Cancers (Mar 2022)

Functional Antagonism of Junctional Adhesion Molecule-A (JAM-A), Overexpressed in Breast Ductal Carcinoma In Situ (DCIS), Reduces HER2-Positive Tumor Progression

  • Yvonne E. Smith,
  • Guannan Wang,
  • Ciara L. Flynn,
  • Stephen F. Madden,
  • Owen MacEneaney,
  • Rodrigo G. B. Cruz,
  • Cathy E. Richards,
  • Hanne Jahns,
  • Marian Brennan,
  • Mattia Cremona,
  • Bryan T. Hennessy,
  • Katherine Sheehan,
  • Alexander Casucci,
  • Faizah A. Sani,
  • Lance Hudson,
  • Joanna Fay,
  • Sri H. Vellanki,
  • Siobhan O’Flaherty,
  • Marc Devocelle,
  • Arnold D. K. Hill,
  • Kieran Brennan,
  • Saraswati Sukumar,
  • Ann M. Hopkins

DOI
https://doi.org/10.3390/cancers14051303
Journal volume & issue
Vol. 14, no. 5
p. 1303

Abstract

Read online

Breast ductal carcinoma in situ (DCIS) is clinically challenging, featuring high diagnosis rates and few targeted therapies. Expression/signaling from junctional adhesion molecule-A (JAM-A) has been linked to poor prognosis in invasive breast cancers, but its role in DCIS is unknown. Since progression from DCIS to invasive cancer has been linked with overexpression of the human epidermal growth factor receptor-2 (HER2), and JAM-A regulates HER2 expression, we evaluated JAM-A as a therapeutic target in DCIS. JAM-A expression was immunohistochemically assessed in patient DCIS tissues. A novel JAM-A antagonist (JBS2) was designed and tested alone/in combination with the HER2 kinase inhibitor lapatinib, using SUM-225 cells in vitro and in vivo as validated DCIS models. Murine tumors were proteomically analyzed. JAM-A expression was moderate/high in 96% of DCIS patient tissues, versus 23% of normal adjacent tissues. JBS2 bound to recombinant JAM-A, inhibiting cell viability in SUM-225 cells and a primary DCIS culture in vitro and in a chick embryo xenograft model. JBS2 reduced tumor progression in in vivo models of SUM-225 cells engrafted into mammary fat pads or directly injected into the mammary ducts of NOD-SCID mice. Preliminary proteomic analysis revealed alterations in angiogenic and apoptotic pathways. High JAM-A expression in aggressive DCIS lesions and their sensitivity to treatment by a novel JAM-A antagonist support the viability of testing JAM-A as a novel therapeutic target in DCIS.

Keywords