1. Academic Validation
  2. Hedgehog signalling pathway orchestrates angiogenesis in triple-negative breast cancers

Hedgehog signalling pathway orchestrates angiogenesis in triple-negative breast cancers

  • Br J Cancer. 2017 May 23;116(11):1425-1435. doi: 10.1038/bjc.2017.116.
Concetta Di Mauro 1 Roberta Rosa 1 Valentina D'Amato 1 Paola Ciciola 1 Alberto Servetto 1 Roberta Marciano 1 Roberta Clara Orsini 1 Luigi Formisano 1 Sandro De Falco 2 Valeria Cicatiello 2 Maurizio Di Bonito 3 Monica Cantile 3 Francesca Collina 3 Angela Chambery 4 Bianca Maria Veneziani 5 Sabino De Placido 1 Roberto Bianco 1
Affiliations

Affiliations

  • 1 Department of Clinical Medicine and Surgery, University of Naples 'Federico II', Naples, Italy.
  • 2 Angiogenesis Laboratory, Institute of Genetics and Biophysics, 'Adriano Buzzati-Traverso', Consiglio Nazionale delle Ricerche (CNR), Naples, Italy.
  • 3 Division of Diagnostic Pathology, Department of Diagnostic and Laboratory Pathology, 'Istituto Nazionale Tumori Fondazione Giovanni Pascale-IRCCS', Via Mariano Semmola, Naples, Italy.
  • 4 Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples, Naples, Italy.
  • 5 Department of Molecular Medicine and Medical Biotechnologies, University of Naples 'Federico II', Naples, Italy.
Abstract

Background: Several evidences suggest a marked angiogenic dependency in triple-negative breast Cancer (TNBC) tumorigenesis and a potential sensitivity to anti-angiogenic agents. Herein, the putative role of Hedgehog (Hh) pathway in regulating TNBC-dependent angiogenesis was investigated.

Methods: Expression and regulation of the Hh pathway transcription factor glioma-associated oncogene homolog1 protein (GLI1) were studied on the endothelial compartment and on TNBC-initiated angiogenesis. To evaluate the translational relevance of our findings, the combination of paclitaxel with the Smo Inhibitor NVP-LDE225 was tested in TNBC xenografted mice.

Results: Tissue microarray analysis on 200 TNBC patients showed GLI1 overexpression paired with vascular endothelial growth factor receptor 2 (VEGFR2/KDR/Flk-1) expression. In vitro, Hh pathway promotes TNBC progression in an autocrine manner, regulating the VEGF/VEGFR2/KDR/Flk-1 loop on Cancer cell surface, and in a paracrine manner, orchestrating tumour vascularisation. These effects were counteracted by Smo pharmacological inhibition. In TNBC xenografted mice, scheduling NVP-LDE225 rather than bevacizumab provided a better sustained inhibition of TNBC cells proliferation and endothelial cells organisation.

Conclusions: This study identifies the Hh pathway as one of the main regulators of tumour angiogenesis in TNBC, thus suggesting Hh inhibition as a potential new anti-angiogenic therapeutic option to be clinically investigated in GLI1 overexpressing TNBC patients.

Figures
Products