Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 25(8): 2403-2413, 2019 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-30425090

RESUMO

PURPOSE: This first-in-human, open-label phase I study evaluated AMG 337, an oral, highly selective small-molecule inhibitor of MET in advanced solid tumors.Patients and Methods: Patients enrolled into dose-escalation cohorts received AMG 337 up to 400 mg once daily or up to 250 mg twice daily, following a modified 3+3+3 design. Dose expansion was conducted in MET-amplified patients at the maximum tolerated dose (MTD). Primary endpoints included assessment of adverse events (AEs), establishment of the MTD, and pharmacokinetics; clinical response was a secondary endpoint. RESULTS: The safety analysis set included 111 patients who received ≥1 dose of AMG 337. Thirteen patients had ≥1 AE qualifying as dose-limiting toxicity. The MTD was determined to be 300 mg once daily; the MTD for twice-daily dosing was not reached. Most frequent treatment-related AEs were headache (63%) and nausea (31%). Grade ≥3 treatment-related AEs occurred in 23 patients (21%), most commonly headache (n = 6) and fatigue (n = 5). Maximum plasma concentration occurred at 3.0 hours following 300-mg once-daily dosing, indicating AMG 337 absorption soon after treatment. Objective response rate was 9.9% (11/111; 95% CI, 5.1%-17.0%) in all patients and 29.6% (8/27; 95% CI, 13.8%-50.2%) in MET-amplified patients; median (range) duration of response was 202 (51-1,430+) days in all patients and 197 (64-1,430+) days in MET-amplified patients. CONCLUSIONS: Oral AMG 337 was tolerated with manageable toxicities, with an MTD and recommended phase II dose of 300 mg once daily. The promising response rate observed in patients with heavily pretreated MET-amplified tumors warrants further investigation.See related commentary by Ma, p. 2375.


Assuntos
Neoplasias , Inibidores de Proteínas Quinases , Humanos , Dose Máxima Tolerável , Piridonas , Resultado do Tratamento , Triazóis
2.
Clin Cancer Res ; 23(15): 3980-3993, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28428191

RESUMO

Treatment of myeloma has benefited from the introduction of more effective and better tolerated agents, improvements in supportive care, better understanding of disease biology, revision of diagnostic criteria, and new sensitive and specific tools for disease prognostication and management. Assessment of minimal residual disease (MRD) in response to therapy is one of these tools, as longer progression-free survival (PFS) is seen consistently among patients who have achieved MRD negativity. Current therapies lead to unprecedented frequency and depth of response, and next-generation flow and sequencing methods to measure MRD in bone marrow are in use and being developed with sensitivities in the range of 10-5 to 10-6 cells. These technologies may be combined with functional imaging to detect MRD outside of bone marrow. Moreover, immune profiling methods are being developed to better understand the immune environment in myeloma and response to immunomodulatory agents while methods for molecular profiling of myeloma cells and circulating DNA in blood are also emerging. With the continued development and standardization of these methodologies, MRD has high potential for use in gaining new drug approvals in myeloma. The FDA has outlined two pathways by which MRD could be qualified as a surrogate endpoint for clinical studies directed at obtaining accelerated approval for new myeloma drugs. Most importantly, better understanding of MRD should also contribute to better treatment monitoring. Potentially, MRD status could be used as a prognostic factor for making treatment decisions and for informing timing of therapeutic interventions. Clin Cancer Res; 23(15); 3980-93. ©2017 AACR.


Assuntos
DNA Tumoral Circulante/sangue , Mieloma Múltiplo/sangue , Mieloma Múltiplo/tratamento farmacológico , Neoplasia Residual/sangue , Biomarcadores Tumorais/genética , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Intervalo Livre de Doença , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Mieloma Múltiplo/complicações , Mieloma Múltiplo/genética , Neoplasia Residual/induzido quimicamente , Neoplasia Residual/genética , Seleção de Pacientes , Prognóstico
3.
Cancer ; 123(6): 1061-1070, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27926778

RESUMO

BACKGROUND: MET gene amplification and Met protein overexpression may be associated with a poor prognosis. The MET/Met status is typically determined with fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC), respectively. Targeted proteomics uses mass spectrometry-based selected reaction monitoring (SRM) to accurately quantitate Met expression. FISH, IHC, and SRM analyses were compared to characterize the prognostic value of MET/Met in gastroesophageal adenocarcinoma (GEC). METHODS: Samples from 447 GEC patients were analyzed for MET gene amplification (FISH) and Met protein expression (IHC and SRM). Cox proportional hazards models and Kaplan-Meier estimates were applied to explore relations between Met, overall survival (OS), and clinical/pathological characteristics. Spearman's rank coefficient was used to assess the correlation between parameters. RESULTS: Patients with MET-amplified tumors had worse OS when: the MET/centromere enumeration probe for chromosome 7 FISH ratio was ≥ 2 (hazard ratio [HR], 3.13; 95% confidence interval [CI], 1.84-5.33), the MET gene copy number was ≥5 (HR, 2.51; 95% CI, 1.45-4.34), or ≥ 10% of the cells had ≥15 copies (HR, 4.28; 95% CI, 2.18-8.39). Similar observations were made with Met protein overexpression by IHC (≥1 + intensity in ≥ 25% of the tumor cell membrane: HR, 1.39; 95% CI, 1.04-1.86) or SRM (≥400 amol/µg: HR, 1.76; 95% CI, 1.06-2.90). A significant correlation was observed between MET FISH/Met IHC, MET FISH/Met SRM, and Met IHC/Met SRM; only MET FISH and Met SRM were independent negative prognostic biomarkers in multivariate analyses. CONCLUSIONS: MET amplification and overexpression, assessed by multiple methods, were associated with a worse prognosis in univariate analyses. However, only MET amplification by FISH and Met expression by SRM were independent prognostic biomarkers. Compared with IHC, SRM may provide an added benefit for informed decisions about Met-targeted therapy. Cancer 2017;123:1061-70. © 2016 American Cancer Society.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/mortalidade , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidade , Amplificação de Genes , Expressão Gênica , Proteínas Proto-Oncogênicas c-met/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidade , Biomarcadores Tumorais , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Espectrometria de Massas , Prognóstico , Modelos de Riscos Proporcionais
4.
PLoS One ; 11(2): e0149405, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26919633

RESUMO

BACKGROUND: Targeted Next Generation Sequencing (NGS) offers a way to implement testing of multiple genetic aberrations in diagnostic pathology practice, which is necessary for personalized cancer treatment. However, no standards regarding input material have been defined. This study therefore aimed to determine the effect of the type of input material (e.g. formalin fixed paraffin embedded (FFPE) versus fresh frozen (FF) tissue) on NGS derived results. Moreover, this study aimed to explore a standardized analysis pipeline to support consistent clinical decision-making. METHOD: We used the Ion Torrent PGM sequencing platform in combination with the Ion AmpliSeq Cancer Hotspot Panel v2 to sequence frequently mutated regions in 50 cancer related genes, and validated the NGS detected variants in 250 FFPE samples using standard diagnostic assays. Next, 386 tumour samples were sequenced to explore the effect of input material on variant detection variables. For variant calling, Ion Torrent analysis software was supplemented with additional variant annotation and filtering. RESULTS: Both FFPE and FF tissue could be sequenced reliably with a sensitivity of 99.1%. Validation showed a 98.5% concordance between NGS and conventional sequencing techniques, where NGS provided both the advantage of low input DNA concentration and the detection of low-frequency variants. The reliability of mutation analysis could be further improved with manual inspection of sequence data. CONCLUSION: Targeted NGS can be reliably implemented in cancer diagnostics using both FFPE and FF tissue when using appropriate analysis settings, even with low input DNA.


Assuntos
Análise Mutacional de DNA/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação , Neoplasias/genética , Formaldeído , Humanos , Neoplasias/patologia , Inclusão em Parafina , Reprodutibilidade dos Testes , Fixação de Tecidos
5.
J Clin Invest ; 121(4): 1298-312, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21436587

RESUMO

HSC homing, quiescence, and self-renewal depend on the bone marrow HSC niche. A large proportion of solid tumor metastases are bone metastases, known to usurp HSC homing pathways to establish footholds in the bone marrow. However, it is not clear whether tumors target the HSC niche during metastasis. Here we have shown in a mouse model of metastasis that human prostate cancer (PCa) cells directly compete with HSCs for occupancy of the mouse HSC niche. Importantly, increasing the niche size promoted metastasis, whereas decreasing the niche size compromised dissemination. Furthermore, disseminated PCa cells could be mobilized out of the niche and back into the circulation using HSC mobilization protocols. Finally, once in the niche, tumor cells reduced HSC numbers by driving their terminal differentiation. These data provide what we believe to be the first evidence that the HSC niche serves as a direct target for PCa during dissemination and plays a central role in bone metastases. Our work may lead to better understanding of the molecular events involved in bone metastases and new therapeutic avenues for an incurable disease.


Assuntos
Neoplasias da Medula Óssea/secundário , Células-Tronco Hematopoéticas/patologia , Neoplasias da Próstata , Animais , Neoplasias da Medula Óssea/patologia , Transplante de Medula Óssea , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Biológicos , Transplante de Neoplasias , Osteoblastos/patologia , Neoplasias da Próstata/patologia , Doadores de Tecidos , Transplante Heterólogo
6.
Neoplasia ; 12(2): 116-27, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20126470

RESUMO

Our recent studies have shown that annexin II, expressed on the cell surface of osteoblasts, plays an important role in the adhesion of hematopoietic stem cells (HSCs) to the endosteal niche. Similarly, prostate cancer (PCa) cells express the annexin II receptor and seem to use the stem cell niche for homing to the bone marrow. The role of the niche is thought to be the induction and sustenance of HSC dormancy. If metastatic PCa cells occupy a similar or the same ecological niche as HSCs, then it is likely that the initial role of the HSC niche will be to induce dormancy in metastatic cells. In this study, we demonstrate that the binding of PCa to annexin II induces the expression of the growth arrest-specific 6 (GAS6) receptors AXL, Sky, and Mer, which, in the hematopoietic system, induce dormancy. In addition, GAS6 produced by osteoblasts prevents PCa proliferation and protects PCa from chemotherapy-induced apoptosis. Our results suggest that the activation of GAS6 receptors on PCa in the bone marrow environment may play a critical role as a molecular switch, establishing metastatic tumor cell dormancy.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Invasividade Neoplásica , Neoplasias da Próstata/metabolismo , Nicho de Células-Tronco/metabolismo , Animais , Anexina A2/metabolismo , Apoptose/fisiologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Osteoblastos/metabolismo , Neoplasias da Próstata/patologia , Receptores Proteína Tirosina Quinases , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nicho de Células-Tronco/citologia , Análise Serial de Tecidos
7.
J Cell Biochem ; 104(5): 1587-97, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18646053

RESUMO

Nearly 85% of the men who will die of prostate cancer (PCa) have skeletal metastases present. The ability of PCa cells to interact with the microenvironment determines the success of the tumor cell to form metastatic lesions. The ability to bind to human bone marrow endothelial (HBME) cells and undergo transendothelial cell migration are key steps in allowing the PCa cell to extravasate from the bone microvasculature and invade the bone stroma. We have previously demonstrated that monoctyte chemoattractant protein 1 (MCP-1; CCL2) is expressed by HBME cells and promotes PCa proliferation and migration. In the current study, we demonstrate that the CCL2 stimulation of PCa cells activates the small GTPase, Rac through the actin-associated protein PCNT1. Activation of Rac GTPase is accompanied by morphologic changes and the ability of the cells to undergo diapedesis through HBME cells. These data suggest a role for HBME-secreted CCL2 in promoting PCa cell extravasation into the bone microenvironment.


Assuntos
Movimento Celular/efeitos dos fármacos , Quimiocina CCL2/farmacologia , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteínas rac de Ligação ao GTP/metabolismo , Actinas/metabolismo , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/genética , Transporte Proteico/efeitos dos fármacos
8.
J Cell Biochem ; 105(2): 370-80, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18636554

RESUMO

One of the most life-threatening complications of prostate cancer is skeletal metastasis. In order to develop treatment for metastasis, it is important to understand its molecular mechanisms. Our work in this field has drawn parallels between hematopoietic stem cell and prostate cancer homing to the marrow. Our recent work demonstrated that annexin II expressed by osteoblasts and endothelial cells plays a critical role in niche selection. In this study, we demonstrate that annexin II and its receptor play a crucial role in establishing metastasis of prostate cancer. Prostate cancer cell lines migrate toward annexin II and the adhesion of prostate cancer to osteoblasts and endothelial cells was inhibited by annexin II. By blocking annexin II or its receptor in animal models, short-term and long-term localization of prostate cancers are limited. Annexin II may also facilitate the growth of prostate cancer in vitro and in vivo by the MAPK pathway. These data strongly suggest that annexin II and its receptor axis plays a central role in prostate cancer metastasis, and that prostate cancer utilize the hematopoietic stem cell homing mechanisms to gain access to the niche.


Assuntos
Anexina A2/fisiologia , Fenômenos Fisiológicos Celulares , Metástase Neoplásica/patologia , Neoplasias da Próstata/patologia , Receptores de Peptídeos/fisiologia , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Técnicas de Cocultura , Células Endoteliais , Humanos , Masculino , Osteoblastos
9.
Am J Physiol Cell Physiol ; 295(3): C836-43, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18650261

RESUMO

Glucose transport is a highly regulated process and is dependent on a variety of signaling events. Glycogen synthase kinase-3 (GSK-3) has been implicated in various aspects of the regulation of glucose transport, but the mechanisms by which GSK-3 activity affects glucose uptake have not been well defined. We report that basal glycogen synthase kinase-3 (GSK-3) activity regulates glucose transport in several cell types. Chronic inhibition of basal GSK-3 activity (8-24 h) in several cell types, including vascular smooth muscle cells, resulted in an approximately twofold increase in glucose uptake due to a similar increase in protein expression of the facilitative glucose transporter 1 (GLUT1). Conversely, expression of a constitutively active form of GSK-3beta resulted in at least a twofold decrease in GLUT1 expression and glucose uptake. Since GSK-3 can inhibit mammalian target of rapamycin (mTOR) signaling via phosphorylation of the tuberous sclerosis complex subunit 2 (TSC2) tumor suppressor, we investigated whether chronic GSK-3 effects on glucose uptake and GLUT1 expression depended on TSC2 phosphorylation and TSC inhibition of mTOR. We found that absence of functional TSC2 resulted in a 1.5-to 3-fold increase in glucose uptake and GLUT1 expression in multiple cell types. These increases in glucose uptake and GLUT1 levels were prevented by inhibition of mTOR with rapamycin. GSK-3 inhibition had no effect on glucose uptake or GLUT1 expression in TSC2 mutant cells, indicating that GSK-3 effects on GLUT1 and glucose uptake were mediated by a TSC2/mTOR-dependent pathway. The effect of GSK-3 inhibition on GLUT1 expression and glucose uptake was restored in TSC2 mutant cells by transfection of a wild-type TSC2 vector, but not by a TSC2 construct with mutated GSK-3 phosphorylation sites. Thus, TSC2 and rapamycin-sensitive mTOR function downstream of GSK-3 to modulate effects of GSK-3 on glucose uptake and GLUT1 expression. GSK-3 therefore suppresses glucose uptake via TSC2 and mTOR and may serve to match energy substrate utilization to cellular growth.


Assuntos
Transportador de Glucose Tipo 1/metabolismo , Glucose/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular , Transportador de Glucose Tipo 1/genética , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Indóis/farmacologia , Maleimidas/farmacologia , Mutação , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fatores de Tempo , Transfecção , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
10.
Neoplasia ; 10(4): 371-80, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18392141

RESUMO

We developed a sensitive real-time polymerase chain reaction (QPCR) assay that allows us to track early lodging/homing events in vivo. We used this technology to develop a metastasis assay of human prostate cancer (PCa) growth in severe combined immunodeficient mice. For this purpose, marked human PCa cell lines were implanted subcutaneously or in the prostate (orthotopically) of severe combined immunodeficient mice as models of primary tumors. Mice were then sacrificed at various time points, and distant tissues were investigated for the presence of metastatic cells. At 3 weeks, a number of tissues were recovered and evaluated by QPCR for the presence of metastatic cells. The data demonstrate that several PCa cell lines are able to spread from the primary lesion and take up residence in distant sites. If the primary tumors were resected at 3 weeks, in several cases, metastatic lesions were identified over the course of 9 months. We propose that this new model may be particularly useful in exploring the molecular events in early metastasis, identifying the metastatic niche, and studying issues pertaining to dormancy.


Assuntos
Neoplasias Ósseas/secundário , Modelos Animais de Doenças , Neoplasias da Próstata/patologia , Animais , Neoplasias Ósseas/genética , DNA/genética , DNA/metabolismo , Humanos , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Células Neoplásicas Circulantes/patologia , Hormônio Paratireóideo/metabolismo , Reação em Cadeia da Polimerase , Neoplasias da Próstata/genética , Transplante Heterólogo , Células Tumorais Cultivadas
11.
J Cell Biochem ; 103(1): 1-8, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17541941

RESUMO

Tumor-associated macrophages (TAMs) have been implicated in promoting tumor growth and development. Here we present evidence that demonstrates that co-inoculation of male athymic nude mice with PC-3 prostate cancer cells and U937 promonocytic cells enhances tumor growth and increases tumor angiogenesis. Male athymic nude mice were co-inoculated with PC-3 and U937 cells (control or IL-4 stimulated) and tumor growth was monitored over time. Immunohistochemical analysis of tumor specimens was performed for proliferation markers (e.g., Ki67) and the effects of IL-4 stimulation on U937 cells were analyzed for chemokine expression. The presence of U937 cells increased the rate of tumor growth in vivo and stimulated increased microvascular density within the tumor bed. Stimulation of U937 cells with IL-4 resulted in a significant increase in several pro-angiogenic and pro-tumor chemokines (e.g., CCL2). Co-inoculation increases prostate cancer growth via upregulation of chemokines that induce angiogenesis within the tumor.


Assuntos
Neovascularização Patológica , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/patologia , Animais , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Técnicas de Cocultura , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-4/farmacologia , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células U937 , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Neoplasia ; 9(12): 1030-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18084610

RESUMO

PURPOSE: Upregulation of Bcl-2 family members is a well-established mechanism in the development of androgen-independent prostate cancer. Inhibition of the antiapoptotic proteins Bcl-2 and Mcl-1 may delay the transition to androgen-independent growth. EXPERIMENTAL DESIGN: We have established a prostate cancer model with VCaP prostate cancer cells in vivo to study the transition to androgen independence. Here, we investigated the efficacy of AT-101 (R-(-)-gossypol acetic acid; a pan small molecule inhibitor of Bcl-2, Bcl-x(L), and Mcl-1) in combination with surgical castration to delay the onset of androgen-independent growth in vivo. RESULTS: AT-101 (15 mg/kg, per os (p.o.) 5 days/week) in combination with surgical castration delayed the onset of androgen-independent prostate cancer growth in vivo. In addition, we demonstrate the induction of caspase-9-and caspase-3-dependent induction of apoptosis following AT-101 treatment in vitro which was accompanied by an AT-101-induced downregulation of Bcl-2 and Mcl-1 mRNA and protein expression. CONCLUSIONS: We conclude that AT-101 in combination with surgical castration delays the onset of androgen-independent prostate cancer in vivo by disrupting the antiapoptotic activity of Bcl-2 upregulation during the transition to androgen independence. Further studies are needed to define the mechanism of action by which AT-101 attenuates the expression of Bcl-2 and Mcl-1 and to characterize the potential for AT-101 in combination with hormone therapy.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Gossipol/análogos & derivados , Orquiectomia , Neoplasias da Próstata/tratamento farmacológico , Adenocarcinoma/cirurgia , Animais , Caspases/fisiologia , Terapia Combinada , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes bcl-2 , Gossipol/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos SCID , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neoplasias da Próstata/cirurgia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Res ; 67(19): 9417-24, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909051

RESUMO

The identification of novel tumor-interactive chemokines and the associated insights into the molecular and cellular basis of tumor-microenvironment interactions have continued to stimulate the development of targeted cancer therapeutics. Recently, we have identified monocyte chemoattractant protein 1 (MCP-1; CCL2) as a prominent regulator of prostate cancer growth and metastasis. Using neutralizing antibodies to human CCL2 (CNTO888) and the mouse homologue CCL2/JE (C1142), we show that treatment with anti-CCL2/JE antibody (2 mg/kg, twice weekly i.p.) attenuated PC-3Luc-mediated overall tumor burden in our in vivo model of prostate cancer metastasis by 96% at 5 weeks postintracardiac injection. Anti-CCL2 inhibition was not as effective as docetaxel (40 mg/kg, every week for 3 weeks) as a single agent, but inhibition of CCL2 in combination with docetaxel significantly reduced overall tumor burden compared with docetaxel alone, and induced tumor regression relative to initial tumor burden. These data suggest an interaction between tumor-derived chemokines and host-derived chemokines acting in cooperation to promote tumor cell survival, proliferation, and metastasis.


Assuntos
Anticorpos/farmacologia , Quimiocina CCL2/antagonistas & inibidores , Imunização Passiva/métodos , Imunoglobulina G/farmacologia , Neoplasias da Próstata/terapia , Animais , Anticorpos/imunologia , Processos de Crescimento Celular , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CCL2/imunologia , Docetaxel , Humanos , Imunoglobulina G/imunologia , Masculino , Camundongos , Camundongos SCID , Metástase Neoplásica , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Receptores CCR2 , Receptores de Quimiocinas/biossíntese , Taxoides/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Clin Invest ; 117(9): 2351-61, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17786228

RESUMO

Since the effectiveness of androgen deprivation for treatment of advanced prostate cancer was first demonstrated, prevention strategies and medical therapies for prostate cancer have been based on understanding the biologic underpinnings of the disease. Prostate cancer treatment is one of the best examples of a systematic therapeutic approach to target not only the cancer cells themselves, but the microenvironment in which they are proliferating. As the population ages and prostate cancer prevalence increases, challenges remain in the diagnosis of clinically relevant prostate cancer as well as the management of the metastatic and androgen-independent metastatic disease states.


Assuntos
Biologia , Neoplasias da Próstata/terapia , Androgênios/metabolismo , Transformação Celular Neoplásica/patologia , Humanos , Masculino , Programas de Rastreamento , Metástase Neoplásica , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/prevenção & controle
15.
Neoplasia ; 9(7): 556-62, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17710158

RESUMO

The ability of CCL2 to influence prostate cancer tumorigenesis and metastasis may occur through two distinct mechanisms: 1) a direct effect on tumor cell growth and function, and 2) an indirect effect on the tumor microenvironment by the regulation of macrophage mobilization and infiltration into the tumor bed. We have previously demonstrated that CCL2 exerts a direct effect on prostate cancer epithelial cells by the regulation of their growth, invasion, and migration, resulting in enhanced tumorigenesis and metastasis. Here we describe an indirect effect of CCL2 on prostate cancer growth and metastasis by regulating monocyte/macrophage infiltration into the tumor microenvironment and by stimulating a phenotypic change within these immune cells to promote tumor growth (tumor-associated macrophages). VCaP prostate cancer cells were subcutaneously injected in male SCID mice and monitored for tumor volume, CD68(+) macrophage infiltration, and microvascular density. Systemic administration of anti-CCL2 neutralizing antibodies (CNTO888 and C1142) significantly retarded tumor growth and attenuated CD68(+) macrophage infiltration, which was accompanied by a significant decrease in microvascular density. These data suggest that CCL2 contributes to prostate cancer growth through the regulation of macrophage infiltration and enhanced angiogenesis within the tumor.


Assuntos
Quimiocina CCL2/fisiologia , Quimiotaxia , Macrófagos/imunologia , Neoplasias da Próstata/patologia , Animais , Anticorpos Monoclonais/farmacologia , Quimiocina CCL2/antagonistas & inibidores , Quimiotaxia/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos SCID , Neoplasias da Próstata/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
CA Cancer J Clin ; 57(4): 225-41, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17626119

RESUMO

The last decade has seen an explosion in knowledge of the molecular basis and treatment of cancer. The molecular events that define the lethal phenotype of various cancers--the genetic and cellular alterations that lead to a cancer with a poor or incurable prognosis--are being defined. While these studies describe the cellular events of the lethal phenotype of cancer in detail, how these events result in the common clinical syndromes that kill the majority of cancer patients is not well understood. It is clear that the central step that makes most cancers incurable is metastasis. Understanding the traits that a cancer acquires to successfully grow and metastasize to distant sites gives insight into how tumors produce multiple factors that result in multiple different clinical syndromes that are lethal for the patient.


Assuntos
Neoplasias/genética , Fenótipo , Antineoplásicos , Caquexia/etiologia , Citocinas , Dispneia/etiologia , Humanos , Biologia Molecular , Metástase Neoplásica/genética , Neoplasias/mortalidade , Neoplasias/fisiopatologia , Prognóstico , Medição de Risco , Fatores de Risco , Trombose/etiologia
17.
J Cell Biochem ; 101(5): 1292-300, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17492768

RESUMO

Patients with advanced prostate cancer often exhibit increased activation of the coagulation system. The key activator of the coagulation cascade is the serine protease thrombin which is capable of eliciting numerous cellular responses. We previously reported that the thrombin receptor PAR1 is overexpressed in prostate cancer. To investigate further the role of PAR1 in prostate cancer metastasis, we examined the effects of thrombin activation on cell adhesion and motility in PC-3 prostate cancer cells. Activation of PAR1-induced dynamic cytoskeletal reorganization and reduced PC-3 binding to collagen I, collagen IV, and laminin (P < 0.01) but not fibronectin. Expression of the cell surface integrin receptors did not change as assessed by flow cytometry. Immunofluorescence microscopy revealed that PAR1 stimulation caused reorganization of the focal adhesions, suggesting that PAR1 activation in PC-3 cells may be modulating cell adhesion through integrin function but not expression. Furthermore, RhoA was activated upon stimulation with thrombin with subsequent cell contraction, decreased cell adhesion, and induced migration towards monocyte chemoattractant protein 1 (MCP-1; CCL2). Thus, it appears that thrombin stimulation plays a role in prostate cancer metastasis by decreasing cell adhesion to the extracellular matrix and positioning the cell in a "ready state" for migration in response to a chemotactic signal. Further exploration is needed to determine whether PAR1 activation affects other signaling pathways involved in prostate cancer.


Assuntos
Quimiocina CCL2/farmacologia , Quimiotaxia/efeitos dos fármacos , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Receptor PAR-1/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/enzimologia , Ativação Enzimática/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/enzimologia , Humanos , Integrinas/metabolismo , Masculino , Trombina/farmacologia , Proteína cdc42 de Ligação ao GTP/metabolismo
18.
Curr Cancer Drug Targets ; 6(8): 659-70, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17168671

RESUMO

The paradigm of cancer development and metastasis has been redefined to encompass a more comprehensive interaction between the tumor and microenvironment within which the tumor cells reside. Despite the realization that this more comprehensive relationship has changed the current paradigm of cancer research, the struggle continues to more completely understand the pathogenesis of the disease and the ability to appropriately identify and design novel targets for therapy. Chemokines and chemokine receptors are being investigated for their role in tumor development and metastasis and may prove to be useful therapeutic targets. The chemokine family is a complex network of molecules that are ubiquitously expressed and perform a variety of functions most notably regulating the immune system. Here we review the importance of chemokines in the tumor-stromal interaction and discuss current concepts for targeting the chemokine network.


Assuntos
Quimiocinas/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Receptores de Quimiocinas/antagonistas & inibidores , Movimento Celular , Proliferação de Células , Quimiocinas/fisiologia , Humanos , Macrófagos/fisiologia , Invasividade Neoplásica , Neoplasias/etiologia , Neoplasias/imunologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/etiologia , Receptores de Quimiocinas/fisiologia
19.
Cancer Metastasis Rev ; 25(4): 611-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17160712

RESUMO

The paradigm of cancer development and metastasis is a comprehensive, complex series of events that ultimately reflects a coordinated interaction between the tumor cell and the microenvironment within which the tumor cell resides. Despite the realization that this relationship has changed the current paradigm of cancer research, the struggle continues to more completely understand the pathogenesis of the disease and the ability to appropriately identify and design novel targets for therapy. A particular area of research that has added a significant understanding to cancer metastasis is the role of chemokines and chemokine receptors. Here we review the current concepts of CCL2 (monocyte chemoattractant protein 1) and its role in tumor metastasis with particular interest to its role in the development of bone metastases.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Quimiocina CCL2/metabolismo , Animais , Medula Óssea/metabolismo , Quimiocina CCL2/deficiência , Quimiocina CCL2/genética , Humanos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptores CCR2 , Receptores de Quimiocinas/metabolismo
20.
Neoplasia ; 8(7): 578-86, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16867220

RESUMO

Tumor cells in the bone interact with the microenvironment to promote tumor cell survival and proliferation, resulting in a lethal phenotype for patients with advanced prostate cancer. Monocyte chemoattractant protein 1 (CCL2) is a member of the CC chemokine family and is known to promote monocyte chemotaxis to sites of inflammation. Here we have shown that human bone marrow endothelial (HBME) cells secrete significantly higher levels of CCL2 compared to human aortic endothelial cells and human dermal microvascular endothelial cells. Furthermore, we demonstrate that CCL2 is a potent chemoattractant of prostate cancer epithelial cells, and that stimulation of PC-3 and VCaP cells resulted in a dose-dependent activation of PI3 kinase/Akt signaling pathway. Activation of the PI3 kinase/Akt pathway was found to be vital to the proliferative effects of CCL2 stimulation of both PC-3 and VCaP cells. Additionally, CCL2 stimulated the phosphorylation of p70-S6 kinase (a downstream target of Akt) and induced actin rearrangement, resulting in a dynamic morphologic change indicative of microspike formation. These data suggest that bone marrow endothelial cells are a major source of CCL2, and that an elevated secretion of CCL2 recruits prostate cancer epithelial cells to the bone microenvironment and regulates their proliferation rate.


Assuntos
Biomarcadores Tumorais , Quimiocina CCL2/biossíntese , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/metabolismo , Idoso , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Quimiotaxia , Células Epiteliais/metabolismo , Humanos , Masculino , Fosfatidilinositol 3-Quinases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...