Your browser doesn't support javascript.
loading
Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring.
Shi, Yu; Gao, Weina; Lytle, Nikki K; Huang, Peiwu; Yuan, Xiao; Dann, Amanda M; Ridinger-Saison, Maya; DelGiorno, Kathleen E; Antal, Corina E; Liang, Gaoyang; Atkins, Annette R; Erikson, Galina; Sun, Huaiyu; Meisenhelder, Jill; Terenziani, Elena; Woo, Gyunghwi; Fang, Linjing; Santisakultarm, Thom P; Manor, Uri; Xu, Ruilian; Becerra, Carlos R; Borazanci, Erkut; Von Hoff, Daniel D; Grandgenett, Paul M; Hollingsworth, Michael A; Leblanc, Mathias; Umetsu, Sarah E; Collisson, Eric A; Scadeng, Miriam; Lowy, Andrew M; Donahue, Timothy R; Reya, Tannishtha; Downes, Michael; Evans, Ronald M; Wahl, Geoffrey M; Pawson, Tony; Tian, Ruijun; Hunter, Tony.
Afiliación
  • Shi Y; Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA. yshi@salk.edu.
  • Gao W; Department of Chemistry, Southern University of Science and Technology, Shenzhen, China.
  • Lytle NK; Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
  • Huang P; Department of Pharmacology, University of California San Diego School of Medicine, La Jolla, CA, USA.
  • Yuan X; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA.
  • Dann AM; Department of Chemistry, Southern University of Science and Technology, Shenzhen, China.
  • Ridinger-Saison M; Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
  • DelGiorno KE; State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China.
  • Antal CE; Department of Chemistry, Southern University of Science and Technology, Shenzhen, China.
  • Liang G; Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
  • Atkins AR; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
  • Erikson G; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Sun H; Trovagene, San Diego, CA, USA.
  • Meisenhelder J; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Terenziani E; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Woo G; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Fang L; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Santisakultarm TP; Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Manor U; Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Xu R; Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Becerra CR; Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Borazanci E; Crown Bioscience San Diego, San Diego, CA, USA.
  • Von Hoff DD; Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Grandgenett PM; Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Hollingsworth MA; Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Leblanc M; Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Umetsu SE; Institute of Oncology, Shenzhen People's Hospital, Shenzhen, China.
  • Collisson EA; Texas Oncology-Baylor University Medical Center, Dallas, TX, USA.
  • Scadeng M; The Translational Genomics Research Institute, Scottsdale, AZ, USA.
  • Lowy AM; HonorHealth, Scottsdale, AZ, USA.
  • Donahue TR; The Translational Genomics Research Institute, Scottsdale, AZ, USA.
  • Reya T; HonorHealth, Scottsdale, AZ, USA.
  • Downes M; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
  • Evans RM; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
  • Wahl GM; Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
  • Pawson T; Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
  • Tian R; Hematology Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
  • Hunter T; Center for Functional MRI, Department of Radiology, University of California San Diego, La Jolla, CA, USA.
Nature ; 569(7754): 131-135, 2019 05.
Article en En | MEDLINE | ID: mdl-30996350
ABSTRACT
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis largely owing to inefficient diagnosis and tenacious drug resistance. Activation of pancreatic stellate cells (PSCs) and consequent development of dense stroma are prominent features accounting for this aggressive biology1,2. The reciprocal interplay between PSCs and pancreatic cancer cells (PCCs) not only enhances tumour progression and metastasis but also sustains their own activation, facilitating a vicious cycle to exacerbate tumorigenesis and drug resistance3-7. Furthermore, PSC activation occurs very early during PDAC tumorigenesis8-10, and activated PSCs comprise a substantial fraction of the tumour mass, providing a rich source of readily detectable factors. Therefore, we hypothesized that the communication between PSCs and PCCs could be an exploitable target to develop effective strategies for PDAC therapy and diagnosis. Here, starting with a systematic proteomic investigation of secreted disease mediators and underlying molecular mechanisms, we reveal that leukaemia inhibitory factor (LIF) is a key paracrine factor from activated PSCs acting on cancer cells. Both pharmacologic LIF blockade and genetic Lifr deletion markedly slow tumour progression and augment the efficacy of chemotherapy to prolong survival of PDAC mouse models, mainly by modulating cancer cell differentiation and epithelial-mesenchymal transition status. Moreover, in both mouse models and human PDAC, aberrant production of LIF in the pancreas is restricted to pathological conditions and correlates with PDAC pathogenesis, and changes in the levels of circulating LIF correlate well with tumour response to therapy. Collectively, these findings reveal a function of LIF in PDAC tumorigenesis, and suggest its translational potential as an attractive therapeutic target and circulating marker. Our studies underscore how a better understanding of cell-cell communication within the tumour microenvironment can suggest novel strategies for cancer therapy.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Neoplasias Pancreáticas / Comunicación Paracrina / Carcinoma Ductal Pancreático / Factor Inhibidor de Leucemia Tipo de estudio: Diagnostic_studies / Prognostic_studies Límite: Animals / Female / Humans / Male Idioma: En Año: 2019 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Neoplasias Pancreáticas / Comunicación Paracrina / Carcinoma Ductal Pancreático / Factor Inhibidor de Leucemia Tipo de estudio: Diagnostic_studies / Prognostic_studies Límite: Animals / Female / Humans / Male Idioma: En Año: 2019 Tipo del documento: Article