Previous Article in Journal
Diagnosis and Management of Pediatric Non-Alcoholic Fatty Liver Disease: An Overview
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glucose Metabolism Modulation as a Strategy to Enhance Cancer Radiotherapy

1
School of Basic Medical Sciences, Hengyang Medical College, University of South China, Hengyang 421001, China
2
Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
*
Author to whom correspondence should be addressed.
Metabolites 2025, 15(12), 793; https://doi.org/10.3390/metabo15120793 (registering DOI)
Submission received: 22 October 2025 / Revised: 10 December 2025 / Accepted: 11 December 2025 / Published: 12 December 2025
(This article belongs to the Section Endocrinology and Clinical Metabolic Research)

Abstract

A systematic literature review of the PubMed database, filtering for publication dates up to and including October 2025, was conducted to identify relevant studies on glucose metabolism and radiotherapy. Radioresistance poses a major therapeutic challenge, in which tumor-associated glucose metabolic reprogramming, characterized by the Warburg effect, supports cellular energy requirements and contributes to radioresistance by facilitating DNA repair and promoting survival pathways. Targeting pivotal glycolytic enzymes, such as hexokinase (HK) and pyruvate kinase M2 (PKM2), and integrating radiotherapy with metabolic modulators have been shown to improve radiosensitivity. Special emphasis is placed on how these interventions remodel the tumor microenvironment and modulate antitumor immunity—emerging factors that influence therapeutic efficacy. This review highlights mechanistic insights and potential therapeutic targets for the development of effective radiosensitization strategies.

1. Introduction

Cancer cells fundamentally rewire their metabolic pathways to sustain proliferation and survival, with altered metabolism now recognized as a core hallmark of malignancy. This metabolic reprogramming critically influences therapeutic responses, including resistance to radiotherapy [1]. Glucose metabolic reprogramming, characterized by increased glycolysis and lactate production under normoxic conditions (Warburg effect), supports tumor adaptation to stress [2,3]. It increases radioresistance by promoting DNA repair and survival pathways. This process affects the response to radiation by altering redox balance and energy metabolism [4,5,6]. Some tumors escape glucose metabolism-mediated radioresistance regulation by activating fatty acid synthesis [7]. Metabolites such as lactate and glutamine also contribute to immune evasion [8,9,10].
In glioblastoma, radiotherapy triggers a metabolic shift toward fatty acid β-oxidation, contributing to radioresistance and promoting immune evasion by regulating CD47 [11]. Inhibiting glycolytic transporters (such as GLUT1/4) or metabolic enzymes (including LDHA, HK2, and PKM2) has been shown to significantly improve radiosensitivity [12,13,14,15,16,17,18,19].
Consequently, targeting glucose metabolic reprogramming has become a crucial strategy for enhancing tumor radiosensitivity. This review systematically elucidates the key mechanistic roles of glucose metabolic reprogramming in tumor radioresistance, focusing on its role in DNA damage repair, cell cycle regulation, and tumor microenvironment remodeling. Furthermore, we critically evaluate potential therapeutic strategies targeting key glycolytic pathways to improve radiosensitivity, providing novel insights for developing more effective radiotherapeutic interventions.

2. Overview of Reprogramming of Glucose Metabolism

2.1. Basic Concepts and Biological Significance of Glucose Metabolic Reprogramming

Glucose metabolic reprogramming, a hallmark of cancer, refers to alterations in metabolic pathways that enable cancer cells to meet heightened demands for energy, biosynthetic precursors, and redox balance to support rapid proliferation and survival [20,21]. A hallmark of this reprogramming is the upregulation of glycolysis coupled with a reduction in mitochondrial oxidative phosphorylation (OXPHOS) [22,23]. This shift plays a critical role in the pathophysiology of multiple disorders, especially cancer and neurodegenerative diseases.
Under physiological conditions, cells predominantly produce ATP through mitochondrial OXPHOS. In contrast, during glycolytic reprogramming, ATP generation shifts toward glycolysis despite adequate oxygen availability. This phenomenon, termed the Warburg effect, is often observed in tumor cells [24]. For example, during the induction of induced pluripotent stem cells (iPSCs), elevated glycolytic activity supports somatic cell reprogramming, while its suppression impairs reprogramming efficiency [25]. Similarly, in ischemic stroke, activated microglia undergo a metabolic transition from OXPHOS to glycolysis, a shift that significantly influences their functional behavior [26].
Reduced mitochondrial OXPHOS is a critical aspect of glycolytic reprogramming. Evidence indicates that OXPHOS activity is significantly diminished in tumor cells and in the context of neurodegenerative diseases [27]. For instance, during ischemic stroke, microglial metabolic reprogramming impairs mitochondrial function, leading to decreased ATP generation and elevated oxidative stress, which collectively aggravate neuroinflammation and neuronal injury [28,29]. These findings provide mechanistic insights into glycolytic reprogramming across diverse pathological conditions and offer a conceptual framework for the development of targeted therapeutic interventions.

2.2. Glucose Metabolism and Tumor Relationship

Altered glucose metabolism facilitates tumor progression by upregulating glycolytic pathways, leading to increased glucose uptake and lactate production to support the energy requirements of rapid proliferation [30]. In certain tumor types, reprogramming of fructose metabolism plays a pivotal role. Tumor cells use GLUT5 to import fructose, which promotes survival, proliferation, and enhanced migratory potential [31]. Furthermore, fructose metabolism contributes to tumor growth and metastasis by stimulating angiogenesis [32].
Altered glucose metabolism is closely linked to tumor progression. A study from Lanzhou University showed that KRAS mutation-driven metabolic alterations, including elevated glycolysis, glutamine utilization, and lipid biosynthesis, promote cancer cell proliferation and survival [33]. Moreover, glucose metabolic reprogramming in tumor-associated macrophages (TAMs) influences tumor cell migration, invasion, and angiogenic activity [34]. These findings underscore the critical role of glucose metabolic reprogramming across all stages of tumor development and highlight its potential as a therapeutic target [35,36].

3. Glucose Metabolism and the Cellular Response to Radiation Damage

Glycolytic reprogramming plays a key role in modulating tumor cell responses to radiation-induced stress. Under hypoxic and nutrient-limited conditions, tumor cells enhance glycolysis and lactate production to maintain proliferation. This metabolic adaptation also promotes resistance to radiotherapy by modulating oxidative stress and facilitating DNA damage repair pathways [36,37]. Therefore, understanding the interplay between glucose metabolism and cellular responses to radiation is essential for advancing radiosensitization strategies.
Recent studies have highlighted the essential role of glycolytic reprogramming in modulating cellular responses to radiation. Investigators from the University of South China and Central South University reported that TAB182 regulates lactate dehydrogenase A (LDHA) transcription, modulating glycolysis and lactate production and thereby contributing to tumor radioresistance [38]. Exposure to low-glucose conditions combined with palmitic acid significantly suppresses colorectal cancer cell proliferation and increases radiosensitivity by promoting reactive oxygen species (ROS) generation and inducing DNA damage.

3.1. Glucose Metabolism and DNA Damage Repair

3.1.1. High Glycolysis Promotes DNA Damage Repair

DNA damage repair involves a series of enzymatic processes that restore damaged DNA, preserving genomic integrity and stability. The primary repair mechanisms include base excision repair, nucleotide excision repair, mismatch repair, homologous recombination (HR), and non-homologous end joining (NHEJ) [39,40,41,42]. These pathways are fundamental to the cellular response to environmental stressors such as radiation and chemical exposure and are vital for maintaining normal physiological functions.
Emerging evidence indicates a strong association between glycolytic reprogramming (particularly hyperglycolysis) and DNA damage repair [43,44]. Increased glycolytic activity enables tumor cells to increase glucose uptake and lactate production, supporting the high energy and biosynthetic demands of rapid proliferation. This metabolic shift not only drives tumor growth but also supports DNA repair by supplying key intermediates such as nucleotides and NAD+. For instance, the glycolytic enzyme aldolase A (ALDOA) has been shown to translocate to the nucleus following ionizing radiation, where it co-localizes with γ-H2AX and directly participates in double-strand break (DSB) repair [45]. Moreover, He Yulong’s group at Sun Yat-sen University demonstrated that lactate-mediated modification of the K388 residue on the DNA repair protein NBS1 enhances its recruitment to DSB sites, therefore promoting HR efficiency [46].

3.1.2. Glucose Metabolic Pathways Regulate the DNA Damage Response

Alterations in glycolytic pathways play a critical role in regulating DNA damage responses, with metabolic enzymes and intermediates directly influencing the activity of DNA repair mechanisms. For example, proteasome activator subunit 3 (PSME3) promotes DNA repair in cervical cancer cells by upregulating glycolytic proteins, whereas PSME3 knockdown impairs glycolysis and elevates radiosensitivity [47]. Similarly, inhibition of integrin β5/focal adhesion kinase signaling following cisplatin treatment reduces glycolytic activity in breast and cervical cancer cells by downregulating glycolytic enzymes [48]. These observations suggest that metabolic reprogramming not only affects energy metabolism but also modulates radiosensitivity by regulating DNA damage repair pathways.
Poly(ADP-ribose) polymerase 1 (PARP1), a key mediator of DNA damage repair, relies on NAD+ as a substrate, the biosynthesis of which is dependent on glycolytic flux [49,50]. Glutamine metabolism also plays a pivotal role in supporting DNA repair processes [51,52]. Fu et al. [53] reported that radiation-resistant tumor cells show decreased glycolytic activity, mitochondrial respiration, and tricarboxylic acid (TCA) cycle flux, accompanied by increased glutamine anabolism. The silencing of glutamine synthetase impairs DNA repair kinetics and disrupts nucleotide biosynthesis, sensitizing tumor cells to radiotherapy.

3.2. Glucose Metabolism and Cell Cycle Regulation

The cell cycle [54,55,56,57] refers to the sequential progression of events from the completion of one cell division to the initiation of the next, a process that underlies cell growth, proliferation, and the preservation of tissue homeostasis. It is divided into four distinct phases: G1 (pre-DNA synthesis), S (DNA synthesis), G2 (post-DNA synthesis), and M (mitosis). Progression through these phases is tightly regulated by a network of molecular regulators, including cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors [58,59]. These factors function through specific complex formation to coordinate phase transitions, ensuring that cell division occurs only under favorable conditions.
Cell cycle progression requires significant energy input and biosynthetic resources. Glycolysis is a major energy-producing pathway, providing ATP through the catabolism of glucose by catalyzing it to pyruvate [60,61]. Throughout all phases of the cell cycle, glycolysis supplies energy and generates key intermediates such as glyceraldehyde-3-phosphate and phosphoenolpyruvate (PEP). These intermediates serve as precursors for the synthesis of nucleic acids, proteins, and lipids, supporting the biosynthetic demands associated with cell cycle progression [62,63,64,65].
Glycolytic metabolites influence cyclin activity and regulate cell cycle progression. For example, elevated lactate levels stabilize cyclin D1, facilitating the G1/S phase transition [66]. Hexokinase 2 (HK2) promotes cell cycle progression by enhancing glycolytic flux, which increases intracellular ATP and NAD+ levels, ultimately activating key cell cycle regulators [67].
Cell cycle regulators also exert control over glycolytic activity, establishing a bidirectional relationship between metabolism and cell proliferation. CDKs modulate the transcription of glycolytic genes, either by influencing transcription factors or through direct phosphorylation of gene promoters. CDK4/6–Cyclin D complexes activate E2F transcription factors, leading to the upregulation of glycolytic enzymes such as HK2 and LDHA, thereby increasing glycolytic flux [68]. Similarly, cell division cycle-associated protein 7 (CDCA7) promotes glycolysis, facilitating pancreatic cancer cell proliferation and chemoresistance [69]. This reciprocal regulation enables flexible metabolic adaptation to meet the energetic and biosynthetic requirements of cell cycle progression.

4. Glucose Metabolism and Tumor Radioresistance

4.1. Tumor Radioresistance and Intrinsic Factors

Radiotherapy remains a cornerstone of cancer treatment. It is utilized in >50% of cancer patients for its ability to achieve local tumor control while preserving organ function [70,71]. Its efficacy is limited by intrinsic or acquired tumor radioresistance, a critical obstacle to curative outcomes [72]. Tumor radioresistance refers to the ability of tumor cells to withstand radiation-induced cytotoxicity and often results in reduced radiotherapy efficacy or treatment failure [73,74,75]. This phenomenon arises from inherent cellular characteristics or adaptive responses triggered by radiation exposure. A major obstacle to successful radiotherapy, radioresistance, is the product of multifaceted mechanisms driven by both intrinsic and extrinsic factors within the TME.
The intrinsic mechanisms of tumor radioresistance include a range of cellular adaptations that diminish the efficacy of radiotherapy (Figure 1). A central mechanism involves tumor cells’ increased capacity for DNA damage repair. Radiotherapy primarily exerts its cytotoxic effects via DNA DSBs, which some tumor cells can efficiently repair by activating key repair pathways, such as HR and NHEJ, increasing their survival rate after radiation exposure [76,77,78]. Dysregulation of the cell cycle also contributes significantly to intrinsic radioresistance. Aberrant activation of CDKs or altered cell cycle distribution may result in a predominance of tumor cells in radiation-insensitive phases during treatment, such as the G2 phase, reducing radiosensitivity [79,80,81]. Cancer stem cells (CSCs) are another intrinsic factor contributing to radioresistance. Characterized by self-renewal and differentiation capabilities, CSCs exhibit heightened resistance to radiation through the maintenance of stemness, the activation of specific signaling pathways, including the PI3K/AKT/mTOR axis, and metabolic reprogramming [82,83,84,85]. Metabolic adaptation is a further determinant of intrinsic radioresistance. Under radiotherapy-induced stress, tumor cells may rewire their metabolism, upregulating glycolysis or promoting fatty acid oxidation to support survival. For example, upregulation of fatty acid synthase (FASN) has been shown to stimulate fatty acid metabolism, thereby increasing resistance to radiation [86]. Genetic alterations also underlie radioresistant phenotypes. Mutations in tumor suppressor genes such as TP53 are frequently observed across tumor types and are closely linked to radioresistance. In head and neck squamous cell carcinoma and colorectal cancer, TP53 mutations are associated with reduced radiosensitivity, likely due to impairment of DNA damage response pathways [87,88]. Aberrant activation of YAP, a key effector of the Hippo signaling pathway, has been shown to increase DNA repair capacity and promote tumor cell proliferation, further contributing to radioresistance [89].

4.2. Role of Glycolysis in Radiotherapy Resistance

4.2.1. Glycolytic Pathways Support Tumor Survival

The Warburg effect supports rapid tumor proliferation and influences signaling pathways through metabolite-mediated mechanisms that contribute to radioresistance [90,91,92]. Elevated glycolytic activity also mitigates radiotherapy-induced apoptosis by modulating redox balance and reducing intracellular ROS levels [93].

4.2.2. Lactate Accumulation and Radiotherapy Resistance

Elevated glycolysis leads to the accumulation of metabolites such as lactate, which plays a regulatory role in DNA damage repair through protein lactylation, ultimately diminishing the efficacy of radiotherapy. In glioblastoma, ALDH1A3 activates PKM2, promoting lactate accumulation. Lactate-induced modification of XRCC1 increases DNA repair capacity, contributing to radioresistance [94]. Lactate dehydrogenase (LDH, which catalyzes the conversion of pyruvate to lactate) is frequently overexpressed in tumors and is associated with radioresistant phenotypes [38,95]. LDH facilitates radioresistance by increasing lactate production, thereby supporting tumor cell survival and proliferation. In therapy-resistant tumors, LDHA expression is significantly elevated and is accompanied by excessive lactate accumulation [96]. By catalyzing the conversion of pyruvate to lactate, LDHA accelerates glycolysis, providing both energy and biosynthetic intermediates necessary for sustaining rapid tumor growth and resisting radiotherapy.

4.2.3. Other Metabolic Intermediates and Radiotherapy Resistance

Beyond increased glucose metabolism and lactate accumulation, glycolytic intermediates such as pyruvate, fructose-6-phosphate (F6P), and fructose-1,6-bisphosphate play critical roles in promoting radioresistance. These metabolites facilitate tumor cell survival and proliferation by regulating energy metabolism [97], increasing DNA repair mechanisms [98], modulating oxidative stress responses [99], and reshaping the immune microenvironment [100].
Pyruvate, a central glycolytic intermediate, plays a pivotal role in regulating tumor radioresistance by modulating the DNA damage response. Pyruvate improves FACT-mediated chromatin loading of γH2AX, promoting chromatin remodeling and facilitating efficient DNA repair. This mechanism contributes to increased glioblastoma resistance to radiation-induced DNA damage [101].
Pyruvate-metabolizing enzymes, including pyruvate kinase M2 (PKM2) and pyruvate dehydrogenase (PDH), are key regulators of metabolic reprogramming and radioresistance. PKM2 catalyzes the conversion of PEP to pyruvate with concurrent ATP generation during glycolysis. Beyond its enzymatic role, PKM2 modulates the accumulation of glycolytic intermediates, sustaining aerobic glycolysis in tumors that preferentially engage glycolysis despite sufficient oxygen availability [102,103,104,105]. Activation of PKM2 also induces the release of paclitaxel and indoleamine 2,3-dioxygenase (IDO) inhibitors, augmenting the efficacy of immunochemotherapy in non-small cell lung cancer (NSCLC) [106]. In comparison, PDH mediates the conversion of pyruvate to acetyl-CoA, facilitating its entry into the TCA cycle. Inhibition of PDH restores pyruvate carboxylase (PC) activity, leading to increased succinate secretion, which activates SUCNR1 signaling and boosts CD8+ T cell cytotoxic function, counteracting tumor-induced immunosuppression [107].
Phosphofructokinase-1 (PFK-1) catalyzes the conversion of F6P to fructose-1,6-bisphosphate, a key rate-limiting step in glycolysis that drives further ATP production [108]. Accumulation of F6P is associated with increased tumor radioresistance, reflecting its role in not only energy metabolism but also the regulation of multiple cellular processes. In hepatocellular carcinoma, radioresistant cells show elevated glucose uptake and increased F6P levels, promoting resistance to radiation through redistribution of glycolytic flux and increased branch-point anabolic activity [109].

4.3. Altered Tumor Microenvironment (TME) and Radiotherapy Resistance

4.3.1. Acidification of the TME

Glycolytic reprogramming leads to excessive lactate production, resulting in acidification of the TME. The acidic TME significantly impairs radiotherapy efficacy by diminishing tumor cell radiosensitivity [110]. This effect is mediated through multiple mechanisms: the acidic TME directly subverts radiation-induced antitumor immunity by suppressing key chemokines such as CCL5, thereby blocking CD8+ T cell recruitment to tumors post-irradiation [111]. Research by Gu et al. revealed that an acidic TME activates the NF-κB pathway, inducing the release of inflammatory factors like IL-6, which promote tumor cell survival while further disrupting the immune microenvironment [112]. Within tumor cells, acidosis cooperates with hypoxia to maintain a state of constitutive ATM/CHK1/CHK2 DNA damage checkpoint activation, conferring markedly enhanced DNA repair capacity [113]. A nanotechnology system developed by Dou et al. showed that altering the TME—for instance, using nitric oxide release to alleviate hypoxia and acidosis—can synergistically regulate HIF1α and p53 expression, offering an effective strategy to reverse TME-mediated radioresistance [114]. Additionally, acidic conditions contribute to aberrant, permeable vasculature that further limits radiotherapy by impairing oxygen and drug delivery [115,116].

4.3.2. Effects of Acidic Environment on Cellular Functions

This acidity adversely affects drug permeability and intratumoral distribution, resulting in subtherapeutic drug concentrations and diminished treatment efficacy. Furthermore, the stability of certain therapeutics is compromised under acidic conditions, further limiting their effectiveness. To address these challenges, pH-responsive drug delivery systems have emerged as a promising strategy. For instance, the CDBi, Mn-FA@CaP complex, a novel radiosensitizer, undergoes acid-triggered degradation, releasing smaller carbon dots capable of deep tumor penetration, thereby significantly increasing the efficacy of radiotherapy [117].
In addition to limiting drug permeability, acidic microenvironments contribute to tumor radioresistance through various mechanisms. Acidic conditions activate signaling pathways and increase DNA repair processes, reducing tumor cell radiosensitivity. In breast cancer, acidosis induces activation of β-catenin/TCF signaling, resulting in an increased proportion of CSCs with elevated stemness and migratory potential. This cellular phenotype facilitates tumor regeneration and recurrence after radiotherapy [118]. Moreover, acidic conditions suppress immune cell function and attenuate antitumor immune responses, further weakening radiotherapy-induced immune activation [119,120,121].

4.3.3. Interaction Between Microenvironment and Metabolism

The interplay between the TME and metabolic reprogramming is pivotal in regulating tumor progression. Tumor cells adapt to hypoxic and nutrient-limited conditions by reprogramming their metabolism, particularly through upregulated glycolysis and glutamine metabolism, which support sustained proliferation and survival [122,123,124]. A central component of this adaptation involves the metabolic reprogramming of TAMs, which is closely linked to immunosuppression and tumor progression within the TME. Modulating TAM metabolism has been shown to augment antitumor immune responses and improve the efficacy of immunotherapy [125]. Tumor-derived metabolites, such as lactate and glutamine, further influence immune cell function, facilitating immune evasion. For example, lactate contributes to tumor progression by altering histone acetylation and suppressing macrophage inflammatory responses [126]. This metabolic–immune crosstalk orchestrates both tumor development and immune modulation within the TME.
Modulating the TME metabolism is a promising strategy to increase the efficacy of radiotherapy. A hallmark of the TME is its acidic pH, which is typically lower than that of normal tissues. pH-responsive nanomaterials have emerged as a potential tool for mitigating these limitations [127,128,129]. Targeting glutamine metabolism also disrupts the tumor energy supply and increases radiosensitivity by impairing tumor cell viability and simultaneously improving immune cell function through modulation of the immune microenvironment [52,130]. Further improvement in radiotherapy outcomes can be achieved by targeting key TME metabolites. Inhibiting LDHA, for example, reduces lactate accumulation and increases tumor cell sensitivity to radiation [131].

4.3.4. Metabolic Crosstalk in the Tumor Microenvironment and Immune Synapse Function

Beyond fueling tumor growth, glucose metabolic reprogramming actively contributes to an immunosuppressive TME via metabolite-driven crosstalk. Impairment of T cell immune synapse function is a critical mechanism of radiotherapy resistance.
As a key glycolytic byproduct, lactate acts as a central mediator of immunosuppression. Elevated lactate concentrations acidify the TME, directly suppressing cytotoxic T lymphocyte (CTL) activity and proliferation. Lactate also promotes broad immunosuppression through epigenetic mechanisms such as histone lactylation, facilitating M2 macrophage polarization and impairing natural killer (NK) cell function [132]. This immunosuppressive environment critically attenuates the antitumor immune response initiated by radiotherapy.
Nutrient competition is another crucial mechanism. Activated T cells depend on glycolysis to support effector functions and clonal expansion. However, high glucose consumption by Warburg-effect-driven tumor cells results in local glucose deprivation. This metabolic stress directly compromises T cell immune synapse formation—an energy-intensive process that requires ATP for cytoskeletal dynamics and polarized granule secretion. Under glucose restriction, T cells display disrupted immune synapse integrity, diminished signaling transduction, impaired cytokine production (e.g., IFN-γ), and reduced tumor-killing activity [133].

4.3.5. Key Glycolytic Enzymes as Hubs Linking Intrinsic Tumor Metabolism to the TME

The metabolic alterations within the TME are driven directly by the upregulated activity of specific glycolytic enzymes in tumor cells. For instance, LDHA overactivity is a primary driver of lactate accumulation, leading to TME acidification and promoting radioresistance through mechanisms such as protein lactylation [46,94]. Similarly, HK2-mediated high glycolytic flux not only fulfills the energy demands of cancer cells but also creates nutrient competition, impairing T cell function within the TME [133,134]. Furthermore, the PKM2 isoform, particularly in its low-activity state, favors metabolic reprogramming that sustains the Warburg effect and associated TME remodeling [102]. This direct link establishes these pivotal enzymes as high-priority therapeutic targets, whose inhibition is anticipated to simultaneously disrupt tumor cell metabolism and reverse the radioresistant TME, providing a foundation for the targeted strategies discussed in the following chapter.

5. Glucose Metabolic Reprogramming and Radiosensitization Targets

Building on the rationale that key glycolytic enzymes, such as HK2, LDHA, and PKM2, are central drivers of both tumor metabolism and the radioresistant TME, the following section focuses on therapeutic strategies directly targeting these molecules. Given the significant therapeutic obstacles posed by tumor complexity and heterogeneity, targeting glycolysis-associated pathways has emerged as a prominent area of investigation for radiosensitization (see Table 1) [135,136,137,138].

5.1. Hexokinase

Hexokinase (HK), the first enzyme in glycolysis, phosphorylates glucose to glucose-6-phosphate (G6P), initiating the glycolytic pathway and acting as a central regulatory hub in tumor metabolic reprogramming [155,156]. HK2, which is overexpressed in various tumors, plays a pivotal role in radiosensitization. By regulating glycolysis, HK2 meets the high-energy demands of tumors, supporting cell survival under radiation stress. Its overexpression accelerates glycolysis, increases ATP production, and maintains tumor viability during irradiation [134]. HK2 also interacts with mitochondrial VDACs to modulate redox homeostasis and limit ROS accumulation, increasing radiotolerance. Through these mechanisms, HK2 coordinates energy metabolism to sustain rapid proliferation and influence tumor radiosensitivity [141].
Bergenin (at 60 μM) suppresses HK2 expression (as confirmed by Western blotting) in radioresistant OSCC cells (CAL27IR and SCC25IR, established by serial exposure to 2 Gy/day of radiation up to a total dose of 80 Gy), thereby restoring radiosensitivity (as evidenced by colony formation assays), reducing proliferation and glycolysis, and promoting apoptosis [142]. The FDA-approved antifungal agent ketoconazole also inhibits HK2, thereby enhancing the efficacy of radiotherapy [143]. Overall, targeting HK2 significantly increases tumor radiosensitivity and shows promise for improving therapeutic outcomes.

5.2. PFK-1

PFK-1, a rate-limiting enzyme in glycolysis, catalyzes the conversion of F6P to fructose-1,6-bisphosphate, functioning as a pivotal regulatory checkpoint [157,158,159]. By controlling the glycolytic flux, PFK-1 fulfills the energy and biosynthetic requirements of tumor cells, driving metabolic reprogramming. Owing to its central role in tumor metabolism, PFK-1 is a promising target for radiosensitization strategies.
PFK-1 inhibitors reprogram tumor metabolism and improve the efficacy of radio- and chemotherapy. Among them, 3-(3-pyridyl)-1-(4-pyridyl)-2-propen-1-one (3PO) selectively inhibits PFKFB3, lowering fructose-2,6-bisphosphate (F2,6BP) levels and reducing glycolytic flux. 3PO preferentially targets Ras-transformed cells, suppressing adenocarcinoma growth. In combination with ascorbic acid, it synergistically induces ROS-dependent apoptosis [144]. Tryptolinamide (TLAM), another PFK-1 inhibitor, shows therapeutic promise in cells harboring mutant mitochondrial DNA by promoting mitochondrial respiration and restoring neural differentiation. TLAM inhibits PFK-1 to activate AMPK-mediated fatty acid oxidation, stimulate OXPHOS, and divert glycolytic intermediates into the pentose phosphate pathway (PPP), increasing antioxidant capacity [145]. Altered PFK-1 activity in tumors diverts glycolytic intermediates to the pentose phosphate pathway (PPP), supporting biosynthetic demands [160].
Genetic factors also influence PFK-1 activity. Regulators such as YY1, synoviolin, shRNA-507, SNAI, miR-520a/b/e, miR-128, and β-miR-6517 suppress tumorigenesis by modulating PFK-1 expression [161]. Furthermore, PFK-1 is regulated by metabolites, including ATP, ADP, AMP, and citrate, and by post-translational modifications. These multifaceted regulatory mechanisms support the development of novel PFK-1 inhibitors [162]. Targeting PFK-1 activity offers a strategic approach to reprogramming tumor metabolism and increasing radiosensitivity.

5.3. Pyruvate Kinase

Pyruvate kinase (PK), the final enzyme in glycolysis, catalyzes the conversion of PEP to pyruvate, generating ATP in the process [163,164]. In tumors, the M2 isoform (PKM2) predominates and plays a central role in metabolic reprogramming [165,166]. PKM2 exists in two conformations: an active tetramer and a less active dimer. The tetrameric form efficiently channels pyruvate into the TCA cycle, whereas the dimeric form favors lactate production, sustaining aerobic glycolysis [91]. The dimeric PKM2 can translocate to the nucleus, where it acts as a protein kinase to phosphorylate target proteins and promote tumor proliferation [167]. Owing to its dual metabolic and signaling functions, PKM2 is a promising target for radiosensitization.
Small-molecule inhibitors, such as shikonin, suppress tumor growth by inhibiting PKM2 and increasing ROS levels [146]. Activators (e.g., TEPP-46, DASA-58) stabilize PKM2 tetramers, elevating enzymatic activity to shift tumors from aerobic glycolysis to OXPHOS, improving radiotherapy efficacy [147,148,168]. PKM2 knockdown via siRNA/shRNA increases radiosensitivity in multiple tumor models, including glioma and lung cancer. shRNA-mediated PKM2 targeting combined with radiotherapy inhibits lung cancer proliferation and increases γ-H2AX expression [149]. PKM2-targeting shRNA increases leukemia cell radiosensitivity by suppressing NF-κB/HIF-1α signaling, decreasing glycolytic activity, and reducing stemness and DNA repair capacity [94]. Thus, modulation of PKM2 activity reprograms tumor metabolism and offers a viable strategy to increase radiosensitivity.

6. Clinical Applications of Glucose Metabolic Reprogramming

Glycolytic reprogramming is a hallmark of tumors and metabolic disorders, in which it promotes disease progression and influences therapeutic outcomes by regulating energy metabolism pathways, such as glycolysis and the pentose phosphate pathway, and key metabolic enzymes. The integration of radiotherapy and glycolytic modulation has become an essential focus in clinical research.

6.1. Glycolytic Pathway Intervention Strategies

Strategies targeting the glycolytic pathway encompass multiple therapeutic approaches. In targeted therapy, nano-delivery systems directed at YAP1/TXNIP, such as verteporfin-loaded PLGA nanoparticles, suppress synoviocyte glycolysis, reduce macrophage infiltration, and mitigate inflammatory microenvironments in diabetic osteoarthritis [150]. Among stem cell technologies, chemically induced pluripotent stem cells (CiPSCs) provide innovative options for treating metabolic diseases. Deng Hongkui’s group generated human CiPSCs in under 10 days and subsequently differentiated them into pancreatic β-cells for transplantation in type 1 diabetes. The transplant recipients achieved insulin independence by day 75, with efficacy sustained for over a year. This approach accelerates epigenetic reprogramming via the histone modifiers KAT3A/B and KAT6A, supporting personalized therapeutic applications [152]. Metabolic–immune synergy is also a viable approach: CRISPR activation (CRISPRa) promotes adipocyte browning, increasing the competitive use of glucose and fatty acids and suppressing breast and pancreatic tumor growth [153]. Such cross-regulatory interventions hold promise for integration with immune checkpoint inhibitors.

6.2. Radiotherapy–Glycolysis Co-Targeting

Co-targeting glycolysis and radiotherapy significantly improves therapeutic efficacy and helps overcome radioresistance. In laryngeal cancer, ionizing radiation induces GLUT-1 overexpression, contributing to radioresistance, whereas GLUT-1 antisense oligonucleotides increase radiosensitivity [169]. However, there are obstacles to clinical translation, including oligonucleotide delivery efficiency and potential off-target effects. Similarly, in HepG2 cells, radiotherapy increases lactate, alanine, and glucose levels, thereby promoting glycolysis and resistance to radiation. These findings require validation in diverse tumor models, given intertumoral metabolic heterogeneity. Metabolic intervention in these contexts significantly augments radiotherapy outcomes [154]. Radiotherapy–glycolysis co-targeting is a potent therapeutic approach that simultaneously modulates tumor energy metabolism and DNA repair capacity. However, potential toxicity to normal tissue resulting from metabolic disruption warrants careful investigation. Further research should focus on optimizing delivery strategies and validating safety profiles during clinical translation to improve patient prognosis.

7. Conclusions and Future Perspectives

Building on this foundation of glycolytic reprogramming, we focus on how the Warburg effect-driven metabolic shift specifically enables radioresistance through modulation of DNA repair, cell cycle dysregulation, and TME remodeling. Elucidating these mechanisms is essential for designing strategies to counteract radiotherapeutic resistance.
Radioresistance driven by glycolytic reprogramming operates through three principal mechanisms: (1) increased glycolysis supplies the energy and metabolic intermediates necessary for proliferation while modulating redox balance and signaling pathways to promote resistance; (2) upregulation of glycolytic enzymes and lactate production increases DNA DSB repair, mitigating radiation-induced damage; and (3) provision of energy and metabolites facilitates cell cycle progression, ultimately increasing radiotolerance.
Targeting glycolytic reprogramming shows significant therapeutic promise. The inhibition of key glycolytic enzymes, such as HK and PKM2, has been shown to elevate tumor radiosensitivity. HK inhibitors, including 3-bromopyruvate (3BP), suppress glycolysis and improve radiotherapeutic outcomes. Combining radiotherapy with metabolic interventions is highly efficacious, with glucose analogs such as 2-deoxy-D-glucose (2-DG) effectively inhibiting glycolysis to potentiate radiosensitivity. Furthermore, nanomaterial-based delivery systems, such as PLGA/PEG polymers carrying anti-glycolytic agents like LDHA- or GLUT1-targeting siRNAs, significantly amplify the effects of radiosensitization.
Future research should focus on developing innovative strategies that target glycolytic reprogramming and tumor heterogeneity concurrently. To this end, it is crucial to utilize technologies such as single-cell metabolomics and spatial transcriptomics to thoroughly analyze and delineate distinct metabolic subtypes within tumors, thereby laying a foundation for the design of interventions tailored to specific subpopulations. Promising research directions include the development of rational combination therapies, such as combining glycolytic inhibitors, like those targeting HK2 or LDHA, with immune checkpoint blockers to simultaneously disrupt the tumor’s energy supply and reverse immunosuppression, or co-targeting alternative pathways such as glutamine metabolism to counteract compensatory resistance. Concurrent advances in precision medicine based on metabolic profiling—using FDG-PET imaging and liquid biopsies for patient stratification—will help identify individuals most likely to benefit from glycolysis-targeted therapies. Furthermore, the development of multi-targeting nanocarriers capable of co-delivering radiosensitizers, metabolic inhibitors, and immunomodulators is a key technological approach to addressing intratumoral metabolic heterogeneity and enhancing drug delivery efficiency. Ultimately, validating the safety and efficacy of these integrated strategies through well-designed clinical trials is essential for successfully translating metabolic interventions into clinical practice.
In conclusion, glycolytic reprogramming contributes to radioresistance by promoting glycolysis, increasing lactate production, and increasing DNA repair capacity. Targeting glycolytic enzymes and integrating metabolic modulation with radiotherapy have strong potential for overcoming resistance. Advancing this field will depend on developing novel strategies, addressing tumor heterogeneity, and facilitating clinical application. This review highlights the critical role of glycolytic reprogramming in radiotherapy resistance and provides a framework for future research.

Author Contributions

Writing—original draft, S.G.; Conceptualization, S.G. and P.Z.; Data curation, S.G.; Investigation, S.G.; Visualization, S.G.; Resources, X.L. and S.C.; Supervision, X.L., S.C., and P.Z.; Funding acquisition, P.Z.; Project administration, P.Z.; Writing—review and editing, P.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (No. 32171238). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Fukushi, A.; Kim, H.D.; Chang, Y.C.; Kim, C.H. Revisited Metabolic Control and Reprogramming Cancers by Means of the Warburg Effect in Tumor Cells. Int. J. Mol. Sci. 2022, 23, 10037. [Google Scholar] [CrossRef]
  2. Vaupel, P.; Schmidberger, H.; Mayer, A. The Warburg effect: Essential part of metabolic reprogramming and central contributor to cancer progression. Int. J. Radiat. Biol. 2019, 95, 912–919. [Google Scholar] [CrossRef]
  3. Vaupel, P.; Multhoff, G. Revisiting the Warburg effect: Historical dogma versus current understanding. J. Physiol. 2021, 599, 1745–1757. [Google Scholar] [CrossRef]
  4. Chen, Y.; Han, Z.; Zhang, L.; Gao, C.; Wei, J.; Yang, X.; Han, Y.; Li, Y.; Zhang, C.; Wei, Y.; et al. TIMELESS promotes reprogramming of glucose metabolism in oral squamous cell carcinoma. J. Transl. Med. 2024, 22, 21. [Google Scholar] [CrossRef]
  5. Turos-Cabal, M.; Sánchez-Sánchez, A.M.; Puente-Moncada, N.; Herrera, F.; Rodriguez-Blanco, J.; Antolin, I.; Alvarez-Vega, M.A.; Rodríguez, C.; Martín, V. Endoplasmic reticulum regulation of glucose metabolism in glioma stem cells. Int. J. Oncol. 2024, 64, 1. [Google Scholar] [CrossRef] [PubMed]
  6. Deng, S.; Du, J.; Gale, R.P.; Wang, L.; Zhan, H.; Liu, F.; Huang, K.; Xu, H.; Zeng, H. Glucose partitioning in the bone marrow micro-environment in acute myeloid leukaemia. Leukemia 2023, 37, 1407–1412. [Google Scholar] [CrossRef] [PubMed]
  7. Benaiges, E.; Ceperuelo-Mallafré, V.; Guaita, S.; Maymó-Masip, E.; Madeira, A.; Gómez, D.; Hernández, V.; Vilaseca, I.; Merma, C.; León, X.; et al. Survivin/BIRC5 as a novel molecular effector at the crossroads of glucose metabolism and radioresistance in head and neck squamous cell carcinoma. Head Neck 2024, 46, 1752–1765. [Google Scholar] [CrossRef] [PubMed]
  8. Brown, T.P.; Ganapathy, V. Lactate/GPR81 signaling and proton motive force in cancer: Role in angiogenesis, immune escape, nutrition, and Warburg phenomenon. Pharmacol. Ther. 2020, 206, 107451. [Google Scholar] [CrossRef]
  9. Damiani, C.; Colombo, R.; Gaglio, D.; Mastroianni, F.; Pescini, D.; Westerhoff, H.V.; Mauri, G.; Vanoni, M.; Alberghina, L. A metabolic core model elucidates how enhanced utilization of glucose and glutamine, with enhanced glutamine-dependent lactate production, promotes cancer cell growth: The WarburQ effect. PLoS Comput. Biol. 2017, 13, e1005758. [Google Scholar] [CrossRef]
  10. Vozza, A.; Parisi, G.; De Leonardis, F.; Lasorsa, F.M.; Castegna, A.; Amorese, D.; Marmo, R.; Calcagnile, V.M.; Palmieri, L.; Ricquier, D.; et al. UCP2 transports C4 metabolites out of mitochondria, regulating glucose and glutamine oxidation. Proc. Natl. Acad. Sci. USA 2014, 111, 960–965. [Google Scholar] [CrossRef]
  11. Jiang, N.; Xie, B.; Xiao, W.; Fan, M.; Xu, S.; Duan, Y.; Hamsafar, Y.; Evans, A.C.; Huang, J.; Zhou, W.; et al. Fatty acid oxidation fuels glioblastoma radioresistance with CD47-mediated immune evasion. Nat. Commun. 2022, 13, 1511. [Google Scholar] [CrossRef]
  12. Yu, B.; Schroeder, A.; Nagy, L.E. Ethanol stimulates glucose uptake and translocation of GLUT-4 in H9c2 myotubes via a Ca2+-dependent mechanism. Am. J. Physiol. Endocrinol. Metab. 2000, 279, E1358–E1365. [Google Scholar] [CrossRef]
  13. Zhang, Y.; Xu, H.; Wang, H.; Yu, W.; Zhao, X.; Xue, Y. Fluorine-18-deoxyglucose positron emission tomography/computed tomography with Ki67 and GLUT-1 immunohistochemistry for evaluation of the radiosensitization effect of oleanolic acid on C6 rat gliomas. Nucl. Med. Commun. 2015, 36, 21–27. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, Y.D.; Li, S.J.; Liao, J.X. Inhibition of glucose transporter 1 (GLUT1) chemosensitized head and neck cancer cells to cisplatin. Technol. Cancer Res. Treat. 2013, 12, 525–535. [Google Scholar] [CrossRef] [PubMed]
  15. Cai, J.; Song, L.; Zhang, F.; Wu, S.; Zhu, G.; Zhang, P.; Chen, S.; Du, J.; Wang, B.; Cai, Y.; et al. Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma. Cancer Commun. 2024, 44, 1231–1260. [Google Scholar] [CrossRef] [PubMed]
  16. Guan, L.; Nambiar, D.K.; Cao, H.; Viswanathan, V.; Kwok, S.; Hui, A.B.; Hou, Y.; Hildebrand, R.; von Eyben, R.; Holmes, B.J.; et al. NFE2L2 Mutations Enhance Radioresistance in Head and Neck Cancer by Modulating Intratumoral Myeloid Cells. Cancer Res. 2023, 83, 861–874. [Google Scholar] [CrossRef]
  17. Cai, Y.; Gao, Q.; Meng, J.H.; Chen, L. Puerarin Suppresses Glycolysis and Increases Cisplatin Chemosensitivity in Oral Squamous Cell Carcinoma via FBXW7/mTOR Signaling. Nutr. Cancer 2023, 75, 1028–1037. [Google Scholar] [CrossRef]
  18. Chang, S.; Chang, M.; Liu, G.; Xu, D.; Wang, H.; Sun, R.; Feng, M. LncRNA OIP5-AS1 reduces renal epithelial cell apoptosis in cisplatin-induced AKI by regulating the miR-144-5p/PKM2 axis. Biomed. J. 2022, 45, 642–653. [Google Scholar] [CrossRef]
  19. Liu, X.; Liu, H.; Zeng, L.; Lv, Y. BRCA1 overexpression attenuates breast cancer cell growth and migration by regulating the pyruvate kinase M2-mediated Warburg effect via the PI3K/AKT signaling pathway. PeerJ 2022, 10, e14052. [Google Scholar] [CrossRef]
  20. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  21. Pavlova, N.N.; Thompson, C.B. The Emerging Hallmarks of Cancer Metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef]
  22. Agostini, M.; Mancini, M.; Candi, E. Long non-coding RNAs affecting cell metabolism in cancer. Biol. Direct 2022, 17, 26. [Google Scholar] [CrossRef]
  23. Wang, Y.; Zhao, M.; Cui, J.; Wu, X.; Li, Y.; Wu, W.; Zhang, X. Ochratoxin A induces reprogramming of glucose metabolism by switching energy metabolism from oxidative phosphorylation to glycolysis in human gastric epithelium GES-1 cells in vitro. Toxicol. Lett. 2020, 333, 232–241. [Google Scholar] [CrossRef]
  24. Grubelnik, V.; Zmazek, J.; Marhl, M. The Synergistic Impact of Glycolysis, Mitochondrial OxPhos, and PEP Cycling on ATP Production in Beta Cells. Int. J. Mol. Sci. 2025, 26, 1454. [Google Scholar] [CrossRef] [PubMed]
  25. Ren, X.; Huang, S.; Xu, J.; Xue, Q.; Xu, T.; Shi, D.; Ma, S.; Li, X. BRG1 improves reprogramming efficiency by enhancing glycolytic metabolism. Cell. Mol. Life Sci. 2024, 81, 482. [Google Scholar] [CrossRef]
  26. Zhang, G.; Zhao, A.; Zhang, X.; Zeng, M.; Wei, H.; Yan, X.; Wang, J.; Jiang, X.; Dai, Y. Glycolytic reprogramming in microglia: A potential therapeutic target for ischemic stroke. Cell. Signal. 2024, 124, 111466. [Google Scholar] [CrossRef]
  27. Ugun-Klusek, A.; Theodosi, T.S.; Fitzgerald, J.C.; Burté, F.; Ufer, C.; Boocock, D.J.; Yu-Wai-Man, P.; Bedford, L.; Billett, E.E. Monoamine oxidase-A promotes protective autophagy in human SH-SY5Y neuroblastoma cells through Bcl-2 phosphorylation. Redox Biol. 2019, 20, 167–181. [Google Scholar] [CrossRef]
  28. Liang, J.; Han, R.; Zhou, B. Metabolic Reprogramming: Strategy for Ischemic Stroke Treatment by Ischemic Preconditioning. Biology 2021, 10, 424. [Google Scholar] [CrossRef]
  29. Madai, S.; Kilic, P.; Schmidt, R.M.; Bas-Orth, C.; Korff, T.; Büttner, M.; Klinke, G.; Poschet, G.; Marti, H.H.; Kunze, R. Activation of the hypoxia-inducible factor pathway protects against acute ischemic stroke by reprogramming central carbon metabolism. Theranostics 2024, 14, 2856–2880. [Google Scholar] [CrossRef] [PubMed]
  30. Dong, X.; Zhang, Q.; Yu, X.; Wang, D.; Ma, J.; Ma, J.; Shi, S.H. Metabolic lactate production coordinates vasculature development and progenitor behavior in the developing mouse neocortex. Nat. Neurosci. 2022, 25, 865–875. [Google Scholar] [CrossRef] [PubMed]
  31. Cui, Y.; Liu, H.; Wang, Z.; Zhang, H.; Tian, J.; Wang, Z.; Song, W.; Guo, H.; Liu, L.; Tian, R.; et al. Fructose promotes angiogenesis by improving vascular endothelial cell function and upregulating VEGF expression in cancer cells. J. Exp. Clin. Cancer Res. CR 2023, 42, 184. [Google Scholar] [CrossRef]
  32. Fang, J.H.; Chen, J.Y.; Zheng, J.L.; Zeng, H.X.; Chen, J.G.; Wu, C.H.; Cai, J.L.; Wang, Z.Y.; Zhuang, S.M. Fructose Metabolism in Tumor Endothelial Cells Promotes Angiogenesis by Activating AMPK Signaling and Mitochondrial Respiration. Cancer Res. 2023, 83, 1249–1263. [Google Scholar] [CrossRef] [PubMed]
  33. Ma, Q.; Zhang, W.; Wu, K.; Shi, L. The roles of KRAS in cancer metabolism, tumor microenvironment and clinical therapy. Mol. Cancer 2025, 24, 14. [Google Scholar] [CrossRef]
  34. Xu, M.; Zhou, C.; Weng, J.; Chen, Z.; Zhou, Q.; Gao, J.; Shi, G.; Ke, A.; Ren, N.; Sun, H.; et al. Tumor associated macrophages-derived exosomes facilitate hepatocellular carcinoma malignance by transferring lncMMPA to tumor cells and activating glycolysis pathway. J. Exp. Clin. Cancer Res. CR 2022, 41, 253. [Google Scholar] [CrossRef]
  35. Afonso, J.; Gonçalves, C.; Costa, M.; Ferreira, D.; Santos, L.; Longatto-Filho, A.; Baltazar, F. Glucose Metabolism Reprogramming in Bladder Cancer: Hexokinase 2 (HK2) as Prognostic Biomarker and Target for Bladder Cancer Therapy. Cancers 2023, 15, 982. [Google Scholar] [CrossRef]
  36. Wu, X.; Fan, Y.; Wang, K.; Miao, Y.; Chang, Y.; Ming, J.; Wang, X.; Lu, S.; Liu, R.; Zhang, F.; et al. NIR-II imaging-guided precise photodynamic therapy for augmenting tumor-starvation therapy by glucose metabolism reprogramming interference. Sci. Bull. 2024, 69, 1263–1274. [Google Scholar] [CrossRef]
  37. Xie, Y.; Liu, X.; Xie, D.; Zhang, W.; Zhao, H.; Guan, H.; Zhou, P.K. Voltage-dependent anion channel 1 mediates mitochondrial fission and glucose metabolic reprogramming in response to ionizing radiation. Sci. Total Environ. 2024, 946, 174246. [Google Scholar] [CrossRef]
  38. Chen, S.; Xie, D.F.; Li, S.; Luo, J.; Han, Y.; Guo, H.; Gao, S.; Huang, X.; Guan, H.; Huang, R.; et al. TAB182 regulates glycolytic metabolism by controlling LDHA transcription to impact tumor radiosensitivity. Cell Death Dis. 2024, 15, 209. [Google Scholar] [CrossRef] [PubMed]
  39. Burdak-Rothkamm, S.; Mansour, W.Y.; Rothkamm, K. DNA Damage Repair Deficiency in Prostate Cancer. Trends Cancer 2020, 6, 974–984. [Google Scholar] [CrossRef] [PubMed]
  40. Chappidi, N.; Quail, T.; Doll, S.; Vogel, L.T.; Aleksandrov, R.; Felekyan, S.; Kühnemuth, R.; Stoynov, S.; Seidel, C.A.M.; Brugués, J.; et al. PARP1-DNA co-condensation drives DNA repair site assembly to prevent disjunction of broken DNA ends. Cell 2024, 187, 945–961.e918. [Google Scholar] [CrossRef]
  41. Gai, X.; Xin, D.; Wu, D.; Wang, X.; Chen, L.; Wang, Y.; Ma, K.; Li, Q.; Li, P.; Yu, X. Pre-ribosomal RNA reorganizes DNA damage repair factors in nucleus during meiotic prophase and DNA damage response. Cell Res. 2022, 32, 254–268. [Google Scholar] [CrossRef] [PubMed]
  42. Huang, R.; Zhou, P.K. DNA damage repair: Historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct. Target. Ther. 2021, 6, 254. [Google Scholar] [CrossRef] [PubMed]
  43. D’Souza, L.C.; Shekher, A.; Challagundla, K.B.; Sharma, A.; Gupta, S.C. Reprogramming of glycolysis by chemical carcinogens during tumor development. Semin. Cancer Biol. 2022, 87, 127–136. [Google Scholar] [CrossRef]
  44. Weyand, C.M.; Goronzy, J.J. The immunology of rheumatoid arthritis. Nat. Immunol. 2021, 22, 10–18. [Google Scholar] [CrossRef]
  45. Sobanski, T.; Suraweera, A.; Burgess, J.T.; Richard, I.; Cheong, C.M.; Dave, K.; Rose, M.; Adams, M.N.; O’Byrne, K.J.; Richard, D.J.; et al. The fructose-bisphosphate, Aldolase A (ALDOA), facilitates DNA-PKcs and ATM kinase activity to regulate DNA double-strand break repair. Sci. Rep. 2023, 13, 15171. [Google Scholar] [CrossRef]
  46. Chen, H.; Li, Y.; Li, H.; Chen, X.; Fu, H.; Mao, D.; Chen, W.; Lan, L.; Wang, C.; Hu, K.; et al. NBS1 lactylation is required for efficient DNA repair and chemotherapy resistance. Nature 2024, 631, 663–669. [Google Scholar] [CrossRef] [PubMed]
  47. Wei, X.; Sun, K.; Li, S.; Lin, C.; Wei, Z. PSME3 induces radioresistance and enhances aerobic glycolysis in cervical cancer by regulating PARP1. Tissue Cell 2023, 83, 102151. [Google Scholar] [CrossRef]
  48. Wang, S.; Xie, J.; Li, J.; Liu, F.; Wu, X.; Wang, Z. Cisplatin suppresses the growth and proliferation of breast and cervical cancer cell lines by inhibiting integrin β5-mediated glycolysis. Am. J. Cancer Res. 2016, 6, 1108–1117. [Google Scholar]
  49. Hopp, A.K.; Grüter, P.; Hottiger, M.O. Regulation of Glucose Metabolism by NAD(+) and ADP-Ribosylation. Cells 2019, 8, 890. [Google Scholar] [CrossRef]
  50. Nagahisa, T.; Yamaguchi, S.; Kosugi, S.; Homma, K.; Miyashita, K.; Irie, J.; Yoshino, J.; Itoh, H. Intestinal Epithelial NAD+ Biosynthesis Regulates GLP-1 Production and Postprandial Glucose Metabolism in Mice. Endocrinology 2022, 163, bqac023. [Google Scholar] [CrossRef]
  51. Jeong, S.M.; Xiao, C.; Finley, L.W.; Lahusen, T.; Souza, A.L.; Pierce, K.; Li, Y.H.; Wang, X.; Laurent, G.; German, N.J.; et al. SIRT4 has tumor-suppressive activity and regulates the cellular metabolic response to DNA damage by inhibiting mitochondrial glutamine metabolism. Cancer Cell 2013, 23, 450–463. [Google Scholar] [CrossRef]
  52. Mukha, A.; Kahya, U.; Dubrovska, A. Targeting glutamine metabolism and autophagy: The combination for prostate cancer radiosensitization. Autophagy 2021, 17, 3879–3881. [Google Scholar] [CrossRef]
  53. Fu, S.; Li, Z.; Xiao, L.; Hu, W.; Zhang, L.; Xie, B.; Zhou, Q.; He, J.; Qiu, Y.; Wen, M.; et al. Glutamine Synthetase Promotes Radiation Resistance via Facilitating Nucleotide Metabolism and Subsequent DNA Damage Repair. Cell Rep. 2019, 28, 1136–1143.e1134. [Google Scholar] [CrossRef] [PubMed]
  54. Cezanne, A.; Foo, S.; Kuo, Y.W.; Baum, B. The Archaeal Cell Cycle. Annu. Rev. Cell Dev. Biol. 2024, 40, 1–23. [Google Scholar] [CrossRef] [PubMed]
  55. Coffman, J.A. Cell cycle development. Dev. Cell 2004, 6, 321–327. [Google Scholar] [CrossRef] [PubMed]
  56. Dalton, S. Linking the Cell Cycle to Cell Fate Decisions. Trends Cell Biol. 2015, 25, 592–600. [Google Scholar] [CrossRef]
  57. Weston, W.A.; Barr, A.R. A cell cycle centric view of tumour dormancy. Br. J. Cancer 2023, 129, 1535–1545. [Google Scholar] [CrossRef]
  58. Jirawatnotai, S.; Dalton, S.; Wattanapanitch, M. Role of cyclins and cyclin-dependent kinases in pluripotent stem cells and their potential as a therapeutic target. Semin. Cell Dev. Biol. 2020, 107, 63–71. [Google Scholar] [CrossRef]
  59. McKenney, C.; Lendner, Y.; Guerrero Zuniga, A.; Sinha, N.; Veresko, B.; Aikin, T.J.; Regot, S. CDK4/6 activity is required during G(2) arrest to prevent stress-induced endoreplication. Science 2024, 384, eadi2421. [Google Scholar] [CrossRef]
  60. Li, Z.; Peng, Y.; Li, J.; Chen, Z.; Chen, F.; Tu, J.; Lin, S.; Wang, H. N(6)-methyladenosine regulates glycolysis of cancer cells through PDK4. Nat. Commun. 2020, 11, 2578. [Google Scholar] [CrossRef]
  61. Luengo, A.; Li, Z.; Gui, D.Y.; Sullivan, L.B.; Zagorulya, M.; Do, B.T.; Ferreira, R.; Naamati, A.; Ali, A.; Lewis, C.A.; et al. Increased demand for NAD(+) relative to ATP drives aerobic glycolysis. Mol. Cell 2021, 81, 691–707.e696. [Google Scholar] [CrossRef]
  62. Daems, S.; Shameer, S.; Ceusters, N.; Sweetlove, L.; Ceusters, J. Metabolic modelling identifies mitochondrial Pi uptake and pyruvate efflux as key aspects of daytime metabolism and proton homeostasis in crassulacean acid metabolism leaves. New Phytol. 2024, 244, 159–175. [Google Scholar] [CrossRef] [PubMed]
  63. Du, D.; Liu, C.; Qin, M.; Zhang, X.; Xi, T.; Yuan, S.; Hao, H.; Xiong, J. Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm. Sin. B 2022, 12, 558–580. [Google Scholar] [CrossRef] [PubMed]
  64. Gouzy, A.; Healy, C.; Black, K.A.; Rhee, K.Y.; Ehrt, S. Growth of Mycobacterium tuberculosis at acidic pH depends on lipid assimilation and is accompanied by reduced GAPDH activity. Proc. Natl. Acad. Sci. USA 2021, 118, e2024571118. [Google Scholar] [CrossRef] [PubMed]
  65. Moreno, J.C.; Rojas, B.E.; Vicente, R.; Gorka, M.; Matz, T.; Chodasiewicz, M.; Peralta-Ariza, J.S.; Zhang, Y.; Alseekh, S.; Childs, D.; et al. Tyr-Asp inhibition of glyceraldehyde 3-phosphate dehydrogenase affects plant redox metabolism. EMBO J. 2021, 40, e106800. [Google Scholar] [CrossRef]
  66. Zhang, Y.; Liu, L.; Qi, Y.; Lou, J.; Chen, Y.; Liu, C.; Li, H.; Chang, X.; Hu, Z.; Li, Y.; et al. Lactic acid promotes nucleus pulposus cell senescence and corresponding intervertebral disc degeneration via interacting with Akt. Cell. Mol. Life Sci. CMLS 2024, 81, 24. [Google Scholar] [CrossRef]
  67. Leng, L.; Yuan, Z.; Pan, R.; Su, X.; Wang, H.; Xue, J.; Zhuang, K.; Gao, J.; Chen, Z.; Lin, H.; et al. Microglial hexokinase 2 deficiency increases ATP generation through lipid metabolism leading to β-amyloid clearance. Nat. Metab. 2022, 4, 1287–1305. [Google Scholar] [CrossRef]
  68. Pellarin, I.; Dall’Acqua, A.; Favero, A.; Segatto, I.; Rossi, V.; Crestan, N.; Karimbayli, J.; Belletti, B.; Baldassarre, G. Cyclin-dependent protein kinases and cell cycle regulation in biology and disease. Signal Transduct. Target. Ther. 2025, 10, 11. [Google Scholar] [CrossRef]
  69. Zheng, D.; Deng, Y.; Deng, L.; He, Z.; Sun, X.; Gong, Y.; Shi, B.; Lu, D.; Yu, C. CDCA7 enhances STAT3 transcriptional activity to regulate aerobic glycolysis and promote pancreatic cancer progression and gemcitabine resistance. Cell Death Dis. 2025, 16, 68. [Google Scholar] [CrossRef]
  70. Delaney, G.; Jacob, S.; Featherstone, C.; Barton, M. The role of radiotherapy in cancer treatment: Estimating optimal utilization from a review of evidence-based clinical guidelines. Cancer 2005, 104, 1129–1137. [Google Scholar] [CrossRef]
  71. Baumann, M.; Krause, M.; Overgaard, J.; Debus, J.; Bentzen, S.M.; Daartz, J.; Richter, C.; Zips, D.; Bortfeld, T. Radiation oncology in the era of precision medicine. Nat. Rev. Cancer 2016, 16, 234–249. [Google Scholar] [CrossRef]
  72. Barker, H.E.; Paget, J.T.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef]
  73. Gao, Y.; Gao, J.; Lin, F.; Wang, T.; Huo, S.; Wu, J.; Zhou, Q.; Zhang, C. CircRNAs in Tumor Radioresistance. Biomolecules 2022, 12, 1586. [Google Scholar] [CrossRef]
  74. Huang, R.X.; Zhou, P.K. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct. Target. Ther. 2020, 5, 60. [Google Scholar] [CrossRef]
  75. Porrazzo, A.; Cassandri, M.; D’Alessandro, A.; Morciano, P.; Rota, R.; Marampon, F.; Cenci, G. DNA repair in tumor radioresistance: Insights from fruit flies genetics. Cell. Oncol. 2024, 47, 717–732. [Google Scholar] [CrossRef] [PubMed]
  76. Jiang, K.; Yin, X.; Zhang, Q.; Yin, J.; Tang, Q.; Xu, M.; Wu, L.; Shen, Y.; Zhou, Z.; Yu, H.; et al. STC2 activates PRMT5 to induce radioresistance through DNA damage repair and ferroptosis pathways in esophageal squamous cell carcinoma. Redox Biol. 2023, 60, 102626. [Google Scholar] [CrossRef] [PubMed]
  77. Velegzhaninov, I.O.; Belykh, E.S.; Rasova, E.E.; Pylina, Y.I.; Shadrin, D.M.; Klokov, D.Y. Radioresistance, DNA Damage and DNA Repair in Cells With Moderate Overexpression of RPA1. Front. Genet. 2020, 11, 855. [Google Scholar] [CrossRef]
  78. Zhang, J.; Wu, Q.; Zhu, L.; Xie, S.; Tu, L.; Yang, Y.; Wu, K.; Zhao, Y.; Wang, Y.; Xu, Y.; et al. SERPINE2/PN-1 regulates the DNA damage response and radioresistance by activating ATM in lung cancer. Cancer Lett. 2022, 524, 268–283. [Google Scholar] [CrossRef] [PubMed]
  79. Pagliari, F.; Jansen, J.; Knoll, J.; Hanley, R.; Seco, J.; Tirinato, L. Cancer radioresistance is characterized by a differential lipid droplet content along the cell cycle. Cell Div. 2024, 19, 14. [Google Scholar] [CrossRef]
  80. Sato, Y.; Yoshino, H.; Sato, K.; Kashiwakura, I.; Tsuruga, E. DAP3-mediated cell cycle regulation and its association with radioresistance in human lung adenocarcinoma cell lines. J. Radiat. Res. 2023, 64, 520–529. [Google Scholar] [CrossRef]
  81. Vlatkovic, T.; Veldwijk, M.R.; Giordano, F.A.; Herskind, C. Targeting Cell Cycle Checkpoint Kinases to Overcome Intrinsic Radioresistance in Brain Tumor Cells. Cancers 2022, 14, 701. [Google Scholar] [CrossRef] [PubMed]
  82. Hoque, S.; Dhar, R.; Kar, R.; Mukherjee, S.; Mukherjee, D.; Mukerjee, N.; Nag, S.; Tomar, N.; Mallik, S. Cancer stem cells (CSCs): Key player of radiotherapy resistance and its clinical significance. Biomark. Biochem. Indic. Exp. Response Susceptibility Chem. 2023, 28, 139–151. [Google Scholar] [CrossRef] [PubMed]
  83. Olivares-Urbano, M.A.; Griñán-Lisón, C.; Marchal, J.A.; Núñez, M.I. CSC Radioresistance: A Therapeutic Challenge to Improve Radiotherapy Effectiveness in Cancer. Cells 2020, 9, 1651. [Google Scholar] [CrossRef]
  84. Zhan, Y.; Fan, S. Multiple Mechanisms Involving in Radioresistance of Nasopharyngeal Carcinoma. J. Cancer 2020, 11, 4193–4204. [Google Scholar] [CrossRef] [PubMed]
  85. Zhou, T.; Zhang, L.Y.; He, J.Z.; Miao, Z.M.; Li, Y.Y.; Zhang, Y.M.; Liu, Z.W.; Zhang, S.Z.; Chen, Y.; Zhou, G.C.; et al. Review: Mechanisms and perspective treatment of radioresistance in non-small cell lung cancer. Front. Immunol. 2023, 14, 1133899. [Google Scholar] [CrossRef]
  86. De Martino, M.; Daviaud, C.; Hajjar, E.; Vanpouille-Box, C. Fatty acid metabolism and radiation-induced anti-tumor immunity. Int. Rev. Cell Mol. Biol. 2023, 376, 121–141. [Google Scholar] [CrossRef]
  87. Chen, Y.; Zhou, S.; Wan, K.; Yu, L.; Zhao, C.; Deng, H.; Ou, Q.; Qin, J.; Hu, J.; Hou, Z. RIOK1 mediates p53 degradation and radioresistance in colorectal cancer through phosphorylation of G3BP2. Oncogene 2022, 41, 3433–3444. [Google Scholar] [CrossRef]
  88. Lambing, S.; Tan, Y.P.; Vasileiadou, P.; Holdenrieder, S.; Müller, P.; Hagen, C.; Garbe, S.; Behrendt, R.; Schlee, M.; van den Boorn, J.G.; et al. RIG-I immunotherapy overcomes radioresistance in p53-positive malignant melanoma. J. Mol. Cell Biol. 2023, 15, mjad001. [Google Scholar] [CrossRef] [PubMed]
  89. Li, J.; Zhang, X.; Hou, Z.; Cai, S.; Guo, Y.; Sun, L.; Li, A.; Li, Q.; Wang, E.; Miao, Y. P130cas-FAK interaction is essential for YAP-mediated radioresistance of non-small cell lung cancer. Cell Death Dis. 2022, 13, 783. [Google Scholar] [CrossRef]
  90. Bailleul, J.; Ruan, Y.; Abdulrahman, L.; Scott, A.J.; Yazal, T.; Sung, D.; Park, K.; Hoang, H.; Nathaniel, J.; Chu, F.I.; et al. M2 isoform of pyruvate kinase rewires glucose metabolism during radiation therapy to promote an antioxidant response and glioblastoma radioresistance. Neuro-Oncology 2023, 25, 1989–2000. [Google Scholar] [CrossRef]
  91. Bie, J.; Li, R.; Li, Y.; Song, C.; Chen, Z.; Zhang, T.; Tang, Z.; Su, L.; Zhu, L.; Wang, J.; et al. PKM2 aggregation drives metabolism reprograming during aging process. Nat. Commun. 2024, 15, 5761. [Google Scholar] [CrossRef]
  92. Qin, C.; Yang, G.; Yang, J.; Ren, B.; Wang, H.; Chen, G.; Zhao, F.; You, L.; Wang, W.; Zhao, Y. Metabolism of pancreatic cancer: Paving the way to better anticancer strategies. Mol. Cancer 2020, 19, 50. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, H.; Jiang, H.; Van De Gucht, M.; De Ridder, M. Hypoxic Radioresistance: Can ROS Be the Key to Overcome It? Cancers 2019, 11, 112. [Google Scholar] [CrossRef] [PubMed]
  94. Li, G.; Wang, D.; Zhai, Y.; Pan, C.; Zhang, J.; Wang, C.; Huang, R.; Yu, M.; Li, Y.; Liu, X.; et al. Glycometabolic reprogramming-induced XRCC1 lactylation confers therapeutic resistance in ALDH1A3-overexpressing glioblastoma. Cell Metab. 2024, 36, 1696–1710.e1610. [Google Scholar] [CrossRef]
  95. Jin, X.; Kuang, Y.; Li, L.; Li, H.; Zhao, T.; He, Y.; Di, C.; Kang, J.; Yuan, L.; Yu, B.; et al. A positive feedback circuit comprising p21 and HIF-1α aggravates hypoxia-induced radioresistance of glioblastoma by promoting Glut1/LDHA-mediated glycolysis. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2022, 36, e22229. [Google Scholar] [CrossRef]
  96. Koukourakis, M.; Tsolou, A.; Pouliliou, S.; Lamprou, I.; Papadopoulou, M.; Ilemosoglou, M.; Kostoglou, G.; Ananiadou, D.; Sivridis, E.; Giatromanolaki, A. Blocking LDHA glycolytic pathway sensitizes glioblastoma cells to radiation and temozolomide. Biochem. Biophys. Res. Commun. 2017, 491, 932–938. [Google Scholar] [CrossRef]
  97. Ning, K.; Liu, S.; Yang, B.; Wang, R.; Man, G.; Wang, D.E.; Xu, H. Update on the effects of energy metabolism in bone marrow mesenchymal stem cells differentiation. Mol. Metab. 2022, 58, 101450. [Google Scholar] [CrossRef]
  98. Liu, F.; Wang, B.; Shan, Y.; Kang, W.; Li, J.; Wang, X.; Liu, J. Quantitative and Selective DNA Detection with Portable Personal Glucose Meter Using Loop-Based DNA Competitive Hybridization Strategy. Methods Mol. Biol. 2022, 2393, 473–478. [Google Scholar] [CrossRef]
  99. Walton, E.L. Oxidative stress and diabetes: Glucose response in the cROSsfire. Biomed. J. 2017, 40, 241–244. [Google Scholar] [CrossRef]
  100. Ye, L.; Jiang, Y.; Zhang, M. Crosstalk between glucose metabolism, lactate production and immune response modulation. Cytokine Growth Factor Rev. 2022, 68, 81–92. [Google Scholar] [CrossRef] [PubMed]
  101. Wu, S.; Cao, R.; Tao, B.; Wu, P.; Peng, C.; Gao, H.; Liang, J.; Yang, W. Pyruvate Facilitates FACT-Mediated γH2AX Loading to Chromatin and Promotes the Radiation Resistance of Glioblastoma. Adv. Sci. 2022, 9, e2104055. [Google Scholar] [CrossRef]
  102. Li, T.; Han, J.; Jia, L.; Hu, X.; Chen, L.; Wang, Y. PKM2 coordinates glycolysis with mitochondrial fusion and oxidative phosphorylation. Protein Cell 2019, 10, 583–594. [Google Scholar] [CrossRef]
  103. Wang, J.; Yang, P.; Yu, T.; Gao, M.; Liu, D.; Zhang, J.; Lu, C.; Chen, X.; Zhang, X.; Liu, Y. Lactylation of PKM2 Suppresses Inflammatory Metabolic Adaptation in Pro-inflammatory Macrophages. Int. J. Biol. Sci. 2022, 18, 6210–6225. [Google Scholar] [CrossRef]
  104. Wu, Y.; Tang, L.; Huang, H.; Yu, Q.; Hu, B.; Wang, G.; Ge, F.; Yin, T.; Li, S.; Yu, X. Phosphoglycerate dehydrogenase activates PKM2 to phosphorylate histone H3T11 and attenuate cellular senescence. Nat. Commun. 2023, 14, 1323. [Google Scholar] [CrossRef]
  105. Yu, S.; Zang, W.; Qiu, Y.; Liao, L.; Zheng, X. Deubiquitinase OTUB2 exacerbates the progression of colorectal cancer by promoting PKM2 activity and glycolysis. Oncogene 2022, 41, 46–56. [Google Scholar] [CrossRef]
  106. Wu, H.; Sun, X.; Li, K.; Li, J.; Jiang, H.; Yan, D.; Lin, Y.; Ding, Y.; Lu, Y.; Zhu, X.; et al. Pyruvate Kinase M2-Responsive Release of Paclitaxel and Indoleamine 2,3-Dioxygenase Inhibitor for Immuno-Chemotherapy of Nonsmall Cell Lung Cancer. Adv. Sci. 2025, 12, e2409790. [Google Scholar] [CrossRef]
  107. Elia, I.; Rowe, J.H.; Johnson, S.; Joshi, S.; Notarangelo, G.; Kurmi, K.; Weiss, S.; Freeman, G.J.; Sharpe, A.H.; Haigis, M.C. Tumor cells dictate anti-tumor immune responses by altering pyruvate utilization and succinate signaling in CD8(+) T cells. Cell Metab. 2022, 34, 1137–1150.e1136. [Google Scholar] [CrossRef] [PubMed]
  108. Bakshi, I.; Suryana, E.; Small, L.; Quek, L.E.; Brandon, A.E.; Turner, N.; Cooney, G.J. Fructose bisphosphatase 2 overexpression increases glucose uptake in skeletal muscle. J. Endocrinol. 2018, 237, 101–111. [Google Scholar] [CrossRef] [PubMed]
  109. Fang, Y.; Zhan, Y.; Xie, Y.; Du, S.; Chen, Y.; Zeng, Z.; Zhang, Y.; Chen, K.; Wang, Y.; Liang, L.; et al. Integration of glucose and cardiolipin anabolism confers radiation resistance of HCC. Hepatology 2022, 75, 1386–1401. [Google Scholar] [CrossRef] [PubMed]
  110. An, L.; Li, M.; Jia, Q. Mechanisms of radiotherapy resistance and radiosensitization strategies for esophageal squamous cell carcinoma. Mol. Cancer 2023, 22, 140. [Google Scholar] [CrossRef] [PubMed]
  111. Chen, H.Y.; Xu, L.; Li, L.F.; Liu, X.X.; Gao, J.X.; Bai, Y.R. Inhibiting the CD8(+) T cell infiltration in the tumor microenvironment after radiotherapy is an important mechanism of radioresistance. Sci. Rep. 2018, 8, 11934. [Google Scholar] [CrossRef]
  112. Gu, H.; Huang, T.; Shen, Y.; Liu, Y.; Zhou, F.; Jin, Y.; Sattar, H.; Wei, Y. Reactive Oxygen Species-Mediated Tumor Microenvironment Transformation: The Mechanism of Radioresistant Gastric Cancer. Oxidative Med. Cell. Longev. 2018, 2018, 5801209. [Google Scholar] [CrossRef]
  113. Kim, H.; Lin, Q.; Yun, Z. The hypoxic tumor microenvironment in vivo selects tumor cells with increased survival against genotoxic stresses. Cancer Lett. 2018, 431, 142–149. [Google Scholar] [CrossRef] [PubMed]
  114. Dou, Y.; Liu, Y.; Zhao, F.; Guo, Y.; Li, X.; Wu, M.; Chang, J.; Yu, C. Radiation-responsive scintillating nanotheranostics for reduced hypoxic radioresistance under ROS/NO-mediated tumor microenvironment regulation. Theranostics 2018, 8, 5870–5889. [Google Scholar] [CrossRef] [PubMed]
  115. Hu, C.; Jiang, X. Role of NRP-1 in VEGF-VEGFR2-Independent Tumorigenesis. Target. Oncol. 2016, 11, 501–505. [Google Scholar] [CrossRef]
  116. Jarosz-Biej, M.; Smolarczyk, R.; Cichoń, T.; Kułach, N. Tumor Microenvironment as A “Game Changer” in Cancer Radiotherapy. Int. J. Mol. Sci. 2019, 20, 3212. [Google Scholar] [CrossRef]
  117. Liu, J.; Li, Q.; Tu, H.; Feng, J.; Fan, X.; Mi, Z.; Han, H.; Ma, X.; Liang, Q.; Zhang, Z.; et al. Bismuth/manganese-doped carbon dots in calcium phosphate matrix: Dynamic conformational nanoparticles enhancing tumor accumulation, deep penetration, and radiosensitivity. Chem. Eng. J. 2025, 504, 158683. [Google Scholar] [CrossRef]
  118. Wang, Z.; Zhao, T.; Zhang, S.; Wang, J.; Chen, Y.; Zhao, H.; Yang, Y.; Shi, S.; Chen, Q.; Liu, K. The Wnt signaling pathway in tumorigenesis, pharmacological targets, and drug development for cancer therapy. Biomark. Res. 2021, 9, 68. [Google Scholar] [CrossRef]
  119. Chen, D.; Zhang, X.; Li, Z.; Zhu, B. Metabolic regulatory crosstalk between tumor microenvironment and tumor-associated macrophages. Theranostics 2021, 11, 1016–1030. [Google Scholar] [CrossRef] [PubMed]
  120. Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef]
  121. Pitt, J.M.; Marabelle, A.; Eggermont, A.; Soria, J.C.; Kroemer, G.; Zitvogel, L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2016, 27, 1482–1492. [Google Scholar] [CrossRef] [PubMed]
  122. Ocaña, M.C.; Martínez-Poveda, B.; Quesada, A.R.; Medina, M. Metabolism within the tumor microenvironment and its implication on cancer progression: An ongoing therapeutic target. Med. Res. Rev. 2019, 39, 70–113. [Google Scholar] [CrossRef]
  123. Zhu, L.; Zhu, X.; Wu, Y. Effects of Glucose Metabolism, Lipid Metabolism, and Glutamine Metabolism on Tumor Microenvironment and Clinical Implications. Biomolecules 2022, 12, 580. [Google Scholar] [CrossRef]
  124. Zou, W.; Green, D.R. Beggars banquet: Metabolism in the tumor immune microenvironment and cancer therapy. Cell Metab. 2023, 35, 1101–1113. [Google Scholar] [CrossRef]
  125. Jin, R.; Neufeld, L.; McGaha, T.L. Linking macrophage metabolism to function in the tumor microenvironment. Nat. Cancer 2025, 6, 239–252. [Google Scholar] [CrossRef]
  126. Chen, L.; Huang, L.; Gu, Y.; Cang, W.; Sun, P.; Xiang, Y. Lactate-Lactylation Hands between Metabolic Reprogramming and Immunosuppression. Int. J. Mol. Sci. 2022, 23, 1943. [Google Scholar] [CrossRef] [PubMed]
  127. Wu, M.; Niu, X.; Zhang, R.; Ping Xu, Z. Two-dimensional nanomaterials for tumor microenvironment modulation and anticancer therapy. Adv. Drug Deliv. Rev. 2022, 187, 114360. [Google Scholar] [CrossRef]
  128. Xing, H.; Li, X. Engineered Nanomaterials for Tumor Immune Microenvironment Modulation in Cancer Immunotherapy. Chemistry 2024, 30, e202400425. [Google Scholar] [CrossRef]
  129. Zhao, L.; Liu, H.; Xing, Y.; Wang, R.; Cheng, Z.; Lv, C.; Lv, Z.; Yu, F. Tumor Microenvironment-Specific Functional Nanomaterials for Biomedical Applications. J. Biomed. Nanotechnol. 2020, 16, 1325–1358. [Google Scholar] [CrossRef]
  130. Roh, J.; Im, M.; Chae, Y.; Kang, J.; Kim, W. The Involvement of Long Non-Coding RNAs in Glutamine-Metabolic Reprogramming and Therapeutic Resistance in Cancer. Int. J. Mol. Sci. 2022, 23, 4808. [Google Scholar] [CrossRef] [PubMed]
  131. Yang, Y.; Chong, Y.; Chen, M.; Dai, W.; Zhou, X.; Ji, Y.; Qiu, G.; Du, X. Targeting lactate dehydrogenase a improves radiotherapy efficacy in non-small cell lung cancer: From bedside to bench. J. Transl. Med. 2021, 19, 170. [Google Scholar] [CrossRef]
  132. Pan, B.S.; Hsu, C.C.; Wu, H.E.; Chen, Y.R.; Zhou, X.; Wang, S.C.; Li, C.Y.; Lin, H.K. Glucose metabolism and its direct action in cancer and immune regulation: Opportunities and challenges for metabolic targeting. J. Biomed. Sci. 2025, 32, 71. [Google Scholar] [CrossRef] [PubMed]
  133. Gähler, A.; Trufa, D.I.; Chiriac, M.T.; Tausche, P.; Hohenberger, K.; Brunst, A.K.; Rauh, M.; Geppert, C.I.; Rieker, R.J.; Krammer, S.; et al. Glucose-Restricted Diet Regulates the Tumor Immune Microenvironment and Prevents Tumor Growth in Lung Adenocarcinoma. Front. Oncol. 2022, 12, 873293. [Google Scholar] [CrossRef]
  134. Zhang, L.; Jiang, C.; Zhong, Y.; Sun, K.; Jing, H.; Song, J.; Xie, J.; Zhou, Y.; Tian, M.; Zhang, C.; et al. STING is a cell-intrinsic metabolic checkpoint restricting aerobic glycolysis by targeting HK2. Nat. Cell Biol. 2023, 25, 1208–1222. [Google Scholar] [CrossRef]
  135. Cook, K.M.; Shen, H.; McKelvey, K.J.; Gee, H.E.; Hau, E. Targeting Glucose Metabolism of Cancer Cells with Dichloroacetate to Radiosensitize High-Grade Gliomas. Int. J. Mol. Sci. 2021, 22, 7265. [Google Scholar] [CrossRef] [PubMed]
  136. Floberg, J.M.; Schwarz, J.K. Manipulation of Glucose and Hydroperoxide Metabolism to Improve Radiation Response. Semin. Radiat. Oncol. 2019, 29, 33–41. [Google Scholar] [CrossRef] [PubMed]
  137. Lin, J.; Xia, L.; Liang, J.; Han, Y.; Wang, H.; Oyang, L.; Tan, S.; Tian, Y.; Rao, S.; Chen, X.; et al. The roles of glucose metabolic reprogramming in chemo- and radio-resistance. J. Exp. Clin. Cancer Res. CR 2019, 38, 218. [Google Scholar] [CrossRef]
  138. Cerna, D.; Camphausen, K.; Tofilon, P.J. Histone deacetylation as a target for radiosensitization. Curr. Top. Dev. Biol. 2006, 73, 173–204. [Google Scholar] [CrossRef]
  139. Dai, L.B.; Zhong, J.T.; Shen, L.F.; Zhou, S.H.; Lu, Z.J.; Bao, Y.Y.; Fan, J. Radiosensitizing effects of curcumin alone or combined with GLUT1 siRNA on laryngeal carcinoma cells through AMPK pathway-induced autophagy. J. Cell. Mol. Med. 2021, 13, 6018–6031. [Google Scholar] [CrossRef]
  140. Hao, J.; Graham, P.; Chang, L.; Ni, J.; Wasinger, V.; Beretov, J.; Deng, J.; Duan, W.; Bucci, J.; Malouf, D.; et al. Proteomic identification of the lactate dehydrogenase A in a radioresistant prostate cancer xenograft mouse model for improving radiotherapy. Oncotarget 2016, 7, 74269–74285. [Google Scholar] [CrossRef]
  141. Pedersen, P.L. 3-Bromopyruvate (3BP) a fast acting, promising, powerful, specific, and effective “small molecule” anti-cancer agent taken from labside to bedside: Introduction to a special issue. J. Bioenerg. Biomembr. 2012, 44, 1–6. [Google Scholar] [CrossRef]
  142. Li, X.; Xie, L.; Zhou, L.; Gan, Y.; Han, S.; Zhou, Y.; Qing, X.; Li, W. Bergenin Inhibits Tumor Growth and Overcomes Radioresistance by Targeting Aerobic Glycolysis. Am. J. Chin. Med. 2023, 51, 1905–1925. [Google Scholar] [CrossRef]
  143. Zheng, Y.; Zhan, Y.; Zhang, Y.; Zhang, Y.; Liu, Y.; Xie, Y.; Sun, Y.; Qian, J.; Ding, Y.; Ding, Y.; et al. Hexokinase 2 confers radio-resistance in hepatocellular carcinoma by promoting autophagy-dependent degradation of AIMP2. Cell Death Dis. 2023, 14, 488. [Google Scholar] [CrossRef]
  144. Morais, M.; Dias, F.; Teixeira, A.L.; Medeiros, R. MicroRNAs and altered metabolism of clear cell renal cell carcinoma: Potential role as aerobic glycolysis biomarkers. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 2175–2185. [Google Scholar] [CrossRef]
  145. Kobayashi, H.; Takase, S.; Nishimura, H.; Matsumoto, K.; Harada, H.; Yoshida, M. RNAi screening reveals a synthetic chemical-genetic interaction between ATP synthase and PFK1 in cancer cells. Cancer Sci. 2023, 114, 1663–1671. [Google Scholar] [CrossRef] [PubMed]
  146. Huang, B.; Wang, Q.; Jiang, L.; Lu, S.; Li, C.; Xu, C.; Wang, C.; Zhang, E.; Zhang, X. Shikonin ameliorated mice colitis by inhibiting dimerization and tetramerization of PKM2 in macrophages. Front. Pharmacol. 2022, 13, 926945. [Google Scholar] [CrossRef] [PubMed]
  147. Chen, D.Q.; Han, J.; Liu, H.; Feng, K.; Li, P. Targeting pyruvate kinase M2 for the treatment of kidney disease. Front. Pharmacol. 2024, 15, 1376252. [Google Scholar] [CrossRef] [PubMed]
  148. Palsson-McDermott, E.M.; Curtis, A.M.; Goel, G.; Lauterbach, M.A.; Sheedy, F.J.; Gleeson, L.E.; van den Bosch, M.W.; Quinn, S.R.; Domingo-Fernandez, R.; Johnston, D.G.; et al. Pyruvate kinase M2 regulates Hif-1α activity and IL-1β induction and is a critical determinant of the warburg effect in LPS-activated macrophages. Cell Metab. 2015, 21, 65–80. [Google Scholar] [CrossRef]
  149. Terlizzi, C.; De Rosa, V.; Iommelli, F.; Pezone, A.; Altobelli, G.G.; Maddalena, M.; Dimitrov, J.; De Rosa, C.; Della Corte, C.M.; Avvedimento, V.E.; et al. ATM inhibition blocks glucose metabolism and amplifies the sensitivity of resistant lung cancer cell lines to oncogene driver inhibitors. Cancer Metab. 2023, 11, 20. [Google Scholar] [CrossRef]
  150. Yang, J.; Li, S.; Li, Z.; Yao, L.; Liu, M.; Tong, K.L.; Xu, Q.; Yu, B.; Peng, R.; Gui, T.; et al. Targeting YAP1-regulated Glycolysis in Fibroblast-Like Synoviocytes Impairs Macrophage Infiltration to Ameliorate Diabetic Osteoarthritis Progression. Adv. Sci. 2024, 11, e2304617. [Google Scholar] [CrossRef]
  151. Raez, L.E.; Papadopoulos, K.; Ricart, A.D.; Chiorean, E.G.; Dipaola, R.S.; Stein, M.N.; Rocha Lima, C.M.; Schlesselman, J.J.; Tolba, K.; Langmuir, V.K.; et al. A phase I dose-escalation trial of 2-deoxy-D-glucose alone or combined with docetaxel in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2013, 71, 523–530. [Google Scholar] [CrossRef]
  152. Wang, S.; Du, Y.; Zhang, B.; Meng, G.; Liu, Z.; Liew, S.Y.; Liang, R.; Zhang, Z.; Cai, X.; Wu, S.; et al. Transplantation of chemically induced pluripotent stem-cell-derived islets under abdominal anterior rectus sheath in a type 1 diabetes patient. Cell 2024, 187, 6152–6164.e6118. [Google Scholar] [CrossRef]
  153. Nguyen, H.P.; An, K.; Ito, Y.; Kharbikar, B.N.; Sheng, R.; Paredes, B.; Murray, E.; Pham, K.; Bruck, M.; Zhou, X.; et al. Implantation of engineered adipocytes suppresses tumor progression in cancer models. Nat. Biotechnol. 2025, 1–17. [Google Scholar] [CrossRef] [PubMed]
  154. Gach, K.; Grądzka, I.; Wasyk, I.; Męczyńska-Wielgosz, S.; Iwaneńko, T.; Szymański, J.; Koszuk, J.; Janecki, T.; Kruszewski, M.; Janecka, A. Anticancer activity and radiosensitization effect of methyleneisoxazolidin-5-ones in hepatocellular carcinoma HepG2 cells. Chem.-Biol. Interact. 2016, 248, 68–73. [Google Scholar] [CrossRef]
  155. Yadav, D.; Yadav, A.; Bhattacharya, S.; Dagar, A.; Kumar, V.; Rani, R. GLUT and HK: Two primary and essential key players in tumor glycolysis. Semin. Cancer Biol. 2024, 100, 17–27. [Google Scholar] [CrossRef]
  156. Zuo, J.; Tang, J.; Lu, M.; Zhou, Z.; Li, Y.; Tian, H.; Liu, E.; Gao, B.; Liu, T.; Shao, P. Glycolysis Rate-Limiting Enzymes: Novel Potential Regulators of Rheumatoid Arthritis Pathogenesis. Front. Immunol. 2021, 12, 779787. [Google Scholar] [CrossRef]
  157. Fernandes, P.M.; Kinkead, J.; McNae, I.W.; Vásquez-Valdivieso, M.; Wear, M.A.; Michels, P.A.M.; Walkinshaw, M.D. Kinetic and structural studies of Trypanosoma and Leishmania phosphofructokinases show evolutionary divergence and identify AMP as a switch regulating glycolysis versus gluconeogenesis. FEBS J. 2020, 287, 2847–2861. [Google Scholar] [CrossRef]
  158. Funato, Y.; Hayashi, T.; Irino, Y.; Takenawa, T.; Miki, H. Nucleoredoxin regulates glucose metabolism via phosphofructokinase 1. Biochem. Biophys. Res. Commun. 2013, 440, 737–742. [Google Scholar] [CrossRef]
  159. Kloos, M.; Brüser, A.; Kirchberger, J.; Schöneberg, T.; Sträter, N. Crystal structure of human platelet phosphofructokinase-1 locked in an activated conformation. Biochem. J. 2015, 469, 421–432. [Google Scholar] [CrossRef] [PubMed]
  160. Song, J.; Sun, H.; Zhang, S.; Shan, C. The Multiple Roles of Glucose-6-Phosphate Dehydrogenase in Tumorigenesis and Cancer Chemoresistance. Life 2022, 12, 271. [Google Scholar] [CrossRef] [PubMed]
  161. Ishaq, A.R.; Younis, T.; Lin, S.; Usman, M.; Wang, T.; Chen, Z.S. Phosphofructokinase-1 in Cancer: A Promising Target for Diagnosis and Therapy. Recent Pat. Anti-Cancer Drug Discov. 2024, 20, 667–680. [Google Scholar] [CrossRef]
  162. Lynch, E.M.; Hansen, H.; Salay, L.; Cooper, M.; Timr, S.; Kollman, J.M.; Webb, B.A. Structural basis for allosteric regulation of human phosphofructokinase-1. Nat. Commun. 2024, 15, 7323. [Google Scholar] [CrossRef]
  163. Horemans, S.; Pitoulias, M.; Holland, A.; Pateau, E.; Lechaplais, C.; Ekaterina, D.; Perret, A.; Soultanas, P.; Janniere, L. Pyruvate kinase, a metabolic sensor powering glycolysis, drives the metabolic control of DNA replication. BMC Biol. 2022, 20, 87. [Google Scholar] [CrossRef]
  164. Israelsen, W.J.; Vander Heiden, M.G. Pyruvate kinase: Function, regulation and role in cancer. Semin. Cell Dev. Biol. 2015, 43, 43–51. [Google Scholar] [CrossRef]
  165. Feng, J.; Dai, W.; Mao, Y.; Wu, L.; Li, J.; Chen, K.; Yu, Q.; Kong, R.; Li, S.; Zhang, J.; et al. Simvastatin re-sensitizes hepatocellular carcinoma cells to sorafenib by inhibiting HIF-1α/PPAR-γ/PKM2-mediated glycolysis. J. Exp. Clin. Cancer Res. CR 2020, 39, 24. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, Y.; Shu, H.; Qu, Y.; Jin, X.; Liu, J.; Peng, W.; Wang, L.; Hao, M.; Xia, M.; Zhao, Z.; et al. PKM2 functions as a histidine kinase to phosphorylate PGAM1 and increase glycolysis shunts in cancer. EMBO J. 2024, 43, 2368–2396. [Google Scholar] [CrossRef] [PubMed]
  167. Zahra, K.; Dey, T.; Ashish; Mishra, S.P.; Pandey, U. Pyruvate Kinase M2 and Cancer: The Role of PKM2 in Promoting Tumorigenesis. Front. Oncol. 2020, 10, 159. [Google Scholar] [CrossRef]
  168. Wu, Y.H.; Yang, Y.; Chen, C.H.; Hsiao, C.J.; Li, T.N.; Liao, K.J.; Watashi, K.; Chen, B.S.; Wang, L.H. Aerobic glycolysis supports hepatitis B virus protein synthesis through interaction between viral surface antigen and pyruvate kinase isoform M2. PLoS Pathog. 2021, 17, e1008866. [Google Scholar] [CrossRef] [PubMed]
  169. Zhong, J.T.; Yu, Q.; Zhou, S.H.; Yu, E.; Bao, Y.Y.; Lu, Z.J.; Fan, J. GLUT-1 siRNA Enhances Radiosensitization Of Laryngeal Cancer Stem Cells Via Enhanced DNA Damage, Cell Cycle Redistribution, And Promotion Of Apoptosis In Vitro And In Vivo. OncoTargets Ther. 2019, 12, 9129–9142. [Google Scholar] [CrossRef]
Figure 1. Factors affecting tumor radioresistance.
Figure 1. Factors affecting tumor radioresistance.
Metabolites 15 00793 g001
Table 1. Summary of glycolysis-targeting therapeutic strategies and clinical applications.
Table 1. Summary of glycolysis-targeting therapeutic strategies and clinical applications.
CategoryTargetStrategy TypeSpecific Strategy/CompoundClinical StageReference
Enzyme-Targeted StrategiesGLUT1Small molecule inhibitorOleanolic acidPreclinical[13]
Genetic interventionsiRNA knockdown of GLUT1Preclinical[139]
LDHANano technology Mostly experimental[140]
Genetic interventionsiRNA knockdown of LDHAMostly experimental[38]
HK2Small molecule inhibitor3BP *, BergeninPreclinical[141,142]
Small molecule inhibitorKetoconazolePreclinical[143]
PFK-1Small molecule inhibitor3PO, Tryptolinamide (TLAM)Preclinical[144,145]
PKM2Small molecule inhibitorShikoninPreclinical[146]
Small molecule activatorTEPP-46, DASA-58Preclinical[147,148]
Genetic interventionsiRNA knockdown of PKM2Mostly experimental[94,149]
Platform and
Combination
Strategies
MultipleNanocarrier deliveryLDHA/GLUT1-targeting siRNAMostly experimental[140,150]
Broad-spectrum inhibition2-DGClinical trials[151]
Stem cell technologyCiPSCsClinical trials[152]
Metabolic–immune synergyCRISPR activation promotes
browning of adipocytes
Mostly experimental[153]
Combination therapyRadiotherapy and glycolysis
inhibitors
Preclinical to clinical translation[154]
* 3BP = 3-bromopyruvate; 3PO = 3-(3-pyridyl)-1-(4-pyridyl)-2-propen-1-one; CiPSCs = chemically induced pluripotent stem cells.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gao, S.; Liu, X.; Chen, S.; Zhou, P. Glucose Metabolism Modulation as a Strategy to Enhance Cancer Radiotherapy. Metabolites 2025, 15, 793. https://doi.org/10.3390/metabo15120793

AMA Style

Gao S, Liu X, Chen S, Zhou P. Glucose Metabolism Modulation as a Strategy to Enhance Cancer Radiotherapy. Metabolites. 2025; 15(12):793. https://doi.org/10.3390/metabo15120793

Chicago/Turabian Style

Gao, Shuaining, Xiaochang Liu, Shi Chen, and Pingkun Zhou. 2025. "Glucose Metabolism Modulation as a Strategy to Enhance Cancer Radiotherapy" Metabolites 15, no. 12: 793. https://doi.org/10.3390/metabo15120793

APA Style

Gao, S., Liu, X., Chen, S., & Zhou, P. (2025). Glucose Metabolism Modulation as a Strategy to Enhance Cancer Radiotherapy. Metabolites, 15(12), 793. https://doi.org/10.3390/metabo15120793

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

Article Metrics

Back to TopTop