Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Nature ; 527(7579): 472-6, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26560033

ABSTRACT

The role of epithelial-to-mesenchymal transition (EMT) in metastasis is a longstanding source of debate, largely owing to an inability to monitor transient and reversible EMT phenotypes in vivo. Here we establish an EMT lineage-tracing system to monitor this process in mice, using a mesenchymal-specific Cre-mediated fluorescent marker switch system in spontaneous breast-to-lung metastasis models. We show that within a predominantly epithelial primary tumour, a small proportion of tumour cells undergo EMT. Notably, lung metastases mainly consist of non-EMT tumour cells that maintain their epithelial phenotype. Inhibiting EMT by overexpressing the microRNA miR-200 does not affect lung metastasis development. However, EMT cells significantly contribute to recurrent lung metastasis formation after chemotherapy. These cells survived cyclophosphamide treatment owing to reduced proliferation, apoptotic tolerance and increased expression of chemoresistance-related genes. Overexpression of miR-200 abrogated this resistance. This study suggests the potential of an EMT-targeting strategy, in conjunction with conventional chemotherapies, for breast cancer treatment.


Subject(s)
Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis/pathology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Cell Lineage , Cell Proliferation/drug effects , Cell Tracking , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Male , Mammary Neoplasms, Experimental/genetics , Mice , MicroRNAs/genetics , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Reproducibility of Results
2.
Proc Natl Acad Sci U S A ; 112(52): 16000-5, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26668367

ABSTRACT

Inflammation is inextricably associated with primary tumor progression. However, the contribution of inflammation to tumor outgrowth in metastatic organs has remained underexplored. Here, we show that extrinsic inflammation in the lungs leads to the recruitment of bone marrow-derived neutrophils, which degranulate azurophilic granules to release the Ser proteases, elastase and cathepsin G, resulting in the proteolytic destruction of the antitumorigenic factor thrombospondin-1 (Tsp-1). Genetic ablation of these neutrophil proteases protected Tsp-1 from degradation and suppressed lung metastasis. These results provide mechanistic insights into the contribution of inflammatory neutrophils to metastasis and highlight the unique neutrophil protease-Tsp-1 axis as a potential antimetastatic therapeutic target.


Subject(s)
Lung Neoplasms/metabolism , Neutrophils/metabolism , Peptide Hydrolases/metabolism , Pneumonia/metabolism , Thrombospondin 1/metabolism , Animals , Blotting, Western , Bone Marrow Transplantation , Cathepsin G/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Gene Expression , Leukocyte Elastase/metabolism , Lipopolysaccharides/administration & dosage , Lung Neoplasms/secondary , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neutrophils/drug effects , Neutrophils/enzymology , Proteolysis , Reverse Transcriptase Polymerase Chain Reaction , Serine Proteases/metabolism , Thrombospondin 1/genetics
3.
Adv Exp Med Biol ; 890: 75-110, 2016.
Article in English | MEDLINE | ID: mdl-26703800

ABSTRACT

The tumor microenvironment (TME) represents a milieu that enables tumor cells to acquire the hallmarks of cancer. The TME is heterogeneous in composition and consists of cellular components, growth factors, proteases, and extracellular matrix. Concerted interactions between genetically altered tumor cells and genetically stable intratumoral stromal cells result in an "activated/reprogramed" stroma that promotes carcinogenesis by contributing to inflammation, immune suppression, therapeutic resistance, and generating premetastatic niches that support the initiation and establishment of distant metastasis. The lungs present a unique milieu in which tumors progress in collusion with the TME, as evidenced by regions of aberrant angiogenesis, acidosis and hypoxia. Inflammation plays an important role in the pathogenesis of lung cancer, and pulmonary disorders in lung cancer patients such as chronic obstructive pulmonary disease (COPD) and emphysema, constitute comorbid conditions and are independent risk factors for lung cancer. The TME also contributes to immune suppression, induces epithelial-to-mesenchymal transition (EMT) and diminishes efficacy of chemotherapies. Thus, the TME has begun to emerge as the "Achilles heel" of the disease, and constitutes an attractive target for anti-cancer therapy. Drugs targeting the components of the TME are making their way into clinical trials. Here, we will focus on recent advances and emerging concepts regarding the intriguing role of the TME in lung cancer progression, and discuss future directions in the context of novel diagnostic and therapeutic opportunities.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Communication/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Lung Diseases, Obstructive/complications , Lung Diseases, Obstructive/drug therapy , Lung Diseases, Obstructive/genetics , Lung Diseases, Obstructive/metabolism , Lung Neoplasms/complications , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Pulmonary Emphysema/complications , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Tumor Microenvironment/genetics
4.
Sci Transl Med ; 8(329): 329ra34, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26962158

ABSTRACT

The vast majority of ovarian cancer-related deaths are caused by metastatic dissemination of tumor cells, resulting in subsequent organ failure. However, despite our increased understanding of the physiological processes involved in tumor metastasis, there are no clinically approved drugs that have made a major impact in increasing the overall survival of patients with advanced, metastatic ovarian cancer. We identified prosaposin (psap) as a potent inhibitor of tumor metastasis, which acts via stimulation of p53 and the antitumorigenic protein thrombospondin-1 (TSP-1) in bone marrow-derived cells that are recruited to metastatic sites. We report that more than 97% of human serous ovarian tumors tested express CD36, the receptor that mediates the proapoptotic activity of TSP-1. Accordingly, we sought to determine whether a peptide derived from psap would be effective in treating this form of ovarian cancer. To that end, we developed a cyclic peptide with drug-like properties derived from the active sequence in psap. The cyclic psap peptide promoted tumor regression in a patient-derived tumor xenograft model of metastatic ovarian cancer. Thus, we hypothesize that a therapeutic agent based on this psap peptide would have efficacy in treating patients with metastatic ovarian cancer.


Subject(s)
Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Peptides, Cyclic/therapeutic use , Saposins/chemistry , Thrombospondin 1/pharmacology , Tumor Microenvironment , Amino Acids/metabolism , Animals , CD36 Antigens/metabolism , Carcinoma, Ovarian Epithelial , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclization , Disease Models, Animal , Female , Humans , Mice, Inbred C57BL , Neoplasm Grading , Neoplasms, Glandular and Epithelial/secondary , Ovarian Neoplasms/secondary , Peptides, Cyclic/pharmacology , Regression Analysis , Tissue Array Analysis , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
5.
PLoS One ; 10(6): e0129123, 2015.
Article in English | MEDLINE | ID: mdl-26046767

ABSTRACT

Lung cancer is the leading cause of cancer related mortality worldwide, with non-small cell lung cancer (NSCLC) as the most prevalent form. Despite advances in treatment options including minimally invasive surgery, CT-guided radiation, novel chemotherapeutic regimens, and targeted therapeutics, prognosis remains dismal. Therefore, further molecular analysis of NSCLC is necessary to identify novel molecular targets that impact prognosis and the design of new-targeted therapies. In recent years, tumor "activated/reprogrammed" stromal cells that promote carcinogenesis have emerged as potential therapeutic targets. However, the contribution of stromal cells to NSCLC is poorly understood. Here, we show increased numbers of bone marrow (BM)-derived hematopoietic cells in the tumor parenchyma of NSCLC patients compared with matched adjacent non-neoplastic lung tissue. By sorting specific cellular fractions from lung cancer patients, we compared the transcriptomes of intratumoral myeloid compartments within the tumor bed with their counterparts within adjacent non-neoplastic tissue from NSCLC patients. The RNA sequencing of specific myeloid compartments (immature monocytic myeloid cells and polymorphonuclear neutrophils) identified differentially regulated genes and mRNA isoforms, which were inconspicuous in whole tumor analysis. Genes encoding secreted factors, including osteopontin (OPN), chemokine (C-C motif) ligand 7 (CCL7) and thrombospondin 1 (TSP1) were identified, which enhanced tumorigenic properties of lung cancer cells indicative of their potential as targets for therapy. This study demonstrates that analysis of homogeneous stromal populations isolated directly from fresh clinical specimens can detect important stromal genes of therapeutic value.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung/pathology , Myeloid Cells/pathology , Transcriptome , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Chemokine CCL7/genetics , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Lung/metabolism , Mice, Inbred C57BL , Myeloid Cells/metabolism , Osteopontin/genetics , RNA, Messenger/genetics
6.
Cancer Discov ; 3(5): 578-89, 2013 May.
Article in English | MEDLINE | ID: mdl-23633432

ABSTRACT

UNLABELLED: Metastatic tumors have been shown to establish permissive microenvironments for metastases via recruitment of bone marrow-derived cells. Here, we show that metastasis-incompetent tumors are also capable of generating such microenvironments. However, in these situations, the otherwise prometastatic Gr1(+) myeloid cells create a metastasis-refractory microenvironment via the induction of thrombospondin-1 (Tsp-1) by tumor-secreted prosaposin. Bone marrow-specific genetic deletion of Tsp-1 abolished the inhibition of metastasis, which was restored by bone marrow transplant from Tsp-1(+) donors. We also developed a 5-amino acid peptide from prosaposin as a pharmacologic inducer of Tsp-1 in Gr1(+) bone marrow cells, which dramatically suppressed metastasis. These results provide mechanistic insights into why certain tumors are deficient in metastatic potential and implicate recruited Gr1(+) myeloid cells as the main source of Tsp-1. The results underscore the plasticity of Gr1(+) cells, which, depending on the context, promote or inhibit metastasis, and suggest that the peptide could be a potential therapeutic agent against metastatic cancer. SIGNIFICANCE: The mechanisms of metastasis suppression are poorly understood. Here, we have identified a novel mechanism whereby metastasis-incompetent tumors generate metastasis-suppressive microenvironments in distant organs by inducing Tsp-1 expression in the bone marrow­derived Gr1+myeloid cells. A 5-amino acid peptide with Tsp-1­inducing activity was identified as a therapeutic agent against metastatic cancer.


Subject(s)
Antigens, Ly/metabolism , CD11b Antigen/metabolism , Neoplasms/metabolism , Thrombospondin 1/metabolism , Animals , Bone Marrow Cells/cytology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Oligopeptides/pharmacology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL