Back to Search Start Over

Targeting CD47 in Anaplastic Thyroid Carcinoma Enhances Tumor Phagocytosis by Macrophages and Is a Promising Therapeutic Strategy

Authors :
Roch-Philippe Charles
Marc-David Ruepp
Matthias A. Roelli
Stefan Forster
Philippe Krebs
Renaud Maire
Frido Brühl
Matthias S. Dettmer
Sergio Di Pancrazio
Roland Giger
Marie-Hélène Wasmer
Aurel Perren
Anja Schmitt
Christian M. Schürch
Source :
Schürch, C M, Roelli, M A, Forster, S, Wasmer, M-H, Brühl, F, Maire, R, Di Pancrazio, S, Ruepp, M-D, Giger, R, Perren, A, Schmitt, A, Krebs, P, Charles, R-P & Dettmer, M S 2019, ' Targeting CD47 in anaplastic thyroid carcinoma enhances tumor phagocytosis by macrophages and is a promising therapeutic strategy ', Thyroid, vol. 29, no. 7, pp. 979-992 . https://doi.org/10.1089/thy.2018.0555, Schürch, Christian M; Rölli, Matthias A.; Forster, Stefan; Wasmer, Marie-Hélène; Brühl, Frido; Maire, Renaud S.; Di Pancrazio, Sergio; Ruepp, Marc-David; Giger, Roland; Perren, Aurel; Schmitt, Anja M.; Krebs, Philippe; Charles, Roch-Philippe; Dettmer, Matthias S. (2019). Targeting CD47 in Anaplastic Thyroid Carcinoma Enhances Tumor Phagocytosis by Macrophages and Is a Promising Therapeutic Strategy. Thyroid, 29(7), pp. 979-992. Mary Ann Liebert 10.1089/thy.2018.0555 , Thyroid
Publication Year :
2019
Publisher :
Mary Ann Liebert Inc, 2019.

Abstract

Background: Anaplastic thyroid carcinoma (ATC) is one of the most aggressive human cancers, with a median survival of only three to six months. Standard treatment options and even targeted therapies have so far failed to improve long-term overall survival. Thus, novel treatment modalities for ATC, such as immunotherapy, are urgently needed. CD47 is a "don't eat me" signal, which prevents cancer cells from phagocytosis by binding to signal regulatory protein alpha on macrophages. So far, the role of macrophages and the CD47-signal regulatory protein alpha signaling axis in ATC is not well understood. Methods: This study analyzed 19 primary human ATCs for macrophage markers, CD47 expression, and immune checkpoints by immunohistochemistry. ATC cell lines and a fresh ATC sample were assessed by flow cytometry for CD47 expression and macrophage infiltration, respectively. CD47 was blocked in phagocytosis assays of co-cultured macrophages and ATC cell lines. Anti-CD47 antibody treatment was administered to ATC cell line xenotransplanted immunocompromised mice, as well as to tamoxifen-induced ATC double-transgenic mice. Results: Human ATC samples were heavily infiltrated by CD68- and CD163-expressing tumor-associated macrophages (TAMs), and expressed CD47 and calreticulin, the dominant pro-phagocytic molecule. In addition, ATC tissues expressed the immune checkpoint molecules programmed cell death 1 and programmed death ligand 1. Blocking CD47 promoted the phagocytosis of ATC cell lines by macrophages in vitro. Anti-CD47 antibody treatment of ATC xenotransplanted mice increased the frequency of TAMs, enhanced the expression of macrophage activation markers, augmented tumor cell phagocytosis, and suppressed tumor growth. In double-transgenic ATC mice, CD47 was expressed on tumor cells, and blocking CD47 increased TAM frequencies. Conclusions: Targeting CD47 or CD47 in combination with programmed cell death 1 may potentially improve the outcomes of ATC patients and may represent a valuable addition to the current standard of care.

Details

ISSN :
15579077 and 10507256
Volume :
29
Database :
OpenAIRE
Journal :
Thyroid
Accession number :
edsair.doi.dedup.....ca805573fdaeb1819e23f6886bd6eb3b