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.
Nature ; 467(7318): 972-6, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20927103

RESUMO

Reciprocity of inflammation, oxidative stress and neovascularization is emerging as an important mechanism underlying numerous processes from tissue healing and remodelling to cancer progression. Whereas the mechanism of hypoxia-driven angiogenesis is well understood, the link between inflammation-induced oxidation and de novo blood vessel growth remains obscure. Here we show that the end products of lipid oxidation, ω-(2-carboxyethyl)pyrrole (CEP) and other related pyrroles, are generated during inflammation and wound healing and accumulate at high levels in ageing tissues in mice and in highly vascularized tumours in both murine and human melanoma. The molecular patterns of carboxyalkylpyrroles are recognized by Toll-like receptor 2 (TLR2), but not TLR4 or scavenger receptors on endothelial cells, leading to an angiogenic response that is independent of vascular endothelial growth factor. CEP promoted angiogenesis in hindlimb ischaemia and wound healing models through MyD88-dependent TLR2 signalling. Neutralization of endogenous carboxyalkylpyrroles impaired wound healing and tissue revascularization and diminished tumour angiogenesis. Both TLR2 and MyD88 are required for CEP-induced stimulation of Rac1 and endothelial migration. Taken together, these findings establish a new function of TLR2 as a sensor of oxidation-associated molecular patterns, providing a key link connecting inflammation, oxidative stress, innate immunity and angiogenesis.


Assuntos
Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Estresse Oxidativo/fisiologia , Pirróis/metabolismo , Receptor 2 Toll-Like/metabolismo , Envelhecimento/metabolismo , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Linhagem Celular , Movimento Celular , Células Endoteliais/metabolismo , Membro Posterior/metabolismo , Humanos , Imunidade Inata/imunologia , Inflamação/metabolismo , Isquemia/metabolismo , Ligantes , Melanoma/irrigação sanguínea , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Oxirredução , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Propionatos , Pirróis/química , Pirróis/farmacologia , Receptores Depuradores/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 2 Toll-Like/agonistas , Receptor 4 Toll-Like/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo
2.
J Biol Chem ; 289(12): 8442-9, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24492617

RESUMO

Notch1 is an evolutionarily conserved signaling molecule required for stem cell maintenance that is inappropriately reactivated in several cancers. We have previously shown that melanomas reactivate Notch1 and require its function for growth and survival. However, no Notch1-activating mutations have been observed in melanoma, suggesting the involvement of other activating mechanisms. Notch1 activation requires two cleavage steps: first by a protease and then by γ-secretase, which releases the active intracellular domain (Notch1(NIC)). Interestingly, although ADAM10 and -17 are generally accepted as the proteases responsible of Notch1 cleavage, here we show that MT1-MMP, a membrane-tethered matrix metalloproteinase involved in the pathogenesis of a number of tumors, is a novel protease required for the cleavage of Notch1 in melanoma cells. We find that active Notch1 and MT1-MMP expression correlate significantly in over 70% of melanoma tumors and 80% of melanoma cell lines, whereas such correlation does not exist between Notch1(NIC) and ADAM10 or -17. Modulation of MT1-MMP expression in melanoma cells affects Notch1 cleavage, whereas MT1-MMP expression in ADAM10/17 double knock-out fibroblasts restores the processing of Notch1, indicating that MT1-MMP is sufficient to promote Notch1 activation independently of the canonical proteases. Importantly, we find that MT1-MMP interacts with Notch1 at the cell membrane, supporting a potential direct cleavage mechanism of MT1-MMP on Notch1, and that MT1-MMP-dependent activation of Notch1 sustains melanoma cell growth. Together, the data highlight a novel mechanism of activation of Notch1 in melanoma cells and identify Notch1 as a new MT1-MMP substrate that plays important biological roles in melanoma.


Assuntos
Metaloproteinase 14 da Matriz/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Receptor Notch1/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM17 , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 14 da Matriz/genética , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/genética , Proteínas de Membrana/metabolismo , Camundongos
4.
J Transl Med ; 10: 241, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23217102

RESUMO

BACKGROUND: Blood biomarkers are needed to monitor anti-angiogenic treatments for cancer. The association of blood levels of microRNAs (miRs) implicated in angiogenesis with circulating endothelial cells (CEC) and with angiogenic proteins was examined in patients administered drugs with anti-angiogenic activity. METHODS: Blood was collected from patients with uveal melanoma enrolled on an adjuvant therapy trial in which they were treated sequentially with dacarbazine and interferon-alfa-2b. Plasma levels of nine angioregulatory miRs, miR-16, 20a, 106a, 125b, 126, 146a, 155, 199a, and 221, were determined by quantitative real time polymerase chain reaction; CEC, by semi-automated immunomagnetic; and plasma angiogenic proteins, by enzyme linked immunosorbent assays. RESULTS: Levels of miR-199a were positively correlated and miR-106a negatively correlated with CEC pre-therapy. Decreases in miR-126 and miR-199a and increases in miR-16 and miR-106a were observed after interferon-alfa-2b, but not after dacarbazine. CEC also increased after treatment with interferon but not after treatment with dacarbazine. Levels of miRs did not correlate with levels of vascular endothelial growth factor, basic fibroblast growth factor, and interleukin-8. Angiogenic proteins also did not change significantly with treatment. CONCLUSIONS: Blood levels of specific angioregulatory miRs are associated with CEC, and changes in specific angioregulatory miRs parallel increases in CEC after treatment with interferon-alfa-2b. Blood levels of specific angioregulatory miRs are not associated with levels of angiogenic proteins. miRs warrant further evaluation as blood biomarkers of angiogenesis.


Assuntos
Proteínas Angiogênicas/sangue , Movimento Celular , Dacarbazina/uso terapêutico , Células Endoteliais/patologia , Interferon-alfa/uso terapêutico , MicroRNAs/sangue , Movimento Celular/efeitos dos fármacos , Dacarbazina/administração & dosagem , Dacarbazina/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Interferon-alfa/farmacologia , Melanoma/sangue , Melanoma/irrigação sanguínea , Melanoma/tratamento farmacológico , Melanoma/patologia , Neovascularização Patológica/sangue , Neovascularização Patológica/tratamento farmacológico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Neoplasias Uveais/sangue , Neoplasias Uveais/irrigação sanguínea , Neoplasias Uveais/tratamento farmacológico , Neoplasias Uveais/patologia
5.
J Clin Invest ; 117(10): 3107-17, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17823654

RESUMO

The effectiveness of IFN-alpha2b for human multiple myeloma has been variable. TRAIL has been proposed to mediate IFN-alpha2b apoptosis in myeloma. In this study we assessed the effects of IFN-alpha2b signaling on the apoptotic activity of TRAIL and human myeloma cell survival. While TRAIL was one of the most potently induced proapoptotic genes in myeloma cells following IFN-alpha2b treatment, less than 20% of myeloma cells underwent apoptosis. Thus, we hypothesized that an IFN-stimulated gene (ISG) with prosurvival activity might suppress TRAIL-mediated apoptosis. Consistent with this, IFN-alpha2b stabilized mitochondria and inhibited caspase-3 activation, which antagonized TRAIL-mediated apoptosis and cytotoxicity after 24 hours of cotreatment in cell lines and in fresh myeloma cells, an effect not evident after 72 hours. Induced expression of G1P3, an ISG with largely unknown function, was correlated with the antiapoptotic activity of IFN-alpha2b. Ectopically expressed G1P3 localized to mitochondria and antagonized TRAIL-mediated mitochondrial potential loss, cytochrome c release, and apoptosis, suggesting specificity of G1P3 for the intrinsic apoptosis pathway. Furthermore, RNAi-mediated downregulation of G1P3 restored IFN-alpha2b-induced apoptosis. Our data identify the direct role of a mitochondria-localized prosurvival ISG in antagonizing the effect of TRAIL. Curtailing G1P3-mediated antiapoptotic signals could improve therapies for myeloma or other malignancies.


Assuntos
Antineoplásicos/farmacologia , Interferon-alfa/farmacologia , Proteínas Mitocondriais/metabolismo , Mieloma Múltiplo/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Apoptose/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Regulação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Humanos , Interferon alfa-2 , Mitocôndrias/química , Mitocôndrias/metabolismo , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Mieloma Múltiplo/genética , Proteínas Recombinantes , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
6.
Cytokine Growth Factor Rev ; 18(5-6): 491-501, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17689283

RESUMO

Increased expression of genes, silenced by methylation of their promoters, could have relevance for increasing effects of not only interferons (IFNs) but also APO2L/TRAIL, cytotoxics and immunotherapeutics for melanoma and other malignancies. A resistant melanoma cell line, A375, lacked APO2L/TRAIL or apoptosis induction by either IFN-alpha2 or IFN-beta. However, apoptosis was induced by IFNs in A375 cells by 5-aza,2'-deoxycytidine (5-Aza-dC), evaluated based upon the postulate that promoter methylation might be silencing pro-apopoptotic IFN-stimulated genes (ISGs). RASSF1A, commonly methylated at high frequency in many tumors including melanoma, which we discovered to be also an IFN-regulated gene, was increased by 5-Aza-dC. RASSF1A was important in enhancing apoptotic effects of not only IFNs and APO2L/TRAIL but also cisplatin. Unraveling epigenetic regulatory mechanisms, as yet only partially identified, will result in new biological insights and improved strategies for therapeutic use of IFNs or ISGs such as APO2L/TRAIL.


Assuntos
Antivirais/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Interferons/farmacologia , Melanoma/genética , Animais , Apoptose , Inativação Gênica , Humanos , Melanoma/tratamento farmacológico , Melanoma/metabolismo
7.
Nat Rev Drug Discov ; 18(3): 219-234, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30679806

RESUMO

Over the past decade, preclinical and clinical research have confirmed the essential role of interferons for effective host immunological responses to malignant cells. Type I interferons (IFNα and IFNß) directly regulate transcription of >100 downstream genes, which results in a myriad of direct (on cancer cells) and indirect (through immune effector cells and vasculature) effects on the tumour. New insights into endogenous and exogenous activation of type I interferons in the tumour and its microenvironment have given impetus to drug discovery and patient evaluation of interferon-directed strategies. When combined with prior observations or with other effective modalities for cancer treatment, modulation of the interferon system could contribute to further reductions in cancer morbidity and mortality. This Review discusses new interferon-directed therapeutic opportunities, ranging from cyclic dinucleotides to genome methylation inhibitors, angiogenesis inhibitors, chemoradiation, complexes with neoantigen-targeted monoclonal antibodies, combinations with other emerging therapeutic interventions and associations of interferon-stimulated gene expression with patient prognosis - all of which are strategies that have or will soon enter translational clinical evaluation.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Fatores Imunológicos/uso terapêutico , Interferon-alfa/uso terapêutico , Interferon beta/uso terapêutico , Neoplasias/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Animais , Humanos , Neoplasias/imunologia
8.
Lung Cancer ; 60(2): 259-63, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18006112

RESUMO

PURPOSE: The purpose of this open-label phase II SWOG study was to evaluate the activity of gemcitabine (Gemzar; Eli Lilly, Indiana, USA) and cisplatin combination therapy, in patients with unresectable malignant mesothelioma of the pleura. PATIENTS AND METHODS: Fifty eligible chemotherapy naïve patients with histologically proven malignant mesothelioma of the pleura, and a SWOG performance status 0-2 were enrolled between February 1999 and August 2000. Treatment consisted of gemcitabine 1000mg/m(2) and cisplatin 30mg/m(2) on days 1, 8 and 15 of a 28-day cycle, until progression of disease or two cycles beyond complete response. RESULTS: Using SWOG response criteria, one patient had a confirmed complete response and five patients had a confirmed partial response, for a total response rate of 12% (95% CI 5-24%). All the responses were seen in patients with epithelioid or unspecified histology. Stable disease was seen in 25 patients (50%). The median overall survival was 10 months (95% CI 7-15 months), with a median progression-free survival of 6 months. Sixteen patients experienced Grade 4 toxicity. Twelve of these Grade 4 toxicities were hematologic. There were no treatment-related deaths. CONCLUSIONS: Cisplatin-gemcitabine combination chemotherapy has modest activity with an acceptable toxicity profile, as first line treatment for patients with malignant mesothelioma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Mesotelioma/tratamento farmacológico , Neoplasias Pleurais/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Desoxicitidina/administração & dosagem , Desoxicitidina/efeitos adversos , Desoxicitidina/análogos & derivados , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Mesotelioma/mortalidade , Mesotelioma/patologia , Pessoa de Meia-Idade , Neoplasias Pleurais/mortalidade , Neoplasias Pleurais/patologia , Gencitabina
9.
Cancer Res ; 66(5): 2785-93, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16510600

RESUMO

Resistance of human renal cell carcinoma (RCC) and melanoma to the apoptosis-inducing effects of IFNs was postulated to result from epigenetic silencing of genes by DNA methylation, a common feature of human cancers. To reverse silencing, 5-AZA-deoxycytidine (5-AZA-dC) or selective depletion of DNA methyltransferase 1 (DNMT1) by phosphorothioate oligonucleotide antisense (DNMT1 AS) were employed in cells resistant (<5% terminal deoxynucleotidyl transferase-mediated nick-end labeling positive) to apoptosis induction by IFN-alpha2 and IFN-beta (ACHN, SK-RC-45, and A375). 5-AZA-dC and DNMT1 AS similarly depleted available DNMT1 protein and, at doses that did not cause apoptosis alone, resulted in apoptotic response to IFNs. The proapoptotic tumor suppressor RASSF1A was reactivated by DNMT1 inhibitors in all three cell lines. This was associated with demethylation of its promoter region. IFNs augmented RASSF1A protein expression after reactivation by DNMT1 inhibition. In IFN-sensitive WM9 melanoma cells, expression of RASSF1A was constitutive but also augmented by IFNs. RASSF1A small interfering RNA reduced IFN-induced apoptosis in WM9 cells and in DNMT1-depleted ACHN cells. Conversely, lentiviral expression of RASSF1A but not transduction with empty virus enabled IFN-induced apoptosis. IFN induced tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and TRAIL-neutralizing antibody inhibited apoptotic response to IFN in RASSF1A-expressing ACHN cells. Accordingly, RASSF1A markedly sensitized to recombinant TRAIL. Normal kidney epithelial cells, although expressing RASSF1A, did not undergo apoptosis in response to IFN or TRAIL but had >400-fold higher TRAIL decoy receptor 1 expression than transduced ACHN cells (real-time reverse transcription-PCR). Results identified RASSF1A as regulated by IFNs and participating in IFN-induced apoptosis at least in part by sensitization to TRAIL.


Assuntos
Apoptose/efeitos dos fármacos , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Proteínas Supressoras de Tumor/biossíntese , Apoptose/genética , Proteínas Reguladoras de Apoptose/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/deficiência , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética , Inativação Gênica , Células HeLa , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Lentivirus/genética , Lentivirus/metabolismo , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Glicoproteínas de Membrana/farmacologia , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética
11.
Cancer Chemother Pharmacol ; 60(3): 341-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17103170

RESUMO

BACKGROUND: Pre-clinical activity of SSG against melanoma and renal cancer has been identified recently although the drug's mechanism of action and activity against tumors of additional histological-types remain undefined. METHODS: The effects of SSG and SSG combination with other agents on DU145 human prostate carcinoma xenograft tumors in mice and on DU145 cell subpopulations of differential SSG sensitivities were evaluated. RESULTS: DU145 tumor growth was inhibited by SSG (69%), IFNalpha2 (33%) or the combination (80%) that induced complete regression of WM9 human melanoma tumors. DU145 cells in culture were also partially growth inhibited by SSG at killing doses (200-800 mug/ml) for WM9 cells, indicating a correlation of SSG inhibition of cancer cell growth in vitro and in vivo. DU145 cells formed multiple micro tumors in mice treated with SSG or SSG/IFNalpha2 in contrast to the single large tumors in the control or IFNalpha2-treated mice, suggesting the existence of an SSG-resistant subpopulation in DU145 cells. Indeed, DU145 but not WM9 cells formed colonies (approximately 4% frequency) when cultured in the presence of SSG. Single cell clone (DU145-7) isolated from DU145 cells showed SSG-resistant growth in culture, unassociated with cross-resistance to IFNalpha2 and converted to SSG-responsive cells by BSO that inhibited intracellular glutathione levels. CONCLUSIONS: These results implicate a role for direct growth inhibition in SSG anti-tumor action, provide novel insights into the mechanism of tumor resistance to the drug and suggest a therapeutic potential for SSG and its combinations with IFNalpha2 or BSO for prostate cancer that warrants further investigation.


Assuntos
Gluconato de Antimônio e Sódio/uso terapêutico , Interferon-alfa/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Animais , Gluconato de Antimônio e Sódio/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Interferon-alfa/administração & dosagem , Masculino , Melanoma , Camundongos , Camundongos Nus , Neoplasias da Próstata/patologia , Transplante Heterólogo
12.
Oncology (Williston Park) ; 21(2): 247-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17396485

RESUMO

Preclinical advances offer an opportunity to further reduce morbidity and mortality from sarcomas over the next decade. Since no single institution or North American cooperative oncology group has the expertise or patient resources for histology-specific clinical and translational research on adult sarcomas, efforts have been made to develop funding from the National Cancer Institute (NCI). One such initiative was the Intergroup Coalition Against Sarcomas (ICAS), which, building upon the strengths of the multimodality cooperative oncology groups, provided an infrastructure for broad participation by investigators from all treatment disciplines in protocol development and patient entry. However, despite an excellent evaluation in formal peer review, the Division of Cancer Treatment of NCI has ended this initiative claiming insufficient available funds--to the detriment of adult sarcoma patients now and in the future.


Assuntos
Ensaios Clínicos como Assunto , Oncologia , Sarcoma , Adulto , Tumores do Estroma Gastrointestinal/epidemiologia , Tumores do Estroma Gastrointestinal/terapia , Coalizão em Cuidados de Saúde , Humanos , National Institutes of Health (U.S.) , Apoio à Pesquisa como Assunto , Sarcoma/epidemiologia , Sarcoma/terapia , Estados Unidos
13.
Int Semin Surg Oncol ; 4: 14, 2007 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-17603895

RESUMO

BACKGROUND: We report a case of pulmonary sarcoma which is a rare cause of the common symptom of dyspnea. CASE PRESENTATION: A fifty-one year old previously healthy male presented to the emergency room with complaints of dyspnea on exertion. A cardiac workup including an exercise stress test was negative but an echocardiography showed pulmonary stenosis. Cardiac MRI showed a large mass extending from the pulmonic valve to both the right and left pulmonary arteries suggestive of sarcoma. A complete resection and repair of the pulmonary artery was done and adjuvant chemotherapy with doxorubicin and ifosfamide was recommended. The patient is currently disease free after eighteen months. CONCLUSION: Pulmonary artery sarcomas are a difficult diagnosis. The diagnosis may remain elusive for some time until the proper imaging techniques are utilized to make a diagnosis. Earlier and accurate diagnosis may lead to earlier interventions and improve survival.

14.
JAMA Oncol ; 3(7): 944-952, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28196207

RESUMO

IMPORTANCE: After identification of activating mutations of the KIT gene in gastrointestinal stromal tumor (GIST)-the most common sarcomaof the gastrointestinal tract-a phase 2 study demonstrated efficacy of imatinib mesylate in patients with metastatic GIST harboring a KIT exon 11 mutation. Initial results of long-term follow-up have found a survival benefit in this subgroup of patients. OBECTIVE: To assess the long-term survival of patients with GIST who were treated in SWOG study S0033 and to present new molecular data regarding treatment outcomes. DESIGN, SETTING, AND PARTICIPANTS: In this follow-up of randomized clinical trial participants (from December 15, 2000, to September 1, 2001), patients were required to have advanced GIST that was not surgically curable. Postprotocol data collection occurred from August 29, 2011, to July 15, 2015. Using modern sequencing technologies, 20 cases originally classified as having wild-type tumors underwent reanalysis. This intergroup study was coordinated by SWOG, a cooperative group member within the National Clinical Trials Network, with participation by member/affiliate institutions. This follow-up was not planned as part of the initial study. INTERVENTIONS: Patients were randomized to 1 of 2 dose levels of imatinib mesylate, including 400 mg once daily (400 mg/d) vs 400 mg twice daily (800 mg/d), and were treated until disease progression or unacceptable toxic effects of the drug occurred. MAIN OUTCOMES AND MEASURES: The primary end point was overall survival. Updated survival data were correlated with clinical and molecular factors, and patterns of postprotocol therapies were enumerated and described in long-term survivors. RESULTS: Of 695 eligible patients (376 men [54.1%]; 319 women [45.9%]; mean [SD] age, 60.1 [14.0] years), 189 survived 8 years or longer, including 95 in the 400-mg/d dose arm and 94 in the 800-mg/d arm. The 10-year estimate of overall survival was 23% (95% CI, 20%-26%). Among 142 long-term survivors, imatinib was the sole therapy administered in 69 (48.6%), with additional systemic agents administered to 54 patients (38.0%). Resequencing studies of 20 cases originally classified as KIT/PDGFRA wild-type GIST revealed that 17 (85.0%) harbored a pathogenic mutation, most commonly a mutation of a subunit of the succinate dehydrogenase complex. CONCLUSIONS AND RELEVANCE: A subset of patients with metastatic GIST experiences durable, long-term overall survival with imatinib treatment. Although this study provides guidance for management of GIST harboring the most common KIT and PDGFRA mutations, optimal management of other genotypic subtypes remains unclear. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00009906.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Gastrointestinais/tratamento farmacológico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Mesilato de Imatinib/uso terapêutico , Proteínas Proto-Oncogênicas c-kit/genética , Idoso , Ensaios Clínicos Fase III como Assunto , Intervalo Livre de Doença , Feminino , Seguimentos , Neoplasias Gastrointestinais/genética , Tumores do Estroma Gastrointestinal/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Modelos de Riscos Proporcionais , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Succinato Desidrogenase/genética , Taxa de Sobrevida , Resultado do Tratamento
15.
J Interferon Cytokine Res ; 26(5): 353-61, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16689662

RESUMO

Interferon-alpha (IFN-alpha) has proved effective in the treatment of hemangiomas, hemangioblastomas, and Kaposi's sarcoma. To investigate the ability of IFNs to inhibit angiosarcoma, we used two transformed murine endothelial cell lines that form angiosarcomas in vivo. SVR and MS1-VEGF cell lines express oncogenic H-ras or vascular endothelial growth factor (VEGF), respectively. IFN-alpha1,8, which is active against murine and human cells, inhibited SVR and MS1-VEGF proliferation in vitro by 40% at 10(3) U/mL (p = 0.028). In vivo, IFN-alpha1,8 inhibited SVR tumor volume by 71% (p = 0.047) and MS1-VEGF volume by 79% (p = 0.003). Tumor-induced angiogenesis was decreased in SVR tumors by 52% (p = 0.005) and in MS1-VEGF tumors by 58% (p = 0.001). Sera from IFN-alpha1,8-treated mice bearing either SVR or MS1-VEGF tumors demonstrated a 5-fold increase in IP-10/CXCL10 (p = 0.001), an IFN-induced antiangiogenic protein. Both recombinant IP-10 and IFN-alpha1,8 inhibited human umbilical vein endothelial cell (HUVEC) vessel formation in the fibrin gel assay, a three-dimensional culture model of angiogenesis, by 56% at 25 ng/mL and 50% at 1.2 ng/mL, respectively (p < 0.001). An IP-10 blocking antibody restored vessel formation to 80% of untreated controls (p = 0.001). Given the magnitude of the in vivo response, these data suggested that the antitumor effects of IFN-alpha1,8 were likely mediated through angiogenesis inhibition rather than solely by direct inhibition of tumor cell proliferation.


Assuntos
Hemangiossarcoma/irrigação sanguínea , Hemangiossarcoma/tratamento farmacológico , Interferon-alfa/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL10 , Quimiocinas CXC/biossíntese , Quimiocinas CXC/sangue , Progressão da Doença , Hemangiossarcoma/metabolismo , Hemangiossarcoma/patologia , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Interferon Cytokine Res ; 26(11): 800-3, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17115898

RESUMO

Preclinical studies had determined that tamoxifen and interferon-alpha2b (IFN-alpha2b) synergistically inhibited growth of both estrogen-receptor positive and negative murine tumor xenografts and had combined antiangiogenic effects and that tamoxifen potentiated IFN-stimulated gene (ISG) expression. A phase I trial in 26 patients was conducted using the combination to define tolerance and potentiation of ISG expression. IFN- alpha2b at a dose of 3 x 10(6) units/m(2) daily was given subcutaneously (s.c.), and tamoxifen was initiated as a loading dose of 150 mg/m(2) and then 60 mg/m(2) twice daily on day 8. At this initial dose, reduction of dose of IFN- alpha2b was required in 4 of 11 patients, primarily because of fatigue. Another group of patients was treated with an identical tamoxifen dose but with IFN-alpha2b reduced to 2 x 10(6)/m(2) U; this was better tolerated. As the projected serum tamoxifen level to reduplicate preclinical effects was 300 mg/m(2), dose escalation in a third cohort was undertaken; it had to be discontinued secondary to grade III or IV toxicity in 2 of 2 patients. Increases in products of transcriptionally regulated ISGs, beta (2)-microglobulin, neopterin, and ISG15 were assessed. All ISGs increased after IFN-alpha2b, but only ISG15 had a further significant rise after initiation of tamoxifen. Because at doses not limited by unacceptable toxicities, no marked potentiation of ISGs by tamoxifen could be identified, clinical evaluation of the combination was terminated.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Antineoplásicos Hormonais/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Interferon-alfa/administração & dosagem , Proteínas de Neoplasias/biossíntese , Neoplasias/tratamento farmacológico , Tamoxifeno/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos Hormonais/efeitos adversos , Antineoplásicos Hormonais/farmacocinética , Relação Dose-Resposta a Droga , Estudos de Avaliação como Assunto , Feminino , Seguimentos , Humanos , Interferon alfa-2 , Interferon-alfa/efeitos adversos , Interferon-alfa/farmacocinética , Masculino , Camundongos , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/mortalidade , Proteínas Recombinantes , Tamoxifeno/efeitos adversos , Tamoxifeno/farmacocinética
17.
Cancer Res ; 62(12): 3453-8, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12067988

RESUMO

The immunobiology of tumor-infiltrating dendritic cells (DCs) can be strongly influenced by the cytokine environment present in the malignant tissue. We have previously identified discrete melanoma lines, inducing E-cadherin expression on monocyte-derived DCs in vitro. We demonstrate here that this effect, independent of cell contact, is not inducible in the presence of tumor lysates and requires the constitutive expression of IFN stimulated gene 15 (ISG15) by malignant cells. High-density oligonucleotide arrays were used to investigate the expression pattern of 7000 genes in RNA from two melanoma cell clones competent for E-cadherin induction and two clones devoid of DC-modulating capacity. A total of 13 genes encoding soluble proteins were expressed at higher magnitude in melanomas able to induce E-cadherin expression on DCs. Combining those data with quantitative protein assays, we could narrow our investigation down to three factors: the chemokine CCL5 and the cytokines ISG15 and type I IFNs. Strikingly, >7 ng/ml of ISG15 could be detected in the corresponding melanoma-conditioned medium and induction of E-cadherin on DCs failed in the presence of antibodies neutralizing ISG15 protein. Most importantly, strong cytoplasmic expression of ISG15 was detected by immunohistochemistry in the original tumor specimen from which the melanoma cell lines under investigation were derived. These data describe a novel property of ISG15 targeting induction of E-cadherin on DCs and possibly influencing their migratory behavior.


Assuntos
Caderinas/biossíntese , Citocinas/fisiologia , Células Dendríticas/metabolismo , Melanoma/metabolismo , Ubiquitinas/análogos & derivados , Anticorpos/imunologia , Anticorpos/farmacologia , Antígenos CD/biossíntese , Antígenos CD/genética , Antígeno B7-2 , Caderinas/genética , Caderinas/imunologia , Comunicação Celular/genética , Comunicação Celular/imunologia , Meios de Cultivo Condicionados , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/imunologia , Perfilação da Expressão Gênica , Humanos , Antígenos CD15/biossíntese , Antígenos CD15/genética , Melanoma/genética , Melanoma/imunologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Células Tumorais Cultivadas , Regulação para Cima
18.
Mol Cancer Ther ; 4(6): 948-55, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15956252

RESUMO

PURPOSE: This study intended to determine the maximum tolerated dose, safety, pharmacokinetic variables, clinical response, and pharmacodynamic markers of daily s.c. administration of Angiozyme. PATIENTS AND METHODS: Patients with refractory solid tumors were enrolled in a dose escalation and expanded cohort design. Dose escalation involved cohorts of patients at doses of 10, 30, 100, or 300 mg/m(2)/d for 29 days. A second component enrolled 15 additional patients at a daily dose of 100 mg/m(2). Patients were eligible to continue on therapy until disease progression. RESULTS: Thirty-one patients were enrolled and 28 were evaluable (range, 29-505 days; median, 89.5 days). A maximum tolerated dose was not defined by toxicity but rather by the maximal deliverable dose of 300 mg/m(2)/d. Grade 1 to 2 injection site reactions were the most common toxicities. One patient in the 300 mg/m(2) group experienced a reversible grade 3 injection site reaction. Angiozyme showed dose-dependent plasma concentrations with good bioavailability. Surrogate markers showed Angiozyme localization in tumor biopsies and a significant increase in serum von Willebrand factor antigen, a marker for endothelial cell dysfunction. Although Angiozyme-reactive antibody production was noted for some patients, no antibody-related adverse events were noted. Seven of 28 (25%) evaluable patients had stable disease for >or =6 months, with the longest treatment duration of > or =16 months. Two patients (nasopharyngeal carcinoma and melanoma) showed minor responses. CONCLUSION: Angiozyme was well tolerated with satisfactory pharmacokinetic variables for daily s.c. dosing. Results have provided the basis for subsequent clinical trials of this first-of-class biologically targeted therapeutic.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , RNA Catalítico/uso terapêutico , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/farmacocinética , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias/irrigação sanguínea , Neoplasias/genética , RNA Catalítico/efeitos adversos , RNA Catalítico/genética , RNA Catalítico/farmacocinética , Resultado do Tratamento , Fator de von Willebrand/metabolismo
19.
J Interferon Cytokine Res ; 25(9): 511-27, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16181052

RESUMO

Interferons (IFNs) remain the most broadly active cytokines for cancer treatment, yet ones for which the full potential is not reached. IFNs have impacted positively on both quality and quantity of life for hundreds of thousands of cancer patients with chronic leukemia, lymphoma, bladder carcinoma, melanoma, and renal carcinoma. The role of the IFN system in malignant pathogenesis continues to enhance understanding of how the IFN system may be modulated for therapeutic advantage. Reaching the full potential of IFNs as therapeutics for cancer will also result from additional understanding of the genes underlying apoptosis induction, angiogenesis inhibition, and influence on immunologic function. Food and Drug Administration (FDA) approval of IFNs occurred less than 20 years ago; after 40 years, third-generation products of early cytotoxics, such as 5- fluorouracil (5FU), are beginning to reach clinical approval. Thus, substantial potential exists for additional application of IFNs and IFN inducers as anticancer therapeutics, particularly when one considers that their pleiotropic cellular and molecular effects have yet to be fully defined.


Assuntos
Antineoplásicos/uso terapêutico , Interferon-alfa/uso terapêutico , Interferon beta/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos , Expressão Gênica/efeitos dos fármacos , Humanos , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Camundongos , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neovascularização Patológica/tratamento farmacológico
20.
Clin Cancer Res ; 9(6): 1941-56, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12796356

RESUMO

Sarcomas--like leukemias, which are also mesodermal malignancies--carry biological significance disproportionate to their clinical frequency. Identification of mutations and translocations associated with these tumors has illuminated aberrant signaling pathways that cause these diseases, determine their behavior, and are therapeutic targets. Activated receptor-associated tyrosine kinase c-kit, mutated in most gastrointestinal stromal tumors, has proven a clinically effective target for enzyme inhibition. A translocation involving a single gene family, consisting of EWS and related genes, has been identified in five different sarcomas, and its chimeric protein products could prove similarly amenable to inhibitors. Resolution of the histopathological complexity is being aided by data from molecular and chromosomal characterization. Improvements in imaging, definition of prognostic factors, and surgical and radiotherapeutic treatment have resulted in improved local control. Continued progress will depend on further adapting the rapidly evolving technologies of genomics and proteomics. It will also depend upon accurate histopathological diagnosis based on validated reagents and consistent methodologies applied to adequate tissue samples derived from patients with complete clinical data. Finally, multicenter, coordinated trials, such as those that occurred with assessment of imatinib mesylate in metastatic gastrointestinal stromal tumors, will assure the most rapid reductions in morbidity and mortality.


Assuntos
Sarcoma/terapia , Adulto , Quimioterapia Adjuvante , Aberrações Cromossômicas , Humanos , Cariotipagem , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Retroperitoneais/terapia , Sarcoma/genética , Sarcoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA