- 1Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- 2Department of Clinical Laboratory, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. Recent advancements have illuminated the intricate metabolic reprogramming that underpins NSCLC progression and resistance to therapy. Beyond the classical Warburg effect, emerging evidence highlights the pivotal roles of altered lipid metabolism, amino acid utilization, and the metabolic crosstalk within the tumor microenvironment (TME). This review delves into the latest discoveries in NSCLC metabolism, emphasizing novel pathways and mechanisms that contribute to tumor growth and survival. We critically assess the interplay between cancer cell metabolism and the TME, explore the impact of metabolic heterogeneity, and discuss how metabolic adaptations confer therapeutic resistance. By integrating insights from cutting-edge technologies such as single-cell metabolomics and spatial metabolomics, we identify potential metabolic vulnerabilities in NSCLC. Finally, we propose innovative therapeutic strategies that target these metabolic dependencies, including combination approaches that enhance the efficacy of existing treatments and pave the way for personalized metabolic therapies.
1 Introduction
Lung cancer remains a global health challenge, being the leading cause of cancer-related mortality worldwide, with an estimated 2.2 million new cases and 1.8 million deaths in 2020 (1). NSCLC accounts for approximately 85% of all lung cancer cases and includes various histological subtypes such as adenocarcinoma, squamous cell carcinoma, and large cell carcinoma (2). Despite significant advances in surgical techniques, chemotherapy, targeted therapies, and immunotherapy, the overall five-year survival rate for NSCLC patients remains low, particularly in advanced stages where it drops below 10% (3). Late diagnosis, tumor heterogeneity, and the development of resistance to conventional treatments contribute to this poor prognosis (4). Therefore, a deeper understanding of the underlying mechanisms driving NSCLC progression and therapeutic resistance is crucial for developing more effective treatments.
One area of intense research is the metabolic reprogramming of cancer cells-a hallmark of cancer that supports rapid proliferation and survival under hostile conditions (5). Metabolic alterations enable cancer cells to meet the increased demands for energy and biosynthetic precursors required for continuous growth and division (6). Warburg effect, characterized by increased glycolysis even in the presence of oxygen, has been a focal point of cancer metabolism research (7), recent studies have uncovered a more complex metabolic landscape in NSCLC. Alterations in lipid metabolism, amino acid utilization, and metabolic interactions with the TME play significant roles in tumor progression, metastasis, and therapeutic resistance (8). Furthermore, metabolic heterogeneity within tumors and the metabolic plasticity of cancer cells allow them to adapt to changing environmental conditions and therapeutic pressures (9).
The TME, comprising stromal cells, immune cells, extracellular matrix (ECM) components, and vasculature, interacts dynamically with cancer cells, influencing their metabolic behavior and contributing to disease progression (10). Cancer-associated fibroblasts (CAFs) can alter the availability of nutrients and secrete metabolic intermediates that fuel tumor growth (11). Immune cells within the TME can have their function modulated by the metabolic activities of cancer cells, leading to immune evasion (12). Hypoxia, a common feature of solid tumors due to abnormal vasculature, further drives metabolic reprogramming by stabilizing hypoxia-inducible factors (HIFs) that regulate genes involved in glycolysis and angiogenesis (13).
This review aims to provide a comprehensive and up-to-date analysis of energy metabolism in NSCLC, highlighting novel insights and potential therapeutic opportunities. We focus on recent discoveries that shed light on the metabolic heterogeneity of NSCLC, the influence of the TME on metabolic adaptations, and the implications for therapy resistance. By integrating emerging technologies such as single-cell metabolomics, CRISPR-based metabolic screens, and systems biology approaches, and by adopting multidisciplinary perspectives, we propose innovative strategies to target metabolic vulnerabilities in NSCLC. Ultimately, we aim to bridge the gap between basic metabolic research and clinical applications, paving the way for more effective and personalized therapies for NSCLC patients.
2 Metabolic reprogramming in NSCLC: beyond the Warburg effect
2.1 The Warburg effect revisited
The Warburg effect, first described by Otto Warburg in the 1920s, refers to the observation that cancer cells preferentially utilize glycolysis for energy production even in the presence of adequate oxygen-a phenomenon known as aerobic glycolysis. This metabolic reprogramming allows cancer cells to rapidly generate ATP and accumulate intermediates for biosynthetic processes essential for proliferation (14). In NSCLC, there is significant upregulation of glycolytic enzymes such as hexokinase 2 (HK2) and pyruvate kinase M2 (PKM2), which facilitate increased glycolytic flux (15). However, this glycolytic shift is only part of the complex metabolic adaptations in NSCLC. Recent studies have revealed that mitochondrial oxidative phosphorylation (OXPHOS) remains active in many cancer cells, including NSCLC (16). This suggests that cancer cells exhibit metabolic flexibility, capable of utilizing both glycolysis and OXPHOS depending on environmental conditions and cellular demands (17). For instance, under hypoxic conditions commonly found within tumors, glycolysis is upregulated, while in oxygen-rich areas, OXPHOS can contribute significantly to adenosine triphosphate (ATP) production (18).
Moreover, the reliance on glycolysis is influenced by oncogenic signaling pathways. Mutations in genes such as kirsten rats arcomaviral oncogene homolog (KRAS) and epidermal growth factor receptor (EGFR), which are prevalent in NSCLC, activate downstream effectors like phosphatidylinositol 3-Kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and mitogen-activated protein kinase (MAPK) pathways (19). These pathways upregulate glucose transporters (e.g., GLUT1) and glycolytic enzymes, enhancing glucose uptake and glycolysis (20). Additionally, HIFs, stabilized under low oxygen conditions, promote the expression of genes involved in glycolysis and suppress OXPHOS (21).
Understanding the nuances of the Warburg effect in NSCLC is crucial for therapeutic development. Targeting glycolytic enzymes has shown promise in preclinical models; however, due to the metabolic plasticity of cancer cells, inhibition of glycolysis alone may lead to compensatory upregulation of OXPHOS or other pathways. Therefore, combination therapies that target multiple metabolic pathways may be more effective in overcoming resistance and achieving sustained antitumor effects.
2.2 Mitochondrial metabolism and OXPHOS
Contrary to the traditional view that cancer cells have impaired mitochondrial function, recent studies have demonstrated that mitochondrial OXPHOS remains active and is essential for the survival and proliferation of NSCLC cells (22). Mitochondria play a pivotal role not only in energy production but also in biosynthesis, redox balance, and regulation of apoptosis (23). NSCLC cells exhibit remarkable metabolic flexibility, enabling them to switch between glycolysis and OXPHOS in response to environmental cues such as nutrient availability, oxygen levels, and therapeutic interventions (24). Figure 1 illustrates the dynamic interplay between these two energy-producing pathways and highlights the compensatory mechanisms that allow NSCLC cells to adapt their metabolism under stress conditions.

Figure 1. Integrated schematic of glycolysis, mitochondrial metabolism, and lipid pathways in NSCLC cells. This figure summarizes key metabolic processes in NSCLC cells, illustrating the interplay and compensatory mechanisms between glycolysis and OXPHOS, as well as the integration of lipid metabolism. Glucose is metabolized through glycolysis to produce pyruvate, which enters mitochondria to fuel the TCA cycle and OXPHOS for ATP production. Under certain conditions, pyruvate is diverted to lactate via LDHA. Glutamine contributes to TCA cycle intermediates through glutaminolysis, supporting biosynthesis and redox balance. Fatty acid metabolism is also reprogrammed in NSCLC: fatty acid synthesis, driven by enzymes such as ACC and FASN, converts citrate and acetyl-CoA into lipids, while FAO via CPT1a provides additional acetyl-CoA for mitochondrial respiration. Together, these interconnected pathways reflect the metabolic flexibility and adaptability of NSCLC cells in response to environmental and therapeutic pressures.
The metabolic adaptability contributes significantly to therapeutic resistance. For instance, when glycolysis is inhibited-either pharmacologically or due to nutrient scarcity-NSCLC cells can upregulate OXPHOS to meet their energy and biosynthetic demands (25). This compensatory increase in OXPHOS allows cancer cells to evade glycolysis-targeted therapies, highlighting the challenge of metabolic plasticity in effective cancer treatment (26). Additionally, some subpopulations of cancer stem cells within NSCLC have been found to rely heavily on OXPHOS, contributing to tumor heterogeneity and resistance to chemotherapy and radiotherapy (27).
Recent research has uncovered that oncogenic drivers common in NSCLC, such as mutations in the KRAS gene, can influence mitochondrial function. KRAS-mutant NSCLC cells demonstrate enhanced mitochondrial biogenesis and elevated OXPHOS activity, which supports their aggressive phenotype (28). Moreover, alterations in mitochondrial dynamics-processes that control mitochondrial fission and fusion-have been implicated in NSCLC progression. Dysregulated expression of proteins like dynamin-related protein 1 (DRP1) and mitofusins (MFN1 and MFN2) affects mitochondrial morphology and function, promoting cancer cell survival and metastasis (29). Targeting mitochondrial metabolism presents a promising therapeutic strategy. Inhibitors of OXPHOS components, such as complex I inhibitor IACS-010759, have shown antitumor activity in preclinical models of NSCLC by inducing energy stress and apoptosis (30). Furthermore, combining OXPHOS inhibitors with agents targeting glycolysis may overcome metabolic compensation mechanisms and enhance therapeutic efficacy (31). Agents that disrupt mitochondrial dynamics or promote mitochondrial dysfunction are also being explored as potential treatments (32).
The interplay between mitochondrial metabolism and the TME further complicates the metabolic landscape. Hypoxic regions within tumors can influence mitochondrial function and promote metabolic reprogramming (33). Additionally, interactions with stromal cells and immune cells can modulate mitochondrial activity in NSCLC cells, affecting tumor growth and response to therapy (34).
In summary, mitochondrial metabolism and OXPHOS play critical roles in NSCLC biology. Understanding the mechanisms underlying metabolic flexibility and mitochondrial function in cancer cells is essential for developing effective therapeutic strategies that can circumvent resistance and target the metabolic vulnerabilities of NSCLC.
2.3 Lipid metabolism: a new frontier
Lipid metabolism has emerged as a critical aspect of NSCLC biology, influencing tumor growth, survival, and metastasis (35). Cancer cells require a continuous supply of lipids for the synthesis of cellular membranes and signaling molecules that support rapid proliferation. Enhanced de novo lipogenesis—the endogenous production of fatty acids from non-lipid precursors like glucose and glutamine—is a hallmark of metabolic reprogramming in cancer (36), and is particularly prominent in NSCLC.
This process is depicted in Figure 1, which illustrates key pathways involved in lipid synthesis and oxidation in NSCLC cells. This process provides not only structural components for membrane biogenesis but also lipid signaling molecules that can activate oncogenic pathways. Enzymes such as fatty acid synthase (FASN) and stearoyl-CoA desaturase-1 (SCD1) play pivotal roles in lipid synthesis and are overexpressed in NSCLC (37). FASN is responsible for the synthesis of palmitate, a saturated fatty acid that serves as a building block for more complex lipids. Overexpression of FASN has been associated with increased tumor aggressiveness, resistance to chemotherapy, and poorer prognosis in NSCLC patients (38). Targeting FASN with small-molecule inhibitors has shown promise in preclinical models, leading to reduced tumor growth and enhanced sensitivity to other therapies (39). SCD1 introduces a double bond into saturated fatty acyl-CoAs to produce MUFAs, which are essential for maintaining membrane fluidity and function. Elevated SCD1 expression has been linked to enhanced tumor growth, metastasis, and reduced survival rates in NSCLC. Inhibition of SCD1 can disrupt membrane composition, induce endoplasmic reticulum (ER) stress, and trigger apoptosis in cancer cells (40). Acetyl-CoA carboxylase (ACC) is a key rate-limiting enzyme in fatty acid synthesis, playing a critical role in cellular metabolism and tumor growth by converting acetyl-CoA to malonyl-CoA. ACC regulates de novo fatty acid synthesis to meet the biosynthetic demands of tumor growth, and its inhibitor ND-646 significantly suppresses the growth and viability of NSCLC cells while enhancing the efficacy of chemotherapy, making it a potential target for cancer metabolism therapy (41).
Beyond lipid synthesis, NSCLC cells can utilize lipid oxidation through fatty acid oxidation (FAO) pathways to meet their energy demands and maintain redox balance (42). FAO involves the breakdown of fatty acids in mitochondria to generate acetyl-CoA, nicotinamide adenine dinucleotide (NADH), and flavin adenine dinucleotide hydrogen (FADH2), which feed into the tricarboxylic acid (TCA) cycle and electron transport chain to produce ATP (43). By relying on FAO, cancer cells can adapt to nutrient-deprived or hypoxic conditions where glycolysis may be less efficient (44). FAO also contributes to the maintenance of redox homeostasis by generating NADPH, a critical reducing agent that helps neutralize reactive oxygen species (ROS) and protect cells from oxidative stress (45). Enzymes like carnitine palmitoyltransferase 1 (CPT1), which regulates the transport of long-chain fatty acids into mitochondria, are often upregulated in NSCLC. Inhibition of CPT1 can impair FAO, leading to energy stress and increased sensitivity to oxidative damage (46).
NSCLC cells can enhance lipid uptake from the microenvironment by overexpressing lipid transporters such as cluster of differentiation 36 (CD36) and fatty acid-binding proteins (FABPs) (47). This uptake allows cancer cells to utilize exogenous fatty acids for energy production and membrane synthesis. Additionally, cancer cells can store excess lipids in lipid droplets, which serve as reservoirs that can be mobilized during times of metabolic stress (48). Alterations in lipid metabolism are often driven by oncogenic signaling pathways common in NSCLC. For example, activation of the PI3K/AKT/mTOR pathway can upregulate lipid synthesis by increasing the expression and activity of lipogenic enzymes (49). Mutations in KRAS, frequently observed in NSCLC, have been shown to enhance lipid metabolism, promoting tumor growth and survival (50). These signaling pathways not only stimulate lipid production but also integrate metabolic cues with cell proliferation and survival mechanisms.
2.4 Amino acid metabolism: beyond glutamine addiction
While glutamine metabolism is well-established in cancer biology due to its role in supporting rapid cell proliferation and survival (51), NSCLC cells also exploit other amino acids to meet their metabolic demands. Recent studies have highlighted alterations in the metabolism of amino acids such as serine, glycine, proline, and branched-chain amino acids (BCAAs), which contribute to nucleotide synthesis, redox balance, and energy production (52).
2.4.1 Serine and glycine metabolism
Serine and glycine are non-essential amino acids that play crucial roles in one-carbon metabolism, which is essential for nucleotide synthesis, methylation reactions, and antioxidant defense (53). NSCLC cells can upregulate enzymes involved in the serine-glycine synthesis pathway, such as phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase (PSAT1), and serine hydroxymethyltransferase (SHMT) (54). Overexpression of PHGDH has been observed in NSCLC and is associated with enhanced tumor growth and poor prognosis (55). Targeting this pathway can disrupt nucleotide biosynthesis and reduce the proliferation of cancer cells. Moreover, serine and glycine contribute to the synthesis of glutathione, a major intracellular antioxidant that helps maintain redox homeostasis (56). By elevating serine and glycine metabolism, NSCLC cells enhance their capacity to detoxify ROS, thereby promoting survival under oxidative stress conditions induced by therapies (57).
2.4.2 Proline metabolism
Proline metabolism is another pathway exploited by NSCLC cells to support tumor growth and metastasis (58). Proline biosynthesis from glutamate involves the enzyme pyrroline-5-carboxylate synthase (P5CS), while its degradation is mediated by proline dehydrogenase (PRODH). Proline can serve as a source of energy and contribute to redox balance by generating NADP+/NADPH (59). Altered proline metabolism aids in the adaptation of cancer cells to hypoxic conditions and nutrient deprivation, facilitating tumor progression (60). Inhibiting key enzymes in proline metabolism may impair cancer cell survival and sensitize tumors to treatment (61).
2.4.3 BCAAs
BCAAs-leucine, isoleucine, and valine-are essential amino acids involved in protein synthesis and signaling pathways that regulate cell growth and metabolism (62). NSCLC cells can exhibit increased uptake and catabolism of BCAAs to fuel the TCA cycle and provide nitrogen for nucleotide and amino acid synthesis (52). Enzymes such as branched-chain amino acid transaminase 1 (BCAT1) are upregulated in NSCLC and have been associated with tumor aggressiveness and poor clinical outcomes (63). Targeting BCAA metabolism may disrupt energy production and biosynthesis, leading to reduced tumor growth.
2.4.4 Amino acid transporters
To support increased amino acid demands, NSCLC cells often upregulate amino acid transporters. Transporters like solute carrier family 1 member 5 (SLC1A5/ASCT2) and SLC7A5 (LAT1) facilitate the uptake of glutamine, serine, leucine, and other amino acids (64, 65). Overexpression of these transporters has been linked to enhanced tumor growth, metastasis, and resistance to chemotherapy (66). Inhibiting amino acid transporters can reduce the intracellular availability of critical nutrients, inducing metabolic stress and apoptosis in cancer cells (67).
2.5 Metabolic heterogeneity and plasticity
NSCLC exhibit significant metabolic heterogeneity, both between different tumors (intertumoral heterogeneity) and within individual tumors (intratumoral heterogeneity)1. This heterogeneity arises from a complex interplay of genetic mutations, epigenetic modifications, tumor microenvironmental factors, and cellular interactions, leading to diverse metabolic phenotypes among cancer cells (68).
2.5.1 Genetic mutations and metabolic diversity
Genetic mutations commonly found in NSCLC, such as alterations in KRAS, EGFR, anaplastic lymphoma kinase (ALK), and liver kinase B1 (LKB1), drive distinct metabolic reprogramming in tumor cells (69). For instance, KRAS-mutant NSCLC cells often exhibit enhanced glucose uptake and glycolysis, whereas EGFR-mutant cells may rely more on glutamine metabolism (70). Loss of LKB1 function is associated with defects in mitochondrial OXPHOS and increased dependency on alternative energy sources (71). These genetic differences contribute to metabolic heterogeneity, influencing how tumor cells utilize nutrients and respond to metabolic stress.
2.5.2 Microenvironmental influences
The TME significantly impacts metabolic heterogeneity. Factors such as hypoxia, nutrient availability, pH changes, and interactions with stromal cells create spatial metabolic gradients within tumors (72). Hypoxic regions often lead to increased glycolysis and lactate production, while well-oxygenated areas may favor OXPHOS (73). Additionally, the availability of nutrients like glucose, amino acids, and lipids can vary within the tumor, forcing cancer cells to adapt their metabolism accordingly (74).
2.5.3 Interactions with stromal cells
CAFs, immune cells, and endothelial cells within the TME modulate cancer cell metabolism through paracrine signaling and direct cell-cell interactions (75). For example, CAFs can secrete metabolites such as lactate, amino acids, and fatty acids, which cancer cells uptake and utilize for energy and biosynthesis (10). Immune cells like tumor-associated macrophages (TAMs) produce cytokines that alter metabolic pathways in cancer cells, promoting survival and proliferation (76). These interactions further enhance metabolic diversity within the tumor.
2.5.4 Metabolic plasticity and therapeutic resistance
Metabolic heterogeneity contributes to therapeutic resistance by enabling subpopulations of cancer cells to survive under treatment-induced stress (77). Cancer cells with different metabolic profiles may respond variably to therapies targeting specific metabolic pathways (78). Metabolic plasticity-the ability of cancer cells to switch between metabolic states-allows them to adapt to environmental changes or therapeutic pressures (79). For instance, inhibiting glycolysis may lead some cancer cells to increase OXPHOS or utilize alternative substrates like fatty acids and amino acids (80).
2.5.5 Implications for personalized medicine
The presence of metabolic heterogeneity underscores the need for personalized metabolic interventions in NSCLC (81). Therapeutic strategies that consider the specific metabolic dependencies of a patient’s tumor may improve treatment efficacy (82). Techniques such as single-cell metabolomics and metabolic imaging can identify metabolic subtypes within tumors, guiding the selection of targeted therapies (83). Additionally, combining metabolic inhibitors with other treatments may overcome resistance by targeting multiple metabolic pathways simultaneously (84).
Understanding the mechanisms driving metabolic heterogeneity and plasticity is crucial for developing effective therapies. Integrating genomic, transcriptomic, and metabolomic data can provide a comprehensive view of tumor metabolism (85). Personalized approaches that tailor treatments based on individual metabolic profiles hold promise for improving outcomes in NSCLC patients.
3 The TME: metabolic crosstalk and therapeutic resistance
3.1 CAFs and metabolic support
CAFs are among the most abundant stromal cells within the TME of NSCLC and play a crucial role in supporting tumor metabolism and progression (86). CAFs undergo significant metabolic reprogramming that enables them to supply essential nutrients and metabolites to cancer cells, thereby promoting tumor growth and survival (51). One of the key phenomena illustrating this supportive role is the “reverse Warburg effect”. Unlike the traditional Warburg effect, where cancer cells preferentially utilize glycolysis for energy production even in the presence of oxygen, the reverse Warburg effect describes how CAFs enhance their glycolytic activity to produce high-energy metabolites such as lactate and pyruvate (87). These metabolites are then secreted into the TME and taken up by cancer cells, which utilize them through OXPHOS to generate ATP and support anabolic processes. This metabolic coupling allows cancer cells to conserve glucose for biosynthetic pathways, thus facilitating rapid proliferation and growth (88).
Beyond lactate production, CAFs secrete a variety of nutrients, including amino acids (e.g., glutamine, alanine) and fatty acids, which cancer cells can exploit. CAF-derived glutamine serves as an anaplerotic substrate replenishing TCA cycle intermediates in cancer cells, supporting energy production and biosynthesis of nucleotides and amino acids (89). Alanine secreted by CAFs can be converted into pyruvate, further fueling the TCA cycle. CAFs can release free fatty acids that cancer cells uptake and utilize for β-oxidation, contributing to ATP generation and membrane synthesis (90).
CAFs also modulate the ECM and secrete cytokines and growth factors that influence cancer cell metabolism and behavior (91). Factors such as transforming growth factor-beta (TGF-β), hepatocyte growth factor (HGF), and interleukins secreted by CAFs can activate signaling pathways (e.g., PI3K/AKT, MAPK) in cancer cells, leading to enhanced glycolysis and survival (92).
3.2 Immune cell metabolism and immune evasion
The metabolic state of immune cells within the TME profoundly affects their function and the overall immune response against NSCLC (93). Effective antitumor immunity relies on the activity of various immune cells, particularly effector T cells and natural killer (NK) cells, which require substantial energy and biosynthetic materials to proliferate and exert their cytotoxic functions (94). However, the TME is often characterized by metabolic competition and deprivation, as rapidly proliferating tumor cells consume large amounts of glucose, amino acids, and other nutrients (95). As illustrated in Figure 2, this metabolic imbalance not only limits nutrient availability for immune cells but also drives the reprogramming of both effector and regulatory immune cell populations, ultimately shaping the immune landscape toward either tumor suppression or immune evasion.

Figure 2. Immunometabolic interactions in the NSCLC TME.This schematic illustrates the dual roles of immune cell metabolism in either promoting or suppressing tumor growth in NSCLC. On the left, antitumor immunometabolism is driven by activated CD4+ and CD8+ T cells, NK cells, and other cytotoxic immune cells exhibiting increased glycolysis, fatty acid (FA) synthesis, OXPHOS, and glutaminolysis, supporting IFN-γ production, angiogenesis, and tumoricidal activity. On the right, tumor-promoting immunometabolism is characterized by immunosuppressive cells such as regulatory T cells (Treg cell), myeloid-derived suppressor cells (MDSCs), TAMs, and neutrophils, which exploit elevated glycolysis, lactic acid production, and altered metabolic intermediates (e.g., methylglyoxal) to inhibit cytotoxic function and promote Treg cell proliferation, PD-L1 expression, and M2 polarization. Tumor cells actively shape the metabolic landscape of the TME by rewiring their own metabolic programs (e.g., enhanced glycolysis, glutaminolysis, and β-oxidation) to suppress immune responses and sustain growth.
3.2.1 Metabolic competition and nutrient deprivation
Tumor cells alter the availability of key nutrients in the TME by upregulating glucose transporters and amino acid transporters, leading to increased uptake and consumption of glucose and amino acids (96). This metabolic competition results in a nutrient-deprived environment for immune cells. Effector T cells rely on glycolysis for energy production and effector functions such as cytokine production and proliferation. Glucose deprivation impairs T cell receptor (TCR) signaling, reduces cytokine production (e.g., interferon-gamma), and diminishes cytotoxic activity (97). Amino acids like glutamine, arginine, and tryptophan are critical for T cell function and proliferation (98). Tumor cells can deplete these amino acids or produce immunosuppressive metabolites (e.g., kynurenine from tryptophan catabolism via indoleamine 2,3-dioxygenase [IDO]) that inhibit T cell activity (99).
3.2.2 Metabolic checkpoints and immune suppression
Metabolic reprogramming in lung cancer plays a pivotal role in immune regulation by inducing metabolic stress and activating metabolic checkpoints in immune cells, leading to immunosuppression. Tumor-induced energy stress activates key metabolic sensors such as AMP-Activated Protein Kinase (AMPK), which modulates T cell metabolism and reduces their effector functions. Similarly, nutrient deprivation within the TME inhibits mTOR signaling, impairing T cell growth and responses. Additionally, hypoxia stabilizes HIFs, which alter immune cell metabolism and promote an immunosuppressive phenotype (100).
Tumor cells exploit these metabolic pathways to evade immune responses through several mechanisms. First, the upregulation of immune checkpoint molecules such as programmed cell death ligand 1 (PD-L1) on tumor cells interacts with programmed cell death protein 1 (PD-1) on T cells, suppressing glucose uptake and glycolysis in T cells, thereby diminishing their effector functions (101). Second, tumor cells secrete immunosuppressive factors like TGF-β and adenosine, which further modulate immune cell metabolism and suppress antitumor immunity. These metabolic interactions underscore the critical role of metabolic reprogramming in shaping the immune landscape of lung cancer, presenting potential targets for therapeutic intervention (102).
3.3 Hypoxia and metabolic adaptations
Hypoxia, or low oxygen conditions, is a hallmark of the TME in solid cancers, including NSCLC (103). Rapid tumor growth often outpaces the development of new blood vessels, leading to regions of insufficient oxygen supply. Hypoxic conditions trigger the stabilization of HIFs, particularly HIF-1α and HIF-2α, which are key transcription factors orchestrating cellular responses to low oxygen levels (104). Under normoxic conditions, HIF-α subunits are hydroxylated by prolyl hydroxylase domain proteins (PHDs), marking them for degradation via the von Hippel-Lindau (VHL) ubiquitin-proteasome pathway (105). Hypoxia inhibits PHD activity, preventing HIF-α degradation. Stabilized HIF-α translocates to the nucleus, dimerizes with HIF-1β, and activates the transcription of target genes involved in crucial processes such as metabolism, angiogenesis, erythropoiesis, and cell survival (106).
HIFs play a pivotal role in reprogramming cancer cell metabolism to adapt to hypoxic conditions (107). HIF-1α upregulates glycolytic enzymes, including HK2, phosphofructokinase 1 (PFK1), and lactate dehydrogenase A (LDHA), shifting the metabolic flux towards glycolysis despite the presence of oxygen (aerobic glycolysis) (108). This shift allows cancer cells to generate ATP efficiently under low oxygen conditions. Upregulation of glucose transporters such as GLUT1 enhances glucose uptake from the extracellular environment, providing substrates for glycolysis (109). HIF-1α induces pyruvate dehydrogenase kinase 1 (PDK1), which inhibits pyruvate dehydrogenase (PDH), decreasing the conversion of pyruvate to acetyl-CoA and thus reducing entry into the TCA cycle (110). This adaptation minimizes oxygen consumption and reduces ROS production from mitochondria. Increased LDHA activity converts pyruvate to lactate, which is exported out of the cell via monocarboxylate transporters (MCTs), contributing to the acidic TME (111).
4 Emerging technologies unveiling metabolic vulnerabilities
The rapid advancement of innovative technologies has significantly enhanced our ability to uncover metabolic vulnerabilities in cancer, providing new insights into tumor biology and therapeutic opportunities. Single-cell metabolomics has emerged as a powerful tool, enabling the analysis of metabolic heterogeneity at an unprecedented resolution (112). By profiling individual cells, researchers can identify distinct subpopulations with unique metabolic dependencies, which can inform the development of more precise and effective targeted therapies. Spatial metabolomics further expands this capability by detecting and imaging metabolites with spatial resolution at the tissue or cellular level (113). By combining mass spectrometry imaging with traditional metabolomics approaches, this technique allows for the visualization of metabolite distribution within biological tissues, offering a deeper understanding of the TME and its metabolic interactions.
The advent of high-resolution mass spectrometry (HRMS) has significantly enhanced the sensitivity and accuracy of metabolite detection. HRMS provides precise molecular weight measurements and detailed structural information, making it a cornerstone technology in metabolomics research (114). Finally, systems biology and computational modeling integrate multi-omics data to construct metabolic networks and predict therapeutic outcomes (115). Computational tools enable the simulation of metabolic interventions and identification of synergistic drug combinations, paving the way for more strategic and effective treatment regimens. Together, these emerging technologies are revolutionizing our understanding of cancer metabolism and driving the discovery of novel therapeutic strategies.
5 Therapeutic implications and strategies
Recent advances in elucidating the metabolic landscape of NSCLC have highlighted several key pathways that are amenable to therapeutic intervention. Targeting metabolic dependencies represents a promising strategy to disrupt tumor growth and overcome resistance mechanisms (Figure 3). In this section, we discuss emerging therapeutic approaches aimed at modulating core metabolic processes in NSCLC. A summary of representative metabolic inhibitors and their targeted pathways is provided in Table 1, which offers a comprehensive overview of current therapeutic agents explored in NSCLC metabolic intervention.

Figure 3. Targetable metabolic pathways and inhibitors in NSCLC.This schematic highlights key metabolic pathways reprogrammed in NSCLC cells and outlines current therapeutic targets under investigation. Central carbon metabolism—including glycolysis, the pentose phosphate pathway (PPP), TCA cycle, and OXPHOS—is regulated by oncogenic signaling pathways such as PI3K/mTOR and MAPK/ERK, often activated by mutations in receptor tyrosine kinases (EGFR, ALK, MET). Metabolic enzymes (e.g., HK2, PFKFB3, LDHA, PHGDH, PKM2, ACLY, FASN, SCD, GLS1) and transporters (e.g., SLC1A5/SLC7A5) are shown with corresponding small-molecule inhibitors (e.g., 2-DG, BAY-876, PFK158, Oxamate, PX-478, CB-839, TVB2640, MF-438, JPH203), many of which are being evaluated in preclinical or clinical settings. The diagram also illustrates amino acid metabolism (e.g., glutaminolysis, serine biosynthesis, arginine and proline metabolism), emphasizing the metabolic plasticity of NSCLC and the opportunities for combinatorial metabolic interventions. This comprehensive map provides a framework for rational design of therapies targeting metabolic vulnerabilities in NSCLC.
5.1 Targeting glycolysis and OXPHOS
The metabolic reprogramming of NSCLC cells presents a strategic opportunity for therapeutic intervention by targeting key energy-producing pathways. Glycolysis and OXPHOS are central to cancer cell metabolism, providing ATP and metabolic intermediates necessary for rapid proliferation and survival. Inhibitors targeting these pathways have shown promise in preclinical models, but due to metabolic plasticity, cancer cells can switch between glycolysis and OXPHOS to compensate when one pathway is inhibited (129). Therefore, combination therapies targeting both pathways may be more effective in preventing metabolic compensation and inducing cancer cell death.
Key glycolytic enzymes such as HK2 and PKM2 are overexpressed in NSCLC and are critical for maintaining the high glycolytic flux observed in cancer cells. HK2 catalyzes the first step of glycolysis, phosphorylating glucose to glucose-6-phosphate. Inhibitors like 2-deoxy-D-glucose (2-DG) mimic glucose but cannot undergo further metabolism, effectively inhibiting HK2 activity (130). Preclinical studies have shown that 2-DG induces apoptosis and enhances the sensitivity of cancer cells to chemotherapy and radiotherapy (116). However, its clinical application is limited by toxicity and lack of specificity. PKM2 controls the final step of glycolysis, converting phosphoenolpyruvate to pyruvate. PKM2 exists in both active tetrameric and less active dimeric forms, with cancer cells favoring the dimeric form to promote anabolic processes. Small-molecule activators like TEPP-46 and DASA-58 stabilize the tetrameric form, enhancing pyruvate production and reducing lactate formation (117, 118). This shift can suppress tumor growth and induce apoptosis.
Cancer cells can compensate for inhibited glycolysis by increasing reliance on mitochondrial OXPHOS. Targeting OXPHOS can disrupt this adaptive mechanism: 1. Complex I Inhibitors: Metformin and phenformin inhibit mitochondrial complex I, reducing ATP production and increasing ROS generation (120, 121). Phenformin has shown greater antitumor activity than metformin in preclinical models of NSCLC but with a higher risk of lactic acidosis (131). 2. ATP Synthase Inhibitors: Oligomycin and its derivatives inhibit ATP synthase, leading to decreased ATP production (122). While effective in vitro, their clinical use is limited due to toxicity in normal cells. 3. B-cell lymphoma-2/-xL (Bcl-2/Bcl-xL) Inhibitors: Agents like venetoclax target anti-apoptotic proteins located on the mitochondrial membrane, promoting apoptosis in cancer cells (123). Combining these with OXPHOS inhibitors can enhance cancer cell death.
Combining glycolysis inhibitors with OXPHOS inhibitors may prevent cancer cells from switching energy sources and enhance therapeutic efficacy. Simultaneous targeting of HK2 and complex I can induce energetic crisis in cancer cells. For example, combining 2-DG with metformin has shown synergistic effects in reducing NSCLC cell viability (132). Inhibiting key enzymes in both pathways limits the ability of cancer cells to adapt metabolically. This approach can lead to increased ROS production, DNA damage, and activation of cell death pathways.
5.2 Targeting lipid metabolism
Lipid metabolism plays a crucial role in the growth and survival of NSCLC cells, as previously discussed. Targeting key enzymes involved in lipid synthesis and desaturation presents a promising therapeutic strategy. FASN and SCD1 are two pivotal enzymes in lipid metabolism that have gained significant attention as potential targets for cancer therapy.
FASN is an essential enzyme responsible for the de novo synthesis of long-chain fatty acids from acetyl-CoA and malonyl-CoA precursors. Overexpression of FASN has been observed in various cancers, including NSCLC, and is associated with poor prognosis and aggressive tumor behavior (133). By promoting lipid synthesis, FASN supports membrane biogenesis, energy storage, and the production of lipid signaling molecules that facilitate tumor growth and metastasis (134). TVB-2640 is a first-in-class, orally bioavailable, small-molecule inhibitor of FASN that has entered clinical trials. It selectively inhibits the enzymatic activity of FASN, leading to reduced fatty acid synthesis and accumulation of malonyl-CoA, which can induce apoptosis and inhibit tumor growth (124). In preclinical studies, TVB-2640 demonstrated significant antitumor activity in NSCLC models, both as a monotherapy and in combination with other agents (125). Clinical trials are currently evaluating the safety and efficacy of TVB-2640 in patients with advanced solid tumors, including NSCLC (124). Preliminary results have shown that TVB-2640 is well-tolerated and exhibits antitumor activity, especially when combined with other treatments such as chemotherapy or targeted therapies. These findings suggest that inhibiting FASN can disrupt lipid homeostasis in cancer cells, leading to growth inhibition and enhanced sensitivity to other anticancer agents.
SCD1 is a rate-limiting enzyme in the synthesis of monounsaturated fatty acids (MUFAs) from saturated fatty acids (SFAs). MUFAs are critical components of cellular membranes and play a role in lipid signaling and energy storage (126). Overexpression of SCD1 has been linked to increased proliferation, survival, and chemoresistance in NSCLC cells (135). Targeting SCD1 disrupts the balance of saturated and unsaturated fatty acids, affecting membrane fluidity and function. Inhibition of SCD1 leads to the accumulation of SFAs, which can cause ER stress and activate the unfolded protein response (UPR) (136). Prolonged ER stress can trigger apoptosis in cancer cells. Additionally, decreased levels of MUFAs impair membrane synthesis and the formation of lipid rafts, which are essential for signal transduction and the activation of oncogenic pathways. Several small-molecule inhibitors of SCD1 have been developed and shown to exhibit antitumor activity in preclinical models of NSCLC. For example, A939572 and MF-438 are potent SCD1 inhibitors that have demonstrated the ability to reduce tumor cell proliferation and induce apoptosis (126, 127). In combination with other treatments, such as chemotherapy or targeted therapies, SCD1 inhibitors may enhance therapeutic efficacy by sensitizing cancer cells to these agents. While targeting lipid metabolism shows promise, clinical outcomes have been variable. Some studies suggest that inhibiting FAO may impair T cell function and exacerbate inflammation, raising concerns about off-target effects. Moreover, conflicting evidence exists regarding the dependency of certain NSCLC subtypes on FAO versus lipogenesis, underscoring the need for subtype-specific therapeutic strategies.
5.3 Targeting amino acid metabolism
Amino acid metabolism plays a pivotal role in the growth and survival of NSCLC cells. Targeting key enzymes and pathways involved in amino acid utilization presents a promising therapeutic strategy. Glutaminase (GLS) inhibitors, such as CB-839 (telaglenastat), and inhibitors of serine and glycine synthesis pathways have shown potential in impairing tumor growth by disrupting critical metabolic processes (128). Glutamine is an essential nutrient for rapidly proliferating cancer cells, serving as a carbon and nitrogen source for nucleotide and amino acid synthesis, as well as maintaining redox balance through glutathione production. GLS catalyzes the conversion of glutamine to glutamate, a key step in glutamine metabolism. Overexpression of GLS has been observed in NSCLC and is associated with increased tumor aggressiveness (137). CB-839 is an orally bioavailable, selective GLS inhibitor that has demonstrated antitumor activity in preclinical models of NSCLC by blocking glutamine utilization (138). By inhibiting GLS, CB-839 reduces the production of glutamate and downstream metabolites, leading to impaired nucleotide synthesis, decreased glutathione levels, and increased oxidative stress (139). This can result in cancer cell death and reduced tumor growth.
Serine and glycine are non-essential amino acids integral to one-carbon metabolism, which is crucial for nucleotide synthesis, methylation reactions, and maintaining redox balance (54). NSCLC cells often upregulate enzymes involved in the serine-glycine synthesis pathway to meet the increased demands of rapid proliferation. PHGDH catalyzes the first step in the de novo serine synthesis pathway. Overexpression of PHGDH has been observed in NSCLC and is associated with enhanced tumor growth (140). Inhibitors targeting PHGDH can disrupt serine production, impair nucleotide biosynthesis, and induce cell cycle arrest. Novel PHGDH inhibitors have shown efficacy in preclinical models, reducing tumor growth and enhancing sensitivity to chemotherapy (141). SHMT converts serine to glycine, contributing to one-carbon units necessary for thymidine and purine synthesis (142). Inhibiting SHMT disrupts DNA synthesis and can induce apoptosis in cancer cells. Agents targeting SHMT have demonstrated antitumor activity by inducing DNA damage and impairing cell proliferation (143).
Proline metabolism is another pathway exploited by NSCLC cells to support tumor growth and metastasis (144). Proline biosynthesis and degradation are linked to energy production and redox balance. Inhibiting key enzymes such as pyrroline-5-carboxylate reductase (PYCR) can disrupt proline metabolism, leading to increased oxidative stress and reduced tumor growth (145). Targeting proline metabolism may also impair the survival of cancer stem cells, which are often resistant to conventional therapies (146).
BCAAs-leucine, isoleucine, and valine-are essential amino acids involved in protein synthesis and signaling pathways that regulate cell growth (147). NSCLC cells may exhibit increased uptake and catabolism of BCAAs to fuel the TCA cycle and provide nitrogen for nucleotide and amino acid synthesis (148). BCAT1 catalyzes the first step in BCAA catabolism. Overexpression of BCAT1 has been associated with tumor aggressiveness and poor prognosis in NSCLC (149). Inhibiting BCAT1 can impair BCAA metabolism, suppress tumor growth, and reduce cancer cell proliferation. To support increased amino acid demands, NSCLC cells often upregulate amino acid transporters (150). Targeting these transporters can reduce the uptake of critical nutrients: ASCT2 is a glutamine transporter overexpressed in many cancers (151). Inhibiting ASCT2 can decrease glutamine uptake, leading to metabolic stress and sensitizing cancer cells to chemotherapy (152). LAT1 transports large neutral amino acids, including leucine (153). Targeting LAT1 can disrupt mTOR signaling pathways, reduce protein synthesis, and inhibit tumor growth (65). Similarly, targeting amino acid metabolism is not without limitations. For instance, while LAT1 inhibitors show antitumor potential, they may also impact immune cell metabolism or lead to resistance via transporter redundancy. Furthermore, some clinical trials targeting amino acid pathways have failed to show significant benefits, highlighting the complexity and redundancy of metabolic networks in cancer.
5.4 Combination therapies and immunometabolism
Combining metabolic inhibitors with immunotherapies has emerged as a promising strategy to enhance antitumor immune responses in NSCLC (154). Modulating tumor metabolism can improve the function of immune cells within the TME, representing a synergistic approach to NSCLC treatment. Tumor cells often create a metabolically hostile environment for immune cells by consuming large amounts of glucose and amino acids, leading to nutrient deprivation for tumor-infiltrating lymphocytes (TILs) (155). Additionally, tumor cells produce immunosuppressive metabolites like lactate and adenosine, which inhibit immune cell function (156). By targeting tumor metabolism, it is possible to alleviate these immunosuppressive conditions and enhance the efficacy of immunotherapies. Inhibiting glycolysis in tumor cells can reduce glucose competition, making it more available for effector T cells that rely on glycolysis for their function (157). Glycolytic inhibitors, such as 2-DG, may improve T-cell activity when combined with immune checkpoint inhibitors (116). IDO is an enzyme overexpressed in some tumors that depletes tryptophan and produces immunosuppressive metabolites (158). IDO inhibitors can restore tryptophan levels and enhance T-cell proliferation. Combining IDO inhibitors with PD-1/PD-L1 blockade has shown synergistic antitumor effects in preclinical models (159). As discussed previously, GLS inhibitors like CB-839 can reduce glutamine availability for tumor cells (128). Since glutamine is less critical for T-cell function than for tumor cells, GLS inhibition may preferentially affect cancer cells and improve immune responses.
Modulating the metabolism of immune cells themselves can also enhance antitumor immunity. The mTOR pathway regulates T-cell metabolism and function (160). Activating mTOR can promote T-cell glycolysis and effector functions. Agents that enhance mTOR signaling in T cells may boost their antitumor activity. AMPK activation can improve the metabolic fitness of T cells, enhancing their survival and function in the nutrient-deprived TME (161).
Combining metabolic inhibitors with immunotherapies aims to create a more favorable metabolic environment for immune cells while directly targeting tumor metabolism. Targeting metabolic checkpoints in tumor cells can sensitize them to immune-mediated killing. For example, inhibiting LDHA reduces lactate production, alleviating acid-mediated immunosuppression (119). Combining immune checkpoint inhibitors (e.g., anti-PD-1/PD-L1 antibodies) with metabolic modulators can enhance T-cell infiltration and activity. Clinical trials are exploring such combinations in NSCLC patients. Epacadostat, an IDO inhibitor, has been evaluated in combination with pembrolizumab (anti-PD-1) in clinical trials, showing promising results in some cancer types (162). However, results have been mixed, and further studies are needed to determine efficacy in NSCLC. Metformin, a complex I inhibitor, has immunomodulatory effects and may enhance responses to immunotherapy (132). Retrospective studies suggest that NSCLC patients taking metformin may have improved outcomes with immune checkpoint inhibitors.
5.5 Personalized metabolic therapies
The heterogeneity of metabolic profiles among NSCLC tumors underscores the need for personalized metabolic therapies (163). Metabolic profiling of tumors can identify patient-specific metabolic dependencies, enabling tailored treatment strategies that target the unique metabolic vulnerabilities of each tumor (164). Advanced technologies such as metabolomics, genomics, transcriptomics, and proteomics allow for comprehensive metabolic profiling of tumors. High-throughput techniques can analyze metabolic enzyme expression levels, metabolite concentrations, and metabolic fluxes within cancer cells (165). These high-throughput techniques include: 1. Mass Spectrometry (MS) and Nuclear Magnetic Resonance (NMR) Spectroscopy: These techniques enable the identification and quantification of a wide range of metabolites in tumor samples. 2. Positron Emission Tomography (PET) Imaging: Metabolic imaging using tracers like 18F-fluorodeoxyglucose (FDG) can assess glucose uptake in tumors, providing insights into glycolytic activity. 3. Gene Expression Profiling: Analyzing the expression of genes involved in metabolic pathways can reveal overactive or dysregulated metabolic enzymes.
6 Challenges and future directions
Cancer cells’ ability to adapt metabolically poses a significant challenge for metabolic therapies. Their metabolic plasticity enables them to switch between different energy sources and metabolic pathways, which can render single-agent treatments less effective. Future research should focus on understanding these resistance mechanisms and developing strategies to prevent or overcome them. Ensuring the safety and selectivity of metabolic inhibitors is also critical; these agents must selectively target cancer cells without harming normal tissues. Strategies to achieve this include exploiting cancer-specific metabolic pathways or employing delivery systems that preferentially target tumor cells.
Integrating metabolic biomarkers into clinical practice is essential for personalizing treatment and improving outcomes. Standardizing these biomarkers and incorporating them into clinical workflows require close collaboration between researchers and clinicians. Validation in large, diverse patient cohorts is necessary to ensure their reliability and effectiveness in clinical settings. Advancing the field further necessitates interdisciplinary collaboration among oncologists, biochemists, pharmacologists, and computational biologists. Integrating diverse expertise will accelerate the translation of metabolic discoveries into effective therapies, ultimately enhancing treatment strategies for NSCLC.
7 Conclusion
The metabolic reprogramming of NSCLC cells extends far beyond the Warburg effect, encompassing alterations in lipid and amino acid metabolism and dynamic interactions with the TME. These metabolic adaptations are not merely bystanders but are integral to tumor growth, survival, and therapeutic resistance. By leveraging emerging technologies and a deeper understanding of metabolic heterogeneity, we can identify novel vulnerabilities in NSCLC. Therapeutic strategies that target these metabolic dependencies offer promising avenues for improving patient outcomes. Combining metabolic inhibitors with existing treatments, such as immunotherapies and targeted therapies, may enhance efficacy and overcome resistance. Personalized metabolic therapies, guided by metabolic profiling, represent the next frontier in precision oncology. As we continue to unravel the complexities of NSCLC metabolism, we move closer to realizing the full potential of metabolic targeting in cancer therapy. The integration of cutting-edge research with clinical practice holds the promise of transforming NSCLC management and improving the lives of patients worldwide.
Author contributions
HC: Investigation, Writing – original draft. FZ: Investigation, Writing – original draft. FX: Investigation, Writing – review & editing. CY: Conceptualization, Funding acquisition, Project administration, Supervision, Writing – review & editing.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by a grant from the Dalian Science and Technology Innovation Fund Program (No. 2024JJ13PT070) and United Foundation for Dalian Institute of Chemical Physics, Chinese Academy of Sciences and the Second Hospital of Dalian Medical University (No. DMU-2&DICP UN202410), and National Natural Science Foundation of China (No. 82002211).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Thai AA, Solomon BJ, Sequist LV, Gainor JF, and Heist RS. Lung cancer. Lancet. (2021) 398:535–54. doi: 10.1016/S0140-6736(21)00312-3
2. Megyesfalvi Z, Gay CM, Popper H, Pirker R, Ostoros G, Heeke S, et al. Clinical insights into small cell lung cancer: Tumor heterogeneity, diagnosis, therapy, and future directions. CA Cancer J Clin. (2023) 73:620–52. doi: 10.3322/caac.21785
3. Siegel RL, Miller KD, Fuchs HE, and Jemal A. Cancer statistics, 2021. CA Cancer J Clin. (2021) 71:7–33. doi: 10.3322/caac.21654
4. Chaft JE, Shyr Y, Sepesi B, and Forde PM. Preoperative and postoperative systemic therapy for operable non-small-cell lung cancer. J Clin Oncol. (2022) 40:546–55. doi: 10.1200/JCO.21.01589
5. Pavlova NN, Zhu J, and Thompson CB. The hallmarks of cancer metabolism: Still emerging. Cell Metab. (2022) 34:355–77. doi: 10.1016/j.cmet.2022.01.007
6. Martínez-Reyes I and Chandel NS. Cancer metabolism: looking forward. Nat Rev Cancer. (2021) 21:669–80. doi: 10.1038/s41568-021-00378-6
7. Warburg O. On the origin of cancer cells. Science. (1956) 123:309–14. doi: 10.1126/science.123.3191.309
8. Faubert B, Solmonson A, and DeBerardinis RJ. Metabolic reprogramming and cancer progression. Science. (2020) 368:eaaw5473. doi: 10.1126/science.aaw5473
9. Salcher S, Sturm G, Horvath L, Untergasser G, Kuempers C, Fotakis G, et al. High-resolution single-cell atlas reveals diversity and plasticity of tissue-resident neutrophils in non-small cell lung cancer. Cancer Cell. (2022) 40:1503–1520.e8. doi: 10.1016/j.ccell.2022.10.008
10. Li Z, Sun C, and Qin Z. Metabolic reprogramming of cancer-associated fibroblasts and its effect on cancer cell reprogramming. Theranostics. (2021) 11:8322–36. doi: 10.7150/thno.62378
11. Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. (2021) 20:131. doi: 10.1186/s12943-021-01428-1
12. Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, et al. The cancer metabolic reprogramming and immune response. Mol Cancer. (2021) 20:28. doi: 10.1186/s12943-021-01316-8
13. Jing X, Yang F, Shao C, Wei K, Xie M, Shen H, et al. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol Cancer. (2019) 18:157. doi: 10.1186/s12943-019-1089-9
14. Vander Heiden MG, Cantley LC, and Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. (2009) 324:1029–33. doi: 10.1126/science.1160809
15. Liang LJ, Yang FY, Wang D, Zhang YF, Yu H, Wang Z, et al. CIP2A induces PKM2 tetramer formation and oxidative phosphorylation in non-small cell lung cancer. Cell Discov. (2024) 10:13. doi: 10.1038/s41421-023-00633-0
16. Gonzalez MA, Lu DR, Yousefi M, Kroll A, Lo CH, Briseño CG, et al. Phagocytosis increases an oxidative metabolic and immune suppressive signature in tumor macrophages. J Exp Med. (2023) 220:e20221472. doi: 10.1084/jem.20221472
17. Rao S, Mondragón L, Pranjic B, Hanada T, Stoll G, Köcher T, et al. AIF-regulated oxidative phosphorylation supports lung cancer development. Cell Res. (2019) 29:579–91. doi: 10.1038/s41422-019-0181-4
18. Zhou Y, Guo Y, Ran M, Shan W, Granchi C, Giovannetti E, et al. Combined inhibition of pyruvate dehydrogenase kinase 1 and lactate dehydrogenase A induces metabolic and signaling reprogramming and enhances lung adenocarcinoma cell killing. Cancer Lett. (2023) 577:216425. doi: 10.1016/j.canlet.2023.216425
19. Ge W, Wang Y, Quan M, Mao T, Bischof EY, Xu H, et al. Activation of the PI3K/AKT signaling pathway by ARNTL2 enhances cellular glycolysis and sensitizes pancreatic adenocarcinoma to erlotinib. Mol Cancer. (2024) 23:48. doi: 10.1186/s12943-024-01965-5
20. Sun S, Zhang Y, Xu W, Yang R, Yang Y, Guo J, et al. Plumbagin reduction by thioredoxin reductase 1 possesses synergy effects with GLUT1 inhibitor on KEAP1-mutant NSCLC cells. BioMed Pharmacother. (2022) 146:112546. doi: 10.1016/j.biopha.2021.112546
21. Huang Y, Chen Z, Lu T, Bi G, Li M, Liang J, et al. HIF-1α switches the functionality of TGF-β signaling via changing the partners of smads to drive glucose metabolic reprogramming in non-small cell lung cancer. J Exp Clin Cancer Res. (2021) 40:398. doi: 10.1186/s13046-021-02188-y
22. Sainero-Alcolado L, Liaño-Pons J, Ruiz-Pérez MV, and Arsenian-Henriksson M. Targeting mitochondrial metabolism for precision medicine in cancer. Cell Death Differ. (2022) 29:1304–17. doi: 10.1038/s41418-022-01022-y
23. Porporato PE, Filigheddu N, Pedro JMB, Kroemer G, and Galluzzi L. Mitochondrial metabolism and cancer. Cell Res. (2018) 28:265–80. doi: 10.1038/cr.2017.155
24. Ashton TM, McKenna WG, Kunz-Schughart LA, and Higgins GS. Oxidative phosphorylation as an emerging target in cancer therapy. Clin Cancer Res. (2018) 24:2482–90. doi: 10.1158/1078-0432.CCR-17-3070
25. Lin Z, Li J, Zhang J, Feng W, Lu J, Ma X, et al. Metabolic reprogramming driven by IGF2BP3 promotes acquired resistance to EGFR inhibitors in non-small cell lung cancer. Cancer Res. (2023) 83:2187–207. doi: 10.1158/0008-5472.CAN-22-3059
26. Thirusangu P, Ray U, Sarkar Bhattacharya S, Oien DB, Jin L, Staub J, et al. PFKFB3 regulates cancer stemness through the Hippo pathway in small cell lung carcinoma. Oncogene. (2022) 41:4003–17. doi: 10.1038/s41388-022-02391-x
27. Han M, Bushong EA, Segawa M, Tiard A, Wong A, Brady MR, et al. Spatial mapping of mitochondrial networks and bioenergetics in lung cancer. Nature. (2023) 615:712–9. doi: 10.1038/s41586-023-05793-3
28. Feng J, Lian Z, Xia X, Lu Y, Hu K, Zhang Y, et al. Targeting metabolic vulnerability in mitochondria conquers MEK inhibitor resistance in KRAS-mutant lung cancer. Acta Pharm Sin B. (2023) 13:1145–63. doi: 10.1016/j.apsb.2022.10.023
29. Rehman J, Zhang HJ, Toth PT, Zhang Y, Marsboom G, Hong Z, et al. Inhibition of mitochondrial fission prevents cell cycle progression in lung cancer. FASEB J. (2012) 26:2175–86. doi: 10.1096/fj.11-196543
30. Chen D, Barsoumian HB, Fischer G, Yang L, Verma V, Younes AI, et al. Combination treatment with radiotherapy and a novel oxidative phosphorylation inhibitor overcomes PD-1 resistance and enhances antitumor immunity. J Immunother Cancer. (2020) 8:e000289. doi: 10.1136/jitc-2019-000289
31. Lissanu Deribe Y, Sun Y, Terranova C, Khan F, Martinez-Ledesma J, Gay J, et al. Mutations in the SWI/SNF complex induce a targetable dependence on oxidative phosphorylation in lung cancer. Nat Med. (2018) 24:1047–57. doi: 10.1038/s41591-018-0019-5
32. Zhang J, Qiao W, and Luo Y. Mitochondrial quality control proteases and their modulation for cancer therapy. Med Res Rev. (2023) 43:399–436. doi: 10.1002/med.21929
33. Joshi SR, Kitagawa A, Jacob C, Hashimoto R, Dhagia V, Ramesh A, et al. Hypoxic activation of glucose-6-phosphate dehydrogenase controls the expression of genes involved in the pathogenesis of pulmonary hypertension through the regulation of DNA methylation. Am J Physiol Lung Cell Mol Physiol. (2020) 318:L773–86. doi: 10.1152/ajplung.00001.2020
34. Parma B, Wurdak H, and Ceppi P. Harnessing mitochondrial metabolism and drug resistance in non-small cell lung cancer and beyond by blocking heat-shock proteins. Drug Resist Updat. (2022) 65:100888. doi: 10.1016/j.drup.2022.100888
35. Lien EC, Westermark AM, Zhang Y, Yuan C, Li Z, Lau AN, et al. Low glycaemic diets alter lipid metabolism to influence tumour growth. Nature. (2021) 599:302–7. doi: 10.1038/s41586-021-04049-2
36. Bartolacci C, Andreani C, Vale G, Berto S, Melegari M, Crouch AC, et al. Targeting de novo lipogenesis and the Lands cycle induces ferroptosis in KRAS-mutant lung cancer. Nat Commun. (2022) 13:4327. doi: 10.1038/s41467-022-31963-4
37. Al-Jawadi A, Patel CR, Shiarella RJ, Romelus E, Auvinen M, Guardia J, et al. Cell-type-specific, ketohexokinase-dependent induction by fructose of lipogenic gene expression in mouse small intestine. J Nutr. (2020) 150:1722–30. doi: 10.1093/jn/nxaa113
38. Lyu Q, Zhu W, Wei T, Ding W, Cao M, Wang Q, et al. High mutations in fatty acid metabolism contribute to a better prognosis of small-cell lung cancer patients treated with chemotherapy. Cancer Med. (2021) 10:7863–76. doi: 10.1002/cam4.4290
39. Cao D, Yang J, Deng Y, Su M, Wang Y, Feng X, et al. Discovery of a mammalian FASN inhibitor against xenografts of non-small cell lung cancer and melanoma. Signal Transduct Target Ther. (2022) 7:273. doi: 10.1038/s41392-022-01099-4
40. Wohlhieter CA, Richards AL, Uddin F, Hulton CH, Quintanal-Villalonga À, Martin A, et al. Concurrent mutations in STK11 and KEAP1 promote ferroptosis protection and SCD1 dependence in lung cancer. Cell Rep. (2020) 33:108444. doi: 10.1016/j.celrep.2020.108444
41. Svensson RU, Parker SJ, Eichner LJ, Kolar MJ, Wallace M, Brun SN, et al. Inhibition of acetyl-CoA carboxylase suppresses fatty acid synthesis and tumor growth of non-small-cell lung cancer in preclinical models. Nat Med. (2016) 22:1108–19. doi: 10.1038/nm.4181
42. Peng F, Lu J, Su K, Liu X, Luo H, He B, et al. Oncogenic fatty acid oxidation senses circadian disruption in sleep-deficiency-enhanced tumorigenesis. Cell Metab. (2024) 36:1598–1618.e11. doi: 10.1016/j.cmet.2024.04.018
43. Ma Y, Temkin SM, Hawkridge AM, Guo C, Wang W, Wang XY, et al. Fatty acid oxidation: An emerging facet of metabolic transformation in cancer. Cancer Lett. (2018) 435:92–100. doi: 10.1016/j.canlet.2018.08.006
44. Nimmakayala RK, Leon F, Rachagani S, Rauth S, Nallasamy P, Marimuthu S, et al. Metabolic programming of distinct cancer stem cells promotes metastasis of pancreatic ductal adenocarcinoma. Oncogene. (2021) 40:215–31. doi: 10.1038/s41388-020-01518-2
45. Gao Y, Song Z, Jia L, Tang Y, Wang C, Zhao X, et al. Self-amplified ROS production from fatty acid oxidation enhanced tumor immunotherapy by atorvastatin/PD-L1 siRNA lipopeptide nanoplexes. Biomaterials. (2022) 291:121902. doi: 10.1016/j.biomaterials.2022.121902
46. Yan R, Zheng C, Qian S, Li K, Kong X, and Liao S. The ZNF263/CPT1B axis regulates fatty acid β-oxidation to affect cisplatin resistance in lung adenocarcinoma. Pharmacogenomics J. (2024) 24:33. doi: 10.1038/s41397-024-00355-w
47. Liu LZ, Wang B, Zhang R, Wu Z, Huang Y, Zhang X, et al. The activated CD36-Src axis promotes lung adenocarcinoma cell proliferation and actin remodeling-involved metastasis in high-fat environment. Cell Death Dis. (2023) 14:548. doi: 10.1038/s41419-023-06078-3
48. Gong Z, Li Q, Shi J, Liu ET, Shultz LD, and Ren G. Lipid-laden lung mesenchymal cells foster breast cancer metastasis via metabolic reprogramming of tumor cells and natural killer cells. Cell Metab. (2022) 34:1960–1976.e9. doi: 10.1016/j.cmet.2022.11.003
49. Chen J, Zhao R, Wang Y, Xiao H, Lin W, Diao M, et al. G protein-coupled estrogen receptor activates PI3K/AKT/mTOR signaling to suppress ferroptosis via SREBP1/SCD1-mediated lipogenesis. Mol Med. (2024) 30:28. doi: 10.1186/s10020-023-00763-x
50. Anthiya S, Öztürk SC, Yanik H, Tavukcuoglu E, Şahin A, Datta D, et al. Targeted siRNA lipid nanoparticles for the treatment of KRAS-mutant tumors. J Control Release. (2023) 357:67–83. doi: 10.1016/j.jconrel.2023.03.016
51. Liu T, Han C, Fang P, Ma Z, Wang X, Chen H, et al. Cancer-associated fibroblast-specific lncRNA LINC01614 enhances glutamine uptake in lung adenocarcinoma. J Hematol Oncol. (2022) 15:141. doi: 10.1186/s13045-022-01359-4
52. Mayers JR, Torrence ME, Danai LV, Papagiannakopoulos T, Davidson SM, Bauer MR, et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science. (2016) 353:1161–5. doi: 10.1126/science.aaf5171
53. Rachedi NS, Tang Y, Tai YY, Zhao J, Chauvet C, Grynblat J, et al. Dietary intake and glutamine-serine metabolism control pathologic vascular stiffness. Cell Metab. (2024) 36:1335–1350.e8. doi: 10.1016/j.cmet.2024.04.010
54. DeNicola GM, Chen PH, Mullarky E, Sudderth JA, Hu Z, Wu D, et al. NRF2 regulates serine biosynthesis in non-small cell lung cancer. Nat Genet. (2015) 47:1475–81. doi: 10.1038/ng.3421
55. Zhu J, Ma J, Wang X, Ma T, Zhang S, Wang W, et al. High expression of PHGDH predicts poor prognosis in non-small cell lung cancer. Transl Oncol. (2016) 9:592–9. doi: 10.1016/j.tranon.2016.08.003
56. He L, Endress J, Cho S, Li Z, Zheng Y, Asara JM, et al. Suppression of nuclear GSK3 signaling promotes serine/one-carbon metabolism and confers metabolic vulnerability in lung cancer cells. Sci Adv. (2022) 8:eabm8786. doi: 10.1126/sciadv.abm8786
57. Zhang B, Zheng A, Hydbring P, Ambroise G, Ouchida AT, Goiny M, et al. PHGDH defines a metabolic subtype in lung adenocarcinomas with poor prognosis. Cell Rep. (2017) 19:2289–303. doi: 10.1016/j.celrep.2017.05.067
58. Jing N, Zhang K, Chen X, Liu K, Wang J, Xiao L, et al. ADORA2A-driven proline synthesis triggers epigenetic reprogramming in neuroendocrine prostate and lung cancers. J Clin Invest. (2023) 133:e168670. doi: 10.1172/JCI168670
59. Elia I, Broekaert D, Christen S, Boon R, Radaelli E, Orth MF, et al. Proline metabolism supports metastasis formation and could be inhibited to selectively target metastasizing cancer cells. Nat Commun. (2017) 8:15267. doi: 10.1038/ncomms15267
60. Liu Y, Mao C, Wang M, Liu N, Ouyang L, Liu S, et al. Cancer progression is mediated by proline catabolism in non-small cell lung cancer. Oncogene. (2020) 39:2358–76. doi: 10.1038/s41388-019-1151-5
61. Mailloux RJ. Proline and dihydroorotate dehydrogenase promote a hyper-proliferative state and dampen ferroptosis in cancer cells by rewiring mitochondrial redox metabolism. Biochim Biophys Acta Mol Cell Res. (2024) 1871:119639. doi: 10.1016/j.bbamcr.2023.119639
62. Xu E, Ji B, Jin K, and Chen Y. Branched-chain amino acids catabolism and cancer progression: focus on therapeutic interventions. Front Oncol. (2023) 13:1220638. doi: 10.3389/fonc.2023.1220638
63. Yao X, Deng Y, Zhou J, Jiang L, and Song Y. Expression pattern and prognostic analysis of branched-chain amino acid catabolism-related genes in non-small cell lung cancer. Front Biosci (Landmark Ed). (2023) 28:107. doi: 10.31083/j.fbl2806107
64. Hassanein M, Hoeksema MD, Shiota M, Qian J, Harris BK, Chen H, et al. SLC1A5 mediates glutamine transport required for lung cancer cell growth and survival. Clin Cancer Res. (2013) 19:560–70. doi: 10.1158/1078-0432.CCR-12-2334
65. Cormerais Y, Giuliano S, LeFloch R, Front B, Durivault J, Tambutté E, et al. Genetic disruption of the multifunctional CD98/LAT1 complex demonstrates the key role of essential amino acid transport in the control of mTORC1 and tumor growth. Cancer Res. (2016) 76:4481–92. doi: 10.1158/0008-5472.CAN-15-3376
66. Liu X, Qin H, Li Z, Lv Y, Feng S, Zhuang W, et al. Inspiratory hyperoxia suppresses lung cancer metastasis through a MYC/SLC1A5-dependent metabolic pathway. Eur Respir J. (2022) 60:2200062. doi: 10.1183/13993003.00062-2022
67. Hassanein M, Qian J, Hoeksema MD, Wang J, Jacobovitz M, Ji X, et al. Targeting SLC1a5-mediated glutamine dependence in non-small cell lung cancer. Int J Cancer. (2015) 137:1587–97. doi: 10.1002/ijc.29535
68. Groves SM, Ildefonso GV, McAtee CO, Ozawa PMM, Ireland AS, Stauffer PE, et al. Archetype tasks link intratumoral heterogeneity to plasticity and cancer hallmarks in small cell lung cancer. Cell Syst. (2022) 13:690–710.e17. doi: 10.1016/j.cels.2022.07.006
69. Li L, Bi Z, Wadgaonkar P, Lu Y, Zhang Q, Fu Y, et al. Metabolic and epigenetic reprogramming in the arsenic-induced cancer stem cells. Semin Cancer Biol. (2019) 57:10–8. doi: 10.1016/j.semcancer.2019.04.003
70. Galan-Cobo A, Sitthideatphaiboon P, Qu X, Poteete A, Pisegna MA, Tong P, et al. LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS-mutant lung adenocarcinoma. Cancer Res. (2019) 79:3251–67. doi: 10.1158/0008-5472.CAN-18-3527
71. Kaufman JM, Amann JM, Park K, Arasada RR, Li H, Shyr Y, et al. LKB1 loss induces characteristic patterns of gene expression in human tumors associated with NRF2 activation and attenuation of PI3K-AKT. J Thorac Oncol. (2014) 9:794–804. doi: 10.1097/JTO.0000000000000173
72. Zhang S, Liu Y, Sun Y, Liu Q, Gu Y, Huang Y, et al. Aberrant R-loop-mediated immune evasion, cellular communication, and metabolic reprogramming affect cancer progression: a single-cell analysis. Mol Cancer. (2024) 23:11. doi: 10.1186/s12943-023-01924-6
73. Chen PH, Cai L, Huffman K, Yang C, Kim J, Faubert B, et al. Metabolic diversity in human non-small cell lung cancer cells. Mol Cell. (2019) 76:838–851.e5. doi: 10.1016/j.molcel.2019.08.028
74. Choi KM, Kim JJ, Yoo J, Kim KS, Gu Y, Eom J, et al. The interferon-inducible protein viperin controls cancer metabolic reprogramming to enhance cancer progression. J Clin Invest. (2022) 132:e157302. doi: 10.1172/JCI157302
75. Gu M, Liu Y, Zheng W, Jing Z, Li X, Guo W, et al. Combined targeting of senescent cells and senescent macrophages: A new idea for integrated treatment of lung cancer. Semin Cancer Biol. (2024) 106-107:43–57. doi: 10.1016/j.semcancer.2024.08.006
76. Li M, Yang Y, Xiong L, Jiang P, Wang J, and Li C. Metabolism, metabolites, and macrophages in cancer. J Hematol Oncol. (2023) 16:80. doi: 10.1186/s13045-023-01478-6
77. Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J Exp Clin Cancer Res. (2015) 34:111. doi: 10.1186/s13046-015-0221-y
78. Morigny P, Boucher J, Arner P, and Langin D. Lipid and glucose metabolism in white adipocytes: pathways, dysfunction and therapeutics. Nat Rev Endocrinol. (2021) 17:276–95. doi: 10.3390/cells9102308
79. You M, Xie Z, Zhang N, Zhang Y, Xiao D, Liu S, et al. Signaling pathways in cancer metabolism: mechanisms and therapeutic targets. Signal Transduct Target Ther. (2023) 8:196. doi: 10.1038/s41392-023-01442-3
80. Yang WH, Qiu Y, Stamatatos O, Janowitz T, and Lukey MJ. Enhancing the efficacy of glutamine metabolism inhibitors in cancer therapy. Trends Cancer. (2021) 7:790–804. doi: 10.1016/j.trecan.2021.04.003
81. Aggarwal C, Thompson JC, Black TA, Katz SI, Fan R, Yee SS, et al. Clinical implications of plasma-based genotyping with the delivery of personalized therapy in metastatic non-small cell lung cancer. JAMA Oncol. (2019) 5:173–80. doi: 10.1001/jamaoncol.2018.4305
82. Tong Y, Gao WQ, and Liu Y. Metabolic heterogeneity in cancer: An overview and therapeutic implications. Biochim Biophys Acta Rev Cancer. (2020) 1874:188421. doi: 10.1016/j.bbcan.2020.188421
83. Lanekoff I, Sharma VV, and Marques C. Single-cell metabolomics: where are we and where are we going? Curr Opin Biotechnol. (2022) 75:102693. doi: 10.1016/j.copbio.2022.102693
84. Spina E and de Leon J. Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin Pharmacol Toxicol. (2007) 100:4–22. doi: 10.1111/j.1742-7843.2007.00017.x
85. Sun C, Wang A, Zhou Y, Chen P, Wang X, Huang J, et al. Spatially resolved multi-omics highlights cell-specific metabolic remodeling and interactions in gastric cancer. Nat Commun. (2023) 14:2692. doi: 10.1038/s41467-023-38360-5
86. Hu H, Piotrowska Z, Hare PJ, Chen H, Mulvey HE, Mayfield A, et al. Three subtypes of lung cancer fibroblasts define distinct therapeutic paradigms. Cancer Cell. (2021) 39:1531–1547.e10. doi: 10.1016/j.ccell.2021.09.003
87. Chaudhri VK, Salzler GG, Dick SA, Buckman MS, Sordella R, Karoly ED, et al. Metabolic alterations in lung cancer-associated fibroblasts correlated with increased glycolytic metabolism of the tumor. Mol Cancer Res. (2013) 11:579–92. doi: 10.1158/1541-7786.MCR-12-0437-T
88. Hwang SH, Yang Y, Jung JH, and Kim Y. Oleic acid from cancer-associated fibroblast promotes cancer cell stemness by stearoyl-CoA desaturase under glucose-deficient condition. Cancer Cell Int. (2022) 22:404. doi: 10.1186/s12935-022-02824-3
89. Yao H, Li Y, Zheng Y, and Lin JM. Stability of the arginine-ornithine-citrulline cycle maintained by tumor-stroma interactions in cell coculture hydrogel microspheres. Anal Chem. (2023) 95:10999–1006. doi: 10.1021/acs.analchem.3c01134
90. Zhang X, Dong Y, Zhao M, Ding L, Yang X, Jing Y, et al. ITGB2-mediated metabolic switch in CAFs promotes OSCC proliferation by oxidation of NADH in mitochondrial oxidative phosphorylation system. Theranostics. (2020) 10:12044–59. doi: 10.7150/thno.47901
91. Bai J, Liu T, Tu B, Yuan M, Shu Z, Fan M, et al. Autophagy loss impedes cancer-associated fibroblast activation via downregulating proline biosynthesis. Autophagy. (2023) 19:632–43. doi: 10.1080/15548627.2022.2093026
92. Shi L, Zhu W, Huang Y, Zhuo L, Wang S, Chen S, et al. Cancer-associated fibroblast-derived exosomal microRNA-20a suppresses the PTEN/PI3K-AKT pathway to promote the progression and chemoresistance of non-small cell lung cancer. Clin Transl Med. (2022) 12:e989. doi: 10.1002/ctm2.989
93. Cong J, Wang X, Zheng X, Wang D, Fu B, Sun R, et al. Dysfunction of natural killer cells by FBP1-induced inhibition of glycolysis during lung cancer progression. Cell Metab. (2018) 28:243–255.e5. doi: 10.1016/j.cmet.2018.06.021
94. La Fleur L, Botling J, He F, Pelicano C, Zhou C, He C, et al. Targeting MARCO and IL37R on immunosuppressive macrophages in lung cancer blocks regulatory T cells and supports cytotoxic lymphocyte function. Cancer Res. (2021) 81:956–67. doi: 10.1158/0008-5472.CAN-20-1885
95. Chen Y, Zhou Y, Ren R, Chen Y, Lei J, and Li Y. Harnessing lipid metabolism modulation for improved immunotherapy outcomes in lung adenocarcinoma. J Immunother Cancer. (2024) 12:e008811. doi: 10.1136/jitc-2024-008811
96. Best SA, Gubser PM, Sethumadhavan S, Kersbergen A, Negrón Abril YL, Goldford J, et al. Glutaminase inhibition impairs CD8 T cell activation in STK11-/Lkb1-deficient lung cancer. Cell Metab. (2022) 34:874–887.e6. doi: 10.1016/j.cmet.2022.04.003
97. Hamaidi I, Zhang L, Kim N, Wang MH, Iclozan C, Fang B, et al. Sirt2 inhibition enhances metabolic fitness and effector functions of tumor-reactive T cells. Cell Metab. (2020) 32:420–436.e12. doi: 10.1016/j.cmet.2020.07.008
98. Azuma K, Xiang H, Tagami T, Kasajima R, Kato Y, Karakawa S, et al. Clinical significance of plasma-free amino acids and tryptophan metabolites in patients with non-small cell lung cancer receiving PD-1 inhibitor: a pilot cohort study for developing a prognostic multivariate model. J Immunother Cancer. (2022) 10:e004420. doi: 10.1136/jitc-2021-004420
99. Abd El-Fattah EE. IDO/kynurenine pathway in cancer: possible therapeutic approaches. J Transl Med. (2022) 20:347. doi: 10.1186/s12967-022-03554-w
100. Zhang H, Li S, Wang D, Liu S, Xiao T, Gu W, et al. Metabolic reprogramming and immune evasion: the interplay in the tumor microenvironment. Biomark Res. (2024) 12:96. doi: 10.1186/s40364-024-00646-1
101. Zheng Y, Li L, Shen Z, Wang L, Niu X, Wei Y, et al. Mechanisms of neural infiltration-mediated tumor metabolic reprogramming impacting immunotherapy efficacy in non-small cell lung cancer. J Exp Clin Cancer Res. (2024) 43:284. doi: 10.1186/s13046-024-03202-9
102. Wang Y, Wang D, Yang L, and Zhang Y. Metabolic reprogramming in the immunosuppression of tumor-associated macrophages. Chin Med J (Engl). (2022) 135:2405–16. doi: 10.1097/CM9.0000000000002426
103. Salem A, Asselin MC, Reymen B, Jackson A, Lambin P, West CML, et al. Targeting hypoxia to improve non-small cell lung cancer outcome. J Natl Cancer Inst. (2018) 110(1). doi: 10.1093/jnci/djx160
104. Wang D, Zhao C, Xu F, Zhang A, Jin M, Zhang K, et al. Cisplatin-resistant NSCLC cells induced by hypoxia transmit resistance to sensitive cells through exosomal PKM2. Theranostics. (2021) 11:2860–75. doi: 10.7150/thno.51797
105. Giatromanolaki A, Koukourakis MI, Pezzella F, Turley H, Sivridis E, Bouros D, et al. Expression of prolyl-hydroxylases PHD-1, 2 and 3 and of the asparagine hydroxylase FIH in non-small cell lung cancer relates to an activated HIF pathway. Cancer Lett. (2008) 262:87–93. doi: 10.1016/j.canlet.2007.11.041
106. Nisar H, Labonté FM, Roggan MD, Schmitz C, Chevalier F, Konda B, et al. Hypoxia modulates radiosensitivity and response to different radiation qualities in A549 non-small cell lung cancer (NSCLC) cells. Int J Mol Sci. (2024) 25:1010. doi: 10.3390/ijms25021010
107. Chen W, Tang D, Lin J, Huang X, Lin S, Shen G, et al. Exosomal circSHKBP1 participates in non-small cell lung cancer progression through PKM2-mediated glycolysis. Mol Ther Oncolytics. (2022) 24:470–85. doi: 10.1016/j.omto.2022.01.012
108. Hua Q, Mi B, Xu F, Wen J, Zhao L, Liu J, et al. Hypoxia-induced lncRNA-AC020978 promotes proliferation and glycolytic metabolism of non-small cell lung cancer by regulating PKM2/HIF-1α axis. Theranostics. (2020) 10:4762–78. doi: 10.7150/thno.43839
109. Kokeza J, Strikic A, Ogorevc M, Kelam N, Vukoja M, Dilber I, et al. The effect of GLUT1 and HIF-1α expressions on glucose uptake and patient survival in non-small-cell lung carcinoma. Int J Mol Sci. (2023) 24:10575. doi: 10.3390/ijms241310575
110. Miao Y, Wang W, Dong Y, Hu J, Wei K, Yang S, et al. Hypoxia induces tumor cell growth and angiogenesis in non-small cell lung carcinoma via the Akt-PDK1-HIF1α-YKL-40 pathway. Transl Cancer Res. (2020) 9:2904–18. doi: 10.21037/tcr.2020.03.80
111. Salaroglio IC, Belisario DC, Akman M, La Vecchia S, Godel M, Anobile DP, et al. Mitochondrial ROS drive resistance to chemotherapy and immune-killing in hypoxic non-small cell lung cancer. J Exp Clin Cancer Res. (2022) 41:243. doi: 10.1186/s13046-022-02447-6
112. de Souza LP, Borghi M, and Fernie A. Plant single-cell metabolomics—challenges and perspectives. Int J Mol Sci. (2020) 21:8987. doi: 10.3390/ijms21238987
113. Planque M, Igelmann S, Ferreira Campos AM, and Fendt SM. Spatial metabolomics principles and application to cancer research. Curr Opin Chem Biol. (2023) 76:102362. doi: 10.1016/j.cbpa.2023.102362
114. Pang Z, Zhou G, Ewald J, Chang L, Hacariz O, Basu N, et al. Using MetaboAnalyst 5.0 for LC-HRMS spectra processing, multi-omics integration and covariate adjustment of global metabolomics data. Nat Protoc. (2022) 17:1735–61. doi: 10.1038/s41596-022-00710-w
115. Baysoy A, Bai Z, Satija R, and Fan R. The technological landscape and applications of single-cell multi-omics. Nat Rev Mol Cell Biol. (2023) 24:695–713. doi: 10.1038/s41580-023-00615-w
116. Guo Z, Liu Y, Li X, Huang Y, Zhou Z, and Yang C. Reprogramming hematopoietic stem cell metabolism in lung cancer: glycolysis, oxidative phosphorylation, and the role of 2-DG. Biol Direct. (2024) 19:73. doi: 10.1186/s13062-024-00514-w
117. Saleme B, Gurtu V, Zhang Y, Kinnaird A, Boukouris AE, Gopal K, et al. Tissue-specific regulation of p53 by PKM2 is redox dependent and provides a therapeutic target for anthracycline-induced cardiotoxicity. Sci Transl Med. (2019) 11:eaau8866. doi: 10.1126/scitranslmed.aau8866
118. Ying ZH, Li HM, Yu WY, and Yu CH. Iridin prevented against lipopolysaccharide-induced inflammatory responses of macrophages via inactivation of PKM2-mediated glycolytic pathways. J Inflammation Res. (2021) 14:341–54. doi: 10.2147/JIR.S292244
119. Chen Y, Yan H, Yan L, Wang X, Che X, Hou K, et al. Hypoxia-induced ALDH3A1 promotes the proliferation of non-small-cell lung cancer by regulating energy metabolism reprogramming. Cell Death Dis. (2023) 14:617. doi: 10.1038/s41419-023-06142-y
120. Dutta S, Shah RB, Singhal S, Dutta SB, Bansal S, Sinha S, et al. Metformin: a review of potential mechanism and therapeutic utility beyond diabetes. Drug Des Devel Ther. (2023) 17:1907–32. doi: 10.2147/DDDT.S409373
121. Kim S, Im JH, Kim WK, Choi YJ, Lee JY, Kim SK, et al. Enhanced sensitivity of non-small cell lung cancer with acquired resistance to epidermal growth factor receptor-tyrosine kinase inhibitors to phenformin: the roles of a metabolic shift to oxidative phosphorylation and redox balance. Oxid Med Cell Longev. (2021) 2021:5428364. doi: 10.1155/2021/5428364
122. Sri-Pathmanathan RM, Plumb JA, and Fearon KC. Clofazimine alters the energy metabolism and inhibits the growth rate of a human lung-cancer cell line in vitro and in vivo. Int J Cancer. (1994) 56:900–5. doi: 10.1002/ijc.2910560624
123. Ramkumar K, Tanimoto A, Della Corte CM, Stewart CA, Wang Q, Shen L, et al. Targeting BCL2 overcomes resistance and augments response to aurora kinase B inhibition by AZD2811 in small cell lung cancer. Clin Cancer Res. (2023) 29:3237–49. doi: 10.1158/1078-0432.CCR-23-0375
124. Falchook G, Infante J, Arkenau HT, Patel MR, Dean E, Borazanci E, et al. First-in-human study of the safety, pharmacokinetics, and pharmacodynamics of first-in-class fatty acid synthase inhibitor TVB-2640 alone and with a taxane in advanced tumors. EClinicalMedicine. (2021) 34:100797. doi: 10.1016/j.eclinm.2021.100797
125. Wang J, Lin W, Li R, Cheng H, Sun S, Shao F, et al. The deubiquitinase USP13 maintains cancer cell stemness by promoting FASN stability in small cell lung cancer. Front Oncol. (2022) 12:899987. doi: 10.3389/fonc.2022.899987
126. Pisanu ME, Noto A, De Vitis C, Morrone S, Scognamiglio G, Botti G, et al. Blockade of Stearoyl-CoA-desaturase 1 activity reverts resistance to cisplatin in lung cancer stem cells. Cancer Lett. (2017) 406:93–104. doi: 10.1016/j.canlet.2017.07.027
127. Hu X, Xiang J, Li Y, Xia Y, Xu S, Gao X, et al. Inhibition of Stearoyl-CoA Desaturase 1 potentiates anti-tumor activity of amodiaquine in non-small cell lung cancer. Biol Pharm Bull. (2022) 45:438–45. doi: 10.1248/bpb.b21-00843
128. Boysen G, Jamshidi-Parsian A, Davis MA, Siegel ER, Simecka CM, Kore RA, et al. Glutaminase inhibitor CB-839 increases radiation sensitivity of lung tumor cells and human lung tumor xenografts in mice. Int J Radiat Biol. (2019) 95:436–42. doi: 10.1080/09553002.2018.1558299
129. Wang Z, Wei D, Bin E, Li J, Jiang K, Lv T, et al. Enhanced glycolysis-mediated energy production in alveolar stem cells is required for alveolar regeneration. Cell Stem Cell. (2023) 30:1028–1042.e7. doi: 10.1016/j.stem.2023.07.007
130. Hou XB, Li TH, Ren ZP, and Liu Y. Combination of 2-deoxy d-glucose and metformin for synergistic inhibition of non-small cell lung cancer: A reactive oxygen species and P-p38 mediated mechanism. BioMed Pharmacother. (2016) 84:1575–84. doi: 10.1016/j.biopha.2016.10.037
131. Krishnamurthy S, Ng VW, Gao S, Tan MH, and Yang YY. Phenformin-loaded polymeric micelles for targeting both cancer cells and cancer stem cells in vitro and in vivo. Biomaterials. (2014) 35:9177–86. doi: 10.1016/j.biomaterials.2014.07.018
132. Berber E and Rouse BT. Controlling herpes simplex virus-induced immunoinflammatory lesions using metabolic therapy: a comparison of 2-deoxy-d-glucose with metformin. J Virol. (2022) 96:e0068822. doi: 10.1128/jvi.00688-22
133. Chang L, Fang S, Chen Y, Yang Z, Yuan Y, Zhang J, et al. Inhibition of FASN suppresses the Malignant biological behavior of non-small cell lung cancer cells via deregulating glucose metabolism and AKT/ERK pathway. Lipids Health Dis. (2019) 18:118. doi: 10.1186/s12944-019-1058-8
134. Tan YJ, Ali A, Tee SY, Teo JT, Xi Y, Go ML, et al. Galloyl esters of trans-stilbenes are inhibitors of FASN with anticancer activity on non-small cell lung cancer cells. Eur J Med Chem. (2019) 182:111597. doi: 10.1016/j.ejmech.2019.111597
135. Zhang J, Song F, Zhao X, Jiang H, Wu X, Wang B, et al. EGFR modulates monounsaturated fatty acid synthesis through phosphorylation of SCD1 in lung cancer. Mol Cancer. (2017) 16:127. doi: 10.1186/s12943-017-0704-x
136. Hess D, Chisholm JW, and Igal RA. Inhibition of stearoyl-CoA desaturase activity blocks cell cycle progression and induces programmed cell death in lung cancer cells. PloS One. (2010) 5:e11394. doi: 10.1371/journal.pone.0011394
137. Kodama M, Oshikawa K, Shimizu H, Yoshioka S, Takahashi M, Izumi Y, et al. A shift in glutamine nitrogen metabolism contributes to the Malignant progression of cancer. Nat Commun. (2020) 11:1320. doi: 10.1038/s41467-020-15136-9
138. Riess JW, Frankel P, Shackelford D, Dunphy M, Badawi RD, Nardo L, et al. Phase 1 trial of MLN0128 (Sapanisertib) and CB-839 HCl (Telaglenastat) in patients with advanced NSCLC (NCI 10327): rationale and study design. Clin Lung Cancer. (2021) 22:67–70. doi: 10.1016/j.cllc.2020.10.006
139. Momcilovic M, Bailey ST, Lee JT, Fishbein MC, Magyar C, Braas D, et al. Targeted inhibition of EGFR and glutaminase induces metabolic crisis in EGFR mutant lung cancer. Cell Rep. (2017) 18:601–10. doi: 10.1016/j.celrep.2016.12.061
140. Wang K, Lu H, Wang X, Liu Q, Hu J, Liu Y, et al. Simultaneous suppression of PKM2 and PHGDH elicits synergistic anti-cancer effect in NSCLC. Front Pharmacol. (2023) 14:1200538. doi: 10.3389/fphar.2023.1200538
141. Hamanaka RB, Nigdelioglu R, Meliton AY, Tian Y, Witt LJ, O’Leary E, et al. Inhibition of phosphoglycerate dehydrogenase attenuates bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. (2018) 58:585–93. doi: 10.1165/rcmb.2017-0186OC
142. Lee HM, Muhammad N, Lieu EL, Cai F, Mu J, Ha YS, et al. Concurrent loss of LKB1 and KEAP1 enhances SHMT-mediated antioxidant defence in KRAS-mutant lung cancer. Nat Metab. (2024) 6:1310–28. doi: 10.1038/s42255-024-01066-z
143. Han T, Wang Y, Cheng M, Hu Q, Wan X, Huang M, et al. Phosphorylated SHMT2 regulates oncogenesis through m6A modification in lung adenocarcinoma. Adv Sci (Weinh). (2024) 11:e2307834. doi: 10.1002/advs.202307834
144. Becirovic T, Zhang B, Lindskog C, Norberg E, Vakifahmetoglu-Norberg H, Kaminskyy VO, et al. Deubiquitinase USP9x regulates the proline biosynthesis pathway in non-small cell lung cancer. Cell Death Discov. (2024) 10:342. doi: 10.1038/s41420-024-02111-2
145. Yang L, Gilbertsen A, Jacobson B, Kratzke R, and Henke CA. Serum splicing factor proline- and glutamine-rich is a diagnostic marker for non-small-cell lung cancer and other solid cancers. Int J Mol Sci. (2024) 25:8766. doi: 10.3390/ijms25168766
146. Lu W, Cui J, Wang W, Hu Q, Xue Y, Liu X, et al. PPIA dictates NRF2 stability to promote lung cancer progression. Nat Commun. (2024) 15:4703. doi: 10.1038/s41467-024-48364-4
147. Yao CC, Sun RM, Yang Y, Zhou HY, Meng ZW, Chi R, et al. Accumulation of branched-chain amino acids reprograms glucose metabolism in CD8+ T cells with enhanced effector function and anti-tumor response. Cell Rep. (2023) 42:112186. doi: 10.1016/j.celrep.2023.112186
148. Li Z, Hao J, Wang T, Guo C, and Liu L. Branched-chain amino acids and risk of lung cancer: insights from mendelian randomization and NHANES III. J Thorac Dis. (2024) 16:5248–61. doi: 10.21037/jtd-24-420
149. Palanca-Ballester C, Hervas D, Villalba M, Valdes-Sanchez T, Garcia D, Alcoriza-Balaguer MI, et al. Translation of a tissue epigenetic signature to circulating free DNA suggests BCAT1 as a potential noninvasive diagnostic biomarker for lung cancer. Clin Epigenetics. (2022) 14:116. doi: 10.1186/s13148-022-01334-3
150. Hu K, Li K, Lv J, Feng J, Chen J, Wu H, et al. Suppression of the SLC7A11/glutathione axis causes synthetic lethality in KRAS-mutant lung adenocarcinoma. J Clin Invest. (2020) 130:1752–66. doi: 10.1172/JCI124049
151. Qin L, Cheng X, Wang S, Gong G, Su H, Huang H, et al. Discovery of novel aminobutanoic acid-based ASCT2 inhibitors for the treatment of non-small-cell lung cancer. J Med Chem. (2024) 67:988–1007. doi: 10.1021/acs.jmedchem.3c01093
152. Cormerais Y, Massard PA, Vucetic M, Giuliano S, Tambutté E, Durivault J, et al. The glutamine transporter ASCT2 (SLC1A5) promotes tumor growth independently of the amino acid transporter LAT1 (SLC7A5). J Biol Chem. (2018) 293:2877–87. doi: 10.1074/jbc.RA117.001342
153. Rajasinghe LD, Hutchings M, and Gupta SV. Delta-tocotrienol modulates glutamine dependence by inhibiting ASCT2 and LAT1 transporters in non-small cell lung cancer (NSCLC) cells: a metabolomic approach. Metabolites. (2019) 9:50. doi: 10.3390/metabo9030050
154. Hensley CT, Faubert B, Yuan Q, Lev-Cohain N, Jin E, Kim J, et al. Metabolic heterogeneity in human lung tumors. Cell. (2016) 164:681–94. doi: 10.1016/j.cell.2015.12.034
155. Aramini B, Masciale V, Samarelli AV, Dubini A, Gaudio M, Stella F, et al. Phenotypic, functional, and metabolic heterogeneity of immune cells infiltrating non-small cell lung cancer. Front Immunol. (2022) 13:959114. doi: 10.3389/fimmu.2022.959114
156. Yu T, Liu Z, Tao Q, Xu X, Li X, Li Y, et al. Targeting tumor-intrinsic SLC16A3 to enhance anti-PD-1 efficacy via tumor immune microenvironment reprogramming. Cancer Lett. (2024) 589:216824. doi: 10.1016/j.canlet.2024.216824
157. Morrissey SM, Zhang F, Ding C, Montoya-Durango DE, Hu X, Yang C, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. (2021) 33:2040–2058.e10. doi: 10.1016/j.cmet.2021.09.002
158. Wang W, Huang L, Jin JY, Jolly S, Zang Y, Wu H, et al. IDO immune status after chemoradiation may predict survival in lung cancer patients. Cancer Res. (2018) 78:809–16. doi: 10.1158/0008-5472.CAN-17-2995
159. Qin R, Zhao C, Wang CJ, Xu W, Zhao JY, Lin Y, et al. Tryptophan potentiates CD8+ T cells against cancer cells by TRIP12 tryptophanylation and surface PD-1 downregulation. J Immunother Cancer. (2021) 9:e002840. doi: 10.1136/jitc-2021-002840
160. Xu S, Xu Y, Solek NC, Chen J, Gong F, Varley AJ, et al. Tumor-tailored ionizable lipid nanoparticles facilitate IL-12 circular RNA delivery for enhanced lung cancer immunotherapy. Adv Mater. (2024) 36:e2400307. doi: 10.1002/adma.202400307
161. Hollstein PE, Eichner LJ, Brun SN, Kamireddy A, Svensson RU, Vera LI, et al. The AMPK-related kinases SIK1 and SIK3 mediate key tumor-suppressive effects of LKB1 in NSCLC. Cancer Discov. (2019) 9:1606–27. doi: 10.1158/2159-8290.CD-18-1261
162. Mitchell TC, Hamid O, Smith DC, Bauer TM, Wasser JS, Olszanski AJ, et al. Epacadostat plus pembrolizumab in patients with advanced solid tumors: phase I results from a multicenter, open-label phase I/II trial (ECHO-202/KEYNOTE-037). J Clin Oncol. (2018) 36:3223–30. doi: 10.1200/JCO.2018.78.9602
163. Matschke J, Larafa S, and Jendrossek V. Metabolic reprogramming of antioxidant defense: a precision medicine perspective for radiotherapy of lung cancer? Biochem Soc Trans. (2021) 49:1265–77. doi: 10.1042/BST20200866
164. Schuler M, Hense J, Darwiche K, Michels S, Hautzel H, Kobe C, et al. Early metabolic response by PET predicts sensitivity to next-line targeted therapy in EGFR-mutated lung cancer with unknown mechanism of acquired resistance. J Nucl Med. (2024) 65:851–5. doi: 10.2967/jnumed.123.266979
Keywords: non-small cell lung cancer, metabolic reprogramming, tumor microenvironment, therapeutic targeting, metabolic vulnerabilities
Citation: Cai H, Zhang F, Xu F and Yang C (2025) Metabolic reprogramming and therapeutic targeting in non-small cell lung cancer: emerging insights beyond the Warburg effect. Front. Oncol. 15:1564226. doi: 10.3389/fonc.2025.1564226
Received: 21 January 2025; Accepted: 02 May 2025;
Published: 21 May 2025.
Edited by:
Ana Luísa De Sousa-Coelho, Algarve Biomedical Center Research Institute (ABC-RI), PortugalReviewed by:
Niki Millward, University of Texas MD Anderson Cancer Center, United StatesJohann Matschke, Essen University Hospital, Germany
Copyright © 2025 Cai, Zhang, Xu and Yang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Fang Xu, eHUyNzAwMkAxMjYuY29t; Chunhui Yang, eWFuZ2NodW5odWk2MjdAMTYzLmNvbQ==
†These authors have contributed equally to this work