Differential Metabolism of Glucose and Acetate in Mitochondria of Early Stage Breast Cancer In Vivo
Elizabeth Maher1, Kumar Pichumani2, Venetia Sarode3, Tomoyuki Mashimo1, Manoj Cheriyan1, Vamsidhara Vemireddy1, Barbara Haley1, Dean Sherry2, Roshni Rao4, Craig Malloy2, and Robert Bachoo5

1Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States, 2Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, United States, 3Pathology, UT Southwestern Medical Center, Dallas, TX, United States, 4Surgery, UT Southwestern Medical Center, Dallas, TX, United States, 5Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, United States

Synopsis

Metabolic reprogramming of bioenergetic substrate utilization was shown in primary and metastatic brain tumors. Whether the use of substrates other than glucose to fuel the citric acid cycle is a property of cancer cell growth in the brain or a fundamental property of a transformed cell is not known. To address this question we studied early stage breast cancer patients using infusion of 13C-glucose or 13C-acetate during initial surgery. 13C-NMR spectra of resected tumors show that acetate but not glucose is oxidized in the citric acid cycle, suggesting that acetate may contribute to energy production in these early stage cancers.

PURPOSE

Metabolic reprogramming is a hallmark of cancer. After infusion of 13C-enriched acetate or glucose, 13C-NMR analysis of biopsies from human primary or metastatic brain tumors has demonstrated oxidation of both substrates in the citric acid cycle [1, 2]. It is not known whether this capacity reflects growth in the microenvironment of the brain or a fundamental feature of cellular transformation. To investigate whether the capacity to oxidize both glucose and acetate in the citric acid cycle is a feature of an early cancer, we studied patients with early stage, non-metastatic breast cancer.

METHODS

After obtaining informed consent, eligible patients were enrolled in an Institutional Review Board (IRB) approved clinical protocol. All patients had standard histological analysis of the diagnostic biopsy prior to enrollment. Patients were infused intravenously with either [U-13C]glucose or [1,2-13C]acetate during tumor resection for 120 – 180 minutes prior to tissue sampling. Tumors were sampled by multiple biopsies or immediately after mastectomy to obtain 200-400 mg of tumor. After freeze-clamping and later aqueous extraction, 1H-decoupled 13C-NMR spectra of the tumor extracts were acquired on an Agilent 14.1T system equipped with a cryogenically – cooled probe. Fractional enrichment in plasma glucose was measured by mass spectrometry. Fractional enrichment in plasma acetate was measured by 1H NMR spectroscopy. Results are mean ± SEM.

RESULTS

Eight patients were infused with 13C-glucose, achieving mean 13C-glucose enrichment in the blood of 56±4%. Figure 1A shows the 13C-NMR spectrum from a good prognosis, estrogen (ER) and progesterone receptor (ER) positive, HER2 negative tumor that has a low proliferation rate (Ki67 10%). The 13C-NMR spectra from the most aggressive and highly proliferative (Ki67 70-90%) triple negative breast cancers (ER-, PR- and HER2-) and HER2 positive cancers were remarkably similar (spectra not shown). Each spectrum showed dominant labeling in lactate with minimal to no labeling of glutamate and glutamine from infused 13C-glucose. In marked contrast, infusion of 13C-acetate led to significant labeling in glutamate and glutamine, with the generation of quartets reflecting multiple turns of the citric acid cycle (representative spectrum in Figure 1B from an ER+, PR+, HER2- tumor, Ki67 12%). This pattern was seen in all patients infused with 13C-acetate, including those with triple negative and HER2+ tumors. Among patients infused with [U-13C]glucose, the fraction of acetyl-CoA derived from glucose was < 5% except in two patients with HER2 + disease (17% and 10%). Among patients infused with [1,2-13C]acetate (n=8), the fraction of acetyl-CoA derived from acetate carbons was 48±4% (p < 0.001). No differences in acetate oxidation were detected among prognostic subgroups that were based on hormone receptor status. Interestingly, among patients infused with acetate, 13C-enriched beta-hydroxybutyrate (BHB) was also observed. The fractional 13C enrichment in BHB in plasma was 18±4%.

DISCUSSION

Under these conditions infused 13C-enriched glucose contributed little to acetyl-CoA in most early stage breast cancers, but HER2+ disease was associated with a modest increase in glucose oxidation. However, 13C from infused 13C-enriched acetate contributed nearly 50% of acetyl-CoA, demonstrating an intact citric acid cycle in early stage breast cancer, regardless of prognostic subgroup or proliferative rate. The appearance of 13C-enriched BHB in both plasma and tumor is due to hepatic ketogenesis from the infused 13C-enriched acetate. Together, these data demonstrate that acetate and possibly ketones, but not glucose, may contribute significantly to energy production via the citric acid cycle in early stage breast cancer.

Acknowledgements

The authors acknowledge funding support from NIH, Cancer Prevention Research Institute of Texas (CPRIT) and the Simmons Comprehensive Cancer Center at UT Southwestern Medical Center.

References

1. Maher, E.A., Marin-Valencia, I., Bachoo, R., et al., Metabolism of [U-13 C]glucose in human brain tumors in vivo. NMR Biomed, 2012. 25(11): p. 1234-44.

2. Mashimo, T., Pichumani, K., Vemireddy, V., et al., Acetate is a bioenergetic substrate for human glioblastoma and brain metastases. Cell, 2014. 159(7): p. 1603-14.

Figures

Differential oxidation of glucose and acetate. 13C-NMR spectra following infusion of [U-13C]glucose, A, or [1,2-13C]acetate, B. Insets - GLU4, glutamate C4; GLN4, glutamine C4. Abbreviations: GLU3, glutamate C3; GLN3, glutamine C3; ACE2, acetate C2; BHB4, beta hydroxybutyrate C4; LAC3, lactate C3; ALA3, alanine C3. D45, doublet 4,5; S, singlet; Q, quartet



Proc. Intl. Soc. Mag. Reson. Med. 24 (2016)
0841