemia Mieloide Aguda/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Aberrações Cromossômicas , Feminino , Inativação Gênica , Humanos , Cariotipagem , Masculino , Pessoa de Meia-Idade , Prognóstico , Fatores de Risco
20.
Methods Mol Med ; 113: 279-91, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15968111

RESUMO

Methylation-specific polymerase chain reaction (MSP) is a method that can rapidly assess the methylation status of virtually any group of CpG sites within a CpG island, independent of the use of methylation-sensitive restriction enzymes. This assay entails the initial modification of DNA by sodium bisulfite, converting all unmethylated cytosines to uracils but leaving the methylated cytosines unchanged, followed by subsequent amplification with primers specific for methylated vs unmethylated DNA. The great sensitivity of this technique allows qualitative methylation analysis from DNA obtained not only from fresh frozen tissues, peripheral blood, bone marrow, or body fluids but also from paraffin-embedded samples. It is a rapid and cost-effective method that does not require radioactive reagents and can be used for the analysis of a large number of clinical samples.


Assuntos
Metilação de DNA , Mieloma Múltiplo/genética , Reação em Cadeia da Polimerase/métodos , Sequência de Bases , Primers do DNA , DNA de Neoplasias/genética , Fosfatos de Dinucleosídeos/genética , Amplificação de Genes , Genes Supressores de Tumor , Humanos , Mieloma Múltiplo/patologia , Moldes Genéticos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa