4.8 Article

Neutrophils escort circulating tumour cells to enable cell cycle progression

Journal

NATURE
Volume 566, Issue 7745, Pages 553-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41586-019-0915-y

Keywords

-

Funding

  1. European Research Council
  2. Swiss National Science Foundation
  3. Swiss Cancer League
  4. Basel Cancer League
  5. two Cantons of Basel through the ETH Zurich
  6. University of Basel

Ask authors/readers for more resources

A better understanding of the features that define the interaction between cancer cells and immune cells is important for the development of new cancer therapies(1). However, focus is often given to interactions that occur within the primary tumour and its microenvironment, whereas the role of immune cells during cancer dissemination in patients remains largely uncharacterized(2,3). Circulating tumour cells (CTCs) are precursors of metastasis in several types of cancer(4-6), and are occasionally found within the bloodstream in association with non-malignant cells such as white blood cells (WBCs)(7,8). The identity and function of these CTC-associated WBCs, as well as the molecular features that define the interaction between WBCs and CTCs, are unknown. Here we isolate and characterize individual CTC-associated WBCs, as well as corresponding cancer cells within each CTC-WBC cluster, from patients with breast cancer and from mouse models. We use single-cell RNA sequencing to show that in the majority of these cases, CTCs were associated with neutrophils. When comparing the transcriptome profiles of CTCs associated with neutrophils against those of CTCs alone, we detect a number of differentially expressed genes that outline cell cycle progression, leading to more efficient metastasis formation. Further, we identify cell-cell junction and cytokine-receptor pairs that define CTC-neutrophil clusters, representing key vulnerabilities of the metastatic process. Thus, the association between neutrophils and CTCs drives cell cycle progression within the bloodstream and expands the metastatic potential of CTCs, providing a rationale for targeting this interaction in treatment of breast cancer.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available