Next Article in Journal
Propranolol and Capecitabine Synergy on Inducing Ferroptosis in Human Colorectal Cancer Cells: Potential Implications in Cancer Therapy
Next Article in Special Issue
Recent Strategies to Attenuate Hepatocellular Carcinoma Recurrence After Liver Transplantation: A Narrative Review
Previous Article in Journal
The Outcome of Colonoscopy-Assisted Laparoscopic Wedge Resections (CAL-WR) for Colon Cancer: A Retrospective Cohort Study
Previous Article in Special Issue
TAMing Gliomas: Unraveling the Roles of Iba1 and CD163 in Glioblastoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunometabolism of Innate Immune Cells in Gastrointestinal Cancer

by
Izabela Siemińska
1,2,* and
Marzena Lenart
2,*
1
Institute of Veterinary Sciences, University Center of Veterinary Medicine JU-AU, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059 Krakow, Poland
2
Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, 30-663 Krakow, Poland
*
Authors to whom correspondence should be addressed.
Cancers 2025, 17(9), 1467; https://doi.org/10.3390/cancers17091467
Submission received: 5 March 2025 / Revised: 11 April 2025 / Accepted: 23 April 2025 / Published: 27 April 2025
(This article belongs to the Special Issue Advanced Research in Oncology in 2025)

Simple Summary

This review explores the connection between immunometabolism and innate immune cells in gastrointestinal cancers. While cancer cells consume large amounts of nutrients, immune cells in the tumor microenvironment also have unique metabolic demands that influence immune responses. The review examines how metabolic pathways such as glycolysis, fatty acid oxidation, and amino acid metabolism contribute to the immunosuppressive functions of myeloid-derived suppressor cells (MDSCs), tumor-associated neutrophils (TANs), tumor-associated macrophages (TAMs), and innate lymphocytes such as NK cells. These cells create a tumor-friendly environment, helping cancer evade immune attack. The review also discusses how dietary interventions can reshape immune cell metabolism and highlights emerging therapies targeting these metabolic weaknesses to boost antitumor immunity. Understanding these processes could lead to new strategies for improving cancer treatment by manipulating immune cell metabolism.

Abstract

Cancer cells are often described as voracious consumers of nutrients, with glucose frequently cited as a key energy source; however, their metabolic plasticity allows them to adapt and utilize various substrates, including lipids and amino acids, to sustain growth and survival. However, the metabolic demands of immune cells within the tumor microenvironment (TME) are less commonly discussed despite their critical role in shaping the immune response. In this review, we explored the intricate interplay between immunometabolism and innate immunity cells in gastrointestinal cancers. We focused on how metabolic pathways, including glycolysis, fatty acid oxidation, and amino acid metabolism, drive the immunosuppressive functions of myeloid-derived suppressor cells (MDSCs) and tumor-associated neutrophils (TANs), tumor-associated macrophages (TAMs) and innate lymphocyte subsets such as NK cells. These cells contribute to a hostile immune landscape, supporting tumor growth and evasion from immune surveillance in a phenomenon of tumor-derived immunosuppression. Additionally, we investigated the influence of dietary interventions on the metabolic reprogramming of these immune cells, highlighting how nutrition can modulate the TME. Finally, we discussed emerging therapeutic strategies that target metabolic vulnerabilities in MDSCs, TANs, NK cells, and monocytes, offering a novel avenue for enhancing antitumor immunity. By dissecting these mechanisms, we aim to provide insights into how metabolic pathways can be harnessed to improve cancer treatment outcomes. This review underscores the importance of understanding immunometabolism not only as a driver of immune suppression but also as a potential therapeutic target in gastrointestinal cancer.

Graphical Abstract

1. Introduction

Tumor progression is promoted by communication between the tumor and infiltrating immune cells via the secretion of cytokines, growth factors, and enzymes that remodel the tumor microenvironment (TME). Within tumor-infiltrating leukocytes (TILs), a crucial component consists mainly of cells exhibiting immunosuppressive functions, such as tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), but also innate lymphocyte subsets that physiologically takes part in antitumor response, such as NK cells [1,2].
Although an extensive interest in immunometabolism association with immunosuppression emerged quite recently, the first implications of the role of metabolism in tumor progression were observed 100 years ago and are known as the “Warburg effect” [3]. The author reported that cancer cells prefer glycolysis rather than oxidative phosphorylation for glucose metabolism and that glucose is consumed in large amounts. Now, it become apparent that excessive glucose consumption is also characteristic of immune cells, predominantly myeloid cells [4], suggesting that potential therapeutic metabolic interventions do not have to focus only on tumor cells but may manipulate immune response as well.
Immunometabolism plays a central role in dictating the activation and function of innate immune cells within the tumor microenvironment of gastrointestinal cancers. Macrophages, dendritic cells, and natural killer (NK) cells undergo dynamic metabolic reprogramming in response to hypoxia, tumor-derived metabolites, and nutrient fluctuations, which directly influence their antitumor immune responses. In gastrointestinal cancers, this immunometabolic reprogramming is orchestrated by key hub genes and pathways, including HIF-1α, mTORC1, AMPK, PPAR-γ, AhR, and STAT3. HIF-1α, activated under hypoxic conditions, promotes glycolysis and the expression of cytokines such as IL-10 and TGF-β [5,6]. TORC1 enhances glycolytic metabolism and inflammatory responses but can also contribute to immunosuppression when persistently activated, particularly through its interactions with lipid metabolism and autophagy [7,8]. Conversely, AMPK activation supports antitumor immunity by promoting oxidative metabolism and inhibiting the Warburg effect [9]. PPAR-γ regulates lipid uptake and storage, influencing immune responses through modulation of cytokine expression and metabolic state [10,11]. AhR, activated by environmental and endogenous ligands, contributes to immune suppression by enhancing glycolysis and regulating tryptophan metabolism via the kynurenine pathway [12]. Lastly, STAT3, frequently activated by cytokines such as IL-6, promotes glycolytic activity, inhibits oxidative metabolism, and modulates lipid regulatory genes such as SREBP1 [13].
In this review, we summarize the latest knowledge on the role of immunometabolism in regulating innate immune cell functions in gastrointestinal cancers, explore potential therapeutic targets, and discuss future perspectives for further research.

2. Tumor-Associated Macrophages (TAMs)

TAMs are the most abundant innate immune cell infiltration tumor and TME, playing an important role in immune suppression and tumor growth progression. TAMs, being a specific type of macrophages, originate from monocytes, both recruited from the blood and tissue monocytic precursors [14]. Macrophages differentiate from resting, naive M0 macrophages either to M1- or M2-type, depending on the microenvironment, which is generally considered pro- and anti-inflammatory, respectively [15,16].
TAMs were shown to regulate glycolytic metabolic processes of tumor cells [17], including gastrointestinal cancers. In pancreatic ductal adenocarcinoma (PDAC), lysosomal glycoprotein CD68-positive TAMs infiltration was associated with glycolysis, measured as FDG maximal standardized uptake (FDG SUVmax) in a mechanism of glucose transporter 3 (GLUT3) expression stimulation in PDAC cells by IL-8 secreted by TAMs. IL-8 stimulates STAT3 phosphorylation and its translocation into the nucleus, resulting in upregulation of GLUT3 gene transcription by its promoter activation [18]. Interestingly, GLUT3 was shown before as an important factor required for M2 macrophage polarization. GLUT3-induced M2 polarization relies on IL-4/STAT6 activation independently from glucose transport [19]. Consistent with this phenomenon, He Z. et al. observed that, in gastric cancer, GLUT-3-dependant glycolysis program activation in macrophages by IL-13 secreted by YAP1-overexpressing tumor cells promotes M2 polarization. As a result, M2 macrophages promoted 5-fluorouracil (5-FU) resistance of tumor cells in a mechanism of CCL8 activation of JAK1/STAT3 signaling pathway in tumor cells [20]. Cancer progression and metastasis were also shown to be promoted by M2-like TAMs capacity to consume intratumoral glucose, which fuels hexosamine biosynthetic pathway-dependent O-GlcNAcylation. Glucose metabolism resulted in O-GlcNAcylation of the lysosome-encapsulated protease Cathepsin B, elevating its level in TAMs and promoting its secretion into TME [21]. Elevated Cathepsin B expression was already associated with cancer progression, including CRC [22].
On the other hand, gastrointestinal cancers showed increased expression of lactate, which resulted from hypoxia and acidosis, which were shown to correlate with elevated risk of tumor metastasis and poor prognosis [23]. In CRC, high lactate levels inhibit TAMs glucose consumption [24]. CRC in vitro studies, in which SW480 and HT29 cell lines were cultured under hypoxic conditions, showed that the cell supernatants containing elevated lactate concentration stimulated M2-like polarization of THP-1 macrophages. Inhibition of lactate both reversed the effect on macrophage polarization and inhibited tumor growth [25]. M2 polarization of macrophages by lactate is mediated through the AKT/ERK signaling pathway [26], and this mechanism was also shown in esophageal cancer [27]. Lactate stimulates high-mobility group box 1 (HMGB1) expression and secretion by macrophages, promoting ERK, EMT, and Wnt signaling pathways activation in tumor cells [25]. Others showed that lactate-stimulated M2 macrophages secreted CCL8 and promoted CRC proliferation and metastasis through the CCL8/CCR5/mTORC1 axis [26].
In cancer progression, a dysregulation of fatty acid (FA) metabolism associated with FA oxidation (FAO) is frequently observed. The stimulation of peroxisome proliferator-activated receptor γ (PPAR-γ) was shown to increase FA metabolism in macrophages isolated from tumors in a murine model, resulting in their polarization towards M2-type [28]. Others showed, using a co-culture model of colorectal cancer (CRC) cells and macrophages, that upregulation of FAO in macrophages is associated with the expression of ALKBH5 and CPT1A genes. ALKBH5 enhances fatty acid metabolism and M2 polarization of macrophages by upregulating CPT1A, promoting CRC development [29]. In this study, CPT1A gene expression stimulation is mediated by removing m6A (N6-methyladenosine) modification, which is a reversible RNA epigenetic modification that regulates RNA processing, shown to take part in tumor development and progression [30].
Miao H. et al. showed that CRC-associated macrophages exhibit ectopic expression of AB-hydrolase containing 5 (ABHD5), which promotes tumor growth [31]. ABHD5 is a co-activator of adipose triglyceride lipase (ATGL), which plays a crucial role in the lipolysis of triglycerides into diglycerides and free fatty acids [32]. ABHD5 suppresses spermidine synthase (SRM)-dependent spermidine production by inhibition of the reactive oxygen species-dependent expression of C/EBPɛ, which, in turn, activates SRM gene expression. Macrophage-specific ABHD5 transgene-induced CRC growth in a subcutaneous xenograft mouse CRC model was suppressed when macrophages were transfected with additional SRM transgene [31].
What is more, tumor-derived lactate stimulates epigenetic modifications, in particular histone lysine lactylation (Kla), a phenomenon that was first documented by Zhang et al. in 2019, which occurs as a lactyl (La) group addition to the lysine amino acid residues of histone proteins [33]. Li X. M. et al. showed that, in CRC, tumor-derived lactate downregulates retinoic acid receptor γ (RARγ) expression in macrophages, which results in upregulation of IL-6 levels in the TME and macrophage tumor-promoting functions via TRAF6-IL-6-STAT3 signaling [24]. Moreover, very recent studies revealed an important role of histone lactylation and m6A methyltransferase METTL3 in KCNK6 in inflammation-associated carcinogenesis in mouse models of colon cancer. Increased Kcnk6 stability and upregulated expression of Kcnk6 gene, in a YTHDF2-dependent manner, mediated by METTL3-mediated m6A modification and histone lactylation, respectively, increases NLRP3 inflammasome activation and inflammation-associated carcinogenesis [34].
In the case of amino acid metabolism, a crucial role seems to be played by tryptofan and its metabolism main sensor, the aryl hydrocarbon receptor (AhR). In PDAC, high AhR activity was associated with TAMs pro-tumor activity. AhR deletion or pharmacological inhibition the efficacy of immune-checkpoint blockage, infiltration of cytotoxic CD8+ T cells, and reduced PDAC growth, while TAMs immunosuppressive activity was downregulated by reduced tryptophan supplementation [35]. Moreover, in adenomatous polyposis coli (APC), a key tumor suppressor gene, deficient CRC, tryptophan 2,3-dioxygenase 2 (TDO2) was identified as an essential effector that, via Kyn-AhR pathway, upregulate glycolysis and anabolic cancer cell growth [36]. Single-cell RNA analysis of CRC patient samples revealed that the presence of highly expressing IDO1 macrophages, in comparison to low-IDO1 expressing macrophages, was associated with higher immune cell infiltration, higher immune checkpoint expression, and lower tumor progression [37].
In sum, in gastrointestinal cancers, tumor-derived factors, such as lactate, promote TAM polarization towards the pro-tumor M2 phenotype, which enhances tumor growth, metastasis, and resistance to treatment. We also discussed above how metabolic pathways such as glucose, lipid, and amino acid metabolism, including the key roles of GLUT3, PPAR-γ, and AhR, contribute to the tumor-promoting activities of TAMs. Moreover, recent findings on epigenetic modifications, such as histone lactylation and m6A methylation, add a new dimension to understanding how TAMs influence tumor progression through metabolic reprogramming. These insights underscore the complex metabolic network that fuels cancer development and the potential for targeting TAM metabolism as a therapeutic approach in gastrointestinal malignancies. The relationships discussed above are shown in Figure 1.
Apart from the common immunometabolic alterations observed across gastrointestinal cancers, the metabolic profiles of TAMs are also shaped by the distinct TME of CRC, PDAC, and gastric cancer. In both CRC and gastric cancer, TAMs primarily rely on glycolysis, with CRC, in particular, exhibiting a strong Warburg effect, leading to significant lactate accumulation that promotes an immunosuppressive phenotype and tumor progression [38]. In contrast, the hypovascularized stroma of PDAC imposes nutrient deprivation, compelling TAMs to favor oxidative phosphorylation (OXPHOS) over glycolysis to sustain their survival and pro-tumorigenic functions. Metabolic dependencies also diverge in terms of lipid and amino acid metabolism. In CRC and gastric cancer, lipid metabolism plays a pivotal role, with TAMs accumulating fatty acids that fuel their immunosuppressive state. However, in PDAC, amino acid metabolism dominates particularly glutamine metabolism. PDAC cells exhibit a high dependency on glutamine consumption, while TAMs actively contribute to this pathway by secreting glutamine-derived metabolites, further sustaining tumor growth and metabolic reprogramming [39]. Additionally, in CRC and gastric cancer, TAMs have been shown to upregulate IDO1, which converts tryptophan into kynurenine, a key immunosuppressive metabolite that dampens T-cell activation and promotes immune evasion. This mechanism is less prominent in PDAC, further emphasizing the metabolic divergence between TAMs in different gastrointestinal cancers. Overall, these observations suggest that TAM metabolism in CRC and gastric cancer shares notable similarities. Understanding these metabolic adaptations is crucial for designing metabolically targeted therapies that can selectively disrupt TAM-mediated immunosuppression in different GI cancers.

3. Neutrophils

Neutrophils constitute the largest population of blood leukocytes and, such as various other immune cells, are highly heterogeneous. Circulating neutrophils can be categorized into normal-density neutrophils (NDNs) and low-density neutrophils (LDNs), the latter frequently identified in cancer as polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Tumor-associated neutrophils (TANs), analogous to macrophages, are further classified into TAN1 and TAN2 subsets based on their pro-inflammatory or immunosuppressive functions, respectively [40]. The neutrophil-to-lymphocyte ratio (NLR) has emerged as a prognostic marker in various cancers, including CRC, where elevated NLR correlates with poor survival [41,42].
Neutrophils metabolic activity undergoes significant changes during maturation. Progenitors and early precursors exhibit higher mitochondrial activity and primarily utilize fatty acid oxidation, the TCA cycle, and oxidative phosphorylation (OXPHOS) to meet their energy demands. In contrast, mature neutrophils demonstrate minimal dependence on mitochondrial respiration, favoring glycolysis as their predominant pathway for ATP generation [43]. Moreover, metabolism not only impacts neutrophil function but also neutrophils may act on the metabolism of cancer cells. Studies in CRC have shown that neutrophil extracellular traps (NETs) can induce tumor mitochondrial density, DNA, and ATP production and subsequently affect the growth of tumor cells [44]. In the hepatocellular cell carcinoma model, it has been shown that NETs formation by tumor-associated neutrophils is induced due to a metabolic switch into glycolysis and pentose phosphate pathway [45]. This mechanism was also confirmed in healthy circulating neutrophils [46]. Lactate, a glycolysis product, triggers the polarization of neutrophils into TAN2, which, via extracellular vesicles, enhances the proliferation of colorectal cancer cells [47]. Lactate metabolism by neutrophils has been linked to both colorectal cancer lesions and PDAC in mouse models and human studies, where its upregulation is associated with a shift toward an immunosuppressive phenotype, ultimately facilitating immune evasion [48,49,50].
Knowledge about the impact of amino acid metabolism on neutrophils is limited and only reveals that leucine may induce an antigen-presenting program in neutrophils, which is associated with favorable survival in most cancers [51]. In the case of the suppressive aspect role of arginase (ARG1), indoleamine-2,3-dioxygenase (IDO) and overall arginine metabolism by PMN-MDSCs will be as present lately. In PDAC, neutralizing ARG1 metabolism targets the NETs of TANs, which helps restore the activity of CD8+ T cells in the tumor microenvironment, ultimately enhancing the effectiveness of immune checkpoint therapies [52].
Tumor-associated neutrophils in patients with colorectal cancer have elevated gene levels and protein levels of molecules related to cholesterol metabolism, including the lipid transport receptor CD36 and lipoprotein receptor-related protein-1 (LRP1) and low-density lipoprotein receptor (LDLR) [53]. Moreover, cholesterol metabolism in the neutrophils was linked with the inhibition of NK cells by interrupting lipid raft formation and blocking antitumor signaling pathways [53]. Furthermore, lipid metabolism is also described as an essential way for TANs to survive under glucose-limited conditions [54].
In pancreatic cancer, a distinct population of TANs is characterized by metabolic reprogramming that enhances the TCA cycle and fatty acid metabolism. This shift boosts OXPHOS and amplifies the immunosuppressive activity of these cells also by upregulating PD-L1 expression [55]. Additionally, in PDAC neutrophil population characterized by relatively low glycolysis and a highly active citrate cycle was associated with an increased risk of liver metastasis [56].
In CRC, neutrophil recruitment and NETs formation are inhibited by enoyl-CoA δ-isomerase 2 (ECI2), which is involved in lipid metabolism and reduces lipid-mediated interleukin 8 (IL-8) expression [57]. Neutrophil metabolism undergoes dynamic reprogramming in cancer, with both glycolysis and lipid metabolism contributing to immunosuppressive functions. Enhanced glycolysis supports NET formation and TAN2 polarization via lactate, promoting tumor growth and immune evasion in CRC and PDAC. Similarly, increased fatty acid oxidation and cholesterol metabolism sustain neutrophil activity in nutrient-deprived tumors and interfere with antitumor immune signaling. These metabolic shifts, characteristic of PMN-MDSCs and TAN2, represent key mechanisms by which neutrophils contribute to cancer progression and therapy resistance. The relationships discussed above are shown in Figure 2.

4. Myeloid-Derived Suppressor Cells (MDSCs)

Based on origin, the population of MDSCs is divided into three subpopulations, namely granulocytic/polymorphonuclear (PMN-MDSCs), monocytic (Mo-MDSCs), and “early-stage” MDSCs (e-MDSCs) [2]. Exhibit a range of immunosuppressive mechanisms, including the inhibition of T cell proliferation, induction of regulatory T cells (Tregs), and the polarization of various cell types, such as monocytes and neutrophils, into immunosuppressive phenotypes [58]. A significant increase in MDSCs was observed in several types of cancer, including gastrointestinal cancers, and this increase serves as a poor prognostic marker [59,60].
MDSCs exhibit an increased demand for and uptake of various metabolites, such as glucose, amino acids, and lipids, which are critical to sustaining their survival, immunosuppressive functions, and metabolic reprogramming within the tumor microenvironment. The immunosuppressive function of MDSCs associated with the development of CRC was linked with the intestinal fungus Candida tropicalis, which may enhance the uptake of glucose by MDSCs with simultaneous production of lactate and extracellular acidification rate (ECAR) level [61]. A similar observation was reported in gastric metaplasia induced by Helicobacter, where MDSCs exhibited high expression of genes such as Hexokinase (Hk), Triose Phosphate Isomerase (Tpi), Lactate Dehydrogenase (Ldh) and Glucose Transporter (Glut), all of which are involved in energy metabolism [62] which may also supply sufficient energy to support the immunosuppressive functions of MDSCs [63]. In PDAC, it has been shown that radiation may induce the activity of MDSCs due to lactate production [49], whereas inhibition of lactate production in a colorectal cancer mouse model led to a decreased presence of MDSCs but also other immunosuppressive cell populations such as regulatory T cells (Tregs) within the TEM [64].
MDSCs play a pivotal role in the metabolism of L-arginine, which is crucial for T-cell proliferation and represents a primary mechanism of their immunosuppressive activity. MDSCs enhance the expression of key enzymes, including ARG1 and inducible nitric oxide synthase (iNOS), while upregulating the cationic amino acid transporter 2 (CAT2) to effectively deplete L-arginine from the TME [65]. L-arginine depletion results in suppressed T-cell activity and a dampened antitumor immune response [66]. Similar influence on T cells have depletion of tryptophan where T-MDSCs in CRC express high IDO level responsible for tryptophan metabolism [67]. Amino acids such as arginine and glutamine are closely associated with MDSCs chemotaxis to tumor sites. Through the ERK/ETS2 signaling pathway, arginine can drive the transcription of CXCL1, a chemokine that acts as a chemoattractant for MDSCs by binding to the CXCR2 receptor [68].
In the case of lipid metabolism, it has been shown that tumor-infiltrating MDSCs (T-MDSCs) in colon cancer models increased fatty acid uptake and activated fatty acid oxidation. This was accompanied by an increased mitochondrial mass, upregulation of key FAO enzymes, and increased oxygen consumption rate [54]. Tumor-infiltrating MDSCs preferentially uses fatty acid β oxidation as a primary source of energy with displayed a significantly elevated expression of genes associated with FAO, including CPT1, ACADM, PGC1β, and HADHA [54]. Moreover, tumor-derived cytokines, such as G-CSF and GM-CSF, activate downstream signaling pathways, particularly STAT3 and STAT5, which drive the expression of lipid transport receptors such as CD36 on MDSCs. This upregulation enhances lipid uptake from the lipid-rich tumor microenvironment, thereby promoting the differentiation and accumulation of highly immunosuppressive human MDSCs in vitro [69]. These results underscore the pivotal role of lipid metabolism in shaping MDSCs function and their contribution to tumor immune evasion. In lipid metabolism is also involved another marker highly expressed on lectin-type oxidized LDL receptor 1 (LOX-1). Its expression on PMN-MDSCs of cancer patients was associated with response to endoplasmic reticulum (ER) stress, which seems to be a specific marker of human PMN-MDSCs [70], which may bind other ligands, including other modified lipoproteins, advanced glycation end products, aged red blood cells, apoptotic cells, and activated platelets [71]. In the context of lipid metabolism, it has been shown that decreased expression of miR-4435 promotes CRC tumorigenesis, in part by increasing the infiltration of PMN-MDSCs into the tumor microenvironment [72]. Moreover, fatty acid transport protein 2 (FATP2) facilitates the uptake of arachidonic acid and the synthesis of prostaglandin E2. Its selective inhibition not only suppresses the immunosuppressive activity of PMN-MDSCs and delays tumor progression but also enhances the efficacy of immune checkpoint inhibitors, positioning FATP2 as a promising therapeutic target for improving cancer treatment through the selective modulation of PMN-MDSCs function [73].
During the metabolism of lipids, carbohydrates, and amino acids vital coenzyme is NAD⁺. It participates in glycolysis and the Krebs cycle, acting as an electron carrier. In lipid metabolism, it is essential for beta-oxidation of fatty acids. In amino acid metabolism, it supports dehydrogenase reactions during their conversion to Krebs cycle intermediates. NAD⁺ can be regenerated through the mitochondrial electron transport chain or synthesized de novo from tryptophan or via salvage pathways using nicotinamide. This highlights its central role in energy production and metabolic regulation [74]. It seems that NAD+ is also important in the regulation of immune cells. In the case of MDSCs, CD38 catalyzes the conversion of NAD+ into ADP-ribose (ADPR) and cyclic ADPR (cADPR), which are pivotal for modulating cellular metabolic processes and signaling. The depletion of NAD+ by CD38 activity not only alters intracellular energy homeostasis but also reshapes the extracellular metabolite milieu within the tumor microenvironment [75]. An increased expression of CD38 was shown on Mo-MDSCs in advanced CRC penitents especially this one after therapy [76]. Whereas murine model esophageal cancer shows that MDSCs with CD38 expression poses more immature features [77]. Given its central role in MDSCs metabolism and immune suppression, CD38 represents a promising therapeutic target for reprogramming the tumor microenvironment and restoring antitumor immunity. Moreover, metformin-induced activation of 5′AMP-activated protein kinase (AMPK) decreased the MDSCs and M2 macrophage fractions by downregulating the mevalonate pathway [78]. Mevalonate production is an essential biosynthetic step that provides the precursors for de novo synthesis of cholesterol, which acts as a precursor for various membrane signal transduction and several other cellular components [79]. Taking it all together, glycolysis and lactate production drive their expansion, while lipid uptake and FAO sustain MDSCs survival and immunosuppressive activity. The relationships discussed above are shown in Figure 3.

5. NK Cells

NK cells play a key role in an anticancer immune response, being able to directly recognize and kill malignant cells. It is widely known that cancer cells and tumor microenvironment, including gastrointestinal cancers, impair NK cell activity as a part of the immune evasion process [80,81,82,83]. NK cell functional inhibition was associated with metabolic alterations, yet still little is known about NK cell immunometabolism in gastrointestinal cancers.
Glycolysis was proved to be essential for NK cell activation and, in hypoxic TME conditions, is driven by HIF-1α as overexpression of HIF-1α expression boosts glycolysis and NK cell activity in vitro and in vivo [84].
Tumor-derived lactate’s role in NK cell function seems indirect, relying mostly on the regulation of other cell subsets that can mediate NK cell dysfunction in TME, such as MDSCs or Treg cells [85,86]. The study of Ge W. showed, though, that NK cell’s functional impairment might be directly associated with lactate via sine oculis homeobox homolog 1 (SIX1)/lactate dehydrogenase A (LDHA) axis in pancreatic cancer. SIX1 expression was overexpressed in pancreatic cancer clinical samples, while NK cell co-culture with SIX-1 overexpressing pancreatic cancer cell lines resulted in downregulation of activation and cytotoxic markers in NK cells. NK cell impairment was reversed by the treatment of tumor cells with LDHA inhibitor and lactate transporter blocker [87].
NK cell defective lipid metabolism was shown to inhibit NK cell killing and dysregulate cytokine production in patients with inflammatory bowel disease, and this phenomenon was associated with aberrant mTORC1 activity. The assessment of mitochondria functions and glycoses revealed a distinct NK cell bioenergetic program relying mainly on the latter [88]. Sheppard S. et al. [89] showed that mice challenged with MHC class I-negative RMA-S lymphoma increased the uptake of fatty acids and the expression of carnitine palmitoyltransferase I (CPT1A). CPT1A is a key enzyme involved in fatty acid oxidation (FTO) in mitochondria, being involved in preventing long-chain fatty acid entry into the mitochondria [90]. CPT1A is required for NK cell cytotoxicity in a mechanism associated with cytoskeleton rearrangements, as CPT1A ablation reduces cytotoxicity due to impaired immune synapse formation [89].
FTO and CPT1A’s key role in NK cell metabolism was also shown in studies of the effect of fasting on NK cell anticancer activity. In the murine model of colorectal adenocarcinoma, cyclic fasting diet caused upregulation of glucocorticoids that mediate upregulation of CPT1A expression and fatty acids oxidation in spleen NK cells, although FA uptake by the cells was unaffected. Cpt1a−/− NK cells were unable to control tumor growth during fasting when compared to WT NK cells, which observation was associated with low Cpt1a−/− NK cells tumor infiltration. During fasting, NK cells were also redistributed from peripheral tissues to the bone marrow, and, in contrast to spleen NK cells, BM NK cells showed reduced FA uptake during fasting and downregulated CPT1A expression [91].
Amino acid metabolism of NK cells in gastrointestinal cancers is poorly understood. In gastric cancer, L-kynurenine, a metabolite of the amino acid l-tryptophan, mainly by IDO from tumor cells, impairs NK cells viability in TME, inducing ferroptosis in NK cells via an AhR-independent way [92]. Moreover, IDO1 induces NK cell dysfunction via downregulation of the activating receptor NKG2D on NK cells by inhibiting NKG2D ligand (NGD2DL) expression on lung cancer cells via the IDO1-Kyn-AhR signaling pathway [93]. Finally, IDO1 promotes tryptophan metabolism in the TME, recruiting and activating MDSCs, which, in turn, limits the availability of tryptophan for NK cells, causing their apoptosis [94]. The relationships discussed above are shown in Figure 4.
In sum, NK cells play a central role in antitumor immunity by directly recognizing and killing malignant cells; however, their activity is often suppressed in the TME of gastrointestinal cancers as part of immune evasion mechanisms. Recent studies have highlighted the importance of metabolic pathways such as glycolysis, fatty acid oxidation, and amino acid metabolism in regulating NK cell activation, cytotoxicity, and survival. However, much of this knowledge remains fragmentary. Glycolysis, lipid metabolism, and amino acid pathways all play important roles in regulating NK cell function within the tumor microenvironment. Factors such as hypoxia, tumor-derived metabolites, and immune-suppressive signals can disrupt these metabolic processes, leading to reduced NK cell activation and cytotoxicity. Altered fatty acid and tryptophan metabolism, in particular, have been linked to impaired NK cell responses in gastrointestinal cancers.
However, many aspects of NK cell immunometabolism in gastrointestinal cancers are unexplored. First of all, there is a notable lack of studies utilizing human tumor samples or patient-derived NK cells. Additionally, while research has predominantly focused on glycolysis and fatty acid oxidation, other metabolic pathways, such as amino acid metabolism, mitochondrial dynamics, and cholesterol metabolism, are underexplored in this context. For instance, the depletion of amino acids such as arginine and tryptophan in the tumor microenvironment has been shown to impair NK cell proliferation and function, highlighting the need for deeper investigation into these pathways [95]. Furthermore, mitochondrial fragmentation has been observed in tumor-infiltrating NK cells within human liver cancers, correlating with reduced cytotoxicity and poor patient prognosis [96]. Future studies should focus on (1) comprehensively profiling NK cell metabolism within gastrointestinal tumor tissue using advanced single-cell multi-omics approaches, (2) investigating how metabolic interventions, such as dietary modulation or metabolic inhibitors, may synergize with existing immunotherapies, and (3) exploring strategies to modulate mitochondrial dynamics to enhance NK cell function and tumor infiltration. Addressing these areas could pave the way for novel NK cell-based metabolic immunotherapies in gastrointestinal cancers.

6. Dendritic Cells

Dendritic cells (DCs) are a metabolically active and functionally diverse group of antigen-presenting cells, crucial for initiating and regulating antitumor immune responses. Among them, we distinguish conventional DCs type 1 (cDC1) and type 2 (cDC2), plasmacytoid DCs (pDCs), as well as monocyte-derived DCs (moDCs), each with distinct metabolic profiles and immunological roles. Among dendritic cells, we distinguish conventional DC type 1 (cDC1) and type 2 (cDC2), plasmacytoid DCs (pDCs), as well as monocyte-derived DCs (moDCs). Antitumor immunity of cDC1 has been emphasized by various studies, as well as colorectal cancer and pancreatic cancer [97,98]. DC2 represents a more heterogeneous population with a complex and still poorly defined role, especially in gastrointestinal cancers. In PDAC, cDC2 showed metabolic reprogramming, characterized by increased glycolysis and fatty acid metabolism, as well as increased activity in pathways related to hypoxia production of reactive oxygen species, which was associated with impaired antitumor immunity [99].
More recently, a higher frequency of tumor-infiltrating pDCs was associated with lower tumor stages and improved survival of colon cancer patients [100]. Moreover, higher level of blood-circulating pDCs was associated with longer survival of pancreatic cancer patients [101]. Mo-DCs, usually absent in a steady state, may arise from monocytes during inflammatory responses, including cancer, and drive antitumor immunity [102].
Glutamine plays a key role in enhancing CD8+ lymphocyte antitumor immunity through activation of DC1. However, in the colorectal cancer model, it was shown that glutamine availability is limited by competition between tumor cells and DC1, which use the SLC38A2 transporter for its uptake. This metabolic interaction affects the regulation of the immune response and may determine the efficacy of antitumor mechanisms [103]. Similarly, neutrophils, DC, and monocytes in colorectal cancer patient samples showed increased IDO expression [104]. Where increased expression of IDO in DCs can lead to the conversion of tryptophan to N-formylcine, which promotes the activation and function of Tregs, leading to suppression of the immune response [105].
Additionally, oxidase lipid storage was observed in DC which impairs their ability to cross-present tumor antigens in mouse CRC cell lines [106]. Moreover, similar observations with restore of DC function and enhance the efficacy of cancer vaccines after pharmacological normalization of lipid level confirm another study [107]. All these findings suggest that the metabolic status of DC subsets has a critical impact on their ability to elicit effective antitumor responses. Abnormal metabolic reprogramming, whether through glutamine deficiency, tryptophan catabolism, or lipid accumulation, can impair antigen presentation and promote immunosuppression. Therefore, targeting DC metabolism may be a promising strategy to restore immune competence and improve the outcome of cancer immunotherapy. The relationships discussed above are shown in Figure 5.

7. Immunometabolism-Associated Therapy Approaches

Immunometabolism-associated therapy concerning innate immune cells is still unavailable, yet it might revolutionize future anticancer immunotherapy. At the moment, some potential therapeutics were suggested for macrophage, neutrophils, DCs and MDSCs metabolic alterations treatment (Table 1), while other populations, such as NK cells, require further studies.
Immunometabolism-associated might become a double-edged sword. For example, the usage of CPT1A as a potential target for cancer immunotherapy. CPT1A-targeted therapy was shown to boost radiation therapy sensitivity of nasopharyngeal carcinoma since CPT1A-Rab14 interaction promotes radiation resistance [111]. CPT1A overexpression was also observed in CRC cells, and its inhibition by the treatment with a secolignan-type compound, 2,6-dihydroxypeperomin B (DHP-B), a compound isolated from the plant Peperomia dindygulensis, suppresses tumor growth and progression [112]. However, CPT1A inhibition might have a negative impact on NK cell functions, as CPT1A upregulation promotes the anticancer activity of these cells. In the context of future therapies targeting the metabolism of immune cells in cancer, including gastrointestinal cancers, it is crucial to consider the safety and systemic implications of these interventions. The metabolic pathways targeted in immune cells, such as glycolysis and lipid and amino acid metabolism, are fundamental for the proper activity of not only immune cells but also numerous other cell types. Therefore, potential therapeutics may inadvertently affect non-immune tissues and organs, leading to side effects such as gastrointestinal mucosal injury, hepatotoxicity, and myelosuppression [113]. This is especially relevant in gastrointestinal cancers, in which the tumor microenvironment is characterized by metabolic competition and hypoxia. These conditions may impair T cell function and promote immune evasion [114,115]. Additionally, the overlap in metabolic dependencies between immune and tumor cells adds complexity—targeting pathways such as glutaminolysis or fatty acid oxidation may promote antitumor immune responses but also risk promoting tumor development [116]. For example, the monocarboxylate transporter 1 (MCT1) inhibitor lonidamine, investigated for its metabolic effects in cancer, has been associated with adverse effects, including alopecia and myelosuppression. This was demonstrated by Ning and Hahn, who reported the cytotoxicity of lonidamine both alone and in combination with other agents in murine and human cell lines [117]. Therefore, it is essential to develop selective immunometabolic therapies that minimize systemic toxicity while effectively enhancing antitumor immunity in gastrointestinal cancers. Moreover, inter-individual factors such as metabolic comorbidities and microbiome composition may further influence the safety and efficacy of these approaches [118], underscoring the need for precision strategies tailored to the unique metabolic landscape of the gastrointestinal tumor microenvironment.

8. Conclusions

We are now starting to understand that immunometabolism plays a pivotal role in antitumor immune response and that targeting metabolic pathways opens new possibilities for cancer immunotherapy.
Promising approaches seem to be associated with dietary interventions, especially with fasting. It was shown that cyclic fasting might restore tumor-associated NK cell functional impairment, and, importantly, this observation comes from piloting the human study [91]. Fasting-mimicking diet impairs the pro-tumor functions of TAMs, especially under hypoxic conditions [119], as well as promotes protective gut microbiota, particularly Lactobacillus johnsonii, and increases CD45+ and CD8+ T cells, thereby suppressing tumor growth [120]. Clinical trials in colorectal cancer patients indicate that preoperative consumption of carbohydrate drinks reduced postoperative NLR values and the incidence and severity of postoperative complications following open colorectal surgery, compared with a preoperative fasting protocol [121]. In liver cancer, the blockage of glutamine uptake eliminates hydroxycarboxylic acid receptor 2, also known as GPR109A, and niacin receptor 1 (NIACR1) suppressive effect on myeloid cells [122].
Targeting MDSCs is particularly interesting, as numerous studies indicate that standard oncological treatments often remain ineffective against the MDSCs population [123]. In addition to the approach discussed in this study—modulating lipid metabolism to reduce the number or activity of MDSCs—inhibition of GLUTS, which is overexpressed on CD205⁺ PMN-MDSCs, has been shown to potentially restore antitumor immunity, as proved in a breast cancer model [124].
Interesting observations come from viral studies due to similarities in NK cell functional impairment in cancer and viral infections [125]. Retroviral studies proved that diet-related low iron levels lead to functional NK cell impairment, while iron supplementation enhances NK cell proliferation and their cytotoxic capacity [126,127]. NK cell nutrient uptake relies on the transcription factor IRF4, which is also important for the differentiation and expansion of cytomegalovirus-specific NK cells [128].
Finally, there are many understudied aspects of immunometabolism in gastrointestinal cancers that would be worth focusing on. For instance, in melanoma, the immunosuppressive activity of macrophages was associated with PERK signaling, which plays a crucial role in mitochondrial respiration and lipid oxidation [129], while NK cell functions and survival is inhibited by lactic acid [130]. Another highly understudied topic is an alteration of NK cell glucose metabolism in gastrointestinal cancers. Poznanski S. et al. showed that suppression of glycolysis in NK cells in ovarian cancer and their metabolic flexibility depending on TME in ovarian cancer [131].
In sum, further studies would help to address and overcome immunometabolic hurdles, supporting the development of effective gastrointestinal cancer immunotherapy.

Author Contributions

Conceptualization, I.S. and M.L.; writing—review and editing, I.S. and M.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Acknowledgments

All figures were created with BioRender.com, accessed on 15 April 2025.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Álvarez-Carrasco, P.; Maldonado-Bernal, C. The Innate Defenders: A Review of Natural Killer Cell Immunotherapies in Cancer. Front. Immunol. 2024, 15, 1482807. [Google Scholar] [CrossRef] [PubMed]
  2. Bronte, V.; Brandau, S.; Chen, S.-H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for Myeloid-Derived Suppressor Cell Nomenclature and Characterization Standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [PubMed]
  3. Warburg, O. The Metabolism of Carcinoma Cells. J. Cancer Res. 1925, 9, 148–163. [Google Scholar] [CrossRef]
  4. Kelly, B.; O’Neill, L.A.J. Metabolic Reprogramming in Macrophages and Dendritic Cells in Innate Immunity. Cell Res. 2015, 25, 771–784. [Google Scholar] [CrossRef]
  5. Semenza, G.L. Hypoxia-Inducible Factors in Physiology and Medicine. Cell 2012, 148, 399–408. [Google Scholar] [CrossRef]
  6. Basheeruddin, M.; Qausain, S.; Basheeruddin, M.; Qausain, S. Hypoxia-Inducible Factor 1-Alpha (HIF-1α): An Essential Regulator in Cellular Metabolic Control. Cureus 2024, 16, e63852. [Google Scholar] [CrossRef]
  7. Saxton, R.A.; Sabatini, D.M. MTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef]
  8. O’Neill, L.A.J.; Kishton, R.J.; Rathmell, J. A Guide to Immunometabolism for Immunologists. Nat. Rev. Immunol. 2016, 16, 553–565. [Google Scholar] [CrossRef]
  9. Faubert, B.; Solmonson, A.; DeBerardinis, R.J. Metabolic Reprogramming and Cancer Progression. Science 2020, 368, eaaw5473. [Google Scholar] [CrossRef]
  10. Tian, X.; Bai, X.; Han, Y.; Ye, Y.; Peng, M.; Cui, H.; Li, K.; Han, Y. PPAR γ Changing ALDH1A3 Content to Regulate Lipid Metabolism and Inhibit Lung Cancer Cell Growth. Mol. Genet. Genomics 2025, 300, 41. [Google Scholar] [CrossRef]
  11. Ricote, M.; Li, A.C.; Willson, T.M.; Kelly, C.J.; Glass, C.K. The Peroxisome Proliferator-Activated Receptor-Gamma Is a Negative Regulator of Macrophage Activation. Nature 1998, 391, 79–82. [Google Scholar] [CrossRef] [PubMed]
  12. Chaudhry, K.A.; Bianchi-Smiraglia, A. The Aryl Hydrocarbon Receptor as a Tumor Modulator: Mechanisms to Therapy. Front. Oncol. 2024, 14, 1375905. [Google Scholar] [CrossRef] [PubMed]
  13. Tolomeo, M.; Cascio, A. The Multifaced Role of STAT3 in Cancer and Its Implication for Anticancer Therapy. Int. J. Mol. Sci. 2021, 22, 603. [Google Scholar] [CrossRef]
  14. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef]
  15. Jumaniyazova, E.; Lokhonina, A.; Dzhalilova, D.; Miroshnichenko, E.; Kosyreva, A.; Fatkhudinov, T. The Role of Macrophages in Various Types of Tumors and the Possibility of Their Use as Targets for Antitumor Therapy. Cancers 2025, 17, 342. [Google Scholar] [CrossRef]
  16. Ngambenjawong, C.; Gustafson, H.H.; Pun, S.H. Progress in Tumor-Associated Macrophage (TAM)-Targeted Therapeutics. Adv. Drug Deliv. Rev. 2017, 114, 206–221. [Google Scholar] [CrossRef]
  17. Jeong, H.; Kim, S.; Hong, B.J.; Lee, C.J.; Kim, Y.E.; Bok, S.; Oh, J.M.; Gwak, S.H.; Yoo, M.Y.; Lee, M.S.; et al. Tumor-Associated Macrophages Enhance Tumor Hypoxia and Aerobic Glycolysis. Cancer Res. 2019, 79, 795–806. [Google Scholar] [CrossRef]
  18. Zhong, Z.; Yang, K.; Li, Y.; Zhou, S.; Yao, H.; Zhao, Y.; Huang, Y.; Zou, J.; Li, Y.; Li, J.; et al. Tumor-Associated Macrophages Drive Glycolysis through the IL-8/STAT3/GLUT3 Signaling Pathway in Pancreatic Cancer Progression. Cancer Lett. 2024, 588, 216784. [Google Scholar] [CrossRef]
  19. Yu, D.M.; Zhao, J.; Lee, E.E.; Kim, D.; Mahapatra, R.; Rose, E.K.; Zhou, Z.; Hosler, C.; El Kurdi, A.; Choe, J.Y.; et al. GLUT3 Promotes Macrophage Signaling and Function via RAS-Mediated Endocytosis in Atopic Dermatitis and Wound Healing. J. Clin. Investig. 2023, 133, e170706. [Google Scholar] [CrossRef]
  20. He, Z.; Chen, D.; Wu, J.; Sui, C.; Deng, X.; Zhang, P.; Chen, Z.; Liu, D.; Yu, J.; Shi, J.; et al. Yes Associated Protein 1 Promotes Resistance to 5-Fluorouracil in Gastric Cancer by Regulating GLUT3-Dependent Glycometabolism Reprogramming of Tumor-Associated Macrophages. Arch. Biochem. Biophys. 2021, 702, 108838. [Google Scholar] [CrossRef]
  21. Shi, Q.; Shen, Q.; Liu, Y.; Shi, Y.; Huang, W.; Wang, X.; Li, Z.; Chai, Y.; Wang, H.; Hu, X.; et al. Increased Glucose Metabolism in TAMs Fuels O-GlcNAcylation of Lysosomal Cathepsin B to Promote Cancer Metastasis and Chemoresistance. Cancer Cell 2022, 40, 1207–1222.e10. [Google Scholar] [CrossRef] [PubMed]
  22. Abdulla, M.H.; Valli-Mohammed, M.A.; Al-Khayal, K.; Al Shkieh, A.; Zubaidi, A.; Ahmad, R.; Al-Saleh, K.; Al-Obeed, O.; McKerrow, J. Cathepsin B Expression in Colorectal Cancer in a Middle East Population: Potential Value as a Tumor Biomarker for Late Disease Stages. Oncol. Rep. 2017, 37, 3175–3180. [Google Scholar] [CrossRef] [PubMed]
  23. Huang, A.; Sun, Z.; Hong, H.; Yang, Y.; Chen, J.; Gao, Z.; Gu, J. Novel Hypoxia- and Lactate Metabolism-Related Molecular Subtyping and Prognostic Signature for Colorectal Cancer. J. Transl. Med. 2024, 22, 587. [Google Scholar] [CrossRef] [PubMed]
  24. Li, X.M.; Yang, Y.; Jiang, F.Q.; Hu, G.; Wan, S.; Yan, W.Y.; He, X.S.; Xiao, F.; Yang, X.M.; Guo, X.; et al. Histone Lactylation Inhibits RARγ Expression in Macrophages to Promote Colorectal Tumorigenesis through Activation of TRAF6-IL-6-STAT3 Signaling. Cell Rep. 2024, 43, 113688. [Google Scholar] [CrossRef]
  25. Gao, X.; Zhou, S.; Qin, Z.; Li, D.; Zhu, Y.; Ma, D. Upregulation of HMGB1 in Tumor-Associated Macrophages Induced by Tumor Cell-Derived Lactate Further Promotes Colorectal Cancer Progression. J. Transl. Med. 2023, 21, 53. [Google Scholar] [CrossRef]
  26. Zhou, H.; Yao, J.; Zhong, Z.; Wei, H.; He, Y.; Li, W.; Hu, K. Lactate-Induced CCL8 in Tumor-Associated Macrophages Accelerates the Progression of Colorectal Cancer through the CCL8/CCR5/MTORC1 Axis. Cancers 2023, 15, 5795. [Google Scholar] [CrossRef]
  27. Zhang, C.; Cheng, W.; Yang, T.; Fang, H.; Zhang, R. Lactate Secreted by Esophageal Cancer Cells Induces M2 Macrophage Polarization via the AKT/ERK Pathway. Thorac. Cancer 2023, 14, 2139–2148. [Google Scholar] [CrossRef]
  28. Liu, S.; Zhang, H.; Li, Y.; Zhang, Y.; Bian, Y.; Zeng, Y.; Yao, X.; Wan, J.; Chen, X.; Li, J.; et al. S100A4 Enhances Protumor Macrophage Polarization by Control of PPAR-γ-Dependent Induction of Fatty Acid Oxidation. J. Immunother. Cancer 2021, 9, e002548. [Google Scholar] [CrossRef]
  29. Sun, M.; Yue, Y.; Wang, X.; Feng, H.; Qin, Y.; Chen, M.; Wang, Y.; Yan, S. ALKBH5-Mediated Upregulation of CPT1A Promotes Macrophage Fatty Acid Metabolism and M2 Macrophage Polarization, Facilitating Malignant Progression of Colorectal Cancer. Exp. Cell Res. 2024, 437, 113994. [Google Scholar] [CrossRef]
  30. Xiong, J.; He, J.; Zhu, J.; Pan, J.; Liao, W.; Ye, H.; Wang, H.; Song, Y.; Du, Y.; Cui, B.; et al. Lactylation-Driven METTL3-Mediated RNA M6A Modification Promotes Immunosuppression of Tumor-Infiltrating Myeloid Cells. Mol. Cell 2022, 82, 1660–1677.e10. [Google Scholar] [CrossRef]
  31. Miao, H.; Ou, J.; Peng, Y.; Zhang, X.; Chen, Y.; Hao, L.; Xie, G.; Wang, Z.; Pang, X.; Ruan, Z.; et al. Macrophage ABHD5 Promotes Colorectal Cancer Growth by Suppressing Spermidine Production by SRM. Nat. Commun. 2016, 7, 11716. [Google Scholar] [CrossRef] [PubMed]
  32. Lass, A.; Zimmermann, R.; Haemmerle, G.; Riederer, M.; Schoiswohl, G.; Schweiger, M.; Kienesberger, P.; Strauss, J.G.; Gorkiewicz, G.; Zechner, R. Adipose Triglyceride Lipase-Mediated Lipolysis of Cellular Fat Stores Is Activated by CGI-58 and Defective in Chanarin-Dorfman Syndrome. Cell Metab. 2006, 3, 309–319. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, D.; Tang, Z.; Huang, H.; Zhou, G.; Cui, C.; Weng, Y.; Liu, W.; Kim, S.; Lee, S.; Perez-Neut, M.; et al. Metabolic Regulation of Gene Expression by Histone Lactylation. Nature 2019, 574, 575–580. [Google Scholar] [CrossRef]
  34. Yuan, X.; Wang, Q.; Zhao, J.; Xie, H.; Pu, Z. The M6A Methyltransferase METTL3 Modifies Kcnk6 Promoting on Inflammation Associated Carcinogenesis Is Essential for Colon Homeostasis and Defense System through Histone Lactylation Dependent YTHDF2 Binding. Int. Rev. Immunol. 2025, 44, 1–16. [Google Scholar] [CrossRef] [PubMed]
  35. Hezaveh, K.; Shinde, R.S.; Klötgen, A.; Halaby, M.J.; Lamorte, S.; Ciudad, M.T.; Quevedo, R.; Neufeld, L.; Liu, Z.Q.; Jin, R.; et al. Tryptophan-Derived Microbial Metabolites Activate the Aryl Hydrocarbon Receptor in Tumor-Associated Macrophages to Suppress Anti-Tumor Immunity. Immunity 2022, 55, 324–340.e8. [Google Scholar] [CrossRef]
  36. Lee, R.; Li, J.; Li, J.; Wu, C.J.; Jiang, S.; Hsu, W.H.; Chakravarti, D.; Chen, P.; Labella, K.A.; Li, J.; et al. Synthetic Essentiality of Tryptophan 2,3-Dioxygenase 2 in APC-Mutated Colorectal Cancer. Cancer Discov. 2022, 12, 1702–1717. [Google Scholar] [CrossRef]
  37. Liu, X.; Yan, G.; Xu, B.; Yu, H.; An, Y.; Sun, M. Evaluating the Role of IDO1 Macrophages in Immunotherapy Using ScRNA-Seq and Bulk-Seq in Colorectal Cancer. Front. Immunol. 2022, 13, 1006501. [Google Scholar] [CrossRef]
  38. Zhong, X.; He, X.; Wang, Y.; Hu, Z.; Huang, H.; Zhao, S.; Wei, P.; Li, D. Warburg Effect in Colorectal Cancer: The Emerging Roles in Tumor Microenvironment and Therapeutic Implications. J. Hematol. Oncol. 2022, 15, 160. [Google Scholar] [CrossRef]
  39. Shen, X.; Chen, Y.; Tang, Y.; Lu, P.; Liu, M.; Mao, T.; Weng, Y.; Yu, F.; Liu, Y.; Tang, Y.; et al. Targeting Pancreatic Cancer Glutamine Dependency Confers Vulnerability to GPX4-Dependent Ferroptosis. Cell Rep. Med. 2025, 6, 101928. [Google Scholar] [CrossRef]
  40. Siemińska, I.; Poljańska, E.; Baran, J. Granulocytes and Cells of Granulocyte Origin-The Relevant Players in Colorectal Cancer. Int. J. Mol. Sci. 2021, 22, 3801. [Google Scholar] [CrossRef]
  41. Grenader, T.; Nash, S.; Adams, R.; Kaplan, R.; Fisher, D.; Maughan, T.; Bridgewater, J. Derived Neutrophil Lymphocyte Ratio Is Predictive of Survival from Intermittent Therapy in Advanced Colorectal Cancer: A Post Hoc Analysis of the MRC COIN Study. Br. J. Cancer 2016, 114, 612–615. [Google Scholar] [CrossRef] [PubMed]
  42. Templeton, A.J.; McNamara, M.G.; Šeruga, B.; Vera-Badillo, F.E.; Aneja, P.; Ocaña, A.; Leibowitz-Amit, R.; Sonpavde, G.; Knox, J.J.; Tran, B.; et al. Prognostic Role of Neutrophil-to-Lymphocyte Ratio in Solid Tumors: A Systematic Review and Meta-Analysis. J. Natl. Cancer Inst. 2014, 106, dju124. [Google Scholar] [CrossRef] [PubMed]
  43. Thind, M.K.; Uhlig, H.H.; Glogauer, M.; Palaniyar, N.; Bourdon, C.; Gwela, A.; Lancioni, C.L.; Berkley, J.A.; Bandsma, R.H.J.; Farooqui, A. A Metabolic Perspective of the Neutrophil Life Cycle: New Avenues in Immunometabolism. Front. Immunol. 2024, 14, 1334205. [Google Scholar] [CrossRef] [PubMed]
  44. Yazdani, H.O.; Roy, E.; Comerci, A.J.; Van Der Windt, D.J.; Zhang, H.; Huang, H.; Loughran, P.; Shiva, S.; Geller, D.A.; Bartlett, D.L.; et al. Neutrophil Extracellular Traps Drive Mitochondrial Homeostasis in Tumors to Augment Growth. Cancer Res. 2019, 79, 5626–5639. [Google Scholar] [CrossRef]
  45. Jiang, Z.Z.; Peng, Z.P.; Liu, X.C.; Guo, H.F.; Zhou, M.M.; Jiang, D.; Ning, W.R.; Huang, Y.F.; Zheng, L.; Wu, Y. Neutrophil Extracellular Traps Induce Tumor Metastasis through Dual Effects on Cancer and Endothelial Cells. Oncoimmunology 2022, 11, 2052418. [Google Scholar] [CrossRef]
  46. Rodríguez-Espinosa, O.; Rojas-Espinosa, O.; Moreno-Altamirano, M.M.B.; López-Villegas, E.O.; Sánchez-García, F.J. Metabolic Requirements for Neutrophil Extracellular Traps Formation. Immunology 2015, 145, 213–224. [Google Scholar] [CrossRef]
  47. Wang, J.; Wang, X.; Guo, Y.; Ye, L.; Li, D.; Hu, A.; Cai, S.; Yuan, B.; Jin, S.; Zhou, Y.; et al. Therapeutic Targeting of SPIB/SPI1-Facilitated Interplay of Cancer Cells and Neutrophils Inhibits Aerobic Glycolysis and Cancer Progression. Clin. Transl. Med. 2021, 11, e588. [Google Scholar] [CrossRef] [PubMed]
  48. Wang, L.; Liu, Y.; Dai, Y.; Tang, X.; Yin, T.; Wang, C.; Wang, T.; Dong, L.; Shi, M.; Qin, J.; et al. Single-Cell RNA-Seq Analysis Reveals BHLHE40-Driven pro-Tumour Neutrophils with Hyperactivated Glycolysis in Pancreatic Tumour Microenvironment. Gut 2023, 72, 958–971. [Google Scholar] [CrossRef]
  49. Yang, X.; Lu, Y.; Hang, J.; Zhang, J.; Zhang, T.; Huo, Y.; Liu, J.; Lai, S.; Luo, D.; Wang, L.; et al. Lactate-Modulated Immunosuppression of Myeloid-Derived Suppressor Cells Contributes to the Radioresistance of Pancreatic Cancer. Cancer Immunol. Res. 2020, 8, 1440–1451. [Google Scholar] [CrossRef]
  50. Niu, N.; Shen, X.; Zhang, L.; Chen, Y.; Lu, P.; Yang, W.; Liu, M.; Shi, J.; Xu, D.; Tang, Y.; et al. Tumor Cell-Intrinsic SETD2 Deficiency Reprograms Neutrophils to Foster Immune Escape in Pancreatic Tumorigenesis. Adv. Sci. 2023, 10, e2202937. [Google Scholar] [CrossRef]
  51. Wu, Y.; Ma, J.; Yang, X.; Nan, F.; Zhang, T.; Ji, S.; Rao, D.; Feng, H.; Gao, K.; Gu, X.; et al. Neutrophil Profiling Illuminates Anti-Tumor Antigen-Presenting Potency. Cell 2024, 187, 1422–1439.e24. [Google Scholar] [CrossRef] [PubMed]
  52. Canè, S.; Barouni, R.M.; Fabbi, M.; Cuozzo, J.; Fracasso, G.; Adamo, A.; Ugel, S.; Trovato, R.; De Sanctis, F.; Giacca, M.; et al. Neutralization of NET-Associated Human ARG1 Enhances Cancer Immunotherapy. Sci. Transl. Med. 2023, 15, eabq6221. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, Y.; Wang, Z.; Lu, Y.; Sanchez, D.J.; Li, J.; Wang, L.; Meng, X.; Chen, J.; Kien, T.T.; Zhong, M.; et al. Region-Specific CD16+ Neutrophils Promote Colorectal Cancer Progression by Inhibiting Natural Killer Cells. Adv. Sci. 2024, 11, 2403414. [Google Scholar] [CrossRef]
  54. Hossain, F.; Al-Khami, A.A.; Wyczechowska, D.; Hernandez, C.; Zheng, L.; Reiss, K.; Valle, L.D.; Trillo-Tinoco, J.; Maj, T.; Zou, W.; et al. Inhibition of Fatty Acid Oxidation Modulates Immunosuppressive Functions of Myeloid-Derived Suppressor Cells and Enhances Cancer Therapies. Cancer Immunol. Res. 2015, 3, 1236–1247. [Google Scholar] [CrossRef]
  55. Wang, X.; Hu, L.P.; Qin, W.T.; Yang, Q.; Chen, D.Y.; Li, Q.; Zhou, K.X.; Huang, P.Q.; Xu, C.J.; Li, J.; et al. Identification of a Subset of Immunosuppressive P2RX1-Negative Neutrophils in Pancreatic Cancer Liver Metastasis. Nat. Commun. 2021, 12, 174. [Google Scholar] [CrossRef]
  56. Xu, W.; Liu, J.; Liu, Q.; Xu, J.; Zhou, L.; Liang, Z.; Huang, H.; Huang, B.; Xiao, G.G.; Guo, J. NFE2-Driven Neutrophil Polarization Promotes Pancreatic Cancer Liver Metastasis Progression. Cell Rep. 2025, 44, 115226. [Google Scholar] [CrossRef]
  57. Chen, L.; Dai, P.; Liu, L.; Chen, Y.; Lu, Y.; Zheng, L.; Wang, H.; Yuan, Q.; Li, X. The Lipid-Metabolism Enzyme ECI2 Reduces Neutrophil Extracellular Traps Formation for Colorectal Cancer Suppression. Nat. Commun. 2024, 15, 7184. [Google Scholar] [CrossRef] [PubMed]
  58. Sieminska, I.; Baran, J. Myeloid-Derived Suppressor Cells in Colorectal Cancer. Front. Immunol. 2020, 11, 1526. [Google Scholar] [CrossRef]
  59. Ma, P.; Beatty, P.L.; McKolanis, J.; Brand, R.; Schoen, R.E.; Finn, O.J. Circulating Myeloid Derived Suppressor Cells (MDSC) That Accumulate in Premalignancy Share Phenotypic and Functional Characteristics with MDSC in Cancer. Front. Immunol. 2019, 10, 1401. [Google Scholar] [CrossRef]
  60. Siemińska, I.; Węglarczyk, K.; Walczak, M.; Czerwińska, A.; Pach, R.; Rubinkiewicz, M.; Szczepanik, A.; Siedlar, M.; Baran, J. Mo-MDSCs Are Pivotal Players in Colorectal Cancer and May Be Associated with Tumor Recurrence after Surgery. Transl. Oncol. 2022, 17, 101346. [Google Scholar] [CrossRef]
  61. Zhang, Z.; Zheng, Y.; Chen, Y.; Yin, Y.; Chen, Y.; Chen, Q.; Hou, Y.; Shen, S.; Lv, M.; Wang, T. Gut Fungi Enhances Immunosuppressive Function of Myeloid-Derived Suppressor Cells by Activating PKM2-Dependent Glycolysis to Promote Colorectal Tumorigenesis. Exp. Hematol. Oncol. 2022, 11, 88. [Google Scholar] [CrossRef]
  62. Ding, L.; Sheriff, S.; Sontz, R.A.; Merchant, J.L. Schlafen4+-MDSC in Helicobacter-Induced Gastric Metaplasia Reveals Role for GTPases. Front. Immunol. 2023, 14, 1139391. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, Y.; Jia, A.; Bi, Y.; Wang, Y.; Liu, G. Metabolic Regulation of Myeloid-Derived Suppressor Cell Function in Cancer. Cells 2020, 9, 1011. [Google Scholar] [CrossRef] [PubMed]
  64. Li, N.; Kang, Y.; Wang, L.; Huff, S.; Tang, R.; Hui, H.; Agrawal, K.; Gonzalez, G.M.; Wang, Y.; Patel, S.P.; et al. ALKBH5 Regulates Anti–PD-1 Therapy Response by Modulating Lactate and Suppressive Immune Cell Accumulation in Tumor Microenvironment. Proc. Natl. Acad. Sci. USA 2020, 117, 20159–20170. [Google Scholar] [CrossRef]
  65. Cimen Bozkus, C.; Elzey, B.D.; Crist, S.A.; Ellies, L.G.; Ratliff, T.L. Expression of Cationic Amino Acid Transporter 2 Is Required for Myeloid-Derived Suppressor Cell-Mediated Control of T Cell Immunity. J. Immunol. 2015, 195, 5237–5250. [Google Scholar] [CrossRef]
  66. Won, W.J.; Deshane, J.S.; Leavenworth, J.W.; Oliva, C.R.; Griguer, C.E. Metabolic and Functional Reprogramming of Myeloid-Derived Suppressor Cells and Their Therapeutic Control in Glioblastoma. Cell Stress 2019, 3, 47. [Google Scholar] [CrossRef]
  67. OuYang, L.Y.; Wu, X.J.; Ye, S.B.; Zhang, R.X.; Li, Z.L.; Liao, W.; Pan, Z.Z.; Zheng, L.M.; Zhang, X.S.; Wang, Z.; et al. Tumor-Induced Myeloid-Derived Suppressor Cells Promote Tumor Progression through Oxidative Metabolism in Human Colorectal Cancer. J. Transl. Med. 2015, 13, 47. [Google Scholar] [CrossRef]
  68. Zhou, Q.; Peng, Y.; Ji, F.; Chen, H.; Kang, W.; Chan, L.S.; Gou, H.; Lin, Y.; Huang, P.; Chen, D.; et al. Targeting of SLC25A22 Boosts the Immunotherapeutic Response in KRAS-Mutant Colorectal Cancer. Nat. Commun. 2023, 14, 4677. [Google Scholar] [CrossRef]
  69. Al-Khami, A.A.; Zheng, L.; Del Valle, L.; Hossain, F.; Wyczechowska, D.; Zabaleta, J.; Sanchez, M.D.; Dean, M.J.; Rodriguez, P.C.; Ochoa, A.C. Exogenous Lipid Uptake Induces Metabolic and Functional Reprogramming of Tumor-Associated Myeloid-Derived Suppressor Cells. Oncoimmunology 2017, 6, e1344804. [Google Scholar] [CrossRef]
  70. Condamine, T.; Dominguez, G.A.; Youn, J.I.; Kossenkov, A.V.; Mony, S.; Alicea-Torres, K.; Tcyganov, E.; Hashimoto, A.; Nefedova, Y.; Lin, C.; et al. Lectin-Type Oxidized LDL Receptor-1 Distinguishes Population of Human Polymorphonuclear Myeloid-Derived Suppressor Cells in Cancer Patients. Sci. Immunol. 2016, 1, aaf8943. [Google Scholar] [CrossRef]
  71. Taye, A.; El-Sheikh, A.A.K. Lectin-like Oxidized Low-Density Lipoprotein Receptor 1 Pathways. Eur. J. Clin. Investig. 2013, 43, 740–745. [Google Scholar] [CrossRef] [PubMed]
  72. Yu, H.; Chen, C.; Han, F.; Tang, J.; Deng, M.; Niu, Y.; Lai, M.; Zhang, H. Long Noncoding RNA MIR4435-2HG Suppresses Colorectal Cancer Initiation and Progression by Reprogramming Neutrophils. Cancer Immunol. Res. 2022, 10, 1095–1110. [Google Scholar] [CrossRef] [PubMed]
  73. Veglia, F.; Tyurin, V.A.; Blasi, M.; De Leo, A.; Kossenkov, A.V.; Donthireddy, L.; To, T.K.J.; Schug, Z.; Basu, S.; Wang, F.; et al. Fatty Acid Transport Protein 2 Reprograms Neutrophils in Cancer. Nature 2019, 569, 73–78. [Google Scholar] [CrossRef] [PubMed]
  74. Xie, N.; Zhang, L.; Gao, W.; Huang, C.; Huber, P.E.; Zhou, X.; Li, C.; Shen, G.; Zou, B. NAD+ Metabolism: Pathophysiologic Mechanisms and Therapeutic Potential. Signal Transduct. Target. Ther. 2020, 5, 227. [Google Scholar] [CrossRef]
  75. Malavasi, F.; Deaglio, S.; Funaro, A.; Ferrero, E.; Horenstein, A.L.; Ortolan, E.; Vaisitti, T.; Aydin, S. Evolution and Function of the ADP Ribosyl Cyclase/CD38 Gene Family in Physiology and Pathology. Physiol. Rev. 2008, 88, 841–886. [Google Scholar] [CrossRef]
  76. Karakasheva, T.A.; Dominguez, G.A.; Hashimoto, A.; Lin, E.W.; Chiu, C.; Sasser, K.; Lee, J.W.; Beatty, G.L.; Gabrilovich, D.I.; Rustgi, A.K. CD38+ M-MDSC Expansion Characterizes a Subset of Advanced Colorectal Cancer Patients. JCI Insight 2018, 3, e97022. [Google Scholar] [CrossRef]
  77. Karakasheva, T.A.; Waldron, T.J.; Eruslanov, E.; Kim, S.B.; Lee, J.S.; O’Brien, S.; Hicks, P.D.; Basu, D.; Singhal, S.; Malavasi, F.; et al. CD38-Expressing Myeloid-Derived Suppressor Cells Promote Tumor Growth in a Murine Model of Esophageal Cancer. Cancer Res. 2015, 75, 4074. [Google Scholar] [CrossRef]
  78. Kang, J.; Lee, D.; Lee, K.J.; Yoon, J.E.; Kwon, J.H.; Seo, Y.; Kim, J.; Chang, S.Y.; Park, J.; Kang, E.A.; et al. Tumor-Suppressive Effect of Metformin via the Regulation of M2 Macrophages and Myeloid-Derived Suppressor Cells in the Tumor Microenvironment of Colorectal Cancer. Cancers 2022, 14, 2881. [Google Scholar] [CrossRef]
  79. Juarez, D.; Fruman, D.A. Targeting the Mevalonate Pathway in Cancer. Trends Cancer 2021, 7, 525. [Google Scholar] [CrossRef]
  80. Fionda, C.; Scarno, G.; Stabile, H.; Molfetta, R.; Di Censo, C.; Gismondi, A.; Paolini, R.; Sozzani, S.; Santoni, A.; Sciumè, G. NK Cells and Other Cytotoxic Innate Lymphocytes in Colorectal Cancer Progression and Metastasis. Int. J. Mol. Sci. 2022, 23, 7859. [Google Scholar] [CrossRef]
  81. Han, B.; He, J.; Chen, Q.; Yuan, M.; Zeng, X.; Li, Y.; Zeng, Y.; He, M.; Zhou, Q.; Feng, D.; et al. ELFN1-AS1 Promotes GDF15-Mediated Immune Escape of Colorectal Cancer from NK Cells by Facilitating GCN5 and SND1 Association. Discov. Oncol. 2023, 14, 56. [Google Scholar] [CrossRef] [PubMed]
  82. Sorrentino, C.; D’Antonio, L.; Fieni, C.; Ciummo, S.L.; Di Carlo, E. Colorectal Cancer-Associated Immune Exhaustion Involves T and B Lymphocytes and Conventional NK Cells and Correlates with a Shorter Overall Survival. Front. Immunol. 2021, 12, 778329. [Google Scholar] [CrossRef] [PubMed]
  83. Tumino, N.; Nava Lauson, C.B.; Tiberti, S.; Besi, F.; Martini, S.; Fiore, P.F.; Scodamaglia, F.; Mingari, M.C.; Moretta, L.; Manzo, T.; et al. The Tumor Microenvironment Drives NK Cell Metabolic Dysfunction Leading to Impaired Antitumor Activity. Int. J. Cancer 2023, 152, 1698–1706. [Google Scholar] [CrossRef] [PubMed]
  84. Pelletier, A.; Nelius, E.; Fan, Z.; Khatchatourova, E.; Alvarado-Diaz, A.; He, J.; Krzywinska, E.; Sobecki, M.; Nagarajan, S.; Kerdiles, Y.; et al. Resting Natural Killer Cell Homeostasis Relies on Tryptophan/NAD+ Metabolism and HIF-1α. EMBO Rep. 2023, 24, e56156. [Google Scholar] [CrossRef]
  85. Husain, Z.; Huang, Y.; Seth, P.; Sukhatme, V.P. Tumor-Derived Lactate Modifies Antitumor Immune Response: Effect on Myeloid-Derived Suppressor Cells and NK Cells. J. Immunol. 2013, 191, 1486–1495. [Google Scholar] [CrossRef]
  86. Angelin, A.; Gil-de-Gómez, L.; Dahiya, S.; Jiao, J.; Guo, L.; Levine, M.H.; Wang, Z.; Quinn, W.J.; Kopinski, P.K.; Wang, L.; et al. Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. Cell Metab. 2017, 25, 1282–1293.e7. [Google Scholar] [CrossRef]
  87. Ge, W.; Meng, L.; Cao, S.; Hou, C.; Zhu, X.; Huang, D.; Li, Q.; Peng, Y.; Jiang, K. The SIX1/LDHA Axis Promotes Lactate Accumulation and Leads to NK Cell Dysfunction in Pancreatic Cancer. J. Immunol. Res. 2023, 2023, 6891636. [Google Scholar] [CrossRef]
  88. Zaiatz Bittencourt, V.; Jones, F.; Tosetto, M.; Doherty, G.A.; Ryan, E.J. Dysregulation of Metabolic Pathways in Circulating Natural Killer Cells Isolated from Inflammatory Bowel Disease Patients. J. Crohns Colitis 2021, 15, 1316–1325. [Google Scholar] [CrossRef]
  89. Sheppard, S.; Srpan, K.; Lin, W.; Lee, M.; Delconte, R.B.; Owyong, M.; Carmeliet, P.; Davis, D.M.; Xavier, J.B.; Hsu, K.C.; et al. Fatty Acid Oxidation Fuels Natural Killer Cell Responses against Infection and Cancer. Proc. Natl. Acad. Sci. USA 2024, 121, e2319254121. [Google Scholar] [CrossRef]
  90. Schoors, S.; Bruning, U.; Missiaen, R.; Queiroz, K.C.S.; Borgers, G.; Elia, I.; Zecchin, A.; Cantelmo, A.R.; Christen, S.; Goveia, J.; et al. Fatty Acid Carbon Is Essential for DNTP Synthesis in Endothelial Cells. Nature 2015, 520, 192–197. [Google Scholar] [CrossRef]
  91. Delconte, R.B.; Owyong, M.; Santosa, E.K.; Srpan, K.; Sheppard, S.; McGuire, T.J.; Abbasi, A.; Diaz-Salazar, C.; Chun, J.; Rogatsky, I.; et al. Fasting Reshapes Tissue-Specific Niches to Improve NK Cell-Mediated Anti-Tumor Immunity. Immunity 2024, 57, 1923–1938.e7. [Google Scholar] [CrossRef] [PubMed]
  92. Cui, J.X.; Xu, X.H.; He, T.; Liu, J.J.; Xie, T.Y.; Tian, W.; Liu, J.Y. L-Kynurenine Induces NK Cell Loss in Gastric Cancer Microenvironment via Promoting Ferroptosis. J. Exp. Clin. Cancer Res. 2023, 42, 52. [Google Scholar] [CrossRef] [PubMed]
  93. Fang, X.; Guo, L.; Xing, Z.; Shi, L.; Liang, H.; Li, A.; Kuang, C.; Tao, B.; Yang, Q. IDO1 Can Impair NK Cells Function against Non-Small Cell Lung Cancer by Downregulation of NKG2D Ligand via ADAM10. Pharmacol. Res. 2022, 177, 106132. [Google Scholar] [CrossRef]
  94. Song, H.; Park, H.; Kim, Y.S.; Kim, K.D.; Lee, H.K.; Cho, D.H.; Yang, J.W.; Hur, D.Y. L-Kynurenine-Induced Apoptosis in Human NK Cells Is Mediated by Reactive Oxygen Species. Int. Immunopharmacol. 2011, 11, 932–938. [Google Scholar] [CrossRef]
  95. Terrén, I.; Orrantia, A.; Vitallé, J.; Zenarruzabeitia, O.; Borrego, F. NK Cell Metabolism and Tumor Microenvironment. Front. Immunol. 2019, 10, 483200. [Google Scholar] [CrossRef]
  96. Zheng, X.; Qian, Y.; Fu, B.; Jiao, D.; Jiang, Y.; Chen, P.; Shen, Y.; Zhang, H.; Sun, R.; Tian, Z.; et al. Mitochondrial Fragmentation Limits NK Cell-Based Tumor Immunosurveillance. Nat. Immunol. 2019, 20, 1656–1667. [Google Scholar] [CrossRef]
  97. Lin, J.H.; Huffman, A.P.; Wattenberg, M.M.; Walter, D.M.; Carpenter, E.L.; Feldser, D.M.; Beatty, G.L.; Furth, E.E.; Vonderheide, R.H. Type 1 Conventional Dendritic Cells Are Systemically Dysregulated Early in Pancreatic Carcinogenesis. J. Exp. Med. 2020, 217, e20190673. [Google Scholar] [CrossRef]
  98. Garris, C.S.; Arlauckas, S.P.; Kohler, R.H.; Trefny, M.P.; Garren, S.; Piot, C.; Engblom, C.; Pfirschke, C.; Siwicki, M.; Gungabeesoon, J.; et al. Successful Anti-PD-1 Cancer Immunotherapy Requires T Cell-Dendritic Cell Crosstalk Involving the Cytokines IFN-γ and IL-12. Immunity 2018, 49, 1148–1161.e7. [Google Scholar] [CrossRef]
  99. Alston James, C.; Baer, J.M.; Zou, C.; Panni, U.Y.; Knolhoff, B.L.; Hogg, G.D.; Kingston, N.L.; Kang, L.I.; Lander, V.E.; Luo, J.; et al. Systemic Alterations in Type-2 Conventional Dendritic Cells Leads to Impaired Tumor Immunity in Pancreatic Cancer. Cancer Immunol. Res. 2023, 11, 1055. [Google Scholar] [CrossRef]
  100. Kießler, M.; Plesca, I.; Sommer, U.; Wehner, R.; Wilczkowski, F.; Müller, L.; Tunger, A.; Lai, X.; Rentsch, A.; Peuker, K.; et al. Tumor-Infiltrating Plasmacytoid Dendritic Cells Are Associated with Survival in Human Colon Cancer. J. Immunother. Cancer 2021, 9, e001813. [Google Scholar] [CrossRef]
  101. Tjomsland, V.; Sandström, P.; Spångeus, A.; Messmer, D.; Emilsson, J.; Falkmer, U.; Falkmer, S.; Magnusson, K.E.; Borch, K.; Larsson, M. Pancreatic Adenocarcinoma Exerts Systemic Effects on the Peripheral Blood Myeloid and Plasmacytoid Dendritic Cells: An Indicator of Disease Severity? BMC Cancer 2010, 10, 87. [Google Scholar] [CrossRef] [PubMed]
  102. Boyette, L.B.; MacEdo, C.; Hadi, K.; Elinoff, B.D.; Walters, J.T.; Ramaswami, B.; Chalasani, G.; Taboas, J.M.; Lakkis, F.G.; Metes, D.M. Phenotype, Function, and Differentiation Potential of Human Monocyte Subsets. PLoS ONE 2017, 12, e0176460. [Google Scholar] [CrossRef]
  103. Guo, C.; You, Z.; Shi, H.; Sun, Y.; Du, X.; Palacios, G.; Guy, C.; Yuan, S.; Chapman, N.M.; Lim, S.A.; et al. SLC38A2 and Glutamine Signalling in CDC1s Dictate Anti-Tumour Immunity. Nature 2023, 620, 200–208. [Google Scholar] [CrossRef] [PubMed]
  104. Elomaa, H.; Härkönen, J.; Väyrynen, S.A.; Ahtiainen, M.; Ogino, S.; Nowak, J.A.; Lau, M.C.; Helminen, O.; Wirta, E.V.; Seppälä, T.T.; et al. Quantitative Multiplexed Analysis of Indoleamine 2,3-Dioxygenase (IDO) and Arginase-1 (ARG1) Expression and Myeloid Cell Infiltration in Colorectal Cancer. Mod. Pathol. 2024, 37, 100450. [Google Scholar] [CrossRef] [PubMed]
  105. Sharma, M.D.; Baban, B.; Chandler, P.; Hou, D.Y.; Singh, N.; Yagita, H.; Azuma, M.; Blazar, B.R.; Mellor, A.L.; Munn, D.H. Plasmacytoid Dendritic Cells from Mouse Tumor-Draining Lymph Nodes Directly Activate Mature Tregs via Indoleamine 2,3-Dioxygenase. J. Clin. Investig. 2007, 117, 2570–2582. [Google Scholar] [CrossRef]
  106. Cao, W.; Ramakrishnan, R.; Tuyrin, V.A.; Veglia, F.; Condamine, T.; Amoscato, A.; Mohammadyani, D.; Johnson, J.J.; Min Zhang, L.; Klein-Seetharaman, J.; et al. Oxidized Lipids Block Antigen Cross-Presentation by Dendritic Cells in Cancer Oxidized Lipids and DCs in Cancer. J. Immunol. 2014, 192, 2920. [Google Scholar] [CrossRef]
  107. Herber, D.L.; Cao, W.; Nefedova, Y.; Novitskiy, S.V.; Nagaraj, S.; Tyurin, V.A.; Corzo, A.; Cho, H.I.; Celis, E.; Lennox, B.; et al. Lipid Accumulation and Dendritic Cell Dysfunction in Cancer. Nat. Med. 2010, 16, 880–886. [Google Scholar] [CrossRef]
  108. Wang, X.; Qu, Y.; Ji, J.; Liu, H.; Luo, H.; Li, J.; Han, X. Colorectal Cancer Cells Establish Metabolic Reprogramming with Cancer-Associated Fibroblasts (CAFs) through Lactate Shuttle to Enhance Invasion, Migration, and Angiogenesis. Int. Immunopharmacol. 2024, 143, 113470. [Google Scholar] [CrossRef]
  109. Chang, Y.; Ou, Q.; Zhou, X.; Nie, K.; Zheng, P.; Liu, J.; Chen, L.; Yan, H.; Guo, D.; Zhang, S. Jianpi Jiedu Decoction Suppresses Colorectal Cancer Growth by Inhibiting M2 Polarization of TAMs through the Tryptophan Metabolism-AhR Pathway. Int. Immunopharmacol. 2024, 138, 112610. [Google Scholar] [CrossRef]
  110. Lee, K.J.; Ko, E.J.; Park, Y.Y.; Park, S.S.; Ju, E.J.; Park, J.; Shin, S.H.; Suh, Y.A.; Hong, S.M.; Park, I.J.; et al. A Novel Nanoparticle-Based Theranostic Agent Targeting LRP-1 Enhances the Efficacy of Neoadjuvant Radiotherapy in Colorectal Cancer. Biomaterials 2020, 255, 120151. [Google Scholar] [CrossRef]
  111. Tan, Z.; Xiao, L.; Tang, M.; Bai, F.; Li, J.; Li, L.; Shi, F.; Li, N.; Li, Y.; Du, Q.; et al. Targeting CPT1A-Mediated Fatty Acid Oxidation Sensitizes Nasopharyngeal Carcinoma to Radiation Therapy. Theranostics 2018, 8, 2329–2347. [Google Scholar] [CrossRef] [PubMed]
  112. Hu, A.; Wang, H.; Xu, Q.; Pan, Y.; Jiang, Z.; Li, S.; Qu, Y.; Hu, Y.; Wu, H.; Wang, X. A Novel CPT1A Covalent Inhibitor Modulates Fatty Acid Oxidation and CPT1A-VDAC1 Axis with Therapeutic Potential for Colorectal Cancer. Redox Biol. 2023, 68, 102959. [Google Scholar] [CrossRef] [PubMed]
  113. DePeaux, K.; Delgoffe, G.M. Metabolic Barriers to Cancer Immunotherapy. Nat. Rev. Immunol. 2021, 21, 785–797. [Google Scholar] [CrossRef] [PubMed]
  114. Scharping, N.E.; Rivadeneira, D.B.; Menk, A.V.; Vignali, P.D.A.; Ford, B.R.; Rittenhouse, N.L.; Peralta, R.; Wang, Y.; Wang, Y.; DePeaux, K.; et al. Mitochondrial Stress Induced by Continuous Stimulation under Hypoxia Rapidly Drives T Cell Exhaustion. Nat. Immunol. 2021, 22, 205–215. [Google Scholar] [CrossRef]
  115. Chang, C.H.; Qiu, J.; O’Sullivan, D.; Buck, M.D.; Noguchi, T.; Curtis, J.D.; Chen, Q.; Gindin, M.; Gubin, M.M.; Van Der Windt, G.J.W.; et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef]
  116. 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. [Google Scholar] [CrossRef]
  117. Ning, S.; Hahn, G. Cytotoxicity of Lonidamine Alone and in Combination with Other Drugs against Murine RIF-1 and Human HT1080 Cells in Vitro—PubMed. Cancer Res. 1990, 50, 7867–7870. [Google Scholar]
  118. Fan, Y.; Pedersen, O. Gut Microbiota in Human Metabolic Health and Disease. Nat. Rev. Microbiol. 2020, 19, 55–71. [Google Scholar] [CrossRef]
  119. Wang, L.; Wang, Y.J.; Wang, R.; Gong, F.L.; Shi, Y.H.; Li, S.N.; Chen, P.P.; Yuan, Y.F. Fasting Mimicking Diet Inhibits Tumor-Associated Macrophage Survival and pro-Tumor Function in Hypoxia: Implications for Combination Therapy with Anti-Angiogenic Agent. J. Transl. Med. 2023, 21, 754. [Google Scholar] [CrossRef]
  120. Luo, M.; Wang, Q.; Sun, Y.; Jiang, Y.; Wang, Q.; Gu, Y.; Hu, Z.; Chen, Q.; Xu, J.; Chen, S.; et al. Fasting-Mimicking Diet Remodels Gut Microbiota and Suppresses Colorectal Cancer Progression. Npj Biofilm. Microbiomes 2024, 10, 53. [Google Scholar] [CrossRef]
  121. Rizvanović, N.; Nesek Adam, V.; KalajdŽija, M.; Čaušević, S.; Dervišević, S.; Smajić, J. Effects of Preoperative Oral Carbohydrate Loading on Neutrophil/Lymphocyte Ratio and Postoperative Complications Following Colorectal Cancer Surgery: A Randomized Controlled Study. Eur. Surg. Res. 2023, 64, 278–285. [Google Scholar] [CrossRef] [PubMed]
  122. Yang, Y.; Pei, T.; Liu, C.; Cao, M.; Hu, X.; Yuan, J.; Chen, F.; Guo, B.; Hong, Y.; Liu, J.; et al. Glutamine Metabolic Competition Drives Immunosuppressive Reprogramming of Intratumour GPR109A+myeloid Cells to Promote Liver Cancer Progression. Gut 2024, 74, 255–269. [Google Scholar] [CrossRef] [PubMed]
  123. Izabela, S.; Edyta, R.-B.; Janusz, J.; Mikołaj, P.; Karolina, B.-S.; Janusz, R.; Jan, D.; Maciej, S.; Jarek, B. The Level of Myeloid Derived-Suppressor Cells in Peripheral Blood of Patients with Prostate Cancerafter Various Types of Therapy. Pol. J. Pathol. 2020, 71, 46–54. [Google Scholar]
  124. Fu, C.; Fu, Z.; Jiang, C.; Xia, C.; Zhang, Y.; Gu, X.; Zheng, K.; Zhou, D.; Tang, S.; Lyu, S.; et al. CD205+ Polymorphonuclear Myeloid-Derived Suppressor Cells Suppress Antitumor Immunity by Overexpressing GLUT3. Cancer Sci. 2021, 112, 1011–1025. [Google Scholar] [CrossRef]
  125. Lenart, M.; Rutkowska-Zapała, M.; Siedlar, M. NK-Cell Receptor Modulation in Viral Infections. Clin. Exp. Immunol. 2024, 217, 151–158. [Google Scholar] [CrossRef]
  126. Littwitz-Salomon, E.; Moreira, D.; Frost, J.N.; Choi, C.; Liou, K.T.; Ahern, D.K.; O’Shaughnessy, S.; Wagner, B.; Biron, C.A.; Drakesmith, H.; et al. Metabolic Requirements of NK Cells during the Acute Response against Retroviral Infection. Nat. Commun. 2021, 12, 5376. [Google Scholar] [CrossRef]
  127. Schimmer, S.; Sridhar, V.; Satan, Z.; Grebe, A.; Saad, M.; Wagner, B.; Kahlert, N.; Werner, T.; Richter, D.; Dittmer, U.; et al. Iron Improves the Antiviral Activity of NK Cells. Front. Immunol. 2024, 15, 1526197. [Google Scholar] [CrossRef]
  128. Santosa, E.K.; Kim, H.; Rückert, T.; Le Luduec, J.B.; Abbasi, A.J.; Wingert, C.K.; Peters, L.; Frost, J.N.; Hsu, K.C.; Romagnani, C.; et al. Control of Nutrient Uptake by IRF4 Orchestrates Innate Immune Memory. Nat. Immunol. 2023, 24, 1685–1697. [Google Scholar] [CrossRef]
  129. Raines, L.N.; Zhao, H.; Wang, Y.; Chen, H.Y.; Gallart-Ayala, H.; Hsueh, P.C.; Cao, W.; Koh, Y.; Alamonte-Loya, A.; Liu, P.S.; et al. PERK Is a Critical Metabolic Hub for Immunosuppressive Function in Macrophages. Nat. Immunol. 2022, 23, 431–445. [Google Scholar] [CrossRef]
  130. Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef]
  131. Poznanski, S.M.; Singh, K.; Ritchie, T.M.; Aguiar, J.A.; Fan, I.Y.; Portillo, A.L.; Rojas, E.A.; Vahedi, F.; El-Sayes, A.; Xing, S.; et al. Metabolic Flexibility Determines Human NK Cell Functional Fate in the Tumor Microenvironment. Cell Metab. 2021, 33, 1205–1220.e5. [Google Scholar] [CrossRef]
Figure 1. The impact of macrophage-tumor interactions on macrophage metabolism in gastrointestinal cancers. Tumor cells promote TAMs development via lactate secretion, GLUT3 production, AhR, and PPAR-γ stimulation, resulting in glucose, lipid, and amino acid metabolism, promoting further tumor growth and development. Arrows which are next to the protein names mean up- or downregulation.
Figure 1. The impact of macrophage-tumor interactions on macrophage metabolism in gastrointestinal cancers. Tumor cells promote TAMs development via lactate secretion, GLUT3 production, AhR, and PPAR-γ stimulation, resulting in glucose, lipid, and amino acid metabolism, promoting further tumor growth and development. Arrows which are next to the protein names mean up- or downregulation.
Cancers 17 01467 g001
Figure 2. The impact of neutrophil metabolic shifts in gastrointestinal cancers. TANs suppress immune responses by metabolizing amino acids such as tryptophan and arginine, mainly by ARG1, IDO, and CAT2. They may be disrupting NK cell function through cholesterol metabolism through CD36, LRP1 and LDLR and promoting tumor growth via lactate-driven polarization into the immunosuppressive TAN2 phenotype by EVs. Moreover, fatty acid metabolism may be associated with increased PD-L1 expression and a higher risk of liver metastases. Additionally, glycolysis and NETs formation enhance tumor cell proliferation by increasing mitochondrial activity and ATP production. Arrows which are next to the protein names/processes mean up- or downregulation.
Figure 2. The impact of neutrophil metabolic shifts in gastrointestinal cancers. TANs suppress immune responses by metabolizing amino acids such as tryptophan and arginine, mainly by ARG1, IDO, and CAT2. They may be disrupting NK cell function through cholesterol metabolism through CD36, LRP1 and LDLR and promoting tumor growth via lactate-driven polarization into the immunosuppressive TAN2 phenotype by EVs. Moreover, fatty acid metabolism may be associated with increased PD-L1 expression and a higher risk of liver metastases. Additionally, glycolysis and NETs formation enhance tumor cell proliferation by increasing mitochondrial activity and ATP production. Arrows which are next to the protein names/processes mean up- or downregulation.
Cancers 17 01467 g002
Figure 3. The metabolic adaptations MDSCs of in the tumor microenvironment of gastrointestinal cancers. MDSCs suppress immune responses by depleting key amino acids such as L-arginine and tryptophan, inhibiting T cell proliferation due to express ARG1, IDO, and CAT2. Pathogens such as Helicobacter pylori and Candida tropicalis further influence MDSCs metabolism, increasing glucose uptake and lactate production, which reinforces immunosuppression. Additionally, CD38-mediated NAD⁺ convention into ADPR and cADPR alters energy homeostasis, highlighting metabolism as a key driver of MDSCs function and tumor immune evasion. Arrows which are next to the protein names/processes mean up- or downregulation.
Figure 3. The metabolic adaptations MDSCs of in the tumor microenvironment of gastrointestinal cancers. MDSCs suppress immune responses by depleting key amino acids such as L-arginine and tryptophan, inhibiting T cell proliferation due to express ARG1, IDO, and CAT2. Pathogens such as Helicobacter pylori and Candida tropicalis further influence MDSCs metabolism, increasing glucose uptake and lactate production, which reinforces immunosuppression. Additionally, CD38-mediated NAD⁺ convention into ADPR and cADPR alters energy homeostasis, highlighting metabolism as a key driver of MDSCs function and tumor immune evasion. Arrows which are next to the protein names/processes mean up- or downregulation.
Cancers 17 01467 g003
Figure 4. Metabolic alterations in NK cells mediated by gastrointestinal cancers. Tumor cells induce NK cell functional impairment via the production of HIF-1α, lactate, IDO, and CPT1A induction. Elevated tryptophan metabolism, AhR-independent, causes downregulation of crucial activating NK cell receptor NKG2D and ferroptosis, while CPT1A upregulates fatty acid uptake and metabolism. Arrows which are next to the protein names or processes mean up- or downregulation.
Figure 4. Metabolic alterations in NK cells mediated by gastrointestinal cancers. Tumor cells induce NK cell functional impairment via the production of HIF-1α, lactate, IDO, and CPT1A induction. Elevated tryptophan metabolism, AhR-independent, causes downregulation of crucial activating NK cell receptor NKG2D and ferroptosis, while CPT1A upregulates fatty acid uptake and metabolism. Arrows which are next to the protein names or processes mean up- or downregulation.
Cancers 17 01467 g004
Figure 5. The metabolic changes in DCs in gastrointestinal cancers. The competition for glutamine between DCs and cancer cells may decrease the antitumor feature of cDC1, which enhances CD8+ T cells response. Moreover, excessive IDO expression cause tryptophan metabolism, resulting in an increase in Tregs activity. Additionally, lipid accumulation significantly affects the function of DCs and their ability to present tumor antigens. cDC2 shows metabolic reprogramming, characterized by increased glycolysis and fatty acid metabolism, as well as the production of reactive oxygen species, which is associated with impaired antitumor immunity. Arrows which are next to the protein names or processes mean up- or downregulation.
Figure 5. The metabolic changes in DCs in gastrointestinal cancers. The competition for glutamine between DCs and cancer cells may decrease the antitumor feature of cDC1, which enhances CD8+ T cells response. Moreover, excessive IDO expression cause tryptophan metabolism, resulting in an increase in Tregs activity. Additionally, lipid accumulation significantly affects the function of DCs and their ability to present tumor antigens. cDC2 shows metabolic reprogramming, characterized by increased glycolysis and fatty acid metabolism, as well as the production of reactive oxygen species, which is associated with impaired antitumor immunity. Arrows which are next to the protein names or processes mean up- or downregulation.
Cancers 17 01467 g005
Table 1. Potential immunometabolism-associated targets and therapeutics for gastrointestinal cancer treatment.
Table 1. Potential immunometabolism-associated targets and therapeutics for gastrointestinal cancer treatment.
CellsPotential TargetPotential or Approved TherapeuticLiterature
MacrophagesABHD5/SRM/spermidine axis-[31]
IL-8/STAT3/GLUT3 signaling axisreparixin[18]
RARγ-dependant TRAF6-IL-6-STAT3 signalingnordihydroguaiaretic acid (NDGA)[24]
lactate transport function of MCT1AZD3965, an MCT1 (monocarboxylate transporter 1) inhibitor[108]
AhR expression and M2-type polarizationJianpiJiedu decoction (traditional Chinese medicine)[109]
NeutrophilsLRP1LRP1 inhibitor[53,110]
MDSCsCD38anti-CD38 monoclonal antibody (Daratumumab) [77]
activation of 5′AMP-activated protein kinase (AMPK) MDSCsmetformin[78]
Dendritic cellsinhibitor of acetylCoA carboxylase5(tetradecycloxy)2furoic acid (TOFA) in the research phase[107]
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

Siemińska, I.; Lenart, M. Immunometabolism of Innate Immune Cells in Gastrointestinal Cancer. Cancers 2025, 17, 1467. https://doi.org/10.3390/cancers17091467

AMA Style

Siemińska I, Lenart M. Immunometabolism of Innate Immune Cells in Gastrointestinal Cancer. Cancers. 2025; 17(9):1467. https://doi.org/10.3390/cancers17091467

Chicago/Turabian Style

Siemińska, Izabela, and Marzena Lenart. 2025. "Immunometabolism of Innate Immune Cells in Gastrointestinal Cancer" Cancers 17, no. 9: 1467. https://doi.org/10.3390/cancers17091467

APA Style

Siemińska, I., & Lenart, M. (2025). Immunometabolism of Innate Immune Cells in Gastrointestinal Cancer. Cancers, 17(9), 1467. https://doi.org/10.3390/cancers17091467

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