Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 18(44): 6021-8, 1999 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-10557091

RESUMO

All cells depend on sterols and isoprenoids derived from mevalonate (MVA) for growth, differentiation, and maintenance of homeostatic functions. In plants, environmental insults like heat and sunlight trigger the synthesis of isoprene, also derived from MVA, and this phenomenon has been associated with enhanced tolerance to heat. Here, we show that in human prostate adenocarcinoma PC-3M cells heat shock leads to activation of the MVA pathway. This is characterized by a dose- and time-dependent elevation in 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR) activity, enhanced sterol and isoprenoid synthesis, and increased protein prenylation. Furthermore, prenylation and subsequent membrane localization of Ras, a central player in cell signaling, was rapidly induced following heat stress. These effects were dose-dependent, augmented with repeated insults, and were prevented by culturing cells in the presence of lovastatin, a competitive inhibitor of HMGR. Enhanced Ras maturation by heat stress was also associated with a heightened activation of extracellular signal-regulated kinase (ERK), a key mediator of both mitogenic and stress signaling pathways, in response to subsequent growth factor stimulation. Thus, activation of the MVA pathway may constitute an important adaptive host response to stress, and have significant implications to carcinogenesis.


Assuntos
Adenocarcinoma/metabolismo , Colesterol/metabolismo , Genes ras , Hidroximetilglutaril-CoA Redutases/metabolismo , Neoplasias da Próstata/metabolismo , Estresse Fisiológico/metabolismo , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/etiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diterpenos/metabolismo , Farneseno Álcool/metabolismo , Resposta ao Choque Térmico/genética , Humanos , Hidroximetilglutaril-CoA Redutases/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hidroximetilglutaril-CoA-Redutases NADP-Dependentes , Lovastatina/farmacologia , Masculino , Ácido Mevalônico/metabolismo , Proteína Quinase 1 Ativada por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/etiologia , Prenilação de Proteína , Esteróis/biossíntese , Estresse Fisiológico/complicações , Proteínas ras/genética , Proteínas ras/metabolismo
2.
Adv Exp Med Biol ; 400A: 501-5, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9547596

RESUMO

Phenylacetate and analogs represent a new class of pleiotropic growth regulators that alter tumor cell biology by affecting gene expression at both the transcriptional and post transcriptional levels. Based on these findings, NaPA and NaPB entered clinical trials at the National Cancer Institute. Ongoing phase I studies with NaPA, involving adults with prostate and brain cancer, have confirmed that therapeutic levels can be achieved with no significant toxicities, and provide preliminary evidence for benefit to patients with advanced disease (Thibault et al., submitted).


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fenilacetatos/farmacologia , Fenilacetatos/uso terapêutico , Fenilbutiratos/farmacologia , Adulto , Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Ensaios Clínicos Fase I como Assunto , Humanos , Masculino , Neoplasias/tratamento farmacológico , Fenilbutiratos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Transcrição Gênica/efeitos dos fármacos , Células Tumorais Cultivadas
3.
Cell Growth Differ ; 7(12): 1609-15, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8959328

RESUMO

Phenylacetate (PA) and related aromatic fatty acids constitute a novel class of relatively nontoxic antineoplastic agents. These compounds induce tumor cytostasis and growth inhibition and differentiation of cancer cells, but little is known regarding the molecular events mediating these biological effects. Using human breast carcinoma MCF-7 cells as a model, we show here that PA-induced growth arrest is associated with enhanced expression of the cyclin-dependent kinase inhibitor p21Waf1/Cip1 and dephosphorylation of the retinoblastoma protein (pRB). The induction of p21WAF1/CIP1 mRNA by PA was independent of the cellular p53 status. To directly assess the contribution of p21Waf1/Cip1 to PA-mediated cytostasis, we compared the effects of PA in parental MCF-7 cells and cells expressing reduced levels of p21Waf1/Cip1 protein (clones AS.3 and AS.4), accomplished through constitutive expression of antisense p21Waf1/Cip1 transcripts. In contrast to parental cells, AS.3 and AS.4 cells did not show reduced pRB phosphorylation following PA treatment, indicating that p21Waf1/Cip1 induction by PA is required for dephosphorylation (inactivation) of pRB, a known mediator of cell cycle control. A prominent role for p21Waf1/Cip1 in mediating PA-induced growth arrest was further supported by the demonstration that embryonal fibroblasts derived from a p21WAF1/CIP1 knockout mouse (p21-/- mouse embryonal fibroblasts) did not growth arrest following PA treatment, whereas PA effectively induced p21WAF1/CIP1 mRNA and growth inhibition of the wild-type mouse embryonal fibroblasts. Taken together, our findings strongly support a role for p21Waf1/Cip1 in the PA-mediated inhibition of cell growth.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Quinases relacionadas a CDC2 e CDC28 , Ciclinas/metabolismo , Inibidores Enzimáticos/metabolismo , Fenilacetatos/farmacologia , Animais , Elementos Antissenso (Genética) , Neoplasias da Mama , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Divisão Celular/efeitos dos fármacos , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/genética , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/metabolismo , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia , Regulação para Cima/efeitos dos fármacos
4.
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
5.
Biochem Pharmacol ; 50(8): 1273-9, 1995 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-7488244

RESUMO

The aromatic fatty acid phenylacetate, a common metabolite of phenylalanine, shows promise as a relatively non-toxic drug for cancer treatment. This slowly metabolized fatty acid alters tumor cell lipid metabolism causing, among other effects, inhibition of protein prenylation critical to malignant growth. In pursuit of more potent analogues, we have examined the activity of related compounds against tumor cell lines established from patients with advanced prostatic carcinoma, glioblastomas, and malignant melanoma. Like phenylacetate, derivatives containing alpha-carbon or ring substitutions induced cytostasis and phenotypic reversion at non-toxic concentrations. Potency was correlated with the degree of calculated lipophilicity of the aromatic fatty acid, and the extent of inhibition of protein prenylation. Remarkably, a parallel cytostatic activity was reported in embryonic plant cells, which respond to phenylacetate and its analogues in the same concentration range and the same rank order of lipophilicity. These data suggest that phenylacetate and its analogues may act through common mechanisms to inhibit the growth of vastly divergent, undifferentiated cell types, and provide a basis for the development of new agents for the treatment of human malignancies.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Fenilacetatos/farmacologia , Prenilação de Proteína/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desenho de Fármacos , Humanos , Metabolismo dos Lipídeos , Fenótipo , Fenilacetatos/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas/efeitos dos fármacos
6.
Int J Cancer ; 63(1): 124-9, 1995 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-7558439

RESUMO

Alterations in the expression of ras oncogenes are characteristic of a wide variety of human neoplasms. Accumulating evidence has linked elevated ras expression with disease progression and with failure of tumors to respond to conventional therapies, including radiotherapy and certain chemotherapies. These observations led us to investigate the response of ras-transformed cells to the differentiation-inducer phenylacetate (PA). Using gene transfer models, we show that PA caused cytostasis in ras-transformed mesenchymal cells, associated with increased expression of 2',5'-oligoadenylate synthetase, an enzyme implicated in negative growth control. PA also induced phenotypic reversion characterized by loss of anchorage-independent growth, reduced invasiveness and increased expression of collagen alpha type I, a marker of cell differentiation. The anti-tumor activity of PA was observed in cases involving either Ha- or Ki-ras and was independent of the mode of oncogene activation. Interestingly, in contrast to their relative resistance to radiation and doxorubicin, ras-transformed cells were significantly more sensitive to PA than their parental cells. The profound changes in tumor cell and molecular biology were associated with reduced isoprenylation of the ras-encoded p21. Our results indicate that PA can suppress the growth of ras-transformed cells, resistant otherwise to free-radical based therapies, through interference with p21ras isoprenylation, critical to signal transduction and maintenance of the malignant phenotype.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Transformação Celular Neoplásica/patologia , Inibidores do Crescimento/farmacologia , Fenilacetatos/farmacologia , Prenilação de Proteína/efeitos dos fármacos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Células 3T3 , Animais , Diferenciação Celular/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Genes ras , Humanos , Camundongos , Fenótipo , Células Tumorais Cultivadas/patologia
7.
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
8.
Int J Cancer ; 62(3): 345-50, 1995 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-7628877

RESUMO

Cinnamic acid, a naturally occurring aromatic fatty acid of low toxicity, has a long history of human exposure. We now show that cinnamic acid induces cytostasis and a reversal of malignant properties of human tumor cells in vitro. The concentration causing a 50% reduction of cell proliferation (IC50) ranged from 1 to 4.5 mM in glioblastoma, melanoma, prostate and lung carcinoma cells. Using melanoma cells as a model, we found that cinnamic acid induces cell differentiation as evidenced by morphological changes and increased melanin production. Moreover, treated cells had reduced invasive capacity associated with modulation of expression of genes implicated in tumor metastasis (collagenase type IV, and tissue inhibitor metalloproteinase 2) and immunogenicity (HLA-A3, class-I major histocompatibility antigen). Further molecular analysis indicated that the anti-tumor activity of cinnamic acid may be due in part to the inhibition of protein isoprenylation known to block mitogenic signal transduction. The results presented here identify cinnamic acid as a new member of the aromatic fatty acid class of differentiation-inducers with potential use in cancer intervention.


Assuntos
Antineoplásicos/farmacologia , Cinamatos/farmacologia , Neoplasias/tratamento farmacológico , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Melanoma/patologia , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Pigmentação/efeitos dos fármacos , Prenilação de Proteína/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos
9.
Int J Cancer ; 60(4): 507-14, 1995 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-7829265

RESUMO

Phenylacetate, a natural metabolite of phenylalanine which was originally described as a plant growth hormone, has recently gained attention as a possible differentiation inducer for a variety of human tumor cell types. This interest prompted us to assess the ability of sodium phenylacetate (NaPA) to promote the differentiation of human neuroblastoma cells, both alone and in combination with retinoic acid (RA), a known inducer of neuroblastoma differentiation and maturation. Using the LA-N-5 cell line, we have determined that NaPA can stimulate the differentiation of neuroblastoma cells, as evidenced by dose-dependent inhibition of cell proliferation, neurite outgrowth, increased acetylcholinesterase activity and reduction of N-myc expression. Furthermore, NaPA and RA synergized in inducing differentiation, in that combination treatment resulted in cessation of cell growth along with morphologic and biochemical changes indicative of the loss of malignant properties. We have determined that NaPA can markedly enhance mRNA levels of the nuclear RA receptor-beta (RAR beta) in LA-N-5 cells prior to morphologic or other phenotypic changes induced by this compound. This effect appeared to be distinct from the ability of NaPA to alter tumor cell lipid metabolism via inhibition of protein isoprenylation. Thus among its varied effects on LA-N-5 cells, NaPA appears to interact with the RA pathway at the nuclear level by up-regulating RAR beta expression.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neuroblastoma/patologia , Fenilacetatos/farmacologia , Tretinoína/farmacologia , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Genes myc , Glutamina/metabolismo , Humanos , Técnicas Imunoenzimáticas , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Prenilação de Proteína , Proteínas Proto-Oncogênicas c-myc/biossíntese , Receptores do Ácido Retinoico/biossíntese , Células Tumorais Cultivadas/efeitos dos fármacos , Regulação para Cima
10.
J Invest Dermatol ; 103(3): 335-40, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8077698

RESUMO

The increasing incidence of melanoma and the poor responsiveness of disseminated disease to conventional treatments call for the development of new therapeutic approaches. Phenylacetate, a nontoxic differentiation inducer, can suppress the growth of other neuroectodermal tumors, i.e., gliomas, in laboratory models and in humans. This finding led us to explore the efficacy of phenylacetate and related aromatic fatty acids in melanoma. Phenylacetate and phenylbutyrate were found to a) induce selective cytostasis and maturation of cultured human melanoma cells, b) modulate the expression of genes implicated in tumor metastasis (type IV collagenase and tissue inhibitor of metalloproteinases-2) and immunogenicity (HLA class I); and c) enhance the efficacy of other agents of clinical interest, including retinoids, interferon-alpha, suramin, and 5-aza-2'-deoxycytidine. Reflecting on the phenotypic heterogeneity of melanoma, the degree of biologic alterations induced by phenylacetate/phenylbutyrate varied significantly among the tumor cell lines tested. Although losing invasive capacity and tumorigenicity in athymic mice, poorly differentiated cells exhibited only a marginal change in morphology, remained amelanotic, and resumed growth after treatment was discontinued. By contrast, treatment of melanoma cells that were in a more advanced stage of maturation resulted in profound alterations in cell growth, morphology, and pigmentation consistent with terminal differentiation. The in vitro antitumor activity was observed with nontoxic, pharmacologic concentrations of phenylacetate and phenylbutyrate, suggesting potential clinical use of these drugs in the treatment of melanomas.


Assuntos
Melanoma/patologia , Fenilacetatos/farmacologia , Fenilbutiratos/farmacologia , Antineoplásicos/farmacologia , Testes de Carcinogenicidade , Diferenciação Celular , Divisão Celular/efeitos dos fármacos , Sinergismo Farmacológico , Ácidos Graxos/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma/genética , Melanoma/fisiopatologia , Invasividade Neoplásica , Pigmentação/efeitos dos fármacos , 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(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
13.
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
14.
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
15.
Br J Haematol ; 79 Suppl 1: 81-3, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1931716

RESUMO

Any attempt to eradicate the heterogeneous cell population of a tumour mass would require the use of appropriate combination treatment protocols. The antitumour effects of interferon alpha (IFN alpha) in combination with AS2-1, the hydrolysis product of 3-phenylacetyl-amino-2,6-piperidinedione, were examined using several human tumour cell lines as a model. These included the malignant melanoma A375, adenocarcinoma of the prostate PC3 (hormone-insensitive bone metastasis), and the erythroleukaemia line K562. AS2-1 suppressed tumour growth through non-toxic mechanisms, with 1 mg/ml causing approximately 50% inhibition of the melanoma and prostate tumour cell proliferation. By contrast, primary normal human skin fibroblasts were significantly less sensitive to the antiproliferative effect of AS2-1. Suppression of tumour growth was seen also with AS2-1 treatment of the erythroleukaemia K562; in these cultures the drug also induced dose-dependent differentiation, as indicated by the increased haemoglobin production. Interestingly, addition of low doses of IFN alpha markedly enhanced the antitumour and differentiating effects observed with AS2-1. Treatment with 200-300 IU/ml of IFN (which caused about 20% inhibition of growth) together with 1 mg/ml of AS2-1 resulted in over 80% inhibition of the melanoma and prostate cancer cell proliferation, suggesting a synergistic activity of the two agents. This was substantiated by quantitative analysis of the differentiation induced in K562 erythroleukaemia. It appears, therefore, that IFN alpha and AS2-1 may act through synergistic mechanisms to effectively inhibit tumour growth and promote differentiation in a variety of human malignant cell lines.


Assuntos
Glutamina/análogos & derivados , Interferon-alfa/farmacologia , Fenilacetatos/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sinergismo Farmacológico , Glutamina/farmacologia , Humanos , Técnicas In Vitro , Células Tumorais Cultivadas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...