2,538
Views
0
CrossRef citations to date
0
Altmetric
Review Article

Glutaminase inhibition as potential cancer therapeutics: current status and future applications

&
Article: 2290911 | Received 01 Aug 2023, Accepted 29 Nov 2023, Published online: 11 Dec 2023

Abstract

Alterations in normal metabolic processes are defining features of cancer. Glutamine, an abundant amino acid in the human blood, plays a critical role in regulating several biosynthetic and bioenergetic pathways that support tumour growth. Glutaminolysis is a metabolic pathway that converts glutamine into various metabolites involved in the tricarboxylic acid (TCA) cycle and generates antioxidants that are vital for tumour cell survival. As glutaminase catalyses the initial step of this metabolic pathway, it is of great significance in cancer metabolism and tumour progression. Inhibition of glutaminase and targeting of glutaminolysis have emerged as promising strategies for cancer therapy. This review explores the role of glutaminases in cancer metabolism and discusses various glutaminase inhibitors developed as potential therapies for tumour regression.

Introduction

Cancer is caused by several alterations in molecular and cellular pathways. Tumour cells undergo neoplastic transformation because of their high energy and building block requirements, which are primarily fulfilled by the oxygen and nutrients available in the tumour microenvironment. Warburg proposed that cancer cells reprogram cellular energy metabolism by modifying aerobic glycolysis to provide increased energy to tumour cells and that aerobic glycolysis is upregulated 200-fold in cancer cells compared with normal cells.Citation1 The Warburg effect results in the conversion of a significant amount of glucose to lactate, which is not utilised in the mitochondrial tricarboxylic acid (TCA) cycle but is used for ATP production.Citation2 Recently, cancer metabolism hallmarks, including the deregulation of glucose and amino acid uptake and the utilisation of alternative nutrient acquisition pathways, were further reviewed.Citation3 Tumour cells upregulate certain amino acid intake, resulting in alternative pathway activation.

Glutamine, an abundant amino acid in blood and muscle, is a crucial source of nitrogen and carbon for lipid, protein, and nucleotide biosynthesis.Citation4 Oncogenes alter the cellular and metabolic transformations during tumorigenesis, highlighting the importance of glutamine in cancer cells. The Warburg effect implies a reduction in the TCA cycle, indicating that tumour cells utilise glutamine as an alternative to carbon and nitrogen sources for cellular biosynthesis.Citation5 Additionally, glutamine deficiency, rather than glucose deficiency, induces Myc-dependent apoptosis in human cells, corroborating the importance of glutamine in tumorigenesis.Citation6 The essential role of glutamine in cancer cell biosynthesis and bioenergetics underscores the need for further studies on glutamine metabolism to improve cancer prognosis and develop agents that target glutamine-related pathways. This review summarises glutamine metabolism in cancer via glutaminases and their structures and provides an overview of glutaminase inhibitors and their potential to induce tumour regression.

Metabolism of glutamine in cancer

In 1990, Lacey et al. challenged the prevailing belief that glutamine is a nonessential amino acid and demonstrated its importance under catabolic stress.Citation7 In tumour cells, the Warburg effect impedes the entry of pyruvate into the TCA cycle. Glutaminolysis involves the use of glutamine as a precursor to activate the TCA cycle by generating α-ketoglutarate intermediates.Citation8

Glutamine plays a vital role as a cellular intermediary metabolite and can act as a carbon or nitrogen donor (α and γ-nitrogen). De novo synthesis of nucleotides requires carbon and nitrogen donor intermediates. With an increased demand for nucleotide synthesis in proliferating cells, glutamine is the preferred carbon source for pyrimidine ring biosynthesis under the hypoxic conditions of tumour cells;Citation9 glutamine can also serve as a nitrogen source in the de novo synthesis of both purine and pyrimidine nucleotides in proliferating cells.Citation10

Reductively metabolised glutamine acts as a major source of carbon for fatty acid synthesis during impaired mitochondrial respiration and hypoxia. Reductive glutamine metabolism is initiated by an increasing α-ketoglutarate/citrate ratio.Citation11 Glutamine also plays a role in acid-base homeostasis in the kidney, redox homeostasis, and reductive oxygen species (ROS) mitigation.Citation12,Citation13

Glutamine plays a critical role in the growth and proliferation of rapidly dividing cells such as immune, gastrointestinal tract, and tumour cells. Glutamine deficiency can cause intestinal mucosal necrosis and apoptosis in human cell lines,Citation6,Citation7 and oral administration of glutamine can improve the quality of life and promote mucosal healing in cancer patients undergoing chemotherapy and radiotherapy.Citation14 Transporters SLC1A5 (ASCT2) and SLC7A5/SLC3A2 mediate glutamine movement across the cell membrane, and the mitochondrial variant of SLC1A5 facilitates glutamine transport across mitochondrial membranes.Citation15,Citation16

Glutaminolysis involves glutamine metabolism in the mitochondria. The first step involves the conversion of glutamine to glutamate by glutaminase (). Glutamate then enters the TCA cycle via glutamate dehydrogenase (GLUD) or contributes to the biosynthesis of nonessential amino acids (NEAAs), such as aspartate, phosphoserine, and alanine, via aminotransferases.Citation8 Glutaminolysis is crucial for maintaining redox homeostasis as it provides glutamate for glutathione (GSH) synthesis.Citation17 The Arf1 GTPase enzyme activates the mTORC1 pathway in the lysosome through glutamine in cancer cells; hence, cancer cells show high glutamine dependence.Citation18 A previous study reviewed the role of glutaminase isoenzymes in cancer metabolic therapy and highlighted various metabolic vulnerabilities in different types of cancers, including the correlation between glutamine metabolism and tumour malignancy.Citation19

Figure 1. Function and structure of human glutaminases. (A) Catalytic function of glutaminase enzyme. (B) Two splice variants of GLS: KGA, the longer isoform with 18 exons, and GAC, the shorter isoform with 15 exons. (C) Two splice variants of GLS2: GAB, the longer isoform with 18 exons and LGA, the shorter isoform with 17 exons. (D, E) Structural domains of the splice variants of GLS and GLS2.

Figure 1. Function and structure of human glutaminases. (A) Catalytic function of glutaminase enzyme. (B) Two splice variants of GLS: KGA, the longer isoform with 18 exons, and GAC, the shorter isoform with 15 exons. (C) Two splice variants of GLS2: GAB, the longer isoform with 18 exons and LGA, the shorter isoform with 17 exons. (D, E) Structural domains of the splice variants of GLS and GLS2.

These findings suggest that glutamine metabolism in cancer cells is essential for tumour growth and progression and paves the way for studying metabolic pathway-targeted therapies for cancer treatment. The following sections provide further discussion on glutamine metabolism by the two isoforms of glutaminase: kidney-type glutaminase (GLS) and liver-type glutaminase (GLS2). These isoforms are encoded by different but related genes. GLS is expressed in various normal tissues, whereas GLS2 is restricted to the brain, pancreas, and liver.Citation20

Glutaminase

Human glutaminase isoenzymes can be categorised into GLS and GLS2 isoforms, each with long and short splice variants. KGA, the long splice variant of GLS, has exons 1–19 with a deletion of exon 15, while the short splice variant has exons 1–15 with a deletion of exons 16–19 (). In contrast, the long splice variant of GLS2 is GAB with exons 1–18 while the short splice variant LGA has exons 2–18 with a deletion of exon 1 ().Citation21 The domains present in these enzymes are signal peptides at the N-terminus, the catalytic site known as the glutaminase domain, and the C-terminus ().Citation22 Expression of these isoforms varies across different regions of the body. The GAC splice variant of GLS is highly expressed in the lungs, placenta, pancreas, and many transformed cells, whereas KGA is expressed in the brain, kidneys, intestines, and immune cells.Citation23 The GLS2 isoform is mainly found in the adult liver; however,Citation24 research has shown its expression in extrahepatic tissues, such as the brain and pancreas.Citation25 At the cellular level, GLS is most likely located in the mitochondria; however, the exact intramitochondrial localisation remains unclear.Citation26 Conversely, GLS2 is localised in the inner mitochondrial membrane.Citation27 Kidney-type glutaminase is involved in maintaining the acid-base balance during metabolic acidosis in the kidney,Citation28 whereas liver-type GLS2 contributes to the provision of nitrogen for the urea cycle in the liver.Citation29 The KGA variant of GLS is responsible for the production of glutamate and GABA for neurotransmission. In the intestinal epithelium, the KGA variant initiates the catabolism of glutamine as the major respiratory fuel.Citation30 In certain cancers, GLS2 is considered to function as a tumour suppressor,Citation31 and its overexpression is associated with poor overall survival. However, further studies are needed to determine the exact role of GLS2 in different cancers. In contrast, recent studies have shown that GLS is frequently overexpressed in several types of cancers.Citation20 The GAC splice variant of GLS is highly expressed in triple-negative breast cancer, acute myeloid leukaemia, and non-small cell lung cancer.Citation32,Citation33 These findings suggest that different glutaminase splice variants play diverse roles in various biological processes and may be involved in the development and progression of different types of cancer.

Regulators of glutaminase

Several factors have been examined to elucidate the regulation of GLS expression. GLS and GLS2 activation is strongly influenced by phosphate concentrations, with high concentrations activating GLS; however, its activity is significantly inhibited by its end product, glutamate. Conversely, GLS2 was active in low phosphate concentrations and was unaffected by the presence of glutamate.Citation34–36 Meanwhile, ammonia activates and inhibits GLS2.Citation20 These results offer novel insights into glutaminase enzyme regulation and indicate that different factors may have distinct effects on GLS and GLS2 activity. Further research is required to elucidate the precise mechanisms underlying these observations and their implications in cellular metabolism.

Glutaminase activity is influenced by oncogenes and epidermal growth factors (EGF). The association of the three kinase components of Rafa-1/Mek2/ERK stimulates KGA.Citation37 Myc, a proto-oncogene and a major regulator of cell proliferation, also plays a crucial role in increasing glutamine uptake and metabolism, including indirect effects on glutaminase activity.Citation38 A recent study summarised the role of Myc in suppressing miR-23a/b expression, leading to increased mitochondrial GLS expression and glutamine metabolism in PC3 prostate cancer cells and P-493B lymphoma cells.Citation39 Additionally, Myc directly binds to the E-box elements of the SLC7A5 and SLC43A1 transporters and enhances the uptake of essential amino acids, which stimulates mitochondrial glutaminolysis, leading to glutamine addiction as a bioenergetic substrate.Citation40,Citation41 In human leukemic Jurkat cells, the NF-κB p65 subunit inhibits miR-23a expression, leading to increased GLS expression.Citation42 Furthermore, the oncogenic transcription factor c-Jun regulates GLS expression.Citation43 These findings highlight the complex interplay between oncogenes and other factors involved in GLS regulation and suggest potential avenues for further research on the role of glutamine metabolism in cancer.

All mammalian cells express three RAS proteins (H, K, and N) that promote oncogenesis; particularly, KRAS mutations are frequently observed in cancer.Citation44 Studies have revealed that the KRAS and Myc oncogenes increase glutamine addiction and glycolysis, which divert glucose from the TCA cycle.Citation45 Additionally, nuclear factor erythroid 2 related factor 2 (NRF2) has been shown to play an important role in the metabolic reprogramming of glutamine in KRAS-mutated cells;Citation46,Citation47 however, the mechanisms underlying glutamine-dependent tumour growth remain largely unknown. In KRAS-mutated non-small cell lung cancer (NSCLC), an increase in KEAP1 mutations activates the KEAP1/NRF2 pathway, leading to the increased expression of genes involved in glutamine metabolism. KRAS mutations result in the loss of serine/threonine kinase 11 (STK11) in NSCLC. The STK11 and KEAP1/NRF2 pathways drive tumour cells to become more sensitive to the glutaminase inhibitor CB-839 in in vivo and in vitro experiments.Citation47 Asparagine synthetase (ASNS) catalyses the formation of L-asparagine from L-aspartic acid, using L-glutamine as the nitrogen source.Citation48 Inhibition of GLS by IACS-6274 induces glutathione (GSH) depletion and altered cell cycle kinetics, owing to intracellular nucleotide pool depletion and DNA damage accumulation.Citation49 Additionally, it was observed that when determining transaminase activity with the IACS-6274 inhibitor, ASNS-low ovarian cell lines showed higher inhibition than ASNS-high cell lines, which showed resistance to the inhibitor.Citation49 Therefore, this study guided to ASNS as a biomarker for GLS inhibitor-based therapeutic regimens.

In many solid tumours, metabolic adaptation to hypoxic conditions is driven by hypoxia-inducible factors (HIF).Citation50 In normal mammalian cells, acetyl coenzyme A, a precursor of fatty acid biosynthesis, is produced from the TCA cycle via glucose-derived pyruvate. However, cell proliferation under hypoxic conditions depends primarily on the reductive carboxylation of glutamine for lipid biosynthesis.Citation51 For example, hypoxia induces GLS expression. Under hypoxic conditions, GLS mRNA and protein expression increases via hypoxia-inducible factor 1 (HIF-1).Citation52 Furthermore, in addition to transcriptional and post-transcriptional modifications, post-translational modifications (PTM) are key factors in GLS activation.Citation37,Citation53 These findings suggest the complex regulation of glutamine metabolism in cancer and highlight the need for further research into its potential therapeutic implications.

In contrast to GLS, GLS2 is a p53 target gene in both normal and tumour cells,Citation54 and human GLS2 contains a p53 consensus DNA-binding element.Citation55 In addition to p53, TAp63, and TAp73 regulate GLS2 expression.Citation56,Citation57 Taken together, these findings highlight the importance of the glutaminase-mediated metabolism of glutamine and its regulation in different cancers. Target-based therapies targeting glutamine-related metabolic pathways may represent a promising strategy for cancer treatment. provides an overview of regulators involved in glutaminase-mediated glutamine metabolism in cancer cells. This review provides a comprehensive understanding of the regulators of glutamine metabolism and their potential targets for cancer therapeutic interventions.

Figure 2. Cancer metabolism involving glutamine. Cancer cells transport glutamine through ASCT2. Glutamine thus entered is converted to glutamate through GLS and GLS2. The glutamate formed then moves into TCA cycle and supports the biosynthesis of nucleotides, proteins, and lipids. The regulators of glutaminase are marked in pink (Permission granted by Wang et al., 2020).

Figure 2. Cancer metabolism involving glutamine. Cancer cells transport glutamine through ASCT2. Glutamine thus entered is converted to glutamate through GLS and GLS2. The glutamate formed then moves into TCA cycle and supports the biosynthesis of nucleotides, proteins, and lipids. The regulators of glutaminase are marked in pink (Permission granted by Wang et al., 2020).

Inhibitors of glutaminase

Cancer cells rely on different metabolic pathways to maintain their unique characteristics such as unchecked cell proliferation, autonomous growth signalling, and resistance to apoptosis. Glutaminolysis plays a vital role in cancer development and is an attractive therapeutic target. The initial step in glutaminolysis is the conversion of glutamine to glutamate by glutaminase, making it a valuable target for cancer therapy. Inhibition of glutamine metabolism leads to a deficiency in glutathione (GSH) levels and the accumulation of mitochondrial ROS (mitoROS), ultimately leading to apoptotic cell death.Citation58–60 Additionally, combination therapy with glutaminase inhibitors and other chemotherapeutic agents has been proven to be effective in inhibiting tumour cell growth in ovarian and pancreatic cancers.Citation46,Citation60,Citation61 These findings highlight the potential of targeting glutaminases in cancer therapy. Several glutaminase inhibitors have been identified and classified based on their binding sites. Inhibitors that target active or catalytic sites are called orthosteric/competitive inhibitors, whereas those that target other sites are called allosteric inhibitors.

The immunosuppressive environment of tumours must be altered to achieve effective antitumor effects. The blockade of glutamine metabolism by an antagonist results in a decrease in hypoxia, acidosis, and nutrient depletion. However, this antagonism results in the upregulation of the oxidative metabolism of effector T cells, which leads to the formation of a highly activated phenotype of T effector cells.Citation62 The divergent roles of glutaminase inhibitors make this target an important point of study. Glutaminase overexpression is a critical factor in tumorigenesis and tumour progression and has been proposed to be a prognostic biomarker for various cancers, including ovarian cancer, osteosarcoma, hepatocellular carcinoma, colorectal cancer, and breast cancer.Citation45 Glutaminase inhibition in these tumours can induce two divergent roles, thereby resulting in an effective antitumor effect. illustrates various inhibitors identified as anticancer agents and their respective timelines of discovery.

Figure 3. Various GLS inhibitors and the respective timeline of their discoveries. The orthosteric or competitive inhibitors, which include DON, acivicin, JHU-083 and DRP-104, are mentioned in the red boxes. BPTES, CB-839, IACS-6274, Compound 13b and Compound 13, which belongs to the class of allosteric inhibitors, are mentioned in the pink boxes. Compound 968 which is a GLS2 selective molecule is mentioned in the blue box.

Figure 3. Various GLS inhibitors and the respective timeline of their discoveries. The orthosteric or competitive inhibitors, which include DON, acivicin, JHU-083 and DRP-104, are mentioned in the red boxes. BPTES, CB-839, IACS-6274, Compound 13b and Compound 13, which belongs to the class of allosteric inhibitors, are mentioned in the pink boxes. Compound 968 which is a GLS2 selective molecule is mentioned in the blue box.

Cancer therapy and GLS modulators

  • Orthosteric/competitive inhibitors

    Orthosteric/competitive inhibitors are also referred to as glutamine mimetics because they interact with the active site of the enzyme through covalent binding. Numerous potent orthosteric inhibitors have been developed, including DON, DRP-104, JHU-083, and acivicin (). The glutamine mimetics DON and acivicin have been isolated from Streptomyces.Citation22 DON and acivicin are glutamine antagonists that inhibit multiple glutamine/glutamate-binding enzymes, leading to severe toxicity in clinical trials due to their non-selective inhibition. To overcome DON toxicity, a prodrug strategy has been developed for efficient drug transfer to tumour tissues.Citation63 DRP-104, a newly synthesised DON prodrug, provided greater exposure to tumour tissues and exhibited a better tolerance profile.Citation64 Other DON prodrugs include JHU-083, Nedelcovych-13d, and Rais-5C.Citation22,Citation65–67 Several crystal structures of human glutaminases with L-glutamate have been reported (PDBs: 3CZD, 3UO9, 3UNW, and 3SS5), which have aided in the development of these orthosteric inhibitors.

  • Allosteric Inhibitors

    Unlike glutamine mimetics, this class of inhibitors selectively targets glutaminases without disrupting other aspects of glutamine metabolism. The allosteric site, located in the solvent region of the enzyme, is distinct from the catalytic site.Citation22 Glutaminases exist in two forms: an inactive dimeric state and an active tetrameric state that triggers glutaminolysis.Citation68 The allosteric pocket, located at the interface of the GLS dimers, is hydrophobic and symmetric and allows symmetrical molecules, such as BPTES, to occupy this regionCitation27 (). Crystal structures (PDBs 3UO9, 5HL1, 3VP1, 3VOZ, 5JYO, and 4JKT) Citation28,Citation37,Citation68–70 have been used to study the allosteric binding pocket and its interactions, leading to extensive development of inhibitors at this site. It has been shown that the interaction of a ligand at the interface of the dimer triggers a significant conformational change at the key loop near the catalytic site, rendering the enzyme inactive.Citation37 BPTES is an early allosteric inhibitor that was developed to specifically target kidney-type glutaminases.Citation71 In vitro studies have shown its high efficiency and specificity in inhibiting cancer cell proliferation; however, poor bioavailability and aqueous solubility prevent the use of BPTES.Citation22 To overcome these limitations, modified BPTES derivatives have been developed. CB-839 and IACS-6274 () are more potent and orally bioavailable and have been tested in clinical trials.Citation32,Citation72 In addition, IACS-6274 exhibits excellent pharmacokinetic and physicochemical properties and is currently undergoing phase I clinical trials for solid tumours.Citation72

Macrocycles have also been reported to inhibit GLS because they are designed based on existing crystal structures of GLS. Compound 13b shows an activity similar to that of the small molecule CB-839 on GLS.Citation73 Studied on ether-linker macrocycles as GLS inhibitors resulted in the discovery of compound 13.Citation74 Further studies on this category of inhibitors are required to identify potential therapeutic agents targeting GLS. lists different GLS inhibitors and their current developmental stages.

Figure 4. Crystal structure of hKGA showing the allosteric and active sites. GLS forms a tetramer and the interface of dimers forms the allosteric site. Glutamine is the substrate for the active site and BPTES, a GLS inhibitor, occupies the allosteric site (Permission granted by the American Chemical Society from L. Chen & Cui, 2015).

Figure 4. Crystal structure of hKGA showing the allosteric and active sites. GLS forms a tetramer and the interface of dimers forms the allosteric site. Glutamine is the substrate for the active site and BPTES, a GLS inhibitor, occupies the allosteric site (Permission granted by the American Chemical Society from L. Chen & Cui, 2015).

Table 1. GLS inhibitors under development.

Resistance of glutaminase inhibitors

Despite the significant efficacy of GLS inhibitors, it has been observed that certain cancer cell lines eventually develop resistance to these inhibitors. Resistance is often attributed to compensatory pathways that allow tumour cells to evade glutaminase dependence. A study revealed that the treatment of pancreatic cells with a GLS inhibitor suppressed the proliferation of tumour cells in the early phase; however, the proliferation of these tumour cells continued both in vivo and in vitro. This change in the proliferation of these cell lines may be due to adaptive metabolic networks.Citation75 Notably, some altered pathways included the upregulation of supply pathways for the TCA cycle, which resulted in less dependence on glutamine.Citation76 Other pathways for glutaminase inhibition resistance include fatty acid oxidation (FAO) and mTORC1 signalling.Citation75 These compensatory pathways should be targeted for effective treatment with a glutaminase inhibitor. This has led to various combination therapy trials that showed a synergizing effect in different cancer models. Another study reported that combination therapy with GLS and PARP inhibitors could effectively treat chemo-resistant ovarian cancer.Citation77 Several target agents, such as erlotinib (EGFR inhibitor),Citation78 metformin (glycolysis inhibitor),Citation79 glutor (glucose uptake inhibitor),Citation80 and MLN128 (mTOR1 inhibitor),Citation81 in combination with glutaminase inhibitors, have shown synergistic effects regarding activity. shows the inhibitors targeting multiple pathways that resulted in the reduction of the TCA cycle, thereby causing tumour suppression as the energy and building blocks required for growth were blocked.

Figure 5. Glutaminase inhibitor resistance through alternate metabolic pathways. Targeting glutaminase and alternate metabolic pathways prevent the TCA cycle, which results in the deprivation of energy and building blocks required for cell growth. Inhibitors are marked in red and the alternate pathways involved in resistance of GLS inhibitors is marked in purple. (Re-drawn with permission from Wang et al., 2020).

Figure 5. Glutaminase inhibitor resistance through alternate metabolic pathways. Targeting glutaminase and alternate metabolic pathways prevent the TCA cycle, which results in the deprivation of energy and building blocks required for cell growth. Inhibitors are marked in red and the alternate pathways involved in resistance of GLS inhibitors is marked in purple. (Re-drawn with permission from Wang et al., 2020).

Mechanistic studies of CB-839 have indicated that myeloma cells undergo anti-proliferative effects due to caspase-dependent autophagy.Citation82 A phase I clinical trial of CB-839 showed that CB-839 monotherapy was well tolerated and efficacious in a subset of patients,Citation83 and a combination therapy with CB-839 and standard chemotherapeutic agents was also studied in a phase I trial (NCT02071862) that showed encouraging clinical activity and tolerability. In a recent phase 1 clinical study (NCT05039801) of the IACS-6274 for the treatment of advanced solid tumours, IACS-6274 was found to have a half-life of 7.56 h, with plasma exposure showing a dose-dependent increase across dosage cohorts. IACS-6274 showed a robust pharmacokinetic and pharmacodynamic relationship across doses and patients (P < 0.001). DRP-104, an orthosteric inhibitor, is currently undergoing a phase 2 clinical trial for the treatment of solid tumours (NCT04471415). These findings suggest that GLS is a viable target for cancer therapy; however, more data are required to evaluate the efficacy of glutaminase inhibitors in clinical settings.

Cancer therapy and GLS2 modulators

GLS2, in contrast to GLS, has been shown to have a tumour-suppressive role. Nuclear translocation of GLS2 in cancer cells was shown to cause cell proliferation arrest and differentiation.Citation84 GLS2, a target gene of p53, suppresses tumour growth through the regulation of antioxidant function, resulting in different ROS levels in cells.Citation85 In contrast, overexpression of GLS2 resulted in tumour proliferation and metastasis in breast cancer.Citation86 GLS2 knockdown also sensitises cervical cancer cells to ionising radiation and reduces glutathione and NADH levels, indicating that GLS2 may be a therapeutic target in certain cancers.Citation87 Additionally, compound 968 was reported as a potential target for glutaminase activation in transformed cancer cells.Citation88 Subsequently, the sensitivity of compound 968 to GLS2 was studied, and it was demonstrated to have moderate selectivity for GLS2 over GLS.Citation89 Furthermore, alkyl benzoquinones inhibit mTORC1 and reduce cell proliferation. These compounds exhibit selectivity for GLS2 over GLS because of the presence of specific amino acid residues in the allosteric pocket.Citation90 In summary, the conflicting roles of GLS2 make it challenging to target it as an anticancer agent, and further investigation is needed to fully understand the role of GLS2 in different cancers.

Further applications

In addition to cancer, irregularities in glutamine metabolism are defining features of metabolic disorders, including obesity, diabetes, and non-alcoholic fatty liver disease.Citation91 By modifying these metabolic pathways and decreasing the associated conditions, glutaminase inhibitors have the potential to offer a therapeutic approach for diverse metabolic disorders. By targeting glutamine metabolism, these inhibitors may aid in re-establishing the metabolic balance and improving patient outcomes.

In addition, disrupted glutamine metabolism has been linked to neurological disorders such as Alzheimer’s and Parkinson’s diseases.Citation92,Citation93 It was demonstrated that neuronal excitotoxicity is caused by increased glutamate concentrations, indicating the need for glutaminase suppression in Alzheimer’s disease.Citation94 The increase in neuronal glutaminase activity is considered the primary reason for the upregulation of glutamate in the cerebral compartments during Parkinson’s disease,Citation95 indicating the need for therapeutic intervention targeting glutaminases in this disease. Glutaminase inhibitors offer a potential intervention strategy for these conditions by regulating glutamine levels and reducing neurotoxicity. However, the precise mechanisms and therapeutic benefits of these inhibitors in neurological diseases require further investigation.

Glutaminase inhibitors also hold promise for immunological regulation, presenting an exciting avenue due to their potential impact.Citation96 It was noted that glutamine metabolism in immune cells could undergo reprogramming, resulting in cells changing their phenotypes and functions.Citation97 These alterations in function and phenotype can contribute to pro- and anti-tumorigenic effects. These inhibitors may modulate immune responses by affecting glutamine-dependent immune cell function. This modulation may be beneficial in conditions characterised by autoimmune response and chronic inflammation, in which immunological dysregulation plays a significant role. A study demonstrated that blocking GLS using an inhibitor could potentially be a new treatment strategy for systematic lupus erythematosus, an autoimmune disease.Citation98

The applications of glutaminase inhibitors in medicine are highly promising and span multiple fields. From cancer treatment to metabolic disorders, neurological conditions, immune modulation, and organ transplantation, these inhibitors present a novel approach to tackling intricate health issues. Ongoing research and clinical trials will play a vital role in fully understanding and harnessing the capabilities of these inhibitors, ultimately paving the way for innovative therapies.

Discussion

Cancer is characterised by a metabolic shift that leads to uncontrolled cell proliferation and increased energy production and metabolite synthesis. Glutamine, an abundant amino acid in the human blood, plays a vital role in regulating biosynthesis and bioenergetic pathways in tumour growth. In addition, glutamine contributes to the production of antioxidants via glutaminolysis in tumour cells. Herein, we examine the role of glutaminolysis in cancer and the activity of glutaminase isoenzymes in this process. Pharmacological inhibition of glutaminases has shown significant activity against several cancers. Clinical trial molecules, including CB-839, IACS-6274, and DRP-104, suggest that inhibition of glutaminases can have a significant impact on tumour cells in terms of cell proliferation and apoptosis. Combination therapy with these inhibitors and other anticancer agents has also shown synergistic effects that can help overcome the resistance to glutaminase inhibitors. Identifying cancers with upregulated glutaminolysis and studying combination therapies are interesting methods for the treatment of certain cancers. The optimisation of glutaminase inhibitors with enhanced efficacy and favourable pharmacokinetic properties may represent a promising therapeutic strategy for inhibiting tumour growth. Combination therapy comprising glutaminase inhibitors and inhibitors targeting compensatory pathways may prove to be an effective treatment for cancers and other diseases that depend on glutamine. Recent studies have revealed the role of glutaminase in neural diseases and the need for glutaminase inhibitors with good permeability through the blood-brain barrier has become a necessity. The multiple roles of glutaminase make it an interesting target for further study, and the development of glutaminase inhibitors with enhanced efficacy and favourable pharmacokinetic properties can play an important role in the treatment of different diseases.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This research was supported by the Korea Drug Development Fund funded by the Ministry of Science and ICT; the Ministry of Trade, Industry, and Energy; the Ministry of Health and Welfare [HN21C1180]; and the Korea Research Institute of Chemical Technology [KK2331-20]. The authors thank Editage (www.editage.co.kr) for English language editing.

References

  • Warburg O. On the origin of cancer cells. Science. 1956;123(3191):309–314.
  • Maurer PH, Klevens HB, Ellenbogen E. On respiratory impairment in cancer cells. Science. 1954;24:267–268.
  • Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016;23(1):27–47.
  • Mayers JR, Vander Heiden MG. Famine versus feast: Understanding the metabolism of tumors in vivo. Trends Biochem Sci. 2015;40(3):130–140.
  • Gaglio D, Metallo CM, Gameiro PA, Hiller K, Danna LS, Balestrieri C, Alberghina L, Stephanopoulos G, Chiaradonna F. Oncogenic K-Ras decouples glucose and glutamine metabolism to support cancer cell growth. Mol Syst Biol. 2011;7(1):523.
  • 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(1):93–105.
  • Lacey JM, Wilmore DW. Is glutamine a conditionally essential amino acid? Nutr Rev. 2009;48(8):297–309.
  • Hosios AM, Hecht VC, Danai LV, Johnson MO, Rathmell JC, Steinhauser ML, Manalis SR, Vander Heiden MG. Amino acids rather than glucose account for the majority of cell mass in proliferating mammalian cells. Dev Cell. 2016;36(5):540–549.
  • Wang Y, Bai C, Ruan Y, Liu M, Chu Q, Qiu L, Yang C, Li B. Coordinative metabolism of glutamine carbon and nitrogen in proliferating cancer cells under hypoxia. Nat Commun. 2019;10(1):201.
  • Lane AN, Fan TWM. Regulation of mammalian nucleotide metabolism and biosynthesis. Nucleic Acids Res. 2015;43(4):2466–2485.
  • Fendt SM, Bell EL, Keibler MA, Olenchock BA, Mayers JR, Wasylenko TM, Vokes NI, Guarente L, Heiden MGV, Stephanopoulos G. Reductive glutamine metabolism is a function of the α-ketoglutarate to citrate ratio in cells. Nat Commun. 2013;4(1):2236.
  • Weiner ID, Hamm LL. Molecular mechanisms of renal ammonia transport. Annu Rev Physiol. 2007;69(1):317–340.
  • Jiang L, Shestov AA, Swain P, Yang C, Parker SJ, Wang QA, Terada LS, Adams ND, McCabe MT, Pietrak B, et al. Reductive carboxylation supports redox homeostasis during anchorage-independent growth. Nature. 2016;532(7598):255–258.
  • Anderson PM, Lalla RV. Glutamine for amelioration of radiation and chemotherapy associated mucositis during cancer therapy. Nutrients. 2020;12(6):1675.
  • Stine ZE, Dang CV. Glutamine skipping the Q into mitochondria. Trends Mol Med. 2020;26(1):6–7.
  • Yoo HC, Park SJ, Nam M, Kang J, Kim K, Yeo JH, Kim JK, Heo Y, Lee HS, Lee MY, et al. A variant of SLC1A5 is a mitochondrial glutamine transporter for metabolic reprogramming in cancer cells. Cell Metab. 2020;31(2):267–283.e12.
  • Jin L, Alesi GN, Kang S. Glutaminolysis as a target for cancer therapy. Oncogene. 2016;35(28):3619–3625.
  • Jewell JL, Kim YC, Russell RC, Yu F-X, Park HW, Plouffe SW, Tagliabracci VS, Guan K-L. Differential regulation of mTORC1 by leucine and glutamine. Science. 2015;347(6218):194–198.
  • Matés JM, Campos-Sandoval JA, Márquez J. Glutaminase isoenzymes in the metabolic therapy of cancer. Biochim Biophys Acta Rev Cancer. 2018;1870(2):158–164.
  • Saha SK, Riazul Islam SM, Abdullah-Al-Wadud M, Islam S, Ali F, Park KS. Multiomics analysis reveals that GLS and GLS2 differentially modulate the clinical outcomes of cancer. J Clin Med. 2019;8(3):355.
  • Katt WP, Lukey MJ, Cerione RA. A tale of two glutaminases: homologous enzymes with distinct roles in tumorigenesis. Future Med Chem. 2017;9(2):223–243.
  • Xu X, Meng Y, Li L, Xu P, Wang J, Li Z, Bian J. Overview of the development of glutaminase inhibitors: achievements and future directions. J Med Chem. 2019;62(3):1096–1115.
  • Hartwick EW, Curthoys NP. BPTES inhibition of hGA124-551, a truncated form of human kidney-type glutaminase. J Enzyme Inhib Med Chem. 2012;27(6):861–867.
  • Shukla K, Ferraris DV, Thomas AG, Stathis M, Duvall B, Delahanty G, Alt J, Rais R, Rojas C, Gao P, et al. Design, synthesis, and pharmacological evaluation of bis-2-(5- phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide 3 (BPTES) analogs as glutaminase inhibitors. J Med Chem. 2012;55(23):10551–10563.
  • Gómez-Fabre PM, Aledo JC, Del Castillo-Olivares A, Alonso FJ, Núñez De Castro I, Campos JA, Márquez J. Molecular cloning, sequencing and expression studies of the human breast cancer cell glutaminase. Biochem J. 2000;345 Pt 2(Pt 2):365–375. PMID:10620514
  • Katt WP, Cerione RA. Glutaminase regulation in cancer cells: a druggable chain of events. Drug Discov Today. 2014;19(4):450–457.
  • Chen L, Cui H. Targeting glutamine induces apoptosis: A cancer therapy approach. Int J Mol Sci. 2015;16(9):22830–22855.
  • Delabarre B, Gross S, Fang C, Gao Y, Jha A, Jiang F, Song J J, Wei W, Hurov JB. Full-length human glutaminase in complex with an allosteric inhibitor. Biochemistry. 2011;50(50):10764–10770.
  • Zhou Y, Eid T, Hassel B, Danbolt NC. Novel aspects of glutamine synthetase in ammonia homeostasis. Neurochem Int. 2020;140:104809.
  • Elgadi KM, Meguid RA, Qian M, Souba WW, Abcouwer SF. Cloning and analysis of unique human glutaminase isoforms generated by tissue-specific alternative splicing. Physiol Genomics. 1999;1(2):51–62.
  • Liu J, Zhang C, Lin M, Zhu W, Liang Y, Hong X, Zhao Y, Young KH, Hu W, Feng Z. Glutaminase 2 negatively regulates the PI3K/AKT signaling and shows tumor suppression activity in human hepatocellular carcinoma. Oncotarget. 2014;5(9):2635–2647.
  • Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, Janes JR, Laidig GJ, Lewis ER, Li J, et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13(4):890–901.
  • Jacque N, Ronchetti AM, Larrue C, Meunier G, Birsen R, Willems L, Saland E, Decroocq J, Maciel TT, Lambert M, et al. Targeting glutaminolysis has antileukemic activity in acute myeloid leukemia and synergizes with BCL-2 inhibition. Blood. 2015;126(11):1346–1356.
  • Krebs HA. 1980. Glutamine metabolism in the animal body. In: Mora J, editor. Glutamine: Metabolism, enzymology, and regulation. 1st ed. New York: Academic Press.
  • Kovacevic Z, McGivan JD. Mitochondrial metabolism of glutamine and glutamate and its physiological significance. Physiol Rev. 1983;63(2):547–605.
  • Campos-Sandoval JA, López de la Oliva AR, Lobo C, Segura JA, Matés JM, Alonso FJ, Márquez J. Expression of functional human glutaminase in baculovirus system: Affinity purification, kinetic and molecular characterization. Int J Biochem Cell Biol. 2007;39(4):765–773.
  • Thangavelu K, Pan CQ, Karlberg T, Balaji G, Uttamchandani M, Suresh V, Schüler H, Low BC, 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 U S A. 2012;109(20):7705–7710.
  • Daye D, Wellen KE. Metabolic reprogramming in cancer: Unraveling the role of glutamine in tumorigenesis. Semin Cell Dev Biol. 2012;23(4):362–369.
  • Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, Zeller KI, De Marzo AM, Van Eyk JE, Mendell JT, et al. C-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458(7239):762–765.
  • Yue M, Jiang J, Gao P, Liu H, Qing G. Oncogenic MYC activates a feedforward regulatory loop promoting essential amino acid metabolism and tumorigenesis. Cell Rep. 2017;21(13):3819–3832.
  • Wise DR, Deberardinis RJ, Mancuso A, Sayed N, Zhang X-Y, Pfeiffer HK, Nissim I, Daikhin E, Yudkoff M, Mcmahon SB, et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc Natl Acad Sci U S A. 2008;105(48):18782–18787.
  • Rathore MG, Saumet A, Rossi JF, De Bettignies C, Tempé D, Lecellier CH, Villalba M. The NF-κB member p65 controls glutamine metabolism through miR-23a. Int J Biochem Cell Biol. 2012;44(9):1448–1456.
  • Lukey MJ, Greene KS, Erickson JW, Wilson KF, Cerione RA. The oncogenic transcription factor c-Jun regulates glutaminase expression and sensitizes cells to glutaminase-targeted therapy. Nat Commun. 2016;7(1):11321.
  • Prior IA, Lewis PD, Mattos C. A comprehensive survey of ras mutations in cancer. Cancer Res. 2012;72(10):2457–2467.
  • Wang Z, Liu F, Fan N, Zhou C, Li D, Macvicar T, Dong Q, Bruns CJ, Zhao Y. Targeting glutaminolysis: new perspectives to understand cancer development and novel strategies for potential target therapies. Front Oncol. 2020;10:589508.
  • Mukhopadhyay S, Goswami D, Adiseshaiah PP, Burgan W, Yi M, Guerin TM, Kozlov SV, Nissley DV, McCormick F. Undermining glutaminolysis bolsters chemotherapy while NRF2 promotes chemoresistance in KRAS-driven pancreatic cancers. Cancer Res. 2020;80(8):1630–1643.
  • Galan-Cobo A, Sitthideatphaiboon P, Qu X, Poteete A, Pisegna MA, Tong P, Chen PH, Boroughs LK, Rodriguez MLM, Zhang W, et al. LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence inKRAS-mutant lung adenocarcinoma. Cancer Res. 2019;79(13):3251–3267.
  • Zhu W, Radadiya A, Bisson C, Nordin BE, Baumann P, Imasaki T, Wenzel SA, Sedelnikova SE, Berry AH, Nomanbhoy TK, et al. Molecular basis of human asparagine synthetase inhibitor specificity. bioRxiv. 2018;PPR:PPR57405.
  • Spencer ND, Bristow CA, Giulani V, Miller MA, Carugo A, Harris AL, Minelli R, Feng N, Chang Q, Soth MJ, et al. Abstract 87: Asparagine synthetase (ASNS) expression predicts response to the GLS1 inhibitor IPN60090 in ovarian cancer through selective modulation of redox homeostasis. Cancer Res. 2021;81(13_Supplement):87–87.
  • Sormendi S, Wielockx B. Hypoxia pathway proteins as central mediators of metabolism in the tumor cells and their microenvironment. Front Immunol. 2018;9:40.
  • Metallo CM, Gameiro PA, Bell EL, Mattaini KR, Yang J, Hiller K, Jewell CM, Johnson ZR, Irvine DJ, Guarente L, et al. Reductive glutamine metabolism by IDH1 mediates lipogenesis under hypoxia. Nature. 2012;481(7381):380–384.
  • Xiang L, Mou J, Shao B, Wei Y, Liang H, Takano N, Semenza GL, Xie G. Glutaminase 1 expression in colorectal cancer cells is induced by hypoxia and required for tumor growth, invasion, and metastatic colonization. Cell Death Dis. 2019;10(2):40.
  • Wilson KF, Erickson JW, Antonyak MA, Cerione RA. Rho GTPases and their roles in cancer metabolism. Trends Mol Med. 2013;19(2):74–82.
  • Suzuki S, Tanaka T, Poyurovsky MV, Nagano H, Mayama T, Ohkubo S, Lokshin M, Hosokawa H, Nakayama T, Suzuki Y, et al. Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc Natl Acad Sci U S A. 2010;107(16):7461–7466.
  • Chen JQ, Russo J. Dysregulation of glucose transport, glycolysis, TCA cycle and glutaminolysis by oncogenes and tumor suppressors in cancer cells. Biochim Biophys Acta Rev Cancer. 2012;1826(2):370–384.
  • Arianna G, Bongiorno-Borbone L, Bernassola F, Terrinoni A, Markert E, Levine AJ, Fen Z, Agostini M, Zolla L, Finazzi Agro’ A, et al. P63 regulates glutaminase 2 expression. Cell Cycle. 2013;12(9):1395–1405.
  • Velletri T, Romeo F, Tucci P, Peschiaroli A, Annicchiarico-Petruzzelli M, Niklison-Chirou MV, Amelio I, Knight RA, Mak TW, Melino G, et al. GLS2 is transcriptionally regulated by p73 and contributes to neuronal differentiation. Cell Cycle. 2013;12(22):3564–3573.
  • Gregory MA, Nemkov T, Park HJ, Zaberezhnyy V, Gehrke S, Adane B, Jordan CT, Hansen KC, D'Alessandro A, DeGregori J. Targeting glutamine metabolism and redox state for leukemia therapy. Clin Cancer Res. 2019;25(13):4079–4090.
  • Le A, Lane AN, Hamaker M, Bose S, Gouw A, Barbi J, Tsukamoto T, Rojas CJ, Slusher BS, Zhang H, et al. Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Cell Metab. 2012;15(1):110–121.
  • Yuan L, Sheng X, Clark LH, Zhang L, Guo H, Jones HM, Willson AK, Gehrig PA, Zhou C, Bae-Jump VL. Original Article Glutaminase inhibitor compound 968 inhibits cell proliferation and sensitizes paclitaxel in ovarian cancer. Am J Transl Res. 2016;8(10):4265–4277. PMID:27830010
  • Masamha CP, LaFontaine P. Molecular targeting of glutaminase sensitizes ovarian cancer cells to chemotherapy. J Cell Biochem. 2018;119(7):6136–6145.
  • Leone RD, Zhao L, Englert JM, Sun I-M, Oh M-H, Sun I-H, Arwood ML, Bettencourt IA, Patel CH, Wen J, et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science. 2019;366(6468):1013–1021.
  • Lemberg KM, Vornov JJ, Rais R, Slusher BS. We’re not “don” yet: Optimal dosing and prodrug delivery of 6-diazo-5-oxo-L-norleucine. Mol Cancer Ther. 2018;17(9):1824–1832.
  • Rais R, Lemberg KM, Tenora L, Arwood ML, Pal A, Alt J, Wu Y, Lam J, Marie J, Aguilar H, et al. Discovery of DRP-104, a tumor-targeted metabolic inhibitor prodrug. Sci Adv. 2022;8(46):eabq5925.
  • Hanaford AR, Alt J, Rais R, Wang SZ, Kaur H, Thorek DLJ, Eberhart CG, Slusher BS, Martin AM, Raabe EH. Orally bioavailable glutamine antagonist prodrug JHU-083 penetrates mouse brain and suppresses the growth of MYC-driven medulloblastoma. Transl Oncol. 2019;12(10):1314–1322.
  • Rais R, Jančařík A, Tenora L, Nedelcovych M, Alt J, Englert J, Rojas C, Le A, Elgogary A, Tan J, et al. Discovery of 6-Diazo-5-oxo- l -norleucine (DON) prodrugs with enhanced CSF delivery in monkeys: A potential treatment for glioblastoma. J Med Chem. 2016;59(18):8621–8633.
  • Nedelcovych MT, Tenora L, Kim BH, Kelschenbach J, Chao W, Hadas E, Jančařík A, Prchalová E, Zimmermann SC, Dash RP, et al. N-(Pivaloyloxy)alkoxy-carbonyl prodrugs of the glutamine antagonist 6-Diazo-5-oxo- l -norleucine (DON) as a potential treatment for HIV associated neurocognitive disorders. J Med Chem. 2017;60(16):7186–7198.
  • Curthoys NP, Watford M. Regulation of glutaminase activity and glutamine metabolism. Annu Rev Nutr. 1995;15(1):133–159.
  • Ramachandran S, Qiurong Pan C, Zimmermann SC, Duvall B, Tsukamoto T, Low BC, Sivaraman J. Structural basis for exploring the allosteric inhibition of human kidney type glutaminase. Oncotarget. 2016;7(36):57943–57954.
  • McDermott LA, Iyer P, Vernetti L, Rimer S, Sun J, Boby M, Yang T, Fioravanti M, O'Neill J, Wang L, et al. Design and evaluation of novel glutaminase inhibitors. Bioorg Med Chem. 2016;24(8):1819–1839.
  • Robinson MM, McBryant SJ, Tsukamoto T, Rojas C, Ferraris DV, Hamilton SK, Hansen JC, Curthoys NP. 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(3):407–414.
  • Soth MJ, Le K, Di Francesco ME, Hamilton MM, Liu G, Burke JP, Carroll CL, Kovacs JJ, Bardenhagen JP, Bristow CA, et al. Discovery of IPN60090, a clinical stage selective Glutaminase-1 (GLS-1) inhibitor with excellent pharmacokinetic and physicochemical properties. J Med Chem. 2020;63(21):12957–12977.
  • Xu X, Wang J, Wang M, Yuan X, Li L, Zhang C, Huang H, Jing T, Wang C, Tong C, et al. Structure-enabled discovery of novel macrocyclic inhibitors targeting glutaminase 1 allosteric binding site. J Med Chem. 2021;64(8):4588–4611.
  • Lee EJ, Duggirala KB, Lee Y, Yun MR, Jang J, Cyriac R, Jung ME, Choi G, Chae CH, Cho BC, et al. Novel allosteric glutaminase 1 inhibitors with macrocyclic structure activity relationship analysis. Bioorg Med Chem Lett. 2022;75:128956.
  • Biancur DE, Paulo JA, Małachowska B, Del Rey MQ, Sousa CM, Wang X, Sohn ASW, Chu GC, Gygi SP, Harper JW, et al. Compensatory metabolic networks in pancreatic cancers upon perturbation of glutamine metabolism. Nat Commun. 2017;8(1):15965.
  • Méndez-Lucas A, Lin W, Driscoll PC, Legrave N, Novellasdemunt L, Xie C, Charles M, Wilson Z, Jones NP, Rayport S, et al. Identifying strategies to target the metabolic flexibility of tumours. Nat Metab. 2020;2(4):335–350.
  • Shen YA, Hong J, Asaka R, Asaka S, Hsu FC, Rahmanto YS, Jung JG, Chen YW, Yen TT, Tomaszewski A, et al. Inhibition of the MYC-regulated glutaminase metabolic axis is an effective synthetic lethal approach for treating chemoresistant ovarian cancers. Cancer Res. 2021;80(20):4514–4526.
  • Momcilovic M, Bailey ST, Lee JT, Fishbein MC, Magyar C, Braas D, Graeber T, Jackson NJ, Czernin J, Emberley E, et al. Targeted inhibition of EGFR and glutaminase induces metabolic crisis in EGFR mutant lung cancer. Cell Rep. 2017;18(3):601–610.
  • Elgogary A, Xu Q, Poore B, Alt J, Zimmermann SC, Zhao L, Fu J, Chen B, Xia S, Liu Y, et al. Combination therapy with BPTES nanoparticles and metformin targets the metabolic heterogeneity of pancreatic cancer. Proc Natl Acad Sci U S A. 2016;113(36):E5328–E5336.
  • Reckzeh ES, Karageorgis G, Schwalfenberg M, Ceballos J, Nowacki J, Stroet MCM, Binici A, Knauer L, Brand S, Choidas A, et al. Inhibition of glucose transporters and glutaminase synergistically impairs tumor cell growth. Cell Chem Biol. 2019;26(9):1214–1228.e25.
  • Momcilovic M, Bailey ST, Lee JT, Fishbein MC, Braas D, Go J, Graeber TG, Parlati F, Demo S, Li R, et al. The GSK3 signaling axis regulates adaptive glutamine metabolism in lung squamous cell carcinoma. Cancer Cell. 2018;33(5):905–921.e5.
  • Das DS, Ravillah D, Ray A, Song Y, Munshi NC, Richardson PG, Chauhan D, Anderson KC. Anti-myeloma activity of a novel glutaminase inhibitor CB-839. Blood. 2014;124(21):3439–3439.
  • Meric-Bernstam F, Tannir NM, Mier JW, DeMichele A, Telli ML, Fan AC, Munster PN, Carvajal RD, Orford KW, Bennett MK, et al. Phase 1 study of CB-839, a small molecule inhibitor of glutaminase (GLS), alone and in combination with everolimus (E) in patients (pts) with renal cell cancer (RCC). JCO. 2016;34(15_suppl):4568–4568.
  • López de la Oliva AR, Campos-Sandoval JA, Gómez-García MC, Cardona C, Martín-Rufián M, Sialana FJ, Castilla L, Bae N, Lobo C, Peñalver A, et al. Nuclear translocation of glutaminase GLS2 in human cancer cells associates with proliferation arrest and differentiation. Sci Rep. 2020;10(1):2259.
  • Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci U S A. 2010;107(16):7455–7460.
  • Dias MM, Adamoski D, dos Reis LM, Ascenção CFR, de Oliveira KRS, Mafra ACP, da Silva Bastos AC, Quintero M, de G. Cassago C, Ferreira IM, et al. GLS2 is protumorigenic in breast cancers. Oncogene. 2020;39(3):690–702.
  • Xiang L, Xie G, Liu C, Zhou J, Chen J, Yu S, Li J, Pang X, Shi H, Liang H. Knock-down of glutaminase 2 expression decreases glutathione, NADH, and sensitizes cervical cancer to ionizing radiation. Biochim Biophys Acta. 2013;1833(12):2996–3005.
  • Wang J-B, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, Wilson KF, Ambrosio ALB, Dias SMG, Dang CV, et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18(3):207–219.
  • Lukey MJ, Cluntun AA, Katt WP, Lin MJ, Druso JE, Ramachandran S, Erickson JW, Le HH, Wang Z-E, Blank B, et al. Liver-type Glutaminase GLS2 is a druggable metabolic node in luminal-subtype breast cancer. Cell Rep. 2019;29(1):76–88.e7.
  • Lee Y-Z, Yang C-W, Chang H-Y, Hsu H-Y, Chen I-S, Chang H-S, Lee C-H, Lee J, Ramesh Kumar C, Qiu Y-Q, et al. Discovery of selective inhibitors of Glutaminase-2, which inhibit mTORC1, activate autophagy and inhibit proliferation in cancer cells. Oncotarget. 2014;5(15):6087–6101.
  • Stine ZE, Schug ZT, Salvino JM, Dang CV. Targeting cancer metabolism in the era of precision oncology. Nat Rev Drug Discov. 2022;21(2):141–162.
  • Talantova M, Sanz-Blasco S, Zhang X, Xia P, Akhtar MW, Okamoto S, Dziewczapolski G, Nakamura T, Cao G, Pratt AE, et al. Aβ induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss. Proc Natl Acad Sci U S A. 2013;110(27):E2518–E2527.
  • Deb S, Dutta A, Phukan BC, Manivasagam T, Justin Thenmozhi A, Bhattacharya P, Paul R, Borah A. Neuroprotective attributes of L-theanine, a bioactive amino acid of tea, and its potential role in Parkinson’s disease therapeutics. Neurochem Int. 2019;129:104478.
  • Bayraktar A, Li X, Kim W, Zhang C, Turkez H, Shoaie S, Mardinoglu A. Drug repositioning targeting glutaminase reveals drug candidates for the treatment of Alzheimer’s disease patients. J Transl Med. 2023;21(1):332.
  • Chassain C, Bielicki G, Donnat J, Renou J, Eschalier A, Durif F. Cerebral glutamate metabolism in Parkinson’s disease: an in vivo dynamic C NMS study in the rat. Exp Neurol. 2005;191(2):276–284.
  • Jin J, Byun J-K, Choi Y-K, Park K-G. Targeting glutamine metabolism as a therapeutic strategy for cancer. Exp Mol Med. 2023;55(4):706–715.
  • Ma G, Zhang Z, Li P, Zhang Z, Zeng M, Liang Z, Li D, Wang L, Chen Y, Liang Y, et al. Reprogramming of glutamine metabolism and its impact on immune response in the tumor microenvironment. Cell Commun Signal. 2022;20(1):114.
  • Kono M, Yoshida N, Maeda K, Suárez‐Fueyo A, Kyttaris VC, Tsokos GC. Glutaminase 1 inhibition reduces glycolysis and ameliorates lupus‐like disease in mrl/lpr mice and experimental autoimmune encephalomyelitis. Arthritis Rheumatol. 2019;71(11):1869–1878.