20
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Dual Inhibition of the Lactate Transporters MCT1 and MCT4 Is Synthetic Lethal with Metformin due to NAD+ Depletion in Cancer Cells

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          Highly glycolytic cancer cells prevent intracellular acidification by excreting the glycolytic end-products lactate and H + via the monocarboxylate transporters 1 (MCT1) and 4 (MCT4). We report that syrosingopine, an anti-hypertensive drug, is a dual MCT1 and MCT4 inhibitor (with 60-fold higher potency on MCT4) that prevents lactate and H + efflux. Syrosingopine elicits synthetic lethality with metformin, an inhibitor of mitochondrial NADH dehydrogenase. NAD+, required for the ATP-generating steps of glycolysis, is regenerated from NADH by mitochondrial NADH dehydrogenase or lactate dehydrogenase. Syrosingopine treatment leads to high intracellular lactate levels and thereby end-product inhibition of lactate dehydrogenase. The loss of NAD+ regeneration capacity due to combined metformin and syrosingopine treatment results in glycolytic blockade, leading to ATP depletion and cell death. Accordingly, ATP levels can be partly restored by exogenously provided NAD+, the NAD precursor nicotinamide mononucleotide (NMN), or vitamin K2. Thus, pharmacological inhibition of MCT1 and MCT4 combined with metformin treatment is a potential cancer therapy.

          Graphical Abstract

          Highlights

          • Syrosingopine inhibits lactate export by MCT1 and MCT4

          • Combined MCT1 and MCT4 inhibition elicits synthetic lethality with metformin

          • Synthetic lethality is due to loss of NAD+-regenerating capacity

          • Syrosingopine reduces metformin concentration required for anti-cancer activity

          Abstract

          Benjamin et al. show that the clinical drug syrosingopine potently inhibits the lactate transporters MCT1 and MCT4 and is thus a clinically relevant MCT4 inhibitor. Intracellular lactate accumulation by syrosingopine elicits synthetic lethality with metformin and potentiates metformin’s anti-cancer efficacy.

          Related collections

          Most cited references28

          • Record: found
          • Abstract: found
          • Article: not found

          LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells.

          Elevated lactate dehydrogenase A (LDHA) expression is associated with poor outcome in tumor patients. Here we show that LDHA-associated lactic acid accumulation in melanomas inhibits tumor surveillance by T and NK cells. In immunocompetent C57BL/6 mice, tumors with reduced lactic acid production (Ldha(low)) developed significantly slower than control tumors and showed increased infiltration with IFN-γ-producing T and NK cells. However, in Rag2(-/-)γc(-/-) mice, lacking lymphocytes and NK cells, and in Ifng(-/-) mice, Ldha(low) and control cells formed tumors at similar rates. Pathophysiological concentrations of lactic acid prevented upregulation of nuclear factor of activated T cells (NFAT) in T and NK cells, resulting in diminished IFN-γ production. Database analyses revealed negative correlations between LDHA expression and T cell activation markers in human melanoma patients. Our results demonstrate that lactic acid is a potent inhibitor of function and survival of T and NK cells leading to tumor immune escape.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Lactate Metabolism in Human Lung Tumors

            Cancer cells consume glucose and secrete lactate in culture. It is unknown whether lactate contributes to energy metabolism in living tumors. We previously reported that human non-small cell lung cancers (NSCLC) oxidize glucose in the tricarboxylic acid (TCA) cycle. Here we show that lactate is also a TCA cycle carbon source for NSCLC. In human NSCLC, evidence of lactate utilization was most apparent in tumors with high 18 fluorodeoxyglucose uptake and aggressive oncological behavior. Infusing human NSCLC patients with 13 C-lactate revealed extensive labeling of TCA cycle metabolites. In mice, deleting monocarboxylate transporter-1 (MCT1) from tumor cells eliminated lactate-dependent metabolite labeling, confirming tumor-cell autonomous lactate uptake. Strikingly, directly comparing lactate and glucose metabolism in vivo indicated that lactate's contribution to the TCA cycle predominates. The data indicate that tumors, including bona fide human NSCLC, can use lactate as a fuel in vivo. Human non-small cell lung cancer preferentially utilizes lactate over glucose to fuel TCA cycle and sustain tumor metabolism in vivo.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Targeting lactate metabolism for cancer therapeutics.

              Lactate, once considered a waste product of glycolysis, has emerged as a critical regulator of cancer development, maintenance, and metastasis. Indeed, tumor lactate levels correlate with increased metastasis, tumor recurrence, and poor outcome. Lactate mediates cancer cell intrinsic effects on metabolism and has additional non-tumor cell autonomous effects that drive tumorigenesis. Tumor cells can metabolize lactate as an energy source and shuttle lactate to neighboring cancer cells, adjacent stroma, and vascular endothelial cells, which induces metabolic reprogramming. Lactate also plays roles in promoting tumor inflammation and in functioning as a signaling molecule that stimulates tumor angiogenesis. Here we review the mechanisms of lactate production and transport and highlight emerging evidence indicating that targeting lactate metabolism is a promising approach for cancer therapeutics.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Rep
                Cell Rep
                Cell Reports
                Cell Press
                2211-1247
                11 December 2018
                11 December 2018
                11 December 2018
                : 25
                : 11
                : 3047-3058.e4
                Affiliations
                [1 ]Biozentrum, University of Basel, 4056 Basel, Switzerland
                [2 ]Basilea Pharmaceutica International Ltd. AG, Basel, Switzerland
                Author notes
                []Corresponding author heidi.lane@ 123456basilea.com
                [∗∗ ]Corresponding author m.hall@ 123456unibas.ch
                [3]

                These authors contributed equally

                [4]

                Lead Contact

                Article
                S2211-1247(18)31806-0
                10.1016/j.celrep.2018.11.043
                6302548
                30540938
                369ba9b1-1067-4daf-9118-6659eada7099
                © 2018 The Authors

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 22 June 2018
                : 17 October 2018
                : 9 November 2018
                Categories
                Article

                Cell biology
                cancer,metformin,syrosingopine,lactate,mct1,mct4,synthetic lethality
                Cell biology
                cancer, metformin, syrosingopine, lactate, mct1, mct4, synthetic lethality

                Comments

                Comment on this article