Next Article in Journal
α-L-Fucosidases from an Alpaca Faeces Metagenome: Characterisation of Hydrolytic and Transfucosylation Potential
Next Article in Special Issue
Polyphenolic Characterization and Anti-Inflammatory Effect of In Vitro Digested Extracts of Echinacea purpurea L. Plant Parts in an Inflammatory Model of Human Colon Cells
Previous Article in Journal
Role of Clay Substrate Molecular Interactions in Some Dairy Technology Applications
Previous Article in Special Issue
Menadione Contribution to the In Vitro Radical Scavenging Potential of Phytochemicals Naringenin and Lignin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Product-Based Glycolysis Inhibitors as a Therapeutic Strategy for Epidermal Growth Factor Receptor–Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer

by
Wonyoung Park
1,2,†,
Jung Ho Han
3,†,
Shibo Wei
4,
Eun-Sun Yang
2,
Se-Yun Cheon
2,
Sung-Jin Bae
5,
Dongryeol Ryu
6,
Hwan-Suck Chung
3,* and
Ki-Tae Ha
1,2,*
1
Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
2
Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea
3
Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea
4
Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
5
Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan 49267, Republic of Korea
6
Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(2), 807; https://doi.org/10.3390/ijms25020807
Submission received: 5 December 2023 / Revised: 2 January 2024 / Accepted: 5 January 2024 / Published: 9 January 2024
(This article belongs to the Special Issue Molecular Mechanism Study of Natural Products for Human Health)

Abstract

:
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide. Targeted therapy against the epidermal growth factor receptor (EGFR) is a promising treatment approach for NSCLC. However, resistance to EGFR tyrosine kinase inhibitors (TKIs) remains a major challenge in its clinical management. EGFR mutation elevates the expression of hypoxia-inducible factor-1 alpha to upregulate the production of glycolytic enzymes, increasing glycolysis and tumor resistance. The inhibition of glycolysis can be a potential strategy for overcoming EGFR-TKI resistance and enhancing the effectiveness of EGFR-TKIs. In this review, we specifically explored the effectiveness of pyruvate dehydrogenase kinase inhibitors and lactate dehydrogenase A inhibitors in combating EGFR-TKI resistance. The aim was to summarize the effects of these natural products in preclinical NSCLC models to provide a comprehensive understanding of the potential therapeutic effects. The study findings suggest that natural products can be promising inhibitors of glycolytic enzymes for the treatment of EGFR-TKI-resistant NSCLC. Further investigations through preclinical and clinical studies are required to validate the efficacy of natural product-based glycolytic inhibitors as innovative therapeutic modalities for NSCLC.

1. Introduction

Lung cancer is the leading cause of cancer-related deaths, with approximately 1.8 million deaths worldwide in 2020 [1]. Non-small cell lung cancer (NSCLC; 82%) and small cell lung cancer (SCLC; 14%) account for the majority of cases of lung cancer [2,3]. Overall, NSCLC is the most frequent type of lung cancer. Based on the histological characteristics, NSCLC is further classified into lung adenocarcinoma (50–60%), squamous cell carcinoma (20–30%), and large cell carcinoma (10–20%) [4].
The epidermal growth factor receptor (EGFR) is one of the most common driving mutations in NSCLC [5]. EGFR mutations are more common in Asian populations (approximately 50%) than in populations from the United States and Europe (approximately 10%) [6]. Consequently, EGFR is one of the most significant targetable mutations in NSCLC and is widely explored in cancer research, medication development, and diagnosis. A decade ago, the average survival of patients with advanced NSCLC and EGFR mutations was less than 2 years [7]. Currently, patients receiving third-generation EGFR tyrosine kinase inhibitor (TKI) treatment have a median survival time of more than 3 years [8]. A third-generation EGFR-TKI (osimertinib) has good treatment efficacy but is also associated with the development of secondary resistance [9]. Therefore, overcoming EGFR-TKI resistance is important.
Emerging evidence highlights the correlation between glycolysis and drug resistance in cancer. Within cancer cells, glycolysis-related enzymes play pivotal roles in enhancing resistance to chemotherapy [10]. Pyruvate dehydrogenase kinase (PDK) is a key enzyme that is involved in the regulation of glucose metabolism and is frequently overexpressed in cancer cells, resulting in increased glycolysis and lactate production [11]. Moreover, PDK inhibition has been demonstrated to reduce cancer cell growth and enhance susceptibility to chemotherapy [12]. Lactate dehydrogenase (LDH) regulates the conversion and production of pyruvate and lactic acid [13]. Inhibition of LDH A (LDHA) increases oxidative stress and reduces chemoresistance [14]. Therefore, targeting glycolytic enzymes such as PDK or LDHA may be a promising strategy to overcome EGFR-TKI resistance in patients with NSCLC.
Natural products have the potential to be effective as therapeutic agents because of their lower toxicity and higher specificity than synthetic compounds [15]. A growing interest has been observed in identifying glycolysis inhibitors from natural sources that can be used as safe and effective alternatives [16,17]. These inhibitors have demonstrated promising results in preclinical models [18,19,20]. Therefore, we compiled natural products-derived inhibitors of enzymes that are involved in glycolysis, including PDKs and LDHA, with the aim of understanding the potential of these compounds in overcoming EGFR-TKI resistance.
This review provides an overview of the current understanding of the role of targeted therapy in NSCLC, delves into the historical processes underlying EGFR-TKI resistance, and explores the potential of utilizing glycolysis inhibitors that are derived from natural products as a strategy for overcoming EGFR-TKI resistance.

2. Targeted Therapy in NSCLC

The treatment for NSCLC is multifaceted and can be tailored to meet the needs of individual patients. Surgical interventions, including lobectomy and wedge resection, are designed to surgically remove tumors and associated lymph nodes. Radiation therapy utilizes high-energy X-rays to target cancer cells, and chemotherapy employs drugs such as carboplatin and cisplatin to disrupt the growth of these cells. A pivotal shift in the treatment paradigm has occurred with the identification of specific targetable mutations in patients with advanced NSCLC [21], and the use of drugs that are designed for these genetic mutations or altered proteins that promote cancer cell growth and spread [22]. The targeted therapies include those directed towards EGFR, anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1, receptor tyrosine kinase (ROS1), v-raf murine sarcoma viral oncogene homolog B1 (BRAF), proto-oncogene, receptor tyrosine kinase (MET), RET proto-oncogene (RET), Kirsten rat sarcoma virus (KRAS), and programmed death-ligand 1 (PD-L1), and they work by disrupting the signaling pathways that are responsible for cancer cell growth [22]. Unlike conventional treatments, targeted therapy allows for a more precise and personalized approach which enhances treatment efficacy while minimizing side effects and provides a valuable alternative for patients who may not respond well to standard therapies. Additionally, targeted therapy plays a key role in overcoming resistance to traditional treatments and recognizes the unique genetic profile of each patient with cancer [23]. The integration of targeted therapy into NSCLC treatment strategies represents a significant advancement in improving patient outcomes and underscores the necessity for a nuanced and tailored therapeutic approach in the era of precision medicine.
The efficacy of targeted therapies for NSCLC varies depending on individual patient characteristics and genetic testing results. To determine the best therapeutic options for individual patients, NSCLC cells are subjected to molecular profiling. In 2022, the National Comprehensive Cancer Network (NCCN) expanded its guidelines for metastatic NSCLC to include “broad molecular profiles including EGFR (Figure 1A), ALK, HER2, MET, NTRK, RET, ROS1 (Figure 1B), KRAS, BRAF (Figure 1C), and PD-L1 (Figure 1D) [24]. The NCCN guidelines recommend several Food and Drug Administration (FDA)-approved targeted therapeutic agents as first-line therapies for patients with specific mutations. Afatinib [25], dacomitinib [26], erlotinib [27], gefitinib [28], and osimertinib [29] are utilized for patients with the EGFR mutation; amivantamab-vmjw [30] and mobocertinib are employed for patients with EGFR exon 20 insertions; targeted therapy for other receptor tyrosine kinases (ALK, HER2, MET, NTRK, RET, ROS1) include alectinib [31], brigatinib [32], capmatinib [33], ceritinib [34], crizotinib [35], entrectinib [36], larotrectinib [37], lorlatinib [38], pralsetinib [39], selpercatinib [40], and tepotinib [41] (Figure 2). Sotorasib [42] has been approved for KRAS mutations, and dabrafenib [43] and trametinib [43] have been approved for BRAF mutations. Immunotherapy targeting PD-L1 is also considered a form of targeted therapy. The FDA has approved atezolizumab [44], bevacizumab [45], ipilimumab [46], nivolumab [47], and pembrolizumab [48] as PD-L1 targeting therapies. Monoclonal antibodies are a type of targeted therapy which were initially developed as a cancer therapy targeting EGFR [49]. Necitumumab, an EGFR monoclonal antibody, has been approved as a combination therapy for metastatic squamous NSCLC [50].

3. EGFR-TKIs in NSCLC Treatment

EGFR, an oncogenic receptor tyrosine kinase (TK) belonging to the ErbB receptor family, is activated upon binding to specific ligands, including epidermal growth factor (EGF) (Figure 3) [51]. Several isotypic ErbB family receptors, such as human epidermal growth factor receptor (HER) 2, HER3, and HER4, play key roles in the development of NSCLC. In normal cells, EGFR activation leads to receptor homo- or hetero-dimerization and autophosphorylation of the intracellular TK domain, which in turn activates signaling pathways that regulate cellular proliferation, migration, and differentiation [52,53]. However, EGFR is frequently altered in tumor cells, and these alterations can lead to abnormal signaling, resulting in cancer cell proliferation, invasion, and metastasis [54]. To attenuate the effects of EGFR mutation-induced aberrant signaling, EGFR-TKIs have been developed to inhibit enzymatic activity by binding to the TK domain of EGFRs [55].
Mutations in kinase-activating EGFRs and overexpression of the EGFR protein are the predominant changes observed in cancer (Figure 1A) [60,61]. The most common EGFR-activating mutations include an in-frame deletion in exon 19 within codons 746–750 (19D; 45–50%) and a single-base substitution of arginine with leucine at codon 858 in exon 21 (L858R; approximately 35–45%) near the adenosine triphosphate (ATP)-binding pocket of the TK domain [54,62]. First-generation EGFR-TKIs (gefitinib and erlotinib), which reversibly bind to the ATP-binding site of the EGFR tyrosine kinase domain, have resulted in considerable improvements in the outcome for patients with EGFR-mutated NSCLC (L858R and 19D) (Table 1) [63,64]. Additionally, less common EGFR mutations such as G719X, L861Q, and S768I have demonstrated responsiveness to first-generation EGFR-TKI treatment [65,66]. According to a recent study, patients who received subsequent EGFR-TKI treatment lived the longest, with a median overall survival (OS) of 31.3 months (95% confidence interval (CI), 23.9–38.7 months) compared to those who received chemotherapy (median OS, 19.4 months; 95% CI, 18.5–20.3 months) or no subsequent treatment (median OS, 2.4 months; 95% CI, 1.3–3.5 months) [67]. This study suggests that patients with EGFR-mutated NSCLC may benefit from further treatment with EGFR-TKIs. However, after treatment with gefitinib, erlotinib, or afatinib (second-generation EGFR-TKIs) for approximately 9–14 months, up to 50% of the patients developed T790M-mediated resistance [68,69]. In comparison to gefitinib, dacomitinib, a second-generation EGFR-TKI, has significantly improved progression-free survival in the first-line treatment of patients with EGFR-mutation-positive NSCLC; however, the drug also has the potential to directly induce secondary mutations such as T790M [70,71]. Osimertinib is a third-generation EGFR-TKI that binds to the C797 residue in the ATP-binding site of EGFR and exhibits high selectivity for both EGFR-activating mutations and the secondary acquired EGFR T790M mutation [9]. Patients treated with osimertinib had a median OS of 38.6 months (95% CI, 34.5–41.8), whereas those in the comparator group had a median OS of 31.8 months [8]. Osimertinib is a promising third-generation EGFR-TKI, and its combination with platinum-based chemotherapy may provide additional treatment options for EGFR-mutated NSCLC [72]. Although the development of targeted therapies including the first-generation (gefitinib and erlotinib), second-generation (afatinib and dacomitinib), and third-generation (osimertinib) EGFR-TKIs has demonstrated substantial improvements in the overall survival of patients with EGFR-mutated NSCLC, the emergence of secondary resistance is challenging [73]. Thus, continued research and exploration of novel therapeutic strategies are imperative to address and overcome these resistance mechanisms and ensure sustained efficacy and prolonged benefits for patients with EGFR-mutated NSCLC.

4. Enhanced Glycolysis in EGFR-TKI-Resistant NSCLC

Glycolysis is a metabolic pathway that converts glucose to lactate. Moreover, glycolysis is orchestrated by a series of glycolytic enzymes, and the dysregulation of the procedure has been implicated in conferring resistance to EGFR-targeted therapies in cancer cells [79]. The cancer cells often favor glycolysis (the Warburg effect), despite the presence of oxygen [80]. The Warburg effect suggests that cancer cells favor glycolysis even when oxygen is available, because glycolysis is advantageous for their rapid division [81]. Although glycolysis is less efficient in terms of energy production, the process allows cancer cells to generate energy quickly and provides essential biosynthetic precursors for the synthesis of various cellular components that are required for cell growth and division [82]. However, the production of free radicals during glycolysis poses a potential challenge, as these reactive oxygen species (ROS) can have damaging effects on cellular components including deoxyribonucleic acid (DNA), proteins, and lipids [83]. However, the NSCLC cells exhibit several adaptive mechanisms for managing proliferation despite oxidative stress. Cells in NSCLC employ a combination of antioxidant defenses and survival signaling, including the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, metabolic adaptations, DNA repair mechanisms, and adaptation to hypoxic conditions, to manage and sustain proliferation in the presence of oxidative stress during glycolysis [84,85,86,87]. The heightened glycolysis and increased lactate production observed in cancer cells significantly contribute to the development of resistance to EGFR-TKIs [88]. Acknowledging that the complex interplay between various glycolytic enzymes plays a pivotal role in mediating resistance to EGFR-TKIs is crucial. A previous study proposed that elevated glycolytic activity could be predictive of gefitinib resistance in patients with EGFR-mutant NSCLC receiving first-line gefitinib treatment [89]. This suggests that targeting the glycolytic pathway and its associated enzymes can be a promising avenue for novel therapeutic approaches aimed at overcoming EGFR-TKI resistance.
In EGFR-TKI-resistant NSCLC cells, increased glucose uptake is primarily facilitated by the upregulation of glucose transporter 1 (GLUT1), which is a critical regulator of glucose entry into cells [90]. This increase in GLUT1 enhances the efficiency of glucose transport across the cell membrane, ensuring a constant supply of glucose to fuel glycolysis (the preferred energy-producing pathway in resistant cells) [91]. A previous study discovered that inhibiting glycolysis using 2-deoxy-d-glucose enhanced sensitivity to afatinib (a second-generation irreversible EGFR-TK) in NSCLC cells with acquired resistance due to the secondary EGFR T790M mutation [92]. Hexokinase plays an important role in the early stages of glycolysis by catalyzing glucose phosphorylation, which is the first step of this metabolic pathway [93]. Additionally, the inhibition of hexokinase 2 (HK2) sensitizes resistant NSCLC cells to gefitinib. This is suggestive of an important role of HK2 in the development of resistance mechanisms [90]. Pyruvate kinase M2 (PKM2) is a crucial enzyme that regulates the final step of the glycolytic pathway and facilitates the transformation of phosphoenolpyruvate into pyruvate [94]. Additionally, PKM2 can translocate to the nucleus and activate the signal transducer and activator of transcription 3, which can cause resistance to gefitinib [95]. Pyruvate is a critical metabolite in cellular metabolism and is involved in several significant metabolic pathways depending on the cellular context and environmental factors [94]. Pyruvate regulation is complex and involves key enzymes, particularly PDK and LDHA. PDK governs the entry of pyruvate into the citric acid cycle and glycolysis [96], whereas LDHA catalyzes the conversion of pyruvate to lactate under anaerobic conditions [97]. Notably, PDK and LDHA are significantly associated with EGFR-TKI resistance. The interaction between PDK and LDHA will be explored in future studies.
The intricate regulation of glycolysis is a critical factor in EGFR-TKI resistance in NSCLC [98]. Targeting key players of the glycolytic pathway, such as GLUT1, HK2, PKM2, PDK, and LDHA, is a promising avenue for novel therapeutic strategies aimed at overcoming EGFR-TKI resistance [90,95,99,100,101]. Building on this understanding, the following section describes the exploration of glycolytic inhibitors that are derived from natural products and provides insights into potential nature-inspired interventions to disrupt this crucial metabolic pathway and enhance the efficacy of EGFR-targeted therapies.

5. Advantages of PDK Inhibition against EGFR-TKI Resistance and Inhibitors from Natural Products

PDK regulates the activity of the pyruvate dehydrogenase complex (PDC), which converts pyruvate to acetyl-CoA for mitochondrial ATP production [102]. PDK inhibits PDC activity via phosphorylation, resulting in a decreased conversion of pyruvate to acetyl-CoA. Instead, pyruvate is diverted toward lactate production via glycolysis [103]. Cancer cells exhibit high levels of PDK1 expression and activity, which promotes glycolysis and facilitates cancer cell survival and proliferation [104]. The induction of PDK by hypoxia-inducible factor-1 alpha (HIF-1α) has been demonstrated to cause chemotherapy resistance in cancer cells [12]. HIF-1α-induced upregulation of PDK1 inhibits PDC activity, causing a shift in the cancer cell metabolism towards anaerobic glycolysis, and decreases the production of ROS [105]. Consequently, cancer cells become resistant to chemotherapeutic drugs that rely on the cytotoxic effects of ROS [106]. In addition, the shift towards anaerobic glycolysis provides cancer cells with a metabolic advantage, allowing them to survive and proliferate in conditions with limited oxygen and nutrients [79]. Inhibiting PDK allows more pyruvate to enter the mitochondria, promoting oxidative phosphorylation (OXPHOS) and enhancing the production of ROS. This, in turn, causes oxidative stress, damages cellular components, and triggers apoptotic pathways, ultimately leading to cancer cell death [18,19,20,107]. Crystal structure studies have indicated that the pyruvate-binding domain (located at the N-terminal regulatory domain), the lipoamide-binding domain, and the nucleotide-binding domain (located at the C-terminal catalytic domain) are all critical for controlling PDK activity [108]. Dichloroacetate (DCA) is an orally available small-molecule PDK inhibitor that shifts the cancer cell metabolism from glycolysis to OXPHOS by inhibiting PDK activity [109]. In a previous study, the combination of DCA with the first-generation EGFR-TKIs erlotinib and gefitinib dramatically reduced the viability of EGFR-mutant NSCLC cells (NCI-H1975 and NCI-H1650) [110]. Another study suggested that DCA in combination with rociletinib (a third-generation EGFR-TKI) along with radiation therapy might be a promising therapeutic strategy for treating NSCLC [111].
Several natural products have been reported to have PDK-inhibiting activity, for example, huzhangoside A isolated from Anemone rivularis, ilimaquinone isolated from Smenospongia cerebriformis, and hemistepsin A isolated from, Hemistepta lyrate (Table 2) [18,19,20]. Although the precise IC50 values for the PDK enzyme activity of these natural products have not been reported, their anticancer effects have been confirmed using in vitro and/or in vivo studies of colon, lung, breast, and liver cancers. Dicoumarol from Melilotus officinalis, cryptotanshinone from Salvia miltiorrhiza, and quercetin from various fruits and vegetables exhibit inhibitory effects on PDK and have been demonstrated to have anticancer activities against hepatocellular carcinoma, pancreatic cancer, and lung cancer [112,113,114]. In addition, several natural products, such as baicalin, β-asarone, betulinic acid, cardamonin, and helichrysetin, are known to inhibit the expression of PDK1 [115,116,117,118,119]. Most of the substances mentioned above inhibit the upstream factors such as cellular-myelocytomatosis oncogene (c-Myc), HIF-1α, and phosphatase and tensin homolog/Akt that regulate the expression of PDK1. Although these natural products are not direct inhibitors of PDK1, they can inhibit glycolysis; therefore, they may exert effects that are similar to those of synthetic PDK inhibitors. Further clinical trials are needed to determine the efficacy and safety of these PDK inhibitors.
Therefore, PDK inhibition may be a useful therapeutic strategy for overcoming EGFR-TKI resistance. Nevertheless, additional studies are necessary to substantiate the mechanisms and clinical efficacy of PDK inhibitors in the treatment of EGFR-TKI-resistant NSCLC.

6. Natural Product-Derived LDHA Inhibitors and Their Advantage against EGFR-TKI Resistance

LDH plays vital roles in cellular respiration [123] by converting lactate to pyruvate or pyruvate to lactate, thereby maintaining an equilibrium between Nicotinamide adenine dinucleotide (NAD+) and its reduced form (NADH), which are essential elements in energy production [124,125]. In humans, LDH utilizes His193 as a proton acceptor and collaborates with coenzyme-binding residues (Arg99 and Asn138) and substrate-binding (Arg106, Arg169, and Thr248) residues [126]. Two main types of subunits are present in LDH, denoted as M (for muscle) and H (for heart) and encoded by genes—LDH-A and LDH-B, respectively [127]. The combination of these M (LDHA) and H (LDHB) subunits leads to the formation of tetrameric LDH of varying compositions (e.g., LDH-1, LDH-2, LDH-3, LDH-4, and LDH-5), each with different kinetic properties and tissue distributions [127]. Furthermore, LDHA is the most abundant isotype in the skeletal muscle and efficiently catalyzes the conversion of pyruvate to lactate and NADH to NAD+ [128]. In contrast, LDHB is predominantly present in the heart, liver, and brain, where it facilitates the conversion of lactate to pyruvate and NAD+ to NADH [127,129,130].
Abnormal LDH activity has been associated with a range of diseases including cancer, metabolic disorders, neurodegenerative diseases, and cardiovascular diseases [131,132,133,134]. In cancers, dysregulated LDH activity influences tumor progression by promoting the Warburg effect [135]. The conversion of pyruvate to lactate, favored by LDHA overexpression, partially contributes to the acidification of the tumor microenvironment [136]. This acidification is linked to the progression and metastasis of cancer and other diseases [79]. Elevated levels of plasma LDH can be used as a prognostic factor in patients with EGFR-mutated NSCLC [137,138]. However, LDHA inhibition may overcome EGFR TKI resistance.
Several LDHA inhibitors have been derived from natural products (Table 3). Apigenin has been reported to reduce LDHA messenger ribonucleic acid (RNA) expression [139]. Berberine improves ischemia/reperfusion injury by downregulating LDHA activity and subsequently decreases lactate production [140]. Capsaicin suppresses the EGF-induced invasion and migration of human fibrosarcoma cells [141]. In our previous study, catechin, which is known to enhance cardiovascular health and reduce oxidative stress and inflammation [142], exhibited a potent inhibitory effect on LDHA by directly binding to the Thr94, Ala95, Gln99, Arg105, Ser136, Arg168, His192, and Thr247 residues of LDHA [14]. Curcumin, extracted from Curcuma longa, inhibits glycolysis by downregulating the expression of HK2 and LDHA, thereby inducing mitochondria-mediated apoptosis in colorectal cancer cells [143]. Curcumin has been reported to overcome the resistance to EGFR-TKI (gefitinib and erlotinib) [144,145]. Epigallocatechin gallate (EGCG), a significant biologically active component of green tea, has been identified to have LDHA-inhibitory activity [146]. EGCG has a synergistic effect when used in combination with EGFR-TKI for head and neck cancer [147,148]. LDHA is a single-stranded DNA-binding protein that stimulates cell transcription [149]. Galloflavin acts as an inhibitor of LDHA, preventing it from binding to single-stranded DNA and reducing RNA production [150]. Leonurine (known for its cardioprotective effect) has been demonstrated to reduce LDH activity and has antioxidant properties [151]. Quercetin, extracted from Quercus, can decrease the activity and expression of LDHA, suppress PI3K/AKT signaling, and regress Dalton’s lymphoma growth [152]. Ursolic acid has antioxidant, antidiabetic, antibacterial, and anticancer effects [153] and can suppress LDHA expression in breast cancer [154].
The diverse range of natural LDHA inhibitors highlighted in this discussion, such as apigenin, berberine, capsaicin, catechin, curcumin, EGCG, galloflavin, leonurine, quercetin, and ursolic acid, demonstrate the potential of natural compounds to modulate LDHA activity and associated pathologies (Table 3). Curcumin and EGCG have emerged as promising candidates demonstrating efficacy as LDHA inhibitors and for overcoming EGFR-TKI resistance, particularly for cancer treatment. The interplay between LDHA and EGFR-TKI resistance presents a fascinating avenue for further investigation. Though the current research on natural remedy treatment options is relatively limited, exploring new options holds promise for advancing our understanding and refining treatment strategies.

7. Natural Products Suppressing Other Glycolytic Enzymes and Their Use for EGFR-TKI Resistance

Cucurbitacin D, a naturally occurring compound known for its anticancer properties, can impede the growth, invasion, and metastasis of prostate cancer cells by orchestrating the reprogramming of their glucose metabolism network [155]. Cucurbitacin D accomplishes this by binding to GLUT1, a membrane protein that facilitates glucose entry into cells and inhibits cell function, consequently diminishing glucose uptake by prostate cancer cells. Additionally, cucurbitacin D hampers ATP production in these cells, leading to apoptosis. Cucurbitacin B, another member of the cucurbitacin family, has exhibited therapeutic potential when combined with the EGFR-TKI gefitinib [156]. Genistein is a natural isoflavone that can directly downregulate HIF-1α, thereby inactivating GLUT1 and HK2 to suppress aerobic glycolysis [157]. α-Hederin, a pentacyclic triterpenoid saponin that is present in the leaves of Hedera helix, is known for anti-inflammatory, antioxidant, and anticancer properties [158]. Moreover, α-Hederin inhibits the growth of lung cancer cell lines (A549, NCI-H460, and NCI-H292) by suppressing glycolysis-related factors including GLUT1, PKM2, LDHA, and HK2 proteins and demonstrates efficacy in inhibiting tumor growth in an A549-injected mouse model [159]. β-elemene is a natural compound that displays antimetastatic efficacy by blocking PKM2 transformation and nuclear translocation [160]. β-elemene can overcome gefitinib resistance by inducing fructose-1,6-bisphosphatase [161]. Licochalcone, a natural compound from Glycyrrhiza uralensis, is a potent HIF-1α inhibitor that can suppress the expression of GLUT1 and PDK1 by inhibiting HIF-1α in HCT116 cells [162]. Additionally, licochalcone-A can overcome mesenchymal-epithelial transition factor (c-Met) overexpression-mediated gefitinib resistance by promoting c-Met ubiquitination [163]. Tanshinone IIA is a natural product extracted from Salvia miltiorrhiza Bunge [164]. Tanshinone IIA inhibits the development and proliferation of oral squamous cell carcinoma cells by suppressing Akt-c-Myc signaling and HK2-mediated glycolysis by diminishing HK2 expression at the transcriptional level [165]. Tanshinone IIA is an EGFR inhibitor that suppresses the growth of NSCLC cells by targeting the EGFR-Akt-myeloid cell leukemia-1 axis. Sulforaphane can modulate HIF-1α stability in human colon cancer cells [166] and downregulates glycolytic enzymes, including HK2, PKM2, and PDH, in bladder cancer [167]. In EGFR-TKI-resistant NSCLC cells, SFN treatment reduces EGFR expression and inhibits tumor growth [168]. The anticancer effects of EGCG have been attributed to multiple molecular mechanisms. EGCG inhibits HK2 expression and induces apoptosis in human tongue carcinomas [169]. Furthermore, EGCG disrupts the binding of EGF to EGFR, leading to the inhibition of EGFR TK activity [170,171]. EGCG can also induce the internalization of EGFR into endosomes, rendering it inaccessible to EGF ligands [172]. In another study, EGCG was demonstrated to overcome gefitinib resistance by inhibiting autophagy and enhancing cell death by targeting the extracellular signal-regulated kinase pathway in NSCLC [173]. Shikonin has been detected in lithospermum erythrorhizon and identified as a glycolysis inhibitor that suppresses PKM2 [174]. Shikonin exhibits a synergistic anticancer effect when combined with gefitinib via putative molecular processes that are associated with PKM2/STAT3/cyclinD1 inhibition.
In summary, a range of natural compounds such as cucurbitacin D, cucurbitacin B, genistein, α-Hederin, β-elemene, licochalcone, tanshinone IIA, sulforaphane, EGCG, and shikonin demonstrate promising glycolysis-inhibiting effects. These compounds target various glycolytic components and represent potential therapeutic avenues for overcoming EGFR-TKI resistance and inhibiting cancer cell growth (Table 4).

8. Perspectives and Conclusions

Drug resistance is a major challenge in cancer treatment, particularly in targeted therapy [175]. Cancer cells can evolve and adapt to the effects of targeted therapies, thereby reducing treatment effectiveness over time. Resistance to drugs can occur through mechanisms such as mutations in the targeted protein or activation of alternative signaling pathways [176]. In addition, the effectiveness of targeted therapy is restricted to a subset of patients with cancer with specific genetic mutations, limiting the utility of targeted therapy across a wide population of individuals with cancer. In this review, we hypothesized that glycolytic enzymes, including GLUT1, HK2, PKM2, PDK1, and LDHA, are promising targets for enhancing the efficacy of EGFR-targeted therapy and overcoming drug resistance in NSCLC. Targeting the glucose metabolism via the Warburg effect has potential advantages over traditional targeted therapies. One advantage of targeting the glucose metabolism in cancer cells is that it may prove to be more effective than traditional targeted therapies for several types of cancers. Although targeted therapy is often specific to certain genetic mutations or cancer types, most cancer cells exhibit a certain degree of the Warburg effect [177]. Another advantage is that targeting the glucose metabolism may result in fewer side effects than those produced by traditional chemotherapies or targeted therapies, because the Warburg effect is a unique characteristic of cancer cells; therefore, drugs targeting this metabolic pathway may be less toxic to healthy cells [79].
PDK inhibitors have demonstrated promising results in overcoming the resistance to EGFR-targeted therapies in NSCLC, suggesting a potential benefit of their use in combination therapies [109,110,111]. Notably, in all studies investigating combination therapies involving PDK inhibitors and EGFR-TKIs to overcome resistance in NSCLC, the PDK inhibitors utilized were of synthetic origin. In a previous study, we discovered that natural product-based PDK inhibitors such as huzhangoside A, Leelamine, and otobaphenol induced PDH activity-dependent cancer cell death [178]. Natural products, shaped by millions of years of evolution, encompass a wide array of chemical compounds exhibiting diverse structures and functions. They stand out as abundant reservoirs of bioactive molecules with potent therapeutic potential [179]. Furthermore, natural products often typically demonstrate superior biocompatibility and reduced toxicity compared to synthetic compounds. This heightened compatibility with biological systems is a result of their natural selection over time [15]. Additionally, natural products often have unique chemical structures that are difficult to replicate using synthetic chemistry, making them valuable sources of novel compounds for drug discovery and development [180]. In light of these compelling attributes, natural product-based PDK inhibitors represent a promising avenue for advancing therapeutic strategies for the treatment of EGFR-TKI-resistant NSCLC.
The upregulation of HIF-1α plays a pivotal role in the adaptation of cancer cells to the tumor microenvironment [181]. Although not a direct glycolytic enzyme, HIF-1α exerts a profound influence on the glucose metabolism by orchestrating the expression of key enzymes and transporters that are involved in glycolysis [181]. In this review, the potential of several natural product-based glycolysis inhibitors that demonstrated the ability to modulate HIF-1α levels was discussed as potential therapeutic strategies for EGFR-TKI-resistant NSCLC (Figure 4). Notably, compounds such as genistein, licochalcone, and sulforaphane exhibited inhibitory effects on HIF-1α, consequently downregulating the expression of crucial glycolytic components including GLUT1, HK2, PKM2, PDK1, and LDHA. Understanding the intricate interplay between HIF-1α and glycolytic pathways is crucial for developing targeted therapeutic approaches, and our findings underscore the promising role of natural product-based glycolysis inhibitors in this context.
Natural products play a pivotal role in cancer therapy, as they offer a vast array of compounds sourced from plants, marine organisms, and microorganisms [182]. These compounds have demonstrated substantial anticancer properties, including the ability to impede cancer cell growth, induce apoptosis, and inhibit angiogenesis [183]. The significance of natural products lies in their unique chemical structures, which often serve as inspiration for the development of novel drugs with improved efficacy and few side effects [183]. Furthermore, natural products exhibit synergistic effects with anticancer drugs and other natural chemicals [184,185].
Some natural product-based glycolysis inhibitors for EGFR-TKI-resistant NSCLC such as apigenin, quercetin, capsaicin, catechin, curcumin, EGCG, leonurine, and sulforaphane have obtained FDA approval for their safety. However, these drugs have been reported to have adverse effects. Capsaicin causes tissue irritation and burning [186]. EGCG, the primary polyphenol in green tea, may manifest side effects such as anxiolytic activity, potential hypoglycemic effects, a risk of hypochromic anemia due to interference with iron absorption, hepatotoxicity, and kidney issues at high doses [187]. High doses of apigenin, particularly from supplements, may cause stomach discomfort, muscle relaxation, and sleepiness [188]. High levels of sulforaphane, catechin, and curcumin can cause digestive difficulties [189,190,191]. Despite FDA approval, these natural glycolysis inhibitors require careful dosing and monitoring because of the potential side effects in therapeutic use. When exploring the pharmacological characteristics of various compounds, factors such as oral bioavailability and water solubility can significantly affect their clinical applicability. Cryptotanshinone and berberine have low oral bioavailability, which limits their clinical applicability [192,193]. Leelamine, α-hederin, and tanshinone IIA demonstrated very low oral bioavailability at 7.6%, 0.14%, and 3.5%, respectively [194,195,196]. Dicoumarol, ursolic acid, genistein, and shikonin face challenges owing to their poor solubility [197,198,199,200]. β-elemene also exhibits poor solubility in water; thus, researchers have synthesized various derivatives to address this issue and enhance its antitumor activities [201]. Licochalcone A, with an oral bioavailability of 3.3% in mice, exhibits poor absorption; however, when loaded onto liposome carriers, its water solubility and oral bioavailability significantly improve [202,203]. Further research is required to explore formulation strategies, (including nanoformulations and lipid-based carriers) to enhance the bioavailability of drugs with poor absorption characteristics and optimize their clinical applicability. More comprehensive research is needed on the pharmacokinetics and pharmacodynamics of hemistepsin A, huzhangoside A, ilimaquinone, and otobaphenol to explore their therapeutic properties. Berberine (NCT03486496), catechin (NCT00573885 and NCT00611650), curcumin (NCT02321293 and NCT01048983), and genistein (NCT01628471 and NCT00769990) are currently undergoing clinical trials for the treatment of NSCLC. Although natural product-based glycolysis inhibitors exhibit diverse drug profile variations in bioavailability, formulation-dependent improvements, and potential side effects, the therapeutic potential of these substances is promising.
In this review, we have presented the potential of combining glycolytic inhibitors with EGFR-TKI to overcome EGFR-TKI resistance. Despite the absence of clinical studies supporting this hypothesis, ongoing research in this field is promising. Further investigation is essential to evaluate the safety, tolerability, and efficacy of the co-administration of glycolytic inhibitors with EGFR-TKIs in a clinical setting. In addition, the effectiveness of glycolytic inhibitors in conjunction with other targeted therapies such as immunotherapy, conventional chemotherapy, and radiotherapy needs to be explored. Furthermore, biomarkers to identify patients who are most likely to benefit from glycolytic inhibitor-based therapies are needed. The identification of biomarkers can create avenues for personalized cancer treatment, thereby enhancing outcomes and mitigating toxicity in patients with cancer. Although the current literature on the utilization of glycolytic inhibitors and EGFR-TKIs is limited, preclinical studies have demonstrated the potential utility of this approach in cancer therapy. As research in this domain progresses, a meticulous evaluation of the safety and efficacy of combination therapy approaches in clinical settings is imperative to enhance the outcomes for patients with cancer.
In conclusion, this review demonstrates that glycolytic inhibitors represent a promising therapeutic strategy for cancer treatment, and that natural products are rich in compounds with glycolytic inhibitory activity. Overall, combining glycolytic inhibitors with EGFR-TKIs can enhance their effectiveness, while minimizing side effects and the risk of resistance.

Author Contributions

H.-S.C., D.R. and K.-T.H. conceived and supervised the project and revised the manuscript. W.P. and J.H.H. wrote the manuscript. S.W. and S.-J.B. collected the data. S.-Y.C. and E.-S.Y. drew the figures and tables. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) and by National Research Foundation of Korea (NRF) grants funded by the Korean government (MIST) (grant No. NRF-2022R1A2C2005130, NRF-2021R1A4A1025662, and RS-2023-00237776).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge the use of BioRender (https://biorender.com, accessed on 28 December 2023) for creating the figures in this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AbbreviationDefinition
2-DG2-deoxy-d-glucose
ALKAnaplastic lymphoma kinase
ATPAdenosine triphosphate
BRAFV-raf murine sarcoma viral oncogene homolog B1
c-MycCellular-myelocytomatosis oncogene
DCADichloroacetate
DNADeoxyribonucleic acid
EGCGEpigallocatechin gallate
EGFEpidermal growth factor
EGFREpidermal growth factor receptor
FBP1Fructose-1,6-bisphosphatase
FDAFood and Drug Administration
GLUT1Glucose transporter 1
HER2Human epidermal growth factor receptor 2
HIF-1αHypoxia-inducible factor-1 alpha
HK2Hexokinase 2
KRASKirsten rat sarcoma virus
LDHLactate dehydrogenase
LDHALactate dehydrogenase A
METProto-oncogene, receptor tyrosine kinase
NADNicotinamide adenine dinucleotide
NCCNNational Comprehensive Cancer Network
NDNot determined
NSCLCNon-small cell lung cancer
NTRKNeurotrophic tyrosine receptor kinase
OXPHOSOxidative phosphorylation
OROverall survival
PDCPyruvate dehydrogenase complex
PDKPyruvate dehydrogenase kinase
PD-L1Programmed cell death ligand 1
PEPPhosphoenolpyruvate
PI3KPhosphatidylinositol 3-kinase
PKM2Pyruvate kinase M2
RETRET proto-oncogene
RNARibonucleic acid
ROSReactive oxygen species
ROS1ROS proto-oncogene 1, receptor tyrosine kinase
SCLCSmall cell lung cancer
SFNSulforaphane
TKTyrosine kinase
TKITyrosine kinase inhibitor

References

  1. Wang, M.; Herbst, R.S.; Boshoff, C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat. Med. 2021, 27, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
  2. Molina, J.R.; Yang, P.; Cassivi, S.D.; Schild, S.E.; Adjei, A.A. Non-Small Cell Lung Cancer: Epidemiology, Risk Factors, Treatment, and Survivorship; Mayo Clinic Proceedings; Elsevier: Amsterdam, The Netherlands, 2008; pp. 584–594. [Google Scholar]
  3. Van Meerbeeck, J.P.; Fennell, D.A.; De Ruysscher, D.K. Small-cell lung cancer. Lancet 2011, 378, 1741–1755. [Google Scholar] [CrossRef] [PubMed]
  4. Halliday, P.R.; Blakely, C.M.; Bivona, T.G. Emerging targeted therapies for the treatment of non-small cell lung cancer. Curr. Oncol. Rep. 2019, 21, 21. [Google Scholar] [CrossRef] [PubMed]
  5. Li, K.; Yang, M.; Liang, N.; Li, S. Determining EGFR-TKI sensitivity of G719X and other uncommon EGFR mutations in non-small cell lung cancer: Perplexity and solution. Oncol. Rep. 2017, 37, 1347–1358. [Google Scholar] [CrossRef] [PubMed]
  6. Zhang, Y.-L.; Yuan, J.-Q.; Wang, K.-F.; Fu, X.-H.; Han, X.-R.; Threapleton, D.; Yang, Z.-Y.; Mao, C.; Tang, J.-L. The prevalence of EGFR mutation in patients with non-small cell lung cancer: A systematic review and meta-analysis. Oncotarget 2016, 7, 78985. [Google Scholar] [CrossRef] [PubMed]
  7. Inoue, A.; Kobayashi, K.; Usui, K.; Maemondo, M.; Okinaga, S.; Mikami, I.; Ando, M.; Yamazaki, K.; Saijo, Y.; Gemma, A. First-line gefitinib for patients with advanced non-small-cell lung cancer harboring epidermal growth factor receptor mutations without indication for chemotherapy. J. Clin. Oncol. 2009, 27, 1394–1400. [Google Scholar] [CrossRef] [PubMed]
  8. Ramalingam, S.S.; Vansteenkiste, J.; Planchard, D.; Cho, B.C.; Gray, J.E.; Ohe, Y.; Zhou, C.; Reungwetwattana, T.; Cheng, Y.; Chewaskulyong, B. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N. Engl. J. Med. 2020, 382, 41–50. [Google Scholar] [CrossRef] [PubMed]
  9. Leonetti, A.; Sharma, S.; Minari, R.; Perego, P.; Giovannetti, E.; Tiseo, M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer 2019, 121, 725–737. [Google Scholar] [CrossRef]
  10. Varghese, E.; Samuel, S.M.; Líšková, A.; Samec, M.; Kubatka, P.; Büsselberg, D. Targeting Glucose Metabolism to Overcome Resistance to Anticancer Chemotherapy in Breast Cancer. Cancers 2020, 12, 2252. [Google Scholar] [CrossRef]
  11. Jeong, J.Y.; Jeoung, N.H.; Park, K.-G.; Lee, I.-K. Transcriptional regulation of pyruvate dehydrogenase kinase. Diabetes Metab. J. 2012, 36, 328–335. [Google Scholar] [CrossRef]
  12. Lu, C.-W.; Lin, S.-C.; Chen, K.-F.; Lai, Y.-Y.; Tsai, S.-J. Induction of pyruvate dehydrogenase kinase-3 by hypoxia-inducible factor-1 promotes metabolic switch and drug resistance. J. Biol. Chem. 2008, 283, 28106–28114. [Google Scholar] [CrossRef] [PubMed]
  13. Han, J.H.; Kim, M.; Choi, H.J.; Jin, J.S.; Lee, S.O.; Bae, S.J.; Ryu, D.; Ha, K.T. The Oral Administration of Sanguisorba officinalis Extract Improves Physical Performance through LDHA Modulation. Molecules 2021, 26, 1579. [Google Scholar] [CrossRef] [PubMed]
  14. Han, J.H.; Kim, M.; Kim, H.J.; Jang, S.B.; Bae, S.J.; Lee, I.K.; Ryu, D.; Ha, K.T. Targeting Lactate Dehydrogenase A with Catechin Resensitizes SNU620/5FU Gastric Cancer Cells to 5-Fluorouracil. Int. J. Mol. Sci. 2021, 22, 5406. [Google Scholar] [CrossRef] [PubMed]
  15. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef] [PubMed]
  16. Zhao, Y.; Chard Dunmall, L.S.; Cheng, Z.; Wang, Y.; Si, L. Natural products targeting glycolysis in cancer. Front. Pharmacol. 2022, 13, 1036502. [Google Scholar] [CrossRef]
  17. Zhao, M.; Wei, F.; Sun, G.; Wen, Y.; Xiang, J.; Su, F.; Zhan, L.; Nian, Q.; Chen, Y.; Zeng, J. Natural compounds targeting glycolysis as promising therapeutics for gastric cancer: A review. Front. Pharmacol. 2022, 13, 1004383. [Google Scholar] [CrossRef] [PubMed]
  18. Jin, L.; Kim, E.-Y.; Chung, T.-W.; Han, C.W.; Park, S.Y.; Han, J.H.; Bae, S.-J.; Lee, J.R.; Kim, Y.W.; Jang, S.B. Hemistepsin A suppresses colorectal cancer growth through inhibiting pyruvate dehydrogenase kinase activity. Sci. Rep. 2020, 10, 21940. [Google Scholar] [CrossRef]
  19. Kwak, C.-H.; Lee, J.-H.; Kim, E.-Y.; Han, C.W.; Kim, K.-J.; Lee, H.; Cho, M.; Jang, S.B.; Kim, C.-H.; Chung, T.-W. Huzhangoside A suppresses tumor growth through inhibition of pyruvate dehydrogenase kinase activity. Cancers 2019, 11, 712. [Google Scholar] [CrossRef]
  20. Kwak, C.-H.; Jin, L.; Han, J.H.; Han, C.W.; Kim, E.; Cho, M.; Chung, T.-W.; Bae, S.-J.; Jang, S.B.; Ha, K.-T. Ilimaquinone induces the apoptotic cell death of cancer cells by reducing pyruvate dehydrogenase kinase 1 activity. Int. J. Mol. Sci. 2020, 21, 6021. [Google Scholar] [CrossRef]
  21. Yuan, M.; Huang, L.-L.; Chen, J.-H.; Wu, J.; Xu, Q. The emerging treatment landscape of targeted therapy in non-small-cell lung cancer. Signal Transduct. Target. Ther. 2019, 4, 61. [Google Scholar] [CrossRef]
  22. Araghi, M.; Mannani, R.; Heidarnejad maleki, A.; Hamidi, A.; Rostami, S.; Safa, S.H.; Faramarzi, F.; Khorasani, S.; Alimohammadi, M.; Tahmasebi, S. Recent advances in non-small cell lung cancer targeted therapy; an update review. Cancer Cell Int. 2023, 23, 162. [Google Scholar] [CrossRef] [PubMed]
  23. Chan, B.A.; Hughes, B.G. Targeted therapy for non-small cell lung cancer: Current standards and the promise of the future. Transl. Lung Cancer Res. 2015, 4, 36–54. [Google Scholar] [PubMed]
  24. Ettinger, D.S.; Wood, D.E.; Aisner, D.L.; Akerley, W.; Bauman, J.R.; Bharat, A.; Bruno, D.S.; Chang, J.Y.; Chirieac, L.R.; D’Amico, T.A. Non–small cell lung cancer, version 3.2022, NCCN clinical practice guidelines in oncology. J. Natl. Compr. Cancer Netw. 2022, 20, 497–530. [Google Scholar] [CrossRef] [PubMed]
  25. Dungo, R.T.; Keating, G.M. Afatinib: First global approval. Drugs 2013, 73, 1503–1515. [Google Scholar] [CrossRef] [PubMed]
  26. Shirley, M. Dacomitinib: First global approval. Drugs 2018, 78, 1947–1953. [Google Scholar] [CrossRef] [PubMed]
  27. Cohen, M.H.; Johnson, J.R.; Chattopadhyay, S.; Tang, S.; Justice, R.; Sridhara, R.; Pazdur, R. Approval summary: Erlotinib maintenance therapy of advanced/metastatic non-small cell lung cancer (NSCLC). Oncologist 2010, 15, 1344–1351. [Google Scholar] [CrossRef] [PubMed]
  28. Kazandjian, D.; Blumenthal, G.M.; Yuan, W.; He, K.; Keegan, P.; Pazdur, R. FDA approval of gefitinib for the treatment of patients with metastatic EGFR mutation–positive non–small cell lung cancer. Clin. Cancer Res. 2016, 22, 1307–1312. [Google Scholar] [CrossRef]
  29. Ramalingam, S.S.; Yang, J.; Lee, C.K.; Kurata, T.; Kim, D.-W.; John, T.; Nogami, N.; Ohe, Y.; Mann, H.; Rukazenkov, Y. Osimertinib as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer. J. Clin. Oncol. 2018, 36, 841–849. [Google Scholar] [CrossRef]
  30. Park, K.; Haura, E.B.; Leighl, N.B.; Mitchell, P.; Shu, C.A.; Girard, N.; Viteri, S.; Han, J.Y.; Kim, S.W.; Lee, C.K.; et al. Amivantamab in EGFR Exon 20 Insertion-Mutated Non-Small-Cell Lung Cancer Progressing on Platinum Chemotherapy: Initial Results From the CHRYSALIS Phase I Study. J. Clin. Oncol. 2021, 39, 3391–3402. [Google Scholar] [CrossRef]
  31. Herden, M.; Waller, C.F. Alectinib. Recent Results Cancer Res. 2018, 211, 247–256. [Google Scholar]
  32. Markham, A. Brigatinib: First global approval. Drugs 2017, 77, 1131–1135. [Google Scholar] [CrossRef] [PubMed]
  33. Vansteenkiste, J.F.; Van De Kerkhove, C.; Wauters, E.; Van Mol, P. Capmatinib for the treatment of non-small cell lung cancer. Expert Rev. Anticancer Ther. 2019, 19, 659–671. [Google Scholar] [CrossRef] [PubMed]
  34. Chuang, J.C.; Neal, J.W. Crizotinib as first line therapy for advanced ALK-positive non-small cell lung cancers. Transl. Lung Cancer Res. 2015, 4, 639. [Google Scholar] [PubMed]
  35. Shaw, A.T.; Yasothan, U.; Kirkpatrick, P. Crizotinib. Nat. Rev. Drug Discov. 2011, 10, 897–898. [Google Scholar] [CrossRef] [PubMed]
  36. Sartore-Bianchi, A.; Pizzutilo, E.G.; Marrapese, G.; Tosi, F.; Cerea, G.; Siena, S. Entrectinib for the treatment of metastatic NSCLC: Safety and efficacy. Expert Rev. Anticancer Ther. 2020, 20, 333–341. [Google Scholar] [CrossRef] [PubMed]
  37. Scott, L.J. Larotrectinib: First global approval. Drugs 2019, 79, 201–206. [Google Scholar] [CrossRef] [PubMed]
  38. Shaw, A.T.; Bauer, T.M.; de Marinis, F.; Felip, E.; Goto, Y.; Liu, G.; Mazieres, J.; Kim, D.-W.; Mok, T.; Polli, A. First-line lorlatinib or crizotinib in advanced ALK-positive lung cancer. N. Engl. J. Med. 2020, 383, 2018–2029. [Google Scholar] [CrossRef]
  39. Markham, A. Pralsetinib: First approval. Drugs 2020, 80, 1865–1870. [Google Scholar] [CrossRef]
  40. Markham, A. Selpercatinib: First approval. Drugs 2020, 80, 1119–1124. [Google Scholar] [CrossRef]
  41. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J. Tepotinib in non–small-cell lung cancer with MET exon 14 skipping mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef]
  42. Nakajima, E.C.; Drezner, N.; Li, X.; Mishra-Kalyani, P.S.; Liu, Y.; Zhao, H.; Bi, Y.; Liu, J.; Rahman, A.; Wearne, E.; et al. FDA Approval Summary: Sotorasib for KRAS G12C-Mutated Metastatic NSCLC. Clin. Cancer Res. 2022, 28, 1482–1486. [Google Scholar] [CrossRef]
  43. Odogwu, L.; Mathieu, L.; Blumenthal, G.; Larkins, E.; Goldberg, K.B.; Griffin, N.; Bijwaard, K.; Lee, E.Y.; Philip, R.; Jiang, X. FDA approval summary: Dabrafenib and trametinib for the treatment of metastatic non-small cell lung cancers harboring BRAF V600E mutations. Oncologist 2018, 23, 740–745. [Google Scholar] [CrossRef] [PubMed]
  44. Akinboro, O.; Larkins, E.; Pai-Scherf, L.H.; Mathieu, L.N.; Ren, Y.; Cheng, J.; Fiero, M.H.; Fu, W.; Bi, Y.; Kalavar, S. FDA Approval summary: Pembrolizumab, atezolizumab, and cemiplimab-rwlc as single agents for first-line treatment of Advanced/Metastatic PD-L1–high NSCLC. Clin. Cancer Res. 2022, 28, 2221–2228. [Google Scholar] [CrossRef] [PubMed]
  45. Cohen, M.H.; Gootenberg, J.; Keegan, P.; Pazdur, R. FDA drug approval summary: Bevacizumab (Avastin®) plus carboplatin and paclitaxel as first-line treatment of advanced/metastatic recurrent nonsquamous non-small cell lung cancer. Oncologist 2007, 12, 713–718. [Google Scholar] [CrossRef] [PubMed]
  46. Vellanki, P.J.; Mulkey, F.; Jaigirdar, A.A.; Rodriguez, L.; Wang, Y.; Xu, Y.; Zhao, H.; Liu, J.; Howe, G.; Wang, J. FDA Approval Summary: Nivolumab with Ipilimumab and Chemotherapy for Metastatic Non–small Cell Lung Cancer, A Collaborative Project Orbis ReviewFDA Approval: Nivolumab with Ipilimumab and Chemotherapy. Clin. Cancer Res. 2021, 27, 3522–3527. [Google Scholar] [CrossRef]
  47. Kazandjian, D.; Suzman, D.L.; Blumenthal, G.; Mushti, S.; He, K.; Libeg, M.; Keegan, P.; Pazdur, R. FDA approval summary: Nivolumab for the treatment of metastatic non-small cell lung cancer with progression on or after platinum-based chemotherapy. Oncologist 2016, 21, 634–642. [Google Scholar] [CrossRef]
  48. Reck, M. Pembrolizumab as first-line therapy for metastatic non-small-cell lung cancer. Immunotherapy 2018, 10, 93–105. [Google Scholar] [CrossRef]
  49. Martinelli, E.; De Palma, R.; Orditura, M.; De Vita, F.; Ciardiello, F. Anti-epidermal growth factor receptor monoclonal antibodies in cancer therapy. Clin. Exp. Immunol. 2009, 158, 1–9. [Google Scholar] [CrossRef]
  50. Garnock-Jones, K.P. Necitumumab: First Global Approval. Drugs 2016, 76, 283–289. [Google Scholar] [CrossRef]
  51. Herbst, R.S. Review of epidermal growth factor receptor biology. Int. J. Radiat. Oncol. Biol. Phys. 2004, 59, S21–S26. [Google Scholar] [CrossRef]
  52. Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2000, 103, 211–225. [Google Scholar] [CrossRef] [PubMed]
  53. Yarden, Y. The EGFR family and its ligands in human cancer: Signalling mechanisms and therapeutic opportunities. Eur. J. Cancer 2001, 37, 3–8. [Google Scholar] [CrossRef] [PubMed]
  54. Ciardiello, F.; Tortora, G. EGFR antagonists in cancer treatment. N. Engl. J. Med. 2008, 358, 1160–1174. [Google Scholar] [CrossRef] [PubMed]
  55. Shah, R.; Lester, J.F. Tyrosine Kinase Inhibitors for the Treatment of EGFR Mutation-Positive Non-Small-Cell Lung Cancer: A Clash of the Generations. Clin. Lung Cancer 2020, 21, e216–e228. [Google Scholar] [CrossRef]
  56. Wieduwilt, M.; Moasser, M. The epidermal growth factor receptor family: Biology driving targeted therapeutics. Cell. Mol. Life Sci. 2008, 65, 1566–1584. [Google Scholar] [CrossRef] [PubMed]
  57. Burgess, A.W.; Cho, H.-S.; Eigenbrot, C.; Ferguson, K.M.; Garrett, T.P.; Leahy, D.J.; Lemmon, M.A.; Sliwkowski, M.X.; Ward, C.W.; Yokoyama, S. An open-and-shut case? Recent insights into the activation of EGF/ErbB receptors. Mol. Cell 2003, 12, 541–552. [Google Scholar] [CrossRef]
  58. Schlessinger, J. Ligand-induced, receptor-mediated dimerization and activation of EGF receptor. Cell 2002, 110, 669–672. [Google Scholar] [CrossRef]
  59. Zhang, X.; Gureasko, J.; Shen, K.; Cole, P.A.; Kuriyan, J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell 2006, 125, 1137–1149. [Google Scholar] [CrossRef]
  60. Siegelin, M.D.; Borczuk, A.C. Epidermal growth factor receptor mutations in lung adenocarcinoma. Lab. Investig. 2014, 94, 129–137. [Google Scholar] [CrossRef]
  61. Huang, S.-F.; Cheng, S.-D.; Chien, H.-T.; Liao, C.-T.; Chen, I.-H.; Wang, H.-M.; Chuang, W.-Y.; Wang, C.-Y.; Hsieh, L.-L. Relationship between epidermal growth factor receptor gene copy number and protein expression in oral cavity squamous cell carcinoma. Oral Oncol. 2012, 48, 67–72. [Google Scholar] [CrossRef]
  62. Sharma, S.V.; Bell, D.W.; Settleman, J.; Haber, D.A. Epidermal growth factor receptor mutations in lung cancer. Nat. Rev. Cancer 2007, 7, 169–181. [Google Scholar] [CrossRef] [PubMed]
  63. Sullivan, I.; Planchard, D. Next-Generation EGFR Tyrosine Kinase Inhibitors for Treating EGFR-Mutant Lung Cancer beyond First Line. Front. Med. 2016, 3, 76. [Google Scholar] [CrossRef]
  64. Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non–small-cell lung cancer to gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, Y.R.; Fu, Y.N.; Lin, C.H.; Yang, S.T.; Hu, S.F.; Chen, Y.T.; Tsai, S.F.; Huang, S.F. Distinctive activation patterns in constitutively active and gefitinib-sensitive EGFR mutants. Oncogene 2006, 25, 1205–1215. [Google Scholar] [CrossRef] [PubMed]
  66. Kancha, R.K.; von Bubnoff, N.; Peschel, C.; Duyster, J. Functional analysis of epidermal growth factor receptor (EGFR) mutations and potential implications for EGFR targeted therapy. Clin. Cancer Res. 2009, 15, 460–467. [Google Scholar] [CrossRef] [PubMed]
  67. Chang, J.W.; Chang, C.F.; Huang, C.Y.; Yang, C.T.; Kuo, C.S.; Fang, Y.F.; Hsu, P.C.; Wu, C.E. The survival after discontinuation of EGFR-TKIs due to intolerable adverse events in patients with EGFR-mutated non-small cell lung cancer. Thorac. Cancer 2023, 14, 348–356. [Google Scholar] [CrossRef] [PubMed]
  68. Yu, H.A.; Arcila, M.E.; Rekhtman, N.; Sima, C.S.; Zakowski, M.F.; Pao, W.; Kris, M.G.; Miller, V.A.; Ladanyi, M.; Riely, G.J. Analysis of Tumor Specimens at the Time of Acquired Resistance to EGFR-TKI Therapy in 155 Patients with EGFR-Mutant Lung CancersMechanisms of Acquired Resistance to EGFR-TKI Therapy. Clin. Cancer Res. 2013, 19, 2240–2247. [Google Scholar] [CrossRef]
  69. Campo, M.; Gerber, D.; Gainor, J.F.; Heist, R.S.; Temel, J.S.; Shaw, A.T.; Fidias, P.; Muzikansky, A.; Engelman, J.A.; Sequist, L.V. Acquired resistance to first-line afatinib and the challenges of prearranged progression biopsies. J. Thorac. Oncol. 2016, 11, 2022–2026. [Google Scholar] [CrossRef]
  70. Wu, Y.L.; Cheng, Y.; Zhou, X.; Lee, K.H.; Nakagawa, K.; Niho, S.; Tsuji, F.; Linke, R.; Rosell, R.; Corral, J.; et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): A randomised, open-label, phase 3 trial. Lancet Oncoll 2017, 18, 1454–1466. [Google Scholar] [CrossRef]
  71. Kobayashi, Y.; Fujino, T.; Nishino, M.; Koga, T.; Chiba, M.; Sesumi, Y.; Ohara, S.; Shimoji, M.; Tomizawa, K.; Takemoto, T.; et al. EGFR T790M and C797S Mutations as Mechanisms of Acquired Resistance to Dacomitinib. J. Thorac. Oncol. 2018, 13, 727–731. [Google Scholar] [CrossRef]
  72. Asahina, H.; Tanaka, K.; Morita, S.; Maemondo, M.; Seike, M.; Okamoto, I.; Oizumi, S.; Kagamu, H.; Takahashi, K.; Kikuchi, T. A Phase II Study of Osimertinib Combined With Platinum Plus Pemetrexed in Patients With EGFR-Mutated Advanced Non–Small-cell Lung Cancer: The OPAL Study (NEJ032C/LOGIK1801). Clin. Lung Cancer 2021, 22, 147–151. [Google Scholar] [CrossRef]
  73. Shi, K.; Wang, G.; Pei, J.; Zhang, J.; Wang, J.; Ouyang, L.; Wang, Y.; Li, W. Emerging strategies to overcome resistance to third-generation EGFR inhibitors. J. Hematol. Oncol. 2022, 15, 94. [Google Scholar] [CrossRef] [PubMed]
  74. Khozin, S.; Blumenthal, G.M.; Jiang, X.; He, K.; Boyd, K.; Murgo, A.; Justice, R.; Keegan, P.; Pazdur, R. US Food and Drug Administration approval summary: Erlotinib for the first-line treatment of metastatic non-small cell lung cancer with epidermal growth factor receptor exon 19 deletions or exon 21 (L858R) substitution mutations. Oncologist 2014, 19, 774–779. [Google Scholar] [CrossRef] [PubMed]
  75. Markham, A. Mobocertinib: First Approval. Drugs 2021, 81, 2069–2074. [Google Scholar] [CrossRef] [PubMed]
  76. Greig, S.L. Osimertinib: First global approval. Drugs 2016, 76, 263–273. [Google Scholar] [CrossRef] [PubMed]
  77. Food and Drug Administration. FDA Approves Osimertinib for First-Line Treatment of Metastatic NSCLC with Most Common EGFR Mutations; Food and Drug Administration: White Oak, MD, USA, 2018.
  78. Koch, A.L.; Vellanki, P.J.; Drezner, N.; Li, X.; Mishra-Kalyani, P.S.; Shen, Y.L.; Xia, H.; Li, Y.; Liu, J.; Zirkelbach, J.F. FDA Approval Summary: Osimertinib for Adjuvant Treatment of Surgically Resected Non–Small Cell Lung Cancer, a Collaborative Project Orbis ReviewFDA Approval: Adjuvant Osimertinib for EGFR-Mutated NSCLC. Clin. Cancer Res. 2021, 27, 6638–6643. [Google Scholar] [CrossRef] [PubMed]
  79. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef]
  80. Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
  81. Pelicano, H.; Martin, D.; Xu, R.; Huang, P. Glycolysis inhibition for anticancer treatment. Oncogene 2006, 25, 4633. [Google Scholar] [CrossRef]
  82. Gatenby, R.A.; Gillies, R.J. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 2004, 4, 891–899. [Google Scholar] [CrossRef]
  83. Phaniendra, A.; Jestadi, D.B.; Periyasamy, L. Free radicals: Properties, sources, targets, and their implication in various diseases. Indian J. Clin. Biochem. 2015, 30, 11–26. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, Y.; Shi, J.; Wang, X.; Zhou, L.; Wang, Q.; Xie, Y.; Peng, C.; Kuang, L.; Yang, D.; Yang, J.; et al. An antioxidant feedforward cycle coordinated by linker histone variant H1.2 and NRF2 that drives nonsmall cell lung cancer progression. Proc. Natl. Acad. Sci. USA 2023, 120, e2306288120. [Google Scholar] [CrossRef] [PubMed]
  85. Cheng, H.; Shcherba, M.; Pendurti, G.; Liang, Y.; Piperdi, B.; Perez-Soler, R. Targeting the PI3K/AKT/mTOR pathway: Potential for lung cancer treatment. Lung Cancer Manag. 2014, 3, 67–75. [Google Scholar] [CrossRef] [PubMed]
  86. Burgess, J.T.; Rose, M.; Boucher, D.; Plowman, J.; Molloy, C.; Fisher, M.; O’Leary, C.; Richard, D.J.; O’Byrne, K.J.; Bolderson, E. The Therapeutic Potential of DNA Damage Repair Pathways and Genomic Stability in Lung Cancer. Front. Oncol. 2020, 10, 1256. [Google Scholar] [CrossRef] [PubMed]
  87. Shi, Y.; Fan, S.; Wu, M.; Zuo, Z.; Li, X.; Jiang, L.; Shen, Q.; Xu, P.; Zeng, L.; Zhou, Y.; et al. YTHDF1 links hypoxia adaptation and non-small cell lung cancer progression. Nat. Commun. 2019, 10, 4892. [Google Scholar] [CrossRef] [PubMed]
  88. Apicella, M.; Giannoni, E.; Fiore, S.; Ferrari, K.J.; Fernández-Pérez, D.; Isella, C.; Granchi, C.; Minutolo, F.; Sottile, A.; Comoglio, P.M. Increased lactate secretion by cancer cells sustains non-cell-autonomous adaptive resistance to MET and EGFR targeted therapies. Cell Metab. 2018, 28, 848–865.e6. [Google Scholar] [CrossRef]
  89. Keam, B.; Lee, S.J.; Kim, T.M.; Paeng, J.C.; Lee, S.H.; Kim, D.W.; Jeon, Y.K.; Chung, D.H.; Kang, K.W.; Chung, J.K.; et al. Total Lesion Glycolysis in Positron Emission Tomography Can Predict Gefitinib Outcomes in Non-Small-Cell Lung Cancer with Activating EGFR Mutation. J. Thorac. Oncol. 2015, 10, 1189–1194. [Google Scholar] [CrossRef]
  90. Suzuki, S.; Okada, M.; Takeda, H.; Kuramoto, K.; Sanomachi, T.; Togashi, K.; Seino, S.; Yamamoto, M.; Yoshioka, T.; Kitanaka, C. Involvement of GLUT1-mediated glucose transport and metabolism in gefitinib resistance of non-small-cell lung cancer cells. Oncotarget 2018, 9, 32667–32679. [Google Scholar] [CrossRef]
  91. Wang, J.; Ye, C.; Chen, C.; Xiong, H.; Xie, B.; Zhou, J.; Chen, Y.; Zheng, S.; Wang, L. Glucose transporter GLUT1 expression and clinical outcome in solid tumors: A systematic review and meta-analysis. Oncotarget 2017, 8, 16875–16886. [Google Scholar] [CrossRef]
  92. Kim, S.M.; Yun, M.R.; Hong, Y.K.; Solca, F.; Kim, J.H.; Kim, H.J.; Cho, B.C. Glycolysis inhibition sensitizes non-small cell lung cancer with T790M mutation to irreversible EGFR inhibitors via translational suppression of Mcl-1 by AMPK activation. Mol. Cancer Ther. 2013, 12, 2145–2156. [Google Scholar] [CrossRef]
  93. Ciscato, F.; Ferrone, L.; Masgras, I.; Laquatra, C.; Rasola, A. Hexokinase 2 in Cancer: A Prima Donna Playing Multiple Characters. Int. J. Mol. Sci. 2021, 22, 4716. [Google Scholar] [CrossRef] [PubMed]
  94. 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]
  95. Li, Q.; Zhang, D.; Chen, X.; He, L.; Li, T.; Xu, X.; Li, M. Nuclear PKM2 contributes to gefitinib resistance via upregulation of STAT3 activation in colorectal cancer. Sci. Rep. 2015, 5, 16082. [Google Scholar] [CrossRef] [PubMed]
  96. Stacpoole, P.W. Therapeutic Targeting of the Pyruvate Dehydrogenase Complex/Pyruvate Dehydrogenase Kinase (PDC/PDK) Axis in Cancer. J. Natl. Cancer Inst. 2017, 109. [Google Scholar] [CrossRef] [PubMed]
  97. Han, J.H.; Lee, E.J.; Park, W.; Ha, K.T.; Chung, H.S. Natural compounds as lactate dehydrogenase inhibitors: Potential therapeutics for lactate dehydrogenase inhibitors-related diseases. Front. Pharmacol. 2023, 14, 1275000. [Google Scholar] [CrossRef] [PubMed]
  98. Xu, J.Q.; Fu, Y.L.; Zhang, J.; Zhang, K.Y.; Ma, J.; Tang, J.Y.; Zhang, Z.W.; Zhou, Z.Y. Targeting glycolysis in non-small cell lung cancer: Promises and challenges. Front. Pharmacol. 2022, 13, 1037341. [Google Scholar] [CrossRef] [PubMed]
  99. Lin, C.; Chen, H.; Han, R.; Li, L.; Lu, C.; Hao, S.; Wang, Y.; He, Y. Hexokinases II-mediated glycolysis governs susceptibility to crizotinib in ALK-positive non-small cell lung cancer. Thorac. Cancer 2021, 12, 3184–3193. [Google Scholar] [CrossRef]
  100. Yang, Z.; Zhang, S.L.; Hu, X.; Tam, K.Y. Inhibition of pyruvate dehydrogenase kinase 1 enhances the anti-cancer effect of EGFR tyrosine kinase inhibitors in non-small cell lung cancer. Eur. J. Pharmacol. 2018, 838, 41–52. [Google Scholar] [CrossRef]
  101. Ma, R.; Li, X.; Gong, S.; Ge, X.; Zhu, T.; Ge, X.; Weng, L.; Tao, Q.; Guo, J. Dual Roles of Lactate in EGFR-TKI-Resistant Lung Cancer by Targeting GPR81 and MCT1. J. Oncol. 2022, 2022, 3425841. [Google Scholar] [CrossRef]
  102. Jeoung, N.H. Pyruvate dehydrogenase kinases: Therapeutic targets for diabetes and cancers. Diabetes Metab. J. 2015, 39, 188. [Google Scholar] [CrossRef]
  103. Schell, J.C.; Olson, K.A.; Jiang, L.; Hawkins, A.J.; Van Vranken, J.G.; Xie, J.; Egnatchik, R.A.; Earl, E.G.; DeBerardinis, R.J.; Rutter, J. A role for the mitochondrial pyruvate carrier as a repressor of the Warburg effect and colon cancer cell growth. Mol. Cell 2014, 56, 400–413. [Google Scholar] [CrossRef]
  104. Liu, T.; Yin, H. PDK1 promotes tumor cell proliferation and migration by enhancing the Warburg effect in non-small cell lung cancer. Oncol. Rep. 2017, 37, 193–200. [Google Scholar] [CrossRef] [PubMed]
  105. Papandreou, I.; Cairns, R.A.; Fontana, L.; Lim, A.L.; Denko, N.C. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006, 3, 187–197. [Google Scholar] [CrossRef] [PubMed]
  106. Viale, A.; Corti, D.; Draetta, G.F. Tumors and Mitochondrial Respiration: A Neglected ConnectionMitochondrial Role in Tumor Progression. Cancer Res. 2015, 75, 3687–3691. [Google Scholar] [CrossRef] [PubMed]
  107. Park, W.; Wei, S.; Kim, B.S.; Kim, B.; Bae, S.J.; Chae, Y.C.; Ryu, D.; Ha, K.T. Diversity and complexity of cell death: A historical review. Exp. Mol. Med. 2023, 55, 1573–1594. [Google Scholar] [CrossRef] [PubMed]
  108. Steussy, C.N.; Popov, K.M.; Bowker-Kinley, M.M.; Sloan, R.B.; Harris, R.A.; Hamilton, J.A. Structure of pyruvate dehydrogenase kinase: Novel folding pattern for a serine protein kinase. J. Biol. Chem. 2001, 276, 37443–37450. [Google Scholar] [CrossRef]
  109. Michelakis, E.; Webster, L.; Mackey, J. Dichloroacetate (DCA) as a potential metabolic-targeting therapy for cancer. Br. J. Cancer 2008, 99, 989–994. [Google Scholar] [CrossRef]
  110. Yang, Z.; Tam, K.Y. Anti-cancer synergy of dichloroacetate and EGFR tyrosine kinase inhibitors in NSCLC cell lines. Eur. J. Pharmacol. 2016, 789, 458–467. [Google Scholar] [CrossRef]
  111. Dyrstad, S.E.; Lotsberg, M.L.; Tan, T.Z.; Pettersen, I.K.; Hjellbrekke, S.; Tusubira, D.; Engelsen, A.S.; Daubon, T.; Mourier, A.; Thiery, J.P. Blocking aerobic glycolysis by targeting pyruvate dehydrogenase kinase in combination with EGFR TKI and ionizing radiation increases therapeutic effect in non-small cell lung cancer cells. Cancers 2021, 13, 941. [Google Scholar] [CrossRef]
  112. Xu, H.; He, Y.; Ma, J.; Zhao, Y.; Liu, Y.; Sun, L.; Su, J. Inhibition of pyruvate dehydrogenase kinase-1 by dicoumarol enhances the sensitivity of hepatocellular carcinoma cells to oxaliplatin via metabolic reprogramming. Int. J. Oncol. 2020, 57, 733–742. [Google Scholar] [CrossRef]
  113. Tambe, Y.; Terado, T.; Kim, C.J.; Mukaisho, K.I.; Yoshida, S.; Sugihara, H.; Tanaka, H.; Chida, J.; Kido, H.; Yamaji, K. Antitumor activity of potent pyruvate dehydrogenase kinase 4 inhibitors from plants in pancreatic cancer. Mol. Carcinog. 2019, 58, 1726–1737. [Google Scholar] [CrossRef] [PubMed]
  114. Zheng, S.-Y.; Li, Y.; Jiang, D.; Zhao, J.; Ge, J.-F. Anticancer effect and apoptosis induction by quercetin in the human lung cancer cell line A-549. Mol. Med. Rep. 2012, 5, 822–826. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, F.; Zhuang, M.; Zhong, C.; Peng, J.; Wang, X.; Li, J.; Chen, Z.; Huang, Y. Baicalein reverses hypoxia-induced 5-FU resistance in gastric cancer AGS cells through suppression of glycolysis and the PTEN/Akt/HIF-1alpha signaling pathway. Oncol. Rep. 2015, 33, 457–463. [Google Scholar] [CrossRef] [PubMed]
  116. Tao, H.; Ding, X.; Wu, J.; Liu, S.; Sun, W.; Nie, M.; Pan, X.; Zou, X. beta-Asarone Increases Chemosensitivity by Inhibiting Tumor Glycolysis in Gastric Cancer. Evid. Based Complement Altern. Med. 2020, 2020, 6981520. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, P.; Jin, J.M.; Liang, X.H.; Yu, M.Z.; Yang, C.; Huang, F.; Wu, H.; Zhang, B.B.; Fei, X.Y.; Wang, Z.T.; et al. Helichrysetin inhibits gastric cancer growth by targeting c-Myc/PDHK1 axis-mediated energy metabolism reprogramming. Acta Pharmacol. Sin. 2022, 43, 1581–1593. [Google Scholar] [CrossRef]
  118. Jiao, L.; Wang, S.; Zheng, Y.; Wang, N.; Yang, B.; Wang, D.; Yang, D.; Mei, W.; Zhao, Z.; Wang, Z. Betulinic acid suppresses breast cancer aerobic glycolysis via caveolin-1/NF-kappaB/c-Myc pathway. Biochem. Pharmacol. 2019, 161, 149–162. [Google Scholar] [CrossRef]
  119. Jin, J.; Qiu, S.; Wang, P.; Liang, X.; Huang, F.; Wu, H.; Zhang, B.; Zhang, W.; Tian, X.; Xu, R.; et al. Cardamonin inhibits breast cancer growth by repressing HIF-1alpha-dependent metabolic reprogramming. J. Exp. Clin. Cancer Res. 2019, 38, 377. [Google Scholar] [CrossRef]
  120. Aicher, T.D.; Damon, R.E.; Koletar, J.; Vinluan, C.C.; Brand, L.J.; Gao, J.; Shetty, S.S.; Kaplan, E.L.; Mann, W.R. Triterpene and diterpene inhibitors of pyruvate dehydrogenase kinase (PDK). Bioorganic Med. Chem. Lett. 1999, 9, 2223–2228. [Google Scholar] [CrossRef]
  121. Ha, K.-T. Composition for Preventing or Treating Cancer, and Containing Otobaphenol as Active Component. WO2018026210A1, 8 February 2018. [Google Scholar]
  122. Dahiya, R.; Mohammad, T.; Roy, S.; Anwar, S.; Gupta, P.; Haque, A.; Khan, P.; Kazim, S.N.; Islam, A.; Ahmad, F. Investigation of inhibitory potential of quercetin to the pyruvate dehydrogenase kinase 3: Towards implications in anticancer therapy. Int. J. Biol. Macromol. 2019, 136, 1076–1085. [Google Scholar] [CrossRef]
  123. Fantin, V.R.; St-Pierre, J.; Leder, P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 2006, 9, 425–434. [Google Scholar] [CrossRef]
  124. Livesey, A.; Garty, F.; Shipman, A.; Shipman, K. Lactate dehydrogenase in dermatology practice. Clin. Exp. Dermatol. 2020, 45, 539–543. [Google Scholar] [CrossRef] [PubMed]
  125. Hols, P.; Ramos, A.; Hugenholtz, J.; Delcour, J.; de Vos, W.M.; Santos, H.; Kleerebezem, M. Acetate utilization in Lactococcus lactis deficient in lactate dehydrogenase: A rescue pathway for maintaining redox balance. J. Bacteriol. 1999, 181, 5521–5526. [Google Scholar] [CrossRef] [PubMed]
  126. Holmes, R.S.; Goldberg, E. Computational analyses of mammalian lactate dehydrogenases: Human, mouse, opossum and platypus LDHs. Comput. Biol. Chem. 2009, 33, 379–385. [Google Scholar] [CrossRef] [PubMed]
  127. Urbańska, K.; Orzechowski, A. Unappreciated Role of LDHA and LDHB to Control Apoptosis and Autophagy in Tumor Cells. Int. J. Mol. Sci. 2019, 20, 2085. [Google Scholar] [CrossRef] [PubMed]
  128. Kane, D.A. Lactate oxidation at the mitochondria: A lactate-malate-aspartate shuttle at work. Front. Neurosci. 2014, 8, 366. [Google Scholar] [CrossRef] [PubMed]
  129. Read, J.; Winter, V.; Eszes, C.; Sessions, R.; Brady, R. Structural basis for altered activity of M- and H-isozyme forms of human lactate dehydrogenase. Proteins Struct. Funct. Bioinform. 2001, 43, 175–185. [Google Scholar] [CrossRef]
  130. Imagawa, T.; Yamamoto, E.; Sawada, M.; Okamoto, M.; Uehara, M. Expression of lactate dehydrogenase-A and-B messenger ribonucleic acids in chick glycogen body. Poult. Sci. 2006, 85, 1232–1238. [Google Scholar] [CrossRef]
  131. Feng, Y.; Xiong, Y.; Qiao, T.; Li, X.; Jia, L.; Han, Y. Lactate dehydrogenase A: A key player in carcinogenesis and potential target in cancer therapy. Cancer Med. 2018, 7, 6124–6136. [Google Scholar] [CrossRef]
  132. Klein, R.; Nagy, O.; Tóthová, C.; Chovanová, F. Clinical and Diagnostic Significance of Lactate Dehydrogenase and Its Isoenzymes in Animals. Vet. Med. Int. 2020, 2020, 5346483. [Google Scholar] [CrossRef]
  133. Yang, C.; Pan, R.Y.; Guan, F.; Yuan, Z. Lactate metabolism in neurodegenerative diseases. Neural Regen Res. 2024, 19, 69–74. [Google Scholar] [CrossRef]
  134. Zhu, W.; Ma, Y.; Guo, W.; Lu, J.; Li, X.; Wu, J.; Qin, P.; Zhu, C.; Zhang, Q. Serum Level of Lactate Dehydrogenase is Associated with Cardiovascular Disease Risk as Determined by the Framingham Risk Score and Arterial Stiffness in a Health-Examined Population in China. Int. J. Gen. Med. 2022, 15, 11–17. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, Z.; Lu, W.; Garcia-Prieto, C.; Huang, P. The Warburg effect and its cancer therapeutic implications. J. Bioenerg. Biomembr. 2007, 39, 267–274. [Google Scholar] [CrossRef]
  136. Mishra, D.; Banerjee, D. Lactate Dehydrogenases as Metabolic Links between Tumor and Stroma in the Tumor Microenvironment. Cancers 2019, 11, 750. [Google Scholar] [CrossRef] [PubMed]
  137. Inomata, M.; Hayashi, R.; Tanaka, H.; Shimokawa, K.; Tokui, K.; Taka, C.; Okazawa, S.; Kambara, K.; Ichikawa, T.; Yamada, T.; et al. Elevated levels of plasma lactate dehydrogenase is an unfavorable prognostic factor in patients with epidermal growth factor receptor mutation-positive non-small cell lung cancer, receiving treatment with gefitinib or erlotinib. Mol. Clin. Oncol. 2016, 4, 774–778. [Google Scholar] [CrossRef]
  138. Gong, T.; Liu, J.; Jiang, J.; Zhai, Y.F.; Wu, C.M.; Ma, C.; Wen, B.L.; Yan, X.Y.; Zhang, X.; Wang, D.M.; et al. The role of lactate deshydrogenase levels on non-small cell lung cancer prognosis: A meta-analysis. Cell. Mol. Biol. (Noisy-Le-Grand) 2019, 65, 89–93. [Google Scholar] [CrossRef] [PubMed]
  139. Korga, A.; Ostrowska, M.; Jozefczyk, A.; Iwan, M.; Wojcik, R.; Zgorka, G.; Herbet, M.; Vilarrubla, G.G.; Dudka, J. Apigenin and hesperidin augment the toxic effect of doxorubicin against HepG2 cells. BMC Pharmacol. Toxicol. 2019, 20, 22. [Google Scholar] [CrossRef] [PubMed]
  140. Zhu, N.; Li, J.; Li, Y.; Zhang, Y.; Du, Q.; Hao, P.; Li, J.; Cao, X.; Li, L. Berberine protects against simulated ischemia/reperfusion injury-induced H9C2 cardiomyocytes apoptosis in vitro and myocardial ischemia/reperfusion-induced apoptosis in vivo by regulating the mitophagy-mediated HIF-1α/BNIP3 pathway. Front. Pharmacol. 2020, 11, 367. [Google Scholar] [CrossRef] [PubMed]
  141. Hwang, Y.P.; Yun, H.J.; Choi, J.H.; Han, E.H.; Kim, H.G.; Song, G.Y.; Kwon, K.i.; Jeong, T.C.; Jeong, H.G. Suppression of EGF-induced tumor cell migration and matrix metalloproteinase-9 expression by capsaicin via the inhibition of EGFR-mediated FAK/Akt, PKC/Raf/ERK, p38 MAPK, and AP-1 signaling. Mol. Nutr. Food Res. 2011, 55, 594–605. [Google Scholar] [CrossRef]
  142. Chen, X.Q.; Hu, T.; Han, Y.; Huang, W.; Yuan, H.B.; Zhang, Y.T.; Du, Y.; Jiang, Y.W. Preventive Effects of Catechins on Cardiovascular Disease. Molecules 2016, 21, 1759. [Google Scholar] [CrossRef]
  143. Wang, K.; Fan, H.; Chen, Q.; Ma, G.; Zhu, M.; Zhang, X.; Zhang, Y.; Yu, J. Curcumin inhibits aerobic glycolysis and induces mitochondrial-mediated apoptosis through hexokinase II in human colorectal cancer cells in vitro. Anticancer Drugs 2015, 26, 15–24. [Google Scholar] [CrossRef]
  144. Chen, P.; Huang, H.P.; Wang, Y.; Jin, J.; Long, W.G.; Chen, K.; Zhao, X.H.; Chen, C.G.; Li, J. Curcumin overcome primary gefitinib resistance in non-small-cell lung cancer cells through inducing autophagy-related cell death. J. Exp. Clin. Cancer Res. 2019, 38, 254. [Google Scholar] [CrossRef] [PubMed]
  145. Li, S.; Liu, Z.; Zhu, F.; Fan, X.; Wu, X.; Zhao, H.; Jiang, L. Curcumin lowers erlotinib resistance in non-small cell lung carcinoma cells with mutated EGF receptor. Oncol. Res. 2013, 21, 137–144. [Google Scholar] [CrossRef] [PubMed]
  146. Lu, Q.Y.; Zhang, L.; Yee, J.K.; Go, V.W.; Lee, W.N. Metabolic Consequences of LDHA inhibition by Epigallocatechin Gallate and Oxamate in MIA PaCa-2 Pancreatic Cancer Cells. Metabolomics 2015, 11, 71–80. [Google Scholar] [CrossRef] [PubMed]
  147. Zhang, X.; Zhang, H.; Tighiouart, M.; Lee, J.E.; Shin, H.J.; Khuri, F.R.; Yang, C.S.; Chen, Z.; Shin, D.M. Synergistic inhibition of head and neck tumor growth by green tea (−)-epigallocatechin-3-gallate and EGFR tyrosine kinase inhibitor. Int. J. Cancer 2008, 123, 1005–1014. [Google Scholar] [CrossRef] [PubMed]
  148. Haque, A.; Rahman, M.A.; Chen, Z.G.; Saba, N.F.; Khuri, F.R.; Shin, D.M.; Ruhul Amin, A.R. Combination of erlotinib and EGCG induces apoptosis of head and neck cancers through posttranscriptional regulation of Bim and Bcl-2. Apoptosis 2015, 20, 986–995. [Google Scholar] [CrossRef]
  149. Grosse, F.; Nasheuer, H.P.; Scholtissek, S.; Schomburg, U. Lactate dehydrogenase and glyceraldehyde-phosphate dehydrogenase are single-stranded DNA-binding proteins that affect the DNA-polymerase-alpha-primase complex. Eur. J. Biochem. 1986, 160, 459–467. [Google Scholar] [CrossRef] [PubMed]
  150. Fiume, L.; Vettraino, M.; Carnicelli, D.; Arfilli, V.; Di Stefano, G.; Brigotti, M. Galloflavin prevents the binding of lactate dehydrogenase A to single stranded DNA and inhibits RNA synthesis in cultured cells. Biochem. Biophys. Res. Commun. 2013, 430, 466–469. [Google Scholar] [CrossRef]
  151. Liu, X.; Pan, L.; Chen, P.; Zhu, Y. Leonurine improves ischemia-induced myocardial injury through antioxidative activity. Phytomedicine 2010, 17, 753–759. [Google Scholar] [CrossRef]
  152. Maurya, A.K.; Vinayak, M. Quercetin regresses Dalton’s lymphoma growth via suppression of PI3K/AKT signaling leading to upregulation of p53 and decrease in energy metabolism. Nutr. Cancer 2015, 67, 354–363. [Google Scholar] [CrossRef]
  153. Mlala, S.; Oyedeji, A.O.; Gondwe, M.; Oyedeji, O.O. Ursolic acid and its derivatives as bioactive agents. Molecules 2019, 24, 2751. [Google Scholar] [CrossRef]
  154. Wang, S.; Chang, X.; Zhang, J.; Li, J.; Wang, N.; Yang, B.; Pan, B.; Zheng, Y.; Wang, X.; Ou, H. Ursolic acid inhibits breast cancer metastasis by suppressing glycolytic metabolism via activating sp1/caveolin-1 signaling. Front. Oncol. 2021, 11, 745584. [Google Scholar] [CrossRef] [PubMed]
  155. Sikander, M.; Malik, S.; Chauhan, N.; Khan, P.; Kumari, S.; Kashyap, V.K.; Khan, S.; Ganju, A.; Halaweish, F.T.; Yallapu, M.M. Cucurbitacin D reprograms glucose metabolic network in prostate cancer. Cancers 2019, 11, 364. [Google Scholar] [CrossRef] [PubMed]
  156. Yar Saglam, A.; Alp, E.; Elmazoglu, Z.; Menevse, S. Treatment with cucurbitacin B alone and in combination with gefitinib induces cell cycle inhibition and apoptosis via EGFR and JAK/STAT pathway in human colorectal cancer cell lines. Hum. Exp. Toxicol. 2016, 35, 526–543. [Google Scholar] [CrossRef] [PubMed]
  157. Li, S.; Li, J.; Dai, W.; Zhang, Q.; Feng, J.; Wu, L.; Liu, T.; Yu, Q.; Xu, S.; Wang, W. Genistein suppresses aerobic glycolysis and induces hepatocellular carcinoma cell death. Br. J. Cancer 2017, 117, 1518–1528. [Google Scholar] [CrossRef] [PubMed]
  158. Keyhanmanesh, R.; Saadat, S.; Mohammadi, M.; Shahbazfar, A.A.; Fallahi, M. The protective effect of α-hederin, the active constituent of Nigella sativa, on lung inflammation and blood cytokines in ovalbumin sensitized Guinea pigs. Phytother. Res. 2015, 29, 1761–1767. [Google Scholar] [CrossRef]
  159. Fang, C.; Liu, Y.; Chen, L.; Luo, Y.; Cui, Y.; Zhang, N.; Liu, P.; Zhou, M.; Xie, Y. α-Hederin inhibits the growth of lung cancer A549 cells in vitro and in vivo by decreasing SIRT6 dependent glycolysis. Pharm. Biol. 2021, 59, 11–20. [Google Scholar] [CrossRef]
  160. Pan, Y.; Wang, W.; Huang, S.; Ni, W.; Wei, Z.; Cao, Y.; Yu, S.; Jia, Q.; Wu, Y.; Chai, C. Beta-elemene inhibits breast cancer metastasis through blocking pyruvate kinase M2 dimerization and nuclear translocation. J. Cell. Mol. Med. 2019, 23, 6846–6858. [Google Scholar] [CrossRef]
  161. Li, J.; Dai, P.; Sun, J.; Yu, W.; Han, W.; Li, K. FBP1 induced by β-elemene enhances the sensitivity of gefitinib in lung cancer. Thorac. Cancer 2023, 14, 371–380. [Google Scholar] [CrossRef]
  162. Park, M.K.; Ji, J.; Haam, K.; Han, T.-H.; Lim, S.; Kang, M.-J.; Lim, S.S.; Ban, H.S. Licochalcone A inhibits hypoxia-inducible factor-1α accumulation by suppressing mitochondrial respiration in hypoxic cancer cells. Biomed. Pharmacother. 2021, 133, 111082. [Google Scholar] [CrossRef]
  163. Han, S.; Li, X.; Gan, Y.; Li, W. Licochalcone A promotes the ubiquitination of c-met to abrogate gefitinib resistance. BioMed Res. Int. 2022, 2022, 5687832. [Google Scholar] [CrossRef]
  164. Yoon, Y.; Kim, Y.-O.; Jeon, W.-K.; Park, H.-J.; Sung, H.J. Tanshinone IIA isolated from Salvia miltiorrhiza BUNGE induced apoptosis in HL60 human premyelocytic leukemia cell line. J. Ethnopharmacol. 1999, 68, 121–127. [Google Scholar] [CrossRef] [PubMed]
  165. Li, M.; Gao, F.; Zhao, Q.; Zuo, H.; Liu, W.; Li, W. Tanshinone IIA inhibits oral squamous cell carcinoma via reducing Akt-c-Myc signaling-mediated aerobic glycolysis. Cell Death Dis. 2020, 11, 381. [Google Scholar] [CrossRef] [PubMed]
  166. Kim, D.H.; Sung, B.; Kang, Y.J.; Hwang, S.Y.; Kim, M.J.; Yoon, J.H.; Im, E.; Kim, N.D. Sulforaphane inhibits hypoxia-induced HIF-1α and VEGF expression and migration of human colon cancer cells. Int. J. Oncol. 2015, 47, 2226–2232. [Google Scholar] [CrossRef] [PubMed]
  167. Huang, L.; He, C.; Zheng, S.; Wu, C.; Ren, M.; Shan, Y. AKT1/HK2 Axis-mediated Glucose Metabolism: A Novel Therapeutic Target of Sulforaphane in Bladder Cancer. Mol. Nutr. Food Res. 2022, 66, e2100738. [Google Scholar] [CrossRef] [PubMed]
  168. Chen, C.-Y.; Yu, Z.-Y.; Chuang, Y.-S.; Huang, R.-M.; Wang, T.-C.V. Sulforaphane attenuates EGFR signaling in NSCLC cells. J. Biomed. Sci. 2015, 22, 38. [Google Scholar] [CrossRef] [PubMed]
  169. Gao, F.; Li, M.; Liu, W.-B.; Zhou, Z.-S.; Zhang, R.; Li, J.-L.; Zhou, K.-C. Epigallocatechin gallate inhibits human tongue carcinoma cells via HK2-mediated glycolysis. Oncol. Rep. 2015, 33, 1533–1539. [Google Scholar] [CrossRef] [PubMed]
  170. Yang, C.S.; Wang, X.; Lu, G.; Picinich, S.C. Cancer prevention by tea: Animal studies, molecular mechanisms and human relevance. Nat. Rev. Cancer 2009, 9, 429–439. [Google Scholar] [CrossRef]
  171. Liang, Y.C.; Lin-shiau, S.Y.; Chen, C.F.; Lin, J.K. Suppression of extracellular signals and cell proliferation through EGF receptor binding by (−)-epigallocatechin gallate in human A431 epidermoid carcinoma cells. J. Cell Biochem. 1997, 67, 55–65. [Google Scholar] [CrossRef]
  172. Adachi, S.; Nagao, T.; To, S.; Joe, A.K.; Shimizu, M.; Matsushima-Nishiwaki, R.; Kozawa, O.; Moriwaki, H.; Maxfield, F.R.; Weinstein, I.B. (−)-Epigallocatechin gallate causes internalization of the epidermal growth factor receptor in human colon cancer cells. Carcinogenesis 2008, 29, 1986–1993. [Google Scholar] [CrossRef]
  173. Meng, J.; Chang, C.; Chen, Y.; Bi, F.; Ji, C.; Liu, W. EGCG overcomes gefitinib resistance by inhibiting autophagy and augmenting cell death through targeting ERK phosphorylation in NSCLC. OncoTargets Ther. 2019, 12, 6033. [Google Scholar] [CrossRef]
  174. Chen, J.; Xie, J.; Jiang, Z.; Wang, B.; Wang, Y.; Hu, X. Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene 2011, 30, 4297–4306. [Google Scholar] [CrossRef] [PubMed]
  175. Labrie, M.; Brugge, J.S.; Mills, G.B.; Zervantonakis, I.K. Therapy resistance: Opportunities created by adaptive responses to targeted therapies in cancer. Nat. Rev. Cancer 2022, 22, 323–339. [Google Scholar] [CrossRef] [PubMed]
  176. Ye, J.; Wu, J.; Liu, B. Therapeutic strategies of dual-target small molecules to overcome drug resistance in cancer therapy. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2023, 1878, 188866. [Google Scholar] [CrossRef] [PubMed]
  177. Hsu, P.P.; Sabatini, D.M. Cancer cell metabolism: Warburg and beyond. Cell 2008, 134, 703–707. [Google Scholar] [CrossRef] [PubMed]
  178. Jin, L.; Cho, M.; Kim, B.-S.; Han, J.H.; Park, S.; Lee, I.-K.; Ryu, D.; Kim, J.H.; Bae, S.-J.; Ha, K.-T. Drug evaluation based on phosphomimetic PDHA1 reveals the complexity of activity-related cell death in A549 non-small cell lung cancer cells. BMB Rep. 2021, 54, 563. [Google Scholar] [CrossRef] [PubMed]
  179. Cragg, G.M.; Newman, D.J. Natural products: A continuing source of novel drug leads. Biochim. Et Biophys. Acta (BBA)-Gen. Subj. 2013, 1830, 3670–3695. [Google Scholar] [CrossRef]
  180. Harvey, A.L. Natural products in drug discovery. Drug Discov. Today 2008, 13, 894–901. [Google Scholar] [CrossRef]
  181. Roy, S.; Kumaravel, S.; Sharma, A.; Duran, C.L.; Bayless, K.J.; Chakraborty, S. Hypoxic tumor microenvironment: Implications for cancer therapy. Exp. Biol. Med. 2020, 245, 1073–1086. [Google Scholar] [CrossRef]
  182. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  183. Demain, A.L.; Vaishnav, P. Natural products for cancer chemotherapy. Microb. Biotechnol. 2011, 4, 687–699. [Google Scholar] [CrossRef]
  184. Tan, G.; Gyllenhaal, C.; Soejarto, D.D. Biodiversity as a source of anticancer drugs. Curr. Drug Targets 2006, 7, 265–277. [Google Scholar] [CrossRef]
  185. Rahmani, A.H.; Babiker, A.Y.; Anwar, S. Hesperidin, a Bioflavonoid in Cancer Therapy: A Review for a Mechanism of Action through the Modulation of Cell Signaling Pathways. Molecules 2023, 28, 5152. [Google Scholar] [CrossRef] [PubMed]
  186. Yenigun, O.M.; Thanassi, M. Capsaicin: An Uncommon Exposure and Unusual Treatment. Clin. Pract. Cases Emerg. Med. 2019, 3, 219–221. [Google Scholar] [CrossRef]
  187. Mereles, D.; Hunstein, W. Epigallocatechin-3-gallate (EGCG) for clinical trials: More pitfalls than promises? Int. J. Mol. Sci. 2011, 12, 5592–5603. [Google Scholar] [CrossRef] [PubMed]
  188. Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kręgiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino, E.; et al. The Therapeutic Potential of Apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef]
  189. Lozanovski, V.J.; Polychronidis, G.; Gross, W.; Gharabaghi, N.; Mehrabi, A.; Hackert, T.; Schemmer, P.; Herr, I. Broccoli sprout supplementation in patients with advanced pancreatic cancer is difficult despite positive effects-results from the POUDER pilot study. Investig. New Drugs 2020, 38, 776–784. [Google Scholar] [CrossRef] [PubMed]
  190. Wu, Q.; Ouyang, Y.; Kong, Y.; Min, Y.; Xiao, J.; Li, S.; Zhou, M.; Feng, N.; Zhang, L. Catechin Inhibits the Release of Advanced Glycation End Products during Glycated Bovine Serum Albumin Digestion and Corresponding Mechanisms In Vitro. J. Agric. Food Chem. 2021, 69, 8807–8818. [Google Scholar] [CrossRef]
  191. Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its Effects on Human Health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
  192. Xing, L.; Tan, Z.R.; Cheng, J.L.; Huang, W.H.; Zhang, W.; Deng, W.; Yuan, C.S.; Zhou, H.H. Bioavailability and pharmacokinetic comparison of tanshinones between two formulations of Salvia miltiorrhiza in healthy volunteers. Sci. Rep. 2017, 7, 4709. [Google Scholar] [CrossRef]
  193. Solnier, J.; Zhang, Y.; Kuo, Y.C.; Du, M.; Roh, K.; Gahler, R.; Wood, S.; Chang, C. Characterization and Pharmacokinetic Assessment of a New Berberine Formulation with Enhanced Absorption In Vitro and in Human Volunteers. Pharmaceutics 2023, 15, 2567. [Google Scholar] [CrossRef]
  194. Song, M.; Lee, D.; Lee, T.; Lee, S. Determination of leelamine in mouse plasma by LC-MS/MS and its pharmacokinetics. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2013, 931, 170–173. [Google Scholar] [CrossRef] [PubMed]
  195. Li, H.-L.; Qin, Z.-M.; Cai, H.-D.; Tan, Y.-F.; Zhang, X.-P.; Luo, Y.-C.; Li, B.; Chen, F.; Zhang, J.-Q. Determination of α-hederin in rat plasma using liquid chromatography electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS) and its application to a pharmacokinetic study. Anal. Methods 2015, 7, 2155–2161. [Google Scholar] [CrossRef]
  196. Hao, H.; Wang, G.; Cui, N.; Li, J.; Xie, L.; Ding, Z. Pharmacokinetics, absorption and tissue distribution of tanshinone IIA solid dispersion. Planta Med. 2006, 72, 1311–1317. [Google Scholar] [CrossRef] [PubMed]
  197. Sun, C.; Zhao, W.; Wang, X.; Sun, Y.; Chen, X. A pharmacological review of dicoumarol: An old natural anticoagulant agent. Pharmacol. Res. 2020, 160, 105193. [Google Scholar] [CrossRef] [PubMed]
  198. Jinhua, W. Ursolic acid: Pharmacokinetics process in vitro and in vivo, a mini review. Arch. Pharm. 2019, 352, e1800222. [Google Scholar] [CrossRef] [PubMed]
  199. Yang, Z.; Kulkarni, K.; Zhu, W.; Hu, M. Bioavailability and pharmacokinetics of genistein: Mechanistic studies on its ADME. Anticancer. Agents Med. Chem. 2012, 12, 1264–1280. [Google Scholar] [CrossRef] [PubMed]
  200. Sun, Q.; Gong, T.; Liu, M.; Ren, S.; Yang, H.; Zeng, S.; Zhao, H.; Chen, L.; Ming, T.; Meng, X.; et al. Shikonin, a naphthalene ingredient: Therapeutic actions, pharmacokinetics, toxicology, clinical trials and pharmaceutical researches. Phytomedicine 2022, 94, 153805. [Google Scholar] [CrossRef] [PubMed]
  201. Zhai, B.; Zeng, Y.; Zeng, Z.; Zhang, N.; Li, C.; Zeng, Y.; You, Y.; Wang, S.; Chen, X.; Sui, X.; et al. Drug delivery systems for elemene, its main active ingredient β-elemene, and its derivatives in cancer therapy. Int. J. Nanomed. 2018, 13, 6279–6296. [Google Scholar] [CrossRef]
  202. Li, T.; Ye, W.; Huang, B.; Lu, X.; Chen, X.; Lin, Y.; Wen, C.; Wang, X. Determination and pharmacokinetic study of echinatin by UPLC-MS/MS in rat plasma. J. Pharm. Biomed. Anal. 2019, 168, 133–137. [Google Scholar] [CrossRef]
  203. Liu, J.; Zhu, Z.; Yang, Y.; Adu-Frimpong, M.; Chen, L.; Ji, H.; Toreniyazov, E.; Wang, Q.; Yu, J.; Xu, X. Preparation, characterization, pharmacokinetics, and antirenal injury activity studies of Licochalcone A-loaded liposomes. J. Food Biochem. 2022, 46, e14007. [Google Scholar] [CrossRef]
Figure 1. Targeted therapies for lung cancer. Illustration of the current targeted therapies used for non-small cell lung cancer (NSCLC), with specific drugs targeting (A) EGFR mutations; (B) ALK, HER2, MET, NTRK, RET, and ROS1 mutations; (C) KRAS and BRAF mutations; and (D) immunotherapy drugs for PD-L1 expression. This figure provides an outline of the targeted therapy choices recommended by the 2022 National Comprehensive Cancer Network guidelines for metastatic NSCLC, emphasizing the importance of personalized genetic testing in determining the optimal treatment strategy. EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; HER2, human epidermal growth factor receptor 2; MET, proto-oncogene, receptor tyrosine kinase; NTRK, neurotrophic tyrosine receptor kinase; RET, RET proto-oncogene; ROS1, ROS proto-oncogene 1, receptor tyrosine kinase; KRAS, Kirsten rat sarcoma virus; BRAF, v-raf murine sarcoma viral oncogene homolog B1; PD-L1, programmed cell death ligand 1.
Figure 1. Targeted therapies for lung cancer. Illustration of the current targeted therapies used for non-small cell lung cancer (NSCLC), with specific drugs targeting (A) EGFR mutations; (B) ALK, HER2, MET, NTRK, RET, and ROS1 mutations; (C) KRAS and BRAF mutations; and (D) immunotherapy drugs for PD-L1 expression. This figure provides an outline of the targeted therapy choices recommended by the 2022 National Comprehensive Cancer Network guidelines for metastatic NSCLC, emphasizing the importance of personalized genetic testing in determining the optimal treatment strategy. EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; HER2, human epidermal growth factor receptor 2; MET, proto-oncogene, receptor tyrosine kinase; NTRK, neurotrophic tyrosine receptor kinase; RET, RET proto-oncogene; ROS1, ROS proto-oncogene 1, receptor tyrosine kinase; KRAS, Kirsten rat sarcoma virus; BRAF, v-raf murine sarcoma viral oncogene homolog B1; PD-L1, programmed cell death ligand 1.
Ijms 25 00807 g001
Figure 2. Timeline of non-small cell lung cancer targeted therapy. Illustration of the timeline of genetic alterations in non-small cell lung cancer (NSCLC) subtypes, including EGFR, ALK, ROS1, KRAS, MET, PD-L1, and other mutations. This figure also indicates major concerns regarding the development of targeted therapy for NSCLC. EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; KRAS, Kirsten rat sarcoma virus; MET, proto-oncogene, receptor tyrosine kinase; ROS1, ROS proto-oncogene 1, receptor tyrosine kinase; PD-L1, programmed cell death ligand 1.
Figure 2. Timeline of non-small cell lung cancer targeted therapy. Illustration of the timeline of genetic alterations in non-small cell lung cancer (NSCLC) subtypes, including EGFR, ALK, ROS1, KRAS, MET, PD-L1, and other mutations. This figure also indicates major concerns regarding the development of targeted therapy for NSCLC. EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; KRAS, Kirsten rat sarcoma virus; MET, proto-oncogene, receptor tyrosine kinase; ROS1, ROS proto-oncogene 1, receptor tyrosine kinase; PD-L1, programmed cell death ligand 1.
Ijms 25 00807 g002
Figure 3. Schematic diagrams of EGFR, HER2, HER3, and HER4. The ErbB protein family includes the EGFR (HER1 and ErbB1), HER2 (Neu and ErbB2), HER3 (ErbB3), and HER4 (ErbB4) proteins. Structurally, EGFR comprises an extracellular domain containing a ligand-binding region, a transmembrane domain, a tyrosine kinase (TK) domain, and a C-terminal phosphorylation domain. Additionally, the binding of growth factors to these receptors is displayed: seven to EGFR, none to HER2, two to HER3, and seven to HER4. Compared with other ErbB protein family members (EGFR, HER2, and HER4), HER3 has little to no TK activity [56,57,58,59]. EGFR, epidermal growth factor receptor; HER, human epidermal growth factor receptor. ‘?‘ means that ‘None to HER2’ indicates there are presently no known HER2 ligands.
Figure 3. Schematic diagrams of EGFR, HER2, HER3, and HER4. The ErbB protein family includes the EGFR (HER1 and ErbB1), HER2 (Neu and ErbB2), HER3 (ErbB3), and HER4 (ErbB4) proteins. Structurally, EGFR comprises an extracellular domain containing a ligand-binding region, a transmembrane domain, a tyrosine kinase (TK) domain, and a C-terminal phosphorylation domain. Additionally, the binding of growth factors to these receptors is displayed: seven to EGFR, none to HER2, two to HER3, and seven to HER4. Compared with other ErbB protein family members (EGFR, HER2, and HER4), HER3 has little to no TK activity [56,57,58,59]. EGFR, epidermal growth factor receptor; HER, human epidermal growth factor receptor. ‘?‘ means that ‘None to HER2’ indicates there are presently no known HER2 ligands.
Ijms 25 00807 g003
Figure 4. Treatment strategy for EGFR-mutated non-small cell lung cancer (NSCLC). Illustration depicting the treatment strategy for NSCLC with EGFR mutations. The figure focuses on the glycolysis pathway, a key metabolic process in cancer cells. The glycolysis pathway is highlighted, with key enzymes, including HK2, PKM2, LDHA, and PDK1, marked in pink to emphasize their significance in the metabolic reprogramming of cancer cells. In the blue box, natural product-based glycolysis inhibitors are indicated, showcasing their potential role in targeting glycolytic pathways. Additionally, the figure underscores the regulatory influence of HIF-1α, depicted as a key factor (red), which can upregulate glycolytic enzymes, further emphasizing the intricate interplay within the glycolysis pathway. EGFR, epidermal growth factor receptor; HK2, hexokinase 2; PKM2, pyruvate kinase M2; LDHA, lactate dehydrogenase A; PDK, pyruvate dehydrogenase kinase; HIF-1α, hypoxia-inducible factor 1-alpha. The upward arrow symbol (↑) indicates upregulation.
Figure 4. Treatment strategy for EGFR-mutated non-small cell lung cancer (NSCLC). Illustration depicting the treatment strategy for NSCLC with EGFR mutations. The figure focuses on the glycolysis pathway, a key metabolic process in cancer cells. The glycolysis pathway is highlighted, with key enzymes, including HK2, PKM2, LDHA, and PDK1, marked in pink to emphasize their significance in the metabolic reprogramming of cancer cells. In the blue box, natural product-based glycolysis inhibitors are indicated, showcasing their potential role in targeting glycolytic pathways. Additionally, the figure underscores the regulatory influence of HIF-1α, depicted as a key factor (red), which can upregulate glycolytic enzymes, further emphasizing the intricate interplay within the glycolysis pathway. EGFR, epidermal growth factor receptor; HK2, hexokinase 2; PKM2, pyruvate kinase M2; LDHA, lactate dehydrogenase A; PDK, pyruvate dehydrogenase kinase; HIF-1α, hypoxia-inducible factor 1-alpha. The upward arrow symbol (↑) indicates upregulation.
Ijms 25 00807 g004
Table 1. Several approved EGFR-TKIs.
Table 1. Several approved EGFR-TKIs.
DrugStructureDrug TypeFDA Approval
Erlotinib
(TarcevaTM)
Ijms 25 00807 i0011st-generation
EGFR-TKI
1st-line, NSCLC with EGFR19D/EGFRL858R [74]
Gefitinib
(IressaTM)
Ijms 25 00807 i0021st-generation
EGFR-TKI
1st-line, NSCLC with EGFR19D/EGFRL858R [28]
Afatinib
(GilotrifTM)
Ijms 25 00807 i0032nd-generation
EGFR-TKI
1st-line, NSCLC with EGFR19D/EGFRL858R [25]
Dacomitinib
(VizimproTM)
Ijms 25 00807 i0042nd-generation
EGFR-TKI
1st-line, NSCLC with EGFR19D/EGFRL858R [26]
Mobocertinib
(ExkivityTM)
Ijms 25 00807 i0053rd-generation
EGFR-TKI
NSCLC with EGFR exon20 insertion [75]
Osimertinib
(TagrissoTM)
Ijms 25 00807 i0063rd-generation
EGFR-TKI
2nd-line, NSCLC with EGFRT790M [76]
1st-line, NSCLC with EGFR19D/EGFRL858R [77]
adjuvant therapy for NSCLC [78]
Table 2. Small-molecule PDK inhibitors derived from natural products.
Table 2. Small-molecule PDK inhibitors derived from natural products.
PDK InhibitorStructurePropertyOriginClinical Trials for NSCLCReference
CryptotanshinoneIjms 25 00807 i007IC50: PDK2 (11 µM), PDK4 (>30 µM)Salvia miltiorrhizaND[113]
DicoumarolIjms 25 00807 i008IC50: PDK1 (19.42 μM)Melilotus officinalisND[112]
Hemistepsin AIjms 25 00807 i009NDHemistepta lyrateND[18]
Huzhangoside AIjms 25 00807 i010NDAnemone rivularisND[19]
IlimaquinoneIjms 25 00807 i011NDSmenospongia cerebriformisND[20]
LeelamineIjms 25 00807 i012IC50: 9.5 µMbark of pine treesND[120]
OtobaphenolIjms 25 00807 i013NDMyristica fragransND[121]
QuercetinIjms 25 00807 i014IC50: PDK3 (~9.5 μM),flavonoid glycosides from fruits and vegetablesND[122]
ND (not determined).
Table 3. LDHA inhibitors from natural products.
Table 3. LDHA inhibitors from natural products.
LDHA InhibitorStructurePropertyOriginClinical Trials for NSCLCReference
ApigeninIjms 25 00807 i015IC50: LDHA (0.042 mM)Flavonoid from fruits, vegetables, and herbsND[139]
BerberineIjms 25 00807 i016NDGoldenseal (Hydrastis canadensis)NCT03486496[140]
CapsaicinIjms 25 00807 i017NDCapsicum annuumND[141]
CatechinIjms 25 00807 i018IC50: LDHA (40.69 μM)Camellia sinensisNCT00573885
NCT00611650
[14]
CurcuminIjms 25 00807 i019NDCurcuma longaNCT02321293
NCT01048983
[143]
Epigallocatechin gallate Ijms 25 00807 i020NDCamellia sinensisND[146]
GalloflavinIjms 25 00807 i021IC50: LDHA (5.46 μM)Flavonoid from food and vegetablesND[150]
LeonurineIjms 25 00807 i022NDLeonurus cardiacaND[151]
QuercetinIjms 25 00807 i023NDQuercus, Flavonoid glycosides from fruits and vegetablesND[152]
Ursolic acidIjms 25 00807 i024NDTriterpenoid from citrus fruits and vegetablesND[154]
ND (not determined).
Table 4. Other glycolysis inhibitors from natural products.
Table 4. Other glycolysis inhibitors from natural products.
Glycolysis InhibitorStructureEffectOriginClinical Trials for NSCLCReference
α-HederinIjms 25 00807 i025GLUT1, PKM2, LDHA,
and HK2↓
Hedera helixND[159]
β-elemeneIjms 25 00807 i026PKM2↓Curcuma aromaticaND[160]
Cucurbitacin DIjms 25 00807 i027Glut1↓CucurbitaceaeND[155]
Epigallocatechin gallate Ijms 25 00807 i028HK2↓Green teaND[169]
GenisteinIjms 25 00807 i029HIF-1α, GLUT1, and HK2↓Lupin, fava beans, soybeans, kudzu, and psoraleaNCT01628471
NCT00769990
[157]
Licochalcone AIjms 25 00807 i030HIF-1α, PDK1, and GLUT1↓Glycyrrhiza uralensisND[162]
ShikoninIjms 25 00807 i031PKM2↓lithospermum erythrorhizonND[174]
SulforaphaneIjms 25 00807 i032HIF-1α, HK2, and PKM2↓BroccoliND[166,167]
Tanshinone IIAIjms 25 00807 i033HK2↓Salvia miltiorrhizaND[165]
ND (not determined). The downward arrow (↓) indicates a decrease.
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

Park, W.; Han, J.H.; Wei, S.; Yang, E.-S.; Cheon, S.-Y.; Bae, S.-J.; Ryu, D.; Chung, H.-S.; Ha, K.-T. Natural Product-Based Glycolysis Inhibitors as a Therapeutic Strategy for Epidermal Growth Factor Receptor–Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2024, 25, 807. https://doi.org/10.3390/ijms25020807

AMA Style

Park W, Han JH, Wei S, Yang E-S, Cheon S-Y, Bae S-J, Ryu D, Chung H-S, Ha K-T. Natural Product-Based Glycolysis Inhibitors as a Therapeutic Strategy for Epidermal Growth Factor Receptor–Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer. International Journal of Molecular Sciences. 2024; 25(2):807. https://doi.org/10.3390/ijms25020807

Chicago/Turabian Style

Park, Wonyoung, Jung Ho Han, Shibo Wei, Eun-Sun Yang, Se-Yun Cheon, Sung-Jin Bae, Dongryeol Ryu, Hwan-Suck Chung, and Ki-Tae Ha. 2024. "Natural Product-Based Glycolysis Inhibitors as a Therapeutic Strategy for Epidermal Growth Factor Receptor–Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer" International Journal of Molecular Sciences 25, no. 2: 807. https://doi.org/10.3390/ijms25020807

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