Your browser doesn't support javascript.
loading
Neutrophil Elastase Remodels Mammary Tumors to Facilitate Lung Metastasis.
Lulla, Amriti R; Akli, Said; Karakas, Cansu; Caruso, Joseph A; Warma, Lucas D; Fowlkes, Natalie W; Rao, Xiayu; Wang, Jing; Hunt, Kelly K; Watowich, Stephanie S; Keyomarsi, Khandan.
Affiliation
  • Lulla AR; Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Akli S; Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Karakas C; Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Caruso JA; Department of Pathology and Helen Diller Cancer Center, University of California, San Francisco, California.
  • Warma LD; Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Fowlkes NW; Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Rao X; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Wang J; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Hunt KK; Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Watowich SS; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Keyomarsi K; Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
Mol Cancer Ther ; 23(4): 492-506, 2024 Apr 02.
Article in En | MEDLINE | ID: mdl-37796181
ABSTRACT
Metastatic disease remains the leading cause of death due to cancer, yet the mechanism(s) of metastasis and its timely detection remain to be elucidated. Neutrophil elastase (NE), a serine protease secreted by neutrophils, is a crucial mediator of chronic inflammation and tumor progression. In this study, we used the PyMT model (NE+/+ and NE-/-) of breast cancer to interrogate the tumor-intrinsic and -extrinsic mechanisms by which NE can promote metastasis. Our results showed that genetic ablation of NE significantly reduced lung metastasis and improved metastasis-free survival. RNA-sequencing analysis of primary tumors indicated differential regulation of tumor-intrinsic actin cytoskeleton signaling pathways by NE. These NE-regulated pathways are critical for cell-to-cell contact and motility and consistent with the delay in metastasis in NE-/- mice. To evaluate whether pharmacologic inhibition of NE inhibited pulmonary metastasis and phenotypically mimicked PyMT NE-/- mice, we utilized AZD9668, a clinically available and specific NE inhibitor. We found AZD9668 treated PyMT-NE+/+ mice showed significantly reduced lung metastases, improved recurrence-free, metastasis-free and overall survival, and their tumors showed similar molecular alterations as those observed in PyMT-NE-/- tumors. Finally, we identified a NE-specific signature that predicts recurrence and metastasis in patients with breast cancer. Collectively, our studies suggest that genetic ablation and pharmacologic inhibition of NE reduces metastasis and extends survival of mouse models of breast cancer, providing rationale to examine NE inhibitors as a treatment strategy for the clinical management of patients with metastatic breast cancer.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pyridones / Sulfones / Breast Neoplasms / Lung Neoplasms Type of study: Prognostic_studies Limits: Animals / Female / Humans Language: En Journal: Mol Cancer Ther Journal subject: ANTINEOPLASICOS Year: 2024 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pyridones / Sulfones / Breast Neoplasms / Lung Neoplasms Type of study: Prognostic_studies Limits: Animals / Female / Humans Language: En Journal: Mol Cancer Ther Journal subject: ANTINEOPLASICOS Year: 2024 Document type: Article