Abstract

Cancer cells expressing constitutively active phosphatidylinositol-3 kinase (PI3K) are proliferative regardless of the absence of insulin, and they form dietary restriction (DR)-resistant tumors in vivo. Because the binding of insulin to its receptors activates the PI3K/AKT/mammalian target of rapamycin (mTOR) signaling cascade, activating mutations in the PIK3CA oncogene may determine tumor response to DR-like pharmacological strategies targeting the insulin and mTOR pathways. The anti-diabetic drug metformin is a stereotypical DR mimetic that exerts its anti-cancer activity through a dual mechanism involving insulin-related (systemic) and mTOR-related (cell-autonomous) effects. However, it remains unclear whether PIK3CA-activating mutations might preclude the anti-cancer activity of metformin in vivo. To model the oncogenic PIK3CA-driven early stages of cancer, we used the clonal breast cancer cell line MCF10DCIS.com, which harbors the gain-of-function H1047R hot-spot mutation in the catalytic domain of the PI3KCA gene and has been shown to form DR-refractory xenotumors. To model PIK3CA-activating mutations in late stages of cancer, we took advantage of the isogenic conversion of a PIK3CA-wild-type tumor into a PIK3CA H1047R-mutated tumor using the highly metastatic colorectal cancer cell line SW48. MCF10DCIS.com xenotumors, although only modestly affected by treatment with oral metformin (approximately 40% tumor growth inhibition), were highly sensitive to the intraperitoneal (i.p.) administration of metformin, the anti-cancer activity of which increased in a time-dependent manner and reached >80% tumor growth inhibition by the end of the treatment. Metformin treatment via the i.p. route significantly reduced the proliferation factor mitotic activity index (MAI) and decreased tumor cellularity in MCF10DCIS.com cancer tissues. Whereas SW48-wild-type (PIK3CA+/+) cells rapidly formed metformin-refractory xenotumors in mice, ad libitum access to water containing metformin significantly reduced the growth of SW48-mutated (PIK3CAH1047R/+) xenotumors by approximately 50%. Thus, metformin can no longer be considered as a bona fide DR mimetic, at least in terms of anti-cancer activity, because tumors harboring the insulin-unresponsive, DR-resistant, PIK3CA-activating mutation H1047R remain sensitive to the anti-tumoral effects of the drug. Given the high prevalence of PIK3CA mutations in human carcinomas and the emerging role of PIK3CA mutation status in the treatment selection process, these findings might have a significant impact on the design of future trials evaluating the potential of combining metformin with targeted therapy.

Highlights

  • The most hotly debated and yet unresolved issue in the area of employing calorie restriction (CR) mimetics (CRMs) as anti-cancer agents with mammalian target of rapamycin-inhibiting activity is the relative effects of nature versus nurture, i.e., the relative contributions of systemic factors versus cancer cellautonomous effects [1,2,3]

  • The notion that metformin suppresses cancer growth through pathways other than insulin/IGF-1-dependent, indirect drug action has recently been supported by experiments in liver IGF-1-deficient (LID) mice, which had naturally decreased lung tumor multiplicity and burden compared with wild-type mice [28, 29]

  • To model the oncogenic PIK3CA-driven early stages of human cancer, we employed the clonal breast cancer cell line MCF10DCIS.com, which was derived from a xenograft originating from premalignant MCF10AT cells that were injected into SCID mice

Read more

Summary

INTRODUCTION

The most hotly debated and yet unresolved issue in the area of employing calorie restriction (CR) mimetics (CRMs) as anti-cancer agents with mammalian target of rapamycin (mTOR)-inhibiting activity is the relative effects of nature versus nurture, i.e., the relative contributions of systemic factors versus cancer cellautonomous effects [1,2,3]. The notion that metformin suppresses cancer growth through pathways other than insulin/IGF-1-dependent, indirect drug action has recently been supported by experiments in liver IGF-1-deficient (LID) mice, which had naturally decreased lung tumor multiplicity and burden compared with wild-type mice [28, 29]. The authors demonstrated that metformin can act through IGF1-independent mechanisms by promoting the systemic inhibition of circulating growth factors and local receptor tyrosine kinase signaling in cancer tissues [28] These findings suggest an insulin-independent, direct anti-tumoral activity for metformin, it remains unknown whether cancer cell-autonomous alterations that are known to drive insulin/IGF-1-independent, DR-resistant tumors www.impactjournals.com/oncotarget. Given the high number of PIK3CA mutations in human cancer, this finding could have a significant impact on the design of metformin-based therapies that aim to influence both the early stages of tumor formation and progression and cancer recurrence in advanced tumors

RESULTS
DISCUSSION
METHODS
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call