Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 198
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Urol ; 190(5): 1846-51, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23727411

RESUMO

PURPOSE: Nephrectomy with lymph node sampling is the recommended treatment for children with unilateral Wilms tumor under the Children's Oncology Group protocols. Using radiological assessment, we determined the feasibility of performing partial nephrectomy in a select group of patients with very low risk unilateral Wilms tumor. MATERIALS AND METHODS: We reviewed imaging studies of 60 patients with a mean age of less than 2 years with very low risk unilateral Wilms tumor (mean weight less than 550 gm) to assess the feasibility of partial nephrectomy. We evaluated percentage of salvageable parenchyma, tumor location and anatomical features preventing a nephron sparing approach. RESULTS: A linear relationship exists between tumor weight and computerized tomography estimated tumor volume. Mean tumor weight in the study population was 315 gm. Partial nephrectomy was deemed feasible in only 5 of 60 patients (8%). CONCLUSIONS: When considering a select population with very low risk unilateral Wilms tumor (lower volume tumor), only a small percentage of nonpretreated patients are candidates for nephron sparing surgery.


Assuntos
Neoplasias Renais/diagnóstico por imagem , Neoplasias Renais/cirurgia , Nefrectomia/métodos , Tumor de Wilms/diagnóstico por imagem , Tumor de Wilms/cirurgia , Estudos de Viabilidade , Feminino , Humanos , Lactente , Masculino , Tratamentos com Preservação do Órgão , Radiografia , Medição de Risco
2.
J Exp Med ; 145(1): 175-86, 1977 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-187717

RESUMO

We have isolated cloned variants in phagocytosis from a cloned continuous murine macrophage-like cell line, J 774.2. A selection procedure against Fc receptor-mediated phagocytosis was devised using IgG-coated SRBC containing a toxic drug, tubercidin, as the lethal agent. A series of variant clones deficient in Fc receptor-mediated phagocytosis were isolated. Such variants occurred at low frequency (approximately 6 X 10(-5)), were stable, and appeared to possess Fc receptors. The degree to which they were defective in phagocytosis of IgG-coated SRBC varied from clone to clone, yet all clones, were able to phagocytize latex particles. The phagocytic defect in some variants could be corrected by the addition of 8 Br-cAMP, in others, the drug was without effect. It is likely, therefore, that different variants are defective in several distinct steps critical to Fc receptor-mediated phagocytosis.


Assuntos
Macrófagos/fisiologia , Fagocitose , Sítios de Ligação , Linhagem Celular , Separação Celular , Células Clonais/fisiologia , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Fragmentos Fc das Imunoglobulinas , Proteínas Opsonizantes , Fagocitose/efeitos dos fármacos , Borracha , Tubercidina
3.
Skin Therapy Lett ; 15(5): 5-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20505896

RESUMO

An actinic keratosis (AK) is a pre-malignant cutaneous lesion that frequently manifests in sun-exposed areas of the skin as a small, rough, scaly erythematous papule. They are one of the most common presenting complaints for dermatologists. AKs should be treated due to their potential to progress into a squamous cell carcinoma (SCC). There are numerous treatments available for managing AKs including those broadly categorized as destructive, topical field, and procedural field therapies. The topical field therapies include 5-fluorouracil, imiquimod, and diclofenac gel. Recently, imiquimod 3.75% (Zyclara TM) has been approved for the treatment AKs on the face and scalp. It is a reasonable alternative to imiquimod 5%, as the approved indication includes a larger surface area for treatment, shorter duration course, and the potential for less severe local skin reactions. There is no widely accepted algorithm for the treatment of AKs, as comparative data is unavailable between all approaches. Therapy choices are guided by efficacy, adverse effects, cosmetic results, and patient compliance.


Assuntos
Carcinoma de Células Escamosas/prevenção & controle , Ceratose Actínica/terapia , Neoplasias Cutâneas/prevenção & controle , Administração Cutânea , Aminoquinolinas/administração & dosagem , Aminoquinolinas/uso terapêutico , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/etiologia , Diclofenaco/administração & dosagem , Diclofenaco/uso terapêutico , Fluoruracila/administração & dosagem , Fluoruracila/uso terapêutico , Humanos , Imiquimode , Ceratose Actínica/complicações , Ceratose Actínica/fisiopatologia , Fotoquimioterapia/métodos , Neoplasias Cutâneas/etiologia
4.
J Clin Invest ; 95(4): 1897-905, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7706497

RESUMO

Both EGF and insulin, or IGF, stimulate the growth of many cell types by activating receptors that contain tyrosine kinase activities. A monoclonal antibody (mAb 225) against the EGF receptor produced in this laboratory has been shown to competitively inhibit EGF binding and block activation of receptor tyrosine kinase. Here we report that a human colorectal carcinoma cell line, DiFi, which expresses high levels of EGF receptors on plasma membranes, can be induced to undergo G1 cell cycle arrest and programmed cell death (apoptosis) when cultured with mAb 225 at concentrations that saturate EGF receptors. Addition of IGF-1 or high concentrations of insulin can delay apoptosis induced by mAb 225, while the G1 arrest cannot be reversed by either IGF-1 or insulin. Insulin/IGF-1 cannot activate EGF receptor tyrosine kinase that has been inhibited by mAb 225. Moreover, an mAb against the IGF-1 receptor, which has little direct effect on DiFi cell growth, can block the capacity of insulin/IGF-1 to delay apoptosis induced by mAb 225, suggesting that the insulin/IGF-1-mediated delay of apoptosis is acting through the IGF-1 receptor. In contrast, insulin/IGF-1 cannot delay the apoptosis caused by the DNA damaging agent, cisplatin. The results indicate that EGF receptor activation is required both for cell cycle progression and for prevention of apoptosis in DiFi cells, and that a signal transduction pathway shared by receptors for insulin/IGF-1 and EGF may be involved in regulating apoptosis triggered by blockade of the EGF receptor.


Assuntos
Apoptose/fisiologia , Carcinoma/metabolismo , Neoplasias Colorretais/metabolismo , Receptores ErbB/metabolismo , Receptor IGF Tipo 1/metabolismo , Anticorpos Monoclonais/farmacologia , Ciclo Celular/fisiologia , Cisplatino/toxicidade , Interações Medicamentosas , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/imunologia , Humanos , Insulina/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Fosforilação , Proteína do Retinoblastoma/metabolismo , Células Tumorais Cultivadas
5.
J Clin Invest ; 86(6): 1806-14, 1990 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2174908

RESUMO

Expression of insulin-like growth factor I (IGF-I) mRNA by some tumor cell lines of neuroectodermal origin has been described. To further explore the significance of IGF-I mRNA expression in these tumors, a more extensive analysis was performed. Most (9 of 10) neuroectodermal tumor cell lines with a t(11;22) translocation (primitive neuroectodermal tumor [PNET], Ewing's sarcoma, esthesioneuroblastoma) expressed IGF-I mRNA, whereas 0 of 15 cell lines without the translocation (PNET, neuroblastoma) expressed IGF-I. Furthermore, inasmuch as all neuroblastoma (12 of 12) cell lines examined expressed IGF-II RNA, the pattern of IGF expression could distinguish between these closely related tumors. CHP-100, a PNET cell line with the t(11;22) translocation, was shown to secrete both IGF-I protein and an IGF binding protein, IGFBP-2. This cell line also expressed the type I IGF receptor mRNA, and blockade of this receptor by a monoclonal antibody (alpha IR3) inhibited serum-free growth. These data demonstrate that IGF-I expression is a property of neuroectodermal tumors with a t(11;22) translocation and that interruption of an IGF-I autocrine loop inhibits the growth of these tumor cells.


Assuntos
Cromossomos Humanos Par 11 , Cromossomos Humanos Par 22 , Fator de Crescimento Insulin-Like I/fisiologia , Neuroblastoma/fisiopatologia , Tumores Neuroectodérmicos Primitivos Periféricos/fisiopatologia , Receptores de Superfície Celular/fisiologia , Divisão Celular , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like I/genética , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , RNA Neoplásico/genética , Receptores de Superfície Celular/genética , Receptores de Somatomedina , Sarcoma/genética , Translocação Genética
6.
Mol Cell Biol ; 9(7): 2983-8, 1989 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2779553

RESUMO

The lck protein tyrosine kinase is normally expressed in a cell type-specific fashion, with mRNA being confined to cells of lymphoid lineage. Despite this highly specific pattern of expression in normal tissues, lck mRNA has also been detected in selected cell lines derived from human nonlymphoid neoplasms. In this study we explored the mechanisms underlying the expression of lck mRNA within human nonlymphoid neoplastic cell lines. We determined that lck mRNA expression was correlated with transcriptional activation and that there was no evidence for genomic rearrangement or amplification within the lck coding region to account for the expression of lck mRNA in the nonlymphoid neoplastic cell lines. The lck gene has previously been shown to contain two distinct promoter elements. In this study, we demonstrated that lck-producing cell lines derived from human nonlymphoid neoplasms expressed transcripts initiated exclusively from the 3'-most promoter element (3' promoter). In contrast, lymphoid cell lines derived from nonmalignant sources expressed lck transcripts exclusively initiated from the 5'-most promoter element (5' promoter). Most cell lines derived from human lymphoid neoplasms express lck transcripts initiated from both the 5' and 3' promoters in various ratios. Thus, lck expression in a variety of malignant cell lines results from a selective induction of transcription from the 3' promoter.


Assuntos
Proteínas Tirosina Quinases/genética , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Sequência de Bases , Northern Blotting , Southern Blotting , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Sondas RNA , RNA Mensageiro/genética , RNA Neoplásico/genética , Ribonucleases , Células Tumorais Cultivadas
7.
Oncogene ; 36(16): 2255-2264, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-27748766

RESUMO

Dysregulated activation of the CDK4/6 kinases is a hallmark of most mammary-derived carcinomas. ATP-competitive inhibitors against this complex have been recently advanced in the clinic and have shown significant activity, particularly against tumors driven by the estrogen receptor (ER). However, resistance to these compounds has begun to emerge often months to years after their initiation. We investigated potential mechanisms of resistance using cell line models that are highly sensitive to this class of drugs. After prolonged exposure to the selective and potent CDK4/6 inhibitor LY2835219, clones emerged and several were found to harbor amplification of the CDK6 kinase. Amplification of CDK6 resulted in a marked increase in CDK6 expression and reduced response of the CDK4/6 target, phospho-Rb (pRb), to CDK4/6 inhibitors. Knockdown of CDK6 restored drug sensitivity, while enforced overexpression of CDK6 was sufficient to mediate drug resistance. Not only did CDK6 overexpression mediate resistance to CDK4/6 inhibitors but it also led to reduced expression of the ER and progesterone receptor (PR), and diminished responsiveness to ER antagonism. The reduced ER/PR expression after CDK4/6 inhibitor resistance was additionally observed in tumor biopsy specimens from patients treated with these drugs. Alternative mechanisms of resistance to CDK4/6 inhibitors such as loss of pRb and cyclin E1 overexpression also exhibited decreased hormone responsiveness, suggesting that the clinical paradigm of sequential endocrine-based therapy may be ineffective in some settings of acquired CDK4/6 resistance.


Assuntos
Neoplasias da Mama/enzimologia , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos , Inibidores de Proteínas Quinases/farmacologia , Receptores de Estrogênio/metabolismo , Aminopiridinas/farmacologia , Benzimidazóis/farmacologia , Neoplasias da Mama/dietoterapia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Quinase 6 Dependente de Ciclina/genética , Feminino , Amplificação de Genes , Humanos , Células MCF-7 , Transdução de Sinais/efeitos dos fármacos
9.
Cancer Res ; 60(14): 3940-6, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10919672

RESUMO

The ansamycin antibiotics, herbimycin A (HA) and geldanamycin (GM), bind to a conserved pocket in heat shock protein 90 (Hsp90) and alter the function of this chaperone protein. Occupancy of this pocket results in the degradation of a subset of signaling molecules. These include proteins known to associate with Hsp90, e.g., the steroid receptors and Raf, as well as certain transmembrane tyrosine kinases, such as the ErbB receptor family. In a variety of tumor cell lines, treatment with HA potently inhibited cellular proliferation by inducing G1 arrest. This arrest was accompanied by hypophosphorylation of the retinoblastoma gene product (RB) and rapid down-regulation of cyclin D- and E-associated kinase activities. Inhibition of kinase activity was found to result from loss in expression of cyclins D1, D3, and E, as well as the associated cyclin-dependent kinases, cyclin-dependent kinase 4 and cyclin-dependent kinase 6. In addition, HA treatment also caused a late induction of p27(Kip1) protein. The loss of cyclin D preceded the other effects of HA, suggesting that it might be the primary cause of G1 arrest. To determine whether the effects of HA are mediated by selective inhibition of the cyclin D-RB pathway, HA was added to tumor cell lines lacking functional RB. HA treatment of Rb-negative tumor cell lines failed to elicit a G1 arrest. In addition, after release from synchronization with nocodazole, Rb-negative but not Rb-positive cell lines were able to progress through G1 into S phase in the presence of HA. Together, these findings suggest that induction of G1 arrest by HA results from down-regulation of cyclin D expression and its associated kinase activity. Furthermore, these findings imply that Hsp90 selectively regulates signaling pathways upstream of RB.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Proteínas de Ciclo Celular , Fase G1/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas Proto-Oncogênicas , Quinonas/farmacologia , Proteína do Retinoblastoma/metabolismo , Proteínas Supressoras de Tumor , Benzoquinonas , Western Blotting , Neoplasias da Mama/metabolismo , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Ciclina A/metabolismo , Ciclina D1/metabolismo , Ciclina D3 , Ciclina E/metabolismo , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Citometria de Fluxo , Humanos , Lactamas Macrocíclicas , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteína do Retinoblastoma/genética , Rifabutina/análogos & derivados , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Células Tumorais Cultivadas
10.
Cancer Res ; 61(19): 7184-8, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11585753

RESUMO

The epidermal growth factor receptor (EGFR) is commonly overexpressed in many human tumors and provides a new target for anticancer drug development. ZD1839 ("Iressa"), a quinazoline tyrosine kinase inhibitor selective for the EGFR, has shown good activity in preclinical studies and in the early phase of clinical trials. However, because it remains unclear which tumor types are the best targets for treatment with this agent, the molecular characteristics that correlate with tumor sensitivity to ZD1839 have been studied. In a panel of human breast cancer and other epithelial tumor cell lines, HER2-overexpressing tumors were particularly sensitive to ZD1839. Growth inhibition of these tumor cell lines was associated with the dephosphorylation of EGFR, HER2, and HER3, accompanied by the loss of association of HER3 with phosphatidylinositol 3-kinase, and down-regulation of Akt activity. These studies suggest that HER2-overexpressing tumors are particularly susceptible to the inhibition of HER family tyrosine kinase signaling and suggest novel strategies to treat these particularly aggressive tumors.


Assuntos
Antineoplásicos/farmacologia , Proteínas Serina-Treonina Quinases , Quinazolinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Regulação para Baixo/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/biossíntese , Gefitinibe , Humanos , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt , Quinazolinas/administração & dosagem , Receptor ErbB-2/metabolismo , Receptor ErbB-2/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trastuzumab , Células Tumorais Cultivadas
11.
Cancer Res ; 61(7): 2945-52, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11306472

RESUMO

17-(Allylamino)-17-demethoxygeldanamycin (17-AAG) is an ansamycin antibiotic that binds to a conserved pocket in Hsp90 and induces the degradation of proteins that require this chaperone for conformational maturation. 17-AAG causes a retinoblastoma (RB)-dependent G1 block in cancer cells and is now in clinical trial. In breast cancer cells, G1 block is accompanied by differentiation and followed by apoptosis. The differentiation is characterized by specific changes in morphology and induction of milk fat proteins and lipid droplets. In cells lacking RB, neither G1 arrest nor differentiation occurs; instead, they undergo apoptosis in mitosis. Introduction of RB into these cells restores the differentiation response to 17-AAG. Inhibitors of the ras, mitogen-activated protein kinase, and phosphatidylinositol 3-kinase pathways cause accumulation of milk fat proteins and induction of lipid droplets when associated with G1 arrest but do not cause morphological changes. Thus, regulation of Hsp90 function by 17-AAG in breast cancer cells induces RB-dependent morphological and functional mammary differentiation. G1 arrest is sufficient for some but not all aspects of the phenotype. Induction of differentiation may be responsible for some of the antitumor effects of this drug.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Quinonas/farmacologia , Rifabutina/farmacologia , Apoptose/efeitos dos fármacos , Benzoquinonas , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Fase G1/efeitos dos fármacos , Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Proteínas de Choque Térmico HSP90/fisiologia , Lactamas Macrocíclicas , Gotículas Lipídicas , Proteínas do Leite/metabolismo , Fenótipo , Fosforilação , Proteína do Retinoblastoma/metabolismo , Proteína do Retinoblastoma/fisiologia , Rifabutina/análogos & derivados , Células Tumorais Cultivadas
12.
Cancer Res ; 50(1): 48-53, 1990 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-2152773

RESUMO

The insulin-like growth factors IGF-I and IGF-II are potent mitogens for several breast tumor cell lines in culture. Additionally, both IGF-I and IGF-II mRNAs are easily detected in the majority of breast tumor specimens examined, while no breast cancer epithelial cell lines we have studied express authentic IGF-I mRNA, and few lines express IGF-II mRNA. Although receptors for insulin, IGF-I, and IGF-II have been described, there is significant cross-reactivity between the various receptors and ligands in the insulin/insulin-like growth factor family, and it is not clear which receptor or receptors are responsible for the biological effects of these growth factors in this system. Using an RNase protection assay, we examined breast tumor specimens and breast cancer epithelial cell lines for expression of mRNA encoding the type I and type II IGF receptors as well as the insulin receptor. Virtually all of the specimens examined expressed mRNA for all three receptors. We then examined estrogen-dependent MCF-7 cells for the mitogenic effects of IGF-I and II in the presence of antibodies to both the type I and type II receptors. alpha IR-3, a monoclonal antibody which blocks the type I receptor, abolished the mitogenic effects of both IGF-I and IGF-II. It did not, however, block the mitogenic effects of insulin. We conclude that type I and type II IGF receptors are ubiquitously expressed in breast cancer, and our experiments with MCF-7 cells suggest the mitogenic effects of both IGF-I and IGF-II are mediated via the type I IGF receptor.


Assuntos
Neoplasias da Mama/genética , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Receptores de Superfície Celular/genética , Somatomedinas/metabolismo , Northern Blotting , Neoplasias da Mama/metabolismo , Divisão Celular , Linhagem Celular , Sondas de DNA , DNA de Neoplasias/genética , Feminino , Humanos , Cinética , RNA Neoplásico/genética , Receptores de Superfície Celular/metabolismo , Receptores de Somatomedina , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/metabolismo
13.
Cancer Res ; 51(18): 4978-85, 1991 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-1893385

RESUMO

Breast tumors are a complex mix of epithelial, stromal, and vascular elements. We examined primary cultures of breast fibroblasts derived from benign and malignant lesions for expression of various growth factors. All fibroblast cultures, regardless of whether they were derived from benign or malignant lesions, expressed platelet-derived growth factor A chain, basic fibroblast growth factor, fibroblast growth factor 5, and transforming growth factor beta 1 mRNA. None expressed platelet-derived growth factor B chain or transforming growth factor alpha mRNA. However, examination of mRNA expression for the insulin-like growth factors revealed that 7 of 8 fibroblasts derived from benign lesions expressed insulin-like growth factor I (IGF-I) mRNA, while only 1 of 9 fibroblasts derived from malignancies expressed IGF-I mRNA. The opposite picture was seen for insulin-like growth factor II (IGF-II) mRNA expression, in which 1 of 9 benign-derived fibroblasts expressed IGF-II mRNA, while 5 of 9 malignant-derived fibroblasts expressed IGF-II. This correlated with previous in situ hybridization data, which showed IGF-I mRNA expression confined to the stroma of benign breast tissue. PDGF treatment of tumor fibroblasts resulted in a 3-fold increase in IGF-II mRNA. Thus there was an apparent dichotomy between IGF-I mRNA expression in the majority of fibroblasts derived from benign lesions and IGF-II mRNA expression in the majority of tumor-derived fibroblasts. Since the insulin-like growth factors are potent mitogens for breast tumor epithelial cells, this further supports the notion of a paracrine growth-promoting role for the insulin-like growth factors in breast lesions and suggests that IGF-II may be the more important growth promoter in malignant lesions.


Assuntos
Doenças Mamárias/genética , Neoplasias da Mama/genética , Fibroblastos/fisiologia , Substâncias de Crescimento/genética , RNA Mensageiro/genética , Doenças Mamárias/patologia , Neoplasias da Mama/patologia , Divisão Celular/fisiologia , Células Cultivadas , Feminino , Fator 5 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/genética , Imunofluorescência , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Derivado de Plaquetas/genética , RNA Mensageiro/biossíntese , Ribonucleases/metabolismo , Fator de Crescimento Transformador alfa/genética , Fator de Crescimento Transformador beta/genética , Células Tumorais Cultivadas
14.
Cancer Res ; 46(8): 4139-42, 1986 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3731081

RESUMO

Many human neuroblastoma tumors and cells in culture contain amplified N-myc DNA and this is associated with tumor stage. We have analyzed pairs of neuroblastoma cell cultures derived from two patients at the time of diagnosis and after tumor progression following the initiation of therapy. Cell cultures derived after progression have increased expression of N-myc RNA. In one pair this increase is associated with increased N-myc DNA amplification; in the other, amplification decreases and activation of N-myc is most likely the result of a regulatory change. Analysis of the pattern of DNA amplification in these cell cultures demonstrates additional changes that might be associated with tumor progression.


Assuntos
Neuroblastoma/genética , Proto-Oncogenes , Sequência de Bases , Linhagem Celular , DNA de Neoplasias/análise , Amplificação de Genes , Humanos , Neuroblastoma/patologia , RNA Mensageiro/análise
15.
Cancer Res ; 55(22): 5302-9, 1995 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-7585592

RESUMO

Farnesyl protein transferase (FPTase) catalyzes the first of a series of posttranslational modifications of Ras required for full biological activity. Peptidomimetic inhibitors of FPTase have been designed that selectively block farnesylation in vivo and in vitro. These inhibitors prevent Ras processing and membrane localization and are effective in reversing the transformed phenotype of Rat1-v-ras cells but not that of cells transformed by v-raf or v-mos. We have tested the effect of the FPTase inhibitor L-744,832 (FTI) on the anchorage-dependent and -independent growth of human tumor cell lines. The growth of over 70% of all tumor cell lines tested was inhibited by 2-20 microM of the FTI, whereas the anchorage-dependent growth of nontransformed epithelial cells was less sensitive to the effects of the compound. No correlation was observed between response to drug and the origin of the tumor cell or whether it contained mutationally activated ras. In fact, cell lines with wild-type ras and active protein tyrosine kinases in which the transformed phenotype may depend on upstream activation of the ras pathway were especially sensitive to the drug. To define the important targets of FTI action, the mechanism of cellular drug resistance was examined. It was not a function of altered drug accumulation or of FPTase insensitivity since, in all cell lines tested, FPTase activity was readily inhibited within 1 h of treatment with the inhibitor. Furthermore, the general pattern of inhibition of cellular protein farnesylation and the specific inhibition of lamin B processing were the same in sensitive and resistant cells. In addition, functional activation of Ras was inhibited to the same degree in sensitive and resistant cell lines. However, the FTI inhibited the epidermal growth factor-induced activation of mitogen-activated protein kinases in sensitive cells but not in two resistant cell lines. These data suggest that the drug does inhibit ras function and that resistance in some cells is associated with the presence of Ras-independent pathways for mitogen-activated protein kinase activation by tyrosine kinases. We conclude that FPTase inhibitors are potent antitumor agents with activity against many types of human cancer cell lines, including those with wild-type ras.


Assuntos
Alquil e Aril Transferases , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Transferases/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Divisão Celular/efeitos dos fármacos , Genes ras , Humanos , Células Tumorais Cultivadas
16.
Cancer Res ; 61(23): 8492-7, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11731433

RESUMO

Histone deacetylase (HDACs) regulate histone acetylation by catalyzing the removal of acetyl groups on the NH(2)-terminal lysine residues of the core nucleosomal histones. Modulation of the acetylation status of core histones is involved in the regulation of the transcriptional activity of certain genes. HDAC activity is generally associated with transcriptional repression. Aberrant recruitment of HDAC activity has been associated with the development of certain human cancers. We have developed a class of HDAC inhibitors, such as suberoylanilide hydroxamic acid (SAHA), that were initially identified based on their ability to induce differentiation of cultured murine erythroleukemia cells. Additional studies have demonstrated that SAHA inhibits the growth of tumors in rodents. In this study we have examined the effects of SAHA on MCF-7 human breast cancer cells. We found that SAHA causes the inhibition of proliferation, accumulation of cells in a dose-dependent manner in G(1) then G(2)-M phase of the cell cycle, and induction of milk fat globule protein, milk fat membrane globule protein, and lipid droplets. Growth inhibition was associated with morphological changes including the flattening and enlargement of the cytoplasm, and a decrease in the nuclear:cytoplasmic ratio. Withdrawal of SAHA led to reentry of cells into the cell cycle and reversal to a less differentiated phenotype. SAHA induced differentiation in the estrogen receptor-negative cell line SKBr-3 and the retinoblastoma-negative cell line MDA-468. We propose that SAHA has profound antiproliferative activity by causing these cells to undergo cell cycle arrest and differentiation that is dependent on the presence of SAHA. SAHA and other HDAC inhibitors are currently in Phase I clinical trials. These findings may impact the clinical use of these drugs.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias da Mama/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Inibidores de Histona Desacetilases , Humanos , Lipídeos/biossíntese , Proteínas do Leite/biossíntese , Receptor ErbB-2/biossíntese , Receptor ErbB-2/fisiologia , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/fisiologia , Proteína do Retinoblastoma/biossíntese , Proteína do Retinoblastoma/fisiologia , Vorinostat
17.
Cancer Res ; 59(24): 6145-52, 1999 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10626805

RESUMO

src kinase activity is elevated in some human tumors, including breast and colon cancers. The precise cellular function of the src family kinases is not clearly understood, but they appear to be involved in numerous signaling pathways. We studied the effects of PD173955, a novel src family-selective tyrosine kinase inhibitor, on cancer cell lines and found that it has significant antiproliferative activity due to a potent arrest of mitotic progression. The mitotic block occurs after chromosome condensation in prophase, before spindle assembly and without loss of cyclin A and B kinase activities. This effect is seen in cancer cell lines of all types with low or high activities of src kinases as well as in untransformed cell lines. In MDA-MB-468 breast cancer cells, this drug produces a rapid inhibition of cellular src and yes kinase activities as well as suppression of the mitotic hyperactivity of these kinases. This compound defines a novel class of antimitotic drugs that work through inhibition of src kinases and possibly other protein kinases that are required for progression through the initial phases of mitosis.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Piridonas/farmacologia , Pirimidinas/farmacologia , Quinases da Família src/antagonistas & inibidores , Genes src , Humanos , Mitose/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-yes , Células Tumorais Cultivadas
18.
Cancer Res ; 48(23): 6691-6, 1988 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-3180080

RESUMO

Insulin-like growth factor II is a growth factor important in fetal development. Several cancer tissues and cell lines have been reported to express IGF-II and rat IGF-II is mitogenic for breast cancer cell lines. Using Northern analysis and ribonuclease protection assays, IGF-II mRNA was detected in normal fibroblasts and in the established breast cancer cell line, T47D. In this cell line, steady state levels of IGF-II message were increased by treatment with estradiol. 10 nM IGF-II, purified from human serum, was mitogenic for breast cancer cell lines. In vitro, IGF-II may act as an autocrine growth factor for some cell lines. RNA derived from breast cancer, pathologically normal breast tissue, and benign breast disease also contained IGF-II mRNA. When paired samples of normal and cancer tissue were obtained from the breast of the same patient, the level of IGF-II mRNA expression in the normal tissue was at least that found in the cancer. This is consistent with previous observations that show IGF-II is expressed in mesenchyme. These findings suggest that in breast cancer IGF-II is produced by stromal tissue elements and potentially by the malignant epithelial cells. Therefore, IGF-II may function as an autocrine or a paracrine growth factor in different breast tumors.


Assuntos
Neoplasias da Mama/metabolismo , Fator de Crescimento Insulin-Like II/genética , RNA Mensageiro/análise , Somatomedinas/genética , Northern Blotting , Neoplasias da Mama/patologia , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fator de Crescimento Insulin-Like II/farmacologia , Células Tumorais Cultivadas
19.
Cancer Res ; 47(19): 5141-8, 1987 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-2441861

RESUMO

An MCF-7 human breast cancer cell line was selected which was 200-fold more resistant to Adriamycin than the wild type cell line. This Adriamycin-resistant (AdrR) cell line exhibited a multidrug-resistant phenotype and was cross-resistant to a wide range of antineoplastic agents including Vinca alkaloids, anthracyclines, and epipodophyllotoxins. Cytogenetic analysis of the AdrR cell line showed the presence of homogeneously staining regions on several chromosomes which were not present in the parental cell line. Using the technique of in-gel renaturation, DNA sequences which were amplified 50- to 100-fold in the AdrR cell line and which covered a total of over 140 kilobases were isolated. In addition, AdrR cells were found to contain amplified and overexpressed sequences which were homologous to hamster P-glycoprotein gene sequences. A hamster cDNA P-glycoprotein gene probe was used to screen a lambda gt10 cDNA library made from human AdrR cell line mRNA and human cDNA sequences homologous to the P-glycoprotein gene were isolated. Hybridization studies with the cloned human cDNA (pADR1) showed that the AdrR MCF-7 cell line contained a 60-fold amplification of this DNA sequence and that polyadenylated mRNA from the AdrR cell line contained a 4.8-kilobase transcript which was overexpressed 45-fold. There was a direct correlation between DNA and RNA copy number of this sequence and level of resistance among several MCF-7 Adriamycin-resistant cell lines. In situ hybridization studies demonstrated that the human P-glycoprotein gene sequence was found on chromosome 7q21.1 in normal human lymphocytes and that amplified DNA sequences isolated from the AdrR MCF-7 cells by the in-gel hybridization technique were linked to the human P-glycoprotein sequences in the homogeneously staining regions in the AdrR cells.


Assuntos
Neoplasias da Mama/genética , DNA de Neoplasias/análise , DNA/análise , Doxorrubicina/farmacologia , Amplificação de Genes , Sequência de Bases , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular , Resistência a Medicamentos , Feminino , Glicoproteínas/genética , Humanos , RNA/análise , Vincristina/farmacologia
20.
Cancer Res ; 60(8): 2090-4, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10786665

RESUMO

Geldanamycin (GM) is a natural antibiotic that binds Hsp90 and induces the degradation of receptor tyrosine kinases, steroid receptors, and Raf. It is a potent inhibitor of cancer cells that overexpress HER-kinases, but its effects on other important proteins may cause significant toxicity and limit its clinical use. We report the synthesis and identification of a GM dimer, GMD-4c, which had selective activity against HER-kinases. Selectivity was a function of linker length and required two intact GM moieties. GMD-4c is a potent inducer of G1 block and apoptosis of breast cancer cell lines that overexpress HER2, but does not appreciably inhibit the growth of 32D cells that lack HER-kinases. GMD-4c could be useful in the treatment of carcinomas dependent on HER-kinases.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/enzimologia , Quinonas/farmacologia , Receptor ErbB-2/metabolismo , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/uso terapêutico , Benzoquinonas , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Dimerização , Regulação para Baixo/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Concentração Inibidora 50 , Lactamas Macrocíclicas , Proteínas Proto-Oncogênicas c-raf/metabolismo , Quinonas/química , Quinonas/uso terapêutico , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/biossíntese , Receptor IGF Tipo 1/metabolismo , Receptores de Estrogênio/metabolismo , Especificidade por Substrato , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA