Metastasis articles within Nature Medicine

Featured

  • Article |

    In a phase 2 trial of patients with untreated, recurrent and progressive brain metastases treated with an anti-programmed cell death protein 1 inhibitor, the overall intracranial benefit rate was 42.1%, which met the prespecified primary endpoint.

    • Priscilla K. Brastianos
    • , Albert E. Kim
    •  & Ryan J. Sullivan
  • Article
    | Open Access

    Genomic analyses in a rapid autopsy cohort study of patients with melanoma identify the genetic and transcriptomic landscape of melanoma with acquired resistance to MAPK inhibitor and immune checkpoint blockade therapies, providing insights for the potential improvement of therapeutic strategies.

    • Sixue Liu
    • , Prashanthi Dharanipragada
    •  & Roger S. Lo
  • Research Highlight |

    The release of breast cancer cells into the circulation occurs mostly during rest — a surprising discovery that may help researchers to better study and target cancer metastasis.

    • Karen O’Leary
  • Research Briefing |

    Secretion of S100A9 by brain metastatic cells activates radioresistance. Blood S100A9 could be used as a biomarker to predict responses to irradiation and to guide a combination therapy with a radiosensitizer.

  • News & Views |

    A recent study investigates the contribution of epigenomic plasticity to lung metastasis in osteosarcoma. Changes in the enhancer landscape were found to be nonrandom and driven by selective forces in the microenvironment.

    • Heinrich Kovar
  • Article |

    Factors secreted by metastatic a primary tumors induce an early phenotypic switch in perivascular cells at distant pre-metastatic niches. By using sophisticated lineage-tracing mouse models, the authors demonstrate that enhanced KLF4 expression in these cells increases their ability to proliferate and migrate away from the vasculature, and augments fibronectin deposition, which contributes to metastatic growth. These findings increase the mechanistic understanding of the metastatic process and uncover a role for perivascular plasticity that could be targeted to prevent metastasis.

    • Meera Murgai
    • , Wei Ju
    •  & Rosandra N Kaplan
  • News & Views |

    In a recent study in mice, researchers combined tumor barcoding with unbiased genomic analysis and identified Cd109 as a hub gene involved in metastatic progression. They show that pharmacological inhibition of its downstream effectors JAK1 and STAT3 curtails metastatic growth.

    • Laura Pisarsky
    • , Jinxiang Dai
    •  & Cyrus M Ghajar
  • Article |

    In vivo screening of pro-metastatic factors in a genetically engineered mouse model of lung cancer uncovered the CD109–JAK–STAT3 axis as a key contributor of metastatic dissemination of lung cancer cells. Activation of this pathway predicts poor outcome in patients with cancer, and its pharmacological inhibition dramatically reduces the metastatic ability of tumor cells, suggesting that it might be an effective intervention in patients.

    • Chen-Hua Chuang
    • , Peyton G Greenside
    •  & Monte M Winslow
  • News & Views |

    A recent study confirms an association between vessel co-option and resistance to bevacizumab, an anti-vascular endothelial growth factor-A (VEGFA) antibody, in patients with liver metastases. The authors suggest a combined therapeutic strategy that reduces co-option in mice.

    • Kyrre E Emblem
    •  & Rakesh K Jain
  • News & Views |

    Cancer cachexia leads to involuntary weight loss resulting from the atrophy of skeletal muscle and adipose tissues. Now, in metastatic mouse models of cancer, investigators reveal a cross talk pathway between bone and muscle that provides a new understanding of wasting in advanced cancers.

    • Denis C Guttridge
  • Resource |

    Understanding tumor metastasis is crucial to developing more effective cancer therapies. Here McCreery et al. analyzed the mutational profile of metastases from chemically induced skin tumors in mice and found that parallel evolution of synchronously disseminated tumor cells underlies most metastasis.

    • Melissa Q McCreery
    • , Kyle D Halliwill
    •  & Allan Balmain
  • Review Article |

    Increasing understanding of how tumor cells metastasize, what secondary organs are the targets of disseminating tumor cells and what molecular mechanisms are involved in the metastatic cascade can provide a road map to translate new biological insights into clinical practice. As tumor metastasis remains the main cause of death for patients with cancer, this is an unmet clinical need that requires a thorough examination of the most recent and relevant translational research.

    • Liling Wan
    • , Klaus Pantel
    •  & Yibin Kang
  • Commentary |

    Promising advances in cancer therapy stemming from an increasing understanding of the molecular and genetic underpinnings of the tumorigenic process have been fueled by a strong, determined scientific community, influential patient advocacy groups and committed funding bodies. Despite these efforts, the development of effective drugs to prevent systemic dissemination of cancer cells or to eliminate overt metastasis in secondary organs remains a challenge to both researchers and physicians. In an attempt to tackle the most relevant and timely translational issues, a meeting held in 2012 as a result of a successful partnership between the Volkswagen Foundation and Nature Medicine brought together a group of metastasis research experts to identify the most important hurdles and help create a framework for potential clinical and translational strategies.

    • Thomas Brabletz
    • , David Lyden
    •  & Zena Werb
  • Between Bedside and Bench |

    Cancer resistance to targeted therapies seems to be a field of active research. But there are still open questions as to what drives drug resistance not only in metastatic tumor cells but also in disseminated tumor cells (DTCs) during adjuvant treatment, before metastases are established in other organs. Targeting this residual cancer disease or keeping these DTCs in a dormant state may be a way to stop progression to metastatic disease. For this, a further understanding of the biology of these cells is necessary. In 'Bedside to Bench', Bernhard Polzer and Christopher Klein put forward several scenarios to explain the different resistant mechanisms that might account for DTCs unresponsiveness to cancer drugs and emphasize the relevance of synchronizing targeted therapies with the changing responsive or dormant state of disseminated cancer cells in the clinic. In 'Bench to Bedside', Julio A Aguirre-Ghiso, Paloma Bragado and Maria Soledad Sosa discuss possible cell-intrinsic and microenvironment-derived signaling pathways that may be exploited to maintain dormancy in DTCs and explore the possibility of using dormancy gene signatures to identify individuals with dormant disease.

    • Bernhard Polzer
    •  & Christoph A Klein
  • Article |

    Whether the molecular drivers of tumor initiation are the same factors that promote metastasis during tumor progression is addressed in this report. In renal carcinoma, common primary driving alterations such as VHL loss do not necessarily correlate with outcome, and the authors show that additional epigenetic adaptations are required to unleash prometastatic behavior. Two important metastastic drivers, CXCR4 and CYTIP, are activated downstream of VHL loss through epigenetic reprogramming involving differential chromatin modification or DNA methylation, exemplifying the complex evolution of tumorigenic traits downstream of driving alterations.

    • Sakari Vanharanta
    • , Weiping Shu
    •  & Joan Massagué
  • News & Views |

    Reactivation of 'metastasis suppressor' genes holds great promise for the treatment of incurable malignancies. To date, only a few of these genes have been identified. A new study shows that breast cancer metastasis can be blunted by leukemia inhibitory factor (LIF) signaling through regulation of the Hippo pathway, linking metastatic growth to the regulation of a pathway involved in building organs during development (pages 1511–1517).

    • Stefano Piccolo
  • Article |

    The authors identify LIFR as a breast cancer metastasis suppressor by showing how its loss promotes metastasis without substantial effect on primary tumor growth. This function of LIFR involves promoting the membrane localization of Scribble and enabling the cytoplasmic sequestration of Hippo pathway transducers, thus involving this signaling pathway in metastasis control. LIFR loss is also observed in human breast tumors, where it correlates with poor prognosis.

    • Dahu Chen
    • , Yutong Sun
    •  & Li Ma
  • Article |

    The authors identify Irf7 and associated interferon signaling as an important factor suppressing bone metastasis of breast cancers. Irf7 is lost in experimental metastasis and human bone metastastic tissue, and this fosters an immunosuppressive environment that facilitates metastasis. Manipulating this innate immune signaling pathway emerging from tumor cells by interferon administration had beneficial effects in mouse models by reducing bone metastasis and increasing survival time.

    • Bradley N Bidwell
    • , Clare Y Slaney
    •  & Belinda S Parker
  • News & Views |

    A new study shows that melanoma-derived exosomes contribute to metastatic invasion by carrying messenger proteins that direct bone marrow–derived cells toward a prometastatic phenotype. This leads to the promotion of proangiogenic events and modification of the extracellular matrix at premetastatic sites (pages 883–891).

    • Rajasekharan Somasundaram
    •  & Meenhard Herlyn
  • News & Views |

    Metastatic colorectal cancer is a largely incurable disease with a pressing need for targeted therapies. A new study sheds light on a surprising interaction between FOXO3a and β-catenin in metastatic colorectal cancer, suggesting new therapeutic avenues for agents targeting the PI3K-AKT pathway (pages 892–901).

    • Yibing Yan
    •  & Mark R Lackner
  • Article |

    Exosomes can transfer proteins and nucleic acids from one cell to another, altering the phenotype of the recipient cell. In the case of cancer, tumor-derived exosomes have been shown to promote tumor cell proliferation. Now, in a mouse model of melanoma, Peinado et al. report that exosomes derived from highly metastatic tumor cells can influence bone marrow cells, resulting in increased recruitment of provasculogenic bone marrow progenitors to sites of metastasis, increased primary tumor growth and metastatic spread.

    • Héctor Peinado
    • , Maša Alečković
    •  & David Lyden
  • Article |

    The crosstalk between the transcriptional activity of β-catenin and FOXO3a reveals unexpected pro-metastatic cooperative effects of these pathways through concurrent modulation of cell adhesion and motility programs. In tumors with high FOXO3a and β-catenin activity, the proapoptotic effect of FOXO3a is subverted and the pro-proliferative effect of β-catenin is also dampened, but pro-metastatic pathways are activated. These findings suggest that caution should be exerted when using targeted inhibitors that activate FOXO3a in tumors with high β-catenin activity, as coactivation of both pathways also correlates with more aggressive disease in humans.

    • Stephan P Tenbaum
    • , Paloma Ordóñez-Morán
    •  & Héctor G Palmer
  • Letter |

    Ovarian tumors preferentially metastasize to the omentum, a peritoneal fat layer. This report proposes that the reasons for this predilection stem from the growth advantage conferred by adipocytes on ovarian cancer cells. Adipocytes seem to promote homing of cancer cells through adipokine secretion, and direct contact of the two cell types promotes metabolic changes that result in lipid transfer from adipocytes to tumor cells. Environmental-driven metabolic adaptations could be exploited to target omental metastasis.

    • Kristin M Nieman
    • , Hilary A Kenny
    •  & Ernst Lengyel
  • Resource |

    By implementing the conditions for orthotopic implantation of different types of human breast tumors, the authors have created a publicly available bank of new mouse models that more faithfully recreate individual tumor properties and provide individualized information about tumor behavior and prognosis.

    • Yoko S DeRose
    • , Guoying Wang
    •  & Alana L Welm
  • News & Views |

    Although the importance of epithelial to mesenchymal transition (EMT) is acknowledged in tumor metastasis, the contribution of the reverse process—MET—to cancer progression has been unclear. A new study shows that the miR-200 family regulates MET and metastatic colonization in breast cancer, suggesting that flexible transitions between EMT and MET, or epithelial-mesenchymal plasticity, may be crucial at different stages of metastasis (pages 1101–1108).

    • E W Thompson
    •  & I Haviv
  • Article |

    Postpartum involution of the mammary gland is a recognized risk factor for breast cancer. This report identifies a mechanism that could be at least partially responsible for the increased risk, involving both the elevated expression of COX-2 and its interaction with extracellular collagen, the deposition of which occurs during postpartum involution. Both these factors promote tumor growth and invasion in mice and correlate with poor prognosis in young women with breast cancer. The data suggest that ibuprofen treatment during involution is a safe and effective approach to diminish pregnancy-associated cancer.

    • Traci R Lyons
    • , Jenean O'Brien
    •  & Pepper Schedin
  • Article |

    The miR200 family regulates EMT through E-cadherin modulation and has been proposed to contribute to metastasis thusly. This report identifies a promoting role of miR-200 in metastatic colonization that involves a novel target, the tumor secretome. The correlation between miR-200 and metastasis in people with cancer supports the relevance of this biphasic, multifaceted role of miR-200.

    • Manav Korpal
    • , Brian J Ell
    •  & Yibin Kang
  • Article |

    Tenascin C is an extracellular matrix protein previously linked to breast cancer metastasis. Here the authors uncover how tenascin C promotes the fitness of metastasis-initiating cells by sustaining the stem and survival signaling pathways NOTCH and Wnt through specific regulation of Msi1 and Lgr5, respectively.

    • Thordur Oskarsson
    • , Swarnali Acharyya
    •  & Joan Massagué

Browse broader subjects

Browse narrower subjects