Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Keywords: Human papillomavirus, Metabolic reprogramming, Fatty acid metabolism, p53, Hypoxia inducible factor 1α, Cellular-MYC, Lactate dehydrogenase
Abbreviations
- CC
Cervical cancer
- HPV
Human papillomavirus
- HIF-1α
Hypoxia inducible factor 1α
- c-MYC
Cellular MYC
- pRB
Retinoblastoma protein
- AKT
Protein kinase B
- AMPK
Adenosine monophosphate activated protein kinase
- TCA
Tricarboxylic acid
- LSIL
Low-grade squamous intraepithelial lesion
- HSIL
Highgrade squamous intraepithelial lesion
- CIS
Carcinoma In Situ
- SCC
Squamous cervical cancer
- HNSCCs
Head and neck squamous cell Carcinomas
- LCRs
Long control regions
- hTERT
human telomerase reverse transcriptase
- BAK
Bcl-2 homologues antagonist/killer1
- WAF1
Wild-type p53-activated fragment 1
- KIP1
cyclindependent kinase inhibitor 1B
- CIP1
cyclin-dependent kinase inhibitor 1 A
- PI3K
PhosphoInositide-3-Kinase
- PPP
Pentose Phosphate Pathway
- ROS
Reactive oxygen species
- CPT
Carnitine palmitoyltransferase
- SREBP-1c
Sterol-regulatory element binding protein 1c
- miR-21
microRNA-21
- MCD
Malonyl-CoA decarboxylase
- FASN
Fatty acid synthase
- FABP5
Fatty acid-binding protein 5
- ACC1
Acetyl-CoA-carboxylase
- CD36
Cluster of differentiation 36
- EGFR
Epidermal growth factor receptor
- JAK/STAT
Janus kinase/Signal transducer and activator of transcription
- PKC
Protein kinase C
- ERK/MAPK
Extracellular signal regulated kinase/mitogen-activated protein kinase
- TME
Tumor micro environment
- MHC
Major histocompatibility complex
- EGF
Epidermal growth factor
- GM1
monosialotetrahexosylganglioside
- FAs
Fatty acid
- HK-2
Hexokinase-2
- PKM2
Pyruvate kinase M2
- GLUT
Glucose transporter
- OXPHOS
Oxidative phosphorylation
- ATP
Adenosine tri phosphate
- INSR
Insulin receptor
- NF-κB
Nuclear factor kappa B
- RRAD
Ras related glycolysis inhibitor and calcium channel regulator
- TIGAR
TP53-induced glycolysis and apoptosis regulators
- PFK-2/FBPase-2
6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase
- PFK-1
Phosphorfructokinase-1
- G6PD
Glucose 6-phosphate dehydrogenase
- CO2
Carbon dioxide
- FADH2
Flavin adenine dinucleotide
- NADPH
Nicotinamide adenine dinucleotide phosphate hydrogen
- GPI
Glucose phosphate isomerase
- PDK1
Pyruvate dehydrogenase kinase-1
- PEP
Phosphoenolpyruvate
- FoxO
Forkhead box O
- CBP
CREB binding protein
- PHD
Prolyl hydroxylase domain
- VHL
Von hippel–lindau
- mTORC1
mammalian Target of the rapamycin complex 1
- HDAC
Histone deacetylases
- HREs
Hypoxia response elements
- LDHA
Lactate dehydrogenase A
- MCT4
Monocarboxylate transporter type 4
- CAIX
Carbonic anhydrase IX
- SN2
System N transporter
- ASCT2
ASC amino acid transporter 2
- GLS1
Glutaminase-1
- G6P
Glucose-6-phosphate
- PDC
Pyruvate dehydrogenase complex
- PDK2
Pyruvate dehydrogenase kinase 2
- FDXR
Ferredoxin reductase
- AIF
Apoptosis-inducing factor
- SCO2
cytochrome c oxidase 2
- SOD
Superoxide dismutase
- GSH
Glutathione
- GPx
Glutathione peroxidase
- NADH
Nicotinamide adenine dinucleotide
- TAFs
Tumor associated fibroblasts
- EMT
Epithelial to mesenchymal transition
- VEGF
Vascular endothelial growth factor
- TGF-β
Transforming growth factor-beta
- IL
Interleukin
- 2-DG
Deoxy-D-glucose
- PPARα
Peroxisome proliferator-activated receptor
- PRR
Pattern recognition receptor
- RNAi
RNA interference
- TALEN
Transcription activator-like effector nucleases
- CRISPR
Clustered regularly interspaced short palindromic repeats
1. Introduction
Cancer is characterized by unchecked division of cells, and oncogenes, such as those that induce changes in growth factor receptors, have long been considered a major etiology of the disease [1]. In one of the earliest studies, Warburg (1956) experimentally demonstrated that aggressive tumors exhibit increased glycolysis—an energetically inefficient process—even in the presence of oxygen [2]. Numerous studies have subsequently linked various metabolic alterations to malignancy (Fig. 1). Both viral replication and oncogenesis must circumvent metabolic regulation, as they depend on the stimulation of anabolic processes to sustain biomass growth and catabolic pathways to produce energy for cell division and virion generation [3]. To overcome this metabolic barrier, viruses have evolved to reorganize and activate key biosynthetic pathways, targeting anabolic processes (such as lipogenesis and amino acid synthesis) while upregulating fundamental catabolic processes (such as glycolysis and the Krebs cycle) in the host. The protein kinase B (AKT) and adenosine monophosphate-activated protein kinase (AMPK) pathways are examples of significant cellular signalling cascades that viruses alter [4]. HPV (human papillomavirus) infection in cervical cancer induces metabolic changes that sustain tumor progression [5].
Fig. 1. Metabolic reprogramming in cancer cell.
Reprogramming can be triggered by many factors, such as viruses, tumor suppressors, non-coding RNAs and oncogenes. Induced reprogramming manifests in different mechanisms such as altered mitochondrial function, immune evasion, altered amino acid, glucose and fatty acid metabolism. https://BioRender.com/d43o653
Cervical cancer (CC), which affects millions of women globally, is one of the most prevalent gynecological cancers in India [6]. In India, CC was responsible for 123,907 new cases and 77,348 deaths in 2020 [7]. CC is significantly influenced by infection with high-risk HPV strains [8]. HIV infection, immunosuppression, smoking, early sexual activity, multiple sexual partners, and oral contraceptives are additional risk factors for cervical cancer [9]. Normal cervical cancer evolves into low-grade squamous intraepithelial lesions (LSIL), high-grade squamous intraepithelial lesions (HSIL), carcinoma in situ (CIS) and metastatic cancer [9]. By incorporating its DNA into the host genome and causing the production of the viral oncoproteins E6 and E7, HPV infection promotes the development of cervical cancer [8]. These proteins promote unchecked cell proliferation by interfering with important tumor suppressor pathways, such as p53 and Rb [9]. HPV-induced transformation causes metabolic reprogramming in cervical cancer cells in addition to encouraging cell cycle impairment [10]. A metabolic shift is represented by increased aerobic glycolysis (Warburg effect), in which glucose is transformed into lactate even when oxygen is present, supplying energy and building blocks for accelerated cell division [2]. To meet the biosynthetic and bioenergetic needs of malignant cells, HPV-infected cells also exhibit modified glutamine metabolism, lipid metabolism, and mitochondrial reprogramming [11]. Moreover, hypoxia in the tumor microenvironment, often induced by rapid cell growth, further enhances (HIF-1α) activation, which drives additional metabolic adaptations to support survival under low-oxygen conditions [12]. These metabolic changes in cervical cancer driven by HPV infection not only contribute to the aggressiveness of the tumor but also present potential targets for novel therapies aimed at disrupting these metabolic pathways. In this review, we focus on associations between HPV oncoproteins and metabolic reprogramming in CC, survival advantages associated with altered metabolism and new strategies for targeting these altered metabolic pathways to improve treatment outcomes and overcoming resistance in CC.
This review highlights the importance of HPV-induced metabolic redirection in cervical cancer and how understanding this process could aid in uncovering molecular specifics, repurposing drugs that target metabolic enzymes, and designing combinatorial treatments that specifically target tumor metabolism, rather than broadly affecting all cells.
2. Metabolic reprogramming in cancer progression
Cancer cells possess distinct metabolic characteristics that provide them with a survival advantage over normal cells, allowing them to compete for the resources required to sustain their metabolism. These characteristics propagate across tumor cells [13]. Otto Warburg first noted that cancer cells consume more glucose and produce more lactate than healthy cells do [2]. This phenomenon, known as the “Warburg effect,” reflects a shift in energy production from oxidative phosphorylation to glycolysis, despite increased oxygen availability (Fig. 2). Metabolic reprogramming is integral to supporting the hallmarks of cancer cells, including sustained growth, evasion of growth inhibitors, resistance to cell death, and enhanced invasiveness [14]. Altered metabolism enables cancer cells to efficiently acquire energy and nutrients, produce vital molecules, and withstand harsh conditions. For example, increased expression of glucose transporters (GLUT1) promotes glucose uptake, fueling the Warburg effect, whereas the pentose phosphate pathway (PPP) aids in the generation of ribose for nucleotide production, supporting cell division [15]. In response to nutrient deprivation, cancer cells also rely on glutamine metabolism to supply intermediates for the tri-carboxylic acid (TCA) cycle and maintain biosynthesis. The shift toward glutamine metabolism also confers antioxidant activity within the tumor microenvironment, promoting tumor survival [16]. Hypoxic conditions stabilize HIF-1α which acivates key glycolytic enzymes such as hexokinase and pyruvate kinase M2 (PKM2), ensuring continued glycolysis and survival. Additionally, activation of the hypoxia pathway induces neoangiogenesis, supporting the continued development of tumors under low oxygen levels [17]. Metabolic reprogramming also facilitates immune evasion by creating an immunosuppressive microenvironment [18]. In cervical cancer, typical metabolic hallmarks, such as elevated lactate levels, lipid accumulation, and glycolytic shifts, have been observed, with these metabolic alterations linked to HPV infection [19].
Fig. 2. Illustration depicting Warburg effect.
Warburg effect/aerobic glycolysis is one of the hallmarks of cancer, and it is a process where lactate is produced from glucose even in the presence of oxygen, resulting in increased glucose uptake followed by an increase in glycolysis and reduction in oxidative phosphorylation and increase in lactate production all of which contributes to tumor progression. An increase in extracellular lactate creates a microenvironment suitable for the progression of tumors. Consecutive arrows indicate multiple steps in between, which are not shown in detail. https://BioRender.com/z11s826
From a therapeutic perspective, understanding these metabolic alterations offers opportunities for targeting specific vulnerabilities within cancer cells. Key enzymes involved in glycolysis, glutamine metabolism, and lipogenesis [e.g., hexokinase, PKM2, fatty acid synthase (FASN) and acetyl-CoA carboxylase (ACC)] represent potential therapeutic targets [20]. Inhibiting mitochondrial reprogramming, the pentose phosphate pathway, or hypoxia-driven pathways (such as HIF-1α) may further impair tumor adaptation [13]. Combining metabolic inhibitors with conventional treatments such as chemotherapy, radiation, or immunotherapy could increase treatment efficacy and overcome resistance [21]. Targeting immune-evasion mechanisms, such as lactate production and angiogenesis, could also improve immune responses. Personalized therapies based on specific metabolic vulnerabilities may offer more effective treatment options for cervical cancer [22]. However, while metabolic reprogramming is crucial for oncogenesis, additional biological changes, including cell cycle progression, metastasis promotion, apoptosis resistance, and immune surveillance evasion, are also necessary for the development and progression of neoplasia [14].
3. Human paillomavirus and the cellular metabolism axis
HPV is a circular double-stranded DNA virus. The role of HPV infection in anogenital tract cancers, including squamous cervical cancers (SCCs), as well as cancers in the penis, anus, vulva, and vagina, has been well documented [23]. Furthermore, HPV has been associated with head and neck squamous cell carcinomas (HNSCCs) [24]. HPV is divided into low risk (HPV6, 11, 40, 42, 43, 44, 53, 54, 61, 72, 73, and 81) and high risk (HPV16, 18, 31, 35, 39, 45, 51, 52, 56, 58, and 59) types on the basis of its ability to induce malignant transformation. HPV has an icosahedral capsid, with a diameter of approximately 55 nm and contains a double-stranded, circular DNA genome of about 8000 base pairs [25]. HPV enters basal epithelial cells through microabrasions. After attaching to receptors on the cell surface, the virus enters the cell via endocytosis. In the nucleus, viral DNA is copied and assembled into newly produced capsids in the cytoplasm. The shedding of infected epithelium releases virions for transmission to new hosts [26]. There are three regions in the HPV genome: the early (E), late (L), and long control regions (LCRs) [27]. The early region encodes proteins (E1–E7) essential for transcription, cell transformation, and viral replication. The structural proteins L1 and L2 are encoded by the late region, whereas the LCR contains regulatory regions that control transcription and viral replication [28]. The viral E1 and E2 proteins maintain viral DNA in the nucleus as an episome or circular DNA. E1 functions as a helicase [29], whereas E2 regulates the expression of the E6 and E7 genes. Since E4 promotes HPV DNA replication and facilitates the generation and dispersal of the virus from cells, it is considered a biomarker of active infection [30,31]. In the early stages of infection, E5 plays a critical role. E5 stimulates cell proliferation by forming a complex with the epidermal growth factor receptor [32]. E6 and E7 are major oncoproteins, with E6 targeting multiple proteins, including p53, human telomerase reverse transcriptase (hTERT), Bcl-2-associated X protein (BAK) and c-MYC, whereas E7 targets pRB, cyclin-dependent kinase inhibitors p21 (WAF1/CIP1), p27 (KIP1), cyclin A, and cyclin E [9]. In addition to their functions in regulating the cell cycle, inhibiting apoptosis, and inducing genomic instability, recent studies have highlighted the involvement of HPV oncoproteins in cellular metabolism (Table 1). Glycolysis, the pentose phosphate pathway, and lipid metabolism are among the metabolic pathways through which HPV is known to rewire to meet the high energy requirements of virus production and to rapidly multiply cancer cells [11]. For example, E6 and E7 upregulate glycolytic enzymes such as hexokinase and PKM2, promoting the Warburg effect even in the presence of oxygen. HPV oncoproteins enhance glucose uptake by upregulating glucose transporters such as GLUT1 and glycolytic enzymes, which increase lactate production and shift metabolism away from oxidative phosphorylation [10]. E6-mediated targeting of p53 further reduces mitochondrial respiration, further driving glycolysis dependence [33]. HPV oncoproteins also influence the cellular redox balance by increasing reactive oxygen species (ROS) production [34]. In addition, the stabilization of HIF-1α by E7 in hypoxic tumor environments activates glycolytic genes to promote the Warburg effect, independent of oxygen availability. Vascular endothelial growth factor (VEGF) stimulation via HIF-1α stabilization enhances angiogenesis, further supporting tumor growth [35]. The PI3K/AKT/mTOR pathway, which is activated by HPV oncoproteins, is a key regulator of growth and metabolism [36]. Additionally, these oncoproteins facilitate glutamine metabolism to support the biosynthesis of nucleotides and amino acids [16]. HPV oncoproteins also upregulate the pentose phosphate pathway, which is crucial for nucleotide biosynthesis [15]. Oncoproteins can also modulate lipid metabolism by activating enzymes such as FASN and ACC, which are critical for de novo lipogenesis, thus supporting lipid accumulation and providing energy for tumor growth [37]. Furthermore, the metabolic changes induced by HPV can contribute to an immunosuppressive tumor microenvironment and are also linked to therapeutic resistance and recurrence [38]. Deeper insights into the HPV-cellular metabolism axis may lead to development of potential new therapeutic strategies. In addition to E6 and E7, E5 also contributes to changes in metabolic state which is discussed further in this review. However, further study is necessary to elucidate the specific mechanisms by which other early oncoproteins, such as E1, E2, and E4, participate in metabolic reprogramming as they are reported to be involved mainly in viral integration, replication and transcription [39].
Table 1. Metabolic targets of E6 and E7 oncoprotein – E6 and E7 are major oncoproteins of HPV.
Table summarizes major targets of E6 and E7 in glucose, fatty acid and amino acid metabolism.
| HPV protein | Target | Downstream target | Influence on metabolism | Reference |
|---|---|---|---|---|
| E6 | ↓p53 | ↑SREBP-1c | ↑Lipogenesis | [44] |
| ↓MCD | ↑CPT and fatty acid uptake | [46] | ||
| ↑GLUT | [70,74,176] | |||
| ↑↓↑miR-34a | ↑Glycolysis | |||
| HIF | ||||
| ↑miR-21 | ↑CD36 | ↑Uptake of fatty acids | [49] | |
| ↑HK2 | – | ↑Glycolysis | [75] | |
| ↑c-MYC | PFK1/2 | ↑Glycolysis | [101,102,106] | |
| ↑LDHA | ↑Lactate production | |||
| ↑ASCT2 ↑SN2 | ↑Amino acid metabolism | |||
| E7 | PKM2 acetylation | Accumulation of phosphoenolpyruvate | Glycolytic intermediates diverted to other metabolic pathways promoting tumor growth | [79] |
| ↓pRB ↑E2F | ↑SREBP-1c | ↑FASN and fatty acid synthesis | [51] | |
| E6/7 | ↑AKT | ↑PI3K/AKT/mTOR | ↑Lipid and protein synthesis | [52] |
| ↑HIF-1α | ↑↑LDHA | ↑Glycolysis | [95,97] | |
| MCT4 | ↑Acidic pH contributing to therapy resistance | |||
| ↑GLUT | ||||
| ↓miR-143 | ↑HK2 | ↑Glycolysis | [76] |
4. Role of HPV in regulating fatty acid metabolism
Rapidly proliferating cells exhibit markedly altered lipid metabolism. In cancer cells, fatty acids (FAs) are primarily used to synthesize glycerophospholipids and sphingolipids, which are subsequently utilized to produce signalling molecules and cell membranes [40]. Fatty acid metabolism is regulated by fatty acid synthesis and β-oxidation. FAs are metabolized into fatty acyl-CoA and subsequently into fatty acylcarnitine. Carnitine palmitoyltransferase (CPT) transports fatty acylcarnitine into mitochondria, where it is converted back to fatty acylCoA, which then undergoes β-oxidation to generate acetyl-CoA [41]. Upon entering the cytoplasm, acetyl-CoA is converted back to fatty acylCoA [42]. HPV oncoproteins target fatty acid metabolism-related transcription factors and enzymes in several ways (Fig. 3).
Fig. 3. Fatty acid metabolism and its regulation by E5, E6 and E7 encoded by HPV oncoproteins.
Oncoproteins E5, E6, and E7 of HPV activate signalling pathway leading to the expression of transcription factor SREBP-1c, an essential transcription factor involved in promoting lipogenesis by activating genes such as fatty acid synthase (FASN), ATP citrate lyase (ACLY). The dotted arrow represents a series of reactions not depicted in detail. https://BioRender.com/v39i304
4.1. E6 oncoproteins
Degradation of p53 by HPV16 E6 leads to increased fatty acid synthesis and decreased fatty acid oxidation [43]. In normal cells, p53 functions not only as a tumor suppressor but also as a lipogenic inhibitor by binding to the promoter region of sterol-regulatory element-binding protein 1c (SREBP-1c) and transcriptionally inhibiting its activity [44]. SREBP-1c is a key transcription factor that regulates glucose metabolism and promotes lipogenesis [45]. The transcription of malonyl-CoA decarboxylase (MCD), which converts malonyl-CoA to acetyl-CoA, is directly induced by activated p53 [46]. A reduction in p53 levels leads to reduced expression of MCD followed by elevated levels of malonyl-CoA, which is an allosteric inhibitor of CPT1 [47], the rate-limiting enzyme in fatty acid β-oxidation. Since β-oxidation is one of the primary pathways for ATP production from fatty acids, its inhibition can result in reduced synthesis of acetyl-CoA. Acetyl-CoA, a major byproduct of β-oxidation, typically enters the citric acid cycle (also known as the Krebs cycle) to contribute to energy production. As a consequence of a decrease in acetyl-CoA, organisms may increasingly rely on glycolysis as a compensatory mechanism to meet their energy demands [48]. Ben et al. (2015) reported that HPV16 E6 upregulates the expression of miR-21 [49], which in turn increases the expression of fatty acid-binding protein 5 (FABP5), acetyl-CoA carboxylase (ACC1), FASN, and other enzymes involved in lipid metabolism through upregulation of cluster of differentiation 36 (CD36) [37]. Increased expression of miR-21 enhances the levels of intracellular phospholipids, neutral lipids, and cellular triglycerides [50].
4.2. E7 oncoproteins
Degradation of pRB by HPV E7 increases the expression of E2F1, which binds to and activates the promoters of several lipogenic genes, including SREBP-1c, which controls genes involved in de novo lipogenesis, such as FASN. FASN catalyzes the biosynthesis of saturated fatty acids, as reported by Denechaud et al. (2015) [51]. Previous studies have shown that E6 and E7 activate AKT [52,53]. Activation of the PI3K/AKT/mTOR pathway promotes lipid and protein synthesis. SREBPs are activated by PI3K/AKT/mTOR, which promotes fatty acid absorption, production, and cholesterol synthesis [36].
4.3. E5 oncoproteins
The E5 protein is an HPV oncoprotein that interacts with the epidermal growth factor receptor (EGFR). E5 stimulates EGFR by increasing the number of receptors on the cell membrane and activating them [54]. According to An et al. (2018), EGFR activation triggers a signalling cascade that activates the PI3K/AKT/mTOR, JAK/STAT, PKC, and ERK/MAPK pathways, which in turn activate SREBP-1c [55]. A study by Bravo et al. (2004) revealed that HPV16 E5 alters the lipid composition of the plasma membrane, including increasing the levels of phosphatidylcholine and phosphatidylserine, and decreasing the levels of phosphatidylglycerol [56]. Impaired major histocompatibility complex (MHC) trafficking and loading are membrane-related functions that might be affected when the E5 protein is present [56]. This could help create a tumor microenvironment (TME) that suppresses the immune system and promotes long-term HPV infection [38]. Furthermore, Suprynowicz et al. (2007) demonstrated that high-risk HPV-16 E5 increases the levels of lipid raft components such as caveolin-1 and ganglioside GM1 in the plasma membrane of cervical cancer cells [57]. The authors assert that the ganglioside-rich membrane triggers the EGF cell signalling pathway and promotes proliferation.
Overall, oncoproteins of HPV mediate a general increase in fatty acid synthesis and a decrease in fatty acid oxidation by targeting major regulatory enzymes involved in fatty acid metabolism. The increase in fatty acid synthesis driven by HPV benefits cervical cancer progression, since lipids are vital parts of the cell membrane, major signalling pathways and a sources of energy [58,59]. For example, lipids such as leukotrienes and prostaglandins produced by cancer cells play significant roles in tumorigenesis, angiogenesis, and metastasis by activating several key signalling pathways. These include the PI3K/AKT/mTOR pathway, which promotes tumor cell growth, survival, and metabolism, enhancing proliferation and resistance to cell death [60]. Additionally, the MAPK/ERK pathway, which is activated by prostaglandins such as PGE2, drives cell migration, proliferation, and invasion, contributing to cancer metastasis [61]. The nuclear factor kappa B (NF-κB) pathway is also triggered by these lipids, promoting inflammation, immune evasion, and the survival of tumor cells, while creating a supportive tumor microenvironment [62]. Furthermore, lipids can influence the Wnt/β-catenin pathway, which regulates cell self-renewal and migration, facilitating tumor growth and spread [63]. Finally, prostaglandins such as PGE2 can upregulate VEGF, stimulating angiogenesis and ensuring that the tumor receives adequate nutrients and oxygen for continued growth and metastasis [64]. Through the activation of these pathways, lipids such as leukotrienes and prostaglandins significantly contribute to the progression of cancer. Additionally, FAs influence membrane stiffness and fluidity by altering membrane lipids, which orchestrates cancer cell survival in a changing microenvironment and promotes invasive properties and metastasis through modifications to membrane structural characteristics [65].
5. Role of HPV in regulating glucose metabolism
According to Sellers et al. (2015), the fact that lung cancer cells, for example, are not dependent on the Warburg effect demonstrates the variety and complexity of cancer metabolism [66]. In contrast, cervical cancer patients appear to adhere to the traditional Warburg effect [19]. Research has shown that HPV downstream targets, such as p53, HIF, c-MYC, PKM2, and HK2, are known metabolic regulators that contribute to the Warburg effect in cervical cancer [10].
5.1. E6 oncoproteins
Glucose is the primary energy source for cells, generating adenosine triphosphate (ATP). The glucose transporter GLUT, with its different isoforms, regulates glucose absorption into cells [67]. E6 oncoproteins bind to various host proteins, either altering their function or accelerating their degradation. p53, a crucial player in metabolic regulation, is one of the major targets of E6 [33]. p53 controls both oxidative phosphorylation (OXPHOS) and glycolysis. By targeting the glucose transporters GLUT1 and GLUT4, p53 governs glycolysis in cells [68]. Additionally, p53 regulates glycolysis by reducing glucose uptake through the inhibition of the insulin receptor (INSR). INSR is essential for the translocation of GLUT4 to intracellular vesicles and ultimately to the plasma membrane [68]. Through the suppression of NF-κB, p53 can indirectly downregulate the expression of GLUT3 [69]. p53 restricts glucose transport by directly activating the transcription of Ras-related glycolysis inhibitor and calcium channel regulator (RRAD), which blocks GLUT1 from translocating to the plasma membrane. [70]. Degradation of p53 by E6 thus increases GLUT expression, suggesting an increase in glucose uptake. In addition to targeting glucose transporters, p53 controls glycolysis in multiple ways, as detailed further (Fig. 4).
Fig. 4. p53 mediated regulation of glucose metabolism.
p53 controls synthesis of glucose transporters (GLUT1 and GLUT4), expression of TP53-induced glycolysis and apoptosis regulators (TIGAR), insulin receptor (INSR), glucose 6-phosphate dehydrogenase (G6PD), and phosphorfructokinase-1 (PFK1), resulting in controlled rate of glucose uptake and glycolysis. https://BioRender.com/u08q386
One of the downstream targets of p53, the TP53-induced glycolysis and apoptosis regulator (TIGAR) protein, is similar to the bisphosphatase domain of PFK-2/FBPase-2 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase). TIGAR functions as an FBPase-2 and decreases the levels of F2,6BP, an allosteric activator of phosphofructokinase-1 (PFK-1). This process adversely regulates PFK1 activity, thereby regulating glycolysis [71]. In short, E6-induced degradation of p53 and subsequent downregulation of TIGAR increase PFK1 activity. Previous studies have demonstrated that cytoplasmic p53 binds to and inhibits the formation of an active dimer of glucose-6-phosphate dehydrogenase (G6PD), a key enzyme in the PPP [72]. The PPP plays a vital role in cancer metabolism by generating essential metabolites, including pentose phosphates for nucleotide biosynthesis, FAs, aromatic amino acids, and nicotinamide adenine dinucleotide phosphate (NADPH) [73]. Through its regulation of the PPP, p53 effectively restricts tumor growth. Moreover, p53 regulates glycolysis through the transactivation of microRNAs (miRNAs). For example, miR-34a, which is activated by p53, inhibits multiple glycolytic enzymes such as HK1, HK2, glucose phosphate isomerase (GPI), and pyruvate dehydrogenase kinase-1 (PDK1) [74], all of which are reversed due to p53 degradation by E6. HK2, a glycolytic enzyme, is activated by E6 through the downregulation of p53. HPV activates HK2 in two ways. First, the MYC transcript, which directly regulates HK2, is upregulated and stabilized by HPV [75]. Second, when E6/E7 levels are reduced, a notable increase in the expression of miR-143-3p occurs, suggesting that E6 and E7 are involved in downregulating the expression of the HK2 inhibitory miRNA miR-143-3p [76,77].
5.2. E7 oncoproteins
In addition to targeting p53, HPV affects several enzymes involved in glucose metabolism. One such enzyme, PKM2, catalyzes the conversion of phosphoenolpyruvate (PEP) to pyruvate while generating ATP [78]. The HPV-E7 oncoprotein promotes the acetylation of the PKM2 isoform [79]. PKM2 exists in both tetrameric and dimeric forms [80], and HPV induces PKM2 to adopt a dimeric conformation, which has a reduced affinity for phosphoenolpyruvate. This results in the accumulation of PEP [79]. As a result, glycolytic intermediates are diverted into other metabolic pathways, thereby promoting tumor cell growth. Forkhead box O (FoxO)-induced glycolytic gene repression is reversed by HPV oncoprotein-induced hyperactivation of AKT. AKT serves as a crucial negative regulator of Forkhead box O (FoxO) transcription factors by phosphorylating FoxO proteins. This phosphorylation prevents FoxO from localizing to the nucleus, thereby inhibiting its transcriptional activity [81]. In addition, AKT increases the expression of the glycolytic enzyme PFK1 and enhances the expression and membrane translocation of glucose transporters [82]. Moreover, the active PI3K/AKT/mTOR pathway promotes lipid synthesis, glucose absorption, and glycolysis by inhibiting glycogen production [83].
Overall, glucose metabolism is disrupted by HPV through E6-mediated p53 degradation, resulting in increased generation of glucose transporters (GLUT1 and GLUT4), reduced TIGAR expression, and increased expression of INSR, HK2, and PFK1, which ultimately increases the rates of glucose uptake and glycolysis. E7 of HPV also contributes to glucose metabolism by targeting PKM2 and AKT, further promoting glycolysis (Fig. 5).
Fig. 5. Overall regulation of glucose metabolism mediated by E6 and E7 oncoprotein.
E6 and E7 oncoprotein targets enzymes such as pyruvate kinase M2 (PKM2) and hexokinase-2 (HK2), transcription factors such as hypoxia-inducible factor 1 (HIF1), cellular-MYC and p53, miRNAs such as miR-143 to regulate glucose metabolism and facilitate HPV induced metabolic reprogramming. https://BioRender.com/a98u037
6. Regulation of HIF-1α by E6 and E7 oncoproteins
Normal cells rely primarily on glucose as a source of pyruvate, which is the energy-producing substance that drives the TCA cycle under normoxic conditions. In hypoxic environments, normal cells transition from the TCA cycle to lactate fermentation for energy production [84]. However, cancer cells generate energy primarily through lactic acid fermentation, independent of oxygen levels [85,86]. The transcription factor HIF-1 is a critical gene that contributes to the Warburg effect in cancer and significantly influences tumor metabolism. In response to hypoxia, HIF-1 functions as a master regulator and transcription factor. It transactivates hypoxia-responsive genes in a complex with the transcriptional coactivators p300 and CREB binding protein (CBP) to facilitate adaptation to hypoxic conditions [87]. HIF-1 typically increases GLUT-1 expression, which facilitates cellular glucose uptake. In addition, HIF-1 upregulates the transcription of genes encoding glycolysis-related enzymes, including glyceraldehyde 3-phosphate dehydrogenase and hexokinases 1 and 2 [88,89]. Furthermore, HIF-1 regulates pyruvate dehydrogenase, which leads to a shift from the TCA cycle to lactate production from pyruvate [90,91].
Active HIF-1 is a heterodimer composed of two subunits: the constitutively produced HIF-1β component and the hypoxia-induced, O2-sensing HIF-1α subunit. Transcription factors such as c-MYC, active HIF-1, the STAT complex, and NF-κB bind to the promoter region of HIF-1α to stimulate its transcription [92]. Members of the prolyl hydroxylase domain (PHD) protein family primarily stabilize both HIF subunits. Under normal oxygen conditions, HIF-1α undergoes rapid degradation through hydroxylation mediated by PHD2, which creates a binding site for the Von Hippel–Lindau (VHL) tumor suppressor protein, initiating ubiquitination [93]. Through this pathway, HIF-1α is prevented from accumulating in the cytoplasm and entering the nucleus, where it normally interacts with HIF-1β to form active HIF (Fig. 6). Further discussion will address how the HPV E6 and E7 oncoproteins modulate this pathway to prevent HIF degradation and maintain its expression even under normoxic conditions.
Fig. 6. Fate of hypoxia-inducible factor 1α (HIF1α) under normoxic conditions and under the influence of E6.
Prolyl hydroxylase domain (PHD) protein family members primarily stabilize both HIF subunits. In the presence of oxygen, PHD2-mediated hydroxylation causes HIF-1α to break down quickly. This creates a binding site where the Von Hippel-Lindau (VHL) tumor suppressor protein can attach and start the ubiquitination process, thus preventing it from entering the nucleus and forming an active dimer with HIF1β. In addition, transcription factors such as hypoxia-inducible factor 1 (HIF1), signal transducer and activator of transcription (STAT), c-MYC, and E6 oncoprotein can stabilize the HIF subunit by preventing its interaction with VHL. Active HIF dimer binds to hypoxia response elements (HREs), which are present in the promoter region of genes such as monocarboxylate transporter type 4 (MCT4), hexokinase-2 (HK2), lactate dehydrogenase A (LDHA) which are involved in metabolic processes. https://BioRender.com/d13z938
HPV E6 can activate HIF-1 by blocking the inhibitory effects of p53, which has been reported to bind to the HIF-1α/p300 complex and reduce its activity [94]. Additionally, the HPV16 E6 protein can directly bind to HIF-1α and stabilize it by reducing its interaction with VHL, thereby preventing its degradation [95]. By stimulating the mammalian target of rapamycin complex 1 (mTORC1) pathway, E6 activates HIF-1α. As a metabolic sensor, the mTORC1 signalling cascade responds to nutrient and growth factor availability, leading to HIF-1α accumulation [53,96]. The HPV E7 oncoprotein also influences HIF-1 expression. In individuals with HPV16 infection in oral squamous cell carcinoma, a noticeable association found between HPV16 E7 expression and HIF-1 [97]. Furthermore, the HPV E7 protein orchestrates the dissociation of histone deacetylases (HDAC) from HIF-1α, thus activating HIF-1 [35]. However, additional research is required to fully understand the precise mechanisms involved. In summary, HPV stabilizes HIF-1α protein expression, and the HIF-1α subunit of active HIF binds to hypoxia response elements (HREs) (Fig. 6). These HREs have been identified in the promoter regions of genes critical for glucose metabolism, such as lactate dehydrogenase A (LDHA), HK2, monocarboxylate transporter type 4 (MCT4), phosphoglycerate kinase, carbonic anhydrase IX (CAIX), and GLUT1 [98,99].
The intricate molecular interaction between HIF and the oncogene c-MYC has been well documented by Gordan et al. (2007) [100]. The HIF-2α isoform activates c-MYC, whereas the HIF-1α isoform has the opposite effect. As a prominent oncogene, c-MYC targets numerous enzymes and is essential for metabolic reprogramming. It stimulates the expression of genes involved in glycolysis, including glucose transporters, PFK1/2, LDHA, enolase A, and HK2 [101]. MYC also contributes to the regulation of amino acid metabolism. It promotes the import of glutamine into cells by increasing the expression of glutamine transporters such as the system N transporter (SN2) and ASC amino acid transporter 2 (ASCT2) [102]. In addition to being a vital source of energy, glutamine serves as a precursor for the antioxidant glutathione and as a building block for protein synthesis. Furthermore, MYC activates glutaminase-1 (GLS1) by transcriptionally inhibiting miR-23a/b, a negative regulator of GLS1, which is a potential oncogene that converts glutamine to glutamate [103]. As glutamate is a precursor of the antioxidant glutathione, it becomes essential for cancer cells to maintain higher levels of ROS-scavengers such as glutathione, as cancer cells have markedly elevated ROS levels due to altered metabolism and mitochondrial dysfunction, resulting in significant buildup of oxidized lipids, proteins, and DNA [104,105].
Several strategies have been proposed for targeting the MYC oncogene in HPV infection. For example, Peter et al. (2006) demonstrated that the aberrant expression of this oncogene could be caused by the integration of the HPV genome into the MYC locus (chromosome band 8q24) [106]. Moreover, the HPV E6 protein has been shown to increase the level of O-GlcNAc transferase, an enzyme that stabilizes c-MYC through O-GlcNAcylation, which in turn increases its turnover [107]. Understanding the mechanisms underlying the HPV-HIF-c-MYC axis in HPV-induced malignancies would be valuable, as HPV may activate both HIF and c-MYC.
In addition to modulating p53, E6, by regulating HIF and c-MYC, increases the rate of glucose metabolism and accelerates cervical cancer cell proliferation. In addition to benefiting cancer cells, increased glucose availability also supports HPV genome replication. During HPV replication, a peak in ATP levels is required for the production of viral DNA. E1 of HPV, which functions as a helicase, requires large amounts of ATP for DNA unwinding and stimulation of DNA synthesis. Increased glycolysis thus benefits HPV replication by supplying the necessary ATP [108]. Additionally, high nucleotide levels, which are necessary for HPV replication, can be obtained by metabolizing glucose-6-phosphate (G6P), a glycolytic intermediate, through the PPP [109]. In this context, the Warburg effect facilitates HPV genome replication by providing the ATP and nucleotides required at various stages of the viral life cycle.
7. Regulation of the TCA cycle and the electron transport chain by HPV oncoprotiens
The pyruvate dehydrogenase complex (PDC) facilitates the metabolic conversion of pyruvate to acetyl-CoA, which then enters the Krebs cycle. The Krebs cycle, also known as the citric acid cycle or TCA cycle, is a sequence of reactions that occur in the mitochondrial matrix and are catalyzed by enzymes [110]. Acetyl-CoA, a critical carbon source derived from fatty acids or glucose, enters the citric acid cycle. This cycle generates carbon dioxide (CO2), nicotinamide adenine dinucleotide (NADH), flavin adenine dinucleotide (FADH2), and ATP. The coenzymes NADH and FADH2 produced during the cycle are subsequently used in oxidative phosphorylation. In this process, electrons from these coenzymes are transferred through a series of five enzyme complexes that constitute the oxidative phosphorylation (OXPHOS) system, which is situated in the inner mitochondrial membrane. A proton gradient is generated by pumping hydrogen ions (H+) from the mitochondrial matrix into the intermembrane space via the energy released during electron transport, with a significant portion of this energy being dissipated as heat. The enzyme ATP synthase then harnesses this proton gradient to synthesize ATP [111].
7.1. E6 oncoproteins
As previously established, p53 is degraded by the HPV E6 oncoprotein. In addition to controlling glycolysis, p53 can also modulate the Krebs cycle and oxidation reactions. P53 increases the activity of PDC by inhibiting the inhibitor of PDC, pyruvate kinase 2 (PDK2). PDC catalyzes the conversion of pyruvate to acetyl-CoA, which enters the Krebs cycle [112]. Upon HPV infection, the E6-mediated degradation of p53 leads to a reduction in PDC activity, causing pyruvate to accumulate and shift from acetyl-CoA synthesis to lactate production. The excess lactic acid generated results in an acidified tumor microenvironment [113]. The mitochondrial enzyme glutaminase 2 (GLS2) catalyzes the hydrolysis of glutamine to glutamate, and p53 modulates glutaminolysis by binding to p53 consensus DNA-binding sites in the promoter of the GLS2 gene [114]. According to Hu et al. (2010), increased GLS2 expression lowers cellular ROS levels while enhancing mitochondrial respiration, ATP synthesis, and glutathione (GSH) production [114]. Because p53 is degraded by E6, all of the aforementioned regulatory effects are reversed. The usual function of p53 in triggering the expression of proteins that maintain mitochondrial integrity and support oxidative phosphorylation, including ferredoxin reductase (FDXR) [115], apoptosis-inducing factor (AIF) [116], and cytochrome c oxidase 2 (SCO2), which are essential for the assembly of complex IV of the electron transport chain [117], is suppressed by E6-induced degradation.
7.2. E2 oncoproteins
HPV E2 can alter mitochondrial metabolism and disrupt the mitochondrial network. Xylas et al. (2014) reported that HPV-induced structural changes in mitochondria may modify metabolic processes, as mitochondria are crucial for various metabolic activities, including the TCA cycle, fatty acid β-oxidation, and oxidative phosphorylation, which produce ATP [118]. E2 interacts with the mitochondrial membrane, and a study by Lai et al. (2013) revealed that the presence of E2 in the mitochondrial membrane alters the shape of cristae and enhances ROS production, which affects cellular respiration [34]. The primary mediators of mitochondrial ROS production are complex III proteins and ATP synthase, which regulate cristae morphology. Therefore, through interactions with these proteins, the E2 protein may regulate ROS release and mitochondrial activity [119]. Reports indicate that the combination of E2 and E1 proteins increases ROS levels in tumor cells, which negatively affects the levels of glutathione and superoxide dismutase [120]. Martínez-Ramírez et al. (2018) further suggested that the suppression of antioxidants, including superoxide dismutase (SOD1/2), GSH, and glutathione peroxidase (GPx), could result in oxidative stress and DNA damage, thereby facilitating viral DNA integration and causing neoplastic transformation [119].
In summary, HPV E6, by degrading p53, and HPV E2 and E1, by influencing mitochondrial metabolism, alters mitochondrial respiration and ATP production. These changes, in addition to modulating ROS levels, induce oxidative stress and DNA damage, which could facilitate HPV genome integration.
8. Indirect effects of HPV oncoproteins on lactate metabolism
Sources of metabolites are reprogrammed by tumors to thrive in a microenvironment that would otherwise be deficient in nutrients [121]. Cancer cells can preferentially convert glucose to lactate through aerobic glycolysis, which is commonly referred to as the “Warburg effect” [122]. Lactate generated and discharged into the environment serves as an alternative metabolic substrate for tumor cells [123]. Lactate has been shown to activate c-MYC [124] and stabilize transcription factors such as HIF; the outcomes of these studies were previously described in this article. One of the most important metabolic enzymes found in the tumor microenvironment is lactate dehydrogenase (LDH), which is crucial for the conversion of pyruvate to lactate and vice versa [125]. As pyruvate is converted to lactate, NADH is reduced to NAD+, which is subsequently regenerated in the glycolytic cycle and assists cancer cells in continuing glycolysis. The glycolytic phenotype of cancer cells is maintained by LDH-A [126]. Numerous HPV targets, such as p53, HIF, and c-MYC, can stimulate the production of the LDHA enzyme. HPV thus indirectly modulates LDH levels through these transcription factors and oncogenes.
As reported by R. Zhang et al. (2016), HPV E6-induced p53 degradation reduces the expression of miR-34a, an inhibitor of LDH-A, which is activated by p53 under normal circumstances [127]. An increase in LDHA activity increases the conversion of pyruvate to lactate. An increase in lactate and its accumulation even in the presence of oxygen explains the aggressive behavior of tumors, as it provides cancer cells with an acidic internal environment that triggers the release of metalloproteinases and hyaluronidases, which causes tumor-associated fibroblasts (TAFs) to degrade the matrix and contribute to tumorigenic characteristics by promoting the remodelling of the extracellular matrix [126]. Additionally, the accumulation of lactate caused by LDH-A induces angiogenesis, metastasis, and immune response evasion (Fig. 7) in the following ways. Accumulated lactate suppresses T-cell and dendritic cell activity, which leads to immunological evasion [128]. Lactic acid builds inside tumor cells even under normoxic conditions as a result of lactate produced from pyruvate. This acidic environment stimulates the release of the angiogenic factor VEGF, epithelial to mesenchymal transition (EMT) factors such as transforming growth factor-beta (TGF-β) [126], and the interleukis; IL-8 [129] and IL-17 [130], which promote proliferation and angiogenesis. Angiogenesis is the process by which preexisting endothelial cells divide to create new blood vessels. These blood vessels provide the necessary substrate for cancer cells to multiply and proliferate. In addition to inhibiting LDH via p53, the E6 oncoprotein also increases the activity of the mTOR signalling pathway, which amplifies the accumulation of lactate by increasing LDH-A levels [131]. LDH-A has also been reported to be a c-MYC responsive gene [132].
Fig. 7. Downstream effects of increased lactate production.
Increased lactate production leads to build-up of H+ ions, resulting in reduced pH in the tumor environment which activates genes associated with angiogenesis, metastasis, and immune evasion. https://BioRender.com/r92n271
Although an increase in lactate has numerous advantages for cancer cells, to maintain a constant glycolytic flow, cancer cells need to promote the release of accumulated lactate [123]. MCT expression is upregulated in response to intracellular acidification. MCTs are responsible for lactate efflux. Only isoforms 1–4 of the MCT family can transport monocarboxylates such as lactate [133]. Under normal circumstances, p53 represses MCT1, which limits the rate of glycolysis by preventing lactate transport and causing lactate accumulation [134]. This process is reversed upon degradation of p53 induced by E6 ultimately resulting in the release of accumulated lactate thus maintaining a steady flux of glycolysis. CAIX, a pH regulator, that catalyzes the conversion of carbon dioxide to bicarbonate, is an important protein driven by hypoxia. It is responsible for acidosis adaptation and is linked to the progression of cancer [135]. CAIX overexpression in cervical cancer has also been documented by Kirkpatrick et al. (2008) [136]. MCT and CIAX are targets of the HIF1α transcription factor, which in turn is under the control of HPV [98], thus suggesting a role for HPV in modulating lactate levels in cervical cancer. Studies have indicated that drug resistance is linked to an acidic extracellular environment, both in in vitro and in vivo models [137]. Weakly basic drugs, including docetaxel, paclitaxel, vincristine, vinblastine, mitoxantrone, and doxorubicin, are essential components of modern chemotherapy [138]. The acidic extracellular environment of tumors causes these drugs to become protonated [138]. In their protonated form, these agents are less permeable to cell membranes, leading to their accumulation outside the cell. As a result, weakly basic drugs may fail to reach intracellular concentrations sufficient to effectively kill cancer cells [139]. This phenomenon is termed “physiological drug resistance,” in contrast to “biochemical drug resistance,” which arises from alterations in signalling pathways or changes in protein expression [140].
In brief, HPV can contribute to drug resistance by modulating lactate levels and maintaining the tumor microenvironment through transcription factors such as p53, c-MYC and HIF, which regulate LDH, MCT4 and CAIX all of which are important proteins linked to the hyperglycolytic and acid-resistant phenotype of cancer cells.
9. Therapeutic approaches
9.1. Targeting HPV Oncoproteins
The development and progression of cervical cancer are driven primarily by HPV-induced transformation, which is mediated by the oncoproteins E6 and E7. Inhibition of these oncoproteins has been shown to reverse HPV-induced oncogenesis [9]. The development of targeted therapeutics has been facilitated by discovery of new biomarkers and improvements in our knowledge of the molecular pathways leading to HPV induced transformation [141]. Immunotherapy, particularly immune checkpoint inhibitors, has demonstrated potential, and future treatments may include T-cell receptor therapies targeting E6/E7, pattern recognition receptor (PRR) agonists such as imiquimod for HPV-related warts, oncolytic adenoviruses, and vaccines such as Cervarix and Gardasil [142,143]. Clinical trials are underway to evaluate their efficacy in cervical cancer, while gene silencing techniques such as RNA interference (RNAi), TALENs, and CRISPR offer new avenues for localized treatment [142] Additional strategies under investigation include E5-targeted vaccines, small-molecule inhibitors, and antibody therapies to disrupt the interaction between the E5 protein and the EGFR pathway, potentially preventing HPV-associated cancer. Rimantadine has shown potential in inhibiting E5 activity in certain cancer cell lines [144]. While most research has focused on the well-established E6 and E7 oncoproteins, emerging studies are exploring small-molecule inhibitors that target E4 activity, revealing a novel approach for treating HPV-related malignancies [145].
Alterations in cellular metabolism induced by HPV oncoproteins are critical drivers of cervical cancer progression and may affect therapeutic outcomes. Consequently, targeting the interaction between HPV onco-proteins and dysregulated metabolic pathways represents a potential therapeutic strategy for cervical cancer [146]. Specifically, targeting the E6 and E7 proteins, their mRNAs, or associated metabolic genes may restore normal metabolic function, contributing to tumor suppression [147]. Approaches such as small-molecule inhibitors or gene-editing techniques could disrupt the interactions between E6/E7 and host cellular proteins [145]. Additionally, pharmacological agents that target metabolic pathways disturbed by HPV oncoproteins, such as those that inhibit glycolysis, lipid metabolism, the pentose phosphate pathway, or glutaminase, hold therapeutic promise [148]. Preclinical and clinical studies have demonstrated the potential benefits of targeting HPV oncoproteins or metabolic reprogramming in cervical cancer [147]. In vitro studies indicate that inhibitors of glycolysis increase the sensitivity of HPV-positive cancer cells and that silencing E6/E7 has been linked to the reversal of metabolic alterations and reduced tumorigenicity [149]. In vivo studies have shown that combining metabolic inhibitors with E6/E7-targeting therapies results in significant tumor growth suppression [150]. Early-phase clinical trials are investigating the efficacy of metabolic inhibitors in HPV-driven cancers, with a focus on combination therapies targeting both HPV oncoproteins and metabolic pathways [141]. Furthermore, restoring the function of tumor suppressor pathways such as p53 and Rb, and targeting the PI3K/AKT/mTOR pathway, HIF-1α, and key metabolic enzymes (e.g., hexokinase, PKM2, and LDH) may offer promising therapeutic strategies for cervical cancer [141].
9.2. Targeting metabolic enzymes as a therapeutic strategy
As discussed in this review, metabolic reprogramming in cancer cells is driven by both the cancer microenvironment and the adaptation of metabolic enzymes within the cancer cells [151]. These distinct metabolic profiles can counteract the harmful effects of anticancer medications, leading to chemotherapy resistance [20,152–154]. Depending on the type and stage of cancer, potential treatment options include surgery, chemotherapy, and radiation therapy. However, owing to the heterogeneous and inconsistent response of cancer cells, additional therapeutic approaches are needed. One promising strategy involves targeting metabolism-related reprogramming in cancer, specifically altering how cancer cells produce and utilize energy. The relationships among oncogenes, tumor suppressors, and metabolism have been increasingly studied, leading to the discovery of numerous therapeutic targets involved in cancer metabolism and the development of related therapeutic drugs [155]. The anticancer properties of certain drugs that target key metabolic pathways, such as lipid, glucose, amino acid, and one-carbon metabolism have been identified. Additionally, drugs repurposed from other therapeutic areas—such as metformin for diabetes and orlistat for obesity—have also demonstrated anticancer potential. Several of these drugs are currently undergoing clinical trials [156]. Researchers worldwide have identified compounds that increase the effectiveness of conventional therapeutic drugs, target various enzymes involved in metabolic pathways, or overcome resistance to radiation or chemotherapy [20]. A few potential metabolic targets that could complement current cervical cancer treatments are discussed in the following section.
According to Velagran et al. (2011), HK2 inhibitors, such as 2-deoxy-D-glucose (2-DG), can suppress the Warburg effect, leading to antitumor activity [157]. Several small chemical inhibitors targeting MCT1 have been reported [158]. Wy14,643, a peroxisome proliferator-activated receptor (PPARα) agonist, suppresses tumor development and chemo-resistance by blocking the mTOR pathway, decreasing glucose absorption, and inhibiting GLUT1 transcriptional activity [159]. The traditional LDHA inhibitor oxamate can effectively increase sensitivity to antitumor medications and radiation therapy by competing with pyruvate, an LDHA substrate, thereby inhibiting LDHA activity and reducing cancer cell growth, invasion, and migration [160]. Telaglenastat (CB-839), a novel GLS inhibitor, has been shown in preclinical studies to reduce the growth of pancreatic, breast, kidney, and lymphoma malignancies through glutaminase inhibition [161]. V-9302, a glutamine analogue, functions as an ASCT2 antagonist and has been shown to inhibit human liver tumor xenograft development when combined with the glutaminase inhibitor CB-839 [162]. Further investigations are needed to evaluate the efficacy of HK2, LDHA, GLUT, and MCT inhibitors in the treatment of cervical cancer, as well as to identify additional targets for future therapeutic strategies.
In tumor lipid metabolism reprogramming, an increase in de novo fatty acid and lipid synthesis, coupled with decreased breakdown, supports cancer cell invasion, migration, elevated membrane biosynthesis, and resistance to treatment [163]. Increasing the effectiveness of antitumor therapy could involve targeting lipid metabolism processes in tumors. Many tumor cells exhibit markedly elevated levels of FASN, a key enzyme responsible for de novo fatty acid synthesis. FASN over-expression has been associated with resistance to anti-neoplastic drugs, including radioresistance in head and neck carcinoma [164], cisplatin resistance in ovarian cancer cells [165], and gemcitabine resistance in pancreatic cells [166]. Numerous FASN inhibitors, such as orlistat, cerulein, and TVB-2640, have been developed. These inhibitors either directly induce cancer cell death or increase the susceptibility of drug-resistant cells to chemotherapeutic treatments [167].
9.3. Targeting metabolic enzymes as a therapeutic strategy in CC
Ping et al. (2023) compiled extensive research on targeting fatty acid metabolism in CC [168]. Several studies have focused on metabolites in cervical cancer, with reports indicating that FASN inhibitors, such as cerulenin and C75, significantly reduce lymph node metastasis in patients with cervical cancer [169]. Orlistat, an FDA-approved anti-obesity medication with demonstrated efficacy in treating tumors, is a FASN inhibitor that has been extensively studied [170,171]. Orlistat dramatically slowed the growth and division of cervical cancer cells in vitro, particularly in HPV16 and HPV18-positive cells. This effect is associated with apoptosis rather than necrosis [172].
To increase pharmaceutical efficacy, research is underway to develop FASN inhibitors with improved selectivity, reversibility, and nonreactivity. For example, TVB-2640 is the only such drug currently in clinical trials [173]. TVB-2640 therapy has demonstrated strong results in treating various solid tumors, including cervical and breast cancer. Additionally, the coadministration of TVB-2640 with paclitaxel has shown successful target binding [174]. An experiment also revealed that cervical cancer cells presented high levels of SREBP-1. Quercetin, a naturally occurring polyphenolic flavonoid, reduces SREBP-1 levels and inhibits cancer cell growth [175]. Future improvements in cervical cancer treatments are expected, and the development of effective targeted therapeutics will require a thorough understanding of the metabolic needs of cancer cells.
10. Conclusion
Cervical cancer, predominantly driven by persistent infection with high-risk HPV, is characterized by profound metabolic reprogramming that supports tumor growth, survival, and resistance to conventional therapies. The HPV-encoded oncoproteins E6 and E7 are central to this process, as they disrupt key cellular pathways and rewire metabolic networks to favour cancer progression. Specifically, E6 and E7 promote alterations in glycolysis, fatty acid metabolism, and the TCA cycle, enabling the tumor to adapt to its microenvironment and sustain rapid proliferation under conditions of stress.
A hallmark of cancer metabolism is the Warburg effect, wherein cancer cells preferentially utilize glycolysis for energy production, even in the presence of sufficient oxygen. As reviewed in this article, HPV oncoproteins play crucial roles in inducing metabolic shifts, which not only provide rapid ATP generation but also supply biosynthetic intermediates essential for cell growth and division. Additionally, increased fatty acid synthesis and altered glucose metabolism contribute to tumor progression, immune evasion, and the development of aggressive phenotypes. These metabolic adaptations are further supported by the upregulation of key enzymes such as FASN and MCTs, which play critical roles in lipid biosynthesis and pH regulation, respectively.
The metabolic reprogramming induced by HPV oncoproteins presents unique therapeutic opportunities. The targeting of metabolic enzymes such as FASN and MCT1 has shown promise in preclinical studies, as their inhibition can disrupt the metabolic dependencies of cancer cells. Furthermore, direct targeting of the HPV oncoproteins E6 and E7 remains a promising strategy. Emerging approaches include immunotherapies, gene silencing techniques (e.g., CRISPR/Cas9 or RNA interference), and small molecule inhibitors designed to neutralize oncoprotein functions. These strategies aim to restore normal cellular regulation and sensitize tumors to existing treatments.
In conclusion, a deeper understanding of HPV-mediated metabolic reprogramming in cervical cancer provides a foundation for development of personalized treatments that address the specific shortcomings of cancer cells. By combining metabolic inhibitors with therapies directed against HPV oncoproteins, it may be possible to overcome therapeutic resistance and improve the clinical outcomes of patients with HPV-associated cervical cancer. This integrated approach holds significant potential for advancing precision medicine in cervical cancer treatment, addressing both tumor progression and resistance mechanisms.
Acknowledgements
We thank DBT/Wellcome Trust India Alliance (Grant No: IA/I/22/1/506240) for funding the study, UGC-NET JRF for Ph.D. fellowship to Ms Mrudula Gore and Manipal Academy of Higher Education for infrastructure support.
We would like to acknowledge the use of Rubriq, a writing assistant tool from American Journal Experts to correct the English language. Additionally, we acknowledge the use of BioRender for creating the figures and diagrams in this manuscript.
Footnotes
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Data availability
No data was used for the research described in the article.
References
- [1].Cline MJ, Slamon DJ, Lipsick JS. Oncogenes: implications for the diagnosis and treatment of cancer. Ann Intern Med. 1984;101(2):223–233. doi: 10.7326/0003-4819-101-2-223. [DOI] [PubMed] [Google Scholar]
- [2].Warburg O. On the origin of cancer cells. Science. 1956;123(3191):309–314. doi: 10.1126/science.123.3191.309. [DOI] [PubMed] [Google Scholar]
- [3].Purdy JG, Luftig MA. Reprogramming of cellular metabolic pathways by human oncogenic viruses. Curr Opin Virol. 2019;39:60–69. doi: 10.1016/j.coviro.2019.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Thaker SK, Ch’ng J, Christofk HR. Viral hijacking of cellular metabolism. BMC Biol. 2019;17(1):59. doi: 10.1186/s12915-019-0678-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Pappa KI, Daskalakis G, Anagnou NP. Metabolic rewiring is associated with HPV-specific profiles in cervical cancer cell lines. Sci Rep. 2021;11(1):17718. doi: 10.1038/s41598-021-96038-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
- [7].Mehrotra R, Yadav K. Cervical cancer: formulation and implementation of Govt of India guidelines for screening and management. Indian J Gynecol Oncol. 2022;20(1):4. doi: 10.1007/s40944-021-00602-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Williams VM, Filippova M, Soto U, Duerksen-Hughes PJ. HPV-DNA integration and carcinogenesis: putative roles for inflammation and oxidative stress. Future Virol. 2011;6(1):45–57. doi: 10.2217/fvl.10.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Pal A, Kundu R. Human papillomavirus E6 and E7: the cervical cancer hallmarks and targets for therapy. Front Microbiol. 2019;10:3116. doi: 10.3389/fmicb.2019.03116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Li J, Yang H, Zhang L, Zhang S, Dai Y. Metabolic reprogramming and interventions in endometrial carcinoma. Biomed Pharmacother. 2023;161:114526. doi: 10.1016/j.biopha.2023.114526. [DOI] [PubMed] [Google Scholar]
- [11].Arizmendi-Izazaga A, Navarro-Tito N, Jimenez-Wences H, Mendoza-Catalan MA, Martinez-Carrillo DN, Zacapala-Gomez AE, et al. Metabolic reprogramming in cancer: role of HPV 16 variants. Pathogens. 2021;10(3) doi: 10.3390/pathogens10030347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Ortmann BM. Hypoxia-inducible factor in cancer: from pathway regulation to therapeutic opportunity. BMJ Oncol. 2024;3(1):e000154. doi: 10.1136/bmjonc-2023-000154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Schiliro C, Firestein BL. Mechanisms of metabolic reprogramming in cancer cells supporting enhanced growth and proliferation. Cells. 2021;10(5) doi: 10.3390/cells10051056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Liu S, Zhang X, Wang W, Li X, Sun X, Zhao Y, et al. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol Cancer. 2024;23(1):261. doi: 10.1186/s12943-024-02165-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, et al. Metabolic reprogramming in cancer: mechanisms and therapeutics. MedComm (2020) 2023;4(2):e218. doi: 10.1002/mco2.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Jin J, Byun JK, Choi YK, Park KG. Targeting glutamine metabolism as a therapeutic strategy for cancer. Exp Mol Med. 2023;55(4):706–715. doi: 10.1038/s12276-023-00971-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Corcoran SE, O’Neill LA. HIF1alpha and metabolic reprogramming in inflammation. J Clin Invest. 2016;126(10):3699–3707. doi: 10.1172/JCI84431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Zhang H, Li S, Wang D, Liu S, Xiao T, Gu W, et al. Metabolic reprogramming and immune evasion: the interplay in the tumor microenvironment. Biomark Res. 2024;12(1):96. doi: 10.1186/s40364-024-00646-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Li B, Sui L. Metabolic reprogramming in cervical cancer and metabolomics perspectives. Nutr Metab (Lond) 2021;18(1):93. doi: 10.1186/s12986-021-00615-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Zhao Y, Butler EB, Tan M. Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis. 2013;4(3):e532. doi: 10.1038/cddis.2013.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Tufail M, Jiang CH, Li N. Altered metabolism in cancer: insights into energy pathways and therapeutic targets. Mol Cancer. 2024;23(1):203. doi: 10.1186/s12943-024-02119-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Jiang M, Wang Y, Zhao X, Yu J. From metabolic byproduct to immune modulator: the role of lactate in tumor immune escape. Front Immunol. 2024;15:1492050. doi: 10.3389/fimmu.2024.1492050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Wang R, Pan W, Jin L, Huang W, Li Y, Wu D, et al. Human papillomavirus vaccine against cervical cancer: opportunity and challenge. Cancer Lett. 2020;471:88–102. doi: 10.1016/j.canlet.2019.11.039. [DOI] [PubMed] [Google Scholar]
- [24].Castellsague X, Alemany L, Quer M, Halec G, Quiros B, Tous S, et al. HPV involvement in head and neck cancers: comprehensive assessment of biomarkers in 3680 patients. J Natl Cancer Inst. 2016;108(6):djv403. doi: 10.1093/jnci/djv403. [DOI] [PubMed] [Google Scholar]
- [25].Haedicke J, Iftner T. Human papillomaviruses and cancer. Radiother Oncol. 2013;108(3):397–402. doi: 10.1016/j.radonc.2013.06.004. [DOI] [PubMed] [Google Scholar]
- [26].Graham SV. The human papillomavirus replication cycle, and its links to cancer progression: a comprehensive review. Clin Sci (Lond) 2017;131(17):2201–2221. doi: 10.1042/CS20160786. [DOI] [PubMed] [Google Scholar]
- [27].Burd EM. Human papillomavirus and cervical cancer. Clin Microbiol Rev. 2003;16(1):1–17. doi: 10.1128/CMR.16.1.1-17.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Munger K, Baldwin A, Edwards KM, Hayakawa H, Nguyen CL, Owens M, et al. Mechanisms of human papillomavirus-induced oncogenesis. J Virol. 2004;78(21):11451–11460. doi: 10.1128/JVI.78.21.11451-11460.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Hughes FJ, Romanos MA. E1 protein of human papillomavirus is a DNA helicase/ATPase. Nucleic Acids Res. 1993;21(25):5817–5823. doi: 10.1093/nar/21.25.5817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Baker CC, Phelps WC, Lindgren V, Braun MJ, Gonda MA, Howley PM. Structural and transcriptional analysis of human papillomavirus type 16 sequences in cervical carcinoma cell lines. J Virol. 1987;61(4):962–971. doi: 10.1128/jvi.61.4.962-971.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Doorbar J, Egawa N, Griffin H, Kranjec C, Murakami I. Human papillomavirus molecular biology and disease association. Rev Med Virol. 2015;25(Suppl 1):2–23. doi: 10.1002/rmv.1822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Hwang ES, Nottoli T, Dimaio D. The HPV16 E5 protein: expression, detection, and stable complex formation with transmembrane proteins in COS cells. Virology. 1995;211(1):227–233. doi: 10.1006/viro.1995.1395. [DOI] [PubMed] [Google Scholar]
- [33].Yapindi L, Bowley T, Kurtaneck N, Bergeson RL, James K, Wilbourne J, et al. Activation of p53-regulated pro-survival signals and hypoxia-independent mitochondrial targeting of TIGAR by human papillomavirus E6 oncoproteins. Virology. 2023;585:1–20. doi: 10.1016/j.virol.2023.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Lai D, Tan CL, Gunaratne J, Quek LS, Nei W, Thierry F, et al. Localization of HPV-18 E2 at mitochondrial membranes induces ROS release and modulates host cell metabolism. PLoS One. 2013;8(9):e75625. doi: 10.1371/journal.pone.0075625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Bodily JM, Mehta KP, Laimins LA. Human papillomavirus E7 enhances hypoxia-inducible factor 1-mediated transcription by inhibiting binding of histone deacetylases. Cancer Res. 2011;71(3):1187–1195. doi: 10.1158/0008-5472.CAN-10-2626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond) 2018;38(1):27. doi: 10.1186/s40880-018-0301-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Glatz JFC, Nabben M, Luiken J. CD36 (SR-B2) as master regulator of cellular fatty acid homeostasis. Curr Opin Lipidol. 2022;33(2):103–111. doi: 10.1097/MOL.0000000000000819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Zhang B, Li P, Wang E, Brahmi Z, Dunn KW, Blum JS, et al. The E5 protein of human papillomavirus type 16 perturbs MHC class II antigen maturation in human foreskin keratinocytes treated with interferon-gamma. Virology. 2003;310(1):100–108. doi: 10.1016/s0042-6822(03)00103-x. [DOI] [PubMed] [Google Scholar]
- [39].Burley M, Roberts S, Parish JL. Epigenetic regulation of human papillomavirus transcription in the productive virus life cycle. Semin Immunopathol. 2020;42(2):159–171. doi: 10.1007/s00281-019-00773-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Koundouros N, Poulogiannis G. Reprogramming of fatty acid metabolism in cancer. Br J Cancer. 2020;122(1):4–22. doi: 10.1038/s41416-019-0650-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Carracedo A, Cantley LC, Pandolfi PP. Cancer metabolism: fatty acid oxidation in the limelight. Nat Rev Cancer. 2013;13(4):227–232. doi: 10.1038/nrc3483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Wakil SJ. Fatty acid synthase, a proficient multifunctional enzyme. Biochemistry. 1989;28(11):4523–4530. doi: 10.1021/bi00437a001. [DOI] [PubMed] [Google Scholar]
- [43].Parrales A, Iwakuma T. p53 as a regulator of lipid metabolism in cancer. Int J Mol Sci. 2016;17(12) doi: 10.3390/ijms17122074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Yahagi N, Shimano H, Matsuzaka T, Najima Y, Sekiya M, Nakagawa Y, et al. p53 activation in adipocytes of obese mice. J Biol Chem. 2003;278(28):25395–25400. doi: 10.1074/jbc.M302364200. [DOI] [PubMed] [Google Scholar]
- [45].Ferre P, Foufelle F. Hepatic steatosis: a role for de novo lipogenesis and the transcription factor SREBP-1c. Diabetes Obes Metab. 2010;12(Suppl 2):83–92. doi: 10.1111/j.1463-1326.2010.01275.x. [DOI] [PubMed] [Google Scholar]
- [46].Liu Y, He Y, Jin A, Tikunov AP, Zhou L, Tollini LA, et al. Ribosomal protein-Mdm2-p53 pathway coordinates nutrient stress with lipid metabolism by regulating MCD and promoting fatty acid oxidation. Proc Natl Acad Sci USA. 2014;111(23):E2414–E2422. doi: 10.1073/pnas.1315605111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Rasmussen BB, Holmback UC, Volpi E, Morio-Liondore B, Paddon-Jones D, Wolfe R. Malonyl coenzyme a and the regulation of functional carnitine palmitoyltransferase-1 activity and fat oxidation in human skeletal muscle. J Clin Invest. 2002;110(11):1687–1693. doi: 10.1172/JCI15715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Chowdhury A, Boshnakovska A, Aich A, Methi A, Vergel Leon AM, Silbern I, et al. Metabolic switch from fatty acid oxidation to glycolysis in knock-in mouse model of Barth syndrome. EMBO Mol Med. 2023;15(9):e17399. doi: 10.15252/emmm.202317399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Ben W, Yang Y, Yuan J, Sun J, Huang M, Zhang D, et al. Human papillomavirus 16 E6 modulates the expression of host microRNAs in cervical cancer, Taiwan. J Obstet Gynecol. 2015;54(4):364–370. doi: 10.1016/j.tjog.2014.06.007. [DOI] [PubMed] [Google Scholar]
- [50].Ni K, Wang D, Xu H, Mei F, Wu C, Liu Z, et al. miR-21 promotes non-small cell lung cancer cells growth by regulating fatty acid metabolism. Cancer Cell Int. 2019;19:219. doi: 10.1186/s12935-019-0941-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Denechaud PD, Lopez-Mejia IC, Giralt A, Lai Q, Blanchet E, Delacuisine B, et al. E2F1 mediates sustained lipogenesis and contributes to hepatic steatosis. J Clin Invest. 2016;126(1):137–150. doi: 10.1172/JCI81542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Pim D, Massimi P, Dilworth SM, Banks L. Activation of the protein kinase B pathway by the HPV-16 E7 oncoprotein occurs through a mechanism involving interaction with PP2A. Oncogene. 2005;24(53):7830–7838. doi: 10.1038/sj.onc.1208935. [DOI] [PubMed] [Google Scholar]
- [53].Spangle JM, Munger K. The human papillomavirus type 16 E6 oncoprotein activates mTORC1 signaling and increases protein synthesis. J Virol. 2010;84(18):9398–9407. doi: 10.1128/JVI.00974-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Ilahi NE, Bhatti A. Impact of HPV E5 on viral life cycle via EGFR signaling. Microb Pathog. 2020;139:103923. doi: 10.1016/j.micpath.2019.103923. [DOI] [PubMed] [Google Scholar]
- [55].An Z, Aksoy O, Zheng T, Fan QW, Weiss WA. Epidermal growth factor receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies. Oncogene. 2018;37(12):1561–1575. doi: 10.1038/s41388-017-0045-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Bravo IG, Crusius K, Alonso A. The E5 protein of the human papillomavirus type 16 modulates composition and dynamics of membrane lipids in keratinocytes. Arch Virol. 2005;150(2):231–246. doi: 10.1007/s00705-004-0420-x. [DOI] [PubMed] [Google Scholar]
- [57].Suprynowicz FA, Disbrow GL, Krawczyk E, Simic V, Lantzky K, Schlegel R. HPV-16 E5 oncoprotein upregulates lipid raft components caveolin-1 and ganglioside GM1 at the plasma membrane of cervical cells. Oncogene. 2008;27(8):1071–1078. doi: 10.1038/sj.onc.1210725. [DOI] [PubMed] [Google Scholar]
- [58].Cheng M, Bhujwalla ZM, Glunde K. Targeting phospholipid metabolism in cancer. Front Oncol. 2016;6:266. doi: 10.3389/fonc.2016.00266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Mollinedo F, Gajate C. Lipid rafts as signaling hubs in cancer cell survival/death and invasion: implications in tumor progression and therapy: thematic review series: biology of lipid rafts. J Lipid Res. 2020;61(5):611–635. doi: 10.1194/jlr.TR119000439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Wang D, Dubois RN. Eicosanoids and cancer. Nat Rev Cancer. 2010;10(3):181–193. doi: 10.1038/nrc2809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Krysan K, Reckamp KL, Dalwadi H, Sharma S, Rozengurt E, Dohadwala M, et al. Prostaglandin E2 activates mitogen-activated protein kinase/Erk pathway signaling and cell proliferation in non-small cell lung cancer cells in an epidermal growth factor receptor-independent manner. Cancer Res. 2005;65(14):6275–6281. doi: 10.1158/0008-5472.CAN-05-0216. [DOI] [PubMed] [Google Scholar]
- [62].Cao Y, Yi Y, Han C, Shi B. NF-kappaB signaling pathway in tumor microenvironment. Front Immunol. 2024;15:1476030. doi: 10.3389/fimmu.2024.1476030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Zhang S, Zhu N, Li HF, Gu J, Zhang CJ, Liao DF, et al. The lipid rafts in cancer stem cell: a target to eradicate cancer. Stem Cell Res Ther. 2022;13(1):432. doi: 10.1186/s13287-022-03111-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Finetti F, Travelli C, Ercoli J, Colombo G, Buoso E, Trabalzini L. Prostaglandin E2 and cancer: insight into tumor progression and immunity. Biology (Basel) 2020;9(12) doi: 10.3390/biology9120434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Martin-Perez M, Urdiroz-Urricelqui U, Bigas C, Benitah SA. The role of lipids in cancer progression and metastasis. Cell Metab. 2022;34(11):1675–1699. doi: 10.1016/j.cmet.2022.09.023. [DOI] [PubMed] [Google Scholar]
- [66].Sellers K, Fox MP, Bousamra M, 2nd, Slone SP, Higashi RM, Miller DM, et al. Pyruvate carboxylase is critical for non-small-cell lung cancer proliferation. J Clin Invest. 2015;125(2):687–698. doi: 10.1172/JCI72873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Navale AM, Paranjape AN. Glucose transporters: physiological and pathological roles. Biophys Rev. 2016;8(1):5–9. doi: 10.1007/s12551-015-0186-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E. The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res. 2004;64(7):2627–2633. doi: 10.1158/0008-5472.can-03-0846. [DOI] [PubMed] [Google Scholar]
- [69].Kawauchi K, Araki K, Tobiume K, Tanaka N. p53 regulates glucose metabolism through an IKK-NF-kappaB pathway and inhibits cell transformation. Nat Cell Biol. 2008;10(5):611–618. doi: 10.1038/ncb1724. [DOI] [PubMed] [Google Scholar]
- [70].Zhang C, Liu J, Wu R, Liang Y, Lin M, Liu J, et al. Tumor suppressor p53 negatively regulates glycolysis stimulated by hypoxia through its target RRAD. Oncotarget. 2014;5(14):5535–5546. doi: 10.18632/oncotarget.2137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Li H, Jogl G. Structural and biochemical studies of TIGAR (TP53-induced glycolysis and apoptosis regulator) J Biol Chem. 2009;284(3):1748–1754. doi: 10.1074/jbc.M807821200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Jiang P, Du W, Wang X, Mancuso A, Gao X, Wu M, et al. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat Cell Biol. 2011;13(3):310–316. doi: 10.1038/ncb2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Jin L, Zhou Y. Crucial role of the pentose phosphate pathway in malignant tumors. Oncol Lett. 2019;17(5):4213–4221. doi: 10.3892/ol.2019.10112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Kim HR, Roe JS, Lee JE, Cho EJ, Youn HD. p53 regulates glucose metabolism by miR-34a. Biochem Biophys Res Commun. 2013;437(2):225–231. doi: 10.1016/j.bbrc.2013.06.043. [DOI] [PubMed] [Google Scholar]
- [75].Zeng Q, Chen J, Li Y, Werle KD, Zhao RX, Quan CS, et al. LKB1 inhibits HPV-associated cancer progression by targeting cellular metabolism. Oncogene. 2017;36(9):1245–1255. doi: 10.1038/onc.2016.290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Konstantopoulos G, Leventakou D, Saltiel DR, Zervoudi E, Logotheti E, Pettas S, et al. HPV16 E6 oncogene contributes to cancer immune evasion by regulating PD-L1 expression through a miR-143/HIF-1a pathway. Viruses. 2024;16(1) doi: 10.3390/v16010113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Fang R, Xiao T, Fang Z, Sun Y, Li F, Gao Y, et al. MicroRNA-143 (miR-143) regulates cancer glycolysis via targeting hexokinase 2 gene. J Biol Chem. 2012;287(27):23227–23235. doi: 10.1074/jbc.M112.373084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Israelsen WJ, Vander Heiden MG. Pyruvate kinase: function, regulation and role in cancer. Semin Cell Dev Biol. 2015;43:43–51. doi: 10.1016/j.semcdb.2015.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Gui C, Ji M, Song Y, Wang J, Zhou Y. Functions and modulation of PKM2 activity by human papillomavirus E7 oncoprotein (review. Oncol Lett. 2023;25(1):7. doi: 10.3892/ol.2022.13593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Zahra K, Dey T, Ashish Mishra SP, Pandey U. Pyruvate kinase M2 and cancer: the role of PKM2 in promoting tumorigenesis. Front Oncol. 2020;10:159. doi: 10.3389/fonc.2020.00159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Tzivion G, Dobson M, Ramakrishnan G. FoxO transcription factors; regulation by AKT and 14-3-3 proteins. Biochim Biophys Acta. 2011;1813(11):1938–1945. doi: 10.1016/j.bbamcr.2011.06.002. [DOI] [PubMed] [Google Scholar]
- [82].Robey RB, Hay N. Is Akt the “Warburg kinase”?-Akt-energy metabolism interactions and oncogenesis. Semin Cancer Biol. 2009;19(1):25–31. doi: 10.1016/j.semcancer.2008.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Han B, Lin X, Hu H. Regulation of PI3K signaling in cancer metabolism and PI3K-targeting therapy. Transl Breast Cancer Res. 2024;5:33. doi: 10.21037/tbcr-24-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Li X, Yang Y, Zhang B, Lin X, Fu X, An Y, et al. Lactate metabolism in human health and disease. Signal Transduct Target Ther. 2022;7(1):305. doi: 10.1038/s41392-022-01151-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even Warburg did not anticipate. Cancer Cell. 2012;21(3):297–308. doi: 10.1016/j.ccr.2012.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J Gen Physiol. 1927;8(6):519–530. doi: 10.1085/jgp.8.6.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Yfantis A, Mylonis I, Chachami G, Nikolaidis M, Amoutzias GD, Paraskeva E, et al. Transcriptional response to hypoxia: the role of HIF-1-associated co-regulators. Cells. 2023;12(5) doi: 10.3390/cells12050798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Semenza GL. Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin Cancer Biol. 2009;19(1):12–16. doi: 10.1016/j.semcancer.2008.11.009. [DOI] [PubMed] [Google Scholar]
- [89].Semenza GL. HIF-1: upstream and downstream of cancer metabolism. Curr Opin Genet Dev. 2010;20(1):51–56. doi: 10.1016/j.gde.2009.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006;3(3):187–197. doi: 10.1016/j.cmet.2006.01.012. [DOI] [PubMed] [Google Scholar]
- [91].Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006;3(3):177–185. doi: 10.1016/j.cmet.2006.02.002. [DOI] [PubMed] [Google Scholar]
- [92].Koyasu S, Kobayashi M, Goto Y, Hiraoka M, Harada H. Regulatory mechanisms of hypoxia-inducible factor 1 activity: two decades of knowledge. Cancer Sci. 2018;109(3):560–571. doi: 10.1111/cas.13483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Haase VH. The VHL tumor suppressor: master regulator of HIF. Curr Pharm Des. 2009;15(33):3895–3903. doi: 10.2174/138161209789649394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Blagosklonny MV, An WG, Romanova LY, Trepel J, Fojo T, Neckers L. p53 inhibits hypoxia-inducible factor-stimulated transcription. J Biol Chem. 1998;273(20):11995–11998. doi: 10.1074/jbc.273.20.11995. [DOI] [PubMed] [Google Scholar]
- [95].Guo Y, Meng X, Ma J, Zheng Y, Wang Q, Wang Y, et al. Human papillomavirus 16 E6 contributes HIF-1alpha induced Warburg effect by attenuating the VHL-HIF-1alpha interaction. Int J Mol Sci. 2014;15(5):7974–7986. doi: 10.3390/ijms15057974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Luo W, Semenza GL. Pyruvate kinase M2 regulates glucose metabolism by functioning as a coactivator for hypoxia-inducible factor 1 in cancer cells. Oncotarget. 2011;2(7):551–556. doi: 10.18632/oncotarget.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Rodolico V, Arancio W, Amato MC, Aragona F, Cappello F, Di Fede O, et al. Hypoxia inducible factor-1 alpha expression is increased in infected positive HPV16 DNA oral squamous cell carcinoma and positively associated with HPV16 E7 oncoprotein. Infect Agent Cancer. 2011;6(1):18. doi: 10.1186/1750-9378-6-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Lee JW, Bae SH, Jeong JW, Kim SH, Kim KW. Hypoxia-inducible factor (HIF-1)alpha: its protein stability and biological functions. Exp Mol Med. 2004;36(1):1–12. doi: 10.1038/emm.2004.1. [DOI] [PubMed] [Google Scholar]
- [99].Wenger RH, Stiehl DP, Camenisch G. Integration of oxygen signaling at the consensus HRE. Sci STKE. 2005;2005(306):re12. doi: 10.1126/stke.3062005re12. [DOI] [PubMed] [Google Scholar]
- [100].Gordan JD, Thompson CB, Simon MC. HIF and c-Myc: sibling rivals for control of cancer cell metabolism and proliferation. Cancer Cell. 2007;12(2):108–113. doi: 10.1016/j.ccr.2007.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Dang CV, O’Donnell KA, Zeller KI, Nguyen T, Osthus RC, Li F. The c-Myc target gene network. Semin Cancer Biol. 2006;16(4):253–264. doi: 10.1016/j.semcancer.2006.07.014. [DOI] [PubMed] [Google Scholar]
- [102].Dejure FR, Eilers M. MYC and tumor metabolism: chicken and egg. EMBO J. 2017;36(23):3409–3420. doi: 10.15252/embj.201796438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458(7239):762–765. doi: 10.1038/nature07823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458(7239):780–783. doi: 10.1038/nature07733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Bansal A, Simon MC. Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol. 2018;217(7):2291–2298. doi: 10.1083/jcb.201804161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Peter M, Rosty C, Couturier J, Radvanyi F, Teshima H, Sastre-Garau X. MYC activation associated with the integration of HPV DNA at the MYC locus in genital tumors. Oncogene. 2006;25(44):5985–5993. doi: 10.1038/sj.onc.1209625. [DOI] [PubMed] [Google Scholar]
- [107].Zeng Q, Zhao RX, Chen J, Li Y, Li XD, Liu XL, et al. O-linked GlcNAcylation elevated by HPV E6 mediates viral oncogenesis. Proc Natl Acad Sci USA. 2016;113(33):9333–9338. doi: 10.1073/pnas.1606801113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Chojnacki M, Melendy T. The human papillomavirus DNA helicase E1 binds, stimulates, and confers processivity to cellular DNA polymerase epsilon. Nucleic Acids Res. 2018;46(1):229–241. doi: 10.1093/nar/gkx1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–1033. doi: 10.1126/science.1160809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Krebs HA. The citric acid cycle: a reply to the criticisms of F. L. Breusch and of J. Thomas. Biochem J. 1940;34(3):460–463. doi: 10.1042/bj0340460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Ahmad M, Wolberg A, Kahwaji CI. Biochemistry, Electron Transport Chain. StatPearls; Treasure Island (FL): 2025. [PubMed] [Google Scholar]
- [112].Contractor T, Harris CR. p53 negatively regulates transcription of the pyruvate dehydrogenase kinase Pdk2. Cancer Res. 2012;72(2):560–567. doi: 10.1158/0008-5472.CAN-11-1215. [DOI] [PubMed] [Google Scholar]
- [113].Choi SY, Collins CC, Gout PW, Wang Y. Cancer-generated lactic acid: a regulatory, immunosuppressive metabolite? J Pathol. 2013;230(4):350–355. doi: 10.1002/path.4218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci USA. 2010;107(16):7455–7460. doi: 10.1073/pnas.1001006107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Liu G, Chen X. The ferredoxin reductase gene is regulated by the p53 family and sensitizes cells to oxidative stress-induced apoptosis. Oncogene. 2002;21(47):7195–7204. doi: 10.1038/sj.onc.1205862. [DOI] [PubMed] [Google Scholar]
- [116].Stambolsky P, Weisz L, Shats I, Klein Y, Goldfinger N, Oren M, et al. Regulation of AIF expression by p53. Cell Death Differ. 2006;13(12):2140–2149. doi: 10.1038/sj.cdd.4401965. [DOI] [PubMed] [Google Scholar]
- [117].Matoba S, Kang JG, Patino WD, Wragg A, Boehm M, Gavrilova O, et al. p53 regulates mitochondrial respiration. Science. 2006;312(5780):1650–1653. doi: 10.1126/science.1126863. [DOI] [PubMed] [Google Scholar]
- [118].Xylas J, Varone A, Quinn KP, Pouli D, McLaughlin-Drubin ME, Thieu HT, et al. Noninvasive assessment of mitochondrial organization in three-dimensional tissues reveals changes associated with cancer development. Int J Cancer. 2015;136(2):322–332. doi: 10.1002/ijc.28992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Martinez-Ramirez I, Carrillo-Garcia A, Contreras-Paredes A, Ortiz-Sanchez E, Cruz-Gregorio A, Lizano M. Regulation of cellular metabolism by high-risk human papillomaviruses. Int J Mol Sci. 2018;19(7) doi: 10.3390/ijms19071839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Cruz-Gregorio A, Manzo-Merino J, Gonzalez-Garcia MC, Pedraza-Chaverri J, Medina-Campos ON, Valverde M, et al. Human papillomavirus types 16 and 18 early-expressed proteins differentially modulate the cellular redox state and DNA damage. Int J Biol Sci. 2018;14(1):21–35. doi: 10.7150/ijbs.21547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Lyssiotis CA, Kimmelman AC. Metabolic interactions in the tumor microenvironment. Trends Cell Biol. 2017;27(11):863–875. doi: 10.1016/j.tcb.2017.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Barba I, Carrillo-Bosch L, Seoane J. Targeting the Warburg effect in cancer: where do we stand? Int J Mol Sci. 2024;25(6) doi: 10.3390/ijms25063142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Perez-Tomas R, Perez-Guillen I. Lactate in the tumor microenvironment: an essential molecule in cancer progression and treatment. Cancers (Basel) 2020;12(11) doi: 10.3390/cancers12113244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Perez-Escuredo J, Dadhich RK, Dhup S, Cacace A, Van Hee VF, De Saedeleer CJ, et al. Lactate promotes glutamine uptake and metabolism in oxidative cancer cells. Cell Cycle. 2016;15(1):72–83. doi: 10.1080/15384101.2015.1120930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Mishra D, Banerjee D. Lactate dehydrogenases as metabolic links between tumor and stroma in the tumor microenvironment. Cancers (Basel) 2019;11(6) doi: 10.3390/cancers11060750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].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(12):6124–6136. doi: 10.1002/cam4.1820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Zhang R, Su J, Xue SL, Yang H, Ju LL, Ji Y, et al. HPV E6/p53 mediated down-regulation of miR-34a inhibits Warburg effect through targeting LDHA in cervical cancer. Am J Cancer Res. 2016;6(2):312–320. [PMC free article] [PubMed] [Google Scholar]
- [128].Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 2007;109(9):3812–3819. doi: 10.1182/blood-2006-07-035972. [DOI] [PubMed] [Google Scholar]
- [129].Li A, Dubey S, Varney ML, Dave BJ, Singh RK. IL-8 directly enhanced endothelial cell survival, proliferation, and matrix metalloproteinases production and regulated angiogenesis. J Immunol. 2003;170(6):3369–3376. doi: 10.4049/jimmunol.170.6.3369. [DOI] [PubMed] [Google Scholar]
- [130].Numasaki M, Fukushi J, Ono M, Narula SK, Zavodny PJ, Kudo T, et al. Interleukin-17 promotes angiogenesis and tumor growth. Blood. 2003;101(7):2620–2627. doi: 10.1182/blood-2002-05-1461. [DOI] [PubMed] [Google Scholar]
- [131].Kindt N, Descamps G, Lechien JR, Remmelink M, Colet JM, Wattiez R, et al. Involvement of HPV infection in the release of macrophage migration inhibitory factor in head and neck squamous cell carcinoma. J Clin Med. 2019;8(1) doi: 10.3390/jcm8010075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Shim H, Dolde C, Lewis BC, Wu CS, Dang G, Jungmann RA, et al. c-Myc transactivation of LDH-A: implications for tumor metabolism and growth. Proc Natl Acad Sci USA. 1997;94(13):6658–6663. doi: 10.1073/pnas.94.13.6658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Halestrap AP, Wilson MC. The monocarboxylate transporter family–role and regulation. IUBMB Life. 2012;64(2):109–119. doi: 10.1002/iub.572. [DOI] [PubMed] [Google Scholar]
- [134].Boidot R, Vegran F, Meulle A, Le Breton A, Dessy C, Sonveaux P, et al. Regulation of monocarboxylate transporter MCT1 expression by p53 mediates inward and outward lactate fluxes in tumors. Cancer Res. 2012;72(4):939–948. doi: 10.1158/0008-5472.CAN-11-2474. [DOI] [PubMed] [Google Scholar]
- [135].Pastorekova S, Gillies RJ. The role of carbonic anhydrase IX in cancer development: links to hypoxia, acidosis, and beyond. Cancer Metastasis Rev. 2019;38(1–2):65–77. doi: 10.1007/s10555-019-09799-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Kirkpatrick JP, Rabbani ZN, Bentley RC, Hardee ME, Karol S, Meyer J, et al. Elevated CAIX expression is associated with an increased risk of distant failure in early-stage cervical cancer. Biomark Insights. 2008;3:45–55. doi: 10.4137/bmi.s570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Wojtkowiak JW, Verduzco D, Schramm KJ, Gillies RJ. Drug resistance and cellular adaptation to tumor acidic pH microenvironment. Mol Pharm. 2011;8(6):2032–2038. doi: 10.1021/mp200292c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Tredan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst. 2007;99(19):1441–1454. doi: 10.1093/jnci/djm135. [DOI] [PubMed] [Google Scholar]
- [139].Li R, Xie L, Zhu Z, Liu Q, Hu Y, Jiang X, et al. Reversion of pH-induced physiological drug resistance: a novel function of copolymeric nanoparticles. PLoS One. 2011;6(9):e24172. doi: 10.1371/journal.pone.0024172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Raghunand N, Gillies RJ. pH and drug resistance in tumors. Drug Resist Updat. 2000;3(1):39–47. doi: 10.1054/drup.2000.0119. [DOI] [PubMed] [Google Scholar]
- [141].Burmeister CA, Khan SF, Prince S. Drugs and drug targets for the treatment of HPV-positive cervical cancer. Tumour Virus Res. 2024;19:200309. doi: 10.1016/j.tvr.2024.200309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Lu Z, Haghollahi S, Afzal M. Potential therapeutic targets for the treatment of HPV-associated malignancies. Cancers (Basel) 2024;16(20) doi: 10.3390/cancers16203474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Wang JW, Roden RB. Virus-like particles for the prevention of human papillomavirus-associated malignancies. Expert Rev Vaccines. 2013;12(2):129–141. doi: 10.1586/erv.12.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Miyauchi S, Sanders PD, Guram K, Kim SS, Paolini F, Venuti A, et al. HPV16 E5 mediates resistance to PD-L1 blockade and can be targeted with rimantadine in head and neck cancer. Cancer Res. 2020;80(4):732–746. doi: 10.1158/0008-5472.CAN-19-1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Bhattacharjee R, Das SS, Biswal SS, Nath A, Das D, Basu A, et al. Mechanistic role of HPV-associated early proteins in cervical cancer: molecular pathways and targeted therapeutic strategies. Crit Rev Oncol Hematol. 2022;174:103675. doi: 10.1016/j.critrevonc.2022.103675. [DOI] [PubMed] [Google Scholar]
- [146].Cruz-Gregorio A, Aranda-Rivera AK, Roviello GN, Pedraza-Chaverri J. Targeting mitochondrial therapy in the regulation of HPV infection and HPV-related cancers. Pathogens. 2023;12(3) doi: 10.3390/pathogens12030402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Tian Y, Zhang S, Ni F. Targeting glucose metabolism for HPV-associated cervical cancer: a sweet poison. Biomed Pharmacother. 2024;180:117519. doi: 10.1016/j.biopha.2024.117519. [DOI] [PubMed] [Google Scholar]
- [148].Stine ZE, Schug ZT, Salvino JM, Dang CV. Targeting cancer metabolism in the era of precision oncology. Nat Rev Drug Discov. 2022;21(2):141–162. doi: 10.1038/s41573-021-00339-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Hu C, Liu T, Han C, Xuan Y, Jiang D, Sun Y, et al. HPV E6/E7 promotes aerobic glycolysis in cervical cancer by regulating IGF2BP2 to stabilize m(6)A-MYC expression. Int J Biol Sci. 2022;18(2):507–521. doi: 10.7150/ijbs.67770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Bader JE, Voss K, Rathmell JC. Targeting metabolism to improve the tumor microenvironment for cancer immunotherapy. Mol Cell. 2020;78(6):1019–1033. doi: 10.1016/j.molcel.2020.05.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Garcia SN, Guedes RC, Marques MM. Unlocking the potential of HK2 in cancer metabolism and therapeutics. Curr Med Chem. 2019;26(41):7285–7322. doi: 10.2174/0929867326666181213092652. [DOI] [PubMed] [Google Scholar]
- [152].Varghese E, Samuel SM, Liskova A, Samec M, Kubatka P, Busselberg D. Targeting glucose metabolism to overcome resistance to anticancer chemotherapy in breast cancer. Cancers (Basel) 2020;12(8) doi: 10.3390/cancers12082252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Rahman M, Hasan MR. Cancer metabolism and drug resistance. Metabolites. 2015;5(4):571–600. doi: 10.3390/metabo5040571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Lin J, Xia L, Liang J, Han Y, Wang H, Oyang L, et al. The roles of glucose metabolic reprogramming in chemo- and radio-resistance. J Exp Clin Cancer Res. 2019;38(1):218. doi: 10.1186/s13046-019-1214-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Zheng P, Lin Z, Ding Y, Duan S. Targeting the dynamics of cancer metabolism in the era of precision oncology. Metabolism. 2023;145:155615. doi: 10.1016/j.metabol.2023.155615. [DOI] [PubMed] [Google Scholar]
- [156].Fu Y, Zou T, Shen X, Nelson PJ, Li J, Wu C, et al. Lipid metabolism in cancer progression and therapeutic strategies. MedComm (2020) 2021;2(1):27–59. doi: 10.1002/mco2.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Vegran F, Boidot R, Michiels C, Sonveaux P, Feron O. Lactate influx through the endothelial cell monocarboxylate transporter MCT1 supports an NF-kappaB/IL-8 pathway that drives tumor angiogenesis. Cancer Res. 2011;71(7):2550–2560. doi: 10.1158/0008-5472.CAN-10-2828. [DOI] [PubMed] [Google Scholar]
- [158].Puri S, Juvale K. Monocarboxylate transporter 1 and 4 inhibitors as potential therapeutics for treating solid tumours: a review with structure-activity relationship insights. Eur J Med Chem. 2020;199:112393. doi: 10.1016/j.ejmech.2020.112393. [DOI] [PubMed] [Google Scholar]
- [159].Gou Q, Dong C, Jin J, Liu Q, Lu W, Shi J, et al. PPARalpha agonist alleviates tumor growth and chemo-resistance associated with the inhibition of glucose metabolic pathway. Eur J Pharmacol. 2019;863:172664. doi: 10.1016/j.ejphar.2019.172664. [DOI] [PubMed] [Google Scholar]
- [160].Koukourakis M, Tsolou A, Pouliliou S, Lamprou I, Papadopoulou M, Ilemosoglou M, et al. Blocking LDHA glycolytic pathway sensitizes glioblastoma cells to radiation and temozolomide. Biochem Biophys Res Commun. 2017;491(4):932–938. doi: 10.1016/j.bbrc.2017.07.138. [DOI] [PubMed] [Google Scholar]
- [161].Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13(4):890–901. doi: 10.1158/1535-7163.MCT-13-0870. [DOI] [PubMed] [Google Scholar]
- [162].Jin L, Alesi GN, Kang S. Glutaminolysis as a target for cancer therapy. Oncogene. 2016;35(28):3619–3625. doi: 10.1038/onc.2015.447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Rohrig F, Schulze A. The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer. 2016;16(11):732–749. doi: 10.1038/nrc.2016.89. [DOI] [PubMed] [Google Scholar]
- [164].Mims J, Bansal N, Bharadwaj MS, Chen X, Molina AJ, Tsang AW, et al. Energy metabolism in a matched model of radiation resistance for head and neck squamous cell cancer. Radiat Res. 2015;183(3):291–304. doi: 10.1667/RR13828.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Papaevangelou E, Almeida GS, Box C, deSouza NM, Chung YL. The effect of FASN inhibition on the growth and metabolism of a cisplatin-resistant ovarian carcinoma model. Int J Cancer. 2018;143(4):992–1002. doi: 10.1002/ijc.31392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Yang Y, Liu H, Li Z, Zhao Z, Yip-Schneider M, Fan Q, et al. Role of fatty acid synthase in gemcitabine and radiation resistance of pancreatic cancers. Int J Biochem Mol Biol. 2011;2(1):89–98. [PMC free article] [PubMed] [Google Scholar]
- [167].Fhu CW, Ali A. Fatty acid synthase: an emerging target in cancer. Molecules. 2020;25(17) doi: 10.3390/molecules25173935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Ping P, Li J, Lei H, Xu X. Fatty acid metabolism: a new therapeutic target for cervical cancer. Front Oncol. 2023;13:1111778. doi: 10.3389/fonc.2023.1111778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Du Q, Liu P, Zhang C, Liu T, Wang W, Shang C, et al. FASN promotes lymph node metastasis in cervical cancer via cholesterol reprogramming and lymphangiogenesis. Cell Death Dis. 2022;13(5):488. doi: 10.1038/s41419-022-04926-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Zhou X, Chang TL, Chen S, Liu T, Wang H, Liang JF. Polydopamine-decorated orlistat-loaded hollow capsules with an enhanced cytotoxicity against cancer cell lines. Mol Pharm. 2019;16(6):2511–2521. doi: 10.1021/acs.molpharmaceut.9b00116. [DOI] [PubMed] [Google Scholar]
- [171].Zhang C, Sheng L, Yuan M, Hu J, Meng Y, Wu Y, et al. Orlistat delays hepatocarcinogenesis in mice with hepatic co-activation of AKT and c-Met. Toxicol Appl Pharmacol. 2020;392:114918. doi: 10.1016/j.taap.2020.114918. [DOI] [PubMed] [Google Scholar]
- [172].Nascimento J, Mariot C, Vianna DRB, Kliemann LM, Chaves PS, Loda M, et al. Fatty acid synthase as a potential new therapeutic target for cervical cancer. An Acad Bras Cienc. 2022;94(2):e20210670. doi: 10.1590/0001-3765202220210670. [DOI] [PubMed] [Google Scholar]
- [173].Singh S, Karthikeyan C, Moorthy N. Recent advances in the development of fatty acid synthase inhibitors as anticancer agents. Mini-Rev Med Chem. 2020;20(18):1820–1837. doi: 10.2174/1389557520666200811100845. [DOI] [PubMed] [Google Scholar]
- [174].Falchook G, Infante J, Arkenau HT, Patel MR, Dean E, Borazanci E, et al. First-in-human study of the safety, pharmacokinetics, and pharmacodynamics of first-in-class fatty acid synthase inhibitor TVB-2640 alone and with a taxane in advanced tumors. EClinicalMedicine. 2021;34:100797. doi: 10.1016/j.eclinm.2021.100797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Ali A, Kim MJ, Kim MY, Lee HJ, Roh GS, Kim HJ, et al. Quercetin induces cell death in cervical cancer by reducing O-GlcNAcylation of adenosine monophosphate-activated protein kinase. Anat Cell Biol. 2018;51(4):274–283. doi: 10.5115/acb.2018.51.4.274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Zhang C, Liu J, Wang J, Zhang T, Xu D, Hu W, et al. The interplay between tumor suppressor p53 and hypoxia signaling pathways in cancer. Front Cell Dev Biol. 2021;9:648808. doi: 10.3389/fcell.2021.648808. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No data was used for the research described in the article.







