Skip to main content
Erschienen in: Journal of Translational Medicine 1/2023

Open Access 01.12.2023 | Review

The potential role of reprogrammed glucose metabolism: an emerging actionable codependent target in thyroid cancer

verfasst von: Sai-li Duan, Min Wu, Zhe-Jia Zhang, Shi Chang

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2023

Abstract

Although the incidence of thyroid cancer is increasing year by year, most patients, especially those with differentiated thyroid cancer, can usually be cured with surgery, radioactive iodine, and thyroid-stimulating hormone suppression. However, treatment options for patients with poorly differentiated thyroid cancers or radioiodine-refractory thyroid cancer have historically been limited. Altered energy metabolism is one of the hallmarks of cancer and a well-documented feature in thyroid cancer. In a hypoxic environment with extreme nutrient deficiencies resulting from uncontrolled growth, thyroid cancer cells utilize “metabolic reprogramming” to satisfy their energy demand and support malignant behaviors such as metastasis. This review summarizes past and recent advances in our understanding of the reprogramming of glucose metabolism in thyroid cancer cells, which we expect will yield new therapeutic approaches for patients with special pathological types of thyroid cancer by targeting reprogrammed glucose metabolism.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
TC
Thyroid cancer
DTC
Differentiated thyroid cancers
PTC
Papillary thyroid cancer
FTC
Follicular thyroid cancer
MTC
Medullary thyroid cancer
ATC
Anaplastic thyroid cancer
RAIR
Radioiodine refractory
ATP
Adenosine triphosphate
PI3K
Phosphatidylinositol 3-kinase
HIF-1α
Hypoxia-inducible factor-1 alpha
TME
Tumor microenvironment
PK
Pyruvate kinase
GLUT1
Glucose transporter 1
TCA cycle
Tricarboxylic acid cycle
LA
Lactic acid
PEP
Phosphoenolpyruvate
GLS
Glutaminase
GDH
Glucose dehydrogenase
PFK
Phosphofructokinase
ROS
Reactive oxygen species
LDHA
Lactate-dehydrogenase A
PD-1
Programmed cell death protein 1
PGC-1
Peroxisome proliferator-activated receptor γ coactivator-1
MAPK
Mitogen-activated protein kinase
MCTs
Proton-linked monocarboxylate transporters
ACF
Acriflavine
CO2
Carbon dioxide
PPP
Pentose phosphate pathway
ECM
Extracellular matrix
BM
Basement membrane
EMT
Epithelial-mesenchymal transition
MMP
Matrix metalloproteinases
PD-L1
Programmed death-ligand 1
FNAB
Fine-needle aspiration biopsy
CT
Computed tomography
MRI
Magnetic resonance imaging
FDG-PET
Fluoro-2-deoxy-D-glucose positron emission tomography
NIS
Na + /I- symporter
ATA
American Translators Association
Tg
Thyroglobulin
DWI
Diffusion-weighted imaging
TKIs
Tyrosine kinase inhibitors
VEGFR2
Vascular endothelial growth factor receptor 2
VEGF
Vascular endothelial growth factor
PDGFR
Platelet-derived growth factor receptors
STAT3
Signal transducer and activator of transcription 3
NAD
Nicotinamide adenine dinucleotide
GAPDH
Glyceraldehyde 3-phosphate dehydrogenase
IGFR
Insulin-like growth factor receptor
TSH
Thyroid stimulating hormone

Introduction

Thyroid cancer (TC) is an endocrine system tumor originating from follicular thyroid cells and parafollicular C cells, and its incidence rate is rising worldwide [1, 2]. Most thyroid malignancies (> 95%) are differentiated thyroid cancers (DTCs), which include papillary thyroid cancer (PTC) and follicular thyroid cancer (FTC) [3]. Lymph node metastasis is a common clinical feature of DTC, and a considerable portion of DTC is prone to early cervical lymph node metastases (approximately 20–70%), which is an important factor affecting the prognosis of DTC patients [4, 5]. Despite the widespread use of multimodality treatment (ie, surgery, chemotherapy, and radiotherapy), survival rates have not improved much over the past few decades, suggesting that new treatment options should be explored [68]. Therefore, we recommend seeking other effective treatments for TC. Probing the molecular mechanism of disease progress, and developing new targeted drugs remain the focus of TC research [9]. Initiation and progression of TC involves multiple genetic and epigenetic alterations, among which common mutations found in TC are point mutations in the BRAF and RAS genes as well as RET/PTC and PAX8/PPARγ chromosomal rearrangements [10, 11]. These alterations often lead to anomalies in the proliferation, differentiation and metabolism patterns of thyroid follicular cells, acting synergistically to amplify their effects on thyroid tumor development. These mutations are crucial for the abnormal activation of the MAPK and PI3K-AKT signaling pathways, which primarily regulate cell proliferation and differentiation, but also directly regulate the activity of oxidative phosphorylation, cellular glucose uptake and aerobic glycolytic processes [12, 13]. Cancer cells frequently undergo a reorganization of metabolism to promote growth, survival, proliferation and long-term maintenance [14]. For some rapidly proliferating cells and tumors, cells use glycolysis to provide cancer cells with adenosine triphosphate (ATP), nucleotides, lipids, and amino acids needed for their growth, so that even under aerobic conditions, the glucose uptake rate is significantly increased and lactate is produced, a phenomenon known as the Warburg effect [15]. This metabolic change occurs in tumors, with marked differences in glucose use between cancer cells and normal cells [16]. However, the mechanism of the Warburg effect in TC has not yet been fully elucidated. Glucose metabolic reprogramming is a primary mode of energy production in TC and has been shown to be closely associated with tumorigenesis [15]. Importantly, these metabolic adaptations appear to be responsive not only to the genotype of the tumor, but also to the biochemical microenvironment [17]. Many studies have demonstrated that glycolysis is involved in the activation of oncogenes, such as phosphatidylinositol 3-kinase (PI3K) and hypoxia-inducible factor-1 alpha (HIF-1α) in the tumor microenvironment (TME), and acts as an energetic source for cancer cells [18]. The unique hypoxic and nutrient-deficient microenvironment further leads TC cells to utilize glucose in a hypoxic manner [15].
Here, our review mainly focuses on the interactions of glycolysis in the development of TC cells. As we understand the role of glycolysis in the growth, proliferation and metastasis of TC cells, we are able to put forward suggestions for better treatment of TC indications. At the same time, we describe the factors that affect the early detection of TC to find treatment methods that can achieve better clinical results. By understanding the process of glycolysis and its relationship with tumor cells, we can further consider the targeted treatment of TC based on the glycolysis pathway in combination with clinical treatment and whether TC can be diagnosed through the relevant factors of the glycolysis process, so as to gain insight for cancer treatment.

Insights from glycolysis in thyroid cancer

Cancer cells are well known for a series of patterns including constant proliferative signaling, growth suppressor’s avoidance, resistance to cell death, replicative immortality, high angiogenesis, reprogrammed energy metabolism, immune-mediated destruction, invasion, and metastasis, by which they can surpass normal cells’ capacity, occupy normal tissues, and even invade into surrounding or even distant area. These characteristics are largely supported by the reprogrammed energy metabolisms, which provide sufficient and instant material for cancer cells’ energy consumption and superfluous anabolism [19]. In normal conditions, cells are dependent on glycolysis rather than oxygen-consuming mitochondrial metabolism for energy supply facing short of oxygen. However, cancer cells prefer glycolysis even when oxygen is on the scene, a phenomenon first observed by Otto Warburg [20]. Such preference is shown because even though glycolysis produce less ATP per molecule of glucose, it can yield energy at a much higher rate [21]. Hence, it satisfies the high demand of cancer cells and becomes the central pathway glucose metabolism. The cancer gene mutations together with altered glycolysis, as well, promote the branches of glucose metabolism such as pentose phosphate pathway (PPP) partly because of the upregulated flux of glucose entering the PPP branch. Besides the dysregulation of glucose metabolism, other metabolism pathways also undergone great change partly ascribe to the upstream glucose metabolism alteration and partly due to the requirements of biosynthesis of biomass, such as nucleotides, amino acids and lipids [22]. Aberrant lipid metabolism, amino acids metabolism, mitochondrial biogenesis, and other bioenergetic metabolism pathways have been gradually uncovered, showing the thorough reformation of metabolism in cancer cells [23, 24].

Differences in glycolysis between cancer and normal cells

There are three main differences in the glycolysis process in TC cells compared with normal cells (see Fig. 1), such as glucose transports, pyruvate kinases, and lactic acid metabolism. In terms of glucose transport, TC has a high demand for glucose, so it overexpresses glucose transporters to transport a large amount of glucose through the membrane. In this process, controlling the expression of pyruvate kinase (PK) will block the final step of glycolysis, leading to the accumulation of many early intermediate metabolites in tumor cells. Otherwise, enhanced glycolysis and reduced oxygen consumption in cancer cells would lead to the expansion of lactate.

Glucose transport

Excessive proliferation is one of the main differences between cancer and normal cells. Therefore, a common characteristic of metabolic changes in tumor cells is increased glucose uptake [25], which can also be observed when mitochondrial functions are complete [2628]. To meet the large nutritional demands in the course of cell proliferation, tumor cells adopt a very uneconomic way of glucose metabolism to ensure that a large amount of glucose enters the cells for decomposition [29]. TC cells usually exhibit a state of hypoxia, which prevents the cells from performing sufficient glycolysis and providing sufficient ATP [30]. However, tumor cells are well adapted to this hypoxic environment [31] due to glycolysis. This behavior has been observed in all sorts of tumors [32]. A defining feature of TC cells is their ability to absorb large amounts of glucose compared to normal thyroid tissues. The upregulation of glucose transporters (GLUTs) has been reported to be an indicator of aggressiveness and loss of tumor differentiation in TC [33]. In most cases, TC cells tend to exhibit GLUTs overexpression, particularly the hypoxic-reactive GLUT1 and GLUT3 proteins [34]. The primary cellular function of the GLUT is to facilitate the entry of glucose molecules into cells [35]. Among GLUTs, GLUT1 is the most frequent isoform in many cancers, such as lung cancer, colorectal cancer, prostate cancer, and hepatocellular carcinoma [3639]. In several cancers, overexpression of GLUT1 is related to invasion and poor survival [40, 41], while increased GLUT1 expression improves glycolysis [42]. Previous studies have demonstrated that the translocation of GLUT1 to the cancer cell membrane is a factor limiting the rate of cellular energy generation [42]. The high expression of GLUT1 in TC is positively correlated with the proliferation index, which is equivalent to malignant characteristics [43]. In particular, overexpression of GLUT1 on cell membranes correlates perfectly with the rate of cell de-differentiation and greater biological aggressiveness of TC [44]. Greater GLUT1 expression can often be detected by immunostaining in TC, but not in benign nodules or normal thyroids [45]. This suggests that GLUT1, as a rate-limiting step in glucose metabolism in cancer cells and a modulator of glucose uptake pathways, is a promising target for the development of anti-cancer strategies.

Pyruvate kinase and pyruvate carboxylase

TC cells consume more glucose compared to normal cells [46], limiting the final step of the glycolysis pathway through their negative feedback mechanism and thus, leading to the accumulation of many early intermediate metabolites in tumor cells [47]. Even in a tumor microenvironment with normal oxygen levels, TC increases glucose absorption, metabolizes glucose to acrylic acid, and then converts the product to lactic acid (LA) rather than allowing it’s to enter the TCA cycle [48]. Pyruvate kinase (PK) facilitates the last step of glycolysis, the exchange of phosphoenolpyruvate (PEP) with pyruvate and is involved in the TCA cycle. The intersection between anabolic and catabolic pathways is primarily conducted by PK [49], mainly PKM1 and PKM2. PKM1 regulates the transport of pyruvate from the cytoplasm to mitochondria, while PKM2 regulates the decomposition of pyruvate to LA in the cytoplasm of tumor cells [50]. Compared with PKM1, the PKM2 isoform has a low catalytic enzyme efficiency, leading to the accumulation of glycolytic intermediates, and is involved in other biochemical synthesis pathways [5153]. During PKM1 activation, anabolic synthesis (or branching pathways in glycolysis) is promoted [54, 55], while phosphoenolpyruvate (PEP) is converted to pyruvate due to PKM2 activation, to produce ATP molecules [49, 56]. The activity of the PKM2 tetramer promotes the complete oxidative decomposition of glucose into ATP through oxidative phosphorylation, while the activity of its dimer promotes the glycolysis [57]. Some studies have suggested that PKM2 is related to the poor prognosis and has been identified as a prognostic marker in many cancers [58, 59]. One study has stated that PKM2 is involved in the progression of TC [60]. PKM2 is significantly overexpressed in PTC, especially in cases harboring BRAF mutations, and its overexpression is closely related to advanced tumor stage and lymph node metastasis [60]; meanwhile, PKM2 knockdown significantly inhibits PTC cell growth, lactic acid and ATP production, and glucose consumption [61]. Additionally, the activation or up-regulation of PKM2 could activate multiple cancer-related pathways such as ERK signaling and STAT3 signaling [62, 63]. Therefore, inhibition of PKM2 may be potential to inhibit glycolysis and thus the proliferation of tumor cells. Moreover, pyruvate carboxylase (PC), a key enzyme at the intersection of glycolysis and the TCA cycle in TC cells, plays an important role in replenishment [64]. It is reported that PC is strongly involved in the tumor aggressiveness of TC via its stimulation of fatty acid synthesis [65]. Hence, PC restraint can significantly reduce TC cell proliferation [65], suggesting that it may be possible to detect the expression of PC in living tissues to reflect the invasive behavior of tumors and provide valuable information for clinical diagnosis and treatment of TC.

Lactic acid metabolism

As a key energetic source, a glucose precursor, and a signal molecule, LA plays a vital role in shuttling between cells in vivo [66]. Previous studies have shown that glycolysis plays a role in cell signal transduction [67, 68], promoting proliferation, invasion, and drug resistance in cancer cells [69]. Aberrant glycolysis in TC involves increased LA production and accumulation, which further promotes pH conditions conducive to growth and invasion [70]. Lactate dehydrogenase (LDH), which includes two isoforms (LDHA and LDHB), plays a decisive role in LA production. LDHA is responsible for converting PA into LA and NAD, whereas LDHB converts LA into PA and promotes oxidative metabolism [66]. The increase of LDH activity leads to tumor immune evasion via inhibiting the function of immune cells [71]. For instance, LDHA-associated LA accumulation in melanoma has been shown to inhibit tumor monitoring by T and NK cells [71]. LDHA can increase acetylation and transcription of interferon-γ (IFNG) to promote T cell effector functions, thereby highlighting the key role of LDH in inflammation [72]. Changes in LDHB expression are often associated with early metabolic adaptation [73]. LDHB-mediated LA use supports autophagy to maintain metabolic health and cancer cells growth [74], indicating that the production and use of LA may be involved in the metabolic adaptation of cancer cells to support the development of metastasis. For example, overexpression of LDHB significantly inhibits the inhibitory effects of HYOU1 silencing on aerobic glycolysis, proliferation, migration, and invasion of PTC cells [75]. In glycolytic tumors, LA levels in cancer cells are increased more than 40-fold and are highly correlated with cancer invasion and low survival rate [76, 77]. Inhibition of the mitochondrial biogenesis pathway will decrease tumor survival and reduce tumor progression [78]. LA inhibits the differentiation of monocyte into dendritic cells [79], suggesting that high LA levels in the TME may hinder the formation and accumulation of dendritic cell. Meanwhile, high LA levels in the TME also inhibit LA efflux from T cells, resulting in decreased cytokine production and cytotoxic activity [80]. Inhibition of LA shuttle has been reported to significantly reduce the proliferation and glycolytic capacity of ATC cells in a low-glucose environment [81]. TC cells rely on glucose to activate the PI3K pathway, which influences many cellular processes, such as metabolism, cancer progression, and metastasis [82]. PI3K signaling can regulate GLUT1 expression through Akt, enhance glucose intake and facilitate phosphofructokinase (PFK) activity [83] to further promote the increase in LA. Thyroid oncogene mutations, such as c-Myc, can also increase GLUT1 expression in cancer cells, affecting glucose metabolism, and driving cell malignant transformation [84]. Therefore, glycolysis can help cancer cells survive, grow, and metastasize and further help cells resist apoptosis and avoid immune system destruction [85]. The expression of LA and LDH can support the metabolic adaptation and tumorigenesis of cancer cells [81]. Therefore, targeted suppression of glycolytic and lactate processing pathways may represent an effective treatment strategy for TC.

The role of glycolysis in thyroid cancer

Abnormal glycolysis in TC can acidify the tumor microenvironment, further leading to the abnormal growth of cancer cells. Acidification leads to changes in biological factors in the environment, which can promote or inhibit further development of TC. The fatal element of TC is metastasis, which is also affected by alterations in the tumor microenvironment also affects tumor metastasis [86].

Thyroid cell carcinogenesis and tumor formation

A necessary condition for cell growth is energy supply, and glycolysis is a crucial method to provide power for cell growth. Compared with normal cells, tumor cells have inefficient energy production, which implies that their growth and reproduction require more glucose to provide power [87]. Tumor cells can exhibit a specific metabolic pattern, which can quickly transport and consume glucose to produce ATP and boost drug excretion [88]. Meanwhile, increased levels of reactive oxygen species (ROS) are also an essential feature of TC cells, and high ROS production may lead to cell damage and cell death [89]. ROS have been demonstrated to play a significant role in cell proliferation, metabolism, angiogenesis, cell growth, and survival in several advanced malignant tumors [90, 91]. In TC, cancer cells preferentially undergo glycolysis even under aerobic conditions [92]. Many genes are upregulated or downregulated to change glycolysis, thereby promoting or inhibiting tumor growth (shown in Table 1).
Table 1
Regulators of glycolysis associated with thyroid cancer cells growth
Regulators
Effects in glycolysis
Effects in TC growth
Downstream molecules
Participation pathway
Mechanism
Negative regulators
PTEN
Negative
Negative
HIF-1, VEGF, PCNA
PI3K/PTEN/AKT, PI3K-AKT-mTOR
Inhibit GLUT1 expression and glucose uptake in TC, downregulate PI3K-AKT-mTOR pathway and affect glucose metabolism
P53
Negative
Negative
AMPK, GLUT1,3,4, PGM, TSC2, RRAD
PI3K-AKT-mTOR, caspase pathway
Shorten glucose uptake and promote mitochondrial oxidation, so as to resist Warburg effect, which also leads to cell cycle arrest and apoptotic cell death
Iodide
Negative
Negative
GLUT1
Oxidation pathway, rate-limiting glucose-facilitated transport system
Inhibit TSH induced stimulation of glucose transport, reduce the number of available carrier sites and inhibit cell growth
BRAFV600E
Negative
Negative
GLUT1
RAF/MEK/ERK
Initiate the glycolytic table associated with GLUT1 overexpression and inhibit mitochondrial respiration in thyroid cells
Positive regulators
HIF-1
Positive
Positive
GLUT1, PDK, PKM2, HKII
PI3K/AKT
Enhance glycolysis, increase GLUTs expression, and promote tumor growth
PI3K/AKT
Positive
Positive
GLUT1, HKII, PDK1
PI3K/AKT
Promote cell carcinogenesis and increase glycolytic flux
TSH
Positive
Positive
mTOR
PI3K.AKT, RAS/MAPK
Promote thymocyte proliferation and thyroid proliferation
c-Myc
Positive
Positive
GLUT1, LDHA, PK, PKM2, MCTs
APC, miR-222-3p/HIPK2/ERK
Promote anaerobic glycolysis, tumor growth and cell proliferation
AMPK
Positive
Negative
HIF-1α, mTOR
AMPK/AKT, AMPT/mTOR
Regulate glycolysis and control cell growth, apoptosis and survival
 
LDHA
Positive
Positive
STAT3
JAK/STAT
Promote the conversion of pyruvate to lactic acid, so as to promote the glycolysis process and tumor growth
 
PD-1
Positive
Positive
SHP2, RAS
SHP2/RAS/MAPK, RAS-MAPK-ERS
Promote the proliferation and vitality of thyroid cancer cells
BRAFV600E mutations are common in TCs [93, 94]. The BRAFV600E mutation can alter the HIF1-Myc-PGC1 axis, leading to inhibition of mitochondrial respiration and enhancement of aerobic glycolysis [95]. Meanwhile, glycolytic enzymes (such as LDHA and PKM2) are regulated by HIF1 and Myc to promote glycolysis, and BRAFV600E can regulate phosphate MEK1/2, thereby reducing mitochondrial metabolism [95]. The role of BRAFV600E signaling in the regulation of tumor metabolism suggests that BRAF can generate biodynamic adaptation by inhibiting oxidative phosphorylation [96]. Other studies have shown that glucose restriction in the cellular environment can restrain the proliferation of ATC cells [97, 98], while programmed cell death protein 1 (PD-1) can promote the proliferation and viability of TC cells [99]. HIF1 inhibits mitochondrial respiration and Myc activity in TC by inhibiting the expression of peroxisome proliferator-activated receptor γ coactivator-1 (PGC-1), indicating that the metabolic reprogramming may be a key step in thyroid carcinogenesis [100].
Mutations in the RAS-MAPK-ERK and PI3K-Akt-mTOR pathways usually exist in highly differentiated tumor components, and most ATCs are developed from these tumor components [101, 102]. In ATC, genetic alterations in the p53 gene are the most common (55%) changes [103]. Approximately 40% of PTC and 22% of FTC have p53 gene changes [104]. Studies have found that pAKT is highly expressed with pERK and low in PTEN in ATC patients, which indicates that the two pathways of RAS-MAPK-ERK and PI3K-AKT-mTOR play a synergistic role in the development of ATC [99]. In addition, the mutation of p53 was negatively correlated with the expression of pAKT, and there was a significant positive correlation between PTEN and pERK [99]. Activation of the PI3K-AKT-mTOR signaling pathway inhibits ERK1/2 activation, which suggests that the RAS-MAPK-ERK or PI3K-AKT-mTOR pathway controls the carcinogenic effects of ATC [105]. mTOR mediator signals are combined with PKB/Akt, HIF1, and AMPK signaling pathways to manage cell proliferation and survival under conditions of nutrient and energy deprivation [106]. mTOR is a central activator of the Warburg effect [2]. mTOR upregulates PKM1 expression through mediated transcriptional activation of HIF1α and c-Myc heteroribonucleoprotein-dependent regulation of PKM2 gene splicing [107]. The destruction of PKM1 inhibits oncogenic mTOR mediated tumorigenesis [108]. Unlike normal cells, mTOR hyperactive cells are more sensitive to the inhibition of mTOR or glycolysis. The dual inhibition of mTOR and glycolysis synergistically passivates the proliferation and tumor development of mTOR hyperactive cells [107]. PD-1 can activate Ras-MAPK signaling cascade in TC cells and enhance the expression of Ras in TC cells [109]. In addition, RET/PTC or BRAF mutations can also lead to active PI3K [110]. In TC cells, downregulation and activation of the Ras-MAPK and PI3K-Akt pathways mainly inhibit cell migration and proliferation [111]. However, inactivation of Ras-MAPK signaling has a positive effect on the mobility of ATC cells [112]. Compared to the inhibition of a single pathway, the dual Ras-MAPK and PI3K-Akt-mTOR pathways can inhibit cell growth and even lead to growth retardation in TC cells in a congenerous manner [113]. In addition to the influence of these factors and pathways, glycolysis leads to acidification of the tumor microenvironment, which also promotes TC progression.

Glycolysis and thyroid cancer microenvironmental acidosis

Glucose is converted to LA in tumor cells and flows extracellularly to form lactate and produce lactate accumulation (Fig. 2) [114]. Through continuous aerobic glycolysis, glucose alters some of the microenvironment, resulting in side effects [115]. LDHA of the glycolysis process promotes the conversion of pyruvate to LA, which is associated with the development of various cancers, including TC [116118]. Proton-linked monocarboxylate transporters (MCTs) transport LA across the plasma membrane, which requires binding of CAIX to the CD147, a widely expressed membrane glycoprotein [119]. Studies have shown that MCT1 is required for CD147 protein expression, causing the MCT/CD147 subunits to assemble and target the plasma membrane [120]. In rat thyroid tissues, MCT4 can output LA through the plasma membrane with the assistance of CD147 [120]. Research has found that Acriflavine (ACF) can disrupt the binding of MCT4 to its essential cofactor basigin [121]. ACF can effectively inhibit the growth of ATC cells in vitro by inhibiting LA output and subsequently inhibiting upstream glycolysis [81]. High levels of acid production result in a sharp local drop in the extracellular pH value. In addition to lactate, carbon dioxide (CO2) manufactured by catalytic pathways, such as the pentose phosphate pathway (PPP), is also conducive to acidifying the TME [122, 123]. In this case, microenvironmental acidification can promote tumor invasion by destroying adjacent normal cells, inducing extracellular matrix (ECM) degradation, and promoting angiogenesis [114]. Long-term exposure of normal cells to an acidic microenvironment leads to cell necrosis or apoptosis depending on p53 and caspase-3 mechanisms [124]. Nevertheless, tumor cells adjust their survival conditions to adapt to the acidic microenvironment [125]. The acidic microenvironment can inhibit the growth of normal cells, but acidosis is an indispensable criterion for cancer cell migration and invasion [124]. High levels of LA in TME will reduce the activity of immune cells, thereby promoting tumors and metastasis [126, 127]. Meanwhile, the accumulation of lactate and acidification of TME will accelerate the remodeling of basement membranes (BM) or boost the progression of epithelial-mesenchymal transition (EMT), contributing to tumor invasion [128]. Acidosis can affect tumor progression, aggression and metastasis, a phenotypic feature of TME markers [129]. The glucose uptake of tumor cells increases due to hypoxia, and glucose restriction in the TME also facilitates the activation of the M2-like phenotype in tumor-infiltrating macrophages, promoting the anti-inflammatory response and tumor growth [130]. In conclusion, aerobic glycolysis in tumor cells produces many lactic acid accumulations, which acidifies the TME, destroys adjacent normal tissues, degrades the extracellular matrix, and promotes angiogenesis, thus promoting tumor invasion and metastasis.

Glucometabolic reprogramming in metastatic thyroid cancer

Tumor metastasis to distant organs is caused by tumor cells with primary heterogeneous tumor diffusion, and the sequential growth and survival of tumor metastasis depend on different metabolic changes [131]. Malignant tumors proliferate indefinitely and have a tendency for distant metastasis. They require large amounts of energy and biosynthetic precursors to promote cell division, invasion, and migration [132, 133]. Secreted lactic acid can affect cell types in TME by activating multiple processes such as tumor cell survival and proliferation [134]. LA accumulation can induce various events in the TME, including the upregulation of hyaluronic acid, which is conducive to tumor migration [135, 136]. Lactate excretion by tumor cells allows acidic degradation of the matrix around healthy tissues, leading to invasive growth [137]. Tumor metastasis is a multistep cascade process, and more than 90% of cancer deaths are not caused by tumors alone, but by tumor metastasis [138]. At the beginning of metastasis, invasion is required. That is, diffuse malignant cells must converge the normal ambient tissues into a collective tissue structure or separate into small cell clusters [139, 140]. As tumor cells reduce cell-to-cell adhesion to relax tight structures, they promote further cell invasion, which is a feature of the EMT process [141, 142]. During the EMT process, the viscosity of tumor cells decreases and the activity of tumor cells increases. After converging normal ambient tissues and forming a new vascular network by secreting vascular factors, tumor cells will connect the small blood vessels, such as veins or capillaries, and lymph nodes and enter the circulation [143]. Interaction with neutrophils in circulation can promote further metastasis and diffusion. Neutrophils may also promote tumor cell extravasation by secreting matrix metalloproteinases (MMPs) [144]. Eventually, tumor cells can leave the blood circulation and invade secondary tissues.
Metastasis occurs in 10% of TC patients (Fig. 3), and approximately half of distant metastases occur in the lung, which may be associated with anti-nest loss apoptosis and pro-invasion signals mediated by LDHA phosphorylation [145, 146]. LDHA phosphorylation provides invasive signals in metastatic cancer cells by regulating redox status, and LDHA can also enhance tumor progression by possessing molecules related to EMT [147]. When LDHA is inhibited, more pyruvate will enter the tricarboxylic acid (TCA) cycle, resulting in an increased oxygen demand [147]. However, cancer cells are overdependent on aerobic glycolysis, which produces ATP rapidly and can use more precursors to meet the metabolic requirements of rapid proliferation. Therefore, when LDHA is inhibited, it can affect the proliferation, invasion, and metastasis of TC cells and prevent TC cells from escaping immunity [100]. In conclusion, LDHA can cause EMT-like changes that promote migration and invasion of TC cells, and can therefore be considered as a target factor for the treatment of TC. Increased GLUT-1 expression is also related to the increased invasive behavior and metastasis characteristics [148, 149]. HIF1, a downstream target of GLUT1, is also involved in tumor metastasis and migration [150]. Under hypoxiaconditions, increased glucose uptake by cancer cells can upregulate the stability of HIF-1α, leading to a weakened antitumor immune response [18]. In addition, programmed death-ligand 1 (PD-L1) is a downstream target of HIF-1 that can bind to PD-1 on T cells. The PD-1/PD-L1 interaction can activate the dephosphorylation of PI3K and block the Akt/mTOR pathway [151, 152].

Potential clinical value of glycolysis in thyroid cancers

Tools for detecting thyroid glycolysis

At present, many methods are available to help diagnose TC, and each of these methods has its own advantages and disadvantages, such as ultrasonography, fine-needle aspiration biopsy (FNAB), computed tomography (CT), and magnetic resonance imaging (MRI) (See Table 2). Among them, the diagnostic method of fluoro-2-deoxy-D-glucose positron emission tomography (FDG-PET) imaging is closely related to the glycolysis pathway. Taking advantage of exploiting the high activity of GLUT1 in tumor cells, FDG-PET imaging accumulates a significant amount of glucose in tumor cells and conducts in vivo detection in humans [153]. The diagnosis of malignant tumor metastasis relies heavily on this technology. In the thyroid gland, follicular epithelial cells exhibit fanatical iodine absorption mediated by the Na+/I symporter (NIS) [154]. Glucose uptake is increased by GLUT1 overexpression during differentiation in TCs. The opposite relationship between iodine absorption and glucose utilization is known as the iodine/FDG ‘turnover phenomenon’, reflecting the cell differentiation state and heterogeneous pattern of NIS expression [155, 156]. FDG-PET imaging is not recommended for the evaluation of patients with newly identified thyroid nodules or thyroid diseases [156]. Nevertheless, skeletal muscle metastasis of thyroid microcarcinoma can be evaluated by 18F-FDG PET/CT [157]. 18F-fluoro-2-deoxy-D-glucose (18F-FDG) is the most commonly used radiotracer in oncology imaging for staging, re-documentation, and assessment of treatment response in several tumors [158]. In DTC patients, lesions with high 18F-FDG and low radioactive iodine uptake are more clinically invasive [159]. Studies have found that malignant cells have the lowest degree of differentiation and the highest ability to absorb 18F-FDG [160]. Quantitative 18F-FDG-PET/CT evaluation can exclude the malignancy of uncertain thyroid nodules [161]. 18F-FDG-PET/CT can also be used to evaluate response to treatment, detect lesions in metastatic patients, and predict the prognosis of high-risk patients [162]. Although the evaluation of node status has reasonable specificity (94%), 18F-FDG PET/CT imaging shows a low sensitivity (30%) [162, 163], and the current American Thyroid Association (ATA) criteria do not recommend 18F-FDG PET/CT as a routine preoperative test [164]. However, 18F-FDG PET/CT is strongly recommended for follow-up of high-risk patients with elevated serum thyroglobulin (Tg) and negative 131I imaging [164, 165]. FDG PET has been shown to be helpful in detecting persistent or recurrent DTC in patients with low Tg; however, when FDG PET-CT is negative, this does not exclude DTC and requires further investigation [166].
Table 2
Detection strategies in thyroid cancers
Method
Principle
Frequency
Advantages
Disadvantages
Ultrasonography
Use ultrasound to present the internal image of opaque objects
Always
Detection of residual thyroid cancer in cervical lymph nodes or soft tissue
Unclear imaging, difficult qualitative, inaccurate quantitative
FNAB
A sterile puncture needle was used to puncture the suspicious part of the nodule, and some nodule components were extracted for cytological and pathological examination
Always
Differential diagnosis between benign and malignant thyroid nodules and diffuse goiter
Too few materials to know whether the blood vessels and capsule are invaded at the same time
CT
The optical signal is changed into an electrical signal, then into a digital signal, and finally into a computer picture
Always
Preoperative staging, monitoring, re staging, location of metastatic disease and continuous monitoring of progression and treatment response of thyroid cancer
Difficult to find small lesions with little or no density change
MRI
Based on the low diffusion coefficient of water molecules in high cell tissues
Often
Helpful to detect lymph node involvement and lymph node metastasis before operation
Expensive equipment, long time to image and limited patients
Radioiodine imaging
TSH stimulates iodine uptake in residual normal and malignant thyroid tissues
Often
Identify, locate and monitor the progress or treatment response of iodine preference metastasis in differentiated thyroid cancer
Needed TSH to stimulate iodine uptake in residual normal and malignant thyroid tissues
123Ι/131Ι/99mTc Thyroid Scintigraphy
Effective concentration of iodine based on thyroid follicular cells
Often
The only evidence of autonomic functional thyroid nodules
Uncertain to hyper-functional nodules
FDG-PET
Based on the mutual annihilation of positrons and electrons, two high-energy 511 keV photons are released in the opposite direction
Often
Evaluation of thyroid cancer recurrence and for systemic and focal dosimetry
Limitation for patients with newly discovered thyroid nodules or thyroid diseases were evaluated
Diffusion-weighted imaging (DWI) provides quantitative and qualitative information based on the assessment of micro movement of water at the cell level, and can be used to distinguish benign and malignant diseases [167]. ATA states that cervical ultrasound is the best method to assess the status of lymph nodes prior to surgery [167]. MRI is a sensitive imaging modality that localizes sites of potential recurrence of DTC in the neck, mediastinum, bones, and liver, although the accuracy of detecting lung lesions is low [168]. MRI significantly reduced the total radiation dose of patients compared to PET/CT [169]. Meanwhile, PET/MRI is a promising tool with great potential to provide complementary data obtained under the same time and conditions. Diagnosis of thyroid nodules by conventional ultrasound relies on image quality, neck coverage, and ultrasound interpretation [170]. The current gold standard for confirming the diagnosis of TC is FNAB, but it remains highly likely to fail to describe micronodules of the thyroid gland [171]. Therefore, it is important to combine the available tools, such as ultrasound, CT and MRI, to establish a correct diagnosis of TC and evaluate the curative effect after treatment.

Therapeutic strategies targeting glycolysis dependence in thyroid cancers

Glycolysis brings many advantages to fast-growing tumor cells [172], and targeting metabolic pathways may be a promising method for tumor therapy [173]. Many targeted treatment methods are available for TCs, but the existing techniques are not systematically integrated. Among them, treatment strategies targeting glycolysis have received considerable attention. Below, we summarize some specific targeted treatment methods for TCs based on crucial factors of the glycolysis process (Fig. 4).

Tyrosine kinase inhibitors (TKIs)

Tyrosine kinase receptors are involved in cancer proliferation, angiogenesis and lymphangiogenesis [8, 174]. Angiogenesis plays a significant role in the occurrence and development of tumors, while lymphangiogenesis is critical for metastasis formation [175]. The expression of VEGF in TC cells can facilitate tumor angiogenesis [176]. Vascular endothelial growth factor receptor 2 (VEGFR2) is a TK receptor expressed by vascular endothelial cells from TME via immune cells, and its activation can initiate HIF1α in tumors and promote VEGF-α overexpression [177, 178]. VEGF-α is mainly expressed in ATC cells but not in normal thyroid tissues and upregulates the PI3K/Akt and MAPK pathways through growth factor signals [179]. In DTC, VEGF and VEGFR2 are overexpressed and can promote tumor progression and invasion. VEGF receptor is also overexpressed in MTC [180]. PI3K/Akt/mTOR and Raf/MEK/ERK are involved in ATC dedifferentiation and tumor growth [181]. TKIs targeting RET and VEGFR2 have shown promising results in phase II trials [182, 183]. Therefore, the study of TKIs in the treatment of TC plays a positive role in improving the current situation of TC patients.
ATA guidelines recommend that patients with stable or minimal progression should not be treated immediately and TKI treatment should be considered in “patients with metastatic, rapidly progressive, symptomatic, and/or imminently threatening disease [184, 185]. Several tyrosine kinase inhibitors have entered clinical trials: (1) sorafenib (BAY 43-9006) can inhibit RAF, VEGFR2, VEGFR3, and KIT kinase, which inhibits TC growth through anti-proliferation and anti-angiogenesis mechanisms [186, 187]; (2) Sunitinib (SU011248) preferentially inhibits VEGFR1-3, KIT, and PDGFR kinase [188]. Sunitinib inhibits the autophosphorylation of RET/PTC and the activation of STAT3, and blocks the transformation ability of RET/PTC [189]; (3) Vandetanib (ZD6474) is an effective inhibitor of VEGFR2, VEGFR3, RET, and epidermal growth factor receptor kinase [190]; (4) Lenvatinib (E7080) inhibits FGFR1-4, PDGFRβ, Vegfr1-3, RET, and supporting element kinase [191]; and (5) Cabotanni (XL184) inhibits c-Met, VEGFR1, 2, and RET kinases [192]. ZD6474 and XL184 have been approved as targeted treatments for advanced MTC with symptoms or high tumor burden [8, 193]. Research has shown that long-term medication cessation in patients may not lead to rapid disease progression. However, it may result in long-term “TKI free” stable diseases in individual patients [194]. Analysis of calcitonin and CDT is necessary during discontinuation to reveal tumor progression. In the event of progress, the same TKI can be used to restart [195]. A large number of studies have demonstrated that TKIs represent a new targeted therapy for invasive, progressive, and refractory TCs [196]. However, there are toxic reactions to inhibiting VEGF treatment, such as hypertension, kidney injury, bleeding, cardiovascular toxicity, etc. [197]. Doctors should closely understand the toxicity, adopt appropriate treatment strategies, and decide on treatment interruption, dosage adjustment, and cessation as needed.

LDHA inhibitors

LDHA, one of the pivotal glycolytic enzymes, promotes the conversion of pyruvate to lactic acid by compensating for the reduction in oxidative mitochondrial function and sustains cell survival under hypoxia [117, 198]. The decrease in glucose uptake caused by LDHA inhibitors is not the cause of decreased cell density or reduced GLUT1 surface expression [199]. LDHA inhibitors inhibit the regeneration of nicotinamide adenine dinucleotide (NAD) and impair the activity of glyceraldehyde 3-phosphate dehydrogenase (GAPDH), which accumulates an intermediate volume of glucose in the initial step of glycolysis, increases the cellular level of unused glucose, and inhibits glucose uptake [200]. The overexpression of LDHA and increased phosphorylation are common findings in thyroid malignancies [201]. Patients with high LDHA expression have a poor prognosis, which is closely connected with metastasis, and high LDHA levels have been demonstrated to be related to lymph node metastasis [202]. STAT3 is a new upstream regulator of LDHA and a key transcription factor involved in many growth factors and cytokines, which can trigger various biological processes, including cell growth, differentiation, and survival [116]. The expression of STAT3 is positively associated with the expression of LDHA [203]. The expression levels of STAT3 and PSTAT3 are higher in the group with lymph node metastasis than in the group without lymph node metastasis [204]. LDHA can increase the proliferation, invasion, and metastasis of TC cells and help TC cells evade immunity [100]. Therefore, LDHA is considered as a promising target for the prevention and treatment of TC. Chemical inhibitors of LDHA are being developed, such as the LDHA inhibitors FX11, GSK 2837808A, sodium oxamate, and pyruvic acid, which significantly inhibit cell proliferation and induce apoptosis [205]. Phosphorylated AMPK levels increases when LDHA is knocked down or inhibited. As the primary downstream target of the AMPK signal, mTOR is involved in cell growth, cell proliferation, and cell survival [206]. LDHA knockdown or inhibition reduces the phosphorylation level of mTOR, which also suggests that mTOR inhibitors can be used in the glycolysis process to inhibit the growth and metastasis of TC [202].

mTOR inhibitors

mTOR is involved in controlling the proliferation of normal and TC cells and regulating iodide absorption in normal thyroid cells; therefore, mTOR inhibition may efficiently reduce cell proliferation and stimulate iodide absorption in TC cells [207]. mTOR inhibition leads to severe impairment of proliferative signals via the PI3K/Akt pathway [208] and cell cycle arrest in the G1 stage, which can be activated by membrane receptors, including the insulin-like growth factor receptor (IGFR) and the thyroid-stimulating hormone (TSH) receptor in thyroid cells. Rapamycin analogs can directly inhibit mTOR signaling, such as LY294002, AY-22989, AZD8055, and temsirolimus (CCI-7790) [209], and reduce cell proliferation in TC cell lines [210]. Genes encoding TK receptors, mitochondrial activated protein kinase (MAPK) and PI3K/Akt pathway, are mutated in almost all ATC cases [102]. Two open-label phase II clinical trials have demonstrated the modest anti-tumor activity of everolimus and the stabilization of TC [211, 212]. However, neither trial demonstrated an association between tumor mutation status and drug response in patients with ATC. Therefore, targeting these two pathways at the same time may be particularly effective in the treatment of ATC. A PI3K inhibitor (LY294002) was found to inhibit mTOR, slow disease progression, eliminate lung metastasis and prolong the survival time in mice due to inhibition of cancer growth and proliferation, increased apoptosis, and decreased cell activity [213]. The growth inhibition of cancer cell lines treated with MAPK kinase (MEK) and mTOR inhibitor was greater than 60% [111]. Another study demonstrated the therapeutic potential of the novel MEK inhibitor RDEA119 in TC and its synergistic effect with the mTOR inhibitor temsirolimus [214].

MEK inhibitors

The MAPK-MEK signaling pathway is often overactivated in ATCs and correlated with the progression of ATCs [215]. MEK inhibitors can induce iodine uptake and retention in TCs, which exhibits G0/G1 arrest by downregulating MEK/ERK phosphorylation and inhibiting the viability of BRAF mutant cells [216, 217]. The MAPK pathway is an evolutionarily preserved signaling cascade that links extracellular and internal stimuli with the control of multiple cellular processes under physiological and pathological conditions, including cell proliferation, survival, invasion, migration, and differentiation [216]. Trametinib, an MEK1/2 inhibitor, has been demonstrated to independently improve survival in patients with metastatic melanoma [218]. Downstream MEK inhibition can not only prevent BRAF resistance in BRAF mutant cells but also block abnormal MAPK activation in BRAF wild-type cells [219]. Pretreatment with MAPK inhibitors improves the reactivity of RAI treatment [220]. MEK inhibitors (such as selumetinib) activate PI3K and MAPK pathways by stimulating HER3 gene expression, and the HER3 inhibitor lapatinib can prevent MAPK rebound and sensitize BRAFv600E positive TC cells to Raf or MAP/ERK inhibitors [221]. The most common adverse effects of selumetinib that were reported are fatigue, diarrhea, and rash [222]. A single-arm multicenter two-phase II clinical trial is currently underway in the UK to evaluate the efficacy of selumetinib in combination with RAI in patients with recurrent thyroid cancer [223]. HER inhibitors combined with BRAF/MEK inhibitors can improve the sensitivity of BRAFv600E positive PTC to BRAF/MEK inhibitors by preventing MAPK rebound and increasing NIS expression [224, 225]. MEK has many unique biochemical and biological characteristics, rendering it an attractive target from the perspective of anticancer drug development.

Other strategies

In addition to the above treatments targeting specific enzymes, some studies have found that aerobic exercise may also be a new treatment. Aerobic exercise, an anti-Warburg maneuver, such as swimming and jogging, can increase mitochondrial function and lactate clearance, which increases fat oxidation, decreases glycolysis and reduces dependence on glycogen and glucose [226]. In addition, exercise can reduce the harmful activity of c-Myc [227]. Hence, aerobic exercise helps counteract the metabolic conversion of cancer cells to glycolytic metabolism and produces epigenetic responses that help restore the oxidative phenotype [228]. Other studies have also proved that diets might affect the tumor growth and be a potential treatment [229]. Tailed diets are based on the nutritional vulnerabilities of tumor. Although lacking well-designed clinical trials, some preclinical studies have demonstrated that tailed diet such as low-carbohydrate diet and restring dietary serine and glycine can starve tumors and boots the effectiveness of cancer therapy [230]. Thus, alteration of cellular metabolism by low-carbohydrate ketogenic diets can be an important therapeutic strategy to selectively kill cancer cells that mainly survive on glycolysis [231]. Calorie-restricted diets enhance ameliorate metabolic pathogenesis and reduce the incidence of cancer [232, 233]. Also, caloric restriction promotes antineoplastic immune responses and suppresses tumor cell proliferation [234, 235]. Hence, metabolic interventions may have a great potential as co-adjuvant therapy in the management of TC.

Conclusions

The increasing incidence rate of TC has been a significant concern in the medical field. The unambiguous pathogenesis of TC is not yet fully understood because of its diversity. Glycolysis is a process that occurs in all cancer cells. Linking it with TC provides some insights for the treatment of TC. Warburg effect, that is, aerobic glycolysis in the presence of oxygen and mitochondria with normal function in principle, constitutes the main driving factor of cancer progression mechanism, resistance to traditional therapy and poor prognosis of patients. The molecular and functional processes associated with tumorigenesis may include: (a) significant acceleration of glycolytic flux; (b) generation of sufficient ATP to provide energy for cancer cells; (c) backup and transfer of glycolytic intermediates, promoting the biosynthesis of nucleotides, nonessential amino acids, lipids and hexosamine; (d) inhibition of pyruvate from entering mitochondria; (e) excessive formation and accumulation of lactate; (f) maintaining cell redox homeostasis and low ROS formation; and (h) HIF-1 overexpression, mutant p53 and mutant PTEN, which inhibit mitochondrial biogenesis and function. The Warburg effect can help cancer cells survive, grow and metastasize, further helping tumors resist apoptosis, and avoid destruction by the immune system. Glycolysis has a complete mechanism. By understanding the process of its occurrence and comparing differences in glycolysis processes between normal cells and cancer cells, we can target the glycolysis pathway to treat TC in follow-up research. The common treatment for TC is surgical resection, but recurrence or deterioration is still possible. For special types of TC, the current treatment cannot achieve a good therapeutic effect, and whether we can target glycolysis to achieve a therapeutic effect requires further exploration.
With improved understanding of “reprogramming of glucose metabolism” in TC, patients with poorly differentiated TC are no longer without effective therapies in terms of the development of new therapies. Novel diagnostic methods based on glycolysis mechanism, such as FDG-PET, as well as targeting drugs, such as FX11, trametinib, and AZD8055, and diets will be of great significance to further deepen our understanding of glycolysis regulation and reasonably design strategies for the diagnosis and treatment of TC, especially for patients with poorly differentiated TC or relapse status. Many inhibitors have entered the stage of clinical experimental research, but no extraordinary evidence shows that they have a good therapeutic effect. Research in this field still needs to be further strengthened. Moreover, how to affect the occurrence and development of TC requires further verification. Whether inhibitors affecting the glycolysis pathway have a definitive inhibitory effect on TC and their safety warrants our attention. Taken together, targeting the cancer metabolism holds great promise as a therapeutic modality in TC.

Acknowledgements

We wish to thank all the authors who participated in this study.

Declarations

This study was approved by the Ethics Review Committee of Xiangya Hospital, Central South University, and followed the Declaration of Helsinki (20211245). The informed consent was waived because of the retrospective and anonymous nature of the study.
All authors gave consent for the publication of this study.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Baloch ZW, Asa SL, Barletta JA, Ghossein RA, Juhlin CC, Jung CK, LiVolsi VA, Papotti MG, Sobrinho-Simoes M, Tallini GJ. Overview of the 2022 WHO classification of thyroid neoplasms. Endocr Pathol. 2022;33:27–63.PubMed Baloch ZW, Asa SL, Barletta JA, Ghossein RA, Juhlin CC, Jung CK, LiVolsi VA, Papotti MG, Sobrinho-Simoes M, Tallini GJ. Overview of the 2022 WHO classification of thyroid neoplasms. Endocr Pathol. 2022;33:27–63.PubMed
2.
Zurück zum Zitat Shi YB, Chen SY, Liu RB. The new insights into autophagy in thyroid cancer progression. J Transl Med. 2023;21(1):1–3. Shi YB, Chen SY, Liu RB. The new insights into autophagy in thyroid cancer progression. J Transl Med. 2023;21(1):1–3.
3.
Zurück zum Zitat van Houten P, Netea-Maier RT, Smit JW. Differentiated thyroid carcinoma: an update. Best Pract Res Clin Endocrinol Metab. 2023;37(1):101687.PubMed van Houten P, Netea-Maier RT, Smit JW. Differentiated thyroid carcinoma: an update. Best Pract Res Clin Endocrinol Metab. 2023;37(1):101687.PubMed
4.
Zurück zum Zitat Cabanillas ME, McFadden DG, Durante C. Thyroid cancer. Lancet. 2016;388:2783–95.PubMed Cabanillas ME, McFadden DG, Durante C. Thyroid cancer. Lancet. 2016;388:2783–95.PubMed
5.
Zurück zum Zitat Filetti S, Durante C, Hartl D, Leboulleux S, Locati LD, Newbold K, Papotti MG, Berruti A. Thyroid cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-updagger. Ann Oncol. 2019;30:1856–83.PubMed Filetti S, Durante C, Hartl D, Leboulleux S, Locati LD, Newbold K, Papotti MG, Berruti A. Thyroid cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-updagger. Ann Oncol. 2019;30:1856–83.PubMed
6.
Zurück zum Zitat Oh JM, Ahn BC. Molecular mechanisms of radioactive iodine refractoriness in differentiated thyroid cancer: Impaired sodium iodide symporter (NIS) expression owing to altered signaling pathway activity and intracellular localization of NIS. Theranostics. 2021;11:6251–77.PubMedPubMedCentral Oh JM, Ahn BC. Molecular mechanisms of radioactive iodine refractoriness in differentiated thyroid cancer: Impaired sodium iodide symporter (NIS) expression owing to altered signaling pathway activity and intracellular localization of NIS. Theranostics. 2021;11:6251–77.PubMedPubMedCentral
7.
Zurück zum Zitat Gild ML, Tsang VHM, Clifton-Bligh RJ, Robinson BG. Multikinase inhibitors in thyroid cancer: timing of targeted therapy. Nat Rev Endocrinol. 2021;17:225–34.PubMed Gild ML, Tsang VHM, Clifton-Bligh RJ, Robinson BG. Multikinase inhibitors in thyroid cancer: timing of targeted therapy. Nat Rev Endocrinol. 2021;17:225–34.PubMed
8.
Zurück zum Zitat Cabanillas ME, Ryder M, Jimenez C. Targeted therapy for advanced thyroid cancer: kinase inhibitors and beyond. Endocr Rev. 2019;40:1573–604.PubMedPubMedCentral Cabanillas ME, Ryder M, Jimenez C. Targeted therapy for advanced thyroid cancer: kinase inhibitors and beyond. Endocr Rev. 2019;40:1573–604.PubMedPubMedCentral
9.
Zurück zum Zitat Lin R. Thyroid cancer stem cells. Nat Rev Endocrinol. 2011;7:609–16.PubMed Lin R. Thyroid cancer stem cells. Nat Rev Endocrinol. 2011;7:609–16.PubMed
10.
Zurück zum Zitat Fallahi P, Ferrari SM, Galdiero MR, Varricchi G, Elia G, Ragusa F, Paparo SR, Benvenga S, Antonelli A. Molecular targets of tyrosine kinase inhibitors in thyroid cancer. Semin Cancer Biol. 2022;79:180–96.PubMed Fallahi P, Ferrari SM, Galdiero MR, Varricchi G, Elia G, Ragusa F, Paparo SR, Benvenga S, Antonelli A. Molecular targets of tyrosine kinase inhibitors in thyroid cancer. Semin Cancer Biol. 2022;79:180–96.PubMed
11.
Zurück zum Zitat Hu J, Yuan IJ, Mirshahidi S, Simental A, Lee SC. Thyroid carcinoma: phenotypic features, underlying biology and potential relevance for targeting therapy. Int J Mol Sci. 2021;22(4):1950.PubMedPubMedCentral Hu J, Yuan IJ, Mirshahidi S, Simental A, Lee SC. Thyroid carcinoma: phenotypic features, underlying biology and potential relevance for targeting therapy. Int J Mol Sci. 2021;22(4):1950.PubMedPubMedCentral
12.
Zurück zum Zitat Bryant KL, Stalnecker CA, Zeitouni D, Klomp JE, Peng S, Tikunov AP, Gunda V, Pierobon M, Waters AM, George SD, et al. Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat Med. 2019;25:628–40.PubMedPubMedCentral Bryant KL, Stalnecker CA, Zeitouni D, Klomp JE, Peng S, Tikunov AP, Gunda V, Pierobon M, Waters AM, George SD, et al. Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat Med. 2019;25:628–40.PubMedPubMedCentral
13.
Zurück zum Zitat Su WY, Tian LY, Guo LP, Huang LQ, Gao WY. PI3K signaling-regulated metabolic reprogramming: from mechanism to application. Biochim Biophys Acta. 2023;25:188952. Su WY, Tian LY, Guo LP, Huang LQ, Gao WY. PI3K signaling-regulated metabolic reprogramming: from mechanism to application. Biochim Biophys Acta. 2023;25:188952.
14.
Zurück zum Zitat Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41:211–8.PubMedPubMedCentral Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41:211–8.PubMedPubMedCentral
15.
16.
Zurück zum Zitat Heydarzadeh S, Moshtaghie AA, Daneshpour M, Hedayati M. Correction to: regulators of glucose uptake in thyroid cancer cell lines. Cell Commun Signal. 2022;20:11.PubMedPubMedCentral Heydarzadeh S, Moshtaghie AA, Daneshpour M, Hedayati M. Correction to: regulators of glucose uptake in thyroid cancer cell lines. Cell Commun Signal. 2022;20:11.PubMedPubMedCentral
18.
Zurück zum Zitat Nagao A, Kobayashi M, Koyasu S, Chow CCT, Harada H. HIF-1-dependent reprogramming of glucose metabolic pathway of cancer cells and its therapeutic significance. Int J Mol Sci. 2019;20:238.PubMedPubMedCentral Nagao A, Kobayashi M, Koyasu S, Chow CCT, Harada H. HIF-1-dependent reprogramming of glucose metabolic pathway of cancer cells and its therapeutic significance. Int J Mol Sci. 2019;20:238.PubMedPubMedCentral
19.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.PubMed Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.PubMed
20.
Zurück zum Zitat Warburg O. Origin of cancer cells. Oncol. 1956;9:75–83. Warburg O. Origin of cancer cells. Oncol. 1956;9:75–83.
21.
Zurück zum Zitat Lunt SY, Vander Heiden MG. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu Rev Cell Dev Biol. 2011;27:441–64.PubMed Lunt SY, Vander Heiden MG. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu Rev Cell Dev Biol. 2011;27:441–64.PubMed
22.
Zurück zum Zitat Khatami F, Payab M, Sarvari M, Gilany K, Larijani B, Arjmand B, Tavangar SM. Oncometabolites as biomarkers in thyroid cancer: a systematic review. Cancer Manag Res. 2019;11:1829–41.PubMedPubMedCentral Khatami F, Payab M, Sarvari M, Gilany K, Larijani B, Arjmand B, Tavangar SM. Oncometabolites as biomarkers in thyroid cancer: a systematic review. Cancer Manag Res. 2019;11:1829–41.PubMedPubMedCentral
23.
Zurück zum Zitat Biswas SK. Metabolic reprogramming of immune cells in cancer progression. Immunity. 2015;43:435–49.PubMed Biswas SK. Metabolic reprogramming of immune cells in cancer progression. Immunity. 2015;43:435–49.PubMed
24.
Zurück zum Zitat Mates JM, Campos-Sandoval JA, Santos-Jimenez JL, Marquez J. Dysregulation of glutaminase and glutamine synthetase in cancer. Cancer Lett. 2019;467:29–39.PubMed Mates JM, Campos-Sandoval JA, Santos-Jimenez JL, Marquez J. Dysregulation of glutaminase and glutamine synthetase in cancer. Cancer Lett. 2019;467:29–39.PubMed
25.
Zurück zum Zitat Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.PubMed Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.PubMed
26.
Zurück zum Zitat Warburg O. On the origin of cancer cells. Science. 1956;123:309–14.PubMed Warburg O. On the origin of cancer cells. Science. 1956;123:309–14.PubMed
27.
Zurück zum Zitat Pascale RM, Calvisi DF, Simile MM, Feo CF, Feo F. The Warburg effect 97 years after its discovery. Cancers (Basel). 2020;12:2819.PubMed Pascale RM, Calvisi DF, Simile MM, Feo CF, Feo F. The Warburg effect 97 years after its discovery. Cancers (Basel). 2020;12:2819.PubMed
28.
Zurück zum Zitat Vaupel P, Multhoff G. Revisiting the Warburg effect: historical dogma versus current understanding. J Physiol. 2021;599:1745–57.PubMed Vaupel P, Multhoff G. Revisiting the Warburg effect: historical dogma versus current understanding. J Physiol. 2021;599:1745–57.PubMed
29.
Zurück zum Zitat Moreno-Sánchez R, Rodríguez-Enríquez S, Marín-Hernández A, Saavedra E. Energy metabolism in tumor cells. FEBS J. 2007;274:1393–418.PubMed Moreno-Sánchez R, Rodríguez-Enríquez S, Marín-Hernández A, Saavedra E. Energy metabolism in tumor cells. FEBS J. 2007;274:1393–418.PubMed
30.
Zurück zum Zitat Bose S, Le A. Glucose metabolism in cancer. Adv Exp Med Biol. 2018;1063:3–12.PubMed Bose S, Le A. Glucose metabolism in cancer. Adv Exp Med Biol. 2018;1063:3–12.PubMed
31.
Zurück zum Zitat Bose S, Zhang C, Le A. Glucose metabolism in cancer: the Warburg effect and beyond. Adv Exp Med Biol. 2021;1311:3–15.PubMedPubMedCentral Bose S, Zhang C, Le A. Glucose metabolism in cancer: the Warburg effect and beyond. Adv Exp Med Biol. 2021;1311:3–15.PubMedPubMedCentral
32.
Zurück zum Zitat Qin C, Yang G, Yang J, Ren B, Wang H, Chen G, Zhao F, You L, Wang W, Zhao Y. Metabolism of pancreatic cancer: paving the way to better anticancer strategies. Mol Cancer. 2020;19:50.PubMedPubMedCentral Qin C, Yang G, Yang J, Ren B, Wang H, Chen G, Zhao F, You L, Wang W, Zhao Y. Metabolism of pancreatic cancer: paving the way to better anticancer strategies. Mol Cancer. 2020;19:50.PubMedPubMedCentral
33.
Zurück zum Zitat Heydarzadeh S, Moshtaghie AA, Daneshpoor M, Hedayati M. Regulators of glucose uptake in thyroid cancer cell lines. Cell Commun Signal. 2020;18:83.PubMedPubMedCentral Heydarzadeh S, Moshtaghie AA, Daneshpoor M, Hedayati M. Regulators of glucose uptake in thyroid cancer cell lines. Cell Commun Signal. 2020;18:83.PubMedPubMedCentral
34.
Zurück zum Zitat Shi Y, Liu S, Ahmad S, Gao Q. Targeting key transporters in tumor glycolysis as a novel anticancer strategy. Curr Top Med Chem. 2018;18:454–66.PubMed Shi Y, Liu S, Ahmad S, Gao Q. Targeting key transporters in tumor glycolysis as a novel anticancer strategy. Curr Top Med Chem. 2018;18:454–66.PubMed
35.
36.
Zurück zum Zitat Meziou S, Ringuette Goulet C, Hovington H, Lefebvre V, Lavallée É, Bergeron M, Brisson H, Champagne A, Neveu B, Lacombe D, et al. GLUT1 expression in high-risk prostate cancer: correlation with (18)F-FDG-PET/CT and clinical outcome. Prostate Cancer Prostatic Dis. 2020;23:441–8.PubMed Meziou S, Ringuette Goulet C, Hovington H, Lefebvre V, Lavallée É, Bergeron M, Brisson H, Champagne A, Neveu B, Lacombe D, et al. GLUT1 expression in high-risk prostate cancer: correlation with (18)F-FDG-PET/CT and clinical outcome. Prostate Cancer Prostatic Dis. 2020;23:441–8.PubMed
37.
Zurück zum Zitat Cho H, Lee YS, Kim J, Chung JY, Kim JH. Overexpression of glucose transporter-1 (GLUT-1) predicts poor prognosis in epithelial ovarian cancer. Cancer Invest. 2013;31:607–15.PubMed Cho H, Lee YS, Kim J, Chung JY, Kim JH. Overexpression of glucose transporter-1 (GLUT-1) predicts poor prognosis in epithelial ovarian cancer. Cancer Invest. 2013;31:607–15.PubMed
38.
Zurück zum Zitat Whitaker RM, Wills LP, Stallons LJ, Schnellmann RG. cGMP-selective phosphodiesterase inhibitors stimulate mitochondrial biogenesis and promote recovery from acute kidney injury. J Pharmacol Exp Ther. 2013;347:626–34.PubMedPubMedCentral Whitaker RM, Wills LP, Stallons LJ, Schnellmann RG. cGMP-selective phosphodiesterase inhibitors stimulate mitochondrial biogenesis and promote recovery from acute kidney injury. J Pharmacol Exp Ther. 2013;347:626–34.PubMedPubMedCentral
39.
Zurück zum Zitat Gonzalez-Menendez P, Hevia D, Alonso-Arias R, Alvarez-Artime A, Rodriguez-Garcia A, Kinet S, Gonzalez-Pola I, Taylor N, Mayo JC, Sainz RM. GLUT1 protects prostate cancer cells from glucose deprivation-induced oxidative stress. Redox Biol. 2018;17:112–27.PubMedPubMedCentral Gonzalez-Menendez P, Hevia D, Alonso-Arias R, Alvarez-Artime A, Rodriguez-Garcia A, Kinet S, Gonzalez-Pola I, Taylor N, Mayo JC, Sainz RM. GLUT1 protects prostate cancer cells from glucose deprivation-induced oxidative stress. Redox Biol. 2018;17:112–27.PubMedPubMedCentral
40.
Zurück zum Zitat Kunkel M, Reichert TE, Benz P, Lehr HA, Jeong JH, Wieand S, Bartenstein P, Wagner W, Whiteside TL. Overexpression of Glut-1 and increased glucose metabolism in tumors are associated with a poor prognosis in patients with oral squamous cell carcinoma. Cancer. 2003;97:1015–24.PubMed Kunkel M, Reichert TE, Benz P, Lehr HA, Jeong JH, Wieand S, Bartenstein P, Wagner W, Whiteside TL. Overexpression of Glut-1 and increased glucose metabolism in tumors are associated with a poor prognosis in patients with oral squamous cell carcinoma. Cancer. 2003;97:1015–24.PubMed
41.
Zurück zum Zitat Kawamura T, Kusakabe T, Sugino T, Watanabe K, Fukuda T, Nashimoto A, Honma K, Suzuki T. Expression of glucose transporter-1 in human gastric carcinoma: association with tumor aggressiveness, metastasis, and patient survival. Cancer. 2001;92:634–41.PubMed Kawamura T, Kusakabe T, Sugino T, Watanabe K, Fukuda T, Nashimoto A, Honma K, Suzuki T. Expression of glucose transporter-1 in human gastric carcinoma: association with tumor aggressiveness, metastasis, and patient survival. Cancer. 2001;92:634–41.PubMed
42.
Zurück zum Zitat Chen JQ, Russo J. Dysregulation of glucose transport, glycolysis, TCA cycle and glutaminolysis by oncogenes and tumor suppressors in cancer cells. Biochim Biophys Acta. 2012;1826:370–84.PubMed Chen JQ, Russo J. Dysregulation of glucose transport, glycolysis, TCA cycle and glutaminolysis by oncogenes and tumor suppressors in cancer cells. Biochim Biophys Acta. 2012;1826:370–84.PubMed
43.
Zurück zum Zitat Bongiovanni M, Paone G, Ceriani L, Pusztaszeri MJC, Imaging T. Cellular and molecular basis for thyroid cancer imaging in nuclear medicine. Clin Transl Imaging. 2013;1:149–61. Bongiovanni M, Paone G, Ceriani L, Pusztaszeri MJC, Imaging T. Cellular and molecular basis for thyroid cancer imaging in nuclear medicine. Clin Transl Imaging. 2013;1:149–61.
44.
Zurück zum Zitat Grabellus F, Nagarajah J, Bockisch A, Schmid KW, Sheu SY. Glucose transporter 1 expression, tumor proliferation, and iodine/glucose uptake in thyroid cancer with emphasis on poorly differentiated thyroid carcinoma. Clin Nucl Med. 2012;37:121–7.PubMed Grabellus F, Nagarajah J, Bockisch A, Schmid KW, Sheu SY. Glucose transporter 1 expression, tumor proliferation, and iodine/glucose uptake in thyroid cancer with emphasis on poorly differentiated thyroid carcinoma. Clin Nucl Med. 2012;37:121–7.PubMed
45.
Zurück zum Zitat Haber RS, Weiser KR, Pritsker A, Reder I, Burstein DE. GLUT1 glucose transporter expression in benign and malignant thyroid nodules. Thyroid. 1997;7:363–7.PubMed Haber RS, Weiser KR, Pritsker A, Reder I, Burstein DE. GLUT1 glucose transporter expression in benign and malignant thyroid nodules. Thyroid. 1997;7:363–7.PubMed
46.
Zurück zum Zitat Liu L, Qi L, Knifley T, Piecoro DW, Rychahou P, Liu J, Mitov MI, Martin J, Wang C, Wu J, et al. S100A4 alters metabolism and promotes invasion of lung cancer cells by up-regulating mitochondrial complex I protein NDUFS2. J Biol Chem. 2019;294:7516–27.PubMedPubMedCentral Liu L, Qi L, Knifley T, Piecoro DW, Rychahou P, Liu J, Mitov MI, Martin J, Wang C, Wu J, et al. S100A4 alters metabolism and promotes invasion of lung cancer cells by up-regulating mitochondrial complex I protein NDUFS2. J Biol Chem. 2019;294:7516–27.PubMedPubMedCentral
47.
Zurück zum Zitat Sokolov SS, Balakireva AV, Markova OV, Severin FF. Negative feedback of glycolysis and oxidative phosphorylation: mechanisms of and reasons for it. Biochemistry (Mosc). 2015;80:559–64.PubMed Sokolov SS, Balakireva AV, Markova OV, Severin FF. Negative feedback of glycolysis and oxidative phosphorylation: mechanisms of and reasons for it. Biochemistry (Mosc). 2015;80:559–64.PubMed
49.
Zurück zum Zitat Mazurek S. Pyruvate kinase M2: a key enzyme of the tumor metabolome and its medical relevance. Br J Nutr. 2012;23:133–41. Mazurek S. Pyruvate kinase M2: a key enzyme of the tumor metabolome and its medical relevance. Br J Nutr. 2012;23:133–41.
50.
Zurück zum Zitat Luo W, Semenza GL. Pyruvate kinase M2 regulates glucose metabolism by functioning as a coactivator for hypoxia-inducible factor 1 in cancer cells. Oncotarget. 2011;2:551–6.PubMedPubMedCentral Luo W, Semenza GL. Pyruvate kinase M2 regulates glucose metabolism by functioning as a coactivator for hypoxia-inducible factor 1 in cancer cells. Oncotarget. 2011;2:551–6.PubMedPubMedCentral
51.
Zurück zum Zitat Zahra K, Dey T, Mishra SP, Pandey U. Pyruvate Kinase M2 and cancer: the role of PKM2 in promoting tumorigenesis. Front Oncol. 2020;10:159.PubMedPubMedCentral Zahra K, Dey T, Mishra SP, Pandey U. Pyruvate Kinase M2 and cancer: the role of PKM2 in promoting tumorigenesis. Front Oncol. 2020;10:159.PubMedPubMedCentral
52.
Zurück zum Zitat Amin S, Yang P, Li Z. Pyruvate kinase M2: a multifarious enzyme in non-canonical localization to promote cancer progression. Biochim Biophys Acta Rev Cancer. 2019;1871:331–41.PubMed Amin S, Yang P, Li Z. Pyruvate kinase M2: a multifarious enzyme in non-canonical localization to promote cancer progression. Biochim Biophys Acta Rev Cancer. 2019;1871:331–41.PubMed
53.
Zurück zum Zitat Wang C, Zhang S, Liu J, Tian Y, Ma B, Xu S, Fu Y, Luo Y. Secreted pyruvate kinase M2 promotes lung cancer metastasis through activating the integrin beta1/FAK signaling pathway. Cell Rep. 2020;30:1780-1797.e1786.PubMed Wang C, Zhang S, Liu J, Tian Y, Ma B, Xu S, Fu Y, Luo Y. Secreted pyruvate kinase M2 promotes lung cancer metastasis through activating the integrin beta1/FAK signaling pathway. Cell Rep. 2020;30:1780-1797.e1786.PubMed
54.
Zurück zum Zitat Ye J, Mancuso A, Tong X, Ward PS, Fan J, Rabinowitz JD, Thompson CB. Pyruvate kinase M2 promotes de novo serine synthesis to sustain mTORC1 activity and cell proliferation. Proc Natl Acad Sci USA. 2012;109:6904–9.PubMedPubMedCentral Ye J, Mancuso A, Tong X, Ward PS, Fan J, Rabinowitz JD, Thompson CB. Pyruvate kinase M2 promotes de novo serine synthesis to sustain mTORC1 activity and cell proliferation. Proc Natl Acad Sci USA. 2012;109:6904–9.PubMedPubMedCentral
55.
Zurück zum Zitat Yang M, Vousden KH. Serine and one-carbon metabolism in cancer. Nat Rev Cancer. 2016;16:650–62.PubMed Yang M, Vousden KH. Serine and one-carbon metabolism in cancer. Nat Rev Cancer. 2016;16:650–62.PubMed
56.
Zurück zum Zitat Shulman RG, Rothman DL. The glycogen shunt maintains glycolytic homeostasis and the Warburg effect in cancer. Trends Cancer. 2017;3:761–7.PubMed Shulman RG, Rothman DL. The glycogen shunt maintains glycolytic homeostasis and the Warburg effect in cancer. Trends Cancer. 2017;3:761–7.PubMed
57.
58.
Zurück zum Zitat Hsu MC, Hung WC. Pyruvate kinase M2 fuels multiple aspects of cancer cells: from cellular metabolism, transcriptional regulation to extracellular signaling. Mol Cancer. 2018;17:35.PubMedPubMedCentral Hsu MC, Hung WC. Pyruvate kinase M2 fuels multiple aspects of cancer cells: from cellular metabolism, transcriptional regulation to extracellular signaling. Mol Cancer. 2018;17:35.PubMedPubMedCentral
59.
Zurück zum Zitat Zhu S, Guo Y, Zhang X, Liu H, Yin M, Chen X, Peng C. Pyruvate kinase M2 (PKM2) in cancer and cancer therapeutics. Cancer Lett. 2021;503:240–8.PubMed Zhu S, Guo Y, Zhang X, Liu H, Yin M, Chen X, Peng C. Pyruvate kinase M2 (PKM2) in cancer and cancer therapeutics. Cancer Lett. 2021;503:240–8.PubMed
60.
Zurück zum Zitat Feng C, Gao Y, Wang C, Yu X, Zhang W, Guan H, Shan Z, Teng W. Aberrant overexpression of pyruvate kinase M2 is associated with aggressive tumor features and the BRAF mutation in papillary thyroid cancer. J Clin Endocrinol Metab. 2013;98:E1524-1533.PubMed Feng C, Gao Y, Wang C, Yu X, Zhang W, Guan H, Shan Z, Teng W. Aberrant overexpression of pyruvate kinase M2 is associated with aggressive tumor features and the BRAF mutation in papillary thyroid cancer. J Clin Endocrinol Metab. 2013;98:E1524-1533.PubMed
61.
Zurück zum Zitat Liu B, Song M, Qin H, Zhang B, Liu Y, Sun Y, Ma Y, Shi T. Phosphoribosyl pyrophosphate amidotransferase promotes the progression of thyroid cancer via regulating pyruvate kinase M2. Onco Targets Ther. 2020;13:7629–39.PubMedPubMedCentral Liu B, Song M, Qin H, Zhang B, Liu Y, Sun Y, Ma Y, Shi T. Phosphoribosyl pyrophosphate amidotransferase promotes the progression of thyroid cancer via regulating pyruvate kinase M2. Onco Targets Ther. 2020;13:7629–39.PubMedPubMedCentral
62.
Zurück zum Zitat Zheng B, Geng L, Zeng L, Liu F, Huang Q. AKT2 contributes to increase ovarian cancer cell migration and invasion through the AKT2-PKM2-STAT3/NF-κB axis. Cell Signal. 2018;45:122–31.PubMed Zheng B, Geng L, Zeng L, Liu F, Huang Q. AKT2 contributes to increase ovarian cancer cell migration and invasion through the AKT2-PKM2-STAT3/NF-κB axis. Cell Signal. 2018;45:122–31.PubMed
63.
Zurück zum Zitat Wang B, Liu S, Fan B, Xu X, Chen Y, Lu R, Xu Z, Liu X. PKM2 is involved in neuropathic pain by regulating ERK and STAT3 activation in rat spinal cord. J Headache Pain. 2018;19:7.PubMedPubMedCentral Wang B, Liu S, Fan B, Xu X, Chen Y, Lu R, Xu Z, Liu X. PKM2 is involved in neuropathic pain by regulating ERK and STAT3 activation in rat spinal cord. J Headache Pain. 2018;19:7.PubMedPubMedCentral
64.
Zurück zum Zitat Strickaert A, Corbet C, Spinette SA, Craciun L, Dom G, Andry G, Larsimont D, Wattiez R, Dumont JE, Feron O, Maenhaut C. Reprogramming of energy metabolism: increased expression and roles of pyruvate carboxylase in papillary thyroid cancer. Thyroid. 2019;29:845–57.PubMed Strickaert A, Corbet C, Spinette SA, Craciun L, Dom G, Andry G, Larsimont D, Wattiez R, Dumont JE, Feron O, Maenhaut C. Reprogramming of energy metabolism: increased expression and roles of pyruvate carboxylase in papillary thyroid cancer. Thyroid. 2019;29:845–57.PubMed
65.
Zurück zum Zitat Liu C, Zhou X, Pan Y, Liu Y, Zhang Y. Pyruvate carboxylase promotes thyroid cancer aggressiveness through fatty acid synthesis. BMC Cancer. 2021;21:722.PubMedPubMedCentral Liu C, Zhou X, Pan Y, Liu Y, Zhang Y. Pyruvate carboxylase promotes thyroid cancer aggressiveness through fatty acid synthesis. BMC Cancer. 2021;21:722.PubMedPubMedCentral
66.
Zurück zum Zitat Certo M, Tsai CH, Pucino V, Ho PC, Mauro C. Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat Rev Immunol. 2021;21:151–61.PubMed Certo M, Tsai CH, Pucino V, Ho PC, Mauro C. Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat Rev Immunol. 2021;21:151–61.PubMed
67.
Zurück zum Zitat Wellen KE, Thompson CB. A two-way street: reciprocal regulation of metabolism and signalling. Nat Rev Mol Cell Biol. 2012;13:270–6.PubMed Wellen KE, Thompson CB. A two-way street: reciprocal regulation of metabolism and signalling. Nat Rev Mol Cell Biol. 2012;13:270–6.PubMed
68.
Zurück zum Zitat Kishton RJ, Sukumar M, Restifo NP. Metabolic regulation of T cell longevity and function in tumor immunotherapy. Cell Metab. 2017;26:94–109.PubMedPubMedCentral Kishton RJ, Sukumar M, Restifo NP. Metabolic regulation of T cell longevity and function in tumor immunotherapy. Cell Metab. 2017;26:94–109.PubMedPubMedCentral
69.
Zurück zum Zitat Lee LJ, Papadopoli D, Jewer M, Del Rincon S, Topisirovic I, Lawrence MG, Postovit LM. Cancer plasticity: the role of mRNA translation. Trends Cancer. 2021;7:134–45.PubMed Lee LJ, Papadopoli D, Jewer M, Del Rincon S, Topisirovic I, Lawrence MG, Postovit LM. Cancer plasticity: the role of mRNA translation. Trends Cancer. 2021;7:134–45.PubMed
70.
Zurück zum Zitat Icard P, Shulman S, Farhat D, Steyaert JM, Alifano M, Lincet H. How the Warburg effect supports aggressiveness and drug resistance of cancer cells? Drug Resist Updat. 2018;38:1–11.PubMed Icard P, Shulman S, Farhat D, Steyaert JM, Alifano M, Lincet H. How the Warburg effect supports aggressiveness and drug resistance of cancer cells? Drug Resist Updat. 2018;38:1–11.PubMed
71.
Zurück zum Zitat Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, Matos C, Bruss C, Klobuch S, Peter K, et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 2016;24:657–71.PubMed Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, Matos C, Bruss C, Klobuch S, Peter K, et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 2016;24:657–71.PubMed
72.
Zurück zum Zitat Peng M, Yin N, Chhangawala S, Xu K, Leslie CS, Li MO. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science. 2016;354:481–4.PubMedPubMedCentral Peng M, Yin N, Chhangawala S, Xu K, Leslie CS, Li MO. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science. 2016;354:481–4.PubMedPubMedCentral
73.
Zurück zum Zitat Kurpińska A, Suraj J, Bonar E, Zakrzewska A, Stojak M, Sternak M, Jasztal A, Walczak M. Proteomic characterization of early lung response to breast cancer metastasis in mice. Exp Mol Pathol. 2019;107:129–40.PubMed Kurpińska A, Suraj J, Bonar E, Zakrzewska A, Stojak M, Sternak M, Jasztal A, Walczak M. Proteomic characterization of early lung response to breast cancer metastasis in mice. Exp Mol Pathol. 2019;107:129–40.PubMed
74.
Zurück zum Zitat Mishra D, Banerjee DJC. Lactate dehydrogenases as metabolic links between tumor and stroma in the tumor microenvironment. Cancers (Basel). 2019;11:750.PubMed Mishra D, Banerjee DJC. Lactate dehydrogenases as metabolic links between tumor and stroma in the tumor microenvironment. Cancers (Basel). 2019;11:750.PubMed
75.
Zurück zum Zitat Wang JM, Jiang JY, Zhang DL, Du X, Wu T, Du ZX. HYOU1 facilitates proliferation, invasion and glycolysis of papillary thyroid cancer via stabilizing LDHB mRNA. J Cell Mol Med. 2021;25:4814–25.PubMedCentral Wang JM, Jiang JY, Zhang DL, Du X, Wu T, Du ZX. HYOU1 facilitates proliferation, invasion and glycolysis of papillary thyroid cancer via stabilizing LDHB mRNA. J Cell Mol Med. 2021;25:4814–25.PubMedCentral
76.
Zurück zum Zitat San-Millán I, Brooks GA. Reexamining cancer metabolism: lactate production for carcinogenesis could be the purpose and explanation of the Warburg effect. Carcinogenesis. 2017;38:119–33.PubMed San-Millán I, Brooks GA. Reexamining cancer metabolism: lactate production for carcinogenesis could be the purpose and explanation of the Warburg effect. Carcinogenesis. 2017;38:119–33.PubMed
77.
Zurück zum Zitat Lis P, Dyląg M, Niedźwiecka K, Ko YH, Pedersen PL, Goffeau A, Ułaszewski S. The HK2 dependent “Warburg effect” and mitochondrial oxidative phosphorylation in cancer: targets for effective therapy with 3-bromopyruvate. Molecules. 2016;21:1730.PubMedPubMedCentral Lis P, Dyląg M, Niedźwiecka K, Ko YH, Pedersen PL, Goffeau A, Ułaszewski S. The HK2 dependent “Warburg effect” and mitochondrial oxidative phosphorylation in cancer: targets for effective therapy with 3-bromopyruvate. Molecules. 2016;21:1730.PubMedPubMedCentral
78.
Zurück zum Zitat Lebelo MT, Joubert AM, Visagie MH. Warburg effect and its role in tumourigenesis. Arch Pharm Res. 2019;42:833–47.PubMed Lebelo MT, Joubert AM, Visagie MH. Warburg effect and its role in tumourigenesis. Arch Pharm Res. 2019;42:833–47.PubMed
79.
Zurück zum Zitat Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A, Kreutz M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood. 2006;107:2013–21.PubMed Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A, Kreutz M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood. 2006;107:2013–21.PubMed
80.
Zurück zum Zitat Xia H, Wang W, Crespo J, Kryczek I, Li W, Wei S, Bian Z, Maj T, He M, Liu RJ, et al. Suppression of FIP200 and autophagy by tumor-derived lactate promotes naïve T cell apoptosis and affects tumor immunity. Sci Immunol. 2017;2:eaan4631.PubMedPubMedCentral Xia H, Wang W, Crespo J, Kryczek I, Li W, Wei S, Bian Z, Maj T, He M, Liu RJ, et al. Suppression of FIP200 and autophagy by tumor-derived lactate promotes naïve T cell apoptosis and affects tumor immunity. Sci Immunol. 2017;2:eaan4631.PubMedPubMedCentral
81.
Zurück zum Zitat Zhao B, Aggarwal A, Im SY, Viswanathan K, Landa I, Nehs MA. Effect of lactate export inhibition on anaplastic thyroid cancer growth and metabolism. J Am Coll Surg. 2022;234:1044–50.PubMed Zhao B, Aggarwal A, Im SY, Viswanathan K, Landa I, Nehs MA. Effect of lactate export inhibition on anaplastic thyroid cancer growth and metabolism. J Am Coll Surg. 2022;234:1044–50.PubMed
82.
Zurück zum Zitat Hoxhaj G, Manning BD. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer. 2020;20:74–88.PubMed Hoxhaj G, Manning BD. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer. 2020;20:74–88.PubMed
83.
Zurück zum Zitat DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7:11–20.PubMed DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7:11–20.PubMed
84.
Zurück zum Zitat Osthus RC, Shim H, Kim S, Li Q, Reddy R, Mukherjee M, Xu Y, Wonsey D, Lee LA, Dang CV. Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J Biol Chem. 2000;275:21797–800.PubMed Osthus RC, Shim H, Kim S, Li Q, Reddy R, Mukherjee M, Xu Y, Wonsey D, Lee LA, Dang CV. Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J Biol Chem. 2000;275:21797–800.PubMed
85.
Zurück zum Zitat Abbaszadeh Z, Çeşmeli S, Biray Avcı Ç. Crucial players in glycolysis: cancer progress. Gene. 2020;726: 144158.PubMed Abbaszadeh Z, Çeşmeli S, Biray Avcı Ç. Crucial players in glycolysis: cancer progress. Gene. 2020;726: 144158.PubMed
86.
Zurück zum Zitat Boedtkjer E, Pedersen SF. The acidic tumor microenvironment as a driver of cancer. Annu Rev Physiol. 2020;82:103–26.PubMed Boedtkjer E, Pedersen SF. The acidic tumor microenvironment as a driver of cancer. Annu Rev Physiol. 2020;82:103–26.PubMed
87.
Zurück zum Zitat Epstein T, Gatenby RA, Brown JS. The Warburg effect as an adaptation of cancer cells to rapid fluctuations in energy demand. PLoS ONE. 2017;12(9): e0185085.PubMedPubMedCentral Epstein T, Gatenby RA, Brown JS. The Warburg effect as an adaptation of cancer cells to rapid fluctuations in energy demand. PLoS ONE. 2017;12(9): e0185085.PubMedPubMedCentral
88.
Zurück zum Zitat Zhang X, Ai Z, Chen J, Yi J, Liu Z, Zhao H, Wei H. Glycometabolic adaptation mediates the insensitivity of drug-resistant K562/ADM leukaemia cells to adriamycin via the AKT-mTOR/c-Myc signalling pathway. Mol Med Rep. 2017;15:1869–76.PubMed Zhang X, Ai Z, Chen J, Yi J, Liu Z, Zhao H, Wei H. Glycometabolic adaptation mediates the insensitivity of drug-resistant K562/ADM leukaemia cells to adriamycin via the AKT-mTOR/c-Myc signalling pathway. Mol Med Rep. 2017;15:1869–76.PubMed
89.
Zurück zum Zitat Ferro F, Servais S, Besson P, Roger S, Dumas JF, Brisson L. Autophagy and mitophagy in cancer metabolic remodelling. Semin Cell Dev Biol. 2020;98:129–38.PubMed Ferro F, Servais S, Besson P, Roger S, Dumas JF, Brisson L. Autophagy and mitophagy in cancer metabolic remodelling. Semin Cell Dev Biol. 2020;98:129–38.PubMed
90.
Zurück zum Zitat Du J, Liu J, Smith BJ, Tsao MS, Cullen JJ. Role of Rac1-dependent NADPH oxidase in the growth of pancreatic cancer. Cancer Gene Ther. 2011;18:135–43.PubMed Du J, Liu J, Smith BJ, Tsao MS, Cullen JJ. Role of Rac1-dependent NADPH oxidase in the growth of pancreatic cancer. Cancer Gene Ther. 2011;18:135–43.PubMed
91.
Zurück zum Zitat Bisevac JP, Djukic M, Stanojevic I, Stevanovic I, Mijuskovic Z, Djuric A, Gobeljic B, Banovic T, Vojvodic D. Association between oxidative stress and melanoma progression. J Med Biochem. 2018;37:12–20.PubMedPubMedCentral Bisevac JP, Djukic M, Stanojevic I, Stevanovic I, Mijuskovic Z, Djuric A, Gobeljic B, Banovic T, Vojvodic D. Association between oxidative stress and melanoma progression. J Med Biochem. 2018;37:12–20.PubMedPubMedCentral
93.
Zurück zum Zitat Nagarajah J, Le M, Knauf JA, Ferrandino G, Montero-Conde C, Pillarsetty N, Bolaender A, Irwin C, Krishnamoorthy GP, Saqcena M, et al. Sustained ERK inhibition maximizes responses of BrafV600E thyroid cancers to radioiodine. J Clin Invest. 2016;126:4119–24.PubMedPubMedCentral Nagarajah J, Le M, Knauf JA, Ferrandino G, Montero-Conde C, Pillarsetty N, Bolaender A, Irwin C, Krishnamoorthy GP, Saqcena M, et al. Sustained ERK inhibition maximizes responses of BrafV600E thyroid cancers to radioiodine. J Clin Invest. 2016;126:4119–24.PubMedPubMedCentral
95.
Zurück zum Zitat Kumar SM, Yu H, Edwards R, Chen L, Kazianis S, Brafford P, Acs G, Herlyn M, Xu X. Mutant V600E BRAF increases hypoxia inducible factor-1alpha expression in melanoma. Cancer Res. 2007;67:3177–84.PubMed Kumar SM, Yu H, Edwards R, Chen L, Kazianis S, Brafford P, Acs G, Herlyn M, Xu X. Mutant V600E BRAF increases hypoxia inducible factor-1alpha expression in melanoma. Cancer Res. 2007;67:3177–84.PubMed
96.
Zurück zum Zitat Haq R, Fisher DE, Widlund HR. Molecular pathways: BRAF induces bioenergetic adaptation by attenuating oxidative phosphorylation. Clin Cancer Res. 2014;20:2257–63.PubMedPubMedCentral Haq R, Fisher DE, Widlund HR. Molecular pathways: BRAF induces bioenergetic adaptation by attenuating oxidative phosphorylation. Clin Cancer Res. 2014;20:2257–63.PubMedPubMedCentral
97.
Zurück zum Zitat Sandulache VC, Skinner HD, Wang Y, Chen Y, Dodge CT, Ow TJ, Bankson JA, Myers JN, Lai SY. Glycolytic inhibition alters anaplastic thyroid carcinoma tumor metabolism and improves response to conventional chemotherapy and radiation. Mol Cancer Ther. 2012;11:1373–80.PubMed Sandulache VC, Skinner HD, Wang Y, Chen Y, Dodge CT, Ow TJ, Bankson JA, Myers JN, Lai SY. Glycolytic inhibition alters anaplastic thyroid carcinoma tumor metabolism and improves response to conventional chemotherapy and radiation. Mol Cancer Ther. 2012;11:1373–80.PubMed
98.
Zurück zum Zitat Li Y, Qin J, He Z, Cui G, Zhang K, Wu B. Knockdown of circPUM1 impedes cell growth, metastasis and glycolysis of papillary thyroid cancer via enhancing MAPK1 expression by serving as the sponge of miR-21-5p. Genes Genomics. 2021;43:141–50.PubMed Li Y, Qin J, He Z, Cui G, Zhang K, Wu B. Knockdown of circPUM1 impedes cell growth, metastasis and glycolysis of papillary thyroid cancer via enhancing MAPK1 expression by serving as the sponge of miR-21-5p. Genes Genomics. 2021;43:141–50.PubMed
99.
Zurück zum Zitat Milosevic Z, Pesic M, Stankovic T, Dinic J, Milovanovic Z, Stojsic J, Dzodic R, Tanic N, Bankovic J. Targeting RAS-MAPK-ERK and PI3K-AKT-mTOR signal transduction pathways to chemosensitize anaplastic thyroid carcinoma. Transl Res. 2014;164:411–23.PubMed Milosevic Z, Pesic M, Stankovic T, Dinic J, Milovanovic Z, Stojsic J, Dzodic R, Tanic N, Bankovic J. Targeting RAS-MAPK-ERK and PI3K-AKT-mTOR signal transduction pathways to chemosensitize anaplastic thyroid carcinoma. Transl Res. 2014;164:411–23.PubMed
100.
Zurück zum Zitat Gao Y, Yang F, Yang XA, Zhang L, Yu H, Cheng X, Xu S, Pan J, Wang K, Li P. Mitochondrial metabolism is inhibited by the HIF1α-MYC-PGC-1β axis in BRAF V600E thyroid cancer. FEBS j. 2019;286:1420–36.PubMed Gao Y, Yang F, Yang XA, Zhang L, Yu H, Cheng X, Xu S, Pan J, Wang K, Li P. Mitochondrial metabolism is inhibited by the HIF1α-MYC-PGC-1β axis in BRAF V600E thyroid cancer. FEBS j. 2019;286:1420–36.PubMed
101.
Zurück zum Zitat Wang HM, Huang YW, Huang JS, Wang CH, Kok VC, Hung CM, Chen HM, Tzen CY. Anaplastic carcinoma of the thyroid arising more often from follicular carcinoma than papillary carcinoma. Ann Surg Oncol. 2007;14:3011–8.PubMed Wang HM, Huang YW, Huang JS, Wang CH, Kok VC, Hung CM, Chen HM, Tzen CY. Anaplastic carcinoma of the thyroid arising more often from follicular carcinoma than papillary carcinoma. Ann Surg Oncol. 2007;14:3011–8.PubMed
102.
Zurück zum Zitat Liu Z, Hou P, Ji M, Guan H, Studeman K, Jensen K, Vasko V, El-Naggar AK, Xing M. Highly prevalent genetic alterations in receptor tyrosine kinases and phosphatidylinositol 3-kinase/akt and mitogen-activated protein kinase pathways in anaplastic and follicular thyroid cancers. J Clin Endocrinol Metab. 2008;93:3106–16.PubMed Liu Z, Hou P, Ji M, Guan H, Studeman K, Jensen K, Vasko V, El-Naggar AK, Xing M. Highly prevalent genetic alterations in receptor tyrosine kinases and phosphatidylinositol 3-kinase/akt and mitogen-activated protein kinase pathways in anaplastic and follicular thyroid cancers. J Clin Endocrinol Metab. 2008;93:3106–16.PubMed
103.
Zurück zum Zitat Smallridge RC, Copland JA. Anaplastic thyroid carcinoma: pathogenesis and emerging therapies. Clin Oncol (R Coll Radiol). 2010;22:486–97.PubMed Smallridge RC, Copland JA. Anaplastic thyroid carcinoma: pathogenesis and emerging therapies. Clin Oncol (R Coll Radiol). 2010;22:486–97.PubMed
104.
Zurück zum Zitat Agrawal N, Akbani R, Aksoy BA, Ally A, Arachchi H, Asa SL, Auman JT, Balasundaram M, Balu S, Baylin SB. Integrated genomic characterization of papillary thyroid carcinoma. Cell. 2014;159:676–90.PubMedCentral Agrawal N, Akbani R, Aksoy BA, Ally A, Arachchi H, Asa SL, Auman JT, Balasundaram M, Balu S, Baylin SB. Integrated genomic characterization of papillary thyroid carcinoma. Cell. 2014;159:676–90.PubMedCentral
105.
Zurück zum Zitat Liu R, Chen Y, Liu G, Li C, Song Y, Cao Z, Li W, Hu J, Lu C, Liu Y. PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis. 2020;11:797.PubMedPubMedCentral Liu R, Chen Y, Liu G, Li C, Song Y, Cao Z, Li W, Hu J, Lu C, Liu Y. PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis. 2020;11:797.PubMedPubMedCentral
106.
Zurück zum Zitat Jiang W, Zhu Z, Thompson HJ. Effects of limiting energy availability via diet and physical activity on mammalian target of rapamycin-related signaling in rat mammary carcinomas. Carcinogenesis. 2013;34:378–87.PubMed Jiang W, Zhu Z, Thompson HJ. Effects of limiting energy availability via diet and physical activity on mammalian target of rapamycin-related signaling in rat mammary carcinomas. Carcinogenesis. 2013;34:378–87.PubMed
107.
Zurück zum Zitat Sun Q, Chen X, Ma J, Peng H, Wang F, Zha X, Wang Y, Jing Y, Yang H, Chen R, et al. Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic glycolysis and tumor growth. Proc Natl Acad Sci USA. 2011;108:4129–34.PubMedPubMedCentral Sun Q, Chen X, Ma J, Peng H, Wang F, Zha X, Wang Y, Jing Y, Yang H, Chen R, et al. Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic glycolysis and tumor growth. Proc Natl Acad Sci USA. 2011;108:4129–34.PubMedPubMedCentral
108.
Zurück zum Zitat Fan H, Wu Y, Yu S, Li X, Wang A, Wang S, Chen W, Lu Y. Critical role of mTOR in regulating aerobic glycolysis in carcinogenesis. Int J Oncol. 2021;58:9–19.PubMed Fan H, Wu Y, Yu S, Li X, Wang A, Wang S, Chen W, Lu Y. Critical role of mTOR in regulating aerobic glycolysis in carcinogenesis. Int J Oncol. 2021;58:9–19.PubMed
109.
Zurück zum Zitat Liotti F, Kumar N, Prevete N, Marotta M, Sorriento D, Ieranò C, Ronchi A, Marino FZ, Moretti S, Colella R, et al. PD-1 blockade delays tumor growth by inhibiting an intrinsic SHP2/Ras/MAPK signalling in thyroid cancer cells. J Exp Clin Cancer Res. 2021;40:22.PubMedPubMedCentral Liotti F, Kumar N, Prevete N, Marotta M, Sorriento D, Ieranò C, Ronchi A, Marino FZ, Moretti S, Colella R, et al. PD-1 blockade delays tumor growth by inhibiting an intrinsic SHP2/Ras/MAPK signalling in thyroid cancer cells. J Exp Clin Cancer Res. 2021;40:22.PubMedPubMedCentral
110.
Zurück zum Zitat Saji M, Ringel MD. The PI3K-Akt-mTOR pathway in initiation and progression of thyroid tumors. Mol Cell Endocrinol. 2010;321:20–8.PubMed Saji M, Ringel MD. The PI3K-Akt-mTOR pathway in initiation and progression of thyroid tumors. Mol Cell Endocrinol. 2010;321:20–8.PubMed
111.
Zurück zum Zitat Jin N, Jiang T, Rosen DM, Nelkin BD, Ball DW. Dual inhibition of mitogen-activated protein kinase kinase and mammalian target of rapamycin in differentiated and anaplastic thyroid cancer. J Clin Endocrinol Metab. 2009;94:4107–12.PubMedPubMedCentral Jin N, Jiang T, Rosen DM, Nelkin BD, Ball DW. Dual inhibition of mitogen-activated protein kinase kinase and mammalian target of rapamycin in differentiated and anaplastic thyroid cancer. J Clin Endocrinol Metab. 2009;94:4107–12.PubMedPubMedCentral
112.
Zurück zum Zitat Liu D, Xing M. Potent inhibition of thyroid cancer cells by the MEK inhibitor PD0325901 and its potentiation by suppression of the PI3K and NF-kappaB pathways. Thyroid. 2008;18:853–64.PubMedPubMedCentral Liu D, Xing M. Potent inhibition of thyroid cancer cells by the MEK inhibitor PD0325901 and its potentiation by suppression of the PI3K and NF-kappaB pathways. Thyroid. 2008;18:853–64.PubMedPubMedCentral
113.
Zurück zum Zitat Glassmann A, Winter J, Kraus D, Veit N, Probstmeier R. Pharmacological suppression of the Ras/MAPK pathway in thyroid carcinoma cells can provoke opposite effects on cell migration and proliferation: the appearance of yin-yang effects and the need of combinatorial treatments. Int J Oncol. 2014;45:2587–95.PubMed Glassmann A, Winter J, Kraus D, Veit N, Probstmeier R. Pharmacological suppression of the Ras/MAPK pathway in thyroid carcinoma cells can provoke opposite effects on cell migration and proliferation: the appearance of yin-yang effects and the need of combinatorial treatments. Int J Oncol. 2014;45:2587–95.PubMed
114.
Zurück zum Zitat Lyssiotis CA, Kimmelman AC. Metabolic interactions in the tumor microenvironment. Trends Cell Biol. 2017;27:863–75.PubMedPubMedCentral Lyssiotis CA, Kimmelman AC. Metabolic interactions in the tumor microenvironment. Trends Cell Biol. 2017;27:863–75.PubMedPubMedCentral
115.
Zurück zum Zitat Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer. 2004;4:891–9.PubMed Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer. 2004;4:891–9.PubMed
116.
Zurück zum Zitat Huo N, Cong R, Sun ZJ, Li WC, Zhu X, Xue CY, Chen Z, Ma LY, Chu Z, Han YC, et al. STAT3/LINC00671 axis regulates papillary thyroid tumor growth and metastasis via LDHA-mediated glycolysis. Cell Death Dis. 2021;12:799.PubMedPubMedCentral Huo N, Cong R, Sun ZJ, Li WC, Zhu X, Xue CY, Chen Z, Ma LY, Chu Z, Han YC, et al. STAT3/LINC00671 axis regulates papillary thyroid tumor growth and metastasis via LDHA-mediated glycolysis. Cell Death Dis. 2021;12:799.PubMedPubMedCentral
117.
Zurück zum Zitat Cai H, Li J, Zhang Y, Liao Y, Zhu Y, Wang C, Hou J. LDHA promotes oral squamous cell carcinoma progression through facilitating glycolysis and epithelial-mesenchymal transition. Front Oncol. 2019;9:1446.PubMedPubMedCentral Cai H, Li J, Zhang Y, Liao Y, Zhu Y, Wang C, Hou J. LDHA promotes oral squamous cell carcinoma progression through facilitating glycolysis and epithelial-mesenchymal transition. Front Oncol. 2019;9:1446.PubMedPubMedCentral
118.
Zurück zum Zitat Ban EJ, Kim D, Kim JK, Kang SW, Lee J, Jeong JJ, Nam KH, Chung WY, Kim K. Lactate dehydrogenase A as a potential new biomarker for thyroid cancer. Endocrinol Metab (Seoul). 2021;36:96–105.PubMed Ban EJ, Kim D, Kim JK, Kang SW, Lee J, Jeong JJ, Nam KH, Chung WY, Kim K. Lactate dehydrogenase A as a potential new biomarker for thyroid cancer. Endocrinol Metab (Seoul). 2021;36:96–105.PubMed
119.
Zurück zum Zitat Cardone RA, Alfarouk KO, Elliott RL, Alqahtani SS, Ahmed SB, Aljarbou AN, Greco MR, Cannone S, Reshkin SJ. The role of sodium hydrogen exchanger 1 in dysregulation of proton dynamics and reprogramming of cancer metabolism as a sequela. Int J Mol Sci. 2019;20:3694.PubMedPubMedCentral Cardone RA, Alfarouk KO, Elliott RL, Alqahtani SS, Ahmed SB, Aljarbou AN, Greco MR, Cannone S, Reshkin SJ. The role of sodium hydrogen exchanger 1 in dysregulation of proton dynamics and reprogramming of cancer metabolism as a sequela. Int J Mol Sci. 2019;20:3694.PubMedPubMedCentral
120.
Zurück zum Zitat Fanelli A, Grollman EF, Wang D, Philp NJ. MCT1 and its accessory protein CD147 are differentially regulated by TSH in rat thyroid cells. Am J Physiol Endocrinol Metab. 2003;285:E1223-1229.PubMed Fanelli A, Grollman EF, Wang D, Philp NJ. MCT1 and its accessory protein CD147 are differentially regulated by TSH in rat thyroid cells. Am J Physiol Endocrinol Metab. 2003;285:E1223-1229.PubMed
122.
Zurück zum Zitat Cassim S, Pouyssegur J. Tumor microenvironment: a metabolic player that shapes the immune response. Int J Mol Sci. 2019;21:157.PubMedPubMedCentral Cassim S, Pouyssegur J. Tumor microenvironment: a metabolic player that shapes the immune response. Int J Mol Sci. 2019;21:157.PubMedPubMedCentral
123.
Zurück zum Zitat Xia A, Wu Y. Joint interactions of carbon and nitrogen metabolism dominated by bicarbonate and nitrogen in Orychophragmus violaceus and Brassica napus under simulated karst habitats. BMC Plant Biol. 2022;22:264.PubMedPubMedCentral Xia A, Wu Y. Joint interactions of carbon and nitrogen metabolism dominated by bicarbonate and nitrogen in Orychophragmus violaceus and Brassica napus under simulated karst habitats. BMC Plant Biol. 2022;22:264.PubMedPubMedCentral
124.
Zurück zum Zitat Park HJ, Lyons JC, Ohtsubo T, Song CW. Acidic environment causes apoptosis by increasing caspase activity. Br J Cancer. 1999;80:1892–7.PubMedPubMedCentral Park HJ, Lyons JC, Ohtsubo T, Song CW. Acidic environment causes apoptosis by increasing caspase activity. Br J Cancer. 1999;80:1892–7.PubMedPubMedCentral
125.
Zurück zum Zitat Maiuri MC, Tasdemir E, Criollo A, Morselli E, Vicencio JM, Carnuccio R, Kroemer G. Control of autophagy by oncogenes and tumor suppressor genes. Cell Death Differ. 2009;16:87–93.PubMed Maiuri MC, Tasdemir E, Criollo A, Morselli E, Vicencio JM, Carnuccio R, Kroemer G. Control of autophagy by oncogenes and tumor suppressor genes. Cell Death Differ. 2009;16:87–93.PubMed
126.
Zurück zum Zitat Kierans SJ, Taylor CT. Regulation of glycolysis by the hypoxia-inducible factor (HIF): implications for cellular physiology. J Physiol. 2021;599:23–37.PubMed Kierans SJ, Taylor CT. Regulation of glycolysis by the hypoxia-inducible factor (HIF): implications for cellular physiology. J Physiol. 2021;599:23–37.PubMed
127.
Zurück zum Zitat Zheng T, Jäättelä M, Liu B. pH gradient reversal fuels cancer progression. Int J Biochem Cell Biol. 2020;125: 105796.PubMed Zheng T, Jäättelä M, Liu B. pH gradient reversal fuels cancer progression. Int J Biochem Cell Biol. 2020;125: 105796.PubMed
128.
Zurück zum Zitat Niu D, Luo T, Wang H, Xia Y, Xie Z. Lactic acid in tumor invasion. Clin Chim Acta. 2021;522:61–9.PubMed Niu D, Luo T, Wang H, Xia Y, Xie Z. Lactic acid in tumor invasion. Clin Chim Acta. 2021;522:61–9.PubMed
129.
Zurück zum Zitat Kato Y, Ozawa S, Miyamoto C, Maehata Y, Suzuki A, Maeda T, Baba Y. Acidic extracellular microenvironment and cancer. Cancer Cell Int. 2013;13:89.PubMedPubMedCentral Kato Y, Ozawa S, Miyamoto C, Maehata Y, Suzuki A, Maeda T, Baba Y. Acidic extracellular microenvironment and cancer. Cancer Cell Int. 2013;13:89.PubMedPubMedCentral
130.
Zurück zum Zitat Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, Chen Q, Gindin M, Gubin MM, van der Windt GJ, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162:1229–41.PubMedPubMedCentral Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, Chen Q, Gindin M, Gubin MM, van der Windt GJ, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162:1229–41.PubMedPubMedCentral
131.
Zurück zum Zitat El Hassouni B, Granchi C, Vallés-Martí A, Supadmanaba IGP, Bononi G, Tuccinardi T, Funel N, Jimenez CR, Peters GJ, Giovannetti E, Minutolo F. The dichotomous role of the glycolytic metabolism pathway in cancer metastasis: interplay with the complex tumor microenvironment and novel therapeutic strategies. Semin Cancer Biol. 2020;60:238–48.PubMed El Hassouni B, Granchi C, Vallés-Martí A, Supadmanaba IGP, Bononi G, Tuccinardi T, Funel N, Jimenez CR, Peters GJ, Giovannetti E, Minutolo F. The dichotomous role of the glycolytic metabolism pathway in cancer metastasis: interplay with the complex tumor microenvironment and novel therapeutic strategies. Semin Cancer Biol. 2020;60:238–48.PubMed
132.
Zurück zum Zitat Bettum IJ, Gorad SS, Barkovskaya A, Pettersen S, Moestue SA, Vasiliauskaite K, Tenstad E, Øyjord T, Risa Ø, Nygaard V, et al. Metabolic reprogramming supports the invasive phenotype in malignant melanoma. Cancer Lett. 2015;366:71–83.PubMed Bettum IJ, Gorad SS, Barkovskaya A, Pettersen S, Moestue SA, Vasiliauskaite K, Tenstad E, Øyjord T, Risa Ø, Nygaard V, et al. Metabolic reprogramming supports the invasive phenotype in malignant melanoma. Cancer Lett. 2015;366:71–83.PubMed
133.
Zurück zum Zitat Yang J, Ren B, Yang G, Wang H, Chen G, You L, Zhang T, Zhao Y. The enhancement of glycolysis regulates pancreatic cancer metastasis. Cell Mol Life Sci. 2020;77:305–21.PubMed Yang J, Ren B, Yang G, Wang H, Chen G, You L, Zhang T, Zhao Y. The enhancement of glycolysis regulates pancreatic cancer metastasis. Cell Mol Life Sci. 2020;77:305–21.PubMed
134.
Zurück zum Zitat Clambey ET, McNamee EN, Westrich JA, Glover LE, Campbell EL, Jedlicka P, de Zoeten EF, Cambier JC, Stenmark KR, Colgan SP, Eltzschig HK. Hypoxia-inducible factor-1 alpha-dependent induction of FoxP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc Natl Acad Sci USA. 2012;109:E2784-2793.PubMedPubMedCentral Clambey ET, McNamee EN, Westrich JA, Glover LE, Campbell EL, Jedlicka P, de Zoeten EF, Cambier JC, Stenmark KR, Colgan SP, Eltzschig HK. Hypoxia-inducible factor-1 alpha-dependent induction of FoxP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc Natl Acad Sci USA. 2012;109:E2784-2793.PubMedPubMedCentral
135.
Zurück zum Zitat Fong GH, Takeda K. Role and regulation of prolyl hydroxylase domain proteins. Cell Death Differ. 2008;15:635–41.PubMed Fong GH, Takeda K. Role and regulation of prolyl hydroxylase domain proteins. Cell Death Differ. 2008;15:635–41.PubMed
136.
Zurück zum Zitat Sasidharan Nair V, Saleh R, Toor SM, Cyprian FS, Elkord E. Metabolic reprogramming of T regulatory cells in the hypoxic tumor microenvironment. Cancer Immunol Immunother. 2021;70:2103–21.PubMedPubMedCentral Sasidharan Nair V, Saleh R, Toor SM, Cyprian FS, Elkord E. Metabolic reprogramming of T regulatory cells in the hypoxic tumor microenvironment. Cancer Immunol Immunother. 2021;70:2103–21.PubMedPubMedCentral
137.
Zurück zum Zitat Beckert S, Farrahi F, Aslam RS, Scheuenstuhl H, Königsrainer A, Hussain MZ, Hunt TK. Lactate stimulates endothelial cell migration. Wound Repair Regen. 2006;14:321–4.PubMed Beckert S, Farrahi F, Aslam RS, Scheuenstuhl H, Königsrainer A, Hussain MZ, Hunt TK. Lactate stimulates endothelial cell migration. Wound Repair Regen. 2006;14:321–4.PubMed
138.
139.
140.
Zurück zum Zitat van Zijl F, Krupitza G, Mikulits W. Initial steps of metastasis: cell invasion and endothelial transmigration. Mutat Res. 2011;728:23–34.PubMedPubMedCentral van Zijl F, Krupitza G, Mikulits W. Initial steps of metastasis: cell invasion and endothelial transmigration. Mutat Res. 2011;728:23–34.PubMedPubMedCentral
141.
Zurück zum Zitat Ye X, Weinberg RA. Epithelial-mesenchymal plasticity: a central regulator of cancer progression. Trends Cell Biol. 2015;25:675–86.PubMedPubMedCentral Ye X, Weinberg RA. Epithelial-mesenchymal plasticity: a central regulator of cancer progression. Trends Cell Biol. 2015;25:675–86.PubMedPubMedCentral
142.
Zurück zum Zitat Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15:178–96.PubMedPubMedCentral Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15:178–96.PubMedPubMedCentral
143.
Zurück zum Zitat Thews O, Riemann A. Tumor pH and metastasis: a malignant process beyond hypoxia. Cancer Metastasis Rev. 2019;38:113–29.PubMed Thews O, Riemann A. Tumor pH and metastasis: a malignant process beyond hypoxia. Cancer Metastasis Rev. 2019;38:113–29.PubMed
144.
Zurück zum Zitat Spiegel A, Brooks MW, Houshyar S, Reinhardt F, Ardolino M, Fessler E, Chen MB, Krall JA, DeCock J, Zervantonakis IK, et al. Neutrophils suppress intraluminal NK cell-mediated tumor cell clearance and enhance extravasation of disseminated carcinoma cells. Cancer Discov. 2016;6:630–49.PubMedPubMedCentral Spiegel A, Brooks MW, Houshyar S, Reinhardt F, Ardolino M, Fessler E, Chen MB, Krall JA, DeCock J, Zervantonakis IK, et al. Neutrophils suppress intraluminal NK cell-mediated tumor cell clearance and enhance extravasation of disseminated carcinoma cells. Cancer Discov. 2016;6:630–49.PubMedPubMedCentral
145.
Zurück zum Zitat Schmid KW. Lymph node and distant metastases of thyroid gland cancer. Metastases in the thyroid glands. Pathologe. 2015;36(Suppl 2):171–5.PubMed Schmid KW. Lymph node and distant metastases of thyroid gland cancer. Metastases in the thyroid glands. Pathologe. 2015;36(Suppl 2):171–5.PubMed
146.
Zurück zum Zitat Hirsch D, Levy S, Tsvetov G, Gorshtein A, Slutzky-Shraga I, Akirov A, Robenshtok E, Shimon I, Benbassat CA. Long-term outcomes and prognostic factors in patients with differentiated thyroid cancer and distant metastases. Endocr Pract. 2017;23:1193–200.PubMed Hirsch D, Levy S, Tsvetov G, Gorshtein A, Slutzky-Shraga I, Akirov A, Robenshtok E, Shimon I, Benbassat CA. Long-term outcomes and prognostic factors in patients with differentiated thyroid cancer and distant metastases. Endocr Pract. 2017;23:1193–200.PubMed
147.
Zurück zum Zitat Hou XM, Yuan SQ, Zhao D, Liu XJ, Wu XA. LDH-A promotes malignant behavior via activation of epithelial-to-mesenchymal transition in lung adenocarcinoma. Biosci Rep. 2019;39:BSR20181476.PubMedPubMedCentral Hou XM, Yuan SQ, Zhao D, Liu XJ, Wu XA. LDH-A promotes malignant behavior via activation of epithelial-to-mesenchymal transition in lung adenocarcinoma. Biosci Rep. 2019;39:BSR20181476.PubMedPubMedCentral
148.
Zurück zum Zitat Wang J, Ye C, Chen C, Xiong H, Xie B, Zhou J, Chen Y, Zheng S, Wang L. Glucose transporter GLUT1 expression and clinical outcome in solid tumors: a systematic review and meta-analysis. Oncotarget. 2017;8:16875–86.PubMedPubMedCentral Wang J, Ye C, Chen C, Xiong H, Xie B, Zhou J, Chen Y, Zheng S, Wang L. Glucose transporter GLUT1 expression and clinical outcome in solid tumors: a systematic review and meta-analysis. Oncotarget. 2017;8:16875–86.PubMedPubMedCentral
149.
Zurück zum Zitat Zhang B, Xie Z, Li B. The clinicopathologic impacts and prognostic significance of GLUT1 expression in patients with lung cancer: a meta-analysis. Gene. 2019;689:76–83.PubMed Zhang B, Xie Z, Li B. The clinicopathologic impacts and prognostic significance of GLUT1 expression in patients with lung cancer: a meta-analysis. Gene. 2019;689:76–83.PubMed
150.
Zurück zum Zitat Zhang JZ, Behrooz A, Ismail-Beigi F. Regulation of glucose transport by hypoxia. Am J Kidney Dis. 1999;34:189–202.PubMed Zhang JZ, Behrooz A, Ismail-Beigi F. Regulation of glucose transport by hypoxia. Am J Kidney Dis. 1999;34:189–202.PubMed
151.
Zurück zum Zitat Chen J, Jiang CC, Jin L, Zhang XD. Regulation of PD-L1: a novel role of pro-survival signalling in cancer. Ann Oncol. 2016;27:409–16.PubMed Chen J, Jiang CC, Jin L, Zhang XD. Regulation of PD-L1: a novel role of pro-survival signalling in cancer. Ann Oncol. 2016;27:409–16.PubMed
152.
Zurück zum Zitat Ruf M, Moch H, Schraml P. PD-L1 expression is regulated by hypoxia inducible factor in clear cell renal cell carcinoma. Int J Cancer. 2016;139:396–403.PubMed Ruf M, Moch H, Schraml P. PD-L1 expression is regulated by hypoxia inducible factor in clear cell renal cell carcinoma. Int J Cancer. 2016;139:396–403.PubMed
153.
Zurück zum Zitat Mihailovic J, Killeen RP, Duignan JA. PET/CT variants and pitfalls in head and neck cancers including thyroid cancer. Semin Nucl Med. 2021;51:419–40.PubMed Mihailovic J, Killeen RP, Duignan JA. PET/CT variants and pitfalls in head and neck cancers including thyroid cancer. Semin Nucl Med. 2021;51:419–40.PubMed
154.
Zurück zum Zitat Dohán O, Carrasco N. Advances in Na(+)/I(−) symporter (NIS) research in the thyroid and beyond. Mol Cell Endocrinol. 2003;213:59–70.PubMed Dohán O, Carrasco N. Advances in Na(+)/I(−) symporter (NIS) research in the thyroid and beyond. Mol Cell Endocrinol. 2003;213:59–70.PubMed
155.
Zurück zum Zitat Garcia D, Singh V. Nuclear medicine PET/CT thyroid cancer assessment, protocols, and interpretation. In: StatPearls. Treasure Island (FL): StatPearls Publishing Copyright © 2021, StatPearls Publishing LLC.; 2021. Garcia D, Singh V. Nuclear medicine PET/CT thyroid cancer assessment, protocols, and interpretation. In: StatPearls. Treasure Island (FL): StatPearls Publishing Copyright © 2021, StatPearls Publishing LLC.; 2021.
156.
Zurück zum Zitat Duarte PS, Marin JFG, de Carvalho JWA, Sapienza MT, Buchpiguel CA. Iodine/FDG “flip-flop” phenomenon inside a large metastatic thyroid cancer lesion better characterized on SPECT/CT and PET/CT studies. Clin Nucl Med. 2018;43:436–8.PubMed Duarte PS, Marin JFG, de Carvalho JWA, Sapienza MT, Buchpiguel CA. Iodine/FDG “flip-flop” phenomenon inside a large metastatic thyroid cancer lesion better characterized on SPECT/CT and PET/CT studies. Clin Nucl Med. 2018;43:436–8.PubMed
157.
Zurück zum Zitat Hitu L, Cainap C, Apostu D, Gabora K, Bonci EA, Badan M, Mester A, Piciu A. Skeletal muscle metastasis in papillary thyroid microcarcinoma evaluated by F18-FDG PET/CT. Diagnostics (Basel). 2020;10:100.PubMed Hitu L, Cainap C, Apostu D, Gabora K, Bonci EA, Badan M, Mester A, Piciu A. Skeletal muscle metastasis in papillary thyroid microcarcinoma evaluated by F18-FDG PET/CT. Diagnostics (Basel). 2020;10:100.PubMed
158.
Zurück zum Zitat Wartski M, Sauvanet A. 18F-FDG PET/CT in pancreatic adenocarcinoma: a role at initial imaging staging? Diagn Interv Imaging. 2019;100:735–41.PubMed Wartski M, Sauvanet A. 18F-FDG PET/CT in pancreatic adenocarcinoma: a role at initial imaging staging? Diagn Interv Imaging. 2019;100:735–41.PubMed
159.
Zurück zum Zitat Kukulska A, Krajewska J, Kołosza Z, Paliczka-Cies Lik E, Puch Z, Gubała E, Król A, Kalemba M, Kropin Ska A, Jarząb B. The role of FDG-PET in localization of recurrent lesions of differentiated thyroid cancer (DTC) in patients with asymptomatic hyperthyroglobulinemia in a real clinical practice. Eur J Endocrinol. 2016;175:379–85.PubMed Kukulska A, Krajewska J, Kołosza Z, Paliczka-Cies Lik E, Puch Z, Gubała E, Król A, Kalemba M, Kropin Ska A, Jarząb B. The role of FDG-PET in localization of recurrent lesions of differentiated thyroid cancer (DTC) in patients with asymptomatic hyperthyroglobulinemia in a real clinical practice. Eur J Endocrinol. 2016;175:379–85.PubMed
160.
Zurück zum Zitat Zhang Y, Zhao H, Liu Y, Zeng M, Zhang J, Hao D. Diagnostic performance of dynamic contrast-enhanced MRI and (18)F-FDG PET/CT for evaluation of soft tissue tumors and correlation with pathology parameters. Acad Radiol. 2022;29:1842–51.PubMed Zhang Y, Zhao H, Liu Y, Zeng M, Zhang J, Hao D. Diagnostic performance of dynamic contrast-enhanced MRI and (18)F-FDG PET/CT for evaluation of soft tissue tumors and correlation with pathology parameters. Acad Radiol. 2022;29:1842–51.PubMed
161.
Zurück zum Zitat de Koster EJ, Noortman WA, Mostert JM, Booij J, Brouwer CB, de Keizer B, de Klerk JMH, Oyen WJG, van Velden FHP, de Geus-Oei LF, Vriens D. Quantitative classification and radiomics of [(18)F]FDG-PET/CT in indeterminate thyroid nodules. Eur J Nucl Med Mol Imaging. 2022;49:2174–88.PubMedPubMedCentral de Koster EJ, Noortman WA, Mostert JM, Booij J, Brouwer CB, de Keizer B, de Klerk JMH, Oyen WJG, van Velden FHP, de Geus-Oei LF, Vriens D. Quantitative classification and radiomics of [(18)F]FDG-PET/CT in indeterminate thyroid nodules. Eur J Nucl Med Mol Imaging. 2022;49:2174–88.PubMedPubMedCentral
162.
Zurück zum Zitat Zampella E, Klain M, Pace L, Cuocolo A. PET/CT in the management of differentiated thyroid cancer. Diagn Interv Imaging. 2021;102:515–23.PubMed Zampella E, Klain M, Pace L, Cuocolo A. PET/CT in the management of differentiated thyroid cancer. Diagn Interv Imaging. 2021;102:515–23.PubMed
163.
Zurück zum Zitat Kim DH, Kim SJ. Diagnostic role of F-18 FDG PET/CT for preoperative lymph node staging in thyroid cancer patients; a systematic review and meta analysis. Clin Imaging. 2020;65:100–7.PubMed Kim DH, Kim SJ. Diagnostic role of F-18 FDG PET/CT for preoperative lymph node staging in thyroid cancer patients; a systematic review and meta analysis. Clin Imaging. 2020;65:100–7.PubMed
164.
Zurück zum Zitat Luster M, Aktolun C, Amendoeira I, Barczyński M, Bible KC, Duntas LH, Elisei R, Handkiewicz-Junak D, Hoffmann M, Jarząb B, et al. European perspective on 2015 American thyroid association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: an interactive international symposium. Thyroid. 2019;29:7–26.PubMed Luster M, Aktolun C, Amendoeira I, Barczyński M, Bible KC, Duntas LH, Elisei R, Handkiewicz-Junak D, Hoffmann M, Jarząb B, et al. European perspective on 2015 American thyroid association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: an interactive international symposium. Thyroid. 2019;29:7–26.PubMed
165.
Zurück zum Zitat Mu X, Huang X, Jiang Z, Li M, Jia L, Lv Z, Fu W, Mao J. [(18)F]FAPI-42 PET/CT in differentiated thyroid cancer: diagnostic performance, uptake values, and comparison with 2-[(18)F]FDG PET/CT. Eur J Nucl Med Mol Imaging. 2023;50:1205–15.PubMed Mu X, Huang X, Jiang Z, Li M, Jia L, Lv Z, Fu W, Mao J. [(18)F]FAPI-42 PET/CT in differentiated thyroid cancer: diagnostic performance, uptake values, and comparison with 2-[(18)F]FDG PET/CT. Eur J Nucl Med Mol Imaging. 2023;50:1205–15.PubMed
166.
Zurück zum Zitat Lebbink CA, de Vries LH, Borel Rinkes IHM, Braat A, van Leeuwaarde RS, Lodewijk L, van Treijen MJC, Vriens MR, Valk GD, van Santen HM, de Keizer B. FDG PET/CT in differentiated thyroid cancer patients with low thyroglobulin levels. Eur J Endocrinol. 2022;187:101–10.PubMed Lebbink CA, de Vries LH, Borel Rinkes IHM, Braat A, van Leeuwaarde RS, Lodewijk L, van Treijen MJC, Vriens MR, Valk GD, van Santen HM, de Keizer B. FDG PET/CT in differentiated thyroid cancer patients with low thyroglobulin levels. Eur J Endocrinol. 2022;187:101–10.PubMed
167.
Zurück zum Zitat Stecco A, Trisoglio A, Soligo E, Berardo S, Sukhovei L, Carriero A. Whole-body MRI with diffusion-weighted imaging in bone metastases: a narrative review. Diagnostics (Basel). 2018;8:45.PubMed Stecco A, Trisoglio A, Soligo E, Berardo S, Sukhovei L, Carriero A. Whole-body MRI with diffusion-weighted imaging in bone metastases: a narrative review. Diagnostics (Basel). 2018;8:45.PubMed
168.
Zurück zum Zitat Klain M, Nappi C, Nicolai E, Romeo V, Piscopo L, Giordano A, Gaudieri V, Zampella E, Pace L, Carlo C, et al. Comparison of simultaneous (18)F-2-[18F] FDG PET/MR and PET/CT in the follow-up of patients with differentiated thyroid cancer. Eur J Nucl Med Mol Imaging. 2020;47:3066–73.PubMed Klain M, Nappi C, Nicolai E, Romeo V, Piscopo L, Giordano A, Gaudieri V, Zampella E, Pace L, Carlo C, et al. Comparison of simultaneous (18)F-2-[18F] FDG PET/MR and PET/CT in the follow-up of patients with differentiated thyroid cancer. Eur J Nucl Med Mol Imaging. 2020;47:3066–73.PubMed
169.
Zurück zum Zitat Alavi A, Saboury B, Nardo L, Zhang V, Wang M, Li H, Raynor WY, Werner TJ, Høilund-Carlsen PF, Revheim ME. Potential and most relevant applications of total body PET/CT imaging. Clin Nucl Med. 2022;47:43–55.PubMed Alavi A, Saboury B, Nardo L, Zhang V, Wang M, Li H, Raynor WY, Werner TJ, Høilund-Carlsen PF, Revheim ME. Potential and most relevant applications of total body PET/CT imaging. Clin Nucl Med. 2022;47:43–55.PubMed
170.
Zurück zum Zitat Shao C, Li Z, Zhang C, Zhang W, He R, Xu J, Cai Y. Optical diagnostic imaging and therapy for thyroid cancer. Mater Today Bio. 2022;17: 100441.PubMedPubMedCentral Shao C, Li Z, Zhang C, Zhang W, He R, Xu J, Cai Y. Optical diagnostic imaging and therapy for thyroid cancer. Mater Today Bio. 2022;17: 100441.PubMedPubMedCentral
172.
Zurück zum Zitat Pfeiffer T, Schuster S, Bonhoeffer S. Cooperation and competition in the evolution of ATP-producing pathways. Science. 2001;292:504–7.PubMed Pfeiffer T, Schuster S, Bonhoeffer S. Cooperation and competition in the evolution of ATP-producing pathways. Science. 2001;292:504–7.PubMed
173.
Zurück zum Zitat Macheda ML, Rogers S, Best JD. Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J Cell Physiol. 2005;202:654–62.PubMed Macheda ML, Rogers S, Best JD. Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J Cell Physiol. 2005;202:654–62.PubMed
174.
Zurück zum Zitat Sammarco G, Varricchi G, Ferraro V, Ammendola M, De Fazio M, Altomare DF, Luposella M, Maltese L, Currò G, Marone G, et al. Mast cells, angiogenesis and lymphangiogenesis in human gastric cancer. Int J Mol Sci. 2019;20:2106.PubMedPubMedCentral Sammarco G, Varricchi G, Ferraro V, Ammendola M, De Fazio M, Altomare DF, Luposella M, Maltese L, Currò G, Marone G, et al. Mast cells, angiogenesis and lymphangiogenesis in human gastric cancer. Int J Mol Sci. 2019;20:2106.PubMedPubMedCentral
176.
Zurück zum Zitat Gulubova M, Ivanova K, Ananiev J, Gerenova J, Zdraveski A, Stoyanov H, Vlaykova T. VEGF expression, microvessel density and dendritic cell decrease in thyroid cancer. Biotechnol Biotechnol Equip. 2014;28:508–17.PubMedPubMedCentral Gulubova M, Ivanova K, Ananiev J, Gerenova J, Zdraveski A, Stoyanov H, Vlaykova T. VEGF expression, microvessel density and dendritic cell decrease in thyroid cancer. Biotechnol Biotechnol Equip. 2014;28:508–17.PubMedPubMedCentral
177.
Zurück zum Zitat Varricchi G, Granata F, Loffredo S, Genovese A, Marone G. Angiogenesis and lymphangiogenesis in inflammatory skin disorders. J Am Acad Dermatol. 2015;73:144–53.PubMed Varricchi G, Granata F, Loffredo S, Genovese A, Marone G. Angiogenesis and lymphangiogenesis in inflammatory skin disorders. J Am Acad Dermatol. 2015;73:144–53.PubMed
178.
Zurück zum Zitat Loffredo S, Borriello F, Iannone R, Ferrara AL, Galdiero MR, Gigantino V, Esposito P, Varricchi G, Lambeau G, Cassatella MA, et al. Group V secreted phospholipase A(2) induces the release of proangiogenic and antiangiogenic factors by human neutrophils. Front Immunol. 2017;8:443.PubMedPubMedCentral Loffredo S, Borriello F, Iannone R, Ferrara AL, Galdiero MR, Gigantino V, Esposito P, Varricchi G, Lambeau G, Cassatella MA, et al. Group V secreted phospholipase A(2) induces the release of proangiogenic and antiangiogenic factors by human neutrophils. Front Immunol. 2017;8:443.PubMedPubMedCentral
179.
Zurück zum Zitat Valerio L, Pieruzzi L, Giani C, Agate L, Bottici V, Lorusso L, Cappagli V, Puleo L, Matrone A, Viola D, et al. Targeted therapy in thyroid cancer: state of the art. Clin Oncol (R Coll Radiol). 2017;29:316–24.PubMed Valerio L, Pieruzzi L, Giani C, Agate L, Bottici V, Lorusso L, Cappagli V, Puleo L, Matrone A, Viola D, et al. Targeted therapy in thyroid cancer: state of the art. Clin Oncol (R Coll Radiol). 2017;29:316–24.PubMed
180.
Zurück zum Zitat Capp C, Wajner SM, Siqueira DR, Brasil BA, Meurer L, Maia AL. Increased expression of vascular endothelial growth factor and its receptors, VEGFR-1 and VEGFR-2, in medullary thyroid carcinoma. Thyroid. 2010;20:863–71.PubMed Capp C, Wajner SM, Siqueira DR, Brasil BA, Meurer L, Maia AL. Increased expression of vascular endothelial growth factor and its receptors, VEGFR-1 and VEGFR-2, in medullary thyroid carcinoma. Thyroid. 2010;20:863–71.PubMed
181.
Zurück zum Zitat Samimi H, Fallah P, Naderi Sohi A, Tavakoli R, Naderi M, Soleimani M, Larijani B, Haghpanah V. Precision medicine approach to anaplastic thyroid cancer: advances in targeted drug therapy based on specific signaling pathways. Acta Med Iran. 2017;55:200–8.PubMed Samimi H, Fallah P, Naderi Sohi A, Tavakoli R, Naderi M, Soleimani M, Larijani B, Haghpanah V. Precision medicine approach to anaplastic thyroid cancer: advances in targeted drug therapy based on specific signaling pathways. Acta Med Iran. 2017;55:200–8.PubMed
182.
Zurück zum Zitat Schlumberger MJ, Elisei R, Bastholt L, Wirth LJ, Martins RG, Locati LD, Jarzab B, Pacini F, Daumerie C, Droz JP, et al. Phase II study of safety and efficacy of motesanib in patients with progressive or symptomatic, advanced or metastatic medullary thyroid cancer. J Clin Oncol. 2009;27:3794–801.PubMed Schlumberger MJ, Elisei R, Bastholt L, Wirth LJ, Martins RG, Locati LD, Jarzab B, Pacini F, Daumerie C, Droz JP, et al. Phase II study of safety and efficacy of motesanib in patients with progressive or symptomatic, advanced or metastatic medullary thyroid cancer. J Clin Oncol. 2009;27:3794–801.PubMed
183.
Zurück zum Zitat Wells SA Jr, Gosnell JE, Gagel RF, Moley J, Pfister D, Sosa JA, Skinner M, Krebs A, Vasselli J, Schlumberger M. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Oncol. 2010;28:767–72.PubMedPubMedCentral Wells SA Jr, Gosnell JE, Gagel RF, Moley J, Pfister D, Sosa JA, Skinner M, Krebs A, Vasselli J, Schlumberger M. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Oncol. 2010;28:767–72.PubMedPubMedCentral
184.
Zurück zum Zitat Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger M, et al. 2015 American thyroid association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: the American thyroid association guidelines task force on thyroid nodules and differentiated thyroid cancer. Thyroid. 2016;26:1–133.PubMedPubMedCentral Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger M, et al. 2015 American thyroid association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: the American thyroid association guidelines task force on thyroid nodules and differentiated thyroid cancer. Thyroid. 2016;26:1–133.PubMedPubMedCentral
185.
Zurück zum Zitat Motzer RJ, Jonasch E, Boyle S, Carlo MI, Manley B, Agarwal N, Alva A, Beckermann K, Choueiri TK, Costello BA, et al. NCCN guidelines insights: kidney cancer, version 1.2021. J Natl Compr Canc Netw. 2020;18:1160–70.PubMedPubMedCentral Motzer RJ, Jonasch E, Boyle S, Carlo MI, Manley B, Agarwal N, Alva A, Beckermann K, Choueiri TK, Costello BA, et al. NCCN guidelines insights: kidney cancer, version 1.2021. J Natl Compr Canc Netw. 2020;18:1160–70.PubMedPubMedCentral
186.
Zurück zum Zitat Fallahi P, Ferrari SM, Santini F, Corrado A, Materazzi G, Ulisse S, Miccoli P, Antonelli A. Sorafenib and thyroid cancer. BioDrugs. 2013;27:615–28.PubMed Fallahi P, Ferrari SM, Santini F, Corrado A, Materazzi G, Ulisse S, Miccoli P, Antonelli A. Sorafenib and thyroid cancer. BioDrugs. 2013;27:615–28.PubMed
187.
Zurück zum Zitat Worden F, Fassnacht M, Shi Y, Hadjieva T, Bonichon F, Gao M, Fugazzola L, Ando Y, Hasegawa Y, Park DJ, et al. Safety and tolerability of sorafenib in patients with radioiodine-refractory thyroid cancer. Endocr Relat Cancer. 2015;22:877–87.PubMedPubMedCentral Worden F, Fassnacht M, Shi Y, Hadjieva T, Bonichon F, Gao M, Fugazzola L, Ando Y, Hasegawa Y, Park DJ, et al. Safety and tolerability of sorafenib in patients with radioiodine-refractory thyroid cancer. Endocr Relat Cancer. 2015;22:877–87.PubMedPubMedCentral
188.
Zurück zum Zitat Ferrari SM, Centanni M, Virili C, Miccoli M, Ferrari P, Ruffilli I, Ragusa F, Antonelli A, Fallahi P. Sunitinib in the treatment of thyroid cancer. Curr Med Chem. 2019;26:963–72.PubMed Ferrari SM, Centanni M, Virili C, Miccoli M, Ferrari P, Ruffilli I, Ragusa F, Antonelli A, Fallahi P. Sunitinib in the treatment of thyroid cancer. Curr Med Chem. 2019;26:963–72.PubMed
189.
Zurück zum Zitat Kim DW, Jo YS, Jung HS, Chung HK, Song JH, Park KC, Park SH, Hwang JH, Rha SY, Kweon GR, et al. An orally administered multitarget tyrosine kinase inhibitor, SU11248, is a novel potent inhibitor of thyroid oncogenic RET/papillary thyroid cancer kinases. J Clin Endocrinol Metab. 2006;91:4070–6.PubMed Kim DW, Jo YS, Jung HS, Chung HK, Song JH, Park KC, Park SH, Hwang JH, Rha SY, Kweon GR, et al. An orally administered multitarget tyrosine kinase inhibitor, SU11248, is a novel potent inhibitor of thyroid oncogenic RET/papillary thyroid cancer kinases. J Clin Endocrinol Metab. 2006;91:4070–6.PubMed
190.
Zurück zum Zitat Carlomagno F, Vitagliano D, Guida T, Ciardiello F, Tortora G, Vecchio G, Ryan AJ, Fontanini G, Fusco A, Santoro M. ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Res. 2002;62:7284–90.PubMed Carlomagno F, Vitagliano D, Guida T, Ciardiello F, Tortora G, Vecchio G, Ryan AJ, Fontanini G, Fusco A, Santoro M. ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Res. 2002;62:7284–90.PubMed
191.
Zurück zum Zitat Nair A, Lemery SJ, Yang J, Marathe A, Zhao L, Zhao H, Jiang X, He K, Ladouceur G, Mitra AK, et al. FDA approval summary: lenvatinib for progressive, radio-iodine-refractory differentiated thyroid cancer. Clin Cancer Res. 2015;21:5205–8.PubMed Nair A, Lemery SJ, Yang J, Marathe A, Zhao L, Zhao H, Jiang X, He K, Ladouceur G, Mitra AK, et al. FDA approval summary: lenvatinib for progressive, radio-iodine-refractory differentiated thyroid cancer. Clin Cancer Res. 2015;21:5205–8.PubMed
192.
Zurück zum Zitat Fallahi P, Ferrari SM, Di Bari F, Materazzi G, Benvenga S, Miccoli P, Antonelli A. Cabozantinib in thyroid cancer. Recent Pat Anticancer Drug Discov. 2015;10:259–69.PubMed Fallahi P, Ferrari SM, Di Bari F, Materazzi G, Benvenga S, Miccoli P, Antonelli A. Cabozantinib in thyroid cancer. Recent Pat Anticancer Drug Discov. 2015;10:259–69.PubMed
193.
Zurück zum Zitat Chau NG, Haddad RI. Vandetanib for the treatment of medullary thyroid cancer. Clin Cancer Res. 2023;19(3):524–9. Chau NG, Haddad RI. Vandetanib for the treatment of medullary thyroid cancer. Clin Cancer Res. 2023;19(3):524–9.
194.
Zurück zum Zitat Brandenburg T, Tiedje V, Muchalla P, Theurer S, Weber F, Schmid KW, Dralle H, Führer D. Continued discontinuation of TKI treatment in medullary thyroid carcinoma—lessons from individual cases with long-term follow-up. Front Endocrinol (Lausanne). 2021;12: 718418.PubMed Brandenburg T, Tiedje V, Muchalla P, Theurer S, Weber F, Schmid KW, Dralle H, Führer D. Continued discontinuation of TKI treatment in medullary thyroid carcinoma—lessons from individual cases with long-term follow-up. Front Endocrinol (Lausanne). 2021;12: 718418.PubMed
195.
Zurück zum Zitat Nylén C, Mechera R, Maréchal-Ross I, Tsang V, Chou A, Gill AJ, Clifton-Bligh RJ, Robinson BG, Sywak MS, Sidhu SB, Glover AR. Molecular markers guiding thyroid cancer management. Cancers (Basel). 2020;12:2164.PubMed Nylén C, Mechera R, Maréchal-Ross I, Tsang V, Chou A, Gill AJ, Clifton-Bligh RJ, Robinson BG, Sywak MS, Sidhu SB, Glover AR. Molecular markers guiding thyroid cancer management. Cancers (Basel). 2020;12:2164.PubMed
196.
Zurück zum Zitat Ferrari SM, Ruffilli I, Centanni M, Virili C, Materazzi G, Alexopoulou M, Miccoli M, Antonelli A, Fallahi P. Lenvatinib in the therapy of aggressive thyroid cancer: state of the art and new perspectives with patents recently applied. Recent Pat Anticancer Drug Discov. 2018;13:201–8.PubMed Ferrari SM, Ruffilli I, Centanni M, Virili C, Materazzi G, Alexopoulou M, Miccoli M, Antonelli A, Fallahi P. Lenvatinib in the therapy of aggressive thyroid cancer: state of the art and new perspectives with patents recently applied. Recent Pat Anticancer Drug Discov. 2018;13:201–8.PubMed
197.
Zurück zum Zitat Enokida T, Tahara M. Management of VEGFR-targeted TKI for thyroid cancer. Cancers (Basel). 2021;13:5536.PubMed Enokida T, Tahara M. Management of VEGFR-targeted TKI for thyroid cancer. Cancers (Basel). 2021;13:5536.PubMed
198.
Zurück zum Zitat Wang C, Li Y, Yan S, Wang H, Shao X, Xiao M, Yang B, Qin G, Kong R, Chen R, Zhang N. Interactome analysis reveals that lncRNA HULC promotes aerobic glycolysis through LDHA and PKM2. Nat Commun. 2020;11:3162.PubMedPubMedCentral Wang C, Li Y, Yan S, Wang H, Shao X, Xiao M, Yang B, Qin G, Kong R, Chen R, Zhang N. Interactome analysis reveals that lncRNA HULC promotes aerobic glycolysis through LDHA and PKM2. Nat Commun. 2020;11:3162.PubMedPubMedCentral
199.
Zurück zum Zitat Boudreau A, Purkey HE, Hitz A, Robarge K, Peterson D, Labadie S, Kwong M, Hong R, Gao M, Del Nagro C, et al. Metabolic plasticity underpins innate and acquired resistance to LDHA inhibition. Nat Chem Biol. 2016;12:779–86.PubMed Boudreau A, Purkey HE, Hitz A, Robarge K, Peterson D, Labadie S, Kwong M, Hong R, Gao M, Del Nagro C, et al. Metabolic plasticity underpins innate and acquired resistance to LDHA inhibition. Nat Chem Biol. 2016;12:779–86.PubMed
200.
Zurück zum Zitat Pathria G, Scott DA, Feng Y, Sang Lee J, Fujita Y, Zhang G, Sahu AD, Ruppin E, Herlyn M, Osterman AL, Ronai ZA. Targeting the Warburg effect via LDHA inhibition engages ATF4 signaling for cancer cell survival. Embo J. 2018;37: e99735.PubMedPubMedCentral Pathria G, Scott DA, Feng Y, Sang Lee J, Fujita Y, Zhang G, Sahu AD, Ruppin E, Herlyn M, Osterman AL, Ronai ZA. Targeting the Warburg effect via LDHA inhibition engages ATF4 signaling for cancer cell survival. Embo J. 2018;37: e99735.PubMedPubMedCentral
201.
Zurück zum Zitat Kachel P, Trojanowicz B, Sekulla C, Prenzel H, Dralle H, Hoang-Vu C. Phosphorylation of pyruvate kinase M2 and lactate dehydrogenase A by fibroblast growth factor receptor 1 in benign and malignant thyroid tissue. BMC Cancer. 2015;15:140.PubMedPubMedCentral Kachel P, Trojanowicz B, Sekulla C, Prenzel H, Dralle H, Hoang-Vu C. Phosphorylation of pyruvate kinase M2 and lactate dehydrogenase A by fibroblast growth factor receptor 1 in benign and malignant thyroid tissue. BMC Cancer. 2015;15:140.PubMedPubMedCentral
202.
Zurück zum Zitat Hou X, Shi X, Zhang W, Li D, Hu L, Yang J, Zhao J, Wei S, Wei X, Ruan X, et al. LDHA induces EMT gene transcription and regulates autophagy to promote the metastasis and tumorigenesis of papillary thyroid carcinoma. Cell Death Dis. 2021;12:347.PubMedPubMedCentral Hou X, Shi X, Zhang W, Li D, Hu L, Yang J, Zhao J, Wei S, Wei X, Ruan X, et al. LDHA induces EMT gene transcription and regulates autophagy to promote the metastasis and tumorigenesis of papillary thyroid carcinoma. Cell Death Dis. 2021;12:347.PubMedPubMedCentral
204.
Zurück zum Zitat Zhang J, Gill A, Atmore B, Johns A, Delbridge L, Lai R, McMullen T. Upregulation of the signal transducers and activators of transcription 3 (STAT3) pathway in lymphatic metastases of papillary thyroid cancer. Int J Clin Exp Pathol. 2011;4:356–62.PubMedPubMedCentral Zhang J, Gill A, Atmore B, Johns A, Delbridge L, Lai R, McMullen T. Upregulation of the signal transducers and activators of transcription 3 (STAT3) pathway in lymphatic metastases of papillary thyroid cancer. Int J Clin Exp Pathol. 2011;4:356–62.PubMedPubMedCentral
205.
Zurück zum Zitat Jafary F, Ganjalikhany MR, Moradi A, Hemati M, Jafari S. Novel peptide inhibitors for lactate dehydrogenase A (LDHA): a survey to inhibit LDHA activity via disruption of protein-protein interaction. Sci Rep. 2019;9:4686.PubMedPubMedCentral Jafary F, Ganjalikhany MR, Moradi A, Hemati M, Jafari S. Novel peptide inhibitors for lactate dehydrogenase A (LDHA): a survey to inhibit LDHA activity via disruption of protein-protein interaction. Sci Rep. 2019;9:4686.PubMedPubMedCentral
206.
Zurück zum Zitat Wang Z, Wang N, Liu P, Xie X. AMPK and cancer. Exp Suppl. 2016;107:203–26.PubMed Wang Z, Wang N, Liu P, Xie X. AMPK and cancer. Exp Suppl. 2016;107:203–26.PubMed
207.
Zurück zum Zitat Souza EC, Ferreira AC, Carvalho DP. The mTOR protein as a target in thyroid cancer. Expert Opin Ther Targets. 2011;15:1099–112.PubMed Souza EC, Ferreira AC, Carvalho DP. The mTOR protein as a target in thyroid cancer. Expert Opin Ther Targets. 2011;15:1099–112.PubMed
208.
Zurück zum Zitat Mita MM, Mita A, Rowinsky EK. Mammalian target of rapamycin: a new molecular target for breast cancer. Clin Breast Cancer. 2003;4:126–37.PubMed Mita MM, Mita A, Rowinsky EK. Mammalian target of rapamycin: a new molecular target for breast cancer. Clin Breast Cancer. 2003;4:126–37.PubMed
209.
Zurück zum Zitat Elit L. CCI-779 Wyeth. Curr Opin Investig Drugs. 2002;3:1249–53.PubMed Elit L. CCI-779 Wyeth. Curr Opin Investig Drugs. 2002;3:1249–53.PubMed
210.
Zurück zum Zitat Liu D, Hou P, Liu Z, Wu G, Xing M. Genetic alterations in the phosphoinositide 3-kinase/Akt signaling pathway confer sensitivity of thyroid cancer cells to therapeutic targeting of Akt and mammalian target of rapamycin. Cancer Res. 2009;69:7311–9.PubMedPubMedCentral Liu D, Hou P, Liu Z, Wu G, Xing M. Genetic alterations in the phosphoinositide 3-kinase/Akt signaling pathway confer sensitivity of thyroid cancer cells to therapeutic targeting of Akt and mammalian target of rapamycin. Cancer Res. 2009;69:7311–9.PubMedPubMedCentral
211.
Zurück zum Zitat Hanna GJ, Busaidy NL, Chau NG, Wirth LJ, Barletta JA, Calles A, Haddad RI, Kraft S, Cabanillas ME, Rabinowits GJ. Genomic correlates of response to everolimus in aggressive radioiodine-refractory thyroid cancer: a phase II study. Clin Cancer Res. 2018;24:1546–53.PubMed Hanna GJ, Busaidy NL, Chau NG, Wirth LJ, Barletta JA, Calles A, Haddad RI, Kraft S, Cabanillas ME, Rabinowits GJ. Genomic correlates of response to everolimus in aggressive radioiodine-refractory thyroid cancer: a phase II study. Clin Cancer Res. 2018;24:1546–53.PubMed
212.
Zurück zum Zitat Schneider TC, de Wit D, Links TP, van Erp NP, van der Hoeven JJM, Gelderblom H, Roozen ICFM, Bos M, Corver WE, van Wezel T, et al. Everolimus in patients with advanced follicular-derived thyroid cancer: results of a phase II clinical trial. J Clin Endocrinol Metab. 2016;102:698–707. Schneider TC, de Wit D, Links TP, van Erp NP, van der Hoeven JJM, Gelderblom H, Roozen ICFM, Bos M, Corver WE, van Wezel T, et al. Everolimus in patients with advanced follicular-derived thyroid cancer: results of a phase II clinical trial. J Clin Endocrinol Metab. 2016;102:698–707.
213.
Zurück zum Zitat Furuya F, Lu C, Willingham MC, Cheng SY. Inhibition of phosphatidylinositol 3-kinase delays tumor progression and blocks metastatic spread in a mouse model of thyroid cancer. Carcinogenesis. 2007;28:2451–8.PubMed Furuya F, Lu C, Willingham MC, Cheng SY. Inhibition of phosphatidylinositol 3-kinase delays tumor progression and blocks metastatic spread in a mouse model of thyroid cancer. Carcinogenesis. 2007;28:2451–8.PubMed
214.
Zurück zum Zitat Liu D, Xing J, Trink B, Xing M. BRAF mutation-selective inhibition of thyroid cancer cells by the novel MEK inhibitor RDEA119 and genetic-potentiated synergism with the mTOR inhibitor temsirolimus. Int J Cancer. 2010;127:2965–73.PubMedPubMedCentral Liu D, Xing J, Trink B, Xing M. BRAF mutation-selective inhibition of thyroid cancer cells by the novel MEK inhibitor RDEA119 and genetic-potentiated synergism with the mTOR inhibitor temsirolimus. Int J Cancer. 2010;127:2965–73.PubMedPubMedCentral
215.
Zurück zum Zitat Zhu X, Park S, Lee WK, Cheng SY. Potentiated anti-tumor effects of BETi by MEKi in anaplastic thyroid cancer. Endocr Relat Cancer. 2019;26:739–50.PubMedPubMedCentral Zhu X, Park S, Lee WK, Cheng SY. Potentiated anti-tumor effects of BETi by MEKi in anaplastic thyroid cancer. Endocr Relat Cancer. 2019;26:739–50.PubMedPubMedCentral
216.
Zurück zum Zitat Zaballos MA, Acuña-Ruiz A, Morante M, Crespo P, Santisteban P. Regulators of the RAS-ERK pathway as therapeutic targets in thyroid cancer. Endocr Relat Cancer. 2019;26:R319-r344.PubMed Zaballos MA, Acuña-Ruiz A, Morante M, Crespo P, Santisteban P. Regulators of the RAS-ERK pathway as therapeutic targets in thyroid cancer. Endocr Relat Cancer. 2019;26:R319-r344.PubMed
217.
Zurück zum Zitat Kurata K, Onoda N, Noda S, Kashiwagi S, Asano Y, Hirakawa K, Ohira M. Growth arrest by activated BRAF and MEK inhibition in human anaplastic thyroid cancer cells. Int J Oncol. 2016;49:2303–8.PubMed Kurata K, Onoda N, Noda S, Kashiwagi S, Asano Y, Hirakawa K, Ohira M. Growth arrest by activated BRAF and MEK inhibition in human anaplastic thyroid cancer cells. Int J Oncol. 2016;49:2303–8.PubMed
218.
Zurück zum Zitat Flaherty KT, Robert C, Hersey P, Nathan P, Garbe C, Milhem M, Demidov LV, Hassel JC, Rutkowski P, Mohr P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med. 2012;367:107–14.PubMed Flaherty KT, Robert C, Hersey P, Nathan P, Garbe C, Milhem M, Demidov LV, Hassel JC, Rutkowski P, Mohr P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med. 2012;367:107–14.PubMed
219.
Zurück zum Zitat Subbiah V, Baik C, Kirkwood JM. Clinical development of BRAF plus MEK inhibitor combinations. Trends Cancer. 2020;6:797–810.PubMed Subbiah V, Baik C, Kirkwood JM. Clinical development of BRAF plus MEK inhibitor combinations. Trends Cancer. 2020;6:797–810.PubMed
220.
Zurück zum Zitat Sabra MM, Dominguez JM, Grewal RK, Larson SM, Ghossein RA, Tuttle RM, Fagin JA. Clinical outcomes and molecular profile of differentiated thyroid cancers with radioiodine-avid distant metastases. J Clin Endocrinol Metab. 2013;98:E829-836.PubMedPubMedCentral Sabra MM, Dominguez JM, Grewal RK, Larson SM, Ghossein RA, Tuttle RM, Fagin JA. Clinical outcomes and molecular profile of differentiated thyroid cancers with radioiodine-avid distant metastases. J Clin Endocrinol Metab. 2013;98:E829-836.PubMedPubMedCentral
221.
Zurück zum Zitat Montero-Conde C, Ruiz-Llorente S, Dominguez JM, Knauf JA, Viale A, Sherman EJ, Ryder M, Ghossein RA, Rosen N, Fagin JA. Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discov. 2013;3:520–33.PubMedPubMedCentral Montero-Conde C, Ruiz-Llorente S, Dominguez JM, Knauf JA, Viale A, Sherman EJ, Ryder M, Ghossein RA, Rosen N, Fagin JA. Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discov. 2013;3:520–33.PubMedPubMedCentral
222.
Zurück zum Zitat Laha D, Nilubol N, Boufraqech M. New therapies for advanced thyroid cancer. Front Endocrinol (Lausanne). 2020;11:82.PubMed Laha D, Nilubol N, Boufraqech M. New therapies for advanced thyroid cancer. Front Endocrinol (Lausanne). 2020;11:82.PubMed
223.
Zurück zum Zitat Brown SR, Hall A, Buckley HL, Flanagan L, Gonzalez de Castro D, Farnell K, Moss L, Gregory R, Newbold K, Du YJ. Investigating the potential clinical benefit of Selumetinib in resensitising advanced iodine refractory differentiated thyroid cancer to radioiodine therapy (SEL-I-METRY): protocol for a multicentre UK single arm phase II trial. BMC Cancer. 2019;19:1–10. Brown SR, Hall A, Buckley HL, Flanagan L, Gonzalez de Castro D, Farnell K, Moss L, Gregory R, Newbold K, Du YJ. Investigating the potential clinical benefit of Selumetinib in resensitising advanced iodine refractory differentiated thyroid cancer to radioiodine therapy (SEL-I-METRY): protocol for a multicentre UK single arm phase II trial. BMC Cancer. 2019;19:1–10.
224.
Zurück zum Zitat Aashiq M, Silverman DA, Na’ara S, Takahashi H, Amit M. Radioiodine-refractory thyroid cancer: molecular basis of redifferentiation therapies, management, and novel therapies. Cancers (Basel). 2019;11:1382.PubMed Aashiq M, Silverman DA, Na’ara S, Takahashi H, Amit M. Radioiodine-refractory thyroid cancer: molecular basis of redifferentiation therapies, management, and novel therapies. Cancers (Basel). 2019;11:1382.PubMed
225.
Zurück zum Zitat Cheng L, Jin Y, Liu M, Ruan M, Chen L. HER inhibitor promotes BRAF/MEK inhibitor-induced redifferentiation in papillary thyroid cancer harboring BRAFV600E. Oncotarget. 2017;8:19843–54.PubMedPubMedCentral Cheng L, Jin Y, Liu M, Ruan M, Chen L. HER inhibitor promotes BRAF/MEK inhibitor-induced redifferentiation in papillary thyroid cancer harboring BRAFV600E. Oncotarget. 2017;8:19843–54.PubMedPubMedCentral
226.
Zurück zum Zitat Turcotte LP, Richter EA, Kiens B. Increased plasma FFA uptake and oxidation during prolonged exercise in trained vs. untrained humans. Am J Physiol. 1992;262:E791-799.PubMed Turcotte LP, Richter EA, Kiens B. Increased plasma FFA uptake and oxidation during prolonged exercise in trained vs. untrained humans. Am J Physiol. 1992;262:E791-799.PubMed
227.
Zurück zum Zitat Gohil K, Brooks GA. Exercise tames the wild side of the Myc network: a hypothesis. Am J Physiol Endocrinol Metab. 2012;303:E18-30.PubMed Gohil K, Brooks GA. Exercise tames the wild side of the Myc network: a hypothesis. Am J Physiol Endocrinol Metab. 2012;303:E18-30.PubMed
228.
Zurück zum Zitat Friedenreich CM, Orenstein MR. Physical activity and cancer prevention: etiologic evidence and biological mechanisms. J Nutr. 2002;132:3456s–64s.PubMed Friedenreich CM, Orenstein MR. Physical activity and cancer prevention: etiologic evidence and biological mechanisms. J Nutr. 2002;132:3456s–64s.PubMed
229.
Zurück zum Zitat Bellastella G, Scappaticcio L, Caiazzo F, Tomasuolo M, Carotenuto R, Caputo M, Arena S, Caruso P, Maiorino MI, Esposito K. Mediterranean diet and thyroid: an interesting alliance. Nutrients. 2022;14:4130.PubMedPubMedCentral Bellastella G, Scappaticcio L, Caiazzo F, Tomasuolo M, Carotenuto R, Caputo M, Arena S, Caruso P, Maiorino MI, Esposito K. Mediterranean diet and thyroid: an interesting alliance. Nutrients. 2022;14:4130.PubMedPubMedCentral
230.
Zurück zum Zitat Aggarwal A, Yuan Z, Barletta JA, Lorch JH, Nehs MA. Ketogenic diet combined with antioxidant N-acetylcysteine inhibits tumor growth in a mouse model of anaplastic thyroid cancer. Surgery. 2020;167:87–93.PubMed Aggarwal A, Yuan Z, Barletta JA, Lorch JH, Nehs MA. Ketogenic diet combined with antioxidant N-acetylcysteine inhibits tumor growth in a mouse model of anaplastic thyroid cancer. Surgery. 2020;167:87–93.PubMed
231.
Zurück zum Zitat Tella SH, Kommalapati A, Esquivel MA, Correa R. Potential role of metabolic intervention in the management of advanced differentiated thyroid cancer. Front Oncol. 2017;7:160.PubMedPubMedCentral Tella SH, Kommalapati A, Esquivel MA, Correa R. Potential role of metabolic intervention in the management of advanced differentiated thyroid cancer. Front Oncol. 2017;7:160.PubMedPubMedCentral
232.
Zurück zum Zitat Sowah SA, Milanese A, Schübel R, Wirbel J, Kartal E, Johnson TS, Hirche F, Grafetstätter M, Nonnenmacher T, Kirsten R, et al. Calorie restriction improves metabolic state independently of gut microbiome composition: a randomized dietary intervention trial. Genome Med. 2022;14:30.PubMedPubMedCentral Sowah SA, Milanese A, Schübel R, Wirbel J, Kartal E, Johnson TS, Hirche F, Grafetstätter M, Nonnenmacher T, Kirsten R, et al. Calorie restriction improves metabolic state independently of gut microbiome composition: a randomized dietary intervention trial. Genome Med. 2022;14:30.PubMedPubMedCentral
233.
Zurück zum Zitat Madeo F, Carmona-Gutierrez D, Hofer SJ, Kroemer G. Caloric restriction mimetics against age-associated disease: targets, mechanisms, and therapeutic potential. Cell Metab. 2019;29:592–610.PubMed Madeo F, Carmona-Gutierrez D, Hofer SJ, Kroemer G. Caloric restriction mimetics against age-associated disease: targets, mechanisms, and therapeutic potential. Cell Metab. 2019;29:592–610.PubMed
234.
Zurück zum Zitat Ferrere G, Tidjani Alou M, Liu P, Goubet AG, Fidelle M, Kepp O, Durand S, Iebba V, Fluckiger A, Daillère R, et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight. 2021;6: e145207.PubMedPubMedCentral Ferrere G, Tidjani Alou M, Liu P, Goubet AG, Fidelle M, Kepp O, Durand S, Iebba V, Fluckiger A, Daillère R, et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight. 2021;6: e145207.PubMedPubMedCentral
235.
Zurück zum Zitat Vernieri C, Fucà G, Ligorio F, Huber V, Vingiani A, Iannelli F, Raimondi A, Rinchai D, Frigè G, Belfiore A, et al. Fasting-mimicking diet is safe and reshapes metabolism and antitumor immunity in patients with cancer. Cancer Discov. 2022;12:90–107.PubMed Vernieri C, Fucà G, Ligorio F, Huber V, Vingiani A, Iannelli F, Raimondi A, Rinchai D, Frigè G, Belfiore A, et al. Fasting-mimicking diet is safe and reshapes metabolism and antitumor immunity in patients with cancer. Cancer Discov. 2022;12:90–107.PubMed
Metadaten
Titel
The potential role of reprogrammed glucose metabolism: an emerging actionable codependent target in thyroid cancer
verfasst von
Sai-li Duan
Min Wu
Zhe-Jia Zhang
Shi Chang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2023
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-023-04617-2

Weitere Artikel der Ausgabe 1/2023

Journal of Translational Medicine 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.