Your browser doesn't support javascript.
loading
Evolution of Cancer Stem-like Cells in Endocrine-Resistant Metastatic Breast Cancers Is Mediated by Stromal Microvesicles.
Sansone, Pasquale; Berishaj, Marjan; Rajasekhar, Vinagolu K; Ceccarelli, Claudio; Chang, Qing; Strillacci, Antonio; Savini, Claudia; Shapiro, Lauren; Bowman, Robert L; Mastroleo, Chiara; De Carolis, Sabrina; Daly, Laura; Benito-Martin, Alberto; Perna, Fabiana; Fabbri, Nicola; Healey, John H; Spisni, Enzo; Cricca, Monica; Lyden, David; Bonafé, Massimiliano; Bromberg, Jacqueline.
Afiliação
  • Sansone P; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Berishaj M; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Rajasekhar VK; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Ceccarelli C; Department of Experimental, Diagnostic and Specialty Medicine, AlmaMater Studiorum, Università di Bologna, Bologna, Italy.
  • Chang Q; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Strillacci A; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Savini C; Department of Biological, Geological and Environmental Sciences, Università di Bologna, Bologna, Italy.
  • Shapiro L; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Bowman RL; Department of Experimental, Diagnostic and Specialty Medicine, AlmaMater Studiorum, Università di Bologna, Bologna, Italy.
  • Mastroleo C; Center for Applied Biomedical Research Laboratory, Policlinico Universitario S. Orsola-Malpighi AlmaMater Studiorum, Università di Bologna, Bologna, Italy.
  • De Carolis S; Department of Radiation Oncology, Kaiser Permanente, Oakland, California.
  • Daly L; Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Benito-Martin A; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Perna F; Department of Experimental, Diagnostic and Specialty Medicine, AlmaMater Studiorum, Università di Bologna, Bologna, Italy.
  • Fabbri N; Center for Applied Biomedical Research Laboratory, Policlinico Universitario S. Orsola-Malpighi AlmaMater Studiorum, Università di Bologna, Bologna, Italy.
  • Healey JH; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Spisni E; Department of Pediatrics, Cell and Developmental Biology, Children's Cancer and Blood Foundation Laboratories, Weill Cornell Medicine, New York, New York.
  • Cricca M; Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Lyden D; Orthopedics Service, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Bonafé M; Orthopedics Service, Memorial Sloan Kettering Cancer Center, New York, New York.
  • Bromberg J; Department of Biological, Geological and Environmental Sciences, Università di Bologna, Bologna, Italy.
Cancer Res ; 77(8): 1927-1941, 2017 04 15.
Article em En | MEDLINE | ID: mdl-28202520
ABSTRACT
The hypothesis that microvesicle-mediated miRNA transfer converts noncancer stem cells into cancer stem cells (CSC) leading to therapy resistance remains poorly investigated. Here we provide direct evidence supporting this hypothesis, by demonstrating how microvesicles derived from cancer-associated fibroblasts (CAF) transfer miR-221 to promote hormonal therapy resistance (HTR) in models of luminal breast cancer. We determined that CAF-derived microvesicles horizontally transferred miR-221 to tumor cells and, in combination with hormone therapy, activated an ERlo/Notchhi feed-forward loop responsible for the generation of CD133hi CSCs. Importantly, microvesicles from patients with HTR metastatic disease expressed high levels of miR-221. We further determined that the IL6-pStat3 pathway promoted the biogenesis of onco-miR-221hi CAF microvesicles and established stromal CSC niches in experimental and patient-derived breast cancer models. Coinjection of patient-derived CAFs from bone metastases led to de novo HTR tumors, which was reversed with IL6R blockade. Finally, we generated patient-derived xenograft (PDX) models from patient-derived HTR bone metastases and analyzed tumor cells, stroma, and microvesicles. Murine and human CAFs were enriched in HTR tumors expressing high levels of CD133hi cells. Depletion of murine CAFs from PDX restored sensitivity to HT, with a concurrent reduction of CD133hi CSCs. Conversely, in models of CD133neg, HT-sensitive cancer cells, both murine and human CAFs promoted de novo HT resistance via the generation of CD133hi CSCs that expressed low levels of estrogen receptor alpha. Overall, our results illuminate how microvesicle-mediated horizontal transfer of genetic material from host stromal cells to cancer cells triggers the evolution of therapy-resistant metastases, with potentially broad implications for their control. Cancer Res; 77(8); 1927-41. ©2017 AACR.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Neoplasias da Mama / Células Estromais / Antineoplásicos Hormonais / Micropartículas Derivadas de Células Tipo de estudo: Observational_studies / Prognostic_studies / Risk_factors_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Neoplasias da Mama / Células Estromais / Antineoplásicos Hormonais / Micropartículas Derivadas de Células Tipo de estudo: Observational_studies / Prognostic_studies / Risk_factors_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article