Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mod Pathol ; 25(5): 651-60, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22301699

RESUMO

Lymphoplasmacytic lymphomas and marginal zone lymphomas of nodal, extra-nodal and splenic types account for 10% of non-Hodgkin lymphomas. They are similar at the cell differentiation level, sometimes making difficult to distinguish them from other indolent non-Hodgkin lymphomas. To better characterize their genetic basis, we performed array-based comparative genomic hybridization in 101 marginal zone lymphomas (46 MALT, 35 splenic and 20 nodal marginal zone lymphomas) and 13 lymphoplasmacytic lymphomas. Overall, 90% exhibited copy-number abnormalities. Lymphoplasmacytic lymphomas demonstrated the most complex karyotype (median=7 copy-number abnormalities), followed by MALT (4), nodal (3.5) and splenic marginal zone lymphomas (3). A comparative analysis exposed a group of copy-number abnormalities shared by several or all the entities with few disease-specific abnormalities. Gain of chromosomes 3, 12 and 18 and loss of 6q23-q24 (TNFAIP3) were identified in all entities. Losses of 13q14.3 (MIRN15A-MIRN16-1) and 17p13.3-p12 (TP53) were found in lymphoplasmacytic and splenic marginal zone lymphomas; loss of 11q21-q22 (ATM) was found in nodal, splenic marginal zone and lymphoplasmacytic lymphomas and loss of 7q32.1-q33 was found in MALT, splenic and lymphoplasmacytic lymphomas. Abnormalities affecting the nuclear factor kappa B pathway were observed in 70% of MALT and lymphoplasmacytic lymphomas and 30% of splenic and nodal marginal zone lymphomas, suggesting distinct roles of this pathway in the pathogenesis/progression of these subtypes. Elucidation of the genetic alterations contributing to the pathogenesis of these lymphomas may guide to design-specific therapeutic approaches.


Assuntos
Linfonodos/patologia , Linfoma de Zona Marginal Tipo Células B/patologia , Macroglobulinemia de Waldenstrom/patologia , Aberrações Cromossômicas , DNA de Neoplasias/análise , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genômica , Humanos , Hibridização in Situ Fluorescente , Linfoma de Zona Marginal Tipo Células B/genética , Macroglobulinemia de Waldenstrom/genética
2.
Eur J Haematol ; 89(2): 136-44, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22553948

RESUMO

INTRODUCTION: Risk stratification is vital for prognostication and informing treatment decisions in multiple myeloma (MM). We study the prognostic values of the International Staging System (ISS) and underlying cytogenetics in the bortezomib era and assess the impacts of an upfront risk-adapted approach in the treatment of MM. METHODS: We compare the overall survival (OS) of 221 patients with MM diagnosed from 2006 to 2009 (era 2) where upfront bortezomib combination was approved for high-risk MM with the OS of 262 patients diagnosed from 2000 to 2005 (era 1) where bortezomib could only be administered at relapse. High-risk MM is defined by the presence of ISS III disease with renal impairment or adverse cytogenetics. RESULTS: Baseline characteristics were comparable between the 2 eras. At median follow-up of 20 months, 0% and 26% of patients had received frontline bortezomib in eras 1 and 2, respectively. The median OS were 4.2 yrs and not reached for eras 1 and 2, respectively (P = 0.03). On multivariate analysis stratified by era, the most significant prognostic factor shifts from cytogenetics in era 1 to the quality of response in era 2. CONCLUSION: Frontline use of bortezomib in a risk-adapted manner may avert early mortality and is better able to overcome adverse risks compared to its sequential use.


Assuntos
Mieloma Múltiplo/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Ácidos Borônicos/administração & dosagem , Bortezomib , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/tratamento farmacológico , Pirazinas/administração & dosagem , Fatores de Risco , Singapura/epidemiologia , Taxa de Sobrevida , Fatores de Tempo
3.
Blood ; 113(3): 635-45, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18974375

RESUMO

We conducted comprehensive gene expression profiling (GEP) of primary pulmonary mucosa-associated lymphoid tissue (MALT) lymphoma (n = 33) and compared the results to GEP of other B- and T-cell lymphomas and normal lymphocytes to identify novel markers and deregulated pathways. MALT has a prominent T-cell signature and a marginal zone/memory B-cell profile. Four novel transcripts were specifically overexpressed in MALT, and 2 of these, MMP7 and SIGLEC6, were validated at the protein level. GEP also revealed distinct molecular subsets in MALT. One subset, characterized by MALT1 translocations, showed overexpression of nuclear factor-kappaB (NF-KB) pathway genes but also was enriched for chemokine signaling pathways. Another subset showed increased plasma cells and a prominent plasma cell gene signature. By analyzing several genes with very high ("spiked") expression in individual cases, we identified clusters with different biologic characteristics, such as samples with MALT1 translocations having high expression of MALT1 and RARA, samples with plasmacytic differentiation having high FKBP11 expression, and samples with high RGS13 expression tending to have trisomy 3 and reactive follicles. In conclusion, MALT subgroups with distinct pathologic features defined by distinct groups of deregulated genes were identified. These genes could represent novel diagnostic and therapeutic targets.


Assuntos
Perfilação da Expressão Gênica , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Linfoma de Zona Marginal Tipo Células B/diagnóstico , Linfoma de Zona Marginal Tipo Células B/genética , Feminino , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Neoplasias Pulmonares/terapia , Linfoma de Zona Marginal Tipo Células B/terapia , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Análise Serial de Tecidos
4.
Cancer Res ; 67(7): 2982-9, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17409404

RESUMO

Hyperdiploid multiple myeloma (H-MM) is the most common form of myeloma. In this gene expression profiling study, we show that H-MM is defined by a protein biosynthesis signature that is primarily driven by a gene dosage mechanism as a result of trisomic chromosomes. Within H-MM, four independently validated patient clusters overexpressing nonoverlapping sets of genes that form cognate pathways/networks that have potential biological importance in multiple myeloma were identified. One prominent cluster, cluster 1, is characterized by high expression of cancer testis antigen and proliferation-associated genes. Tumors from these patients were more proliferative than tumors in other clusters (median plasma cell labeling index, 3.8; P < 0.05). Another cluster, cluster 3, is characterized by genes involved in tumor necrosis factor/nuclear factor-kappaB signaling and antiapoptosis. These patients have better response to bortezomib as compared with patients within other clusters (70% versus 29%; P = 0.02). Furthermore, for a group of patients generally thought to have better prognosis, a cluster of patients with short survival (cluster 1; median survival, 27 months) could be identified. This analysis illustrates the heterogeneity within H-MM and the importance of defining specific cytogenetic prognostic factors. Furthermore, the signatures that defined these clusters may provide a basis for tailoring treatment to individual patients.


Assuntos
Mieloma Múltiplo/genética , Trissomia , Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Bortezomib , Estudos de Coortes , Dexametasona/uso terapêutico , Diploide , Perfilação da Expressão Gênica , Humanos , Hibridização in Situ Fluorescente , Família Multigênica , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Prognóstico , Biossíntese de Proteínas/genética , Pirazinas/uso terapêutico
5.
Front Pharmacol ; 9: 365, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29773987

RESUMO

Several lines of evidence have demonstrated that deregulated activation of NF-κB plays a pivotal role in the initiation and progression of a variety of cancers including multiple myeloma (MM). Therefore, novel molecules that can effectively suppress deregulated NF-κB upregulation can potentially reduce MM growth. In this study, the effect of celastrol (CSL) on patient derived CD138+ MM cell proliferation, apoptosis, cell invasion, and migration was investigated. In addition, we studied whether CSL can potentiate the apoptotic effect of bortezomib, a proteasome inhibitor in MM cells and in a xenograft mouse model. We found that CSL significantly reduced cell proliferation and enhanced apoptosis when used in combination with bortezomib and upregulated caspase-3 in these cells. CSL also inhibited invasion and migration of MM cells through the suppression of constitutive NF-κB activation and expression of downstream gene products such as CXCR4 and MMP-9. Moreover, CSL when administered either alone or in combination with bortezomib inhibited MM tumor growth and decreased serum IL-6 and TNF-α levels. Overall, our results suggest that CSL can abrogate MM growth both in vitro and in vivo and may serve as a useful pharmacological agent for the treatment of myeloma and other hematological malignancies.

6.
Blood ; 116(7): 1021-2, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20724547
7.
Mol Oncol ; 11(8): 965-980, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28397399

RESUMO

There have been advances in personalized therapy directed by molecular profiles in lung adenocarcinoma, but not in lung squamous cell carcinoma (SCC). The lack of actionable driver oncogenes in SCC has restricted the use of small-molecule inhibitors. Here, we show that SCC cell lines displayed differential sensitivities to belinostat, a pan-histone deacetylase inhibitor. Phosphoproteomic analysis of belinostat-treated SCC cells revealed significant downregulation of the MAPK pathway, along with the induction of apoptosis. In cisplatin-resistant cells that demonstrated aberrant MAPK activation, combined treatment with belinostat significantly inhibited cisplatin-induced ERK phosphorylation and exhibited strong synergistic cytotoxicity. Furthermore, belinostat transcriptionally upregulated the F-box proteins FBXO3 and FBXW10, which directly targeted son of sevenless (SOS), an upstream regulator of the MAPK pathway, for proteasome-mediated degradation. Supporting this, suppression of SOS/ERK pathway by belinostat could be abrogated by inhibiting proteasomal activity either with bortezomib or with siRNA knockdown of FBXO3/FBXW10. Taken together, these preclinical data offer a novel understanding of the epigenetic mechanism by which belinostat exerts its cytotoxicity and supports the combination with cisplatin in clinical settings for chemorefractory SCC tumors.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Ácidos Hidroxâmicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Sulfonamidas/farmacologia , Ubiquitina/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia
8.
Pharmacogenomics ; 6(6): 563-73, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16142997

RESUMO

The advent of new techniques, such as interphase fluorescence in situ hybridization, and, more recently, global array-based gene expression profiling, has accelerated genomic research in myeloma. Distinct biologic subtypes, characterized by unique genetic abnormalities with differing clinical outcomes, have been identified. The identification of these primary genetic defects, and the deregulated oncogenes and pathways in myeloma, has allowed for the development of more targeted therapies. This has led to the discovery of an increased number of active agents in the treatment of myeloma. Genetics also have prognostic importance in myeloma. Recent studies have elucidated a genetic prognostic hierarchy, and have enabled improved definition of the prognostic significance of their interactions. The current challenges are to: improve the dissection of the genetic heterogeneity of the disease; better define progression events; improve the risk stratification of patients; more accurately select patients who will respond well to a particular treatment; and develop more rational combinations of treatment. Genomics will have an important role to play in all of these goals.


Assuntos
Genômica/métodos , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/genética , Progressão da Doença , Quimioterapia Combinada , Previsões , Heterogeneidade Genética , Humanos , Modelos Biológicos , Mieloma Múltiplo/classificação , Mieloma Múltiplo/tratamento farmacológico , Seleção de Pacientes , Prognóstico , Fatores de Risco , Resultado do Tratamento
9.
Clin Lymphoma Myeloma ; 6(3): 200-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16354325

RESUMO

Marked clinical and biologic heterogeneity exists in multiple myeloma (MM). Over the years, many prognostic factors have been identified and several prognostic systems have been proposed. The integration of data from international groups including patients treated with common modalities such as chemotherapy and high-dose therapy culminated in the International Staging System. Recently, genetic information has also been shown to include powerful prognostic factors across different treatment modalities. These advances have facilitated categorization of patients into different risk groups, particularly a subset of patients at high risk with short survival times after current standard therapy. The expanding armamentarium of effective treatments in MM also means that it is now possible to select treatments for patients based on their risk categories. This review will summarize the important prognostic factors identified to date, how they can be used to identify patients at high risk, and their clinical utility in relation to treatment optimization at diagnosis.


Assuntos
Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/patologia , Humanos , Mieloma Múltiplo/tratamento farmacológico , Estadiamento de Neoplasias , Prognóstico , Fatores de Risco
10.
Blood Coagul Fibrinolysis ; 16(1): 75-8, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15650550

RESUMO

Diagnosis of von Willebrand disease requires a combination of quantitative and qualitative tests including the von Willebrand factor ristocetin cofactor assay (vWF:RCo), the von Willebrand factor collagen binding assay (vWF:CB) and von Willebrand factor antigen quantification (vWF:Ag). Genetic factors, especially the ABO blood group, contribute significantly to the variation in vWF levels and function. Recent studies suggest that ethnicity may be another important modulator. We investigated the effect of the ABO blood group on these tests in 52 blood group O and 54 non-group O Chinese blood donors who were well-matched for sex and age distribution. Group O donors had significantly lower vWF:RCo, vWF:Ag and vWF:CB (P < 0.0001). Reduction in vWF:CB was greater than vWF:RCo in group O donors (53 versus 27%). This led to a lower vWF:CB/vWF:Ag ratio (P < 0.0001) in group O donors whereas the vWF:RCo/vWF:Ag ratio was unaffected (P = 0.97). These variations should be taken into consideration when interpreting these results in the Chinese.


Assuntos
Sistema ABO de Grupos Sanguíneos/química , Colágeno/química , Testes Hematológicos , Ristocetina/química , Fator de von Willebrand/análise , Povo Asiático , Coagulação Sanguínea , Feminino , Testes Hematológicos/normas , Humanos , Masculino
12.
Am J Kidney Dis ; 41(3): 692-5, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12612995

RESUMO

Human recombinant erythropoietin is the main treatment for anemia in renal patients. Recently, there have been case reports of pure red blood cell aplasia (PRCA) developing in renal patients administered erythropoietin, probably because of neutralizing antibodies detected in all these patients. All reports were from the West, and most patients were treated with erythropoietin-alpha. Cyclosporine is an immunosuppressive agent used to treat a spectrum of autoimmune conditions. We report a series of Chinese renal patients who developed PRCA after treatment with erythropoietin-alpha, suggesting that this is a problem worldwide. They were treated successfully with cyclosporine and became transfusion independent.


Assuntos
Ciclosporina/uso terapêutico , Eritropoetina/efeitos adversos , Eritropoetina/uso terapêutico , Falência Renal Crônica/tratamento farmacológico , Aplasia Pura de Série Vermelha/induzido quimicamente , Aplasia Pura de Série Vermelha/tratamento farmacológico , Idoso , Povo Asiático , Epoetina alfa , Humanos , Masculino , Proteínas Recombinantes , Aplasia Pura de Série Vermelha/patologia
17.
Environ Mol Mutagen ; 50(8): 697-707, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19739237

RESUMO

Multiple myeloma is the second most common hematological malignancy in the United States. The disease is characterized by an accumulation of clonal plasma cells. Clinically, patients present with anemia, lytic bone lesions, hypercalcaemia, or renal impairment. The genome of the malignant plasma cells is extremely unstable and is typically aneuploid and characterized by a complex combination of structure and numerical abnormalities. The basis of the genomic instability underlying myeloma is unclear. In this regard, centrosome amplification is present in about a third of myeloma and may represent a mechanism leading to genomic instability in myeloma. Centrosome amplification is associated with high-risk features and poor prognosis. Understanding the underlying etiology of centrosome amplification in myeloma may lead to new therapeutic avenues.


Assuntos
Aneuploidia , Proliferação de Células , Centrossomo , Mieloma Múltiplo/patologia , Animais , Humanos , Mieloma Múltiplo/genética
18.
Exp Hematol ; 37(2): 234-44, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19135901

RESUMO

OBJECTIVE: Chromosome 13 deletions (del[13]), detected by metaphase cytogenetics, predict poor outcomes in multiple myeloma (MM), but the gene(s) responsible have not been conclusively identified. We sought to identify tumor-suppressor genes on chromosome 13 using a novel array comparative genomic hybridization (aCGH) strategy. MATERIALS AND METHODS: We identified DNA copy number losses on chromosome 13 using genomic DNA isolated from CD138-enriched bone marrow cells (tumor) from 20 patients with MM, monoclonal gammopathy of undetermined significance, or amyloidosis. We used matched skin biopsy (germline) genomic DNA to control for copy number polymorphisms and a novel aCGH array dedicated to chromosome 13 to map somatic DNA gains and losses at ultra-high resolution (>385,000 probes; median probe spacing 60 bp). We analyzed microarray expression data from an additional 262 patient samples both with and without del[13]. RESULTS: Two distinct minimally deleted regions at 13q14 and 13q13 were defined that affected the RB1 and NBEA genes, respectively. RB1 is a canonical tumor suppressor previously implicated in MM. NBEA is implicated in membrane trafficking in neurons, protein kinase A binding, and has no known role in cancer. Noncoding RNAs on chromosome 13 were not affected by interstitial deletions. Both the RB1 and NBEA genes were deleted in 40% of cases (8 of 20; 5 patients with del[13] detected by traditional methods and 3 patients with interstitial deletions detected by aCGH). Forty-one additional MM patient samples were used for complete exonic sequencing of RB1, but no somatic mutations were found. Along with RB1, NBEA gene expression was significantly reduced in cases with del[13]. CONCLUSIONS: The NBEA gene at 13q13, and its expression are frequently disrupted in MM. Additional studies are warranted to evaluate the role of NBEA as a novel candidate tumor-suppressor gene.


Assuntos
Sequência de Bases/genética , Proteínas de Transporte/genética , Deleção Cromossômica , Cromossomos Humanos Par 13/genética , Mieloma Múltiplo/genética , Proteínas do Tecido Nervoso/genética , Deleção de Sequência/genética , Proteínas Supressoras de Tumor/genética , Idoso , Células da Medula Óssea/metabolismo , Proteínas de Transporte/biossíntese , Mapeamento Cromossômico/métodos , Cromossomos Humanos Par 13/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteínas Supressoras de Tumor/biossíntese
19.
Blood ; 111(3): 1603-9, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18006703

RESUMO

Centrosome amplification is common in myeloma and may be involved in disease pathogenesis. We have previously derived a gene expression-based centrosome index (CI) that correlated with centrosome amplification and was an independent prognostic factor in a small cohort of heterogeneously treated patients. In this study, we validated the prognostic significance of the CI in 2 large cohorts of patients entered into clinical trials and showed that a high CI is a powerful independent prognostic factor in both newly diagnosed and relapsed patients, whether treated by intensive therapy (total therapy II) or novel agents (bortezomib). Tumors with high CI overexpressed genes coding for proteins involved in cell cycle, proliferation, DNA damage, and G(2)-M checkpoints, and associated with the centrosome and kinetochore/ microtubules. In particular, aurora kinases are significantly overexpressed in patients with high CI, with concordant increase in protein expression. Human myeloma cell lines with higher CI are more responsive to treatment with a novel aurora kinase inhibitor. Aurora kinase may represent novel therapeutic targets in these patients with very poor prognosis.


Assuntos
Biomarcadores Tumorais/metabolismo , Centrossomo/efeitos dos fármacos , Centrossomo/enzimologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/enzimologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Aurora Quinases , Biomarcadores Tumorais/genética , Estudos de Coortes , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Instabilidade Genômica/genética , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Prognóstico , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Risco , Taxa de Sobrevida
20.
Blood ; 109(3): 1228-32, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16840723

RESUMO

The biological and clinical implications of p16 gene methylation in multiple myeloma (MM) are still unclear despite previous studies. In this comprehensive study, using methylation-specific PCR (MS-PCR), we show that p16 methylation is relatively common and occurs in monoclonal gammopathy of undetermined significance (MGUS; n=17), smoldering multiple myeloma (SMM; n=40), and MM (n=522) at a prevalence of 24%, 28%, and 34%, respectively. However, p16 methylation does not appear to affect gene expression level. In a large cohort of patients with long-term follow-up information (n=439), there was no difference in overall survival between patients with or without p16 methylation. We also found no association between p16 methylation and the main cytogenetic categories, although it was more common among patients with 17p13.1 deletions (p53 locus), a genetic progression event in MM. In addition, p16 methylation has no apparent effect on the cycle because there was also no difference in the plasma cell labeling index (a direct measurement of proliferation) between patients with and without p16 methylation. Our results question a major role for p16 methylation in the oncogenesis of the PC neoplasm, and we now believe p16 methylation may be a marker for overall epigenetic changes associated with disease progression, with no obvious direct biological or clinical consequences.


Assuntos
Metilação de DNA , Genes p16 , Mieloma Múltiplo/genética , Ciclo Celular , Progressão da Doença , Epigênese Genética , Humanos , Pessoa de Meia-Idade , Mieloma Múltiplo/mortalidade , Taxa de Sobrevida , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA