Abstract
Introduction
Anaplastic thyroid carcinoma (ATC) is the rarest subtype of thyroid cancer, however it disproportionately accounts for a large percentage of all thyroid cancer related deaths and is considered one of the most lethal solid tumors in humans, having a median survival of only a few months upon diagnosis. While a variety of treatment options are available including surgery, radiation and targeted therapies, response rates are low, due in part to the drug-resistant nature of this disease; therefore, new avenues for therapeutic intervention are surely needed. Recent investigation into the metabolic profile of ATC has revealed a tumor-specific dependency for increased de novo lipogenesis, offering new insight into the molecular mechanisms that govern disease initiation and progression.
Areas Covered
Herein we summarize known oncogenic signaling pathways and current therapeutic strategies for the treatment of ATC. We further discuss the unique expression pattern of lipid metabolism constituents in this disease. Additionally, the current literature correlating aberrant lipogenesis with carcinogenesis is reviewed, and the implications of targeting this pathway as an innovative approach for treating ATC and other malignancies is discussed. As stearoyl-CoA desaturase (SCD) is the most differentially expressed constituent of lipid metabolism in ATC, an additional focus on this enzyme as a novel therapeutic target is applied.
Expert Opinion
This section is used to summarize the current research efforts underway in defining the role of lipid metabolism specifically in thyroid carcinoma. Included is a brief summary of lipid metabolism factors for which inhibitors have been generated and are under current investigation as anti-cancer agents. Finally, research limitations regarding the use of these inhibitors against components of this pathway are discussed.
Keywords: De novo lipogenesis, lipid metabolism, anaplastic thyroid carcinoma, stearoyl-CoA desaturase, fatty acid synthase, sterol regulatory-element binding protein, carcinogenesis, chemoresistance, fatty acids
1. Common genetic anomalies in ATC
Anaplastic thyroid carcinoma (ATC) is thought to arise from pre-existing well-differentiated thyroid carcinomas (WDTC) through the acquisition of additional genetic deviations. This has been deciphered through multiple genetically engineered mouse models that progress from follicular and papillary WDTC to undifferentiated ATC1. Currently, numerous transformations in molecular mechanisms governing the development of thyroid malignancy and subsequent progression into poorly differentiated and ATC have been identified. These include activation of receptor tyrosine kinases, proliferative or pro-survival intracellular signaling pathways, abnormalities in cell cycle constituents, alterations in cytoskeletal components and regulators of mitosis, and overexpression of apoptotic inhibitors2-4. A summary of characterized mutations in ATC is listed in Table 1.
Table 1.
Common mutations in ATC
Activation of MAPK signaling has been demonstrated in well-differentiated as well as ATC tumors, facilitated by a number of mechanisms. Several RET-PTC translocations have been characterized in papillary thyroid carcinoma (PTC), and in particular RET/PTC3 is often correlated with advanced disease and more aggressive phenotype6. Activating point mutations in RET are frequently observed in medullary thyroid carcinoma (MTC)7. Oncogenic RET signaling results in increased tyrosine kinase activity of this receptor and subsequent stimulation of MAPK and PI3K/AKT pathways 7. ATC lesions arising from WDTC may also demonstrate RET hyperactivation2-5. Mutations in BRAF are also observed in both PTC and ATC, leading to activation of its downstream effector MEK.
The small GTPase RAS, also mutated in follicular thyroid carcinoma (FTC) and ATC, is known to govern a variety of pro-survival pathways including both MAPK and PI3K/AKT. Dysregulation of PI3K/AKT has also been demonstrated in all thyroid malignancies via genetic silencing or inactivating mutations in the tumor suppressor PTEN or activating point mutations in PI3KCA2-5. AKT activation conveys tumor cell survival and growth through a multitude of molecular mechanisms including mTOR signaling.
TP53 gene alterations are frequently observed in poorly differentiated thyroid carcinomas and ATC 8, 9, likely leading to disruption of cell cycle checkpoints and DNA repair machinery, resulting in tumor cell tolerance of accumulating genetic instabilities. Loss of molecular mechanisms governing cell polarity such as mutations in CTNNB1 are frequently observed in ATC2, and likely facilitate tumor cell dissemination and invasion.
Finally, vascular homeostasis is lost in ATC though tumor-associated overexpression of VEGF, PDGFR and other angiogenic factors2, 5. This neovascularization results in the formation of tortuous vessels, abnormal perivascular coverage, and irregular extracellular matrix deposition. Not only does this contribute to tumor cell growth, but also enhances intravassation, metastasis, and drug resistance due to increased oncotic pressure resulting from leaky vasculature10.
2. Current targeted therapies and their efficacy in ATC
Several targeted therapies are currently under clinical investigation to evaluate their efficacy against ATC, and are largely based on the genetic profile of ATC tumors. These include tyrosine kinase blockade of RET, VEGFR1-2, PDGFRA, BRAF and EGFR using either small molecule or antibody mediated inhibition. Vascular disrupting agents as well as other novel compounds are also in clinical testing. A summary of agents investigated in a clinical setting are provided in Table 2.
Table 2.
Summary of Targeted Therapy in ATC
| Targeted therapy |
Trade Name |
Molecular Target | Clinical Evaluation |
No. ATC Patients |
PR | SD | PD | Median Survival (months) |
Reference |
|---|---|---|---|---|---|---|---|---|---|
| Sorafenib | Nexavar® | BRAF, VEGFR, c-KIT, RET, PDGFRA | Phase II | 2/30 | 2 | 11 | |||
| Phase II | 4/58 | 1 | 3 | 12 | |||||
| Phase II | 16 | 3.5 | 13 | ||||||
| Phase II | 20 | 2 | 5 | 3.9 | 14 | ||||
| Dabrafenib | Tafinlar® | BRAF | Phase I | 1/14 | 1 | 11.3 | 15 | ||
| Pazopanib | Votrient® | VEGFR, PDGFR, RET, c-KIT | Phase II | 15 | 3.6 | 16 | |||
| Imatinib | Gleevec® | ABL, c-KIT, PDGFR | Phase II | 11 | 6 | 17 | |||
| Axitinib | Inlyta® | VEGRF1-3, PDGFR, c-KIT | Phase II | 2/60 | 1 | 18 | |||
| Fosbretabulin | Zybrestat® | Vascular disrupting agent | Phase II | 26 | 0 | 7 | 15 | 4.7 | 19 |
| Fosbretabulin in combination with Paclitaxel/Carboplatin | Zybrestat® | Vascular disrupting agent, microtubule stabilizer, DNA crosslinker | Phase I | 80 | 5.2 | 20 | |||
| Bardoxolone methyl | NRF2 agonist, NFKB | Phase I | 4/47 | 1 | 21 | ||||
| Gefitinib | Iressa® | EGFR | Phase II | 5/27 | 1 | 22 | |||
| Efatutazone in combination with Paclitaxel | PPARƔ agonist, microtubule stabilizer | Phase I | 15 | 1 | 7 | 4 | 3.2-4.4 | 23 | |
* PR-partial response; SD- stable disease; PD-progressive disease
Unfortunately ATC is largely chemo and radiation resistant, and develops drug-resistance rapidly despite the current availability of targeted therapeutics24. Furthermore, while surgery often conveys a survival advantage in patients with ATC, it is not curative, and advanced local and distant metastasis often renders ATC prodigally unresectable2, 25, 26. For patients who can tolerate aggressive treatment, tumor debulking followed by radiation and chemotherapy is the current recommended course of therapy. It is clear that even current targeted therapies only modestly prolong patient survival, and therefore new avenues of targeted therapeutics must be explored.
3. De novo lipid biosynthesis in ATC
Recently, a renewed emphasis on cancer-associated metabolic changes has shed light on dysregulated activation of de novo fatty acid and cholesterol biosynthesis. Normal adult mammalian tissues derive the majority of lipid molecules from the diet through the blood stream either in the form of free fatty acids or lipoproteins27. De novo lipid biosynthesis is tightly regulated, predominantly occurring in the liver, intestines, adipose tissue, and lactating mammary tissues27. It was established over 60 years ago that neoplastic tissues demonstrate a higher lipogenic need, suggestive of either increased uptake from the host or accelerated biosynthesis to support tumorigenesis28. Upregulation of fatty acid transport proteins has been observed in cancer, allowing these cells to provision free fatty acids from the extracellular environment29. More recent publications highlight specific overexpression of various constituents of lipid biosynthesis in a variety of tumors as well as tumor cell sensitivity to targeted inhibition of these components, thereby demonstrating a tumor-specific requirement for lipid metabolism27, 30-33. Tumors that demonstrate a lipogenic phenotype are often correlated with disease aggressiveness, and frequently exhibit poorer patient outcomes30, 34.
Gene array analysis of ATC tissue compared to normal thyroid tissue reveals increased expression of machinery that facilitates de novo fatty acid biosynthesis. These include acetyl-CoA carboxylase alpha (ACC), fatty acid synthase (FASN), and several fatty acid elongase and desaturase enzymes including both human isoforms of stearoyl-CoA desaturase (SCD1, SCD5) 35. In addition, a variety of proteins whose roles include fatty acid uptake, transport, and metabolism are upregulated including several fatty acid binding proteins (FABP) and solute carrier family 27 (SLC27A) proteins35. Taken together, this suggests an increased need for lipid bioavailability in this malignancy.
4. Lipid metabolism and the tumorigenic phenotype
Currently it is not well understood what specific elements of lipid biosynthesis support neoplastic transformation and tumor progression, however alterations in any number of lipid metabolism enzymes, transcription, factors, etc. involved in normal cellular processes can plausibly affect cell growth, signaling, and motility. Synthesis of saturated fatty acid molecules by FASN, and subsequent modification by a variety of elongase and desaturase enzymes, including SCD, produces a variety of saturated and unsaturated fatty acids that are substrates for membrane phosphoglyceride and triacylglyceride synthesis27. Membrane phosphoglycerides are critical for membrane synthesis, influence membrane fluidity and permeability, regulate membrane protein docking, incorporation and turnover, are involved in the synthesis of prostaglandins and other inflammatory species, and are important in numerous signaling pathways that regulate cell survival, mobility, and growth27, 36.
Triacylglycerides can be used for long-term energy storage, or can be transported to the mitochondria where they are subject to β-oxidation, producing NADH, FADH2, and acetyl-CoA which can re-enter the TCA cycle27. Activation of the mevalonate pathway in cancer cells leads to the production of cholesterol. As with fatty acids, cholesterol molecules can be incorporated into lipid bilayer structures where they influence membrane fluidity, receptor clustering, and endo/exocytosis27. Cholesterol also serves as a building block for numerous steroid-based signaling molecules. In addition, the mevalonate pathway drives isoprenoid synthesis which is important for the prenylation of a wide variety of proteins. One notable example of this is the oncoprotein Ras, which is frequently activated in thyroid malignancies. Addition of lipid moieties subsequently modifies its ability to interact with and activate target proteins or substrates36.
Several malignancies, such as glioblastoma and prostate cancer, demonstrate oncogenic activation of sterol regulatory element-binding proteins (SREBP)37, 38, a transcription factor that is critical in regulating multiple facets of lipid metabolism through controlled expression of a large number of lipogenic factors involved in cholesterol and fatty acid metabolism, triacylglyceride synthesis, and plasma lipoprotein metabolism39, 40. SREBP activity is typically governed by intracellular levels of cholesterol and fatty acids39, 40. It has been recently reported that aberrant upstream signaling through receptor tyrosine kinases, AKT, and mTOR- pathways commonly altered in cancer, may promote SREBP processing and activation thereby driving de novo lipogenesis29, 37. Guo et al. demonstrated glioblastoma cell sensitivity to an inhibitor of SREBP-1 processing, resulting in decreased proliferation. In addition, this group presented anti-tumor efficacy in a preclinical model of glioblastoma using a pharmacological inhibitor of FASN, a transcriptional target of SREBP-141, strongly supporting the need for increased lipid metabolism in this cancer.
FASN over-expression and activity has been extensively studied in breast cancer, and is correlated with numerous functions that enhance tumorigenesis. These include membrane phospholipid and lipid raft synthesis, receptor tyrosine kinase activation, DNA replication, fatty acid β-oxidation, and activation of survival signaling pathways such as Akt30. Elegant work by Rysman et al, demonstrates that activation of de novo lipogenesis and resulting accumulation of saturated and mono-unsaturated lipid species (specifically the products of FASN and SCD enzymatic activity) modulate the biophysical properties of cancer cell lipid membranes by promoting their saturation, resulting in reduced sensitivity to lipid peroxidation as well as treatment with H202 and chemotherapy 42. These findings suggest that lipogenic tumors may demonstrate increased resistance to chemotherapy and are also protected against oxidative stress. Paton et al. 2010 similarly correlated SCD desaturase activity with membrane homeostasis in breast cancer cells, where targeted inhibition of SCD led to increased free cholesterol levels, decreased cholesterol esterification, and affected cholesterol efflux. Increased lipid raft formation due to membrane incorporation of free cholesterol was observed, and was predicted to affect membrane fluidity43.
Several groups have also correlated oleic acid production with increased tumor cell migration and invasiveness44, 45. Work done by Vazquez-Martin et al. found that de novo lipogenesis is activated in induced pluripotent stem cells (iPS), and that pharmacological inhibition of FASN or ACC hinders reprogramming efficiency46. This suggests a potential role for lipid biosynthesis in tumor stem cell generation and proliferation. A role for SCD activity in autophagosome formation has also been reported47. As autophagy is commonly associated with cancer cell survival and drug resistance48, this further implicates a role for lipogenesis in tumor viability and progression. A brief summary of lipid metabolism is provided in Figure 1, including upstream signaling implicated in tumor-associated lipogenesis, and biological function resultant of specific components of this pathway.
Figure 1. Summary of lipid metabolism.
A brief summary of lipid metabolism signaling including fatty acid synthesis and components of the mevalonate pathway thought to be important in the pathogenesis of cancer. Activation of Receptor Tyrosine Kinases (RTKs) such as EGFR or HER2/neu through ligand binding, receptor overexpression, or activating mutations are thought to promote a variety of intracellular signaling pathways such as PI3K/AKT. AKT activation in turn stimulates mTOR, resulting in SREBP processing and activation through blockade of LIPIN1. SREBP is a potent transcription factor that recognizes and binds to sterol regulatory elements (SRE) within the genome, inducing transcription of these target genes that include numerous factors involved in lipid metabolism and fatty acid uptake. Tumor cells also may increase lipid bioavailability through increased Free Fatty Acid (FFA) uptake via upregulation of fatty acid transport receptors and chaperones such as CD36, Solute Carrier Family 27/Fatty Acid Transporter (SLC27A/FATP), and Fatty Acid Binding Proteins (FABP). In addition, metabolic reprograming that facilitates glycolysis, a common phenomenon in tumorigenesis known as the Warburg effect, can activate de novo lipogenesis. Acetyl-CoA derived from citrate can be further processed into a variety of lipid species via the activity of various enzymes. Cholesterol and free fatty acids are important for a variety of cellular functions including membrane synthesis, inflammation, fatty acid β-oxidation, energy storage, cholesterol synthesis, protection from cellular stress, post-translational modification of proteins through prenylation, and cellular communication. Lipid metabolism enzymes that are current therapeutic targets of interest for the treatment of cancer are bold-faced. Enzymes are italicized. Abbreviations: ACL- ATP Citrate Lyase ; ACC- Acetyl-CoA Carboxylase Alpha; EGF- Epidermal Growth Factor; FGF- Fibroblast Growth Factor; FASN- Fatty Acid Synthase; FFA- Free Fatty Acid; GLUT- Glucose Transporter; HMGCR- Hydroxymethylglutaryl-CoA Reductase; IGF- Insulin-like Growth Factor; LDLR- Low Density Lipoprotein Receptor; PDH-Pyruvate Dehydrogenase; RTK- Receptor Tyrosine Kinase; SCD- Stearoyl-CoA Desaturase; TGF- Transforming Growth Factor. A small ‘p’ indicates a phosphorylation event.
5. Stearoyl-CoA desaturase as a therapeutic target in ATC
Of the lipogenic enzymes upregulated in ATC, SCD is the most significantly differentially expressed compared to normal thyroid tissues35. Additionally, protein expression of this enzyme is increased in WDTCs including PTC and FTC. SCD is a fatty acyl desaturase enzyme whose functional role is to catalyze the conversion of saturated fatty acids produced by FASN activity into mono-unsaturated fatty acids through insertion of a cis-double bond at the Δ9 position of the carbon chain49. To date, five isoforms of this enzyme have been characterized, of which two are expressed in humans- SCD1 and SCD550. SCD1-4 were identified and characterized in the mouse genome50. Each of these isoforms demonstrates differential tissue distribution and expression patterns, yet maintains conserved enzymatic function50. In ATC, therapeutic and genetic targeted inhibition of SCD enzymatic activity facilitated a robust decrease in cell proliferation and induced cell death where normal thyroid cells remained unaffected 35. Interestingly, tumor cells derived from well-differentiated lesions demonstrate resistance against SCD inhibition, and suggest a specific vulnerability in the more aggressive thyroid malignancy to this treatment.
Loss of SCD enzymatic activity in ATC correlates with an induction of the endoplasmic reticulum (ER) stress response, resulting in the activation of a variety of chaperone, degradation, and pro-apoptotic proteins. This is consistent with other research studies focusing on SCD in cancer as well as metabolic disorders. In cancer, SCD inhibition demonstrates activation of ER stress and inflammation in a variety of tumor models including colorectal adenocarcinoma51, non-small cell lung carcinoma52, and renal cell carcinoma33, resulting in tumor cell death. SCD activity has also been shown to play a key role in modulating diabetes by reducing ER stress, inflammation, and insulin resistance by converting lipotoxic substrates such as stearate and palmitate into monounsaturated fatty acid species in primary patient-derived myotube models53. Green et al. reported that expression of SCD1 and the fatty acid elongase ELOVL6 could protect pancreatic β-cells from ER stress and apoptosis induced by exposure to elevated free fatty acids- a potent risk factor for type 2 diabetes and insulin resistance in a rodent model of diabetes54. Given the numerous reports linking the loss SCD enzymatic activity with heightened ER stress and inflammation, it is plausible that these intracellular responses are culpable for increased cancer cell death as a result of tumor-specific dependency on this pathway.
Considering the reported biological diversity involving SCD activity and other constituents of de novo lipogenesis in tumor cell chemoresistance, protection from cellular stress, proliferation and migration, and energy storage and metabolism, it stands to reason that targeted inhibition of this pathway may be used successfully in combination with a variety of existing anti-cancer regimens for the treatment of ATC. When combined with the proteasome inhibitor carfilzomib, the SCD inhibitor MF-438 showed reduced tumor growth in a preclinical model of ATC 35. In addition, several recent examples of successful combinatorial therapy involving agents targeting lipid metabolism in models of cancer have been established. FASN inhibitors demonstrate good anti-tumor efficacy in cell models of prostate cancer when used in combination with thiazolidinediones (TZDs), a family of PPARγ activators55. In a preclinical model of colorectal carcinoma, the FASN inhibitor cerulenin when used in combination with oxaliplatin chemotherapy significantly decreased tumor growth when compared to monotherapy56. The SCD inhibitor A939572 when tested in a preclinical model of clear cell renal cell carcinoma combined with temsirolimus, an mTOR inhibitor, demonstrated anti-tumor synergy33.
6. Expert Opinion
The disequilibrium in the expression of lipid metabolism constituents in ATC is strongly suggestive of increased lipogenesis in these tumors. Our work and that of others support that various components of this pathway may serve as attractive therapeutic targets for the treatment of a variety of cancers. Currently, little to no investigation into the role of lipogenesis as a pro-tumorigenic mechanism specifically in ATC, or other subtypes of thyroid cancer has been done. Wojakowska et al. recently published on the application of metabolomics as a useful tool in the investigation of thyroid malignancy, summarizing notable discrepancies in the expression of various lipid constituents between normal and malignant thyroid tissue 57. Lovastatin, an inhibitor of HMG-CoA reductase (HMGCR), has been shown to demonstrate anti-tumor efficacy in ATC through targeted inhibition of mevalonate synthesis and blockade of geranylgeranylation and subsequent Rho GTPase activation58. Recently published work by our group represents one of the first comprehensive analyses of lipid metabolism profiling in ATC35. As such, there is a need for additional investigation of lipid metabolism in the context of ATC and thyroid carcinoma in order to improve our understanding of its contribution toward the initiation and progression of this disease, and define additional appropriate therapeutic targets.
Given the tumor-specific over-expression of particular lipid metabolism genes in ATC, and the strict regulation of this pathway in normal tissue, it stands to reason that targeted inhibitors may be predicted to produce discriminative therapeutic responses in diseased tissue. This presents an entirely new venue in which several candidate therapeutic targets may be explored and identified. Currently inhibitors against a variety of enzymes involved in de novo lipogenesis have been produced and are under investigation for anti-tumorigenic efficacy including ACC, ATP citrate lyase (ACL), carnitine palmitoyltransferase 1 (CPT-1), FASN, monoacylglycerol lipase (MAGL), HMGCR, SCD, and SREBP136, 59-61.
Acquired resistance to monotherapeutic applications remains a challenge given the heterogeneous nature of ATC and other aggressive malignancies. While limited efficacy was observed in a preclinical model of ATC treated solely with an SCD inhibitor, a significant combinatorial effect with a proteasome inhibitor was observed35. Thus, while various inhibitors of lipid metabolism should be individually investigated for anti-tumor efficacy, combinatorial applications with complementary regimens including radiation, chemotherapy, and other targeted agents should also be explored.
One of the most notable impediments in the progression of lipid metabolism research in cancer is the use of rodents as preclinical models. While generally observed as convenient tools to evaluate therapeutic efficacy, the vast discrepancies in genetic variation, tissue distribution, expression, and regulatory mechanisms governing lipid metabolism limit their translational relevance in humans 62. For example, murine models express four isoforms of SCD, only one being homologous to human SCD50. Therefore, adverse events observed in rodent models in response to SCD inhibitors may not necessarily be predictive of toxicity profiles in humans. Alternative preclinical models that more accurately reflect lipid metabolism observed in humans may be necessary in order to enhance the prognostic value of targeting this pathway not only in cancer, but other diseases as well.
Article Highlights.
This article provides a brief review of the known oncogenic factors in ATC.
This article summarizes the current targeted therapies for ATC and results of clinical evaluation.
A brief review of cancer-associated lipid metabolism is provided, and pro-tumorigenic mechanisms of aberrant lipogenesis are highlighted.
Constituents of lipid metabolism currently under investigation as novel therapeutic targets are discussed.
Abnormal expression of de novo lipogenesis in ATC is examined.
Targeting SCD as well as other metabolic enzymes as a treatment for this variant of thyroid cancer, either alone or in combination with other therapies, is discussed
Acknowledgments
CA von Roemeling and JA Copland were supported by the National Cancer Institute (NCI) of the National Institutes of Health (NIH) under award number R01CA136665; the Florida Department of Health under Bankhead-Coley Cancer Research Program grant number FL09B202.
Footnotes
Financial and competing interests disclosure
The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
Contributor Information
Christina A. von Roemeling, The Mayo Clinic Graduate School, Rochester, MN 55905. Vonroemeling.christina@mayo.edu, Phone: 904-953-6045.
John A. Copland, The Department of Cancer Biology, Mayo Clinic Jacksonville, Jacksonville, FL 32224.
References
Reference annotations
* Of interest
** Of considerable interest
- 1.Champa D, Di Cristofano A. Modeling anaplastic thyroid carcinoma in the mouse. Horm Cancer. 2015 Feb;6(1):37–44. doi: 10.1007/s12672-014-0208-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Smallridge RC, Copland JA. Anaplastic thyroid carcinoma: pathogenesis and emerging therapies. Clin Oncol (R Coll Radiol) 2010 Aug;22(6):486–97. doi: 10.1016/j.clon.2010.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rusinek D, Szpak-Ulczok S, Jarzab B. Gene expression profile of human thyroid cancer in relation to its mutational status. J Mol Endocrinol. 2011 Dec;47(3):R91–103. doi: 10.1530/JME-11-0023. [DOI] [PubMed] [Google Scholar]
- 4.Guerra A, Di Crescenzo V, Garzi A, Cinelli M, Carlomagno C, Tonacchera M, et al. Genetic mutations in the treatment of anaplastic thyroid cancer: a systematic review. BMC Surg. 2013;13(Suppl 2):S44. doi: 10.1186/1471-2482-13-S2-S44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ragazzi M, Ciarrocchi A, Sancisi V, Gandolfi G, Bisagni A, Piana S. Update on anaplastic thyroid carcinoma: morphological, molecular, and genetic features of the most aggressive thyroid cancer. Int J Endocrinol. 2014;2014:790834. doi: 10.1155/2014/790834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Romei C, Elisei R. RET/PTC Translocations and Clinico-Pathological Features in Human Papillary Thyroid Carcinoma. Front Endocrinol (Lausanne) 2012;3:54. doi: 10.3389/fendo.2012.00054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mulligan LM. RET revisited: expanding the oncogenic portfolio. Nat Rev Cancer. 2014 Mar;14(3):173–86. doi: 10.1038/nrc3680. [DOI] [PubMed] [Google Scholar]
- 8.Patel KN, Shaha AR. Poorly differentiated thyroid cancer. Curr Opin Otolaryngol Head Neck Surg. 2014 Apr;22(2):121–6. doi: 10.1097/MOO.0000000000000037. [DOI] [PubMed] [Google Scholar]
- 9.Perri F, Pezzullo L, Chiofalo MG, Lastoria S, Di Gennaro F, Scarpati GD, et al. Targeted therapy: A new hope for thyroid carcinomas. Crit Rev Oncol Hematol. 2015 Apr;94(1):55–63. doi: 10.1016/j.critrevonc.2014.10.012. [DOI] [PubMed] [Google Scholar]
- 10.Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005 Jan 7;307(5706):58–62. doi: 10.1126/science.1104819. [DOI] [PubMed] [Google Scholar]
- 11.Gupta-Abramson V, Troxel AB, Nellore A, Puttaswamy K, Redlinger M, Ransone K, et al. Phase II trial of sorafenib in advanced thyroid cancer. J Clin Oncol. 2008 Oct 10;26(29):4714–9. doi: 10.1200/JCO.2008.16.3279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kloos RT, Ringel MD, Knopp MV, Hall NC, King M, Stevens R, et al. Phase II trial of sorafenib in metastatic thyroid cancer. J Clin Oncol. 2009 Apr 1;27(10):1675–84. doi: 10.1200/JCO.2008.18.2717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Nagaiah GF,P, Wasman JK, et al. Phase II trial of sorafenib (bay 43-9006) in patients wtih advanced anaplastic carcinoma of the thyroid (ATC). J Clin Oncol. 2009;27:15s. [Google Scholar]
- 14.Savvides P, Nagaiah G, Lavertu P, Fu P, Wright JJ, Chapman R, et al. Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid. Thyroid. 2013 May;23(5):600–4. doi: 10.1089/thy.2012.0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Falchook GS, Millward M, Hong D, Naing A, Piha-Paul S, Waguespack SG, et al. BRAF inhibitor dabrafenib in patients with metastatic BRAF-mutant thyroid cancer. Thyroid. 2015 Jan;25(1):71–7. doi: 10.1089/thy.2014.0123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bible KC, Suman VJ, Menefee ME, Smallridge RC, Molina JR, Maples WJ, et al. A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. J Clin Endocrinol Metab. 2012 Sep;97(9):3179–84. doi: 10.1210/jc.2012-1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ha HT, Lee JS, Urba S, Koenig RJ, Sisson J, Giordano T, et al. A phase II study of imatinib in patients with advanced anaplastic thyroid cancer. Thyroid. 2010 Sep;20(9):975–80. doi: 10.1089/thy.2010.0057. [DOI] [PubMed] [Google Scholar]
- 18.Cohen EE, Rosen LS, Vokes EE, Kies MS, Forastiere AA, Worden FP, et al. Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: results from a phase II study. J Clin Oncol. 2008 Oct 10;26(29):4708–13. doi: 10.1200/JCO.2007.15.9566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mooney CJ, Nagaiah G, Fu P, Wasman JK, Cooney MM, Savvides PS, et al. A phase II trial of fosbretabulin in advanced anaplastic thyroid carcinoma and correlation of baseline serum-soluble intracellular adhesion molecule-1 with outcome. Thyroid. 2009 Mar;19(3):233–40. doi: 10.1089/thy.2008.0321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sosa JA, Elisei R, Jarzab B, Balkissoon J, Lu SP, Bal C, et al. Randomized safety and efficacy study of fosbretabulin with paclitaxel/carboplatin against anaplastic thyroid carcinoma. Thyroid. 2014 Feb;24(2):232–40. doi: 10.1089/thy.2013.0078. [DOI] [PubMed] [Google Scholar]
- 21.Hong DS, Kurzrock R, Supko JG, He X, Naing A, Wheler J, et al. A phase I first-in-human trial of bardoxolone methyl in patients with advanced solid tumors and lymphomas. Clin Cancer Res. 2012 Jun 15;18(12):3396–406. doi: 10.1158/1078-0432.CCR-11-2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pennell NA, Daniels GH, Haddad RI, Ross DS, Evans T, Wirth LJ, et al. A phase II study of gefitinib in patients with advanced thyroid cancer. Thyroid. 2008 Mar;18(3):317–23. doi: 10.1089/thy.2007.0120. [DOI] [PubMed] [Google Scholar]
- 23.Smallridge RC, Copland JA, Brose MS, Wadsworth JT, Houvras Y, Menefee ME, et al. Efatutazone, an oral PPAR-gamma agonist, in combination with paclitaxel in anaplastic thyroid cancer: results of a multicenter phase 1 trial. J Clin Endocrinol Metab. 2013 Jun;98(6):2392–400. doi: 10.1210/jc.2013-1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Perri F, Lorenzo GD, Scarpati GD, Buonerba C. Anaplastic thyroid carcinoma: A comprehensive review of current and future therapeutic options. World J Clin Oncol. 2011 Mar 10;2(3):150–7. doi: 10.5306/wjco.v2.i3.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Brown RF, Ducic Y. Aggressive surgical resection of anaplastic thyroid carcinoma may provide long-term survival in selected patients. Otolaryngol Head Neck Surg. 2013 Apr;148(4):564–71. doi: 10.1177/0194599813477364. [DOI] [PubMed] [Google Scholar]
- 26.Nagaiah G, Hossain A, Mooney CJ, Parmentier J, Remick SC. Anaplastic thyroid cancer: a review of epidemiology, pathogenesis, and treatment. J Oncol. 2011;2011:542358. doi: 10.1155/2011/542358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27*.Santos CR, Schulze A. Lipid metabolism in cancer. FEBS J. 2012 Aug;279(15):2610–23. doi: 10.1111/j.1742-4658.2012.08644.x. [Provides an in depth look at oncogenic signaling pathways responsible for promoting tumor-associated lipogensis. Described are pro-tumorogenic biological responses as a result of aberrant lipid metabolism.] [DOI] [PubMed] [Google Scholar]
- 28.Medes G, Thomas A, Weinhouse S. Metabolism of neoplastic tissue. IV. A study of lipid synthesis in neoplastic tissue slices in vitro. Cancer Res. 1953 Jan;13(1):27–9. [PubMed] [Google Scholar]
- 29.Currie E, Schulze A, Zechner R, Walther TC, Farese RV., Jr Cellular fatty acid metabolism and cancer. Cell Metab. 2013 Aug 6;18(2):153–61. doi: 10.1016/j.cmet.2013.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30**.Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer. 2007 Oct;7(10):763–77. doi: 10.1038/nrc2222. [Discusses the role of FASN, one of the first candidate targets in tumor-associated lipogenesis, in tumor initiation and progression. Reviewed within are oncogenic signaling mechanisms correlated with FASN activity.] [DOI] [PubMed] [Google Scholar]
- 31.Abramson HN. The lipogenesis pathway as a cancer target. J Med Chem. 2011 Aug 25;54(16):5615–38. doi: 10.1021/jm2005805. [DOI] [PubMed] [Google Scholar]
- 32.Ackerman D, Simon MC. Hypoxia, lipids, and cancer: surviving the harsh tumor microenvironment. Trends Cell Biol. 2014 Aug;24(8):472–8. doi: 10.1016/j.tcb.2014.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.von Roemeling CA, Marlow LA, Wei JJ, Cooper SJ, Caulfield TR, Wu K, et al. Stearoyl-CoA desaturase 1 is a novel molecular therapeutic target for clear cell renal cell carcinoma. Clin Cancer Res. 2013 May 1;19(9):2368–80. doi: 10.1158/1078-0432.CCR-12-3249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34**.Igal RA. Stearoyl-CoA desaturase-1: a novel key player in the mechanisms of cell proliferation, programmed cell death and transformation to cancer. Carcinogenesis. 2010 Sep;31(9):1509–15. doi: 10.1093/carcin/bgq131. [Provides an in depth review of SCD1 as a key player in cellular transformation and carcinogenesis, and prospects as a therapeutic target.] [DOI] [PubMed] [Google Scholar]
- 35.von Roemeling CA, Marlow LA, Pinkerton AB, Crist A, Miller J, Tun HW, et al. Aberrant Lipid Metabolism in Anaplastic Thyroid Carcinoma Reveals Stearoyl CoA Desaturase 1 as a Novel Therapeutic Target. J Clin Endocrinol Metab. 2015 May;100(5):E697–709. doi: 10.1210/jc.2014-2764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zhang F, Du G. Dysregulated lipid metabolism in cancer. World J Biol Chem. 2012 Aug 26;3(8):167–74. doi: 10.4331/wjbc.v3.i8.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Guo D, Bell EH, Mischel P, Chakravarti A. Targeting SREBP-1-driven lipid metabolism to treat cancer. Curr Pharm Des. 2014;20(15):2619–26. doi: 10.2174/13816128113199990486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ettinger SL, Sobel R, Whitmore TG, Akbari M, Bradley DR, Gleave ME, et al. Dysregulation of sterol response element-binding proteins and downstream effectors in prostate cancer during progression to androgen independence. Cancer Res. 2004 Mar 15;64(6):2212–21. doi: 10.1158/0008-5472.can-2148-2. [DOI] [PubMed] [Google Scholar]
- 39.Shimano H. Sterol regulatory element-binding proteins (SREBPs): transcriptional regulators of lipid synthetic genes. Prog Lipid Res. 2001 Nov;40(6):439–52. doi: 10.1016/s0163-7827(01)00010-8. [DOI] [PubMed] [Google Scholar]
- 40.Peterson TR, Sengupta SS, Harris TE, Carmack AE, Kang SA, Balderas E, et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell. 2011 Aug 5;146(3):408–20. doi: 10.1016/j.cell.2011.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Guo D, Prins RM, Dang J, Kuga D, Iwanami A, Soto H, et al. EGFR signaling through an Akt-SREBP-1-dependent, rapamycin-resistant pathway sensitizes glioblastomas to antilipogenic therapy. Sci Signal. 2009;2(101):ra82. doi: 10.1126/scisignal.2000446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Rysman E, Brusselmans K, Scheys K, Timmermans L, Derua R, Munck S, et al. De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation. Cancer Res. 2010 Oct 15;70(20):8117–26. doi: 10.1158/0008-5472.CAN-09-3871. [DOI] [PubMed] [Google Scholar]
- 43.Paton CM, Ntambi JM. Loss of stearoyl-CoA desaturase activity leads to free cholesterol synthesis through increased Xbp-1 splicing. Am J Physiol Endocrinol Metab. 2010 Dec;299(6):E1066–75. doi: 10.1152/ajpendo.00388.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Noto A, Raffa S, De Vitis C, Roscilli G, Malpicci D, Coluccia P, et al. Stearoyl-CoA desaturase-1 is a key factor for lung cancer-initiating cells. Cell Death Dis. 2013;4:e947. doi: 10.1038/cddis.2013.444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Navarro-Tito N, Soto-Guzman A, Castro-Sanchez L, Martinez-Orozco R, Salazar EP. Oleic acid promotes migration on MDA-MB-231 breast cancer cells through an arachidonic acid-dependent pathway. Int J Biochem Cell Biol. 2010 Feb;42(2):306–17. doi: 10.1016/j.biocel.2009.11.010. [DOI] [PubMed] [Google Scholar]
- 46.Vazquez-Martin A, Corominas-Faja B, Cufi S, Vellon L, Oliveras-Ferraros C, Menendez OJ, et al. The mitochondrial H(+)-ATP synthase and the lipogenic switch: new core components of metabolic reprogramming in induced pluripotent stem (iPS) cells. Cell Cycle. 2013 Jan 15;12(2):207–18. doi: 10.4161/cc.23352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ogasawara Y, Itakura E, Kono N, Mizushima N, Arai H, Nara A, et al. Stearoyl-CoA desaturase 1 activity is required for autophagosome formation. J Biol Chem. 2014 Aug 22;289(34):23938–50. doi: 10.1074/jbc.M114.591065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yang ZJ, Chee CE, Huang S, Sinicrope FA. The role of autophagy in cancer: therapeutic implications. Mol Cancer Ther. 2011 Sep;10(9):1533–41. doi: 10.1158/1535-7163.MCT-11-0047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ntambi JM. Regulation of stearoyl-CoA desaturase by polyunsaturated fatty acids and cholesterol. J Lipid Res. 1999 Sep;40(9):1549–58. [PubMed] [Google Scholar]
- 50.Liu X, Strable MS, Ntambi JM. Stearoyl CoA desaturase 1: role in cellular inflammation and stress. Adv Nutr. 2011 Jan;2(1):15–22. doi: 10.3945/an.110.000125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Minville-Walz M, Pierre AS, Pichon L, Bellenger S, Fevre C, Bellenger J, et al. Inhibition of stearoyl-CoA desaturase 1 expression induces CHOP-dependent cell death in human cancer cells. PLoS One. 2010;5(12):e14363. doi: 10.1371/journal.pone.0014363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Roongta UV, Pabalan JG, Wang X, Ryseck RP, Fargnoli J, Henley BJ, et al. Cancer cell dependence on unsaturated fatty acids implicates stearoyl-CoA desaturase as a target for cancer therapy. Mol Cancer Res. 2011 Nov;9(11):1551–61. doi: 10.1158/1541-7786.MCR-11-0126. [DOI] [PubMed] [Google Scholar]
- 53.Peter A, Weigert C, Staiger H, Machicao F, Schick F, Machann J, et al. Individual stearoyl-coa desaturase 1 expression modulates endoplasmic reticulum stress and inflammation in human myotubes and is associated with skeletal muscle lipid storage and insulin sensitivity in vivo. Diabetes. 2009 Aug;58(8):1757–65. doi: 10.2337/db09-0188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Green CD, Olson LK. Modulation of palmitate-induced endoplasmic reticulum stress and apoptosis in pancreatic beta-cells by stearoyl-CoA desaturase and Elovl6. Am J Physiol Endocrinol Metab. 2011 Apr;300(4):E640–9. doi: 10.1152/ajpendo.00544.2010. [DOI] [PubMed] [Google Scholar]
- 55.Mansour M, Schwartz D, Judd R, Akingbemi B, Braden T, Morrison E, et al. Thiazolidinediones/PPARgamma agonists and fatty acid synthase inhibitors as an experimental combination therapy for prostate cancer. Int J Oncol. 2011 Feb;38(2):537–46. doi: 10.3892/ijo.2010.877. [DOI] [PubMed] [Google Scholar]
- 56.Shiragami R, Murata S, Kosugi C, Tezuka T, Yamazaki M, Hirano A, et al. Enhanced antitumor activity of cerulenin combined with oxaliplatin in human colon cancer cells. Int J Oncol. 2013 Aug;43(2):431–8. doi: 10.3892/ijo.2013.1978. [DOI] [PubMed] [Google Scholar]
- 57.Wojakowska A, Chekan M, Widlak P, Pietrowska M. Application of metabolomics in thyroid cancer research. Int J Endocrinol. 2015;2015:258763. doi: 10.1155/2015/258763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhong WB, Liang YC, Wang CY, Chang TC, Lee WS. Lovastatin suppresses invasiveness of anaplastic thyroid cancer cells by inhibiting Rho geranylgeranylation and RhoA/ROCK signaling. Endocr Relat Cancer. 2005 Sep;12(3):615–29. doi: 10.1677/erc.1.01012. [DOI] [PubMed] [Google Scholar]
- 59*.Zhao Y, Butler EB, Tan M. Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis. 2013;4:e532. doi: 10.1038/cddis.2013.60. [comprehensive review of aberrant metabolism in cancer including glycolysis, fatty acid synthesis, glutaminolysis and their potential for targeted inhibition as anti-tumor strategies.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Beckers A, Organe S, Timmermans L, Scheys K, Peeters A, Brusselmans K, et al. Chemical inhibition of acetyl-CoA carboxylase induces growth arrest and cytotoxicity selectively in cancer cells. Cancer Res. 2007 Sep 1;67(17):8180–7. doi: 10.1158/0008-5472.CAN-07-0389. [DOI] [PubMed] [Google Scholar]
- 61.Flavin R, Peluso S, Nguyen PL, Loda M. Fatty acid synthase as a potential therapeutic target in cancer. Future Oncol. 2010 Apr;6(4):551–62. doi: 10.2217/fon.10.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Bergen WG, Mersmann HJ. Comparative aspects of lipid metabolism: impact on contemporary research and use of animal models. J Nutr. 2005 Nov;135(11):2499–502. doi: 10.1093/jn/135.11.2499. [DOI] [PubMed] [Google Scholar]

