Metabolic Rewiring in Cancer: Small Molecule Inhibitors in Colorectal Cancer Therapy
Abstract
:1. Introduction
2. Metabolism-Based Inhibitors
2.1. Glycolytic Metabolism in Cancer
Lactate Dehydrogenase Inhibitors
- Sodium oxamate (1) (2-amino-2-oxoacetic acid sodium salt, Figure 3) is a structural analogue of pyruvate that is able to inhibit both LDHA/LDHB and aspartate aminotransferase (AAT). It competitively inhibits human LDHA (KI = 136 μM), binding at the pyruvate site and leading to the formation of an inactive complex [28]. Notably, several studies have shown the anticancer properties of compound 1 on different cancer cell lines, including those obtained from CRC, breast cancer, ovarian cancer, and lymphoma, highlighting the correlation with the inhibition of aerobic glycolysis due to its ability to significantly inhibit the growth of cancer cells exclusively under glucose-treated conditions [29,30,31,32,33]. In addition, in gastric cancer, where an overexpression of LDHA has been confirmed, sodium oxamate was observed to decrease lactate production, resulting in a dose-dependent inhibition of cancer cell proliferation in the presence of glucose.
- 2.
- Gossypol (2) ((2,2′-binaphthalene)-8,8′-dicarboxaldehyde,1,1′,6,6′,7,7′-hexahydroxy-5,5′-diisopropyl-3,3′-dimethyl, Figure 3) is the most important natural polyphenolic pigment present in cotton (Gossypium hirsutum L.); first isolated in its pure form by Marchlewski in 1899 [41], it is the only compound with anti-LDH activity that has been evaluated clinically to date. Originally evaluated for use as a male antifertility agent, 2 showed broad-spectrum antitumor, antiviral, antibacterial, and antioxidant activities [42]. Despite displaying a wide array of potential biological activities, compound 2 has a poor applicability in drug development due to its toxicity, which is possibly attributed to the presence of reactive aldehyde and catechol hydroxyl functional groups [43].
- 3.
- FX11 (3) (3-dihydroxy-6-methyl-7-(phenylmethyl)-4-propylnaphthalene-1-carboxylic acid, Figure 3) is a synthetic drug-like small molecule that is structurally related to gossypol, which was initially designed as an antimalarial agent and discovered through a Heparin-induced thrombocytopenia (HIT) assay. It is well known that compound 3 represents a sensitizer in chemotherapy-resistant tumor cells, and it acts as an NADH-competitive, -selective, and -reversible hLDH5 inhibitor with a Ki value of 0.05 μM [20]. Furthermore, preclinical studies have shown its efficacy in inhibiting the progression of various adult cancers, including pancreatic cancer, prostate cancer, lymphoma, and osteosarcoma.
- 4.
- Galloflavin (4, Figure 3), a flavone-like compound synthesized by gallic acid, is a non-selective inhibitor of both LDHA and LDHB that preferentially binds to the free enzyme without competing with the substrate (pyruvate) or with the cofactor (NADH). The calculated Ki values for its inhibitory activity were 5.46 μM and 56.0 μM against LDHA and LDHB, respectively. Notably, its ability to inhibit LDHA was found to be beneficial in overcoming acquired resistance to taxol and trastuzumab in breast cancer cells, which are both drugs that are widely used to treat this disease. Moreover, 4 showed the inhibition of aerobic glycolysis, without interference with cell respiration, with the consequent reduction in the growth of different cell lines such as colon, breast, liver, Burkitt lymphoma, and endometrial cancer cells [53]. Compound 4 exerted a similar growth inhibition in MCF-7, MDA-MB-231, and MCF-Tam cells, for which high glucose uptake has been observed. MDA-MB-231 and MCF-Tam cells, which exhibited different LDH expression levels, also experienced growth inhibition due to a constitutively activated stress response. Farabegoli et al. emphasized that the antiproliferative effect of compound 4 against different cell lines was attributable to different mechanisms, such as the downregulation of the ERα-mediated signaling pathway in MCF-7 cells and the induction of an oxidative stress condition in MDA-MB-231 and MCF-Tam cells [54].
- 5.
- 3-((3-carbamoyl-7-(3,5-dimethylisoxazol-4-yl)-6-methoxyquinolin-4-yl) amino) benzoic acid (GSK 2837808A, 5, Figure 3) is a quinoline 3-sulfonamides-based compound, obtained following lead optimization studies, that exhibits an inhibitory potency against LDHA of less than 2 nM and has selectivity over LDHB ranging from 10 to 80-fold. Its effect is exerted in the rate of lactate production in different cancer cell lines, including hepatocellular and breast carcinomas [26]. In hepatocellular carcinoma cells (HCCs), Billiard et al. demonstrated the ability of compound 5 to increase oxygen levels at doses of up to 3 μM in HCCs, while at a higher concentration, mitochondrial function was directly inhibited. In addition, in wider terms, a metabolite analysis in Snu398 cells, upon treatment with a quinoline-3-sulfonamide LDHA inhibitor, revealed an increased intracellular concentration of intermediates of glycolysis and the citric acid cycle, which was consistent with the enhanced activity of the Krebs cycle and with the blockage of cytosolic glycolysis. This resulted in an enhanced activity of pyruvate kinase isozymes M2 (PKM2) and in the induction of apoptosis in Snu398 cells [50,51].
- 6.
- N-Hydroxyindole-2-carboxylate derivatives (NHIs, 6, Figure 3). There has recently been a growing interest in developing NHIs as potential therapeutic agents due to their low likelihood of generating reactive intermediates [56,57]. Granchi et al. [58] reported the design and synthesis of a selected series of NHIs, with a particular focus on their inhibitory activity against LDHA, particularly targeting hLDH5. The importance of targeting the isoform 5 of LDHA is gathered from studies on LDHA-deprived cancer cell lines suggesting that targeting hLDH5 can significantly prevent tumor growth and invasiveness, especially under hypoxic conditions [59,60]. In addition, studies on individuals with hereditary LDHA deficiency revealed that the lack of this enzyme did not cause symptoms under normal circumstances, hence supporting the notion that hLDH5 could be a safe target for anti-tumor agents.
- 7.
- Pyrazole-based derivatives. In 2020, structure-guided SAR optimization studies of pyrazole-based derivatives [63] led Rai and coworkers [64] to the identification of two lead compounds, named NCATS-SM1440 (9, Figure 3) and NCATS-SM1441 (10, Figure 3), with robust in vivo LDH inhibition. Indeed, both compounds showed nearly identical potency against both LDHA and LDHB, with an IC50 value of 0.04 μM. Notably, their impact on the glycolytic pathway was assessed in human Ewing’s sarcoma (A673) cell lines through the glycolysis stress test (GST), which measures the extracellular acidification rate (ECAR) of the media. In this assay, the decrease in ECAR, which results from glycolysis-dependent proton production by the cells, indicated a depletion of NAD+ and, consequently, an inhibition of the entire pathway.
- 8.
- Pyran-4-one derivatives. The screening of a proprietary compound library containing 5000 small molecules led to the identification of 16 pyran-4-one-based compounds endowed with anti-LDHA activity. Among them, compound 12 (Figure 3), identified through hit-to-lead optimization studies by Zhou and co-workers [66], emerged as a potent LDHA inhibitor with an EC50 value of 90 nM.
- 9.
- RS6212 (13, Figure 3). Pyrimidine derivatives showed significant in vitro cell growth inhibition of medulloblastoma or pancreatic tumor cells [67]. A tetrahydropyrimidine-5-carboxamide derivative that is able to induce cancer regression in mice bearing human tumors without significant side effects was recently patented [68]. RS6212 represents a novel specific pyridazine LDHA inhibitor, which was discovered, starting from the available enzyme crystal structure, through structure-based virtual screening studies based on docking and pharmacophore filters [69]. A preliminary in vitro test highlighted the ability of compound 13 to exhibit micromolar anticancer activity against Med1-MB (IC50 of 81 μM), a cell line that is dependent on aerobic glycolysis, as well as against HCT116 and SW620 CRC cell lines, A549 lung cancer, and PANC-1 pancreatic cancer cells. As an LDHA inhibitor, 13 was remarkably superior to compound 1. Consistently, 13 increased the NADH content, decreased the lactate levels in tumor cells, and failed to inhibit the cell growth of HCT116 cells with LDHA deficiency. Compound 13 exhibited antitumor synergistic activity with rotenone, a proven mitochondrial complex I inhibitor that showed anti-carcinogenic activity in several studies [70].
2.2. Lipid Metabolism in Cancer
2.2.1. Fatty Acid Synthase Inhibitors
- Cerulein ((2S,3R)-2,3-epoxy-4-oxo-7,10-dodecadienamide, 14, Figure 6) is a natural decapeptide that is isolated from Cephalosporium caerulens, and it is endowed with structural elements common to the natural gastrointestinal peptide hormone cholecystokinin, which stimulates secretions on the stomach, bile, pancreas, and on some smooth muscles. It was the first antibiotic reported to inhibit FASN [86,87]. Compound 14 induced apoptosis in both Colon 26 and CMT 93 murine CRC cell lines. In Colon-26 cells, 14 significantly reduced the numbers and sizes of liver metastatic CRC tumors, and its activity was associated with reduced levels of phosphorylated Akt [88].
- 2.
- (-)-Epigallocatechin gallate (EGCG, 15, Figure 6), formally the ester of gallic acid with the (3R)-hydroxy group of (-)-epigallocatechin, is a polyphenol extracted from green tea and has been proven to be a potent inhibitor of FASN, along with (-)-gallocatechin gallate, another component of green tea extract, whereas ungallated catechins showed a weaker inhibition [93]. In a study that aimed to investigate the effects in CRC SW480, SW620, and LS411N cell lines, 15 significantly inhibited cell proliferation and induced apoptosis through the activation of apoptosis-related proteins, such as caspase-3, caspase-9, and PARP-1. Signaling and transcriptional activation factor 3 (STAT3) is involved in cancer propagation: it activates the transcription of downstream target genes, inhibits apoptosis, and regulates members of the Bcl-2 family [94,95]. In the three cell lines, 15 significantly downregulated both STAT3 and phosphorylated STAT3 (p-STAT3); decreased Bcl-2, MCL-1, and Vimentin; and increased E-cadherin [96]. Additionally, 15 showed to downregulate STAT3 expression in breast cancer stem cell phenotype MDA-MB-231 cells and MCF7 cells [97] and in nasopharyngeal carcinoma [98].
- 3.
- Luteolin (3′,4′,5,7-tetrahydroxyflavone, 16, Figure 6) is a naturally occurring 4H-chromen-4-one derivative belonging to the family of flavonoids that exhibits several therapeutic properties, such as anti-proliferative, antioxidant, anti-inflammatory, and anti-infective activities. Compound 16 showed anticancer activity against CRC by interfering with the Wnt/β-catenin signaling pathway [103] and by reducing oxidative stress [104]. Compound 16 proved to be a potent FASN inhibitor capable of reducing DNA synthesis and inhibiting HT-29 cell growth and proliferation through the induction of cell cycle arrest at the G1 and G2/M phases, leading to a dose-dependent apoptotic effect. In addition, compound 16 upregulated microRNA-384, a short non-coding RNA that is involved in the post-transcriptional regulation of gene expression, and it downregulated the expression of pleiotrophin, a potent mitogenic cytokine [105], both in CRC cells and tissues, suggesting that the anticancer activity exerted by 16 against CRC cells went through the modulation of the heparin-binding growth factor pleiotrophin (PTN) via microRNA 384 [106]. Apoptosis induced by 16 was mediated through the expression of death receptor 5 (DR5), the cleavage of Bcl-2-interacting domain, and the activation of caspases 3 and 9–10, suggesting that DR5 plays a role in 16-induced apoptosis [107].
- 4.
- TVB-3664 (17, Figure 6) is a potent FASN inhibitor developed by 3-V Biosciences. TVB-3664 demonstrated anti-tumor activity in multiple cancers, including CRC, in vitro and in vivo and has a favorable tolerability profile in phase I clinical trials [108,109]. CRC cells and patient-derived xenografts (PDXs) from patients with CRC have shown a wide range of sensitivity to FASN inhibitors. In nine CRC cells (PDXs), treatment with 17 yielded a significant reduction in the tumor volume in 30% of cases. The anti-tumor treatment with 17 caused significant reductions in the pool of adenine nucleotides, which play key metabolic functions [110], and in lipids, fatty acids, and phospholipids and caused an increase in sphingolipids.
2.2.2. ATP-Citrate Lyase Inhibitors
- SB204990 (18, Figure 8), a prodrug derived after the lactonization of the hydroxy-acid SB201076, represents a potent and specific ACLy inhibitor. In 2005, Hatzivassiliou et al. [119] demonstrated the ability of ACLy inhibition to suppress tumor growth, correlating with the glycolytic phenotype of the tumor. Their study included three human tumor cell lines—A549, PC3, and SKOV3—treated with compound 18, revealing dose- and time-dependent sensitivity. This treatment resulted in a notable reduction in the total acetyl-CoA level across all three treated cells compared to the controls. Notably, the SKOV3 cells exhibited resistance to compound 18, which was possibly attributed to their lower glycolysis rate compared to the other cell lines, resulting in reduced glucose utilization and lactate production.
- 2.
- GSK165 (2-hydroxy-N-arylbenzenesulfonamide, 19, Figure 8) was identified as a novel ACLy inhibitor with an in vitro IC50 value of 0.13 μM. In high-fat-diet-fed mice, 19 lowered plasma cholesterol, triglyceride, and glucose and inhibited weight gain [121]. Zhou et al. [118] reported that compound 19 exhibited growth inhibition in cancer cells as a standalone agent, with an IC50 value of ~30 μM, whereas at a concentration of 40 μM, compound 19 demonstrated its ability to sensitize HT29 cells to SN38.
2.2.3. Acetyl-CoA Carboxylase-α Inhibitors
- TOFA (5-tetradecyloxy-2-furoic acid, 20, Figure 10) is an allosteric inhibitor of the ACCA that represents the rate-limiting enzyme of the FA synthesis pathway. More precisely, inside adipocytes and hepatocytes, TOFA is converted to TOFyl-CoA (5-tetradecyloxy-2-furoyl-CoA), exerting the allosteric inhibition on ACCA [125]. It showed strong cytotoxicity and induced apoptosis in human NCI-H460 lung cancer, HCT-8, and HCT-15 colorectal cancer cells exposed to 20 in a dose-dependent fashion [126,127]. Moreover, it was reported that compound 20 inhibited fatty acid synthesis, increased the oxidation of fatty acid and ketogenesis, and decreased the synthesis of triglycerides and the production of very low-density lipoprotein (VLD) [128]. In addition, compound 20 inhibited the growth of ovarian cancer cell lines COC1 and COC1/DDP; arrested the cells in the G0/G1 cell cycle phase; induced apoptosis; inhibited the expressions of Cyclin D1, CDK4, and Bcl-2 proteins; and activated cleaved caspase-3 [129].
2.3. Glutaminolysis in Cancer
Glutaminolysis Inhibitors
- Compound 968 (21, Figure 13) is a small molecule that acts as an allosteric regulator of isoform 1 of glutaminase (GLS1) and inhibits the activity of both major splice GLS variants: long form KGA and short form GAC [142]. Compound 21 demonstrated antitumor activity in vitro in several types of cancer, including lymphoma, breast cancer, glioblastoma, and lung cancer [143,144,145]. In fibroblasts, 21 inhibited oncogenic transformation caused by various Rho family GTPases without exhibiting toxic effects on normal cells [146]. Compound 21 inhibited the proliferation of HEY, SKOV3, and IGROV-1 cells upon 5-day treatment and induced apoptosis [147]. In addition, 21, in combination with a low concentration of paclitaxel, showed stronger inhibitory effects. The treatment of endometrial cancer with 21 downregulated the expressions of GLS1 and cyclin D1 and upregulated the expressions of P21 and E-cadherin. In xenograft mouse models of endometrial cancer, 21 significantly suppressed tumor growth [148]. The combination of compound 21 with metformin effectively suppressed CSCs in SW620 cells, and this effect was further enhanced in HT29 cells. Notably, SW620 cells exhibited higher expression levels of GLU1 and the glutamine transporter ASCT2 compared to HT29 cells [149].
- BPTES (bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide, 22, Figure 13) was reported as a potent and allosterically selective inhibitor of GLS, consequently inhibiting glutaminolysis. A crystal structure analysis showed that compound 22 binds to the dimer interface of GLS, consequently stabilizing the inactive tetrameric form of the enzyme [150]. Compound 22 at 300 nM decreased the glutamate in intestinal epithelial and neuronal cells with highly specific binding to the enzyme and enzyme–substrate complex to form a stable but inactive tetramer. Despite its poor physiochemical properties, such as its poor aqueous solubility, it has been used as key molecular template for the identification of other allosteric GLS inhibitors with more potent and enhanced drug-like properties.
- 3.
- CB-839 (23, Figure 13) is a potent and selective inhibitor of glutaminase that showed in vitro antitumor activity against a panel of TNBC cell lines. Compound 23 showed in vivo efficacy in breast cancer xenograft models, both alone and in combination with paclitaxel [153]. Compound 23 exhibited cytotoxicity in tumor-infiltrating lymphocytes (TILs) isolated from patient-derived melanoma cells. Notably, it demonstrated a more potent decrease in the conversion of glutamine to α-KG in tumor cells compared to TILs in co-cultures. This suggests that 23 may have the potential to selectively modulate the metabolism of tumor and immune cells, potentially enhancing immune function in the tumor microenvironment. In vivo treatment with compound 23 activated melanoma antigen-specific T cells. Furthermore, in combination with anti-PD1 or anti-CTLA4 antibodies, it synergized their activity [154].
- 4.
- GPD-20 (24, Figure 13) and GPD-23 (25, Figure 13) are selenadiazole analogues of compound 23. They showed an enhanced inhibition of cancer cells and of the KGA isoform, demonstrated higher potency in inducing ROS, and showed better cell accumulation. However, in both the glutamine-dependent HCT116 and aggressive H22 liver cancer xenograft models, 24–25 only resulted in a partial reduction in the tumor size [157,158].
- 5.
- CDP-3B (Hexylselen, 26, Figure 13) is a seleno derivative inhibitor of KGA/glutamate dehydrogenase (GDH) that completely inhibited the cancer cell growth of many aggressive tumors, reduced the tumor size in an aggressive liver cancer xenograft model, and showed no toxic effects on normal cells at a concentration of up to 10 μM. Compound 26 also inhibited thioredoxin reductase (TrxR) and glutamine amidotransferase (GatCAB), thus affecting the Akt/Erk/caspase-9 signaling pathways [159].
2.4. Other Target Inhibitors
- Lonidamine (27, Figure 14) is a derivative of indazole-3-carboxylic acid that blocks glycolysis in cancer cells, resulting in a reduction in cellular ATP. It has progressed into clinical trials and gained approval for the treatment of lung, breast, prostate, and brain cancers either alone or in combination with other anticancer agents. Compound 27 has been explored for the treatment of several types of cancer, including CRC [160].
- 2.
- Mito-lonidamine (28, Figure 14) was synthesized by conjugating compound 27 with TPP+ via an aliphatic chain linker to enhance its potential mitochondrial activity. At low micromolar concentrations, compound 28 was more effective than 27 in primary lung tumors and lung cancer brain metastases in an orthotopic mouse model. It inhibited mitochondrial complexes I and II and stimulated ROS production through the induction of autophagic cell death [163]. In a phase II study involving patients with metastatic colorectal adenocarcinoma who had undergone previous chemotherapy for metastatic disease, the majority of those treated with compound 28 exhibited incomplete or partial remissions [167].
- 3.
- 2-deoxy-D-glucose (2-DG, 29, Figure 14) has demonstrated the ability to enhance the antitumor effects of radio- and chemotherapy both in vitro and in vivo across various solid tumors, including lung, breast, pancreas, head, neck, and gastric tumors, with weak damage to normal cells [168]. In cancer cells under hypoxic conditions, 29 is recognized as a glycolysis inhibitor that hampers ATP production. When applied to chemoresistant CRC cells expressing drug resistance-related proteins, compound 29 treatment led to a reduced expression of glycolysis-related enzymes and the secretion of cytokines associated with epithelial-to-mesenchymal transition (EMT) [169]. Moreover, 29 inhibited the activation of A disintegrin and metalloproteinase (ADAM)10 and, potentially, ADAM17, both of which play a central role in generating soluble CD137 (sCD137), which is considered a splice variant of CD137 within the TNF receptor family [170].
3. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
5-FU | 5-fluorouracil |
α-KG | α-ketoglutarate |
ACACA | acetyl-CoA carboxylase alpha |
ACLy | ATP-citrate lyase |
ADAM | A disintegrin and metalloproteinase |
ATP | adenosine triphosphate |
BRAF | B-Raf protooncogene |
CCR | colon-rectal cancer |
CoA | coenzyme A |
CRC | colorectal cancer |
CSCs | cancer stem cells |
DMEs | drug-metabolizing enzymes |
ECAR | extracellular acidification rate |
ER | endoplasmic reticulum |
FAs | fatty acids |
FASN | fatty acid synthase |
GA3PDH | glucelaldehyde-3-phosphate dehydrogenase |
GatCAB | glutamine amidotransferase |
GDH | glutamate dehydrogenase |
GLS | glutaminase |
GLS1 | glutaminase isoform 1 |
GLUD1 | glutamate dehydrogenase |
GMB | glioblastoma |
GnRH | gonadotropin-releasing hormone |
GST | glycolysis stress test |
HCC | hepatocellular carcinoma |
HIF-1 | hypoxia-inducible factor 1 |
HIT | heparin-induced thrombocytopenia |
ICDH | isocitrate dehydrogenase |
KGA | kidney mitochondrial glutaminase |
KRAS | Kirsten rat sarcoma viral oncogene homolog |
LDH | lactate dehydrogenase |
LDHA | lactate dehydrogenase A |
LDHB | lactate dehydrogenase B |
mCRC | metastatic colorectal cancer |
MDH | malate dehydrogenase |
miRNA-106a | microRNA-106a |
NSCLC | non-small-cell lung cancer |
OxPhos | oxidative phosphorylation |
PH1 | primary hyperoxaluria type 1 |
PK | pharmacokinetic |
PPAR | peroxisome-proliferator activated receptor |
p-STAT3 | phosphorylated STAT3 |
ROS | reactive oxygen species |
STAT3 | signaling and transcriptional activation factor 3 |
TCA | tricarboxylic acid |
TPP+ | triphenyl phosphonium cation |
TrxR | thioredoxin reductase |
VLDL | very-low-density lipoprotein |
References
- World Cancer Research Fund International Colorectal Cancer Statistics. Available online: https://www.wcrf.org/cancer-trends/colorectal-cancer-statistics (accessed on 22 September 2023).
- Macrae, F.A. Colorectal Cancer: Epidemiology, Risk Factors, and Protective Factors. Litterature Review. Available online: https://lifewithnogallbladder.org/article/colorectal-cancer-epidemiology-risk-factors-and-protective-factors/ (accessed on 22 September 2023).
- Burt, R.W.; DiSario, J.A.; Cannon-Albright, L. Genetics of Colon Cancer: Impact of Inheritance on Colon Cancer Risk. Annu. Rev. Med. 1995, 46, 371–379. [Google Scholar] [CrossRef] [PubMed]
- Lynch, H.T.; Smyrk, T.C.; Watson, P.; Lanspa, S.J.; Lynch, J.F.; Lynch, P.M.; Cavalieri, R.J.; Boland, C.R. Genetics, Natural History, Tumor Spectrum, and Pathology of Hereditary Nonpolyposis Colorectal Cancer: An Updated Review. Gastroenterology 1993, 104, 1535–1549. [Google Scholar] [CrossRef] [PubMed]
- Ponz de Leon, M.; Sassatelli, R.; Benatti, P.; Roncucci, L. Identification of Hereditary Nonpolyposis Colorectal Cancer in the General Population. The 6-Year Experience of a Population-Based Registry. Cancer 1993, 71, 3493–3501. [Google Scholar] [CrossRef] [PubMed]
- Botteri, E.; Iodice, S.; Bagnardi, V.; Raimondi, S.; Lowenfels, A.B.; Maisonneuve, P. Smoking and Colorectal Cancer: A Meta-Analysis. JAMA 2008, 300, 2765–2778. [Google Scholar] [CrossRef] [PubMed]
- Todoroki, I.; Friedman, G.D.; Slattery, M.L.; Potter, J.D.; Samowitz, W. Cholecystectomy and the Risk of Colon Cancer. Am. J. Gastroenterol. 1999, 94, 41–46. [Google Scholar] [CrossRef] [PubMed]
- Gillessen, S.; Templeton, A.; Marra, G.; Kuo, Y.-F.; Valtorta, E.; Shahinian, V.B. Risk of Colorectal Cancer in Men on Long-Term Androgen Deprivation Therapy for Prostate Cancer. J. Natl. Cancer Inst. 2010, 102, 1760–1770. [Google Scholar] [CrossRef] [PubMed]
- Fedirko, V.; Tramacere, I.; Bagnardi, V.; Rota, M.; Scotti, L.; Islami, F.; Negri, E.; Straif, K.; Romieu, I.; La Vecchia, C.; et al. Alcohol Drinking and Colorectal Cancer Risk: An Overall and Dose-Response Meta-Analysis of Published Studies. Ann. Oncol. 2011, 22, 1958–1972. [Google Scholar] [CrossRef] [PubMed]
- National Cancer Institute. Surveillance, E. and E.R. Program. Cancer Stat Facts: Colorectal Cancer. Available online: https://seer.cancer.gov/statfacts/html/colorect.html (accessed on 22 September 2023).
- Biller, L.H.; Schrag, D. Diagnosis and Treatment of Metastatic Colorectal Cancer: A Review. JAMA 2021, 325, 669–685. [Google Scholar] [CrossRef] [PubMed]
- Fouad, Y.A.; Aanei, C. Revisiting the Hallmarks of Cancer. Am. J. Cancer Res. 2017, 7, 1016–1036. [Google Scholar]
- Asgari, Y.; Zabihinpour, Z.; Salehzadeh-Yazdi, A.; Schreiber, F.; Masoudi-Nejad, A. Alterations in Cancer Cell Metabolism: The Warburg Effect and Metabolic Adaptation. Genomics 2015, 105, 275–281. [Google Scholar] [CrossRef]
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed]
- Lu, J.; Tan, M.; Cai, Q. The Warburg Effect in Tumor Progression: Mitochondrial Oxidative Metabolism as an Anti-Metastasis Mechanism. Cancer Lett. 2015, 356, 156–164. [Google Scholar] [CrossRef] [PubMed]
- Warburg, O.; Wind, F.; Negelein, E. The Metabolism of Tumors in the Body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef] [PubMed]
- WARBURG, O. On Respiratory Impairment in Cancer Cells. Science 1956, 124, 269–270. [Google Scholar] [CrossRef] [PubMed]
- Warburg, O. On the Origin of Cancer Cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef] [PubMed]
- Vaupel, P.; Multhoff, G. Revisiting the Warburg Effect: Historical Dogma versus Current Understanding. J. Physiol. 2021, 599, 1745–1757. [Google Scholar] [CrossRef] [PubMed]
- Mishra, D.; Banerjee, D. Lactate Dehydrogenases as Metabolic Links between Tumor and Stroma in the Tumor Microenvironment. Cancers 2019, 11, 750. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.; Qiao, L.; Zuo, D.; Qin, D.; Xiao, J.; An, H.; Wang, Y.; Zhang, X.; Jin, Y.; Ren, L. Aberrant Lactate Dehydrogenase A Signaling Contributes Metabolic Signatures in Pancreatic Cancer. Ann. Transl. Med. 2021, 9, 358. [Google Scholar] [CrossRef] [PubMed]
- Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef]
- Rani, R.; Kumar, V. Recent Update on Human Lactate Dehydrogenase Enzyme 5 (HLDH5) Inhibitors: A Promising Approach for Cancer Chemotherapy. J. Med. Chem. 2016, 59, 487–496. [Google Scholar] [CrossRef]
- Claps, G.; Faouzi, S.; Quidville, V.; Chehade, F.; Shen, S.; Vagner, S.; Robert, C. The Multiple Roles of LDH in Cancer. Nat. Rev. Clin. Oncol. 2022, 19, 749–762. [Google Scholar] [CrossRef] [PubMed]
- Forkasiewicz, A.; Dorociak, M.; Stach, K.; Szelachowski, P.; Tabola, R.; Augoff, K. The Usefulness of Lactate Dehydrogenase Measurements in Current Oncological Practice. Cell Mol. Biol. Lett. 2020, 25, 35. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.R.; Jain, S.; Bamezai, R.N.K.; Tiwari, P.K. Utility of Serum LDH Isoforms in the Assessment of Mycobacterium Tuberculosis Induced Pathology in TB Patients of Sahariya Tribe. Indian J. Clin. Biochem. 2010, 25, 57–63. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.-L.; He, Y.; Tam, K.Y. Targeting Cancer Metabolism to Develop Human Lactate Dehydrogenase (HLDH)5 Inhibitors. Drug Discov. Today 2018, 23, 1407–1415. [Google Scholar] [CrossRef] [PubMed]
- Thornburg, J.M.; Nelson, K.K.; Clem, B.F.; Lane, A.N.; Arumugam, S.; Simmons, A.; Eaton, J.W.; Telang, S.; Chesney, J. Targeting Aspartate Aminotransferase in Breast Cancer. Breast Cancer Res. 2008, 10, R84. [Google Scholar] [CrossRef] [PubMed]
- Fiume, L.; Manerba, M.; Vettraino, M.; Di Stefano, G. Impairment of Aerobic Glycolysis by Inhibitors of Lactic Dehydrogenase Hinders the Growth of Human Hepatocellular Carcinoma Cell Lines. Pharmacology 2010, 86, 157–162. [Google Scholar] [CrossRef] [PubMed]
- Qiao, T.; Xiong, Y.; Feng, Y.; Guo, W.; Zhou, Y.; Zhao, J.; Jiang, T.; Shi, C.; Han, Y. Inhibition of LDH-A by Oxamate Enhances the Efficacy of Anti-PD-1 Treatment in an NSCLC Humanized Mouse Model. Front. Oncol. 2021, 11, 632364. [Google Scholar] [CrossRef]
- Zhao, Y.; Liu, H.; Liu, Z.; Ding, Y.; LeDoux, S.P.; Wilson, G.L.; Voellmy, R.; Lin, Y.; Lin, W.; Nahta, R.; et al. Overcoming Trastuzumab Resistance in Breast Cancer by Targeting Dysregulated Glucose Metabolism. Cancer Res. 2011, 71, 4585–4597. [Google Scholar] [CrossRef] [PubMed]
- Zhou, M.; Zhao, Y.; Ding, Y.; Liu, H.; Liu, Z.; Fodstad, O.; Riker, A.I.; Kamarajugadda, S.; Lu, J.; Owen, L.B.; et al. Warburg Effect in Chemosensitivity: Targeting Lactate Dehydrogenase-A Re-Sensitizes Taxol-Resistant Cancer Cells to Taxol. Mol. Cancer 2010, 9, 33. [Google Scholar] [CrossRef]
- Manerba, M.; Di Ianni, L.; Fiume, L.; Roberti, M.; Recanatini, M.; Di Stefano, G. Lactate Dehydrogenase Inhibitors Sensitize Lymphoma Cells to Cisplatin without Enhancing the Drug Effects on Immortalized Normal Lymphocytes. Eur. J. Pharm. Sci. 2015, 74, 95–102. [Google Scholar] [CrossRef]
- Karsli-Uzunbas, G.; Guo, J.Y.; Price, S.; Teng, X.; Laddha, S.V.; Khor, S.; Kalaany, N.Y.; Jacks, T.; Chan, C.S.; Rabinowitz, J.D.; et al. Autophagy Is Required for Glucose Homeostasis and Lung Tumor Maintenance. Cancer Discov. 2014, 4, 914–927. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Su, D.; Zhao, L.; Zhang, D.; Xu, J.; Wan, J.; Fan, S.; Chen, M. Different Effects of LDH-A Inhibition by Oxamate in Non-Small Cell Lung Cancer Cells. Oncotarget 2014, 5, 11886–11896. [Google Scholar] [CrossRef] [PubMed]
- Salgado-García, R.; Coronel-Hernández, J.; Delgado-Waldo, I.; Cantú de León, D.; García-Castillo, V.; López-Urrutia, E.; Gutiérrez-Ruiz, M.C.; Pérez-Plasencia, C.; Jacobo-Herrera, N. Negative Regulation of ULK1 by MicroRNA-106a in Autophagy Induced by a Triple Drug Combination in Colorectal Cancer Cells In Vitro. Genes 2021, 12, 245. [Google Scholar] [CrossRef] [PubMed]
- Hao, H.; Xia, G.; Wang, C.; Zhong, F.; Liu, L.; Zhang, D. MiR-106a Suppresses Tumor Cells Death in Colorectal Cancer through Targeting ATG7. Med. Mol. Morphol. 2017, 50, 76–85. [Google Scholar] [CrossRef] [PubMed]
- Tschan, M.P.; Jost, M.; Batliner, J.; Fey, M.F. The Autophagy Gene ULK1 Plays a Role In AML Differentiation and Is Negatively Regulated by the Oncogenic MicroRNA 106a. Blood 2010, 116, 503. [Google Scholar] [CrossRef]
- Peng, Q.; Shen, Y.; Zhao, P.; Cheng, M.; Zhu, Y.; Xu, B. Biomarker Roles Identification of MiR-106 Family for Predicting the Risk and Poor Survival of Colorectal Cancer. BMC Cancer 2020, 20, 506. [Google Scholar] [CrossRef]
- Goldberg, E.B.; Nitowsky, H.M.; Colowick, S.P. The role of glycolysis in the growth of tumor cells. Iv. The basis of glucose toxicity in oxamate-treated, cultured cells. J. Biol. Chem. 1965, 240, 2791–2796. [Google Scholar] [CrossRef] [PubMed]
- Marchlewski, L. Gossypol, Ein Bestandtheil Der Baumwollsamen. J. Für Prakt. Chem. 1899, 60, 84–90. [Google Scholar] [CrossRef]
- Dodou, K. Investigations on Gossypol: Past and Present Developments. Expert. Opin. Investig. Drugs 2005, 14, 1419–1434. [Google Scholar] [CrossRef]
- Xiong, J.; Li, J.; Yang, Q.; Wang, J.; Su, T.; Zhou, S. Gossypol Has Anti-Cancer Effects by Dual-Targeting MDM2 and VEGF in Human Breast Cancer. Breast Cancer Res. 2017, 19, 27. [Google Scholar] [CrossRef]
- Koppaka, V.; Thompson, D.C.; Chen, Y.; Ellermann, M.; Nicolaou, K.C.; Juvonen, R.O.; Petersen, D.; Deitrich, R.A.; Hurley, T.D.; Vasiliou, V. Aldehyde Dehydrogenase Inhibitors: A Comprehensive Review of the Pharmacology, Mechanism of Action, Substrate Specificity, and Clinical Application. Pharmacol. Rev. 2012, 64, 520–539. [Google Scholar] [CrossRef] [PubMed]
- Flack, M.R.; Pyle, R.G.; Mullen, N.M.; Lorenzo, B.; Wu, Y.W.; Knazek, R.A.; Nisula, B.C.; Reidenberg, M.M. Oral Gossypol in the Treatment of Metastatic Adrenal Cancer. J. Clin. Endocrinol. Metab. 1993, 76, 1019–1024. [Google Scholar] [CrossRef] [PubMed]
- Bushunow, P.; Reidenberg, M.M.; Wasenko, J.; Winfield, J.; Lorenzo, B.; Lemke, S.; Himpler, B.; Corona, R.; Coyle, T. Gossypol Treatment of Recurrent Adult Malignant Gliomas. J. Neurooncol. 1999, 43, 79–86. [Google Scholar] [CrossRef] [PubMed]
- Van Poznak, C.; Seidman, A.D.; Reidenberg, M.M.; Moasser, M.M.; Sklarin, N.; Van Zee, K.; Borgen, P.; Gollub, M.; Bacotti, D.; Yao, T.J.; et al. Oral Gossypol in the Treatment of Patients with Refractory Metastatic Breast Cancer: A Phase I/II Clinical Trial. Breast Cancer Res. Treat. 2001, 66, 239–248. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Liu, H.; Guo, R.; Ling, Y.; Wu, X.; Li, B.; Roller, P.P.; Wang, S.; Yang, D. Molecular Mechanism of Gossypol-Induced Cell Growth Inhibition and Cell Death of HT-29 Human Colon Carcinoma Cells. Biochem. Pharmacol. 2003, 66, 93–103. [Google Scholar] [CrossRef] [PubMed]
- Le, A.; Cooper, C.R.; Gouw, A.M.; Dinavahi, R.; Maitra, A.; Deck, L.M.; Royer, R.E.; Vander Jagt, D.L.; Semenza, G.L.; Dang, C.V. Inhibition of Lactate Dehydrogenase A Induces Oxidative Stress and Inhibits Tumor Progression. Proc. Natl. Acad. Sci. USA 2010, 107, 2037–2042. [Google Scholar] [CrossRef] [PubMed]
- Billiard, J.; Dennison, J.B.; Briand, J.; Annan, R.S.; Chai, D.; Colón, M.; Dodson, C.S.; Gilbert, S.A.; Greshock, J.; Jing, J.; et al. Quinoline 3-Sulfonamides Inhibit Lactate Dehydrogenase A and Reverse Aerobic Glycolysis in Cancer Cells. Cancer Metab. 2013, 1, 19. [Google Scholar] [CrossRef]
- Xian, Z.-Y.; Liu, J.-M.; Chen, Q.-K.; Chen, H.-Z.; Ye, C.-J.; Xue, J.; Yang, H.-Q.; Li, J.-L.; Liu, X.-F.; Kuang, S.-J. Inhibition of LDHA Suppresses Tumor Progression in Prostate Cancer. Tumour Biol. 2015, 36, 8093–8100. [Google Scholar] [CrossRef]
- Rajeshkumar, N.V.; Dutta, P.; Yabuuchi, S.; de Wilde, R.F.; Martinez, G.V.; Le, A.; Kamphorst, J.J.; Rabinowitz, J.D.; Jain, S.K.; Hidalgo, M.; et al. Therapeutic Targeting of the Warburg Effect in Pancreatic Cancer Relies on an Absence of P53 Function. Cancer Res. 2015, 75, 3355–3364. [Google Scholar] [CrossRef]
- Manerba, M.; Vettraino, M.; Fiume, L.; Di Stefano, G.; Sartini, A.; Giacomini, E.; Buonfiglio, R.; Roberti, M.; Recanatini, M. Galloflavin (CAS 568-80-9): A Novel Inhibitor of Lactate Dehydrogenase. ChemMedChem 2012, 7, 311–317. [Google Scholar] [CrossRef]
- Farabegoli, F.; Vettraino, M.; Manerba, M.; Fiume, L.; Roberti, M.; Di Stefano, G. Galloflavin, a New Lactate Dehydrogenase Inhibitor, Induces the Death of Human Breast Cancer Cells with Different Glycolytic Attitude by Affecting Distinct Signaling Pathways. Eur. J. Pharm. Sci. 2012, 47, 729–738. [Google Scholar] [CrossRef] [PubMed]
- Guo, L.; Yang, Y.; Sheng, Y.; Wang, J.; Li, W.; Zhou, X.; Ruan, S.; Han, C. Galloflavin Relieves the Malignant Behavior of Colorectal Cancer Cells in the Inflammatory Tumor Microenvironment. Front. Pharmacol. 2021, 12, 752118. [Google Scholar] [CrossRef]
- Granchi, C.; Calvaresi, E.C.; Tuccinardi, T.; Paterni, I.; Macchia, M.; Martinelli, A.; Hergenrother, P.J.; Minutolo, F. Assessing the Differential Action on Cancer Cells of LDH-A Inhibitors Based on the N-Hydroxyindole-2-Carboxylate (NHI) and Malonic (Mal) Scaffolds. Org. Biomol. Chem. 2013, 11, 6588. [Google Scholar] [CrossRef]
- Rani, R.; Granchi, C. Bioactive Heterocycles Containing Endocyclic N-Hydroxy Groups. Eur. J. Med. Chem. 2015, 97, 505–524. [Google Scholar] [CrossRef] [PubMed]
- Granchi, C.; Roy, S.; De Simone, A.; Salvetti, I.; Tuccinardi, T.; Martinelli, A.; Macchia, M.; Lanza, M.; Betti, L.; Giannaccini, G.; et al. N-Hydroxyindole-Based Inhibitors of Lactate Dehydrogenase against Cancer Cell Proliferation. Eur. J. Med. Chem. 2011, 46, 5398–5407. [Google Scholar] [CrossRef] [PubMed]
- Minutolo, F.; Macchia, M.; Granchi, C.; Roy, S.; Giannaccini, G.; Lucacchini, A. N-Hydroxyazole derivatives as lactate dehydrogenase inhibitors and their preparation and use for the treatment of diseases. WO2011054525, 12 May 2011. [Google Scholar]
- Minutolo, F.; Macchia, M.; Granchi, C.; Di Bussolo, V.; Giannaccini, G.; Lucacchini, A.; Hergenrother, P.J.; Calvaresi, E.C. Preparation of Indole Derivatives as Inhibitors of Enzyme Lactate Dehydrogenase (LDH). WO2013092753, 27 June 2013. [Google Scholar]
- Granchi, C.; Roy, S.; Giacomelli, C.; Macchia, M.; Tuccinardi, T.; Martinelli, A.; Lanza, M.; Betti, L.; Giannaccini, G.; Lucacchini, A.; et al. Discovery of N-Hydroxyindole-Based Inhibitors of Human Lactate Dehydrogenase Isoform A (LDH-A) as Starvation Agents against Cancer Cells. J. Med. Chem. 2011, 54, 1599–1612. [Google Scholar] [CrossRef]
- Daniele, S.; Giacomelli, C.; Zappelli, E.; Granchi, C.; Trincavelli, M.L.; Minutolo, F.; Martini, C. Lactate Dehydrogenase-A Inhibition Induces Human Glioblastoma Multiforme Stem Cell Differentiation and Death. Sci. Rep. 2015, 5, 15556. [Google Scholar] [CrossRef]
- Rai, G.; Brimacombe, K.R.; Mott, B.T.; Urban, D.J.; Hu, X.; Yang, S.-M.; Lee, T.D.; Cheff, D.M.; Kouznetsova, J.; Benavides, G.A.; et al. Discovery and Optimization of Potent, Cell-Active Pyrazole-Based Inhibitors of Lactate Dehydrogenase (LDH). J. Med. Chem. 2017, 60, 9184–9204. [Google Scholar] [CrossRef]
- Rai, G.; Urban, D.J.; Mott, B.T.; Hu, X.; Yang, S.-M.; Benavides, G.A.; Johnson, M.S.; Squadrito, G.L.; Brimacombe, K.R.; Lee, T.D.; et al. Pyrazole-Based Lactate Dehydrogenase Inhibitors with Optimized Cell Activity and Pharmacokinetic Properties. J. Med. Chem. 2020, 63, 10984–11011. [Google Scholar] [CrossRef]
- Chinook Therapeutics, CHK-336 Is a First-In-Class, Liver-Targeted, Oral Small Molecule LDHA Inhibitor Being Developed for the Treatment of Primary and Idiopathic Hyperoxaluria. Available online: https://www.chinooktx.com/pipeline/CHK-336/ (accessed on 20 August 2023).
- Zhou, Y.; Tao, P.; Wang, M.; Xu, P.; Lu, W.; Lei, P.; You, Q. Development of Novel Human Lactate Dehydrogenase A Inhibitors: High-Throughput Screening, Synthesis, and Biological Evaluations. Eur. J. Med. Chem. 2019, 177, 105–115. [Google Scholar] [CrossRef]
- Frati, L. Reconsidering Otto Warburg’s Glycolytic Shift: Pyrimidine Derivatives Are Effective for the Treatment of Tumors Exerting Aerobic Glycolysis. Panminerva Med. 2022, 64, 567–568. [Google Scholar] [CrossRef] [PubMed]
- Frati, L. Pyrimidines Derivatives and Their Use in the Treatment of Tumors. IT202100022682A1, 30 August 2022. Available online: https://patents.google.com/patent/IT202100022682A1/en (accessed on 28 April 2024).
- Di Magno, L.; Coluccia, A.; Bufano, M.; Ripa, S.; La Regina, G.; Nalli, M.; Di Pastena, F.; Canettieri, G.; Silvestri, R.; Frati, L. Discovery of Novel Human Lactate Dehydrogenase Inhibitors: Structure-Based Virtual Screening Studies and Biological Assessment. Eur. J. Med. Chem. 2022, 240, 114605. [Google Scholar] [CrossRef] [PubMed]
- Heinz, S.; Freyberger, A.; Lawrenz, B.; Schladt, L.; Schmuck, G.; Ellinger-Ziegelbauer, H. Mechanistic Investigations of the Mitochondrial Complex I Inhibitor Rotenone in the Context of Pharmacological and Safety Evaluation. Sci. Rep. 2017, 7, 45465. [Google Scholar] [CrossRef] [PubMed]
- Lydic, T.A.; Goo, Y. Lipidomics Unveils the Complexity of the Lipidome in Metabolic Diseases. Clin. Transl. Med. 2018, 7, 4. [Google Scholar] [CrossRef] [PubMed]
- Matsushita, Y.; Nakagawa, H.; Koike, K. Lipid Metabolism in Oncology: Why It Matters, How to Research, and How to Treat. Cancers 2021, 13, 474. [Google Scholar] [CrossRef] [PubMed]
- Röhrig, F.; Schulze, A. The Multifaceted Roles of Fatty Acid Synthesis in Cancer. Nat. Rev. Cancer 2016, 16, 732–749. [Google Scholar] [CrossRef] [PubMed]
- Medes, G.; Thomas, A.; Weinhouse, S. Metabolism of Neoplastic Tissue. IV. A Study of Lipid Synthesis in Neoplastic Tissue Slices in Vitro. Cancer Res. 1953, 13, 27–29. [Google Scholar] [PubMed]
- Currie, E.; Schulze, A.; Zechner, R.; Walther, T.C.; Farese, R.V. Cellular Fatty Acid Metabolism and Cancer. Cell Metab. 2013, 18, 153–161. [Google Scholar] [CrossRef] [PubMed]
- Pakiet, A.; Kobiela, J.; Stepnowski, P.; Sledzinski, T.; Mika, A. Changes in Lipids Composition and Metabolism in Colorectal Cancer: A Review. Lipids Health Dis. 2019, 18, 29. [Google Scholar] [CrossRef]
- Tian, Y.; Xu, T.; Huang, J.; Zhang, L.; Xu, S.; Xiong, B.; Wang, Y.; Tang, H. Tissue Metabonomic Phenotyping for Diagnosis and Prognosis of Human Colorectal Cancer. Sci. Rep. 2016, 6, 20790. [Google Scholar] [CrossRef]
- Butler, L.M.; Perone, Y.; Dehairs, J.; Lupien, L.E.; de Laat, V.; Talebi, A.; Loda, M.; Kinlaw, W.B.; Swinnen, J.V. Lipids and Cancer: Emerging Roles in Pathogenesis, Diagnosis and Therapeutic Intervention. Adv. Drug Deliv. Rev. 2020, 159, 245–293. [Google Scholar] [CrossRef] [PubMed]
- Beloribi-Djefaflia, S.; Vasseur, S.; Guillaumond, F. Lipid Metabolic Reprogramming in Cancer Cells. Oncogenesis 2016, 5, e189. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Puxeddu, M.; Wu, J.; Bai, R.; D’Ambrosio, M.; Nalli, M.; Coluccia, A.; Manetto, S.; Ciogli, A.; Masci, D.; Urbani, A.; et al. Induction of Ferroptosis in Glioblastoma and Ovarian Cancers by a New Pyrrole Tubulin Assembly Inhibitor. J. Med. Chem. 2022, 65, 15805–15818. [Google Scholar] [CrossRef] [PubMed]
- Zaytseva, Y. Lipid Metabolism as a Targetable Metabolic Vulnerability in Colorectal Cancer. Cancers 2021, 13, 301. [Google Scholar] [CrossRef] [PubMed]
- Zaytseva, Y.Y.; Harris, J.W.; Mitov, M.I.; Kim, J.T.; Butterfield, D.A.; Lee, E.Y.; Weiss, H.L.; Gao, T.; Evers, B.M. Increased Expression of Fatty Acid Synthase Provides a Survival Advantage to Colorectal Cancer Cells via Upregulation of Cellular Respiration. Oncotarget 2015, 6, 18891–18904. [Google Scholar] [CrossRef] [PubMed]
- You, M.; Xie, Z.; Zhang, N.; Xiao, D.; Liu, S.; Zhuang, W.; Li, L.; Tao, Y. Signaling pathways in cancer metabolism: Mechanisms and therapeutic targets. Sig Transduct Target Ther 2023, 8, 196. [Google Scholar] [CrossRef] [PubMed]
- Zaytseva, Y.Y.; Elliott, V.A.; Rychahou, P.; Mustain, W.C.; Kim, J.T.; Valentino, J.; Gao, T.; O’Connor, K.L.; Neltner, J.M.; Lee, E.Y.; et al. Cancer Cell-Associated Fatty Acid Synthase Activates Endothelial Cells and Promotes Angiogenesis in Colorectal Cancer. Carcinogenesis 2014, 35, 1341–1351. [Google Scholar] [CrossRef] [PubMed]
- Vance, D.; Goldberg, I.; Mitsuhashi, O.; Bloch, K.; Ōmura, S.; Nomura, S. Inhibition of Fatty Acid Synthetases by the Antibiotic Cerulenin. Biochem. Biophys. Res. Commun. 1972, 48, 649–656. [Google Scholar] [CrossRef]
- Schweizer, E.; Werkmeister, K.; Jain, M.K. Fatty Acid Biosynthesis in Yeast. Mol. Cell Biochem. 1978, 21, 95–107. [Google Scholar] [CrossRef]
- Murata, S.; Yanagisawa, K.; Fukunaga, K.; Oda, T.; Kobayashi, A.; Sasaki, R.; Ohkohchi, N. Fatty Acid Synthase Inhibitor Cerulenin Suppresses Liver Metastasis of Colon Cancer in Mice. Cancer Sci. 2010, 101, 1861–1865. [Google Scholar] [CrossRef]
- Kang, L.; Tian, Y.; Xu, S.; Chen, H. Oxaliplatin-Induced Peripheral Neuropathy: Clinical Features, Mechanisms, Prevention and Treatment. J. Neurol. 2021, 268, 3269–3282. [Google Scholar] [CrossRef] [PubMed]
- Murata, S.; Shiragami, R.; Maruyama, T.; Koda, K.; Ohkohchi, N. Fatty Acid Synthase Inhibitor Cerulenin Suppresses Colorectal Cancer in Combination with Oxaliplatin. Austin J. Surg. 2014, 1, 1011. [Google Scholar]
- Shiragami, R.; Murata, S.; Kosugi, C.; Tezuka, T.; Yamazaki, M.; Hirano, A.; Yoshimura, Y.; Suzuki, M.; Shuto, K.; Koda, K. Enhanced Antitumor Activity of Cerulenin Combined with Oxaliplatin in Human Colon Cancer Cells. Int. J. Oncol. 2013, 43, 431–438. [Google Scholar] [CrossRef] [PubMed]
- Kim, H. Cerulein Pancreatitis: Oxidative Stress, Inflammation, and Apoptosis. Gut Liver 2008, 2, 74–80. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Song, K.-S.; Guo, Q.-X.; Tian, W.-X. The Galloyl Moiety of Green Tea Catechins Is the Critical Structural Feature to Inhibit Fatty-Acid Synthase. Biochem. Pharmacol. 2003, 66, 2039–2047. [Google Scholar] [CrossRef] [PubMed]
- Alshamsan, A.; Hamdy, S.; Samuel, J.; El-Kadi, A.O.S.; Lavasanifar, A.; Uludağ, H. The Induction of Tumor Apoptosis in B16 Melanoma Following STAT3 SiRNA Delivery with a Lipid-Substituted Polyethylenimine. Biomaterials 2010, 31, 1420–1428. [Google Scholar] [CrossRef] [PubMed]
- Zhao, M.; Jiang, B.; Gao, F.-H. Small Molecule Inhibitors of STAT3 for Cancer Therapy. Curr. Med. Chem. 2011, 18, 4012–4018. [Google Scholar] [CrossRef] [PubMed]
- Luo, K.-W.; Xia, J.; Cheng, B.-H.; Gao, H.-C.; Fu, L.-W.; Luo, X.-L. Tea Polyphenol EGCG Inhibited Colorectal-Cancer-Cell Proliferation and Migration via Downregulation of STAT3. Gastroenterol. Rep. 2021, 9, 59–70. [Google Scholar] [CrossRef]
- Chung, S.S.; Vadgama, J.V. Curcumin and Epigallocatechin Gallate Inhibit the Cancer Stem Cell Phenotype via Down-Regulation of STAT3-NFκB Signaling. Anticancer. Res. 2015, 35, 39–46. [Google Scholar]
- Lin, C.-H.; Chao, L.-K.; Hung, P.-H.; Chen, Y.-J. EGCG Inhibits the Growth and Tumorigenicity of Nasopharyngeal Tumor-Initiating Cells through Attenuation of STAT3 Activation. Int. J. Clin. Exp. Pathol. 2014, 7, 2372–2381. [Google Scholar] [PubMed]
- Jin, H.; Gong, W.; Zhang, C.; Wang, S. Epigallocatechin Gallate Inhibits the Proliferation of Colorectal Cancer Cells by Regulating Notch Signaling. Onco Targets Ther. 2013, 6, 145–153. [Google Scholar] [CrossRef] [PubMed]
- Granja, A.; Neves, A.R.; Sousa, C.T.; Pinheiro, M.; Reis, S. EGCG Intestinal Absorption and Oral Bioavailability Enhancement Using Folic Acid-Functionalized Nanostructured Lipid Carriers. Heliyon 2019, 5, e02020. [Google Scholar] [CrossRef] [PubMed]
- Toden, S.; Tran, H.-M.; Tovar-Camargo, O.A.; Okugawa, Y.; Goel, A. Epigallocatechin-3-Gallate Targets Cancer Stem-like Cells and Enhances 5-Fluorouracil Chemosensitivity in Colorectal Cancer. Oncotarget 2016, 7, 16158–16171. [Google Scholar] [CrossRef] [PubMed]
- Shin, C.M.; Lee, D.H.; Seo, A.Y.; Lee, H.J.; Kim, S.B.; Son, W.-C.; Kim, Y.K.; Lee, S.J.; Park, S.-H.; Kim, N.; et al. Green Tea Extracts for the Prevention of Metachronous Colorectal Polyps among Patients Who Underwent Endoscopic Removal of Colorectal Adenomas: A Randomized Clinical Trial. Clin. Nutr. 2018, 37, 452–458. [Google Scholar] [CrossRef] [PubMed]
- Nalli, M.; Masci, D.; Urbani, A.; La Regina, G.; Silvestri, R. Emerging Direct Targeting β-Catenin Agents. Molecules 2022, 27, 7735. [Google Scholar] [CrossRef] [PubMed]
- Pandurangan, A.K.; Esa, N.M. Luteolin, a Bioflavonoid Inhibits Colorectal Cancer through Modulation of Multiple Signaling Pathways: A Review. Asian Pac. J. Cancer Prev. 2014, 15, 5501–5508. [Google Scholar] [CrossRef] [PubMed]
- Wang, X. Pleiotrophin: Activity and Mechanism. In Advances in Clinical Chemistry; Academic Press: London, UK, 2020; pp. 51–89. [Google Scholar]
- Yao, Y.; Rao, C.; Zheng, G.; Wang, S. Luteolin Suppresses Colorectal Cancer Cell Metastasis via Regulation of the MiR-384/Pleiotrophin Axis. Oncol. Rep. 2019, 42, 131–141. [Google Scholar] [CrossRef] [PubMed]
- Horinaka, M.; Yoshida, T.; Shiraishi, T.; Nakata, S.; Wakada, M.; Nakanishi, R.; Nishino, H.; Matsui, H.; Sakai, T. Luteolin Induces Apoptosis via Death Receptor 5 Upregulation in Human Malignant Tumor Cells. Oncogene 2005, 24, 7180–7189. [Google Scholar] [CrossRef]
- Brenner, A.J.; Von Hoff, D.D.; Infante, J.R.; Patel, M.R.; Jones, S.F.; Burris, H.A.; Rubino, C.; McCulloch, W.; Zhukova-Harrill, V.; Kemble, G. First-in-Human Investigation of the Oral First-in-Class Fatty Acid Synthase (FASN) Inhibitor, TVB-2640. J. Clin. Oncol. 2015, 33, TPS2615. [Google Scholar] [CrossRef]
- Falchook, G.; Infante, J.; Arkenau, H.-T.; Patel, M.R.; Dean, E.; Borazanci, E.; Brenner, A.; Cook, N.; Lopez, J.; Pant, S.; et al. First-in-Human Study of the Safety, Pharmacokinetics, and Pharmacodynamics of First-in-Class Fatty Acid Synthase Inhibitor TVB-2640 Alone and with a Taxane in Advanced Tumors. EClinicalMedicine 2021, 34, 100797. [Google Scholar] [CrossRef] [PubMed]
- Lane, A.N.; Fan, T.W.-M. Regulation of Mammalian Nucleotide Metabolism and Biosynthesis. Nucleic Acids Res. 2015, 43, 2466–2485. [Google Scholar] [CrossRef] [PubMed]
- Drury, J.; Rychahou, P.G.; He, D.; Jafari, N.; Wang, C.; Lee, E.Y.; Weiss, H.L.; Evers, B.M.; Zaytseva, Y.Y. Inhibition of Fatty Acid Synthase Upregulates Expression of CD36 to Sustain Proliferation of Colorectal Cancer Cells. Front. Oncol. 2020, 10, 1185. [Google Scholar] [CrossRef] [PubMed]
- Glatz, J.C.; Luiken, J.F. Dynamic Role of the Transmembrane Glycoprotein CD36 (SR-B2) in Cellular Fatty Acid Uptake and Utilization. J. Lipid Res. 2018, 59, 1084–1093. [Google Scholar] [CrossRef] [PubMed]
- Zaytseva, Y.Y.; Rychahou, P.G.; Le, A.-T.; Scott, T.L.; Flight, R.M.; Kim, J.T.; Harris, J.; Liu, J.; Wang, C.; Morris, A.J.; et al. Preclinical Evaluation of Novel Fatty Acid Synthase Inhibitors in Primary Colorectal Cancer Cells and a Patient-Derived Xenograft Model of Colorectal Cancer. Oncotarget 2018, 9, 24787–24800. [Google Scholar] [CrossRef] [PubMed]
- Zu, X.-Y.; Zhang, Q.-H.; Liu, J.-H.; Cao, R.-X.; Zhong, J.; Yi, G.-H.; Quan, Z.-H.; Pizzorno, G. ATP Citrate Lyase Inhibitors as Novel Cancer Therapeutic Agents. Recent. Pat. Anticancer Drug Discov. 2012, 7, 154–167. [Google Scholar] [CrossRef] [PubMed]
- Menendez, J.A.; Lupu, R. Fatty Acid Synthase and the Lipogenic Phenotype in Cancer Pathogenesis. Nat. Rev. Cancer 2007, 7, 763–777. [Google Scholar] [CrossRef] [PubMed]
- Liang, J.-J.; Zhou, X.-F.; Long, H.; Li, C.-Y.; Wei, J.; Yu, X.-Q.; Guo, Z.-Y.; Zhou, Y.-Q.; Deng, Z.-S. Recent advance of ATP citrate lyase inhibitors for the treatment of cancer and related diseases. Bioorganic Chemistry 2024, 142, 106933. [Google Scholar] [CrossRef] [PubMed]
- Berwick, D.C.; Hers, I.; Heesom, K.J.; Moule, S.K.; Tavareá, J.M. The Identification of ATP-Citrate Lyase as a Protein Kinase B (Akt) Substrate in Primary Adipocytes. J. Biol. Chem. 2002, 277, 33895–33900. [Google Scholar] [CrossRef]
- Zhou, Y.; Bollu, L.R.; Tozzi, F.; Ye, X.; Bhattacharya, R.; Gao, G.; Dupre, E.; Xia, L.; Lu, J.; Fan, F.; et al. ATP Citrate Lyase Mediates Resistance of Colorectal Cancer Cells to SN38. Mol. Cancer Ther. 2013, 12, 2782–2791. [Google Scholar] [CrossRef]
- Hatzivassiliou, G.; Zhao, F.; Bauer, D.E.; Andreadis, C.; Shaw, A.N.; Dhanak, D.; Hingorani, S.R.; Tuveson, D.A.; Thompson, C.B. ATP Citrate Lyase Inhibition Can Suppress Tumor Cell Growth. Cancer Cell 2005, 8, 311–321. [Google Scholar] [CrossRef] [PubMed]
- Pearce, N.J.; Yates, J.W.; Berkhout, T.A.; Jackson, B.; Tew, D.; Boyd, H.; Camilleri, P.; Sweeney, P.; Gribble, A.D.; Shaw, A.; et al. The Role of ATP Citrate-Lyase in the Metabolic Regulation of Plasma Lipids. Biochem. J. 1998, 334, 113–119. [Google Scholar] [CrossRef] [PubMed]
- Li, J.J.; Wang, H.; Tino, J.A.; Robl, J.A.; Herpin, T.F.; Lawrence, R.M.; Biller, S.; Jamil, H.; Ponticiello, R.; Chen, L.; et al. 2-Hydroxy-N-Arylbenzenesulfonamides as ATP-Citrate Lyase Inhibitors. Bioorg. Med. Chem. Lett. 2007, 17, 3208–3211. [Google Scholar] [CrossRef] [PubMed]
- Milgraum, L.Z.; Witters, L.A.; Pasternack, G.R.; Kuhajda, F.P. Enzymes of the Fatty Acid Synthesis Pathway Are Highly Expressed in in Situ Breast Carcinoma. Clin. Cancer Res. 1997, 3, 2115–2120. [Google Scholar] [PubMed]
- Yahagi, N.; Shimano, H.; Hasegawa, K.; Ohashi, K.; Matsuzaka, T.; Najima, Y.; Sekiya, M.; Tomita, S.; Okazaki, H.; Tamura, Y.; et al. Co-Ordinate Activation of Lipogenic Enzymes in Hepatocellular Carcinoma. Eur. J. Cancer 2005, 41, 1316–1322. [Google Scholar] [CrossRef] [PubMed]
- Swinnen, J.V.; Heemers, H.; Van de Sande, T.; De Schrijver, E.; Brusselmans, K.; Heyns, W.; Verhoeven, G. Androgens, Lipogenesis and Prostate Cancer. J. Steroid Biochem. Mol. Biol. 2004, 92, 273–279. [Google Scholar] [CrossRef] [PubMed]
- Halvorson, D.L.; McCune, S.A. Inhibition of Fatty Acid Synthesis in Isolated Adipocytes by 5-(Tetradecyloxy)-2-furoic Acid. Lipids 1984, 19, 851–856. [Google Scholar] [CrossRef] [PubMed]
- McCune, S.A.; Harris, R.A. Mechanism Responsible for 5-(Tetradecyloxy)-2-Furoic Acid Inhibition of Hepatic Lipogenesis. J. Biol. Chem. 1979, 254, 10095–10101. [Google Scholar] [CrossRef] [PubMed]
- He, D.; Sun, X.; Yang, H.; Li, X.; Yang, D. TOFA induces cell cycle arrest and apoptosis in ACHN and 786-O cells through inhibiting PI3K/Akt/mTOR pathway. J Cancer 2018, 9, 2734–2742. [Google Scholar] [CrossRef]
- Wang, C.; Xu, C.; Sun, M.; Luo, D.; Liao, D.; Cao, D. Acetyl-CoA Carboxylase-α Inhibitor TOFA Induces Human Cancer Cell Apoptosis. Biochem. Biophys. Res. Commun. 2009, 385, 302–306. [Google Scholar] [CrossRef]
- Li, S.; Qiu, L.; Wu, B.; Shen, H.; Zhu, J.; Zhou, L.; Gu, L.; Di, W. TOFA Suppresses Ovarian Cancer Cell Growth in Vitro and in Vivo. Mol. Med. Rep. 2013, 8, 373–378. [Google Scholar] [CrossRef] [PubMed]
- DeBerardinis, R.J.; Cheng, T. Q’s next: The Diverse Functions of Glutamine in Metabolism, Cell Biology and Cancer. Oncogene 2010, 29, 313–324. [Google Scholar] [CrossRef] [PubMed]
- Dang, C.V. Links between Metabolism and Cancer. Genes. Dev. 2012, 26, 877–890. [Google Scholar] [CrossRef] [PubMed]
- Reitzer, L.J.; Wice, B.M.; Kennell, D. Evidence That Glutamine, Not Sugar, Is the Major Energy Source for Cultured HeLa Cells. J. Biol. Chem. 1979, 254, 2669–2676. [Google Scholar] [CrossRef] [PubMed]
- Martínez-Reyes, I.; Chandel, N.S. Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun. 2020, 3, 102. [Google Scholar] [CrossRef] [PubMed]
- Knox, W.E.; Horowitz, M.L.; Friedell, G.H. The Proportionality of Glutaminase Content to Growth Rate and Morphology of Rat Neoplasms. Cancer Res. 1969, 29, 669–680. [Google Scholar] [PubMed]
- Linder-Horowitz, M.; Knox, W.E.; Morris, H.P. Glutaminase Activities and Growth Rates of Rat Hepatomas. Cancer Res. 1969, 29, 1195–1199. [Google Scholar]
- Yuneva, M.; Zamboni, N.; Oefner, P.; Sachidanandam, R.; Lazebnik, Y. Deficiency in Glutamine but Not Glucose Induces MYC-Dependent Apoptosis in Human Cells. J. Cell Biol. 2007, 178, 93–105. [Google Scholar] [CrossRef]
- Wise, D.R.; DeBerardinis, R.J.; Mancuso, A.; Sayed, N.; Zhang, X.-Y.; Pfeiffer, H.K.; Nissim, I.; Daikhin, E.; Yudkoff, M.; McMahon, S.B.; et al. Myc Regulates a Transcriptional Program That Stimulates Mitochondrial Glutaminolysis and Leads to Glutamine Addiction. Proc. Natl. Acad. Sci. USA 2008, 105, 18782–18787. [Google Scholar] [CrossRef]
- Bhutia, Y.D.; Ganapathy, V. Glutamine Transporters in Mammalian Cells and Their Functions in Physiology and Cancer. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2016, 1863, 2531–2539. [Google Scholar] [CrossRef]
- Yang, L.; Venneti, S.; Nagrath, D. Glutaminolysis: A Hallmark of Cancer Metabolism. Annu. Rev. Biomed. Eng. 2017, 19, 163–194. [Google Scholar] [CrossRef] [PubMed]
- Lobo, C.; Ruiz-Bellido, M.A.; Aledo, J.C.; Márquez, J.; Núñez De Castro, I.; Alonso, F.J. Inhibition of Glutaminase Expression by Antisense MRNA Decreases Growth and Tumourigenicity of Tumour Cells. Biochem. J. 2000, 348 Pt 2, 257–261. [Google Scholar] [CrossRef] [PubMed]
- Gao, P.; Tchernyshyov, I.; Chang, T.-C.; Lee, Y.-S.; Kita, K.; Ochi, T.; Zeller, K.I.; De Marzo, A.M.; Van Eyk, J.E.; Mendell, J.T.; et al. C-Myc Suppression of MiR-23a/b Enhances Mitochondrial Glutaminase Expression and Glutamine Metabolism. Nature 2009, 458, 762–765. [Google Scholar] [CrossRef]
- Katt, W.P.; Cerione, R.A. Glutaminase Regulation in Cancer Cells: A Druggable Chain of Events. Drug Discov. Today 2014, 19, 450–457. [Google Scholar] [CrossRef] [PubMed]
- Xie, C.; Jin, J.; Bao, X.; Zhan, W.-H.; Han, T.-Y.; Gan, M.; Zhang, C.; Wang, J. Inhibition of Mitochondrial Glutaminase Activity Reverses Acquired Erlotinib Resistance in Non-Small Cell Lung Cancer. Oncotarget 2016, 7, 610–621. [Google Scholar] [CrossRef] [PubMed]
- Simpson, N.E.; Tryndyak, V.P.; Beland, F.A.; Pogribny, I.P. An in Vitro Investigation of Metabolically Sensitive Biomarkers in Breast Cancer Progression. Breast Cancer Res. Treat. 2012, 133, 959–968. [Google Scholar] [CrossRef] [PubMed]
- Katt, W.P.; Ramachandran, S.; Erickson, J.W.; Cerione, R.A. Dibenzophenanthridines as Inhibitors of Glutaminase C and Cancer Cell Proliferation. Mol. Cancer Ther. 2012, 11, 1269–1278. [Google Scholar] [CrossRef]
- Wang, J.-B.; Erickson, J.W.; Fuji, R.; Ramachandran, S.; Gao, P.; Dinavahi, R.; Wilson, K.F.; Ambrosio, A.L.B.; Dias, S.M.G.; Dang, C.V.; et al. Targeting Mitochondrial Glutaminase Activity Inhibits Oncogenic Transformation. Cancer Cell 2010, 18, 207–219. [Google Scholar] [CrossRef]
- Yuan, L.; Sheng, X.; Clark, L.H.; Zhang, L.; Guo, H.; Jones, H.M.; Willson, A.K.; Gehrig, P.A.; Zhou, C.; Bae-Jump, V.L. Glutaminase Inhibitor Compound 968 Inhibits Cell Proliferation and Sensitizes Paclitaxel in Ovarian Cancer. Am. J. Transl. Res. 2016, 8, 4265–4277. [Google Scholar]
- Guo, H.; Li, W.; Pan, G.; Wang, C.; Li, D.; Liu, N.; Sheng, X.; Yuan, L. The Glutaminase Inhibitor Compound 968 Exhibits Potent In Vitro and In Vivo Anti-Tumor Effects in Endometrial Cancer. Anticancer Agents Med. Chem. 2023, 23, 210–221. [Google Scholar] [CrossRef]
- Kim, J.H.; Lee, K.J.; Seo, Y.; Kwon, J.-H.; Yoon, J.P.; Kang, J.Y.; Lee, H.J.; Park, S.J.; Hong, S.P.; Cheon, J.H.; et al. Effects of Metformin on Colorectal Cancer Stem Cells Depend on Alterations in Glutamine Metabolism. Sci. Rep. 2018, 8, 409. [Google Scholar] [CrossRef] [PubMed]
- Thangavelu, K.; Pan, C.Q.; Karlberg, T.; Balaji, G.; Uttamchandani, M.; Suresh, V.; Schüler, H.; Low, B.C.; Sivaraman, J. Structural Basis for the Allosteric Inhibitory Mechanism of Human Kidney-Type Glutaminase (KGA) and Its Regulation by Raf-Mek-Erk Signaling in Cancer Cell Metabolism. Proc. Natl. Acad. Sci. USA 2012, 109, 7705–7710. [Google Scholar] [CrossRef] [PubMed]
- Nagana Gowda, G.A.; Barding, G.A.; Dai, J.; Gu, H.; Margineantu, D.H.; Hockenbery, D.M.; Raftery, D. A Metabolomics Study of BPTES Altered Metabolism in Human Breast Cancer Cell Lines. Front. Mol. Biosci. 2018, 5, 49. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.-Y.; Zhang, H.-S.; Liu, M.-Y.; Li, H.-M.; Wang, X.-Y.; Wang, M. GLS1 Depletion Inhibited Colorectal Cancer Proliferation and Migration via Redox/Nrf2/Autophagy-Dependent Pathway. Arch. Biochem. Biophys. 2021, 708, 108964. [Google Scholar] [CrossRef] [PubMed]
- Gross, M.I.; Demo, S.D.; Dennison, J.B.; Chen, L.; Chernov-Rogan, T.; Goyal, B.; Janes, J.R.; Laidig, G.J.; Lewis, E.R.; Li, J.; et al. Antitumor Activity of the Glutaminase Inhibitor CB-839 in Triple-Negative Breast Cancer. Mol. Cancer Ther. 2014, 13, 890–901. [Google Scholar] [CrossRef] [PubMed]
- Varghese, S.; Pramanik, S.; Williams, L.J.; Hodges, H.R.; Hudgens, C.W.; Fischer, G.M.; Luo, C.K.; Knighton, B.; Tan, L.; Lorenzi, P.L.; et al. The Glutaminase Inhibitor CB-839 (Telaglenastat) Enhances the Antimelanoma Activity of T-Cell–Mediated Immunotherapies. Mol. Cancer Ther. 2021, 20, 500–511. [Google Scholar] [CrossRef] [PubMed]
- Hao, Y.; Samuels, Y.; Li, Q.; Krokowski, D.; Guan, B.-J.; Wang, C.; Jin, Z.; Dong, B.; Cao, B.; Feng, X.; et al. Oncogenic PIK3CA Mutations Reprogram Glutamine Metabolism in Colorectal Cancer. Nat. Commun. 2016, 7, 11971. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Feng, X.; Chen, Y.; Selfridge, J.E.; Gorityala, S.; Du, Z.; Wang, J.M.; Hao, Y.; Cioffi, G.; Conlon, R.A.; et al. 5-Fluorouracil Enhances the Antitumor Activity of the Glutaminase Inhibitor CB-839 against PIK3CA -Mutant Colorectal Cancers. Cancer Res. 2020, 80, 4815–4827. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.; Li, D.; Xu, N.; Fang, J.; Yu, Y.; Hou, W.; Ruan, H.; Zhu, P.; Ma, R.; Lu, S.; et al. Novel 1,3,4-Selenadiazole-Containing Kidney-Type Glutaminase Inhibitors Showed Improved Cellular Uptake and Antitumor Activity. J. Med. Chem. 2019, 62, 589–603. [Google Scholar] [CrossRef]
- Li, L.; Meng, Y.; Li, Z.; Dai, W.; Xu, X.; Bi, X.; Bian, J. Discovery and Development of Small Molecule Modulators Targeting Glutamine Metabolism. Eur. J. Med. Chem. 2019, 163, 215–242. [Google Scholar] [CrossRef]
- Ruan, J.J.; Yu, Y.; Hou, W.; Chen, Z.; Fang, J.; Zhang, J.; Ni, M.; Li, D.; Lu, S.; Rui, J.; et al. Kidney-Type Glutaminase Inhibitor Hexylselen Selectively Kills Cancer Cells via a Three-Pronged Mechanism. ACS Pharmacol. Transl. Sci. 2019, 2, 18–30. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.; Sun, G.; Sun, X.; Li, F.; Zhao, L.; Zhong, R.; Peng, Y. The Potential of Lonidamine in Combination with Chemotherapy and Physical Therapy in Cancer Treatment. Cancers 2020, 12, 3332. [Google Scholar] [CrossRef] [PubMed]
- Gourdier, I.; Del Rio, M.; Crabbé, L.; Candeil, L.; Copois, V.; Ychou, M.; Auffray, C.; Martineau, P.; Mechti, N.; Pommier, Y.; et al. Drug Specific Resistance to Oxaliplatin Is Associated with Apoptosis Defect in a Cellular Model of Colon Carcinoma. FEBS Lett. 2002, 529, 232–236. [Google Scholar] [CrossRef] [PubMed]
- Floridi, A.; Bruno, T.; Miccadei, S.; Fanciulli, M.; Federico, A.; Paggi, M.G. Enhancement of Doxorubicin Content by the Antitumor Drug Lonidamine in Resistant Ehrlich Ascites Tumor Cells through Modulation of Energy Metabolism. Biochem. Pharmacol. 1998, 56, 841–849. [Google Scholar] [CrossRef] [PubMed]
- Ben-Yoseph, O.; Lyons, J.C.; Song, C.W.; Ross, B.D. Mechanism of Action of Lonidamine in the 9L Brain Tumor Model Involves Inhibition of Lactate Efflux and Intracellular Acidification. J. Neurooncol. 1998, 36, 149–157. [Google Scholar] [CrossRef] [PubMed]
- Cheng, G.; Zhang, Q.; Pan, J.; Lee, Y.; Ouari, O.; Hardy, M.; Zielonka, M.; Myers, C.R.; Zielonka, J.; Weh, K.; et al. Targeting Lonidamine to Mitochondria Mitigates Lung Tumorigenesis and Brain Metastasis. Nat. Commun. 2019, 10, 2205. [Google Scholar] [CrossRef] [PubMed]
- Guo, L.; Shestov, A.A.; Worth, A.J.; Nath, K.; Nelson, D.S.; Leeper, D.B.; Glickson, J.D.; Blair, I.A. Inhibition of Mitochondrial Complex II by the Anticancer Agent Lonidamine. J. Biol. Chem. 2016, 291, 42–57. [Google Scholar] [CrossRef] [PubMed]
- Zielonka, J.; Joseph, J.; Sikora, A.; Hardy, M.; Ouari, O.; Vasquez-Vivar, J.; Cheng, G.; Lopez, M.; Kalyanaraman, B. Mitochondria-Targeted Triphenylphosphonium-Based Compounds: Syntheses, Mechanisms of Action, and Therapeutic and Diagnostic Applications. Chem. Rev. 2017, 117, 10043–10120. [Google Scholar] [CrossRef] [PubMed]
- Passalacqua, R.; Bisagni, G.; Bertusi, M.; Donati, D.; Buzzi, F.; Di Costanzo, F.; Basurto, C.; Gori, S. Lonidamine in Advanced Colorectal Cancer: A Phase II Study of the Italian Oncology Group for Clinical Research (Goirc). Tumori J. 1989, 75, 277–279. [Google Scholar] [CrossRef]
- Farooque, A.; Afrin, F.; Adhikari, J.; Dwarakanath, B. Protection of Normal Cells and Tissues during Radio- and Chemosensitization of Tumors by 2-Deoxy-D-Glucose. J. Cancer Res. Ther. 2009, 5, 32. [Google Scholar] [CrossRef]
- Park, G.B.; Chung, Y.H.; Kim, D. 2-Deoxy-D-Glucose Suppresses the Migration and Reverses the Drug Resistance of Colon Cancer Cells through ADAM Expression Regulation. Anticancer Drugs 2017, 28, 410–420. [Google Scholar] [CrossRef] [PubMed]
- Seidel, J.; Leitzke, S.; Ahrens, B.; Sperrhacke, M.; Bhakdi, S.; Reiss, K. Role of ADAM10 and ADAM17 in Regulating CD137 Function. Int. J. Mol. Sci. 2021, 22, 2730. [Google Scholar] [CrossRef] [PubMed]
- Kurtoglu, M.; Gao, N.; Shang, J.; Maher, J.C.; Lehrman, M.A.; Wangpaichitr, M.; Savaraj, N.; Lane, A.N.; Lampidis, T.J. Under Normoxia, 2-Deoxy-D-Glucose Elicits Cell Death in Select Tumor Types Not by Inhibition of Glycolysis but by Interfering with N-Linked Glycosylation. Mol. Cancer Ther. 2007, 6, 3049–3058. [Google Scholar] [CrossRef]
- Nenkov, M.; Ma, Y.; Gaßler, N.; Chen, Y. Metabolic Reprogramming of Colorectal Cancer Cells and the Microenvironment: Implication for Therapy. Int. J. Mol. Sci. 2021, 22, 6262. [Google Scholar] [CrossRef] [PubMed]
- Robinson, M.M.; Mcbryant, S.J.; Tsukamoto, T.; Rojas, C.; Ferraris, D.V.; Hamilton, S.K.; Hansen, J.C.; Curthoys, N.P. Novel Mechanism of Inhibition of Rat Kidney-Type Glutaminase by Bis-2-(5-Phenylacetamido-1,2,4-Thiadiazol-2-Yl)Ethyl Sulfide (BPTES). Biochem. J. 2007, 406, 407–414. [Google Scholar] [CrossRef]
Category | Compound | Targets | Ref. |
---|---|---|---|
Oxamate | LDHA, AAT | [36,37,38,39,40] | |
Gossypol | LDH, GA3PDH, MDH, ICDH | [42] | |
FX11 with Daporinad | LDHA, NMPRTase | [49,51] | |
Galloflavin | LDHA, LDHB | [53] | |
Quinoline-3-sulfonamide | LDHA, PKM2 1 | [50] | |
N-Hydroxyindole-2-carboxylate derivatives | hLDH5 | [59,60] | |
NCATS-SM1441 | LDHA, LDHB | [64] | |
CHK-336 | LDHA | [65] | |
Pyran-4-one | LDHA | [66] | |
Pyrimidine-5-carboxamide | LDHA | [68] 2 | |
RS6212 | LDHA | [70] | |
Lipid metabolism | Cerulein | FASN, Akt 1 | [86,87,88] |
Cerulein, oxaliplatin | Induction p53-p21, p38 | [91] | |
(-)-Epigallocatechin gallate | FASN, STAT3 | [93,96] | |
Luteolin | Wnt/β-catenin, PTN | [104,106] | |
TVB-3664 | FASN, Akt 1, Erk1/2 1, AMPK 1 | [108,109,113] | |
SB204990 | ACLy | [119] | |
GSK165 | ACLy | [121] | |
TOFA | ACCA | [126,127] | |
Glutaminolysis | Compd 968 | GLS1 KGA GAC, Rho 1 | [142,146] |
Compd 968, metformin | GLS1, ASCT2 1 | [149] | |
BTES | GLS1 KGA | [151,173] | |
CB-839 | GLS, αKGA | [153] | |
CB-839, capecitabine | GLS, 5-FU target | [156] | |
CDP-3B | KGA, GDH, TrxR, GatCAB | [159] | |
Other | Lonidamine | Hexokinase, complex II | [162,164] |
Mito-lonidamine | Hexokinase, complex I and II | [163,167] | |
2-deoxy-D-glucose | Glycolysis, sCD1371 | [169] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Masci, D.; Puxeddu, M.; Silvestri, R.; La Regina, G. Metabolic Rewiring in Cancer: Small Molecule Inhibitors in Colorectal Cancer Therapy. Molecules 2024, 29, 2110. https://doi.org/10.3390/molecules29092110
Masci D, Puxeddu M, Silvestri R, La Regina G. Metabolic Rewiring in Cancer: Small Molecule Inhibitors in Colorectal Cancer Therapy. Molecules. 2024; 29(9):2110. https://doi.org/10.3390/molecules29092110
Chicago/Turabian StyleMasci, Domiziana, Michela Puxeddu, Romano Silvestri, and Giuseppe La Regina. 2024. "Metabolic Rewiring in Cancer: Small Molecule Inhibitors in Colorectal Cancer Therapy" Molecules 29, no. 9: 2110. https://doi.org/10.3390/molecules29092110
APA StyleMasci, D., Puxeddu, M., Silvestri, R., & La Regina, G. (2024). Metabolic Rewiring in Cancer: Small Molecule Inhibitors in Colorectal Cancer Therapy. Molecules, 29(9), 2110. https://doi.org/10.3390/molecules29092110