Skip to main content
Cancer Medicine logoLink to Cancer Medicine
. 2023 Oct 20;12(21):20573–20589. doi: 10.1002/cam4.6623

Metabolic reprogramming, autophagy, and ferroptosis: Novel arsenals to overcome immunotherapy resistance in gastrointestinal cancer

Xiangwen Wang 1, Liwen Zhou 2, Hongpeng Wang 1, Wei Chen 1, Lei Jiang 1, Guangtao Ming 1, Jun Wang 1,
PMCID: PMC10660574  PMID: 37860928

Abstract

Background

Gastrointestinal cancer poses a serious health threat owing to its high morbidity and mortality. Although immune checkpoint blockade (ICB) therapies have achieved meaningful success in most solid tumors, the improvement in survival in gastrointestinal cancers is modest, owing to sparse immune response and widespread resistance. Metabolic reprogramming, autophagy, and ferroptosis are key regulators of tumor progression.

Methods

A literature review was conducted to investigate the role of the metabolic reprogramming, autophagy, and ferroptosis in immunotherapy resistance of gastrointestinal cancer.

Results

Metabolic reprogramming, autophagy, and ferroptosis play pivotal roles in regulating the survival, differentiation, and function of immune cells within the tumor microenvironment. These processes redefine the nutrient allocation blueprint between cancer cells and immune cells, facilitating tumor immune evasion, which critically impacts the therapeutic efficacy of immunotherapy for gastrointestinal cancers. Additionally, there exists profound crosstalk among metabolic reprogramming, autophagy, and ferroptosis. These interactions are paramount in anti‐tumor immunity, further promoting the formation of an immunosuppressive microenvironment and resistance to immunotherapy.

Conclusions

Consequently, it is imperative to conduct comprehensive research on the roles of metabolic reprogramming, autophagy, and ferroptosis in the resistance of gastrointestinal tumor immunotherapy. This understanding will illuminate the clinical potential of targeting these pathways and their regulatory mechanisms to overcome immunotherapy resistance in gastrointestinal cancers.

Keywords: autophagy, ferroptosis, gastrointestinal cancer, immunotherapy resistance, metabolic reprogramming

1. INTRODUCTION

Gastrointestinal cancer is a major public health problem worldwide, with 26% and 35% of the total global cancer incidence and mortality, respectively. 1 Although conventional treatments such as radiotherapy, chemotherapy, and surgery have been widely applied in treating gastrointestinal cancers, these strategies have modestly improved survival, especially in patients with advanced disease. 2 In the past few years, the advent of immunotherapy has significantly changed the landscape of solid tumor treatment, and immune checkpoint blockade (ICB) therapies (e.g., anti‐PD‐1, anti‐PD‐L1, and anti‐CTLA‐4) have achieved breakthrough results in many tumors, such as melanoma and non‐small cell lung cancer, which caused a paradigm shift of cancer treatment. 3 , 4 The US Food and Drug Administration (FDA) has approved PD‐1 inhibitors (e.g., pembrolizumab) for gastrointestinal cancers, including advanced esophageal, gastric, colorectal, hepatic, and pancreatic cancers. 5 However, the efficacy of current ICB therapies remains a clinical challenge for gastrointestinal cancers owing to the insufficient response to ICB‐based immunotherapy. 6 To date, ICB therapies are appropriate for gastrointestinal cancers of rare subtypes, such as those having microsatellite instability high (MSI‐H) and DNA mismatch repair deficiency (dMMR), and these molecular subtypes occur in only 5%–10% of all gastrointestinal cancers, with benefits for only a small subset of patients. 7 Recent clinical trials have revealed that the efficacy of ICB therapies is limited in patients with microsatellite stability, particularly in colorectal and pancreatic cancers. 8 , 9 , 10 Moreover, approximately 50% of cancers with the MSI‐H subtype are intrinsically resistant to anti‐PD‐1 therapies. 11 Therefore, the small applicability and widespread resistance to ICB therapies drastically limit the efficacy of immunotherapy in gastrointestinal cancers.

Accumulated evidence has revealed the tumor‐intrinsic and tumor‐extrinsic mechanisms of resistance to immunotherapy. Intrinsic factors are characteristic of genetic T‐cell exclusion, absence of antigenic proteins, and antigen presentation, whereas extrinsic factors are immunosuppressive cells, inhibitory immune checkpoints, and absence of T cells, 12 , 13 which implies that finding ways to modulate both intrinsic and extrinsic factors is significant for overcoming immunotherapy resistance in gastrointestinal cancers. Metabolic reprogramming, autophagy, and ferroptosis have been demonstrated as critical factors in tumorigenesis and progression of gastrointestinal cancers, 14 , 15 , 16 and are involved in the regulation of tumor trafficking, death, differentiation, activation, and efficacy of immune cell subsets. 17 , 18 , 19 Moreover, metabolic reprogramming can support the unrestrained proliferative and malignant phenotype of cancer cells, while autophagy and ferroptosis can alter the death modalities of tumor and immune cells, enhancing their malignancy. These three pathways play crucial roles in the malignancy of cancer cells, immune cell activity, and the establishment of an immunosuppressive microenvironment. 17 , 18 , 19 Targeting these pathways presents a promising strategy for enhancing the regulation of antitumor immune responses and overcoming resistance to immunotherapy.

There are close interactions between metabolic remodeling, autophagy, and ferroptosis, understanding the crosstalk among these processes is valuable for developing new strategies for gastrointestinal immunotherapy resistance.

In this review, we discuss the effects of metabolic remodeling, autophagy, and ferroptosis on antitumor immune responses in gastrointestinal cancers, the roles of their interactions in immunotherapy, and the potential of these targets in immunotherapy. Improving our understanding of the regulation of these factors and antitumor immunity responses will help develop combination therapies to overcome resistance to gastrointestinal cancer immunotherapy.

2. METABOLIC REPROGRAMMING, AUTOPHAGY, AND FERROPTOSIS IN ANTITUMOR IMMUNITY

2.1. Metabolic reprogramming in cancer and immune cells

Metabolic reprogramming, a hallmark of cancer, supports unrestricted proliferation, division, and metastasis of tumor cells by rewiring many metabolic pathways, such as glucose, amino acids, and lipids. 20 Previously excellent reviews have also illustrated that metabolic rewiring promotes tumorigenesis, progression, metastasis, and treatment resistance in gastrointestinal cancers. 21 , 22 The metabolic pattern of tumor cells is complex, with obvious heterogeneity, and different metabolic adaptation phenotypes appear owing to changes in the external environment. For example, Metastasis associated in colon cancer 1 (MACC1) is significantly upregulated to facilitate the Warburg effect and ensure gastric cancer growth in glucose deprivation‐induced metabolic stress. 23 Under nutrient‐deprived conditions, hepatic cancer cells activate the serine biosynthesis pathway to promote cancer progression by upregulating oncogene cMyc. 24 In addition, this complex and variable metabolic pattern also exists in immune cells, which determines their differentiation and function. 25 For T‐cell subsets, effector T cells prefer the glycolytic pathway for effect killing function, 26 whereas other T cells (naive, Treg, memory) usually use fatty acid oxidation (FAO) and oxidative phosphorylation (OXPHOS) pathways to maintain their survival. 27 Moreover, neutrophils, natural killer (NK) cells, B cells, M1 macrophages, and active dendritic cells (DCs) use the glycolytic pathway as energy supply, whereas M2 macrophages and resting DCs rely on the OXPHOS pathway. 28 , 29 , 30 , 31 Both tumor and immune cells need to obtain sufficient energy to survive or function through metabolic rewiring, especially glycolysis, implying that competition for nutrients between tumor and immune cells promotes immunosuppressive phenotypes.

2.2. Glycolysis

Aberrant glucose metabolism is one of the main causes of ineffective antitumor immunity (Figure 1). Chang et al. 32 revealed that competitive tumor glucose consumption limits the functions of CD8+ T effector cells, which leads to the inefficiency of interferon‐gamma (IFNγ) production and failure of tumor clearance. Furthermore, hypoxia‐mediated metabolic rewiring upregulates the glucose metabolism of pancreatic cancer, and glucose consumption inhibits the activity of antitumor T cells. 33 Additionally, glucose consumption induced by accelerated glycolysis in cancer cells further inhibits effector T‐cell function by inducing Forkhead box protein 3 (FOXP3) expression and promoting infiltration of myeloid‐derived suppressor cells (MDSCs). 34 , 35 Thus, targeting gastrointestinal cancer‐specific energy metabolism to suppress the nutrient deprivation of immune cells by tumors is a promising approach to improve the efficacy of immunotherapy.

FIGURE 1.

FIGURE 1

Impact of metabolic reprogramming on antitumor immunity. Tumor metabolic reprogramming, through nutrient competition involving glucose, glutamine, lipids, and other substances, coupled with the export of metabolic byproducts like lactate into the microenvironment, fosters an immunosuppressive tumor microenvironment (TME). This landscape is characterized by diminished effector immune cell activity, reduced antigen presentation, and an increased presence of immunosuppressive cells and immune checkpoints.

Glucose transporter proteins (GLUT) are responsible for glucose transport and uptake. As a member of the GLUT family, GLUT‐1 is overexpressed in pancreatic cancer and correlates with a higher density of PD‐1+ T cells. 36 GLUT3 is overexpressed in gastrointestinal cancers 37 and contributes to the T helper 17 cell responses to M2 macrophage subtype transition. 38 , 39 Hexokinase (HK), phosphofructokinase (PFK), and pyruvate kinases (PKs) are rate‐limiting enzymes of the glycolysis pathway and are closely associated with the progression of gastrointestinal cancers. 40 The overexpression of HK2 is positively correlated with immune infiltration in esophageal cancer; this downregulates the human leukocyte antigen (HLA) of hepatic cancer and impairs the sensitivity of NK cells and innate immune responses. 41 , 42 , 43 The enzyme fructose‐2,6‐bisphosphatase‐3 (PFKFB‐3) plays an essential role in the progression and resistance to sorafenib and upregulates PD‐L1 expression of macrophages to confer immune tolerance in hepatic cancer. 44 , 45 Pyruvate kinase isoform M2 (PKM2), the last rate‐limiting enzyme and driver of gastrointestinal cancer, promotes hepatic cancer progression by inducing an immunosuppressive microenvironment, 46 and the inhibition of PKM2 suppresses PD‐L1 expression on tumor cells. 47 Moreover, an increase in glycolysis accelerates the production of lactate and decreases OXPHOS. Lactate is then transported into the extracellular environment by monocarboxylate transporter 4 (MCT4), 47 resulting in an acidic tumor microenvironment (TME) and PD‐1‐overexpressing Treg, which inhibits antitumor immunity and immunosurveillance by T and NK Cells. 48 In addition, many proteins, long noncoding RNAs (lncRNA), and microRNAs were identified as regulators of glycolysis that modulate antitumor immune response in gastrointestinal cancers. The characteristics of these targets are summarized in Table 1.

TABLE 1.

Characteristics of molecules that regulate antitumor immune response by modulating metabolism, autophagy and ferroptosis in gastrointestinal cancers.

Metabolic pathways Cancer type Phenotype Target References
Glycolysis GC Activity of γδT cells, macrophages/CD8+ T cells exhaustion H19/CXCL1 49, 50
CRC T‐cell proliferation and activation/CD8+ T‐cell effector function ZFP91/NIK 51, 52
Treg identity and function MondoA 53
HCC Improvement of anti‐PD‐1 therapy CircRHBDD1 54
Upregulation of PD‐L1 miR‐675‐5p 43
M2 macrophage differentiation/MDSCs recruitment ECT2/PIWIL1 55, 56
PC Function of CD4+ T cells DKK3 58
Immunosuppression in TME Bmi1‐UPF1‐HK2 axis/BHLHE40 59, 60
Glutamine metabolism GC Immune infiltrates in TME SLC1A5 61
HCC Immune Infiltration defect SLC38A1 62
PD‐L1+ exosomes activity HMGB1/RICTOR 46
Lipid metabolism ESC Macrophage alternative activation PPARγ 63
CRC Regulation of IL‐10‐producing regulatory B cells LXA4 64
Regulation of IFN‐γ FABP1 65
Regulation of PD‐L1 USP19 66
HCC Regulation of IFN‐γ SREBF1/miR‐27a 57, 67
Regulation of IFNGR1 and MHC‐I expression Optineurin 68
MDSCs recruitment CCRK 69
Regulation of Treg cells/HLA‐C and B2M expression MARC2 70
CD8+ T‐cell proliferation/T‐cell infiltration and activation CD137/ARF1 71, 72
PC T‐cell survival and persistence ACADVL 73
Autophagy GC Tregs infiltration BECLIN‐1 74
CRC Function of DCs HMGB1 75
Tregs infiltration SQSTM1 76
Regulation of IFN‐I ATG16L1 76
HCC CD8+ T‐cell suppression GOLM1 77
Induction of acidic/immunostimulatory TME ASIC1a/LIF 78, 79
PC MHC‐I expression NBR1 80, 81
CD8+ T‐cell infiltration and effector function RNF31 82
PD‐L1 expression/M2 macrophage differentiation SEMA3C 83
Ferroptosis ESC CD8+ T Cells apoptosis LncRNA‐OIP5‐AS1 84
GC Regulation of T/B cell reporter/immune checkpoints SLC2A3 85
CRC Immune infiltration in TME SLC2A1/MT1G 86, 87
Leukocytes recruitment, T‐cell accumulation OTUD1 88
HCC Immune infiltration in TME BTBD10/HSPB1 89, 90
PC Immune infiltration in TME, tumor mutation burden Linc02432, Hsa‐miR‐98‐5p, HK2 91

Abbreviations: CRC, colorectal cancer; ESC, esophageal cancer; GC, gastric cancer; HCC, hepatocellular carcinoma; PC, pancreatic cancer.

2.3. Glutamine metabolism

Another striking feature of cancer metabolic reprogramming is glutamine addiction. Glutamine is essential for redox balance, de novo synthesis of nucleotides, efficient DNA replication, and cell proliferation in cancer, 61 and increased glutamine consumption can exhaust nutrients within the tumor, leading to metabolic stress that may affect the progression of gastrointestinal cancer. 92 , 93 In addition, immune cells require glutamine uptake for survival, activation, and differentiation. Glutamine can promote the transcription of proliferation‐related genes by activating the ERK and JNK signaling pathways, resulting in the rapid proliferation of immune cells. 94 Glutamine metabolism is also essential to regulate the activation of macrophages and facilitate the differentiation of M2 macrophages. 94 In TME, tumor and immune cells also compete for glutamine uptake, similar to the competition between glucose. Selective inhibition of glutamine metabolism in tumor cells can increase glutamine uptake of T cells and enhance their antitumor activity, 95 and the activation of the MAPK/ERK pathway modulates glutamine uptake between tumor and T cells. 96 A recent study reveals that glutamine allocation in TME is programmed, with cell‐intrinsic programs driving tumor cells to preferentially consume glutamine, whereas immune cells consume glucose. 97 This nutrient distribution strategy is induced by mTORC1 signaling‐mediated expression of glucose‐ and glutamine‐related genes. Thus, these cell‐intrinsic programs may be potential clinical translation targets to overcome immunotherapy resistance in gastrointestinal cancers.

In addition to glutamine status in the TME, glutamine metabolic reprogramming in tumor cells also significantly affects antitumor immune responses. Glutamine deprivation in tumor cells activates EGFR/ERK/C‐JUN signaling and then upregulates the expression of PD‐L1 in tumor cells, which weakens the antitumor immune response. 98 Glutamine metabolic reprogramming promotes tumorigenesis of hepatic cancer, upregulates the expression of lymphocyte activation gene 3 protein (LAG3) in infiltrating γδ T cells, 99 and impairs immunotherapy by PD‐L1. 98 Moreover, in a mouse model of colon cancer, glutamine blockade decreases hypoxia, acidosis, and nutrient depletion, and destroys immunosuppressive TME to overcome tumor immune evasion. 99 Sharma et al. 100 have shown that targeting glutamine‐utilizing enzymes using a glutamine analog (6‐diazo‐5‐oxo‐l‐norleucine) can remodel the extracellular matrix (ECM) and elevate infiltration of CD8+ T cells, leading to sensitization of pancreatic cancer to anti‐PD1 therapy. Glutamine transporter SLC1A5 is also positively associated with immune infiltrates in gastric cancer, 101 and inhibition of SLC1A5 improves antitumor immunity in TME. 102 Therefore, targeting cancer‐specific glutamine metabolism is a promising strategy to improve the efficacy of immunotherapy. Other molecules involved in the antitumor immune response by modulating glutamine metabolic reprogramming in gastrointestinal cancers are shown in Table 1.

2.4. Lipid metabolism

Tumor cells usually exhibit a high affinity for fatty acids and cholesterol to support their growth, and many lipogenic enzymes, such as fatty acid synthase (FASN), ATP‐citrate lyase (ACLY), and acetyl‐CoA carboxylase (ACC), are overexpressed in gastrointestinal cancers. 103 In contrast, fatty acids and lipid accumulation result in general immunosuppression in immune cells. For example, high amounts of lipids stimulate M2 macrophages, leading to protumorigenic polarization in gastric cancer, 104 long‐chain fatty acid metabolism reinforces the suppression by tumor‐associated macrophages of tumor immune surveillance. 105 Moreover, lipid accumulation in DCs inhibits its capacity to process antigens. 106 Fatty acids are essential for the differentiation of Treg cells, and the overexpression of lipid transporter CD36 contributes to the survival and functions of Treg cells. 107 The accumulation of long‐chain fatty acids also damages CD8+ T‐cell functions in pancreatic cancer. 73 Moreover, pharmacological inhibition of FAS using an ACC inhibitor reduces lipid levels in DCs and rescues their antitumor immunity activity, 106 and inhibition of FASN protects CD4+ effector T cells from restimulation‐induced cell death and enhances T‐cell immunity. 108 FAO blockade also limits the immunosuppressive effect of M2 macrophages and Tregs. 109 , 110

The metabolism of cholesterol represents a pivotal pathway in modulating antitumor immunity. High levels of cholesterol induce overexpression of PD‐1, LAG‐3, and TIM3 in immune cells, which perturbs its normal lipid metabolism, promotes its apoptosis, and reduces its proliferation. 111 , 112 Free cholesterol in the T‐cell membrane is an integral component of the T‐cell receptor (TCR) and the immunological synapse of T cells, directly orchestrating signaling pathways and effector functions. 113 Gu et al. 114 found that cholesterol esterase acetyl‐CoA acetyltransferase 1 (ACAT1), an oncogene, promotes hepatocarcinogenesis by remodeling lipid metabolism, and pharmacological inhibition using avasimibe could have an antitumor effect on CD8+ T cells and improve the efficacy of PD‐1 inhibitors by regulating cholesterol metabolism. 115 Within the tumor microenvironment, lactate suppresses the expression of PPARγ in intratumoral iNKT cells, thereby diminishing their cholesterol synthesis and production of IFN‐γ, attenuating the antitumor efficacy of iNKT cells in hepatocellular carcinoma. 57 Conversely, ATP‐binding cassette transporter A9 promotes cholesterol accumulation in macrophages, amplifying antitumor immunity against peritoneal metastasis in colorectal cancer. 116 Elevated serum cholesterol levels enhance the antitumor capabilities of natural killer cells and decelerate the growth of murine liver tumors. 117 Absence of Apolipoprotein E (ApoE) results in a reduced proportion of MDSCs in the body, leading to accelerated therapeutic growth. 118 Notably, both an increase or decrease in cholesterol can inhibit T‐cell function. 113 These findings underscore a dual role of cholesterol metabolism in resistance to immunotherapy for gastrointestinal tumors, suggesting that targeting cholesterol metabolic imbalances might be a potential avenue to rejuvenate antitumor immune efficacy.

Moreover, lipid metabolism‐related genes are also closely associated with immune infiltration. The modulation of lipid metabolism through ADP ribosylation factor 1 (ARF1) curtails the release of damage‐associated molecular patterns (DAMPs), which compromise antitumor immune surveillance by suppressing T‐cell infiltration and activation in CRC. 119 Gu et al. 114 and Hu et al. 120 have established lipid metabolism‐based prediction models that characterize the relationship between lipid metabolism‐related genes, immune infiltration, and prognosis in colon cancer and hepatocellular carcinoma. We generalized the mechanisms by which lipid metabolism regulates antitumor immunity and summarized the molecules that enhance or weaken antitumor immunity by regulating lipid metabolism (Table 1). Together, this complex fatty acid and cholesterol metabolism offers an opportunity to modulate the immunosuppressive TME and enhance antitumor immunity.

2.5. Autophagy

Autophagy is a highly conserved multistep catabolic pathway dominated by macroautophagy (hereafter referred to as autophagy) to maintain metabolic adaptation, which is dependent on intracellular lysosomes and is regulated by the ATG family of proteins. Autophagy, as a “double‐edged sword,” plays a complex role in gastrointestinal tumor biology. For instance, autophagy as a suppressor delays cancer cell transformation during pancreatic and liver tumorigenesis, whereas in transformed cancer cells, it promotes the progression, treatment, and treatment resistance of hepatic and pancreatic cancer. 121 , 122 , 123 In addition, autophagy regulates the activity of immune cells and stromal cells in the TME and exacerbates immune evasion and tolerance of tumor cells (Figure 2). Therefore, autophagy plays an important role in regulating resistance to immunotherapy.

FIGURE 2.

FIGURE 2

Impact of autophagy on antitumor immunity. Autophagy modulates the functions of naive T cells, T cells, dendritic cells (DC), natural killer (NK) cells, myeloid‐derived suppressor cells (MDSC), and regulatory T cells (Treg). Additionally, it promotes the secretion of cytokines by stromal cells and facilitates MHC‐I degradation and PD‐L1 downregulation in tumor cells, culminating in immune evasion and immune tolerance.

2.6. Autophagy in the TME

Autophagy can promote or inhibit tumor progression by regulating the survival, activation, proliferation, and differentiation of immune cells in the TME. Xia et al. 124 have reported that autophagy deficiency induces naive T‐cell apoptosis and inhibits tumor immunity, and autophagy‐related protein phosphatidylinositol 3‐kinase catalytic subunit type 3 (PIK3C3) deficiency impairs T‐cell mitochondrial metabolism, resulting in the failure of CD4+ T cells to differentiate into effector T cells. 125 Moreover, defective proliferation is observed in ATG3/ATG5/PIK3C3‐deficient T cells by facilitating the accumulation of cell‐cycle inhibitor cyclin‐dependent kinase inhibitor 1B (CDKN1B), 126 and Autophagy‐related 5/7 (ATG5/ATG7) deficiency causes functional deficit of memory CD8+ T cells. 127 Autophagy also plays a critical role in the antigen processing and presentation functions of DCs. ATG5 deficiency in DCs upregulates the scavenger receptor CD36 and positively regulates major histocompatibility complex class II (MHC‐II) antigen presentation, 128 and autophagy‐related protein Vps34 is essential for functions of antigen cross‐presenting CD8α+ DCs. 129 As a major innate effector component, NK cells exert powerful antitumor immunity. Phosphorylated Forkhead box O1 (FOXO1) interacts with ATG7 and induces autophagy in immature NK cells, and this FOXO1‐mediated autophagy is essential for NC cell‐induced antitumor immunity. 130 As professional antigen‐presenting cells, B cells require autophagy to regulate antigen presentation, cross‐presentation, and memory maintenance functions. 131 The inhibition of autophagy promotes the polarization of hepatoma‐related M2 macrophages and elevates antitumor phagocytosis and cytotoxicity of macrophages. 132 Moreover, autophagy is essential to regulate the survival and function of immunosuppressive Tregs and MDSCs. For example, the inhibition of autophagy promotes the activation of STAT3 signaling, resulting in the accumulation and immunosuppressive function of MDSCs. 133 Autophagy also enhances the functional integrity of Tregs by maintaining homeostasis between environmental stress and metabolic adaptations. 134 In addition, stromal cells in the TME secrete specific cytokines through autophagy to promote autophagy of tumor cells and accelerate tumor progression. Li et al. 135 revealed that hypoxia‐induced autophagy of stellate cells reduces stromal lumican in the TME of pancreatic cancer, thereby promoting cancer progression. Wang et al. 136 also found that cancer‐associated fibroblasts promote the survival of irradiated cancer cells by secretory factor‐induced autophagy. Furthermore, an intricate dance exists between DAMPs and autophagy. By thwarting autophagy utilizing hydroxychloroquine, the vigor of DAMPs is heightened, galvanizing dendritic cell activation. In a counter‐play, DAMPs might spur autophagy, thus advancing tumor malignancy. 137 Therefore, targeting autophagy in the TME may be a novel approach to circumvent TME‐mediated immunotherapy resistance.

2.7. Autophagy in tumor cells

In addition to autophagy in the TME, autophagy in tumor cells can also affect antitumor immunity. Altered expression of MHC‐I in tumor cells usually helps them evade immune surveillance, which causes immunotherapy resistance. Among gastrointestinal tumors, pancreatic cancer is the least sensitive to immunotherapy. Recent studies have illustrated that autophagy facilitates immune evasion of pancreatic cancer by lysosomal‐mediated degradation of MHC‐I, and inhibition of autophagy using chloroquine rescues the expression of MHC‐I, improves antigen presentation, and synergizes with immunotherapy (anti‐PD1 or anti‐CTLA4 antibodies). 80 , 81 Pharmacological inhibition of MEK and autophagy activate STING/TNF‐1 signaling and paracrine signaling, in turn promoting tumor‐associated macrophages toward an immunogenic M1‐like phenotype, leading to the activation of immune recognition in pancreatic cancer. 138 Moreover, during chemotherapy‐induced immunogenic cell death in colon cancer cells, ATG5/ATG7 deficiency impairs antitumor immunity by reducing the release of adenosine triphosphate. 139 Autophagy has also been shown to regulate the expression of immune checkpoints in tumor cells to promote immune tolerance. For example, the inhibition of autophagy upregulates the expression of PD‐L1 in gastric cancer cells and enhances the efficacy of PD‐L1 blockade. 140 Thus, autophagy plays critical roles in the regulation of the antitumor immune response by modulating MHC‐I‐antigen complexes, immunogenic tumor cell death, and expression of immune checkpoints in gastrointestinal cancers. In addition, autophagy has a twofold function in both the initial emergence and advanced stages of cancer. In the early stages, autophagy suppresses the process of carcinogenesis by degrading toxic substances within cells and by regulating intercellular communication mediated by proteins and hormones. However, in advanced stages, autophagy promotes cancer progression by recycling cellular components to provide metabolic substrates, facilitating the metabolic reprogramming of cancer cells. 141 We summarize the characteristics of molecules that regulate autophagy and antitumor immunity in Table 1.

2.8. Ferroptosis

Ferroptosis is a form of programmed cell death characterized by lethal levels of iron‐metabolism‐dependent accumulation of lipid reactive oxygen species (ROS). Recently, numerous studies have shown that ferroptosis is closely related to the antitumor immune regulation of gastrointestinal cancers. A predictive model of immune response in hepatocellular, pancreatic, colon, and gastric cancers has been constructed based on ferroptosis‐related LncRNAs. 142 , 143 , 144 , 145 Ferroptosis act as a key regulator of survival and activation of immune cells in the TME. The selenoenzyme glutathione peroxidase (GPX4) is a central repressor of ferroptosis, and overexpression of GPX4 renders CD8+ T cells ferroptosis‐resistant and protects against death induced by excess lipid accumulation. The inhibition of ferroptosis in CD8+ T cells effectively restores their antitumor activity. 146 , 147 Hou et al. 84 reported that GPX4 reduces the proportion of CD8+ T cells and promotes immune evasion of esophageal cancer cells by alleviating ferroptosis. Moreover, immunosuppressive MDSCs and Tregs both exhibit high ferroptosis resistance that reduces antitumor immunity. 148 Wang et al. 149 first explicitly reported the phenomenon of crosstalk between the ferroptosis pathway of immune cells and tumor cells; immune‐activated CD8+ T cells downregulate the expression of glutamate‐cystine anti‐transport system proteins (SLC3A2 and SLC7A11) by releasing IFN‐γ, inhibiting the uptake of cystine by tumor cells, and promoting tumor cell lipid peroxidation and ferroptosis, resulting in enhanced antitumor immunity. The induction of ferroptosis in tumor cells also increases their immunogenicity and improves the efficacy of anti‐PD‐1 therapy. 150 Zhang et al. 151 found that dihydroartemisinin induces ferroptosis in pancreatic cancer and contributes to an increase in the population of CD8+T, NK, and NKT cells. Additionally, early cellular ferroptosis induces immunogenic cell death, and concurrently, the release of DAMPs leads to the maturation of DC. 137 Therefore, the induction of ferroptosis enhances the killing effect of immune cells on tumor cells and inhibits the survival of immunosuppressive cells while reducing the antitumor activity of effector T cells (Figure 3). However, tumor cells are more sensitive to ferroptosis than effector T cells, 148 suggesting that this approach is a promising antitumor strategy. The features of molecules that regulate ferroptosis and modulate antitumor immunity are shown in Table 1.

FIGURE 3.

FIGURE 3

Impact of ferroptosis on antitumor immunity. Activated CD8+ T cells can downregulate the expression of SLC3A2 and SLC7A11 via the release of IFN‐γ, inhibiting the uptake of cystine by tumor cells. This promotes ferroptosis in tumor cells, enhancing their immunogenicity. CPX4 fosters immune evasion by attenuating the sensitivity to ferroptosis.

3. CROSSTALK BETWEEN METABOLIC REPROGRAMMING, AUTOPHAGY, AND FERROPTOSIS

Metabolic reprogramming, autophagy, and ferroptosis play critical roles in the regulation of cancer progression and antitumor immunity, and crosstalk exists between these three processes in antitumor immunity. Autophagy recycles intracellular components under metabolic stress and regulates the energy metabolism of immune cells in the TME. Recent results suggest that induction of ferroptosis is dependent on autophagy and metabolic reprogramming. 152 , 153 Therefore, an in‐depth understanding of the crosstalk among the three is conducive to the discovery of new antitumor immunotherapy strategies.

3.1. Autophagy and metabolic reprogramming

The main role of autophagy is to provide sufficient substrate for anabolism. Autophagy‐deficient tumor cells are incapable of macromolecular degradation and Krebs cycle substrate support, which prevents aspartate production and nucleotide synthesis. 141 Autophagy also promotes mitochondrial metabolism and removes mitochondria with excessive levels of oxidative stress, and this crosstalk is convenient for tumor growth. 154 Autophagy is activated in the presence of hypoxia and nutrient deprivation in the TME. 155 Under hypoxia, tumor cells evade immune surveillance by T cells by inducing autophagy through HIF‐1α/STAT3 signaling, 156 and hypoxia‐induced autophagy reduces NK cell killing by degrading the NK‐derived serine protease granzyme B (GZMB) during transport within cancer cells. 157 Moreover, autophagy of cancer‐associated fibroblasts (CAFs) directly supplies lactate, ketone bodies, fatty acids, and glutamine for cancer cell proliferation in the TME. 158 In addition, the phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumor suppressor, suppresses glycolysis, the pentose phosphate pathway, lipid synthesis, and pyrimidine synthesis by blocking AKT and mTOR signaling. 159 PTEN also induces autophagy by inhibiting mTOR signaling from regulating epithelial to mesenchymal transition (EMT) and invasion of cancer cells. 159 As a common switch for autophagy and metabolic reprogramming, PTEN achieves tumor suppression and immune protection by blocking mTOR signaling, inducing T‐cell infiltration, inhibiting M2 macrophage polarization, and decreasing PD‐L1 expression in the TME, 159 , 160 , 161 suggesting that PTEN is a promising therapeutic target to enhance antitumor immunity.

3.2. Metabolic reprogramming and ferroptosis

A recent study has demonstrated a dynamic interaction between ferroptosis and glycolysis, lipid metabolism, and amino acid metabolism in the TME, which affects the efficacy of immunotherapy. 162 Increased glycolysis in tumor cells and CAFs produces excess lactate, which creates an acidic TME, leading to immune escape, tumor metastasis, and therapy resistance. Pucino et al. 163 reported that lactate accumulation regulates ferroptosis by modulating the production of lipids. Inhibition of hydroxycarboxylic acid receptor 1 (HCAR1) or monocarboxylate transporter 1 (MCT1) blocks lactate uptake and reduces ATP production in hepatocellular carcinoma cells, resulting in the activation of AMPK signaling and ferroptosis. 164 This suggests that this crosstalk between lactate and ferroptosis reshapes the TME and may lead to immunosuppressive and immune evasion phenotypes. Lipid metabolism is closely related to the sensitivity of cells to ferroptosis and plays an important role in antitumor immune regulation. Cancer cells in ferroptosis regulate the activity of immune cells in the TME by releasing oxidized lipid metabolites such as prostaglandin E2 (PGE2). 165 As an immunosuppressive factor, PGE2 inhibits the antitumor effect of NK cells, DCs, and CD8+ T cells in the TME. 166 In addition, amino acid metabolism is an important step in regulating ferroptosis; it enhances the antioxidant capacity of cancer cells by providing cysteine, cystine, and glycine to combat ferroptosis. Cystine deficiency impairs mitochondrial OXPHOS by inhibiting cytosolic aspartate aminotransaminase (GOT1), inducing ferroptosis‐mediated cell death in pancreatic cancer. 167 The cystine‐glutamate antiporter xCT protein SLC7a11 regulates cellular sensitivity to ferroptosis by regulating glutathione synthesis, and inhibition of SLC7a11 improves the efficacy of anti‐CTLA‐4 in the colon and pancreatic cancers. 168

3.3. Autophagy and ferroptosis

Autophagy is an important pathway for cells to maintain homeostasis, but excessive or no autophagy induces the appearance of the “autophagy‐dependent death” phenotype. Ferroptosis is an iron‐dependent form of lipid peroxidation‐mediated cell death that requires autophagy‐mediated regulatory mechanisms. 169 Remarkably, several key regulators of ferroptosis, including SLC7A11, GPX4, NRF2, and p53, also hold crucial roles in the orchestration of autophagic processes. For instance, GPX4 has been delineated to directly engage with copper, consequentially inducing the aggregation of GPX4 and its subsequent degradation via autophagic pathways—a pivotal process steering ferroptosis. 170 Chaperone‐mediated autophagy can further catalyze the degradation of GPX4, serving as an ignition for ferroptosis. 171 This crosstalk between autophagy and ferroptosis is closely associated with cancer progression and antitumor immune response. Cells in ferroptosis release proteoglycan decorin (DCN) dependent on autophagy, leading to pro‐inflammatory pathology, and the inhibition of DCN release limits the ability of pancreatic cancer cells in ferroptosis to induce a cancer‐protective immune response. 172 Chen et al. 173 revealed that the interplay between autophagy and ferroptosis is vital for the regulation of TME immunity, treatment resistance, and prognosis. Liu et al. 174 also identified transmembrane protein 164 (TMEM164) as a key regulator of autophagy‐dependent ferroptosis, and the overexpression of TMEM164 is closely associated with the improvement of survival outcome and immune infiltration in patients with pancreatic cancer. In addition, pancreatic cancer cells with autophagy‐dependent ferroptosis release the KRASG12D protein, which is taken up by macrophages, resulting in the polarization of M2 macrophages, which promotes the progression of pancreatic cancer. 175

The intricate dance of autophagy, metabolic reprogramming, and ferroptosis intricately weaves a tapestry that establishes an immunosuppressive TME. Within this complex nexus, metabolic perturbations in the TME actuate the autophagic machinery in CAFs, nudging them toward a glycolytic metabolic predisposition. 176 This metabolic pivot, evoking echoes of a reverse Warburg effect, orchestrates a concerted extrusion of lactate into the TME, providing tumor cells with an essential metabolic lifeline. 176 Particularly in hepatocellular carcinoma, an intrinsic autophagic drive fosters lipid droplet accretion and curtails the presence of CD4+ T cells, embedding the immunosuppressive tenor of the TME even further. 177 Furthermore, a heightened orchestration of lipid metabolism resonates synergistically with autophagy, collaboratively modulating the energetic harmonics of the TME. 178 Central to this, the nuanced choreography between autophagy and metabolic reprogramming potentially creates a reverberating feedback loop, amplifying the immunosuppressive ambiance of the TME, paving the way for immunotherapeutic recalcitrance. As tumors evolve, cells conspicuously adopt a heightened glycolytic metabolic signature, catalyzing a prolific release of lactic acid that subsequently sculpts the TME's architecture. This amassed lactic acid serves a pivotal role as a molecular activator, zeroing in on and invigorating the hydroxycarboxylic acid receptor 1 (HCAR1) ensconced on the tumor cell surface. The ensuing activation of HCAR1 champions the synthesis of monounsaturated fatty acids (MUFA), staunch defenders against ferroptosis. It is imperative to underscore that ferroptotic cells exude lipid metabolites that possess the capability to hamstring the functionality of immune sentinel cells, thereby nurturing the seeds of an immunosuppressive microcosm. 179 In parallel, autophagy‐fueled ferroptosis shepherds the alignment of tumor‐associated macrophages into the M2 paradigm via release and uptake of oncogenic KRAS protein, potentially ushering in adaptive immune subjugation. 175 In summation, the orchestrated symphony among these tripartite pathways crescendos into an immunosuppressive microenvironmental anthem, unveiling novel prospects for strategizing against immunotherapy resistance (Figure 4).

FIGURE 4.

FIGURE 4

Interplay of metabolic reprogramming, autophagy, and ferroptosis in promoting an immunosuppressive TME. Autophagy instigates the glycolytic pathway in CAFs, mediating the reverse Warburg effect, and further establishes a feedback loop with lipid metabolism. Metabolic reprogramming, through lactic acid release, activates HCAR1, promoting MUFA synthesis, leading to ferroptosis. The lipids released subsequently dampen the activity of immune cells. Autophagy‐mediated cell death further accelerates ferroptosis in tumor cells, resulting in the release of KRASG12D protein and activation of M2 macrophages. These cascading events collectively sculpt an immunosuppressive TME.

4. TARGETING METABOLIC REPROGRAMMING, AUTOPHAGY, AND FERROPTOSIS

We summarize the antitumor immunity effect of inhibitors targeting the metabolic reprogramming/autophagy/ferroptosis processes in gastrointestinal cancers in clinical development in Table 2. AZD3965, a selective inhibitor targeting MCT1, significantly reduces colon cancer growth by increasing immune infiltration of DC and NK cells. 180 Sasaki et al. 181 reported that deoxy‐D‐glucose (2‐DG) induces antitumor immunity by regulating glucose metabolism in the TME of hepatocellular carcinoma. A glutamine analog (6‐diazo‐5‐oxo‐l‐norleucine, DON) and its prodrug DRP‐104 inhibit glutamine anabolism and improve the efficacy of immunotherapy in gastrointestinal cancers. 109 , 182 In a patient‐derived xenograft (PDX) model of colorectal cancer, simvastatin reduces cholesterol biosynthesis and elevates antitumor immunity by downregulating PD‐L1 expression. 183 Pharmacological inhibition of fatty acid transporter protein 2 (FATP2) using lipofermata abrogates the activity of polymorphonuclear MDSC and substantially inhibits tumor progression in a mouse model of pancreatic and colorectal cancers. 184 Chloroquine (CQ) and hydroxychloroquine (HCQ), autophagy inhibitors, inhibit lysosome acidification and have been demonstrated as regulators of antitumor immunity in pancreatic and colorectal cancers. 80 , 81 , 185 Two phase II clinical studies (NCT04214418 and NCT03344172) also use HCQ as sensitizer of anti‐PD‐1 in gastrointestinal cancer. Erastin is an inhibitor targeting system Xc, a ferroptosis regulator consisting of SLC3A2 and SLC7A11, that induces powerful antitumor immunity by promoting differentiation of Th17 and activating the efficacy of CD8+ T cells. 186 , 187 Moreover, a recent study also found that sorafenib induces ferroptosis of hepatoma carcinoma cells and enhances antitumor immunity. Therefore, these studies indicate that targeting metabolic reprogramming, autophagy, and ferroptosis are promising strategies for improving the efficacy of immunotherapy in gastrointestinal cancer.

TABLE 2.

Metabolism/autophagy/ferroptosis inhibitors in clinical development combined with immunotherapy.

Metabolic pathways Inhibitor Target Immune effect Cancer types Status/Model References
Glycolysis AZD3965 MCT1 Increase of tumor immune infiltration CRC Preclinical/CDX 180
2‐DG GLUT1 Increase of CD8+ T‐cell chemotaxis HCC Preclinical/CDX and syngeneic mouse 181
Glutamine metabolism DON GFAT1 Sensitization for anti‐PD1 therapy PC Preclinical/syngeneic mouse 100
DPR‐104 GFAT1 Improvement of immune cell infiltration and effect CRC and HCC Preclinical/CDX 182
Lipid metabolism Simvastatin HMGCR Downregulation of PD‐L1 expression CRC Preclinical/PDX 183
Lipofermata FATP2 Inactivation of MDSCs PC, CRC Preclinical/CDX and syngeneic mouse 184
Autophagy HCQ Lysosome Combination with atezolizumab Gastrointestinal cancer Clinical trial/Phase II NCT04214418
HCQ Lysosome Combination with avelumab PC Clinical trial/Phase II NCT03344172
CQ Lysosome Degradation of MHC‐I, Sensitization for anti‐PD1/CTLA‐4 therapy PC Preclinical/CDX and syngeneic mouse 80, 81
CQ Lysosome Induction of DC maturation and T‐cell responses CRC CDX 185
Ferroptosis Erastin System Xc Differentiation of Th17 cell HCC CDX 186
Erastin System Xc Activation of CD8+ T cells HCC and CRC CDX 187
Sorafenib System Xc Infiltration of CD8+ T cells HCC CDX 188

Abbreviations: CDX, cell‐derived xenografts; CRC, colorectal cancer; HCC, hepatocellular carcinoma; PC, pancreatic cancer; PDX, patient‐derived xenografts.

5. CONCLUSIONS AND PERSPECTIVES

Weak immune response and widespread resistance severely limit the application of immunotherapy in gastrointestinal cancer. Metabolic reprogramming, autophagy, and ferroptosis have recently been identified as key regulators of antitumor immunity. This review delineates the synergistic roles of metabolic reprogramming, autophagy, and ferroptosis in shaping antitumor immunity in gastrointestinal malignancies and unravels the intricate interplay between these processes. While metabolic reprogramming can diminish antitumor immune responses by reshaping the behavior of tumor cells, stromal cells, and immune cells within the tumor microenvironment, its systemic nature necessitates the identification of more specific metabolic patterns in gastrointestinal tumors to counteract immunotherapeutic resistance. Both autophagy and ferroptosis profoundly influence the survival capacities of tumor cells and the activity of immune cells in the microenvironment, but they can antagonize while concurrently synergizing antitumor immune responses. The crosstalk between these pathways plays a critical role in advancing an immune‐suppressive tumor milieu. However, the mechanisms by which this interplay fosters resistance to immunotherapeutic strategies in gastrointestinal cancers remain elusive. Deciphering the nuances of tumor immune evasion and tolerance is pivotal for devising combined therapeutic approaches.

From a clinical vantage point, a diverse array of immunotherapeutic strategies, complemented by drug regimens targeting metabolic reprogramming, autophagy, and ferroptosis, have undergone rigorous evaluation in both preclinical models and advanced clinical trials. These endeavors have manifested in early‐phase results that are not only promising but also indicative of the therapeutic potential these avenues harbor. However, navigating the intricate lattice of these molecular pathways presents inherent challenges. To truly leverage their capabilities and overcome the formidable barriers of immunotherapeutic resistance inherent to gastrointestinal tumors, there is an imperative to delve deeper into the discovery of highly specific regulatory nodes and checkpoints. By doing so, we can mitigate the unintended consequences arising from inadvertently disrupting immune cell dynamics within the tumor microenvironment. Such an approach also promises to illuminate the molecular landscapes that are optimally attuned to these therapeutic strategies, enhancing not only the specificity but also the overall efficacy of these targeted interventions.

AUTHOR CONTRIBUTIONS

Xiangwen Wang: Conceptualization (equal); data curation (lead); formal analysis (equal); investigation (lead); software (lead); validation (lead); writing – original draft (lead). Liwen Zhou: Data curation (lead); validation (supporting). Hongpeng Wang: Data curation (supporting); software (lead). Wei Chen: Data curation (lead); validation (equal). Lei Jiang: Data curation (lead); investigation (equal). Guangtao Ming: Data curation (equal). Jun Wang: Conceptualization (lead); data curation (lead); formal analysis (lead); project administration (lead); writing – review and editing (lead).

CONFLICT OF INTEREST STATEMENT

The authors declare that they have no conflict of interest.

ETHICS STATEMENT

Ethics approval statement is not applicable to this article.

ACKNOWLEDGMENTS

Illustrations were created in Biorender.com.

Wang X, Zhou L, Wang H, et al. Metabolic reprogramming, autophagy, and ferroptosis: Novel arsenals to overcome immunotherapy resistance in gastrointestinal cancer. Cancer Med. 2023;12:20573‐20589. doi: 10.1002/cam4.6623

Xiangwen Wang and Liwen Zhou contributed equally.

DATA AVAILABILITY STATEMENT

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

REFERENCES

  • 1. Arnold M, Abnet CC, Neale RE, et al. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology. 2020;159:335‐349.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Procaccio L, Schirripa M, Fassan M, et al. Immunotherapy in gastrointestinal cancers. Biomed Res Int. 2017;2017:4346576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8:1069‐1086. [DOI] [PubMed] [Google Scholar]
  • 4. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450‐461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Wang ZX, Pan YQ, Li X, Tsubata T, Xu RH. Immunotherapy in gastrointestinal cancers: advances, challenges, and countermeasures. Sci Bull. 2023;68:763‐766. [DOI] [PubMed] [Google Scholar]
  • 6. Petricevic B, Kabiljo J, Zirnbauer R, Walczak H, Laengle J, Bergmann M. Neoadjuvant immunotherapy in gastrointestinal cancers—the new standard of care? Semin Cancer Biol. 2022;86(Pt 2):834‐850. [DOI] [PubMed] [Google Scholar]
  • 7. Hazama S, Tamada K, Yamaguchi Y, Kawakami Y, Nagano H. Current status of immunotherapy against gastrointestinal cancers and its biomarkers: perspective for precision immunotherapy. Ann Gastroenterol Surg. 2018;2:289‐303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair‐deficient or microsatellite instability‐high colorectal cancer (CheckMate 142): an open‐label, multicentre, phase 2 study. Lancet Oncol. 2017;18:1182‐1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. le DT, Uram JN, Wang H, et al. PD‐1 blockade in tumors with mismatch‐repair deficiency. N Engl J Med. 2015;372:2509‐2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. O'Reilly EM, Oh DY, Dhani N, et al. Durvalumab with or without Tremelimumab for patients with metastatic pancreatic ductal adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol. 2019;5:1431‐1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Kwon M, An M, Klempner SJ, et al. Determinants of response and intrinsic resistance to PD‐1 blockade in microsatellite instability‐high gastric cancer. Cancer Discov. 2021;11:2168‐2185. [DOI] [PubMed] [Google Scholar]
  • 12. Vesely MD, Zhang T, Chen L. Resistance mechanisms to anti‐PD cancer immunotherapy. Annu Rev Immunol. 2022;40:45‐74. [DOI] [PubMed] [Google Scholar]
  • 13. Sharma P, Hu‐Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707‐723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Nannini G, Meoni G, Amedei A, Tenori L. Metabolomics profile in gastrointestinal cancers: update and future perspectives. World J Gastroenterol. 2020;26:2514‐2532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Chen HT, Liu H, Mao MJ, et al. Crosstalk between autophagy and epithelial‐mesenchymal transition and its application in cancer therapy. Mol Cancer. 2019;18:101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Xu C, Liu Z, Xiao J. Ferroptosis: a double‐edged sword in gastrointestinal disease. Int J Mol Sci. 2021;22:12403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Bader JE, Voss K, Rathmell JC. Targeting metabolism to improve the tumor microenvironment for cancer immunotherapy. Mol Cell. 2020;78:1019‐1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Xia H, Green DR, Zou W. Autophagy in tumour immunity and therapy. Nat Rev Cancer. 2021;21:281‐297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Zheng S, Guan XY. Ferroptosis: promising approach for cancer and cancer immunotherapy. Cancer Lett. 2023;561:216152. [DOI] [PubMed] [Google Scholar]
  • 20. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12:31‐46. [DOI] [PubMed] [Google Scholar]
  • 21. Ganguly K, Kimmelman AC. Reprogramming of tissue metabolism during cancer metastasis. Trends Cancer. 2023;9:461‐471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Sedlak JC, Yilmaz ÖH, Roper J. Metabolism and colorectal cancer. Annu Rev Pathol. 2023;18:467‐492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Lin L, Huang H, Liao W, et al. MACC1 supports human gastric cancer growth under metabolic stress by enhancing the Warburg effect. Oncogene. 2015;34:2700‐2710. [DOI] [PubMed] [Google Scholar]
  • 24. Zhao L, Liu Y, Zhang S, et al. Impacts and mechanisms of metabolic reprogramming of tumor microenvironment for immunotherapy in gastric cancer. Cell Death Dis. 2022;13:378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Kabat AM, Pearce EL, Pearce EJ. Metabolism in type 2 immune responses. Immunity. 2023;56(4):723‐741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Pearce EL, Poffenberger MC, Chang CH, Jones RG. Fueling immunity: insights into metabolism and lymphocyte function. Science. 2013;342:1242454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Beier UH, Angelin A, Akimova T, et al. Essential role of mitochondrial energy metabolism in Foxp3+ T‐regulatory cell function and allograft survival. FASEB J. 2015;29(6):2315‐2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kobayashi T, Lam PY, Jiang H, et al. Increased lipid metabolism impairs NK cell function and mediates adaptation to the lymphoma environment. Blood. 2020;136:3004‐3017. [DOI] [PubMed] [Google Scholar]
  • 29. Ogawa K, Asano K, Yotsumoto S, et al. Frontline science: conversion of neutrophils into atypical Ly6G+ SiglecF+ immune cells with neurosupportive potential in olfactory neuroepithelium. J Leukoc Biol. 2021;109:481‐496. [DOI] [PubMed] [Google Scholar]
  • 30. Yu Y, Cai W, Zhou J, et al. Anti‐arthritis effect of berberine associated with regulating energy metabolism of macrophages through AMPK/ HIF‐1α pathway. Int Immunopharmacol. 2020;87:106830. [DOI] [PubMed] [Google Scholar]
  • 31. Williford JM, Ishihara J, Ishihara A, et al. Recruitment of CD103+ dendritic cells via tumor‐targeted chemokine delivery enhances efficacy of checkpoint inhibitor immunotherapy. Sci Adv. 2019;5:eaay1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Chang CH, Qiu J, O'Sullivan D, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162:1229‐1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. 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. Biomed Pharmacother. 2021;141:111798. [DOI] [PubMed] [Google Scholar]
  • 34. Macintyre AN, Gerriets VA, Nichols AG, et al. The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. Cell Metab. 2014;20:61‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Li W, Tanikawa T, Kryczek I, et al. Aerobic glycolysis controls myeloid‐derived suppressor cells and tumor immunity via a specific CEBPB isoform in triple‐negative breast cancer. Cell Metab. 2018;28:87‐103.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Cortese N, Capretti G, Barbagallo M, et al. Metabolome of pancreatic juice delineates distinct clinical profiles of pancreatic cancer and reveals a link between glucose metabolism and PD‐1+ cells. Cancer Immunol Res. 2020;8:493‐505. [DOI] [PubMed] [Google Scholar]
  • 37. Dai W, Xu Y, Mo S, et al. GLUT3 induced by AMPK/CREB1 axis is key for withstanding energy stress and augments the efficacy of current colorectal cancer therapies. Signal Transduct Target Ther. 2020;5(1):177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Schlößer HA, Drebber U, Urbanski A, et al. Glucose transporters 1, 3, 6, and 10 are expressed in gastric cancer and glucose transporter 3 is associated with UICC stage and survival. Gastric Cancer. 2017;20:83‐91. [DOI] [PubMed] [Google Scholar]
  • 39. He Z, Chen D, Wu 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. [DOI] [PubMed] [Google Scholar]
  • 40. Feng J, Li J, Wu L, et al. Emerging roles and the regulation of aerobic glycolysis in hepatocellular carcinoma. J Exp Clin Cancer Res. 2020;39(1):126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Liu XS, Liu JM, Chen YJ, et al. Comprehensive analysis of hexokinase 2 immune infiltrates and m6A related genes in human esophageal carcinoma. Front Cell Dev Biol. 2021;9:715883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Perrin‐Cocon L, Vidalain PO, Jacquemin C, et al. A hexokinase isoenzyme switch in human liver cancer cells promotes lipogenesis and enhances innate immunity. Commun Biol. 2021;4:217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Liu Z, Ning F, Cai Y, et al. The EGFR‐P38 MAPK axis up‐regulates PD‐L1 through miR‐675‐5p and down‐regulates HLA‐ABC via hexokinase‐2 in hepatocellular carcinoma cells. Cancer Commun (Lond). 2021;41:62‐78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Li S, Dai W, Mo W, et al. By inhibiting PFKFB3, aspirin overcomes sorafenib resistance in hepatocellular carcinoma. Int J Cancer. 2017;141:2571‐2584. [DOI] [PubMed] [Google Scholar]
  • 45. Chen DP, Ning WR, Jiang ZZ, et al. Glycolytic activation of peritumoral monocytes fosters immune privilege via the PFKFB3‐PD‐L1 axis in human hepatocellular carcinoma. J Hepatol. 2019;71:333‐343. [DOI] [PubMed] [Google Scholar]
  • 46. Li TE, Wang S, Shen XT, et al. PKM2 drives hepatocellular carcinoma progression by inducing immunosuppressive microenvironment. Front Immunol. 2020;11:589997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Palsson‐McDermott EM, Dyck L, Zasłona Z, et al. Pyruvate kinase M2 is required for the expression of the immune checkpoint PD‐L1 in immune cells and tumors. Front Immunol. 2017;8:1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Kumagai S, Koyama S, Itahashi K, et al. Lactic acid promotes PD‐1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell. 2022;40:201‐218.e9. [DOI] [PubMed] [Google Scholar]
  • 49. Sun L, Li J, Yan W, et al. H19 promotes aerobic glycolysis, proliferation, and immune escape of gastric cancer cells through the microRNA‐519d‐3p/lactate dehydrogenase a axis. Cancer Sci. 2021. Jun;112(6):2245‐2259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Zhou X, Fang D, Liu H, et al. PMN‐MDSCs accumulation induced by CXCL1 promotes CD8+ T cells exhaustion in gastric cancer. Cancer Lett. 2022;532:215598. [DOI] [PubMed] [Google Scholar]
  • 51. Wang F, Zhang Y, Yu X, et al. ZFP91 disturbs metabolic fitness and antitumor activity of tumor‐infiltrating T cells. J Clin Invest. 2021;131:e144318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Gu M, Zhou X, Sohn JH, et al. NF‐κB‐inducing kinase maintains T cell metabolic fitness in antitumor immunity. Nat Immunol. 2021;22:193‐204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Lu Y, Li Y, Liu Q, et al. MondoA‐thioredoxin‐interacting protein Axis maintains regulatory T‐cell identity and function in colorectal cancer microenvironment. Gastroenterology. 2021;161:575‐591.e16. [DOI] [PubMed] [Google Scholar]
  • 54. Cai J, Chen Z, Zhang Y, et al. CircRHBDD1 augments metabolic rewiring and restricts immunotherapy efficacy via m6A modification in hepatocellular carcinoma. Mol Ther Oncolytics. 2022;24:755‐771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Xu D, Wang Y, Wu J, et al. ECT2 overexpression promotes the polarization of tumor‐associated macrophages in hepatocellular carcinoma via the ECT2/PLK1/PTEN pathway. Cell Death Dis. 2021;12:162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Wang N, Tan HY, Lu Y, et al. PIWIL1 governs the crosstalk of cancer cell metabolism and immunosuppressive microenvironment in hepatocellular carcinoma. Signal Transduct Target Ther. 2021;6:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Fu S, He K, Tian C, et al. Impaired lipid biosynthesis hinders anti‐tumor efficacy of intratumoral iNKT cells. Nat Commun. 2020;11:438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Guo Q, Chu Y, Li H, et al. Dickkopf‐related protein 3 alters aerobic glycolysis in pancreatic cancer BxPC‐3 cells, promoting CD4+ T‐cell activation and function. Eur J Med Res. 2021;26:93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Wu S, Zhang H, Gao C, et al. Hyperglycemia enhances immunosuppression and aerobic glycolysis of pancreatic cancer through upregulating Bmi1‐UPF1‐HK2 pathway. Cell Mol Gastroenterol Hepatol. 2022;14:1146‐1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Wang L, Liu Y, Dai Y, et al. Single‐cell RNA‐seq analysis reveals BHLHE40‐driven pro‐tumour neutrophils with hyperactivated glycolysis in pancreatic tumour microenvironment. Gut. 2022;72:958‐971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Kodama M, Oshikawa K, Shimizu H, et al. A shift in glutamine nitrogen metabolism contributes to the malignant progression of cancer. Nat Commun. 2020;11:1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Liu Y, Yang Y, Jiang L, Xu H, Wei J. High expression levels of SLC38A1 are correlated with poor prognosis and defective immune infiltration in hepatocellular carcinoma. J Oncol. 2021;2021:5680968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Xu M, Wang X, Li Y, et al. Arachidonic acid metabolism controls macrophage alternative activation through regulating oxidative phosphorylation in PPARγ dependent manner. Front Immunol. 2021;12:618501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Wang Z, Cheng Q, Tang K, et al. Lipid mediator lipoxin A4 inhibits tumor growth by targeting IL‐10‐producing regulatory B (Breg) cells. Cancer Lett. 2015;364:118‐124. [DOI] [PubMed] [Google Scholar]
  • 65. Wood SM, Gill AJ, Brodsky AS, et al. Fatty acid‐binding protein 1 is preferentially lost in microsatellite instable colorectal carcinomas and is immune modulated via the interferon γ pathway. Mod Pathol. 2017;30:123‐133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Zhu Y, Gu L, Lin X, et al. USP19 exacerbates lipogenesis and colorectal carcinogenesis by stabilizing ME1. Cell Rep. 2021;37:110174. [DOI] [PubMed] [Google Scholar]
  • 67. Shirasaki T, Honda M, Shimakami T, et al. MicroRNA‐27a regulates lipid metabolism and inhibits hepatitis C virus replication in human hepatoma cells. J Virol. 2013;87:5270‐5286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Du W, Hua F, Li X, et al. Loss of optineurin drives cancer immune evasion via palmitoylation‐dependent IFNGR1 lysosomal sorting and degradation. Cancer Discov. 2021;11:1826‐1843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Sun H, Yang W, Tian Y, et al. An inflammatory‐CCRK circuitry drives mTORC1‐dependent metabolic and immunosuppressive reprogramming in obesity‐associated hepatocellular carcinoma. Nat Commun. 2018;9:5214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Wu D, Liang S, Guo H, et al. Downregulation of MARC2 promotes immune escape and is associated with immunosuppression of hepatocellular carcinoma. Front Genet. 2022;12:790093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Choi BK, Lee DY, Lee DG, et al. 4‐1BB signaling activates glucose and fatty acid metabolism to enhance CD8+ T cell proliferation. Cell Mol Immunol. 2017;14:748‐757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Wang G, Xu J, Zhao J, et al. Arf1‐mediated lipid metabolism sustains cancer cells and its ablation induces anti‐tumor immune responses in mice. Nat Commun. 2020;11:220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Manzo T, Prentice BM, Anderson KG, et al. Accumulation of long‐chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J Exp Med. 2020;217:e20191920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Song MJ, Park S, Won KY. Expression of Beclin‐1, an autophagy‐related protein, is associated with tumoral FOXP3 expression and Tregs in gastric adenocarcinoma: the function of Beclin‐1 expression as a favorable prognostic factor in gastric adenocarcinoma. Pathol Res Pract. 2020;216:152927. [DOI] [PubMed] [Google Scholar]
  • 75. Zheng Y, Zhu G. HMGB1 suppresses colon carcinoma cell apoptosis triggered by co‐culture with dendritic cells via an ER stress‐associated autophagy pathway. Mol Med Rep. 2018;17:3123‐3132. [DOI] [PubMed] [Google Scholar]
  • 76. Kosumi K, Masugi Y, Yang J, et al. Tumor SQSTM1 (p62) expression and T cells in colorectal cancer. Onco Targets Ther. 2017;6:e1284720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Grimm WA, Messer JS, Murphy SF, et al. The Thr300Ala variant in ATG16L1 is associated with improved survival in human colorectal cancer and enhanced production of type I interferon. Gut. 2016;65:456‐464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Wang Y, Zhang J, Jiang P, Li K, Sun Y, Huang Y. ASIC1a promotes acidic microenvironment‐induced HCC cells migration and invasion by inducing autophagy. Eur J Pharmacol. 2021;907:174252. [DOI] [PubMed] [Google Scholar]
  • 79. Yin S, Jin W, Qiu Y, Fu L, Wang T, Yu H. Solamargine induces hepatocellular carcinoma cell apoptosis and autophagy via inhibiting LIF/miR‐192‐5p/CYR61/Akt signaling pathways and eliciting immunostimulatory tumor microenvironment. J Hematol Oncol. 2022;15:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Yamamoto K, Venida A, Yano J, et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC‐I. Nature. 2020;581:100‐105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Yamamoto K, Venida A, Perera RM, Kimmelman AC. Selective autophagy of MHC‐I promotes immune evasion of pancreatic cancer. Autophagy. 2020;16:1524‐1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Frey N, Tortola L, Egli D, et al. Loss of Rnf31 and Vps4b sensitizes pancreatic cancer to T cell‐mediated killing. Nat Commun. 2022;13:1804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Zhang D, Lindstrom A, Kim EJ, et al. SEMA3C supports pancreatic cancer progression by regulating the autophagy process and tumor immune microenvironment. Front Oncol. 2022;12:890154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Hou J, Huang Q, Fan Z, et al. LncRNA OIP5‐AS1 knockdown facilitated the ferroptosis and immune evasion by modulating the GPX4 in oesophageal carcinoma. Comput Math Methods Med. 2022;2022:8103198. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 85. Lin L, Que R, Wang J, Zhu Y, Liu X, Xu R. Prognostic value of the ferroptosis‐related gene SLC2A3 in gastric cancer and related immune mechanisms. Front Genet. 2022;13:919313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Liu XS, Yang JW, Zeng J, et al. SLC2A1 is a diagnostic biomarker involved in immune infiltration of colorectal cancer and associated with m6A modification and ceRNA. Front Cell Dev Biol. 2022;10:853596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Peng B, Peng J, Kang F, Zhang W, Peng E, He Q. Ferroptosis‐related gene MT1G as a novel biomarker correlated with prognosis and immune infiltration in colorectal cancer. Front Cell Dev Biol. 2022;10:881447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Song J, Liu T, Yin Y, et al. The deubiquitinase OTUD1 enhances iron transport and potentiates host antitumor immunity. EMBO Rep. 2021;22:e51162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Li J, Tian X, Nie Y, et al. BTBD10 is a prognostic biomarker correlated with immune infiltration in hepatocellular carcinoma. Front Mol Biosci. 2022;8:762541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Long S, Peng F, Song B, Wang L, Chen J, Shang B. Heat shock protein Beta 1 is a prognostic biomarker and correlated with immune infiltrates in hepatocellular carcinoma. Int J Gen Med. 2021;14:5483‐5492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Tan P, Li M, Liu Z, Li T, Zhao L, Fu W. Glycolysis‐related LINC02432/Hsa‐miR‐98‐5p/HK2 Axis inhibits ferroptosis and predicts immune infiltration, tumor mutation burden, and drug sensitivity in pancreatic adenocarcinoma. Front Pharmacol. 2022;13:937413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Recouvreux MV, Moldenhauer MR, Galenkamp KMO, et al. Glutamine depletion regulates slug to promote EMT and metastasis in pancreatic cancer. J Exp Med. 2020;217:e20200388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Wong CC, Xu J, Bian X, et al. In colorectal cancer cells with mutant KRAS, SLC25A22‐mediated Glutaminolysis reduces DNA demethylation to increase WNT signaling, stemness, and drug resistance. Gastroenterology. 2020;159:2163‐2180.e6. [DOI] [PubMed] [Google Scholar]
  • 94. Cruzat V, Macedo Rogero M, Noel Keane K, Curi R, Newsholme P. Glutamine: metabolism and immune function, supplementation and clinical translation. Nutrients. 2018;10:1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Edwards DN, Ngwa VM, Raybuck AL, et al. Selective glutamine metabolism inhibition in tumor cells improves antitumor T lymphocyte activity in triple‐negative breast cancer. J Clin Invest. 2021;131:e140100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Carr EL, Kelman A, Wu GS, et al. Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol. 2010;185:1037‐1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Reinfeld BI, Madden MZ, Wolf MM, et al. Cell‐programmed nutrient partitioning in the tumour microenvironment. Nature. 2021;593:282‐288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Wei Y, Tang X, Ren Y, et al. An RNA‐RNA crosstalk network involving HMGB1 and RICTOR facilitates hepatocellular carcinoma tumorigenesis by promoting glutamine metabolism and impedes immunotherapy by PD‐L1+ exosomes activity. Signal Transduct Target Ther. 2021;6:421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Leone RD, Zhao L, Englert JM, et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science. 2019;366:1013‐1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Sharma NS, Gupta VK, Garrido VT, et al. Targeting tumor‐intrinsic hexosamine biosynthesis sensitizes pancreatic cancer to anti‐PD1 therapy. J Clin Invest. 2020;130:451‐465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Zhu D, Wu S, Li Y, et al. Ferroptosis‐related gene SLC1A5 is a novel prognostic biomarker and correlates with immune infiltrates in stomach adenocarcinoma. Cancer Cell Int. 2022;22:124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Nachef M, Ali AK, Almutairi SM, Lee SH. Targeting SLC1A5 and SLC3A2/SLC7A5 as a potential strategy to strengthen anti‐tumor immunity in the tumor microenvironment. Front Immunol. 2021;12:624324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Bleve A, Durante B, Sica A, Consonni FM. Lipid metabolism and cancer immunotherapy: immunosuppressive myeloid cells at the crossroad. Int J Mol Sci. 2020;21:5845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Luo Q, Zheng N, Jiang L, et al. Lipid accumulation in macrophages confers protumorigenic polarization and immunity in gastric cancer. Cancer Sci. 2020;111:4000‐4011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Wu H, Han Y, Rodriguez Sillke Y, et al. Lipid droplet‐dependent fatty acid metabolism controls the immune suppressive phenotype of tumor‐associated macrophages. EMBO Mol Med. 2019;11:e10698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Herber DL, Cao W, Nefedova Y, et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat Med. 2010;16:880‐886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Wang H, Franco F, Tsui YC, et al. CD36‐mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat Immunol. 2020;21:298‐308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Voss K, Luthers CR, Pohida K, Snow AL. Fatty acid synthase contributes to Restimulation‐induced cell death of human CD4 T cells. Front Mol Biosci. 2019;6:106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Michalek RD, Gerriets VA, Jacobs SR, et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011;186:3299‐3303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Zhang Q, Wang H, Mao C, et al. Fatty acid oxidation contributes to IL‐1β secretion in M2 macrophages and promotes macrophage‐mediated tumor cell migration. Mol Immunol. 2018;94:27‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Perrone F, Minari R, Bersanelli M, et al. The prognostic role of high blood cholesterol in advanced cancer patients treated with immune checkpoint inhibitors. J Immunother. 2020;43:196‐203. [DOI] [PubMed] [Google Scholar]
  • 112. Jiang C, Liu Y, Wen S, Xu C, Gu L. In silico development and clinical validation of novel 8 gene signature based on lipid metabolism related genes in colon adenocarcinoma. Pharmacol Res. 2021;169:105644. [DOI] [PubMed] [Google Scholar]
  • 113. King RJ, Singh PK, Mehla K. The cholesterol pathway: impact on immunity and cancer. Trends Immunol. 2022;43:78‐92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Gu L, Zhu Y, Lin X, Tan X, Lu B, Li Y. Stabilization of FASN by ACAT1‐mediated GNPAT acetylation promotes lipid metabolism and hepatocarcinogenesis. Oncogene. 2020;39:2437‐2449. [DOI] [PubMed] [Google Scholar]
  • 115. Yang W, Bai Y, Xiong Y, et al. Potentiating the antitumour response of CD8(+) T cells by modulating cholesterol metabolism. Nature. 2016;531:651‐655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Shi R, Zhao K, Wang T, et al. 5‐aza‐2′‐deoxycytidine potentiates anti‐tumor immunity in colorectal peritoneal metastasis by modulating ABC A9‐mediated cholesterol accumulation in macrophages. Theranostics. 2022;12:875‐890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Qin WH, Yang ZS, Li M, et al. High serum levels of cholesterol increase antitumor functions of nature killer cells and reduce growth of liver tumors in mice. Gastroenterology. 2020;158(6):1713‐1727. [DOI] [PubMed] [Google Scholar]
  • 118. Tavazoie MF, Pollack I, Tanqueco R, et al. LXR/ApoE activation restricts innate immune suppression in cancer. Cell. 2018;172:825‐840.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Zhang Q, Kang R, Zeh HJ III, et al. DAMPs and autophagy: cellular adaptation to injury and unscheduled cell death. Autophagy. 2013;9:451‐458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Hu B, Yang XB, Sang XT. Construction of a lipid metabolism‐related and immune‐associated prognostic signature for hepatocellular carcinoma. Cancer Med. 2020;9:7646‐7662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Li J, Chen X, Kang R, Zeh H, Klionsky DJ, Tang D. Regulation and function of autophagy in pancreatic cancer. Autophagy. 2021;17:3275‐3296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Chao X, Qian H, Wang S, Fulte S, Ding WX. Autophagy and liver cancer. Clin Mol Hepatol. 2020;26:606‐617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Yu S, Wang Y, Jing L, et al. Autophagy in the "inflammation‐carcinogenesis" pathway of liver and HCC immunotherapy. Cancer Lett. 2017;411:82‐89. [DOI] [PubMed] [Google Scholar]
  • 124. Xia H, Wang W, Crespo J, et al. Suppression of FIP200 and autophagy by tumor‐derived lactate promotes naïve T cell apoptosis and affects tumor immunity. Sci Immunol. 2017;2:eaan4631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Yang G, Song W, Postoak JL, et al. Autophagy‐related protein PIK3C3/VPS34 controls T cell metabolism and function. Autophagy. 2021;17:1193‐1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Jia W, He MX, McLeod IX, et al. Autophagy regulates T lymphocyte proliferation through selective degradation of the cell‐cycle inhibitor CDKN1B/p27Kip1. Autophagy. 2015;11:2335‐2345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Xu X, Araki K, Li S, et al. Autophagy is essential for effector CD8(+) T cell survival and memory formation. Nat Immunol. 2014;15:1152‐1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Lee HK, Mattei LM, Steinberg BE, et al. In vivo requirement for Atg5 in antigen presentation by dendritic cells. Immunity. 2010;32:227‐239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Parekh VV, Pabbisetty SK, Wu L, et al. Autophagy‐related protein Vps34 controls the homeostasis and function of antigen cross‐presenting CD8α+ dendritic cells. Proc Natl Acad Sci USA. 2017;114:E6371‐E6380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Wang S, Xia P, Huang G, et al. FoxO1‐mediated autophagy is required for NK cell development and innate immunity. Nat Commun. 2016;7:11023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Chen M, Hong MJ, Sun H, et al. Essential role for autophagy in the maintenance of immunological memory against influenza infection. Nat Med. 2014;20:503‐510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Zhang X, Fan J, Wang S, et al. Targeting CD47 and autophagy elicited enhanced antitumor effects in non‐small cell lung cancer. Cancer Immunol Res. 2017;5:363‐375. [DOI] [PubMed] [Google Scholar]
  • 133. Dong G, Si C, Zhang Q, et al. Autophagy regulates accumulation and functional activity of granulocytic myeloid‐derived suppressor cells via STAT3 signaling in endotoxin shock. Biochim Biophys Acta Mol Basis Dis. 2017;1863:2796‐2807. [DOI] [PubMed] [Google Scholar]
  • 134. Wei J, Long L, Yang K, et al. Autophagy enforces functional integrity of regulatory T cells by coupling environmental cues and metabolic homeostasis. Nat Immunol. 2016;17:277‐285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Li X, Lee Y, Kang Y, et al. Hypoxia‐induced autophagy of stellate cells inhibits expression and secretion of lumican into microenvironment of pancreatic ductal adenocarcinoma. Cell Death Differ. 2019;26:382‐393. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 136. Wang Y, Gan G, Wang B, et al. Cancer‐associated fibroblasts promote irradiated cancer cell recovery through autophagy. EBioMedicine. 2017;17:45‐56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Lin SY, Hsieh SY, Fan YT, et al. Necroptosis promotes autophagy‐dependent upregulation of DAMP and results in immunosurveillance. Autophagy. 2018;14:778‐795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Jiang H, Courau T, Borison J, et al. Activating immune recognition in pancreatic ductal adenocarcinoma via autophagy inhibition, MEK blockade, and CD40 agonism. Gastroenterology. 2022;162:590‐603.e14. [DOI] [PubMed] [Google Scholar]
  • 139. Michaud M, Martins I, Sukkurwala AQ, et al. Autophagy‐dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573‐1577. [DOI] [PubMed] [Google Scholar]
  • 140. Wang X, Wu WKK, Gao J, et al. Autophagy inhibition enhances PD‐L1 expression in gastric cancer. J Exp Clin Cancer Res. 2019;38:140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Debnath J, Gammoh N, Ryan KM. Autophagy and autophagy‐related pathways in cancer. Nat Rev Mol Cell Biol. 2023;24:1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Tang R, Wu Z, Rong Z, et al. Ferroptosis‐related lncRNA pairs to predict the clinical outcome and molecular characteristics of pancreatic ductal adenocarcinoma. Brief Bioinform. 2022;23:bbab388. [DOI] [PubMed] [Google Scholar]
  • 143. Wu Z, Lu Z, Li L, et al. Identification and validation of ferroptosis‐related LncRNA signatures as a novel prognostic model for colon cancer. Front Immunol. 2022;12:783362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Xu Z, Peng B, Liang Q, et al. Construction of a ferroptosis‐related nine‐lncRNA signature for predicting prognosis and immune response in hepatocellular carcinoma. Front Immunol. 2021;12:719175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Li J, Xiang R, Song W, Wu J, Kong C, Fu T. A novel ferroptosis‐related LncRNA pair prognostic signature predicts immune landscapes and treatment responses for gastric cancer patients. Front Genet. 2022;13:899419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Drijvers JM, Gillis JE, Muijlwijk T, et al. Pharmacologic screening identifies metabolic vulnerabilities of CD8+ T cells. Cancer Immunol Res. 2021;9:184‐199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Ma X, Xiao L, Liu L, et al. CD36‐mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab. 2021;33:1001‐1012.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Xu H, Ye D, Ren M, Zhang H, Bi F. Ferroptosis in the tumor microenvironment: perspectives for immunotherapy. Trends Mol Med. 2021;27:856‐867. [DOI] [PubMed] [Google Scholar]
  • 149. Wang W, Green M, Choi JE, et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569:270‐274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Yu B, Choi B, Li W, Kim DH. Magnetic field boosted ferroptosis‐like cell death and responsive MRI using hybrid vesicles for cancer immunotherapy. Nat Commun. 2020;11:3637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Zhang H, Zhuo Y, Li D, et al. Dihydroartemisinin inhibits the growth of pancreatic cells by inducing ferroptosis and activating antitumor immunity. Eur J Pharmacol. 2022;926:175028. [DOI] [PubMed] [Google Scholar]
  • 152. Kang R, Tang D. Autophagy and ferroptosis—What's the connection? Curr Pathobiol Rep. 2017;5:153‐159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Wu Y, Zhang S, Gong X, et al. The epigenetic regulators and metabolic changes in ferroptosis‐associated cancer progression. Mol Cancer. 2020;19:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Anderson CM, Macleod KF. Autophagy and cancer cell metabolism. Int Rev Cell Mol Biol. 2019;347:145‐190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Viry E, Paggetti J, Baginska J, et al. Autophagy: an adaptive metabolic response to stress shaping the antitumor immunity. Biochem Pharmacol. 2014;92(1):31‐42. [DOI] [PubMed] [Google Scholar]
  • 156. Noman MZ, Buart S, Van Pelt J, et al. The cooperative induction of hypoxia‐inducible factor‐1 alpha and STAT3 during hypoxia induced an impairment of tumor susceptibility to CTL‐mediated cell lysis. J Immunol. 2009;182:3510‐3521. [DOI] [PubMed] [Google Scholar]
  • 157. Baginska J, Viry E, Berchem G, et al. Granzyme B degradation by autophagy decreases tumor cell susceptibility to natural killer‐mediated lysis under hypoxia. Proc Natl Acad Sci USA. 2013;110:17450‐17455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Wu D, Zhuo L, Wang X. Metabolic reprogramming of carcinoma‐associated fibroblasts and its impact on metabolic heterogeneity of tumors. Semin Cell Dev Biol. 2017;64:125‐131. [DOI] [PubMed] [Google Scholar]
  • 159. Lee YR, Chen M, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor: new modes and prospects. Nat Rev Mol Cell Biol. 2018;19:547‐562. [DOI] [PubMed] [Google Scholar]
  • 160. Peng W, Chen JQ, Liu C, et al. Loss of PTEN promotes resistance to T cell‐mediated immunotherapy. Cancer Discov. 2016;6:202‐216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Sahin E, Haubenwallner S, Kuttke M, et al. Macrophage PTEN regulates expression and secretion of arginase I modulating innate and adaptive immune responses. J Immunol. 2014;193:1717‐1727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Liu Y, Duan C, Dai R, Zeng Y. Ferroptosis‐mediated crosstalk in the tumor microenvironment implicated in cancer progression and therapy. Front Cell Dev Biol. 2021;9:739392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Pucino V, Certo M, Bulusu V, et al. Lactate buildup at the site of chronic inflammation promotes disease by inducing CD4+ T cell metabolic rewiring. Cell Metab. 2019;30:1055‐1074.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. de la Cruz‐López KG, Castro‐Muñoz LJ, Reyes‐Hernández DO, García‐Carrancá A, Manzo‐Merino J. Lactate in the regulation of tumor microenvironment and therapeutic approaches. Front Oncol. 2019;9:1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer‐acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19:405‐414. [DOI] [PubMed] [Google Scholar]
  • 166. Johnson AM, Kleczko EK, Nemenoff RA. Eicosanoids in cancer: new roles in immunoregulation. Front Pharmacol. 2020;11:595498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Kremer DM, Nelson BS, Lin L, et al. GOT1 inhibition promotes pancreatic cancer cell death by ferroptosis. Nat Commun. 2021;12:4860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Arensman MD, Yang XS, Leahy DM, et al. Cystine‐glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proc Natl Acad Sci USA. 2019;116:9533‐9542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Zhang L, Cui T, Wang X. The interplay between autophagy and regulated necrosis. Antioxid Redox Signal. 2023;38(7–9):550‐580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Xue Q, Yan D, Chen X, et al. Copper‐dependent autophagic degradation of GPX4 drives ferroptosis. Autophagy. 2023;19:1982‐1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Wu Z, Geng Y, Lu X, et al. Chaperone‐mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci USA. 2019;116:2996‐3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Liu J, Zhu S, Zeng L, et al. DCN released from ferroptotic cells ignites AGER‐dependent immune responses. Autophagy. 2022;18:2036‐2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Chen L, Niu X, Qiao X, et al. Characterization of interplay between autophagy and ferroptosis and their synergistical roles on manipulating immunological tumor microenvironment in squamous cell carcinomas. Front Immunol. 2022;12:739039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Liu J, Liu Y, Wang Y, et al. TMEM164 is a new determinant of autophagy‐dependent ferroptosis. Autophagy. 2022;19:1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Dai E, Han L, Liu J, et al. Autophagy‐dependent ferroptosis drives tumor‐associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 2020;16:2069‐2083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Zhang P, Cheng S, Sheng X, Dai H, He K, du Y. The role of autophagy in regulating metabolism in the tumor microenvironment. Genes Dis. 2021;10:447‐456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Ma C, Kesarwala AH, Eggert T, et al. NAFLD causes selective CD4(+) T lymphocyte loss and promotes hepatocarcinogenesis. Nature. 2016;531:253‐257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Singh R, Kaushik S, Wang Y, et al. Autophagy regulates lipid metabolism. Nature. 2009;458:1131‐1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Zhao Y, Li M, Yao X, et al. HCAR1/MCT1 regulates tumor ferroptosis through the lactate‐mediated AMPK‐SCD1 activity and its therapeutic implications. Cell Rep. 2020;33:108487. [DOI] [PubMed] [Google Scholar]
  • 180. Beloueche‐Babari M, Casals Galobart T, Delgado‐Goni T, et al. Monocarboxylate transporter 1 blockade with AZD3965 inhibits lipid biosynthesis and increases tumour immune cell infiltration. Br J Cancer. 2020;122:895‐903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Sasaki K, Nishina S, Yamauchi A, et al. Nanoparticle‐mediated delivery of 2‐deoxy‐D‐glucose induces antitumor immunity and cytotoxicity in liver tumors in mice. Cell Mol Gastroenterol Hepatol. 2021;11:739‐762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Yokoyama Y, Wild R, Estok TM. Sirpiglenastat (DRP‐104) induces anti‐tumor efficacy through direct, broad antagonism of glutamine metabolism and stimulation of the innate & adaptive immune systems. Mol Cancer Ther. 2022;21:1561‐1572. [DOI] [PubMed] [Google Scholar]
  • 183. Ni W, Mo H, Liu Y, et al. Targeting cholesterol biosynthesis promotes anti‐tumor immunity by inhibiting long noncoding RNA SNHG29‐mediated YAP activation. Mol Ther. 2021;29(10):2995‐3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Veglia F, Tyurin VA, Blasi M, et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature. 2019;569:73‐78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Zamame Ramirez JA, Romagnoli GG, Falasco BF, et al. Blocking drug‐induced autophagy with chloroquine in HCT‐116 colon cancer cells enhances DC maturation and T cell responses induced by tumor cell lysate. Int Immunopharmacol. 2020;84:106495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Tang B, Zhu J, Li J, et al. The ferroptosis and iron‐metabolism signature robustly predicts clinical diagnosis, prognosis and immune microenvironment for hepatocellular carcinoma. Cell Commun Signal. 2020;18:174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Liu W, Chen H, Zhu Z, et al. Ferroptosis inducer improves the efficacy of oncolytic virus‐mediated cancer immunotherapy. Biomedicine. 2022;10:1425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Li Z, Bu J, Zhu X, et al. Anti‐tumor immunity and ferroptosis of hepatocellular carcinoma are enhanced by combined therapy of sorafenib and delivering modified GO‐based PD‐L1 siRNAs. Biomater Adv. 2022;136:212761. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.


Articles from Cancer Medicine are provided here courtesy of Wiley

RESOURCES