Yonsei Med J.  2018 Dec;59(10):1143-1149. 10.3349/ymj.2018.59.10.1143.

Cancer Metabolism as a Mechanism of Treatment Resistance and Potential Therapeutic Target in Hepatocellular Carcinoma

Affiliations
  • 1Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea. yunmijin@yuhs.ac
  • 2Division of Life Science, College of Life Science and Bioengineering, Incheon National University, Incheon, Korea.

Abstract

Various molecular targeted therapies and diagnostic modalities have been developed for the treatment of hepatocellular carcinoma (HCC); however, HCC still remains a difficult malignancy to cure. Recently, the focus has shifted to cancer metabolism for the diagnosis and treatment of various cancers, including HCC. In addition to conventional diagnostics, the measurement of enhanced tumor cell metabolism using F-18 fluorodeoxyglucose (18F-FDG) for increased glycolysis or C-11 acetate for fatty acid synthesis by positron emission tomography/computed tomography (PET/CT) is well established for clinical management of HCC. Unlike tumors displaying the Warburg effect, HCCs vary substantially in terms of 18F-FDG uptake, which considerably reduces the sensitivity for tumor detection. Accordingly, C-11 acetate has been proposed as a complementary radiotracer for detecting tumors that are not identified by 18F-FDG. In addition to HCC diagnosis, since the degree of 18F-FDG uptake converted to standardized uptake value (SUV) correlates well with tumor aggressiveness, 18F-FDG PET/CT scans can predict patient outcomes such as treatment response and survival with an inverse relationship between SUV and survival. The loss of tumor suppressor genes or activation of oncogenes plays an important role in promoting HCC development, and might be involved in the "metabolic reprogramming" of cancer cells. Mutations in various genes such as TERT, CTNNB1, TP53, and Axin1 are responsible for the development of HCC. Some microRNAs (miRNAs) involved in cancer metabolism are deregulated in HCC, indicating that the modulation of genes/miRNAs might affect HCC growth or metastasis. In this review, we will discuss cancer metabolism as a mechanism for treatment resistance, as well as an attractive potential therapeutic target in HCC.

Keyword

Hepatocellular carcinoma; cancer metabolism; positron emission tomography/computed tomography (PET/CT); drug resistance

MeSH Terms

Carcinoma, Hepatocellular*
Diagnosis
Drug Resistance
Electrons
Fluorodeoxyglucose F18
Genes, Tumor Suppressor
Glycolysis
Humans
Metabolism*
MicroRNAs
Molecular Targeted Therapy
Neoplasm Metastasis
Oncogenes
Positron-Emission Tomography and Computed Tomography
Fluorodeoxyglucose F18
MicroRNAs

Figure

  • Fig. 1 Hepatocellular carcinoma positive for F-18 fluorodeoxyglucose (A), but negative for C-11 acetate (B).

  • Fig. 2 Hepatocellular carcinoma negative for F-18 fluorodeoxyglucose (A), but positive C-11 acetate (B).

  • Fig. 3 Differences in the expression of glucose transport 1 (A and C) and monocarboxylate transporter 1 (B and D) in hepatocellular carcinoma (HCC) samples, based on 18F-fluorodeoxyglucose and 11C-acetate uptake. Human HCC samples were used. Immunohistochemistry (IHC) was performed as described previously.16 After antigen retrieval, IHC was performed using indicated antibodies. Scale bars: 40 µm.


Reference

1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010; 127:2893–2917. PMID: 21351269.
Article
2. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015; 136:E359–E386. PMID: 25220842.
Article
3. Velázquez RF, Rodríguez M, Navascués CA, Linares A, Pérez R, Sotorríos NG, et al. Prospective analysis of risk factors for hepatocellular carcinoma in patients with liver cirrhosis. Hepatology. 2003; 37:520–527. PMID: 12601348.
Article
4. Fattovich G, Stroffolini T, Zagni I, Donato F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology. 2004; 127(5 Suppl 1):S35–S50. PMID: 15508101.
Article
5. Bruix J, Sherman M. American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma: an update. Hepatology. 2011; 53:1020–1022. PMID: 21374666.
Article
6. Song PP, Xia JF, Inagaki Y, Hasegawa K, Sakamoto Y, Kokudo N, et al. Controversies regarding and perspectives on clinical utility of biomarkers in hepatocellular carcinoma. World J Gastroenterol. 2016; 22:262–274. PMID: 26755875.
Article
7. Cameron AM. Screening for viral hepatitis and hepatocellular cancer. Surg Clin North Am. 2015; 95:1013–1021. PMID: 26315520.
Article
8. Phelps ME, Huang SC, Hoffman EJ, Selin C, Sokoloff L, Kuhl DE. Tomographic measurement of local cerebral glucose metabolic rate in humans with (F-18)2-fluoro-2-deoxy-D-glucose: validation of method. Ann Neurol. 1979; 6:371–388. PMID: 117743.
Article
9. Gambhir SS, Czernin J, Schwimmer J, Silverman DH, Coleman RE, Phelps ME. A tabulated summary of the FDG PET literature. J Nucl Med. 2001; 42(5 Suppl):1S–93S. PMID: 11483694.
10. Stokkel MP, Draisma A, Pauwels EK. Positron emission tomography with 2-[18F]-fluoro-2-deoxy-D-glucose in oncology. Part IIIb: therapy response monitoring in colorectal and lung tumours, head and neck cancer, hepatocellular carcinoma and sarcoma. J Cancer Res Clin Oncol. 2001; 127:278–285. PMID: 11355142.
11. Iwata Y, Shiomi S, Sasaki N, Jomura H, Nishiguchi S, Seki S, et al. Clinical usefulness of positron emission tomography with fluorine-18-fluorodeoxyglucose in the diagnosis of liver tumors. Ann Nucl Med. 2000; 14:121–126. PMID: 10830530.
Article
12. Khan MA, Combs CS, Brunt EM, Lowe VJ, Wolverson MK, Solomon H, et al. Positron emission tomography scanning in the evaluation of hepatocellular carcinoma. J Hepatol. 2000; 32:792–797. PMID: 10845666.
Article
13. Böhm B, Voth M, Geoghegan J, Hellfritzsch H, Petrovich A, Scheele J, et al. Impact of positron emission tomography on strategy in liver resection for primary and secondary liver tumors. J Cancer Res Clin Oncol. 2004; 130:266–272. PMID: 14767761.
14. Medina RA, Owen GI. Glucose transporters: expression, regulation and cancer. Biol Res. 2002; 35:9–26. PMID: 12125211.
Article
15. Amann T, Maegdefrau U, Hartmann A, Agaimy A, Marienhagen J, Weiss TS, et al. GLUT1 expression is increased in hepatocellular carcinoma and promotes tumorigenesis. Am J Pathol. 2009; 174:1544–1552. PMID: 19286567.
Article
16. Lee M, Jeon JY, Neugent ML, Kim JW, Yun M. 18F-Fluorodeoxyglucose uptake on positron emission tomography/computed tomography is associated with metastasis and epithelial-mesenchymal transition in hepatocellular carcinoma. Clin Exp Metastasis. 2017; 34:251–260. PMID: 28429188.
Article
17. Hwang SH, Lee JW, Cho HJ, Kim KS, Choi GH, Yun M. Prognostic value of metabolic tumor volume and total lesion glycolysis on preoperative 18F-FDG PET/CT in patients with very early and early hepatocellular carcinoma. Clin Nucl Med. 2017; 42:34–39. PMID: 27775949.
Article
18. Schug ZT, Vande Voorde J, Gottlieb E. The metabolic fate of acetate in cancer. Nat Rev Cancer. 2016; 16:708–717. PMID: 27562461.
Article
19. Ho CL, Yu SC, Yeung DW. 11C-acetate PET imaging in hepatocellular carcinoma and other liver masses. J Nucl Med. 2003; 44:213–221. PMID: 12571212.
20. Bjornson E, Mukhopadhyay B, Asplund A, Pristovsek N, Cinar R, Romeo S, et al. Stratification of hepatocellular carcinoma patients based on acetate utilization. Cell Rep. 2015; 13:2014–2026. PMID: 26655911.
21. Jeon J, Lee M, Whang S, Kim JW, Cho A, Yun M. Regulation of acetate utilization by monocarboxylate transporter 1 (MCT1) in hepatocellular carcinoma (HCC). Oncol Res. 2018; 26:71–81. PMID: 28390113.
Article
22. Breuhahn K, Gores G, Schirmacher P. Strategies for hepatocellular carcinoma therapy and diagnostics: lessons learned from high throughput and profiling approaches. Hepatology. 2011; 53:2112–2121. PMID: 21433041.
Article
23. Thorgeirsson SS. Genomic decoding of hepatocellular carcinoma. Gastroenterology. 2006; 131:1344–1346. PMID: 17030203.
Article
24. Wang K, Lim HY, Shi S, Lee J, Deng S, Xie T, et al. Genomic landscape of copy number aberrations enables the identification of oncogenic drivers in hepatocellular carcinoma. Hepatology. 2013; 58:706–717. PMID: 23505090.
Article
25. Kan Z, Zheng H, Liu X, Li S, Barber TD, Gong Z, et al. Whole-genome sequencing identifies recurrent mutations in hepatocellular carcinoma. Genome Res. 2013; 23:1422–1433. PMID: 23788652.
Article
26. Totoki Y, Tatsuno K, Covington KR, Ueda H, Creighton CJ, Kato M, et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat Genet. 2014; 46:1267–1273. PMID: 25362482.
Article
27. Schulze K, Imbeaud S, Letouzé E, Alexandrov LB, Calderaro J, Rebouissou S, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet. 2015; 47:505–511. PMID: 25822088.
Article
28. Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH, et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 2012; 44:760–764. PMID: 22634756.
Article
29. Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012; 44:694–698. PMID: 22561517.
Article
30. Huang J, Deng Q, Wang Q, Li KY, Dai JH, Li N, et al. Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma. Nat Genet. 2012; 44:1117–1121. PMID: 22922871.
Article
31. Cleary SP, Jeck WR, Zhao X, Chen K, Selitsky SR, Savich GL, et al. Identification of driver genes in hepatocellular carcinoma by exome sequencing. Hepatology. 2013; 58:1693–1702. PMID: 23728943.
Article
32. Ahn SM, Jang SJ, Shim JH, Kim D, Hong SM, Sung CO, et al. Genomic portrait of resectable hepatocellular carcinomas: implications of RB1 and FGF19 aberrations for patient stratification. Hepatology. 2014; 60:1972–1982. PMID: 24798001.
33. Nault JC, Zucman-Rossi J. TERT promoter mutations in primary liver tumors. Clin Res Hepatol Gastroenterol. 2016; 40:9–14. PMID: 26336998.
Article
34. Jones RG, Thompson CB. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev. 2009; 23:537–548. PMID: 19270154.
Article
35. Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell. 2006; 126:107–120. PMID: 16839880.
Article
36. Dang CV, Le A, Gao P. MYC-induced cancer cell energy metabolism and therapeutic opportunities. Clin Cancer Res. 2009; 15:6479–6483. PMID: 19861459.
Article
37. Shim H, Dolde C, Lewis BC, Wu CS, Dang G, Jungmann RA, et al. c-Myc transactivation of LDH-A: implications for tumor metabolism and growth. Proc Natl Acad Sci U S A. 1997; 94:6658–6663. PMID: 9192621.
Article
38. Levine AJ, Puzio-Kuter AM. The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science. 2010; 330:1340–1344. PMID: 21127244.
Article
39. Méndez-Lucas A, Li X, Hu J, Che L, Song X, Jia J, et al. Glucose catabolism in liver tumors induced by c-MYC can be sustained by various PKM1/PKM2 ratios and pyruvate kinase activities. Cancer Res. 2017; 77:4355–4364. PMID: 28630053.
Article
40. Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol. 1992; 12:5447–5454. PMID: 1448077.
Article
41. Semenza GL, Roth PH, Fang HM, Wang GL. Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J Biol Chem. 1994; 269:23757–23763. PMID: 8089148.
Article
42. Firth JD, Ebert BL, Pugh CW, Ratcliffe PJ. Oxygen-regulated control elements in the phosphoglycerate kinase 1 and lactate dehydrogenase A genes: similarities with the erythropoietin 3’ enhancer. Proc Natl Acad Sci U S A. 1994; 91:6496–6500. PMID: 8022811.
Article
43. Rigiracciolo DC, Scarpelli A, Lappano R, Pisano A, Santolla MF, De Marco P, et al. Copper activates HIF-1α/GPER/VEGF signalling in cancer cells. Oncotarget. 2015; 6:34158–34177. PMID: 26415222.
Article
44. Xia X, Lemieux ME, Li W, Carroll JS, Brown M, Liu XS, et al. Integrative analysis of HIF binding and transactivation reveals its role in maintaining histone methylation homeostasis. Proc Natl Acad Sci U S A. 2009; 106:4260–4265. PMID: 19255431.
Article
45. Hamaguchi T, Iizuka N, Tsunedomi R, Hamamoto Y, Miyamoto T, Iida M, et al. Glycolysis module activated by hypoxia-inducible factor 1alpha is related to the aggressive phenotype of hepatocellular carcinoma. Int J Oncol. 2008; 33:725–731. PMID: 18813785.
Article
46. Daskalow K, Rohwer N, Raskopf E, Dupuy E, Kühl A, Loddenkemper C, et al. Role of hypoxia-inducible transcription factor 1alpha for progression and chemosensitivity of murine hepatocellular carcinoma. J Mol Med (Berl). 2010; 88:817–827. PMID: 20383692.
47. Lau CK, Yang ZF, Ho DW, Ng MN, Yeoh GC, Poon RT, et al. An Akt/hypoxia-inducible factor-1alpha/platelet-derived growth factor-BB autocrine loop mediates hypoxia-induced chemoresistance in liver cancer cells and tumorigenic hepatic progenitor cells. Clin Cancer Res. 2009; 15:3462–3471. PMID: 19447872.
48. Krishnamachary B, Zagzag D, Nagasawa H, Rainey K, Okuyama H, Baek JH, et al. Hypoxia-inducible factor-1-dependent repression of E-cadherin in von Hippel-Lindau tumor suppressor-null renal cell carcinoma mediated by TCF3, ZFHX1A, and ZFHX1B. Cancer Res. 2006; 66:2725–2731. PMID: 16510593.
Article
49. Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, et al. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat Cell Biol. 2008; 10:295–305. PMID: 18297062.
50. Zhang L, Huang G, Li X, Zhang Y, Jiang Y, Shen J, et al. Hypoxia induces epithelial-mesenchymal transition via activation of SNAI1 by hypoxia-inducible factor-1α in hepatocellular carcinoma. BMC Cancer. 2013; 13:108. PMID: 23496980.
Article
51. Wang L, Wang WL, Zhang Y, Guo SP, Zhang J, Li QL. Epigenetic and genetic alterations of PTEN in hepatocellular carcinoma. Hepatol Res. 2007; 37:389–396. PMID: 17441812.
Article
52. Peyrou M, Bourgoin L, Foti M. PTEN in liver diseases and cancer. World J Gastroenterol. 2010; 16:4627–4633. PMID: 20872961.
53. Rahman MA, Kyriazanos ID, Ono T, Yamanoi A, Kohno H, Tsuchiya M, et al. Impact of PTEN expression on the outcome of hepatitis C virus-positive cirrhotic hepatocellular carcinoma patients: possible relationship with COX II and inducible nitric oxide synthase. Int J Cancer. 2002; 100:152–157. PMID: 12115563.
Article
54. Zundel W, Schindler C, Haas-Kogan D, Koong A, Kaper F, Chen E, et al. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Dev. 2000; 14:391–396. PMID: 10691731.
Article
55. Tian T, Nan KJ, Wang SH, Liang X, Lu CX, Guo H, et al. PTEN regulates angiogenesis and VEGF expression through phosphatase-dependent and -independent mechanisms in HepG2 cells. Carcinogenesis. 2010; 31:1211–1219. PMID: 20430845.
Article
56. Chen C, Pore N, Behrooz A, Ismail-Beigi F, Maity A. Regulation of glut1 mRNA by hypoxia-inducible factor-1. Interaction between H-ras and hypoxia. J Biol Chem. 2001; 276:9519–9525. PMID: 11120745.
57. Sakamoto T, Niiya D, Seiki M. Targeting the Warburg effect that arises in tumor cells expressing membrane type-1 matrix metalloproteinase. J Biol Chem. 2011; 286:14691–14704. PMID: 21372132.
Article
58. Wang H, Zhang C, Xu L, Zang K, Ning Z, Jiang F, et al. Bufalin suppresses hepatocellular carcinoma invasion and metastasis by targeting HIF-1α via the PI3K/AKT/mTOR pathway. Oncotarget. 2016; 7:20193–20208. PMID: 26958938.
Article
59. Wang Y, Ren J, Gao Y, Ma JZ, Toh HC, Chow P, et al. MicroRNA-224 targets SMAD family member 4 to promote cell proliferation and negatively influence patient survival. PLoS One. 2013; 8:e68744. PMID: 23922662.
Article
60. Wang Y, Lee CG. MicroRNA and cancer--focus on apoptosis. J Cell Mol Med. 2009; 13:12–23. PMID: 19175697.
61. Fornari F, Gramantieri L, Ferracin M, Veronese A, Sabbioni S, Calin GA, et al. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene. 2008; 27:5651–5661. PMID: 18521080.
Article
62. Lee J, Kemper JK. Controlling SIRT1 expression by microRNAs in health and metabolic disease. Aging (Albany NY). 2010; 2:527–534. PMID: 20689156.
Article
63. Lee J, Padhye A, Sharma A, Song G, Miao J, Mo YY, et al. A pathway involving farnesoid X receptor and small heterodimer partner positively regulates hepatic sirtuin 1 levels via microRNA-34a inhibition. J Biol Chem. 2010; 285:12604–12611. PMID: 20185821.
Article
64. Qin Y, Lu Y, Wang R, Li W, Qu X. SL1122-37, a novel derivative of sorafenib, has greater effects than sorafenib on the inhibition of human hepatocellular carcinoma (HCC) growth and prevention of angiogenesis. Biosci Trends. 2013; 7:237–244. PMID: 24241174.
Article
65. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008; 359:378–390. PMID: 18650514.
Article
66. Aft RL, Zhang FW, Gius D. Evaluation of 2-deoxy-D-glucose as a chemotherapeutic agent: mechanism of cell death. Br J Cancer. 2002; 87:805–812. PMID: 12232767.
Article
67. Zhang Y, Huang F, Wang J, Luo H, Wang Z. 2-DG-regulated RIP and c-FLIP effect on liver cancer cell apoptosis induced by TRAIL. Med Sci Monit. 2015; 21:3442–3448. PMID: 26552967.
Article
68. Tesori V, Piscaglia AC, Samengo D, Barba M, Bernardini C, Scatena R, et al. The multikinase inhibitor Sorafenib enhances glycolysis and synergizes with glycolysis blockade for cancer cell killing. Sci Rep. 2015; 5:9149. PMID: 25779766.
Article
69. Geschwind JF, Georgiades CS, Ko YH, Pedersen PL. Recently elucidated energy catabolism pathways provide opportunities for novel treatments in hepatocellular carcinoma. Expert Rev Anticancer Ther. 2004; 4:449–457. PMID: 15161443.
Article
70. Ko YH, Verhoeven HA, Lee MJ, Corbin DJ, Vogl TJ, Pedersen PL. A translational study “case report” on the small molecule “energy blocker” 3-bromopyruvate (3BP) as a potent anticancer agent: from bench side to bedside. J Bioenerg Biomembr. 2012; 44:163–170. PMID: 22328020.
Article
71. Schimmer AD, Thomas MP, Hurren R, Gronda M, Pellecchia M, Pond GR, et al. Identification of small molecules that sensitize resistant tumor cells to tumor necrosis factor-family death receptors. Cancer Res. 2006; 66:2367–2375. PMID: 16489043.
Article
72. Wood TE, Dalili S, Simpson CD, Hurren R, Mao X, Saiz FS, et al. A novel inhibitor of glucose uptake sensitizes cells to FAS-induced cell death. Mol Cancer Ther. 2008; 7:3546–3555. PMID: 19001437.
Article
73. Liu Y, Cao Y, Zhang W, Bergmeier S, Qian Y, Akbar H, et al. A small-molecule inhibitor of glucose transporter 1 downregulates glycolysis, induces cell-cycle arrest, and inhibits cancer cell growth in vitro and in vivo. Mol Cancer Ther. 2012; 11:1672–1682. PMID: 22689530.
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr