1. Academic Validation
  2. Targeting Glutamine Metabolism in Breast Cancer with Aminooxyacetate

Targeting Glutamine Metabolism in Breast Cancer with Aminooxyacetate

  • Clin Cancer Res. 2015 Jul 15;21(14):3263-73. doi: 10.1158/1078-0432.CCR-14-1200.
Preethi Korangath 1 Wei Wen Teo 1 Helen Sadik 1 Liangfeng Han 1 Noriko Mori 2 Charlotte M Huijts 1 Flonne Wildes 2 Santosh Bharti 2 Zhe Zhang 1 Cesar A Santa-Maria 1 Hualing Tsai 1 Chi V Dang 3 Vered Stearns 1 Zaver M Bhujwalla 2 Saraswati Sukumar 4
Affiliations

Affiliations

  • 1 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • 2 Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • 3 Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.
  • 4 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. [email protected].
Abstract

Purpose: Glutamine addiction in c-MYC-overexpressing breast Cancer is targeted by the aminotransferase inhibitor, aminooxyacetate (AOA). However, the mechanism of ensuing cell death remains unresolved.

Experimental design: A correlation between glutamine dependence for growth and c-Myc expression was studied in breast Cancer cell lines. The cytotoxic effects of AOA, its correlation with high c-Myc expression, and effects on enzymes in the glutaminolytic pathway were investigated. AOA-induced cell death was assessed by measuring changes in metabolite levels by magnetic resonance spectroscopy (MRS), the effects of amino acid depletion on nucleotide synthesis by cell-cycle and bromodeoxyuridine (BrdUrd) uptake analysis, and activation of the endoplasmic reticulum (ER) stress-mediated pathway. Antitumor effects of AOA with or without common chemotherapies were determined in breast Cancer xenografts in immunodeficient mice and in a transgenic MMTV-rTtA-TetO-myc mouse mammary tumor model.

Results: We established a direct correlation between c-Myc overexpression, suppression of glutaminolysis, and AOA sensitivity in most breast Cancer cells. MRS, cell-cycle analysis, and BrdUrd uptake measurements indicated depletion of aspartic acid and alanine leading to cell-cycle arrest at S-phase by AOA. Activation of components of the ER stress-mediated pathway, initiated through GRP78, led to apoptotic cell death. AOA inhibited growth of SUM159, SUM149, and MCF-7 xenografts and c-myc-overexpressing transgenic mouse mammary tumors. In MDA-MB-231, AOA was effective only in combination with chemotherapy.

Conclusions: AOA mediates its cytotoxic effects largely through the stress response pathway. The preclinical data of AOA's effectiveness provide a strong rationale for further clinical development, particularly for c-MYC-overexpressing breast cancers.

Figures
Products