Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6–dependent manner

AW Studebaker, G Storci, JL Werbeck, P Sansone… - Cancer research, 2008 - AACR
AW Studebaker, G Storci, JL Werbeck, P Sansone, AK Sasser, S Tavolari, T Huang…
Cancer research, 2008AACR
Common sites of breast cancer metastasis include the lung, liver, and bone, and of these
secondary metastatic sites, estrogen receptor α (ERα)–positive breast cancer often favors
bone. Within secondary organs, cancer cells would predictably encounter tissue-specific
fibroblasts or their soluble factors, yet our understanding of how tissue-specific fibroblasts
directly affect cancer cell growth rates and survival remains largely unknown. Therefore, we
tested the hypothesis that mesenchymal fibroblasts isolated from common sites of breast …
Abstract
Common sites of breast cancer metastasis include the lung, liver, and bone, and of these secondary metastatic sites, estrogen receptor α (ERα)–positive breast cancer often favors bone. Within secondary organs, cancer cells would predictably encounter tissue-specific fibroblasts or their soluble factors, yet our understanding of how tissue-specific fibroblasts directly affect cancer cell growth rates and survival remains largely unknown. Therefore, we tested the hypothesis that mesenchymal fibroblasts isolated from common sites of breast cancer metastasis provide a more favorable microenvironment with respect to tumor growth rates. We found a direct correlation between the ability of breast, lung, and bone fibroblasts to enhance ERα-positive breast cancer cell growth and the level of soluble interleukin-6 (IL-6) produced by each organ-specific fibroblast, and fibroblast-mediated growth enhancement was inhibited by the removal or inhibition of IL-6. Interestingly, mice coinjected with MCF-7 breast tumor cells and senescent skin fibroblasts, which secrete IL-6, developed tumors, whereas mice coinjected with presenescent skin fibroblasts that produce little to no IL-6 failed to form xenograft tumors. We subsequently determined that IL-6 promoted growth and invasion of breast cancer cells through signal transducer and activator of transcription 3–dependent up-regulation of Notch-3, Jagged-1, and carbonic anhydrase IX. These data suggest that tissue-specific fibroblasts and the factors they produce can promote breast cancer disease progression and may represent attractive targets for development of new therapeutics. [Cancer Res 2008;68(21):9087–95]
AACR