IMR Press / FBL / Volume 29 / Issue 4 / DOI: 10.31083/j.fbl2904137
Open Access Review
Promising Therapeutic Approach in Pancreatic Cancer: Metabolism-Related Genes
Show Less
1 Department of Medical and Biological Sciences, The Catholic University of Korea, 14662 Bucheon, Republic of Korea
2 Department of Biotechnology, The Catholic University of Korea, 14662 Bucheon, Republic of Korea
3 Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
4 Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 06591 Seoul, Republic of Korea
*Correspondence: hjkoh92@naver.com (Hyun Jung Koh); hyoon@catholic.ac.kr (Hyunho Yoon)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2024, 29(4), 137; https://doi.org/10.31083/j.fbl2904137
Submitted: 1 December 2023 | Revised: 12 March 2024 | Accepted: 19 March 2024 | Published: 2 April 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Most pancreatic cancers are pancreatic ductal adenocarcinomas. This is an extremely lethal disease with poor prognosis and almost no treatment choices. Considering the profound role of the pancreas in the human body, malfunction of this organ can significantly affect quality of life. Although multiple metabolic pathways are altered in cancer cells, certain metabolic gene signatures may be critical for immunotherapy. The reprogrammed metabolism of glucose, amino acids, and lipids can nourish the tumor microenvironment (TME). Previous studies have also shown that reprogrammed metabolism influences immune responses. Tumor-infiltrating immune cells in the TME can adapt their metabolism to blunt the immune system, leading to immunosuppression and tumor progression. The identification of metabolism-related genes (MRGs) associated with immune reactions in pancreatic cancer may lead to improved treatments. This review highlights the characteristics of MRGs in pancreatic cancer and suggests that enhanced anti-cancer therapies could be used to overcome resistance to immunotherapy.

Keywords
pancreatic cancer
metabolism
metabolites
immunotherapy
1. Introduction

Pancreatic cancer is the fourth leading cause of cancer-related deaths worldwide, with a five-year overall survival rate of less than 8% [1]. It typically arises in the pancreatic duct, and the known risk factors for pancreatic ductal adenocarcinoma (PDAC) include smoking, obesity, and diabetes. The incidence of PDAC is higher in women than men. Excessive alcohol intake and a family history of PDAC may also contribute to increased risk [2, 3, 4, 5, 6]. Patients with pancreatic cancer often present with advanced disease due to the lack of early detection [7, 8, 9]. Although surgery remains the best treatment option, PDAC is often discovered at an advanced stage when lesions cannot be removed. Consequently, a multidisciplinary therapeutic approach has been implemented for PDAC [8]. Adjuvant chemotherapy has improved long-term outcomes in patients with advanced cancer, although the response rates remain low. Up to 80% of patients with pancreatic cancer have a poor initial prognosis [10]. Even patients who undergo successful surgery eventually develop local recurrence or metastasis, which then greatly worsens the disease prognosis [11]. Although successful in some cancer types, immunotherapy has not been effective in the treatment of pancreatic cancer, with most clinical trials for PDAC giving negative results [8]. Therefore, alternative therapeutic approaches are urgently required for PDAC.

Metabolic reprogramming determines the function and viability of cancer cells and plays an important role in tumor initiation and progression, including PDAC [12, 13, 14]. Tumors undergo dramatic changes in glucose, lipid, and amino acid metabolism compared with normal tissues [15]. Growing evidence has shown the metabolic requirements of immune cells in the tumor microenvironment (TME) have a significant impact on the success of immunotherapy [12, 16]. Despite the presence of many immune cells in pancreatic cancer tissue, immune dysfunction is observed when the TME is immunosuppressive, thereby hindering the activation of immune effectors [17]. In order to develop effective treatment strategies, it is therefore critical to uncover the mechanisms that underlie abnormal metabolism in cancer [15].

Glucose metabolism has been implicated in cancer development and metastasis and is the primary source of carbon chains for biosynthesis and energy metabolism [18]. In normal cells, glucose metabolism involves the synthesis of adenosine triphosphate (ATP) through anaerobic glycolysis and cellular respiration. However, glucose metabolism in cancer cells is aerobic and is regulated by epigenetic mechanisms and cooperation with the TME, which can lead to the loss of tumor suppressors [19, 20]. The rapid proliferation of cancer cells is facilitated by lactate production and glucose metabolism, which lead to cancer progression, chemoresistance, and local depletion of oxygen. This results in hypoxic regions containing high levels of lactate [21, 22, 23], which then restricts immune cell function by switching to an acidic environment [24]. Pancreatic cancers show upregulated expression of rate-limiting glycolytic enzymes including hexokinase 1/2 (HK1/2), phosphofructokinase 1 (PFK1), and lactate dehydrogenase A (LDHA, a subunit of LDH), thus contributing to the Warburg effect and improving lactate-mediated glycolysis [22].

Non-glucose-mediated metabolism, such as amino acids and lipids, is also necessary for cancer cell survival and growth. The main function of amino acids in normal cells is the synthesis of new proteins, and amino acid metabolism in cancer cells is also an essential factor for cell proliferation [25]. Glutamine is important for the production of metabolic intermediates, energy, and cellular functions [26, 27]. Importantly, dysregulation of glutamine metabolism in the tricarboxylic acid (TCA) cycle, the central metabolic hub of cells, is an important driver of cancer cells [28]. Cancer cell growth and proliferation depend on TCA cycle intermediates and amino acids to generate purine and pyrimidine nucleotides [18]. Amino acids are involved in biosynthesis and in the maintenance of redox balance, as well as contributing to immune responses associated with tumor development and metastasis via epigenetic regulation [29].

Lipids are required for membrane biosynthesis during rapid cell proliferation. In addition, they serve as energy stores during metabolic stress, and participate in various signaling pathways [21]. While most normal cells preferentially use extracellular lipids to synthesize new structural lipids, cancer cells are able to synthesize fatty acids (FAs) to maintain proliferation in lipid-poor microenvironments and in the absence of extracellular lipids [30]. Lipid metabolism is reprogrammed in cancer cells and contributes to rapid tumor growth through increased lipid uptake, storage, and lipogenesis [31, 32, 33, 34]. These observations suggest that targeting of dysregulated lipid metabolism may be a promising approach for cancer treatment.

Cell metabolism and metabolites were recently reported to be crucial regulators of the immune system. Therefore, understanding the differential metabolic requirements of various cell types provides an opportunity to highlight metabolic vulnerabilities and expose therapeutic windows that may improve immunotherapy [13]. Immunotherapy is currently shifting the paradigm of cancer therapy. It is important to target metabolism in the normal immune system when treating cancer, particularly for solid tumors such as pancreatic cancer which often contain more non-cancer cells than cancer cells [18].

2. Glucose Metabolism in Pancreatic Cancer

Glycolysis is a vital metabolic process that produces ATP from glucose to maintain cellular growth. Pancreatic cancers exhibit dysregulated cellular metabolism, which is one of the hallmarks of cancer (Fig. 1) [9]. The Warburg effect is a representative feature of this rewired glucose metabolism that provides adequate ATP for tumor cells to grow, proliferate, and survive [35]. Anaerobic glycolysis is preferred even under normoxic conditions in cancer cells, as it allows the rapid production of metabolites [36]. Mutations in the Kristen Rat Sarcoma Viral oncogene homolog (KRAS) and its downstream pathways regulate glucose metabolism-related genes (MRGs) in PDAC, leading to an increased need for glucose and modulation of the TME [8]. As an example, the rate-limiting enzymes and glucose transporters in glycolysis, PFK1, HK2, and LDHA, are all controlled by oncogenic KRAS [9].

Fig. 1.

Altered metabolism of glucose, glutamine, and lipids in pancreatic cancer. Abnormal metabolism of essential nutrients is a hallmark feature in cancer cells compared to normal cells. Enhanced uptake of glucose in cancer preferentially undergoes anaerobic glycolysis (Warburg effect), resulting in elevated lactate production to generate more ATP. Increased influx of glutamine is facilitated by metabolism-related genes (MRGs) and participates in the tricarboxylic acid (TCA) cycle by undergoing conversion into glutamate and α-KG. Malonyl-CoA is generated from acetyl-CoA and produces more lipids in cancer cells, thereby supporting the progression of pancreatic cancer. GLUT1, glucose transporter 1; Ac-CoA, acetyl-CoA; α-KG, α-ketoglutarate.

2.1 Correlation between Glucose Metabolism and Immunotherapy in Pancreatic Cancer

Resistance to immunotherapy remains a major challenge in PDAC treatment, despite extensive investigations with this anti-cancer therapy. Metabolic interactions with the immune response may provide some clues to address this limitation. In this regard, reprogrammed metabolic processes in pancreatic cancer have been shown to influence immune cells in the TME, resulting in tumor immunosuppression and resistance to immunotherapy [37]. Since glucose is continuously consumed by the Warburg effect, immune cells compete with cancer cells for nutrition [37]. Moreover, elevated levels of glycolysis induce acidic conditions in the TME due to the accumulated lactate. This allows cancer cells to avoid immune surveillance, but also debilitates the immune response [38]. Lactate accumulation in PDAC reduces the efficacy of immunotherapy by creating an immunosuppressive microenvironment. T cell activation is especially important for functional immunity and is impacted by metabolic changes in the TME. Due to the massive uptake of glucose by cancer cells, the level becomes insufficient for T cell activation [27]. This leads to T cell exhaustion, which inhibits appropriate binding between the immune checkpoint and its ligands in PDAC [39]. As a consequence, the elevated glucose metabolism in PDAC results in immune evasion.

2.2 Glucose Metabolism-Related Genes in Pancreatic Cancer
2.2.1 GLUT1

Several MRGs form a vital link between glucose metabolism and the immune response in PDAC. Glucose transporter 1 (GLUT1) mainly contributes to the glycolytic process as a membrane protein. It is highly upregulated in many cancer cell types due to their increased demand for glucose [40, 41]. The crucial role of GLUT1 in generating ATP from glucose makes it a potential biomarker for pancreatic cancer and a target for therapy. Immune cells such as T cells, B cells and macrophages, as well as tumor cells require considerable amounts of energy for optimal function [42]. A glucose-depleted TME can therefore suppress T cells [43]. GLUT1 also participates in immune escape by PDAC through a positive relationship with programmed cell death ligand 1 (PD-L1) and an inverse relationship with programmed cell death 1 (PD-1), both of which are immune checkpoints [41]. Moreover, binding of PD-1/PD-L1 in the TME inversely hinders T cell function, leading to immune evasion [44].

2.2.2 HK2

HK2 is a key enzyme in the first step of glycolysis and is highly expressed in most tumors [45]. Together with GLUT1, increased HK2 expression supports the Warburg effect. It serves as an altered glycolytic process and produces sufficient ATP in the TME even under hypoxic conditions [46]. Furthermore, patients with high HK2 expression show anchorage-independent cell growth, metastatic signatures, and poor overall survival [47, 48]. HK2 is negatively associated with immune processes in PDAC. Elevated glycolysis by HK2 generates lactate and glucose, resulting in immunosuppression. Lactic acid can also be utilized by cancer cells as a nutrient substitute [49]. Acidification of the TME by accumulated lactate can suppress cytotoxic T cells and natural killer (NK) cells, which are vital for impeding tumorigenesis [49].

2.2.3 IGFBPs

Insulin-like growth factor-binding proteins (IGFBPs) respond to insulin signaling by binding and stabilizing insulin-like growth factor (IGF). Of note, the expression of IGF receptor (IGFR) is elevated in severe-grade tumors such as PDAC [50]. Insulin is produced in the pancreas and transforms glucose into the glycogen storage form. It is therefore strongly associated with both glucose metabolism and pancreatic cancer. IGFBP-1/2 can be downregulated by insulin to activate free IGF, thereby promoting cancer progression [51]. IGF-1 modulates apoptosis, proliferation, metastasis, and the immune system [50, 51, 52]. It also allows CCL5 secreted from peripheral cells to gather in anti-tumor immune cells and tumor-associated M2 macrophages. Moreover, cancer-associated fibroblasts (CAFs) in PDAC can produce IGF ligands [50, 53]. IGFBP-6 plays a role in immunosuppression and chemoresistance, both of which are related to T cell malfunction [54]. Thus, IGFBPs could be used as prognostic biomarkers and therapeutic targets in pancreatic cancer.

2.2.4 ALDH1A3

The enzyme aldehyde dehydrogenase 1 family member A3 (ALDH1A3) alters the glucose flux and is associated with aggressive PDAC [55]. It also increases HK2 expression, leading to enhance glycolysis [56]. ALDH1A3 is expressed in cancer stem cells (CSCs), which can initiate cancer through differentiation and self-renewal [57]. Elevated levels of ALDH1A3 therefore indicate stemness features of pancreatic cancer, including metastasis and progression [58]. CSCs participate in the immunosuppressive TME as ‘latency competent’ CSCs, which refers to their ability to decrease NK cells while transitioning into a dormant state and evading cytolysis [59]. The plasticity of CSCs in PDAC allows them to escape immunotherapy because they persist for extended periods and are promoted by hyperglycemia [60].

2.2.5 ALDOA

Aldolase A (ALDOA) catalyzes fructose 1,6-bisphosphate into glyceraldehyde 3-phosphate (G3P) and dihydroxyacetone phosphate in the glucose metabolic pathway [61]. This enzyme is highly expressed in most cancer types. In pancreatic cancer, ALDOA is associated with malignant characteristics such as cell proliferation, invasion and migration, and worse patient survival [40, 62]. ALDOA is thus closely associated with PDAC metastasis and progression. Silencing of ALDOA in PDAC cell has been shown to downregulate crucial molecules in glycolysis, including hypoxia-inducible factor 1 (HIF-1) [63]. Moreover, ALDOA is associated with HIF-1-related genes and reduces CD8+ T cell infiltration, thereby causing immunosuppression [64]. HIF-1 can regulate both innate and adaptive immune responses in the hypoxic TME and trigger immune evasion by inducing immune checkpoint inhibitors, such as PD-1/PD-L1 [65].

3. Glutamine Metabolism in Pancreatic Cancer

Amino acids are crucial nutrients for most cells, including cancer cells, with glutamine being the main reprogrammed amino acid in cancer [66]. Glutamine has multifaceted functions that affect PDAC progression. It can support the TCA cycle by converting to α-ketoglutarate (α-KG) and sustaining cellular redox homeostasis [67]. Glutamine is also an essential nutrient source, especially for the proliferation of cancer cells and the synthesis of cellular products. Various cancer cell types, including PDAC, therefore exhibit glutamine addiction [68]. In contrast to normal cells, PDAC have an elevated requirement for glutamine. Furthermore, KRAS oncogenic mutation can regulate glutamine MRGs such as glutamine transporters [69, 70]. Glutamine metabolism is also associated with the glycolytic process and the TCA cycle. In addition, cancer cells utilize more glutamine and somewhat more lipids than glucose compared with immune cells, resulting in an immunosuppressive TME [71].

3.1 Correlation between Glutamine Metabolism and Immunotherapy in Pancreatic Cancer

Given its multiple roles, glutamine is able to coordinate various immune reactions. A sophisticated TME organized by dysregulated glutamine metabolism can attenuate anti-tumor immune processes in PDAC [72]. T cell proliferation is enhanced by increasing the levels of glutamine transporters, glutaminolysis-associated enzymes, and Myc transcription factor via T cell receptor (TCR)-stimulated activation [73]. Elevated expression of these proteins can meet the demands of anabolic mechanisms and immune cells [73]. However, excessive glutamine consumption by cancer cells interferes with the appropriate utilization of glutamine by immune cells, leading to the reprogramming of immune cell metabolism [74]. This affects tumor immune cell infiltration by inhibiting T cell activation through the upregulation of PD-L1 expression [73, 75]. Hence, the targeting of PD-L1 in combination with glutamine MRGs may address the challenges associated with immune checkpoint blockade.

3.2 Glutamine Metabolism-Related Genes in Pancreatic Cancer
3.2.1 TFEB

Transcription factor EB (TFEB) is a critical molecule in autophagy and lysosomal function. It forms part of the coordinated lysosomal expression and regulation (CLEAR) gene network response to nutrient availability, including amino acids [76]. In PDAC, TFEB shows increased expression following glutamine metabolism [77]. TFEB regulates the cellular process of autophagy, which is responsible for the degradation and recycling of intracellular components. Autophagy plays a vital role in cancer cells by providing alternative energy sources for cell growth. Since cancer cells utilize glutamine and TFEB modulates glutaminase expression, the regulation of TFEB is a significant factor in PDAC growth [77]. Moreover, TFEB is linked to innate immunity and inflammation through its activation of macrophages and promotion of autophagy [78]. TFEB can also enhance PD-L1 expression and help cancer cells evade immune responses by inhibiting mammalian target of rapamycin (mTOR) [79].

3.2.2 GFPT2

Metabolic pathways for cellular biosynthetic sources interact with various metabolic genes. For example, glutamine-fructose-6-phosphate transaminase 2 (GFPT2) is a rate-limiting enzyme in the hexosamine biosynthesis pathway (HBP) that promotes glucose uptake in cancer cells [80]. Increased GFPT2 expression in hypoxic pancreatic cancer is associated with sufficient access to both glucose and glutamine via the HBP [37]. GFPT2 favors the epithelial-mesenchymal transition (EMT) and cell invasion and migration, leading to poor prognosis [80, 81]. Additionally, GFPT2 is closely associated with immune infiltration in the TME, resulting in the progression of cancer through the release of growth factors from immune cells, including T cells, B cells, and macrophages [82]. Interestingly, GFPT2 is also linked to T cell exhaustion and immunosuppressive factors, such as macrophages and fibroblasts. This occurs via the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway and infiltrated immune cells [83]. Thus, GFPT2 is a promising MRG for the targeting of immune escape in cancer.

3.2.3 EAAT

Excitatory amino acid transporters (EAATs) can maintain the glutamate status and transform glutamine-derived glutamate into α-KG to support the TCA cycle [84]. Upregulated EAATs can induce cancer invasion and growth by accumulating glutamate and promoting hypoxia [85]. Malignant pancreatic cancer cells express high levels of EAAT, causing resistance to anti-tumor treatment, cell proliferation, migration, and invasion [66, 86]. Although glutamate is a major neurotransmitter in the nervous system, some immune cells (e.g., T cells) also have glutamate receptors and regulate immune cell functions by signaling through these receptors [85]. Glutamine deprivation in the TME can influence immune cell activity and potentially contribute to immunosuppressive reactions [75, 87]. Therefore, dysregulated glutamine metabolism due to EAAT can adversely affect PDAC by modulating the immune response.

3.2.4 LAT1 (SLC7A5)

The L-type amino acid transporter LAT1, also known as SLC7A5, is essential for metabolism during the progression of cancer through the provision of sufficient amino acids [88]. LAT1 is able to transport amino acids even under conditions of low affinity, and can fully activate CD8+ T cells [89]. TCR stimulation can increase LAT1 expression via the extracellular signal-regulated kinase 1/2-mitogen-activated protein kinase (ERK/MAPK) pathway. Low levels of LAT1 therefore lead to compromised T cell function and cytokine release [89]. LAT1 expression is elevated in pancreatic cancer and correlates with cell proliferation and poor outcome, as well as the induction of PD-L1 expression [90]. LAT1 is also required for immune cells to function in the presence of sufficient nutrients, especially glutamine [91].

4. Lipid Metabolism in Pancreatic Cancer

Lipids are an essential nutrient source for cancer and normal cells. In cancer cells, lipids provide fuel for cell membranes, ATP production, and signaling pathways [92]. Therefore, altered lipid metabolic processes contribute to the metastasis, invasion, and proliferation of tumors [93]. Increased de novo lipid synthesis leads to the accumulation of fatty acids (FAs), resulting in tumor progression [94]. KRAS mutations occur in most pancreatic cancers and have been shown to influence FA oxidation and lipid droplet (LD) storage by repressing hormone-sensitive lipase (HSL), thereby favoring PDAC cell invasion and migration [94]. Because the requirement for lipids is elevated in advanced pancreatic cancer, the lipolysis-related enzymes fatty acid synthase (FASN) and acetyl-CoA carboxylase (ACC) are upregulated [95]. In addition, an abundance of lipids and immune complexes can lead to inflammatory reactions [33].

4.1 Correlation between Lipid Metabolism and the Response to Immunotherapy in Pancreatic Cancer

A deeper understanding of the connection between immune responses and metabolic processes is required to fully appreciate the impact of the immune system on cancer cells (Fig. 2). Enhanced lipid metabolism leads to greater demand for lipids, FA oxidation and FAS, and results in the creation of an immunosuppressive niche [96]. While lipolysis exhibits a tendency converse to lipogenesis to support excessive provision of lipids. Downregulation of HSL suppresses the breakdown of stored lipids and triggers PDAC metastasis [97]. Furthermore, reprogrammed lipid metabolism in tumor-associated macrophages (TAM) in the TME helps to regulate cancer immunity [98]. Lipids accumulate in TAM and promote M2 macrophage traits [99]. Lipid metabolism subsequently releases anti-inflammatory chemokines and cytokines that attract regulatory T (Treg) cells and present the PD-L1 and CD80 ligands to the immune checkpoint inhibitors PD-1 and cytotoxic T-lymphocyte-associated antigen 4 (CTLA4). This leads to diminished anti-cancer immunity related to T cell function [98, 100]. Accordingly, abnormal lipid metabolism is relevant not only for the immune response in the TME, but also as a target of immunotherapy resistance.

Fig. 2.

Impact of MRGs on the immunosuppressive Tumor microenvironment (TME) in pancreatic cancer. Glucose, glutamine, and lipid metabolism contribute to cancer cells through the TCA cycle. They are connected to cellular energy production and synthesis, thereby influencing the overall cellular metabolic landscape and supporting the biosynthetic demands of rapidly proliferating cancer cells. Moreover, MRGs induce an immunosuppressive environment in the TME and connect metabolism with the immune response in PDAC. TME, tumor microenvironment; GLUT1, glucose transporter 1; HK2, hexokinase 2; IGFBPs, insulin-like growth factor binding proteins; ALDH1A3, Aldehyde dehydrogenase 1 family member A3; ALDOA, Aldolase A; TFEB, Transcription factor EB; GFPT2, Glutamine-fructose-6-phosphate transaminase 2; EAAT, Excitatory amino acid transporters; LAT1, L-type amino acid transporter 1; HSDL2, Hydroxysteroid dehydrogenase like protein 2; HILPDA, Hypoxia inducible lipid droplet associated; ACSL4, Long chain acyl CoA synthetase 4; Ac-CoA, acetyl CoA; α-KG, α-ketoglutarate; ATP, adenosine triphosphate.

4.2 Lipid Metabolism-Related Genes in Pancreatic Cancer
4.2.1 HSDL2

Hydroxysteroid dehydrogenase-like protein 2 (HSDL2) is a significant FA enzyme in lipid metabolic pathways relevant to tumorigenesis [101]. The expression of HSDL2 is increased in PDAC compared to adjacent tissues and is associated with poor prognosis [101, 102]. HSDL2 also promotes the first stage of cancer progression involving increased cell proliferation and migration via EMT [102, 103]. Moreover, HSDL2 interacts with peroxisome proliferator-activated receptors (PPARs) to induce Tregs and immunosuppressive activity, which are crucial for regulating innate immune responses and inflammation [104]. HSDL2 has drawn attention as a candidate gene for immunosuppression in pancreatic cancer treatment due to its association with immune cell infiltration.

4.2.2 HILPDA

Hypoxia-inducible lipid droplet-associated (HILPDA) reacts to oxygen or glucose starvation in PDAC and macrophages by increasing the capacity of LDs to store neutral lipids [105]. The modulation of triglyceride and LD development through attachment and prevention of Adipose Triglyceride Lipase (ATGL) activity, a rate-limiting lipase, is critical for tumor growth [105]. HILPDA is highly expressed in cancer cells, and increased lipid metabolic processes result in the accumulation of incoming lipids into LDs. Consequently, HILPDA expression tends to promote tumor growth, leading to poor survival of pancreatic cancer patients [106]. HILPDA expression correlates with infiltrated immune cells, such as TAM, and also with immunosuppressive genes, including PD-L1/PD-1 [107]. Abnormal HILPDA function in cancer cells may therefore impede the efficacy of anti-cancer treatment.

4.2.3 CD36

CD36 contributes to FA uptake under dysregulated metabolic conditions, especially under high lipid requirements [108]. CD36 establishes lipid homeostasis as an FA transporter and accelerates the progression and metastasis of pancreatic cancer [109]. Interestingly, CD36 expression in PDAC is lower than in normal tissues as an unfavorable prognostic marker but shows shorter overall survival, indicating a more severe tumor [109, 110]. Downregulated CD36 expression reduces cell adhesion to the Extracellular matrix (ECM) and induces cell motility, resulting in increased metastasis [109]. Additionally, CD36 expression is associated with immune cell infiltration and with checkpoint inhibitors involving PD-L1 [111]. Along with CD36, lipids can also regulate immunity in cancer cells, and altered lipid metabolism leads to both tumorigenesis and immune tolerance [111].

4.2.4 ACSL4

Long-chain acyl-CoA synthetase 4 (ACSL4) controls de novo lipogenesis by increasing the levels of lipogenic regulators in cancer cells [112]. ACSL4 expression in cancer cells leads to the accumulation of lipids from aberrant metabolic states. This occurs sequentially via increased levels of Myc, sterol regulatory element binding protein 1 (SREBP1), and FA synthesis, which then eventually induces cancer cell growth and metastasis [112]. Furthermore, ACSL4 induces an immunosuppressive TME in PDAC, resulting in poor prognosis [113]. ACSL4 is also associated with ferroptosis, a form of regulated cell death, and its downregulation promotes cancer progression [113]. This process functions in an anti-tumor immune response, followed by CD8+ T cell-mediated immunity, along with interferon gamma (IFN-γ) and immune checkpoint inhibitors [114].

5. Diverse Implications of Metabolism-Related Genes in Pancreatic Cancer

The significant progress in tumor biology has led to new methods for treating multifarious cancers. Treatment with immune checkpoint inhibitors has been widely used as a novel strategy, but has significant limitations. Abnormal metabolic changes are a distinct feature of malignant cells, and hence the ability to regulate cell metabolism may give rise to new treatment options (Fig. 3).

Fig. 3.

Implications of MRGs in pancreatic cancer. The targeting of glucose, glutamine, and lipid MRGs may be a promising strategy to treat PDAC. Each metabolic pathway can promote or suppress crucial functions in cancer cells to modulate tumor development and progression. Inhibitors mainly regulate the major MRGs in metabolic processes and exacerbate immunosuppression. Hence, they could also be utilized in combination therapy. PDAC, pancreatic ductal adenocarcinoma; CANA, canagliflozin; 3-BP, 3-bromopyruvate; DON, 6-diazo-5-oxo-L-norleucine.

Modulation of glucose uptake is important for metabolism-associated treatment. Of the genes mentioned earlier, GLUT1 is a well-known enzyme involved in glucose metabolism and is currently the subject of active research. Canagliflozin (CANA) is an anti-diabetic drug that acts by inhibiting sodium-glucose cotransporter 2 (SGLT2), which manages glucose flux. CANA has reportedly shown efficacy against hepatocellular carcinoma (HCC) [115] and can reduce the viability and growth of cancer cells through necrosis, both in vitro and in vivo [116, 117]. CANA can promote apoptosis and suppress glycolysis in pancreatic cancer by decreasing GLUT1 expression through the phosphoinositide 3 kinase/AKT/mTOR (PI3K/AKT/mTOR) pathway [117]. CANA also improves the efficacy of the chemotherapeutic drug gemcitabine, indicating a synergetic effect when used in combination with current medication [117, 118]. WZB117 is a competitive GLUT1 inhibitor that targets glycolysis. However, these inhibitors are still undergoing Phase II clinical trials [47, 119]. WZB117 decreases cancer progression in vivo by reducing ATP and glycolytic enzyme levels in a dose-dependent manner [116]. Combination therapy with GLUT inhibitors and immunotherapy appears to weaken tumor cell lysis and overcome resistance [120].

Given that glutamine contributes to tumor growth and survival, targeted therapy against glutamine MRGs holds promise for overcoming resistance to immunotherapy. The LAT1 inhibitor JPH203 has completed first-in-human Phase I trials and is currently undergoing randomized Phase II clinical trials [121, 122]. JPH203 prevents the in vitro and in vivo progression of PDAC by blocking transport of bulky neutral amino acids [122, 123]. SKN103 is another LAT1 inhibitor that can inhibit pancreatic cancer cell growth in vitro [124]. Moreover, the glutamine antagonist 6-diazo-5-oxo-L-norleucine (DON) enhances the efficacy of intra-tumoral MBTA immunotherapy [125]. MBTA refers to the combination of Mannan-BAM, Toll-like receptor (TLR) ligands, and Anti-CD40 antibody. This immunotherapy can activate not only innate immunity but also the adaptive immune response in pancreatic cancer [126]. Therefore, DON can sensitize PDAC to immunotherapy within the TME [125].

Cancer cells facilitate the production of lipids, glucose, and glutamine. This has led to the suggestion of novel therapeutic approaches for pancreatic cancer that involve lipid-related MRGs. Lipids mediate the immune system in the TME in several ways. Cyclooxygenase-2 (COX-2) inhibitors favor the immune response by decreasing myeloid-derived suppressor cells (MDSCs). This results in a permissive TME for T cells rather than the immunosuppressive TME in pancreatic cancer [127]. COX-2 inhibitors can potentially be co-administered with immune checkpoint blockers to reduce the adverse effects of immunotherapy. Combination treatment with COX-2 inhibitor and different drugs, such as celecoxib and aspirin, is still undergoing clinical trials in various cancer types [127]. Disulfiram targets aldehyde dehydrogenase (ALDH), which is involved in both lipid and glutamine metabolism, and induces anti-tumor activity in pancreatic cancer both in vitro and in vivo [128, 129, 130]. In particular, disulfiram inhibits PDAC growth by suppressing cell proliferation [128]. This suggests that combined treatment with MRGs may be an effective anti-cancer therapy to address the current challenge of immunotherapy resistance. Because aberrant metabolism mostly results in immunosuppression of the TME, targeting these MRGs has the potential to increase the efficacy of pancreatic cancer treatment (Table 1, Ref. [116, 117, 122, 131, 132, 133, 134, 135, 136, 137]).

Table 1.Inhibitors that target MRGs in pancreatic cancer.
Related metabolism Cellular target Drug Function FDA-approval Reference
Glucose GLUT1 CANA Enhances apoptosis and inhibits glycolysis Yes [117]
WZB117 Suppresses cancer growth No [116]
HK2 3-BP Restrains ATP generation and induces apoptosis in cancer No [131]
Glutamine LAT1 JPH203 Represses PDAC progression by obstructing amino acids transporters No [122]
SKN103 Downregulates cancer development No [132]
Glutaminase DON Inactivates tumor metabolism and alleviates hypoxia No [133]
Lipid ALDH Disulfiram Upregulates anti-tumor effect Yes [134]
ACSL4 Triacsin C Arrests ACSL4 via competing with FAs and induces ferroptosis No [135, 136]
Thiazolidinediones Suppresses ACSL4 and reduces cancer cell growth No [136, 137]

GLUT1, glucose transporter 1; CANA, canagliflozin; HK2, hexokinase 2; 3-BP, 3-bromopyruvate; ATP, adenosine triphosphate; LAT1, L-type amino acid transporter 1; PDAC, pancreatic ductal adenocarcinoma; DON, 6-diazo-5-oxo-L-norleucine; ALDH, aldehyde dehydrogenase; ACSL4, long chain acyl CoA synthetase 4; FAs, fatty acids; FDA, U.S. Food and Drug Administration.

6. Limitations and Future Perspectives in Pancreatic Cancer Metabolism

Although the targeting of specific metabolites may be a promising treatment approach, several concerns remain. Cancer cells exhibit considerable dependence on nutrients for their growth. Several metabolic pathways in tumor cells function in tandem with the surrounding cells [138]. Therefore, metabolic changes that occur after targeting MRGs might lead to unwanted interactions between the heterogeneous TME. In other words, changes to MRGs only may result in unsatisfactory outcomes in PDAC patients, and application of combination therapy may be required. Currently, clinical trials of combination therapies with simvastatin, digoxin and metformin aim to assess their therapeutic efficacy in PDAC and other advanced solid tumors [8]. Future studies aimed at developing metabolism-related strategies must overcome conventional obstacles, including immunosuppression. This review provides some useful insights for the investigation of MRGs as possible targets.

7. Conclusions

The alteration of various metabolites, including glucose, amino acids, and lipids, is a conspicuous feature of cancer progression. Cancer cells consume essential nutrients to fuel the excessive growth of tumors and to modify the TME in order to survive. Immunotherapy, primarily against the immune checkpoints PD-L1/PD-1 and CTLA4, is used in pancreatic cancer treatment. Although various remedies to cure cancer have been investigated, the outcome of PDAC exhibits little promise in terms of the detrimental effects of immunotherapy. Targeted therapies against MRGs may overcome this limitation, and combination treatments with existing strategies could result in improved outcomes. Numerous studies have demonstrated the potential of MRG-associated therapies for pancreatic cancer. Recent developments with single cell analysis platforms allow the study of targeted genes in the PDAC phenotype [139, 140]. However, clinical trials have yet to be performed, and it is uncertain whether anti-cancer treatments using MRG will be beneficial. In view of the complexity of abnormal metabolic processes and their interactions within the TME, further careful research is necessary. Resistance to immunotherapy is a critical barrier that must be overcome to obtain effective anti-tumor treatments in various cancer types. MRGs have been identified as significant therapeutic targets and as potentially useful in combination therapies for PDAC.

Author Contributions

SC, WK, EK, SS, MS, HJK, and HY conceptualized the study. SC, WK, EK, SS, MS, HJK, and HY prepared the original draft of the manuscript. SC, WK, EK, SS, MS, HJK, and HY reviewed and edited the manuscript. HY supervised the study. HY was responsible for funding acquisition. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

The authors wish to acknowledge the financial support of the Catholic Medical Center Research Foundation made in the program year of 2019.

Funding

The authors wish to acknowledge the financial support of the Catholic Medical Center Research Foundation made in the program year of 2019, and funded by the Brain Korea 21 (BK21), grant number M2022B002600003, and the Ministry of Food and Drug Safety in Korea, grant number 22213MFDS421.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Yang L, TeSlaa T, Ng S, Nofal M, Wang L, Lan T, et al. Ketogenic diet and chemotherapy combine to disrupt pancreatic cancer metabolism and growth. Med (New York, N.Y.). 2022; 3: 119–136.
[2]
Padoan A, Plebani M, Basso D. Inflammation and Pancreatic Cancer: Focus on Metabolism, Cytokines, and Immunity. International Journal of Molecular Sciences. 2019; 20: 676.
[3]
Koyanagi YN, Matsuo K, Ito H, Tamakoshi A, Sugawara Y, Hidaka A, et al. Body-Mass Index and Pancreatic Cancer Incidence: A Pooled Analysis of Nine Population-Based Cohort Studies With More Than 340,000 Japanese Subjects. Journal of Epidemiology. 2018; 28: 245–252.
[4]
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA: a Cancer Journal for Clinicians. 2018; 68: 7–30.
[5]
Jiao L, Chen L, White DL, Tinker L, Chlebowski RT, Van Horn LV, et al. Low-fat Dietary Pattern and Pancreatic Cancer Risk in the Women’s Health Initiative Dietary Modification Randomized Controlled Trial. Journal of the National Cancer Institute. 2018; 110.
[6]
Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet (London, England). 2016; 388: 73–85.
[7]
Yao W, Maitra A, Ying H. Recent insights into the biology of pancreatic cancer. EBioMedicine. 2020; 53: 102655.
[8]
Qin C, Yang G, Yang J, Ren B, Wang H, Chen G, et al. Metabolism of pancreatic cancer: paving the way to better anticancer strategies. Molecular Cancer. 2020; 19: 50.
[9]
Yan L, Raj P, Yao W, Ying H. Glucose Metabolism in Pancreatic Cancer. Cancers. 2019; 11: 1460.
[10]
Yin X, Xu R, Song J, Ruze R, Chen Y, Wang C, et al. Lipid metabolism in pancreatic cancer: emerging roles and potential targets. Cancer Communications (London, England). 2022; 42: 1234–1256.
[11]
Li D, Frazier M, Evans DB, Hess KR, Crane CH, Jiao L, et al. Single nucleotide polymorphisms of RecQ1, RAD54L, and ATM genes are associated with reduced survival of pancreatic cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2006; 24: 1720–1728.
[12]
Martínez-Reyes I, Chandel NS. Cancer metabolism: looking forward. Nature Reviews. Cancer. 2021; 21: 669–680.
[13]
Leone RD, Powell JD. Metabolism of immune cells in cancer. Nature Reviews. Cancer. 2020; 20: 516–531.
[14]
Singer K, Cheng WC, Kreutz M, Ho PC, Siska PJ. Immunometabolism in cancer at a glance. Disease Models & Mechanisms. 2018; 11: dmm034272.
[15]
Yu T, Wang Y, Fan Y, Fang N, Wang T, Xu T, et al. CircRNAs in cancer metabolism: a review. Journal of Hematology & Oncology. 2019; 12: 90.
[16]
Guerra L, Bonetti L, Brenner D. Metabolic Modulation of Immunity: A New Concept in Cancer Immunotherapy. Cell Reports. 2020; 32: 107848.
[17]
Duan Q, Li H, Gao C, Zhao H, Wu S, Wu H, et al. High glucose promotes pancreatic cancer cells to escape from immune surveillance via AMPK-Bmi1-GATA2-MICA/B pathway. Journal of Experimental & Clinical Cancer Research: CR. 2019; 38: 192.
[18]
Stine ZE, Schug ZT, Salvino JM, Dang CV. Targeting cancer metabolism in the era of precision oncology. Nature Reviews. Drug Discovery. 2022; 21: 141–162.
[19]
Kocianova E, Piatrikova V, Golias T. Revisiting the Warburg Effect with Focus on Lactate. Cancers. 2022; 14: 6028.
[20]
Vaupel P, Multhoff G. Revisiting the Warburg effect: historical dogma versus current understanding. The Journal of Physiology. 2021; 599: 1745–1757.
[21]
Bandi DSR, Sarvesh S, Farran B, Nagaraju GP, El-Rayes BF. Targeting the metabolism and immune system in pancreatic ductal adenocarcinoma: Insights and future directions. Cytokine & Growth Factor Reviews. 2023; 71-72: 26–39.
[22]
O’Sullivan D, Sanin DE, Pearce EJ, Pearce EL. Metabolic interventions in the immune response to cancer. Nature Reviews. Immunology. 2019; 19: 324–335.
[23]
Bose S, Le A. Glucose metabolism in cancer. The Heterogeneity of Cancer Metabolism. 2018; 3–12.
[24]
Ye L, Jiang Y, Zhang M. Crosstalk between glucose metabolism, lactate production and immune response modulation. Cytokine & Growth Factor Reviews. 2022; 68: 81–92.
[25]
Vettore L, Westbrook RL, Tennant DA. New aspects of amino acid metabolism in cancer. British Journal of Cancer. 2020; 122: 150–156.
[26]
Zhu L, Zhu X, Wu Y. Effects of Glucose Metabolism, Lipid Metabolism, and Glutamine Metabolism on Tumor Microenvironment and Clinical Implications. Biomolecules. 2022; 12: 580.
[27]
DePeaux K, Delgoffe GM. Metabolic barriers to cancer immunotherapy. Nature Reviews. Immunology. 2021; 21: 785–797.
[28]
Halama A, Suhre K. Advancing Cancer Treatment by Targeting Glutamine Metabolism-A Roadmap. Cancers. 2022; 14: 553.
[29]
Lieu EL, Nguyen T, Rhyne S, Kim J. Amino acids in cancer. Experimental & Molecular Medicine. 2020; 52: 15–30.
[30]
Broadfield LA, Pane AA, Talebi A, Swinnen JV, Fendt SM. Lipid metabolism in cancer: New perspectives and emerging mechanisms. Developmental Cell. 2021; 56: 1363–1393.
[31]
Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Communications (London, England). 2018; 38: 27.
[32]
Bian X, Liu R, Meng Y, Xing D, Xu D, Lu Z. Lipid metabolism and cancer. The Journal of Experimental Medicine. 2021; 218: e20201606.
[33]
Long J, Zhang CJ, Zhu N, Du K, Yin YF, Tan X, et al. Lipid metabolism and carcinogenesis, cancer development. American Journal of Cancer Research. 2018; 8: 778–791.
[34]
Zhu Y, Aupperlee MD, Zhao Y, Tan YS, Kirk EL, Sun X, et al. Pubertal and adult windows of susceptibility to a high animal fat diet in Trp53-null mammary tumorigenesis. Oncotarget. 2016; 7: 83409–83423.
[35]
Schiliro C, Firestein BL. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells. 2021; 10: 1056.
[36]
Pouysségur J, Marchiq I, Parks SK, Durivault J, Ždralević M, Vucetic M. ‘Warburg effect’ controls tumor growth, bacterial, viral infections and immunity - Genetic deconstruction and therapeutic perspectives. Seminars in Cancer Biology. 2022; 86: 334–346.
[37]
Hao X, Ren Y, Feng M, Wang Q, Wang Y. Metabolic reprogramming due to hypoxia in pancreatic cancer: Implications for tumor formation, immunity, and more. Biomedicine & Pharmacotherapy. 2021; 141: 111798.
[38]
Shamsi M, Saghafian M, Dejam M, Sanati-Nezhad A. Mathematical Modeling of the Function of Warburg Effect in Tumor Microenvironment. Scientific Reports. 2018; 8: 8903.
[39]
Zarour HM. Reversing T-cell Dysfunction and Exhaustion in Cancer. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2016; 22: 1856–1864.
[40]
Curcio C, Brugiapaglia S, Bulfamante S, Follia L, Cappello P, Novelli F. The Glycolytic Pathway as a Target for Novel Onco-Immunology Therapies in Pancreatic Cancer. Molecules (Basel, Switzerland). 2021; 26: 1642.
[41]
Li F, He C, Yao H, Liang W, Ye X, Ruan J, et al. GLUT1 Regulates the Tumor Immune Microenvironment and Promotes Tumor Metastasis in Pancreatic Adenocarcinoma via ncRNA-mediated Network. Journal of Cancer. 2022; 13: 2540–2558.
[42]
Renner K, Singer K, Koehl GE, Geissler EK, Peter K, Siska PJ, et al. Metabolic Hallmarks of Tumor and Immune Cells in the Tumor Microenvironment. Frontiers in Immunology. 2017; 8: 248.
[43]
Gun SY, Lee SWL, Sieow JL, Wong SC. Targeting immune cells for cancer therapy. Redox Biology. 2019; 25: 101174.
[44]
Dong S, Li W, Li X, Wang Z, Chen Z, Shi H, et al. Glucose metabolism and tumour microenvironment in pancreatic cancer: A key link in cancer progression. Frontiers in Immunology. 2022; 13: 1038650.
[45]
Zhang Y, Li Q, Huang Z, Li B, Nice EC, Huang C, et al. Targeting Glucose Metabolism Enzymes in Cancer Treatment: Current and Emerging Strategies. Cancers. 2022; 14: 4568.
[46]
Liu C, Jin Y, Fan Z. The Mechanism of Warburg Effect-Induced Chemoresistance in Cancer. Frontiers in Oncology. 2021; 11: 698023.
[47]
Penny HL, Sieow JL, Gun SY, Lau MC, Lee B, Tan J, et al. Targeting Glycolysis in Macrophages Confers Protection Against Pancreatic Ductal Adenocarcinoma. International Journal of Molecular Sciences. 2021; 22: 6350.
[48]
Anderson M, Marayati R, Moffitt R, Yeh JJ. Hexokinase 2 promotes tumor growth and metastasis by regulating lactate production in pancreatic cancer. Oncotarget. 2016; 8: 56081–56094.
[49]
Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, et al. The cancer metabolic reprogramming and immune response. Molecular Cancer. 2021; 20: 28.
[50]
Mutgan AC, Besikcioglu HE, Wang S, Friess H, Ceyhan GO, Demir IE. Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer. Molecular Cancer. 2018; 17: 66.
[51]
Ghanavat M, Shahrouzian M, Deris Zayeri Z, Banihashemi S, Kazemi SM, Saki N. Digging deeper through glucose metabolism and its regulators in cancer and metastasis. Life Sciences. 2021; 264: 118603.
[52]
Quoc Lam B, Shrivastava SK, Shrivastava A, Shankar S, Srivastava RK. The Impact of obesity and diabetes mellitus on pancreatic cancer: Molecular mechanisms and clinical perspectives. Journal of Cellular and Molecular Medicine. 2020; 24: 7706–7716.
[53]
Thomas D, Radhakrishnan P. Role of Tumor and Stroma-Derived IGF/IGFBPs in Pancreatic Cancer. Cancers. 2020; 12: 1228.
[54]
Liso A, Venuto S, Coda ARD, Giallongo C, Palumbo GA, Tibullo D. IGFBP-6: At the Crossroads of Immunity, Tissue Repair and Fibrosis. International Journal of Molecular Sciences. 2022; 23: 4358.
[55]
Nie S, Qian X, Shi M, Li H, Peng C, Ding X, et al. ALDH1A3 Accelerates Pancreatic Cancer Metastasis by Promoting Glucose Metabolism. Frontiers in Oncology. 2020; 10: 915.
[56]
Stanciu S, Ionita-Radu F, Stefani C, Miricescu D, Stanescu-Spinu II, Greabu M, et al. Targeting PI3K/AKT/mTOR Signaling Pathway in Pancreatic Cancer: From Molecular to Clinical Aspects. International Journal of Molecular Sciences. 2022; 23: 10132.
[57]
Kasai T, Tamori S, Takasaki Y, Matsuoka I, Ozaki A, Matsuda C, et al. High expression of PKCλ and ALDH1A3 indicates a poor prognosis, and PKCλ is required for the asymmetric cell division of ALDH1A3-positive cancer stem cells in PDAC. Biochemical and Biophysical Research Communications. 2023; 669: 85–94.
[58]
Ciccone V, Morbidelli L, Ziche M, Donnini S. How to conjugate the stemness marker ALDH1A1 with tumor angiogenesis, progression, and drug resistance. Cancer Drug Resistance (Alhambra, Calif.). 2020; 3: 26–37.
[59]
Mortezaee K. Enriched cancer stem cells, dense stroma, and cold immunity: Interrelated events in pancreatic cancer. Journal of Biochemical and Molecular Toxicology. 2021; 35: e22708.
[60]
Liu Z, Hayashi H, Matsumura K, Uemura N, Shiraishi Y, Sato H, et al. Biological and Clinical Impacts of Glucose Metabolism in Pancreatic Ductal Adenocarcinoma. Cancers. 2023; 15: 498.
[61]
Tian W, Zhou J, Chen M, Qiu L, Li Y, Zhang W, et al. Bioinformatics analysis of the role of aldolase A in tumor prognosis and immunity. Scientific Reports. 2022; 12: 11632.
[62]
Tang Y, Yang X, Feng K, Hu C, Li S. High expression of aldolase A is associated with tumor progression and poor prognosis in hepatocellular carcinoma. Journal of Gastrointestinal Oncology. 2021; 12: 174–183.
[63]
Ji S, Zhang B, Liu J, Qin Y, Liang C, Shi S, et al. ALDOA functions as an oncogene in the highly metastatic pancreatic cancer. Cancer Letters. 2016; 374: 127–135.
[64]
Zhuang H, Wang S, Chen B, Zhang Z, Ma Z, Li Z, et al. Prognostic Stratification Based on HIF-1 Signaling for Evaluating Hypoxic Status and Immune Infiltration in Pancreatic Ductal Adenocarcinomas. Frontiers in Immunology. 2021; 12: 790661.
[65]
You L, Wu W, Wang X, Fang L, Adam V, Nepovimova E, et al. The role of hypoxia-inducible factor 1 in tumor immune evasion. Medicinal Research Reviews. 2021; 41: 1622–1643.
[66]
Xu R, Yang J, Ren B, Wang H, Yang G, Chen Y, et al. Reprogramming of Amino Acid Metabolism in Pancreatic Cancer: Recent Advances and Therapeutic Strategies. Frontiers in Oncology. 2020; 10: 572722.
[67]
Yang S, Hwang S, Kim M, Seo SB, Lee JH, Jeong SM. Mitochondrial glutamine metabolism via GOT2 supports pancreatic cancer growth through senescence inhibition. Cell Death & Disease. 2018; 9: 55.
[68]
Feng M, Xiong G, Cao Z, Yang G, Zheng S, Qiu J, et al. LAT2 regulates glutamine-dependent mTOR activation to promote glycolysis and chemoresistance in pancreatic cancer. Journal of Experimental & Clinical Cancer Research: CR. 2018; 37: 274.
[69]
Recouvreux MV, Moldenhauer MR, Galenkamp KMO, Jung M, James B, Zhang Y, et al. Glutamine depletion regulates Slug to promote EMT and metastasis in pancreatic cancer. The Journal of Experimental Medicine. 2020; 217: e20200388.
[70]
Miguel XF, José MP. Inhibition of glutamine metabolism as a therapeutic approach against pancreatic ductal adenocarcinoma. Journal of Molecular and Clinical Medicine. 2019; 2: 97–110.
[71]
Niu Y, Mayr T, Muders MH. Competition for nutrients or cell intrinsic programming? - Metabolic mechanisms behind the tumor promoting immune microenvironment in cancer. Signal Transduction and Targeted Therapy. 2021; 6: 279.
[72]
Ali A, Chianese U, Papulino C, Toraldo A, Abakar MEA, Passaro E, et al. Metabolic Pathways as a Novel Landscape in Pancreatic Ductal Adenocarcinoma. Cancers. 2022; 14: 3799.
[73]
Jin J, Byun JK, Choi YK, Park KG. Targeting glutamine metabolism as a therapeutic strategy for cancer. Experimental & Molecular Medicine. 2023; 55: 706–715.
[74]
Yoo HC, Yu YC, Sung Y, Han JM. Glutamine reliance in cell metabolism. Experimental & Molecular Medicine. 2020; 52: 1496–1516.
[75]
Ma G, Zhang Z, Li P, Zhang Z, Zeng M, Liang Z, et al. Reprogramming of glutamine metabolism and its impact on immune response in the tumor microenvironment. Cell Communication and Signaling: CCS. 2022; 20: 114.
[76]
He R, Wang M, Zhao C, Shen M, Yu Y, He L, et al. TFEB-driven autophagy potentiates TGF-β induced migration in pancreatic cancer cells. Journal of Experimental & Clinical Cancer Research: CR. 2019; 38: 340.
[77]
Kim JH, Lee J, Cho YR, Lee SY, Sung GJ, Shin DM, et al. TFEB Supports Pancreatic Cancer Growth through the Transcriptional Regulation of Glutaminase. Cancers. 2021; 13: 483.
[78]
Brady OA, Martina JA, Puertollano R. Emerging roles for TFEB in the immune response and inflammation. Autophagy. 2018; 14: 181–189.
[79]
Zhang C, Duan Y, Xia M, Dong Y, Chen Y, Zheng L, et al. TFEB Mediates Immune Evasion and Resistance to mTOR Inhibition of Renal Cell Carcinoma via Induction of PD-L1. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2019; 25: 6827–6838.
[80]
Chao D, Ariake K, Sato S, Ohtsuka H, Takadate T, Ishida M, et al. Stomatin like protein 2 induces metastasis by regulating the expression of a rate limiting enzyme of the hexosamine biosynthetic pathway in pancreatic cancer. Oncology Reports. 2021; 45: 90.
[81]
Zhou L, Luo M, Cheng LJ, Li RN, Liu B, Linghu H. Glutamine-fructose-6-phosphate transaminase 2 (GFPT2) promotes the EMT of serous ovarian cancer by activating the hexosamine biosynthetic pathway to increase the nuclear location of β-catenin. Pathology, Research and Practice. 2019; 215: 152681.
[82]
Zhang J, Wang T, Wei S, Chen S, Bi J. GFPT2 pan-cancer analysis and its prognostic and tumor microenvironment associations. Oncology and Translational Medicine. 2021; 7: 286–293.
[83]
Ding X, Liu H, Yuan Y, Zhong Q, Zhong X. Roles of GFPT2 Expression Levels on the Prognosis and Tumor Microenvironment of Colon Cancer. Frontiers in Oncology. 2022; 12: 811559.
[84]
Magi S, Piccirillo S, Amoroso S, Lariccia V. Excitatory Amino Acid Transporters (EAATs): Glutamate Transport and Beyond. International Journal of Molecular Sciences. 2019; 20: 5674.
[85]
Koda S, Hu J, Ju X, Sun G, Shao S, Tang RX, et al. The role of glutamate receptors in the regulation of the tumor microenvironment. Frontiers in Immunology. 2023; 14: 1123841.
[86]
García-Gaytán AC, Hernández-Abrego A, Díaz-Muñoz M, Méndez I. Glutamatergic system components as potential biomarkers and therapeutic targets in cancer in non-neural organs. Frontiers in Endocrinology. 2022; 13: 1029210.
[87]
Bader JE, Voss K, Rathmell JC. Targeting Metabolism to Improve the Tumor Microenvironment for Cancer Immunotherapy. Molecular Cell. 2020; 78: 1019–1033.
[88]
Lu X. The Role of Large Neutral Amino Acid Transporter (LAT1) in Cancer. Current Cancer Drug Targets. 2019; 19: 863–876.
[89]
Weiss HJ, Angiari S. Metabolite Transporters as Regulators of Immunity. Metabolites. 2020; 10: 418.
[90]
Kurozumi S, Kaira K, Matsumoto H, Kurosumi M, Yokobori T, Kanai Y, et al. Association of L-type amino acid transporter 1 (LAT1) with the immune system and prognosis in invasive breast cancer. Scientific Reports. 2022; 12: 2742.
[91]
Lopes C, Pereira C, Medeiros R. ASCT2 and LAT1 Contribution to the Hallmarks of Cancer: From a Molecular Perspective to Clinical Translation. Cancers. 2021; 13: 203.
[92]
Wolrab D, Jirásko R, Cífková E, Höring M, Mei D, Chocholoušková M, et al. Lipidomic profiling of human serum enables detection of pancreatic cancer. Nature Communications. 2022; 13: 124.
[93]
Lee JH, Cho YR, Kim JH, Kim J, Nam HY, Kim SW, et al. Branched-chain amino acids sustain pancreatic cancer growth by regulating lipid metabolism. Experimental & Molecular Medicine. 2019; 51: 1–11.
[94]
Rozeveld CN, Johnson KM, Zhang L, Razidlo GL. KRAS Controls Pancreatic Cancer Cell Lipid Metabolism and Invasive Potential through the Lipase HSL. Cancer Research. 2020; 80: 4932–4945.
[95]
Biancur DE, Kimmelman AC. The plasticity of pancreatic cancer metabolism in tumor progression and therapeutic resistance. Biochimica et Biophysica Acta. Reviews on Cancer. 2018; 1870: 67–75.
[96]
Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K, et al. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics. Journal of Hematology & Oncology. 2023; 16: 103.
[97]
Vasseur S, Guillaumond F. Lipids in cancer: a global view of the contribution of lipid pathways to metastatic formation and treatment resistance. Oncogenesis. 2022; 11: 46.
[98]
Bleve A, Durante B, Sica A, Consonni FM. Lipid Metabolism and Cancer Immunotherapy: Immunosuppressive Myeloid Cells at the Crossroad. International Journal of Molecular Sciences. 2020; 21: 5845.
[99]
Qiao X, Hu Z, Xiong F, Yang Y, Peng C, Wang D, et al. Lipid metabolism reprogramming in tumor-associated macrophages and implications for therapy. Lipids in Health and Disease. 2023; 22: 45.
[100]
Martinez-Bosch N, Vinaixa J, Navarro P. Immune Evasion in Pancreatic Cancer: From Mechanisms to Therapy. Cancers. 2018; 10: 6.
[101]
Han A, Xu R, Liu Y, Yin X, Lin Z, Yang W. HSDL2 Acts as a Promoter in Pancreatic Cancer by Regulating Cell Proliferation and Lipid Metabolism. OncoTargets and Therapy. 2021; 14: 435–444.
[102]
Yang Y, Han A, Wang X, Yin X, Cui M, Lin Z. Lipid metabolism regulator human hydroxysteroid dehydrogenase-like 2 (HSDL2) modulates cervical cancer cell proliferation and metastasis. Journal of Cellular and Molecular Medicine. 2021; 25: 4846–4859.
[103]
Jia LH, Hu MD, Liu Y, Xiong X, Wang WJ, Wang JG, et al. HSDL2 Promotes Bladder Cancer Growth In Vitro and In Vivo. International Journal of Medical Sciences. 2019; 16: 654–659.
[104]
Zhao Q, Zhong J, Lu P, Feng X, Han Y, Ling C, et al. DOCK4 Is a Platinum-Chemosensitive and Prognostic-Related Biomarker in Ovarian Cancer. PPAR Research. 2021; 2021: 6629842.
[105]
Grachan JJ, Kery M, Giaccia AJ, Denko NC, Papandreou I. Lipid droplet storage promotes murine pancreatic tumor growth. Oncology Reports. 2021; 45: 21.
[106]
Bai R, Rebelo A, Kleeff J, Sunami Y. Identification of prognostic lipid droplet-associated genes in pancreatic cancer patients via bioinformatics analysis. Lipids in Health and Disease. 2021; 20: 58.
[107]
Liu C, Zhou X, Zeng H, Wu D, Liu L. HILPDA Is a Prognostic Biomarker and Correlates With Macrophage Infiltration in Pan-Cancer. Frontiers in Oncology. 2021; 11: 597860.
[108]
Li Y, Huang X, Yang G, Xu K, Yin Y, Brecchia G, et al. CD36 favours fat sensing and transport to govern lipid metabolism. Progress in Lipid Research. 2022; 88: 101193.
[109]
Tanase C, Gheorghisan-Galateanu AA, Popescu ID, Mihai S, Codrici E, Albulescu R, et al. CD36 and CD97 in Pancreatic Cancer versus Other Malignancies. International Journal of Molecular Sciences. 2020; 21: 5656.
[110]
Jia S, Zhou L, Shen T, Zhou S, Ding G, Cao L. Down-expression of CD36 in pancreatic adenocarcinoma and its correlation with clinicopathological features and prognosis. Journal of Cancer. 2018; 9: 578–583.
[111]
Chen YJ, Liao WX, Huang SZ, Yu YF, Wen JY, Chen J, et al. Prognostic and immunological role of CD36: A pan-cancer analysis. Journal of Cancer. 2021; 12: 4762–4773.
[112]
Chen J, Ding C, Chen Y, Hu W, Yu C, Peng C, et al. ACSL4 reprograms fatty acid metabolism in hepatocellular carcinoma via c-Myc/SREBP1 pathway. Cancer Letters. 2021; 502: 154–165.
[113]
Yang Y, Zhu T, Wang X, Xiong F, Hu Z, Qiao X, et al. ACSL3 and ACSL4, Distinct Roles in Ferroptosis and Cancers. Cancers. 2022; 14: 5896.
[114]
Liao P, Wang W, Wang W, Kryczek I, Li X, Bian Y, et al. CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell. 2022; 40: 365–378.e6.
[115]
Kaji K, Nishimura N, Seki K, Sato S, Saikawa S, Nakanishi K, et al. Sodium glucose cotransporter 2 inhibitor canagliflozin attenuates liver cancer cell growth and angiogenic activity by inhibiting glucose uptake. International Journal of Cancer. 2018; 142: 1712–1722.
[116]
Pliszka M, Szablewski L. Glucose Transporters as a Target for Anticancer Therapy. Cancers. 2021; 13: 4184.
[117]
Xu D, Zhou Y, Xie X, He L, Ding J, Pang S, et al. Inhibitory effects of canagliflozin on pancreatic cancer are mediated via the downregulation of glucose transporter 1 and lactate dehydrogenase A. International Journal of Oncology. 2020; 57: 1223–1233.
[118]
Henriksen A, Dyhl-Polk A, Chen I, Nielsen D. Checkpoint inhibitors in pancreatic cancer. Cancer Treatment Reviews. 2019; 78: 17–30.
[119]
Poonprasartporn A, Xiao J, Chan KLA. A study of WZB117 as a competitive inhibitor of glucose transporter in high glucose treated PANC-1 cells by live-cell FTIR spectroscopy. Talanta. 2024; 266: 125031.
[120]
Tilekar K, Upadhyay N, Iancu CV, Pokrovsky V, Choe JY, Ramaa CS. Power of two: combination of therapeutic approaches involving glucose transporter (GLUT) inhibitors to combat cancer. Biochimica et Biophysica Acta. Reviews on Cancer. 2020; 1874: 188457.
[121]
Okano N, Naruge D, Kawai K, Kobayashi T, Nagashima F, Endou H, et al. First-in-human phase I study of JPH203, an L-type amino acid transporter 1 inhibitor, in patients with advanced solid tumors. Investigational New Drugs. 2020; 38: 1495–1506.
[122]
Nishikubo K, Ohgaki R, Okanishi H, Okuda S, Xu M, Endou H, et al. Pharmacologic inhibition of LAT1 predominantly suppresses transport of large neutral amino acids and downregulates global translation in cancer cells. Journal of Cellular and Molecular Medicine. 2022; 26: 5246–5256.
[123]
Altan B, Kaira K, Watanabe A, Kubo N, Bao P, Dolgormaa G, et al. Relationship between LAT1 expression and resistance to chemotherapy in pancreatic ductal adenocarcinoma. Cancer Chemotherapy and Pharmacology. 2018; 81: 141–153.
[124]
Häfliger P, Charles RP. The L-Type Amino Acid Transporter LAT1-An Emerging Target in Cancer. International Journal of Molecular Sciences. 2019; 20: 2428.
[125]
Frejlachova A, Lencova R, Venhauerova A, Skalickova M, Uher O, Caisova V, et al. The combination of immunotherapy and a glutamine metabolism inhibitor represents an effective therapeutic strategy for advanced and metastatic murine pancreatic adenocarcinoma. International Immunopharmacology. 2023; 118: 110150.
[126]
Lookian PP, Zhao D, Medina R, Wang H, Zenka J, Gilbert MR, et al. Mannan-BAM, TLR Ligands, Anti-CD40 Antibody (MBTA) Vaccine Immunotherapy: A Review of Current Evidence and Applications in Glioblastoma. International Journal of Molecular Sciences. 2021; 22: 3455.
[127]
Zheng M, Zhang W, Chen X, Guo H, Wu H, Xu Y, et al. The impact of lipids on the cancer-immunity cycle and strategies for modulating lipid metabolism to improve cancer immunotherapy. Acta Pharmaceutica Sinica. B. 2023; 13: 1488–1497.
[128]
Xu Y, Lu L, Luo J, Wang L, Zhang Q, Cao J, et al. Disulfiram Alone Functions as a Radiosensitizer for Pancreatic Cancer Both In Vitro and In Vivo. Frontiers in Oncology. 2021; 11: 683695.
[129]
Flor AC, Wolfgeher D, Wu D, Kron SJ. A signature of enhanced lipid metabolism, lipid peroxidation and aldehyde stress in therapy-induced senescence. Cell Death Discovery. 2017; 3: 17075.
[130]
Sunami Y, Rebelo A, Kleeff J. Lipid Metabolism and Lipid Droplets in Pancreatic Cancer and Stellate Cells. Cancers. 2017; 10: 3.
[131]
Roy S, Dukic T, Bhandary B, Tu KJ, Molitoris J, Ko YH, et al. 3-Bromopyruvate inhibits pancreatic tumor growth by stalling glycolysis, and dismantling mitochondria in a syngeneic mouse model. American Journal of Cancer Research. 2022; 12: 4977–4987.
[132]
Zhang J, Xu Y, Li D, Fu L, Zhang X, Bao Y, et al. Review of the Correlation of LAT1 With Diseases: Mechanism and Treatment. Frontiers in Chemistry. 2020; 8: 564809.
[133]
Yang WH, Qiu Y, Stamatatos O, Janowitz T, Lukey MJ. Enhancing the Efficacy of Glutamine Metabolism Inhibitors in Cancer Therapy. Trends in Cancer. 2021; 7: 790–804.
[134]
Lu C, Li X, Ren Y, Zhang X. Disulfiram: a novel repurposed drug for cancer therapy. Cancer Chemotherapy and Pharmacology. 2021; 87: 159–172.
[135]
Li D, Li Y. The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduction and Targeted Therapy. 2020; 5: 108.
[136]
Zhang L, Wang X. Characteristics of long‐chain acyl‐CoA synthetases in metabolism and cancer. Clinical and Translational Discovery. 2023; 3: e200.
[137]
Kwon MJ, Lee YJ, Jung HS, Shin HM, Kim TN, Lee SH, et al. The direct effect of lobeglitazone, a new thiazolidinedione, on pancreatic beta cells: A comparison with other thiazolidinediones. Diabetes Research and Clinical Practice. 2019; 151: 209–223.
[138]
Elia I, Haigis MC. Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism. Nature Metabolism. 2021; 3: 21–32.
[139]
Mi H, Sivagnanam S, Betts CB, Liudahl SM, Jaffee EM, Coussens LM, et al. Quantitative Spatial Profiling of Immune Populations in Pancreatic Ductal Adenocarcinoma Reveals Tumor Microenvironment Heterogeneity and Prognostic Biomarkers. Cancer Research. 2022; 82: 4359–4372.
[140]
Liudahl SM, Betts CB, Sivagnanam S, Morales-Oyarvide V, da Silva A, Yuan C, et al. Leukocyte Heterogeneity in Pancreatic Ductal Adenocarcinoma: Phenotypic and Spatial Features Associated with Clinical Outcome. Cancer Discovery. 2021; 11: 2014–2031.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top