Next Article in Journal
Circulating Tumor Cell-Free DNA as Prognostic Biomarker in Non-Small Cell Lung Cancer Patients Undergoing Immunotherapy: The CORELAB Experience
Previous Article in Journal
Enhancing Soybean Salt Tolerance with GSNO and Silicon: A Comprehensive Physiological, Biochemical, and Genetic Study
Previous Article in Special Issue
Enhanced CRC Growth in Iron-Rich Environment, Facts and Speculations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Pentose Phosphate Pathway: From Mechanisms to Implications for Gastrointestinal Cancers

1
Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
2
Cancer Institute (A Key Laboratory for Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
3
Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(2), 610; https://doi.org/10.3390/ijms26020610
Submission received: 28 October 2024 / Revised: 27 December 2024 / Accepted: 8 January 2025 / Published: 13 January 2025

Abstract

:
The pentose phosphate pathway (PPP), traditionally recognized for its role in generating nicotinamide adenine dinucleotide phosphate (NADPH) and ribose-5-phosphate (R5P), has emerged as a critical metabolic hub with involvements in various gastrointestinal (GI) cancers. The PPP plays crucial roles in the initiation, development, and tumor microenvironment (TME) of GI cancers by modulating redox homeostasis and providing precursors for nucleotide biosynthesis. Targeting PPP enzymes and their regulatory axis has been a potential strategy in anti-GI cancer therapies. In this review, we summarize the regulatory mechanisms of PPP enzymes, elucidate the relationships between the PPP and TME’s elements, and discuss the therapeutic potential of targeting the PPP in GI cancers.

1. Introduction

Gastrointestinal (GI) cancers are prevalent and have a detrimental impact on patients’ quality of life and their capacity for productivity, leading to substantial healthcare utilization [1]. According to the global cancer statistics by world region for the year 2022, GI cancers account for nearly one-quarter of the global cancer incidence and one-third of all cancer-related deaths [2].
Metabolic reprogramming, a hallmark of cancer, is described as alterations in key metabolic pathways to meet the metabolic needs of tumor cells [3]. The Warburg effect refers to the preference of cancer cells to convert glucose to lactate through glycolysis, even in the presence of sufficient oxygen and functional mitochondria, which is closely linked to rapid cancer cell proliferation [4]. This effect not only enhances cellular energy production and generates reducing power but also provides carbon precursors for the synthesis of nucleotides, lipids, and amino acids, thus promoting cancer progression [5]. As a glucose-oxidizing pathway that runs in parallel to upper glycolysis, the role of the pentose phosphate pathway (PPP) in the occurrence and development of malignant cancers is receiving more attention [6,7,8,9].
Briefly, the PPP is divided into two branches: the oxidative branch (oxPPP) and the non-oxidative branch (non-oxPPP). The oxPPP generates nicotinamide adenine dinucleotide phosphate (NADPH), a crucial electron donor for biosynthetic reactions and a protector against oxidative stress [10]. The non-oxPPP contributes to the production of ribose-5-phosphate (R5P), a precursor for nucleotide biosynthesis [11]. Thus, it integrates the biosynthetic demands with the maintenance of redox homeostasis, playing a pivotal role in supporting anabolic growth and protecting cells from oxidative stress [12]. During this process, glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme of the PPP, while transketolase (TKT) is the key enzyme in the non-oxPPP. A summary of the metabolic flux of the PPP is shown in Figure 1. However, whether and how the key enzymes or flux of the PPP function in GI cancer initiation and progression remain to be explored further.
In this review, we summarize the regulatory mechanisms of critical enzymes in the PPP during GI cancer pathogenesis. We also discuss the relationships between the PPP and elements of the tumor microenvironment (TME) in GI cancers. Furthermore, the role of the PPP in current pharmaceutical agents and therapeutic strategies for GI cancer treatment is highlighted.

2. Regulatory Mechanisms of the PPP in GI Cancers

The PPP plays an important role in GI cancers. Generally, enzymes like G6PD and TKT regulate cell proliferation, invasion, and metastasis, thereby promoting tumorigenesis, progression, and treatment resistance. Abnormal activation of the PPP is related to genetic mutations, further driving cancer progression [13]. Additionally, the PPP generates NADPH necessary for redox homeostasis and influences inflammation progression, which is also related to the development of GI cancers [14]. The multifaceted regulatory mechanisms of the PPP in various GI cancers are elucidated as follows.

2.1. Esophageal Cancer

In esophageal squamous cell carcinoma (ESCC), G6PD is an independent prognostic factor [15]. Polo-like kinase 1 (PLK1) coordinates biosynthesis during cell cycle progression and promotes cancer cell growth by directly increasing G6PD phosphorylation and activating the PPP [16]. Research has also established that the knock-out of PLK1 promoted ferroptosis through the inhibition of the PPP in ESCC [17]. In rat duodenal reflux models, chronic bile acid exposure triggers G6PD overexpression and nuclear factor kappa B (NF-κB) activation, potentially inducing genetic mutations and facilitating ESCC progression [13]. DNA polymerase ι promotes ESCC proliferation by activating G6PD and shunting glucose flux towards the PPP through O-GlcNAc transferase (OGT)-promoted O-GlcNAcylation [18].
Transketolase-like-1 (TKTL1), one of the three isoforms of TKT, is involved in the regulation of multiple cancer-related events [19]. TKTL1 overexpression is linked to the heightened aggressiveness of ESCC. Specifically, TKTL1 expression is positively correlated with the expression of several cell proliferation-related genes and pro-metastasis genes and is negatively correlated with the expression of apoptosis-related genes from the BCL-2 family and anti-metastasis genes [20]. In addition, Liu et al. found that high-mobility group AT-hook 1 (HMGA1), a structural transcriptional factor, upregulates the expression of TKT by enhancing the binding of specificity protein 1 (Sp1) to TKT promoter, thereby promoting ESCC tumorigenesis [21].

2.2. Gastric Cancer

In gastric cancer (GC), LINC00242 competitively combined miR-1-3p, thus relieving miR-1-3p-mediated suppression on G6PD and promoting aerobic glycolysis and GC progression [22]. As a nuclear receptor, Rev-erbα binds to the promoters of the 6-phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) and G6PD genes, thereby inhibiting their transcription. Consequently, reduced expressions of Rev-erbα significantly increase the proliferation of GC and are positively correlated with the advanced TMN stage and poorer prognosis of patients [23].
TKTL1 is a biomarker for the poor prognosis of GC patients, and an elevated expression of TKTL1 in patients is associated with reduced chemosensitivity to docetaxel, oxaliplatin, and S-1 [24,25,26]. In addition, with shRNA silencing TKTL1 in GC cells, their proliferation and tumor growth were inhibited, while the cell cycle was delayed in the G0/G1 phase [27].

2.3. Colorectal Cancer

In colorectal cancer (CRC), G6PD maintains redox balance and shields cancer cells from oxidative stress, contributing to cancer progression [28]. Zhang et al. highlighted the ability of p21-activated kinase 4 (PAK4) to boost G6PD activity through enhanced murine double minute 2 (MDM2)-mediated p53 ubiquitination degradation [29]. Furthermore, G6PD phosphorylation, controlled by neuronal differentiation 1 (NeuroD1) and tyrosine kinase c-Src, activates the PPP [30,31]. The Ras-related C3 botulinum toxin substrate 1 (Rac1) upregulates the expression of the sex-determining region Y-box 9 (SOX9) through the PI3K/AKT signaling pathway. SOX9 directly binds to the promoters of Hexokinase 2 (HK2) and G6PD, enhancing their transcription. This increases glycolysis and promotes the PPP, ultimately promoting the proliferation, invasion, and migration of CRC [32]. Pre-B-cell leukemia transcription factor 3 (PBX3) has been found to bind directly to the G6PD promoter, leading to PPP stimulation and enhancing the production of nucleotides and NADPH [33]. Otherwise, circNOLC1 interacts with AZGP1 under Yin Yang 1 (YY1) regulation to activate the mTOR/SREBP1 signaling pathway, resulting in c-Met overexpression and G6PD activation [34]. Interestingly, YY1 plays multifaceted roles in CRC. First, the YY1/ELFN1-AS1/TP53/G6PD axis is identified as a regulator axis of G6PD, linking its oncogenic activity with tumor cell metabolic reprogramming [35]. Second, another YY1-mediated regulation of the PPP in CRC is not through p53 but rather through the direct activation of G6PD transcription by YY1 [36]. Lastly, circAGFG1 was found to drive metastasis and stemness in CRC by modulating the YY1/CTNNB1 axis, enhancing G6PD activity [37]. Furthermore, the overexpression of ATPase cation transporting 13A2 (ATP13A2) not only enhances the nuclear localization of transcription factor EB (TFEB) but also inhibits its phosphorylation, leading to an increased expression of 6-phosphogluconate dehydrogenase (6PGD) and elevated activity of the PPP [38].
TKT interacts with glucose-regulated protein 78 (GRP78) and promotes CRC metastasis by regulating Akt phosphorylation [39]. Sirtuin 5 (SIRT5), a member of the NAD+-dependent class III histone deacetylase family, directly interacts with TKT, leading to its demalonylation and activation, which enhances the PPP and DNA replication, ultimately delivering poor prognosis [40]. Additionally, the high expression of TKT is linked to the upregulation of the Notch signaling pathway, thereby enhancing the proliferation and migration of CRC cells [41]. Elevated TKTL1 expression in CRC enhances glucose metabolism independent of oxygen and matrix degradation, which is fueled by lactate production, thus promoting metastasis [42]. When oncogenic signals suppress p16, it leads to uninhibited cell cycle progression and allows cells to bypass oncogene-induced senescence. p16 suppression activates the mechanistic target of rapamycin complex 1 (mTORC1) signaling, which in turn increases the expression of ribose-5-phosphate isomerase A (RPIA), a key enzyme in the PPP. The upregulation of RPIA enhances nucleotide synthesis, leading to an increased production of deoxyribonucleotides, which are essential for DNA replication and proliferation [43]. Importantly, RPIA transcends its traditional enzymatic role by entering the nucleus to form a complex with adenomatous polyposis coli (APC) and β-catenin. This interaction prevents the phosphorylation, ubiquitination, and degradation of β-catenin, leading to its upregulation [44]. This process is important as the aberrant activation of the Wnt/β-catenin signaling pathway is a common oncogenic event in CRC. Persistent Wnt/β-catenin signaling facilitates the epithelial–mesenchymal transition (EMT), accelerating CRC invasion and metastasis [45]. Moreover, the TP53-induced glycolysis and apoptosis regulator (TIGAR) primarily reduces glycolysis and increases PPP flux, thereby boosting the production of R5P and NADPH, which are important for intestinal regeneration and are associated with the prevalence of CRC [46,47].

2.4. Liver Cancer

In liver cancer, elevated G6PD expression is significantly associated with metastasis and poor prognosis of hepatocellular carcinoma (HCC) in patients. Mechanically, G6PD promotes EMT by upregulating the STAT3 signaling pathway, which ultimately enhances migration and invasion in HCC [48]. In a rat model of hepatocarcinogenesis, where early preneoplastic foci and nodules progressed towards HCC, metabolic changes were found to be characterized by enhanced activity of the PPP and reduced oxidative phosphorylation (OXPHOS) through the upregulation of nuclear factor erythroid 2-related factor 2 (NRF2), leading to the overexpression of G6PD [49]. Interestingly, NRF2 activation is also evident in hepatitis B virus (HBV) infection. In hepatocytes, HBV stimulates G6PD expression through HBV X protein (HBx) in an NRF2 activation-dependent pathway. HBx binds to the UBA and PB1 domains of the adaptor protein p62, enhancing the interaction between p62 and the NRF2 repressor kelch-like ECH-associated protein 1 (Keap1), forming an HBx–p62–Keap1 complex in the cytoplasm. This complex sequesters Keap1, reducing its inhibition of NRF2, leading to NRF2 activation and, subsequently, increasing G6PD transcription [50]. Another study confirmed that RNA interference targeting G6PD significantly reduced HBV replication, decreasing it by fivefold via the IFN pathway [51]. This indicates that the mechanism related to the NRF2/G6PD axis plays an important role in HBV-related liver cancer development and progression. In HCC, phosphatase and tensin homolog located on chromosome 10 (PTEN) has also been reported to negatively regulate G6PD through three distinct mechanisms. First, PTEN activates glycogen synthase kinase-3β (GSK3β), which phosphorylates T cell leukemia 1 (Tcl1), thereby inhibiting Tcl1’s interaction with heterogeneous nuclear ribonucleoprotein K (hnRNPK). Second, PTEN binds to hnRNPK, preventing the cleavage of G6PD pre-mRNA. Third, PTEN directly interacts with G6PD, inhibiting its dimerization and activity. These findings suggest that the PTEN/Tcl1/hnRNPK/G6PD axis could be a potential therapeutic target to improve the prognosis for HCC patients [52]. In HCC, aldolase B (Aldob) potentiates the p53-mediated inhibition of G6PD through the formation of an Aldob–G6PD–p53 complex, which is independent of Aldob enzymatic activity [53]. Furthermore, Bcl-2-associated athanogene 3 (BAG3) directly interacts with G6PD and suppresses PPP flux, de novo DNA synthesis, and cell growth in HCC [54]. Interestingly, some researchers have observed that although both G6PD and malic enzyme mRNA expression are increased, G6PD activity is inhibited in both hyperplastic liver and HCC by peroxisome proliferators, while malic enzyme activity is elevated to support NADPH production and cholesterol synthesis [55]. Additionally, the amplification of c-MYC has been observed in HCC patients, which drives cholesterol synthesis [56]. Hu et al. found that c-MYC promoted a positive feedback loop between cholesterol synthesis and the PPP, which drove the proliferation of malignant hepatocytes. In the meantime, mR-206 directly repressed the expression of HMGCR and G6PD, thereby disrupting the positive feedback loop [57]. Moreover, 6PGD has been identified as an independent prognostic factor for HCC patients, with high expression levels correlating with worse prognosis and beneficial efficacy of immunotherapy [58]. NRF2 is overexpressed in HCC and directly binds to the antioxidant response element in the 6PGD promoter region, enhancing its expression. Increased 6PGD expression, in turn, upregulates NRF2, forming a positive feedback loop between NRF2 and 6PGD. This loop ultimately leads to increased cell proliferation, survival, and migration in HCC [59].
TKT has been identified as a driver of HCC development by counteracting oxidative stress through the NRF2/KEAP1/BTB and CNC homolog 1 (BACH1) pathway [60]. Except for its metabolic function, the nuclear localization of TKT has been demonstrated to promote HCC through the activation of the epidermal growth factor receptor (EGFR) pathway in a non-metabolic manner [61]. As a Ser-Thr kinase, vaccinia-related kinase 2 (VRK2) can promote Thr287 phosphorylation of TKT and then facilitate E3 ubiquitin ligase F-Box and Leucine-Rich Repeat Protein 6 (FBXL6)-mediated ubiquitination and activation of TKT. Activated TKT further upregulated programmed death-ligand 1 (PD-L1) and VRK2 expression by decreasing reactive oxygen species (ROS) accumulation and mTOR activation, resulting in immune evasion and HCC metastasis [62]. Moreover, Zheng et al. revealed an interaction between TKT and SH2 domain-containing 5 (SH2D5), induced by HBx, which promotes HCC cell proliferation [63]. The overexpression of RPIA in HCC leads to ERK phosphorylation, increased lipid synthesis, and the activation of the β-catenin signaling pathway, ultimately prom oting cancer progression [64]. Compared to normal hepatocytes, HCC cells have a significantly lower fructose metabolism rate and ROS level due to the expression of the high-activity ketohexokinase (KHK)-A isoform. As a protein kinase, KHK-A phosphorylates and activates phosphoribosyl pyrophosphate synthetase 1 (PRPS1) to promote PPP-dependent de novo nucleic acid synthesis and HCC formation [65].

2.5. Pancreatic Cancer

In pancreatic ductal adenocarcinoma (PDAC), patients with high glucose metabolism levels have a worse prognosis. Mechanically, these patients have high glucose transporter 1 (GLUT1) and low Aldob expression, leading to increased glycolytic flux, G6PD activity, and pyrimidine biosynthesis [66]. In addition, PDAC distant metastases have resulted in a core pentose conversion pathway, which converts glucose-derived metabolites into 6PGD substrate, thereby hyperactivating 6PGD to support tumor growth [67].
Histone H3 lysine 4 trimethylation (H3K4me3) protein was specifically recruited to the promoter of TKT, which is facilitated by S100 calcium-binding protein A11 (S100A11) interacting with SET and MYND domain-containing 3 (SMYD3), thus enhancing PPP flux and further promoting PDAC progression [68]. The short isoform of the prolactin receptor (PRLR) activates the Hippo pathway by interacting with NIMA-related kinase 9 (NEK9), thus inhibiting G6PD and TKT expression and contributing to proliferation inhibition in PDAC [69]. Moreover, oncogenic KRAS has been found to activate a MAPK-dependent signaling pathway, leading to MYC upregulation and transcription of the RPIA, which facilitates nucleotide biosynthesis to support PDAC growth [70]. The long non-coding RNA (lncRNA) growth arrest-specific 5 (GAS5) plays a vital role in the emergence of the CD133+ population, representing tumor-initiating cells that result in tumor relapse. In PDAC, the CD133+ cell population was found to redirect glucose to the PPP, which was predominantly biosynthetic. Despite being quiescent in nature, these cells did not use it immediately for nucleotide synthesis [71].

2.6. Other Cancers

In gastrointestinal stromal tumors (GISTs), when HIF-1α binds to the 6PGD promoter sequence, it upregulates 6PGD expression, leading to increased NADPH production. This rise in NADPH counteracts ROS-induced oxidative damage, stimulating GIST cells to progress from the G1 phase to the S phase of the cell cycle [72]. In cholangiocarcinoma, the overexpression of G6PD is also associated with decreased mitochondrial ROS and increased cisplatin resistance, which could be reversed by chloroquine via the inhibition of the autophagy lysosome pathway [73].
The regulatory axis and function of G6PD and other key enzymes in the PPP in GI cancers are shown in Figure 2 and Table 1.

3. The PPP and the TME in GI Cancers

The TME encompasses a range of cells, including immune cells, cancer-associated fibroblasts (CAFs), endothelial cells, pericytes, and other tissue-residing cells [79]. While previously seen as bystanders to tumorigenesis, these host cells are now recognized as key players in the development and advancement of cancer. The PPP plays a significant role in the dynamic interplay between tumor cells and the TME. Tumor cells within the TME can adapt to the intricate microenvironmental conditions by reprogramming the PPP. Therefore, exploring the relationships between the PPP and TME’s elements can provide insights into the crosstalk between tumor cells and the complex TME, which is summarized in Figure 3.

3.1. The PPP and Nutrient Deprivation

The progression of tumor growth is accompanied by an enlargement in volume, potentially resulting in restricted nutritional support for the tumor cells [80]. Hence, the deficiency of glucose, serum, and amino acids is a characteristic of the TME.
Glucose deprivation enhances the binding of coactivator-associated arginine methyltransferase 1 (CARM1) and RPIA to induce the arginine 42 methylation of RPIA, thereby increasing the activity of RPIA and amplifying oxPPP flux, which contributes to the survival of CRC cells [74]. In addition, glucose deprivation and hydroxyethyl disulfide (HEDS) trigger p53-independent metabolic stress, including the loss of oxPPP function, thiol homeostasis, and sensitivity to radiation-induced oxidative stress in CRC cells [81]. Furthermore, PPP flux is increased when proline oxidase (POX) activity is enhanced due to glucose deprivation, leading to a boost in NADPH production. This increase in NADPH, in turn, contributes to the elevation of ATP levels through the proline cycle [82].
Numerous cancer cells rely upon glutamine, the most abundant amino acid in the blood, to replenish intermediates used for macromolecule biosynthesis [83]. Glutamine deprivation can increase the expression of G6PD through NRF2 activation, suggesting that targeting oxPPP enzymes and glutamine catabolism together is a strategy to combat CRC [84]. Moreover, De Falco et al. found that N-acetyltransferase 8-like (NAT8L) silenced HCC cells acquired proliferative advantage depending on glutamine oxidation. Specifically, the downregulation of NAT8L increases cytosolic aspartate levels to promote glucose flux into the PPP, thus boosting purine biosynthesis and ensuring HCC cell proliferation [85].
As for serum deprivation, it was found that macroH2A1-depleted HepG2 cells were insensitive to serum exhaustion. Specifically, the depletion of macroH2A1 in HCC cells leads to a significant enhancement of the PPP, which is crucial for providing precursors for nucleotide synthesis and supports the cancer stem cell-like metabolic phenotype [86].
LncRNAs also participate in the metabolic reprogramming of GI cancers under serum deprivation. For example, the stability and level of LINC01615 increase in a m6A-dependent manner under serum deprivation, thereby enhancing the expression of G6PD, the activation of PPP, and CRC cell survival [87].

3.2. The PPP and Hypoxia

Hypoxia within tumors arises due to the rapid proliferation of cancer cells and the insufficient balance between blood vessel development and oxygen availability. This state of low oxygen is a common, constant, and complex condition in the TME [88]. Facing the shift from normoxia to hypoxia, cells predominantly depend on the increased expression of hypoxia-inducible factors (HIFs) and the activation of HIF signaling pathways [89].
Singh et al. found that MUC1 physically interacts with HIF-1α and p300 in a hypoxia-dependent manner and facilitates the recruitment of HIF-1α and p300 on glycolytic gene promoters [90]. Consequently, PPP flux is enhanced by MUC1 overexpression, which contributes to nucleotide synthesis, resulting in radiation and gemcitabine resistance in PDAC [91,92]. In HCC, hypoxia is an extrinsic factor inducing phosphofructokinase-fructose bisphosphatase 4 (PFKFB4) expression in HCC in a HIF-1-dependent manner, which shifts the equilibrium from glycolysis to the PPP and alleviates cellular stress response, thus supporting HCC progression [93]. In addition, the nuclear translocation of TIGAR under genome stress or hypoxia increases, thereby activating the PPP and protecting HCC cells from DNA damage [94]. As a cell surface marker for cancer stem cells, CD44 ablation weakens the glycolytic phenotype of p53-deficient or hypoxic CRC cells and decreases metabolic flux to the PPP and glutathione (GSH) levels, which in turn increases the chemotherapy sensitivity of CRC cells [95].

3.3. The PPP and Acidosis

With a higher intracellular pH but a lower extracellular pH, the acidic niche is described as the acidosis of a tumor and its microenvironment, which is closely related to the hypoxia niche and lactate metabolism [96]. An acidic microenvironment redirects glucose away from lactate production and towards the oxPPP in order to produce NADPH and counter the increase in ROS present under acidosis. In terms of the mechanism, acidosis activates p53, which promotes the PPP, partly through the induction of G6PD expression [97]. Dichloroacetate, a kind of pyruvate dehydrogenase kinase (PDK) inhibitor, is more effective in decreasing the cell proliferation of acidic pH-adapted CRC cells compared to native CRC cells, which is related to a greater decrease in PPP activity [98]. In acidosis-adapted PDAC cells, PPP activity is enhanced compared to control cells, along with an increase in the inactivation of AMP-activated protein kinase (AMPK) and upregulation of matrix metalloproteinase-1 (MMP1), leading to higher proliferation, invasion, and metastasis ability [99].

3.4. The PPP and Tumor-Infiltrating Immunocytes

Throughout the stages of tumor progression, there is a dynamic and continuous interaction between tumor cells and the tumor-infiltrating immunocytes within the tumor immune microenvironment. These tumor-infiltrating immune cells include T lymphocytes, B lymphocytes, natural killer cells, neutrophils, tumor-associated macrophages, myeloid-derived suppressor cells, and dendritic cells. Each plays a distinct role in either promoting or inhibiting the progression of tumors within the microenvironment [100].
Within the inflammatory process, immune cells such as M1 macrophages and T-helper 17 cells undergo metabolic alterations characterized by an increased glucose uptake, reliance on glycolysis, and upregulated PPP. Conversely, cells with anti-inflammatory functions, like M2 macrophages, regulatory T cells, and quiescent memory T cells, exhibit decreased glycolytic activity and increased oxidative metabolism [101]. Modulating carbohydrate kinase-like (CARKL) intricately links the metabolic pathways and functional outcomes of M1 and M2 macrophages. When CARKL is downregulated, it decreases sedoheptulose-7-phosphate (S7P) and increases G6PD activity. This shift favors the pro-inflammatory M1 macrophage phenotype, enhancing the production of tumor necrosis factor α (TNFα) and interleukin-6 (IL-6), promoting oxPPP flux, and increasing NADPH production and ROS detoxification. Conversely, the upregulation of CARKL increases S7P levels, supporting the anti-inflammatory M2 macrophage phenotype [102].
Furthermore, the expression of PPP enzymes in immune cells is pivotal to their function. Tumor cells induce H3K9me3 deposition at the promoter of G6PD, resulting in decreased G6PD and granzyme B expression in tumor-specific cytotoxic T cells [103]. Although T cell proliferation depends on glycolysis, the differentiation of T effector cells requires ROS signaling. Genetic ablation or pharmacologic inhibition of the PPP enzyme 6PGD in the oxPPP promotes differentiation towards CD8+ T effector cells, enhancing tumoricidal activity and immunotherapy [104]. Considering that high levels of NADPH and GSH are essential for the formation and maintenance of CD8+ T memory cells, the cytosolic phosphoenolpyruvate carboxykinase (PCK1) increases glycogenesis to fuel PPP flux [105]. Additionally, a deficiency of TKT in regulatory T cells impairs their suppressive capability, induced by uncontrolled OXPHOS, lower α-ketoglutarate levels, and DNA hypermethylation [106].

3.5. The PPP and the Mechanical Microenvironment

The mechanical microenvironment within tumors, particularly concerning CAFs, plays a crucial role in shaping the TME and influencing tumor growth, metastasis, and resistance to therapy. CAFs significantly contribute to the deposition and remodeling of the extracellular matrix (ECM), altering the physical and biochemical TME in which cancer cells interact [107].
G6PD is highly expressed and activates the NF-κB signaling pathway, thereby promoting the production of hepatocyte growth factor (HGF) in gastric cancer-associated mesenchymal stem cells (GCMSCs). HGF, in turn, enhances GC cell proliferation and metastasis by upregulating HK2 [108]. In PDAC, the absence of focal adhesion kinase (FAK) in a subset of CAFs escalates tumor growth and boosts glycolysis in cancer cells due to the enrichment of the cytokine signaling pathway. Proteomics analysis has shown elevated levels of two key enzymes in the oxPPP, G6PD, and 6PGD in PDAC cells exposed to a FAK-depleted and CAF-conditioned medium [109]. As prominent stromal cells in PDAC, pancreatic stellate cells (PSCs) promote tumor progression through stromal-derived factor-1α (SDF-1α) and IL-6 secretion, which triggers cell proliferation via NRF2-mediated metabolic reprogramming and ROS detoxification. Moreover, G6PD downregulation or inhibition can attenuate this PSC-induced proliferation [77]. The loss of attachment to the ECM can initiate a range of cellular responses that influence cell survival. Detachment from the ECM triggers anoikis, a form of caspase-mediated apoptosis, characterized by significant metabolic disruptions. Upon ECM detachment, cells experience a notable reduction in ATP levels due to impaired glucose uptake, thus diminishing PPP flux and resulting in elevated ROS levels. The lack of glucose transport further compounds this metabolic impairment, which is crucial for ATP production through both glycolysis and the PPP. In matrix-detached cells, the downregulation of G6PD exacerbates oxidative stress by reducing NADPH production and increasing ROS accumulation. However, the overexpression of oncogenes like ERBB2 can rescue these cells by stabilizing EGFR and activating the PI3K pathway, restoring glucose uptake and PPP flux, reducing ROS levels, and promoting cell survival through enhanced ATP generation and fatty acid oxidation [110]. The activation of serum and glucocorticoid kinase-1 (SGK1) is instrumental in anchorage-independent growth during ECM detachment. Mechanically, SGK1 activation promotes glucose uptake, PPP flux, glyceraldehyde-3-phosphate (G3P) production, and ATP generation, thereby enhancing CRC cell survival [111].

4. The Role of the PPP in Therapeutic Strategies for GI Cancers

The PPP is instrumental in influencing therapeutic outcomes in GI cancers, impacting chemoradiotherapy, targeted therapy, immunotherapy, combination therapy, and emerging therapy. Below, we will elaborate on these categories, detailing the underlying mechanisms of therapeutic strategies related to the PPP. Meanwhile, a detailed discussion of the impact of non-coding RNAs (ncRNAs) and therapeutic agents in the treatment of GI cancers is presented in Table 2 and Table 3, respectively.

4.1. Chemoradiotherapy

Personalized medicine is being used increasingly in cancer treatment, with a significant focus on discovering new biomarkers and therapeutic targets. Both chemotherapy and radiotherapy are now more precisely tailored to fit each patient’s unique profile. In ESCC, reduced expression of PLK1 can inhibit the PPP, leading to decreased levels of NADPH and GSH. This disruption triggers ferroptosis and enhances the sensitivity of cancer cells to paclitaxel, cisplatin, and radiotherapy [17]. Another study further demonstrated that silencing PKM2 increased cisplatin sensitivity in ESCC by inhibiting the PPP [112]. SET-domain-containing 2 (SETD2) deficiency leads to the upregulation of GLUT1 to meet the high glucose demand of PDAC cells. Concurrently, SETD2 deficiency directly inhibits the transcription of TKT, compromising nucleotide synthesis. The synergistic effect of these alterations increases the sensitivity of SETD2-deficient PDAC cells to gemcitabine when glycolysis is restricted [113]. In addition, PRLR reduces nucleotide synthesis through miRNA-induced G6PD and TKT inhibition, thus enhancing the sensitivity of PDAC cells to gemcitabine [114].
Table 2. Non-coding RNA related to the PPP in GI cancers.
Table 2. Non-coding RNA related to the PPP in GI cancers.
GI Cancer TypeNcRNARoleFunctionsMechanismsReference
CRCLINC01615Oncogene↑Survival, ↑nucleotide and lipid synthesis, ↓ROS production, ↑oxaliplatin resistance, and ↑PPP fluxSerum starvation/↓METTL3/↑LINC01615/competitive binding with hnRNPA1/↑G6PD[87]
CircNOLC1Oncogene↑Proliferation, ↑migration, ↑liver metastasis, and ↑PPP fluxYY1/↑CircNOLC1/AZGP1/↑mTOR/SREBP1 signaling/↑G6PD;
YY1/↑CircNOLC1/↓miR-212-5p/↑c-Met/↑G6PD
[34]
Circ_0003215Tumor suppressor gene↓Proliferation, ↓migration, ↓invasion, ↓metastasis, and ↓PPP fluxCirc_0003215/↓miR-663b/↑DLG4/↓G6PD[115]
ELFN1-AS1Oncogene↑Proliferation, ↑migration, ↑invasion, ↓apoptosis, and↑PPP fluxYY1/↑ELFN1-AS1/↓TP53/↑G6PD [35]
Lnc-APTumor suppressor gene↓Oxaliplatin resistance, ↑ROS accumulation, ↑apoptosis, and ↓PPP fluxLnc-AP encoded pep-AP/↓TAL [116]
miR-124Tumor suppressor gene↓Growth, ↓nucleotide synthesis, and ↓PPP fluxmiR-124/↓PRPS1 and RPIA.[117]
HCCmiR-206Tumor suppressor gene↓Proliferation, ↓lipid accumulation, and ↓PPP fluxmiR-206/↓G6PD[118]
miR-206Tumor suppressor gene↓Growth, ↓cholesterol synthesis, and ↓PPP fluxmiR-206/↓G6PD and HMGCR[57]
miR-122, miR-1Tumor suppressor gene↓Viability and ↓PPP fluxmiR-122 and miR-1/↓G6PD [119]
PDACGAS5Oncogene↓Proliferation,↑quiescence, ↑metastasis, ↑invasion, and ↑PPP fluxSox2/↑GAS5/↓glucocorticoid receptor transcriptional activity[71]
miR-4763-3p, miR-3663-5pTumor suppressor gene↓Nucleotide synthesis, ↑gemcitabine sensitivity, and ↓PPP fluxPRLR/↑miR-4763-3p/↓G6PD
PRLR/↑miR-3663-5p/↓TKT
[114]
GCLINC00242Oncogene↑Aerobic glycolysis, ↑proliferation, ↓apoptosis, and ↓PPP fluxLINC00242/↓miR-1-3p/↑G6PD[22]
“↑” means upregulation and “↓” means downregulation.
Research shows that inhibitor of differentiation 1 (ID1) promotes G6PD expression and PPP activation via the Wnt/β-catenin/c-MYC signaling pathway, contributing to oxaliplatin resistance and poor prognosis in patients [75]. Inhibiting G6PD reduces NADPH and GSH production, impairing their ability to clear ROS. This ROS-mediated damage subsequently enhances the apoptosis of CRC cells induced by oxaliplatin [28]. POU domain class 2 transcription factor 1 (POU2F1) binds directly to the aldolase A (ALDOA) promoter, thereby strengthening PPP activity in CRC. Silencing POU2F1, however, significantly increased CRC sensitivity to oxaliplatin [120]. Treatment with diallyl disulfide (DADS) disrupts the PPP, leading to decreased production of 5-phosphate ribose-1-pyrophosphate (PRPP). This results in increased DNA damage, enhanced cell apoptosis, and reduced growth of CRC cells, which is related to ubiquitination and degradation of POU2F1 [121].
As for radiotherapy, MUC1 expression reduced radiation-induced cytotoxicity and DNA damage in PDAC by promoting glycolysis, the PPP, and nucleotide biosynthesis. Pretreatment with the glycolysis inhibitor 3-bromopyruvate overcame MUC1-mediated radiation resistance, both in vitro and in vivo, by decreasing glucose entry into nucleotide synthesis pathways and increasing DNA damage [91]. At the same time, high TKT expression leads to radiotherapy resistance in clinical HCC patients, demonstrating its role in enhancing the auto-PARylation of poly (ADP-ribose) polymerase 1 (PARP1) in response to DNA double-strand breaks [122].

4.2. Targeted Therapy

Since their invention in the early 2000s, tyrosine kinase inhibitors (TKIs) have gained prominence as one of the most effective pathway-directed anti-cancer agents. Some TKIs have been taken to clinical trials in GI cancers, while imatinib is the mainstay of medical treatment of GISTs in the first-line metastatic setting [123,124].
PDAC resistance to the EGFR inhibitor erlotinib is closely linked to the upregulation of the PPP. Erlotinib-resistant PDAC cells show increased G6PD levels, reduced glycolytic activity, and lower glycolytic metabolites, while elevated G6PD levels are attributed to the upregulation of the ID1 [78]. Furthermore, erlotinib treatment could suppress the development of HCC by blocking the effect of TKT nuclear localization and inhibiting the EGFR pathway [61]. PTEN binds to G6PD in HCC, preventing the formation of active G6PD dimers, whereas Tcl1 can counteract this inhibition. Importantly, knocking down Tcl1 increases HCC sensitivity to sorafenib. This indicates that the PTEN–Tcl1 interaction plays a role in sorafenib resistance in HCC through the PPP [52]. Similarly, a positive feedback loop between the PPP and PI3K/AKT signaling upregulates G6PD activity, contributing to regorafenib resistance in HCC. Consequently, the PI3K/AKT pathway inhibitor MK-2206 or G6PD inhibitor 6-aminonicotinamide (6AN) can counteract this resistance [125]. In GISTs, resistance to imatinib is closely related to the PPP. Prolonged imatinib treatment increases intracellular ROS levels, which then causes an adaptive rise in HIF-1α protein levels. This rise in HIF-1α is accompanied by an increased expression of 6PGD, leading to the upregulation of the PPP. The upregulation of the PPP mitigates ROS-induced damage, promotes cell proliferation, and inhibits apoptosis, promoting the growth of resistant tumor cells [72].
As a novel TKT inhibitor, oroxylin A directly binds to TKT in HCC, decreasing its activity and expression. This results in the accumulation of non-oxPPP substrates and the activation of the p53 signaling pathway, ultimately inducing apoptosis and causing cell cycle arrest [126].
Table 3. Therapeutic agents related to the PPP in GI cancers.
Table 3. Therapeutic agents related to the PPP in GI cancers.
GI Cancer TypeAgentTargetCharacteristicsMechanismsReference
CRCM4IDPG6PDZoledronic acid derivative↑Unprenylated Rap1A, RhoA and CDC42,↓G6PD,↑ROS, ↓NADPH and GSH, ↓mevalonate pathway, and ↓PPP flux[127]
Ankaferd hemostat6PGDPlant extracts of Thymus vulgaris, Glycyrrhiza glabra, Vitis vinifera, Alpinia officinarum, and Urtica dioica↓6PGD,↑oxidative stress, and ↓PPP flux[128]
GO-203MUC1 C-terminal subunitD-amino acid cell-penetrating peptide↓AKT-S6K1-elF4A pathway, ↓TIGAR,↓GSH/mitochondrial transmembrane potential, ↑ROS, and ↓PPP flux[129]
Piperlongumine/auranofinGlutathione S-transferase π/Thioredoxin reductaseNatural alkaloid from piper longum L/trialkylphosphine gold complex↑NRF2 target genes (G6PD), ↓CD44v9-positive fraction, ↓tumor formation and growth, and ↓PPP flux[130]
INK128/AvemarmTORmTOR1/2 inhibitor/fermented wheat germ extract↓PPP enzymes (G6PD, PGD, TKT), ↓NADPH/NADP+ and GSH/GSSG ratios, and ↓PPP flux[131]
HalofuginoneAkt/mTORC1 signalingDerivative of the febrifugine↓G6PD, ↑ROS, ↓NADPH, ↓glycolysis and lipid biosynthesis, and ↓PPP flux[132]
Epicatechin gallateG6PD, TKTCatechins in green tea and grape↓Enzymatic activity of G6PD and TKT, ↓de novo synthesis of RNA ribose, and ↓PPP flux[133]
AspirinG6PD, TKTSalicylate↑Acetylation of G6PD and TKT,↓activity of G6PD, and ↓PPP flux[134]
Cu2O@Au nanocompositesFerroptosis pathwayNanoparticles↓GSH, ↑H2O2, ↑ferroptosis, ↑immune therapy response, and ↓PPP flux[135]
Phy@PLGdH nanosheets6PGDNanoparticles↓NADPH and nucleotide synthesis, ↑radiation-therapy-mediated oxidative stress and DNA damage, ↑immunogenic cell death, and ↓PPP flux[136]
HCCOroxylin ATKTHerbal extracts↓TKT activity, ↑non-oxPPP substrates, ↑p53 signaling, and ↓PPP flux[126]
CP-91149Glycogen phosphorylaseIndole carboxamide↑Effect of 6AN, ↑phosphorylation of AMPK, and ↓PPP flux[137]
ZerumbonePI3K/AKT/mTOR and STAT3 signaling pathwaysSesquiterpene derived from the ginger plant zingiber zerumbet↓Enzymes in PPP (G6PD, RPIA, RPE, TKT, and TAL), and ↓PPP flux[138]
PDACHypericinG6PDNaphthodianthrone, anthraquinone derivative, and active constituents of Hypericum↓G6PD, ↓GSH, ↑ROS, ↑effect of gemcitabine, and ↓PPP flux[139]
CholangiocarcinomaChloroquineAutophagy lysosome pathwayAntimalarials drug↓G6PD, ↑mitochondrial ROS, ↑cisplatin-induced apoptosis, and ↓PPP flux[73]
“↑” means upregulation and “↓” means downregulation.

4.3. Immunotherapy

Enzymes in the PPP participate in the survival, function, activation, and differentiation of T cells, which demonstrates their potential as a novel metabolic checkpoint for immunotherapy applications [103,104,140,141]. Dimethyl fumarate (DMF), a glyceraldehyde-3-phosphate dehydrogenase (GAPDH) inhibitor, which has been shown to treat autoimmune diseases, was found to promote oxPPP by increasing G6PD expression in tumor cells. In addition, DMF enhanced the efficiency of interleukin-2 (IL-2) therapy while eliminating severe toxicity induced by IL-2 therapy [142]. A recent study has demonstrated that two DNA-binding agents, trabectedin (TRB) and lurbinectedin (LUR), can prompt human macrophages toward a pro-inflammatory state and enhance their anti-tumor activity through metabolic reprogramming, involving ROS production, changes to the mitochondrial inner membrane potential, and the activation of the oxPPP [143].
The development of nano-formulations has increased attention to nano-targeted drug-delivery systems. One study demonstrated the creation of Cu2O@Au nanocomposites, which induce oxidative stress pathways, including the PPP. Cu2O@Au releases Cu2O and Au nanoparticles upon delivery to cancer cells, promoting H2O2 production and disrupting GSH generation via the PPP. This leads to lipid peroxide accumulation and ferroptosis induction. Ferroptosis triggers immunogenic cell death, dendritic cell maturation, and T cell infiltration and enhances PD-L1 antibody efficacy [135]. Glutaryl-CoA dehydrogenase (GCDH) suppresses HCC by enhancing the crotonylation of key enzymes in the PPP and reducing R5P and lactate production, thereby limiting the Warburg effect. The suppression of the PPP induces oxidative stress and cell senescence, creating an environment antagonistic to tumor growth. The depletion of GCDH renders HCC cells more vulnerable to anti-PD-1 therapy, suggesting that targeting GCDH and the PPP is a promising approach for HCC treatment [144].

4.4. Combination Therapy

HIF-1α-induced glucose uptake in PDAC cells enhances both glycolysis and the non-oxPPP, resulting in increased pyrimidine biosynthesis and higher cytoplasmic deoxycytidine triphosphate (dCTP) levels. This elevation in dCTP can diminish gemcitabine’s effectiveness in inhibiting DNA replication in PDAC. In contrast, combining the HIF-1α inhibitors YC1 or digoxin with gemcitabine has been shown to induce apoptosis in PDAC and enhance mouse survival without causing weight loss. Thus, targeting the HIF-1α pathway or pyrimidine biosynthesis may overcome gemcitabine resistance in PDAC and offer a potential combination therapy option with gemcitabine for treating PDAC [92]. Inhibiting G6PD with 6AN has been shown to increase ROS levels, induce cell cycle arrest, and enhance the sensitivity of resistant cells to erlotinib. This highlights the potential of combining 6AN with erlotinib as a promising strategy to address drug resistance in PDAC [78].
Researchers have developed Phy@PLGdH nanosheets, which have been shown to enhance immunogenic cell death, induced by a combination of a 6PGD inhibitor physcion and radiation therapy, thus creating a potent in situ tumor vaccination and amplifying oxidative stress and DNA damage [136]. Furthermore, combining chemotherapy drugs with physcion is more effective in suppressing HCC growth and survival compared to chemotherapy alone [145].
Additionally, the overexpression of LINC01615 competitively binds hnRNPA1 to promote the splicing of G6PD precursor mRNA and upregulate G6PD expression. Elevated G6PD increases nucleotide and lipid synthesis, reduces ROS production, and minimizes oxidative damage, thereby promoting cell survival under nutrient starvation and oxaliplatin treatment in CRC, leading to chemoresistance. Therefore, targeting LINC01615 in combination with oxaliplatin treatment is proposed as a potential alternative strategy against chemoresistance in CRC [87]. Another short peptide encoded by lnc-AP, namely, pep-AP, also exhibits potential for similar combination therapy with oxaliplatin. Specifically, pep-AP interacts with the transaldolase (TAL) protein to inhibit its expression. This action weakens the PPP, leading to ROS accumulation and apoptosis, thereby sensitizing CRC to oxaliplatin both in vitro and in vivo [116].

4.5. Emerging Therapy

As the next-generation sequencing and omics technologies have advanced and been integrated into medical treatment, the field of gene therapy for cancer has seen remarkable progress over the past few decades. Gene therapy utilizes vectors to introduce genetic material into host cells for therapeutic gene modification [146]. Considering that some key PPP enzymes play an important role in the development of GI cancers, silencing these genes in cancer cells is an attractive treatment option. For example, silencing G6PD with lentivirus or non-viral gene delivery vector enhances oxaliplatin anti-tumor effects in CRC xenografts and PDX models [28]. The activation of a tumor suppressor gene like p53, PTEN, and some ncRNAs that regulate PPP is another promising gene therapy strategy [52,147,148].
To date, a significant proportion of gene therapy research in oncology has been integrated with immunotherapeutic approaches. Among these, chimeric antigen receptor (CAR)-T cell therapy stands out as a promising technique within cancer immunotherapy [149]. In the context of CAR-T cell therapy for cancer, metabolic reprogramming following the pharmacological or genetic ablation of isocitrate dehydrogenase 2 (IDH2) redirects glucose metabolism towards the PPP. This metabolic shift enhances the antioxidative capacity and attenuates CAR-T cell exhaustion, which is particularly beneficial under nutrient-restricted conditions, thereby improving tumor eradication and sustaining CAR-T cell persistence [150].

5. Conclusions and Perspectives

Since Horecker et al. fully elucidated the entire PPP in the 1950s, the PPP has been gaining more attention regarding its roles in health and disease [151]. Many studies have shown that cancer cells modulate PPP flux either directly or indirectly to meet their growth demand. The cancer cells need increased PPP flux to generate high NADPH levels to counteract ROS and maintain redox homeostasis. However, ROS can promote tumorigenesis during cancer initiation, but after the tumor is established, it can restrain cancer cell survival and growth. For example, inflammatory bowel disease (IBD) is recognized as a significant risk factor for CRC [152]. TKT ablation in intestinal epithelium leads to extensive mucosal erosion, aberrant tight junctions, impaired barrier function, and increased inflammatory cell infiltration and ROS production in mice, showing similar phenotypes to IBD [153]. In contrast, high expression of TKT promotes the proliferation of CRC cells and increases CRC migration and invasion abilities [39]. The different expression levels of TKT indicate that manipulating PPP enzymes at different stages of GI cancers may have different effects. It is crucial to transition from broad generalizations about the impact of the PPP on cancer development to the time-dependent, location-dependent, and molecule-specific roles that the PPP plays throughout cancer progression.
Over the past decade, significant advancements in the study of metabolic enzymes have revolutionized our comprehension of their roles. Historically, these enzymes were known to catalyze metabolic reactions and were exclusively tied to metabolism-related pathways. Recent insights have expanded this view, revealing a more complex and multifaceted involvement of metabolic enzymes in cellular processes [154]. As mentioned earlier, the nuclear translocation of TKT activates the EGFR pathway, thereby promoting HCC development [61]. Consequently, the non-metabolic function of the PPP enzymes elicited by pathological, especially oncogenic signals or gene mutations, should be taken into consideration.
Considering that the inhibition of the PPP could overcome chemoresistance and radioresistance in GI cancer cells, the synergistic and additive effects of PPP inhibition and other therapeutic strategies are promising. Unfortunately, there are still no specific PPP enzyme inhibitors applied in clinical practice due to their potential toxicity, low efficacy, and off-target effects [155]. Inhibiting the PPP at the organismal level to selectively restrain tumorigenesis without significant adverse physiological consequences remains a challenge. Thus, there is a need for the development of safe and effective PPP inhibitors. Gaining a better understanding of the regulatory mechanisms governing the PPP and the selective advantages it confers to GI cancer cells could unlock novel therapeutic opportunities in GI cancer treatment.

Author Contributions

J.Q., Z.Y., J.Y. and H.W. conceived the manuscript; J.Q., Z.Y., H.Z. and W.W. searched for the references and made the outline; J.Q., Z.Y., H.Z. and W.W. wrote the initial draft; J.Q. and Z.Y. edited the figures and tables; J.Q., Z.Y., H.Z. and W.W. checked all the references and formatted the manuscript; and J.Y. and H.W. revised and supervised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (No. 81773065 and No. 82173113) and the Natural Science Foundation of Zhejiang Province (LY21H160023).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No datasets were generated or analyzed during the current study.

Acknowledgments

The figures were created using BioRender (Biorender.com).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

6AN: 6-aminonicotinamide; 6PGD: 6-phosphogluconate dehydrogenase; ALDOA: aldolase A; Aldob: aldolase B; ATP13A2: ATPase cation transporting 13A2; AMPK: AMP-activated protein kinase; APC: adenomatous polyposis coli; BACH1: BTB and CNC homolog 1; BAG3: Bcl-2 associated athanogene 3; CAF: cancer-associated fibroblast; CAR: chimeric antigen receptor; CARM1: coactivator-associated arginine methyltransferase 1; CARKL: carbohydrate kinase-like; CRC: colorectal cancer; DADS: diallyl disulfide; dCTP: deoxycytidine triphosphate; DMF: dimethyl fumarate; ECM: extracellular matrix; EGFR: epidermal growth factor receptor; EMT: epithelial-mesenchymal transition; ESCC: esophageal squamous cell carcinoma; FAK: focal adhesion kinase; G3P: glyceraldehyde-3-phosphate; G6PD: glucose-6-phosphate dehydrogenase; GAS5: growth arrest-specific 5; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GC: gastric cancer; GCDH: glutaryl-CoA dehydrogenase; GCMSC: gastric cancer-associated mesenchymal stem cells; GI: gastrointestinal; GIST: gastrointestinal stromal tumor; GLUT1: glucose transporter 1; GRP78: glucose-regulated protein 78; GSH: glutathione; GSK3β: glycogen synthase kinase-3β; H3K4me3: histone H3 lysine 4 trimethylation; HBV: hepatitis B virus; HBx: HBV X protein; HCC: hepatocellular carcinoma; HEDS: hydroxyethyl disulfide; HMGA1: high-mobility group AT-hook 1; hnRNPK: heterogeneous nuclear ribonucleoprotein K; HGF: hepatocyte growth factor; HK2: hexokinase 2; HIF: hypoxia-inducible factor; Keap1: Kelch-like ECH-associated protein 1; KHK: ketohexokinase; IBD: inflammatory bowel disease; ID1: inhibitor of differentiation 1; IDH2: isocitrate dehydrogenase 2; IL-2: interleukin-2; IL-6: interleukin-6; LUR: lurbinectedin; MDM2: murine double minute 2; MMP1: matrix metalloproteinase-1; mTORC1: mechanistic target of rapamycin complex 1; NADPH: nicotinamide adenine dinucleotide phosphate; NAT8L: N-acetyltransferase 8-like; NEK9: NIMA-related kinase 9;NeuroD1: neuronal differentiation 1; NF-κB: nuclear factor kappa B; Non-oxPPP: non-oxidative branch of pentose phosphate pathway; NRF2: nuclear factor erythroid 2-related factor 2; OGT: O-GlcNAc transferase; OxPPP: oxidative branch of pentose phosphate pathway; OXPHOS: oxidative phosphorylation; PAK4: p21-activated kinase 4; PARP1: poly (ADP-ribose) polymerase 1; PBX3: pre-B-cell leukemia transcription factor 3; PCK1: phosphoenolpyruvate carboxykinase; PDAC: pancreatic ductal adenocarcinoma; PDK: pyruvate dehydrogenase kinase; PLK1: polo-like kinase 1; PFKFB3: 6-phosphofructokinase-2/fructose-2,6-bisphosphatase 3; PFKFB4: phosphofructokinase-fructose bisphosphatase 4; POU2F1: POU domain class 2 transcription factor 1; POX: proline oxidase; PPP: pentose phosphate pathway; PRLR: prolactin receptor; PRPS1: phosphoribosyl pyrophosphate synthetase 1; PRPP: 5-phosphate ribose-1-pyrophosphate; PSC: pancreatic stellate cell; PTEN: phosphatase and tensin homolog located on chromosome 10; R5P: ribose-5-phosphate; Rac1: ras-related C3 botulinum toxin substrate 1; ROS: reactive oxygen species; RPIA: ribose-5-phosphate isomerase A; S7P: sedoheptulose-7-phosphate; S100A11: S100 calcium-binding protein A11; SETD2: SET-domain-containing 2; SGK1: serum and glucocorticoid kinase-1; SH2D5: SH2 domain-containing 5; SIRT5: sirtuin 5; SMYD3: SET and MYND domain-containing 3; Sp1: specificity protein 1; SOX9: sex-determining region Y-box 9; TAL: transaldolase; Tcl1: T cell leukemia 1; TIGAR: TP53-induced glycolysis and apoptosis regulator; TFEB: transcription factor EB; TNFα: tumor necrosis factor α; TKIs: tyrosine kinase inhibitors; TKT: transketolase; TKTL1: transketolase-like-1; TME: tumor microenvironment; TRB: trabectedin; and YY1: Yin Yang 1.

References

  1. Peery, A.F.; Crockett, S.D.; Murphy, C.C.; Jensen, E.T.; Kim, H.P.; Egberg, M.D.; Lund, J.L.; Moon, A.M.; Pate, V.; Barnes, E.L.; et al. Burden and Cost of Gastrointestinal, Liver, and Pancreatic Diseases in the United States: Update 2021. Gastroenterology 2022, 162, 621–644. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  3. Pavlova, N.N.; Thompson, C.B. The Emerging Hallmarks of Cancer Metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef] [PubMed]
  4. DeBerardinis, R.J.; Chandel, N.S. We need to talk about the Warburg effect. Nat. Metab. 2020, 2, 127–129. [Google Scholar] [CrossRef] [PubMed]
  5. Hsu, P.P.; Sabatini, D.M. Cancer cell metabolism: Warburg and beyond. Cell 2008, 134, 703–707. [Google Scholar] [CrossRef]
  6. Jin, L.; Zhou, Y. Crucial role of the pentose phosphate pathway in malignant tumors. Oncol. Lett. 2019, 17, 4213–4221. [Google Scholar] [CrossRef] [PubMed]
  7. Jiang, P.; Du, W.; Wu, M. Regulation of the pentose phosphate pathway in cancer. Protein Cell 2014, 5, 592–602. [Google Scholar] [CrossRef] [PubMed]
  8. Ghanem, N.; El-Baba, C.; Araji, K.; El-Khoury, R.; Usta, J.; Darwiche, N. The Pentose Phosphate Pathway in Cancer: Regulation and Therapeutic Opportunities. Chemotherapy 2021, 66, 179–191. [Google Scholar] [CrossRef]
  9. Patra, K.C.; Hay, N. The pentose phosphate pathway and cancer. Trends Biochem. Sci. 2014, 39, 347–354. [Google Scholar] [CrossRef] [PubMed]
  10. Wamelink, M.M.; Struys, E.A.; Jakobs, C. The biochemistry, metabolism and inherited defects of the pentose phosphate pathway: A review. J. Inherit. Metab. Dis. 2008, 31, 703–717. [Google Scholar] [CrossRef]
  11. Stincone, A.; Prigione, A.; Cramer, T.; Wamelink, M.M.; Campbell, K.; Cheung, E.; Olin-Sandoval, V.; Grüning, N.M.; Krüger, A.; Tauqeer Alam, M.; et al. The return of metabolism: Biochemistry and physiology of the pentose phosphate pathway. Biol. Rev. Camb. Philos. Soc. 2015, 90, 927–963. [Google Scholar] [CrossRef]
  12. TeSlaa, T.; Ralser, M.; Fan, J.; Rabinowitz, J.D. The pentose phosphate pathway in health and disease. Nat. Metab. 2023, 5, 1275–1289. [Google Scholar] [CrossRef] [PubMed]
  13. Munemoto, M.; Mukaisho, K.I.; Miyashita, T.; Oyama, K.; Haba, Y.; Okamoto, K.; Kinoshita, J.; Ninomiya, I.; Fushida, S.; Taniura, N.; et al. Roles of the hexosamine biosynthetic pathway and pentose phosphate pathway in bile acid-induced cancer development. Cancer Sci. 2019, 110, 2408–2420. [Google Scholar] [CrossRef]
  14. Perl, A.; Hanczko, R.; Telarico, T.; Oaks, Z.; Landas, S. Oxidative stress, inflammation and carcinogenesis are controlled through the pentose phosphate pathway by transaldolase. Trends Mol. Med. 2011, 17, 395–403. [Google Scholar] [CrossRef]
  15. Wang, X.; Li, X.; Zhang, X.; Fan, R.; Gu, H.; Shi, Y.; Liu, H. Glucose-6-phosphate dehydrogenase expression is correlated with poor clinical prognosis in esophageal squamous cell carcinoma. Eur. J. Surg. Oncol. 2015, 41, 1293–1299. [Google Scholar] [CrossRef] [PubMed]
  16. Ma, X.; Wang, L.; Huang, D.; Li, Y.; Yang, D.; Li, T.; Li, F.; Sun, L.; Wei, H.; He, K.; et al. Polo-like kinase 1 coordinates biosynthesis during cell cycle progression by directly activating pentose phosphate pathway. Nat. Commun. 2017, 8, 1506. [Google Scholar] [CrossRef] [PubMed]
  17. Zhao, M.; Lu, T.; Bi, G.; Hu, Z.; Liang, J.; Bian, Y.; Feng, M.; Zhan, C. PLK1 regulating chemoradiotherapy sensitivity of esophageal squamous cell carcinoma through pentose phosphate pathway/ferroptosis. Biomed. Pharmacother. 2023, 168, 115711. [Google Scholar] [CrossRef]
  18. Su, Z.; Gao, A.; Li, X.; Zou, S.; He, C.; Wu, J.; Ding, W.Q.; Zhou, J. DNA Polymerase Iota Promotes Esophageal Squamous Cell Carcinoma Proliferation Through Erk-OGT-Induced G6PD Overactivation. Front. Oncol. 2021, 11, 706337. [Google Scholar] [CrossRef] [PubMed]
  19. Hao, S.; Meng, Q.; Sun, H.; Li, Y.; Li, Y.; Gu, L.; Liu, B.; Zhang, Y.; Zhou, H.; Xu, Z.; et al. The role of transketolase in human cancer progression and therapy. Biomed. Pharmacother. 2022, 154, 113607. [Google Scholar] [CrossRef]
  20. Li, J.; Zhu, S.C.; Li, S.G.; Zhao, Y.; Xu, J.R.; Song, C.Y. TKTL1 promotes cell proliferation and metastasis in esophageal squamous cell carcinoma. Biomed. Pharmacother. 2015, 74, 71–76. [Google Scholar] [CrossRef] [PubMed]
  21. Liu, M.J.; Zhao, Y.; Li, Q.T.; Lei, X.Y.; He, K.Y.; Guo, J.R.; Yang, J.Y.; Yan, Z.H.; Wu, D.H.; Zhang, L.; et al. HMGA1 promotes the progression of esophageal squamous cell carcinoma by elevating TKT-mediated upregulation of pentose phosphate pathway. Cell Death Dis. 2024, 15, 541. [Google Scholar] [CrossRef]
  22. Deng, P.; Li, K.; Gu, F.; Zhang, T.; Zhao, W.; Sun, M.; Hou, B. LINC00242/miR-1-3p/G6PD axis regulates Warburg effect and affects gastric cancer proliferation and apoptosis. Mol. Med. 2021, 27, 9. [Google Scholar] [CrossRef] [PubMed]
  23. Tao, L.; Yu, H.; Liang, R.; Jia, R.; Wang, J.; Jiang, K.; Wang, Z. Rev-erbα inhibits proliferation by reducing glycolytic flux and pentose phosphate pathway in human gastric cancer cells. Oncogenesis 2019, 8, 57. [Google Scholar] [CrossRef] [PubMed]
  24. Staiger, W.I.; Coy, J.F.; Grobholz, R.; Hofheinz, R.D.; Lukan, N.; Post, S.; Schwarzbach, M.H.; Willeke, F. Expression of the mutated transketolase TKTL1, a molecular marker in gastric cancer. Oncol. Rep. 2006, 16, 657–661. [Google Scholar] [CrossRef] [PubMed]
  25. Song, Y.; Liu, D.; He, G. TKTL1 and p63 are biomarkers for the poor prognosis of gastric cancer patients. Cancer Biomark. 2015, 15, 591–597. [Google Scholar] [CrossRef]
  26. Li, Y.; Xu, C.; Wang, B.; Xu, F.; Ma, F.; Qu, Y.; Jiang, D.; Li, K.; Feng, J.; Tian, S.; et al. Proteomic characterization of gastric cancer response to chemotherapy and targeted therapy reveals new therapeutic strategies. Nat. Commun. 2022, 13, 5723. [Google Scholar] [CrossRef] [PubMed]
  27. Yuan, W.; Wu, S.; Guo, J.; Chen, Z.; Ge, J.; Yang, P.; Hu, B.; Chen, Z. Silencing of TKTL1 by siRNA inhibits proliferation of human gastric cancer cells in vitro and in vivo. Cancer Biol. Ther. 2010, 9, 710–716. [Google Scholar] [CrossRef] [PubMed]
  28. Ju, H.Q.; Lu, Y.X.; Wu, Q.N.; Liu, J.; Zeng, Z.L.; Mo, H.Y.; Chen, Y.; Tian, T.; Wang, Y.; Kang, T.B.; et al. Disrupting G6PD-mediated Redox homeostasis enhances chemosensitivity in colorectal cancer. Oncogene 2017, 36, 6282–6292. [Google Scholar] [CrossRef]
  29. Zhang, X.; Zhang, X.; Li, Y.; Shao, Y.; Xiao, J.; Zhu, G.; Li, F. PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis. 2017, 8, e2820. [Google Scholar] [CrossRef]
  30. Li, Z.; He, Y.; Li, Y.; Li, J.; Zhao, H.; Song, G.; Miyagishi, M.; Wu, S.; Kasim, V. NeuroD1 promotes tumor cell proliferation and tumorigenesis by directly activating the pentose phosphate pathway in colorectal carcinoma. Oncogene 2021, 40, 6736–6747. [Google Scholar] [CrossRef]
  31. Ma, H.; Zhang, F.; Zhou, L.; Cao, T.; Sun, D.; Wen, S.; Zhu, J.; Xiong, Z.; Tsau, M.T.; Cheng, M.L.; et al. c-Src facilitates tumorigenesis by phosphorylating and activating G6PD. Oncogene 2021, 40, 2567–2580. [Google Scholar] [CrossRef]
  32. Liang, J.; Liu, Q.; Xia, L.; Lin, J.; Oyang, L.; Tan, S.; Peng, Q.; Jiang, X.; Xu, X.; Wu, N.; et al. Rac1 promotes the reprogramming of glucose metabolism and the growth of colon cancer cells through upregulating SOX9. Cancer Sci. 2023, 114, 822–836. [Google Scholar] [CrossRef] [PubMed]
  33. Luo, X.; Wei, M.; Li, W.; Zhao, H.; Kasim, V.; Wu, S. PBX3 promotes pentose phosphate pathway and colorectal cancer progression by enhancing G6PD expression. Int. J. Biol. Sci. 2023, 19, 4525–4538. [Google Scholar] [CrossRef] [PubMed]
  34. Yuan, M.; Zhang, X.; Yue, F.; Zhang, F.; Jiang, S.; Zhou, X.; Lv, J.; Zhang, Z.; Sun, Y.; Chen, Z.; et al. CircNOLC1 Promotes Colorectal Cancer Liver Metastasis by Interacting with AZGP1 and Sponging miR-212-5p to Regulate Reprogramming of the Oxidative Pentose Phosphate Pathway. Adv. Sci. 2023, 10, e2205229. [Google Scholar] [CrossRef]
  35. Wu, F.; Zhang, W.; Wei, H.; Ma, H.; Leng, G.; Zhang, Y. lncRNA ELFN1-AS1 promotes proliferation, migration and invasion and suppresses apoptosis in colorectal cancer cells by enhancing G6PD activity. Acta Biochim. Biophys. Sin. 2023, 55, 649–660. [Google Scholar] [CrossRef]
  36. Wu, S.; Wang, H.; Li, Y.; Xie, Y.; Huang, C.; Zhao, H.; Miyagishi, M.; Kasim, V. Transcription Factor YY1 Promotes Cell Proliferation by Directly Activating the Pentose Phosphate Pathway. Cancer Res. 2018, 78, 4549–4562. [Google Scholar] [CrossRef] [PubMed]
  37. Zhang, L.; Dong, X.; Yan, B.; Yu, W.; Shan, L. CircAGFG1 drives metastasis and stemness in colorectal cancer by modulating YY1/CTNNB1. Cell Death Dis. 2020, 11, 542. [Google Scholar] [CrossRef]
  38. Zhang, F.; Wu, Z.; Yu, B.; Ning, Z.; Lu, Z.; Li, L.; Long, F.; Hu, Q.; Zhong, C.; Zhang, Y.; et al. ATP13A2 activates the pentose phosphate pathway to promote colorectal cancer growth though TFEB-PGD axis. Clin. Transl. Med. 2023, 13, e1272. [Google Scholar] [CrossRef]
  39. Li, M.; Zhao, X.; Yong, H.; Xu, J.; Qu, P.; Qiao, S.; Hou, P.; Li, Z.; Chu, S.; Zheng, J.; et al. Transketolase promotes colorectal cancer metastasis through regulating AKT phosphorylation. Cell Death Dis. 2022, 13, 99. [Google Scholar] [CrossRef] [PubMed]
  40. Wang, H.L.; Chen, Y.; Wang, Y.Q.; Tao, E.W.; Tan, J.; Liu, Q.Q.; Li, C.M.; Tong, X.M.; Gao, Q.Y.; Hong, J.; et al. Sirtuin5 protects colorectal cancer from DNA damage by keeping nucleotide availability. Nat. Commun. 2022, 13, 6121. [Google Scholar] [CrossRef]
  41. Zhang, L.; Huang, Z.; Cai, Q.; Zhao, C.; Xiao, Y.; Quan, X.; Tang, C.; Gao, J. Inhibition of Transketolase Improves the Prognosis of Colorectal Cancer. Front. Med. 2022, 9, 837143. [Google Scholar] [CrossRef]
  42. Langbein, S.; Zerilli, M.; Zur Hausen, A.; Staiger, W.; Rensch-Boschert, K.; Lukan, N.; Popa, J.; Ternullo, M.P.; Steidler, A.; Weiss, C.; et al. Expression of transketolase TKTL1 predicts colon and urothelial cancer patient survival: Warburg effect reinterpreted. Br. J. Cancer 2006, 94, 578–585. [Google Scholar] [CrossRef]
  43. Buj, R.; Chen, C.W.; Dahl, E.S.; Leon, K.E.; Kuskovsky, R.; Maglakelidze, N.; Navaratnarajah, M.; Zhang, G.; Doan, M.T.; Jiang, H.; et al. Suppression of p16 Induces mTORC1-Mediated Nucleotide Metabolic Reprogramming. Cell Rep. 2019, 28, 1971–1980.e78. [Google Scholar] [CrossRef] [PubMed]
  44. Chou, Y.T.; Jiang, J.K.; Yang, M.H.; Lu, J.W.; Lin, H.K.; Wang, H.D.; Yuh, C.H. Identification of a noncanonical function for ribose-5-phosphate isomerase A promotes colorectal cancer formation by stabilizing and activating β-catenin via a novel C-terminal domain. PLoS Biol. 2018, 16, e2003714. [Google Scholar] [CrossRef] [PubMed]
  45. Sun, L.; Xing, J.; Zhou, X.; Song, X.; Gao, S. Wnt/β-catenin signalling, epithelial-mesenchymal transition and crosslink signalling in colorectal cancer cells. Biomed. Pharmacother. 2024, 175, 116685. [Google Scholar] [CrossRef]
  46. Tang, J.; Chen, L.; Qin, Z.H.; Sheng, R. Structure, regulation, and biological functions of TIGAR and its role in diseases. Acta Pharmacol. Sin. 2021, 42, 1547–1555. [Google Scholar] [CrossRef] [PubMed]
  47. Cheung, E.C.; Athineos, D.; Lee, P.; Ridgway, R.A.; Lambie, W.; Nixon, C.; Strathdee, D.; Blyth, K.; Sansom, O.J.; Vousden, K.H. TIGAR is required for efficient intestinal regeneration and tumorigenesis. Dev. Cell 2013, 25, 463–477. [Google Scholar] [CrossRef] [PubMed]
  48. Lu, M.; Lu, L.; Dong, Q.; Yu, G.; Chen, J.; Qin, L.; Wang, L.; Zhu, W.; Jia, H. Elevated G6PD expression contributes to migration and invasion of hepatocellular carcinoma cells by inducing epithelial-mesenchymal transition. Acta Biochim. Biophys. Sin. 2018, 50, 370–380. [Google Scholar] [CrossRef] [PubMed]
  49. Kowalik, M.A.; Guzzo, G.; Morandi, A.; Perra, A.; Menegon, S.; Masgras, I.; Trevisan, E.; Angioni, M.M.; Fornari, F.; Quagliata, L.; et al. Metabolic reprogramming identifies the most aggressive lesions at early phases of hepatic carcinogenesis. Oncotarget 2016, 7, 32375–32393. [Google Scholar] [CrossRef] [PubMed]
  50. Liu, B.; Fang, M.; He, Z.; Cui, D.; Jia, S.; Lin, X.; Xu, X.; Zhou, T.; Liu, W. Hepatitis B virus stimulates G6PD expression through HBx-mediated Nrf2 activation. Cell Death Dis. 2015, 6, e1980. [Google Scholar] [CrossRef] [PubMed]
  51. Hu, H.; Ding, X.; Yang, Y.; Zhang, H.; Li, H.; Tong, S.; An, X.; Zhong, Q.; Liu, X.; Ma, L.; et al. Changes in glucose-6-phosphate dehydrogenase expression results in altered behavior of HBV-associated liver cancer cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 307, G611–G622. [Google Scholar] [CrossRef]
  52. Hong, X.; Song, R.; Song, H.; Zheng, T.; Wang, J.; Liang, Y.; Qi, S.; Lu, Z.; Song, X.; Jiang, H.; et al. PTEN antagonises Tcl1/hnRNPK-mediated G6PD pre-mRNA splicing which contributes to hepatocarcinogenesis. Gut 2014, 63, 1635–1647. [Google Scholar] [CrossRef]
  53. Li, M.; He, X.; Guo, W.; Yu, H.; Zhang, S.; Wang, N.; Liu, G.; Sa, R.; Shen, X.; Jiang, Y.; et al. Aldolase B suppresses hepatocellular carcinogenesis by inhibiting G6PD and pentose phosphate pathways. Nat. Cancer 2020, 1, 735–747. [Google Scholar] [CrossRef]
  54. Kong, D.H.; Li, S.; Du, Z.X.; Liu, C.; Liu, B.Q.; Li, C.; Zong, Z.H.; Wang, H.Q. BAG3 elevation inhibits cell proliferation via direct interaction with G6PD in hepatocellular carcinomas. Oncotarget 2016, 7, 700–711. [Google Scholar] [CrossRef] [PubMed]
  55. Rao, K.N.; Elm, M.S.; Kelly, R.H.; Chandar, N.; Brady, E.P.; Rao, B.; Shinozuka, H.; Eagon, P.K. Hepatic hyperplasia and cancer in rats: Metabolic alterations associated with cell growth. Gastroenterology 1997, 113, 238–248. [Google Scholar] [CrossRef]
  56. Ally, A.; Balasundaram, M.; Carlsen, R.; Chuah, E.; Clarke, A.; Dhalla, N.; Ferguson, M.L. Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell 2017, 169, 1327–1341.e23. [Google Scholar] [CrossRef] [PubMed]
  57. Hu, J.; Liu, N.; Song, D.; Steer, C.J.; Zheng, G.; Song, G. A positive feedback between cholesterol synthesis and the pentose phosphate pathway rather than glycolysis promotes hepatocellular carcinoma. Oncogene 2023, 42, 2892–2904. [Google Scholar] [CrossRef]
  58. Liu, T.; Qi, J.; Wu, H.; Wang, L.; Zhu, L.; Qin, C.; Zhang, J.; Zhu, Q. Phosphogluconate dehydrogenase is a predictive biomarker for immunotherapy in hepatocellular carcinoma. Front. Oncol. 2022, 12, 993503. [Google Scholar] [CrossRef] [PubMed]
  59. Ong, A.J.; Saeidi, S.; Chi, N.H.K.; Kim, S.J.; Kim, D.H.; Kim, S.H.; Park, S.A.; Cha, Y.N.; Na, H.K.; Surh, Y.J. The positive feedback loop between Nrf2 and phosphogluconate dehydrogenase stimulates proliferation and clonogenicity of human hepatoma cells. Free Radic. Res. 2020, 54, 906–917. [Google Scholar] [CrossRef]
  60. Xu, I.M.; Lai, R.K.; Lin, S.H.; Tse, A.P.; Chiu, D.K.; Koh, H.Y.; Law, C.T.; Wong, C.M.; Cai, Z.; Wong, C.C.; et al. Transketolase counteracts oxidative stress to drive cancer development. Proc. Natl. Acad. Sci. USA 2016, 113, E725–E734. [Google Scholar] [CrossRef] [PubMed]
  61. Qin, Z.; Xiang, C.; Zhong, F.; Liu, Y.; Dong, Q.; Li, K.; Shi, W.; Ding, C.; Qin, L.; He, F. Transketolase (TKT) activity and nuclear localization promote hepatocellular carcinoma in a metabolic and a non-metabolic manner. J. Exp. Clin. Cancer Res. 2019, 38, 154. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, J.; Lin, X.T.; Yu, H.Q.; Fang, L.; Wu, D.; Luo, Y.D.; Zhang, Y.J.; Xie, C.M. Elevated FBXL6 expression in hepatocytes activates VRK2-transketolase-ROS-mTOR-mediated immune evasion and liver cancer metastasis in mice. Exp. Mol. Med. 2023, 55, 2162–2176. [Google Scholar] [CrossRef]
  63. Zheng, Y.; Ming, P.; Zhu, C.; Si, Y.; Xu, S.; Chen, A.; Wang, J.; Zhang, B. Hepatitis B virus X protein-induced SH2 domain-containing 5 (SH2D5) expression promotes hepatoma cell growth via an SH2D5-transketolase interaction. J. Biol. Chem. 2019, 294, 4815–4827. [Google Scholar] [CrossRef]
  64. Chou, Y.T.; Chen, L.Y.; Tsai, S.L.; Tu, H.C.; Lu, J.W.; Ciou, S.C.; Wang, H.D.; Yuh, C.H. Ribose-5-phosphate isomerase A overexpression promotes liver cancer development in transgenic zebrafish via activation of ERK and β-catenin pathways. Carcinogenesis 2019, 40, 461–473. [Google Scholar] [CrossRef]
  65. Li, X.; Qian, X.; Peng, L.X.; Jiang, Y.; Hawke, D.H.; Zheng, Y.; Xia, Y.; Lee, J.H.; Cote, G.; Wang, H.; et al. A splicing switch from ketohexokinase-C to ketohexokinase-A drives hepatocellular carcinoma formation. Nat. Cell Biol. 2016, 18, 561–571. [Google Scholar] [CrossRef]
  66. Li, Y.; Tang, S.; Shi, X.; Lv, J.; Wu, X.; Zhang, Y.; Wang, H.; He, J.; Zhu, Y.; Ju, Y.; et al. Metabolic classification suggests the GLUT1/ALDOB/G6PD axis as a therapeutic target in chemotherapy-resistant pancreatic cancer. Cell Rep. Med. 2023, 4, 101162. [Google Scholar] [CrossRef] [PubMed]
  67. Bechard, M.E.; Word, A.E.; Tran, A.V.; Liu, X.; Locasale, J.W.; McDonald, O.G. Pentose conversions support the tumorigenesis of pancreatic cancer distant metastases. Oncogene 2018, 37, 5248–5256. [Google Scholar] [CrossRef]
  68. Zeng, X.; Guo, H.; Liu, Z.; Qin, Z.; Cong, Y.; Ren, N.; Zhang, Y.; Zhang, N. S100A11 activates the pentose phosphate pathway to induce malignant biological behaviour of pancreatic ductal adenocarcinoma. Cell Death Dis. 2022, 13, 568. [Google Scholar] [CrossRef] [PubMed]
  69. Nie, H.; Huang, P.Q.; Jiang, S.H.; Yang, Q.; Hu, L.P.; Yang, X.M.; Li, J.; Wang, Y.H.; Li, Q.; Zhang, Y.F.; et al. The short isoform of PRLR suppresses the pentose phosphate pathway and nucleotide synthesis through the NEK9-Hippo axis in pancreatic cancer. Theranostics 2021, 11, 3898–3915. [Google Scholar] [CrossRef] [PubMed]
  70. Santana-Codina, N.; Roeth, A.A.; Zhang, Y.; Yang, A.; Mashadova, O.; Asara, J.M.; Wang, X.; Bronson, R.T.; Lyssiotis, C.A.; Ying, H.; et al. Oncogenic KRAS supports pancreatic cancer through regulation of nucleotide synthesis. Nat. Commun. 2018, 9, 4945. [Google Scholar] [CrossRef] [PubMed]
  71. Sharma, N.S.; Gnamlin, P.; Durden, B.; Gupta, V.K.; Kesh, K.; Garrido, V.T.; Dudeja, V.; Saluja, A.; Banerjee, S. Long non-coding RNA GAS5 acts as proliferation “brakes” in CD133+ cells responsible for tumor recurrence. Oncogenesis 2019, 8, 68. [Google Scholar] [CrossRef]
  72. Xu, K.; He, Z.; Chen, M.; Wang, N.; Zhang, D.; Yang, L.; Xu, Z.; Xu, H. HIF-1α regulates cellular metabolism, and Imatinib resistance by targeting phosphogluconate dehydrogenase in gastrointestinal stromal tumors. Cell Death Dis. 2020, 11, 586. [Google Scholar] [CrossRef]
  73. Qu, X.; Sheng, J.; Shen, L.; Su, J.; Xu, Y.; Xie, Q.; Wu, Y.; Zhang, X.; Sun, L. Autophagy inhibitor chloroquine increases sensitivity to cisplatin in QBC939 cholangiocarcinoma cells by mitochondrial ROS. PLoS ONE 2017, 12, e0173712. [Google Scholar] [CrossRef] [PubMed]
  74. Guo, J.; Zhang, Q.; Su, Y.; Lu, X.; Wang, Y.; Yin, M.; Hu, W.; Wen, W.; Lei, Q.Y. Arginine methylation of ribose-5-phosphate isomerase A senses glucose to promote human colorectal cancer cell survival. Sci. China Life Sci. 2020, 63, 1394–1405. [Google Scholar] [CrossRef]
  75. Yin, X.; Tang, B.; Li, J.H.; Wang, Y.; Zhang, L.; Xie, X.Y.; Zhang, B.H.; Qiu, S.J.; Wu, W.Z.; Ren, Z.G. ID1 promotes hepatocellular carcinoma proliferation and confers chemoresistance to oxaliplatin by activating pentose phosphate pathway. J. Exp. Clin. Cancer Res. 2017, 36, 166. [Google Scholar] [CrossRef] [PubMed]
  76. Ciou, S.C.; Chou, Y.T.; Liu, Y.L.; Nieh, Y.C.; Lu, J.W.; Huang, S.F.; Chou, Y.T.; Cheng, L.H.; Lo, J.F.; Chen, M.J.; et al. Ribose-5-phosphate isomerase A regulates hepatocarcinogenesis via PP2A and ERK signaling. Int. J. Cancer 2015, 137, 104–115. [Google Scholar] [CrossRef]
  77. Wu, Y.S.; Looi, C.Y.; Subramaniam, K.S.; Masamune, A.; Chung, I. Soluble factors from stellate cells induce pancreatic cancer cell proliferation via Nrf2-activated metabolic reprogramming and ROS detoxification. Oncotarget 2016, 7, 36719–36732. [Google Scholar] [CrossRef]
  78. Sharma, N.; Bhushan, A.; He, J.; Kaushal, G.; Bhardwaj, V. Metabolic plasticity imparts erlotinib-resistance in pancreatic cancer by upregulating glucose-6-phosphate dehydrogenase. Cancer Metab. 2020, 8, 19. [Google Scholar] [CrossRef]
  79. de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef]
  80. Parlani, M.; Jorgez, C.; Friedl, P. Plasticity of cancer invasion and energy metabolism. Trends Cell Biol. 2023, 33, 388–402. [Google Scholar] [CrossRef] [PubMed]
  81. Li, J.; Ward, K.M.; Zhang, D.; Dayanandam, E.; Denittis, A.S.; Prendergast, G.C.; Ayene, I.S. A bioactive probe of the oxidative pentose phosphate cycle: Novel strategy to reverse radioresistance in glucose deprived human colon cancer cells. Toxicol. In Vitro 2013, 27, 367–377. [Google Scholar] [CrossRef] [PubMed]
  82. Pandhare, J.; Donald, S.P.; Cooper, S.K.; Phang, J.M. Regulation and function of proline oxidase under nutrient stress. J. Cell Biochem. 2009, 107, 759–768. [Google Scholar] [CrossRef] [PubMed]
  83. Altman, B.J.; Stine, Z.E.; Dang, C.V. From Krebs to clinic: Glutamine metabolism to cancer therapy. Nat. Rev. Cancer 2016, 16, 619–634. [Google Scholar] [CrossRef]
  84. Polat, I.H.; Tarrado-Castellarnau, M.; Benito, A.; Hernandez-Carro, C.; Centelles, J.; Marin, S.; Cascante, M. Glutamine Modulates Expression and Function of Glucose 6-Phosphate Dehydrogenase via NRF2 in Colon Cancer Cells. Antioxidants 2021, 10, 1349. [Google Scholar] [CrossRef] [PubMed]
  85. De Falco, P.; Lazzarino, G.; Felice, F.; Desideri, E.; Castelli, S.; Salvatori, I.; Ciccarone, F.; Ciriolo, M.R. Hindering NAT8L expression in hepatocellular carcinoma increases cytosolic aspartate delivery that fosters pentose phosphate pathway and purine biosynthesis promoting cell proliferation. Redox Biol. 2023, 59, 102585. [Google Scholar] [CrossRef]
  86. Lo Re, O.; Douet, J.; Buschbeck, M.; Fusilli, C.; Pazienza, V.; Panebianco, C.; Castracani, C.C.; Mazza, T.; Li Volti, G.; Vinciguerra, M. Histone variant macroH2A1 rewires carbohydrate and lipid metabolism of hepatocellular carcinoma cells towards cancer stem cells. Epigenetics 2018, 13, 829–845. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, Y.; Xu, L.; Ren, Z.; Liu, X.; Song, J.; Zhang, P.; Zhang, C.; Gong, S.; Wu, N.; Zhang, X.; et al. LINC01615 maintains cell survival in adaptation to nutrient starvation through the pentose phosphate pathway and modulates chemosensitivity in colorectal cancer. Cell Mol. Life Sci. 2022, 80, 20. [Google Scholar] [CrossRef] [PubMed]
  88. Qiu, G.Z.; Jin, M.Z.; Dai, J.X.; Sun, W.; Feng, J.H.; Jin, W.L. Reprogramming of the Tumor in the Hypoxic Niche: The Emerging Concept and Associated Therapeutic Strategies. Trends Pharmacol. Sci. 2017, 38, 669–686. [Google Scholar] [CrossRef] [PubMed]
  89. Schito, L.; Semenza, G.L. Hypoxia-Inducible Factors: Master Regulators of Cancer Progression. Trends Cancer 2016, 2, 758–770. [Google Scholar] [CrossRef]
  90. Chaika, N.V.; Gebregiworgis, T.; Lewallen, M.E.; Purohit, V.; Radhakrishnan, P.; Liu, X.; Zhang, B.; Mehla, K.; Brown, R.B.; Caffrey, T.; et al. MUC1 mucin stabilizes and activates hypoxia-inducible factor 1 alpha to regulate metabolism in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2012, 109, 13787–13792. [Google Scholar] [CrossRef]
  91. Gunda, V.; Souchek, J.; Abrego, J.; Shukla, S.K.; Goode, G.D.; Vernucci, E.; Dasgupta, A.; Chaika, N.V.; King, R.J.; Li, S.; et al. MUC1-Mediated Metabolic Alterations Regulate Response to Radiotherapy in Pancreatic Cancer. Clin. Cancer Res. 2017, 23, 5881–5891. [Google Scholar] [CrossRef] [PubMed]
  92. Shukla, S.K.; Purohit, V.; Mehla, K.; Gunda, V.; Chaika, N.V.; Vernucci, E.; King, R.J.; Abrego, J.; Goode, G.D.; Dasgupta, A.; et al. MUC1 and HIF-1alpha Signaling Crosstalk Induces Anabolic Glucose Metabolism to Impart Gemcitabine Resistance to Pancreatic Cancer. Cancer Cell 2017, 32, 71–87.e77. [Google Scholar] [CrossRef]
  93. Kam, C.S.; Ho, D.W.; Ming, V.S.; Tian, L.; Sze, K.M.; Zhang, V.X.; Tsui, Y.M.; Husain, A.; Lee, J.M.; Wong, C.C.; et al. PFKFB4 Drives the Oncogenicity in TP53-Mutated Hepatocellular Carcinoma in a Phosphatase-Dependent Manner. Cell Mol. Gastroenterol. Hepatol. 2023, 15, 1325–1350. [Google Scholar] [CrossRef] [PubMed]
  94. Yu, H.P.; Xie, J.M.; Li, B.; Sun, Y.H.; Gao, Q.G.; Ding, Z.H.; Wu, H.R.; Qin, Z.H. TIGAR regulates DNA damage and repair through pentosephosphate pathway and Cdk5-ATM pathway. Sci. Rep. 2015, 5, 9853. [Google Scholar] [CrossRef]
  95. Tamada, M.; Nagano, O.; Tateyama, S.; Ohmura, M.; Yae, T.; Ishimoto, T.; Sugihara, E.; Onishi, N.; Yamamoto, T.; Yanagawa, H.; et al. Modulation of glucose metabolism by CD44 contributes to antioxidant status and drug resistance in cancer cells. Cancer Res. 2012, 72, 1438–1448. [Google Scholar] [CrossRef] [PubMed]
  96. Kato, Y.; Ozawa, S.; Miyamoto, C.; Maehata, Y.; Suzuki, A.; Maeda, T.; Baba, Y. Acidic extracellular microenvironment and cancer. Cancer Cell Int. 2013, 13, 89. [Google Scholar] [CrossRef]
  97. Lamonte, G.; Tang, X.; Chen, J.L.; Wu, J.; Ding, C.K.; Keenan, M.M.; Sangokoya, C.; Kung, H.N.; Ilkayeva, O.; Boros, L.G.; et al. Acidosis induces reprogramming of cellular metabolism to mitigate oxidative stress. Cancer Metab. 2013, 1, 23. [Google Scholar] [CrossRef] [PubMed]
  98. Schoonjans, C.A.; Joudiou, N.; Brusa, D.; Corbet, C.; Feron, O.; Gallez, B. Acidosis-induced metabolic reprogramming in tumor cells enhances the anti-proliferative activity of the PDK inhibitor dichloroacetate. Cancer Lett. 2020, 470, 18–28. [Google Scholar] [CrossRef]
  99. Chen, S.; Ning, B.; Song, J.; Yang, Z.; Zhou, L.; Chen, Z.; Mao, L.; Liu, H.; Wang, Q.; He, S.; et al. Enhanced pentose phosphate pathway activity promotes pancreatic ductal adenocarcinoma progression via activating YAP/MMP1 axis under chronic acidosis. Int. J. Biol. Sci. 2022, 18, 2304–2316. [Google Scholar] [CrossRef]
  100. Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef]
  101. O’Neill, L.A.; Hardie, D.G. Metabolism of inflammation limited by AMPK and pseudo-starvation. Nature 2013, 493, 346–355. [Google Scholar] [CrossRef] [PubMed]
  102. Haschemi, A.; Kosma, P.; Gille, L.; Evans, C.R.; Burant, C.F.; Starkl, P.; Knapp, B.; Haas, R.; Schmid, J.A.; Jandl, C.; et al. The sedoheptulose kinase CARKL directs macrophage polarization through control of glucose metabolism. Cell Metab. 2012, 15, 813–826. [Google Scholar] [CrossRef] [PubMed]
  103. Lu, C.; Yang, D.; Klement, J.D.; Colson, Y.L.; Oberlies, N.H.; Pearce, C.J.; Colby, A.H.; Grinstaff, M.W.; Ding, H.F.; Shi, H.; et al. G6PD functions as a metabolic checkpoint to regulate granzyme B expression in tumor-specific cytotoxic T lymphocytes. J. Immunother. Cancer 2022, 10, e003543. [Google Scholar] [CrossRef] [PubMed]
  104. Daneshmandi, S.; Cassel, T.; Lin, P.; Higashi, R.M.; Wulf, G.M.; Boussiotis, V.A.; Fan, T.W.; Seth, P. Blockade of 6-phosphogluconate dehydrogenase generates CD8(+) effector T cells with enhanced anti-tumor function. Cell Rep. 2021, 34, 108831. [Google Scholar] [CrossRef]
  105. Ma, R.; Ji, T.; Zhang, H.; Dong, W.; Chen, X.; Xu, P.; Chen, D.; Liang, X.; Yin, X.; Liu, Y.; et al. A Pck1-directed glycogen metabolic program regulates formation and maintenance of memory CD8(+) T cells. Nat. Cell Biol. 2018, 20, 21–27. [Google Scholar] [CrossRef]
  106. Liu, Q.; Zhu, F.; Liu, X.; Lu, Y.; Yao, K.; Tian, N.; Tong, L.; Figge, D.A.; Wang, X.; Han, Y.; et al. Non-oxidative pentose phosphate pathway controls regulatory T cell function by integrating metabolism and epigenetics. Nat. Metab. 2022, 4, 559–574. [Google Scholar] [CrossRef]
  107. Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, B.; Cai, T.; Huang, C.; Zang, X.; Sun, L.; Guo, S.; Wang, Q.; Chen, Z.; Zhao, Y.; Han, Z.; et al. G6PD-NF-κB-HGF Signal in Gastric Cancer-Associated Mesenchymal Stem Cells Promotes the Proliferation and Metastasis of Gastric Cancer Cells by Upregulating the Expression of HK2. Front. Oncol. 2021, 11, 648706. [Google Scholar] [CrossRef] [PubMed]
  109. Demircioglu, F.; Wang, J.; Candido, J.; Costa, A.S.H.; Casado, P.; de Luxan Delgado, B.; Reynolds, L.E.; Gomez-Escudero, J.; Newport, E.; Rajeeve, V.; et al. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat. Commun. 2020, 11, 1290. [Google Scholar] [CrossRef]
  110. Schafer, Z.T.; Grassian, A.R.; Song, L.; Jiang, Z.; Gerhart-Hines, Z.; Irie, H.Y.; Gao, S.; Puigserver, P.; Brugge, J.S. Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment. Nature 2009, 461, 109–113. [Google Scholar] [CrossRef]
  111. Mason, J.A.; Cockfield, J.A.; Pape, D.J.; Meissner, H.; Sokolowski, M.T.; White, T.C.; Valentín López, J.C.; Liu, J.; Liu, X.; Martínez-Reyes, I.; et al. SGK1 signaling promotes glucose metabolism and survival in extracellular matrix detached cells. Cell Rep. 2021, 34, 108821. [Google Scholar] [CrossRef] [PubMed]
  112. Fukuda, S.; Miyata, H.; Miyazaki, Y.; Makino, T.; Takahashi, T.; Kurokawa, Y.; Yamasaki, M.; Nakajima, K.; Takiguchi, S.; Mori, M.; et al. Pyruvate Kinase M2 Modulates Esophageal Squamous Cell Carcinoma Chemotherapy Response by Regulating the Pentose Phosphate Pathway. Ann. Surg. Oncol. 2015, 22 (Suppl. S3), S1461–S1468. [Google Scholar] [CrossRef] [PubMed]
  113. Shen, X.; Chen, Y.; Liu, M.; Shi, J.; Tang, Y.; Yang, X.; Xu, D.; Yao, H.; Lu, P.; Sun, Y.; et al. Glycolysis addiction compensating for a defective pentose phosphate pathway confers gemcitabine sensitivity in SETD2-deficient pancreatic cancer. Biochem. Biophys. Res. Commun. 2022, 615, 9–16. [Google Scholar] [CrossRef] [PubMed]
  114. Liao, Y.N.; Huang, P.Q.; Pan, H.; Gai, Y.Z.; Zhan, Y.F.; Li, S.X.; Nie, H.Z. Prolactin receptor potentiates chemotherapy through miRNAs-induced G6PD/TKT inhibition in pancreatic cancer. Faseb J. 2024, 38, e23705. [Google Scholar] [CrossRef]
  115. Chen, B.; Hong, Y.; Gui, R.; Zheng, H.; Tian, S.; Zhai, X.; Xie, X.; Chen, Q.; Qian, Q.; Ren, X.; et al. N6-methyladenosine modification of circ_0003215 suppresses the pentose phosphate pathway and malignancy of colorectal cancer through the miR-663b/DLG4/G6PD axis. Cell Death Dis. 2022, 13, 804. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, X.; Zhang, H.; Yin, S.; Yang, Y.; Yang, H.; Yang, J.; Zhou, Z.; Li, S.; Ying, G.; Ba, Y. lncRNA-encoded pep-AP attenuates the pentose phosphate pathway and sensitizes colorectal cancer cells to Oxaliplatin. EMBO Rep. 2022, 23, e53140. [Google Scholar] [CrossRef]
  117. Qiu, Z.; Guo, W.; Wang, Q.; Chen, Z.; Huang, S.; Zhao, F.; Yao, M.; Zhao, Y.; He, X. MicroRNA-124 reduces the pentose phosphate pathway and proliferation by targeting PRPS1 and RPIA mRNAs in human colorectal cancer cells. Gastroenterology 2015, 149, 1587–1598.e1511. [Google Scholar] [CrossRef] [PubMed]
  118. Wang, A.; Chen, B.; Jian, S.; Cai, W.; Xiao, M.; Du, G. miR-206-G6PD axis regulates lipogenesis and cell growth in hepatocellular carcinoma cell. Anticancer. Drugs 2021, 32, 508–516. [Google Scholar] [CrossRef]
  119. Barajas, J.M.; Reyes, R.; Guerrero, M.J.; Jacob, S.T.; Motiwala, T.; Ghoshal, K. The role of miR-122 in the dysregulation of glucose-6-phosphate dehydrogenase (G6PD) expression in hepatocellular cancer. Sci. Rep. 2018, 8, 9105. [Google Scholar] [CrossRef]
  120. Lin, J.; Xia, L.; Oyang, L.; Liang, J.; Tan, S.; Wu, N.; Yi, P.; Pan, Q.; Rao, S.; Han, Y.; et al. The POU2F1-ALDOA axis promotes the proliferation and chemoresistance of colon cancer cells by enhancing glycolysis and the pentose phosphate pathway activity. Oncogene 2022, 41, 1024–1039. [Google Scholar] [CrossRef] [PubMed]
  121. Xia, L.; Lin, J.; Peng, M.; Jiang, X.; Peng, Q.; Cui, S.; Zhang, W.; Li, S.; Wang, J.; Oyang, L.; et al. Diallyl disulfide induces DNA damage and growth inhibition in colorectal cancer cells by promoting POU2F1 ubiquitination. Int. J. Biol. Sci. 2024, 20, 1125–1141. [Google Scholar] [CrossRef]
  122. Geng, L.; Zhu, M.; Luo, D.; Chen, H.; Li, B.; Lao, Y.; An, H.; Wu, Y.; Li, Y.; Xia, A.; et al. TKT-PARP1 axis induces radioresistance by promoting DNA double-strand break repair in hepatocellular carcinoma. Oncogene 2024, 43, 682–692. [Google Scholar] [CrossRef] [PubMed]
  123. Hammel, P.; Huguet, F.; van Laethem, J.L.; Goldstein, D.; Glimelius, B.; Artru, P.; Borbath, I.; Bouché, O.; Shannon, J.; André, T.; et al. Effect of Chemoradiotherapy vs Chemotherapy on Survival in Patients With Locally Advanced Pancreatic Cancer Controlled After 4 Months of Gemcitabine With or Without Erlotinib: The LAP07 Randomized Clinical Trial. JAMA 2016, 315, 1844–1853. [Google Scholar] [CrossRef] [PubMed]
  124. Blay, J.Y.; Kang, Y.K.; Nishida, T.; von Mehren, M. Gastrointestinal stromal tumours. Nat. Rev. Dis. Primers 2021, 7, 22. [Google Scholar] [CrossRef] [PubMed]
  125. Yang, H.; Chen, D.; Wu, Y.; Zhou, H.; Diao, W.; Liu, G.; Li, Q. A feedback loop of PPP and PI3K/AKT signal pathway drives regorafenib-resistance in HCC. Cancer Metab. 2023, 11, 27. [Google Scholar] [CrossRef]
  126. Jia, D.; Liu, C.; Zhu, Z.; Cao, Y.; Wen, W.; Hong, Z.; Liu, Y.; Liu, E.; Chen, L.; Chen, C.; et al. Novel transketolase inhibitor oroxylin A suppresses the non-oxidative pentose phosphate pathway and hepatocellular carcinoma tumour growth in mice and patient-derived organoids. Clin. Transl. Med. 2022, 12, e1095. [Google Scholar] [CrossRef]
  127. Peng, Y.; Liu, Q.Z.; Xu, D.; Fu, J.Y.; Zhang, L.X.; Qiu, L.; Lin, J.G. M(4)IDP stimulates ROS elevation through inhibition of mevalonate pathway and pentose phosphate pathway to inhibit colon cancer cells. Biochem. Pharmacol. 2023, 217, 115856. [Google Scholar] [CrossRef] [PubMed]
  128. Koçak, E.; Çelebier, M.; Haznedaroglu, I.C.; Altınöz, S. Analysis of the Antiproliferative Effect of Ankaferd Hemostat on Caco-2 Colon Cancer Cells via LC/MS Shotgun Proteomics Approach. Biomed. Res. Int. 2019, 2019, 5268031. [Google Scholar] [CrossRef] [PubMed]
  129. Ahmad, R.; Alam, M.; Hasegawa, M.; Uchida, Y.; Al-Obaid, O.; Kharbanda, S.; Kufe, D. Targeting MUC1-C inhibits the AKT-S6K1-elF4A pathway regulating TIGAR translation in colorectal cancer. Mol. Cancer 2017, 16, 33. [Google Scholar] [CrossRef]
  130. Tanaka, G.; Inoue, K.; Shimizu, T.; Akimoto, K.; Kubota, K. Dual pharmacological inhibition of glutathione and thioredoxin systems synergizes to kill colorectal carcinoma stem cells. Cancer Med. 2016, 5, 2544–2557. [Google Scholar] [CrossRef]
  131. Shibuya, N.; Inoue, K.; Tanaka, G.; Akimoto, K.; Kubota, K. Augmented pentose phosphate pathway plays critical roles in colorectal carcinomas. Oncology 2015, 88, 309–319. [Google Scholar] [CrossRef]
  132. Chen, G.Q.; Tang, C.F.; Shi, X.K.; Lin, C.Y.; Fatima, S.; Pan, X.H.; Yang, D.J.; Zhang, G.; Lu, A.P.; Lin, S.H.; et al. Halofuginone inhibits colorectal cancer growth through suppression of Akt/mTORC1 signaling and glucose metabolism. Oncotarget 2015, 6, 24148–24162. [Google Scholar] [CrossRef]
  133. Sánchez-Tena, S.; Alcarraz-Vizán, G.; Marín, S.; Torres, J.L.; Cascante, M. Epicatechin gallate impairs colon cancer cell metabolic productivity. J. Agric. Food Chem. 2013, 61, 4310–4317. [Google Scholar] [CrossRef]
  134. Marimuthu, S.; Chivukula, R.S.; Alfonso, L.F.; Moridani, M.; Hagen, F.K.; Bhat, G.J. Aspirin acetylates multiple cellular proteins in HCT-116 colon cancer cells: Identification of novel targets. Int. J. Oncol. 2011, 39, 1273–1283. [Google Scholar] [CrossRef] [PubMed]
  135. Lin, X.; Chen, H.; Deng, T.; Cai, B.; Xia, Y.; Xie, L.; Wang, H.; Huang, C. Improved Immune Response for Colorectal Cancer Therapy Triggered by Multifunctional Nanocomposites with Self-Amplifying Antitumor Ferroptosis. ACS Appl. Mater. Interfaces 2024, 16, 13481–13495. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, Y.; Chen, J.; Duan, R.; Gu, R.; Wang, W.; Wu, J.; Lian, H.; Hu, Y.; Yuan, A. High-Z-Sensitized Radiotherapy Synergizes with the Intervention of the Pentose Phosphate Pathway for In Situ Tumor Vaccination. Adv. Mater. 2022, 34, e2109726. [Google Scholar] [CrossRef] [PubMed]
  137. Barot, S.; Stephenson, O.J.; Priya Vemana, H.; Yadav, A.; Bhutkar, S.; Trombetta, L.D.; Dukhande, V.V. Metabolic alterations and mitochondrial dysfunction underlie hepatocellular carcinoma cell death induced by a glycogen metabolic inhibitor. Biochem. Pharmacol. 2022, 203, 115201. [Google Scholar] [CrossRef] [PubMed]
  138. Wani, N.A.; Zhang, B.; Teng, K.Y.; Barajas, J.M.; Motiwala, T.; Hu, P.; Yu, L.; Brüschweiler, R.; Ghoshal, K.; Jacob, S.T. Reprograming of Glucose Metabolism by Zerumbone Suppresses Hepatocarcinogenesis. Mol. Cancer Res. 2018, 16, 256–268. [Google Scholar] [CrossRef]
  139. Sun, L.; Shang, H.; Wu, Y.; Xin, X. Hypericin-mediated photodynamic therapy enhances gemcitabine induced Capan-2 cell apoptosis via inhibiting NADPH level. J. Pharm. Pharmacol. 2022, 74, 596–604. [Google Scholar] [CrossRef] [PubMed]
  140. Daneshmandi, S.; Cassel, T.; Higashi, R.M.; Fan, T.W.; Seth, P. 6-Phosphogluconate dehydrogenase (6PGD), a key checkpoint in reprogramming of regulatory T cells metabolism and function. eLife 2021, 10, e67476. [Google Scholar] [CrossRef]
  141. Gu, M.; Zhou, X.; Sohn, J.H.; Zhu, L.; Jie, Z.; Yang, J.Y.; Zheng, X.; Xie, X.; Yang, J.; Shi, Y.; et al. NF-κB-inducing kinase maintains T cell metabolic fitness in antitumor immunity. Nat. Immunol. 2021, 22, 193–204. [Google Scholar] [CrossRef]
  142. Lei, J.; Yang, Y.; Lu, Z.; Pan, H.; Fang, J.; Jing, B.; Chen, Y.; Yin, L. Taming metabolic competition via glycolysis inhibition for safe and potent tumor immunotherapy. Biochem. Pharmacol. 2022, 202, 115153. [Google Scholar] [CrossRef] [PubMed]
  143. Povo-Retana, A.; Fariñas, M.; Landauro-Vera, R.; Mojena, M.; Alvarez-Lucena, C.; Fernández-Moreno, M.A.; Castrillo, A.; de la Rosa Medina, J.V.; Sánchez-García, S.; Foguet, C.; et al. Immunometabolic actions of trabectedin and lurbinectedin on human macrophages: Relevance for their anti-tumor activity. Front. Immunol. 2023, 14, 1211068. [Google Scholar] [CrossRef]
  144. Lao, Y.; Cui, X.; Xu, Z.; Yan, H.; Zhang, Z.; Zhang, Z.; Geng, L.; Li, B.; Lu, Y.; Guan, Q.; et al. Glutaryl-CoA dehydrogenase suppresses tumor progression and shapes an anti-tumor microenvironment in hepatocellular carcinoma. J. Hepatol. 2024, 81, 847–861. [Google Scholar] [CrossRef]
  145. Chen, H.; Wu, D.; Bao, L.; Yin, T.; Lei, D.; Yu, J.; Tong, X. 6PGD inhibition sensitizes hepatocellular carcinoma to chemotherapy via AMPK activation and metabolic reprogramming. Biomed. Pharmacother. 2019, 111, 1353–1358. [Google Scholar] [CrossRef]
  146. Dunbar, C.E.; High, K.A.; Joung, J.K.; Kohn, D.B.; Ozawa, K.; Sadelain, M. Gene therapy comes of age. Science 2018, 359, eaan4672. [Google Scholar] [CrossRef]
  147. Jiang, P.; Du, W.; Wang, X.; Mancuso, A.; Gao, X.; Wu, M.; Yang, X. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat. Cell Biol. 2011, 13, 310–316. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, K.; Lu, Y.; Li, H.; Zhang, J.; Ju, Y.; Ouyang, M. Role of long non-coding RNAs in metabolic reprogramming of gastrointestinal cancer cells. Cancer Cell Int. 2024, 24, 15. [Google Scholar] [CrossRef] [PubMed]
  149. Vincent, R.L.; Gurbatri, C.R.; Li, F.; Vardoshvili, A.; Coker, C.; Im, J.; Ballister, E.R.; Rouanne, M.; Savage, T.; de Los Santos-Alexis, K.; et al. Probiotic-guided CAR-T cells for solid tumor targeting. Science 2023, 382, 211–218. [Google Scholar] [CrossRef] [PubMed]
  150. Si, X.; Shao, M.; Teng, X.; Huang, Y.; Meng, Y.; Wu, L.; Wei, J.; Liu, L.; Gu, T.; Song, J.; et al. Mitochondrial isocitrate dehydrogenase impedes CAR T cell function by restraining antioxidant metabolism and histone acetylation. Cell Metab. 2024, 36, 176–192.e110. [Google Scholar] [CrossRef] [PubMed]
  151. Horecker, B.L. The pentose phosphate pathway. J. Biol. Chem. 2002, 277, 47965–47971. [Google Scholar] [CrossRef] [PubMed]
  152. Shah, S.C.; Itzkowitz, S.H. Colorectal Cancer in Inflammatory Bowel Disease: Mechanisms and Management. Gastroenterology 2022, 162, 715–730.e713. [Google Scholar] [CrossRef] [PubMed]
  153. Tian, N.; Hu, L.; Lu, Y.; Tong, L.; Feng, M.; Liu, Q.; Li, Y.; Zhu, Y.; Wu, L.; Ji, Y.; et al. TKT maintains intestinal ATP production and inhibits apoptosis-induced colitis. Cell Death Dis. 2021, 12, 853. [Google Scholar] [CrossRef] [PubMed]
  154. Xu, D.; Shao, F.; Bian, X.; Meng, Y.; Liang, T.; Lu, Z. The Evolving Landscape of Noncanonical Functions of Metabolic Enzymes in Cancer and Other Pathologies. Cell Metab. 2021, 33, 33–50. [Google Scholar] [CrossRef] [PubMed]
  155. Meskers, C.J.W.; Franczak, M.; Smolenski, R.T.; Giovannetti, E.; Peters, G.J. Are we still on the right path(way)?: The altered expression of the pentose phosphate pathway in solid tumors and the potential of its inhibition in combination therapy. Expert. Opin. Drug Metab. Toxicol. 2022, 18, 61–83. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic drawing of the metabolic flux of glycolysis, oxPPP, and non-oxPPP. Abbreviations: 2PG: 2-Phosphoglycerate; 3PG: 3-phosphoglycerate; 6PDGL: 6-phosphogluconolactone; 6PG: 6-phosphogluconate; DHAP: dihydroxyacetone phosphate; E4P: erythrose 4-phosphate; F1,6BP: Fructose-1,6-bisphosphate; F6P: fructose-6-phosphate; G6P: glucose-6-phosphate; GADP: glyceraldehyde 3-phosphate; GLUT: glucose transporter; HK: hexokinase; LDH: lactate dehydrogenase; NAD: nicotinamide adenine dinucleotide; NADPH: nicotinamide adenine dinucleotide phosphate; PEP: phosphoenolpyruvate; PGAM: phosphoglycerate mutase; PFK1: phosphofructokinase-1; R5P: ribose 5-phosphate; Ru5P: ribulose 5-phosphate; S7P: sedoheptulose 7-phosphate; TAL: transaldolase; TKT: transketolase; TPI: triosephosphate isomerase; and Xu5P: xylulose 5-phosphate.
Figure 1. Schematic drawing of the metabolic flux of glycolysis, oxPPP, and non-oxPPP. Abbreviations: 2PG: 2-Phosphoglycerate; 3PG: 3-phosphoglycerate; 6PDGL: 6-phosphogluconolactone; 6PG: 6-phosphogluconate; DHAP: dihydroxyacetone phosphate; E4P: erythrose 4-phosphate; F1,6BP: Fructose-1,6-bisphosphate; F6P: fructose-6-phosphate; G6P: glucose-6-phosphate; GADP: glyceraldehyde 3-phosphate; GLUT: glucose transporter; HK: hexokinase; LDH: lactate dehydrogenase; NAD: nicotinamide adenine dinucleotide; NADPH: nicotinamide adenine dinucleotide phosphate; PEP: phosphoenolpyruvate; PGAM: phosphoglycerate mutase; PFK1: phosphofructokinase-1; R5P: ribose 5-phosphate; Ru5P: ribulose 5-phosphate; S7P: sedoheptulose 7-phosphate; TAL: transaldolase; TKT: transketolase; TPI: triosephosphate isomerase; and Xu5P: xylulose 5-phosphate.
Ijms 26 00610 g001
Figure 2. Overview of the regulatory network involving G6PD in GI cancers. Abbreviations: Aldob: aldolase B; BAG3: Bcl-2 associated athanogene 3; CRC: colorectal cancer; ESCC: esophageal squamous cell carcinoma; G6PD: glucose-6-phosphate dehydrogenase; GC: gastric cancer; GSK-3β: glycogen synthase kinase-3β; GLUT1: glucose transporter 1; HBx: hepatitis B virus X protein; HCC: hepatocellular carcinoma; HnRNPK: heterogeneous nuclear ribonuclear protein K; KEAP1: kelch-like ECH-associated protein 1; ID1: inhibitor of differentiation 1; IL-6: interleukin-6; MDM2: murine double minute 2; NeuroD1: neuronal differentiation 1; Nrf2: nuclear factor erythroid 2-related factor 2; OGT: O-GlcNAc transferase; PAK4: p21-activated kinase 4; PBX3: Pre-B-cell leukemia transcription factor 3; PDAC: pancreatic ductal adenocarcinoma; PLK1: polo-like kinase 1; Pol ι: DNA polymerase iota; PSC: pancreatic stellate cell; PTEN: phosphatase and tensin homolog located on chromosome 10; Rac1: ras-related C3 botulinum toxin substrate 1; RF: regulatory factor; SDF-1α: stromal-derived factor-1 alpha; SOX9: sex-determining region Y-box 9; Tcl1: T cell leukemia 1; TF: transcription factor; TP53: tumor protein 53; and YY1: Yin Yang 1.
Figure 2. Overview of the regulatory network involving G6PD in GI cancers. Abbreviations: Aldob: aldolase B; BAG3: Bcl-2 associated athanogene 3; CRC: colorectal cancer; ESCC: esophageal squamous cell carcinoma; G6PD: glucose-6-phosphate dehydrogenase; GC: gastric cancer; GSK-3β: glycogen synthase kinase-3β; GLUT1: glucose transporter 1; HBx: hepatitis B virus X protein; HCC: hepatocellular carcinoma; HnRNPK: heterogeneous nuclear ribonuclear protein K; KEAP1: kelch-like ECH-associated protein 1; ID1: inhibitor of differentiation 1; IL-6: interleukin-6; MDM2: murine double minute 2; NeuroD1: neuronal differentiation 1; Nrf2: nuclear factor erythroid 2-related factor 2; OGT: O-GlcNAc transferase; PAK4: p21-activated kinase 4; PBX3: Pre-B-cell leukemia transcription factor 3; PDAC: pancreatic ductal adenocarcinoma; PLK1: polo-like kinase 1; Pol ι: DNA polymerase iota; PSC: pancreatic stellate cell; PTEN: phosphatase and tensin homolog located on chromosome 10; Rac1: ras-related C3 botulinum toxin substrate 1; RF: regulatory factor; SDF-1α: stromal-derived factor-1 alpha; SOX9: sex-determining region Y-box 9; Tcl1: T cell leukemia 1; TF: transcription factor; TP53: tumor protein 53; and YY1: Yin Yang 1.
Ijms 26 00610 g002
Figure 3. The relationships between the PPP and TME’s elements in GI cancers. Abbreviations: AMPK: AMP-activated protein kinase; AMOTL1: angiomotin like 1; ATP: adenosine triphosphate; CAFs: cancer-associated fibroblasts; CARM1: coactivator-associated arginine methyltransferase 1; CM: conditioned medium; FAK: focal adhesion kinase; G3P: glycerol-3-phosphate; G6P: glucose-6-phosphate; G6PD: glucose-6-phosphate dehydrogenase; HIF1α: hypoxia-inducible factors 1α; IFNγ: interferon-γ; IL-6: interleukin-6; MMP1: matrix metalloproteinase-1; MUC1: mucin 1; NRF2: nuclear factor erythroid 2-related factor 2; OXPHOS: oxidative phosphorylation; PCK1: phosphoenolpyruvate carboxykinase 1; PFKFB4: phosphofructokinase-fructose bisphosphatase 4; PI3K: phosphoinositide 3-kinase; POX: proline oxidase; PPP: pentose phosphate pathway; PFK1: phosphofructokinase-1; RPIA: ribose-5-phosphate isomerase A; ROS: reactive oxygen species; SDF-1α: stromal-derived factor-1α; SGK1: serum and glucocorticoid kinase-1; S7P: sedoheptulose-7-phosphate; TME: tumor microenvironment; TNFα: tumor necrosis factor α; and YAP/TAZ: yes-associated protein and transcriptional coactivator with PDZ-binding motif.
Figure 3. The relationships between the PPP and TME’s elements in GI cancers. Abbreviations: AMPK: AMP-activated protein kinase; AMOTL1: angiomotin like 1; ATP: adenosine triphosphate; CAFs: cancer-associated fibroblasts; CARM1: coactivator-associated arginine methyltransferase 1; CM: conditioned medium; FAK: focal adhesion kinase; G3P: glycerol-3-phosphate; G6P: glucose-6-phosphate; G6PD: glucose-6-phosphate dehydrogenase; HIF1α: hypoxia-inducible factors 1α; IFNγ: interferon-γ; IL-6: interleukin-6; MMP1: matrix metalloproteinase-1; MUC1: mucin 1; NRF2: nuclear factor erythroid 2-related factor 2; OXPHOS: oxidative phosphorylation; PCK1: phosphoenolpyruvate carboxykinase 1; PFKFB4: phosphofructokinase-fructose bisphosphatase 4; PI3K: phosphoinositide 3-kinase; POX: proline oxidase; PPP: pentose phosphate pathway; PFK1: phosphofructokinase-1; RPIA: ribose-5-phosphate isomerase A; ROS: reactive oxygen species; SDF-1α: stromal-derived factor-1α; SGK1: serum and glucocorticoid kinase-1; S7P: sedoheptulose-7-phosphate; TME: tumor microenvironment; TNFα: tumor necrosis factor α; and YAP/TAZ: yes-associated protein and transcriptional coactivator with PDZ-binding motif.
Ijms 26 00610 g003
Table 1. The regulatory mechanisms of enzymes in the PPP in GI cancers.
Table 1. The regulatory mechanisms of enzymes in the PPP in GI cancers.
GI Cancer TypeEnzymeBranch of PPPRegulatory AxisFunctionsReference
ESCCG6PDOxPPPYY1/↑PLK1/↑G6PD↓Ferroptosis
↓Chemoradiotherapy sensitivity
[17]
Pol ι/↑ERK/↑OGT/↑O-GlcNAc of G6PD↑Proliferation[18]
TKTNon-oxPPPHMGA1/↑Sp1/↑TKT↑Proliferation
↑PPP flux
↑NADPH and GSH
[21]
GCG6PDOxPPPRev-erbα/↓G6PD↑Proliferation
↑Glycolysis
[23]
CRCG6PDOxPPPPAK4/↑MDM2-mediated p53 ubiquitination/↑G6PD ↑Glucose consumption
↑NADPH production
[29]
NeuroD1/↑G6PD↑Proliferation
↓Apoptosis
↑NADPH production
↓ROS level
[30]
c-Src/↑G6PD Tyr 112 phosphorylation↑Tumor growth
↑NADPH production
↑Nucleotides synthesis
↑Lipid biosynthesis
[31]
Rac1/↑PI3K-AKT/↑SOX9/↑G6PD↑Proliferation
↑Migration
↑Invasion
↑Tumor growth
[32]
PBX3/↑G6PD↑Viability
↑Proliferation
↓Apoptosis
↑NADPH production
↓ROS level
↑Lipid biosynthesis
↑Tumor growth
[33]
YY1/↑G6PD↑Proliferation
↑Nucleotides synthesis
↑Lipid biosynthesis
↑NADPH production
[36]
6PGDATP13A2/↑TFEB nuclear localization/↑6PGD↑Proliferation
↑PPP activity
↑Tumor growth
[38]
TKTNon-oxPPPTKT/↑GRP78/↑AKT phosphorylation↑Proliferation
↑Metastasis
↑Aerobic glycolysis
[39]
RPIANuclear localization of RPIA/↑β-catenin↑Proliferation
↑Tumor growth
[44]
p16/↓mTORC1/↓RPIA↑Proliferation
↓Senescence
↑Nucleotide synthesis
[43]
Glucose deprivation/↑CARM1-RPIA interaction/↑RPIA R42 methylation↑PPP flux
↑ROS clearance
↑Cell growth
[74]
HCCG6PDOxPPPG6PD/↑STAT3 phosphorylation↑Proliferation
↑Migration
↑Invasion
↑Tumor growth
↑EMT
[48]
PTEN/↑GSK3β/↓Tcl1/↑hnRNPK/↓G6PD pre-mRNA splicing↓G6PD dimer formation
↓Proliferation
↑Senescence
↑Sensitivity of HCC to sorafenib
[52]
Aldob–G6PD–p53 protein complex/↓G6PD activity↓Tumorigenesis[53]
BAG3/↓G6PD↓Proliferation[54]
HBx–p62–Keap1 complex/↑Nrf2/↑G6PD↑Proliferation[50]
ID1/↑Wnt/β-catenin pathway/↑c-MYC/↑G6PD↑Proliferation
↓Apoptosis
↑Oxaliplatin resistance
[75]
6PGDNrf2/↑6PGD,
6PGD/↓Keap1/↑Nrf2
↑Proliferation
↑Migration
[59]
TKTNon-oxPPPTKT nuclear localization/↑EGFR pathway↑Proliferation
↑Viability
↑Migration
↑Invasion
↑Metastasis
[61]
HBx/↑SH2D5/↑interaction of SH2D5 and TKT/↑STAT3 pathway↑Proliferation
↑Migration
↑Invasion
[63]
VRK2/↑TKT phosphorylation/↑FBXL6/↑TKT ubiquitination and activation/↑ROS-mTOR axis/↑PD-L1↑Tumorigenesis
↑Immune evasion
↑Metastasis
[62]
RPIARPIA/↓PP2A activity/↑ERK signaling ↑Proliferation
↑Tumor growth
[76]
PDACG6PDOxPPP↑GLUT1/↓Aldob/↑G6PD activity↑Chemoresistance[66]
PSC-CM/↑SDF-1α, IL-6/↑Nrf2/↑G6PD↑Proliferation
↑Glucose metabolism
↑Glutaminolysis
↑Glutathione biosynthesis
↓ROS level
[77]
ID1/↑c-MYC/↑G6PD↓Glycolysis
↑PPP flux
↑Erlotinib resistance
[78]
TKTNon-oxPPP↑S100A11 expression/↑H3K4me3 on TKT promoter/↑TKT expression↑Proliferation
↑Tumor growth
[68]
GIST6PGDOxPPPLong-term imatinib exposure/↑HIF-1α/↑6PGD↑Proliferation
↓Apoptosis
↑Imatinib-resistant
[72]
“↑” means upregulation and “↓” means downregulation.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Qiao, J.; Yu, Z.; Zhou, H.; Wang, W.; Wu, H.; Ye, J. The Pentose Phosphate Pathway: From Mechanisms to Implications for Gastrointestinal Cancers. Int. J. Mol. Sci. 2025, 26, 610. https://doi.org/10.3390/ijms26020610

AMA Style

Qiao J, Yu Z, Zhou H, Wang W, Wu H, Ye J. The Pentose Phosphate Pathway: From Mechanisms to Implications for Gastrointestinal Cancers. International Journal of Molecular Sciences. 2025; 26(2):610. https://doi.org/10.3390/ijms26020610

Chicago/Turabian Style

Qiao, Jincheng, Zhengchen Yu, Han Zhou, Wankun Wang, Hao Wu, and Jun Ye. 2025. "The Pentose Phosphate Pathway: From Mechanisms to Implications for Gastrointestinal Cancers" International Journal of Molecular Sciences 26, no. 2: 610. https://doi.org/10.3390/ijms26020610

APA Style

Qiao, J., Yu, Z., Zhou, H., Wang, W., Wu, H., & Ye, J. (2025). The Pentose Phosphate Pathway: From Mechanisms to Implications for Gastrointestinal Cancers. International Journal of Molecular Sciences, 26(2), 610. https://doi.org/10.3390/ijms26020610

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop