Endocrinol Metab.  2022 Feb;37(1):53-61. 10.3803/EnM.2022.1402.

Development of Metabolic Synthetic Lethality and Its Implications for Thyroid Cancer

Affiliations
  • 1Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
  • 2Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea

Abstract

Cancer therapies targeting genetic alterations are a topic of great interest in the field of thyroid cancer, which frequently harbors mutations in the RAS, RAF, and RET genes. Unfortunately, U.S. Food and Drug Administration-approved BRAF inhibitors have relatively low therapeutic efficacy against BRAF-mutant thyroid cancer; in addition, the cancer often acquires drug resistance, which prevents effective treatment. Recent advances in genomics and transcriptomics are leading to a more complete picture of the range of mutations, both driver and messenger, present in thyroid cancer. Furthermore, our understanding of cancer suggests that oncogenic mutations drive tumorigenesis and induce rewiring of cancer cell metabolism, which promotes survival of mutated cells. Synthetic lethality (SL) is a method of neutralizing mutated genes that were previously considered untargetable by traditional genotype-targeted treatments. Because these metabolic events are specific to cancer cells, we have the opportunity to develop new therapies that target tumor cells specifically without affecting healthy tissue. Here, we describe developments in metabolism-based cancer therapy, focusing on the concept of metabolic SL in thyroid cancer. Finally, we discuss the essential implications of metabolic reprogramming and its role in the future direction of SL for thyroid cancer.

Keyword

Synthetic lethal mutations; Thyroid neoplasms; Metabolic reprogramming

Figure

  • Fig. 1 The principles of synthetic lethality (SL) in cancer. Loss or inhibition of either of the protein products of gene A or B alone, or overexpression of gene A, is viable. However, pharmacological intervention by the partner gene product will result in an SL interaction in tumor cells with a loss-of-function mutation in a tumor suppressor gene (TSG) (A). In addition, pharmacological intervention by the partner gene product will result in a synthetic dosage lethality (SDL) interaction in tumor cells with a gain-of-function mutation or overexpression of the oncogene (B). The yellow star denotes a mutation. The thicker arrow denotes overexpression. The crossed line denotes inhibition of the gene product by pharmacological intervention.

  • Fig. 2 Application of metabolic synthetic lethality (SL) to cancer. The main metabolic pathways involved in metabolic reprogramming of cancer cells harboring mutant genes that may provide a target for SL.


Cited by  1 articles

The Role of De novo Serine Biosynthesis from Glucose in Papillary Thyroid Cancer
Seong Eun Lee, Na Rae Choi, Jin-Man Kim, Mi Ae Lim, Bon Seok Koo, Yea Eun Kang
Int J Thyroidol. 2023;16(2):175-183.    doi: 10.11106/ijt.2023.16.2.175.


Reference

1. Bridges CB. The origin of variations in sexual and sex-limited characters. Am Nat. 1922; 56:51–63.
Article
2. Zhang B, Tang C, Yao Y, Chen X, Zhou C, Wei Z, et al. The tumor therapy landscape of synthetic lethality. Nat Commun. 2021; 12:1275.
Article
3. Ashworth A, Lord CJ. Synthetic lethal therapies for cancer: what’s next after PARP inhibitors? Nat Rev Clin Oncol. 2018; 15:564–76.
Article
4. O’Neil NJ, Bailey ML, Hieter P. Synthetic lethality and cancer. Nat Rev Genet. 2017; 18:613–23.
Article
5. Chen M, Cai X. Synthetic lethality is a novel and potential paradigm for precision medicine in advanced hepatocellular carcinoma. Liver Cancer. 2020; 9:225–6.
Article
6. Sajesh BV, Guppy BJ, McManus KJ. Synthetic genetic targeting of genome instability in cancer. Cancers (Basel). 2013; 5:739–61.
Article
7. Ashworth A, Lord CJ, Reis-Filho JS. Genetic interactions in cancer progression and treatment. Cell. 2011; 145:30–8.
Article
8. Hartwell LH, Szankasi P, Roberts CJ, Murray AW, Friend SH. Integrating genetic approaches into the discovery of anticancer drugs. Science. 1997; 278:1064–8.
Article
9. Luo J, Solimini NL, Elledge SJ. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell. 2009; 136:823–37.
Article
10. Dhoonmoon A, Schleicher EM, Clements KE, Nicolae CM, Moldovan GL. Genome-wide CRISPR synthetic lethality screen identifies a role for the ADP-ribosyltransferase PARP14 in DNA replication dynamics controlled by ATR. Nucleic Acids Res. 2020; 48:7252–64.
Article
11. Parrish P, Thomas JD, Gabel AM, Kamlapurkar S, Bradley RK, Berger AH. Discovery of synthetic lethal and tumor suppressor paralog pairs in the human genome. Cell Rep. 2021; 36:109597.
Article
12. Haince JF, Rouleau M, Hendzel MJ, Masson JY, Poirier GG. Targeting poly(ADP-ribosyl)ation: a promising approach in cancer therapy. Trends Mol Med. 2005; 11:456–63.
Article
13. Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005; 434:917–21.
Article
14. Vieri M, Preisinger C, Schemionek M, Salimi A, Patterson JB, Samali A, et al. Targeting of BCR-ABL1 and IRE1α induces synthetic lethality in Philadelphia-positive acute lymphoblastic leukemia. Carcinogenesis. 2021; 42:272–84.
Article
15. Kim D, Hwang JH, Suh JM, Kim H, Song JH, Hwang ES, et al. RET/PTC (rearranged in transformation/papillary thyroid carcinomas) tyrosine kinase phosphorylates and activates phosphoinositide-dependent kinase 1 (PDK1): an alternative phosphatidylinositol 3-kinase-independent pathway to activate PDK1. Mol Endocrinol. 2003; 17:1382–94.
Article
16. Wells SA Jr, Santoro M. Update: the status of clinical trials with kinase inhibitors in thyroid cancer. J Clin Endocrinol Metab. 2014; 99:1543–55.
Article
17. Eiring AM, Page B, Kraft IL, Mason CC, Vellore NA, Resetca D, et al. Combined STAT3 and BCR-ABL1 inhibition induces synthetic lethality in therapy-resistant chronic myeloid leukemia. Leukemia. 2015; 29:586–97.
Article
18. Aguirre AJ, Hahn WC. Synthetic lethal vulnerabilities in KRAS-mutant cancers. Cold Spring Harb Perspect Med. 2018; 8:a031518.
19. Hu K, Li K, Lv J, Feng J, Chen J, Wu H, et al. Suppression of the SLC7A11/glutathione axis causes synthetic lethality in KRAS-mutant lung adenocarcinoma. J Clin Invest. 2020; 130:1752–66.
Article
20. Schulten HJ, Salama S, Al-Ahmadi A, Al-Mansouri Z, Mirza Z, Al-Ghamdi K, et al. Comprehensive survey of HRAS, KRAS, and NRAS mutations in proliferative thyroid lesions from an ethnically diverse population. Anticancer Res. 2013; 33:4779–84.
21. Zhu X, Zhao L, Park JW, Willingham MC, Cheng SY. Synergistic signaling of KRAS and thyroid hormone receptor β mutants promotes undifferentiated thyroid cancer through MYC up-regulation. Neoplasia. 2014; 16:757–69.
Article
22. McKeown MR, Bradner JE. Therapeutic strategies to inhibit MYC. Cold Spring Harb Perspect Med. 2014; 4:a014266.
Article
23. Cermelli S, Jang IS, Bernard B, Grandori C. Synthetic lethal screens as a means to understand and treat MYC-driven cancers. Cold Spring Harb Perspect Med. 2014; 4:a014209.
Article
24. Kessler JD, Kahle KT, Sun T, Meerbrey KL, Schlabach MR, Schmitt EM, et al. A SUMOylation-dependent transcriptional subprogram is required for Myc-driven tumorigenesis. Science. 2012; 335:348–53.
Article
25. Liu L, Ulbrich J, Muller J, Wustefeld T, Aeberhard L, Kress TR, et al. Deregulated MYC expression induces dependence upon AMPK-related kinase 5. Nature. 2012; 483:608–12.
Article
26. Toyoshima M, Howie HL, Imakura M, Walsh RM, Annis JE, Chang AN, et al. Functional genomics identifies therapeutic targets for MYC-driven cancer. Proc Natl Acad Sci U S A. 2012; 109:9545–50.
Article
27. Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016; 23:27–47.
Article
28. Lee J, Chang JY, Kang YE, Yi S, Lee MH, Joung KH, et al. Mitochondrial energy metabolism and thyroid cancers. Endocrinol Metab (Seoul). 2015; 30:117–23.
Article
29. Bajrami I, Walker C, Krastev DB, Weekes D, Song F, Wicks AJ, et al. Sirtuin inhibition is synthetic lethal with BRCA1 or BRCA2 deficiency. Commun Biol. 2021; 4:1270.
Article
30. Villanueva MT. Anticancer therapy: metabolic synthetic lethality. Nat Rev Drug Discov. 2018; 17:543.
31. Molina JR, Sun Y, Protopopova M, Gera S, Bandi M, Bristow C, et al. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat Med. 2018; 24:1036–46.
Article
32. Zecchini V, Frezza C. Metabolic synthetic lethality in cancer therapy. Biochim Biophys Acta Bioenerg. 2017; 1858:723–31.
Article
33. Ban EJ, Kim D, Kim JK, Kang SW, Lee J, Jeong JJ, et al. Lactate dehydrogenase A as a potential new biomarker for thyroid cancer. Endocrinol Metab (Seoul). 2021; 36:96–105.
Article
34. Bao L, Xu T, Lu X, Huang P, Pan Z, Ge M. Metabolic reprogramming of thyroid cancer cells and crosstalk in their microenvironment. Front Oncol. 2021; 11:773028.
Article
35. Pathria G, Scott DA, Feng Y, Sang Lee J, Fujita Y, Zhang G, et al. Targeting the Warburg effect via LDHA inhibition engages ATF4 signaling for cancer cell survival. EMBO J. 2018; 37:e99735.
36. Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, et al. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci U S A. 2010; 107:2037–42.
Article
37. Scaglia N, Chisholm JW, Igal RA. Inhibition of stearoylCoA desaturase-1 inactivates acetyl-CoA carboxylase and impairs proliferation in cancer cells: role of AMPK. PLoS One. 2009; 4:e6812.
Article
38. Cheong JH, Park ES, Liang J, Dennison JB, Tsavachidou D, Nguyen-Charles C, et al. Dual inhibition of tumor energy pathway by 2-deoxyglucose and metformin is effective against a broad spectrum of preclinical cancer models. Mol Cancer Ther. 2011; 10:2350–62.
Article
39. Benjamin D, Robay D, Hindupur SK, Pohlmann J, Colombi M, El-Shemerly MY, et al. Dual inhibition of the lactate transporters MCT1 and MCT4 is synthetic lethal with metformin due to NAD+ depletion in cancer cells. Cell Rep. 2018; 25:3047–58.
Article
40. Benjamin D, Colombi M, Hindupur SK, Betz C, Lane HA, El-Shemerly MY, et al. Syrosingopine sensitizes cancer cells to killing by metformin. Sci Adv. 2016; 2:e1601756.
Article
41. Durai L, Ravindran S, Arvind K, Karunagaran D, Vijayalakshmi R. Synergistic effect of metformin and vemurufenib (PLX4032) as a molecular targeted therapy in anaplastic thyroid cancer: an in vitro study. Mol Biol Rep. 2021; 48:7443–56.
Article
42. Yang M, Vousden KH. Serine and one-carbon metabolism in cancer. Nat Rev Cancer. 2016; 16:650–62.
Article
43. Sun WY, Kim HM, Jung WH, Koo JS. Expression of serine/glycine metabolism-related proteins is different according to the thyroid cancer subtype. J Transl Med. 2016; 14:168.
Article
44. Favaro E, Bensaad K, Chong MG, Tennant DA, Ferguson DJ, Snell C, et al. Glucose utilization via glycogen phosphorylase sustains proliferation and prevents premature senescence in cancer cells. Cell Metab. 2012; 16:751–64.
Article
45. Jariyal H, Weinberg F, Achreja A, Nagarath D, Srivastava A. Synthetic lethality: a step forward for personalized medicine in cancer. Drug Discov Today. 2020; 25:305–20.
Article
46. Xing M. BRAF mutation in thyroid cancer. Endocr Relat Cancer. 2005; 12:245–62.
Article
47. Garnett MJ, Rana S, Paterson H, Barford D, Marais R. Wild-type and mutant B-RAF activate C-RAF through distinct mechanisms involving heterodimerization. Mol Cell. 2005; 20:963–9.
Article
48. Knauf JA, Ma X, Smith EP, Zhang L, Mitsutake N, Liao XH, et al. Targeted expression of BRAFV600E in thyroid cells of transgenic mice results in papillary thyroid cancers that undergo dedifferentiation. Cancer Res. 2005; 65:4238–45.
Article
49. Begum S, Rosenbaum E, Henrique R, Cohen Y, Sidransky D, Westra WH. BRAF mutations in anaplastic thyroid carcinoma: implications for tumor origin, diagnosis and treatment. Mod Pathol. 2004; 17:1359–63.
Article
50. Fallahi P, Ferrari SM, Galdiero MR, Varricchi G, Elia G, Ragusa F, et al. Molecular targets of tyrosine kinase inhibitors in thyroid cancer. Semin Cancer Biol. 2022; 79:180–96.
Article
51. Jurchott K, Kuban RJ, Krech T, Bluthgen N, Stein U, Walther W, et al. Identification of Y-box binding protein 1 as a core regulator of MEK/ERK pathway-dependent gene signatures in colorectal cancer cells. PLoS Genet. 2010; 6:e1001231.
Article
52. Stelniec-Klotz I, Legewie S, Tchernitsa O, Witzel F, Klinger B, Sers C, et al. Reverse engineering a hierarchical regulatory network downstream of oncogenic KRAS. Mol Syst Biol. 2012; 8:601.
Article
53. Klotz-Noack K, Klinger B, Rivera M, Bublitz N, Uhlitz F, Riemer P, et al. SFPQ depletion is synthetically lethal with BRAFV600E in colorectal cancer cells. Cell Rep. 2020; 32:108184.
Article
54. Bi O, Anene CA, Nsengimana J, Shelton M, Roberts W, Newton-Bishop J, et al. SFPQ promotes an oncogenic transcriptomic state in melanoma. Oncogene. 2021; 40:5192–203.
Article
55. Wang X, Zhang Y, Han ZG, He KY. Malignancy of cancers and synthetic lethal interactions associated with mutations of cancer driver genes. Medicine (Baltimore). 2016; 95:e2697.
Article
56. Li Y, Su X, Feng C, Liu S, Guan H, Sun Y, et al. CYP2S1 is a synthetic lethal target in BRAFV600E-driven thyroid cancers. Signal Transduct Target Ther. 2020; 5:191.
Article
57. Kang HB, Fan J, Lin R, Elf S, Ji Q, Zhao L, et al. Metabolic rewiring by oncogenic BRAF V600E links ketogenesis pathway to BRAF-MEK1 signaling. Mol Cell. 2015; 59:345–58.
Article
58. Kaplon J, Zheng L, Meissl K, Chaneton B, Selivanov VA, Mackay G, et al. A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence. Nature. 2013; 498:109–12.
Article
59. Scortegagna M, Lau E, Zhang T, Feng Y, Sereduk C, Yin H, et al. PDK1 and SGK3 contribute to the growth of BRAF-mutant melanomas and are potential therapeutic targets. Cancer Res. 2015; 75:1399–412.
Article
60. Porchia LM, Guerra M, Espinosa AV, Saji M, Kulp SK, Ringel MD, et al. OSU03012, a novel PDK1 inhibitor, decreases thyroid cancer proliferation and migration via multiple downstream pathways. Cancer Res. 2014; 66(8 Suppl):1205.
61. Yeh JJ, Lunetta KL, van Orsouw NJ, Moore FD Jr, Mutter GL, Vijg J, et al. Somatic mitochondrial DNA (mtDNA) mutations in papillary thyroid carcinomas and differential mtDNA sequence variants in cases with thyroid tumours. Oncogene. 2000; 19:2060–6.
Article
62. Yi HS, Chang JY, Kim KS, Shong M. Oncogenes, mitochondrial metabolism, and quality control in differentiated thyroid cancer. Korean J Intern Med. 2017; 32:780–9.
Article
63. Raimundo N, Baysal BE, Shadel GS. Revisiting the TCA cycle: signaling to tumor formation. Trends Mol Med. 2011; 17:641–9.
Article
64. Sun Y, Bandi M, Lofton T, Smith M, Bristow CA, Carugo A, et al. Functional genomics reveals synthetic lethality between phosphogluconate dehydrogenase and oxidative phosphorylation. Cell Rep. 2019; 26:469–82.
Article
Full Text Links
  • ENM
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