Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 5665, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38969631

RESUMEN

The paradigm for macrophage characterization has evolved from the simple M1/M2 dichotomy to a more complex model that encompasses the broad spectrum of macrophage phenotypic diversity, due to differences in ontogeny and/or local stimuli. We currently lack an in-depth pan-cancer single cell RNA-seq (scRNAseq) atlas of tumour-associated macrophages (TAMs) that fully captures this complexity. In addition, an increased understanding of macrophage diversity could help to explain the variable responses of cancer patients to immunotherapy. Our atlas includes well established macrophage subsets as well as a number of additional ones. We associate macrophage composition with tumour phenotype and show macrophage subsets can vary between primary and metastatic tumours growing in sites like the liver. We also examine macrophage-T cell functional cross talk and identify two subsets of TAMs associated with T cell activation. Analysis of TAM signatures in a large cohort of immune checkpoint inhibitor-treated patients (CPI1000 + ) identify multiple TAM subsets associated with response, including the presence of a subset of TAMs that upregulate collagen-related genes. Finally, we demonstrate the utility of our data as a resource and reference atlas for mapping of novel macrophage datasets using projection. Overall, these advances represent an important step in both macrophage classification and overcoming resistance to immunotherapies in cancer.


Asunto(s)
Inmunoterapia , Neoplasias , Macrófagos Asociados a Tumores , Humanos , Inmunoterapia/métodos , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/patología , Neoplasias/genética , Microambiente Tumoral/inmunología , Análisis de la Célula Individual , Linfocitos T/inmunología , RNA-Seq , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Macrófagos/inmunología , Regulación Neoplásica de la Expresión Génica
2.
Nanomedicine ; 40: 102506, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34875352

RESUMEN

Oncolytic viruses (OVs) selectively replicate in and destroy cancer cells resulting in anti-tumor immunity. However, clinical use remains a challenge because of virus clearance upon intravenous delivery. OV packaging using a nanomedicine approach could overcome this. Here we encapsulate an oncolytic adenovirus (Ad[I/PPT-E1A]) into CCL2-coated liposomes in order to exploit recruitment of CCR2-expressing circulating monocytes into tumors. We demonstrate successful encapsulation of Ad[I/PPT-E1A] into CCL2-coated liposomes that were preferentially taken up by CCR2-expressing monocytes. No complex-related toxicities were observed following incubation with prostate tumor cells and the encapsulation did not affect virus oncolytic activity in vitro. Furthermore, intravenous administration of our nanomedicine resulted in a significant reduction in tumor size and pulmonary metastasis in prostate cancer-bearing mice whereby a 1000-fold less virus was needed compared to Ad[I/PPT-E1A] alone. Taken together our data provide an opportunity to target OVs via circulation to inaccessible tumors using liposome-assisted drug delivery.


Asunto(s)
Adenoviridae , Viroterapia Oncolítica , Adenoviridae/genética , Animales , Línea Celular Tumoral , Quimiocina CCL2/genética , Vectores Genéticos , Humanos , Liposomas , Masculino , Ratones , Monocitos , Viroterapia Oncolítica/métodos
3.
Cancer Res ; 81(21): 5399-5400, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725133

RESUMEN

Androgen deprivation therapy (ADT) is the front-line treatment for early and metastatic prostate cancer, and the development of tumor resistance to it has major clinical consequences. Cancer cells start to proliferate and tumors begin to regrow, requiring the administration of more generic anticancer treatments like surgery, radiotherapy, and/or chemotherapy. Tumor-associated macrophages are known to drive tumor resistance to a number of anti-cancer therapies. El-Kenawi and colleagues now demonstrate a novel mechanism underpinning their ability to do so in prostate tumors during ADT. This involves the accumulation of cholesterol by macrophages in tumors and its transfer to cancer cells, where it acts as a precursor for androgen biosynthesis and results in the activation of androgen receptors.See related article by El-Kenawi and colleagues, p. 5477.


Asunto(s)
Antagonistas de Andrógenos , Neoplasias de la Próstata Resistentes a la Castración , Antagonistas de Andrógenos/uso terapéutico , Humanos , Macrófagos , Masculino , Receptores Androgénicos
5.
Mol Cancer Ther ; 20(3): 589-601, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33298589

RESUMEN

Oncolytic viruses (OV) have been shown to activate the antitumor functions of specific immune cells like T cells. Here, we show OV can also reprogram tumor-associated macrophage (TAM) to a less immunosuppressive phenotype. Syngeneic, immunocompetent mouse models of primary breast cancer were established using PyMT-TS1, 4T1, and E0771 cell lines, and a metastatic model of breast cancer was established using the 4T1 cell line. Tumor growth and overall survival was assessed following intravenous administration of the OV, HSV1716 (a modified herpes simplex virus). Infiltration and function of various immune effector cells was assessed by NanoString, flow cytometry of dispersed tumors, and immunofluorescence analysis of tumor sections. HSV1716 administration led to marked tumor shrinkage in primary mammary tumors and a decrease in metastases. This was associated with a significant increase in the recruitment/activation of cytotoxic T cells, a reduction in the presence of regulatory T cells and the reprograming of TAMs towards a pro-inflammatory, less immunosuppressive phenotype. These findings were supported by in vitro data demonstrating that human monocyte-derived macrophages host HSV1716 replication, and that this led to immunogenic macrophage lysis. These events were dependent on macrophage expression of proliferating cell nuclear antigen (PCNA). Finally, the antitumor effect of OV was markedly diminished when TAMs were depleted using clodronate liposomes. Together, our results show that TAMs play an essential role in support of the tumoricidal effect of the OV, HSV1716-they both host viral replication via a novel, PCNA-dependent mechanism and are reprogramed to express a less immunosuppressive phenotype.


Asunto(s)
Macrófagos/metabolismo , Virus Oncolíticos/patogenicidad , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Mamarias Animales , Ratones , Transfección
6.
Oncogene ; 38(28): 5670-5685, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31043706

RESUMEN

The hypoxia-inducible transcription factor HIF-1 is appreciated as a promising target for cancer therapy. However, conditional deletion of HIF-1 and HIF-1 target genes in cells of the tumor microenvironment can result in accelerated tumor growth, calling for a detailed characterization of the cellular context to fully comprehend HIF-1's role in tumorigenesis. We dissected cell type-specific functions of HIF-1 for intestinal tumorigenesis by lineage-restricted deletion of the Hif1a locus. Intestinal epithelial cell-specific Hif1a loss reduced activation of Wnt/ß-catenin, tumor-specific metabolism and inflammation, significantly inhibiting tumor growth. Deletion of Hif1a in myeloid cells reduced the expression of fibroblast-activating factors in tumor-associated macrophages resulting in decreased abundance of tumor-associated fibroblasts (TAF) and robustly reduced tumor formation. Interestingly, hypoxia was detectable only sparsely and without spatial association with HIF-1α, arguing for an importance of hypoxia-independent, i.e., non-canonical, HIF-1 stabilization for intestinal tumorigenesis that has not been previously appreciated. This adds a further layer of complexity to the regulation of HIF-1 and suggests that hypoxia and HIF-1α stabilization can be uncoupled in cancer. Collectively, our data show that HIF-1 is a pivotal pro-tumorigenic factor for intestinal tumor formation, controlling key oncogenic programs in both the epithelial tumor compartment and the tumor microenvironment.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Oncogenes , Estabilidad Proteica , Microambiente Tumoral
7.
Nat Cell Biol ; 21(2): 190-202, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30598531

RESUMEN

Cytotoxic chemotherapy is an effective treatment for invasive breast cancer. However, experimental studies in mice also suggest that chemotherapy has pro-metastatic effects. Primary tumours release extracellular vesicles (EVs), including exosomes, that can facilitate the seeding and growth of metastatic cancer cells in distant organs, but the effects of chemotherapy on tumour-derived EVs remain unclear. Here we show that two classes of cytotoxic drugs broadly employed in pre-operative (neoadjuvant) breast cancer therapy, taxanes and anthracyclines, elicit tumour-derived EVs with enhanced pro-metastatic capacity. Chemotherapy-elicited EVs are enriched in annexin A6 (ANXA6), a Ca2+-dependent protein that promotes NF-κB-dependent endothelial cell activation, Ccl2 induction and Ly6C+CCR2+ monocyte expansion in the pulmonary pre-metastatic niche to facilitate the establishment of lung metastasis. Genetic inactivation of Anxa6 in cancer cells or Ccr2 in host cells blunts the pro-metastatic effects of chemotherapy-elicited EVs. ANXA6 is detected, and potentially enriched, in the circulating EVs of breast cancer patients undergoing neoadjuvant chemotherapy.


Asunto(s)
Doxorrubicina/uso terapéutico , Vesículas Extracelulares/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Paclitaxel/uso terapéutico , Animales , Anexina A6/metabolismo , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos
8.
Cancer Res ; 78(19): 5492-5503, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30206177

RESUMEN

Tumor-associated macrophages are a major constituent of malignant tumors and are known to stimulate key steps in tumor progression. In our review in this journal in 2006, we postulated that functionally distinct subsets of these cells exist in different areas within solid tumors. Here, we review the many experimental and clinical studies conducted since then to investigate the function(s), regulation, and clinical significance of macrophages in these sites. The latter include three sites of cancer cell invasion, tumor nests, the tumor stroma, and areas close to, or distant from, the tumor vasculature. A more complete understanding of macrophage diversity in tumors could lead to the development of more selective therapies to restore the formidable, anticancer functions of these cells. Cancer Res; 78(19); 5492-503. ©2018 AACR.


Asunto(s)
Macrófagos/patología , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral , Animales , Ensayos Clínicos como Asunto , Humanos , Hipoxia , Macrófagos/metabolismo , Ratones , Necrosis , Invasividad Neoplásica , Oncogenes , Fenotipo
9.
Nat Rev Immunol ; 18(11): 689-702, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30127389

RESUMEN

Macrophages are a heterogeneous group of cells that are capable of carrying out distinct functions in different tissues, as well as in different locations within a given tissue. Some of these tissue macrophages lie on, or close to, the outer (abluminal) surface of blood vessels and perform several crucial activities at this interface between the tissue and the blood. In steady-state tissues, these perivascular macrophages maintain tight junctions between endothelial cells and limit vessel permeability, phagocytose potential pathogens before they enter tissues from the blood and restrict inappropriate inflammation. They also have a multifaceted role in diseases such as cancer, Alzheimer disease, multiple sclerosis and type 1 diabetes. Here, we examine the important functions of perivascular macrophages in various adult tissues and describe how these functions are perturbed in a broad array of pathological conditions.


Asunto(s)
Células Endoteliales/citología , Endotelio Vascular/citología , Macrófagos/citología , Macrófagos/inmunología , Uniones Estrechas/inmunología , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Humanos , Ratones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Neoplasias/inmunología , Neoplasias/patología , Fagocitosis/inmunología
10.
11.
Cancer Cell ; 30(1): 18-25, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27411586

RESUMEN

Evidence has emerged for macrophages in the perivascular niche of tumors regulating important processes like angiogenesis, various steps in the metastatic cascade, the recruitment and activity of other tumor-promoting leukocytes, and tumor responses to frontline therapies like irradiation and chemotherapy. Understanding the mechanisms controlling the recruitment, retention, and function of these cells could identify important targets for anti-cancer therapeutics.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias/terapia , Animales , Progresión de la Enfermedad , Humanos , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica , Resultado del Tratamiento , Microambiente Tumoral
12.
Cancer Res ; 75(17): 3479-91, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26269531

RESUMEN

Tumor relapse after chemotherapy-induced regression is a major clinical problem, because it often involves inoperable metastatic disease. Tumor-associated macrophages (TAM) are known to limit the cytotoxic effects of chemotherapy in preclinical models of cancer. Here, we report that an alternatively activated (M2) subpopulation of TAMs (MRC1(+)TIE2(Hi)CXCR4(Hi)) accumulate around blood vessels in tumors after chemotherapy, where they promote tumor revascularization and relapse, in part, via VEGF-A release. A similar perivascular, M2-related TAM subset was present in human breast carcinomas and bone metastases after chemotherapy. Although a small proportion of M2 TAMs were also present in hypoxic tumor areas, when we genetically ablated their ability to respond to hypoxia via hypoxia-inducible factors 1 and 2, tumor relapse was unaffected. TAMs were the predominant cells expressing immunoreactive CXCR4 in chemotherapy-treated mouse tumors, with the highest levels expressed by MRC1(+) TAMs clustering around the tumor vasculature. Furthermore, the primary CXCR4 ligand, CXCL12, was upregulated in these perivascular sites after chemotherapy, where it was selectively chemotactic for MRC1(+) TAMs. Interestingly, HMOX-1, a marker of oxidative stress, was also upregulated in perivascular areas after chemotherapy. This enzyme generates carbon monoxide from the breakdown of heme, a gas known to upregulate CXCL12. Finally, pharmacologic blockade of CXCR4 selectively reduced M2-related TAMs after chemotherapy, especially those in direct contact with blood vessels, thereby reducing tumor revascularization and regrowth. Our studies rationalize a strategy to leverage chemotherapeutic efficacy by selectively targeting this perivascular, relapse-promoting M2-related TAM cell population.


Asunto(s)
Neoplasias de la Mama/genética , Macrófagos/patología , Recurrencia Local de Neoplasia/genética , Neovascularización Patológica/genética , Receptores CXCR4/biosíntesis , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Macrófagos/metabolismo , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neovascularización Patológica/tratamiento farmacológico , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética , Transducción de Señal/efectos de los fármacos , Tamoxifeno/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
13.
Oncotarget ; 5(24): 12472-508, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25537519

RESUMEN

During the past decades, anticancer immunotherapy has evolved from a promising therapeutic option to a robust clinical reality. Many immunotherapeutic regimens are now approved by the US Food and Drug Administration and the European Medicines Agency for use in cancer patients, and many others are being investigated as standalone therapeutic interventions or combined with conventional treatments in clinical studies. Immunotherapies may be subdivided into "passive" and "active" based on their ability to engage the host immune system against cancer. Since the anticancer activity of most passive immunotherapeutics (including tumor-targeting monoclonal antibodies) also relies on the host immune system, this classification does not properly reflect the complexity of the drug-host-tumor interaction. Alternatively, anticancer immunotherapeutics can be classified according to their antigen specificity. While some immunotherapies specifically target one (or a few) defined tumor-associated antigen(s), others operate in a relatively non-specific manner and boost natural or therapy-elicited anticancer immune responses of unknown and often broad specificity. Here, we propose a critical, integrated classification of anticancer immunotherapies and discuss the clinical relevance of these approaches.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Animales , Humanos
14.
Angiogenesis ; 17(1): 207-19, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24129822

RESUMEN

The binding of vascular endothelial growth factor (VEGF) to VEGF receptor-2 (VEGFR-2) on the surface of vascular endothelial cells stimulates many steps in the angiogenic pathway. Inhibition of this interaction is proving of value in moderating the neovascularization accompanying age-related macular degeneration and in the treatment of cancer. Tissue inhibitor of metalloproteinases-3 (TIMP-3) has been shown to be a natural VEGFR-2 specific antagonist-an activity that is independent of its ability to inhibit metalloproteinases. In this investigation we localize this activity to the C-terminal domain of the TIMP-3 molecule and characterize a short peptide, corresponding to part of this domain, that not only inhibits all three VEGF-family receptors, but also fibroblast growth factor and platelet-derived growth factor receptors. This multiple-receptor inhibition may explain why the peptide was also seen to be a powerful inhibitor of tumour growth and also a partial inhibitor of arthritic joint inflammation in vivo.


Asunto(s)
Artritis/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Péptidos/farmacología , Inhibidor Tisular de Metaloproteinasa-3/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Artritis/metabolismo , Artritis/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Degeneración Macular/patología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Péptidos/química , Inhibidor Tisular de Metaloproteinasa-3/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Cancer Cell ; 23(3): 277-86, 2013 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-23518347

RESUMEN

Tumor-associated macrophages (TAMs) promote key processes in tumor progression, like angiogenesis, immunosuppression, invasion, and metastasis. Increasing studies have also shown that TAMs can either enhance or antagonize the antitumor efficacy of cytotoxic chemotherapy, cancer-cell targeting antibodies, and immunotherapeutic agents--depending on the type of treatment and tumor model. TAMs also drive reparative mechanisms in tumors after radiotherapy or treatment with vascular-targeting agents. Here, we discuss the biological significance and clinical implications of these findings, with an emphasis on novel approaches that effectively target TAMs to increase the efficacy of such therapies.


Asunto(s)
Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Anticuerpos Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Progresión de la Enfermedad , Humanos , Inmunoterapia , Activación de Macrófagos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Neovascularización Patológica/tratamiento farmacológico
16.
Genesis ; 51(1): 41-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22927121

RESUMEN

Macrophages play an essential role in tissue homeostasis, innate immunity, inflammation, and wound repair. Macrophages are also essential during development, severely limiting the use of mouse models in which these cells have been constitutively deleted. Consequently, we have developed a transgenic model of inducible macrophage depletion in which macrophage-specific induction of the cytotoxic diphtheria toxin A chain (DTA) is achieved by administration of doxycycline. Induction of the DTA protein in transgenic animals resulted in a significant 50% reduction in CD68+ macrophages of the liver, spleen, and bone over a period of 6 weeks. Pertinently, the macrophages remaining after doxycycline treatment were substantially smaller and are functionally impaired as shown by reduced inflammatory cytokine production in response to lipopolysaccharide. This inducible model of macrophage depletion can now be utilized to determine the role of macrophages in both development and animal models of chronic inflammatory diseases.


Asunto(s)
Macrófagos/fisiología , Ratones Transgénicos , Modelos Animales , Animales , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Huesos/citología , Citocinas/inmunología , Toxina Diftérica/genética , Doxiciclina/toxicidad , Terapia de Inmunosupresión , Lipopolisacáridos/inmunología , Hígado/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Fragmentos de Péptidos/genética , Bazo/citología
17.
Cancer Res ; 73(2): 490-5, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23172310

RESUMEN

Frontline anticancer therapies such as chemotherapy and irradiation often slow tumor growth, but tumor regrowth and spread to distant sites usually occurs after the conclusion of treatment. We recently showed that macrophages could be used to deliver large quantities of a hypoxia-regulated, prostate-specific oncolytic virus (OV) to prostate tumors. In the current study, we show that administration of such OV-armed macrophages 48 hours after chemotherapy (docetaxel) or tumor irradiation abolished the posttreatment regrowth of primary prostate tumors in mice and their spread to the lungs for up to 27 or 40 days, respectively. It also significantly increased the lifespan of tumor-bearing mice compared with those given docetaxel or irradiation alone. These new findings suggest that such a novel, macrophage-based virotherapy could be used to markedly increase the efficacy of chemotherapy and irradiation in patients with prostate cancer.


Asunto(s)
Macrófagos/virología , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Taxoides/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Terapia Combinada , Docetaxel , Humanos , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Masculino , Ratones , Neoplasias de la Próstata/radioterapia , Recurrencia , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Clin Invest ; 121(5): 1969-73, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21490397

RESUMEN

Vascular-disrupting agents (VDAs) such as combretastatin A4 phosphate (CA4P) selectively disrupt blood vessels in tumors and induce tumor necrosis. However, tumors rapidly repopulate after treatment with such compounds. Here, we show that CA4P-induced vessel narrowing, hypoxia, and hemorrhagic necrosis in murine mammary tumors were accompanied by elevated tumor levels of the chemokine CXCL12 and infiltration by proangiogenic TIE2-expressing macrophages (TEMs). Inhibiting TEM recruitment to CA4P-treated tumors either by interfering pharmacologically with the CXCL12/CXCR4 axis or by genetically depleting TEMs in tumor-bearing mice markedly increased the efficacy of CA4P treatment. These data suggest that TEMs limit VDA-induced tumor injury and represent a potential target for improving the clinical efficacy of VDA-based therapies.


Asunto(s)
Macrófagos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Estilbenos/farmacología , Animales , Antineoplásicos Fitogénicos/farmacología , Separación Celular , Quimiocina CXCL12/metabolismo , Femenino , Citometría de Flujo , Neoplasias Mamarias Animales , Ratones , Ratones Transgénicos , Necrosis/patología , Trasplante de Neoplasias , Receptor TIE-2 , Receptores CXCR4/metabolismo
19.
Cancer Cell ; 19(4): 431-3, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21481783

RESUMEN

In this issue of Cancer Cell, Mazzieri, Pucci, and colleagues describe the marked effects of inhibiting the proangiogenic cytokine, Angiopoietin-2, on tumor angiogenesis and progression in spontaneous tumor models, as well as the proangiogenic functions of TIE2-expressing macrophages.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...