Isotope tracing reveals glycolysis and oxidative metabolism in childhood tumors of multiple histologies

Kendra Johnston, Panayotis Pachnis, Alpaslan Tasdogan, Brandon Faubert, Lauren G Zacharias, Hieu Sy Vu, Laurie Rodgers-Augustyniak, Allison Johnson, Fang Huang, Sean Ricciardo, Zhiyu Zhao, Thomas P Mathews, Tanya Watt, Patrick Leavey, Ralph J DeBerardinis, Kendra Johnston, Panayotis Pachnis, Alpaslan Tasdogan, Brandon Faubert, Lauren G Zacharias, Hieu Sy Vu, Laurie Rodgers-Augustyniak, Allison Johnson, Fang Huang, Sean Ricciardo, Zhiyu Zhao, Thomas P Mathews, Tanya Watt, Patrick Leavey, Ralph J DeBerardinis

Abstract

Background: Survival among children with high-risk solid tumors remains poor. Reprogrammed metabolism promotes tumor growth and may contain therapeutic liabilities. Tumor metabolism has been assessed in adults using intra-operative 13C-glucose infusions. Pediatric tumors differ from adult cancers in their low mutational burden and derivation from embryonic tissues. Here we used 13C infusions to examine tumor metabolism in children, comparing phenotypes among tumor types and between childhood and adult cancers.

Methods: Patients recruited to study NCT03686566 received an intra-operative infusion of [U-13C]glucose during tumor resection to evaluate central carbon pathways in the tumor, with concurrent metabolomics to provide a broad overview of metabolism. Differential characteristics were determined using multiple comparison tests and mixed effect analyses.

Findings: We studied 23 tumors from 22 patients. All tumors analyzed by [U-13C]glucose contained labeling in glycolytic and tricarboxylic acid (TCA) cycle intermediates. Labeling in the TCA cycle indicated activity of pyruvate dehydrogenase (PDH) and pyruvate carboxylase (PC), with PDH predominating. Neuroblastomas had high lactate labeling relative to other childhood cancers and lung cancer, and were distinguished by abundant tyrosine catabolites consistent with catecholamine synthesis.

Conclusions: Intra-operative [U13C]glucose infusions are safe and informative in pediatric cancer. Tumors of various histologies use glycolysis and oxidative metabolism, with subtype-selective differences evident from this small cohort. Expanding this cohort may uncover predictive biomarkers and therapeutic targets from tumor metabolism.

Funding: N.C.I grants to P.L. (R21CA220090-01A1) and R.J.D. (R35CA22044901); H.H.M.I. funding to R.J.D.; Children's Clinical Research Advisory Committee funding to K.J.

Keywords: Cancer; glucose; isotopes; metabolism; metabolomics; neuroblastoma; sarcoma.

Figures

Figure 1:. Method for intra-operative [U- 13…
Figure 1:. Method for intra-operative [U-13C]glucose infusions in children.
(A) Two weight-based regimens for infusion of [U-13C]glucose. (B) Plasma glucose enrichment levels for pediatric patients on Regimen 1 or Regimen 2.
Figure 2:. 13 C labeling in tumor…
Figure 2:. 13C labeling in tumor metabolites from two children with neuroblastoma.
(A) Expected labeling of glycolytic and TCA cycle metabolites after infusion with [U-13C]glucose. In this scheme, the two labeled carbons from acetyl-CoA are transferred to citrate positions 1 and 2 and then to positions 4 and 5 of α-KG and carried further around the TCA cycle. Labeled OAA exchanges with labeled aspartate. Further metabolism of labeled OAA into subsequent turns of cycle is not illustrated. (B) Computed tomography of Patient #1, a 9-year old boy with a non-MYCN-amplified neuroblastoma (red box). (C) Time-dependent labeling of glucose, lactate and pyruvate in Patient #1’s plasma during the [U-13C]glucose infusion. (D) Labeling in metabolites extracted from Patient #1’s tumor. Labeling is normalized to the glucose m+6 fraction in plasma at the time of tumor resection. (E) Computed tomography of Patient #3, a 2-year old girl with a non-MYCN-amplified neuroblastoma (red box). (F) Time-dependent labeling of glucose, lactate and pyruvate in Patient #3’s plasma during the [U-13C]glucose infusion. (G) Labeling in metabolites extracted from Patient #3’s tumor. Labeling is normalized to the glucose m+6 fraction in plasma at the time of tumor resection.
Figure 3:. Metabolite labeling reveals evidence of…
Figure 3:. Metabolite labeling reveals evidence of metabolic differences between neuroblastoma and other tumor types.
(A) Fractional enrichment of metabolites extracted from all pediatric tumors in the cohort. Labeling is normalized to the glucose m+6 fraction in plasma at the time of tumor resection. The bar indicates that in neuroblastomas, relative labeling in lactate is higher than relative labeling in 3PG. (B) Fractional enrichment of metabolites extracted from all pediatric tumors in the cohort. These data are the same as in panel (A), but sarcomas are considered as a separate group. The bar indicates the difference in relative lactate labeling between neuroblastoma and other non-sarcoma tumors. (C) Fractional enrichment in tumor lactate, normalized to plasma glucose m+6, in pediatric tumors, adult NSCLC and adult ccRCC. NSCLC and ccRCC data include previously-reported patients , and are displayed here for the purposes of comparing to the new data from pediatric cancers. (D) Lactate m+3/3PG m+3 labeling ratios in the tumors displayed in (C). (E) Summary of labeling surrogates for PC activity (citrate m+3/pyruvate m+3 ratio) and PDH activity (citrate m+2/pyruvate m+3 ratio) for all pediatric tumors subjected to [U-13C]glucose infusion. (F) Comparison of PC and PDH labeling surrogates in different pediatric tumor types. Abbreviations: Low grade fibromyxoid sarcoma (LGF), osteosarcoma (OS), clear cell sarcoma of the kidney (CCS), rhabdomyosarcoma (RMS), synovial sarcoma (SS).
Figure 4:. Metabolomic features differentiate neuroblastomas from…
Figure 4:. Metabolomic features differentiate neuroblastomas from other pediatric tumors.
(A) Partial least squares discriminant analysis (PLS-DA) on metabolomics data from neuroblastomas and other pediatric tumors. (B) Volcano plot demonstrating metabolomic differences between neuroblastomas and other pediatric tumors. Several catecholamine-related metabolites accumulated in the neuroblastomas are indicated in red. (C) Metabolism of tyrosine to fumarate and acetoacetate or into catecholamine synthesis. (D) Volcano plot demonstrating gene expression differences between neuroblastomas and other tumors. Tumors used in this analysis include those shown in (E). Genes encoding several enzymes of catecholamine synthesis are indicated in red. (E) Expression of genes involved in tyrosine catabolism and catecholamine synthesis in neuroblastomas and several other forms of cancer. ****p

References

    1. Society, A.C. (2020). Cancer Facts & Figures 2020. .
    1. Matthay KK, Maris JM, Schleiermacher G, Nakagawara A, Mackall CL, Diller L, and Weiss WA (2016). Neuroblastoma . Nat Rev Dis Primers 2, 16078.
    1. Maris JM (2010). Recent advances in neuroblastoma. The New England journal of medicine 362, 2202–2211.
    1. Park JR, and Bagatell R (2016). Chapter 24 - Neuroblastoma. In Lanzkowsky’s Manual of Pediatric Hematology and Oncology (Sixth Edition), Lanzkowsky P, Lipton JM, and Fish JD, eds. (Academic Press; ), pp. 473–490.
    1. Pinto NR, Applebaum MA, Volchenboum SL, Matthay KK, London WB, Ambros PF, Nakagawara A, Berthold F, Schleiermacher G, Park JR, et al. (2015). Advances in Risk Classification and Treatment Strategies for Neuroblastoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 33, 3008–17.
    1. Sangkhathat S (2015). Current management of pediatric soft tissue sarcomas. World J Clin Pediatr 4, 94–105.
    1. Smith MA, Altekruse SF, Adamson PC, Reaman GH, and Seibel NL (2014). Declining childhood and adolescent cancer mortality. Cancer 120, 2497–2506.
    1. Hanahan D, and Weinberg RA (2011). Hallmarks of cancer: the next generation. Cell 144, 646–74.
    1. DeBerardinis RJ, and Chandel NS (2016). Fundamentals of cancer metabolism. Sci Adv 2, e1600200.
    1. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, et al. (2009). IDH1 and IDH2 mutations in gliomas. The New England journal of medicine 360, 765–73.
    1. Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, et al. (2010). Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 18, 553–67.
    1. Golub D, Iyengar N, Dogra S, Wong T, Bready D, Tang K, Modrek AS, and Placantonakis DG (2019). Mutant Isocitrate Dehydrogenase Inhibitors as Targeted Cancer Therapeutics. Front Oncol 9, 417–417.
    1. Popovici-Muller J, Lemieux RM, Artin E, Saunders JO, Salituro FG, Travins J, Cianchetta G, Cai Z, Zhou D, Cui D, et al. (2018). Discovery of AG-120 (Ivosidenib): A First-in-Class Mutant IDH1 Inhibitor for the Treatment of IDH1 Mutant Cancers. ACS medicinal chemistry letters 9, 300–305.
    1. Clavell LA, Gelber RD, Cohen HJ, Hitchcock-Bryan S, Cassady JR, Tarbell NJ, Blattner SR, Tantravahi R, Leavitt P, and Sallan SE (1986). Four-agent induction and intensive asparaginase therapy for treatment of childhood acute lymphoblastic leukemia. The New England journal of medicine 315, 657–63.
    1. Egler RA, Ahuja SP, and Matloub Y (2016). L-asparaginase in the treatment of patients with acute lymphoblastic leukemia. J Pharmacol Pharmacother 7, 62–71.
    1. Kim J, and DeBerardinis RJ (2019). Mechanisms and Implications of Metabolic Heterogeneity in Cancer. Cell Metab 30, 434–446.
    1. Yuneva MO, Fan TW, Allen TD, Higashi RM, Ferraris DV, Tsukamoto T, Mates JM, Alonso FJ, Wang C, Seo Y, et al. (2012). The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type. Cell Metab 15, 157–70.
    1. Faubert B, Solmonson A, and DeBerardinis RJ (2020). Metabolic reprogramming and cancer progression. Science 368, eaaw5473.
    1. Hu J, Locasale JW, Bielas JH, O’Sullivan J, Sheahan K, Cantley LC, Vander Heiden MG, and Vitkup D (2013). Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat Biotechnol 31, 522–9.
    1. Pugh TJ, Morozova O, Attiyeh EF, Asgharzadeh S, Wei JS, Auclair D, Carter SL, Cibulskis K, Hanna M, Kiezun A, et al. (2013). The genetic landscape of high-risk neuroblastoma. Nat Genet 45, 279–84.
    1. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, Schrock A, Campbell B, Shlien A, Chmielecki J, et al. (2017). Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 9, 34.
    1. Federico S, Brennan R, and Dyer MA (2011). Childhood cancer and developmental biology a crucial partnership. Curr Top Dev Biol 94, 1–13.
    1. Cheung NK, and Dyer MA (2013). Neuroblastoma: developmental biology, cancer genomics and immunotherapy. Nat Rev Cancer 13, 397–411.
    1. Faubert B, Li KY, Cai L, Hensley CT, Kim J, Zacharias LG, Yang C, Do QN, Doucette S, Burguete D, et al. (2017). Lactate Metabolism in Human Lung Tumors. Cell 171, 358–371.e9.
    1. Fan TWM, Lane AN, Higashi RM, Farag MA, Gao H, Bousamra M, and Miller DM (2009). Altered regulation of metabolic pathways in human lung cancer discerned by (13)C stable isotope-resolved metabolomics (SIRM). Mol Cancer 8, 41–41.
    1. Sellers K, Fox MP, Bousamra M II, Slone SP, Higashi RM, Miller DM, Wang Y, Yan J, Yuneva MO, Deshpande R, et al. (February 2). Pyruvate carboxylase is critical for non-small-cell lung cancer proliferation. The Journal of Clinical Investigation 125, 687–698.
    1. Hensley CT, Faubert B, Yuan Q, Lev-Cohain N, Jin E, Kim J, Jiang L, Ko B, Skelton R, Loudat L, et al. (2016). Metabolic Heterogeneity in Human Lung Tumors. Cell 164, 681–694.
    1. Courtney KD, Bezwada D, Mashimo T, Pichumani K, Vemireddy V, Funk AM, Wimberly J, McNeil SS, Kapur P, Lotan Y, et al. (2018). Isotope Tracing of Human Clear Cell Renal Cell Carcinomas Demonstrates Suppressed Glucose Oxidation In Vivo. Cell metabolism 28, 793–800.e2.
    1. Maher EA, Marin-Valencia I, Bachoo RM, Mashimo T, Raisanen J, Hatanpaa KJ, Jindal A, Jeffrey FM, Choi C, Madden C, et al. (2012). Metabolism of [U-13 C]glucose in human brain tumors in vivo. NMR Biomed 25, 1234–44.
    1. Tasdogan A, Faubert B, Ramesh V, Ubellacker JM, Shen B, Solmonson A, Murphy MM, Gu Z, Gu W, Martin M, et al. (2020). Metabolic heterogeneity confers differences in melanoma metastatic potential. Nature 577, 115–120.
    1. Bier DM, Leake RD, Haymond MW, Arnold KJ, Gruenke LD, Sperling MA, and Kipnis DM (1977). Measurement of “true” glucose production rates in infancy and childhood with 6,6-dideuteroglucose. Diabetes 26, 1016–23.
    1. Lau AN, Li Z, Danai LV, Westermark AM, Darnell AM, Ferreira R, Gocheva V, Sivanand S, Lien EC, Sapp KM, et al. (2020). Dissecting cell-type-specific metabolism in pancreatic ductal adenocarcinoma. Elife 9.
    1. Markert CL, Shaklee JB, and Whitt GS (1975). Evolution of a gene. Multiple genes for LDH isozymes provide a model of the evolution of gene structure, function and regulation. Science 189, 102–14.
    1. Chatham JC, Des Rosiers C, and Forder JR (2001). Evidence of separate pathways for lactate uptake and release by the perfused rat heart. Am J Physiol Endocrinol Metab 281, E794–802.
    1. Svoboda LK, Teh SSK, Sud S, Kerk S, Zebolsky A, Treichel S, Thomas D, Halbrook CJ, Lee HJ, Kremer D, et al. (2018). Menin regulates the serine biosynthetic pathway in Ewing sarcoma. J Pathol 245, 324–336.
    1. Issaq SH, Mendoza A, Kidner R, Rosales TI, Duveau DY, Heske CM, Rohde JM, Boxer MB, Thomas CJ, DeBerardinis RJ, et al. (2020). EWS-FLI1-regulated serine synthesis and exogenous serine are necessary for Ewing sarcoma cellular proliferation and tumor growth. Mol Cancer Ther.
    1. Tanner JM, Bensard C, Wei P, Krah NM, Schell JC, Gardiner J, Schiffman J, Lessnick SL, and Rutter J (2017). EWS/FLI is a Master Regulator of Metabolic Reprogramming in Ewing Sarcoma. Mol Cancer Res 15, 1517–1530.
    1. Qing G, Li B, Vu A, Skuli N, Walton ZE, Liu X, Mayes PA, Wise DR, Thompson CB, Maris JM, et al. (2012). ATF4 regulates MYC-mediated neuroblastoma cell death upon glutamine deprivation. Cancer Cell 22, 631–44.
    1. Chen P-H, Cai L, Huffman K, Yang C, Kim J, Faubert B, Boroughs L, Ko B, Sudderth J, McMillan EA, et al. (2019). Metabolic Diversity in Human Non-Small Cell Lung Cancer Cells. Molecular Cell 76, 838–851.e5.
    1. Davidson SM, Papagiannakopoulos T, Olenchock BA, Heyman JE, Keibler MA, Luengo A, Bauer MR, Jha AK, O’Brien JP, Pierce KA, et al. (2016). Environment Impacts the Metabolic Dependencies of Ras-Driven Non-Small Cell Lung Cancer. Cell Metab 23, 517–28.
    1. Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, Joseph J, Lopez M, Kalyanaraman B, Mutlu GM, Budinger GR, and Chandel NS (2010). Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci U S A 107, 8788–93.
    1. Molina JR, Sun Y, Protopopova M, Gera S, Bandi M, Bristow C, McAfoos T, Morlacchi P, Ackroyd J, Agip AA, et al. (2018). An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat Med 24, 1036–1046.
    1. Kim J, Hu Z, Cai L, Li K, Choi E, Faubert B, Bezwada D, Rodriguez-Canales J, Villalobos P, Lin Y-F, et al. (2017). CPS1 maintains pyrimidine pools and DNA synthesis in KRAS/LKB1-mutant lung cancer cells. Nature 546, 168–172.
    1. Wilson M, Davies NP, Brundler MA, McConville C, Grundy RG, and Peet AC (2009). High resolution magic angle spinning 1H NMR of childhood brain and nervous system tumours. Mol Cancer 8, 6.
    1. Kohe SE, Bennett CD, Gill SK, Wilson M, McConville C, and Peet AC (2018). Metabolic profiling of the three neural derived embryonal pediatric tumors retinoblastoma, neuroblastoma and medulloblastoma, identifies distinct metabolic profiles. Oncotarget 9, 11336–11351.
    1. LaBrosse EH, Comoy E, Bohuon C, Zucker JM, and Schweisguth O (1976). Catecholamine metabolism in neuroblastoma. Journal of the National Cancer Institute 57, 633–8.
    1. Rokita JL, Rathi KS, Cardenas MF, Upton KA, Jayaseelan J, Cross KL, Pfeil J, Egolf LE, Way GP, Farrel A, et al. (2019). Genomic Profiling of Childhood Tumor Patient-Derived Xenograft Models to Enable Rational Clinical Trial Design. Cell Rep 29, 1675–1689 e9.

Source: PubMed

3
Subscribe