Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Clin Invest ; 91(5): 2288-95, 1993 May.
Artigo em Inglês | MEDLINE | ID: mdl-8486788

RESUMO

Differentiation therapy may provide an alternative for treatment of cancers that do not respond to cytotoxic chemotherapy or hormonal manipulations. This hypothesis led us to evaluate the effect of a nontoxic differentiation inducer, sodium phenylacetate (NaPA), on hormone-refractory prostate cancer, the second most common cause of cancer deaths in men. NaPA treatment of androgen-independent PC3 and DU145 prostate cell lines, like that of hormone-responsive LNCaP cultures, resulted in dose-dependent inhibition of cell proliferation. Similar treatments were not significantly inhibitory to replicating normal endothelial cells and skin fibroblasts. In addition to the selective cytostatic effect, NaPA induced reversion of the prostatic cells to a nonmalignant phenotype, evidenced by their reduced invasiveness and loss of tumorigenicity in athymic mice. Phenotypic reversion was accompanied by alterations in gene expression, including selective reduction in tumor growth factor-beta 2 mRNA levels and increased amounts of class I major histocompatibility complex HLA transcripts. Furthermore, there was a decrease in tumor-associated proteolysis mediated by urokinase plasminogen activator, a molecular marker of disease progression in humans. When tumor cells were treated with NaPA together with suramin, a drug with demonstrable activity in patients, there was complete abrogation of cell growth under conditions in which each treatment alone produced only a partial effect. The in vitro antineoplastic activity was observed with drug concentrations that have been achieved in humans with no significant toxicities, suggesting that PA, used alone or in combination with other antitumor agents, warrants evaluation in the treatment of advanced prostatic cancer.


Assuntos
Divisão Celular/efeitos dos fármacos , Fenilacetatos/farmacologia , Neoplasias da Próstata/patologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Feminino , Glutamina/farmacologia , Antígeno HLA-A3/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia , Transplante de Neoplasias , Fenótipo , Neoplasias da Próstata/genética , RNA Neoplásico/genética , RNA Neoplásico/isolamento & purificação , Suramina/farmacologia , Fator de Crescimento Transformador beta/genética , Transplante Heterólogo , Células Tumorais Cultivadas
2.
Mol Cell Biol ; 7(6): 2196-200, 1987 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2439904

RESUMO

Prolonged alpha/beta interferon (IFN-alpha/beta) treatment of NIH 3T3 cells transformed by a long terminal repeat-activated Ha-ras proto-oncogene resulted in revertants that maintained a nontransformed phenotype long after IFN treatment had been discontinued. Cloned persistent revertants (PRs) produced large amounts of the ras-encoded p21 and were refractile to transformation by EJras DNA and by transforming retroviruses which carried the v-Ha-ras, v-Ki-ras, v-abl, or v-fes oncogene. Transient treatment either in vitro or in vivo with cytidine analogs that alter gene expression by inhibiting DNA methylation resulted in transformation of PR, but not of NIH 3T3, cells. The PR retransformants reverted again with IFN, suggesting that DNA methylation is involved in IFN-induced persistent reversion.


Assuntos
Azacitidina/farmacologia , Transformação Celular Neoplásica , Interferon Tipo I/farmacologia , Proto-Oncogenes , Animais , Transformação Celular Neoplásica/efeitos dos fármacos , Células Cultivadas , Células Clonais , Células L , Camundongos , Camundongos Endogâmicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Proto-Oncogenes/efeitos dos fármacos , RNA Mensageiro/genética , Retroviridae/genética
3.
Mol Cell Biol ; 13(7): 4416-22, 1993 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8321241

RESUMO

Alterations in intracellular glutathione (GSH) content are known to affect intrinsic responses to ionizing radiation. More recently, it became apparent that radiation responses may depend also on the expression of specific oncogenes, including ras. These findings, suggesting a possible link between GSH and ras, led us to examine the effect of various GSH modulators on ras expression. Treatment of c-Ha-ras-transformed NIH 3T3 cells with L-buthionine S'R'-sulfoximine, dimethylfumarate, or N',N'-1,3-bis(trans-4-hydroxycyclohexyl)-N'-nitrosourea resulted in dose- and time-dependent reduction in ras mRNA steady-state levels followed by a decrease in ras-encoded p21 protein production. The effect on ras correlated with the extent of GSH decline, was common to different members of the ras family, and was independent of the mode of oncogene activation or cell phenotype. Indeed, similar drug effects were observed with murine cells in which overexpression of the c-Ha-ras proto-oncogene was due to transcriptional activation (PR4, nontumorigenic) or gene amplification (NIH 136, tumorigenic) and with malignant cells expressing a mutated Ha-ras (RS504). Moreover, N-ras, EJras, and Ki-ras in human tumor cells were similarly affected. Molecular analysis revealed a significant decrease in ras mRNA half-life in cells subjected to GSH inhibition, an effect that required de novo protein synthesis, but there was no change in the rate of gene transcription. These results indicate that pharmacological manipulation of cellular GSH content can down-regulate ras expression at the posttranscriptional level by destabilizing ras transcripts. The potential clinical implications are discussed.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes ras , Glutationa/metabolismo , Processamento Pós-Transcricional do RNA , Células 3T3 , Animais , Northern Blotting , Butionina Sulfoximina , Ciclo Celular , Cicloeximida/farmacologia , Regulação para Baixo , Humanos , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacologia , Camundongos , Proteína Oncogênica p21(ras)/genética , Proteína Oncogênica p21(ras)/metabolismo , Proto-Oncogene Mas , Transcrição Gênica , Células Tumorais Cultivadas
4.
Mol Cell Biol ; 15(11): 6150-9, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7565767

RESUMO

The Epstein-Barr Virus (EBV) latency C promoter (Cp) is the origin of transcripts for six viral proteins. The promoter is active in lymphoblastoid B-cell lines but silent in many EBV-associated tumors and tumor cell lines. In these latter cell lines, the viral episome is hypermethylated in the vicinity of this promoter. We show that in such a cell line (Rael, a Burkitt's lymphoma line), 5-azacytidine inhibits DNA methyltransferase, brings about demethylation of EBV genomes, activates Cp transcription, and induces the expression of EBNA-2. Investigation of the phenomenon demonstrates the importance of the methylation status of a particular CpG site for the regulation of the Cp: (i) genomic sequencing shows that this site is methylated when the Cp is inactive and is not methylated when the promoter is active; (ii) methylation or transition mutation at this site abolishes complex formation with a cellular binding activity (CBF2) as determined by electrophoretic mobility shift analyses, competition binding analyses, and DNase I footprinting; and (iii) a single C --> T transition mutation at this site is associated with a marked reduction (> 50-fold) of transcriptional activity in a reporter plasmid. Thus, the CBF2 binding activity is shown to be methylation sensitive and crucial to EBNA-2-mediated activation of the Cp.


Assuntos
Azacitidina/farmacologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Herpesvirus Humano 4/genética , Latência Viral , Antígenos Virais/genética , Sequência de Bases , Pegada de DNA , DNA Viral/química , DNA Viral/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr , Metilação , Dados de Sequência Molecular , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica/efeitos dos fármacos
5.
Cancer Res ; 52(7): 1988-92, 1992 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1372534

RESUMO

Sodium phenylacetate was found to affect the growth and differentiation of tumor cells in vitro at concentrations that have been achieved in humans with no significant adverse effects. Treatment of promyelocytic leukemia HL-60 cells resulted in the rapid decline of myc oncogene expression followed by growth arrest and granulocyte differentiation. Phenylacetate also induced highly efficient adipocyte conversion in immortalized mesenchymal C3H 10T1/2 cultures; yet, unlike the differentiating chemotherapeutic drug 5-aza-2'-deoxycytidine, phenylacetate did not cause neoplastic transformation in these susceptible cells. The results indicate that phenylacetate is both effective in inducing tumor cell maturation and free of cytotoxic and carcinogenic effects, a combination that warrants attention to its potential use in cancer intervention.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Transformação Celular Neoplásica , Fenilacetatos/farmacologia , Tecido Adiposo/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Linhagem Celular , Decitabina , Expressão Gênica/efeitos dos fármacos , Genes myc/efeitos dos fármacos , Humanos , Leucemia Promielocítica Aguda , Camundongos , Camundongos Nus , RNA Ribossômico/efeitos dos fármacos , RNA Ribossômico/genética
6.
Cancer Res ; 48(18): 5157-62, 1988 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-2457435

RESUMO

Elevation in Ha-ras expression, due to transcriptional activation or gene amplification, is associated with oncogenic transformation of NIH 3T3 cells. We have previously shown that murine interferon (IFN)-alpha/beta induced phenotypic reversion of NIH 3T3 cells transformed by long terminal repeat (LTR)-activated c-Ha-ras. The revertants produced decreased amounts of ras-encoded Mr 21,000 protein. We have now determined the molecular level at which LTR-ras regulation occurred. Nuclear run-on experiments revealed a selective inhibition of ras transcription in IFN-treated revertants. There was no apparent additional posttranscriptional control by IFN as judged by the unchanged half-life of ras transcripts. Inhibition of ras RNA synthesis was seen only in conjunction with long-term IFN treatment and was limited to the revertants, a population of cells that maintained sensitivity to IFN during the prolonged exposure. The reduction in ras activity appeared responsible in part for the loss of tumorigenicity in treated cells and was stable for several weeks after treatment had been discontinued.


Assuntos
Transformação Celular Neoplásica/patologia , Genes ras , Interferons/farmacologia , Transcrição Gênica/efeitos dos fármacos , Animais , Linhagem Celular , Cicloeximida/farmacologia , Amplificação de Genes , Regulação da Expressão Gênica , Meia-Vida , Camundongos , Peso Molecular , Fenótipo , Sequências Repetitivas de Ácido Nucleico
7.
Cancer Res ; 51(1): 324-30, 1991 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-1703037

RESUMO

The hypomethylating chemotherapeutic drug 5-aza-2'-deoxycytidine (5AzadC) has been shown to induce cell differentiation in some systems, while promoting neoplastic transformation in others. Using both in vitro and in vivo models, we have explored the relationship between oncogene expression and the susceptibility of cells to malignant transformation by 5AzadC. The study involved several nontumorigenic subclones of NIH3T3 fibroblasts, including cells transfected with deregulated c-myc, as well as phenotypic revertants expressing v-Ki-ras or long terminal repeat-activated c-Ha-ras. Transient 5AzadC treatment of the oncogene-bearing cell lines was associated with a rapid and efficient neoplastic transformation. In some cases, over 50% of the cell population exhibited loss of contact inhibition of growth within 1 week of treatment. The transformants were capable of forming s.c. tumors and experimental lung metastases in recipient nude mice. In contrast, 5AzadC failed to induce malignant properties in control 3T3 cultures transfected with the bacterial neor gene; rather, treatment of these cells was associated with differentiation into adipocytes and myotubes. The differential response to 5AzadC was also observed in vivo, in mice first inoculated s.c. with the premalignant cells and then treated with 5AzadC 24 h later. In agreement with the in vitro model, tumor development in mice correlated with the presence of cells with activated ras or myc oncogenes. Cytidine analogs that do not inhibit DNA methylation (i.e., 6-azacytidine and 1-beta-D-arabinofuranosyl cytosine) had no effect on cell phenotype. The results indicate that exposure of cells to 5AzadC can lead to tumor progression both in vitro and in vivo and suggest that preexisting alterations in oncogene expression may facilitate the evolution of cancerous growth induced by hypomethylating agents.


Assuntos
Azacitidina/análogos & derivados , Transformação Celular Neoplásica/efeitos dos fármacos , Genes ras , Proteínas Proto-Oncogênicas c-myc/genética , Proto-Oncogenes , Animais , Azacitidina/farmacologia , Northern Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cocarcinogênese , DNA/metabolismo , Decitabina , Metaloproteinase 3 da Matriz , Metaloendopeptidases/genética , Metilação , Camundongos , Camundongos Nus , Metástase Neoplásica , RNA Mensageiro/genética
8.
Cancer Res ; 53(20): 4890-5, 1993 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8402677

RESUMO

There is a pressing need for in vivo models in which potential antitumor agents can be tested for their ability to inhibit the growth and metastatic spread of human sarcomas. A recent advance in this regard has been the development of a v-Ki-ras-oncogene-transformed human osteosarcoma cell line (KRIB) that efficiently colonizes the lungs of athymic nude mice when cells (1 x 10(5)) are administered by i.v. injection. In the present study, we have utilized this cell line to develop a spontaneous metastasis model in which a small number of tumor cells are injected into the tibial bones of athymic mice. When as few as 1000 KRIB cells are orthotopically implanted into the tibial bones of nude mice, bone tumors, which are radiographically and histologically similar to primary human osteosarcoma, develop within 4 weeks. Furthermore, as in the human disease, cells from these primary tumors subsequently seed the animals' lungs, resulting in reproducible and quantifiable pulmonary metastasis evident both upon gross inspection of the lungs and histologically 6 weeks after tumor inoculation. Surgical amputation of the tumor inoculation site up to 2 weeks after tumor injection prevents pulmonary metastasis, indicating that substantial local (tibial) growth and invasion of the primary tumor for at least 2 weeks is required for subsequent metastasis. Implantation of s.c. 5000 KRIB cells fails to produce local or metastatic tumors. We anticipate that this model will prove to be a powerful tool with which to study the mechanisms of human osteosarcoma growth and pulmonary metastasis, and to assess the efficacy of promising therapeutic agents.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias Pulmonares/secundário , Osteossarcoma/secundário , Animais , Linhagem Celular , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Metástase Neoplásica , Osteossarcoma/patologia , Tíbia , Transplante Heterólogo , Células Tumorais Cultivadas
9.
Cancer Res ; 54(7): 1766-71, 1994 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-7511051

RESUMO

MAGE-1 is a gene that encodes an antigen on a melanoma cell line that is recognized by cytolytic T-cells. We have used a reverse transcription-polymerase chain reaction assay to analyze expression of the MAGE-1 gene by cell lines from different types of tumors, melanomas from different stages of disease progression, normal diploid cell lines, and melanocyte and nevus tissue from which malignant melanomas are derived. MAGE-1 is expressed by melanoma tissue from all stages of disease, but not melanocytes, nevus tissue, or any normal diploid cell line tested. A fraction of tumor lines derived from various epithelial and neuroectodermal malignancies expressed MAGE-1 but not peripheral blood cells from patients with melanoma. 5-Aza-2'-deoxycytidine (DAC), a demethylating agent, was capable of inducing MAGE-1 expression by a MAGE-1-negative melanoma cell line 888-mel as well as by a number of other melanoma cell lines. At an optimum concentration of 1 microM DAC, MAGE-1 expression was detectable by 24 h, plateaued by 72 h, but remained high for two weeks after removal of DAC from treated 888-mel cells, consistent with induction by demethylation. With the exception of tumor-infiltrating leukocytes, no normal diploid cell line could be induced with DAC to upregulate MAGE-1 expression. DAC-treated 888-mel cells were lysed by a MAGE-1-specific major histocompatibility complex restricted cytolytic T-cell clone, whereas control untreated cells were not, suggesting that production of the antigen encoded by the MAGE-1 gene was induced by DAC and that it was presented in association with major histocompatibility complex class I molecules at the cell surface for T-cell recognition.


Assuntos
Antígenos de Neoplasias/biossíntese , Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Citotoxicidade Imunológica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Melanoma/metabolismo , Proteínas de Neoplasias , Antígenos de Neoplasias/genética , Azacitidina/farmacologia , Sequência de Bases , Linhagem Celular , Primers do DNA , Decitabina , Feminino , Gliceraldeído-3-Fosfato Desidrogenases/biossíntese , Gliceraldeído-3-Fosfato Desidrogenases/genética , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/metabolismo , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Melanócitos/citologia , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Melanoma/patologia , Antígenos Específicos de Melanoma , Dados de Sequência Molecular , Metástase Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Reação em Cadeia da Polimerase/métodos , RNA Neoplásico/genética , RNA Neoplásico/isolamento & purificação , Células Tumorais Cultivadas
10.
Cancer Res ; 54(4): 891-5, 1994 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8313377

RESUMO

Phenylacetate, a deaminated metabolite of phenylalanine, has been implicated in damage to immature brain in phenylketonuria. Because primary brain tumors are highly reminiscent of the immature central nervous system, these neoplasms should be equally vulnerable. We show here that sodium phenylacetate can induce cytostasis and reversal of malignant properties of cultured human glioblastoma cells, when used at pharmacological concentrations that are well tolerated by children and adults. Treated tumor cells exhibited biochemical alterations similar to those observed in phenylketonuria-like conditions, including selective decline in de novo cholesterol synthesis from mevalonate. Because gliomas, but not mature normal brain cells, are highly dependent on mevalonate for production of sterols and isoprenoids vital for cell growth, sodium phenylacetate would be expected to affect tumor growth in vivo while sparing normal tissues. Systemic treatment of rats bearing intracranial gliomas resulted in significant tumor suppression with no apparent toxicity to the host. The data indicate that phenylacetate, acting through inhibition of protein prenylation and other mechanisms, may offer a safe and effective novel approach to treatment of malignant gliomas and perhaps other neoplasms as well.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Glioma/tratamento farmacológico , Fenilacetatos/farmacologia , Fenilcetonúrias/patologia , Animais , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Feminino , Glioma/patologia , Humanos , Ácido Mevalônico/metabolismo , Camundongos , Camundongos Nus , Fenilacetatos/uso terapêutico , Prenilação de Proteína , Ratos , Ratos Endogâmicos F344 , Células Tumorais Cultivadas
11.
Cancer Res ; 54(11): 2923-7, 1994 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8187079

RESUMO

Phenylacetate is a naturally occurring plasma component that suppresses the growth of tumor cells and induces differentiation in vitro. To evaluate the in vivo potential and preventive and therapeutic antitumor efficacy of sodium phenylacetate against malignant brain tumors, Fischer 344 rats (n = 50) bearing cerebral 9L gliosarcomas received phenylacetate by continuous s.c. release starting on the day of tumor inoculation (n = 10) using s.c. osmotic minipumps (550 mg/kg/day for 28 days). Rats with established brain tumors (n = 12) received continuous s.c. phenylacetate supplemented with additional daily i.p. dose (300 mg/kg). Control rats (n = 25) were treated in a similar way with saline. Rats were sacrificed during treatment for electron microscopic studies of their tumors, in vivo proliferation assays, and measurement of phenylacetate levels in the serum and cerebrospinal fluid. Treatment with phenylacetate extended survival when started on the day of tumor inoculation (P < 0.01) or 7 days after inoculation (P < 0.03) without any associated adverse effects. In the latter group, phenylacetate levels in pooled serum and cerebrospinal fluid samples after 7 days of treatment were in the therapeutic range as determined in vitro (2.45 mM in serum and 3.1 mM in cerebrospinal fluid). Electron microscopy of treated tumors demonstrated marked hypertrophy and organization of the rough endoplasmic reticulum, indicating cell differentiation, in contrast to the scant and randomly distributed endoplasmic reticulum in tumors from untreated animals. In addition, in vitro studies demonstrated dose-dependent inhibition of the rate of tumor proliferation and restoration of anchorage dependency, a marker of phenotypic reversion. Phenylacetate, used at clinically achievable concentrations, prolongs survival of rats with malignant brain tumors through induction of tumor differentiation. Its role in the treatment of brain tumors and other cancers should be explored further.


Assuntos
Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/prevenção & controle , Gliossarcoma/mortalidade , Gliossarcoma/prevenção & controle , Fenilacetatos/uso terapêutico , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Divisão Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Gliossarcoma/metabolismo , Gliossarcoma/patologia , Gliossarcoma/ultraestrutura , Microscopia Eletrônica , Transplante de Neoplasias , Fenilacetatos/sangue , Fenilacetatos/líquido cefalorraquidiano , Ratos , Ratos Endogâmicos F344 , Ensaio Tumoral de Célula-Tronco
12.
Cancer Res ; 54(7): 1690-4, 1994 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8137283

RESUMO

Phenylacetate has recently been shown to suppress tumor growth and promote differentiation in experimental models. A phase I trial of phenylacetate was conducted in 17 patients with advanced solid tumors. Each patient received a single i.v. bolus dose followed by a 14-day continuous i.v. infusion of the drug. Twenty-one cycles of therapy were administered at four dose levels, achieved by increasing the rate of the continuous i.v. infusion. Phenylacetate displayed nonlinear pharmacokinetics [Km = 105.1 +/- 44.5 (SD) microgram/ml, Vmax = 24.1 +/- 5.2 mg/kg/h and Vd = 19.2 +/- 3.3 L]. There was also evidence for induction of drug clearance. Ninety-nine % of phenylacetate elimination was accounted for by conversion to phenylacetylglutamine, which was excreted in the urine. Continuous i.v. infusion rates resulting in serum phenylacetate concentrations exceeding Km often resulted in rapid drug accumulation and dose-limiting toxicity, which consisted of reversible central nervous system depression, preceded by emesis. Three of nine patients with metastatic, hormone-refractory prostate cancer maintained stable prostatic specific antigen levels for more than 2 months; another had less bone pain. One of six patients with glioblastoma multiforme, whose steroid dosage has remained unchanged for the duration of therapy, has sustained functional improvement for more than 9 months. The use of adaptive control with feedback for the dosing of each patient enabled us to safely maintain stable phenylacetate concentrations up to the range of 200-300 micrograms/ml, which resulted in clinical improvement in some patients with advanced disease.


Assuntos
Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Neoplasias/tratamento farmacológico , Fenilacetatos/farmacocinética , Fenilacetatos/toxicidade , Adulto , Idoso , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Esquema de Medicação , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glutamina/sangue , Humanos , Infusões Intravenosas , Injeções Intravenosas , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/patologia , Fenilacetatos/sangue
13.
Leukemia ; 13(8): 1243-53, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10450753

RESUMO

The aromatic fatty acid phenylbutyrate (PB) induces cytostasis, differentiation, and apoptosis in primary myeloid leukemic cells at clinically achievable concentrations. In the present study, we have investigated the structural and cellular basis for PB-induced cytostasis, using the ML-1 human myeloid leukemia cell line as a model system. PB induced a dose-dependent increase in cells in G1 with a corresponding decrease in cells in S-phase of the cell cycle. At comparable doses, PB induced expression of CD11b, indicating myeloid differentiation. At higher doses, the drug induced apoptosis. The antitumor activity was independent of the aromatic ring, as butyric acid (BA) was of equal or greater potency at producing these biological changes. In contrast, shortening of the fatty acid carbon chain length, as demonstrated with phenylacetate (PA), significantly diminished drug potency. Consistent with their effects on cell cycle, PB and BA, but not PA, induced the cyclin-dependent kinase inhibitor, p21(WAF1/CIP1), and led to the appearance of hypophosphorylated Rb, suggesting a role for p21(WAF1/CIP1) in PB-induced cytostasis. Therefore, it appears that the fatty acid moiety of PB, rather than its aromatic ring, is critical for its activity in myeloid leukemic cells. These data provide a potential mechanistic basis for the increased potency of PB over PA previously demonstrated in primary leukemic samples, and support the further clinical development of PB in the treatment of hematologic malignancies.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/patologia , Fenilbutiratos/farmacologia , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Fenilbutiratos/química , Fenilbutiratos/uso terapêutico , Relação Estrutura-Atividade , Células Tumorais Cultivadas
14.
Clin Cancer Res ; 3(10): 1755-62, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9815560

RESUMO

Sodium phenylacetate (PA) and sodium phenylbutyrate (PB) are aromatic fatty acids that can effect differentiation in a variety of cell lines at doses that may be clinically attainable. We have studied the impact of these two agents on lineage- and differentiation stage-specific antigen expression, proliferation, apoptosis, and clonogenic cell survival in primary cultures of bone marrow samples from patients with myeloid neoplasms at presentation and in remission and from normal volunteers. PB inhibited the proliferation of primary acute myeloid leukemia cells in suspension culture with an ID50 of 6.6 mM, similar to its ED50 in cell lines. At higher doses (>/=5 mM), PB also induced apoptosis. PB inhibited clonogenic leukemia cell growth with a median ID50 of less than 2 mM; however, colony-forming units-granulocyte/macrophage from patients with myelodysplasia and normal volunteers were inhibited with a similar ID50. In contrast to PB, its metabolite PA had no significant effect on either acute myeloid leukemia proliferation or apoptosis. Expression of the monocytic marker CD14 was increased in monocytic and myelomonocytic leukemias in response to PB, and to a lesser extent, PA. Surprisingly, both agents appeared to increase expression of the progenitor cell antigen CD34, as well as the DR locus of the human leukocyte antigen. These data indicate that PB, but not its metabolite PA, has significant cytostatic and differentiating activity against primary neoplastic myeloid cells at doses that may be achievable clinically.


Assuntos
Leucemia Mieloide/patologia , Fenilacetatos/farmacologia , Fenilbutiratos/farmacologia , Doença Aguda , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Apoptose/efeitos dos fármacos , Medula Óssea/patologia , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem da Célula , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Síndromes Mielodisplásicas/patologia , Células Tumorais Cultivadas/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
15.
Clin Cancer Res ; 1(8): 865-71, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9816056

RESUMO

Differentiation inducers selected for their low cytotoxic and genotoxic potential could be of major value in chemoprevention and maintenance therapy. We focus here on phenylacetate, a naturally occurring plasma component recently shown to affect the growth and differentiation of established neoplasms in experimental models. The ability of phenylacetate to prevent carcinogenesis by the chemotherapeutic hypomethylating drug 5-aza-2'-deoxycytidine (5AzadC) was tested in vitro and in mice. Transient exposure of immortalized, but poorly tumorigenic ras-transformed 4C8 fibroblasts to 5AzadC resulted in neoplastic transformation manifested by loss of contact inhibition of growth, acquired invasiveness, and increased tumorigenicity in athymic mice. The latter was associated with elevation in ras expression and a decline in collagen biosynthesis. These profound phenotypic and molecular changes were prevented by a simultaneous treatment with phenylacetate. Protection from 5AzadC carcinogenesis by phenylacetate was: (a) highly efficient despite DNA hypomethylation by both drugs, (b) free of cytotoxic and genotoxic effects, (c) stable after treatment was discontinued, and (d) reproducible in vivo. Whereas athymic mice bearing 4C8 cells developed fibrosarcomas following a single i.p. injection with 5AzadC, tumor development was significantly inhibited by systemic treatment with nontoxic doses of phenylacetate. Phenylacetate and its precursor suitable for oral administration, phenylbutyrate, may thus represent a new class of chemopreventive agents, the efficacy and safety of which should be further evaluated.


Assuntos
Anticarcinógenos/farmacologia , Azacitidina/análogos & derivados , Carcinógenos/toxicidade , Transformação Celular Neoplásica/efeitos dos fármacos , Quimioprevenção , Genes ras , Neoplasias Experimentais/patologia , Fenilacetatos/farmacologia , Células 3T3 , Animais , Azacitidina/toxicidade , Divisão Celular/efeitos dos fármacos , Células Clonais , Colágeno , Metilação de DNA/efeitos dos fármacos , Decitabina , Combinação de Medicamentos , Feminino , Laminina , Camundongos , Camundongos Nus , Invasividade Neoplásica/prevenção & controle , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/prevenção & controle , Proteoglicanas
16.
Clin Cancer Res ; 6(3): 933-41, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10741718

RESUMO

Aromatic fatty acids, of which phenylacetate is a prototype, constitute a class of low toxicity drugs with demonstrated antitumor activity in experimental models and in humans. Using in vitro models, we show here a tight correlation between tumor growth arrest by phenylacetate and activation of peroxisome proliferator-activated receptor gamma (PPARgamma), a member of the nuclear receptor superfamily. In support are the following observations: (a) the efficacy of phenylacetate as a cytostatic agent was correlated with pre-treatment levels of PPARgamma, as documented using established tumor lines and forced expression models; (b) in responsive tumor cells, PPARgamma expression was up-regulated within 2-9 h of treatment preceding increases in p21waf1, a marker of cell cycle arrest; (c) inhibition of mitogen-activated protein kinase, a negative regulator of PPARgamma, enhanced drug activity; and (d) phenylacetate interacted directly with the ligand-binding site of PPARgamma and activated its transcriptional function. The ability to bind and activate PPARgamma was common to biologically active analogues of phenylacetate and corresponded to their potency as antitumor agents (phenylacetate < phenylbutyrate < p-chloro-phenylacetate < p-iodo-phenylbutyrate), whereas an inactive derivative, phenylacetylglutamine, had no effect on PPARgamma. These findings point to PPARgamma as a novel target in cancer therapy and provide the first identification of ligands that have selective antitumor activity in patients.


Assuntos
Neoplasias/prevenção & controle , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Animais , Antimetabólitos Antineoplásicos/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Fenilacetatos/metabolismo , Fenilacetatos/farmacologia , Fosforilação , Ligação Proteica , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ensaio Radioligante , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional/efeitos dos fármacos , Células Tumorais Cultivadas , Regulação para Cima
17.
Clin Cancer Res ; 5(6): 1509-16, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10389940

RESUMO

Although Burkitt's lymphoma (BL) is a readily treated malignancy, recurrences, as well as disease arising in immunosuppressed patients, are notoriously resistant to conventional therapeutic approaches. The EBV is associated with a significant proportion of these lymphomas that evade immune surveillance through decreased expression of both viral and cellular antigens. Increasing the immunogenicity of BL cells may, therefore, represent a potentially beneficial therapeutic maneuver. Using in vitro models of EBV-transformed lymphoblastoid as well as BL cell lines, we demonstrate increased expression of genes coding for HLA class I and EBV latent proteins by the differentiation inducer phenylbutyrate (PB). The aromatic fatty acid also caused cytostasis associated with sustained declines in c-myc expression, a direct antitumor effect that was independent of the EBV status. We conclude, therefore, that differentiation therapy of BL with PB may lead to growth arrest with increased tumor immunogenicity in vivo. The findings may have clinical relevance because the in vitro activity has been observed with PB concentrations that are well tolerated and nonimmunosuppressive in humans, a desirable feature for the different patient populations afflicted with this disease.


Assuntos
Linfoma de Burkitt/patologia , Linfoma de Burkitt/virologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Herpesvirus Humano 4/genética , Fenilbutiratos/farmacologia , Linfoma de Burkitt/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , DNA Viral/biossíntese , Relação Dose-Resposta a Droga , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Herpesvirus Humano 4/crescimento & desenvolvimento , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Células Tumorais Cultivadas , Regulação para Cima , Proteínas da Matriz Viral/biossíntese , Ativação Viral/efeitos dos fármacos
18.
Clin Cancer Res ; 2(5): 865-72, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-9816242

RESUMO

Cytotoxic chemotherapies often give rise to multidrug resistance, which remains a major problem in cancer management. In pursuit of alternative treatments for chemoresistant tumor cells, we tested the response of multidrug-resistant (MDR) tumor cell lines to the aromatic fatty acids phenylacetate (PA) and phenylbutyrate (PB), two differentiation inducers currently in clinical trials. Both compounds induced cytostasis and maturation of multidrug-resistant breast, ovarian, and colon carcinoma cells with no significant effect on cell viability. In contrast to their poor response to doxorubicin, the MDR cells were generally more sensitive to growth arrest by PA and PB than their parental counterparts. The aromatic fatty acids, like the differentiation-inducing aliphatic fatty acid butyrate, up-regulated mdr-1 gene expression. However, while butyrate increased multidrug resistance, PA and PB potentiated the cytotoxic activity of doxorubicin against MDR cells. The latter was associated with time-dependent declines in glutathione levels and in the activity of superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase, and glutathione S-transferase, the antioxidant enzymes implicated in cell resistance to free radical-based therapies. Taken together, our in vitro data indicate that PA and PB, differentiation inducers of the aromatic fatty acid class, may provide an alternative approach to the treatment of MDR tumors.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Fenilacetatos/farmacologia , Fenilbutiratos/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Catalase/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Resistência a Múltiplos Medicamentos , Glutationa/metabolismo , Humanos , Células Tumorais Cultivadas , Verapamil/farmacologia
19.
Clin Cancer Res ; 2(3): 483-91, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9816194

RESUMO

Lovastatin, an inhibitor of the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase (the major regulatory enzyme of the mevalonate pathway of cholesterol synthesis), displays antitumor activity in experimental models. We therefore conducted a Phase I trial to characterize the tolerability of lovastatin administered at progressively higher doses to cancer patients. From January 1992 to July 1994, 88 patients with solid tumors (median age, 57 +/- 14 years) were treated p.o. with 7-day courses of lovastatin given monthly at doses ranging from 2 to 45 mg/kg/day. The inhibitory effects of lovastatin were monitored through serum concentrations of cholesterol and ubiquinone, two end products of the mevalonate pathway. Concentrations of lovastatin and its active metabolites were also determined, by bioassay, in the serum of selected patients. Cyclical treatment with lovastatin markedly inhibited the mevalonate pathway, evidenced by reductions in both cholesterol and ubiquinone concentrations, by up to 43 and 49% of pretreatment values, respectively. The effect was transient, however, and its magnitude appeared to be dose independent. Drug concentrations reached up to 3.9 micrometer and were in the range associated with antiproliferative activity in vitro. Myopathy was the dose-limiting toxicity. Other toxicities included nausea, diarrhea, and fatigue. Treatment with ubiquinone was associated with reversal of lovastatin-induced myopathy, and its prophylactic administration prevented the development of this toxicity in a cohort of 56 patients. One minor response was documented in a patient with recurrent high-grade glioma. Lovastatin given p.o. at a dose of 25 mg/kg daily for 7 consecutive days is well tolerated. The occurrence of myopathy, the dose-limiting toxicity, can be prevented by ubiquinone supplementation. To improve on the transient inhibitory activity of this dosing regimen on the mevalonate pathway, alternative schedules based on uninterrupted administration of lovastatin should also be studied.


Assuntos
Antineoplásicos/efeitos adversos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Lovastatina/efeitos adversos , Neoplasias/tratamento farmacológico , Adulto , Idoso , Feminino , Humanos , Lovastatina/farmacologia , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Ubiquinona/sangue
20.
Eur J Cell Biol ; 49(2): 221-4, 1989 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2673787

RESUMO

EK-3 cells, previously isolated by us from cultures of NIH 3T3, require both ras and myc oncogenes for efficient transformation, while their parent cells are readily transformed by ras alone. We transfected the EK-3 cells with the v-Ha-ras oncogene and obtained several sublines which integrated this gene and transcribed it successfully. The ras-NIH 3T3 formed foci of multilayered cells that were piling up in culture, while the ras-EK-3 cells remained contact inhibited. Furthermore, when the growth of the cells in soft agar was examined, a clear difference was observed. Cells of the ras-NIH 3T3 clonal lines showed high efficiency of growth (10%), while the ras-EK-3 cells exhibited low efficiency (0.2%). The latter being quite similar to that of the non-transfected NIH 3T3 and EK-3 cells (0.05%). The results presented now, showing that ras-EK-3 cells are more anchorage dependent than the ras-NIH 3T3 cells, clearly indicate that differences, previously shown to exist between EK-3 and NIH 3T3 cells, persist in their daughter cell lines derived following transfection with the Ha-ras oncogene.


Assuntos
Divisão Celular , Genes ras , Transfecção , Ágar , Animais , Sequência de Bases , Linhagem Celular , Linhagem Celular Transformada , DNA/genética , Immunoblotting , Camundongos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA