Abstract

Abstract Metastases arising in distant tissues continue to cause the majority of all cancer-related deaths. The uncertainty of whether and when metastases will occur poses a major challenge to treatment effectiveness. In many cancer patients, persistent disseminated tumor cells (DTCs) enter a state of dormancy upon arrival at the distant tissue, only to awaken years or even decades afterwards and initiate deadly metastases. How are DTCs kept dormant, what prompts them to awaken, and how can dormant DTCs be targeted are pressing questions in cancer research. Because cancer dormancy provides a unique therapeutic window for preventing metastatic disease, a comprehensive understanding of the distribution, composition and dynamics of reservoirs of dormant DTCs is imperative. Recently, we developed a tool to follow dormant DTCs, and surveyed the anatomical distribution of dormant reservoirs in models of breast cancer metastasis. We discovered that DTCs laid preferentially dormant in the liver, and spatially distinct liver sub-microenvironments allow coexistence of dormant DTCs and rare growing metastases even within the same tissue. To explore whether the distinct fates of DTCs stemmed from intra-organ microenvironmental differences, we performed global transcriptomic profiling on hepatic milieus corresponding to dormancy and metastasis. We found that stromal cells clustered on the basis of disease stage, and genes that were upregulated in dormancy samples were associated with host defense processes and natural killer (NK) cell-mediated response. A thorough examination of viable populations of immune cells in each hepatic milieu revealed that NK cells were the only type of immune cell that increased in dormancy milieus. Gain- and loss-of-function experiments in vivo further indicated that the pool of NK cells encountered by DTCs is decisive on their awakening. Remarkably, adjuvant interleukin-15-based immunotherapy ensured an abundant pool of NK cells that sustained dormancy, thereby preventing hepatic metastases and prolonging survival. In addition, we demonstrated in vivo and in liver-like co-cultures that NK cells control dormant DTCs through IFN-γ-induced quiescence. Thus, we uncovered an alternative cytostatic IFNγ-mediated mechanism of NK cell immunity that is essential to the control of breast cancer dormancy. This finding reroutes the importance of IFNγ to much earlier stages of disease, and encourages its therapeutic use for establishing the non-permissive cytokine milieu that is necessary to limit the emergence of DTCs from dormancy. Another key finding of our work is that liver injury limits NK cell expansion, thus triggering the switch from dormancy to liver metastasis. Using a model of chemically-induced liver injury, we found that activated hepatic stellate cells (aHSCs, which activate from quiescent HSCs to proliferative myofibroblasts) orchestrate a stromal response that hampers NK cell-mediated immunity and precipitates metastasis. Mechanistically, we show that aHSCs secrete the chemokine CXCL12, inducing NK cell quiescence via its cognate receptor CXCR4. This NK cell-inhibitory function of CXCL12 adds to its canonical effect as an inducer of DTC proliferation, and contributes to metastatic outgrowth. Our findings revive clinical interest on the use of CXCR4 inhibitors in the treatment of patients with cancer, but suggest that they should be repurposed to earlier stages to prevent progression of dormant disease. Because the size of the NK cell pool can itself determine metastatic outgrowth, a decrease of NK cells in patients with cancer who are apparently disease-free might identify individuals who are at risk of recurrence and who would benefit from CXCR4 inhibition therapy - a question that we will address in a clinical trial that we are currently setting up at the University Hospital Basel. All this work, which I am the first and corresponding author of, was recently published in Nature (Correia et al. Nature, 2021). In summary, our study shows that DTC dormancy is achieved by preserving normal tissue physiology—particularly tissue innate immunity—and that disruptions of the latter present DTCs with an opportunity for reactivation. In patients, this is reflected in the invariable decline in host defense capabilities that occurs during ageing, but also in the increased risk that recurrent infections and lifestyle-related injury triggers (such as alcohol, obesity and smoking) bring to the sprouting of site-specific metastases. We envision adjuvant NK cell immunotherapy as a means to preserve tissue physiology and prevent metastatic disease. Citation Format: Ana Luisa Correia. Revisiting NK cell immunity to prevent metastasis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr SY31-02.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call