International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Metabolic reprogramming is a fundamental characteristic of cancer development and progression (1), and it is primarily marked by a notable increase in aerobic glycolysis (2). This atypical activation of aerobic glycolysis not only meets the energy and biosynthetic demands of rapidly proliferating malignant tumor cells but also contributes to the formation of a hypoxic, acidic and nutrient-deprived tumor microenvironment (TME). Such an environment facilitates tumor immune evasion and presents a notable challenge to the antitumor immune response (3,4). Hexokinase 2 (HK2), the initial and critical rate-limiting enzyme in glycolysis, is markedly upregulated in various solid tumors [such as colorectal cancer (5), triple-negative breast cancer (6), hepatocellular carcinoma (7), pancreatic cancer (8), esophageal cancer (9), etc.], where it serves a pivotal role in catalyzing glycolytic processes and influencing the malignant behavior of tumors; it is also closely related to poor patient prognosis (10–18). The regulatory function of HK2 in immune evasion has increasingly attracted attention for research (3,19–21). Previous research indicates that the functions of HK2 encompass multiple immune escape mechanisms (3,20,22). The expression and post-translational modification of HK2 are modulated by a variety of signal transduction pathways and oncogenic factors, which further facilitate immune evasion through both glycolysis-dependent and independent mechanisms. These mechanisms impact immune cell distribution (23), modulate the expression of immune checkpoint molecules (20) and contribute to the establishment of an immunosuppressive microenvironment (3). Furthermore, HK2-mediated hyperactive glycolysis in tumor cells competes with immune cells for glucose, induces metabolic reprogramming in immune cells, suppresses the activation and function of immune cells, and releases large quantities of metabolites to establish an immunosuppressive niche, thereby impairing immune molecule expression and further weakening the antitumor immune response (24,25). Although small-molecule inhibitors [such as 3-bromopyruvic acid (26) and lonidamine (27)] and gene-editing tools that target HK2 can reverse metabolism-dependent immunosuppression, their clinical application is constrained by off-target toxicity, compensatory metabolism and delivery efficiency (21,28–30). Emerging strategies focusing on subcellular localization and non-catalytic intervention may offer novel directions to overcome these clinical translation bottlenecks (3).
At present, an expanding body of research has substantiated the multifaceted role of HK2 in bridging tumor metabolic reprogramming and immune evasion; however, comprehensive analyses remain limited. In light of this, the present review aims to summarize the research advancements regarding the regulation of immune escape in malignant tumors by HK2 and the role of HK2 in metabolic-immune interactions, with the objective of providing references and theoretical underpinnings for the development of novel immunotherapeutic strategies.
HK serves as the rate-limiting enzyme that initiates the glycolytic pathway by catalyzing the phosphorylation of glucose to glucose-6-phosphate (G-6-P), thereby facilitating energy production and biosynthesis (31). In mammals, five isoforms of HK have been identified, HK1-4 and HK domain-containing 1, with HK2 being markedly upregulated in malignancies, and strongly associated with tumor aerobic glycolysis and malignant progression (14,32). Unlike other isomers, HK2 has unique structural features: Although its secondary structure is very similar to that of HK1 and HK3, both composed of two symmetrical α-helical regions linked to glucokinase-catalyzed equivalent domains, HK2 is the only isomer that maintains catalytic activity in both domains (2), which allows for more efficient glycolysis (3). The N-terminal hydrophobic domain of HK2 directly anchors to the voltage-dependent anion channel (VDAC) located on the outer mitochondrial membrane. This crucial interaction results in the formation of a transmembrane-coupled complex with the adenine nucleotide translocator on the inner membrane. Such a configuration effectively prevents inhibition by G-6-P and establishes a ‘metabolic compartmentalization’ effect, thereby facilitating efficient coordination between glycolysis and mitochondrial energy metabolism within localized cellular regions (33). As a result, ATP produced by the mitochondria is directly utilized by HK2 to initiate glycolysis, creating a metabolic relay. This mechanism rapidly supplies the energy, metabolic intermediates and precursors necessary for tumor cell proliferation within a short period (34), thereby serving a pivotal role in cancer cell proliferation, drug resistance and immune evasion (35). Concurrently, HK2 localized to the mitochondrial outer membrane serves a critical role in inhibiting the opening of the mitochondrial permeability transition pore, stabilizing the mitochondrial membrane potential, suppressing the release of cytochrome c and blocking apoptotic signaling pathways, thereby facilitating tumor cell survival (36,37). Furthermore, HK2 is subject to O-GlcNAcylation, a process that enhances cell proliferation, migration and invasion in non-small cell lung cancer, and mitigates mitochondrial damage, and thus, facilitates tumor progression (38).
Activation of the PI3K/AKT signaling pathway facilitates cellular metabolic reprogramming (4) and is frequently associated with immunosuppressive effects (21). Research indicates that the AKT-induced nuclear translocation of HK2 enhances the transcription of programmed death-ligand 1 (PD-L1) through hypoxia-inducible factor 1-α (HIF-1α), which promotes immune evasion in gastric cancer cells (39). AKT further activates the downstream mechanistic target of rapamycin complex 1 (mTORC1), leading to the stabilization of HIF-1α, which subsequently binds to hypoxia-response elements (HREs) within the HK2 promoter. This interaction establishes a positive feedback loop that upregulates HK2 transcription, induces M2 macrophage polarization and suppresses the functions of T cells and natural killer (NK) cells (40). The inhibition of PI3K activity or the application of 2-deoxy-D-glucose (2-DG) can reverse these effects (41). A clinical study has corroborated a positive association between the expression of PI3K/AKT and HK2 in hepatocellular carcinoma (HCC), with a marked increase in PD-L1 positivity observed in groups exhibiting high co-expression levels (42). The programmed cell death protein 1 (PD-1)/PD-L1 pathway inhibits T cell glycolysis and promotes autophagy through negative regulation of HK2 activity (43). Inhibitors of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit β, such as AZD8186, disrupt the AKT-HK2 signaling pathway, leading to decreased lactate production and the restoration of T cell functionality (44). In glioma models, the combination of HK2 inhibitors with PD-1 antibodies has been demonstrated to enhance the infiltration of CD8+ T cells (3,5). Future research should focus on developing precise stratification strategies based on the subcellular localization of HK2 and investigate the potential of PI3K isoform-specific inhibitors.
The MAPK signaling pathway facilitates signal transduction through a hierarchical kinase cascade, predominantly involving the extracellular signal-regulated kinases 1 and 2 (ERK1/2), c-Jun amino-terminal kinases 1 to 3 (JNK1 to −3), p38 (α, β, γ and δ), and ERK5 families (45). HK2 activates ERK1/2 via the Raf/MEK pathway, leading to the induction of cyclin A1 expression, the downregulation of p27, the initiation of DNA replication stress, the release of damage-associated molecular patterns, the activation of chronic inflammatory responses and the recruitment of immunosuppressive cells (46,47). Furthermore, HK2 enhances the expression of pyruvate dehydrogenase kinase 1 through the JNK-c-Jun pathway, thereby inhibiting CD8+ T cell infiltration, or modulates the nucleocytoplasmic shuttling of heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) via p38 MAPKβ (48). hnRNPA1 interacts with the 3′ untranslated region of PD-L1 mRNA to maintain its stability, consequently suppressing T cell activity (49) and fostering the development of an immunosuppressive microenvironment. In the context of colorectal cancer, fructose attenuates MAPK/signal transducer and activator of transcription (STAT)1 signaling through the HK2- inositol 1,4,5-trisphosphate receptor type 3-Ca2+ axis, thereby inhibiting the polarization of M1-type tumor-associated macrophages (TAMs) and further diminishing antitumor immune responses (50). In melanoma, the splicing of hnRNPA1 regulated by HK2 leads to the production of immunosuppressive human leukocyte antigen G mRNA isoforms, which subsequently diminish the cytotoxic efficacy of NK cells (51). Additionally, the pro-inflammatory mediator, macrophage migration inhibitory factor, enhances HK2 expression through the MAPK-ERK signaling pathway, establishing an immunosuppressive positive feedback loop that synergistically promotes immune evasion by the tumor (52).
The NF-κB signaling pathway is activated by chronic inflammation, oncogenic mutations or microenvironmental stressors (53). HK2 interacts with the NF-κB pathway through mechanisms that are both dependent and independent of its metabolic functions. A seminal study conducted by Guo et al (3) demonstrated that HK2 possessed non-metabolic roles in the cytoplasm, functioning as a protein kinase to directly phosphorylate the threonine 291 residue of the NF-κB inhibitor nuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor α (IκBα). This phosphorylation event facilitated the degradation of IκBα via the µ-calpain-proteasome pathway, thereby releasing the NF-κB dimer (RelA/p50) to translocate into the nucleus and initiate the transcription of immune checkpoint proteins, including PD-L1 (3). Concurrently, G-6-P, produced through HK2 catalysis, activates the mTORC1 signaling pathway, which promotes the phosphorylation of the IκB kinase complex and indirectly increases NF-κB activity (54). Simultaneously, NF-κB directly interacts with the HK2 promoter region, thereby augmenting its transcriptional activity and activating downstream glycolysis-related genes, which collectively constitute a self-reinforcing regulatory network known as the ‘NF-κB-HK2-Warburg effect’ (42).
In pancreatic ductal adenocarcinoma models, the HK2-IκBα-NF-κB axis markedly upregulates PD-L1 expression, diminishes CD8+ T cell infiltration and stabilizes β-catenin, thereby enhancing the self-renewal capacity of CD133+ tumor stem cells and further mediating immunosuppression (55). In sarcoma, the canonical NF-κB pathway upregulates HK2 expression, promoting lactate production and suppressing mitochondrial oxidative phosphorylation, which results in the acidification of the TME (56). Lactate contributes to the establishment of an immunosuppressive microenvironment by activating regulatory T cells (Tregs) and M2 macrophages, while inhibiting dendritic cell (DC) maturation (57,58). Preclinical studies have demonstrated that a combined therapeutic approach utilizing the HK2-specific inhibitor 2-DG and the NF-κB pathway inhibitor BAY11-7082 enhanced antitumor immune responses through dual mechanisms: The suppression of PD-L1 protein expression and the reversal of T cell exhaustion phenotypes (59,60). This synergistic therapeutic effect lays the theoretical groundwork for the clinical advancement of innovative immunometabolic combination therapies.
The TGF-β pathway facilitates the differentiation of immunosuppressive cells and diminishes immune cell cytotoxicity through mechanisms that are both SMAD-dependent and independent, thereby promoting an immune-tolerant phenotype (61). In models characterized by energy deficiency induced by oligomycin A-mediated inhibition of oxidative phosphorylation, highly metastatic HCT116 cells stimulated by TGF-β contribute to the formation of an immunosuppressive microenvironment through the upregulation of HK2 expression (62). Furthermore, TGF-β modulates HK2 expression by activating the SMAD3 pathway, while HK2 directly interacts with TGF-β receptor I (TβRI) via its structural domain, stabilizing the TβRI-TβRII complex and enhancing the phosphorylation efficiency of SMAD2/3 (31,63). This receptor-kinase interaction creates a positive feedback loop that intensifies TGF-β-induced epithelial-mesenchymal transition and stromal fibrosis, thereby physically obstructing T cell infiltration into tumor beds (64–66). Furthermore, the TGF-β/SMAD signaling pathway directly inhibits the transcription of major histocompatibility complex class I (MHC-I) heavy chains and antigen processing-associated transporters (67). Concurrently, HK2 activates histone deacetylases through metabolic byproducts, leading to further silencing of the epigenetic regulatory regions of MHC-I genes and impairment of the recognition and binding of tumor-specific antigens by immune cells (68).
The JAK/STAT pathway, a pivotal axis for cytokine signal transduction, facilitates tumor immunosuppression through aberrant activation of STAT3/STAT5 and alters the TME via metabolic reprogramming (69). In colorectal cancer, upregulation of HK2 enhances STAT3 phosphorylation, reduces the infiltration of immune cells such as CD8+ T cells and NK cells, and markedly increases PD-L1 levels, and these effects are reversed by JAK/STAT3 inhibitors (5). Mechanistically, phosphorylated STAT3 directly binds to the PD-L1 promoter, and HK2 indirectly promotes PD-L1 transcription by augmenting STAT3 activity (70).
SUMOylation has been identified as a crucial factor in the cellular response to mitochondrial stress, acting as a sensor and mediator of stress signals to maintain mitochondrial homeostasis (65). This regulatory mechanism is particularly relevant in the context of HK2, suggesting that deSUMOylation likely enhances the interaction between HK2 and VDAC1, thereby activating JAK2 kinase activity and establishing an HK2-JAK2/STAT3-immunosuppressive positive feedback loop (71,72). Epigenetic modifications serve a notable role in this process. Specifically, methyltransferase-like 3 stabilizes HK2 mRNA through N6-methyladenosine modification, and its upregulation enhances HK2 translation via insulin-like growth factor 2 mRNA-binding protein 2, while concurrently activating STAT3-dependent PD-L1 transcription (73). Furthermore, the accumulation of lactate mediated by HK2 inhibits glycolysis in T cells, diminishes interferon γ (IFN-γ) secretion and activates the JAK-STAT pathway through paracrine signaling, thereby further impairing antitumor immunity (74) (Table I).
The phenomenon of tumor immune escape is intricately linked to the immune subtypes present within the TME (75). However, the specific role of HK2 within these subtypes remains poorly understood. A comprehensive analysis utilizing data from The Cancer Genome Atlas and the International Union of Immunological Societies Immunology Databases revealed that HK2 expression is markedly elevated in immunosuppressive tumors, including pancreatic and liver cancers. This elevated expression is associated with the accumulation of Tregs and M2-type macrophages (76). Such an immunosuppressive milieu may further facilitate tumor cell immune escape through metabolic modulation mediated by HK2 (38). Conversely, in immune-active tumors, such as melanoma, a negative association between HK2 expression and T helper type 1 (Th1) cytokines (such as IFN-γ and C-X-C motif chemokine ligand 9) suggests that HK2 may alter the composition and functionality of immune cells by modulating the expression of chemokines and antigen-presenting molecules (77). Furthermore, research has demonstrated that knockout of HK2 can markedly increase the expression of MHC-II and its antigen-presenting capabilities in DCs, while also modulating the Th17/Treg balance in CD4+ T cells. This modulation subsequently influences the specificity and intensity of the immune response (78,79). These observations underscore the pivotal role of HK2 in determining immune cell subtypes. Consequently, these findings imply that HK2 is not only critical within tumor cells but also potentially modulates immune responses by affecting the functionality of immune cells within the tumor immune microenvironment.
Lactate, a central metabolic product of HK2-driven glycolysis, has transitioned from being considered merely a ‘metabolic waste product’ (80) to a multifunctional mediator, acting as a signaling molecule, energy substrate and immunoregulatory agent that governs tumor immune evasion (81,82). Tumor cells continuously produce lactate through HK2 hyperactivation, which increases extracellular acidification rates, contributes to the formation of an acidic TME and establishes an immunosuppressive niche through multiple pathways (such as inhibiting the function of effector immune cells, promoting the expansion of inhibitory immune cells, disrupting antigen presentation and upregulating checkpoints) (30). As shown in Fig. 1, the elevated lactate levels mediated by HK2 result in a reduction in the calcium influx in T cells, inhibition of IFN-γ secretion and prevention of the nuclear translocation of activated T cell nuclear factors, as well as the expression of the autophagy factor focal adhesion kinase family interacting protein of 200 kDa (57,83,84). These processes contribute to T cell exhaustion, apoptosis and a diminished potential for memory T cell differentiation (85,86). Application of the HK2 inhibitor 2-DG counteracts these effects, thereby restoring the antitumor immune function of T cells (87). HK2-driven glycolysis creates intracellular and extracellular pH gradients, which directly induce the expansion of Tregs and upregulate PD-1 expression. Furthermore, pH gradients downregulate major MHC-II/CD80 expression through HIF-1α, thereby impairing the antigen-processing capacity of DCs and the polarization of helper T cells (88–91). Lactate also upregulates interleukin (IL)-10 and downregulates acidification-dependent natural killer group 2D (NKG2D) and DNAX accessory molecule-1 (DNAM-1) by suppressing the activation of NF-κB (92), leading to suppressed cytotoxicity in NK cells (93,94). Additionally, lactate reduces the expression of inducible nitric oxide synthase, C-C motif chemokine ligand (CCL)2 and IL-6 in M1 macrophages by inhibiting the activity of the NF-κB signaling pathway, while the STAT3/HIF-1α axis promotes macrophage M2 polarization, facilitating immune escape (95,96). Furthermore, the elevation of lactate mediated by HK2 upregulates inhibitory molecules such as PD-L1, cytotoxic T-lymphocyte-associated protein 4 and TGF-β (97–99), induces histone lysine lactylation, and establishes a ‘glycolysis-lactylation-immunosuppression’ positive feedback loop (100,101).
The interaction of HK2 with mitochondrial VDAC1 leads to localized microenvironmental hypoxia, which accelerates lactate accumulation and abnormal angiogenesis, thereby further exacerbating hypoxia (100,102). As shown in Fig. 2, intratumoral hypoxia predominantly inhibits the prolyl hydroxylase-mediated stabilization of HIF-1α (103). Activation of HIF-1α also enhances HK2 expression (104). The hypoxia-HIF-1α axis directly interacts with HREs in the HK2 promoter, amplifying the hypoxia process in the TME and forming an immunosuppressive feedforward loop (105,106). Clinical investigations have demonstrated that the HK2-mediated upregulation of HIF-1α in breast and lung cancer is positively associated with PD-L1 expression, and patients with high HK2 expression exhibit reduced response rates to immune checkpoint inhibitors (107,108). Mechanistically, the activation of HIF-1α by HK2 facilitates its binding to HREs within the PD-L1 promoter, leading to upregulation of PD-L1 transcription (109). Additionally, this activation promotes the post-translational modifications of PD-L1 through the activation of the PI3K/AKT/mTOR signaling pathway (110). Furthermore, HK2-mediated activation of HIF-1α results in the upregulation of microRNA-224, which suppresses the natural cytotoxicity triggering receptor 1/NKp46 pathway and thereby protects tumors from NK cell-mediated attack (111). HIF-1α also induces the secretion of CCL28 and TGF-β by tumor cells, facilitating the recruitment of Tregs to tumor sites (112,113), and sustains immunosuppressive function of CCL28 and TGF-β by enhancing the expression of forkhead box P3 (Foxp3) (114). Concurrently, HIF-1α promotes the polarization of TAMs towards the M2 phenotype through the upregulation of vascular endothelial growth factor (VEGF) and IL-10. These M2 TAMs subsequently secrete arginase 1, IL-10 and other factors (such as TGF-β, PD-L1) that inhibit the activity of CD8+ T cells (115). Furthermore, HIF-1α activates the STAT3 pathway, leading to the expansion of myeloid-derived suppressor cells, which further suppress the antitumor effects of NK and T cells (116). HK2-activated HIF-1α directly interacts with HREs in the MHC-I gene promoter or recruits histone deacetylases to induce chromatin compaction, thereby inhibiting MHC-I transcription (30). HIF-1α inhibits the IFN-γ-mediated upregulation of MHC-I by suppressing the activity of IFN regulatory factors (117). Additionally, HIF-1α impairs DC differentiation and antigen-presenting capabilities through the induction of VEGF and IL-6, resulting in inadequate T cell priming (118). HK2 further enhances the expression of HIF-1α and monocarboxylate transporters, facilitating HK2-mediated lactate efflux and exacerbating the immunosuppressive TME (119). HIF-1α also upregulates CD73 expression, which catalyzes the conversion of ATP to adenosine, subsequently inhibiting T cell activation and NK cell cytotoxicity via adenosine A2A receptor interaction (120–122). Furthermore, HIF-1α induces the expression of indoleamine 2,3-dioxygenase, leading to tryptophan depletion and kynurenine acid production in the microenvironment, thereby suppressing T cell proliferation and promoting Treg differentiation (123,124).
The synergistic effects of the HK2-mediated glycolytic enhancement and impaired vascular exchange result in limited glucose availability within the TME, causing intratumoral hypoglycemia (125). Under these glucose-deprivation conditions, the TME selectively impairs glucose-dependent immune cells (126). As shown in Fig. 3, the hypoglycemic TME driven by HK2 activates AMP-activated protein kinase (AMPK), inhibits the signal of MTORC1-activated cell metabolic synthesis, downregulates HIF-1α expression, reduces IL-12 secretion and fails to effectively activate naïve T cells (127–129). The secretion of effector cytokines such as IFN-γ and tumor necrosis factor-α is also diminished (130). Under conditions of HK2-induced glucose deprivation, T cells are compelled to shift towards fatty acid oxidation and mitochondrial respiration. However, this metabolic shift is inefficient due to the suppression of carnitine palmitoyltransferase 1A translation mediated by glucose deprivation, ultimately leading to T cell exhaustion (131). Hypoglycemia induces DCs to adopt a tolerogenic phenotype characterized by elevated PD-L1 and IL-10 expression, further inhibiting T cell responses (128). By contrast, Tregs within the hypoglycemic TME have enhanced oxidative phosphorylation through Foxp3-mediated metabolic reprogramming, relying on fatty acid and lactate metabolism to maintain their immunosuppressive functions. This metabolic adaptation provides Tregs with a competitive advantage in nutrient competition, thereby exacerbating antitumor immune suppression (132). Furthermore, glucose deprivation triggers the activation of the AMPK pathway in tumor cells. AMPK phosphorylates Hu antigen R (HuR) at Ser318, facilitating its translocation from the nucleus to the cytoplasm (133). In melanoma cells, the cytoplasmic localization of HuR increases from 20 to 60% under hypoglycemic conditions, which enhances its binding affinity to PD-L1 mRNA, and thus, stabilizes PD-L1 mRNA (134).
Advancements in the development of novel HK2 inhibitors have reached the clinical research phase, in which they have shown notable antitumor activity (Table II). Small molecule inhibitors, such as benserazide and benitrobenrazide (BNBZ), have been shown to decrease glucose uptake, lactic acid production and intracellular ATP levels by specifically binding to HK2 and inhibiting its enzymatic function (135,136). This mechanism induces apoptosis in tumor cells and suppresses tumor cell proliferation. BNBZ has exhibited notable antitumor effects in xenograft mouse models using SW1990 and SW480 cells, highlighting its potential as a promising HK2 inhibitor (136). Similarly, benserazide has been shown to effectively reduce tumor growth in a SW480 cell xenograft mouse model without significant toxic effects (135). The nanoparticle formulation of benserazide further enhances its antitumor efficacy and specificity in targeting tumors (135).
VDA-1102 (Almavid™), the first small molecule targeting the HK2-VDAC1 interaction, disrupts HK2 mitochondrial localization to restore cancer cell apoptosis. In a phase II trial for cutaneous T-cell lymphoma and actinic keratosis, VDA-1102 achieved an objective response rate (ORR) >60% with manageable toxicity (137). The subcutaneous formulation of VDA-1102 exhibited a stable plasma concentration (>24 h) and dose-linear pharmacokinetics in pediatric patients with brain tumors, indicating broad-spectrum potential in solid tumors (138).
Ketoconazole, a United States Food and Drug Administration-approved antifungal agent and HK2 inhibitor, disrupts the interaction between HK2 and aminoacyl transfer RNA synthetase complex-interacting multifunctional protein 2 (AIMP2). This reduces autophagic lysosome-dependent degradation of AIMP2, restoring AIMP2-mediated apoptosis and synergizing with radiotherapy to suppress HCC growth. This drug-repurposing strategy overcomes radioresistance with translational advantages in terms of safety and cost (139).
A novel series of 2,6-disubstituted glucosamines has been identified as potent and selective HK2 inhibitors. These compounds demonstrate significant selectivity over HK1 and have been shown to inhibit glycolysis in cancer cell lines, underscoring their potential as therapeutic agents (140). Ikarugamycin, a polycyclic tetramide macrolide compound isolated from Actinomycetes associated with mangroves, has been shown to inhibit the glycolytic flux in pancreatic cancer cells by targeting HK2 and exhibits antitumor activity in murine models without exhibiting significant cytotoxicity (141). Another promising development in the field is the identification of natural HK2 inhibitors from Ganoderma sinense. For example, a specific steroid compound, (22E,24R)-6β-methoxyergosta-7,9(11),22-triene-3β,5α-diol, has been identified as having a high binding affinity for HK2, suggesting its potential as a drug candidate for cancer therapy (142). This discovery is complemented by a broader study that systematically reviews the characteristics of HK2 inhibitors, emphasizing the need for compounds with improved efficacy and selectivity due to the poor performance of existing inhibitors when used alone (143).
RNA interference-based therapy, which silences the expression of HK2, has been shown to inhibit tumor cell proliferation and metastasis, thereby demonstrating notable therapeutic potential (144–148). The clinical potential of HK2 inhibitors is further supported by studies on their role in inducing apoptosis and endoplasmic reticulum stress in cancer cells. For instance, the HK2 inhibitor 3-bromopyruvate has been shown to inhibit the survival and proliferation of colon cancer cells, suggesting its utility in combination therapies (149). Additionally, the use of azole antifungals, such as ketoconazole and posaconazole, has demonstrated efficacy in targeting HK2-expressing glioblastoma cells, providing a basis for repurposing these compounds in clinical trials (150). These findings collectively underscore the therapeutic promise of HK2 inhibitors and their potential to advance to clinical applications (Table II).
As the role of HK2 in tumor immune evasion becomes clearer, its utility as a predictive biomarker for immunotherapy is gaining attention. In bladder cancer research, the immune expression of glycolysis-related proteins, including HK2, has been assessed. The findings indicated that high HK2 expression was closely associated with independent prognostic factors for progression-free survival and overall survival, suggesting its utility as a prognostic biomarker for patients with this disease (151). In colorectal cancer, HK2 expression is associated with tumor size, depth of invasion, liver metastasis and TNM stage, effectively predicting the recurrence risk and overall mortality. Thus, HK2 serves as an important prognostic biomarker (12). Additionally, in patients with lung cancer, HK2 expression serves as a biomarker for identifying a novel subset of circulating tumor cells associated with poor prognosis. This finding is instrumental for prognostic evaluation prior to treatment (152). Positron emission tomography utilizing 18F-fluorodeoxyglucose (18F-FDG) is a widely employed technique for assessing tumor glucose metabolism. HK2 expression is associated with the uptake of 18F-FDG by tumors (153). Consequently, HK2 detection can facilitate the early diagnosis, staging and evaluation of the treatment response in tumors (10). Furthermore, the development of a ternary scoring system incorporating HK2, PD-L1 and HIF-1α for predicting the response of patients with liver cancer is a promising approach that leverages the interplay between metabolic pathways, immune checkpoints and hypoxic conditions within the TME (154). This model is substantiated by several studies that highlight the critical roles of these biomarkers in HCC prognosis and treatment response (155–157).
The integration of single-cell transcriptomics and spatial transcriptomics has revolutionized the current understanding of cell dynamics and tissue structure, providing unprecedented insights into the study of gene expression patterns and cell interactions in natural spatial environments (158,159). This two-pronged research strategy is particularly important when monitoring the changes in HK2; it can achieve single-cell resolution analysis of HK2 expression profiles while maintaining spatial information, thereby deeply analyzing the expression distribution of HK2 in complex tissue environments and its interaction with other factors. This strategy can also provide in-depth insights into the metabolic heterogeneity within tumors (160,161). Based on single-cell transcriptome and spatial transcriptome techniques, the HK2-Glycolytic Activity Index shows good stability in various tumor types and is associated with the characteristics of immune infiltration in the tumor microenvironment, having the potential to guide the selection of CAR-T cell therapy (20). Furthermore, advanced computational tools, such as the Significant Process Inference Algorithm, enhance the detection of significant expression patterns and biological processes, even the subtle changes in HK2 expression across different cell populations (162). Furthermore, the application of deep learning models, such as KanCell, enhances the analysis of cellular heterogeneity by integrating single-cell RNA-sequencing and spatial transcriptomics data (163). These models can effectively capture non-linear relationships and optimize computational efficiency, providing a more accurate depiction of HK2 expression patterns and their implications in disease microenvironments (164). By leveraging these advanced technologies, researchers can gain a deeper understanding of the molecular mechanisms underlying HK2 regulation, and the impact on cellular metabolism and disease progression.
Current research on tumor immune escape is focused on several advanced fields. A comprehensive investigation into the interactions among diverse cellular and molecular mechanisms within the TME, particularly the metabolic interactions between immune cells and tumor cells, will provide a theoretical foundation for the development of more effective therapeutic strategies. Studies have identified that TAMs facilitate tumor immune escape through multiple signaling pathways, such as IL-4/STAT6, MAPK/ERK and TLR-NF-κB (165–167). Therapeutic strategies aimed at targeting TAMs, including colony stimulating factor 1 receptor inhibitors and CD40 agonists, have progressed to clinical trials (nos. NCT04169672 and NCT04059588), illustrating their potential to mitigate immunosuppression and augment the effectiveness of immunotherapy (78,168,169). Conversely, the utilization of advanced technologies, such as single-cell sequencing and high-dimensional flow cytometry, offers a deeper understanding of the heterogeneity inherent in tumor immune evasion, thereby providing a more precise foundation for personalized treatment approaches (170).
The multifaceted role of HK2 in linking tumor metabolic reprogramming and immune escape is becoming increasingly evident. HK2, with its distinctive dual catalytic active domain and N-terminal mitochondrial anchoring capability, facilitates the ‘metabolic compartmentalization’ effect and ‘metabolic relay’, thereby mediating efficient glycolysis. HK2 has a bidirectional regulatory network with critical oncogenic pathways, including the PI3K/AKT, MAPK, NF-κB, TGF-β and JAK/STAT pathways, thereby enhancing the glycolysis-dependent immune evasion of tumors. The metabolic competition induced by the Warburg effect facilitates the influence of HK2 on the function of immune cells in the TME, leading to the formation of hypoxic and hypoglycemic niches. This process is characterized by abundant lactic acid excretion, which collectively influences immune cell distribution, alters immune cell function and modulates the expression of immune checkpoints, thereby further facilitating tumor immune escape. The protein kinase function, subcellular localization regulation of HK2 and its involvement in different immune subtypes have gained notable attention, offering a novel perspective for a comprehensive analysis of its non-glycolytic functions. Emerging HK2 inhibitors, along with multi-dimensional predictive models and their integration with immunotherapy, have demonstrated promising potential for clinical translation. Future research should focus on elucidating immune subtype-specific mechanisms, developing highly selective targeted therapeutics and conducting comprehensive analyses of HK2-related molecular characteristics. These efforts should aim to advance the clinical implementation of HK2-targeted intervention strategies in precision immunotherapy.
Not applicable.
The present study was supported by the National Natural Science Foundation of China (grant no. 82505450), the Top Talent Support Program for Young and Middle-Aged People of Wuxi Health Committee (grant no. HB2023067), the Natural Science Foundation of Jiangsu Province (grant no. BK20240309), and the Natural Science Foundation of Nanjing University of Chinese Medicine (grant no. XZR2023095).
Not applicable.
YQ participated in conceptualization, data curation and writing of the original draft. XZ participated in conceptualization, acquisition of funding, and reviewing and editing of the manuscript. DN and QT participated in reviewing and editing the manuscript. CJ participated in reviewing and editing the manuscript, and acquiring resources. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
During the preparation of this work, artificial intelligence tools (ChatGPT) were used to improve the readability and language of the manuscript, and subsequently, the authors revised and edited the content produced by the artificial intelligence tools as necessary, taking full responsibility for the ultimate content of the present manuscript.
|
Galassi C, Chan TA, Vitale I and Galluzzi L: The hallmarks of cancer immune evasion. Cancer Cell. 42:1825–1863. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Paul S, Ghosh S and Kumar S: Tumor glycolysis, an essential sweet tooth of tumor cells. Semin Cancer Biol. 86:1216–1230. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Guo D, Tong Y, Jiang X, Meng Y, Jiang H, Du L, Wu Q, Li S, Luo S, Li M, et al: Aerobic glycolysis promotes tumor immune evasion by hexokinase2-mediated phosphorylation of IκBα. Cell Metab. 34:1312–1324. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu L, Jin Y, Zhao X, Tang K, Zhao Y, Tong L, Yu X, Xiong K, Luo C, Zhu J, et al: Tumor aerobic glycolysis confers immune evasion through modulating sensitivity to T cell-mediated bystander killing via TNF-α. Cell Metab. 35:1580–1596. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Qing S and Shen Z: High expression of hexokinase 2 promotes proliferation, migration and invasion of colorectal cancer cells by activating the JAK/STAT pathway and regulating tumor immune microenvironment. Nan Fang Yi Ke Da Xue Xue Bao. 45:542–553. 2025.(In Chinese). PubMed/NCBI | |
|
Li C, Tang Y, Zhang R, Shi L, Chen J, Zhang P, Zhang N and Li W: Inhibiting glycolysis facilitated checkpoint blockade therapy for triple-negative breast cancer. Discov Oncol. 16:5502025. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng M, Wang B, Duan L, Jin Y, Zhang W and Li N: HOTAIR knockdown increases the sensitivity of hepatocellular carcinoma cells to sorafenib by disrupting miR-145-5p/HK2 axis-mediated mitochondrial function and glycolysis. Front Biosci (Landmark Ed). 30:373682025. View Article : Google Scholar : PubMed/NCBI | |
|
Cui R, Wang G, Liu F, Wang Y, Zhao Z, Mutailipu M, Mu H, Jiang X, Le W, Yang L and Chen B: Neurturin-induced activation of GFRA2-RET axis potentiates pancreatic cancer glycolysis via phosphorylated hexokinase 2. Cancer Lett. 621:2175832025. View Article : Google Scholar : PubMed/NCBI | |
|
Lyu SI, Simon AG, Jung JO, Fretter C, SchrÖder W, Bruns CJ, Schmidt T, Quaas A and Knipper K: Hexokinase 2 as an independent risk factor for worse patient survival in esophageal adenocarcinoma and as a potential therapeutic target protein: A retrospective, single-center cohort study. Oncol Lett. 28:4952024. View Article : Google Scholar : PubMed/NCBI | |
|
Guo W, Kuang Y, Wu J, Wen D, Zhou A, Liao Y, Song H, Xu D, Wang T, Jing B, et al: Hexokinase 2 depletion confers sensitization to metformin and inhibits glycolysis in lung squamous cell carcinoma. Front Oncol. 10:522020. View Article : Google Scholar : PubMed/NCBI | |
|
Jin Z, Gu J, Xin X, Li Y and Wang H: Expression of hexokinase 2 in epithelial ovarian tumors and its clinical significance in serous ovarian cancer. Eur J Gynaecol Oncol. 35:519–524. 2014.PubMed/NCBI | |
|
Katagiri M, Karasawa H, Takagi K, Nakayama S, Yabuuchi S, Fujishima F, Naitoh T, Watanabe M, Suzuki T, Unno M and Sasano H: Hexokinase 2 in colorectal cancer: A potent prognostic factor associated with glycolysis, proliferation and migration. Histol Histopathol. 32:351–360. 2017.PubMed/NCBI | |
|
Yoshino H, Enokida H, Itesako T, Kojima S, Kinoshita T, Tatarano S, Chiyomaru T, Nakagawa M and Seki N: Tumor-suppressive microRNA-143/145 cluster targets hexokinase-2 in renal cell carcinoma. Cancer Sci. 104:1567–1574. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Botzer LE, Maman S, Sagi-Assif O, Meshel T, Nevo I, Yron I and Witz IP: Hexokinase 2 is a determinant of neuroblastoma metastasis. Br J Cancer. 114:759–766. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Gao H, Zhou Y and Chen X: Tregs and platelets play synergistic roles in tumor immune escape and inflammatory diseases. Crit Rev Immunol. 42:59–69. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Wu K, Shi L, Xiang F, Tao K and Wang G: Prognostic significance of the metabolic marker hexokinase-2 in various solid tumors: A meta-analysis. PLoS One. 11:e01662302016. View Article : Google Scholar : PubMed/NCBI | |
|
Kwee SA, Hernandez B, Chan O and Wong L: Choline kinase alpha and hexokinase-2 protein expression in hepatocellular carcinoma: Association with survival. PLoS One. 7:e465912012. View Article : Google Scholar : PubMed/NCBI | |
|
Suh DH, Kim MA, Kim H, Kim MK, Kim HS, Chung HH, Kim YB and Song YS: Association of overexpression of hexokinase II with chemoresistance in epithelial ovarian cancer. Clin Exp Med. 14:345–353. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wang XT, Xie L, Hu YT, Zhao YY, Wang RY, Yan Y, Zhu XZ and Liu LL: T. pallidum achieves immune evasion by blocking autophagic flux in microglia through hexokinase 2. Microb Pathog. 199:1072162025. View Article : Google Scholar : PubMed/NCBI | |
|
Lin J, Fang W, Xiang Z, Wang Q, Cheng H, Chen S, Fang J, Liu J, Wang Q, Lu Z and Ma L: Glycolytic enzyme HK2 promotes PD-L1 expression and breast cancer cell immune evasion. Front Immunol. 14:11899532023. View Article : Google Scholar : PubMed/NCBI | |
|
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. 25:1208–1222. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Xu S, Zhan Y, Lv X, Sun Z, Man L, Yang D, Sun Y and Ding S: CircRUNX1 enhances the Warburg effect and immune evasion in non-small cell lung cancer through the miR-145/HK2 pathway. Cancer Lett. 28:2176392025. View Article : Google Scholar : PubMed/NCBI | |
|
Li R, Mei S, Ding Q, Wang Q, Yu L and Zi F: A pan-cancer analysis of the role of hexokinase II (HK2) in human tumors. Sci Rep. 12:188072022. View Article : Google Scholar : PubMed/NCBI | |
|
Ho PC, Bihuniak JD, Macintyre AN, Staron M, Liu X, Amezquita R, Tsui YC, Cui G, Micevic G, Perales JC, et al: Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell. 162:1217–1228. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Chang CH, Qiu J, O'Sullivan D, Buck MD, Noguchi T, Curtis JD, Chen Q, Gindin M, Gubin MM, van der Windt GJ, et al: Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 162:1229–1241. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Shi ZY, Yang C, Lu LY, Lin CX, Liang S, Li G, Zhou HM and Zheng JM: Inhibition of hexokinase 2 with 3-BrPA promotes MDSCs differentiation and immunosuppressive function. Cell Immunol. 385:1046882023. View Article : Google Scholar : PubMed/NCBI | |
|
İpek ӦS, Sucu BO, Selvi S, Alkan FK, Tiryaki B, Alkan HK, Sayyah E, Tolu İ, Güzel M, Durdağı S, et al: Anti-cancer efficacy of novel lonidamine derivatives: Design, synthesis, in vitro, in vivo, and computational studies targeting hexokinase-2. Eur J Med Chem. 296:1178902025. View Article : Google Scholar : PubMed/NCBI | |
|
Gu QL, Zhang Y, Fu XM, Lu ZL, Yu Y, Chen G, Ma R, Kou W and Lan YM: Toxicity and metabolism of 3-bromopyruvate in Caenorhabditis elegans. J Zhejiang Univ Sci B. 21:77–86. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Pajak B, Siwiak E, Sołtyka M, Priebe A, Zieliński R, Fokt I, Ziemniak M, Jaśkiewicz A, Borowski R, Domoradzki T and Priebe W: 2-Deoxy-d-glucose and its analogs: From diagnostic to therapeutic agents. Int J Mol Sci. 21:2342019. View Article : Google Scholar : PubMed/NCBI | |
|
Rho H, Terry AR, Chronis C and Hay N: Hexokinase 2-mediated gene expression via histone lactylation is required for hepatic stellate cell activation and liver fibrosis. Cell Metab. 35:1406–1423. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Patra KC, Wang Q, Bhaskar PT, Miller L, Wang Z, Wheaton W, Chandel N, Laakso M, Muller WJ, Allen EL, et al: Hexokinase 2 is required for tumor initiation and maintenance and its systemic deletion is therapeutic in mouse models of cancer. Cancer Cell. 24:213–228. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Li M, Zhang Y, Wu C, Yang K, Gao S, Zheng M, Li X, Li H and Chen L: Structure based discovery of novel hexokinase 2 inhibitors. Bioorg Chem. 96:1036092020. View Article : Google Scholar : PubMed/NCBI | |
|
Bustamante E and Pedersen PL: Mitochondrial hexokinase of rat hepatoma cells in culture: Solubilization and kinetic properties. Biochemistry. 19:4972–4977. 1980. View Article : Google Scholar : PubMed/NCBI | |
|
Deberardinis RJ, Sayed N, Ditsworth D and Thompson CB: Brick by brick: Metabolism and tumor cell growth. Curr Opin Genet Dev. 18:54–61. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ganapathy-Kanniappan S and Geschwind JF: Tumor glycolysis as a target for cancer therapy: Progress and prospects. Mol Cancer. 12:1522013. View Article : Google Scholar : PubMed/NCBI | |
|
Nawaz MH, Ferreira JC, Nedyalkova L, Zhu H, Carrasco-López C, Kirmizialtin S and Rabeh WM: The catalytic inactivation of the N-half of human hexokinase 2 and structural and biochemical characterization of its mitochondrial conformation. Biosci Rep. 38:BSR201716662018. View Article : Google Scholar : PubMed/NCBI | |
|
Lee HJ, Li CF, Ruan D, He J, Montal ED, Lorenz S, Girnun GD and Chan CH: Non-proteolytic ubiquitination of Hexokinase 2 by HectH9 controls tumor metabolism and cancer stem cell expansion. Nat Commun. 10:26252019. View Article : Google Scholar : PubMed/NCBI | |
|
Panpan SI, Wei GE, Kaiming WU and Zhang R: O-GlcNAcylation of hexokinase 2 modulates mitochondrial dynamics and enhances the progression of lung cancer. Mol Cell Biochem. 480:2633–2643. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Huang C, Chen B, Wang X, Xu J, Sun L, Wang D, Zhao Y, Zhou C, Gao Q, Wang Q, et al: Gastric cancer mesenchymal stem cells via the CXCR2/HK2/PD-L1 pathway mediate immunosuppression. Gastric Cancer. 26:691–707. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ranjbar A, Soltanshahi M, Taghiloo S and Asgarian-Omran H: Glucose metabolism in acute myeloid leukemia cell line is regulated via combinational PI3K/AKT/mTOR pathway inhibitors. Iran J Pharm Res. 22:e1405072023.PubMed/NCBI | |
|
Collins NB, Al Abosy R, Milsler BC, Bi K, Zhao Q, Quigley M, Ishizuka JJ, Yates KB, Pope HW, Manguso R, et al: PI3K activation allows immune evasion by promoting an inhibitory myeloid tumor microenvironment. J Immunother Cancer. 10:e0034022022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Lin X, Lei Y, Xu X, Zhou Q, Chen Y, Liu H, Jiang J, Yang Y, Zheng F and Wu B: Aerobic glycolysis enhances HBx-initiated hepatocellular carcinogenesis via NF-κBp65/HK2 signalling. J Exp Clin Cancer Res. 41:3292022. View Article : Google Scholar : PubMed/NCBI | |
|
Kawasaki Y, Sato K, Mashima K, Nakano H, Ikeda T, Umino K, Morita K, Izawa J, Takayama N, Hayakawa H, et al: Mesenchymal stromal cells inhibit aerobic glycolysis in activated t cells by negatively regulating hexokinase II activity through PD-1/PD-L1 interaction. Transplant Cell Ther. 27:231.e231–231.e238. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lynch JT, Polanska UM, Delpuech O, Hancox U, Trinidad AG, Michopoulos F, Lenaghan C, McEwen R, Bradford J, Polanski R, et al: Inhibiting PI3Kβ with AZD8186 regulates key metabolic pathways in PTEN-null tumors. Clin Cancer Res. 23:7584–7595. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Cargnello M and Roux PP: Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 75:50–83. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Ji P, Bäumer N, Yin T, Diederichs S, Zhang F, Beger C, Welte K, Fulda S, Berdel WE, Serve H, et al: DNA damage response involves modulation of Ku70 and Rb functions by cyclin A1 in leukemia cells. Int J Cancer. 121:706–713. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Cui N, Li L, Feng Q, Ma HM, Lei D and Zheng PS: Hexokinase 2 promotes cell growth and tumor formation through the raf/mek/erk signaling pathway in cervical cancer. Front Oncol. 10:5812082020. View Article : Google Scholar : PubMed/NCBI | |
|
Courteau L, Crasto J, Hassanzadeh G, Baird SD, Hodgins J, Liwak-Muir U, Fung G, Luo H, Stojdl DF, Screaton RA and Holcik M: Hexokinase 2 controls cellular stress response through localization of an RNA-binding protein. Cell Death Dis. 6:e18372015. View Article : Google Scholar : PubMed/NCBI | |
|
Lian C, Zhang C, Tian P, Tan Q, Wei Y, Wang Z, Zhang Q, Zhang Q, Zhong M, Zhou L, et al: Epigenetic reader ZMYND11 noncanonical function restricts HNRNPA1-mediated stress granule formation and oncogenic activity. Signal Transduct Target Ther. 9:2582024. View Article : Google Scholar : PubMed/NCBI | |
|
Yan H, Wang Z, Teng D, Chen X, Zhu Z, Chen H, Wang W, Wei Z, Wu Z, Chai Q, et al: Hexokinase 2 senses fructose in tumor-associated macrophages to promote colorectal cancer growth. Cell Metab. 36:2449–2467. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Dunnett L, Das S, Venditti V and Prischi F: Enhanced identification of small molecules binding to hnRNPA1 via cryptic pockets mapping coupled with X-ray fragment screening. J Biol Chem. 301:1083352025. View Article : Google Scholar : PubMed/NCBI | |
|
Yang S, Tang W, Azizian A, Gaedcke J, Ohara Y, Cawley H, Hanna N, Ghadimi M, Lal T, Sen S, et al: MIF/NR3C2 axis regulates glucose metabolism reprogramming in pancreatic cancer through MAPK-ERK and AP-1 pathways. Carcinogenesis. 45:582–594. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang T, Ma C, Zhang Z, Zhang H and Hu H: NF-κB signaling in inflammation and cancer. MedComm (2020). 2:618–653. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Q, Xu J and Wang H: Intestine-derived exosomes regulates inflammatory response of HK-2 cells through miR-146a-regulated NF-κB pathway. Drug Eval Res. 47:2326–2333. 2024. | |
|
Tong Y, Liu X, Wu L, Xiang Y, Wang J, Cheng Y, Zhang C, Han B, Wang L and Yan D: Hexokinase 2 nonmetabolic function-mediated phosphorylation of IκBα enhances pancreatic ductal adenocarcinoma progression. Cancer Sci. 115:2673–2685. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Londhe P, Yu PY, Ijiri Y, Ladner KJ, Fenger JM, London C, Houghton PJ and Guttridge DC: Classical NF-κB metabolically reprograms sarcoma cells through regulation of hexokinase 2. Front Oncol. 8:1042018. View Article : Google Scholar : PubMed/NCBI | |
|
Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, Matos C, Bruss C, Klobuch S, Peter K, et al: LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24:657–671. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM, et al: Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 513:559–563. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Sharan L, Pal A, Babu SS, Kumar A and Banerjee S: Bay 11-7082 mitigates oxidative stress and mitochondrial dysfunction via NLRP3 inhibition in experimental diabetic neuropathy. Life Sci. 359:1232032024. View Article : Google Scholar : PubMed/NCBI | |
|
Dent AT and Wilks A: Contributions of the heme coordinating ligands of the Pseudomonas aeruginosa outer membrane receptor HasR to extracellular heme sensing and transport. J Biol Chem. 295:10456–10467. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Derynck R, Turley SJ and Akhurst RJ: TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol. 18:9–34. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong C, Wang W, Yao Y, Lian S, Xie X, Xu J, He S, Luo L, Ye Z, Zhang J, et al: TGF-β secreted by cancer cells-platelets interaction activates cancer metastasis potential by inducing metabolic reprogramming and bioenergetic adaptation. J Cancer. 16:1310–1323. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Roh JI, Kim Y, Oh J, Kim Y, Lee J, Lee J, Chun KH and Lee HW: Hexokinase 2 is a molecular bridge linking telomerase and autophagy. PLoS One. 13:e01931822018. View Article : Google Scholar : PubMed/NCBI | |
|
Lu J, Liu Q, Wang L, Tu W, Chu H, Ding W, Jiang S, Ma Y, Shi X, Pu W, et al: Increased expression of latent TGF-β-binding protein 4 affects the fibrotic process in scleroderma by TGF-β/SMAD signaling. Lab Invest. 97:591–601. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang YL, Li X, Tan YW, Fang YJ, Liu KY, Wang YF, Ma T, Ou QJ and Zhang CX: Docosahexaenoic acid inhibits the invasion and migration of colorectal cancer by reversing EMT through the TGF-β1/Smad signaling pathway. Food Funct. 15:9420–9433. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, Kadel EE III, Koeppen H, Astarita JL, Cubas R, et al: TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 554:544–548. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jeon HS and Jen J: TGF-beta signaling and the role of inhibitory Smads in non-small cell lung cancer. J Thorac Oncol. 5:417–419. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Santarpia M, González-Cao M, Viteri S, Karachaliou N, Altavilla G and Rosell R: Programmed cell death protein-1/programmed cell death ligand-1 pathway inhibition and predictive biomarkers: Understanding transforming growth factor-beta role. Transl Lung Cancer Res. 4:728–742. 2015.PubMed/NCBI | |
|
Ayele TM, Muche ZT, Teklemariam AB, Kassie AB and Abebe EC: Role of JAK2/STAT3 signaling pathway in the tumorigenesis, chemotherapy resistance, and treatment of solid tumors: A systemic review. J Inflamm Res. 15:1349–1364. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ren D, Hua Y, Yu B, Ye X, He Z, Li C, Wang J, Mo Y, Wei X, Chen Y, et al: Predictive biomarkers and mechanisms underlying resistance to PD1/PD-L1 blockade cancer immunotherapy. Mol Cancer. 19:192020. View Article : Google Scholar : PubMed/NCBI | |
|
Shangguan X, He J, Ma Z, Zhang W, Ji Y, Shen K, Yue Z, Li W, Xin Z, Zheng Q, et al: SUMOylation controls the binding of hexokinase 2 to mitochondria and protects against prostate cancer tumorigenesis. Nat Commun. 12:18122021. View Article : Google Scholar : PubMed/NCBI | |
|
Xue YN, Yu BB, Li JL, Guo R, Zhang LC, Sun LK, Liu YN and Li Y: Zinc and p53 disrupt mitochondrial binding of HK2 by phosphorylating VDAC1. Exp Cell Res. 374:249–258. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Guo X, Li L, Gao Z, Su X, Ji M and Liu J: N6-methyladenosine METTL3 promotes cervical cancer tumorigenesis and Warburg effect through YTHDF1/HK2 modification. Cell Death Dis. 11:9112020. View Article : Google Scholar : PubMed/NCBI | |
|
Klinke DJ II, Cheng N and Chambers E: Quantifying crosstalk among interferon-γ, interleukin-12, and tumor necrosis factor signaling pathways within a TH1 cell model. Sci Signal. 5:ra322012. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Li S, Wang D, Liu S, Xiao T, Gu W, Yang H, Wang H, Yang M and Chen P: Metabolic reprogramming and immune evasion: The interplay in the tumor microenvironment. Biomark Res. 12:962024. View Article : Google Scholar : PubMed/NCBI | |
|
Ferreira JC, Khrbtli AR, Shetler CL, Mansoor S, Ali L, Sensoy O and Rabeh WM: Linker residues regulate the activity and stability of hexokinase 2, a promising anticancer target. J Biol Chem. 296:1000712021. View Article : Google Scholar : PubMed/NCBI | |
|
Peláez R, Fernández-García P, Herrero P and Moreno F: Nuclear import of the yeast hexokinase 2 protein requires α/β-importin-dependent pathway. J Biol Chem. 287:3518–3529. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Zhang M, Gao Y, Zhao C, Liao S, Zhao X, Ning Q and Tang S: Mechanisms of tumor-associated macrophages promoting tumor immune escape. Carcinogenesis. 46:bgaf0232025. View Article : Google Scholar : PubMed/NCBI | |
|
Gou Q, Che S, Chen M, Chen H, Shi J and Hou Y: PPARγ inhibited tumor immune escape by inducing PD-L1 autophagic degradation. Cancer Sci. 114:2871–2881. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Rabinowitz JD and Enerbäck S: Lactate: The ugly duckling of energy metabolism. Nat Metab. 2:566–571. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Brooks GA: The science and translation of lactate shuttle theory. Cell Metab. 27:757–785. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W, Wang G, Xu ZG, Tu H, Hu F, Dai J, Chang Y, Chen Y, Lu Y, Zeng H, et al: Lactate is a natural suppressor of rlr signaling by targeting MAVS. Cell. 178:176–189. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chang CH, Curtis JD, Maggi LB Jr, Faubert B, Villarino AV, O'Sullivan D, Huang SC, van der Windt GJ, Blagih J, Qiu J, et al: Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell. 153:1239–1251. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Xia H, Wang W, Crespo J, Kryczek I, Li W, Wei S, Bian Z, Maj T, He M, Liu RJ, et al: Suppression of FIP200 and autophagy by tumor-derived lactate promotes naïve T cell apoptosis and affects tumor immunity. Sci Immunol. 2:eaan46312017. View Article : Google Scholar : PubMed/NCBI | |
|
Arruga F, Gyau BB, Iannello A, Vitale N, Vaisitti T and Deaglio S: Immune response dysfunction in chronic lymphocytic leukemia: Dissecting molecular mechanisms and microenvironmental conditions. Int J Mol Sci. 21:18252020. View Article : Google Scholar : PubMed/NCBI | |
|
Corbet C and Feron O: Tumour acidosis: From the passenger to the driver's seat. Nat Rev Cancer. 17:577–593. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Sukumar M, Liu J, Ji Y, Subramanian M, Crompton JG, Yu Z, Roychoudhuri R, Palmer DC, Muranski P, Karoly ED, et al: Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J Clin Invest. 123:4479–4488. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Nasi A, Fekete T, Krishnamurthy A, Snowden S, Rajnavölgyi E, Catrina AI, Wheelock CE, Vivar N and Rethi B: Dendritic cell reprogramming by endogenously produced lactic acid. J Immunol. 191:3090–3099. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, Gottfried E, Schwarz S, Rothe G, Hoves S, et al: Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 109:3812–3819. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Lin S, Sun L, Lyu X, Ai X, Du D, Su N, Li H, Zhang L, Yu J and Yuan S: Lactate-activated macrophages induced aerobic glycolysis and epithelial-mesenchymal transition in breast cancer by regulation of CCL5-CCR5 axis: A positive metabolic feedback loop. Oncotarget. 8:110426–110443. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zappasodi R, Serganova I, Cohen IJ, Maeda M, Shindo M, Senbabaoglu Y, Watson MJ, Leftin A, Maniyar R, Verma S, et al: CTLA-4 blockade drives loss of T(reg) stability in glycolysis-low tumours. Nature. 591:652–658. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu N, Luo J, Kuang D, Xu S, Duan Y, Xia Y, Wei Z, Xie X, Yin B, Chen F, et al: Lactate inhibits ATP6V0d2 expression in tumor-associated macrophages to promote HIF-2α-mediated tumor progression. J Clin Invest. 129:631–646. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gao Y, Zhou H, Liu G, Wu J, Yuan Y and Shang A: Tumor microenvironment: Lactic acid promotes tumor development. J Immunol Res. 2022:31193752022. View Article : Google Scholar : PubMed/NCBI | |
|
Langin D: Adipose tissue lipolysis revisited (again!): Lactate involvement in insulin antilipolytic action. Cell Metab. 11:242–243. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Pan Y, Yu Y, Wang X and Zhang T: Corrigendum: Tumor-associated macrophages in tumor immunity. Front Immunol. 12:7757582021. View Article : Google Scholar : PubMed/NCBI | |
|
El-Kenawi A, Gatenbee C, Robertson-Tessi M, Bravo R, Dhillon J, Balagurunathan Y, Berglund A, Vishvakarma N, Ibrahim-Hashim A, Choi J, et al: Acidity promotes tumour progression by altering macrophage phenotype in prostate cancer. Br J Cancer. 121:556–566. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Q, Du F, Huang W, Ding X, Wang Z, Yan F and Wu Z: Epigenetic control of Foxp3 in intratumoral T-cells regulates growth of hepatocellular carcinoma. Aging (Albany NY). 11:2343–2351. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yu J, Chai P, Xie M, Ge S, Ruan J, Fan X and Jia R: Histone lactylation drives oncogenesis by facilitating m6A reader protein YTHDF2 expression in ocular melanoma. Genome Biol. 22:852021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang J, Huang D, Jiang Y, Hou J, Tian M, Li J, Sun L, Zhang Y, Zhang T, Li Z, et al: Lactate modulates cellular metabolism through histone lactylation-mediated gene expression in non-small cell lung cancer. Front Oncol. 11:6475592021. View Article : Google Scholar : PubMed/NCBI | |
|
Certo M, Tsai CH, Pucino V, Ho PC and Mauro C: Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat Rev Immunol. 21:151–161. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang N, Wang W, Wang X, Mang G, Chen J, Yan X, Tong Z, Yang Q, Wang M, Chen L, et al: Histone lactylation boosts reparative gene activation post-myocardial infarction. Circ Res. 131:893–908. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Mathupala SP, Ko YH and Pedersen PL: Hexokinase II: Cancer's double-edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria. Oncogene. 25:4777–4786. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Majmundar AJ, Wong WJ and Simon MC: Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell. 40:294–309. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang LF, Lou JT, Lu MH, Gao C, Zhao S, Li B, Liang S, Li Y, Li D and Liu MF: Suppression of miR-199a maturation by HuR is crucial for hypoxia-induced glycolytic switch in hepatocellular carcinoma. EMBO J. 34:2671–2685. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Bao MH and Wong CC: Hypoxia, metabolic reprogramming, and drug resistance in liver cancer. Cells. 10:17152021. View Article : Google Scholar : PubMed/NCBI | |
|
Reyes A, Duarte LF, Farías MA, Tognarelli E, Kalergis AM, Bueno SM and González PA: Impact of hypoxia over human viral infections and key cellular processes. Int J Mol Sci. 22:79542021. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng H, Ning Y, Zhan Y, Liu S, Yang Y, Wen Q and Fan S: Co-expression of PD-L1 and HIF-1α predicts poor prognosis in patients with non-small cell lung cancer after surgery. J Cancer. 12:2065–2072. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Bandopadhyay S and Patranabis S: Mechanisms of HIF-driven immunosuppression in tumour microenvironment. J Egypt Natl Canc Inst. 35:272023. View Article : Google Scholar : PubMed/NCBI | |
|
You L, Wang X, Wu W, Nepovimova E, Wu Q and Kuca K: HIF-1α inhibits T-2 toxin-mediated ‘immune evasion’ process by negatively regulating PD-1/PD-L1. Toxicology. 480:1533242022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Z, Su W, Wei X, Pan Y, Xing M, Niu L, Feng B, Kong W, Ren X, Huang F, et al: Hypoxia inducible factor-1α drives cancer resistance to cuproptosis. Cancer Cell. 43:937–954. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Chen CH, Li SX, Xiang LX, Mu HQ, Wang SB and Yu KY: HIF-1α induces immune escape of prostate cancer by regulating NCR1/NKp46 signaling through miR-224. Biochem Biophys Res Commun. 503:228–234. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Facciabene A, Peng X, Hagemann IS, Balint K, Barchetti A, Wang LP, Gimotty PA, Gilks CB, Lal P, Zhang L and Coukos G: Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature. 475:226–230. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Peng J, Wang X, Ran L, Song J, Luo R and Wang Y: Hypoxia-inducible factor 1α regulates the transforming growth factor β1/SMAD family member 3 pathway to promote breast cancer progression. J Breast Cancer. 21:259–266. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Clambey ET, McNamee EN, Westrich JA, Glover LE, Campbell EL, Jedlicka P, de Zoeten EF, Cambier JC, Stenmark KR, Colgan SP and Eltzschig HK: Hypoxia-inducible factor-1 alpha-dependent induction of FoxP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc Natl Acad Sci USA. 109:E2784–E2793. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Sormendi S and Wielockx B: Hypoxia pathway proteins as central mediators of metabolism in the tumor cells and their microenvironment. Front Immunol. 9:402018. View Article : Google Scholar : PubMed/NCBI | |
|
Balamurugan K: HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int J Cancer. 138:1058–1066. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Murthy A, Gerber SA, Koch CJ and Lord EM: Intratumoral hypoxia reduces IFN-γ-mediated immunity and mhc class I induction in a preclinical tumor model. Immunohorizons. 3:149–160. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Bhandari T, Olson J, Johnson RS and Nizet V: HIF-1α influences myeloid cell antigen presentation and response to subcutaneous OVA vaccination. J Mol Med (Berl). 91:1199–1205. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Ruiz-Iglesias A and Mañes S: The importance of mitochondrial pyruvate carrier in cancer cell metabolism and tumorigenesis. Cancers (Basel). 13:14882021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang T, Yu-Jing L and Ma T: The immunomodulatory function of adenosine in sepsis. Front Immunol. 13:9365472022. View Article : Google Scholar : PubMed/NCBI | |
|
Haskó G, Linden J, Cronstein B and Pacher P: Adenosine receptors: Therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 7:759–770. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Chambers AM and Matosevic S: Immunometabolic dysfunction of natural killer cells mediated by the hypoxia-CD73 axis in solid tumors. Front Mol Biosci. 6:602019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao F, Xiao C, Evans KS, Theivanthiran T, DeVito N, Holtzhausen A, Liu J, Liu X, Boczkowski D, Nair S, et al: Paracrine Wnt5a-β-catenin signaling triggers a metabolic program that drives dendritic cell tolerization. Immunity. 48:147–160. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Godin-Ethier J, Hanafi LA, Piccirillo CA and Lapointe R: Indoleamine 2,3-dioxygenase expression in human cancers: Clinical and immunologic perspectives. Clin Cancer Res. 17:6985–6991. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Irshad Z, Xue M, Ashour A, Larkin JR, Thornalley PJ and Rabbani N: Activation of the unfolded protein response in high glucose treated endothelial cells is mediated by methylglyoxal. Sci Rep. 9:78892019. View Article : Google Scholar : PubMed/NCBI | |
|
Siska PJ, Beckermann KE, Mason FM, Andrejeva G, Greenplate AR, Sendor AB, Chiang YJ, Corona AL, Gemta LF, Vincent BG, et al: Mitochondrial dysregulation and glycolytic insufficiency functionally impair CD8 T cells infiltrating human renal cell carcinoma. JCI Insight. 2:e934112017. View Article : Google Scholar : PubMed/NCBI | |
|
Blagih J, Coulombe F, Vincent EE, Dupuy F, Galicia-Vázquez G, Yurchenko E, Raissi TC, van der Windt GJ, Viollet B, Pearce EL, et al: The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity. 42:41–54. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Zhao Y, Wu X, Liu Z and Liu X: A novel strategy to fuel cancer immunotherapy: Targeting glucose metabolism to remodel the tumor microenvironment. Front Oncol. 12:9311042022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Mo W, Ye J, Li L, Zhang Y, Hsueh EC, Hoft DF and Peng G: Regulatory T cells trigger effector T cell DNA damage and senescence caused by metabolic competition. Nat Commun. 9:2492018. View Article : Google Scholar : PubMed/NCBI | |
|
Scharping NE and Delgoffe GM: Tumor microenvironment metabolism: A new checkpoint for anti-tumor immunity. Vaccines (Basel). 4:462016. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Y, Li Y and Zhu B: T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 6:e17922015. View Article : Google Scholar : PubMed/NCBI | |
|
Bader JE, Voss K and Rathmell JC: Targeting metabolism to improve the tumor microenvironment for cancer immunotherapy. Mol Cell. 78:1019–1033. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Geraghty KM, Chen S, Harthill JE, Ibrahim AF, Toth R, Morrice NA, Vandermoere F, Moorhead GB, Hardie DG and MacKintosh C: Regulation of multisite phosphorylation and 14-3-3 binding of AS160 in response to IGF-1, EGF, PMA and AICAR. Biochem J. 407:231–241. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Q, Yang Z, Hao X, Dandreo LJ, He L, Zhang Y, Wang F, Wu X and Xu L: Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling. Cell Biosci. 13:1922023. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Zheng M, Wu S, Gao S, Yang M, Li Z, Min Q, Sun W, Chen L, Xiang G and Li H: Benserazide, a dopadecarboxylase inhibitor, suppresses tumor growth by targeting hexokinase 2. J Exp Clin Cancer Res. 36:582017. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng M, Wu C, Yang K, Yang Y, Liu Y, Gao S, Wang Q, Li C, Chen L and Li H: Novel selective hexokinase 2 inhibitor Benitrobenrazide blocks cancer cells growth by targeting glycolysis. Pharmacol Res. 164:1053672021. View Article : Google Scholar : PubMed/NCBI | |
|
VDA-1102, a first-in-class VDAC/HK modulator entering phase 1/2 drug development for treatment of actinic keratosis, cutaneous squamous cell carcinoma. J AM ACAD DERMATOL. 74:2016. | |
|
Lozzi F, Lanna C, Mazzeo M, Garofalo V, Palumbo V, Mazzilli S, Diluvio L, Terrinoni A, Bianchi L and Campione E: Investigational drugs currently in phase II clinical trials for actinic keratosis. Expert Opin Investig Drugs. 28:629–642. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng Y, Zhan Y, Zhang Y, Zhang Y, Liu Y, Xie Y, Sun Y, Qian J, Ding Y, Ding Y and Fang Y: Hexokinase 2 confers radio-resistance in hepatocellular carcinoma by promoting autophagy-dependent degradation of AIMP2. Cell Death Dis. 14:4882023. View Article : Google Scholar : PubMed/NCBI | |
|
Lin H, Zeng J, Xie R, Schulz MJ, Tedesco R, Qu J, Erhard KF, Mack JF, Raha K, Rendina AR, et al: Discovery of a novel 2,6-disubstituted glucosamine series of potent and selective hexokinase 2 inhibitors. ACS Med Chem Lett. 7:217–222. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang SH, Dong FY, Da LT, Yang XM, Wang XX, Weng JY, Feng L, Zhu LL, Zhang YL, Zhang ZG, et al: Ikarugamycin inhibits pancreatic cancer cell glycolysis by targeting hexokinase 2. FASEB J. 34:3943–3955. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Bao F, Yang K, Wu C, Gao S, Wang P, Chen L and Li H: New natural inhibitors of hexokinase 2 (HK2): Steroids from Ganoderma sinense. Fitoterapia. 125:123–129. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Zhuang Y, Xu J, Tong Y, Li X and Dong C: Advances in the study of hexokinase 2 (HK2) inhibitors. Anticancer Agents Med Chem. 23:736–746. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Z, Zhang D, Zhang Z, Luo W, Shi R, Yao J, Li D, Wang L and Liao B: MicroRNA-505, suppressed by oncogenic long non-coding RNA LINC01448, acts as a novel suppressor of glycolysis and tumor progression through inhibiting HK2 expression in pancreatic cancer. Front Cell Dev Biol. 8:6250562020. View Article : Google Scholar : PubMed/NCBI | |
|
Ye J, Xiao X, Han Y, Fan D, Zhu Y and Yang L: MiR-3662 suppresses cell growth, invasion and glucose metabolism by targeting HK2 in hepatocellular carcinoma cells. Neoplasma. 67:773–781. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng C, Li R, Zheng S, Fang H, Xu M and Zhong L: The knockdown of lncRNA DLGAP1-AS2 suppresses osteosarcoma progression by inhibiting aerobic glycolysis via the miR-451a/HK2 axis. Cancer Sci. 114:4747–4762. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu W, Huang Y, Pan Q, Xiang P, Xie N and Yu H: MicroRNA-98 suppress warburg effect by targeting HK2 in colon cancer cells. Dig Dis Sci. 62:660–668. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Guo Y, Liang F, Zhao F and Zhao J: Resibufogenin suppresses tumor growth and Warburg effect through regulating miR-143-3p/HK2 axis in breast cancer. Mol Cell Biochem. 466:103–115. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong J, Lu S, Jia X, Li Q, Liu L, Xie P, Wang G, Lu M, Gao W, Zhao T, et al: Role of endoplasmic reticulum stress in apoptosis induced by HK2 inhibitor and its potential as a new drug combination strategy. Cell Stress Chaperones. 27:273–283. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Agnihotri S, Mansouri S, Burrell K, Li M, Mamatjan Y, Liu J, Nejad R, Kumar S, Jalali S, Singh SK, et al: Ketoconazole and posaconazole selectively target HK2-expressing glioblastoma cells. Clin Cancer Res. 25:844–855. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Afonso J, Gonçalves C, Costa M, Ferreira D, Santos L, Longatto-Filho A and Baltazar F: Glucose metabolism reprogramming in bladder cancer: Hexokinase 2 (HK2) as prognostic biomarker and target for bladder cancer therapy. Cancers (Basel). 15:9822023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L, Yan X, Chen J, Zhan Q, Hua Y, Xu S, Li Z, Wang Z, Dong Y, Zuo D, et al: Hexokinase 2 discerns a novel circulating tumor cell population associated with poor prognosis in lung cancer patients. Proc Natl Acad Sci USA. 118:e20122281182021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu S, Catapang A, Doh HM, Bayley NA, Lee JT, Braas D, Graeber TG and Herschman HR: Hexokinase 2 is targetable for HK1 negative, HK2 positive tumors from a wide variety of tissues of origin. J Nucl Med. 60:212–217. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Shurin MR and Umansky V: Cross-talk between HIF and PD-1/PD-L1 pathways in carcinogenesis and therapy. J Clin Invest. 132:e1594732022. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu J, Zhong F, Zhang Z, Pan B, Ye D, Zhang X, Yao Y, Luo Y, Wang X and Tang N: Hypoxia-responsive lncRNA MIR155HG promotes PD-L1 expression in hepatocellular carcinoma cells by enhancing HIF-1α mRNA stability. Int Immunopharmacol. 136:1124152024. View Article : Google Scholar : PubMed/NCBI | |
|
Xu H, Chen Y, Li Z, Zhang H, Liu J and Han J: The hypoxia-inducible factor 1 inhibitor LW6 mediates the HIF-1α/PD-L1 axis and suppresses tumor growth of hepatocellular carcinoma in vitro and in vivo. Eur J Pharmacol. 930:1751542022. View Article : Google Scholar : PubMed/NCBI | |
|
Dai X, Pi G, Yang SL, Chen GG, Liu LP and Dong HH: Association of PD-L1 and HIF-1α coexpression with poor prognosis in hepatocellular carcinoma. Transl Oncol. 11:559–566. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Desta GM and Birhanu AG: Advancements in single-cell RNA sequencing and spatial transcriptomics: Transforming biomedical research. Acta Biochim Pol. 72:139222025. View Article : Google Scholar : PubMed/NCBI | |
|
Shen X, Zhao Y, Wang Z and Shi Q: Recent advances in high-throughput single-cell transcriptomics and spatial transcriptomics. Lab Chip. 22:4774–4791. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Elosua-Bayes M, Nieto P, Mereu E, Gut I and Heyn H: SPOTlight: Seeded NMF regression to deconvolute spatial transcriptomics spots with single-cell transcriptomes. Nucleic Acids Res. 49:e502021. View Article : Google Scholar : PubMed/NCBI | |
|
Kleshchevnikov V, Shmatko A, Dann E, Aivazidis A, King HW, Li T, Elmentaite R, Lomakin A, Kedlian V, Gayoso A, et al: Cell2location maps fine-grained cell types in spatial transcriptomics. Nat Biotechnol. 40:661–671. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Biran H, Hashimshony T, Lahav T, Efrat O, Mandel-Gutfreund Y and Yakhini Z: Detecting significant expression patterns in single-cell and spatial transcriptomics with a flexible computational approach. Sci Rep. 14:261212024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Dai R, Wang M, Lei L, Zhang Z, Han K, Wang Z and Guo Q: KanCell: Dissecting cellular heterogeneity in biological tissues through integrated single-cell and spatial transcriptomics. J Genet Genomics. 52:689–705. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Ravindran U and Gunavathi C: Deep learning assisted cancer disease prediction from gene expression data using WT-GAN. BMC Med Inform Decis Mak. 24:3112024. View Article : Google Scholar : PubMed/NCBI | |
|
Hu B, Wang Z, Zeng H, Qi Y, Chen Y, Wang T, Wang J, Chang Y, Bai Q, Xia Y, et al: Blockade of DC-SIGN(+) tumor-associated macrophages reactivates antitumor immunity and improves immunotherapy in muscle-invasive bladder cancer. Cancer Res. 80:1707–1719. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang H, Zhang Q, Xu M, Wang L, Chen X, Feng Y, Li Y, Zhang X, Cui W and Jia X: CCL2-CCR2 axis recruits tumor associated macrophages to induce immune evasion through PD-1 signaling in esophageal carcinogenesis. Mol Cancer. 19:412020. View Article : Google Scholar : PubMed/NCBI | |
|
Wolf-Dennen K, Gordon N and Kleinerman ES: Exosomal communication by metastatic osteosarcoma cells modulates alveolar macrophages to an M2 tumor-promoting phenotype and inhibits tumoricidal functions. Oncoimmunology. 9:17476772020. View Article : Google Scholar : PubMed/NCBI | |
|
Rodriguez-Perdigon M, Haeni L, Rothen-Rutishauser B and Rüegg C: Dual CSF1R inhibition and CD40 activation demonstrates anti-tumor activity in a 3D macrophage- HER2(+) breast cancer spheroid model. Front Bioeng Biotechnol. 11:11598192023. View Article : Google Scholar : PubMed/NCBI | |
|
Kinouchi M, Miura K, Mizoi T, Ishida K, Fujibuchi W, Sasaki H, Ohnuma S, Saito K, Katayose Y, Naitoh T, et al: Infiltration of CD40-positive tumor-associated macrophages indicates a favorable prognosis in colorectal cancer patients. Hepatogastroenterology. 60:83–88. 2013.PubMed/NCBI | |
|
Shvefel SC, Pai JA, Cao Y, Pal LR, Bartok O, Levy R, Zemanek MJ, Weller C, Herzog E, Yao W, et al: Temporal genomic analysis of homogeneous tumor models reveals key regulators of immune evasion in melanoma. Cancer Discov. 28:OF1–OF25. 2024. |