Critical role of mTOR in regulating aerobic glycolysis in carcinogenesis (Review)

  • Authors:
    • Hui Fan
    • Yuanyuan Wu
    • Suyun Yu
    • Xiaoman Li
    • Aiyun Wang
    • Shijun Wang
    • Wenxing Chen
    • Yin Lu
  • View Affiliations

  • Published online on: November 25, 2020     https://doi.org/10.3892/ijo.2020.5152
  • Pages: 9-19
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mammalian target of rapamycin (mTOR) serves an important role in regulating various biological processes, including cell proliferation, metabolism, apoptosis and autophagy. Among these processes, energy metabolism is the dominant process. The metabolism of not only amino acids, fatty acids and lipids, but also that of nucleotides and glucose has been indicated to be regulated by mTOR. Aerobic glycolysis, which is a specific form of glucose metabolism, is prevalent in carcinomas, and it has been considered to be a potential target for cancer therapy. In reviewing the complexity of the mTOR pathway, it is important to elucidate the central role and detailed pathway via which mTOR regulates glycolysis. In the present study, the complex mechanisms via which mTOR regulates aerobic glycolysis were comprehensively reviewed to highlight the potential of drug development via targeting the molecules associated with mTOR and glycolysis and to further provide strategies for the clinical treatment of cancer.

1. Introduction

To adapt to the severe nutrient deficient tumor microenvironment, cancer cells have been indicated to adjust their metabolic processes, including the abnormal function of signaling path-ways and cellular metabolism (1). Cancer cells can express certain key glycolysis enzymes that act on the mammalian target of rapamycin (mTOR) signaling pathway to maintain clonality and promote migration (2,3). In recent years, increasing evidence has suggested that mTOR is a core network in cancer cells, regulating several key enzymes to maintain the balance between tumor growth and nutrition outside the cancer cells (4) (Fig. 1). The activation of mTOR has been indicated to promote mRNA transcription, protein synthesis, glucose metabolism and lipid synthesis that is necessary for cell growth (5,6). mTOR is an essential serine/threonine protein kinase that belongs to the PI3K family, and it includes two different catalytic subunit protein complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) (7-9). Both complexes have been revealed to serve important roles in regulating the metabolism of cancer cells (9).

2. mTOR is central to metabolism in cancer

Carcinomas are often accompanied by metabolic alterations that are recognized as hallmarks of cancer during tumor development, and mTOR has been indicated to serve a vital role in cancer metabolism (4). There are several types of metabolism in cancer cells, including amino acid, glucose, nucleotide and lipid metabolism (10) (Fig. 2).

Figure 2

mTOR signaling pathway regulates cancer cell metabolism. mTOR regulates the metabolism of fatty acids and lipids, mediating the uptake of fatty acids via CD36. Citrate is converted to palmitate via multiple steps that are catalyzed by ACLY and FASN, which are both regulated by mTOR signaling. Palmitate serves as a pre-substrate of phosphatidylcholine by catalyzing the formation of phosphocholine CCTα or glucosylceramide through GCS. Both CCTα and GCS are regulated by mTOR signaling. mTOR regulates glycolysis, a complex process that involves glucose transporters and various catalytic enzymes, including HK2, PFK, PKM2 and LDH. Gln, Gly, Asp and CO2 react with PRPP via GARFT catalysis to form the pre-substrate of purine nucleotides. Gln, Asp, HCO3- and ATP are converted to DHO by CAD, and after several steps pyrimidine nucleotides are produced. On the one hand, mTOR signaling regulates various amino acid transporters to increase the uptake of amino acids; on the other hand, mTOR aids in the acquisition of amino acids via macropinocytosis of extracellular proteins to synthesize the required proteins. mTOR, mammalian target of rapamycin; mTORC, mTOR complex; ACLY, ATP citrate lyase; FASN, fatty acid synthase; CCTα, cytidylyltransferase-α; GCS, glucosylceramide synthase; HK2, hexokinase 2; PFK, phosphofructokinase; PKM2, pyruvate kinase muscle isoform 2; LDH, lactate dehydrogenase; Gln, glutamine; Gly, glycine; Asp, aspartate; PRPP, ribose-5-phosphoribosyl-1-pyrophosphate; GARFT, glycinamide ribonucleotide formyltransferase; DHO, dihydroorotase; CAD, aspartate transcarbamylase and dihydroorotase; G6P, glucose-6-phosphate; F1,6BP, fructose-1,6-bisphosphate; PEP, phosphoenolpyruvate; GLUT1, glucose transporter 1; TSC, tuberous sclerosis complex; OMP, orotidine monophosphate; Rheb, Ras homolog enriched in brain.

Amino acid metabolism

Amino acids are vital nutrient substrates for protein synthesis, and this process has been indicated to be closely regulated by mTOR in cancer cells. Both essential and nonessential amino acid metabolism has been associated with the mTOR signaling pathway (4). L-type amino acid transporter (LAT) 1, which transports the essential amino acid leucine, has been associated with the activation of mTOR pathway in colorectal cancer (11). Glutamine, a nonessential amino acid, contributes to the synthesis of other amino acids, lipids and nucleotides (4), and glutaminolysis has been indicated to be promoted by the mTORC1 and mTORC2 pathways (12,13).

Nucleotide metabolism

Cancer cells necessitate nucleic acids to support their own proliferation (14). mTOR promotes purine synthesis by regulating enzymes that mediate the synthetic process. For example, phosphoribosyl pyrophosphate synthase 2 is the rate-limiting enzyme in purine synthesis, and its expression has been revealed to be upregulated by the mTORC2/AKT signaling pathway (4). Pyrimidine synthesis is also regulated by mTOR, as the mTORC1/S6 kinase pathway has been indicated to regulate implicated enzymes, including carbamoyl phosphate synthetase II, phosphorylation and oligomerization of aspartate transcarbamylase and dihydroorotase (4).

Lipid metabolism

Cancer cells can synthesize fatty acids intracellularly or incorporate extracellular fatty acids, and mTOR has been indicated to regulate fatty acid transporters, such as CD36 and synthesis enzymes, including ATP citrate lyase and fatty acid synthase (4). The transmembrane glycoprotein CD147 has been reported to reprogram fatty acid metabolism via the AKT/mTOR/sterol regulatory element binding protein-1c and p38/peroxisome proliferator-activated receptor α pathways in hepatocellular carcinoma cells (15).

Glucose metabolism

Glucose, which is the main cellular energy source, has been demonstrated to promote growth, proliferation and metastasis of cancer cells; therefore, the sense, uptake and utilization of glucose are essential for maintaining life, and the mTOR signaling pathway has been indicated to participate in these processes (16). Otto Warburg has revealed that cancer cells often rely on glycolysis even in the presence of oxygen, and this phenomenon is also called aerobic glycolysis or the Warburg effect (17).

Given the significance and complexity of aerobic glycolysis and mTOR in cancer cells, it is necessary to review the relationship between mTOR and aerobic glycolysis to suggest a potential novel strategy for clinical cancer therapy that depends on both these factors.

3. Glycolysis is a complex process

Glycolysis is a process in which glucose is metabolized into several products, including pyruvate, lactate and hydrogen ions, via multistep enzymatic reactions in the cytoplasm (18). The detailed glycolytic process is depicted in Figs. 2 and 3. Glycolytic enzymes have been indicated to be closely associated with the mTOR signaling pathway (4,19). In a first step, cancer cells overexpress glucose transporters, such as glucose transporter (GLUT)1 and sodium-glucose linked trans-porter 1, transporting glucose into the cytoplasm to maintain high levels of glucose consumption during glycolysis (20,21). Subsequently, hexokinase (HK) phosphorylates intracellular glucose to form glucose 6-phosphate (22,23). In a second step, glucose 6-phosphate isomerase catalyzes the production of fructose-6-phosphate (24). In a third step, fructose-6-phos-phate is phosphorylated by phosphofructokinase (PFK)-1 and PFK-2 into the unstable fructose-1,6-biphosphate and the relatively stable fructose-2,6-biphosphate (19), respectively. Glucose 6-phosphate is also converted into glyceraldehyde-3-phosphate (25), which is subsequently converted into glycerate-1,3-biphosphate, and glycerate-1,3-biphosphate is converted into 3-phosphoglycerate by phosphoglycerate kinase (19). Subsequently, phosphoglyceromutase drives the isomerization of 3-phosphoglycerate to phosphoglycerate 2-phosphate, followed by the formation of phosphoenolpyruvate (PEP). In the last step of glycolysis, PEP is converted into pyruvate by pyruvate kinase (PK) (26).

4. mTOR signaling pathway regulates glycolysis

As aforementioned, mTOR has been associated with several fundamental cellular processes (4), such as protein synthesis (27), autophagy (28) and cancer (29). Moreover, glycolysis is considered a principal driver of carcinoma metastasis (30) due to the energy support provided by glycolysis (30). Ka et al (28) revealed that pharmacological inhibition of phos-phofructokinase-2/fructose-2,6-bisphosphatase 3 suppressed tumor growth and alleviated metastasis in head and neck squamous cell carcinoma. In vitro and in vivo experiments have demonstrated that glycolysis promoted breast cancer growth and metastasis via the regulation of miR-30a-5p (31). These results suggested that targeting glycolysis may be a promising strategy to inhibit cancer growth. The present review subsequently focused on the effects of the mTOR signaling pathway on energy metabolism, especially cancer cell glycolysis (Fig. 3).

Upstream pathways of mTORC1 and glycolysis

mTORC1 integrates four major regulatory inputs: Nutrients, growth factors, energy and stress (32). mTORC1 has been indicated to be frequently overactivated in glycolytic cancer cells, suggesting that the inhibition of factors upstream of mTORC1 may be more effective compared with the inhibition of downstream factors to inhibit glycolysis in cancer cells (4).

Nutrients, mTORC1 and glycolysis

Previous evidence has suggested that amino acids are critical to cancer cell growth by stimulating the mTORC1 signaling pathway, and among these, leucine and glutamine have been revealed to be the most important amino acids (33). Therefore, targeting transporters may be a reasonable strategy to inhibit tumor growth. LAT2, which mainly imports neutral amino acids, including leucine and glutamine, has been indicated to bind to phosphorylated (p)-mTORSer2448 and regulate the glutamine/p-mTORSer2448/glutamine synthetase feedback loop, maintaining mTORC1 activation, which activates glycolysis (34).

Growth factors, mTORC1 and glycolysis

Previous studies have revealed that mTORC1 is a downstream mediator of growth factors (9,35). Insulin, one of these growth factors, has been indicated to upregulate pyruvate kinase M (PKM)2 expression via the PI3K/mTOR-mediated hypoxia inducible factor-1α (HIF-1α) induction, but reduce PKM2 activity independent of this pathway (36,37). Insulin-induced PKM2 upregulation has been revealed to enhance aerobic glycolysis, but the reduction in PKM2 activity may result in a characteristic pooling of glycolytic intermediates and the accumulation of NADPH (37).

Energy and stress regulate mTORC1

mTORC1 can be activated by stress (9), such as low ATP levels (38), hypoxia (39) and DNA damage (40). Mitosis serves a vital role in cellular maintenance and metabolism, and DNA replication is the most vital step of mitosis (41), as cells should check for any DNA damage before proceeding to the subsequent step. mTORC1 has been identified as the determinant for G2/M checkpoint recovery (42) and has been indicated to be required to induce DNA damage repair and cell survival, resulting in increased cancer cell survival in the presence of DNA damage (43). These findings suggested that checkpoint inhibitors present great potential in the treatment of mTORC1-hyperactivated carcinoma.

Upstream pathways of mTORC2

A previous study has indicated that mTORC2 can be activated by growth factors, such as insulin (32). Recent studies have revealed that mTORC2 can also be stimulated by glucose (44), glutamine and mTORC1 (9,45). The mTORC2 pathway is essential for normal glucose homeostasis and metabolic regulation in the body, and the overactivation of mTORC2 may be linked to tumor growth (46). The activation of insulin-like growth factor 1-TORC2 has been indicated to drive the metastasis of nasopharyngeal carcinoma with reprogrammed glucose metabolism (47).

Two facets of AMP kinase (AMPK) and glycolysis

AMPK serves as an energy sensitive receptor in the context of energy stress and can be viewed as a negative regulator of the Warburg effect in human cancers, suggesting that a high level of AMPK indicates a better prognosis (48). Given that both AMPK and mTOR serve important roles in regulating glucose metabolism, it has been demonstrated that AMPK is indeed a regulator of mTOR and phosphorylates regulatory-associated protein of mTOR, which is a component of mTORC1, inhibiting its activity (49). The levels of aerobic glycolysis and tumor growth were indicated to be downregulated by AMPK in vivo, and Myc-induced lymphomagenesis was accelerated when the α1 catalytic subunit of AMPK was ablated (50). Moreover, a metabolic shift to aerobic glycolysis has been observed in both transformed and untransformed cells when AMPK was inactivated, which increased the allocation of glucose-derived carbon into lipids and biomass accumulation (50). However, the role of AMPK in regulating glycolysis may be different in other contexts. For example, tamoxifen has been indicated to inhibit oxygen consumption via inhibiting mitochondrial complex I, increasing the AMP/ATP ratio and activating the AMPK signaling pathway, thereby promoting glycolysis (51).

Moreover, glucose promotes glycolysis by regulating the AMPK/AKT/mTOR/S6 kinase and MAPK pathways (52). The small polyphenol resveratrol has been indicated to inhibit glycolysis in female ovarian cancer cells via activating the AMPK/mTOR signaling pathway (53). However, AMPK has also been demonstrated to regulate glycolysis via phosphorylating and activating 6-phosphofructo-2-kinase/fruc-tose-2,6-bisphosphatase (PFKFB)3, thereby increasing the level of fructose-2,6-bisphosphate (54).

PI3K/AKT/mTOR signaling pathway regulates glycolysis in cancer

Glycolysis, which is a hallmark of cancer cells (4), exhibits a faster ATP production rate compared with oxidative phosphorylation to provide a carbon source for anabolic processes, including the synthesis of amino acids, lipids and nucleotides, although glycolysis produces less energy compared with oxidative phosphorylation (17). The PI3K/AKT/mTOR signaling pathway has been indicated to be a critical pathway during the process of cancer glycolysis (55). It has been demonstrated that targeting the PI3K/AKT/mTOR signaling pathway during metabolic processes presents great potential as an anticancer strategy (55). The expres-sion of glycolysis-related genes, including GLUT1, GLUT3, lactic dehydrogenase (LDH)A, LDHB, HK1, HK2, PKM and HIF-1α, has been indicated to be higher in carcinoma compared with surrounding tissues in bladder cancer (56). Moreover, inhibition of miR-21, which has been revealed to regulate aerobic glycolysis in bladder cancer cells via the PTEN/PI3K/AKT/mTOR pathway (57,58), resulted in a reduction in aerobic glycolysis in cancer cells (56). It has been also demonstrated that inhibitors of PI3K/mTOR suppressed the glycolysis level of EGFR-mutant lung adenocarcinoma (LAD) cells and the levels of early metabolites of glycolysis (59). Moreover, 6-phosphogluconate was revealed to be decreased in response to PI3K/AKT/mTOR pathway inhibition, indicating that PI3K/AKT/mTOR signaling is indispensable for the regulation of aerobic glycolysis in EGFR-mutant LAD cells (59). Furthermore, inhibition of PI3K/mTOR has been indicated to effectively suppress the membrane localization of GLUT1, which is critical for glycolysis (59). However, the molecular mechanism by which PI3K/AKT/mTOR regulates GLUT1 remains to be further elucidated (60).

The upstream and downstream pathways of the PI3K/AKT/mTOR signaling pathway have been revealed to regulate cancer cell glycolysis. For example, knockdown of Forkhead box protein O (FOXO)6, a FOXO family member, has been indicated to inhibit PI3K/AKT/mTOR pathway activation and alter cellular metabolism via inhibiting glycolysis and promoting mitochondrial respiration in colorectal cancer (CRC), indicating that FOXO6 may serve as a potential mTOR-dependent target for CRC therapy (61). Krüppel-like factor 5 (KLF5) silencing has been reported to suppress HIF-1α-dependent glycolysis in non-small cell lung cancer (NSCLC) via inactivating the PI3K/AKT/mTOR pathway (62). Knockdown of glutamate receptor, iono-tropic, N-methyl D-aspartate-associated protein 1, which is also known as GRINA, has been demonstrated to inhibit PI3K/AKT/mTOR signaling and glycolytic metabolism in gastric cancer cells (63). CD147 has been indicated to promote glycolytic metabolism in hepatocellular carcinoma cells via the PI3K/AKT/mTOR signaling pathway (64). CD276 has been reported to enhance the Warburg effect via promoting the PI3K/AKT/mTOR/HIF-1α pathway, as well as its downstream targets GLUT1 and PFKFB3, in oral squamous carcinoma (65). Tripartite motif-containing protein 59 may enhance glycolysis via the PI3K/AKT/mTOR signaling pathway, ultimately contributing to pancreatic cancer progression (66). Type Iγ phosphatidylinositol phosphate kinase has been indicated to upregulate c-Myc and HIF-1α levels by stimulating the PI3K/AKT/mTOR signaling pathway, thereby regulating the expression of glycolytic enzymes to enhance glycolysis in CRC (67).

PI3K/AKT/mTOR signaling pathway and drug resistance

The PI3K/AKT/mTOR signaling pathway is one of the most commonly activated signaling pathways associated with drug resistance in various cancers, such as breast cancer, ovarian cancer and NSCLC (62,68,69). A recent study on the drug resistance of breast cancer has revealed that the PI3K/AKT/mTOR signaling pathway serves an important role in endocrine resistance, since it was indicated to be activated in response to CC-chemokine ligand 2 secreted by tumor-associated macrophages, which promoted an endocrine resistance feedback loop in the tumor microenvironment (68). In addition to breast cancer, the PI3K/AKT/mTOR signaling pathway has also been revealed to facilitate the chemoresistance of ovarian cancer. For example, this pathway has been indicated to trigger the expression of cancer stem cell markers, including CD44v6, CD117, aldehyde dehydrogenase 1 family member A1 and Snail, resulting in cisplatin resistance in epithelial ovarian cancer (70). Hypoxia-induced cisplatin resistance in NSCLC has been demonstrated to be suppressed after knockdown of KLF5, which may occur following inhibition of HIF-1α-dependent glycolysis via PI3K/AKT/mTOR inactivation (62).

Key steps in glycolysis: One input and three processing tools

There are various types of glucose transporters and three key rate-limiting enzymes in glycolysis, HK, PFK and PK (71), and all of these transporters and enzymes have been reported to be associated with the mTOR signaling pathway (23,72).

mTOR signaling pathway and glucose transporters

Glucose transporters act as gatekeepers of glycolysis and have been reported to be elevated to facilitate glucose uptake due to the low efficiency of glycolysis in various types of cancers, such as NSCLC (73), hepatocellular carcinoma and breast cancer (74). A previous study has revealed that glucose uptake and GLUT1 expression were increased in multiple cell types due to the absence of the downstream target protein of mTOR, tuberous sclerosis (TSC)2 protein (75). Moreover, mTORC1 has been indicated to be activated in renal angiomyolipomas as a result of the loss of TSC1/2 function, and TSC2 has been reported to regulate the membrane localization of various glucose transporter proteins, such as GLUT1, GLUT2 and GLUT4, ultimately affecting glucose uptake (76). In addition, the proliferation of CRC has been indicated to be inhibited following GLUT1 gene silencing, which was mediated by the TGF-β/PI3K/AKT/mTOR signaling pathway, providing a novel basis for targeted therapy and the development of novel drugs to treat CRC (77). It has been demonstrated that GLUT1 translocation may be regulated by the PI3K/AKT/mTOR-mediated activation of the RhoA/Rho-associated protein kinase 1 pathway or by stimulating glycolysis, but this remains to be elucidated in the future (59). Certain researchers have focused on the effects of oncogenes that are downstream of the mTOR signaling pathway, including c-Myc and HIF-1α, on the expression of glucose transporters (78,79). In addition to GLUT1, the effects of other members of the glucose family, such as GLUT2 and GLUT5, have also been examined in the context of cancer (74). In studying the role of microRNA (miRNA/miR)-21 in glycolysis and tumorigenesis in T24 bladder cancer cells, it has been revealed that GLUT3 was downregulated when miRNA-21 was sponged and that the associated mechanism may implicate the PI3K/AKT/mTOR signaling pathway (56). Similarly, the expression of GLUT3 has been indicated to be positively regulated by mTORC1 via the activation of IKK/NF-κB signaling, and reduction in aerobic glycolysis, inhibition of cell proliferation, suppression of colony formation and delay in tumor growth have been revealed to occur following GLUT3 knockdown (80).

mTOR signaling pathway and HK2

Glucose phosphorylation, which is the first step in glucose metabolism, is under the control of HK (81). There are five different types of hexokinase isoforms that have been discovered in mammalian cells (82). HK1 is widely expressed in human cells and is considered to be a housekeeping isoform, while HK2 is only expressed in certain tissues, including skeletal muscle, cardiac muscle and adipose tissues (83,84), as well as in cancer cells (85). HK3 is less characterized compared with the other isoforms due to its low levels, and it is considered to be inhibited by glucose under physiological conditions (86). Moreover, recent research has indicated that the upregulation of HK3 is closely associated with the occurrence of EMT in CRC and may be of importance in metabolic adaptation for the rapid proliferation, survival and metastasis of CRC cells (87). HK4, which is also called glucokinase, is expressed in the liver and pancreas (88) and is involved in the migration of regulatory T cells via a PI3K-mTORC2-mediated pathway (89). The importance of HK2 in cancer cells and its relationship with mTOR is highlighted below. HK2 has also been indicated to bind to mitochondrial porins and catalyze the first step of glycolysis (90). AKT can activate HK2 associated with mitochondria, thereby promoting glycolysis (91). In addition, HK2 expression has been revealed to be enhanced by HIF-1α (92,93), which is regulated by the PI3K/AKT/mTORC1 pathway (4,94). To investigate the contributions of c-Src, a proto-oncogene closely associated with mTOR (95,96), to the metabolic reprogramming of cancer cells, Conde et al (92) demonstrated that c-Src phosphorylated HK1 at Tyr732 and HK2 at Tyr686, strengthening their catalytic activity and enhancing glycolysis. Cell glycolysis, proliferation and metastasis were diminished when the c-Src phosphorylation site of either HK1 or HK2 was mutated (97). In addition, HK may be affected by miRNAs. miR-214 downregulation has been revealed to inhibit glycolysis by decreasing the expression of both HK2 and PKM2 via the PTEN/AKT/mTOR pathway in NSCLC cells, revealing the significance of miR-214 and the importance of mTOR-mediated HK-regulated glycolysis in NSCLC treatment (98).

mTOR signaling pathway and PFK

HIF-1 and c-Myc, which are both downstream mediators of the mTOR signaling pathway, are two major regulators of glycolytic enzymes, including HK2, PFK and LDHA (96,99). Fructose 6-phos-phate is converted to fructose 1,6-bisphosphate by PFK-1, which is a well-known 'gatekeeper' of glycolysis, and this constitutes the commitment step of glycolysis (100). PFK-1 is considered to be a key regulator of metabolic repro-gramming in various types of cancers (101,102), including glioblastoma (103), lung cancer (104) and breast cancer (105). It has been indicated that c-Myc directly transactivated genes encoding PFK and increased glucose uptake in Rat-1 fibroblasts (106). A previous study has revealed that PFK-1 platelet isoform (PFKP), which is the main PFK-1 isoform, was overexpressed in human glioblastoma due to the loss of PTEN- and EGFR-mediated PI3K activation, contributing to glycolysis and tumorigenesis (103). These results highlighted the potential role and regulation of PFKP and mTOR-related AKT signaling in human glioblastoma development (103). It has been revealed that PFKFB is a key glycolysis regulator that modulates fructose 2,6-bisphosphate levels and glucose uptake (107). PFKFB3 has been reported to contribute to the development, metastasis and chemotherapy resistance of cancer cells (108-111). Previous studies have indicated that mTORC2 activated AKT by phosphorylating S473, leading to the allosteric activation of PFK-1 (108,110). Moreover, PFKFB3 has been demonstrated to be upregulated by mTORC1, which was dependent on HIF-1α in acute myeloid leukemia (112), and has been indicated to be activated by AKT in another study, which was downstream of mTORC2, thereby contributing to tumor angiogenesis (113). In addition to PFKFB3, PFKFB4 has also been reported to be associated with certain malignancies (114,115). PFKFB4 expression in human bladder cancer has been demonstrated to be associated with hypoxia via HIF-1α (116). The aforementioned results have indicated that mTOR-mediated PFK signaling is tightly associated with carcinogenesis.

mTOR signaling pathway and PK

There are two genes encoding four PK isoforms, where two PK isoforms correspond to each gene (117). The PKLR gene encodes PKL and PKR, while PKM1 and PKM2 are encoded by the PKM gene (118). It has been indicated that there is a potential relationship between PK and mTOR. The final step of glycolysis is catalyzed by PKM2, which converts PEP to pyruvate via the transfer of a phosphate group to ADP (4). PKM2 has been widely studied in past decades, and both inhibitors and activators have been developed for anticancer purposes in different contexts due to the multifaceted characteristics of cancers (119,120). Certain PKM2 activators have been developed to induce the tetramerization of PKM2, causing PKM2 to function as PKM1, which is the dominant glyco-lytic enzyme in healthy adult tissues (121,122). A number of scientists believe that the inhibition of PKM2 occurs as a result of its high expression in various types of cancers, including lung (123), breast (124) and gastric cancer (125). The PI3K/AKT/mTOR pathway has been implicated in regulating PKM2 expression levels, and evidence has indicated that rapamycin decreased PKM2 expression, highlighting the role of mTOR in regulating PKM2 (126,127). HIF-1α and c-Myc have also both been closely associated with the activity of PKM2 (128,129). It has been demonstrated that when mTOR was activated or inhibited, the expression levels of PKM2 and HIF-1α, as well as glycolysis, were altered in esophageal squamous cell carcinoma, indicating that mTOR promoted aerobic glycolysis in esophageal squamous cell carcinoma by upregulating PKM2 expression (130). Yes-associated protein is a powerful regulator that has been indicated to be overexpressed in various types of cancers, such as hepatocellular carcinoma (131), binds to HIF-1α in the nucleus to maintain the stability of HIF-1α protein and also binds to PKM2 gene and directly activates PKM2 transcription to accelerate glycolysis, providing a novel therapeutic target based on HIF-1α (131). Moreover, the interaction of PKM2 with the HIF-1α subunit stimulates PKM2 transactivation via a feedback loop (132). Following ligand stimulation and post-translational modification, PKM2 is transferred into the nucleus activating HIF-1α, thereby promoting the glycolytic pathway (122). The correlation between c-Myc expression and increased PKM2 synthesis in various cancers, such as gastric (133), head and neck (129), liver (134) and breast cancer (135), highlights the possible role of c-Myc in PKM2 expression (134). A recent study has revealed that c-Myc expression was upregulated by p21-activated kinase 2 (PAK2) and that c-Myc bound to the PKM promoter to induce PKM2 expression, which provided a potential framework for head and neck cancer therapy by targeting the PAK2-cMyc-PKM2 axis (129). Furthermore, mTOR has been indicated to regulate TSC1/2 to control the expression of PAK2, but the regulation of glycolysis via PKM2 remains to be explored (136).

mTOR, monocarboxylate transporters (MCTs) and metabolic symbiosis

After the bidirectional enzyme LDH converts pyruvate to lactate, certain cancer cells have been indicated to retain lactate as a metabolic substrate, although others secrete it (4,137,138). Glycolytic cancer cells have been revealed to overexpress MCT-4 to export lactate, maintaining a proper pH for tumor growth, while MCT-1 has been indicated to be highly expressed in normoxic cancer cells in close proximity to blood vessels to import lactate for the synthesis of amino acids (139,140). The phenomenon of lactate re-use in two different types of cells is called metabolic symbiosis, and it is regarded as a potent therapeutic target for cancer (139,141,142). Notably, MCT-1 and MCT-4 have been demonstrated to be critical in metabolic symbiosis (143,144). In arsenite-induced carcinogenesis, it has been reported that arsenite caused high HIF-1α-mediated expression of MCT-4 in liver cells, enhancing glycolysis and resulting in hepatoxicity (145).

5. Perspectives

Cancer cell metabolism shifts from oxidative phosphorylation to glycolysis even in the presence of oxygen, which is also called the Warburg effect, to accommodate the rapid increase in the energy requirements of cancer cells (4). Cancer cells often overexpress catalytic glycolysis enzymes to promote energy production, which is often associated with the abnormal activation of the mTOR signaling pathway (146). All molecules of the entire glycolytic pathway from GLUT to PK are indispensable for glycolysis, and disruption of any factor can lead to glycolysis disorders (4). More importantly, the molecules involved in glycolysis do not function alone, but are regulated by other molecules (147). mTOR-mediated pathways have been revealed to act as central regulators to control the entire glycolytic process (4). Currently, several drugs have been indicated to affect glycolytic proteins, and certain of these drugs will undergo clinical testing (148). Although the Warburg effect has been identified in the past century, its application in cancer has been investigated until recent decades (149). mTOR inhibitors that have been used in the clinic to treat cancer have also been revealed to exert unsatisfactory effects (148). Therefore, in reviewing the importance of aerobic glycolysis and mTOR in carcino-genesis and the limitations of a single application in cancer treatment, we hypothesize that simultaneously blocking glycolysis-associated proteins and the mTOR pathway may be a better alternative for cancer therapy compared with the single-agent inhibition of glycolysis.

Funding

The present study was funded by National Natural Science Foundation of China (grant nos. 81673648, 81673725 and 81673795) and the Natural Science Foundation of Higher School of Jiangsu Province (grant no. 17KJA360003).

Availability of data and materials

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Authors' contributions

WXC, YL and SJW conceived and supervised the study; HF and YYW wrote the original draft; HF, SYY, XML and AYW wrote and edited the review; HF prepared the figures; WXC and YL acquired funding. All the authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Abbreviations:

AMPK

AMP kinase

CRC

colorectal cancer

FOXO6

forkhead box protein O6

GLUT

glucose transporter

HIF

hypoxia inducible factor

HK

hexokinase

KLF5

krüppel-like factor 5

LAD

lung adenocarcinoma

LAT

L-type amino acid transporter

LDH

lactate dehydrogenase

MCT

monocarboxylate transporter

mTOR

mammalian target of rapamycin

NSCLC

non-small cell lung cancer

PAK2

p21-activated kinase 2

PEP

phosphoenolpyruvate

PFK

phosphofructokinase

PK

pyruvate kinase

PKM

PK isozyme M

TSC

tuberous sclerosis complex

References

1 

Vander Heiden MG and DeBerardinis RJ: Understanding the intersections between metabolism and cancer biology. Cell. 168:657–669. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Vijayakrishnapillai LMK, Desmarais JS, Groeschen MN and Perlin MH: Deletion of ptn1, a PTEN/TEP1 orthologue, in ustilago maydis reduces pathogenicity and teliospore development. J Fungi (Basel). 5:12018. View Article : Google Scholar

3 

Huang S, Yang C, Li M, Wang B, Chen H, Fu D and Chong T: Effect of dual mTOR inhibitor on TGFβ1-induced fibrosis in primary human urethral scar fibroblasts. Biomed Pharmacother. 106:1182–1187. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Mossmann D, Park S and Hall MN: mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat Rev Cancer. 18:744–757. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Kenerson HL, Aicher LD, True LD and Yeung RS: Activated mammalian target of rapamycin pathway in the pathogenesis of tuberous sclerosis complex renal tumors. Cancer Res. 62:5645–5650. 2002.PubMed/NCBI

6 

Dowling RJ, Topisirovic I, Fonseca BD and Sonenberg N: Dissecting the role of mTOR: Lessons from mTOR inhibitors. Biochim Biophys Acta. 1804:433–439. 2010. View Article : Google Scholar

7 

Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument-Bromage H, Tempst P and Sabatini DM: mTOR inter-acts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell. 110:163–175. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Hara K, Maruki Y, Long X, Yoshino KI, Oshiro N, Hidayat S, Tokunaga C, Avruch J and Yonezawa K: Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell. 110:177–189. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Saxton RA and Sabatini DM: mTOR signaling in growth, metabolism, and disease. Cell. 169:361–371. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Liu GY and Sabatini DM: mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 21:183–203. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Hayase S, Kumamoto K, Saito K, Kofunato Y, Sato Y, Okayama H, Miyamoto K, Ohki S and Takenoshita S: L-type amino acid transporter 1 expression is upregulated and associated with cellular proliferation in colorectal cancer. Oncol Lett. 14:7410–7416. 2017.

12 

Villar VH, Nguyen TL, Delcroix V, Terés S, Bouchecareilh M, Salin B, Bodineau C, Vacher P, Priault M, Soubeyran P and Durán RV: mTORC1 inhibition in cancer cells protects from glutaminolysis-mediated apoptosis during nutrient limitation. Nat Commun. 8:141242017. View Article : Google Scholar : PubMed/NCBI

13 

van der Vos KE, Eliasson P, Proikas-Cezanne T, Vervoort SJ, van Boxtel R, Putker M, van Zutphen IJ, Mauthe M, Zellmer S, Pals C, et al: Modulation of glutamine metabolism by the PI(3) K-PKB-FOXO network regulates autophagy. Nat Cell Biol. 14:829–837. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Wise DR and Thompson CB: Glutamine addiction: A new therapeutic target in cancer. Trends Biochem Sci. 35:427–433. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Li J, Huang Q, Long X, Zhang J, Huang X, Aa J, Yang H, Chen Z and Xing J: CD147 reprograms fatty acid metabolism in hepatocellular carcinoma cells through Akt/mTOR/SREBP1c and P38/PPARα pathways. J Hepatol. 63:1378–1389. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Harachi M, Masui K, Okamura Y, Tsukui R, Mischel PS and Shibata N: mTOR complexes as a nutrient sensor for driving cancer progression. Int J Mol Sci. 19:32672018. View Article : Google Scholar :

17 

Vander Heiden MG, Cantley LC and Thompson CB: Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 324:1029–1033. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Ganapathy-Kanniappan S: Molecular intricacies of aerobic glycolysis in cancer: Current insights into the classic metabolic phenotype. Crit Rev Biochem Mol Biol. 53:667–682. 2018. View Article : Google Scholar

19 

Chen XS, Li LY, Guan YD, Yang JM and Cheng Y: Anticancer strategies based on the metabolic profile of tumor cells: Therapeutic targeting of the Warburg effect. Acta Pharmacol Sin. 37:1013–1019. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Lei S, Yang J, Chen C, Sun J, Yang L, Tang H, Yang T, Chen A, Zhao H, Li Y and Du X: FLIP(L) is critical for aerobic glycolysis in hepatocellular carcinoma. J Exp Clin Cancer Res. 35:792016. View Article : Google Scholar : PubMed/NCBI

21 

Shi Y, Liu S, Ahmad S and Gao Q: Targeting key transporters in tumor glycolysis as a novel anticancer strategy. Curr Top Med Chem. 18:454–466. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Liu B and Yu S: Amentoflavone suppresses hepatocellular carcinoma by repressing hexokinase 2 expression through inhibiting JAK2/STAT3 signaling. Biomed Pharmacother. 107:243–253. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Gao X and Han H: Jolkinolide B inhibits glycolysis by down-regulating hexokinase 2 expression through inactivating the Akt/mTOR pathway in non-small cell lung cancer cells. J Cell Biochem. 119:4967–4974. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Liu B, Huang ZB, Chen X, See YX, Chen ZK and Yao HK: Mammalian target of rapamycin 2 (MTOR2) and C-MYC modulate glucosamine-6-phosphate synthesis in glioblastoma (GBM) cells through glutamine: Fructose-6-phosphate aminotransferase 1 (GFAT1). Cell Mol Neurobiol. 39:415–434. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Ghashghaeinia M, Koberle M, Mrowietz U and Bernhardt I: Proliferating tumor cells mimick glucose metabolism of mature human erythrocytes. Cell Cycle. 18:1316–1334. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Prakasam G, Singh RK, Iqbal MA, Saini SK, Tiku AB and Bamezai RNK: Pyruvate kinase M knockdown-induced signaling via AMP-activated protein kinase promotes mitochondrial biogenesis, autophagy, and cancer cell survival. J Biol Chem. 292:15561–15576. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Wang R, Jiao H, Zhao J, Wang X and Lin H: L-arginine enhances protein synthesis by phosphorylating mTOR (Thr 2446) in a nitric oxide-dependent manner in C2C12 cells. Oxid Med Cell Longev. 2018:75691272018. View Article : Google Scholar : PubMed/NCBI

28 

Ka M, Smith AL and Kim WY: MTOR controls genesis and autophagy of GABAergic interneurons during brain development. Autophagy. 13:1348–1363. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Caron A, Briscoe DM, Richard D and Laplante M: DEPTOR at the nexus of cancer, metabolism, and immunity. Physiol Rev. 98:1765–1803. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Payen VL, Porporato PE, Baselet B and Sonveaux P: Metabolic changes associated with tumor metastasis, part 1: Tumor pH, glycolysis and the pentose phosphate pathway. Cell Mol Life Sci. 73:1333–1348. 2016. View Article : Google Scholar

31 

Li L, Kang L, Zhao W, Feng Y, Liu W, Wang T, Mai H, Huang J, Chen S, Liang Y, et al: miR-30a-5p suppresses breast tumor growth and metastasis through inhibition of LDHA-mediated Warburg effect. Cancer Lett. 400:89–98. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Zoncu R, Efeyan A and Sabatini DM: mTOR: From growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 12:21–35. 2011. View Article : Google Scholar

33 

Wyant GA, Abu-Remaileh M, Wolfson RL, Chen WW, Freinkman E, Danai LV, Vander Heiden MG and Sabatini DM: mTORC1 activator SLC38A9 is required to efflux essential amino acids from lysosomes and use protein as a nutrient. Cell. 171:642–654 e612. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Feng M, Xiong G, Cao Z, Yang G, Zheng S, Qiu J, You L, Zheng L, Zhang T and Zhao Y: LAT2 regulates glutamine-dependent mTOR activation to promote glycolysis and chemoresistance in pancreatic cancer. J Exp Clin Cancer Res. 37:2742018. View Article : Google Scholar : PubMed/NCBI

35 

Yoshida S, Pacitto R, Yao Y, Inoki K and Swanson JA: Growth factor signaling to mTORC1 by amino acid-laden macropinosomes. J Cell Biol. 211:159–172. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese CB and Cohen P: Characterization of a 3-phos-phoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr Biol. 7:261–269. 1997. View Article : Google Scholar : PubMed/NCBI

37 

Iqbal MA, Siddiqui FA, Gupta V, Chattopadhyay S, Gopinath P, Kumar B, Manvati S, Chaman N and Bamezai RNK: Insulin enhances metabolic capacities of cancer cells by dual regulation of glycolytic enzyme pyruvate kinase M2. Mol Cancer. 12:722013. View Article : Google Scholar : PubMed/NCBI

38 

Neil J, Shannon C, Mohan A, Laurent D, Murali R and Jhanwar-Uniyal M: ATP-site binding inhibitor effectively targets mTORC1 and mTORC2 complexes in glioblastoma. Int J Oncol. 48:1045–1052. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Yang H, Jiang X, Li B, Yang HJ, Miller M, Yang A, Dhar A and Pavletich NP: Mechanisms of mTORC1 activation by RHEB and inhibition by PRAS40. Nature. 552:368–373. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Ma Y, Vassetzky Y and Dokudovskaya S: mTORC1 pathway in DNA damage response. Biochim Biophys Acta Mol Cell Res. 1865:1293–1311. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Dewar JM and Walter JC: Mechanisms of DNA replication termination. Nat Rev Mol Cell Biol. 18:507–516. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Hsieh HJ, Zhang W, Lin SH, Yang WH, Wang JZ, Shen J, Zhang Y, Lu Y, Wang H, Yu J, et al: Systems biology approach reveals a link between mTORC1 and G2/M DNA damage check-point recovery. Nat Commun. 9:39822018. View Article : Google Scholar

43 

Silvera D, Ernlund A, Arju R, Connolly E, Volta V, Wang J and Schneider RJ: mTORC1 and -2 coordinate transcriptional and translational reprogramming in resistance to DNA damage and replicative stress in breast cancer cells. Mol Cell Biol. 37:e005772017. View Article : Google Scholar :

44 

Javary J, Allain-Courtois N, Saucisse N, Costet P, Heraud C, Benhamed F, Pierre R, Bure C, Pallares-Lupon N, Do Cruzeiro M, et al: Liver reptin/RUVBL2 controls glucose and lipid metabolism with opposite actions on mTORC1 and mTORC2 signalling. Gut. 67:2192–2203. 2018. View Article : Google Scholar

45 

Byun JK, Choi YK, Kim JH, Jeong JY, Jeon HJ, Kim MK, Hwang I, Lee SY, Lee YM, Lee IK and Park KG: A positive feedback loop between sestrin2 and mTORC2 is required for the survival of glutamine-depleted lung cancer cells. Cell Rep. 20:586–599. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Yuan T, Lupse B, Maedler K and Ardestani A: mTORC2 signaling: A path for pancreatic β cell's growth and function. J Mol Biol. 430:904–918. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Zhang J, Jia L, Liu T, Yip YL, Tang WC, Lin W, Deng W, Lo KW, You C, Lung ML, et al: mTORC2-mediated PDHE1α nuclear translocation links EBV-LMP1 reprogrammed glucose metabolism to cancer metastasis in nasopharyngeal carcinoma. Oncogene. 38:4669–4684. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Li W, Wong CC, Zhang X, Kang W, Nakatsu G, Zhao Q, Chen H, Go MYY, Chiu PWY, Wang X, et al: CAB39L elicited an anti-Warburg effect via a LKB1-AMPK-PGC1α axis to inhibit gastric tumorigenesis. Oncogene. 37:6383–6398. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Varshney R, Gupta S and Roy P: Cytoprotective effect of kaempferol against palmitic acid-induced pancreatic β-cell death through modulation of autophagy via AMPK/mTOR signaling pathway. Mol Cell Endocrinol. 448:1–20. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Faubert B, Boily G, Izreig S, Griss T, Samborska B, Dong Z, Dupuy F, Chambers C, Fuerth BJ, Viollet B, et al: AMPK is a negative regulator of the Warburg effect and suppresses tumor growth in vivo. Cell Metab. 17:113–124. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Daurio NA, Tuttle SW, Worth AJ, Song EY, Davis JM, Snyder NW, Blair IA and Koumenis C: AMPK activation and metabolic repro-gramming by tamoxifen through estrogen receptor-independent mechanisms suggests new uses for this therapeutic modality in cancer treatment. Cancer Res. 76:3295–3306. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Han J, Zhang L, Guo H, Wysham WZ, Roque DR, Willson AK, Sheng X, Zhou C and Bae-Jump VL: Glucose promotes cell proliferation, glucose uptake and invasion in endometrial cancer cells via AMPK/mTOR/S6 and MAPK signaling. Gynecol Oncol. 138:668–675. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Liu Y, Tong L, Luo Y, Li X, Chen G and Wang Y: Resveratrol inhibits the proliferation and induces the apoptosis in ovarian cancer cells via inhibiting glycolysis and targeting AMPK/mTOR signaling pathway. J Cell Biochem. 119:6162–6172. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Liang J and Mills GB: AMPK: A contextual oncogene or tumor suppressor? Cancer Res. 73:2929–2935. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Hauge M, Bruserud O and Hatfield KJ: Targeting of cell metabolism in human acute myeloid leukemia-more than targeting of isocitrate dehydrogenase mutations and PI3K/AKT/mTOR signaling? Eur J Haematol. 96:211–221. 2016. View Article : Google Scholar

56 

Yang X, Cheng Y, Li P, Tao J, Deng X, Zhang X, Gu M, Lu Q and Yin C: A lentiviral sponge for miRNA-21 dimin-ishes aerobic glycolysis in bladder cancer T24 cells via the PTEN/PI3K/AKT/mTOR axis. Tumour Biol. 36:383–391. 2015. View Article : Google Scholar

57 

Wang P, Guan Q, Zhou D, Yu Z, Song Y and Qiu W: miR-21 inhibitors modulate biological functions of gastric cancer cells via PTEN/PI3K/mTOR pathway. DNA Cell Biol. 37:38–45. 2018. View Article : Google Scholar

58 

Wang WJ, Yang W, Ouyang ZH, Xue JB, Li XL, Zhang J, He WS, Chen WK, Yan YG and Wang C: MiR-21 promotes ECM degradation through inhibiting autophagy via the PTEN/akt/mTOR signaling pathway in human degenerated NP cells. Biomed Pharmacother. 99:725–734. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Makinoshima H, Takita M, Saruwatari K, Umemura S, Obata Y, Ishii G, Matsumoto S, Sugiyama E, Ochiai A, Abe R, et al: Signaling through the phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) axis is responsible for aerobic glycolysis mediated by glucose transporter in epidermal growth factor receptor (EGFR)-mutated lung adeno-carcinoma. J Biol Chem. 290:17495–17504. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Fruman DA and Rommel C: PI3K and cancer: Lessons, challenges and opportunities. Nat Rev Drug Discov. 13:140–156. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Li Q, Tang H, Hu F and Qin C: Silencing of FOXO6 inhibits the proliferation, invasion, and glycolysis in colorectal cancer cells. J Cell Biochem. 120:3853–3860. 2019. View Article : Google Scholar

62 

Gong T, Cui L, Wang H, Wang H and Han N: Knockdown of KLF5 suppresses hypoxia-induced resistance to cisplatin in NSCLC cells by regulating HIF-1α-dependent glycolysis through inactivation of the PI3K/Akt/mTOR pathway. J Transl Med. 16:1642018. View Article : Google Scholar

63 

Xu DH, Li Q, Hu H, Ni B, Liu X, Huang C, Zhang ZZ and Zhao G: Transmembrane protein GRINA modulates aerobic glycolysis and promotes tumor progression in gastric cancer. J Exp Clin Cancer Res. 37:3082018. View Article : Google Scholar : PubMed/NCBI

64 

Li X, Zhang Y, Ma W, Fu Q, Liu J, Yin G, Chen P, Dai D, Chen W, Qi L, et al: Enhanced glucose metabolism mediated by CD147 contributes to immunosuppression in hepatocellular carcinoma. Cancer Immunol Immunother. 69:535–548. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Li Z, Liu J, Que L and Tang X: The immunoregulatory protein B7-H3 promotes aerobic glycolysis in oral squamous carcinoma via PI3K/Akt/mTOR pathway. J Cancer. 10:5770–5784. 2019. View Article : Google Scholar : PubMed/NCBI

66 

Li R, Weng L, Liu B, Zhu L, Zhang X, Tian G, Hu L, Li Q, Jiang S and Shang M: TRIM59 predicts poor prognosis and promotes pancreatic cancer progression via the PI3K/AKT/mTOR-glycolysis signaling axis. J Cell Biochem. 121:1986–1997. 2020. View Article : Google Scholar

67 

Peng W, Huang W, Ge X, Xue L, Zhao W and Xue J: Type Ig phosphatidylinositol phosphate kinase promotes tumor growth by facilitating Warburg effect in colorectal cancer. EBioMedicine. 44:375–386. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Li D, Ji H, Niu X, Yin L, Wang Y, Gu Y, Wang J, Zhou X, Zhang H and Zhang Q: Tumor-associated macrophages secrete CC-chemokine ligand 2 and induce tamoxifen resistance by activating PI3K/Akt/mTOR in breast cancer. Cancer Sci. 111:47–58. 2020. View Article : Google Scholar

69 

Gasparri ML, Besharat ZM, Farooqi AA, Khalid S, Taghavi K, Besharat RA, Sabato C, Papadia A, Panici PB, Mueller MD and Ferretti E: MiRNAs and their interplay with PI3K/AKT/mTOR pathway in ovarian cancer cells: A potential role in platinum resistance. J Cancer Res Clin Oncol. 144:2313–2318. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Deng J, Bai X, Feng X, Ni J, Beretov J, Graham P and Li Y: Inhibition of PI3K/Akt/mTOR signaling pathway alleviates ovarian cancer chemoresistance through reversing epithelial-mesenchymal transition and decreasing cancer stem cell marker expression. BMC Cancer. 19:6182019. View Article : Google Scholar : PubMed/NCBI

71 

Massari F, Ciccarese C, Santoni M, Iacovelli R, Mazzucchelli R, Piva F, Scarpelli M, Berardi R, Tortora G, Lopez-Beltran A, et al: Metabolic phenotype of bladder cancer. Cancer Treat Rev. 45:46–57. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Wang Y, Han X, Fu M, Wang J, Song Y, Liu Y, Zhang J, Zhou J and Ge J: Qiliqiangxin attenuates hypoxia-induced injury in primary rat cardiac microvascular endothelial cells via promoting HIF-1α-dependent glycolysis. J Cell Mol Med. 22:2791–2803. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Koh YW, Lee SJ and Park SY: Differential expression and prognostic significance of GLUT1 according to histologic type of non-small-cell lung cancer and its association with volume-dependent parameters. Lung Cancer. 104:31–37. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Hamann I, Krys D, Glubrecht D, Bouvet V, Marshall A, Vos L, Mackey JR, Wuest M and Wuest F: Expression and function of hexose transporters GLUT1, GLUT2, and GLUT5 in breast cancer-effects of hypoxia. FASEB J. 32:5104–5118. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Buller CL, Loberg RD, Fan MH, Zhu Q, Park JL, Vesely E, Inoki K, Guan KL and Brosius FC III: A GSK-3/TSC2/mTOR pathway regulates glucose uptake and GLUT1 glucose trans-porter expression. Am J Physiol Cell Physiol. 295:C836–C843. 2008. View Article : Google Scholar : PubMed/NCBI

76 

Jiang X, Kenerson H, Aicher L, Miyaoka R, Eary J, Bissler J and Yeung RS: The tuberous sclerosis complex regulates trafficking of glucose transporters and glucose uptake. Am J Pathol. 172:1748–1756. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Wu XL, Wang LK, Yang DD, Qu M, Yang YJ, Guo F, Han L and Xue J: Effects of Glut1 gene silencing on proliferation, differentiation, and apoptosis of colorectal cancer cells by targeting the TGF-β/PI3K-AKT-mTOR signaling pathway. J Cell Biochem. 119:2356–2367. 2018. View Article : Google Scholar

78 

Barron CC, Bilan PJ, Tsakiridis T and Tsiani E: Facilitative glucose transporters: Implications for cancer detection, prognosis and treatment. Metabolism. 65:124–139. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Do SK, Jeong JY, Lee SY, Choi JE, Hong MJ, Kang HG, Lee WK, Seok Y, Lee EB, Shin KM, et al: Glucose transporter 1 gene variants predict the prognosis of patients with early-stage non-small cell lung cancer. Ann Surg Oncol. 25:3396–3403. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Zha X, Hu Z, Ji S, Jin F, Jiang K, Li C, Zhao P, Tu Z, Chen X, Di L, et al: NFκB up-regulation of glucose transporter 3 is essential for hyperactive mammalian target of rapamycin-induced aerobic glycolysis and tumor growth. Cancer Lett. 359:97–106. 2015. View Article : Google Scholar : PubMed/NCBI

81 

DeWaal D, Nogueira V, Terry AR, Patra KC, Jeon SM, Guzman G, Au J, Long CP, Antoniewicz MR and Hay N: Hexokinase-2 depletion inhibits glycolysis and induces oxidative phosphorylation in hepatocellular carcinoma and sensitizes to metformin. Nat Commun. 9:4462018. View Article : Google Scholar : PubMed/NCBI

82 

Hay N: Reprogramming glucose metabolism in cancer: Can it be exploited for cancer therapy? Nat Rev Cancer. 16:635–649. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Esteves JV, Yonamine CY, Pinto-Junior DC, Gerlinger-Romero F, Enguita FJ and Machado UF: Diabetes modulates MicroRNAs 29b-3p, 29c-3p, 199a-5p and 532-3p expression in muscle: Possible role in GLUT4 and HK2 repression. Front Endocrinol (Lausanne). 9:5362018. View Article : Google Scholar

84 

Marampon F, Antinozzi C, Corinaldesi C, Vannelli GB, Sarchielli E, Migliaccio S, Di Luigi L, Lenzi A and Crescioli C: The phosphodiesterase 5 inhibitor tadalafil regulates lipidic homeostasis in human skeletal muscle cell metabolism. Endocrine. 59:602–613. 2018. View Article : Google Scholar

85 

DeWaal D, Nogueira V, Terry AR, Patra KC, Jeon SM, Guzman G, Au J, Long CP, Antoniewicz MR and Hay N: Author correction: Hexokinase-2 depletion inhibits glycolysis and induces oxidative phosphorylation in hepatocellular carcinoma and sensitizes to metformin. Nat Commun. 9:25392018. View Article : Google Scholar : PubMed/NCBI

86 

Kudryavtseva AV, Fedorova MS, Zhavoronkov A, Moskalev AA, Zasedatelev AS, Dmitriev AA, Sadritdinova AF, Karpova IY, Nyushko KM, Kalinin DV, et al: Effect of lentivirus-mediated shRNA inactivation of HK1, HK2, and HK3 genes in colorectal cancer and melanoma cells. BMC Genet. 17(Suppl 3): S1562016. View Article : Google Scholar

87 

Pudova EA, Kudryavtseva AV, Fedorova MS, Zaretsky AR, Shcherbo DS, Lukyanova EN, Popov AY, Sadritdinova AF, Abramov IS, Kharitonov SL, et al: HK3 overexpression associated with epithelial-mesenchymal transition in colorectal cancer. BMC Genomics. 19(Suppl 3): S1132018. View Article : Google Scholar

88 

Fujieda H, Kogami M, Sakairi M, Kato N, Makino M, Takahashi N, Miyazawa T, Harada S and Yamashita T: Discovery of a potent glucokinase activator with a favorable liver and pancreas distribution pattern for the treatment of type 2 diabetes mellitus. Eur J Med Chem. 156:269–294. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Kishore M, Cheung KCP, Fu H, Bonacina F, Wang G, Coe D, Ward EJ, Colamatteo A, Jangani M, Baragetti A, et al: Regulatory T cell migration is dependent on glucokinase-mediated glycolysis. Immunity. 47:875–889 e10. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Yeung SJ, Pan J and Lee MH: Roles of p53, MYC and HIF-1 in regulating glycolysis - the seventh hallmark of cancer. Cell Mol Life Sci. 65:3981–3999. 2008. View Article : Google Scholar : PubMed/NCBI

91 

Roberts DJ and Miyamoto S: Hexokinase II integrates energy metabolism and cellular protection: Akting on mitochondria and TORCing to autophagy. Cell Death Differ. 22:3642015. View Article : Google Scholar : PubMed/NCBI

92 

Conde E, Giménez-Moyano S, Martín-Gómez L, Rodríguez M, Ramos ME, Aguado-Fraile E, Blanco-Sanchez I, Saiz A and García-Bermejo ML: HIF-1α induction during reperfusion avoids maladaptive repair after renal ischemia/reperfusion involving miR127-3p. Sci Rep. 7:410992017. View Article : Google Scholar

93 

Zhang T, Zhu X, Wu H, Jiang K, Zhao G, Shaukat A, Deng G and Qiu C: Targeting the ROS/PI3K/AKT/HIF-1α/HK2 axis of breast cancer cells: Combined administration of polydatin and 2-deoxy-d-glucose. J Cell Mol Med. 23:3711–3723. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Miyazaki M, Miyazaki K, Chen S, Chandra V, Wagatsuma K, Agata Y, Rodewald HR, Saito R, Chang AN, Varki N, et al: The E-Id protein axis modulates the activities of the PI3K-AKT-mTORC1-Hif1a and c-myc/p19Arf pathways to suppress innate variant TFH cell development, thymocyte expansion, and lymphomagenesis. Genes Dev. 29:409–425. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Sudhagar S, Sathya S and Lakshmi BS: Rapid non-genomic signalling by 17β-oestradiol through c-Src involves mTOR-dependent expression of HIF-1α in breast cancer cells. Br J Cancer. 105:953–960. 2011. View Article : Google Scholar : PubMed/NCBI

96 

Huang C, Bruggeman LA, Hydo LM and Miller RT: Shear stress induces cell apoptosis via a c-Src-phospholipase D-mTOR signaling pathway in cultured podocytes. Exp Cell Res. 318:1075–1085. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Zhang J, Wang S, Jiang B, Huang L, Ji Z, Li X, Zhou H, Han A, Chen A, Wu Y, et al: c-Src phosphorylation and activation of hexokinase promotes tumorigenesis and metastasis. Nat Commun. 8:137322017. View Article : Google Scholar : PubMed/NCBI

98 

Zhang K, Zhang M, Jiang H, Liu F, Liu H and Li Y: Down-regulation of miR-214 inhibits proliferation and glycolysis in non-small-cell lung cancer cells via down-regulating the expression of hexokinase 2 and pyruvate kinase isozyme M2. Biomed Pharmacother. 105:545–552. 2018. View Article : Google Scholar : PubMed/NCBI

99 

Singh D, Arora R, Kaur P, Singh B, Mannan R and Arora S: Overexpression of hypoxia-inducible factor and metabolic path-ways: Possible targets of cancer. Cell Biosci. 7:622017. View Article : Google Scholar

100 

Webb BA, Forouhar F, Szu FE, Seetharaman J, Tong L and Barber DL: Structures of human phosphofructokinase-1 and atomic basis of cancer-associated mutations. Nature. 523:111–114. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Yi W, Clark PM, Mason DE, Keenan MC, Hill C, Goddard WA III, Peters EC, Driggers EM and Hsieh-Wilson LC: Phosphofructokinase 1 glycosylation regulates cell growth and metabolism. Science. 337:975–980. 2012. View Article : Google Scholar : PubMed/NCBI

102 

Moreno-Sánchez R, Marin-Hernández A, Gallardo-Pérez JC, Quezada H, Encalada R, Rodríguez-Enríquez S and Saavedra E: Phosphofructokinase type 1 kinetics, isoform expression, and gene polymorphisms in cancer cells. J Cell Biochem. 113:1692–1703. 2012.PubMed/NCBI

103 

Lee JH, Liu R, Li J, Zhang C, Wang Y, Cai Q, Qian X, Xia Y, Zheng Y, Piao Y, et al: Stabilization of phosphofructokinase 1 platelet isoform by AKT promotes tumorigenesis. Nat Commun. 8:9492017. View Article : Google Scholar : PubMed/NCBI

104 

Tang H, Lee M, Sharpe O, Salamone L, Noonan EJ, Hoang CD, Levine S, Robinson WH and Shrager JB: Oxidative stress-responsive microRNA-320 regulates glycolysis in diverse biological systems. FASEB J. 26:4710–4721. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Gomez LS, Zancan P, Marcondes MC, Ramos-Santos L, Meyer-Fernandes JR, Sola-Penna M and Da Silva D: Resveratrol decreases breast cancer cell viability and glucose metabolism by inhibiting 6-phosphofructo-1-kinase. Biochimie. 95:1336–1343. 2013. View Article : Google Scholar : PubMed/NCBI

106 

Holmes B, Lee J, Landon KA, Benavides-Serrato A, Bashir T, Jung ME, Lichtenstein A and Gera J: Mechanistic target of rapamycin (mTOR) inhibition synergizes with reduced internal ribosome entry site (IRES)-mediated translation of cyclin D1 and c-MYC mRNAs to treat glioblastoma. J Biol Chem. 291:14146–14159. 2016. View Article : Google Scholar : PubMed/NCBI

107 

Bartrons R, Simon-Molas H, Rodríguez-Garcia A, Castaño E, Navarro-Sabaté À, Manzano A and Martinez-Outschoorn UE: Fructose 2,6-bisphosphate in cancer cell metabolism. Front Oncol. 8:3312018. View Article : Google Scholar : PubMed/NCBI

108 

Wang C, Qu J, Yan S, Gao Q, Hao S and Zhou D: PFK15, a PFKFB3 antagonist, inhibits autophagy and proliferation in rhabdomyosarcoma cells. Int J Mol Med. 42:359–367. 2018.PubMed/NCBI

109 

Ros S and Schulze A: Balancing glycolytic flux: The role of 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatases in cancer metabolism. Cancer Metab. 1:82013. View Article : Google Scholar : PubMed/NCBI

110 

Cantelmo AR, Conradi LC, Brajic A, Goveia J, Kalucka J, Pircher A, Chaturvedi P, Hol J, Thienpont B, Teuwen LA, et al: Inhibition of the glycolytic activator PFKFB3 in endothelium induces tumor vessel normalization, impairs metastasis, and improves chemotherapy. Cancer Cell. 30:968–985. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Atsumi T, Chesney J, Metz C, Leng L, Donnelly S, Makita Z, Mitchell R and Bucala R: High expression of inducible 6-phos-phofructo-2-kinase/fructose-2,6-bisphosphatase (iPFK-2; PFKFB3) in human cancers. Cancer Res. 62:5881–5887. 2002.PubMed/NCBI

112 

Feng Y and Wu L: mTOR up-regulation of PFKFB3 is essential for acute myeloid leukemia cell survival. Biochem Biophys Res Commun. 483:897–903. 2017. View Article : Google Scholar : PubMed/NCBI

113 

Ziegler ME, Hatch MM, Wu N, Muawad SA and Hughes CC: mTORC2 mediates CXCL12-induced angiogenesis. Angiogenesis. 19:359–371. 2016. View Article : Google Scholar : PubMed/NCBI

114 

Shi L, Pan H, Liu Z, Xie J and Han W: Roles of PFKFB3 in cancer. Signal Transduct Target Ther. 2:170442017. View Article : Google Scholar : PubMed/NCBI

115 

Dasgupta S, Rajapakshe K, Zhu B, Nikolai BC, Yi P, Putluri N, Choi JM, Jung SY, Coarfa C, Westbrook TF, et al: Metabolic enzyme PFKFB4 activates transcriptional coactivator SRC-3 to drive breast cancer. Nature. 556:249–254. 2018. View Article : Google Scholar : PubMed/NCBI

116 

Zhang H, Lu C, Fang M, Yan W, Chen M, Ji Y, He S, Liu T, Chen T and Xiao J: HIF-1α activates hypoxia-induced PFKFB4 expression in human bladder cancer cells. Biochem Biophys Res Commun. 476:146–152. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Zhang Z, Deng X, Liu Y, Liu Y, Sun L and Chen F: PKM2, function and expression and regulation. Cell Biosci. 9:522019. View Article : Google Scholar : PubMed/NCBI

118 

Nguyen A, Loo JM, Mital R, Weinberg EM, Man FY, Zeng Z, Paty PB, Saltz L, Janjigian YY, de Stanchina E and Tavazoie SF: PKLR promotes colorectal cancer liver colonization through induction of glutathione synthesis. J Clin Invest. 126:681–694. 2016. View Article : Google Scholar : PubMed/NCBI

119 

Adem S, Comakli V and Uzun N: Pyruvate kinase activators as a therapy target: A patent review 2011-2017. Expert Opin Ther Pat. 28:61–68. 2018. View Article : Google Scholar

120 

Liu VM and Vander Heiden MG: The role of pyruvate kinase M2 in cancer metabolism. Brain Pathol. 25:781–783. 2015. View Article : Google Scholar : PubMed/NCBI

121 

Warner SL, Carpenter KJ and Bearss DJ: Activators of PKM2 in cancer metabolism. Future Med Chem. 6:1167–1178. 2014. View Article : Google Scholar : PubMed/NCBI

122 

Wang HJ, Hsieh YJ, Cheng WC, Lin CP, Lin YS, Yang SF, Chen CC, Izumiya Y, Yu JS, Kung HJ and Wang WC: JMJD5 regulates PKM2 nuclear translocation and reprograms HIF-1α-mediated glucose metabolism. Proc Natl Acad Sci USA. 111:279–284. 2014. View Article : Google Scholar

123 

Kim DJ, Park YS, Kim ND, Min SH, You YM, Jung Y, Koo H, Noh H, Kim JA, Park KC and Yeom YI: A novel pyruvate kinase M2 activator compound that suppresses lung cancer cell viability under hypoxia. Mol Cells. 38:373–379. 2015. View Article : Google Scholar : PubMed/NCBI

124 

Huang L, Yu Z, Zhang Z, Ma W, Song S and Huang G: Interaction with pyruvate kinase M2 destabilizes tristetraprolin by proteasome degradation and regulates cell proliferation in breast cancer. Sci Rep. 6:224492016. View Article : Google Scholar : PubMed/NCBI

125 

Wang C, Jiang J, Ji J, Cai Q, Chen X, Yu Y, Zhu Z and Zhang J: PKM2 promotes cell migration and inhibits autophagy by mediating PI3K/AKT activation and contributes to the malignant development of gastric cancer. Sci Rep. 7:28862017. View Article : Google Scholar : PubMed/NCBI

126 

van Niekerk G and Engelbrecht AM: Role of PKM2 in directing the metabolic fate of glucose in cancer: A potential therapeutic target. Cell Oncol (Dordr). 41:343–351. 2018. View Article : Google Scholar

127 

Nemazanyy I, Espeillac C, Pende M and Panasyuk G: Role of PI3K, mTOR and Akt2 signalling in hepatic tumorigenesis via the control of PKM2 expression. Biochem Soc Trans. 41:917–922. 2013. View Article : Google Scholar : PubMed/NCBI

128 

Moloughney JG, Kim PK, Vega-Cotto NM, Wu CC, Zhang S, Adlam M, Lynch T, Chou PC, Rabinowitz JD, Werlen G and Jacinto E: mTORC2 responds to glutamine catabolite levels to modulate the hexosamine biosynthesis enzyme GFAT1. Mol Cell. 63:811–826. 2016. View Article : Google Scholar : PubMed/NCBI

129 

Gupta A, Ajith A, Singh S, Panday RK, Samaiya A and Shukla S: PAK2-c-Myc-PKM2 axis plays an essential role in head and neck oncogenesis via regulating Warburg effect. Cell Death Dis. 9:8252018. View Article : Google Scholar : PubMed/NCBI

130 

Xiaoyu H, Yiru Y, Shuisheng S, Keyan C, Zixing Y, Shanglin C, Yuan W, Dongming C, Wangliang Z, Xudong B and Jie M: The mTOR pathway regulates PKM2 to affect glycolysis in esophageal squamous cell carcinoma. Technol Cancer Res Treat. 17:15330338187800632018. View Article : Google Scholar : PubMed/NCBI

131 

Zhang X, Li Y, Ma Y, Yang L, Wang T, Meng X, Zong Z, Sun X, Hua X and Li H: Yes-associated protein (YAP) binds to HIF-1 α and sustains HIF-1α protein stability to promote hepatocellular carcinoma cell glycolysis under hypoxic stress. J Exp Clin Cancer Res. 37:2162018. View Article : Google Scholar

132 

Demaria M and Poli V: PKM2, STAT3 and HIF-1α: The Warburg's vicious circle. JAKSTAT. 1:194–196. 2012.PubMed/NCBI

133 

Gao S, Chen M, Wei W, Zhang X, Zhang M, Yao Y, Lv Y, Ling T, Wang L and Zou X: Crosstalk of mTOR/PKM2 and STAT3/c-Myc signaling pathways regulate the energy metabolism and acidic microenvironment of gastric cancer. J Cell Biochem. 2018.Epub ahead of print.

134 

Mendez-Lucas A, Li X, Hu J, Che L, Song X, Jia J, Wang J, Xie C, Driscoll PC, Tschaharganeh DF, et al: Glucose catabolism in liver tumors induced by c-MYC can be sustained by various PKM1/PKM2 ratios and pyruvate kinase activities. Cancer Res. 77:4355–4364. 2017. View Article : Google Scholar : PubMed/NCBI

135 

Yu P, Li AX, Chen XS, Tian M, Wang HY, Wang XL, Zhang Y, Wang KS and Cheng Y: PKM2-c-Myc-survivin cascade regulates the cell proliferation, migration, and tamoxifen resistance in breast cancer. Front Pharmacol. 11:5504692020. View Article : Google Scholar : PubMed/NCBI

136 

Alves MM, Fuhler GM, Queiroz KC, Scholma J, Goorden S, Anink J, Spek CA, Hoogeveen-Westerveld M, Bruno MJ, Nellist M, et al: PAK2 is an effector of TSC1/2 signaling independent of mTOR and a potential therapeutic target for tuberous sclerosis complex. Sci Rep. 5:145342015. View Article : Google Scholar : PubMed/NCBI

137 

Hui S, Ghergurovich JM, Morscher RJ, Jang C, Teng X, Lu W, Esparza LA, Reya T, Zhan L, Guo JY, et al: Glucose feeds the TCA cycle via circulating lactate. Nature. 551:115–118. 2017. View Article : Google Scholar : PubMed/NCBI

138 

Faubert B, Li KY, Cai L, Hensley CT, Kim J, Zacharias LG, Yang C, Do QN, Doucette S, Burguete D, et al: Lactate metabolism in human lung tumors. Cell. 171:358–371 e359. 2017. View Article : Google Scholar : PubMed/NCBI

139 

Allen E, Mieville P, Warren CM, Saghafinia S, Li L, Peng MW and Hanahan D: Metabolic symbiosis enables adaptive resistance to anti-angiogenic therapy that is dependent on mTOR signaling. Cell Rep. 15:1144–1160. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Kim HK, Lee I, Bang H, Kim HC, Lee WY, Yun SH, Lee J, Lee SJ, Park YS, Kim KM and Kang WK: MCT4 expression is a potential therapeutic target in colorectal cancer with peritoneal carcinomatosis. Mol Cancer Ther. 17:838–848. 2018. View Article : Google Scholar : PubMed/NCBI

141 

Pisarsky L, Bill R, Fagiani E, Dimeloe S, Goosen RW, Hagmann J, Hess C and Christofori G: Targeting metabolic symbiosis to overcome resistance to anti-angiogenic therapy. Cell Rep. 15:1161–1174. 2016. View Article : Google Scholar : PubMed/NCBI

142 

Morrot A, da Fonseca LM, Salustiano EJ, Gentile LB, Conde L, Filardy AA, Franklim TN, da Costa KM, Freire-de-Lima CG and Freire-de-Lima L: Metabolic symbiosis and immunomodulation: How tumor cell-derived lactate may disturb innate and adaptive immune responses. Front Oncol. 8:812018. View Article : Google Scholar : PubMed/NCBI

143 

Pavlova NN and Thompson CB: The emerging hallmarks of cancer metabolism. Cell Metab. 23:27–47. 2016. View Article : Google Scholar : PubMed/NCBI

144 

Curry JM, Tuluc M, Whitaker-Menezes D, Ames JA, Anantharaman A, Butera A, Leiby B, Cognetti DM, Sotgia F, Lisanti MP and Martinez-Outschoorn UE: Cancer metabolism, stemness and tumor recurrence: MCT1 and MCT4 are functional biomarkers of metabolic symbiosis in head and neck cancer. Cell Cycle. 12:1371–1384. 2013. View Article : Google Scholar : PubMed/NCBI

145 

Luo F, Zou Z, Liu X, Ling M, Wang Q, Wang Q, Lu L, Shi L, Liu Y, Liu Q and Zhang A: Enhanced glycolysis, regulated by HIF-1α via MCT-4, promotes inflammation in arsenite-induced carcinogenesis. Carcinogenesis. 38:615–626. 2017. View Article : Google Scholar : PubMed/NCBI

146 

Tan FH, Bai Y, Saintigny P and Darido C: mTOR signalling in head and neck cancer: Heads up. Cells. 8:3332019. View Article : Google Scholar :

147 

Jewell JL and Guan KL: Nutrient signaling to mTOR and cell growth. Trends Biochem Sci. 38:233–242. 2013. View Article : Google Scholar : PubMed/NCBI

148 

Martelli AM, Buontempo F and McCubrey JA: Drug discovery targeting the mTOR pathway. Clin Sci (Lond). 132:543–568. 2018. View Article : Google Scholar

149 

Liberti MV and Locasale JW: The Warburg effect: How does it benefit cancer cells? Trends Biochem Sci. 41:211–218. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 58 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fan H, Wu Y, Yu S, Li X, Wang A, Wang S, Chen W and Lu Y: Critical role of mTOR in regulating aerobic glycolysis in carcinogenesis (Review). Int J Oncol 58: 9-19, 2021
APA
Fan, H., Wu, Y., Yu, S., Li, X., Wang, A., Wang, S. ... Lu, Y. (2021). Critical role of mTOR in regulating aerobic glycolysis in carcinogenesis (Review). International Journal of Oncology, 58, 9-19. https://doi.org/10.3892/ijo.2020.5152
MLA
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 (Review)". International Journal of Oncology 58.1 (2021): 9-19.
Chicago
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 (Review)". International Journal of Oncology 58, no. 1 (2021): 9-19. https://doi.org/10.3892/ijo.2020.5152