Phospho-ibuprofen (MDC-917) suppresses breast cancer growth: an effect controlled by the thioredoxin system

Breast Cancer Res. 2012 Jan 31;14(1):R20. doi: 10.1186/bcr3105.

Abstract

Introduction: We have recently synthesized phospho-ibuprofen (P-I; MDC-917), a safer derivative of ibuprofen, which has shown anti-cancer activity. We investigated its efficacy and mechanism of action in the treatment of breast cancer in preclinical models.

Methods: We evaluated the anti-breast-cancer efficacy of P-I alone or incorporated into liposomes (Lipo-P-I) in human estrogen receptor-positive (MCF-7) and triple-negative, i.e., estrogen receptor-negative, progesterone receptor-negative and HER2-negative (MDA-MB231) breast cancer cell lines - as they represent the most frequent (estrogen receptor-positive) and the most difficult-to-treat (triple-negative) subtypes of breast cancer - and their xenografts in nude mice. We assessed the effect of P-I on the levels of reactive oxygen nitrogen species in response to P-I using molecular probes, on the thioredoxin system (expression and redox status of thioredoxin-1 (Trx-1) and thioredoxin reductase activity), on cyclooxygenase 2, NF-κB and mitogen-activated protein kinase cell signaling; and on the growth of xenografts with stably knocked-down Trx-1.

Results: Compared with controls, P-I 400 mg/kg/day inhibited the growth of MDA-MB231 xenografts by 266%, while the growth of MCF-7 xenografts was inhibited 51% byP-I 300 mg/kg/day and 181% by Lipo-P-I 300 mg/kg/day. In both cell lines, P-I induced oxidative stress and suppressed the thioredoxin system (oxidized Trx-1 and decreased its expression; inhibited thioredoxin reductase activity). These changes triggered downstream redox signaling: the activity of NF-κB was suppressed and the Trx-1-ASK1 complex was dissociated, activating the p38 and JNK mitogen-activated protein kinase cascades. Trx-1 knockdown abrogated the anti-cancer effect of P-I in vitro and in vivo.

Conclusion: P-I is safe and effective against breast cancer. Liposomal formulation enhances its efficacy; the effect is heavily dependent on the induction of oxidative stress and the suppression of the thioredoxin system. P-I merits further evaluation as an agent for the treatment of breast cancer.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Antineoplastic Agents / administration & dosage
  • Antineoplastic Agents / pharmacology*
  • Apoptosis / drug effects
  • Breast Neoplasms / drug therapy*
  • Cell Proliferation / drug effects
  • Cell Survival / drug effects
  • Cyclooxygenase 2 / metabolism
  • Female
  • Glutathione / metabolism
  • Humans
  • Ibuprofen / administration & dosage
  • Ibuprofen / analogs & derivatives*
  • Ibuprofen / pharmacology
  • Liposomes
  • MAP Kinase Kinase Kinase 5 / metabolism
  • MCF-7 Cells
  • Mice
  • Mice, Inbred BALB C
  • Mice, Nude
  • Organophosphates / administration & dosage
  • Organophosphates / pharmacology*
  • Oxidation-Reduction
  • Protein Binding
  • Reactive Nitrogen Species / metabolism
  • Reactive Oxygen Species / metabolism
  • Signal Transduction
  • Thioredoxin-Disulfide Reductase / metabolism
  • Thioredoxins / genetics
  • Thioredoxins / metabolism*
  • Xenograft Model Antitumor Assays

Substances

  • 2-(4-isobutylphenyl)propionic acid 4-(diethoxyphosphoryloxy)butyl ester
  • Antineoplastic Agents
  • Liposomes
  • Organophosphates
  • Reactive Nitrogen Species
  • Reactive Oxygen Species
  • Thioredoxins
  • Ptgs2 protein, mouse
  • Cyclooxygenase 2
  • Thioredoxin-Disulfide Reductase
  • MAP Kinase Kinase Kinase 5
  • MAP3K5 protein, human
  • Glutathione
  • Ibuprofen