Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Nov 15, 2023; 15(11): 1852-1863
Published online Nov 15, 2023. doi: 10.4251/wjgo.v15.i11.1852
Glutamine addiction and therapeutic strategies in pancreatic cancer
Lin-Lin Ren, Tao Mao, Pin Meng, Hong-Yun Wei, Zi-Bin Tian, Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
Li Zhang, Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
ORCID number: Lin-Lin Ren (0000-0002-0735-4958); Tao Mao (0000-0003-1635-5335); Zi-Bin Tian (0000-0002-3211-2934).
Author contributions: Ren LL wrote the manuscript; Ren LL, Meng P, Zhang L and Wei HY performed the literature retrieval; Tian ZB and Mao T revised the manuscript; all authors have read and approve the final manuscript.
Supported by the National Natural Science Foundation of China, No. 81602056 and No. 82273393; the Natural Science Foundation of Shandong Province, No. ZR2016HQ45 and No. ZR2020LZL004; and the Shandong Traditional Chinese Medicine Science and Technology Project, No. 2021M161.
Conflict-of-interest statement: The authors declare no conflict of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Tao Mao, Doctor, MD, PhD, Associate Chief Physician, Doctor, Teacher, Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, Shandong Province, China. maotao@qdu.edu.cn
Received: June 27, 2023
Peer-review started: June 27, 2023
First decision: August 25, 2023
Revised: September 6, 2023
Accepted: October 23, 2023
Article in press: October 23, 2023
Published online: November 15, 2023

Abstract

Pancreatic cancer remains one of the most lethal diseases worldwide owing to its late diagnosis, early metastasis, and poor prognosis. Because current therapeutic options are limited, there is an urgent need to investigate novel targeted treatment strategies. Pancreatic cancer faces significant metabolic challenges, principally hypoxia and nutrient deprivation, due to specific microenvironmental constraints, including an extensive desmoplastic stromal reaction. Pancreatic cancer cells have been shown to rewire their metabolism and energy production networks to support rapid survival and proliferation. Increased glucose uptake and glycolytic pathway activity during this process have been extensively described. However, growing evidence suggests that pancreatic cancer cells are glutamine addicted. As a nitrogen source, glutamine directly (or indirectly via glutamate conversion) contributes to many anabolic processes in pancreatic cancer, including amino acids, nucleobases, and hexosamine biosynthesis. It also plays an important role in redox homeostasis, and when converted to α-ketoglutarate, glutamine serves as an energy and anaplerotic carbon source, replenishing the tricarboxylic acid cycle intermediates. The present study aims to provide a comprehensive overview of glutamine metabolic reprogramming in pancreatic cancer, focusing on potential therapeutic approaches targeting glutamine metabolism in pancreatic cancer.

Key Words: Pancreatic cancer, Glutamine metabolism, Cancer treatment, Therapeutic strategies

Core Tip: Most pancreatic ductal adenocarcinomas (PDAC) are diagnosed at an advanced stage, missing the opportunity for surgical treatment and responding poorly to radiotherapy and targeted therapies. Glutamine is a non-essential amino acid that is found in high levels in normal humans. Glutamine metabolism could provide raw material for the synthesis of important molecules and meet the needs of rapid growth and proliferation of tumor cells. The study of glutamine metabolic pathways targeting PDAC may provide new strategies for pancreatic cancer treatment.



INTRODUCTION

Pancreatic cancer is a lethal malignancy with a five-year survival rate of 3%-5%[1,2]. This high mortality rate can be mainly attributed to late disease presentation when patients are no longer candidates for surgical resection[3]. Moreover, because pancreatic cancer responds poorly to radiotherapy, chemotherapy and targeted therapy, new treatment methods are urgently needed to improve the current treatment dilemma. The proliferation of pancreatic connective tissue and severe blood supply deficiency results in local tumor cells being exposed to a microenvironment of nutrient deprivation, hypoxia, and high oxidative stress.

Cancer cells must undergo metabolic reprogramming and reshape their energy metabolic pathways to meet the energy requirements for metabolism, proliferation, and growth, which are primarily manifested as increased aerobic glycolysis of glucose, active catabolism of glutamine, and enhanced autophagy[4,5]. Glutamine is believed to have a significant role in pancreatic cancer cells as it undergoes deamination to α-ketoglutarate (aKG) to support the tricarboxylic acid (TCA) cycle via glutaminolysis, using glutamine as a fuel source[6].

Glutamine is a non-essential amino acid with a high content in the normal human body that can provide a nitrogen source for the synthesis of purine and pyrimidine nucleotides, participate in the TCA cycle, and contribute to its carbon backbone, particularly when carbon is diverted to glycolytic pathways. Glutamine also promotes essential amino acid uptake, activates the mammalian target of rapamycin (mTOR), aids in the recycling of excess ammonia and glutamate, and regulates redox homeostasis[7-9]. Relevant studies have confirmed that glutamine demand is significantly increased in pancreatic cancer, and "Glutamine Addiction" exists in tumor cells[10-12]. Glutamine metabolism produces energy and is an important synthetic raw material for rapid tumor cell growth and proliferation.

In addition, the NADPH and glutathione produced during metabolism can help tumor cells maintain redox homeostasis[13]. Targeting specific metabolic pathways in cancer cells, particularly the glutamine pathway, may be a novel approach to cancer, according to a number of recent studies[14-16]. In this review, we systematically summarize the metabolic status of glutamine in pancreatic cancer, as well as the research progress in targeting the glutamine metabolic pathway in pancreatic cancer treatment.

METABOLISM OF GLUTAMINE IN THE HUMAN BODY

Glutamine accounts for nearly 4.7% of the amino acids that make up human proteins. In some special functional proteins, such as the protein involved in forming the epidermal barrier structure, the proportion can reach 25%. Because glutamine is in high demand in the body, it is also known as the conditionally essential amino acid[17]. The initial production of glutamine takes place in in the skeletal muscle, in the pulmonary, and in the adipose tissue. This process is coordinated by glutamate ammonia ligase (GLUL), a cytosolic enzyme that catalyzes glutamine synthesis from glutamate and ammonium ions (NH4+). Glutaminase (GLS) located in mitochondria is responsible for catalyzing the conversion of glutamine into glutamate in intestinal mucosa, immune cells, renal tubule cells, and liver[18].

Glutamine is not able to cross the plasma membrane and has to be transported into the cells by transporters. There are 14 transporters in total, which are divided into four families: SLC1, SLC6, SLC7 and SLC38[19]. When glutamine enters the cell, it is incorporated into various metabolic pathways as a nitrogen or carbon source. As a nitrogen source, glutamine mainly synthesizes nucleotides (pyrimidine and purine), non-essential amino acids (NEAAs), and glucosamine. For DNA and RNA, however, de novo nucleotide synthesis is essential. It is supported by glutamine, which together with nitrogen, is involved in the production of purines and pyrimidines[20,21]. The carbon in glutamine is used as a carbon source in gluconeogenesis, TCA cycle, and glutathione metabolism.

Gluconeogenesis is an important process for maintaining glucose homeostasis under various conditions, and glutamine is a major gluconeogenic precursor[22]. To maintain redox homeostasis, NADPH and reduced glutathione molecules are primarily responsible for neutralizing reactive oxygen species (ROS). The reaction catalyzed by cysteine ligase produces γ-glutamylcysteine from glutamate and cysteine in the first step of glutathione synthesis. And Gln is a vital substrate in this first step[23]. Other NEAAs can be synthesized from Gln-derived glutamate, including alanine, aspartate, proline, serine, and acetyl-serine[24].

DYSREGULATION OF GLUTAMINE METABOLISM IN PANCREATIC CANCER

Glutamine is more abundant in rapidly proliferating tumor cells and some common cells, such as intestinal epithelial and immune cells. This cell-intrinsic regulation of nutrient partitioning is accomplished through the mTORC1 signaling pathway and the expression of genes involved in glucose and glutamine metabolism[24]. Numerous studies have described the dysregulation of glutamine metabolism in various cancer contexts[25-34] after Roberts et al[25] described the first observation of enhanced glutamine metabolism.

The molecules involved in glutamine metabolism dysregulation differ between cancer types. Alanine, serine, cysteine-preferring transporter 2 (ASCT2), GLUL, and GLS genes related to glutamine metabolism are highly expressed in triple-negative breast cancer patients[35]. The expression of the GLS II pathway in prostate cancer cells increased with tumor cell aggressiveness[36]. Similarly, glutamine metabolism changes are observed in pancreatic cancer patients[5,37-41]. The amino acid transporter SLC6A14 was significantly overexpressed in pancreatic cancer cells, tissues, and patient-derived xenografts in comparison to normal pancreatic tissue or normal pancreatic epithelial cells.

α-Methyltryptophan, a SLC6A14 pharmacological blocker, decreased pancreatic cancer cell growth in vitro and in vivo by inducing amino acid starvation[37]. The decreased blood vessel density in pancreatic cancer favors its resistance to chemotherapy and contributes to its aggressiveness due to the high degree of hypoxia[42,43]. Glutamine is metabolized by hypoxic pancreatic tumor cells in order to survive. The hexosamine biosynthetic pathway links glucose and glutamine metabolism, allowing the protein modification of O-linked N-acetylglucosamine. Hypoxia increases hexosamine biosynthetic pathway gene transcription and O-glycosylated protein levels, and the O-linked N-acetylglucosaminylation of proteins is necessary for the survival of hypoxic pancreatic cancer cells[38].

Cancer cell progression is inhibited by cellular senescence, which causes cell cycle arrest in damaged or dysfunctional cells[44]. The generation of NADPH and maintenance of the cellular redox state in pancreatic cancer depend on glutamine carbon flowing through mitochondrial aspartate transaminase (GOT2). Elevated ROS following GOT2 downregulation can produce p27-mediated senescence, a cyclin-dependent kinase inhibitor[40].

NON-CANONICAL GLUTAMINE METABOLIC PATHWAY IN PANCREATIC CANCER

Pancreatic cancer growth requires glucose. Furthermore, pancreatic cancer cells are highly sensitive to glutamine deprivation, indicating that Gln is important for pancreatic cancer growth. Pancreatic cancer can be significantly inhibited by impaired GLS activity[39]. To replenish TCA cycle metabolites, glutamine can be converted into αKG via two mechanisms: glutamate dehydrogenase (GLUD1) or transaminases[45]. In contrast, most cells use GLUD1 in the mitochondria to convert Gln-derived glutamate to αKG to fuel the TCA cycle[46,47]. Pancreatic cancer is dependent on a specific pathway for the fuel of the TCA cycle, in which aspartate derived from glutamine is transported to the cytoplasm where it is processed to oxaloacetate (OAA) by aspartate transaminase (GOT1). This OAA is then converted to malate and pyruvate, which is thought to increase the ratio of NADPH to NADP+ and possibly maintain the redox state of the cell. The viability of pancreatic cancer cells is significantly reliant on this sequence of reactions, as the knockdown of any component enzyme in this series significantly suppresses pancreatic cancer growth in vitro and in vivo[39] (Figure 1).

Figure 1
Figure 1 Metabolism of glutamine in pancreatic cancer. Glutamine enters the cells through four receptors (SLC1, SLC6, SLC7, and SLC38) and can be incorporated into various metabolic pathways either as a nitrogen source or as a carbon source, which is important for the synthesis of nucleotides, non-essential amino acids, glucosamine and gluconeogenesis, the tricarboxylic acid (TCA) cycle, and glutathione metabolism. Under normal physiological conditions, glutamine can be converted to α-ketoglutarate by canonical pathway to replenish the TCA cycle metabolites, which is differ from the non-canonical pathway of glutamine using in pancreatic cancer. TCA: Tricarboxylic acid; αKG: α-ketoglutarate; GOT1: Aspartate transaminase; OAA: Oxaloacetate; GLUD1: Glutamate dehydrogenase; NEAAs: Non-essential amino acids.
GLUTAMINE METABOLIC REPROGRAMMING IN PANCREATIC CANCER

Several studies have found that oncogenic alterations in cancer cells reprogram glutamine metabolism[39]. The transcriptional regulatory capabilities of the oncogene Myc facilitate the coordination of gene expression necessary for cells to participate in glutamine catabolism, which exceeds the demands of cancer cells for protein and nucleotide biosynthesis. The reliance of mitochondrial metabolic reprogramming on glutamine catabolism for the maintenance of cellular viability and replenishment of the TCA cycle results from Myc-dependent glutaminolysis. And PI3K or AKT activation is not required for the ability of Myc-expressing cells to engage in glutaminolysis.

Transcriptional binding of the proto-oncogene c-MYC to the promoter regions of high-affinity glutamine importers, including the sodium-dependent neutral amino acid transporter type 2 and its isoform, results in enhanced glutamine uptake and reduced contribution of glucose to mitochondrial-dependent phospholipid synthesis[48].

Kras mutations are a common type of mutation in pancreatic cancer, with KrasG12D being the most common oncogenic form[49]. Loss of KrasG12D heterozygosity (KrasG12D-LOH) leads to the increasement of proliferation and invasion capacity in pancreatic cancer cells[50].

As a stress-response gene, regulated in DNA damage and development 1 (REDD1) is expressed in response to hypoxic condition, nutrient deficiency, and other stress factors. Moreover, REDD1 is a vital upstream regulator of the mTOR[51], which is a serine/threonine protein kinase involved in non-canonical anaplerotic glutamine metabolism[52]. KrasG12D-LOH increased non-canonical glutamine metabolism and REDD1 expression. Moreover, REDD1 knockdown inhibited KrasG12D-LOH-induced upregulation of Gln metabolism, effectively impairing KrasG12D-LOH pancreatic cancer cell growth, motility, and invasion[53]. The oncogenic Kras mutation reactivates glutamine metabolism, increases the production of reduced NADPH, and restores cellular redox homeostasis through macromolecule synthesis.

To produce metabolic precursors for NADPH production, mitochondrial glutamine-derived aspartic acid must be transported into the cytoplasm. Mitochondrial uncoupling protein 2 (UCP2) catalyzes the transportation process and significantly promotes tumor growth. Compared to the wild-type cell lines, UCP2-silenced Kras mutant cell lines have lower glutaminolysis, NADPH/NADP+, and glutathione/glutathione disulfide ratios and higher ROS levels. Furthermore, the growth of Kras mutant pancreatic cancer cells was strongly inhibited by UCP2 silencing in vitro and in vivo[54].

p53 is an important tumor suppressor that orchestrates specific cellular responses to various stress signals associated with tumor suppression, including transient cell cycle arrest, cellular senescence, and cell apoptosis. Other important cellular processes, including metabolism, maintenance of the stem cell, tumor invasion and metastasis, and communication within the tumor microenvironment, have also been reported to be under p53 regulation[55]. The metabolic functions of p53 help cells adapt to and survive limited periods of metabolic stress by resisting the switch to glycolysis in cancers[56].

GLS2, a p53 target gene, encodes mitochondrial GLS that catalyzes glutamine hydrolysis. The expression of GLS2 is upregulated by p53 in unstressed and stressed states. GLS2 regulates energy metabolism via increasing the production of glutamate and αKG, leading to increased mitochondrial respiration and ATP generation. Moreover, GLS2 can modulate the antioxidant defense function via upregulation of the glutathione (GSH) and downregulation of the ROS levels in cells, protecting cells from apoptosis caused by oxidative stress. Consistent with these functions of GLS2, p53 activation elevates glutamate and αKG levels, mitochondrial respiration rate, and GSH levels while decreasing ROS levels in cells[57]. Due to poor vascularization in the pancreatic cancer microenvironment and increased glutamine catabolism during rapid tumors growth, cancer cells are often exposed to a glutamine depleted microenvironment.

A conserved role for the protein phosphatase 2A (PP2A)-associated protein α4 in the sensing of glutamine has been reported. Upon depletion of glutamine, the assembly of an adaptive PP2A complex which contains the B55α regulatory subunit (Ppp2r2a) is promoted by α4 via providing the catalytic subunit. And B55α is specifically induced by glutamine deprivation in a ROS-dependent manner to activate p53 via direct interaction and dephosphorylation of EDD (a B55α-interacting protein) and to promote cell survival[58].

Arginine methylation of proteins is a common post-translational modification that is involved in a variety of cellular processes such as signal transduction, gene transcription, and metabolism. Malate dehydrogenase 1 (MDH1), a necessary enzyme for glutamine metabolism, is regulated by R248 site methylation and protein arginine methyltransferase 4 mediated MDH1 methylation. Methylation at R248 inhibits MDH1 post-translational mechanisms by disrupting MDH1 dimerization, which modulates mitochondrial respiration and glutamine-dependent production of NADPH in pancreatic cancer cells. MDH1 downregulation inhibits mitochondrial respiration, glutamine metabolism, and the sensitization of pancreatic cancer cells to oxidative stress and cell proliferation. Wild-type MDH1 re-expression protects pancreatic cancer cells from oxidative damage and restores cell growth and cloning activity. When the clinical sample was examined, it was found that the pancreatic cancer sample was in a hypomethylated state of MDH1 at R248[59].

Metabolomic comparisons revealed that pancreatic cancer tissue had low glucose levels, higher glycolytic intermediates, creatine phosphate, and the amino acids glutamine and serine, which are two main metabolic substrates. However, pancreatic cancer cells accumulate essential amino acids. Further research revealed that the accumulation of essential amino acids may occur from extracellular proteins which are degraded through macropinocytosis, resulting in an excess of most other amino acids[41]. These extracellular proteins are in sufficient quantities to meet the cellular requirement of glutamine. Ras-transformed pancreatic cancer cells transport extracellular proteins into the cell via macropinocytosis. The transported protein in the cell is degraded proteolytically, releasing amino acids, such as glutamine[60].

Glutamine is a key nutrient in tumorigenesis and a significant carbon and nitrogen source for biosynthetic reactions. A recent study indicated that tumor microenvironment-hosting cells do not utilize the nutrient like cancer cells do[61]. Liu et al[62] demonstrated that pancreatic stellate cells, the resident mesenchymal cells of the pancreas, express more glutamine synthetase than pancreatic cancer cells. Moreover, depletion of glutamine synthetase in pancreatic stellate cells impairs pancreatic cancer cell proliferation in vivo and in vitro. Mechanistically, researchers found that the β-catenin/TCF 7 complex directly binds to the glutamine synthetase promoter and upregulates glutamine synthetase to promote glutamine synthesis capacity and pancreatic stellate cell pro-tumor effect[62] (Figure 2).

Figure 2
Figure 2 Glutamine metabolic reprogramming. Glutamine is a key nutrient in tumourgenesis and Glutamine metabolic reprogramming in pancreatic cancer help enhance metabolic use of glutamine in cancer cells. Pancreatic cancer cells affect glutamate metabolism through Myc, Kras mutations, regulated in DNA damage and development 1, p53, Protein arginine methyltransferase 4, Ras transform, β-catenin/TCF 7 complex. REDD1: Regulated in DNA damage and development 1; ROS: Reactive oxygen species; GLS: Glutaminase; mTOR: the mammalian target of rapamycin; MDH1: Malate dehydrogenase 1; GSH: Glutathione.
GLUTAMINE METABOLISM AND CHEMO-RESISTANCE

With limited and ineffective medical and surgical treatment, pancreatic cancer is a most aggressive disease. Pancreatic cancer patients with inoperable lesions are treated with systemic chemotherapy, a therapeutic intervention to which most develop resistance. In tumor cells, the necessary raw materials can be provided from glutamine metabolism for over-activated glycolysis and oxidative phosphorylation reaction. Glutamine metabolism can also directly induce chemo-resistance in tumor cells by affecting the homeostasis of glucose, lipid, and protein metabolism.

In humans, the SLC1A5 gene, which consists of eight exons, has two transcribed variants (NM_005628.2 and NM_001145145.1) that differ from each other in the transcription initiation site. The long transcript of SLC1A5, which is also known as ASCT2, lacks exon 2 encodes 541 amino acids, whereas the short variant that lacks exon 1 encodes 339 amino acids. The SLC1A5 variant plays an important role in metabolic reprogramming. SLC1A5 can transport glutamine into the mitochondria via a mitochondrial-targeting signal at its N-terminal. HIF-2α mediates hypoxia-induced gene expression of the SLC1A5 variant. SLC1A5 variant overexpression in pancreatic cancer cells is involved in glutamine-induced ATP production and the synthesis of glutathione and confers resistance to gemcitabine (GEM)[63].

In addition, SLC7A8 gene-encoded L-type amino acid transporter 2 (LAT2) may promote glycolysis and decrease GEM sensitivity in pancreatic cancer by regulating two glutamine-dependent positive feedback loops (the LAT2/p-mTORSer2448 loop and the glutamine/p-mTORSer2448/glutamine synthetase loop). The study also showed that the reduced chemotherapy sensitivity caused by LAT2 upregulation in pancreatic cancer cells could be reversed by GEM in combination with an mTOR inhibitor (RAD001)[64].

Pancreatic cancer is characterized by severe hypoxia, which can reduce chemotherapy sensitivity[65]. Recent studies have demonstrated that an increase in glutamine catabolism in pancreatic cancer is the primary mechanism inducing hypoxia, and an in vitro 3D cell printing model of hypoxia based on extracellular matrix components has confirmed that glutamine catabolism is significantly increased in chemically resistant pancreatic cancer cells compared to that in chemically reactive pancreatic cancer cells[66]. Further studies have revealed that the rate of oxygen consumption rate is increased by elevated glutamine metabolic flux through oxidative phosphorylation (OXPHOS) in the mitochondrial, promoting hypoxia and chemical resistance. A glutamine antidote can alleviate hypoxia and improve chemotherapy efficacy in vitro and in vivo[66].

TCGA database revealed that wild-type isocitrate dehydrogenase (wtIDH1) was highly expressed in pancreatic cancer samples. Additionally, higher isocitrate dehydrogenase (IDH1) expression in pancreatic cancer patients is related to a poorer survival rate[67]. Cancer cells rely on wtIDH1 to produce NADPH and αKG for adaptive survival under nutritional constraints. These products support cancer cells under metabolic stress via antioxidant defense mechanisms (NADPH) and mitochondrial function (αKG). The experimental results suggest that wtIDH1 is actived by cellular NADPH and αKG, which is of great importance for the chemotherapy efficacy against pancreatic cancer.

5-Fluorouracil (5-FU), a common component of multi-drug treatment strategies in pancreatic cancer, works by converting intracellularly to its active metabolites via disrupting the synthesis of RNA and thymidylate synthase, resulting in cytotoxicity[68]. 5-FU treatment of pancreatic cancer cells increases ROS and apoptosis while inducing wtIDH1. An increase in ROS usually accompanied by accumulation of HuR and NRF-2. These compensations can protect cells from ROS damage via activation antioxidant defense mechanisms, leading to chemical resistance[69,70].

Kras mutations are most common in pancreatic cancer, and transcriptional reprogramming associated with Kras and abnormal signaling are thought to have a vital role in the development of chemotherapy resistance[71]. To meet the high anabolic needs of pancreatic cancer, carcinogenic Kras up-regulates glycolytic pathways and reprograms glutamine metabolism (manifested as glutamine dependence). GEM and other nucleotide analogs bind to replicated DNA, impairing the synthesis and repair of DNA and conferring cytotoxicity. In order to overcome the competitive inhibition of DNA synthesis, de novo synthesis is employed in tumor cells and thus resistance to nucleotide analogs is developed.

Therefore, limitation of the intracellular glutamine availability and inhibition of the glutamine synthetic nucleotides utilization by tumor cells can inhibit the natural nucleotide pool of tumor cells and effectively enhance the chemical sensitivity of tumor cells[72]. Capsid-optimized adenovirus-associated virus 8 vectors was used to deliver the CRISPR-CasRx system to pancreatic cancer in situ tumors and patient-derived tumor xenografts, demonstrating that CasRx accurately and efficiently silenced the expression of KrasG12D mutants in pancreatic ductal adenocarcinoma cells guided by KrasG12D-specific gRNA. CasRx knockdown of KrasG12D mutants can prevent abnormal activation of downstream signaling induced by KrasG12D mutants, thereby inhibiting tumor growth and improving GEM sensitivity in pancreatic cancer[73].

Glutamine metabolism may also contribute to tumor resistance by influencing protein post-translational modifications. The glutamine analog 6-diazo-5-oxo-L-norleucine treatment can make pancreatic cancer more sensitive to GEM. It has been identified that 6-diazo-5-oxo-L-norleucine can inhibit the hexosamine pathway, causing changes in the level of glycosylation of the entire cell proteome and thus affecting tumor sensitivity to chemotherapy[74]. Based on TCGA dataset analysis, Ganguly et al[75] observed mucin 5AC (MUC5AC) depletion in native mouse organoids and human pancreatic cancer cells which increased GEM sensitivity in vitro and in vivo.

Further analysis revealed that MUC5AC mediated β-catenin nuclear accumulation, which caused c-Myc transcription upregulation, increasing the uptake and utilization of glutamine, deoxycytidine triphosphate biosynthesis, and GEM resistance. Drug inhibitors and gene silencing of the MUC5AC/beta-catenin/c-Myc axis inhibited glutamine metabolism and sensitized MUC5AC-expressing pancreatic cancer cells and tumor organoids to GEM therapy[75].

When nab-paclitaxel was added to GEM, the median overall survival increased to 8.5-9.4 mo, however the median overall survival is 6.7 mo in GEM alone group[76]. Continuous induction of c-Myc plays an important role in nab-paclitaxel-resistant cells, and loss of c-Myc restores the sensitivity to nab-paclitaxel. However, the exact regulatory pathway involved remains unknown[77]. Drug resistance to chemotherapy is an important cause of poor prognosis in pancreatic cancer patients, and glutamine dependence caused by local metabolic reprogramming plays an important regulatory role in the occurrence of chemical drug resistance. Therefore, targeting the glutamine metabolic pathway may improve the chemotherapy effect on pancreatic cancer patients, which merits further investigation.

TARGETING GLUTAMINE METABOLISM IN PANCREATIC CANCER

Because glutamine metabolism is dependent on tumor cells and plays a role in tumor chemotherapy resistance, making it a potential anti-cancer target, many compounds have become the focus of research targeting glutamine metabolism from initial transport to a subsequent conversion to alpha-ketoglutaric acid[78,79].

GLS converts glutamine to glutamate during glutamine catabolism. Inhibiting GLS activity can reduce the antioxidant capacity of pancreatic cancer cells and inhibit cell proliferation. There are two GLS subtypes in mammalian cells: renal GLS1 and hepatic GLS2. GLS1 shows a widely expression in normal tissues, whereas GLS2 is primarily expressed in the liver, brain, pituitary gland and pancreas[80]. Inhibiting GLS, which inhibits the catalytic hydrolysis of glutamine to glutamate, is the most direct way to target glutamine breakdown. Bis-2-(5-phenylacetamido-1, 2, 4-thiadiazol-2-yl) ethyl sulfide (BPTES) is a specific GLS1 inhibitor with an allosteric, time-dependent property[81]. BPTES uniquely binds to the GLS tetramer oligomerisation interface[82]. Monotherapy with BPTES nanoparticles, a GLS inhibitor, produced significant tumor inhibition in a orthotopic transplantation mouse model[83].

The main glutamine transporter that determines intracellular glutamine levels is ASCT2, which is a key member of the amino acid transport system ASC family[84]. The mRNA level of ASCT2 was dramatically increased in both pancreatic cancer tissue samples and cancer cells. Kaplan-Meier mapping analysis revealed that high ASCT2 expression in pancreatic cancer was significantly associated with TNM stage and poor prognosis in these individuals[85]. Pancreatic cancer cell growth was significantly inhibited by ASCT2 activity blocking, either with L-γ-glutamyl-p-nitroaniline, a ASCT2 inhibitor, or with a specific shRNA silencing ASCT2 expression. Furthermore, ASCT2 knockdown may induce apoptosis through the Akt/mTOR signaling pathway[12]. Rajeshkumar et al[86] conducted an unbiased study to determine the anti-tumor efficacy of drugs targeting the metabolic pathways of multiple pancreatic cancer patient-derived xenografts and to determine whether genomic alterations are associated with specific metabolic patterns and thus sensitivity to specific metabolic inhibitors. The results indicated that the GLS inhibitor BPTES and aminotransferase inhibitor aminoacetate inhibited tumor growth, but no clear pattern was associated with tumor genomic status[86].

Ivosidenib (AG-120), a recently developed IDH1 inhibitor, disrupts the redox balance in local tissues, resulting in elevated ROS levels and enhanced chemotherapy-induced apoptosis in pancreatic cancer cells[67]. The United States Food and Drug Administration has approved Ivosidenib (AG-120) for treating mutant IDH1 cholangiocarcinoma and certain hematological malignancies[87]. Recent studies have confirmed that AG-120 is an effective wtIDH1 inhibitor in the tumor microenvironment (such as low nutrient levels), and inhibition of wtIDH1 can disrupt the redox balance, increase ROS levels, and enhance chemotherapy-induced apoptosis of pancreatic cancer cells, all of which play a role in pancreatic cancer inhibition[67,88].

GOT1 contributes to the antioxidant capacity of pancreatic cancer[39], and the antioxidative capacity of cancer cells supports their fitness and survival[89]. The present study identified that Aspulvinone O (AO), a novel inhibitor of GOT1 and glutamine metabolism, can sensitize pancreatic cancer cells to oxidative stress and inhibit cell proliferation. Moreover, selective AO inhibition of GOT1 dramatically reduced the proliferation of pancreatic cancer in vivo and in vitro[90]. Similarly, studies have demonstrated that a covalent small-molecule inhibitor (PF-04859989), a known kynurenine aminotransferase 2 inhibitor, has pyridoxal-5'-phosphate-dependent inhibitory activity against GOT1 and selective growth inhibition in pancreatic cancer cell lines[91].

In addition, various strategies have been tried in pancreatic cancer patients to treat the Kras mutation and its downstream signaling pathway. Attempts have been made to block the localization of Ras protein on the plasma membrane, to inhibit downstream oncogenic signaling via Kras effectors (including MEK1/2, Erk1/2 or Akt) targeting alone or in combination, and to inhibit Kras proteins directly. Due to compensatory mechanisms and toxicity associated with small therapeutic windows, the majority of clinical trials have been directed at targeting downstream pathways, with limited clinical benefit[92].

C-Myc is located downstream of Kras and is associated with a number of oncogenic and proliferative pathways in various cancers, including pancreatic cancer. C-Myc provides adequate energy for cancer metabolism and substrate for the synthesis of organic molecules by increasing aerobic glycolysis and modulating the biosynthesis of glutamate biosynthesis in glutamine. Overexpression of C-Myc is correlated with resistance to chemotherapy in pancreatic cancer, and small molecules accelerating the ubiquitination and degradation of C-Myc have shown efficacy in the treatment of pancreatic cancer in preclinical studies[93,94].

However, cancer cells demonstrate strong metabolic compensation ability when a single drug to inhibit key enzymes in the glutamine metabolic pathway is used, making long-term tumor suppression difficult[95,96]. Metabolomic analysis revealed that tumor cells could survive by relying on glycolysis and the synthesis of glycogen following GLS inhibition. Metformin was chosen for application with BPTES nanoparticles based on this finding, and the reduction in pancreatic tumors was significantly more significant than treatment alone[83].

It is suggested that combining glutamine-metabolizing enzyme inhibitors with other anti-metabolic drugs is a promising avenue for further exploration. In addition, pharmacological inhibition of IDH1 by AG-120 is an attractive option for combination therapy with cytotoxic chemotherapy in pancreatic cancer patients, and a phase Ib trial of AG-120 in combination with multi-agent chemotherapy in pancreatic cancer patients has been initiated[67] (Figure 3).

Figure 3
Figure 3 Targeting glutamine metabolism in pancreatic cancer. A few compounds that target glutamine metabolism by inhibiting glutaminase 1, Ivosidenib (AG-120) or aspartate transaminase (GOT1), blocking activity of alanine-serine-cysteine transporter 2 (ASCT2), ASCT2 knockdown, affecting Kas/c-Myc are studied for the treatment of pancreatic cancer. ROS: Reactive oxygen species; GLS: Glutaminase; BPTES: Bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl) ethyl sulfide; AO: Aspulvinone O; mTOR: Mammalian target of rapamycin; IDH1: Isocitrate dehydrogenase.
CONCLUSION

Owing to the high incidence of metastatic disease and limited treatment responses, pancreatic cancer is expected to be the second most lethal cancer by 2040[97]. Currently, there are few effective treatment options for pancreatic cancer. Although new combination chemotherapy has improved, the prognosis is often poor due to the rapid development of resistance to chemotherapy. Therefore, there is an urgent need for new treatment strategies to improve the prognosis of the patients[98]. Metabolic reprogramming has attracted renewed interest in recent years. Cancer cells undergo metabolic reprogramming to promote cell growth and proliferation.

The vital role of glutamine in cell metabolism determines its importance in cancer research. Recently, we have gained a new understanding of the changes in pancreatic cancer metabolism, targeting key enzymes of glutamine metabolism and glutamine transporters and supplementing glutamine metabolism inhibitors during chemoradiotherapy have shown certain anti-tumor effects, but there are still many challenges to developing targeted metabolism into an effective anticancer therapy for clinical application. Owing to the complex feedback loops and parallel and interacting energy supply networks in pancreatic cancer cells, it may be difficult to obtain satisfactory results by inhibiting a single pathway in the energy metabolic pathway of cancer cells. The combined application of multiple anti-metabolic drugs and multi-target intervention has a broad development potential, and individualized treatment based on tumor cell metabolic patterns may further improve the therapeutic effect in pancreatic cancer.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Isaji S, Japan; Koganti SB, United States; Tantau AI, Romania S-Editor: Yan JP L-Editor: A P-Editor: Zhang XD

References
1.  Cai J, Chen H, Lu M, Zhang Y, Lu B, You L, Zhang T, Dai M, Zhao Y. Advances in the epidemiology of pancreatic cancer: Trends, risk factors, screening, and prognosis. Cancer Lett. 2021;520:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 100]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
2.  Klein AP. Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 2021;18:493-502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 334]  [Article Influence: 111.3]  [Reference Citation Analysis (0)]
3.  Wood LD, Canto MI, Jaffee EM, Simeone DM. Pancreatic Cancer: Pathogenesis, Screening, Diagnosis, and Treatment. Gastroenterology. 2022;163:386-402.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 176]  [Article Influence: 88.0]  [Reference Citation Analysis (0)]
4.  Qin C, Yang G, Yang J, Ren B, Wang H, Chen G, Zhao F, You L, Wang W, Zhao Y. Metabolism of pancreatic cancer: paving the way to better anticancer strategies. Mol Cancer. 2020;19:50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 169]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]
5.  Bott AJ, Shen J, Tonelli C, Zhan L, Sivaram N, Jiang YP, Yu X, Bhatt V, Chiles E, Zhong H, Maimouni S, Dai W, Velasquez S, Pan JA, Muthalagu N, Morton J, Anthony TG, Feng H, Lamers WH, Murphy DJ, Guo JY, Jin J, Crawford HC, Zhang L, White E, Lin RZ, Su X, Tuveson DA, Zong WX. Glutamine Anabolism Plays a Critical Role in Pancreatic Cancer by Coupling Carbon and Nitrogen Metabolism. Cell Rep. 2019;29:1287-1298.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 95]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
6.  Altman BJ, Stine ZE, Dang CV. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer. 2016;16:619-634.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1122]  [Cited by in F6Publishing: 1098]  [Article Influence: 137.3]  [Reference Citation Analysis (0)]
7.  Cluntun AA, Lukey MJ, Cerione RA, Locasale JW. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer. 2017;3:169-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 411]  [Cited by in F6Publishing: 421]  [Article Influence: 60.1]  [Reference Citation Analysis (0)]
8.  Sharma S, Agnihotri N, Kumar S. Targeting fuel pocket of cancer cell metabolism: A focus on glutaminolysis. Biochem Pharmacol. 2022;198:114943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
9.  Kao TW, Chuang YC, Lee HL, Kuo CC, Shen YA. Therapeutic Targeting of Glutaminolysis as a Novel Strategy to Combat Cancer Stem Cells. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
10.  Blum R, Kloog Y. Metabolism addiction in pancreatic cancer. Cell Death Dis. 2014;5:e1065.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 103]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
11.  Cohen R, Neuzillet C, Tijeras-Raballand A, Faivre S, de Gramont A, Raymond E. Targeting cancer cell metabolism in pancreatic adenocarcinoma. Oncotarget. 2015;6:16832-16847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 91]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
12.  Wang W, Pan H, Ren F, Chen H, Ren P. Targeting ASCT2-mediated glutamine metabolism inhibits proliferation and promotes apoptosis of pancreatic cancer cells. Biosci Rep. 2022;42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
13.  Ying M, You D, Zhu X, Cai L, Zeng S, Hu X. Lactate and glutamine support NADPH generation in cancer cells under glucose deprived conditions. Redox Biol. 2021;46:102065.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 58]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
14.  Li X, Wenes M, Romero P, Huang SC, Fendt SM, Ho PC. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat Rev Clin Oncol. 2019;16:425-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 279]  [Article Influence: 55.8]  [Reference Citation Analysis (0)]
15.  Halama A, Suhre K. Advancing Cancer Treatment by Targeting Glutamine Metabolism-A Roadmap. Cancers (Basel). 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 32]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
16.  Yang X, Li Z, Ren H, Peng X, Fu J. New progress of glutamine metabolism in the occurrence, development, and treatment of ovarian cancer from mechanism to clinic. Front Oncol. 2022;12:1018642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
17.  Lacey JM, Wilmore DW. Is glutamine a conditionally essential amino acid? Nutr Rev. 1990;48:297-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 553]  [Cited by in F6Publishing: 521]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
18.  Curthoys NP, Weiss RF. Regulation of renal ammoniagenesis. Subcellular localization of rat kidney glutaminase isoenzymes. J Biol Chem. 1974;249:3261-3266.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Bhutia YD, Ganapathy V. Glutamine transporters in mammalian cells and their functions in physiology and cancer. Biochim Biophys Acta. 2016;1863:2531-2539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 188]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
20.  Hartman SC, Buchanan JM. Nucleic acids, purines, pyrimidines (nucleotide synthesis). Annu Rev Biochem. 1959;28:365-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 92]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
21.  Hartman SC, Buchanan JM. Biosynthesis of the purines. XXI. 5-Phosphoribosylpyrophosphate amidotransferase. J Biol Chem. 1958;233:451-455.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Nurjhan N, Bucci A, Perriello G, Stumvoll M, Dailey G, Bier DM, Toft I, Jenssen TG, Gerich JE. Glutamine: a major gluconeogenic precursor and vehicle for interorgan carbon transport in man. J Clin Invest. 1995;95:272-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 123]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
23.  Snoke JE, Bloch K. Formation and utilization of gamma-glutamylcysteine in glutathione synthesis. J Biol Chem. 1952;199:407-414.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Abdul Kader S, Dib S, Achkar IW, Thareja G, Suhre K, Rafii A, Halama A. Defining the landscape of metabolic dysregulations in cancer metastasis. Clin Exp Metastasis. 2022;39:345-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
25.  Roberts E, Frankel S. Free amino acids in normal and neoplastic tissues of mice as studied by paper chromatography. Cancer Res. 1949;9:645-648, 3 pl.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Hassanein M, Hoeksema MD, Shiota M, Qian J, Harris BK, Chen H, Clark JE, Alborn WE, Eisenberg R, Massion PP. SLC1A5 mediates glutamine transport required for lung cancer cell growth and survival. Clin Cancer Res. 2013;19:560-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 200]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
27.  Daemen A, Liu B, Song K, Kwong M, Gao M, Hong R, Nannini M, Peterson D, Liederer BM, de la Cruz C, Sangaraju D, Jaochico A, Zhao X, Sandoval W, Hunsaker T, Firestein R, Latham S, Sampath D, Evangelista M, Hatzivassiliou G. Pan-Cancer Metabolic Signature Predicts Co-Dependency on Glutaminase and De Novo Glutathione Synthesis Linked to a High-Mesenchymal Cell State. Cell Metab. 2018;28:383-399.e9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 45]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
28.  Zhang Z, Liu R, Shuai Y, Huang Y, Jin R, Wang X, Luo J. ASCT2 (SLC1A5)-dependent glutamine uptake is involved in the progression of head and neck squamous cell carcinoma. Br J Cancer. 2020;122:82-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 62]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
29.  Wang R, Xiang W, Xu Y, Han L, Li Q, Dai W, Cai G. Enhanced glutamine utilization mediated by SLC1A5 and GPT2 is an essential metabolic feature of colorectal signet ring cell carcinoma with therapeutic potential. Ann Transl Med. 2020;8:302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
30.  Ramachandran S, R Sennoune S, Sharma M, Thangaraju M, V Suresh V, Sneigowski T, D Bhutia Y, Pruitt K, Ganapathy V. Expression and function of SLC38A5, an amino acid-coupled Na+/H+ exchanger, in triple-negative breast cancer and its relevance to macropinocytosis. Biochem J. 2021;478:3957-3976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
31.  Smith DK, Kates L, Durinck S, Patel N, Stawiski EW, Kljavin N, Foreman O, Sipos B, Solloway MJ, Allan BB, Peterson AS. Elevated Serum Amino Acids Induce a Subpopulation of Alpha Cells to Initiate Pancreatic Neuroendocrine Tumor Formation. Cell Rep Med. 2020;1:100058.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
32.  Najumudeen AK, Ceteci F, Fey SK, Hamm G, Steven RT, Hall H, Nikula CJ, Dexter A, Murta T, Race AM, Sumpton D, Vlahov N, Gay DM, Knight JRP, Jackstadt R, Leach JDG, Ridgway RA, Johnson ER, Nixon C, Hedley A, Gilroy K, Clark W, Malla SB, Dunne PD, Rodriguez-Blanco G, Critchlow SE, Mrowinska A, Malviya G, Solovyev D, Brown G, Lewis DY, Mackay GM, Strathdee D, Tardito S, Gottlieb E; CRUK Rosetta Grand Challenge Consortium, Takats Z, Barry ST, Goodwin RJA, Bunch J, Bushell M, Campbell AD, Sansom OJ. The amino acid transporter SLC7A5 is required for efficient growth of KRAS-mutant colorectal cancer. Nat Genet. 2021;53:16-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 106]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
33.  Myint ZW, Sun RC, Hensley PJ, James AC, Wang P, Strup SE, McDonald RJ, Yan D, St Clair WH, Allison DB. Evaluation of Glutaminase Expression in Prostate Adenocarcinoma and Correlation with Clinicopathologic Parameters. Cancers (Basel). 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
34.  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;10:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 113]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
35.  van Geldermalsen M, Wang Q, Nagarajah R, Marshall AD, Thoeng A, Gao D, Ritchie W, Feng Y, Bailey CG, Deng N, Harvey K, Beith JM, Selinger CI, O'Toole SA, Rasko JE, Holst J. ASCT2/SLC1A5 controls glutamine uptake and tumour growth in triple-negative basal-like breast cancer. Oncogene. 2016;35:3201-3208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 329]  [Cited by in F6Publishing: 373]  [Article Influence: 46.6]  [Reference Citation Analysis (0)]
36.  Dorai T, Dorai B, Pinto JT, Grasso M, Cooper AJL. High Levels of Glutaminase II Pathway Enzymes in Normal and Cancerous Prostate Suggest a Role in 'Glutamine Addiction'. Biomolecules. 2019;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
37.  Coothankandaswamy V, Cao S, Xu Y, Prasad PD, Singh PK, Reynolds CP, Yang S, Ogura J, Ganapathy V, Bhutia YD. Amino acid transporter SLC6A14 is a novel and effective drug target for pancreatic cancer. Br J Pharmacol. 2016;173:3292-3306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 82]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
38.  Guillaumond F, Leca J, Olivares O, Lavaut MN, Vidal N, Berthezène P, Dusetti NJ, Loncle C, Calvo E, Turrini O, Iovanna JL, Tomasini R, Vasseur S. Strengthened glycolysis under hypoxia supports tumor symbiosis and hexosamine biosynthesis in pancreatic adenocarcinoma. Proc Natl Acad Sci U S A. 2013;110:3919-3924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 295]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
39.  Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, Perera RM, Ferrone CR, Mullarky E, Shyh-Chang N, Kang Y, Fleming JB, Bardeesy N, Asara JM, Haigis MC, DePinho RA, Cantley LC, Kimmelman AC. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1227]  [Cited by in F6Publishing: 1400]  [Article Influence: 127.3]  [Reference Citation Analysis (0)]
40.  Yang S, Hwang S, Kim M, Seo SB, Lee JH, Jeong SM. Mitochondrial glutamine metabolism via GOT2 supports pancreatic cancer growth through senescence inhibition. Cell Death Dis. 2018;9:55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 63]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
41.  Kamphorst JJ, Nofal M, Commisso C, Hackett SR, Lu W, Grabocka E, Vander Heiden MG, Miller G, Drebin JA, Bar-Sagi D, Thompson CB, Rabinowitz JD. Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res. 2015;75:544-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 498]  [Cited by in F6Publishing: 596]  [Article Influence: 66.2]  [Reference Citation Analysis (0)]
42.  Koong AC, Mehta VK, Le QT, Fisher GA, Terris DJ, Brown JM, Bastidas AJ, Vierra M. Pancreatic tumors show high levels of hypoxia. Int J Radiat Oncol Biol Phys. 2000;48:919-922.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 423]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
43.  Bristow RG, Hill RP. Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nat Rev Cancer. 2008;8:180-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 822]  [Cited by in F6Publishing: 808]  [Article Influence: 50.5]  [Reference Citation Analysis (0)]
44.  Collado M, Serrano M. Senescence in tumours: evidence from mice and humans. Nat Rev Cancer. 2010;10:51-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 786]  [Cited by in F6Publishing: 806]  [Article Influence: 57.6]  [Reference Citation Analysis (0)]
45.  Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell. 2012;21:297-308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2205]  [Cited by in F6Publishing: 2292]  [Article Influence: 191.0]  [Reference Citation Analysis (0)]
46.  Shanware NP, Mullen AR, DeBerardinis RJ, Abraham RT. Glutamine: pleiotropic roles in tumor growth and stress resistance. J Mol Med (Berl). 2011;89:229-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 138]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
47.  Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, Wilson KF, Ambrosio AL, Dias SM, Dang CV, Cerione RA. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 2010;18:207-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 605]  [Cited by in F6Publishing: 632]  [Article Influence: 45.1]  [Reference Citation Analysis (0)]
48.  Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang XY, Pfeiffer HK, Nissim I, Daikhin E, Yudkoff M, McMahon SB, Thompson CB. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc Natl Acad Sci U S A. 2008;105:18782-18787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1313]  [Cited by in F6Publishing: 1439]  [Article Influence: 89.9]  [Reference Citation Analysis (0)]
49.  di Magliano MP, Logsdon CD. Roles for KRAS in pancreatic tumor development and progression. Gastroenterology. 2013;144:1220-1229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 297]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
50.  Shen X, Chang LG, Hu MY, Yan D, Zhou LN, Ma Y, Ling SK, Fu YQ, Zhang SY, Kong B, Huang PL. KrasG12D-LOH promotes malignant biological behavior and energy metabolism of pancreatic ductal adenocarcinoma cells through the mTOR signaling pathway. Neoplasma. 2018;65:81-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
51.  Saxton RA, Sabatini DM. mTOR Signaling in Growth, Metabolism, and Disease. Cell. 2017;169:361-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1089]  [Cited by in F6Publishing: 1260]  [Article Influence: 180.0]  [Reference Citation Analysis (0)]
52.  Xu P, Oosterveer MH, Stein S, Demagny H, Ryu D, Moullan N, Wang X, Can E, Zamboni N, Comment A, Auwerx J, Schoonjans K. LRH-1-dependent programming of mitochondrial glutamine processing drives liver cancer. Genes Dev. 2016;30:1255-1260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
53.  Ma Y, Li Y, Ling S, Li X, Kong B, Hu M, Huang P. Loss of heterozygosity for Kras(G12D) promotes REDD1-dependent, non-canonical glutamine metabolism in pancreatic ductal adenocarcinoma. Biochem Biophys Res Commun. 2020;526:880-888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
54.  Raho S, Capobianco L, Malivindi R, Vozza A, Piazzolla C, De Leonardis F, Gorgoglione R, Scarcia P, Pezzuto F, Agrimi G, Barile SN, Pisano I, Reshkin SJ, Greco MR, Cardone RA, Rago V, Li Y, Marobbio CMT, Sommergruber W, Riley CL, Lasorsa FM, Mills E, Vegliante MC, De Benedetto GE, Fratantonio D, Palmieri L, Dolce V, Fiermonte G. KRAS-regulated glutamine metabolism requires UCP2-mediated aspartate transport to support pancreatic cancer growth. Nat Metab. 2020;2:1373-1381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 60]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
55.  Bieging KT, Mello SS, Attardi LD. Unravelling mechanisms of p53-mediated tumour suppression. Nat Rev Cancer. 2014;14:359-370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 924]  [Cited by in F6Publishing: 949]  [Article Influence: 94.9]  [Reference Citation Analysis (0)]
56.  Cheung EC, Vousden KH. The role of p53 in glucose metabolism. Curr Opin Cell Biol. 2010;22:186-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 70]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
57.  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;107:7455-7460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 561]  [Cited by in F6Publishing: 612]  [Article Influence: 43.7]  [Reference Citation Analysis (0)]
58.  Reid MA, Wang WI, Rosales KR, Welliver MX, Pan M, Kong M. The B55α subunit of PP2A drives a p53-dependent metabolic adaptation to glutamine deprivation. Mol Cell. 2013;50:200-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 126]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
59.  Wang YP, Zhou W, Wang J, Huang X, Zuo Y, Wang TS, Gao X, Xu YY, Zou SW, Liu YB, Cheng JK, Lei QY. Arginine Methylation of MDH1 by CARM1 Inhibits Glutamine Metabolism and Suppresses Pancreatic Cancer. Mol Cell. 2016;64:673-687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 121]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
60.  Commisso C, Davidson SM, Soydaner-Azeloglu RG, Parker SJ, Kamphorst JJ, Hackett S, Grabocka E, Nofal M, Drebin JA, Thompson CB, Rabinowitz JD, Metallo CM, Vander Heiden MG, Bar-Sagi D. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature. 2013;497:633-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1020]  [Cited by in F6Publishing: 1137]  [Article Influence: 103.4]  [Reference Citation Analysis (0)]
61.  Reinfeld BI, Madden MZ, Wolf MM, Chytil A, Bader JE, Patterson AR, Sugiura A, Cohen AS, Ali A, Do BT, Muir A, Lewis CA, Hongo RA, Young KL, Brown RE, Todd VM, Huffstater T, Abraham A, O'Neil RT, Wilson MH, Xin F, Tantawy MN, Merryman WD, Johnson RW, Williams CS, Mason EF, Mason FM, Beckermann KE, Vander Heiden MG, Manning HC, Rathmell JC, Rathmell WK. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature. 2021;593:282-288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 373]  [Cited by in F6Publishing: 445]  [Article Influence: 148.3]  [Reference Citation Analysis (0)]
62.  Liu H, Zhang H, Liu X, Guo W, Liu Q, Chen L, Pang J, Li R, Tong WM, Wu H, Dai M, Liang Z. Pancreatic stellate cells exploit Wnt/β-catenin/TCF7-mediated glutamine metabolism to promote pancreatic cancer cells growth. Cancer Lett. 2023;555:216040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 11]  [Reference Citation Analysis (0)]
63.  Yoo HC, Park SJ, Nam M, Kang J, Kim K, Yeo JH, Kim JK, Heo Y, Lee HS, Lee MY, Lee CW, Kang JS, Kim YH, Lee J, Choi J, Hwang GS, Bang S, Han JM. A Variant of SLC1A5 Is a Mitochondrial Glutamine Transporter for Metabolic Reprogramming in Cancer Cells. Cell Metab. 2020;31:267-283.e12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 199]  [Article Influence: 49.8]  [Reference Citation Analysis (0)]
64.  Feng M, Xiong G, Cao Z, Yang G, Zheng S, Qiu J, You L, Zheng L, Zhang T, Zhao Y. LAT2 regulates glutamine-dependent mTOR activation to promote glycolysis and chemoresistance in pancreatic cancer. J Exp Clin Cancer Res. 2018;37:274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 74]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
65.  Akman M, Belisario DC, Salaroglio IC, Kopecka J, Donadelli M, De Smaele E, Riganti C. Hypoxia, endoplasmic reticulum stress and chemoresistance: dangerous liaisons. J Exp Clin Cancer Res. 2021;40:28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 57]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
66.  Park SJ, Yoo HC, Ahn E, Luo E, Kim Y, Sung Y, Yu YC, Kim K, Min DS, Lee HS, Hwang GS, Ahn T, Choi J, Bang S, Han JM. Enhanced Glutaminolysis Drives Hypoxia-Induced Chemoresistance in Pancreatic Cancer. Cancer Res. 2023;83:735-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
67.  Zarei M, Hajihassani O, Hue JJ, Graor HJ, Rothermel LD, Winter JM. Targeting wild-type IDH1 enhances chemosensitivity in pancreatic cancer. bioRxiv. 2023;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Reference Citation Analysis (0)]
68.  Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer. 2003;3:330-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3255]  [Cited by in F6Publishing: 3306]  [Article Influence: 157.4]  [Reference Citation Analysis (0)]
69.  Zarei M, Lal S, Parker SJ, Nevler A, Vaziri-Gohar A, Dukleska K, Mambelli-Lisboa NC, Moffat C, Blanco FF, Chand SN, Jimbo M, Cozzitorto JA, Jiang W, Yeo CJ, Londin ER, Seifert EL, Metallo CM, Brody JR, Winter JM. Posttranscriptional Upregulation of IDH1 by HuR Establishes a Powerful Survival Phenotype in Pancreatic Cancer Cells. Cancer Res. 2017;77:4460-4471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 74]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
70.  DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C, Frese K, Mangal D, Yu KH, Yeo CJ, Calhoun ES, Scrimieri F, Winter JM, Hruban RH, Iacobuzio-Donahue C, Kern SE, Blair IA, Tuveson DA. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 2011;475:106-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1492]  [Cited by in F6Publishing: 1618]  [Article Influence: 124.5]  [Reference Citation Analysis (0)]
71.  Tao S, Wang S, Moghaddam SJ, Ooi A, Chapman E, Wong PK, Zhang DD. Oncogenic KRAS confers chemoresistance by upregulating NRF2. Cancer Res. 2014;74:7430-7441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 212]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
72.  Chen X, Chen S, Yu D. Metabolic Reprogramming of Chemoresistant Cancer Cells and the Potential Significance of Metabolic Regulation in the Reversal of Cancer Chemoresistance. Metabolites. 2020;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 63]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
73.  Jiang W, Li H, Liu X, Zhang J, Zhang W, Li T, Liu L, Yu X. Precise and efficient silencing of mutant Kras(G12D) by CRISPR-CasRx controls pancreatic cancer progression. Theranostics. 2020;10:11507-11519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
74.  Chen R, Lai LA, Sullivan Y, Wong M, Wang L, Riddell J, Jung L, Pillarisetty VG, Brentnall TA, Pan S. Disrupting glutamine metabolic pathways to sensitize gemcitabine-resistant pancreatic cancer. Sci Rep. 2017;7:7950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 62]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
75.  Ganguly K, Bhatia R, Rauth S, Kisling A, Atri P, Thompson C, Vengoji R, Ram Krishn S, Shinde D, Thomas V, Kaur S, Mallya K, Cox JL, Kumar S, Batra SK. Mucin 5AC Serves as the Nexus for β-Catenin/c-Myc Interplay to Promote Glutamine Dependency During Pancreatic Cancer Chemoresistance. Gastroenterology. 2022;162:253-268.e13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 28]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
76.  Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, Harris M, Reni M, Dowden S, Laheru D, Bahary N, Ramanathan RK, Tabernero J, Hidalgo M, Goldstein D, Van Cutsem E, Wei X, Iglesias J, Renschler MF. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691-1703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4035]  [Cited by in F6Publishing: 4354]  [Article Influence: 395.8]  [Reference Citation Analysis (0)]
77.  Parasido E, Avetian GS, Naeem A, Graham G, Pishvaian M, Glasgow E, Mudambi S, Lee Y, Ihemelandu C, Choudhry M, Peran I, Banerjee PP, Avantaggiati ML, Bryant K, Baldelli E, Pierobon M, Liotta L, Petricoin E, Fricke ST, Sebastian A, Cozzitorto J, Loots GG, Kumar D, Byers S, Londin E, DiFeo A, Narla G, Winter J, Brody JR, Rodriguez O, Albanese C. The Sustained Induction of c-MYC Drives Nab-Paclitaxel Resistance in Primary Pancreatic Ductal Carcinoma Cells. Mol Cancer Res. 2019;17:1815-1827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
78.  Yang WH, Qiu Y, Stamatatos O, Janowitz T, Lukey MJ. Enhancing the Efficacy of Glutamine Metabolism Inhibitors in Cancer Therapy. Trends Cancer. 2021;7:790-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 87]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
79.  Mukha A, Kahya U, Dubrovska A. Targeting glutamine metabolism and autophagy: the combination for prostate cancer radiosensitization. Autophagy. 2021;17:3879-3881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
80.  Jiang B, Zhang J, Zhao G, Liu M, Hu J, Lin F, Wang J, Zhao W, Ma H, Zhang C, Wu C, Yao L, Liu Q, Chen X, Cao Y, Zheng Y, Han A, Lin D, Li Q. Filamentous GLS1 promotes ROS-induced apoptosis upon glutamine deprivation via insufficient asparagine synthesis. Mol Cell. 2022;82:1821-1835.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
81.  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;109:7705-7710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 149]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
82.  Thomas AG, Rojas C, Tanega C, Shen M, Simeonov A, Boxer MB, Auld DS, Ferraris DV, Tsukamoto T, Slusher BS. Kinetic characterization of ebselen, chelerythrine and apomorphine as glutaminase inhibitors. Biochem Biophys Res Commun. 2013;438:243-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
83.  Elgogary A, Xu Q, Poore B, Alt J, Zimmermann SC, Zhao L, Fu J, Chen B, Xia S, Liu Y, Neisser M, Nguyen C, Lee R, Park JK, Reyes J, Hartung T, Rojas C, Rais R, Tsukamoto T, Semenza GL, Hanes J, Slusher BS, Le A. Combination therapy with BPTES nanoparticles and metformin targets the metabolic heterogeneity of pancreatic cancer. Proc Natl Acad Sci U S A. 2016;113:E5328-E5336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 161]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
84.  Jiang H, Zhang N, Tang T, Feng F, Sun H, Qu W. Target the human Alanine/Serine/Cysteine Transporter 2(ASCT2): Achievement and Future for Novel Cancer Therapy. Pharmacol Res. 2020;158:104844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
85.  Kaira K, Sunose Y, Arakawa K, Sunaga N, Shimizu K, Tominaga H, Oriuchi N, Nagamori S, Kanai Y, Oyama T, Takeyoshi I. Clinicopathological significance of ASC amino acid transporter-2 expression in pancreatic ductal carcinoma. Histopathology. 2015;66:234-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
86.  Rajeshkumar NV, Yabuuchi S, Pai SG, De Oliveira E, Kamphorst JJ, Rabinowitz JD, Tejero H, Al-Shahrour F, Hidalgo M, Maitra A, Dang CV. Treatment of Pancreatic Cancer Patient-Derived Xenograft Panel with Metabolic Inhibitors Reveals Efficacy of Phenformin. Clin Cancer Res. 2017;23:5639-5647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 63]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
87.  Zarei M, Hue JJ, Hajihassani O, Graor HJ, Katayama ES, Loftus AW, Bajor D, Rothermel LD, Vaziri-Gohar A, Winter JM. Clinical development of IDH1 inhibitors for cancer therapy. Cancer Treat Rev. 2022;103:102334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
88.  Vaziri-Gohar A, Cassel J, Mohammed FS, Zarei M, Hue JJ, Hajihassani O, Graor HJ, Srikanth YVV, Karim SA, Abbas A, Prendergast E, Chen V, Katayama ES, Dukleska K, Khokhar I, Andren A, Zhang L, Wu C, Erokwu B, Flask CA, Wang R, Rothermel LD, Romani AMP, Bowers J, Getts R, Tatsuoka C, Morton JP, Bederman I, Brunengraber H, Lyssiotis CA, Salvino JM, Brody JR, Winter JM. Limited nutrient availability in the tumor microenvironment renders pancreatic tumors sensitive to allosteric IDH1 inhibitors. Nat Cancer. 2022;3:852-865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 35]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
89.  Hawk MA, McCallister C, Schafer ZT. Antioxidant Activity during Tumor Progression: A Necessity for the Survival of Cancer Cells? Cancers (Basel). 2016;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
90.  Sun W, Luan S, Qi C, Tong Q, Yan S, Li H, Zhang Y. Aspulvinone O, a natural inhibitor of GOT1 suppresses pancreatic ductal adenocarcinoma cells growth by interfering glutamine metabolism. Cell Commun Signal. 2019;17:111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
91.  Yoshida T, Yamasaki S, Kaneko O, Taoka N, Tomimoto Y, Namatame I, Yahata T, Kuromitsu S, Cantley LC, Lyssiotis CA. A covalent small molecule inhibitor of glutamate-oxaloacetate transaminase 1 impairs pancreatic cancer growth. Biochem Biophys Res Commun. 2020;522:633-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
92.  Choi M, Bien H, Mofunanya A, Powers S. Challenges in Ras therapeutics in pancreatic cancer. Semin Cancer Biol. 2019;54:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
93.  Jin X, Fang R, Fan P, Zeng L, Zhang B, Lu X, Liu T. PES1 promotes BET inhibitors resistance and cells proliferation through increasing c-Myc expression in pancreatic cancer. J Exp Clin Cancer Res. 2019;38:463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
94.  Pan Y, Fei Q, Xiong P, Yang J, Zhang Z, Lin X, Pan M, Lu F, Huang H. Synergistic inhibition of pancreatic cancer with anti-PD-L1 and c-Myc inhibitor JQ1. Oncoimmunology. 2019;8:e1581529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 42]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
95.  Halama A, Kulinski M, Dib SS, Zaghlool SB, Siveen KS, Iskandarani A, Zierer J, Prabhu KS, Satheesh NJ, Bhagwat AM, Uddin S, Kastenmüller G, Elemento O, Gross SS, Suhre K. Accelerated lipid catabolism and autophagy are cancer survival mechanisms under inhibited glutaminolysis. Cancer Lett. 2018;430:133-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
96.  Reis LMD, Adamoski D, Ornitz Oliveira Souza R, Rodrigues Ascenção CF, Sousa de Oliveira KR, Corrêa-da-Silva F, Malta de Sá Patroni F, Meira Dias M, Consonni SR, Mendes de Moraes-Vieira PM, Silber AM, Dias SMG. Dual inhibition of glutaminase and carnitine palmitoyltransferase decreases growth and migration of glutaminase inhibition-resistant triple-negative breast cancer cells. J Biol Chem. 2019;294:9342-9357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 48]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
97.  Rahib L, Wehner MR, Matrisian LM, Nead KT. Estimated Projection of US Cancer Incidence and Death to 2040. JAMA Netw Open. 2021;4:e214708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 500]  [Article Influence: 166.7]  [Reference Citation Analysis (0)]
98.  Nevala-Plagemann C, Hidalgo M, Garrido-Laguna I. From state-of-the-art treatments to novel therapies for advanced-stage pancreatic cancer. Nat Rev Clin Oncol. 2020;17:108-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 215]  [Article Influence: 53.8]  [Reference Citation Analysis (0)]