Metastases, which are the leading cause of death in patients with cancer, have metabolic vulnerabilities. Alterations in metabolism fuel the energy and biosynthetic needs of metastases but are also needed to activate cell state switches in cells leading to invasion, migration, colonization, and outgrowth in distant organs. Specifically, metabolites can activate protein kinases as well as receptors and they are crucial substrates for posttranslational modifications on histone and nonhistone proteins. Moreover, metabolic enzymes can have moonlighting functions by acting catalytically, mainly as protein kinases, or noncatalytically through protein–protein interactions. Here, we summarize the current knowledge on metabolic signaling in cancer metastasis.

Significance:

Effective drugs for the prevention and treatment of metastases will have an immediate impact on patient survival. To overcome the current lack of such drugs, a better understanding of the molecular processes that are an Achilles heel in metastasizing cancer cells is needed. One emerging opportunity is the metabolic changes cancer cells need to undergo to successfully metastasize and grow in distant organs. Mechanistically, these metabolic changes not only fulfill energy and biomass demands, which are often in common between cancer and normal but fast proliferating cells, but also metabolic signaling which enables the cell state changes that are particularly important for the metastasizing cancer cells.

Metastasis formation is a multistep process that requires cancer cells to dynamically change their phenotype. The early metastatic steps include invasion into the surrounding tissue and dissemination via the blood and lymphatic circulation to distant organs. Once the cancer cells have settled outside the primary tumor, they have to colonize and outgrow in the new organ. In recent years, it has emerged that alterations in metabolism are a prerequisite for the early and later steps of metastasis formation because metabolism responds to but also enables the plasticity and heterogeneity of metastasizing cancer cells (1, 2). Thus, there is a rising interest in exploiting the metabolic vulnerabilities of metastasizing cancer cells for metastases prevention and treatment.

Classic activities of metabolism provide energy, an antioxidant defense, and biomass building blocks for the survival, proliferation, and motility of cancer cells. Research on these more classic functions of the metabolic network in cancer cells was initiated by Otto Warburg 100 years ago when he observed that malignant tumors produce significantly more lactate than non-transformed tissues (3). Thus, we nowadays can rely on a wealth of information regarding the structure and players in the metabolic network and how its activity fuels cancer cells by converting metabolites in enzyme-catalyzed reactions into ATP (energy), amino acids, nucleotides, and fatty acids (precursors for proteins, DNA/RNA and lipids) as well as antioxidants such as glutathione. Yet, in more recent years, it has also become evident that metabolites are not only intermediates of biochemical reactions but that they are also sensed to activate receptor and protein kinase signaling and that they are substrates for posttranslational modifications (PTM) that change protein localization, activity, interaction, and stability (Fig. 1). In addition, for more and more enzymes moonlighting functions have been detected. These nonclassical functions of enzymes include altered catalytic activities, often in the nucleus, where the catalyzed reaction does not involve a metabolite but a protein substrate. Furthermore, also noncatalytic moonlighting functions of enzymes have been observed in which enzyme-protein interactions change the activity, localization, or stability of the partner protein; or enzyme-receptor interactions where the enzyme acts as an agonist or antagonist of the associated receptor. These nonclassical functions of metabolism thus trigger metabolic signaling, here defined as the involvement of metabolites or metabolic enzymes in any noncanonical cellular process. Thus, metabolic signaling constitutes one important intersection of metabolism, with the cellular processes that enable plasticity and heterogeneity of (cancer) cells.

Figure 1.

Metabolism supplies the cell with energy, antioxidant defense, and building blocks (classic functions). Concurrently, metabolites are sensed by the cell via receptors and sensing protein kinases and act as substrates for PTMs, whereas metabolic enzymes exert catalytic and noncatalytic moonlighting functions. P, phosphate group; PTMs, posttranslational modifications. Created with BioRender.com.

Figure 1.

Metabolism supplies the cell with energy, antioxidant defense, and building blocks (classic functions). Concurrently, metabolites are sensed by the cell via receptors and sensing protein kinases and act as substrates for PTMs, whereas metabolic enzymes exert catalytic and noncatalytic moonlighting functions. P, phosphate group; PTMs, posttranslational modifications. Created with BioRender.com.

Close modal

Here, we provide an overview of how metabolic signaling contributes to metastasis formation and where promising strategies may be explored to leverage metabolic signaling for metastasis prevention and treatment.

Protein kinases and receptors allow cells to respond to their internal cell state, neighboring cells, and their environment. classic triggers for the signaling cascades downstream of protein kinases and receptors are cytokines, growth factors, and hormones, but accumulating evidence also highlights the importance of metabolites in triggering and regulating receptor and protein kinase–mediated signaling cascades during metastasis formation (Fig. 2).

Figure 2.

Metabolites transduce signals in both cancer and stromal cells modulating tumor progression and metastasis. They can be sensed by their cognate cell-surface receptors or intracellular protein kinases. Affected early steps of the metastatic cascade are depicted in green, whereas late steps are depicted in orange. Transcription factors are depicted in dark blue, whereas all other proteins are depicted in light blue ellipses. 3PG, 3-phosphoglyceric acid; 3POH-pyr, 3-phospho-hydroxypyruvate; 4-EBP1, eukaryotic translation initiation factor 4E binding protein 1; α-KG, α-ketoglutarate; AMPK, 5′-AMP-activated protein kinase; AKT, protein kinase B; ATP, adenosine triphosphate; BC, breast cancer; Ca2+, bivalent calcium cation; CAMK, calcium/calmodulin dependent protein kinase; cAMP, cyclic adenosine monophosphate; CREB, cAMP responsive element-binding protein; DC, dendritic cell; E2, 17β-estradiol; EC, endothelial cell; ECM, extracellular matrix remodeling; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; EA, elaidic acid; EMT, epithelial–mesenchymal transition; ERK, extracellular signal–regulated kinase; FATP5, fatty acid transport protein-5; Fe2+, ferrous cation; FFA, free fatty acid; FMRP (FMR1), fragile X messenger ribonucleoprotein 1; GKAP, DLG associated protein 1; Glc, glucose; Glu, glutamate; GPER, G protein–coupled estrogen receptor; GPR, G protein–coupled receptor; GRIN2B, glutamate ionotropic receptor NMDA type subunit 2B; H3K9, histone 3 lysine 9; HSF1, heat shock transcription factor 1; HFD, high-fat diet; HIF1α, hypoxia-inducible factor 1 subunit alpha; KDM3B, lysine demethylase 3B; L, lactate; LAT1, L-type amino acid transporter 1; LAT3, L-type amino acid transporter 3; MAPK, mitogen-activated protein kinase; Me2, dimethyl group; MEK (MAP2K1), mitogen-activated protein kinase kinase; MHCII, major histocompatibility complex class II; mTORC1, mammalian target of rapamycin complex 1; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NMDAR, N-methyl-D-aspartate receptor; P, phosphate group; PHGDH, phosphoglycerate dehydrogenase; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; PKA, protein kinase A; PSAT1, phosphoserine aminotransferase 1; RPTOR/RAPTOR, regulatory associated protein of mTOR complex 1; S6, ribosomal protein S6; Ser, serine; SLC6A14, solute carrier family 6 member 14; Src, SRC proto-oncogene nonreceptor tyrosine kinase; STAT3, signal transducer and activator of transcription 3; S, succinate; SUNCR1, succinate receptor 1; TAM, tumor-associated macrophage. Created with BioRender.com.

Figure 2.

Metabolites transduce signals in both cancer and stromal cells modulating tumor progression and metastasis. They can be sensed by their cognate cell-surface receptors or intracellular protein kinases. Affected early steps of the metastatic cascade are depicted in green, whereas late steps are depicted in orange. Transcription factors are depicted in dark blue, whereas all other proteins are depicted in light blue ellipses. 3PG, 3-phosphoglyceric acid; 3POH-pyr, 3-phospho-hydroxypyruvate; 4-EBP1, eukaryotic translation initiation factor 4E binding protein 1; α-KG, α-ketoglutarate; AMPK, 5′-AMP-activated protein kinase; AKT, protein kinase B; ATP, adenosine triphosphate; BC, breast cancer; Ca2+, bivalent calcium cation; CAMK, calcium/calmodulin dependent protein kinase; cAMP, cyclic adenosine monophosphate; CREB, cAMP responsive element-binding protein; DC, dendritic cell; E2, 17β-estradiol; EC, endothelial cell; ECM, extracellular matrix remodeling; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; EA, elaidic acid; EMT, epithelial–mesenchymal transition; ERK, extracellular signal–regulated kinase; FATP5, fatty acid transport protein-5; Fe2+, ferrous cation; FFA, free fatty acid; FMRP (FMR1), fragile X messenger ribonucleoprotein 1; GKAP, DLG associated protein 1; Glc, glucose; Glu, glutamate; GPER, G protein–coupled estrogen receptor; GPR, G protein–coupled receptor; GRIN2B, glutamate ionotropic receptor NMDA type subunit 2B; H3K9, histone 3 lysine 9; HSF1, heat shock transcription factor 1; HFD, high-fat diet; HIF1α, hypoxia-inducible factor 1 subunit alpha; KDM3B, lysine demethylase 3B; L, lactate; LAT1, L-type amino acid transporter 1; LAT3, L-type amino acid transporter 3; MAPK, mitogen-activated protein kinase; Me2, dimethyl group; MEK (MAP2K1), mitogen-activated protein kinase kinase; MHCII, major histocompatibility complex class II; mTORC1, mammalian target of rapamycin complex 1; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NMDAR, N-methyl-D-aspartate receptor; P, phosphate group; PHGDH, phosphoglycerate dehydrogenase; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; PKA, protein kinase A; PSAT1, phosphoserine aminotransferase 1; RPTOR/RAPTOR, regulatory associated protein of mTOR complex 1; S6, ribosomal protein S6; Ser, serine; SLC6A14, solute carrier family 6 member 14; Src, SRC proto-oncogene nonreceptor tyrosine kinase; STAT3, signal transducer and activator of transcription 3; S, succinate; SUNCR1, succinate receptor 1; TAM, tumor-associated macrophage. Created with BioRender.com.

Close modal

mTORC1 and AMPK

mTORC1

The mammalian target of rapamycin complex 1 (mTORC1) signaling, which is induced by metabolites and growth factors, has been studied for decades in cancer proliferation. Although amino acids are not the sole metabolites involved, they play a pivotal role as the primary metabolites activating mTORC1 signaling. Recent studies showed that amino acid activation of mTORC1 signaling is similar between primary tumors and metastases, but differences may exist for activation of mTORC1 by other metabolites such as α-ketoglutarate (Fig. 2).

mTOR is activated by intracellular amino acids. Therefore, amino acid transporters become very relevant to mTORC1 activation. In the context of mTORC1 signaling in metastasis mainly the role of L-type amino acid transporters has been studied and similar effects on primary tumors and metastasis were observed. Genetic LAT1 (SLC7A5) or LAT3 (SLC7A6) loss suppressed primary tumor growth and spontaneous metastasis formation in prostate cancer xenograft (4) and colorectal cancer (5) mouse models. Moreover, it was recently shown that LAT3 expression depends on sufficient iron levels (6), which could explain the correlation of high-circulating iron levels with poor prognosis in some cancers (7). Similarly, deletion of the general amino acid transporter Slc6a14, which can transport 18 out of 20 amino acids, decreased primary tumor growth as well as metastasis formation and consequently improved overall survival in pancreatic ductal adenocarcinoma KPC mouse models (8). All the aforementioned studies proposed a direct or indirect involvement of mTORC1 activation in metastasis formation via the SLC-transported amino acids. Yet, the observation that simultaneous loss of Lat1 and targeting of mTORC1 activity with rapamycin in murine Kras-mutant colorectal cancer significantly extended lifespan compared with only Lat1 loss suggests roles of LAT1 in tumor growth and metastasis that go beyond direct activation of mTORC1 (5).

There is also evidence that mTORC1 activation may differ between primary tumors and the corresponding metastases. In patients with breast cancer, a discordance between mTORC1 activity in primary tumors and matched metastases assessed by phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1 (p-4E-BP1) was observed (9). In line, a higher mTORC1 activity in lymph node metastases and general metastases compared with primary tumors was seen for medullary thyroid carcinoma (assessment of p-S6) and renal cell carcinoma (assessment of p-mTOR) samples from patients, respectively (10, 11). Consistently, increased mTORC1 activity based on p-S6 and p-4-EBP1 was observed in lung metastases compared with primary breast cancer of mice (12). Mechanistically, it was found that mTORC1 was activated in lung metastases by α-ketoglutarate derived from an increased de novo serine biosynthesis (12, 13). In contrast, serine biosynthesis was not needed for mTOR activation in primary breast tumors (12). Accordingly, lung metastases only responded to rapamycin when the tumors expressed phosphoglycerate dehydrogenase (Phgdh), which is the first enzyme of the serine biosynthesis pathway (12).

AMPK

mTORC1 is negatively regulated by AMP-activated protein kinase (AMPK) activity, which is a master regulator of energy homeostasis and accordingly has implications for cancer progression (Fig. 2). AMPK activation is triggered by elevated AMP/ATP and ADP/ATP ratios. Thus, the loss of Ampk catalytic subunit alpha 1 and 2 (AMPKα1 and 2, i.e., PRKAA1 and 2) has been shown to promote lung and brain metastasis from breast cancer (14) and melanoma mouse models (15), respectively. Moreover, decreased expression of the fatty acid transport protein 5 (FATP5/SLC27A5) enhanced both, hepatocellular carcinoma growth and metastasis, by suppressing AMPK activation (16). Mechanistically, loss of FATP5 increased glucose uptake and consequently glucose-derived ATP production leading to AMPK inhibition and eventually mTORC1 activation in HCC cells (16). In line, the reactivation of AMPK using metformin impaired the metastatic capacity of FATP5-depleted hepatocellular carcinoma (HCC) cells (16). In triple-negative breast cancer, however, the disruption of mitochondrial ATP production and the subsequent activation of AMPK signaling, achieved by chelating copper with tetramolybdate, only hindered lung metastasis formation without affecting the growth of the primary tumor (17). This suggests that the same metabolic effect of impaired ATP production may have different effects depending on the tumor context and the trigger inducing the ATP shortage (Fig. 2).

In summary, although mTORC1 and AMPK signaling commonly hold significance in both primary tumors and metastases, there are instances and cancer types where the metabolic activation of the AMPK-mTORC1 axis diverts between metastases and primary tumors.

GPCR Signaling

G-protein–coupled receptors (GPR or GPCR) constitute a cell-surface receptor superfamily of over 800 members, for many of them odors, chemokines, neurotransmitters, and hormones act as agonists. So far only a small fraction of GPRs have been identified that use small molecules as activators and some of them have been implicated in the cancer cell–stroma interaction during metastasis formation (Fig. 2).

Free Fatty Acids

Free fatty acids are known agonists of GPR40 and 120. In patients, little is known about the concentrations and origin of the free fatty acids that activate these GPRs. Yet, GPR40 expression was found to be correlated with the occurrence of metastasis in colorectal cancer (18) and melanoma (19) patients. Moreover, GPR120 expression correlated with lymph node metastasis and poor prognosis in PDAC patients (20). Multiple fatty acids including elaidic acid can activate GPR120 and GPR40. Interestingly, blood concentrations of palmitoleic acid and elaidic acid have been associated with colorectal cancer in patients (21). In line, it was found that treatment of colorectal cancer cells with elaidic acid promoted in vitro sphere formation, which was abrogated when both GPR40 and 120 were silenced (22). Moreover, oral administration of elaidic acid to mice promoted colorectal cancer xenograft and subsequent metastasis growth (22). A prometastatic influence of GPR40 and 120 signaling observed in colorectal cancer and papillary renal cell carcinoma (pRCC) cells converged into the activation of the epidermal growth factor receptor (EGFR) signaling pathway (22, 23).

Succinate

Succinate has been identified as an agonist of GPR91, which was later renamed to succinate receptor 1 (SUCNR1). Plasma succinate levels are not only a biomarker for cancers with succinate dehydrogenase (SDH) mutation but can also distinguish patients with non–small cell lung cancer (NSCLC) from healthy individuals, likely because some cancer cells secrete succinate due to reduced SDH activity (24). In this respect, it was recently shown that besides the canonical succinate transporter SLC13A3, the monocarboxylate transporter 1 (MCT1) can shuffle succinate across the plasma membrane (25). Moreover, tumor reoccurrence in patients with head and squamous cell carcinoma correlated with high SUCNR1 expression (26). Activation of Sucnr1 by succinate mainly occurred in stromal cells such as macrophages, CD8+ T cells, and endothelial cells of the tumor microenvironment (27–29), but also autocrine activation of the receptor in cancer cells has been observed (24). In stromal cells, the antitumor activity of CD8+ T cells required autocrine Sucnr1 activation (28), whereas protumor M2-polarized macrophages and angiogenesis were stimulated upon Sucnr1 activation by cancer cell–derived succinate (27, 29). Treatment of murine lung cancers with succinate was shown to increase metastasis formation in multiple organs including lung and liver, whereas primary tumor growth was not affected (24). Moreover, exosomes from human umbilical cord mesenchymal stem cells reduced SUCNR1 protein expression via the miR-1827 in cancer cells, which decreased primary tumor growth and metastasis formation of colorectal cancer xenografts. The effect of this exosome treatment was rescued by overexpression of SUCNR1 in cancer cells (30). Mechanistically, it was shown that autocrine activation of SUCNR1 by succinate activated an epithelial-to-mesenchymal transition via phosphoinositide 3-kinase (PI3K) and hypoxia-inducible factor 1 subunit alpha (HIF1α) cascade in NSCLC cells (24).

Lactate

GPR81 and GPR132 have been reported as lactate receptors. Cancer cell–derived lactate promoted autocrine or, in the tumor microenvironment, paracrine receptor activation that has been linked in breast cancer to metastasis formation. Accordingly, lower GPR132 expression correlated with increased metastasis-free survival in patients with breast cancer (31). Moreover, injection of murine breast cancer cells to Gpr132 knockout mice and crossing of the genetic PyMT breast cancer mouse model into Gpr81-null mice resulted in reduced lung metastasis and, in the case of the PyMT/Gpr81−/− mice, also delayed tumor onset (31, 32).

Mechanistically, the observed outcomes in both cases were associated with paracrine signaling where cancer cell–derived lactate interacted with Gpr132 on macrophages or Gpr81 on dendritic cells, ultimately contributing to protumor microenvironments. These microenvironments were characterized by a reduced presentation of tumor-specific antigens by dendritic cells (Gpr81-null mice) and the polarization of macrophages toward the M2 phenotype (Gpr132-null mice). The latter observation may be breast cancer-specific because, in lung cancers growing in Gpr132-null mice, no differential macrophage polarization was observed (33). Notably, Gpr81 activation during sensory neuron excitation has been linked to bone pain induced by metastatic breast cancer hinting at a potential link between this metabolic signaling and tumor innervation (34).

Also, cancer cell–intrinsic effects of GPR81 silencing have been observed. In multiple cell lines including breast cancer cells, GPR81 silencing reduced in vitro cancer cell survival and motility (35, 36). Moreover, the knockdown of GPR81 in human breast cancer cells implanted in the bone showed reduced growth suggesting a cancer cell–intrinsic role of lactate signaling in metastasis formation (37).

In summary, GPRs activated by metabolites are understudied in cancer, and current evidence shows that cancer cell–intrinsic but also paracrine GPR signaling contributes to metastasis formation. Preclinical antagonists against SUCNR1 have been developed (38, 39), yet it was recently discovered that extracellular succinate-SUCNR1 signaling was important for inducing transcriptional responses for muscle remodeling upon exercise (40) and adipose tissue thermogenesis (41). This suggests that further studies are needed to evaluate GPRs as drug targets in cancer metastasis.

NMDA Receptor

Presynaptic neurons, in the brain parenchyma, secrete vast amounts of glutamate, which acts as an excitatory neurotransmitter in the central nervous system by activating the ionotropic N-methyl-D-aspartate (NMDA) receptor. Cancer cells can hijack this neuron-specific signaling cascade.

Specifically, it was shown that a human brain tropic triple-negative breast cancer cell line highly expressed the NMDA receptor subunit GRIN2B compared with the parental cell line and that the corresponding brain metastases had an elevated GRIN2B expression compared with lung metastases and the primary breast tumor (42). Moreover, genetically engineered mouse models of pancreatic neuroendocrine tumor (PNET), pancreatic adenocarcinoma, and luminal breast cancer showed GRIN2B expression predominantly in the invasive front of the tumors (43). Supporting these data, increased GRIN2B expression was observed in PDAC, breast cancer, ovarian cancer, and glioma patient samples (43, 44). Underscoring the importance of this finding, it was shown that silencing GRIN2B reduced breast cancer–derived brain metastases in mice (42). The NMDA receptor inhibitor MK801 prevented and regressed PNET tumors in mice, whereas the weaker clinically approved inhibitor memantine only regressed them (43). Both inhibitors prolonged the survival of mice harboring PDAC tumors (45) and MK801 treatment arrested breast cancer xenograft growth (46) as well as lung metastasis (47). Accordingly, in patients with breast cancer, elevated GRIN2B expression was associated with poor distant-relapse-free survival and a gene-expression signature indicative of low NMDA receptor activity correlated with improved survival in PDAC, brain cancer, kidney cancer, and uveal melanoma (42, 45).

Glutamate is an agonist of the NMDA receptor. It was found that the glutamate activating the NMDA receptor in breast cancer–derived brain metastases from mice was provided through the formation of pseudotripartite synapses between cancer cells and glutamatergic neurons (42). Yet, also an activation of the NMDA receptor by glutamate released from the cancer cells themselves was observed in PNET cell lines. This glutamate release was triggered by low interstitial fluid pressure, which is found at the invasive front of a tumor (43). Moreover, increased secretion of glutamate was also triggered by 17-β estradiol (E2)-mediated activation of G protein–coupled estrogen receptor (GPER) in triple-negative breast cancer cells (47). Once glutamate activated the NMDA receptor, the associated Ca2+ influx promoted cAMP response element-binding protein (CREB) phosphorylation and the downstream expression of a proinvasive and prometastatic gene signatures through a multistep signaling cascade in breast and PNET cancer cells (43, 47). Moreover, there are indications that activation of the NMDA receptor also promoted the translation of proinvasive genes via activation of the heat shock transcription factor 1 (HSF1) and Fragile X Messenger Ribonucleoprotein 1 (FMR1) axis in PNET cancer cells (45). Finally, the activity of the matrix metalloproteinase 2 (MMP2), which can promote invasion, was correlated with NMDA receptor activity in glioma cell lines (48).

Thus, cancer cells exploit the activation of the NMDA receptor by autocrine- or paracrine-released glutamate to gain invasive capacities promoting metastasis formation. Clinically, it is interesting to target the NMDA receptor in cancer metastasis because several approved inhibitors exist. Esketamine and dextromethorphan are approved for major depressive disorder, whereas memantine is approved for dementia and amantadine for the treatment of Parkinson's disease. Epidemiologic studies analyzing the metastasis risk of patients taking NMDA receptor antagonists, and especially memantine, for which antitumor effects have been reported in mouse models (43, 49, 50), are currently missing.

Metabolites are substrates for PTMs of proteins. Enzymes known as transferases facilitate the covalent attachment of metabolites to proteins, whereas erasers undo these modifications. Furthermore, the extent of protein modification is intricately linked to the concentration of available metabolite substrates. Metabolite substrate concentrations and transferase/eraser expression modulate PTMs in metastasis formation (Figs. 3 and 4). Although transferases and the corresponding erasers are currently explored as drug targets, approaches to modulate metabolite concentrations in metastasizing cancer cells are mostly indirect. Moreover, how diet influences meta­bolite concentrations and consequently PTMs in metastasis formation is only emerging.

Figure 3.

Glycosylation in cancer metastasis. Posttranslational glycosylation events mostly promote the initial steps of metastasis formation by modulating proteins, both in the cancer and stromal cells, through A, GalNAc-mediated O-linked protein glycosylation, B, sialylation, and C, O-GlcNAcylation and N-linked glycosylation. Affected early steps of the metastatic cascade are depicted in green. Transcription factors are depicted in dark blue and writers and erasers of PTMs in yellow, whereas all other proteins are depicted in light blue ellipses. AXL, AXL receptor tyrosine kinase; CatB, cathepsin B; CD82, cluster of differentiation 82; CDK4, cyclin-dependent kinase 4; CMAS, cytidine monophosphate N-acetylneuraminic acid synthetase; CMP-sialic acid, cytidine monophosphate-sialic acid; Cnx, calnexin; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EMT, epithelial–mesenchymal transition; ER, endoplasmic reticulum; ERp57, endoplasmic reticulum protein 57; EV, extracellular vesicle; F6P, fructose 6-phosphate; FN, fibronectin; FOXA1, forkhead box A1; GalNAc, N-acetylgalactosamine; GALNT, polypeptide N-acetylgalactosaminyltransferase; GFAT1, glutamine–fructose-6-phosphate transaminase 1; Glc, glucose; Gln, glutamine; GLUT1, glucose transporter type 1; MCAM, melanoma cell adhesion molecule; MECP2, methyl-CpG binding protein 2; MGAT, mannosyl (alpha-1,3-)-glycoprotein beta-1,2-N-acetylglucosaminyltransferase; MMP, matrix metalloproteinase; NADK, nicotinamide adenine dinucleotide kinase; NEU, sialidase; RB1, retinoblastoma 1; ST3/6GAL, beta-galactoside alpha-2,3-sialyltransferase; TAF, tumor-associated fibroblast; TAM, tumor-associated macrophage, UDP-GlcNAc, uridine diphosphate N-acetylglucosamine. Created with BioRender.com.

Figure 3.

Glycosylation in cancer metastasis. Posttranslational glycosylation events mostly promote the initial steps of metastasis formation by modulating proteins, both in the cancer and stromal cells, through A, GalNAc-mediated O-linked protein glycosylation, B, sialylation, and C, O-GlcNAcylation and N-linked glycosylation. Affected early steps of the metastatic cascade are depicted in green. Transcription factors are depicted in dark blue and writers and erasers of PTMs in yellow, whereas all other proteins are depicted in light blue ellipses. AXL, AXL receptor tyrosine kinase; CatB, cathepsin B; CD82, cluster of differentiation 82; CDK4, cyclin-dependent kinase 4; CMAS, cytidine monophosphate N-acetylneuraminic acid synthetase; CMP-sialic acid, cytidine monophosphate-sialic acid; Cnx, calnexin; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EMT, epithelial–mesenchymal transition; ER, endoplasmic reticulum; ERp57, endoplasmic reticulum protein 57; EV, extracellular vesicle; F6P, fructose 6-phosphate; FN, fibronectin; FOXA1, forkhead box A1; GalNAc, N-acetylgalactosamine; GALNT, polypeptide N-acetylgalactosaminyltransferase; GFAT1, glutamine–fructose-6-phosphate transaminase 1; Glc, glucose; Gln, glutamine; GLUT1, glucose transporter type 1; MCAM, melanoma cell adhesion molecule; MECP2, methyl-CpG binding protein 2; MGAT, mannosyl (alpha-1,3-)-glycoprotein beta-1,2-N-acetylglucosaminyltransferase; MMP, matrix metalloproteinase; NADK, nicotinamide adenine dinucleotide kinase; NEU, sialidase; RB1, retinoblastoma 1; ST3/6GAL, beta-galactoside alpha-2,3-sialyltransferase; TAF, tumor-associated fibroblast; TAM, tumor-associated macrophage, UDP-GlcNAc, uridine diphosphate N-acetylglucosamine. Created with BioRender.com.

Close modal
Figure 4.

Metabolite-driven posttranslational modifications in cancer metastasis. A, Metabolite-derived direct (enzymatic) and indirect (metabolite availability) acetylation modulate metastasis initiation by regulating the expression of pro- and antitumoral genes. Left, acetyl-CoA- and palmitate-derived epigenetic modulation influence both early and late steps of the metastatic cascade. Acetylation-induced EMT processes are reviewed by (93). Right, expression of ACACA in the lung fibroblasts of the premetastatic niche (PMN) is modulated by breast cancer primary tumor. Downregulated ACACA levels promote metastasis formation by regulating senescence and immunosuppression processes in the PMN. B, The effect of protein palmitoylation is context specific, as it drives both pro- and antimetastatic effects. C, Increased levels of the metabolite lactate alter protein lactylation processes, driving early metastatic events. D, Nutrient abundances modulate prometastatic histone methylation. E, Dysregulated balance between SAM/SAH levels, mediated by the enzyme NNMT, regulates metastatic events by blocking antimetastatic histone methylation. Affected early steps of the metastatic cascade are depicted in green, whereas late steps are depicted in orange. Transcription factors are depicted in dark blue and writers and erasers of PTMs in yellow, whereas all other proteins are depicted in light blue ellipses. 1NMA, 1-methylnicotinamide; ACACA, acetyl-CoA carboxylase alpha; ACAT1, acetyl-CoA acetyltransferase 1; Ac, acetyl group; ACOX1, Acyl-CoA oxidase 1; AK2, adenylate kinase 2; ATP, adenosine triphosphate; β-cat, beta-catenin; BC, breast cancer; CD36, cluster of differentiation 36; CDH1, cadherin-1/E-cadherin; CoA, coenzyme A; DCBLD1, discoidin, CUB and LCCL domain containing 1; DHHC, DHHC-type palmitoyltransferase; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EMT, epithelial–mesenchymal transition; ERK, extracellular-signal regulated kinase; FAK, focal adhesion kinase; FA, fatty acid; FASN, fatty acid synthase; FLOT1, flotillin-1; GAL, galanin; Glc, glucose; GLUT1, glucose transporter type 1; Gly, glycine; HDAC, histone deacetylase; HFD, high-fat diet; H3K4, histone 3 lysine 4; H3K9, histone 3 lysine 9; H3K27, histone 3 lysine 27; IGF-1R, insulin-like growth factor 1 receptor; JAM3, junctional adhesion molecule 3; KAT, lysine acetyltransferase; L, lactate; La, lactylation, LDH, lactate dehydrogenase; LDHA, lactate dehydrogenase A; LYPLA1, lysophospholipase 1; MAT2A, methionine adenosyltransferase 2A; MCT1, monocarboxylate transporter 1; MEK, mitogen-activated protein kinase kinase; Me, methyl group; Met, methionine; METTL3, methyltransferase like 3; NAM, nicotinamide; NEDD9, neural precursor cell expressed, developmentally downregulated 9; NNMT, nicotinamide N-methyltransferase; NUSAP1, nucleolar and spindle associated protein 1; pFAO, peroxisomal fatty acid oxidation; P, phosphate group; P300, E1A binding protein P300; p65, transcription factor p65 (RelA); PP2A, protein phosphatase 2A; PPP, pentose phosphate pathway; Pyr, pyruvate; RhoU, Ras homolog family member U; SAH, S-adenosylhomocysteine; SAM, S-adenosylmethionine; Ser, serine; SET1A, histone-lysine N-methyltransferase SET1A; SIRT, sirtuin; SMAD3, SMAD family member 3; SOD2, superoxide dismutase 2; STAT3, signal transducer and activator of transcription 3; Ub, ubiquitin; ZMYND8, zinc finger MYND-type containing 8. Created with BioRender.com.

Figure 4.

Metabolite-driven posttranslational modifications in cancer metastasis. A, Metabolite-derived direct (enzymatic) and indirect (metabolite availability) acetylation modulate metastasis initiation by regulating the expression of pro- and antitumoral genes. Left, acetyl-CoA- and palmitate-derived epigenetic modulation influence both early and late steps of the metastatic cascade. Acetylation-induced EMT processes are reviewed by (93). Right, expression of ACACA in the lung fibroblasts of the premetastatic niche (PMN) is modulated by breast cancer primary tumor. Downregulated ACACA levels promote metastasis formation by regulating senescence and immunosuppression processes in the PMN. B, The effect of protein palmitoylation is context specific, as it drives both pro- and antimetastatic effects. C, Increased levels of the metabolite lactate alter protein lactylation processes, driving early metastatic events. D, Nutrient abundances modulate prometastatic histone methylation. E, Dysregulated balance between SAM/SAH levels, mediated by the enzyme NNMT, regulates metastatic events by blocking antimetastatic histone methylation. Affected early steps of the metastatic cascade are depicted in green, whereas late steps are depicted in orange. Transcription factors are depicted in dark blue and writers and erasers of PTMs in yellow, whereas all other proteins are depicted in light blue ellipses. 1NMA, 1-methylnicotinamide; ACACA, acetyl-CoA carboxylase alpha; ACAT1, acetyl-CoA acetyltransferase 1; Ac, acetyl group; ACOX1, Acyl-CoA oxidase 1; AK2, adenylate kinase 2; ATP, adenosine triphosphate; β-cat, beta-catenin; BC, breast cancer; CD36, cluster of differentiation 36; CDH1, cadherin-1/E-cadherin; CoA, coenzyme A; DCBLD1, discoidin, CUB and LCCL domain containing 1; DHHC, DHHC-type palmitoyltransferase; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EMT, epithelial–mesenchymal transition; ERK, extracellular-signal regulated kinase; FAK, focal adhesion kinase; FA, fatty acid; FASN, fatty acid synthase; FLOT1, flotillin-1; GAL, galanin; Glc, glucose; GLUT1, glucose transporter type 1; Gly, glycine; HDAC, histone deacetylase; HFD, high-fat diet; H3K4, histone 3 lysine 4; H3K9, histone 3 lysine 9; H3K27, histone 3 lysine 27; IGF-1R, insulin-like growth factor 1 receptor; JAM3, junctional adhesion molecule 3; KAT, lysine acetyltransferase; L, lactate; La, lactylation, LDH, lactate dehydrogenase; LDHA, lactate dehydrogenase A; LYPLA1, lysophospholipase 1; MAT2A, methionine adenosyltransferase 2A; MCT1, monocarboxylate transporter 1; MEK, mitogen-activated protein kinase kinase; Me, methyl group; Met, methionine; METTL3, methyltransferase like 3; NAM, nicotinamide; NEDD9, neural precursor cell expressed, developmentally downregulated 9; NNMT, nicotinamide N-methyltransferase; NUSAP1, nucleolar and spindle associated protein 1; pFAO, peroxisomal fatty acid oxidation; P, phosphate group; P300, E1A binding protein P300; p65, transcription factor p65 (RelA); PP2A, protein phosphatase 2A; PPP, pentose phosphate pathway; Pyr, pyruvate; RhoU, Ras homolog family member U; SAH, S-adenosylhomocysteine; SAM, S-adenosylmethionine; Ser, serine; SET1A, histone-lysine N-methyltransferase SET1A; SIRT, sirtuin; SMAD3, SMAD family member 3; SOD2, superoxide dismutase 2; STAT3, signal transducer and activator of transcription 3; Ub, ubiquitin; ZMYND8, zinc finger MYND-type containing 8. Created with BioRender.com.

Close modal

Protein Glycosylation

Glycosylation is the attachment of a carbohydrate to a protein (or lipid). Various modifications exist such as glucuronidation, O-GlcNAcylation, and sialylation, which use UDP-glucuronic acid, UDP-acetylglucosamine (O-GlcNAc), and CMP-sialic acid as metabolite substrate, respectively. It has been consistently observed across various cancers that increased glycosylation predominantly enhances the metastasis initiation capacity of cancer cells, with only a few exceptions (Fig. 3). In this respect, mainly the expression of glycosylating enzymes has been studied rather than the effect of metabolite substrate availability for driving this posttranslational modification. Yet, there is some evidence that increased production of the metabolites needed for glycosylation promotes metastasis initiation (51).

GalNAc (O-GalNAcylation)

N-acetylgalactosaminyltransferase (GALNT) transfers GalNAc to the serine and threonine residues of proteins resulting in O-linked protein glycosylation in the Golgi. Mostly invasiveness-promoting consequences of O-glycosylation have been recorded in cancer cells (Fig. 3A).

Several proteins, crucial for the initiation of metastasis by facilitating invasive properties and extracellular matrix (ECM) degradation in cancer cells, undergo O-linked glycosylation. Calnexin, integrin α5 and β1, EGFR, AXL receptor tyrosine kinase (AXL), and matrix metalloproteinase 14 (MMP14) were observed to be O-glycosylated in human and mouse liver tumors (calnexin) as well as human liver (calnexin, integrin β1, MMP14), oral (EGFR), colorectal cancer (AXL), skin (MMP14), and NSCLC (integrin α5) cancer cell lines supporting invasiveness (52–58). MMP14 and AXL/integrin α5 O-glycosylation were dependent on GALNT1 and GALNT2, respectively (53, 55, 58). Moreover, GALNT2 expression indicated poor prognosis in patients with colorectal cancer and GALNT14 promoted breast cancer–derived lung metastasis (53, 59), while conversely, GALNT8 inhibited the migratory capacity of breast cancer cells because the corresponding glycosylation impaired EGFR activity (60). Interestingly, shifting the localization of GALNT1 from the Golgi to the endoplasmic reticulum in a mouse liver tumor model resulted in MMP14 glycosylation and advanced tumor development as well as metastasis in multiple organs compared with no metastases in control tumors. Moreover, overexpression of Golgi-localized GALNT1 only moderately promoted metastases (58). This implies that the subcellular localization of O-glycosylating enzymes plays a significant role in determining the specificity of their protein targets.

CMP-Sialic Acid (Sialylation)

Sialylation is the attachment of CMP-sialic acid via β-Galacto­side sialyltransferases (STGAL) to N-glycans. In general, increa­sing sialylation either through increasing CMP-sialic acid availability and increased sialyltransferase or decreased sialidase activity promotes metastasis initiation in various cancer types (Fig. 3B).

In patients with breast cancer, it was observed that the gly­cosylation patterns of metastases were mostly organ dependent but that some N-glycans increased with metastatic progression (61). Moreover, ST3GAL6, ST3GAL1 gene, and ST3GAL1 protein expression were indicative of poor prognosis in urinary bladder cancer (62), melanoma progression (63), and decreased disease-free survival in clear cell renal cell carcinoma (ccRCC) (64), respectively. Consistent with this, altered expression of six sialylated N-glycans was found to be correlated with lymph node metastasis in patients with oral squamous cell carcinoma (OSCC) (65). Mechanistically, elevated expression of ST3GAL1 and ST3GAL3 induced the activation of the receptor tyrosine kinase AXL in melanoma cells (63) and integrin β1 and MMP2/9 in breast cancer cells (66), respectively, promoting invasion in vitro. Moreover, extracellular vesicles from B16 melanoma cells expressing high levels of ST3GAL5 induced premetastatic niche formation and subsequent peritoneal dissemination in mice (67). Conversely, ST6GAL1 was downregulated in HCC tissues and overexpression of ST6GAL1 inhibited HCC metastasis through impaired melanoma cell adhesion molecule (MCAM) expression at the cell surface (68).

CMP-sialic acid is the crucial substrate for sialylation, and it is produced by a side pathway of glycolysis with cytidine monophosphate N-acetylneuraminic acid synthetase (CMAS) being the last enzyme of the pathway. Cmas deletion reduced lung metastasis formation in a breast cancer mouse model (69). Moreover, diverging carbon flux toward CMP-sialic acid production increased metastasis initiation in different breast cancer mouse models and invasion in triple-negative breast cancer cells via integrin β3 sialylation, which was abrogated by Cmas silencing or blockage of integrin αvβ3 activity, respectively (51).

The erasers of sialylation are less studied than the transferases. So far only four sialidases (NEU) have been identified. Decreased NEU1 expression correlated with bladder cancer progression (70). NEU4 was found to be downregulated in HCC and inhibited cell motility at least in part through cleavage of sialic acid residuals from the cell-surface adhesion receptor CD44 (71). Moreover, NEU1 expression impaired a proinvasive fibronectin-integrin α5β1 interaction in bladder cancer cells (70). In contrast, NEU1 and NEU3 expression was increased in melanoma and bladder cancer tissues from patients, respectively (72, 73). Accordingly, NEU3 knockdown reduced bladder cancer cell invasion in vitro suggesting a protumoral function (73). Hence, sialidases exhibit both anti- and prometastatic effects, with potential specificity to certain cancer types.

UDP-GlcNAc (O-GlcNAcylation and N-linked Glycosylation)

UDP-GlcNAc is the substrate for the enzyme O-linked N-acetylglucosaminyltransferase (OGT) which catalyzes O-GlcNAcylation of proteins. Moreover, UDP-GlcNAc is required for N-linked glycosylation by N-acetylglucosaminyltransferases (MGAT). Current evidence suggests that O-GlcNAcylation in the tumor stroma and cancer cells promotes early metastatic steps by changing cell–cell and cell–matrix interactions (Fig. 3C). Moreover, based on the type of N-glycosylation, observed pro- or antimetastatic phenotypes are promoted in cancer cells (Fig. 3C).

O-GlcNAcylation

mRNA expression of OGT was positively correlated with metastasis status in patients with lung adenocarcinoma (74) and colon cancer (75), whereas increased O-GlcNAcylation levels were correlated with poor prognosis in patients with breast cancer (76). Moreover, the metabolic pathways leading to UDP-GlcNAc production increased during PDAC progression in the KPC mouse model (77). This effect was based on glutamine–fructose aminotransferase 1 (GFAT1) expression, which is the first step of UDP-GlcNAc production using glutamine and fructose-6-phosphate as substrates (77). Interestingly, 6-diazo-5-oxo-l-norleucine (DON), which inhibits all glutamine-catalyzing reactions, decreased UDP-GlcNAc synthesis and impaired PDAC xenograft growth and lymph node metastasis (78). Moreover, UDP-GlcNAc levels can also be affected by the hexosamine salvage pathway. This was shown to support primary tumor growth in PDAC (79); however, no link to metastasis has been established yet.

Mechanistically, it was found that increased O-GlcNAcyla­tion of cathepsin B, a lysosomal protease of the papain family from macrophages, promoted its secretion into the tumor microenvironment, leading to ECM degradation and increased melanoma-derived lung metastasis (80). In breast cancer, O-GlcNAcylation of the transcription factor Forkhead box protein A1 (FOXA1) was shown to shape its interactome, recruiting the transcriptional repressor methyl-CpG–binding protein 2 (MECP2) (76). This resulted in the suppression of adhesion-related genes, thereby promoting invasion in vitro (76). In lung adenocarcinoma patient samples, tumor-adjacent fibroblasts from the invasive edge displayed an altered glycosylation pattern compared with tumor core fibroblasts. Although the exact cross-talk is unclear, glycoproteomics revealed that glycosylation altered the cyclin-dependent kinase 4 (CDK4)—phosphorylated retinoblastoma protein (pRB) axis in the stroma, indirectly promoting an epithelial-to-mesenchymal transition (EMT) in lung cancer cells (81). Thus, there is evidence that O-GlcNAcylation is promoting metastasis formation.

N-linked Glycosylation

The expression of the N-glycosyltransferase MGAT1, 2, and 4A was found to be increased in glioblastoma versus adjacent brain tissue (82). Moreover, silencing of MGAT1 decreased the proliferation and migration of glioblastoma cells through decreased N-glycosylation and expression of GLUT1 (82). In line, mutations in MGAT5 impaired N-glycan branching and migration of glioblastoma stem-like cells (83). In contrast, it was observed that MGAT3 protein or mRNA was downregulated in metastatic ovarian and/or breast cancers compared with primary tumors and nonmetastatic breast cancer tissues (84, 85). Moreover, MGAT3-mediated N-glycosylation of CD82 blocked integrin α5β1-mediated cellular adhesion, migration, and metastasis of ovarian cancers (84). In line, overexpression of MGAT3 suppressed cervical cancer cell migration and invasion and induced a more epithelial phenotype (86). A similar block of an EMT by MGAT3 activity was observed in OSCC cell lines (87). Thus, MGAT3-mediated N-glycosylation has antimetastatic effects, whereas the expression of the other MGAT enzymes leads to prometastatic N-glycosylation patterns on proteins.

In summary, the majority of studies underscore the critical role of glycosylation in the early steps of metastasis and underscore the importance of glycosylation for the cross-talk of cancer cells with the ECM and stromal cells. At the same time, fewer investigations have demonstrated its significance in the later stage of colonization (88, 89). Although the later stages of metastasis are clinically easier to target, several inhibitors have been developed, for example, against ST3GAL3 and ST3GAL6 (90) Moreover, not only the lack of protein glycosylation but also the accumulation of the required metabolite substrates such as UDP-glucuronic acid may be a therapeutic vulnerability of cancers (91). In addition, in studies regarding congenital disorder of glycosylation, it has been shown that providing nutrients that feed into the substrate production for glycosylation alleviates some symptoms in patients (92). Thus, studies on the role of diet in cancer cell glycosylation may provide novel therapeutic strategies.

Protein Acetylation

Acetyl coenzyme A (acetyl-CoA) is involved in energy production and lipid synthesis, yet it can also serve as a substrate for the acetylation of histone and nonhistone proteins. There is ample evidence that acetylation promotes an EMT in cancer cells as reviewed by Kong and colleagues (93). Yet, also additional mechanisms exist in how acetylation, including histone acetylation and deacetylation, fosters metastasis formation within the same tumor type. Here, we summarize these additional mechanisms for the example of breast cancer (Fig. 4A).

Histone deacetylase (HDAC) increases the global acetylation of histones. The two FDA-approved pan-HDAC inhibitors SAHA and LBH589 were observed to promote breast cancer metastasis in a mouse model (94). In line, genetic and pharmacologic targeting of HDAC11 in mice increased breast cancer cell migration and distant metastasis although lymph node metastasis was reduced (95). Molecularly, HDAC4 inhibition enhanced H3K9 acetylation at the promoter of metastasis marker NEDD9 (neural precursor cell expressed developmentally downregulated 9), increasing its expression and consequently activating invasion via focal adhesion kinase (FAK) phosphorylation (94).

Also, non-histone proteins are frequently acetylated. Zinc finger MYND-type containing 8 (ZMYND8), which affects transcriptional regulation and chromatin remodeling, and the superoxide dismutase SOD2 were observed to be acetylated in breast cancer cells promoting migration and invasion as well as stemness, respectively (96, 97). Mutation of the p300 (histone acetyltransferase P300) acetylation site in ZMYND8 was sufficient to impair primary tumor growth and metastasis (97). Moreover, acetylation of the signal transduction protein SMAD family member 3 (SMAD3) by the acetyltransferase KAT6A promoted breast cancer cell stemness and metastasis formation (98, 99). In patients with breast cancer, KAT2A expression was elevated in metastatic lesions growing in palmitate-enriched organs, whose oxidation provides the acetylation substrate acetyl-CoA, compared with organs with low palmitate content (99). Mechanistically, palmitate oxidation to acetyl-CoA requiring carnitine palmitoyltransferase 1A (Cpt1a) activity promoted the acetylation of the NFκB protein p65 via KAT2A (99). Targeting CPT1A abrogated high-fat diet-induced lung metastasis growth in breast cancer mouse models (99). This is consistent with the observation that inhibiting palmitate uptake using a blocking antibody against CD36 impaired diet-induced metastasis in various mouse models (100, 101).

Notably, not only acetylation changes in cancer cells promote metastasis. In line, it was found that senescent lung fibroblasts that recruit immune cells during premetastatic niche formation show low expression of the enzyme acetyl-CoA carboxylase α (ACACA), which consumes acetyl-CoA and that overexpression of this enzyme reduced metastasis formation (102).

Although numerous studies underscore the importance of acetylation in promoting metastasis, the deacetylation of certain proteins can also be prometastatic in breast cancer. Acetylation of the methyltransferase METTL3 via acetyl-CoA acetyltransferase 1 (ACAT1) expression or downregulation of sirtuin (SIRT) 1 activity, impaired migration and invasion of breast cancer cells (103, 104). Yet, in tissue samples from patients with breast cancer, it was found that the deacetylases SIRT1 and SIRT3 decreased in invasive tumors and lymph node metastasis (compared with primary tumors), respectively (96, 105). Thus, these observations require further investigation to link the cell line data to clinical observations.

Currently, four histone deacetylase inhibitors are clinically approved, and others are passing through clinical trials (106). Yet, inhibitors blocking protein acetylation are less advanced with a drug targeting CREB-binding protein (CBP) and p300, which are highly homologous acetyltransferases, currently in clinical phase I in patients with metastatic prostate cancer (107). Despite these promising advancements in translation, data from preclinical models raise concerns regarding potential adverse effects, given the dual role of acetylation and deacetylation in cancer metastasis. However, a generally positive effect may be observed through dietary interventions or targeting the uptake of the acetyl-CoA precursor palmitate. Accordingly, clinical trials blocking CD36 are about to start (108). Notably, targeting palmitate uptake will have various effects beyond blocking acetylation as reviewed by (109).

Palmitoylation

Lipids play a crucial role as signaling molecules by posttranslationally modifying proteins and influencing their functions. One common protein modification is palmitoylation, which involves the covalent addition of a palmitate moiety. In humans 23 and in mice 24 palmitoyl acyl transferases (DHHCs) have been annotated, whereas 7 depalmitoylating enzymes have been identified. Strikingly, the gene-expression patterns of these enzymes are cancer-type dependent when analyzed in tumor samples from patients and may correlate with both overall and progression-free survival (110). The resulting changes in palmitoylation can influence the localization, interaction, and activity of the substrate proteins with implications in metastasis formation (Fig. 4B).

Glioblastomas are highly invasive cancers. DHHC5 and 9 expression has been associated with an EMT and colony-forming capacity in glioblastoma (111, 112). Mechanistically, DHHC5 promoted FAK palmitoylation sustaining its membrane localization needed for interaction with the ECM (111), whereas DHHC9 palmitoylated the glucose transporter GLUT1 which subsequently promoted glycolysis (112). In HCC, DHHC7 was important for STAT3 palmitoylation, and the DHHC7 inhibitor S-(2-acetamidoethyl) 2-bromohexadecanethioate (MY-D-4; ref. 113) impaired colony growth in HCC cell lines (114). In contrast, DHHC7-mediated palmitoylation of junctional adhesion molecule 3 (JAM3) increased tight junctions and impaired lung cancer cell migration (115). Cervical cancer cell migration, invasion, and EMT were dependent on a dynamic palmitoylation and depalmitoylation of flotillin 1 (FLOT1) by DHHC19 and LYPLA1, which resulted in sustained insulin-like growth factor-1 receptor (IGF-1R) tyrosine kinase activation (116, 117).

Furthermore, it was observed that palmitate levels regulate protein palmitoylation in colorectal cancer and prostate cancer cells (118, 119). Acyl-CoA oxidase 1 (ACOX1), the first enzyme of the peroxisomal fatty acid oxidation pathway, was found to be progressively downregulated in tissue samples from patients with colorectal cancer comparing normal tissue, primary tumors, and metastases (118). In cell lines, the loss of ACOX1 resulted in elevated palmitate levels which fostered β-catenin stability because the associated palmitoylation prevented degradation via ubiquitination (118). Similarly, the loss of fatty acid synthase (FASN) decreased Ras homolog family member U (RhoU) palmitoylation and cell migration in prostate cancer cells (119). Moreover, it was shown that palmitoylation of CD36 by DHHC4/5 led to monounsaturated fatty acid uptake balancing lipotoxicity in high-fat diet-induced breast cancer–derived lung metastasis (120, 121).

Palmitoylation is a widespread posttranslational modification that is only starting to be explored in cancer and metastasis formation. Besides the general inhibitor 2-bromopalmitate (2-BP), several more specific inhibitors have been developed for use in cell lines including inhibitors against DHHC3, 7, and 20 (113). Moreover, molecular docking studies predict favorable binding activity of the polo-like kinase 1 inhibitor BI-2536 to all DHHC enzymes except DHHC3, which could be further explored to develop selective inhibitors against the DHHC family members (110).

Protein Lactylation

Lactylation is a recently discovered protein modification of lysine residues that is regulated by the lactate concentration in cells (122) and has implications for the early steps of metastasis formation (Fig. 4C).

A global lactylome analysis in HCC cells identified almost 10,000 lactylated proteins including many enzymes such as adenylate kinase 2 (AK2), which was inhibited by lactylation (123). In line, the lactylation of AK2, but not AK2 levels itself, was identified as an unfavorable prognostic marker in HCC patients (123). In cervical cancer cell lines, it was shown that lactylation increased the stability of discoidin, CUB, and LCCL domain containing 1 (DCBLD1), which in turn activated the pentose phosphate pathway resulting in elevated migration, invasion, and growth (124). In addition, nucleolar and spindle-associated protein 1 (NUSAP1) was observed to be lactylated promoting its protein stability, and elevated NUSAP1 levels were associated with a worse prognosis in PDAC patients (125). In line, NUSAP1 deletion decreased migration and metastasis of PDAC cells (125). Thus, lactylation emerges as an additional regulator of protein abundance in cancer cells. Moreover, the direct regulation of lactylation by lactate concentrations raises the question of whether the metastatic potential of tumor cells gained through the expression of the lactate transporter MCT1 (126) is also in part mediated through lactylation.

Protein Methylation

S-Adenosyl methionine (SAM) is the substrate for methylation reactions, and it is produced in the methionine cycle that in turn intersects with the folate cycle and NAD+ metabolism. Thereby, methionine and serine can both increase SAM availability, whereas nicotinamide N-methyltransferase (NNMT), which converts SAM and NAM (nicotinamide) to SAH (S-adenosyl-L-homocysteine) and 1NMA (N1-methylnicotinamide) decreases it. Whether elevated SAM levels and methylation promote or inhibit metastasis formation may be dependent on the tumor context, the biochemical alteration, and the specific methylation mark that is affected (Fig. 4D and E).

Diet and Methionine Cycle–Mediated Increase in SAM

Tumoral and blood SAM levels were found to be correlated with metastatic reoccurrence in patients with colorectal cancer (127). It was also shown that efficiently metastasizing melanoma PDX models show elevated levels of methylation-related metabolites and H3K9 and H3K27 trimethylation levels (128), indicating a role of increased methylation in metastasis formation. Moreover, methionine restriction suppressed the invasiveness of triple-negative breast cancer and gastric cancer cells, which led to impaired lung metastasis in orthotopic triple-negative breast cancer (129) and a gastric cancer metastasis mouse model (130), respectively. This is in line with patient data showing that the levels of the methionine adenosyltransferase 2A (MAT2A), which catalyzes the synthesis of SAM from methionine and ATP, correlated with higher-grade tumors and metastases in patients with lung cancer (131). Moreover, palmitate and a palm-oil-rich diet increased—through an unknown mechanism—methyltransferase Set1A–mediated H3K4 trimethylation and metastasis initiation in oral cancers and melanoma (100). Thus, some diets may have metastasis-promoting effects by changing protein methylation.

NNMT-Mediated Decrease in SAM

Most of the evidence that methylation and elevated SAM levels are antimetastatic stems from the study of NNMT, whose expression decreases SAM levels. A meta-analysis including over 2,500 patients with cancer found that high NNMT expression was correlated with worse tumor differentiation, earlier lymph node, and distant metastasis (132). Accordingly, NNMT expression was observed to be high in cancer stem cells of different tumor types and indicated poor outcomes in patients with breast cancer and patients with triple-negative breast cancer undergoing chemotherapy (133–135). NNMT loss inhibited metastasis formation in breast cancer mouse models (134, 135). Mechanistically, the loss of NNMT promoted histone H3K9 trimethylation (H3K9me3) and DNA methylation at multiple promoters leading to decreased expression of collagen and ECM genes in breast cancer cells (135). Moreover, the methylation of protein phosphatase 2 (PP2A), which enhances activity, increased upon NNMT silencing in triple-negative breast cancer cells leading to decreased phosphorylation and activity of the MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase) pathway blocking a downstream EMT (134). Metastases in intrahepatic cholangiocarcinoma mouse models were promoted by NNMT activity because of decreased H3K27 and H3K9 trimethylation leading to elevated EGFR expression (136). Thus, NNMT is emerging as an interesting drug target, and several inhibitors for the use in cell lines have been developed (137–140) although currently no clinical trials have been conducted yet.

Many enzymes have besides their canonical catalytic functions in metabolism also moonlighting functions of catalytic or noncatalytic nature. These moonlighting functions have been extensively studied in primary cancers as reviewed by (141), yet their role in metastasis formation is only emerging with most of them suggesting roles in the early steps of the metastatic cascade (Fig. 5).

Figure 5.

Moonlighting functions of metabolic enzymes. A, The noncanonical catalytic functions of glycolysis-related enzymes drive the early steps of metastasis formation by acting as protein kinases in the nucleus. B, Noncatalytic moonlighting functions of metabolic enzymes regulate metastasis formation by modulating chromatin organization, transcription, translation, posttranslational modifications, and cellular surface architecture. Affected early steps of the metastatic cascade are depicted in green, whereas late steps are depicted in orange. Transcription factors are depicted in dark blue and metabolic enzymes in red, whereas all other proteins are depicted in light blue ellipses. AIMP2, aminoacyl tRNA synthetase complex interacting multifunctional protein 2; ALDOA, aldolase A; ATCG1, actin gamma 1; ATF4, activating transcription factor 4; BMP, bone morphogenetic protein; BMPR, BMP receptor; CMP-sialic acid, cytidine monophosphate-sialic acid; CTR9, Cln three recruiting 9; EC, endothelial cell; eIF4G, eukaryotic initiation factor 4G; EMT, epithelial-to-mesenchymal transition; ENO1, enolase 1; F6P, fructose-6-phosphate; FAK, focal adhesion kinase; Fru, fructose; G6P, glucose-6-phosphate; Glc, glucose; GSK-3β, glycogen synthase kinase-3 beta; HGF, hepatocyte growth factor; HIF-1A, hypoxia-inducible factor-1 alpha; HK2, hexokinase 2; IGF2BP1, insulin-like growth factor 2 mRNA-binding protein 1; IQGAP1, IQ motif containing GTPase activating protein 1; IWS1, interact with SUPT6H; KHK-A, ketohexokinase A; MAX, MYC associated factor 1; NADK, nicotinamide adenine dinucleotide kinase; P, phosphate group; PFK, phosphofructokinase 1; PFKFB4, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4; PGAM1, phosphoglycerate mutase 1; PHGDH, phosphoglycerate dehydrogenase; PKA, proteinkinase A; PKM2, pyruvate kinase M2; PSAT1, phosphoserine aminotransferase 1; R1A, PKA regulatory subunit 1; SIRT1, sirtuin-1; SMAD, suppressor of mothers against decapentaplegic; SMURF1, SMAD-specific E3 ubiquitin protein ligase 1; SPIN1, spindlin 1; SRC, nuclear receptor coactivator; STAT3, signal transducer and activator of transcription 3; TAM, tumor-associated macrophage; Ub, ubiquitin; YWHAH, tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein eta. Created with BioRender.com.

Figure 5.

Moonlighting functions of metabolic enzymes. A, The noncanonical catalytic functions of glycolysis-related enzymes drive the early steps of metastasis formation by acting as protein kinases in the nucleus. B, Noncatalytic moonlighting functions of metabolic enzymes regulate metastasis formation by modulating chromatin organization, transcription, translation, posttranslational modifications, and cellular surface architecture. Affected early steps of the metastatic cascade are depicted in green, whereas late steps are depicted in orange. Transcription factors are depicted in dark blue and metabolic enzymes in red, whereas all other proteins are depicted in light blue ellipses. AIMP2, aminoacyl tRNA synthetase complex interacting multifunctional protein 2; ALDOA, aldolase A; ATCG1, actin gamma 1; ATF4, activating transcription factor 4; BMP, bone morphogenetic protein; BMPR, BMP receptor; CMP-sialic acid, cytidine monophosphate-sialic acid; CTR9, Cln three recruiting 9; EC, endothelial cell; eIF4G, eukaryotic initiation factor 4G; EMT, epithelial-to-mesenchymal transition; ENO1, enolase 1; F6P, fructose-6-phosphate; FAK, focal adhesion kinase; Fru, fructose; G6P, glucose-6-phosphate; Glc, glucose; GSK-3β, glycogen synthase kinase-3 beta; HGF, hepatocyte growth factor; HIF-1A, hypoxia-inducible factor-1 alpha; HK2, hexokinase 2; IGF2BP1, insulin-like growth factor 2 mRNA-binding protein 1; IQGAP1, IQ motif containing GTPase activating protein 1; IWS1, interact with SUPT6H; KHK-A, ketohexokinase A; MAX, MYC associated factor 1; NADK, nicotinamide adenine dinucleotide kinase; P, phosphate group; PFK, phosphofructokinase 1; PFKFB4, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4; PGAM1, phosphoglycerate mutase 1; PHGDH, phosphoglycerate dehydrogenase; PKA, proteinkinase A; PKM2, pyruvate kinase M2; PSAT1, phosphoserine aminotransferase 1; R1A, PKA regulatory subunit 1; SIRT1, sirtuin-1; SMAD, suppressor of mothers against decapentaplegic; SMURF1, SMAD-specific E3 ubiquitin protein ligase 1; SPIN1, spindlin 1; SRC, nuclear receptor coactivator; STAT3, signal transducer and activator of transcription 3; TAM, tumor-associated macrophage; Ub, ubiquitin; YWHAH, tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein eta. Created with BioRender.com.

Close modal

Noncanonical Catalytic Functions of Glycolysis-Related Enzymes

Metastasis-related noncanonical functions of glycolysis-related enzymes have been mainly linked to protein phosphorylation in the nucleus resulting in the activation of signaling pathways leading to cancer cell migration and invasion (Fig. 5A).

Fructose is besides glucose a major carbohydrate in the Western diet. Dietary and polyol pathway (glucose to fructose conversion)–derived fructose is converted by ketohexokinase (KHK) to fructose-1-phosphate. In breast and gastric cancer cells, it was found that nuclear KHK-A suppressed the mRNA expression of the repressive EMT marker CDH1 (E-cadherin) by phosphorylating YWHAH (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein eta), which recruits snail family transcriptional repressor 2 (SLUG) to the promotor of CDH1 (142, 143). In mice, Khk-A promoted fructose-induced breast cancer metastasis to the lung and a phospho-deficient mutant of the Khk-A site in Ywhah (S25A) abolished the metastasis-promoting effect of fructose (142).

6-Phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (Pfkfb4) interconverts fructose-6-phosphate and fructose-2,6-phosphate, which is an allosteric activator of the glycolysis enzyme phosphofructokinase (PFK). Two studies in breast cancer showed that PFKFB4 also acted in the nucleus by phosphorylating nuclear receptor coactivator 3 (Src-3) which activated collagen remodeling and purine biosynthesis (144, 145). In lung adenocarcinoma cells, PFKFB4 could phosphorylate SRC-2 (146). In breast cancer mouse models, Pfkfb4 silencing decreased systemic metastasis but, dependent on the mouse model, not primary tumor growth. A phospho-deficient mutant of the PFKFB4 site in SRC-3 (S857A) did not induce lung metastasis (144). In addition, PFKFB4 expression was higher in lung, liver, and brain metastases compared with primary tumors of triple-negative breast cancer patients (145), and PFKFB4 expression and a PFKFB4-SRC-3 proteomics signature correlated with poor prognosis in basal and triple-negative breast cancer whereby patients with intense nuclear staining showed a worse prognosis (144, 145).

Pyruvate kinase M2 (PKM2) converts phosphoenolpyruvate (PEP) to pyruvate. The functional role of PKM2 in phosphorylating proteins is debated (147). Yet, two studies suggest that PKM2 has a nuclear function in HCC and colorectal cancer metastasis (148, 149). Thereby, a PKM2 mutant that cannot enter the nucleus (R399/400A) but can still form a tetramer does not induce metastasis (149). Molecularly, it was suggested that PKM2 phosphorylates STAT3 inducing EMT needed for migration and invasion in HCC cells (148).

Noncatalytic Functions

Metastasis-initiating capacity in cancer cells can be promoted by protein–protein interactions of enzymes. In most described cases, an increase in this interaction promoted metastasis but also the opposite, namely, the loss of an interaction, has been linked to metastasis formation (Fig. 5B).

Glycolysis

Several glycolytic enzymes have noncatalytic functions in early metastasis formation. Hexokinase 2 (HK2), the enzyme converting glucose to glucose-6-phosphate (G6P), was recently shown to have a noncatalytic function in promoting breast cancer metastasis in two mouse models (150). Mechanistically, it was observed that HK2 interacts with glycogen synthase kinase 3 (GSK3) and the regulatory subunit of protein kinase A (R1A), which enabled PKA to phosphorylate GSK3. This led to the degradation of GSK3 and subsequent snail family transcriptional repressor 1 (SNAIL) expression promoting an EMT in cancer cells (150). Further, it was shown that HK2 interacts with proteins of chromatin organization in the nucleus increasing the transcription of stemness genes and DNA repair in AML cells (151). Meanwhile, HK2 interaction with AIMP2 (proapoptotic protein aminoacyl tRNA synthetase complex interacting multifunctional protein 2) enhanced its degradation in HCC cells (152). These additional noncatalytic functions of HK2 may potentially foster metastasis-initiating capacities in cancer cells by increasing stemness and decreasing cell death. In line, it was observed that elevated HK2 expression was associated with decreased reoccurrence-free survival in HCC patients (152).

Fructose-1,6-bisphosphate (FBP) is converted to dihydroxyacetone phosphate (DHAP) and glyceraldehyde-3-phosphate (G3P) by aldolase (ALDO), whereas triosephosphate isomerase 1 (TPI1) interconverts DHAP and G3P. In HCC cells, it was seen that ALDOA interacted with insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) enhancing eukaryotic translation initiation factor 4G (eIF4G) translation and subsequently global mRNA translation (153). A catalytically inactive mutant of ALDO and TPI1 induced, like the wildtypes, colony formation and migration in HCC and lung cancer cells, respectively (153, 154). ALDOA and TPI1 expression were associated with poor outcomes in HCC and lung adenocarcinoma patients, respectively, suggesting a role of the noncatalytic functions of these enzymes in metastasis initiation (153, 154).

3-Phosphoglycerate (3PG) is converted to 2-phosphoglycerate (2PG) which then is converted to PEP by phosphoglycerate mutase 1 (PGAM1) and enolase (ENO), respectively. It was found that PGAM1 in exosomes from prostate cancer cells was taken up by endothelial cells where it induced angiogenesis, migration, and proliferation through an interaction with actin gamma 1 (ACTG1; ref. 155). Catalytically inactive ENO1 promoted, like wild-type, lung cancer cell invasion presumably through an interaction of ENO1 with hepatocyte growth factor receptor (HGFR) which can promote an EMT via decreased GSK3 activity (156). Moreover, treatment of mice with PGAM1-containing exosomes promoted prostate cancer cell–derived metastasis formation compared with treatment with PGAM1-deficient exosomes. Moreover, patients with metastatic prostate cancer displayed more PGAM1 expression and endothelial cells compared with nonmetastatic patients (155).

Finally, it was found that in lung cancer cells, autocrine secreted, and in breast cancer cells, macrophage secreted, PKM2 binds to integrin β1, consequently triggering an EMT through SRC signaling (157, 158). In line, breast and lung cancer cells treated with recombinant PKM2 and challenged with a wound closure assay or intravenously injected into mice resulted in more migratory cells and enhanced lung and liver metastasis (157, 158).

Serine Pathway

The serine biosynthesis pathway branches off glycolysis converting 3PG to serine in a three-step enzymatic reaction by PHGDH, phosphoserine aminotransferase 1 (PSAT1), and phosphoserine phosphatase (PSPH). The acquisition of metastatic initiation capability in a triple-negative breast cancer mouse model and lung cancer cells was driven by the loss of a noncatalytic function of PHGDH (51) or an increase in the noncatalytic function of PSAT1 (159), respectively. These alterations induced migration and invasion, underlining their pivotal roles in promoting metastasis. Mechanistically, the loss of PHGDH disrupted a protein–protein interaction with the glycolytic enzyme Pfk, which decreased its activity redirecting carbon flux to CMP-sialic acid driving sialylation of integrin αvβ3 in triple-negative breast cancer cells (51). In line, overexpression of a catalytically inactive mutant of PHGDH decreased metastasis to the same extent as the wild-type in Phgdh knockdown cells. In lung cancer cells PSAT1 was found to interact with IQ motif containing GTPase activating protein 1 (IQGAP1), which recruited STAT3 and increased its phosphorylation and function in inducing MMP9, C–X–C motif chemokine ligand 8 (CXCL8), and serum amyloid A1 (SAA1) expression (159). Interestingly, heterogeneous and/or low expression of PHGDH correlated with decreased metastasis-free survival in patients with triple-negative breast cancer indicating a role not only for the level of expression but also for heterogeneous expression within a tumor (51).

NADK

Nicotinamide adenine dinucleotide kinase (NADK) phosphorylates NAD+ to NADP+, and its catalytic function has been shown to promote metastasis formation in breast cancer (160). Also, a noncatalytic function of NADK in lymph node metastasis was observed in an NSCLC mouse model (161). Thereby, NADK interacted with the ubiquitination regulating SMAD-specific E3 ubiquitin protein ligase 1 (SMURF1). Subsequently, the ubiquitination and degradation of bone morphogenetic protein receptor type 1A (BMPR1A) by SMURF1 was impaired leading to BMP signaling-dependent migration and invasion. In patients with NSCLC, NADK expression correlated with lymph node metastasis and poor prognosis (161).

Moonlighting functions of enzymes are more and more frequently detected. However, their targeting may be more difficult especially when catalytic and noncatalytic functions or metabolic and nuclear activities have opposite effects on metastasizing cancer cells. For example, targeting only the catalytic function but not protein stability would be important in the case of leveraging PHGDH as a drug target against metastasis (51, 162).

Metastasis formation is the leading cause of death in patients with cancer and innovative approaches to prevent and target metastases are urgently needed. Functions of metabolites and enzymes outside their canonical role in the metabolic network are emerging as vulnerabilities of metastasizing cancer cells. In this respect, most of our knowledge is currently limited to the early metastatic steps where cancer cells gain metastasis-initiating capacities through increased motility and ECM remodeling capabilities leading to invasiveness. Much less is currently known about metabolic signaling in metastases outgrowth and persistence in distant organs.

Whether and how intercepting the early steps of metastases is beneficial in clinical practice depends on the tumor type (Fig. 6). In glioblastoma, a neoadjuvant treatment reducing the invasiveness of the primary tumor may lead to patient benefit because clear resection margins can be easier achieved, and less tissue will need to be resected from the brain to remove the tumor potentially sparing cognitive functions. In breast cancer, metabolic gene and protein signatures indicating metastasis-initiating capacity (51) and thus potential early dissemination of cancer cells from the primary tumor could be further developed to improve the prediction of patients with high metastasis risk. This may benefit the patients by receiving follow-up treatments and checkups according to their predicted metastasis risk. Moreover, also in pancreatic cancer, it was shown that neoadjuvant treatment before surgery extends median survival by about 1.8-fold (163). Recently, two studies in preclinical mouse models of PDAC were published showing that targeting multiple enzymes that use glutamine as a substrate, rather than only targeting glutaminase, decreased primary tumor growth, metastasis incidence, and metastasis size (164, 165). This increased efficacy may be due to the inhibition of noncanonical functions of glutamine metabolism because an earlier study suggested that the beneficial effect of using an inhibitor that targets multiple glutamine-using enzymes in PDAC mouse models is mainly driven by its inhibitory effect on glycosylation (77).

Figure 6.

Treatment of classic and nonclassical functions of metabolism may be explored to target metastases. Exploiting metabolic gene or protein signatures associated with metastasis-initiating capacity of cancer cells in primary tumors (PT) may be used to predict the metastasis risk in patients. Treatment with therapies regressing invasiveness may increase patient survival and life quality by improving resection precision through clearer margins. Currently, treatments targeting already disseminated cancer cells in distant organs would affect patient survival the most. Created with BioRender.com.

Figure 6.

Treatment of classic and nonclassical functions of metabolism may be explored to target metastases. Exploiting metabolic gene or protein signatures associated with metastasis-initiating capacity of cancer cells in primary tumors (PT) may be used to predict the metastasis risk in patients. Treatment with therapies regressing invasiveness may increase patient survival and life quality by improving resection precision through clearer margins. Currently, treatments targeting already disseminated cancer cells in distant organs would affect patient survival the most. Created with BioRender.com.

Close modal

Currently, most patients will benefit from treatments that can target already disseminated cancer cells growing in distant organs (Fig. 6). An arising opportunity may be to target the metabolic signaling that is induced in metastases growing in palmitate-rich organs (99). Yet, more knowledge is needed on the canonical and particular noncanonical functions of metabolites and metabolic enzymes in distant metastases to find innovative treatment strategies. Moreover, a higher acceptance of therapeutic strategies only targeting metastases and their formation is needed at all levels of translation ranging from drug-developing companies to regulatory bodies to enable nonclassical approaches to tackle metastases. A first step in this direction was the 2018 FDA acceptance of two clini­cal trials with metastasis-free survival as the primary clinical endpoint (166).

S.-M. Fendt reports grants from Gilead, other support from Auron Tx, Blackbelt Tx, and Alesta Tx outside the submitted work.

We apologize to all colleagues whose excellent work we could not cite due to text and reference limits. S. Krieg is supported by an EMBO Postdoctoral Fellowship (ALTF 164-2023). S.I. Fernandes acknowledges funding from Stichting tegen Kanker. C. Kolliopoulos acknowledges funding from the Swedish Research Council (VR, Vetenskapsrådet). M. Liu acknowledges funding from the China Scholarship Council (CSC). S.-M. Fendt acknowledges funding from FWO and iBOF (INTERCEPt) Projects, the Beug Foundation, Fonds Baillet Latour, KU Leuven, Francqui Stichting, Foundation ARC, Wereld Kanker Onderzoek Fonds (WKOF) as part of the World Cancer Research Fund International grant programs and Stichting tegen Kanker.

1.
Demicco
M
,
Liu
XZ
,
Leithner
K
,
Fendt
SM
.
Metabolic heterogeneity in cancer
.
Nat Metab
2024
;
6
:
18
38
.
2.
Bergers
G
,
Fendt
SM
.
The metabolism of cancer cells during metastasis
.
Nat Rev Cancer
2021
;
21
:
162
80
.
3.
Warburg
O
.
über den Stoffwechsel der Carcinomzelle
.
Klin Wochenschr
1925
;
4
:
534
6
.
4.
Wang
Q
,
Tiffen
J
,
Bailey
CG
,
Lehman
ML
,
Ritchie
W
,
Fazli
L
, et al
.
Targeting amino acid transport in metastatic castration-resistant prostate cancer: effects on cell cycle, cell growth, and tumor development
.
J Natl Cancer Inst
2013
;
105
:
1463
73
.
5.
Najumudeen
AK
,
Ceteci
F
,
Fey
SK
,
Hamm
G
,
Steven
RT
,
Hall
H
, et al
.
The amino acid transporter SLC7A5 is required for efficient growth of KRAS-mutant colorectal cancer
.
Nat Genet
2021
;
53
:
16
26
.
6.
Shapiro
JS
,
Chang
H-C
,
Tatekoshi
Y
,
Zhao
Z
,
Waxali
ZS
,
Hong
BJ
, et al
.
Iron drives anabolic metabolism through active histone demethylation and mTORC1
.
Nat Cell Biol
2023
;
25
:
1478
94
.
7.
Chua
AC
,
Knuiman
MW
,
Trinder
D
,
Divitini
ML
,
Olynyk
JK
.
Higher concentrations of serum iron and transferrin saturation but not serum ferritin are associated with cancer outcomes
.
Am J Clin Nutr
2016
;
104
:
736
42
.
8.
Schniers
BK
,
Wachtel
MS
,
Sharma
M
,
Korac
K
,
Rajasekaran
D
,
Yang
S
, et al
.
Deletion of Slc6a14 reduces cancer growth and metastatic spread and improves survival in KPC mouse model of spontaneous pancreatic cancer
.
Biochem J
2022
;
479
:
719
30
.
9.
Akcakanat
A
,
Sahin
A
,
Shaye
AN
,
Velasco
MA
,
Meric
-
Bernstam
F
. Comparison of Akt/mTOR signaling in primary breast tumors and matched distant metastases
.
Cancer
2008
;
112
:
2352
8
.
10.
Rausch
S
,
Schollenberger
D
,
Hennenlotter
J
,
Stühler
V
,
Kruck
S
,
Stenzl
A
, et al
.
mTOR and mTOR phosphorylation status in primary and metastatic renal cell carcinoma tissue: differential expression and clinical relevance
.
J Cancer Res Clin Oncol
2019
;
145
:
153
63
.
11.
Tamburrino
A
,
Molinolo
AA
,
Salerno
P
,
Chernock
RD
,
Raffeld
M
,
Xi
L
, et al
.
Activation of the mTOR pathway in primary medullary thyroid carcinoma and lymph node metastases
.
Clin Cancer Res
2012
;
18
:
3532
40
.
12.
Rinaldi
G
,
Pranzini
E
,
Van Elsen
J
,
Broekaert
D
,
Funk
CM
,
Planque
M
, et al
.
In vivo evidence for serine biosynthesis-defined sensitivity of lung metastasis, but not of primary breast tumors, to mTORC1 inhibition
.
Mol Cell
2021
;
81
:
386
97
.
13.
Durán
RV
,
Oppliger
W
,
Robitaille
AM
,
Heiserich
L
,
Skendaj
R
,
Gottlieb
E
, et al
.
Glutaminolysis activates Rag-mTORC1 signaling
.
Mol Cell
2012
;
47
:
349
58
.
14.
Yi
Y
,
Chen
D
,
Ao
J
,
Zhang
W
,
Yi
J
,
Ren
X
, et al
.
Transcriptional suppression of AMPKα1 promotes breast cancer metastasis upon oncogene activation
.
Proc Natl Acad Sci U S A
2020
;
117
:
8013
21
.
15.
Yuan
P
,
Teng
D
,
de Groot
E
,
Li
M
,
Trousil
S
,
Shen
C-H
, et al
.
Loss of AMPKα2 promotes melanoma tumor growth and brain metastasis
.
iScience
2023
;
26
:
106791
.
16.
Wang
M-D
,
Wang
N-Y
,
Zhang
H-L
,
Sun
L-Y
,
Xu
Q-R
,
Liang
L
, et al
.
Fatty acid transport protein-5 (FATP5) deficiency enhances hepatocellular carcinoma progression and metastasis by reprogramming cellular energy metabolism and regulating the AMPK-mTOR signaling pathway
.
Oncogenesis
2021
;
10
:
74
.
17.
Ramchandani
D
,
Berisa
M
,
Tavarez
DA
,
Li
Z
,
Miele
M
,
Bai
Y
, et al
.
Copper depletion modulates mitochondrial oxidative phosphorylation to impair triple-negative breast cancer metastasis
.
Nat Commun
2021
;
12
:
7311
.
18.
Nakashima
C
,
Shingo
K
,
Fujiwara-Tani
R
,
Luo
Y
,
Kawahara
I
,
Goto
K
, et al
.
Expression of long-chain fatty acid receptor GPR40 is associated with cancer progression in colorectal cancer: a retrospective study
.
Oncol Lett
2018
;
15
:
8641
6
.
19.
Kleemann
J
,
Hrgovic
I
,
Kleimann
P
,
Ter-Nedden
J
,
Glaser
M
,
Steinhorst
K
, et al
.
G protein-coupled receptor 40 expression in human melanoma: correlation with tumour thickness, AJCC stage and survival
.
J Eur Acad Dermatol Venereol
2020
;
34
:
285
92
.
20.
Sun
X
,
Chu
H
,
Lei
K
,
Ci
Y
,
Lu
H
,
Wang
J
, et al
.
GPR120 promotes metastasis but inhibits tumor growth in pancreatic ductal adenocarcinoma
.
Pancreatology
2022
;
22
:
749
59
.
21.
Pickens
CA
,
Lane-Elliot
A
,
Comstock
SS
,
Fenton
JI
.
Altered saturated and monounsaturated plasma phospholipid fatty acid profiles in adult males with colon adenomas
.
Cancer Epidemiol Biomarkers Prev
2016
;
25
:
498
506
.
22.
Fujii
K
,
Luo
Y
,
Fujiwara-Tani
R
,
Kishi
S
,
He
S
,
Yang
S
, et al
.
Pro-metastatic intracellular signaling of the elaidic trans fatty acid
.
Int J Oncol
2017
;
50
:
85
92
.
23.
Karmokar
PF
,
Moniri
NH
.
Free-fatty acid receptor-4 (FFA4/GPR120) differentially regulates migration, invasion, proliferation and tumor growth of papillary renal cell carcinoma cells
.
Biochem Pharmacol
2023
;
213
:
115590
.
24.
Wu
J-Y
,
Huang
T-W
,
Hsieh
Y-T
,
Wang
Y-F
,
Yen
C-C
,
Lee
G-L
, et al
.
Cancer-derived succinate promotes macrophage polarization and cancer metastasis via succinate receptor
.
Mol Cell
2020
;
77
:
213
27
.
25.
Reddy
A
,
Winther
S
,
Tran
N
,
Xiao
H
,
Jakob
J
,
Garrity
R
, et al
.
Monocarboxylate transporters facilitate succinate uptake into brown adipocytes
.
Nat Metab
2024
. doi: .
26.
Terra
X
,
Ceperuelo-Mallafré
V
,
Merma
C
,
Benaiges
E
,
Bosch
R
,
Castillo
P
, et al
.
Succinate pathway in head and neck squamous cell carcinoma: potential as a diagnostic and prognostic marker
.
Cancers
2021
;
13
:
1653
.
27.
Trauelsen
M
,
Hiron
TK
,
Lin
D
,
Petersen
JE
,
Breton
B
,
Husted
AS
, et al
.
Extracellular succinate hyperpolarizes M2 macrophages through SUCNR1/GPR91-mediated Gq signaling
.
Cell Rep
2021
;
35
:
109246
.
28.
Elia
I
,
Rowe
JH
,
Johnson
S
,
Joshi
S
,
Notarangelo
G
,
Kurmi
K
, et al
.
Tumor cells dictate anti-tumor immune responses by altering pyruvate utilization and succinate signaling in CD8+ T cells
.
Cell Metab
2022
;
34
:
1137
50
.
29.
Mu
X
,
Zhao
T
,
Xu
C
,
Shi
W
,
Geng
B
,
Shen
J
, et al
.
Oncometabolite succinate promotes angiogenesis by upregulating VEGF expression through GPR91-mediated STAT3 and ERK activation
.
Oncotarget
2017
;
8
:
13174
85
.
30.
Chen
J
,
Li
Z
,
Yue
C
,
Ma
J
,
Cao
L
,
Lin
J
, et al
.
Human umbilical cord mesenchymal stem cell-derived exosomes carrying miR-1827 downregulate SUCNR1 to inhibit macrophage M2 polarization and prevent colorectal liver metastasis
.
Apoptosis
2023
;
28
:
549
65
.
31.
Chen
P
,
Zuo
H
,
Xiong
H
,
Kolar
MJ
,
Chu
Q
,
Saghatelian
A
, et al
.
Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis
.
Proc Natl Acad Sci USA
2017
;
114
:
580
5
.
32.
Brown
TP
,
Bhattacharjee
P
,
Ramachandran
S
,
Sivaprakasam
S
,
Ristic
B
,
Sikder
MOF
, et al
.
The lactate receptor GPR81 promotes breast cancer growth via a paracrine mechanism involving antigen-presenting cells in the tumor microenvironment
.
Oncogene
2020
;
39
:
3292
304
.
33.
Geeraerts
X
,
Fernández-Garcia
J
,
Hartmann
FJ
,
de Goede
KE
,
Martens
L
,
Elkrim
Y
, et al
.
Macrophages are metabolically heterogeneous within the tumor microenvironment
.
Cell Rep
2021
;
37
:
110171
.
34.
Okui
T
,
Hiasa
M
,
Hasegawa
K
,
Nakamura
T
,
Ono
K
,
Ibaragi
S
, et al
.
Lactate secreted via MCT4 from bonecolonizing breast cancer excites sensory neurons via GPR81
.
Int J Oncol
2023
;
62
:
39
.
35.
Roland
CL
,
Arumugam
T
,
Deng
D
,
Liu
SH
,
Philip
B
,
Gomez
S
, et al
.
Cell surface lactate receptor GPR81 is crucial for cancer cell survival
.
Cancer Res
2014
;
74
:
5301
10
.
36.
Lundø
K
,
Dmytriyeva
O
,
Spøhr
L
,
Goncalves-Alves
E
,
Yao
J
,
Blasco
LP
, et al
.
Lactate receptor GPR81 drives breast cancer growth and invasiveness through regulation of ECM properties and Notch ligand DLL4
.
BMC Cancer
2023
;
23
:
1136
.
37.
Ishihara
S
,
Hata
K
,
Hirose
K
,
Okui
T
,
Toyosawa
S
,
Uzawa
N
, et al
.
The lactate sensor GPR81 regulates glycolysis and tumor growth of breast cancer
.
Sci Rep
2022
;
12
:
6261
.
38.
Velcicky
J
,
Wilcken
R
,
Cotesta
S
,
Janser
P
,
Schlapbach
A
,
Wagner
T
, et al
.
Discovery and optimization of novel SUCNR1 inhibitors: design of zwitterionic derivatives with a salt bridge for the improvement of oral exposure
.
J Med Chem
2020
;
63
:
9856
75
.
39.
Bhuniya
D
,
Umrani
D
,
Dave
B
,
Salunke
D
,
Kukreja
G
,
Gundu
J
, et al
.
Discovery of a potent and selective small molecule hGPR91 antagonist
.
Bioorg Med Chem Lett
2011
;
21
:
3596
602
.
40.
Reddy
A
,
Bozi
LHM
,
Yaghi
OK
,
Mills
EL
,
Xiao
H
,
Nicholson
HE
, et al
.
pH-gated succinate secretion regulates muscle remodeling in response to exercise
.
Cell
2020
;
183
:
62
75
.
41.
Mills
EL
,
Pierce
KA
,
Jedrychowski
MP
,
Garrity
R
,
Winther
S
,
Vidoni
S
, et al
.
Accumulation of succinate controls activation of adipose tissue thermogenesis
.
Nature
2018
;
560
:
102
6
.
42.
Zeng
Q
,
Michael
IP
,
Zhang
P
,
Saghafinia
S
,
Knott
G
,
Jiao
W
, et al
.
Synaptic proximity enables NMDAR signalling to promote brain metastasis
.
Nature
2019
;
573
:
526
31
.
43.
Li
L
,
Hanahan
D
.
Hijacking the neuronal NMDAR signaling circuit to promote tumor growth and invasion
.
Cell
2013
;
153
:
86
100
.
44.
North
WG
,
Liu
F
,
Tian
R
,
Abbasi
H
,
Akerman
B
.
NMDA receptors are expressed in human ovarian cancer tissues and human ovarian cancer cell lines
.
Clin Pharmacol
2015
;
7
:
111
7
.
45.
Li
L
,
Zeng
Q
,
Bhutkar
A
,
Galván
JA
,
Karamitopoulou
E
,
Noordermeer
D
, et al
.
GKAP acts as a genetic modulator of NMDAR signaling to govern invasive tumor growth
.
Cancer Cell
2018
;
33
:
736
51
.
46.
North
WG
,
Gao
G
,
Memoli
VA
,
Pang
RH
,
Lynch
L
.
Breast cancer expresses functional NMDA receptors
.
Breast Cancer Res Treat
2010
;
122
:
307
14
.
47.
Yin
J
,
Tu
G
,
Peng
M
,
Zeng
H
,
Wan
X
,
Qiao
Y
, et al
.
GPER-regulated lncRNA-Glu promotes glutamate secretion to enhance cellular invasion and metastasis in triple-negative breast cancer
.
FASEB J
2020
;
34
:
4557
72
.
48.
Ramaswamy
P
,
Aditi Devi
N
,
Hurmath Fathima
K
,
Dalavaikodihalli Nanjaiah
N
.
Activation of NMDA receptor of glutamate influences MMP-2 activity and proliferation of glioma cells
.
Neurol Sci
2014
;
35
:
823
9
.
49.
Albayrak
G
,
Korkmaz
FD
,
Bali
EB
,
Bagriacik
EU
.
Antitumorigenic effect of memantine via interfering glutamate metabolism in mouse 4T1 breast tumor model
.
Anticancer Agents Med Chem
2021
;
21
:
1575
80
.
50.
Albayrak
G
,
Bali
EB
,
Korkmaz
FD
,
Bağrıaçık
.
Does memantine show chemopreventive effect against mice 4T1 breast tumor model?
Turk J Med Sci
2022
;
52
:
841
7
.
51.
Rossi
M
,
Altea-Manzano
P
,
Demicco
M
,
Doglioni
G
,
Bornes
L
,
Fukano
M
, et al
.
PHGDH heterogeneity potentiates cancer cell dissemination and metastasis
.
Nature
2022
;
605
:
747
53
.
52.
Liu
C-H
,
Hu
R-H
,
Huang
M-J
,
Lai
I-R
,
Chen
C-H
,
Lai
H-S
, et al
.
C1GALT1 promotes invasive phenotypes of hepatocellular carcinoma cells by modulating integrin β1 glycosylation and activity
.
PLoS One
2014
;
9
:
e94995
.
53.
Liao
Y-Y
,
Chuang
Y-T
,
Lin
H-Y
,
Lin
N-Y
,
Hsu
T-W
,
Hsieh
S-C
, et al
.
GALNT2 promotes invasiveness of colorectal cancer cells partly through AXL
.
Mol Oncol
2023
;
17
:
119
33
.
54.
Lin
M-C
,
Huang
M-J
,
Liu
C-H
,
Yang
T-L
,
Huang
M-C
.
GALNT2 enhances migration and invasion of oral squamous cell carcinoma by regulating EGFR glycosylation and activity
.
Oral Oncol
2014
;
50
:
478
84
.
55.
Hu
Q
,
Tian
T
,
Leng
Y
,
Tang
Y
,
Chen
S
,
Lv
Y
, et al
.
The O-glycosylating enzyme GALNT2 acts as an oncogenic driver in non-small cell lung cancer
.
Cell Mol Biol Lett
2022
;
27
:
71
.
56.
Ros
M
,
Nguyen
AT
,
Chia
J
,
Le Tran
S
,
Le Guezennec
X
,
McDowall
R
, et al
.
ER-resident oxidoreductases are glycosylated and trafficked to the cell surface to promote matrix degradation by tumour cells
.
Nat Cell Biol
2020
;
22
:
1371
81
.
57.
Khosrowabadi
E
,
Wenta
T
,
Keskitalo
S
,
Manninen
A
,
Kellokumpu
S
.
Altered glycosylation of several metastasis-associated glycoproteins with terminal GalNAc defines the highly invasive cancer cell phenotype
.
Oncotarget
2022
;
13
:
73
89
.
58.
Nguyen
AT
,
Chia
J
,
Ros
M
,
Hui
KM
,
Saltel
F
,
Bard
F
.
Organelle specific O-glycosylation drives MMP14 activation, tumor growth, and metastasis
.
Cancer Cell
2017
;
32
:
639
53
.
59.
Song
K-H
,
Park
MS
,
Nandu
TS
,
Gadad
S
,
Kim
S-C
,
Kim
M-Y
.
GALNT14 promotes lung-specific breast cancer metastasis by modulating self-renewal and interaction with the lung microenvironment
.
Nat Commun
2016
;
7
:
13796
.
60.
Huang
T
,
Meng
F
,
Huang
H
,
Wang
L
,
Wang
L
,
Liu
Y
, et al
.
GALNT8 suppresses breast cancer cell metastasis potential by regulating EGFR O-GalNAcylation
.
Biochem Biophys Res Commun
2022
;
601
:
16
23
.
61.
Ščupáková
K
,
Adelaja
OT
,
Balluff
B
,
Ayyappan
V
,
Tressler
CM
,
Jenkinson
NM
, et al
.
Clinical importance of high-mannose, fucosylated, and complex N-glycans in breast cancer metastasis
.
JCI Insight
2021
;
6
:
e146945
.
62.
Dalangood
S
,
Zhu
Z
,
Ma
Z
,
Li
J
,
Zeng
Q
,
Yan
Y
, et al
.
Identification of glycogene-type and validation of ST3GAL6 as a biomarker predicts clinical outcome and cancer cell invasion in urinary bladder cancer
.
Theranostics
2020
;
10
:
10078
91
.
63.
Pietrobono
S
,
Anichini
G
,
Sala
C
,
Manetti
F
,
Almada
LL
,
Pepe
S
, et al
.
ST3GAL1 is a target of the SOX2-GLI1 transcriptional complex and promotes melanoma metastasis through AXL
.
Nat Commun
2020
;
11
:
5865
.
64.
Bai
Q
,
Liu
L
,
Xia
Y
,
Long
Q
,
Wang
J
,
Xu
J
, et al
.
Prognostic significance of ST3GAL-1 expression in patients with clear cell renal cell carcinoma
.
BMC Cancer
2015
;
15
:
880
.
65.
Carnielli
CM
,
Melo de Lima Morais
T
,
Malta de Sá Patroni
F
,
Prado Ribeiro
AC
,
Brandão
TB
,
Sobroza
E
, et al
.
Comprehensive glycoprofiling of oral tumors associates N-glycosylation with lymph node metastasis and patient survival
.
Mol Cell Proteomics
2023
;
22
:
100586
.
66.
Cui
H-X
,
Wang
H
,
Wang
Y
,
Song
J
,
Tian
H
,
Xia
C
, et al
.
ST3Gal III modulates breast cancer cell adhesion and invasion by altering the expression of invasion-related molecules
.
Oncol Rep
2016
;
36
:
3317
24
.
67.
Horie
M
,
Takagane
K
,
Itoh
G
,
Kuriyama
S
,
Yanagihara
K
,
Yashiro
M
, et al
.
Exosomes secreted by ST3GAL5high cancer cells promote peritoneal dissemination by establishing a premetastatic microenvironment
.
Mol Oncol
2024
;
18
:
21
43
.
68.
Zou
X
,
Lu
J
,
Deng
Y
,
Liu
Q
,
Yan
X
,
Cui
Y
, et al
.
ST6GAL1 inhibits metastasis of hepatocellular carcinoma via modulating sialylation of MCAM on cell surface
.
Oncogene
2023
;
42
:
516
29
.
69.
Teoh
ST
,
Ogrodzinski
MP
,
Ross
C
,
Hunter
KW
,
Lunt
SY
.
Sialic acid metabolism: a key player in breast cancer metastasis revealed by metabolomics
.
Front Oncol.
2018
;
8
:
174
.
70.
Zhou
X
,
Zhai
Y
,
Liu
C
,
Yang
G
,
Guo
J
,
Li
G
, et al
.
Sialidase NEU1 suppresses progression of human bladder cancer cells by inhibiting fibronectin-integrin α5β1 interaction and Akt signaling pathway
.
Cell Commun Signal
2020
;
18
:
44
.
71.
Zhang
X
,
Dou
P
,
Akhtar
ML
,
Liu
F
,
Hu
X
,
Yang
L
, et al
.
NEU4 inhibits motility of HCC cells by cleaving sialic acids on CD44
.
Oncogene
2021
;
40
:
5427
40
.
72.
Peng
Q
,
Gao
L
,
Cheng
HB
,
Wang
JS
,
Wang
J
.
Sialidase NEU1 may serve as a potential biomarker of proliferation, migration and prognosis in melanoma
.
World J Oncol
2022
;
13
:
222
34
.
73.
Tatsuta
T
,
Ito
J
,
Yamamoto
K
,
Sugawara
S
,
Hosono
M
,
Sato
M
, et al
.
Sialidase NEU3 contributes to the invasiveness of bladder cancer
.
Biomedicines
2024
;
12
:
192
.
74.
Chen
L
,
Zhou
Q
,
Zhang
P
,
Tan
W
,
Li
Y
,
Xu
Z
, et al
.
Direct stimulation of de novo nucleotide synthesis by O-GlcNAcylation
.
Nat Chem Biol
2024
;
20
:
19
29
.
75.
Xu
D
,
Wang
W
,
Bian
T
,
Yang
W
,
Shao
M
,
Yang
H
.
Increased expression of O-GlcNAc transferase (OGT) is a biomarker for poor prognosis and allows tumorigenesis and invasion in colon cancer
.
Int J Clin Exp Pathol
2019
;
12
:
1305
14
.
76.
Liu
Y
,
Yu
K
,
Kong
X
,
Zhang
K
,
Wang
L
,
Zhang
N
, et al
.
FOXA1 O-GlcNAcylation-mediated transcriptional switch governs metastasis capacity in breast cancer
.
Sci Adv
2023
;
9
:
eadg7112
.
77.
Sharma
NS
,
Gupta
VK
,
Garrido
VT
,
Hadad
R
,
Durden
BC
,
Kesh
K
, et al
.
Targeting tumor-intrinsic hexosamine biosynthesis sensitizes pancreatic cancer to anti–PD-1 therapy
.
J Clin Invest
2019
;
130
:
451
65
.
78.
Liu
C
,
Deng
S
,
Xiao
Z
,
Lu
R
,
Cheng
H
,
Feng
J
, et al
.
Glutamine is a substrate for glycosylation and CA19-9 biosynthesis through hexosamine biosynthetic pathway in pancreatic cancer
.
Discover Oncology
2023
;
14
:
20
.
79.
Campbell
S
,
Mesaros
C
,
Izzo
L
,
Affronti
H
,
Noji
M
,
Schaffer
BE
, et al
.
Glutamine deprivation triggers NAGK-dependent hexosamine salvage
.
eLife
2021
;
10
:
e62644
.
80.
Shi
Q
,
Shen
Q
,
Liu
Y
,
Shi
Y
,
Huang
W
,
Wang
X
, et al
.
Increased glucose metabolism in TAMs fuels O-GlcNAcylation of lysosomal Cathepsin B to promote cancer metastasis and chemoresistance
.
Cancer Cell
2022
;
40
:
1207
22
.
81.
Bouchard
G
,
Garcia-Marques
FJ
,
Karacosta
LG
,
Zhang
W
,
Bermudez
A
,
Riley
NM
, et al
.
Multiomics analysis of spatially distinct stromal cells reveals tumor-induced O-glycosylation of the CDK4–pRB axis in fibroblasts at the invasive tumor edge
.
Cancer Res
2022
;
82
:
648
64
.
82.
Li
Y
,
Liu
Y
,
Zhu
H
,
Chen
X
,
Tian
M
,
Wei
Y
, et al
.
N-acetylglucosaminyltransferase I promotes glioma cell proliferation and migration through increasing the stability of the glucose transporter GLUT1
.
FEBS Lett
2020
;
594
:
358
66
.
83.
Marhuenda
E
,
Fabre
C
,
Zhang
C
,
Martin-Fernandez
M
,
Iskratsch
T
,
Saleh
A
, et al
.
Glioma stem cells invasive phenotype at optimal stiffness is driven by MGAT5 dependent mechanosensing
.
J Exp Clin Cancer Res
2021
;
40
:
139
.
84.
Li
J
,
Xu
J
,
Li
L
,
Ianni
A
,
Kumari
P
,
Liu
S
, et al
.
MGAT3-mediated glycosylation of tetraspanin CD82 at asparagine 157 suppresses ovarian cancer metastasis by inhibiting the integrin signaling pathway
.
Theranostics
2020
;
10
:
6467
82
.
85.
Tan
Z
,
Cao
L
,
Wu
Y
,
Wang
B
,
Song
Z
,
Yang
J
, et al
.
Bisecting GlcNAc modification diminishes the pro-metastatic functions of small extracellular vesicles from breast cancer cells
.
J Extracell Vesicles
2020
;
10
:
e12005
.
86.
You
X
,
Wang
Y
,
Meng
J
,
Han
S
,
Liu
L
,
Sun
Y
, et al
.
Exosomal miR663b exposed to TGFβ1 promotes cervical cancer metastasis and epithelialmesenchymal transition by targeting MGAT3
.
Oncol Rep
2021
;
45
:
12
.
87.
Wang
Y
,
Li
Q
,
Niu
L
,
Xu
L
,
Guo
Y
,
Wang
L
, et al
.
Suppression of G6PD induces the expression and bisecting GlcNAc-branched N-glycosylation of E-Cadherin to block epithelial-mesenchymal transition and lymphatic metastasis
.
Br J Cancer
2020
;
123
:
1315
25
.
88.
Reticker-Flynn
NE
,
Bhatia
SN
.
Aberrant glycosylation promotes lung cancer metastasis through adhesion to galectins in the metastatic niche
.
Cancer Discov
2015
;
5
:
168
81
.
89.
Esposito
M
,
Mondal
N
,
Greco
TM
,
Wei
Y
,
Spadazzi
C
,
Lin
S-C
, et al
.
Bone vascular niche E-selectin induces mesenchymal–epithelial transition and Wnt activation in cancer cells to promote bone metastasis
.
Nat Cell Biol
2019
;
21
:
627
39
.
90.
Al Saoud
R
,
Hamrouni
A
,
Idris
A
,
Mousa
WK
,
Abu Izneid
T
.
Recent advances in the development of sialyltransferase inhibitors to control cancer metastasis: a comprehensive review
.
Biomed Pharmacother
2023
;
165
:
115091
.
91.
Doshi
MB
,
Lee
N
,
Tseyang
T
,
Ponomarova
O
,
Goel
HL
,
Spears
M
, et al
.
Disruption of sugar nucleotide clearance is a therapeutic vulnerability of cancer cells
.
Nature
2023
;
623
:
625
32
.
92.
Radenkovic
S
,
Bird
MJ
,
Emmerzaal
TL
,
Wong
SY
,
Felgueira
C
,
Stiers
KM
, et al
.
The metabolic map into the pathomechanism and treatment of PGM1-CDG
.
Am J Hum Genet
2019
;
104
:
835
46
.
93.
Kong
F
,
Ma
L
,
Wang
X
,
You
H
,
Zheng
K
,
Tang
R
.
Regulation of epithelial-mesenchymal transition by protein lysine acetylation
.
Cell Commun Signal
2022
;
20
:
57
.
94.
Hu
Z
,
Wei
F
,
Su
Y
,
Wang
Y
,
Shen
Y
,
Fang
Y
, et al
.
Histone deacetylase inhibitors promote breast cancer metastasis by elevating NEDD9 expression
.
Signal Transduct Target Ther
2023
;
8
:
11
.
95.
Leslie
PL
,
Chao
YL
,
Tsai
Y-H
,
Ghosh
SK
,
Porrello
A
,
Van Swearingen
AED
, et al
.
Histone deacetylase 11 inhibition promotes breast cancer metastasis from lymph nodes
.
Nat Commun
2019
;
10
:
4192
.
96.
He
C
,
Danes
JM
,
Hart
PC
,
Zhu
Y
,
Huang
Y
,
de Abreu
AL
, et al
.
SOD2 acetylation on lysine 68 promotes stem cell reprogramming in breast cancer
.
Proc Natl Acad Sci U S A
2019
;
116
:
23534
41
.
97.
Chen
Y
,
Zhang
B
,
Bao
L
,
Jin
L
,
Yang
M
,
Peng
Y
, et al
.
ZMYND8 acetylation mediates HIF-dependent breast cancer progression and metastasis
.
J Clin Invest
2018
;
128
:
1937
55
.
98.
Yu
B
,
Luo
F
,
Sun
B
,
Liu
W
,
Shi
Q
,
Cheng
S-Y
, et al
.
KAT6A acetylation of SMAD3 regulates myeloid-derived suppressor cell recruitment, metastasis, and immunotherapy in triple-negative breast cancer
.
Adv Sci (Weinh)
2021
;
8
:
e2100014
.
99.
Altea-Manzano
P
,
Doglioni
G
,
Liu
Y
,
Cuadros
AM
,
Nolan
E
,
Fernández-García
J
, et al
.
A palmitate-rich metastatic niche enables metastasis growth via p65 acetylation resulting in pro-metastatic NF-κB signaling
.
Nat Cancer
2023
;
4
:
344
64
.
100.
Pascual
G
,
Domínguez
D
,
Elosúa-Bayes
M
,
Beckedorff
F
,
Laudanna
C
,
Bigas
C
, et al
.
Dietary palmitic acid promotes a prometastatic memory via Schwann cells
.
Nature
2021
;
599
:
485
90
.
101.
Pascual
G
,
Avgustinova
A
,
Mejetta
S
,
Martín
M
,
Castellanos
A
,
Attolini
CS-O
, et al
.
Targeting metastasis-initiating cells through the fatty acid receptor CD36
.
Nature
2017
;
541
:
41
5
.
102.
Huang
Y-C
,
Hou
M-F
,
Tsai
Y-M
,
Pan
Y-C
,
Tsai
P-H
,
Lin
Y-S
, et al
.
Involvement of ACACA (acetyl-CoA carboxylase α) in the lung pre-metastatic niche formation in breast cancer by senescence phenotypic conversion in fibroblasts
.
Cell Oncol (Dordr)
2023
;
46
:
643
60
.
103.
Li
Y
,
He
X
,
Lu
X
,
Gong
Z
,
Li
Q
,
Zhang
L
, et al
.
METTL3 acetylation impedes cancer metastasis via fine-tuning its nuclear and cytosolic functions
.
Nat Commun
2022
;
13
:
6350
.
104.
Zhang
G
,
Huang
R
,
Zhao
H
,
Xia
Y
,
Huang
H
,
Qian
M
, et al
.
ACAT1-mediated METTL3 acetylation inhibits cell migration and invasion in triple negative breast cancer
.
Genes Immun
2023
;
24
:
99
107
.
105.
Jiang
Y
,
Luo
Z
,
Gong
Y
,
Fu
Y
,
Luo
Y
.
NAD+ supplementation limits triple-negative breast cancer metastasis via SIRT1-P66Shc signaling
.
Oncogene
2023
;
42
:
808
24
.
106.
Bondarev
AD
,
Attwood
MM
,
Jonsson
J
,
Chubarev
VN
,
Tarasov
VV
,
Schiöth
HB
.
Recent developments of HDAC inhibitors: emerging indications and novel molecules
.
Br J Clin Pharmacol
2021
;
87
:
4577
97
.
107.
Armstrong
AJ
,
Gordon
MS
,
Reimers
MA
,
Sedkov
A
,
Lipford
K
,
Snavely-Merhaut
J
, et al
.
The Courage study: a first-in-human phase 1 study of the CBP/p300 inhibitor FT-7051 in men with metastatic castration-resistant prostate cancer
.
J Clin Oncol
2021
;
39
:
TPS5085
.
108.
Dolgin
E
.
A fat-blocking drug could help to fight metastatic cancer
.
Nature
2023
. doi: .
109.
Martin-Perez
M
,
Urdiroz-Urricelqui
U
,
Bigas
C
,
Benitah
SA
.
The role of lipids in cancer progression and metastasis
.
Cell Metab
2022
;
34
:
1675
99
.
110.
Kong
Y
,
Liu
Y
,
Li
X
,
Rao
M
,
Li
D
,
Ruan
X
, et al
.
Palmitoylation landscapes across human cancers reveal a role of palmitoylation in tumorigenesis
.
J Transl Med
2023
;
21
:
826
.
111.
Wang
Y
,
Shen
N
,
Yang
Y
,
Xia
Y
,
Zhang
W
,
Lu
Y
, et al
.
ZDHHC5-mediated S-palmitoylation of FAK promotes its membrane localization and epithelial-mesenchymal transition in glioma
.
Cell Commun Signal
2024
;
22
:
46
.
112.
Zhang
Z
,
Li
X
,
Yang
F
,
Chen
C
,
Liu
P
,
Ren
Y
, et al
.
DHHC9-mediated GLUT1 S-palmitoylation promotes glioblastoma glycolysis and tumorigenesis
.
Nat Commun
2021
;
12
:
5872
.
113.
Hong
JY
,
Malgapo
MIP
,
Liu
Y
,
Yang
M
,
Zhu
C
,
Zhang
X
, et al
.
High-throughput enzyme assay for screening inhibitors of the ZDHHC3/7/20 acyltransferases
.
ACS Chem Biol
2021
;
16
:
1318
24
.
114.
Jiang
Y
,
Xu
Y
,
Zhu
C
,
Xu
G
,
Xu
L
,
Rao
Z
, et al
.
STAT3 palmitoylation initiates a positive feedback loop that promotes the malignancy of hepatocellular carcinoma cells in mice
.
Sci Signal
2023
;
16
:
eadd2282
.
115.
Aramsangtienchai
P
,
Spiegelman
NA
,
Cao
J
,
Lin
H
.
S-palmitoylation of junctional adhesion molecule C regulates its tight junction localization and cell migration
.
J Biol Chem
2017
;
292
:
5325
34
.
116.
Jang
D
,
Kwon
H
,
Jeong
K
,
Lee
J
,
Pak
Y
.
Essential role of flotillin-1 palmitoylation in the intracellular localization and signaling function of IGF-1 receptor
.
J Cell Sci
2015
;
128
:
2179
90
.
117.
Kwon
H
,
Choi
M
,
Ahn
Y
,
Jang
D
,
Pak
Y
.
Flotillin-1 palmitoylation turnover by APT-1 and ZDHHC-19 promotes cervical cancer progression by suppressing IGF-1 receptor desensitization and proteostasis
.
Cancer Gene Ther
2023
;
30
:
302
12
.
118.
Zhang
Q
,
Yang
X
,
Wu
J
,
Ye
S
,
Gong
J
,
Cheng
WM
, et al
.
Reprogramming of palmitic acid induced by dephosphorylation of ACOX1 promotes β-catenin palmitoylation to drive colorectal cancer progression
.
Cell Discov
2023
;
9
:
26
.
119.
De Piano
M
,
Manuelli
V
,
Zadra
G
,
Otte
J
,
Edqvist
P-HD
,
Pontén
F
, et al
.
Lipogenic signalling modulates prostate cancer cell adhesion and migration via modification of Rho GTPases
.
Oncogene
2020
;
39
:
3666
79
.
120.
Terry
AR
,
Nogueira
V
,
Rho
H
,
Ramakrishnan
G
,
Li
J
,
Kang
S
, et al
.
CD36 maintains lipid homeostasis via selective uptake of monounsaturated fatty acids during matrix detachment and tumor progression
.
Cell Metab
2023
;
35
:
2060
76
.
121.
Hao
J-W
,
Wang
J
,
Guo
H
,
Zhao
Y-Y
,
Sun
H-H
,
Li
Y-F
, et al
.
CD36 facilitates fatty acid uptake by dynamic palmitoylation-regulated endocytosis
.
Nat Commun
2020
;
11
:
4765
.
122.
Zhang
D
,
Tang
Z
,
Huang
H
,
Zhou
G
,
Cui
C
,
Weng
Y
, et al
.
Metabolic regulation of gene expression by histone lactylation
.
Nature
2019
;
574
:
575
80
.
123.
Yang
Z
,
Yan
C
,
Ma
J
,
Peng
P
,
Ren
X
,
Cai
S
, et al
.
Lactylome analysis suggests lactylation-dependent mechanisms of metabolic adaptation in hepatocellular carcinoma
.
Nat Metab
2023
;
5
:
61
79
.
124.
Meng
Q
,
Sun
H
,
Zhang
Y
,
Yang
X
,
Hao
S
,
Liu
B
, et al
.
Lactylation stabilizes DCBLD1 activating the pentose phosphate pathway to promote cervical cancer progression
.
J Exp Clin Cancer Res
2024
;
43
:
36
.
125.
Chen
M
,
Cen
K
,
Song
Y
,
Zhang
X
,
Liou
Y-C
,
Liu
P
, et al
.
NUSAP1-LDHA-glycolysis-lactate feedforward loop promotes Warburg effect and metastasis in pancreatic ductal adenocarcinoma
.
Cancer Lett
2023
;
567
:
216285
.
126.
Tasdogan
A
,
Faubert
B
,
Ramesh
V
,
Ubellacker
JM
,
Shen
B
,
Solmonson
A
, et al
.
Metabolic heterogeneity confers differences in melanoma metastatic potential
.
Nature
2020
;
577
:
115
20
.
127.
Zhang
Y
,
Yu
H
,
Zhang
J
,
Gao
H
,
Wang
S
,
Li
S
, et al
.
Cul4A-DDB1-mediated monoubiquitination of phosphoglycerate dehydrogenase promotes colorectal cancer metastasis via increased S-adenosylmethionine
.
J Clin Invest
2021
;
131
:
e146187
.
128.
Shi
X
,
Tasdogan
A
,
Huang
F
,
Hu
Z
,
Morrison
SJ
,
DeBerardinis
RJ
.
The abundance of metabolites related to protein methylation correlates with the metastatic capacity of human melanoma xenografts
.
Sci Adv
2017
;
3
:
eaao5268
.
129.
Jeon
H
,
Kim
JH
,
Lee
E
,
Jang
YJ
,
Son
JE
,
Kwon
JY
, et al
.
Methionine deprivation suppresses triple-negative breast cancer metastasis in vitro and in vivo
.
Oncotarget
2016
;
7
:
67223
34
.
130.
Li
Y
,
Liu
C
,
Xin
L
,
Liu
C
,
Cao
J
,
Yue
Z
, et al
.
Upregulation of E-cadherin by the combination of methionine restriction and HDAC2 intervention for inhibiting gastric carcinoma metastasis
.
Acta Biochim Biophys Sin (Shanghai)
2023
;
56
:
62
70
.
131.
Wang
Z
,
Yip
LY
,
Lee
JHJ
,
Wu
Z
,
Chew
HY
,
Chong
PKW
, et al
.
Methionine is a metabolic dependency of tumor-initiating cells
.
Nat Med
2019
;
25
:
825
37
.
132.
Dang
L
,
Wang
Y
.
Prognostic value of nicotinamide N-methyltransferase in human cancers: evidence from a meta-analysis and database validation
.
Open Med (Wars)
2022
;
17
:
292
303
.
133.
Pozzi
V
,
Salvolini
E
,
Lucarini
G
,
Salvucci
A
,
Campagna
R
,
Rubini
C
, et al
.
Cancer stem cell enrichment is associated with enhancement of nicotinamide N-methyltransferase expression
.
IUBMB Life
2020
;
72
:
1415
25
.
134.
Wang
Y
,
Zhou
X
,
Lei
Y
,
Chu
Y
,
Yu
X
,
Tong
Q
, et al
.
NNMT contributes to high metastasis of triple negative breast cancer by enhancing PP2A/MEK/ERK/c-Jun/ABCA1 pathway mediated membrane fluidity
.
Cancer Lett
2022
;
547
:
215884
.
135.
Couto
JP
,
Vulin
M
,
Jehanno
C
,
Coissieux
M-M
,
Hamelin
B
,
Schmidt
A
, et al
.
Nicotinamide N-methyltransferase sustains a core epigenetic program that promotes metastatic colonization in breast cancer
.
EMBO J
2023
;
42
:
e112559
.
136.
Lu
S
,
Ke
S
,
Wang
C
,
Xu
Y
,
Li
Z
,
Song
K
, et al
.
NNMT promotes the progression of intrahepatic cholangiocarcinoma by regulating aerobic glycolysis via the EGFR-STAT3 axis
.
Oncogenesis
2022
;
11
:
39
.
137.
Ruf
S
,
Rajagopal
S
,
Kadnur
SV
,
Hallur
MS
,
Rani
S
,
Kristam
R
, et al
.
Novel tricyclic small molecule inhibitors of nicotinamide N-methy­ltransferase for the treatment of metabolic disorders
.
Sci Rep
2022
;
12
:
15440
.
138.
Kannt
A
,
Rajagopal
S
,
Kadnur
SV
,
Suresh
J
,
Bhamidipati
RK
,
Swaminathan
S
, et al
.
A small molecule inhibitor of Nicotinamide N-methyltransferase for the treatment of metabolic disorders
.
Sci Rep
2018
;
8
:
3660
.
139.
Li
P
,
Xia
C
,
Kong
X
,
Zhang
J
.
Enhancing nicotinamide N-methyltransferase bisubstrate inhibitor activity through 7-deazaadenosine and linker modifications
.
Bioorg Chem
2024
;
143
:
106963
.
140.
Gao
Y
,
van Haren
MJ
,
Buijs
N
,
Innocenti
P
,
Zhang
Y
,
Sartini
D
, et al
.
Potent inhibition of nicotinamide N-methyltransferase by Alkene-linked bisubstrate mimics bearing electron deficient aromatics
.
J Med Chem
2021
;
64
:
12938
63
.
141.
Xu
D
,
Shao
F
,
Bian
X
,
Meng
Y
,
Liang
T
,
Lu
Z
.
The evolving landscape of noncanonical functions of metabolic enzymes in cancer and other pathologies
.
Cell Metab
2021
;
33
:
33
50
.
142.
Kim
J
,
Kang
J
,
Kang
Y-L
,
Woo
J
,
Kim
Y
,
Huh
J
, et al
.
Ketohexokinase-A acts as a nuclear protein kinase that mediates fructose-induced metastasis in breast cancer
.
Nat Commun
2020
;
11
:
5436
.
143.
Kang
Y-L
,
Kim
J
,
Kwak
S-B
,
Kim
Y-S
,
Huh
J
,
Park
J-W
.
The polyol pathway and nuclear ketohexokinase A signaling drive hyperglycemia-induced metastasis of gastric cancer
.
Exp Mol Med
2024
;
56
:
220
34
.
144.
Dasgupta
S
,
Rajapakshe
K
,
Zhu
B
,
Nikolai
BC
,
Yi
P
,
Putluri
N
, et al
.
Metabolic enzyme PFKFB4 activates transcriptional coactivator SRC-3 to drive breast cancer
.
Nature
2018
;
556
:
249
54
.
145.
Dai
T
,
Rosario
SR
,
Katsuta
E
,
Sawant Dessai
A
,
Paterson
EJ
,
Novickis
AT
, et al
.
Hypoxic activation of PFKFB4 in breast tumor microenvironment shapes metabolic and cellular plasticity to accentuate metastatic competence
.
Cell Rep
2022
;
41
:
111756
.
146.
Meng
J
,
Chen
X
,
Han
Z
.
PFKFB4 promotes lung adenocarcinoma progression via phosphorylating and activating transcriptional coactivator SRC-2
.
BMC Pulm Med
2021
;
21
:
60
.
147.
Hosios
AM
,
Fiske
BP
,
Gui
DY
,
Vander Heiden
MG
.
Lack of evidence for pkm2 protein kinase activity
.
Mol Cell
2015
;
59
:
850
7
.
148.
Zhou
Q
,
Yin
Y
,
Yu
M
,
Gao
D
,
Sun
J
,
Yang
Z
, et al
.
GTPBP4 promotes hepatocellular carcinoma progression and metastasis via the PKM2 dependent glucose metabolism
.
Redox Biol
2022
;
56
:
102458
.
149.
Zhao
G
,
Yuan
H
,
Li
Q
,
Zhang
J
,
Guo
Y
,
Feng
T
, et al
.
DDX39B drives colorectal cancer progression by promoting the stability and nuclear translocation of PKM2
.
Signal Transduct Target Ther
2022
;
7
:
275
.
150.
Blaha
CS
,
Ramakrishnan
G
,
Jeon
S-M
,
Nogueira
V
,
Rho
H
,
Kang
S
, et al
.
A non-catalytic scaffolding activity of hexokinase 2 contributes to EMT and metastasis
.
Nat Commun
2022
;
13
:
899
.
151.
Thomas
GE
,
Egan
G
,
García-Prat
L
,
Botham
A
,
Voisin
V
,
Patel
PS
, et al
.
The metabolic enzyme hexokinase 2 localizes to the nucleus in AML and normal haematopoietic stem and progenitor cells to maintain stemness
.
Nat Cell Biol
2022
;
24
:
872
84
.
152.
Zheng
Y
,
Zhan
Y
,
Zhang
Y
,
Zhang
Y
,
Liu
Y
,
Xie
Y
, et al
.
Hexokinase 2 confers radio-resistance in hepatocellular carcinoma by promoting autophagy-dependent degradation of AIMP2
.
Cell Death Dis
2023
;
14
:
488
.
153.
Song
J
,
Li
H
,
Liu
Y
,
Li
X
,
Shi
Q
,
Lei
Q-Y
, et al
.
Aldolase A accelerates cancer progression by modulating mRNA translation and protein biosynthesis via noncanonical mechanisms
.
Adv Sci (Weinh)
2023
;
10
:
e2302425
.
154.
Liu
P
,
Sun
S-J
,
Ai
Y-J
,
Feng
X
,
Zheng
Y-M
,
Gao
Y
, et al
.
Elevated nuclear localization of glycolytic enzyme TPI1 promotes lung adenocarcinoma and enhances chemoresistance
.
Cell Death Dis
2022
;
13
:
205
.
155.
Luo
J-Q
,
Yang
T-W
,
Wu
J
,
Lai
H-H
,
Zou
L-B
,
Chen
W-B
, et al
.
Exosomal PGAM1 promotes prostate cancer angiogenesis and metastasis by interacting with ACTG1
.
Cell Death Dis
2023
;
14
:
502
.
156.
Li
H-J
,
Ke
F-Y
,
Lin
C-C
,
Lu
M-Y
,
Kuo
Y-H
,
Wang
Y-P
, et al
.
ENO1 promotes lung cancer metastasis via HGFR and WNT signaling-driven epithelial-to-mesenchymal transition
.
Cancer Res
2021
;
81
:
4094
109
.
157.
Hamza
A
,
Cho
JY
,
Cap
KC
,
Hossain
AJ
,
Kim
J-G
,
Park
J-B
.
Extracellular pyruvate kinase M2 induces cell migration through p-Tyr42 RhoA-mediated superoxide generation and epithelial-mesenchymal transition
.
Free Radic Biol Med
2023
;
208
:
614
29
.
158.
Wang
C
,
Zhang
S
,
Liu
J
,
Tian
Y
,
Ma
B
,
Xu
S
, et al
.
Secreted pyruvate kinase m2 promotes lung cancer metastasis through activating the integrin Beta1/FAK signaling pathway
.
Cell Rep
2020
;
30
:
1780
97
.
159.
Luo
M-Y
,
Zhou
Y
,
Gu
W-M
,
Wang
C
,
Shen
N-X
,
Dong
J-K
, et al
.
Metabolic and nonmetabolic functions of PSAT1 coordinate signaling cascades to confer EGFR inhibitor resistance and drive progression in lung adenocarcinoma
.
Cancer Res
2022
;
82
:
3516
31
.
160.
Ilter
D
,
Drapela
S
,
Schild
T
,
Ward
NP
,
Adhikari
E
,
Low
V
, et al
.
NADK-mediated de novo NADP(H) synthesis is a metabolic adaptation essential for breast cancer metastasis
.
Redox Biol
2023
;
61
:
102627
.
161.
Zeng
Z
,
Gao
J
,
Chen
T
,
Zhang
Z
,
Li
M
,
Fan
Q
, et al
.
Nicotinamide adenine dinucleotide kinase promotes lymph node metastasis of NSCLC via activating ID1 expression through BMP pathway
.
Int J Biol Sci
2023
;
19
:
3184
99
.
162.
Ngo
B
,
Kim
E
,
Osorio-Vasquez
V
,
Doll
S
,
Bustraan
S
,
Liang
RJ
, et al
.
Limited environmental serine and glycine confer brain metastasis sensitivity to PHGDH inhibition
.
Cancer Discov
2020
;
10
:
1352
73
.
163.
Artinyan
A
,
Anaya
DA
,
McKenzie
S
,
Ellenhorn
JDI
,
Kim
J
.
Neoadjuvant therapy is associated with improved survival in resectable pancreatic adenocarcinoma
.
Cancer
2011
;
117
:
2044
9
.
164.
Encarnación-Rosado
J
,
Sohn
ASW
,
Biancur
DE
,
Lin
EY
,
Osorio-Vasquez
V
,
Rodrick
T
, et al
.
Targeting pancreatic cancer metabolic dependencies through glutamine antagonism
.
Nat Cancer
2024
;
5
:
85
99
.
165.
Recouvreux
MV
,
Grenier
SF
,
Zhang
Y
,
Esparza
E
,
Lambies
G
,
Galapate
CM
, et al
.
Glutamine mimicry suppresses tumor progression through asparagine metabolism in pancreatic ductal adenocarcinoma
.
Nat Cancer
2024
;
5
:
100
13
.
166.
Beaver
JA
,
Kluetz
PG
,
Pazdur
R
.
Metastasis-free survival: a new end point in prostate cancer trials
.
N Engl J Med
2018
;
378
:
2458
60
.