Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Nat Cell Biol ; 21(4): 511-521, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30886344

RESUMO

Recent studies have revealed a role for macrophages and neutrophils in limiting chemotherapy efficacy; however, the mechanisms underlying the therapeutic benefit of myeloid-targeting agents in combination with chemotherapy are incompletely understood. Here, we show that targeting tumour-associated macrophages by colony-stimulating factor-1 receptor (CSF-1R) blockade in the K14cre;Cdh1F/F;Trp53F/F transgenic mouse model for breast cancer stimulates intratumoural type I interferon (IFN) signalling, which enhances the anticancer efficacy of platinum-based chemotherapeutics. Notably, anti-CSF-1R treatment also increased intratumoural expression of type I IFN-stimulated genes in patients with cancer, confirming that CSF-1R blockade is a powerful strategy to trigger an intratumoural type I IFN response. By inducing an inflamed, type I IFN-enriched tumour microenvironment and by further targeting immunosuppressive neutrophils during cisplatin therapy, antitumour immunity was activated in this poorly immunogenic breast cancer mouse model. These data illustrate the importance of breaching multiple layers of immunosuppression during cytotoxic therapy to successfully engage antitumour immunity in breast cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Interferon Tipo I/fisiologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/secundário , Camundongos , Camundongos Knockout , Camundongos Transgênicos
2.
J Clin Neurosci ; 62: 207-211, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30678836

RESUMO

Advances in therapies for breast cancer with cerebral metastases has been slow, despite this being a common diagnosis, due to limited drug delivery by the blood brain barrier. Improvements in drug delivery for brain metastasis to target the metastases and bypass the blood brain barrier are necessary. In our study, we delivered an inhibitor of chemokine receptor 4 by convection enhanced delivery in a hyperosmotic solution to prevent brain metastasis in a mouse model of metastatic breast cancer. We found the inhibitor limited metastatic disease and more interestingly, the hyperosmotic solution targeted tumor tissue allowing for a higher accumulation of the therapy into tumor tissue. This finding has the potential to improve delivery of chemotherapeutic agents to brain metastases.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/secundário , Sistemas de Liberação de Medicamentos/métodos , Neoplasias Mamárias Experimentais/secundário , Receptores CXCR4/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Injeções Intraventriculares , Camundongos , Camundongos Nus , Concentração Osmolar , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Biochem Cell Biol ; 96(6): 825-831, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29894647

RESUMO

Despite the great progress in recent years, many aspects of the pathogenesis and progression of breast cancer remain unclear. A better understanding on the molecular mechanisms underlying metastasis and recurrence is crucial to improve the treatment of this lethal disease. MCF-7 cells were xenografted into mice until visible tumors developed, and the cells from tumor tissue and adjacent normal tissue were cultured with 3 passages as mass tumor (MT) cells and invasive tumor (IT) cells, respectively. Microarray analysis was performed to detect several viable microRNAs in these 2 types of cells. Further, miR-30 knockdown was used to investigate its role in tumor aggression. Relative levels of miR-30 were significantly higher in IT cells than MT cells. Knockdown of miR-30 in both MT and IT cells lowered cell proliferation and cell invasion abilities, and thus increased the survival time of mice xenografted with tumor cells. This study suggested that the knockdown of miR-30 decreased proliferation and invasion of carcinoma cells, giving rise to the potential of miR-30 as a tumor target or marker candidate for breast cancer therapy.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , MicroRNAs/metabolismo , Animais , Proliferação de Células , Feminino , Humanos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/secundário , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica/genética , Metástase Neoplásica/genética
4.
Bone ; 113: 77-88, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29753718

RESUMO

The tumor-cell microenvironment is recognized as a dynamic place where critical cell interactions occur and play an important role in altering tumorigenesis. While many studies have investigated the effects of cellular cross-talk within distinct tumor microenvironments, these interactions have yet to be fully examined in bone. It is well-established that many common cancers metastasize to bone, resulting in the development of tumor-induced bone disease (TIBD), a multi-facetted illness that is driven by complex cell interactions within the bone marrow. Our group has previously published that myeloid progenitor cells expand in the presence of tumors in bone, aligning with the notion that myeloid cells can act as tumor promotors. Several groups, including ours, have established that transforming growth factor ß (TGF-ß), an abundant growth factor in bone, can regulate both TIBD and myeloid expansion. TGF-ß inhibitors have been shown to increase bone volume, decrease bone destruction, and reduce but not eliminate tumor. Therefore, we hypothesize that inhibiting TGF-ß will reduce myeloid expansion leading to a reduction of tumor burden in bone and osteoclast-mediated bone loss, causing to an overall reduction in TIBD. To address this hypothesis, two different mouse models of breast cancer bone colonization were pre-treated with the TGF-ß neutralizing antibody, 1D11, prior to tumor inoculation (athymic: MDA-MB-231, BALB/c: 4T1) and continuously treated until sacrifice. Additionally, a genetically modified mouse model with a myeloid specific deletion of transforming growth factor beta receptor II (TGF-ßRII) (TGF-ßRIIMyeKO) was utilized in our studies. Systemic inhibition of TGF-ß lead to fewer osteolytic lesions, and reduced tumor burden in bone as expected from previous studies. Additionally, early TGF-ß inhibition affected expansion of distinct myeloid populations and shifted the cytokine profile of pro-tumorigenic factors in bone, 4T1 tumor cells, and bone-marrow derived macrophages. Similar observations were seen in tumor-bearing TGF-ßRIIMyeKO mice, where these mice contained fewer bone lesions and significantly less tumor burden in bone, suggesting that TGF-ß inhibition regulates myeloid expansion leading to a significant reduction in TIBD.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias Mamárias Experimentais/secundário , Células Progenitoras Mieloides/patologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Microambiente Tumoral/fisiologia , Animais , Feminino , Humanos , Camundongos , Camundongos Knockout
5.
Am J Med Sci ; 354(1): 54-63, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28755734

RESUMO

BACKGROUND: Cancer stem cells are a subpopulation of tumor cells that are capable of self-renewal, capable of tumor recurrence and metastasis, and in addition are resistant to current cancer therapies. Epigallocatechin-3-gallate is a type of catechin found in green tea that is known for its powerful chemoprotective ability. Hence, this study aimed to investigate the effect of epigallocatechin-3-gallate on 7, 12 dimethylbenzanthracene-induced tumor metastasis, angiogenesis and cancer stem cells. MATERIALS AND METHODS: For this purpose, 3 groups of virgin femal rats with 7,12 dimethylbenzanthracene-induced mammary cancer were treated using epigallocatechin-3-gallate, paclitaxel or their combination. RESULTS: It was found that epigallocatechin-3-gallate exhibited significant chemopreventive effects and anti-cancer stem cell activity through several pathways, including a significant decrease in the size and number of tumors per rat, significant amelioration of the oxidative stress markers' alterations and significant inhibition of CD44, VEGF, Ki-67 and MMP-2 expression associated with a significantly increased expression of caspase-3. Histopathologically, therapy with epigallocatechin-3-gallate resulted in marked necrosis of the neoplastic cells and the tumor masses were mostly replaced by proliferated fibrous tissue so that histological confirmation of a previous tumor was not possible at that site. However, in the combination therapy the neoplastic cells showed marked vacuolation, haphazard arrangement and extensive nuclear pyknosis accompanied with many apoptotic bodies. Therapy with the sole paclitaxel caused variable degrees of necrosis among the neoplastic cells. Additionally, the combination of epigallocatechin-3-gallate and paclitaxel significantly enhanced the later anticancer efficacy. CONCLUSIONS: Epigallocatechin-3-gallatecould be offered as an unprecedented curative strategy to eradicate cancer.


Assuntos
Anticarcinógenos/farmacologia , Catequina/análogos & derivados , Neoplasias Mamárias Experimentais/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , 9,10-Dimetil-1,2-benzantraceno/farmacologia , Animais , Carcinógenos/farmacologia , Catequina/farmacologia , Feminino , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/secundário , Ratos , Ratos Sprague-Dawley
6.
Acta Pharmacol Sin ; 38(6): 924-930, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28504249

RESUMO

Cancer metastasis is the primary cause of high mortality in breast cancer patients. In this study, we loaded an anti-cancer drug, cabazitaxel (CTX), into polymeric micelles (CTX-loaded polymeric micelles, PCMs), and explored their therapeutic efficacy in breast cancer metastasis. The characteristics of PCMs were investigated, and their anti-metastatic efficacy was assessed using in vitro and in vivo evaluations. PCMs had an average diameter of 50.13±11.96 nm with a CTX encapsulation efficiency of 97.02%±0.97%. PCMs could be effectively internalized into metastatic 4T1 breast cancer cells in vitro. CTX (10 ng/mL) or an equivalent concentration in PCMs did not significantly affected the viability of 4T1 cells, but dramatically decreased the cell migration activities. In an orthotopic metastatic breast cancer model, intravenously administered PCMs could be efficiently delivered to the tumor sites, resulting in a 71.6% inhibition of tumor growth and a 93.5% reduction of lung metastases. Taken together, our results verify the anti-metastatic efficacy of PCMs, thus providing an encouraging strategy for treating breast cancer metastasis.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Lactatos/química , Polietilenoglicóis/química , Taxoides/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Neoplasias da Mama/patologia , Neoplasias da Mama/secundário , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Lactatos/administração & dosagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Micelas , Tamanho da Partícula , Polietilenoglicóis/administração & dosagem , Relação Estrutura-Atividade , Propriedades de Superfície , Taxoides/administração & dosagem , Taxoides/química
7.
Sci Rep ; 6: 36173, 2016 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-27812027

RESUMO

This study aimed at evaluating the reliability and precision of Diffuse Luminescent Imaging Tomography (DLIT) for monitoring primary tumor and metastatic spreading in breast cancer mice, and to develop a biomathematical model to describe the collected data. Using orthotopic mammary fat pad model of breast cancer (MDAMB231-Luc) in mice, we monitored tumor and metastatic spreading by three-dimensional (3D) bioluminescence and cross-validated it with standard bioluminescence imaging, caliper measurement and necropsy examination. DLIT imaging proved to be reproducible and reliable throughout time. It was possible to discriminate secondary lesions from the main breast cancer, without removing the primary tumor. Preferential metastatic sites were lungs, peritoneum and lymph nodes. Necropsy examinations confirmed DLIT measurements. Marked differences in growth profiles were observed, with an overestimation of the exponential phase when using a caliper as compared with bioluminescence. Our mathematical model taking into account the balance between living and necrotic cells proved to be able to reproduce the experimental data obtained with a caliper or DLIT imaging, because it could discriminate proliferative living cells from a more composite mass consisting of tumor cells, necrotic cell, or inflammatory tissues. DLIT imaging combined with mathematical modeling could be a powerful and informative tool in experimental oncology.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/secundário , Neoplasias Mamárias Experimentais/secundário , Animais , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Imageamento Tridimensional , Medições Luminescentes , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Camundongos , Modelos Biológicos , Reprodutibilidade dos Testes , Tomografia Óptica/métodos
8.
Adv Exp Med Biol ; 904: 41-58, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26900062

RESUMO

In recent years, our serial investigations focused on the role of cancer cells-derived endogenous formaldehyde in bone cancer pain. We found that cancer cells produced formaldehyde through demethylation process by serine hydroxymethyltransferase (SHMT1 and SHMT2) and lysine-specific histone demethylase 1 (LSD1). When the cancer cells metastasized into bone marrow, the elevated endogenous formaldehyde induced bone cancer pain through activation on the transient receptor potential vanilloid subfamily member 1 (TRPV1) in the peripheral nerve fibers. More interestingly, TRPV1 expressions in the peripheral fibers were upregulated by the local insulin-like growth factor I (IGF-I) produced by the activated osteoblasts. In conclusion, tumor tissue-derived endogenous formaldehyde induced bone cancer pain via TRPV1 activation.


Assuntos
Neoplasias Ósseas/metabolismo , Dor do Câncer/fisiopatologia , Formaldeído/metabolismo , Proteínas de Neoplasias/fisiologia , Nervos Periféricos/metabolismo , Canais de Cátion TRPV/fisiologia , Animais , Neoplasias Ósseas/patologia , Neoplasias Ósseas/fisiopatologia , Dor do Câncer/tratamento farmacológico , Capsaicina/farmacologia , Células Cultivadas , Glicina Hidroximetiltransferase/metabolismo , Histona Desmetilases/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Neoplasias Mamárias Experimentais/fisiopatologia , Neoplasias Mamárias Experimentais/secundário , Osteoblastos/metabolismo , Nervos Periféricos/fisiopatologia , Podofilotoxina/análogos & derivados , Podofilotoxina/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/biossíntese , Canais de Cátion TRPV/genética
9.
Proc Natl Acad Sci U S A ; 113(8): 2223-8, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26858439

RESUMO

Circulating cancer cells can putatively colonize distant organs to form metastases or to reinfiltrate primary tumors themselves through a process termed "tumor self-seeding." Here we exploit this biological attribute to deliver tumor necrosis factor alpha (TNF), a potent antitumor cytokine, directly to primary and metastatic tumors in a mechanism that we have defined as "tumor self-targeting." For this purpose, we genetically engineered mouse mammary adenocarcinoma (TSA), melanoma (B16-F10), and Lewis lung carcinoma cells to produce and release murine TNF. In a series of intervention trials, systemic administration of TNF-expressing tumor cells was associated with reduced growth of both primary tumors and metastatic colonies in immunocompetent mice. We show that these malignant cells home to tumors, locally release TNF, damage neovascular endothelium, and induce massive cancer cell apoptosis. We also demonstrate that such tumor-cell-mediated delivery avoids or minimizes common side effects often associated with TNF-based therapy, such as acute inflammation and weight loss. Our study provides proof of concept that genetically modified circulating tumor cells may serve as targeted vectors to deliver anticancer agents. In a clinical context, this unique paradigm represents a personalized approach to be translated into applications potentially using patient-derived circulating tumor cells as self-targeted vectors for drug delivery.


Assuntos
Neoplasias Experimentais/terapia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Apoptose , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/secundário , Carcinoma Pulmonar de Lewis/terapia , Engenharia Celular , Linhagem Celular Tumoral , Terapia Baseada em Transplante de Células e Tecidos , Sistemas de Liberação de Medicamentos , Endotélio Vascular/patologia , Feminino , Humanos , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/secundário , Neoplasias Mamárias Experimentais/terapia , Melanoma Experimental/patologia , Melanoma Experimental/secundário , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/patologia , Neoplasias Experimentais/secundário , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapêutico , Transdução Genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
10.
PLoS One ; 10(7): e0134458, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26222311

RESUMO

The tumor microenvironment encompasses several stressful conditions for cancer cells such as hypoxia, oxidative stress and pH alterations. Galectin-3, a well-studied member of the beta-galactoside-binding animal family of lectins has been implicated in multiple steps of metastasis as cell-cell and cell-ECM adhesion, promotion of angiogenesis, cell proliferation and resistance to apoptosis. However, both its aberrantly up- and down-regulated expression was observed in several types of cancer. Thus, the mechanisms that regulate galectin-3 expression in neoplastic settings are not clear. In order to demonstrate the putative role of hypoxia in regulating galectin-3 expression in canine mammary tumors (CMT), in vitro and in vivo studies were performed. In malignant CMT cells, hypoxia was observed to induce expression of galectin-3, a phenomenon that was almost completely prevented by catalase treatment of CMT-U27 cells. Increased galectin-3 expression was confirmed at the mRNA level. Under hypoxic conditions the expression of galectin-3 shifts from a predominant nuclear location to cytoplasmic and membrane expressions. In in vivo studies, galectin-3 was overexpressed in hypoxic areas of primary tumors and well-established metastases. Tumor hypoxia thus up-regulates the expression of galectin-3, which may in turn increase tumor aggressiveness.


Assuntos
Galectina 3/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Cães , Feminino , Galectina 3/genética , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Hipóxia/metabolismo , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/secundário , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/secundário , Camundongos Nus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Transplante Heterólogo , Microambiente Tumoral , Regulação para Cima
11.
PLoS One ; 9(9): e105079, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25188396

RESUMO

Circulating tumor cells (CTCs) have been detected in the bloodstream of both early-stage and advanced cancer patients. However, very little is know about the dynamics of CTCs during cancer progression and the clinical relevance of longitudinal CTC enumeration. To address this, we developed a simple bioluminescence imaging assay to detect CTCs in mouse models of metastasis. In a 4T1 orthotopic metastatic mammary carcinoma mouse model, we demonstrated that this quantitative method offers sensitivity down to 2 CTCs in 0.1-1mL blood samples and high specificity for CTCs originating from the primary tumor, independently of their epithelial status. In this model, we simultaneously monitored blood CTC dynamics, primary tumor growth, and lung metastasis progression over the course of 24 days. Early in tumor development, we observed low numbers of CTCs in blood samples (10-15 cells/100 µL) and demonstrated that CTC dynamics correlate with viable primary tumor growth. To our knowledge, these data represent the first reported use of bioluminescence imaging to detect CTCs and quantify their dynamics in any cancer mouse model. This new assay is opening the door to the study of CTC dynamics in a variety of animal models. These studies may inform clinical decision on the appropriate timing of blood sampling and value of longitudinal CTC enumeration in cancer patients.


Assuntos
Medições Luminescentes/métodos , Neoplasias Mamárias Experimentais/sangue , Células Neoplásicas Circulantes/patologia , Animais , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Hemólise , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/secundário , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia
13.
Proc Natl Acad Sci U S A ; 111(32): 11774-9, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25071169

RESUMO

Impressive responses have been observed in patients treated with checkpoint inhibitory anti-programmed cell death-1 (PD-1) or anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) antibodies. However, immunotherapy against poorly immunogenic cancers remains a challenge. Here we report that treatment with both anti-PD-1 and anti-CTLA-4 antibodies was unable to eradicate large, modestly immunogenic CT26 tumors or metastatic 4T1 tumors. Cotreatment with epigenetic-modulating drugs and checkpoint inhibitors markedly improved treatment outcomes, curing more than 80% of the tumor-bearing mice. Functional studies revealed that the primary targets of the epigenetic modulators were myeloid-derived suppressor cells (MDSCs). A PI3K inhibitor that reduced circulating MDSCs also eradicated 4T1 tumors in 80% of the mice when combined with immune checkpoint inhibitors. Thus, cancers resistant to immune checkpoint blockade can be cured by eliminating MDSCs.


Assuntos
Imunoterapia/métodos , Células Mieloides/imunologia , Metástase Neoplásica/imunologia , Metástase Neoplásica/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Azacitidina/administração & dosagem , Benzamidas/administração & dosagem , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/secundário , Neoplasias Colorretais/terapia , Terapia Combinada , Epigênese Genética/efeitos dos fármacos , Feminino , Inibidores de Histona Desacetilases/administração & dosagem , Humanos , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/secundário , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/efeitos dos fármacos , Metástase Neoplásica/genética , Inibidores de Fosfoinositídeo-3 Quinase , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Piridinas/administração & dosagem
14.
Sci Transl Med ; 6(242): 242ra84, 2014 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-24964992

RESUMO

Antiangiogenic agents that block vascular endothelial growth factor (VEGF) signaling are important components of current cancer treatment modalities but are limited by alternative ill-defined angiogenesis mechanisms that allow persistent tumor vascularization in the face of continued VEGF pathway blockade. We identified prostaglandin E2 (PGE2) as a soluble tumor-derived angiogenic factor associated with VEGF-independent angiogenesis. PGE2 production in preclinical breast and colon cancer models was tightly controlled by cyclooxygenase-2 (COX-2) expression, and COX-2 inhibition augmented VEGF pathway blockade to suppress angiogenesis and tumor growth, prevent metastasis, and increase overall survival. These results demonstrate the importance of the COX-2/PGE2 pathway in mediating resistance to VEGF pathway blockade and could aid in the rapid development of more efficacious anticancer therapies.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Neoplasias Hepáticas/prevenção & controle , Neoplasias Hepáticas/secundário , Neoplasias Mamárias Experimentais/prevenção & controle , Neoplasias Mamárias Experimentais/secundário , Ensaios Antitumorais Modelo de Xenoenxerto , Inibidores da Angiogênese/farmacologia , Animais , Axitinibe , Carcinogênese/patologia , Linhagem Celular Tumoral , Células Clonais , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/metabolismo , Feminino , Humanos , Imidazóis/farmacologia , Indazóis/farmacologia , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Terapia Neoadjuvante , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Mol Cell Biol ; 34(17): 3229-43, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24934442

RESUMO

While the E2F transcription factors (E2Fs) have a clearly defined role in cell cycle control, recent work has uncovered new functions. Using genomic signature methods, we predicted a role for the activator E2F transcription factors in the mouse mammary tumor virus (MMTV)-polyomavirus middle T oncoprotein (PyMT) mouse model of metastatic breast cancer. To genetically test the hypothesis that the E2Fs function to regulate tumor development and metastasis, we interbred MMTV-PyMT mice with E2F1, E2F2, or E2F3 knockout mice. With the ablation of individual E2Fs, we noted alterations of tumor latency, histology, and vasculature. Interestingly, we noted striking reductions in metastatic capacity and in the number of circulating tumor cells in both the E2F1 and E2F2 knockout backgrounds. Investigating E2F target genes that mediate metastasis, we found that E2F loss led to decreased levels of vascular endothelial growth factor (Vegfa), Bmp4, Cyr61, Nupr1, Plod 2, P4ha1, Adamts1, Lgals3, and Angpt2. These gene expression changes indicate that the E2Fs control the expression of genes critical to angiogenesis, the remodeling of the extracellular matrix, tumor cell survival, and tumor cell interactions with vascular endothelial cells that facilitate metastasis to the lungs. Taken together, these results reveal that the E2F transcription factors play key roles in mediating tumor development and metastasis in addition to their well-characterized roles in cell cycle control.


Assuntos
Fatores de Transcrição E2F/fisiologia , Neoplasias Mamárias Experimentais/etiologia , Animais , Antígenos Transformantes de Poliomavirus , Fatores de Transcrição E2F/deficiência , Fatores de Transcrição E2F/genética , Fator de Transcrição E2F1/deficiência , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/fisiologia , Fator de Transcrição E2F2/deficiência , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F2/fisiologia , Fator de Transcrição E2F3/deficiência , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/fisiologia , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/secundário , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Knockout , Células Neoplásicas Circulantes/patologia , Neovascularização Patológica/genética , Infecções por Retroviridae/etiologia , Infecções por Retroviridae/patologia , Transdução de Sinais , Microambiente Tumoral , Infecções Tumorais por Vírus/etiologia , Infecções Tumorais por Vírus/patologia
16.
Mol Ther ; 21(11): 1996-2007, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24081029

RESUMO

The ERBB receptors are a family of heterodimerization partners capable of driving transformation and metastasis. While the therapeutic targeting of single receptors has proven efficacious, optimal targeting of this receptor family should target all oncogenic members simultaneously. The juxtamembrane domains of ERBB1, ERBB2, and ERBB3 are highly conserved and control various aspects of ERBB-dependent biology. In an effort to block those functions, we have targeted this domain with decoy peptides synthesized in tandem with a cell-penetrating peptide, termed EJ1. Treatment with EJ1 induces cell death, promotes the formation of inactive ERBB multimers, and results in simultaneous reduction of ERBB1, ERBB2, and ERBB3 activation. Treatment also results in the activation of myosin light chain-dependent cell blebbing while inactivating CaMKII signaling, coincident with the induction of cell death. EJ1 also directly translocates to mitochondria, correlating with a loss of mitochondrial membrane potential and production of reactive oxygen species. Finally, treatment of a mouse model of breast cancer with EJ1 results in the inhibition of tumor growth and metastasis without associated toxicities in normal cells. Overall, these data demonstrate that a portion of the ERBB jxm domain, when used as an intracellular decoy, can inhibit tumor growth and metastasis, representing a novel anticancer therapeutic.


Assuntos
Antineoplásicos/farmacologia , Peptídeos Penetradores de Células/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Metástase Neoplásica/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/uso terapêutico , Progressão da Doença , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Mamárias Experimentais/secundário , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Multimerização Proteica/efeitos dos fármacos , Receptores Proteína Tirosina Quinases/química , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Receptor ErbB-3/antagonistas & inibidores , Receptor ErbB-3/química , Receptor ErbB-3/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Anticancer Drugs ; 24(4): 344-54, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23407059

RESUMO

Emend, an NK1 antagonist, and dexamethasone are used to treat complications associated with metastatic brain tumours and their treatment. It has been suggested that these agents exert anticancer effects apart from their current use. The effects of the NK1 antagonists, Emend and N-acetyl-L-tryptophan, and dexamethasone on tumour growth were investigated in vitro and in vivo at clinically relevant doses. For animal experiments, a stereotaxic injection model of Walker 256 rat breast carcinoma cells into the striatum of Wistar rats was used. Emend treatment led to a decrease in tumour cell viability in vitro, although this effect was not replicated by N-acetyl-L-tryptophan. Dexamethasone did not decrease tumour cell viability in vitro but decreased tumour volume in vivo, likely to be through a reduction in tumour oedema, as indicated by the increase in tumour cell density. None of the agents investigated altered tumour cell replication or apoptosis in vivo. Inoculated animals showed increased glial fibrillary acidic protein and ionized calcium-binding adapter molecule 1 immunoreactivity indicative of astrocytes and microglia in the peritumoral area, whereas treatment with Emend and dexamethasone reduced the labelling for both glial cells. These results do not support the hypothesis that NK1 antagonists or dexamethasone exert a cytotoxic action on tumour cells, although these conclusions may be specific to this model and cell line.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/secundário , Carcinoma/secundário , Dexametasona/farmacologia , Neoplasias Mamárias Experimentais/secundário , Morfolinas/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Antagonistas dos Receptores de Neurocinina-1 , Triptofano/análogos & derivados , Animais , Aprepitanto , Astrócitos/patologia , Edema Encefálico/etiologia , Edema Encefálico/prevenção & controle , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Proteínas de Ligação ao Cálcio/análise , Carcinoma/tratamento farmacológico , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/transplante , Corpo Estriado/patologia , Dexametasona/administração & dosagem , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Proteína Glial Fibrilar Ácida/análise , Humanos , Técnicas In Vitro , Masculino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Proteínas dos Microfilamentos/análise , Microglia/patologia , Modelos Biológicos , Morfolinas/administração & dosagem , Morfolinas/uso terapêutico , Proteínas de Neoplasias/fisiologia , Distribuição Aleatória , Ratos , Ratos Wistar , Receptores da Neurocinina-1/fisiologia , Triptofano/administração & dosagem , Triptofano/farmacologia , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral
18.
J Clin Invest ; 122(10): 3718-30, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22945631

RESUMO

A promising strategy for cancer immunotherapy is to disrupt key pathways regulating immune tolerance, such as cytotoxic T lymphocyte-associated protein 4 (CTLA-4). However, the determinants of response to anti-CTLA-4 mAb treatment remain incompletely understood. In murine models, anti-CTLA-4 mAbs alone fail to induce effective immune responses to poorly immunogenic tumors but are successful when combined with additional interventions, including local ionizing radiation (IR) therapy. We employed an established model based on control of a mouse carcinoma cell line to study endogenous tumor-infiltrating CD8+ T lymphocytes (TILs) following treatment with the anti-CTLA-4 mAb 9H10. Alone, 9H10 monotherapy reversed the arrest of TILs with carcinoma cells in vivo. In contrast, the combination of 9H10 and IR restored MHC class I-dependent arrest. After implantation, the carcinoma cells had reduced expression of retinoic acid early inducible-1 (RAE-1), a ligand for natural killer cell group 2D (NKG2D) receptor. We found that RAE-1 expression was induced by IR in vivo and that anti-NKG2D mAb blocked the TIL arrest induced by IR/9H10 combination therapy. These results demonstrate that anti-CTLA-4 mAb therapy induces motility of TIL and that NKG2D ligation offsets this effect to enhance TILs arrest and antitumor activity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD8-Positivos/efeitos dos fármacos , Antígeno CTLA-4/antagonistas & inibidores , Imunoterapia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Neoplasias Mamárias Experimentais/terapia , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos da radiação , Linhagem Celular Tumoral/imunologia , Linhagem Celular Tumoral/transplante , Movimento Celular , Terapia Combinada , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos da radiação , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Antígenos H-2/imunologia , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/efeitos da radiação , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/radioterapia , Neoplasias Mamárias Experimentais/secundário , Camundongos , Camundongos Endogâmicos BALB C , Subfamília K de Receptores Semelhantes a Lectina de Células NK/antagonistas & inibidores , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Associadas à Matriz Nuclear/biossíntese , Proteínas Associadas à Matriz Nuclear/genética , Proteínas de Transporte Nucleocitoplasmático/biossíntese , Proteínas de Transporte Nucleocitoplasmático/genética , Receptores CXCR/genética , Receptores CXCR6 , Microambiente Tumoral/imunologia
19.
Int J Dev Biol ; 55(7-9): 671-84, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22161824

RESUMO

The present review describes molecular and cellular mechanisms of cancer invasion and metastasis as compared to mammary gland development considering communication inside and between ecosystems. At the level of the individual cell, invasion programs are written by an ecosystem of signalling pathways each of which steers several invasion-related cellular activities. At the supracellular level, communication within the epithelial compartment involves cells of the same origin, but with different phenotypes including stem cells. A similar interaction occurs between the various cells of the stromal compartment. Crucial for our understanding of tumor or mammary gland ecosystems are the mutual interactions between cells of the epithelial and cells of the stromal compartment. An update is provided for endothelial cells, cancer-associated fibroblasts and macrophages that are implicated in angiogenesis, desmoplasia and inflammation respectively. At the level of the organism, distant ecosystems, comprising primary tumor site, sites of metastasis, bone marrow and endocrine glands among others, are in continuous contact through circulating cells and soluble ligands. Our review suggests consideration of these ecosystems when designing therapeutic strategies.


Assuntos
Neoplasias da Mama/patologia , Mama/crescimento & desenvolvimento , Animais , Neoplasias da Mama/fisiopatologia , Neoplasias da Mama/secundário , Comunicação Celular , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Feminino , Humanos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/fisiopatologia , Neoplasias Mamárias Experimentais/secundário , Camundongos , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica/patologia , Metástase Neoplásica/fisiopatologia , Neovascularização Patológica , Transdução de Sinais , Nicho de Células-Tronco , Células Estromais/patologia , Células Estromais/fisiologia , Microambiente Tumoral
20.
Int J Hyperthermia ; 27(6): 563-72, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21846192

RESUMO

PURPOSE: The purpose of this study was to explore the effects of MIH and radiotherapy alone or combined on metastatic breast cancer and the underlying mechanisms. MATERIALS AND METHODS: A murine 4T1 metastatic breast cancer model was established and randomly assigned into four treatment groups: C (control), R (radiotherapy), MIH, and MIH+R. Tumour volume, lung metastasis, the expression of Bax and MMP-9, T cell subsets, serum cytokine levels, and mouse survival were evaluated. RESULTS: Group MIH + R showed significantly reduced tumour volume, lung metastasis, improved survival and increased Bax expression compared to group R or MIH (P<0.05). MMP-9 expression in the primary tumour tissue was significantly increased in group R compared to the other groups (P<0.05), which could be brought down by combined MIH treatment. Group MIH +R showed significantly higher CD4(+) T cell percentage as well as CD4(+)/CD8(+) cell ratio than group R (P<0.05). Group MIH+R showed significantly higher serum levels of TNF-α, IFN-γ and IL-2 than group R (P<0.05). CONCLUSIONS: MIH not only promotes the tumour-cell killing effect of radiotherapy through Bax-mediated cell death, but also improves cellular immunity in mice under radiotherapy and decreases the potential of radiotherapy to enhance MMP-9 expression, which leads to significant improvement in lung metastasis and overall survival of mice under combined treatment of MIH and R. This study is the first to have explored the effect of combined hyperthermia and radiotherapy on tumour metastasis and the underlying mechanisms. It provides insights into the application of MIH as an adjuvant to radiotherapy for metastatic breast cancer.


Assuntos
Hipertermia Induzida/métodos , Magnetismo , Neoplasias Mamárias Experimentais/terapia , Animais , Linhagem Celular Tumoral , Terapia Combinada , Citocinas/sangue , Feminino , Imunidade Celular , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/radioterapia , Neoplasias Mamárias Experimentais/secundário , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Análise de Sobrevida , Subpopulações de Linfócitos T/efeitos da radiação , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA