Effects of obesity on transcriptomic changes and cancer hallmarks in estrogen receptor–positive breast cancer

E Fuentes-Mattei, G Velazquez-Torres… - JNCI: Journal of the …, 2014 - academic.oup.com
E Fuentes-Mattei, G Velazquez-Torres, L Phan, F Zhang, PC Chou, JH Shin, HH Choi
JNCI: Journal of the National Cancer Institute, 2014academic.oup.com
Background Obesity increases the risk of cancer death among postmenopausal women with
estrogen receptor–positive (ER+) breast cancer, but the direct evidence for the mechanisms
is lacking. The purpose of this study is to demonstrate direct evidence for the mechanisms
mediating this epidemiologic phenomenon. Methods We analyzed transcriptomic profiles of
pretreatment biopsies from a prospective cohort of 137 ER+ breast cancer patients. We
generated transgenic (MMTV-TGFα; A y/a) and orthotopic/syngeneic (A y/a) obese mouse …
Background
Obesity increases the risk of cancer death among postmenopausal women with estrogen receptor–positive (ER+) breast cancer, but the direct evidence for the mechanisms is lacking. The purpose of this study is to demonstrate direct evidence for the mechanisms mediating this epidemiologic phenomenon.
Methods
We analyzed transcriptomic profiles of pretreatment biopsies from a prospective cohort of 137 ER+ breast cancer patients. We generated transgenic (MMTV- TGFα;Ay/a ) and orthotopic/syngeneic ( Ay/a ) obese mouse models to investigate the effect of obesity on tumorigenesis and tumor progression and to determine biological mechanisms using whole-genome transcriptome microarrays and protein analyses. We used a coculture system to examine the impact of adipocytes/adipokines on breast cancer cell proliferation. All statistical tests were two-sided.
Results
Functional transcriptomic analysis of patients revealed the association of obesity with 59 biological functional changes ( P < .05) linked to cancer hallmarks. Gene enrichment analysis revealed enrichment of AKT-target genes ( P = .04) and epithelial–mesenchymal transition genes ( P = .03) in patients. Our obese mouse models demonstrated activation of the AKT/mTOR pathway in obesity-accelerated mammary tumor growth (3.7- to 7.0-fold; P < .001; n = 6–7 mice per group). Metformin or everolimus can suppress obesity-induced secretion of adipokines and breast tumor formation and growth (0.5-fold, P = .04; 0.3-fold , P < .001, respectively; n = 6–8 mice per group). The coculture model revealed that adipocyte-secreted adipokines (eg, TIMP-1) regulate adipocyte-induced breast cancer cell proliferation and invasion. Metformin suppress adipocyte-induced cell proliferation and adipocyte-secreted adipokines in vitro.
Conclusions
Adipokine secretion and AKT/mTOR activation play important roles in obesity-accelerated breast cancer aggressiveness in addition to hyperinsulinemia, estrogen signaling, and inflammation. Metformin and everolimus have potential for therapeutic interventions of ER+ breast cancer patients with obesity.
Oxford University Press