Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Cell ; 184(9): 2471-2486.e20, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33878291

ABSTRACT

Metastasis has been considered as the terminal step of tumor progression. However, recent genomic studies suggest that many metastases are initiated by further spread of other metastases. Nevertheless, the corresponding pre-clinical models are lacking, and underlying mechanisms are elusive. Using several approaches, including parabiosis and an evolving barcode system, we demonstrated that the bone microenvironment facilitates breast and prostate cancer cells to further metastasize and establish multi-organ secondary metastases. We uncovered that this metastasis-promoting effect is driven by epigenetic reprogramming that confers stem cell-like properties on cancer cells disseminated from bone lesions. Furthermore, we discovered that enhanced EZH2 activity mediates the increased stemness and metastasis capacity. The same findings also apply to single cell-derived populations, indicating mechanisms distinct from clonal selection. Taken together, our work revealed an unappreciated role of the bone microenvironment in metastasis evolution and elucidated an epigenomic reprogramming process driving terminal-stage, multi-organ metastases.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Tumor Microenvironment , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Dev Cell ; 56(8): 1100-1117.e9, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33878299

ABSTRACT

Estrogen receptor-positive (ER+) breast cancer exhibits a strong bone tropism in metastasis. How the bone microenvironment (BME) impacts ER signaling and endocrine therapy remains poorly understood. Here, we discover that the osteogenic niche transiently and reversibly reduces ER expression and activities specifically in bone micrometastases (BMMs), leading to endocrine resistance. As BMMs progress, the ER reduction and endocrine resistance may partially recover in cancer cells away from the osteogenic niche, creating phenotypic heterogeneity in macrometastases. Using multiple approaches, including an evolving barcoding strategy, we demonstrated that this process is independent of clonal selection, and represents an EZH2-mediated epigenomic reprogramming. EZH2 drives ER+ BMMs toward a basal and stem-like state. EZH2 inhibition reverses endocrine resistance. These data exemplify how epigenomic adaptation to BME promotes phenotypic plasticity of metastatic seeds, fosters intra-metastatic heterogeneity, and alters therapeutic responses. Our study provides insights into the clinical enigma of ER+ metastatic recurrences despite endocrine therapies.


Subject(s)
Adaptation, Physiological , Bone and Bones/pathology , Breast Neoplasms/pathology , Receptors, Estrogen/metabolism , Tumor Microenvironment , Animals , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Cell Communication , Clonal Evolution , Disease Models, Animal , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gap Junctions/metabolism , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , MCF-7 Cells , Mice , Neoplasm Micrometastasis , Osteogenesis , Signal Transduction
4.
Oncogene ; 40(5): 997-1011, 2021 02.
Article in English | MEDLINE | ID: mdl-33323970

ABSTRACT

Estrogen receptor alpha gene (ESR1) mutations occur frequently in ER-positive metastatic breast cancer, and confer clinical resistance to aromatase inhibitors. Expression of the ESR1 Y537S mutation induced an epithelial-mesenchymal transition (EMT) with cells exhibiting enhanced migration and invasion potential in vitro. When small subpopulations of Y537S ESR1 mutant cells were injected along with WT parental cells, tumor growth was enhanced with mutant cells becoming the predominant population in distant metastases. Y537S mutant primary xenograft tumors were resistant to the antiestrogen tamoxifen (Tam) as well as to estradiol (E2) withdrawal. Y537S ESR1 mutant primary tumors metastasized efficiently in the absence of E2; however, Tam treatment significantly inhibited metastasis to distant sites. We identified a nine-gene expression signature, which predicted clinical outcomes of ER-positive breast cancer patients, as well as breast cancer metastasis to the lung. Androgen receptor (AR) protein levels were increased in mutant models, and the AR agonist dihydrotestosterone significantly inhibited estrogen-regulated gene expression, EMT, and distant metastasis in vivo, suggesting that AR may play a role in distant metastatic progression of ESR1 mutant tumors.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Receptor alpha/genetics , Receptors, Androgen/genetics , Tamoxifen/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Estradiol/metabolism , Estrogens/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mutation/genetics , Neoplasm Metastasis , Receptors, Androgen/drug effects , Xenograft Model Antitumor Assays
5.
Nat Cancer ; 1(7): 709-722, 2020 07.
Article in English | MEDLINE | ID: mdl-35122036

ABSTRACT

Polyclonal metastases frequently arise from clusters of circulating tumor cells (CTCs). CTC clusters metastasize better than single CTCs, but the underlying molecular mechanisms are poorly understood. Here, we show that polyclonal metastatic seeds exhibit higher resistance to natural killer (NK) cell killing. Using breast cancer models, we observed higher proportions of polyclonal lung metastasis in immunocompetent mice compared with mice lacking NK cells. Depleting NK cells selectively increased monoclonal but not polyclonal metastases, suggesting that CTC clusters are less sensitive to NK-mediated suppression. Transcriptional analyses revealed that clusters have elevated expression of cell-cell adhesion and epithelial genes, which is associated with decreased expression of NK cell activating ligands. Furthermore, perturbing tumor cell epithelial status altered NK ligand expression and sensitivity to NK-mediated killing. Collectively, our findings show that NK cells can determine the fate of CTCs of different epithelial and mesenchymal states, and impact metastatic clonal evolution by favoring polyclonal seeding.


Subject(s)
Lung Neoplasms , Neoplastic Cells, Circulating , Animals , Cell Count , Killer Cells, Natural , Lung Neoplasms/metabolism , Mice , Monitoring, Immunologic
6.
Nat Cell Biol ; 21(9): 1113-1126, 2019 09.
Article in English | MEDLINE | ID: mdl-31451770

ABSTRACT

Cancer-induced immune responses affect tumour progression and therapeutic response. In multiple murine models and clinical datasets, we identified large variations of neutrophils and macrophages that define 'immune subtypes' of triple-negative breast cancer (TNBC), including neutrophil-enriched (NES) and macrophage-enriched subtypes (MES). Different tumour-intrinsic pathways and mutual regulation between macrophages (or monocytes) and neutrophils contribute to the development of a dichotomous myeloid compartment. MES contains predominantly macrophages that are CCR2-dependent and exhibit variable responses to immune checkpoint blockade (ICB). NES exhibits systemic and local accumulation of immunosuppressive neutrophils (or granulocytic myeloid-derived suppressor cells), is resistant to ICB, and contains a minority of macrophages that seem to be unaffected by CCR2 knockout. A MES-to-NES conversion mediated acquired ICB resistance of initially sensitive MES models. Our results demonstrate diverse myeloid cell frequencies, functionality and potential roles in immunotherapies, and highlight the need to better understand the inter-patient heterogeneity of the myeloid compartment.


Subject(s)
Immunotherapy , Myeloid Cells/immunology , Triple Negative Breast Neoplasms/therapy , Tumor Microenvironment/immunology , Animals , Disease Models, Animal , Female , Granulocytes/immunology , Immunotherapy/methods , Macrophages/immunology , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Neutrophils/immunology , Neutrophils/pathology , Triple Negative Breast Neoplasms/pathology
7.
Trends Cancer ; 5(2): 95-110, 2019 02.
Article in English | MEDLINE | ID: mdl-30755309

ABSTRACT

Metastasis to bones is determined by both intrinsic traits of metastatic tumor cells and properties appertaining to the bone microenvironment. Bone marrow niches are critical for all major steps of metastasis, including the seeding of disseminated tumor cells (DTCs) to bone, the survival of DTCs and microscopic metastases under dormancy, and the eventual outgrowth of overt metastases. In this review, we discuss the role of bone marrow niches in bone colonization. The emphasis is on complicated and dynamic nature of cancer cells-niche interaction, which may underpin the long-standing mystery of metastasis dormancy, and represent a therapeutic target for elimination of minimal residue diseases and prevention of life-taking, overt metastases.


Subject(s)
Bone Neoplasms/complications , Stem Cell Niche/genetics , Humans , Neoplasm Metastasis
8.
Dev Cell ; 45(6): 663-665, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29920271

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) is a key driver of cancer metastasis. In this issue of Developmental Cell, Aiello et al. (2018) demonstrate that an EMT mechanism involving protein internalization impacts cell migration, while Reichert et al. (2018) identify epithelial plasticity as a determinant of metastatic organotropism in pancreatic cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Pancreatic Neoplasms , Cell Movement , Humans , Neoplasm Metastasis
9.
Cancer Res ; 78(4): 845-852, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29437766

ABSTRACT

Circulating tumor cell (CTC) clusters may represent one of the key mechanisms initiating the metastasis process. However, the series of pathophysiologic events by which CTC clusters originate, enter the circulation, and reach the distant sites remain to be identified. The cellular and molecular mechanisms that provide survival advantage for CTC clusters during the transit in the blood stream are also still largely unknown. Understanding the biology of CTC clusters is critical to assess this unified scheme employed by cancer and to device strategies to overcome key pathways responsible for their improved metastatic potential. CTC clusters remain an underdeveloped area of research begging the attention of multidisciplinary cancer research teams. Here, we provide insight on existing preclinical evidence on the potential mechanisms leading to CTC cluster formation and dissemination and on processes that may offer survival advantage. We also offer our perspective on future directions to delineate the role of CTC clusters in metastatic cascade and discuss their clinical significance. Cancer Res; 78(4); 845-52. ©2018 AACR.


Subject(s)
Neoplastic Cells, Circulating/metabolism , Humans , Neoplastic Cells, Circulating/pathology
10.
Sci China Life Sci ; 60(10): 1125-1132, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29027156

ABSTRACT

Breast cancer cells may disseminate early, before tumor diagnosis. Disseminated tumor cells, or DTCs, reside in the bone marrow, and may persist for years or even decades. Some of these cells may be re-activated to resume aggressive growth, and eventually become overt bone metastases. Recent studies have begun to shed light on this complicated process and revealed multiple steps and intermediate states of colonizing DTCs. However, how cancer-host interactions evolve during this process needs to be further understood. Most of our current knowledge of the bone microenvironment is obtained through studies looking for the hematopoietic stem cell (HSC) niche. Although this long-standing question has not yet been resolved, our search for the HSC niche has resulted in a detailed map of various cell types in the bone marrow. Furthermore, various techniques used to find the HSC niche may also be adapted for finding the cancer cell niche. In this article, we will review the recent progress in both the DTC and HSC areas with a focus on their potential microenvironment niches. We will also discuss how to apply what we have learned from HSC studies to map DTCs in the bone context. We hope to stimulate thoughts and ideas to further elucidate the bone colonization process, and develop potential therapeutic interventions.


Subject(s)
Bone Marrow Cells/pathology , Bone Marrow/pathology , Neoplasms/pathology , Stem Cell Niche , Animals , Cell Movement , Hematopoietic Stem Cells/pathology , Humans , Models, Biological , Neoplasm Metastasis
11.
Nat Commun ; 8: 15045, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28429794

ABSTRACT

The majority of breast cancer models for drug discovery are based on orthotopic or subcutaneous tumours. Therapeutic responses of metastases, especially microscopic metastases, are likely to differ from these tumours due to distinct cancer-microenvironment crosstalk in distant organs. Here, to recapitulate such differences, we established an ex vivo bone metastasis model, termed bone-in-culture array or BICA, by fragmenting mouse bones preloaded with breast cancer cells via intra-iliac artery injection. Cancer cells in BICA maintain features of in vivo bone micrometastases regarding the microenvironmental niche, gene expression profile, metastatic growth kinetics and therapeutic responses. Through a proof-of-principle drug screening using BICA, we found that danusertib, an inhibitor of the Aurora kinase family, preferentially inhibits bone micrometastases. In contrast, certain histone methyltransferase inhibitors stimulate metastatic outgrowth of indolent cancer cells, specifically in the bone. Thus, BICA can be used to investigate mechanisms involved in bone colonization and to rapidly test drug efficacies on bone micrometastases.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinases/antagonists & inhibitors , Benzamides/pharmacology , Bone Neoplasms/drug therapy , Bone and Bones/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Animals , Aurora Kinases/genetics , Aurora Kinases/metabolism , Benzamides/adverse effects , Biphenyl Compounds , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Bone and Bones/enzymology , Bone and Bones/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression , High-Throughput Screening Assays , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Morpholines , Pyridones/adverse effects , Tissue Culture Techniques , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...