Skip to main content

Emerging paradigms: unmasking the role of oxidative stress in HPV-induced carcinogenesis

Abstract

The contribution of the human papillomavirus (HPV) to cancer is significant but not exclusive, as carcinogenesis involves complex mechanisms, notably oxidative stress. Oxidative stress and HPV can independently cause genome instability and DNA damage, contributing to tumorigenesis. Oxidative stress-induced DNA damage, especially double-strand breaks, aids in the integration of HPV into the host genome and promotes the overexpression of two viral proteins, E6 and E7. Lifestyle factors, including diet, smoking, alcohol, and psychological stress, along with genetic and epigenetic modifications, and viral oncoproteins may influence oxidative stress, impacting the progression of HPV-related cancers. This review highlights various mechanisms in oxidative-induced HPV-mediated carcinogenesis, including altered mitochondrial morphology and function leading to elevated ROS levels, modulation of antioxidant enzymes like Superoxide Dismutase (SOD), Glutathione (GSH), and Glutathione Peroxidase (GPx), induction of chronic inflammatory environments, and activation of specific cell signaling pathways like the Phosphoinositide 3-kinase, Protein kinase B, Mammalian target of rapamycin (PI3K/AKT/mTOR) and the Extracellular signal-regulated kinase (ERK) signaling pathway. The study highlights the significance of comprehending and controlling oxidative stress in preventing and treating cancer. We suggested that incorporating dietary antioxidants and targeting cancer cells through mechanisms involving ROS could be potential interventions to mitigate the impact of oxidative stress on HPV-related malignancies.

Introduction

The human papillomavirus (HPV), a virus with double-stranded DNA, has a high tendency to infect humans. HPV infections are common and typically clear up within 12–24 months [1]. Nevertheless, a minority of infected individuals may lead to cancer, accounting for nearly 4.5% of global cancer instances [2]. HPV has a particular tropism towards squamous epithelium cells [3]. Chronic infections with high-risk HPV strains, such as HPV-16, HPV-18, HPV-31, and HPV-33, could contribute to the malignancies [4]. HPV plays a major role in cervical, vaginal, vulvar, anal, and penile cancers. Furthermore, HPV is also linked to head and neck cancers (HNC), specifically oral cavity, oropharyngeal, and laryngeal cancer [5]. In 2020, as reported by the World Health Organization, there were 604,000 recent cervical cancer diagnoses and 342,000 associated fatalities [6]. Cancer development is a complex, multistage process involving various cellular and molecular events leading to the converting a normal cell into a malignant neoplastic cell [7]. The oncoproteins E6 and E7 of HPV are responsible for HPV oncogenic properties as they inhibit the tumor suppressors p53 and pRB [1]. However, as an alternative mechanism, simultaneous episomal expression of HPV E2, E4, and E5 can also enhance vulnerability to cancer initiation, particularly in cases of Head and Neck Squamous Cell Carcinoma (HNSCC) [3].

Reactive oxygen species (ROS) are released as a byproduct of regular cellular metabolism and play a critical role in normal cellular signaling [8]. The impact of ROS on a cell is determined not only by their concentration within the cell but also through the equilibrium of ROS and endogenous antioxidants. When this balance is disrupted, oxidative stress (OS) occurs, damaging and altering various intracellular molecules, including DNA, RNA, proteins, and lipids [9]. OS facilitated by ROS might contribute to the development of cancer by changing cell redox balance, activating proinflammatory pathways, damaging DNA and promoting cell proliferation through protein oxidation [10]. Failure to detoxify ROS with antioxidants can cause a surge in OS. This heightened OS can potentially change the activity and structure of vital cellular macromolecules, such as DNA, leading to abnormal cell growth, mutation, and/or chromosome instability. Ultimately, these changes can result in the formation of neoplasms [11]. E6* expression, which is a shorter isoform of HPV E6 oncoprotein can cause elevated levels of ROS, consequently leading to increasing DNA damage. This effect could be attributed to the reduction in antioxidant capacity, as E6* expression diminished antioxidant enzymes such as superoxide dismutase isoform 2 (SOD2) and glutathione peroxidase (GPx) [12]. These findings provide evidence for a sequence of events that begins with the onset of OS, followed by DNA damage, viral integration, and, ultimately, carcinogenesis [13].

OS is gaining attention as a co-factor in HPV-mediated carcinogenesis. This article provides an overview of ROS generation and its biological effects. It also summarizes current knowledge regarding OS's impact on HPV-mediated carcinogenesis, including epigenetic modification, signaling pathways, and inflammatory responses. We will also review factors influencing the transition from HPV infection to carcinogenesis, such as dietary antioxidant components, alcohol consumption, smoking, and psychological stressors in this complex process.

HPV infection and carcinogenesis

HPV infections are usually symptomless and clear up naturally with a robust immune response. However, under specific conditions, like immunosuppression, latent infections can reactivate and, in some instances (especially with HPV types 16 and 18) may progress to cancer [14, 15]. The HPV DNA contains eight open-reading frames, categorized into early (E1, E2, E4, E5, E6, and E7) and late (L1 and L2) regions [16]. The HPV E1 protein is crucial for amplifying the viral episome within host cells, as it is the only enzyme in HPV with ATP-dependent helicase activity, facilitating the replication of the HPV genome [16, 17]. As shown in Fig. 1 in HPV-mediated carcinogenesis, the integration of the viral DNA into the host disrupts the reading frame of E1 and E2, resulting in overexpressed E6 and E7 oncogenes [18]. E2 protein functions as a regulator for the expression of the E6/E7 [19]. E6 interferes with p53 (tumor suppressor) and BAK (pro-apoptotic protein), inhibiting their function and preventing apoptosis, enabling viral DNA replication [18]. Proteasomal degradation of p53 and its inhibition is constituted through E6 binding to the ubiquitin ligase enzyme, referred to as E6 associated protein (E6AP) [20, 21]. Consequently, cells expressing E6 experience a significant reduction in p53 levels, making them prone to the aggregation of chromosomal abnormalities. The primary function of p53 is to protect the integrity of the cell's genome. When DNA damage is present, p53 is crucial in triggering cell-cycle arrest or promoting programmed cell death (apoptosis) [22]. E6 also possesses numerous p53-independent targets that actively contribute to its transformative capabilities. These targets contain a range of crucial factors, such as PDZ proteins that manage cell signaling and adhesion, the p300/CBP transcriptional activators that participate in differentiation regulation and cell cycle control, as well as proteins involved in apoptosis [23]. E6 also triggers the transcription of telomerase reverse transcriptase (TERT), which plays key role in cell immortality [24].

Fig. 1
figure 1

Various processes are involved in HPV-mediated carcinogenesis and the impact of oncoproteins on oxidative stress [21, 22, 52, 53]. In HPV-mediated carcinogenesis, integration of HPV DNA into the host leads to the disruption of E1 and E2 reading frames, resulting in overexpression of E6 and E7 oncogenes. E6 inhibits tumor suppressor protein (p53) and pro-apoptotic protein (BAK), preventing apoptosis and facilitating viral DNA replication. E6 also targets PDZ proteins, enhancing transformative capabilities. E7 interacts with the tumor suppressor (pRb), promoting carcinogenesis by disrupting cell cycle regulation. E7 facilitates E2F-induced transcription by releasing E2F from pRb-E2F complexes, increasing CDK2 activity and centrosome amplification. E7's CD3 domain interacts with proteins like p21 and p27, suppressing their activity and allowing cells to bypass DNA damage-induced cell cycle arrest. These mechanisms collectively create an environment conducive to viral DNA replication and malignant cell transformation [18, 20,21,22,23, 25, 26, 28, 54]

E7, another major HPV oncoprotein, interacts with pRb (tumor suppressor) to develop carcinogenesis [25]. pRb assumes a pivotal function in the cell cycle by regulating the shift of cells from the G1 to the S phase. In the absence of phosphorylation during the early G1 phase, pRb engages with E2F transcription factors and suppresses the transcription of the E2F-regulated genes. However, as the cell moves closer to the S phase of the cell cycle and in late G1, pRb undergoes phosphorylation, releasing E2F. Genes under the control of E2F encompass cell cycle regulators like Cyclin E and Cdc25A, as well as genes responsible for DNA replication [26]. E7 connects with unphosphorylated pRb using the LXCXE motif, facilitating its ubiquitination and subsequent degradation via the proteasome. By disrupting the pRb-E2F complex, E2F is released, triggering E2F-induced transcription. This, in turn, leads to elevated levels of cyclin-dependent kinase-2 (CDK2), as well as cyclins A and E [25]. E7's deregulation of CDK2 function sparks the amplification of centrosomes, a distinct characteristic observed in malignant cells [27]. The E7-dependent inhibition of pRb activity emerges as a pivotal factor in assisting with the shift from one stage to another in the cell cycle during the process of epithelial differentiation. This process creates an ideal environment for viral DNA replication while simultaneously driving malignant cell transformation. E7 could interacting with various cellular proteins, such as the CDK inhibitors p21 and p27. By suppressing their activity, E7 enables cells to evade cell cycle arrest stimulated by DNA damage. Moreover, E7 activates DNA methyltransferase, resulting in an uncontrollable rise in DNA methylation levels. This disruption further interferes with the epigenetic regulation of cellular processes [20, 21, 28].

In addition, the HPV E5 protein could also be involved in the development of carcinogenesis. Research has revealed that the E5 oncoprotein of HPV16 is associated with cervical lesions and may play a role in carcinogenesis by affecting cell proliferation and differentiation as well as apoptosis [29]. Lastly, E4, another HPV protein, was thought to facilitate the release of virions from superficial cell layers by disrupting keratin fibers [30].

Creation and consequences of oxidative stress

ROS can originate from both endogenous and exogenous sources. Inflammatory cells (eosinophils, neutrophils, and macrophages), mitochondria, and peroxisomes are endogenous origins of ROS [31, 32]. The exogenous origins of ROS are exposure to ionizing radiation (IR), ultraviolet (UV) radiation, biological organisms, pollutants, food, alcohol, and tobacco [33, 34]. OS is characterized by the disproportion between the production of ROS and RNS (reactive nitrogen species) and antioxidants [35]. During the process of oxygen reduction, intermediate products attack to cellular DNA, lipids, and proteins, creating reactive compounds that can subsequently interact with DNA bases [36]. ROS also are involved in the onset, promotion, and advancement phases of tumor growth. The participation of ROS in cancer development is evidenced by the presence of oxidative DNA alteration in tumor tissues. During the promotion stage, cells undergo identical mutations due to the stimulation of cell growth and/or suppression of apoptosis [37]. In this phase, ROS crucially promotes the expansion of mutant cell clones. This is accomplished by temporarily adjusting the activity of genes responsible for either cell growth or cell death [38]. Excessive ROS levels can induce transcription factors like nuclear factor-kappa B (NF-kB), which is accountable for regulating cell growth and oncogenesis [37, 39]. Elevated DNA modification caused by oxidative damage may contribute to genetic instability and potentially significantly impact tumor metastases [36, 37]. OS can potentially trigger damage to the host DNA, which in turn can make it easier for HPV DNA to integrate into infected cells [40].

Carcinogenesis of HPV through induction of ROS

Mitochondrial dysfunction

HPV can lead to carcinogenesis by disrupting mitochondria, a cellular powerhouse responsible for metabolism, cell growth, cell survival, apoptosis, and generating superoxide anions as byproducts during their oxidative phosphorylation (OXPHOS) in energy production process [40,41,42]. A study found that E1 and E4 proteins of HPV16 can attach to mitochondria, causing them to detach from microtubules. This results in the clustering of mitochondria near the cell's nucleus, leading to a decline in mitochondrial membrane potential and the initiation of programmed cell death [41]. The localization of HPV18 E2 within the mitochondrial membrane leads to morphological changes of cristae, resulting in an amplified release of ROS from mitochondria. ROS promotes an increase in glycolysis in cellular respiration, shifting away from OXPHOS, which may contribute to tumorigenesis by triggering the Warburg effect [42]. Additionally, Gregorio et al. demonstrated that HR-HPV18 E2 amplifies the production of ROS and causes a diminish in cellular glutathione (GSH) levels. Moreover, the combined expression of E2 and E1 proteins from HR-HPV18 triggers even higher levels of OS and DNA damage compared to E2 alone. This finding implies that E1 and E2 collaborate to enhance OS during the replicative cycle of HR-HPV, potentially leading to apoptosis [43].

The HR-HPV16 E2 protein engages with a mitochondrial protein (gC1qR: a receptor responsible for binding to the globular heads of C1q) that plays a role in inflammation and immune response. This interaction leads to mitochondrial abnormal function, elevated ROS generation, and triggers apoptosis through the activation of the p38 MAPK/c-jun N-terminal kinases (JNK) signaling cascade in cervical cancer cells [44]. Villota et al. observed a distinctive expression pattern in human mitochondrial noncoding ribonucleic acids (ncRNAs) among normal and HPV-infected tumor cells. Specifically, HPV E2 was found to suppress the expression of ASncmtRNAs (antisense noncoding mitochondrial tRNA). Furthermore, the oncoproteins E6 and E7 were found to stimulate SncmtRNA-2 expression, thereby promoting oncogenesis initiated by HR-HPV [45]. Similarly, in another study, the HR-HPV18 E2 protein appears to contribute to the progression of cervical cancer by suppressing the expression of ASncmtRNA. ASncmtRNA expression is upregulated in normal proliferating cells but diminished in tumor cells [46]. Recent studies revealed that HR-HPV16/18 E6 significantly impacts mitochondrial function in a model of HNSCC. These oncoproteins enhanced mitochondrial metabolism by promoting cellular respiration and the activity of mitochondrial complexes (I-V). Although there was an increase in mitochondrial function, there was no increase in ATP production. Instead, this heightened activity led to an elevated parameter of leak mitochondrial respiration, resulting in the formation of ROS within both the mitochondria and the whole cells. Consequently, this OS caused DNA damage [47]. Evans et al. revealed that the elevated expression of HPV16 E6* led to mitochondrial impairment, increased generation of ROS, and reduced GSH levels. These effects were found to be associated with the modification of pathways related to mitochondrial malfunction in cervical cancer cells [48]. The research proved that the oncoprotein E7 from HR-HPV16 stimulated both the expression and activity of catalase by reducing the production of ROS and providing a safeguarding effect against OS; the presence of E7 from HR-HPV16 oncoprotein was observed to prevent apoptosis through the mitochondrial pathway in HaCaT keratinocytes. (HaCaT cells are immortalized human keratinocytes widely utilized for studying epidermal homeostasis and its pathophysiology) [43, 49, 50]. In addition, in HPV + head and neck cancer (HNC), E6 and E7 induce OS through NADPH oxidase (NOX) enzymes, leading to DNA damage, oxidative base lesions, and also DNA SSB chromosomal irregularities [51].

HPV oncoproteins disrupt mitochondrial function, leading to OS and DNA damage, promoting carcinogenesis. The intricate interplay between HPV and mitochondrial processes highlights potential therapeutic targets for HPV-associated cancers. Understanding these mechanisms is crucial for developing targeted interventions to counteract HPV-induced mitochondrial dysfunction and halt tumorigenesis.

Inflammatory responses

The inflammatory reaction begins when the immune system receptors detect the presence of infecting pathogens or any cellular damage [55]. The immune system receptors known as pattern recognition receptors (PRRs) are specific transmembrane receptors found in immune system cells. These receptors recognize conserved molecular components in infecting microorganisms and internal injuries, which are identified as pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), respectively [56, 57]. Several PRRs have a selective ability to detect PAMPs, such as Toll-like receptors (TLRs) and also retinoic acid-inducible gene 1 (RIG 1)-like receptors (RLRs) [58]. The RLRs were shown to recognize viral-infected cells through the detection of cytoplasmic dsRNAs [59]. TLRs are present in both cell membranes (TLR1, 2, 4, 5, and 6) and endosomes (TLR3 and 9) and are pivotal in host cell recognition and antiviral immunity [60]. Activation of these PRRs induces the activity of NF-κB as a major transcription factor involved in inflammation and OS responses. NF-κB triggers the production of inflammatory cytokines and accelerates immune responses by modifying the vascular endothelial permeability and facilitating the recruitment of macrophages and neutrophils as well as natural killer (NK) cells in the infection location [61, 62]. ROS triggered NF-κB inducing inflammatory cytokines. Inflammation, in turn, can cause OS through neutrophil-generated ROS [63]. The NF-κB also can impact the level of ROS by modulating the expression of SOD1/SOD2, Glutathione S-transferase pi (GST-pi) and Gpx1 [64]. Acute inflammation resolves after removing the infecting pathogens and repairing damaged tissues, but if these disorders are not repaired quickly, inflammation will become chronic. Consistently, persistent viral infection promotes chronic inflammation, accumulating of prostaglandins, leukotrienes, and ROS that can increase the risk of DNA damage, mutation, and carcinogenesis [65, 66].

HPV has developed numerous strategies to circumvent or inhibit immune responses by regulating the expression of the inherent immune system components and also neutralizing the acquired immune responses, HPV persistent infection and progress of multiple malignancies, including cervical, vulvar, and vaginal cancers, occur [67, 68]. During replication of HPV, the virus can trigger chronic inflammatory microenvironment via IL-1β, IL-6, and also attracting immune cells to the lesion area [69]. In HPV infection, TLR9 induces the NF-κB-dependent production of type-1 IFNs by promoting several nuclear transcription factors, such as interferon regulatory factor 3 (IRF3) and IRF7 [70]. Recent studies on the antiviral effects of IRFs in HPV indicate that upregulation of these regulatory factors enhances the expression of IFNs and induces the immune clearance of HPV [71, 72]. Further results indicate that the type-1 IFNs enhance the nuclear translocation of the ISGF3 complex comprising STAT1, STAT2, and IRF3 [73]. The ISGF3 complex inhibits viral replication and decreases the proliferation of viral infected cells by facilitating the expression of various antiviral agents, including 2′,5′-oligoadenylate synthase (OAS), and protein kinase R (PKR) [74, 75]. To discern the molecular process engaged in the viral immune escape, it has been shown that infection with HPV16 inhibits the expression of TLR9 by E6 and E7, resulting in the downregulation of inflammatory cytokines like type-1 IFNs and inhibition of immune responses [76]. In addition, the study on HPV E6 and E7 in cervical cancer found that they regulate the expression of CXCL1, CXCL2, and CXCL3, as well as CXCL8 chemokines [77]. Therefore, the immune system targets viral oncogenes expression through OS; however, excessive OS may facilitate HPV DNA integration [78]. Williams et al. suggested that inflammation, through the production of ROS, can trigger DNA strand breaks in both host and virus, facilitating the integration of the HPV genome into the host cell chromatin [40]. In a study examining the impact of HPV infection on inflammation and OS, elevated levels of cytokines (IFN-γ, IL-1β, and IL-6), lipid peroxidation, and 8-hydroxydeoxyguanosine (8-OHdG) were observed alongside decreased total antioxidant capacity. 8-OHdG functioned as an indicator of OS, signifying the presence of DNA oxidation [79]. Similarly, elevated levels of cytokines were accompanied by OS marker (lipid peroxidation) and decreased levels of catalase and superoxide dismutase antioxidant enzymes in high-risk HPV patients. These results imply that HPV infection triggers a persistent inflammatory reaction and plays a role in generating an oxidative environment [80].

Inflammation initiated by HPV triggers NF-κB-mediated cytokine production and immune cell recruitment, leading to chronic inflammation and oxidative stress. HPV employs strategies to evade immune responses, promoting viral persistence and progression of malignancies, including cervical cancer. Excessive oxidative stress may facilitate HPV DNA integration, exacerbating inflammation and contributing to carcinogenesis.

The interplay between HPV infection and cellular signaling pathways

In HPV-related cancers, multiple oncogenic signaling pathways are involved in metabolism, differentiation, and cell proliferation [53]. As mention earlier, persistent infection of HPV is essential to induce cancer. It has been documented that the upregulation of HPV E6, and E7 induces diverse cellular signaling cascades implicated in cells becoming cancerous and immune escape [81]. It has been shown that HPV oncoproteins induce cell cycle progression by inhibiting several tumor suppressors, including p53, PDZ, and retinoblastoma, resulting in tumor initiation and development [81, 82]. PI3K/AKT/mTOR pathway, as one of the central signaling routes regulated in HPV‐related cancers, was implicated in regulating cell proliferation, migration, invasion, and angiogenesis [83]. The over-activation of PI3K/AKT signaling was reported in cervical squamous cell carcinomas like cervical cancer [84]. PI3K signaling was also reported to be activated by HR‐HPV16 E7 in oral cancer. Upregulation of AKT promoted the expression of Pirin and subsequent activation of nuclear factor erythroid 2‐related factor 2 (Nrf2) and the NF-κB signaling cascade, protecting cells from OS effects on viability [53]. Pirin is an OS sensor that is upregulated in various cancers and acts as a potent transcriptional activator of NF-κB, resulting in OS, cell migration, and metastasis [85, 86]. Moreover, it has been shown that HR‐HPV16 downregulates the mTOR inhibitor tuberous sclerosis complex 2 (TSC2) and promotes the activity of mTOR complex 1 (mTORC1), resulting in cell growth, metabolism, and proliferation [87]. Another signaling route that HPV promoted was the extracellular signal-regulated protein kinase (ERK) pathway that engaged in cell growth, EMT, and metastasis [88]. Upregulation of ERK signaling induces various EMT-related proteins, including Snail and Twist, and matrix metalloproteins (MMPs) implicated in cellular migration, invasion, and the spread of cancer cells [89]. Carrillo et al. examined the oncogenic mechanism of HPV in oral and cervical cancer. Their findings indicate that HR‐HPV16 E7 induces the activity of the EGFR/MEK/ERK axis and upregulates the OS sensor Pirin protein, resulting in cell motility and EMT [85]. Further studies on the relationship between ROS levels and ERK signaling in HPV16-positive cervical cancer revealed that decreased ROS levels cause reduced activity of the ERK signaling pathway. Mechanistically, ROS maintains ERK signaling activity by inhibiting ERK phosphatase dual‐specific phosphatase 3 (DUSP3) resulting in increased cell migration and EMT [53, 90].

The wnt/β-catenin pathway is another signaling route that is upregulated in HPV-related cancers. The Wnt/β-catenin axis significantly influences cell proliferation, migration, and differentiation [91,92,93]. In HPV-induced cancers, the Wnt signaling is activated by HPV E6 and E7 oncoproteins through inhibiting various regulatory factors such as p53, human telomerase reverse transcriptase (hTERT), and protein phosphatase 2A (PP2A) [94]. Further studies on the regulatory effects of HPV on Wnt/β-catenin signaling indicate that HPV E6 and E7 oncoproteins stabilize β-catenin proteins in the cytoplasm by inhibiting the degradation complex of β-catenin and PP2A in cancer cells [95]. Additionally, the E6 facilitates the nuclear translocation of β-catenin and induces the expression of Wnt signaling downstream target genes like cyclin D1 and c-MYC, leading to cell growth and proliferation [96]. Recent studies on the Wnt signaling effects on reducing immune responses against HPV-induced cancer cells indicate that c-MYC upregulates PDL1 on the surface of cancer cells. Interaction between PDL1 in cancer cells and PD1 in T cells causes apoptosis and suppresses immune responses against tumor cells [97]. Therefore, the downregulation of Wnt/β-catenin signaling and PDL-1 probebly have therapeutic effect against HPV-induced cancers.

HPV oncoproteins intricately manipulate multiple signaling pathways to drive tumorigenesis and evade immune responses, underscoring the significance of targeted therapeutic interventions in HPV-associated cancers.

DNA damage

ROS is among the most prevalent causes of DNA damage produced during inflammatory processes, cellular metabolism, infections, and chemical or mechanical stress [98]. ROS generates around 10,000 modifications in the DNA bases, resulting in dsDNA/ssDNA breaks, DNA intrastrand adducts, and cross-links to other molecules [40]. In cervical cancer, the levels of 8-nitroguanine and 8-oxodG, which are two main forms of oxidative DNA damage, were linked to higher grade of cervical intraepithelial neoplasia (CIN) [99]. In addition, HR-HPV infection cases exhibited higher serum status of malondialdehyde (MDA), a serum marker of lipid peroxidation, compared to uninfected controls [100]. MDA, which is formed through the process of lipid peroxidation, can cause DNA impairment by creating exocyclic adducts [101].

Generally, the cells activate the DNA damage response (DDR) as a result of DNA impairment [102]. This process entails a complex gene network responsible for sensing and repairing errors, ensuring genome integrity and cell survival [103]. Interestingly, HPV-infected cells adapt to OS situations by inhibiting OS-induced apoptosis and modulating antioxidant activity. These mechanisms are mediated by HPV oncogenes that have a significant impact on bypassing DDR pathways [104]. E6 protein can create a ternary complex with p53 and ubiquitin ligase (E6AP), causing to the ubiquitination and proteasomal degradation of p53 [105]. This protein is needed to detect base excision repair machinery (BER), and its degradation induces apoptosis and disturbance of cell cycle regulation [106]. E6 can also function at the mRNA level to trigger the expression of human telomerase reverse transcriptase (hTERT), contributing to maintaining telomeres length and immortalization [107]. Williams et al. showed that E6* isoform, a truncated form of E6, raises ROS levels and DNA damage by lowering the SOD 2 and GPx 1/2 antioxidant proteins [12]. The attachment of E7 to pRb inhibits its interaction with the E2F transcription factors, eliciting the transcription of S-phase related genes and an uncontrolled proliferation [21]. E7 protein also interacts with the ATR DNA damage pathway to increase the degradation of claspin, a crucial controller of the ATR/CHK1 signaling axis, thereby reducing the DNA damage checkpoint control in the G2 phase [108]. Liu et al. proved that HPV infection could shift cancer cells from homologous-recombination DNA double-strand-break (HR) repair to alternative end-joining (altEJ) by impairing TGFβ signaling. This repair pathway acts in an error-prone manner, probably promoting chromosomal aberrations [109]. Thus, the HPV employs the DDR machinery to trigger its replication in the presence of DNA damage, particularly DS breaks of the host genome, allowing multiple viral integrations [110]. The integration occurrence results in the elimination of the repressive role of E2 and persistent overexpression of the E6 and E7, driving cellular functions toward a carcinogenic process [111]. Figure 2 displays the mechanism of HPV-mediated carcinogenesis following OS and DNA damage.

Fig. 2
figure 2

Mechanism of HPV-mediated carcinogenesis following oxidative stress and DNA damage. HPV oncoproteins regulate the redox system in different ways. E1/E2 co-expression induces ROS production by decreasing the GSH/GSSG rate (reduced vs oxidized form of GSH) and the activity of SOD1/2 [43]. E6*, an isoform of E6, diminishes the levels of GPx and SOD2 proteins, thereby raising OS levels [12]. E6/E7 oncogenes lead to an increase in ROS levels by inhibiting Nrf2 activity, one of the critical transcription factors contributing to the antioxidant response, and activating Nox2 oxidase [51, 112]. ROS-induced DNA damage, especially DSB facilitates the integration of HPV into the human genome and promotes the overexpression of E6 and E7. E6 oncoprotein induces the apoptosis inhibition and immortalization of host cells through the degradation of p53 and NFX1 proteins. NFX1 is a transcriptional repressor binding to the TERT promoter and represses its expression [113, 114]. On the other hand, E7 oncoprotein promotes uncontrolled proliferation and genomic instability by degradation of pRB and claspin, a primary regulator of the ATR repair pathway [108, 115]. E5 is also capable of inhibiting apoptosis by proteasome-mediated degradation of proapoptotic BAX [116]. Glutathione (GSH), glutathione disulfide (GSSG); superoxide dismutase (SOD); glutathione peroxidase (GPx); nuclear factor erythroid 2–related factor 2 (Nrf2); NADPH oxidase 2 (Nox2); nuclear transcription factor X box-binding protein 1 (NFX1); Telomerase reverse transcriptase (TERT)

Consequently, ROS-induced DNA damage, mediated by HPV infection, underscores the intricate interplay between oxidative stress and genomic instability, promoting carcinogenesis via dysregulation of DNA repair pathways and evasion of cell cycle checkpoints. Targeting these mechanisms presents potential avenues for therapeutic intervention in HPV-associated cancers.

Epigenetic modifications

Epigenetic modifications are closely intertwined with gene expression, as they encompass changes to gene function that occur independently in alterations in the DNA sequence [117]. These modifications could regulate the activation or deactivation of genes [118]. The diverse range of mechanisms involved in epigenetic changes encompass DNA methylation, histone alterations, and post-transcriptional modifications facilitated by non-coding RNAs such as miRNA [119]. Many studies have highlighted how epigenetic abnormality in host and viral DNA, resulting from viral integration into the human genome, drives HPV's progression towards carcinogenesis. These changes affect the expression of genes like tumor suppressors and E6/E7 oncogene, as well as the DNA repair process [120,121,122].

Histone modifications, including acetylation via histone acetyltransferases (HAT) and deacetylases (HDAC), ubiquitination, methylation, and phosphorylation, serve as factors in remodeling chromatin, tied to epithelial differentiation [123, 124]. Oxidative genome damage in HPV can lead to epigenetic changes associated with tumorigenesis [110]. In the presence of OS, HPV undergoes a cascade of epigenetic alterations to regulate cellular redox equilibrium and modulate cell proliferation. In mouse cervical tumors, lactate dehydrogenase A (LDHA), as an enzyme involved in energy metabolism, is abnormally translocated to the cell nucleus due to the increase of intracellular ROS caused by HPV-16 E7. Subsequently, LDHA generates α-hydroxybutyrate and initiates histone modification by methylating H3K79 through DOT1L as a disruptor of telomeric silencing 1-like [125].

The interplay between epigenetic regulation via histone methyltransferase and the presence of immune checkpoint molecules, specifically Tim-3 and galectin-9, holds significance in cervical cancer cases, particularly those linked to HPV-18 infection. These molecules are involved in immune evasion strategies [126]. Within this context, Enhancer of Zeste Homolog 2 (EZH2) mRNA emerges as a histone methyltransferase gene responsible for adding methyl groups to histone proteins and raised in cervical cancer [127]. Notably, two transcription factors, E2F-1 and FOXM1, are activated by HPV18 E6 and E7 proteins, leading to an augmentation in EZH2 activity and subsequent suppression of DNA methyltransferase 3A (DNMT3A) expression [126].

Topchu et al. show the impact of reduced function of histone methyltransferase, such as NSD1 and NSD2, accountable for catalyzing the dimethylation of lysine 36 on histone H3 (H3K36me2) in HNSCC cells [128]. Histone modifications like H3K36me2 are vital for controlling the structure and function of chromatin, which, in turn, affects DNA repair and transcription processes [129]. An investigation on HNSCC revealed a decrease in H3K36me2 levels, a substantial diminishment in cell growth, and heightened apoptosis among HNSCC cells. Additionally, the study proved that the downregulation of NSD1 led to a decrease in E2F transcription factor gene expression [128]. Two histone acetyltransferases, Tip60, and p300, had distinct effects on histone modifications, particularly on histone H3K9, in the LCR of HPV-18. The LCR of HPV-18 exhibited a bivalent chromatin, bearing both the active histone modification of H3K9 acetylation and the inhibitory histone H3K9 trimethylation. In HeLa cells, the equilibrium between these marks was intricately controlled by Tip60 and p300, where Tip60 serves as a co-repressor, while p300 was responsible for the activation of E6 and E7 [130].

DNA methylation has been observed in both the LCR, a non-coding region controlling viral gene expression, and the E2 gene [131, 132]. Due to methylation in the LCR region, the E2 becomes unable to bind to this segment of the HPV promoter. Consequently, the regulatory influence of E2 on the E6 and E7 oncoproteins is no longer effective [133]. In patients infected with HPV-16, a positive relationship was noted in the methylation of the L1 gene within CpG regions and the presence of cervical cancer. This methylation was found to increase significantly as the severity of lesions increased [121].

Methylation in host DNA, especially in genes pivotal for cell cycle regulation, apoptosis, DNA repair, and the Wnt signaling cascade, occurs during HPV-mediated carcinogenesis [110]. Khan et al. linked the downregulated expression of the p53 gene in cancer patients to its epigenetic changes, including hypermethylation and deacetylation, shedding light on the role of these modifications in the progression of cervical cancer [134]. Further studies have established a connection between methylation in six host genes (GHSR, SST, ZIC1, ASCL1, LHX8, and ST6GALNAC5) and cervical cancer [135, 136]. Van Keer et al. stated that identifying these hypermethylation of host DNA in urine samples could serve as a non-invasive diagnostic method for low and high-grade cervical diseases [137].

Noncoding RNAs, like microRNAs (miRNAs) and small interfering RNAs (siRNAs), exert their function by interacting with the 3'-UTR (3' untranslated region) of target gene mRNA molecules to the modulation of gene expression [25]. OS can downregulate antiviral genes and miRNAs by affecting transcription factors, thus negatively influencing their expression [138, 139]. miR-34a affects thioredoxin reductase 2 (Trxrd2), which is vital for redox regulation and cancer progression. HPV's E6 degrades p53, suppressing miR-34a, leading to Trxrd2 activity. miR-34a also targets uncoupling protein 2 (UCP2), a ROS regulator. Silencing UCP2 increases ROS, and miR-34a downregulates UCP2, elevating ROS levels. miR-34a lowers SIRT1 levels, enhancing cell sensitivity to apoptosis caused by OS. During HPV infection, E6's inactivation of p53 prevents miR-34a from decreasing, thus allowing SIRT1 to remain functional [139, 140]. Gregorio et al. examined the participate of HPV oncoproteins in cellular redox regulation. They reported that the E6 oncoprotein of HR-HPV led to a diminish in cellular glutathione (GSH) levels, a rise in ROS, and a reduction in catalase levels and activity. The decrease in catalase activity suggests impaired antioxidant defence, which may be linked to epigenetic changes, including miRNA modulation, driven by the E6 oncoprotein [43]. A study investigated the role of miR-206 and its influence on G6PD in cervical cancer [141]. The Glucose-6-phosphate dehydrogenase, or G6PD, produces NADPH, which is vital for preserving decreased glutathione (GSH). GSH, in turn, acts as an antioxidant that neutralizes ROS, thereby safeguarding cancer cells from oxidative harm [142]. A study demonstrated that decreased miRNA-206 expression is indeed connected to tumorigenesis and increased cell proliferation in cervical cancer. Cui et al. reported that in cervical cancer, miR-206 exhibits a specific binding affinity for the 3' untranslated region (UTR) of G6PD. Therefore, miR-206 was found to be downregulated, while G6PD exhibited an increase in expression [141].

Consequently, Epigenetic modifications, encompassing DNA methylation, histone alterations, and non-coding RNA interactions, intricately regulate gene expression, playing a pivotal role in HPV-induced carcinogenesis by modulating oncogene expression, DNA repair processes, and immune evasion strategies. Targeting these epigenetic mechanisms holds promise for developing novel therapeutic interventions in HPV-associated cancers, offering potential avenues for precision medicine approaches.

Exogenous sources of OS and interaction with HPV

Smoking and HPV-induced carcinogenesis

Cigarette smoking with phenolic substances, quinones, and heavy metals as well as free radicals, acts, as a source of ROS, promote OS, which in turn triggers the inflammation response and probably promotes carcinogenesis [143]. Smoking can boost the risk of developing HPV-related cancers, particularly HNSCC, CIN, and cervical cancer [144, 145]. A systematic study documented that individual who smoked had 1.67 times (95% CI = 1.37–2.04) higher likelihood of developing CIN and 2.65 times (95% CI = 1.81–3.88) higher likelihood of developing cervical cancer compared to non-smokers [144]. Moreover, in a study involving 1,473 women, it was discovered that smoking heightened the chance of high-risk HPV infection, and the intensity of smoking was linked to this increase [146]. Current smokers with high-risk HPV types are less likely to clear the infection compared to non-smokers, heightened risk of persistent HPV infection associated with tobacco use [147]. However, smoking was not found to be associated with an elevated likelihood of E7 protein positivity among women who were HR-HPV positive [146].

Smoking can influence HPV virus infectivity through multiple mechanisms, including its impact on virus replication, the heightened produce of E6 and E7 oncogenes, and alterations in immune responses against HPV. Furthermore, smoking can contribute to the advancement of HPV infection towards carcinogenesis by promoting DNA damage and modifying the expression of cellular genes [148]. The HPV protein's dependence on DNA damage responses in infected cells to facilitate the replication of its genome may represent a joining between smoking-induced DNA damage and HPV pathogenesis [149]. The study exposed HPV16-transformed cervical cells to cigarette smoke condensate (CSC) for 72 h. The results showed that exposure to smoking and free radicals led to a dose-dependent increase in DNA damage, affecting both SSBs and DSBs [150]. Further study by Carrillo et al. revealed an elevation in SOD2 status and DNA damage when oral cells, which expressed HPV16 E6 and E7, were subjected to tobacco smoke [151]. SOD2 is an enzyme that involved in defending cells against OS by converting superoxide radicals into less harmful molecules [152]. Moreover, the E6 oncoprotein could increasing SOD2 levels independently of ATM and the PI3K/AKT pathway [151]. Lee et al. reported that the SOD2 gene exhibited significantly higher expression levels in HNSCC patients with a greater in heavy smokers. The SOD2 gene's expression was connected to a reduction in the peroxisome proliferator-activated receptors (PPARs) signaling pathway [153], which plays a role in regulating inflammation [154]. In an in vitro study, Shishodia et al. employed 4-Nitroquinoline N-oxide (4NQO), a known mutagen that mimics the effects of smoking and generates oxygen free radicals, to induce DNA damage in HPV-positive cells. They documented an elevation in double-strand breaks (DSBs) and the activation of homologous recombination (HR) DNA repair proteins like BRCA1 and Rad51 within HPV-positive cells [155].

Exposure to heavy metals from smoking, like cadmium and lead, may induce OS by disrupting cellular thiol buffering systems, depleting antioxidants (e.g., GSH, TXN, PRDX), and generating ROS [63, 156]. Moreover, increased serum chromium in smokers contributes to the production of O2·−, H2O2, and OH [63, 157]. Benzo[a]pyrene (BaP) in Tobacco exerts a direct influence on HPV viral replication by stimulating the MAPK/ERK pathway and subsequently activating CDK1 [98]. Kashyap et al. showed that benzo[a]pyrene (B[a]P) of cigarette smoke resulted in alterations in the expression of E6 and E7. The impact of these risk factors not only enhances clonogenesis and invasion of cervical cancer cells but also induces inflammation by affecting TNF-α and NF-kB signaling, resulting in the modulation of IL-6 and the activation of vascular endothelial growth factor (VEGFA) [158]. Therefore, aside from the DNA damage resulting from the promotion of OS due to tobacco, smoking initiates pathways influenced by ROS, connecting to RAS/MAPK, NF-κB, and AP-1, contributing to inflammation, programmed cell death, proliferation, and cell specialization. [159, 160]. Encounter with tobacco carcinogen 4-(methylnitrosamino)-1-[3-pyridyl]-1-butanone (NNK) was influential on gene expression and cellular transformation in HPV-16-immortalized human cervical cells [161]. Moreover, exposing HPV18 to NNK showed an influence on the creation of pyridyloxybutylated DNA adducts, furthering the process of neoplastic transformation in esophageal squamous epithelial cells [162]. Smoking has been associated with a potential weakening of the immune system in the cervix. This weakening is perhaps manifested through a decrease in Langerhans cells and CD4 lymphocytes [163]. The decreased clearance of high-risk HPV infection in people currently smoking compared to non-smokers, indicating an elevated risk of infection persistence linked to tobacco use, is more likely due to the impression of smoking on the immune system. In this situation, host immunity plays a role in the persistence of infection, which is an important risk factor for emerging cervical cancer [147]. Within a research investigation of women with HPV16 DNA, smokers were more likely to lack HPV16-specific antibodies, which reinforces earlier findings that smoking could elevate the likelihood of enduring infection and long-term cancer susceptibility [164]. Smoking-induced OS activates inflammation and initiates ROS production. This dual process, intended to combat damage and pathogens, can exacerbate OS, potentially contributing to cancer development [143].

In a cross-sectional study involving 3,833 participants, the connection between contact with tobacco smoke and high-risk HPV infection was examined, revealing an odds ratio of 1.32 [165]. Based on the preceding study and numerous investigations demonstrating the link between smoking and HPV infection [146, 150, 165, 166], few studies have contradicted this association [167]. For instance, a systematic study stated that smokers in the HPV (+) the group had a lower likelihood of developing HNSCC compared to those in the HPV (−) group, implying a limited role of smoking in HPV-positive HNSCC cancer [168]. The reporting inconsistency could be because smoking status was determined either through self-reports by the subjects or by identifying the presence of specific cigarette metabolites [165].

Smoking exacerbates HPV-induced carcinogenesis by promoting DNA damage, altering immune responses, and enhancing HPV infectivity through oxidative stress. While strongly linked to increased cancer risk, the interaction between smoking, HPV infection, and cancer development warrants further research due to some conflicting findings.

Alcohol and HPV-induced carcinogenesis

Alcoholic beverage consumption, recognized as a group 1 carcinogen risk factor by the International Agency for Research on Cancer (IARC) [169], contributed to 4.1% of all cancer cases in 2022, including oropharynx, colon, liver, and breast cancer [170]. A multivariate mendelian randomization examination revealed an independent role of alcohol in oral and oropharyngeal cancer with an odds ratio of 2.1 [171]. Alcohol and smoking together heighten squamous cell carcinoma risk by causing ongoing irritation and inflammation of the esophageal mucosa. This combination can also trigger changes in oncogenes like RAS mutations, which encode p21, suppress tumor suppressor genes, and cause DNA damage, ultimately leading to esophageal squamous cell carcinoma (ESCC) [172]. Notably, KRAS mutations are connected with HPV positivity in head and neck cancer and can contribute to its carcinogenesis [173]. Moreover, alcohol in aggregation with smoking and HPV infection, has been established as a contributing factor in the advancement of HNSCC [174]. This association with head and neck cancers may be attributed to free radical damage, interference with DNA repair mechanisms, and the presence of carcinogenic compounds like N-nitrosodiethylamine found in beer [171]. HPV infection may trigger specific anti-apoptotic, proliferative, and malignant cellular reactions, which can be further intensified when combined with the impact of alcohol and tobacco [172]. Alcohol consumption may notably link to a heightened risk of high-risk HPV infection. A study conducted on HNSCC patients reported a noteworthy correlation between alcohol consumption and high-risk HPV infections [175]. Furthermore, alcohol drinker was associated with a higher likelihood of persistent high-risk HPV infection among women with low-grade squamous lesions or lower cytological findings after 1 to 2 years of follow-up, compared to non-drinkers [176]. Similarly, in a cohort study involving 1243 participants, alcohol consumption was linked to over twice the odds (OR = 2.18, 95% CI 1.22–3.89) of developing CIN1 [177]. Therefore, alcohol could play a role in the early stages of HPV infection and carcinogenesis [176]. Moreover, according to Lai and his colleagues, HPV-positive patients with oropharyngeal squamous cell carcinoma (OPSCC) had a worse disease-free survival (DFS) outcome if they drank alcohol, causing 26.1% elevated risk of disease relapse, which consequently reduced the positive effect of HPV status on their prognosis [178]. A prospective study revealed that current drinkers and those with a drinking history of 5 years or more had a enhanced risk of 2-year HR-HPV persistence, which potentially contributed to cervical cancer. Variations in the levels of amines and amine oxidases in cervical mucus can lead to a diverse host response to viral infections, potentially influenced by cervical ROS [179].

Drinking alcohol leads to a low-oxygen environment with higher levels of ROS, primarily caused by the action of CYP2E1, leading to damage to DNA. This OS and the breakdown of acetaldehyde can result in the creation of DNA adducts and may be associated with epigenetic modifications of DNA, like methylation [180]. The breakdown of ethanol via alcohol dehydrogenase (ADH) results in the creation of acetaldehyde and the onset of OS, which, when combined, synergistically contribute to an increase in HPV virus load. Consequently, this contributes to both the initiation and development of cervical tumorigenesis [181]. OS can contribute to HPV- mediated carcinogenesis by increasing the risk of infection and establishment of a long-lasting infection, as well as influencing the integration of the HPV DNA into the host genome [98]. Prolonged and excessive alcohol consumption activates the hepatic microsomal ethanol-oxidizing system, including the cytochrome P-450 (CYP) enzyme, which metabolizes alcohol and generates ROS like hydroxyethyl radical, ethoxy radical, acetyl radical, and others [182]. Ethanol-induced CYP2E1 expression generates ROS, leading to OS and DNA damage. This can result in genome mutations and cell immortality as well as clonal expansion, ultimately contributing to cancer development. ROS can oxidize DNA, causing alterations like oxidized bases, SSB damage, and DNA adducts [180]. Additionally, consuming alcohol has the potential to damage mitochondria and trigger OS [183]. Ethanol may have the capacity to elevate the activity of nicotinamide adenine dinucleotide phosphate oxidase (NOX) or NADPH Oxidase enzymes, which, in turn, can lead to the formation of ROS [180].

Acetaldehyde is considered a ROS that can be formed as a outcome of the metabolism of ethyl alcohol (ethanol) through the action of the ADH enzyme [169, 184]. Acetaldehyde, present in premalignant as well as malignant lesions, induces DNA damage by forming bulky adducts and inhibiting DNA repair mechanisms like O6-methylguanine transferase (MGMT) [184, 185]. It also enhances cellular permeability, causes DNA damage through the generation of bulky adducts like N2-acetaldehyde deoxyguanine, alters the activity of enzyme convertases, and disrupts cellular oxidative balance by binding to glutathione reductase, resulting in lipid peroxidation and the formation of hydroxyl radicals [184]. Alcohol dehydrogenase (ADH) first converts ethanol to acetaldehyde, which is then further metabolized by aldehyde dehydrogenases (ALDH) to acetate [186]. ALDH2 can play a role in tumorigenesis and cancer progression mechanisms [187]. Gui et al. explored the joining between p53 and ALDH in cancer development and discovered greater diversity in ALDH isoform expression in p53-inactivated (p53WT) cases due to the HPV16 E6 oncogene in HNSCC patients. This diversity in ALDH isoforms influenced the prognosis of HNSCC in p53WT cases but not in p53HRmut cases [188].

Unlike studies suggesting a potential link between alcohol consumption and high-risk HPV infection in cancer [175,176,177,178], recently several studies have presented contrasting findings [189, 190]. Auguste et al. investigated the role of tobacco and alcohol in HNSCC and their interaction with HPV infection. They found that the effects of tobacco and alcohol, both individually and when combined, were less significant in HPV-positive HNSCC patients compared to HPV-negative cases [189]. Similarly, A systematic study found that the risk of progression of OPSCC was lower among alcohol drinkers with positive HPV compared to alcohol drinkers with negative HPV [190]. These discrepancies could be attributed to variations in data collection methods and reporting formats [190].

In summary, alcohol consumption, a recognized carcinogen, synergistically heightens the risk of HPV-induced carcinogenesis, particularly in head and neck cancers and cervical cancer. It induces oxidative stress, DNA damage, and epigenetic modifications, fostering viral infection, persistence, and tumor development. While some studies suggest a significant association between alcohol use and HPV infection, conflicting findings underscore the need for further research to clarify the interaction's complexity and its implications for cancer prevention and management.

Nutrition and HPV-induced carcinogenesis

In cervical cancer, maintaining a healthy diet can both prevent and counter HPV infection. This is achieved by safeguarding DNA from damage through the action of antioxidant vitamins that neutralize free radicals [191]. Regarding the antioxidant impact of vitamins, ascorbic acid (vitamin C) is a scavenger of free radicals, reducing O2· and OH· as well as lipid hydroperoxides [192]. Vitamin E (tocopherol), a powerful lipid-soluble antioxidant, safeguards immune cell membranes by countering ROS and reducing PUFAs' oxidation. It also indirectly regulates the immune system by inhibiting PGE2 synthesis, potentially aiding in HPV prevention [193]. Riboflavin (vitamin B2) participates in metabolic redox processes, and the metabolism of 1-carbon, and its deficiency can lead to changes in DNA methylation [194]. Vitamin B12 and folic acid hinder HPV genome methylation, reducing viral replication and persistence of infection; however, their deficiency can facilitate HPV DNA integration through DNA fragmentation [195]. Folic acid can also reduce the risk of HPV infection [196]. It has been found that vitamin E exerts its antioxidant function by eliminating ROS and oxidation of unsaturated fatty acids in the membrane of immune cells to prevent DNA damage [197]. Their isoforms and metabolic products also have anti-inflammatory and anti-cancer impacts [198]. Although the exact mechanism of its effect on HPV has not yet been determined, the use of this vitamin in combination with other micronutrients has been reported to be beneficial. In recent years, the relationship between the risk of cervical cancer and the consumption of carotenoids, vitamin E, and folate has been investigated [199]. Letafati et al. reviewed the antioxidant role of vitamins C, A, D, E, B1, B2, and folate, as well as components like Alpha lipoic acid, carotenoids, and the polyphenols of Olive Leaf in antioxidant defense against HPV-induced carcinogenesis and cervical cancer [200]. A study in American women provides the first reports of an connect between serum vitamin C levels and HPV infection. They claim that although there was no relationship between vitamin C serum levels and infection in women under 25 years of age, a U-shaped relationship was seen in older women with HPV. This means that those who had a serum level of 69.5 µmol/L had the lowest risk of infection, and those who had a serum level lower or higher than this amount had a higher risk [201]. Research indicated that antioxidants can reduce the proliferation of the virus by modulating the immune response and thus control the advancement of cervical cancer [199]. The results of a study showed that women who have a higher composite dietary antioxidant index (CDAI) are less prone to HR-HPV infection than others [202]. In a study, researchers investigated the concentration of folate while controlling the consumption of beneficial micronutrients (vitamin E, B12C, and total carotene) in women exposed to cervical neoplasia. High-risk HPV status with the hybrid absorption method 2 (HC-2) was at least three times in women. The results showed that a higher concentration of folate can reduce the possibility of getting high-risk HPV [203]. EGCG is a catechin with antiviral effects that target different stages of virus entry to fusion [204]. In a study, the effect of Epigallocatechin-3-gallate (EGCG), which is a primary bioactive polyphenol in green tea, was investigated on the growth rate and differentiation of keratinocytes transformed by the HPV virus. In this study, it was suggested that EGCG acts as an anti-virus against E6 and E7 oncoproteins; the destruction of these two is done by proteasome in cells. As a result, the expression of p53, p21WAF1, and pRB and the reduction of cell proliferation are especially beneficial. Accumulation of p53, as well as p21WAF1, can inhibit cell proliferation [205]. Another study was conducted to investigate the expression of antiviral IFN-stimulated genes (ISGs) in keratinocytes transfected with HPV-2 in skin warts. Pretreatment (and not posttreatment) with EGCG can hinder HPV-2 E7 transfection. EGCG can probably, maintain the expression of ISGs and IFN-1 signaling cascade components. The impact of pretreatment with EGCG have been reported in many viral diseases, including HIV-1, WNV, and influenza [206]. Savini et al. showed that folate deficiency was linked to elevate in the level of intracellular homocysteine, which leads to the regulation of the level of hnRNP E1 and interferes with the expression of L2, resulting in the absence of complete virus production. The lack and then the replenishment of folate levels in keratinocyte cells immortalized with HPV16 can enhance the carcinogenic effects of HPV through the change of cell phenotype [207]. Analyzing the effect of folate deficiency on the integration of HPV16 in keratinocytes of an animal model, Xiao Suhong and the authors showed that the cells of folate-deficient mouse models express more HPV 16 E6 and E7 than mice with a higher folate diet. Also, in these cells, the integration of the virus gene with genomic DNA directly or in the vicinity of the cell was about twice as high [208]. A study in American women aged 30–45 with persistent HPV infection showed that combined treatment with hyaluronic acid (HA), folic acid, EGCG, and vitamin B12 for 12 weeks reduced the persistence of the virus. This combination showed positive and effective results for the treatment of patients with (low-grade squamous intraepithelial lesion) LSIL and (atypical squamous cells- undetermined significance) ASC-US [209]. In a case report in 2023, a 39-year-old patient with severe intraepithelial neoplasia of the cervix (CIN3), with positive HPV 16 and diagnosis of the need for hysterectomy, was included in a study to investigate the effects of vitamin B12, EGCG, hyaluronic acid, and folic acid instead of surgery or before surgery. After eight weeks, there was no trace of cellular and nuclear atypia and abnormal proliferation. There was only chronic cervicitis without malignant signs. Examinations six months later also showed negative colposcopy, Pap test, and an HPV DNA test, which can confirm the hypothesis of the effectiveness of the mentioned oral compound on internal cervical lesions and HPV status [195]. Table 1 shows the role of dietary antioxidants such as vitamins C, A, E, B12, K, and folate in HPV infection and carcinogenesis. It seems that the effectiveness of all compounds can provide a clear perspective on the prevention of HPV infection and the treatment of wounds, warts, and cancers caused by this virus in the future.

Table 1 Role of dietary antioxidants such as vitamin C, A, E, B12, K and folate in HPV infection and carcinogenesis

Psychological stress and HPV-induced carcinogenesis

In psychological stress, corticotropin-releasing factor (CRF) regulates the stress response by initiating the hypothalamic–pituitary–adrenal axis, trigger the release of glucocorticoids [219]. Stress hormones, such as glucocorticoids (cortisol and epinephrine), trigger the DNA damage response by generating OS (induction of ROS, RNS) via binding to glucocorticoid receptor (GR) [220, 221]. This process involves the activation of kinases ATM and ATR, which result in cell-cycle arrest, DNA repair, and apoptosis. Glucocorticoids also interfere with DNA repair that is controlled by RAD51 and BRCA1, causing to the buildup of DNA damage [221]. In reaction to psychological stress, cortisol displayed heightened nitric oxide synthase (iNOS) expression, and glucocorticoid receptor-associated Src kinase was related to cortisol-induced production of RNS [220]. Psychological stress can indeed play a role in inducing OS and the production of O2·−, OH, and H2O2 in the brain [222]. In a mouse model study, psychological stress-induced oxidative damage disrupted the granulocyte-lymphocyte balance, increasing granulocyte count, elevating ROS production, and raising peroxynitrite levels and lipid peroxidation in the brain, heart, liver, and spleen [223].

Psychological stress can protract HPV's severity and duration through the activation of stress hormones, which may reawaken latent viruses, boost the expression of HPV oncogene, decrease the release of interferon, and hinder the body's antiviral protection [224, 225]. Exposure to HR-HPV to glucocorticoids and catecholamine may increase oncogene expression, interact with host cell proto-oncogenes, and escape detection by the immune system through the suppression of HLA molecule expression [225]. Acute stress can boost peripheral blood lymphocyte levels, particularly NK cells. Short-term stress is linked to changes in cytokine secretion and a shift in immunity response from Th1 toward Th2. The decline in NK cell activity and lymph proliferation shifts the immune response toward Th2 and decreases secretion of IFN-γ and IL-2, resulting in weakened cellular immunity in chronic stress, potentially increasing the risk of infections and cancer [226]. Elevated levels of psychological stress have been accompanying to a compromised immunity response to HPV in women who have dysplasia of the cervix. This diminished response appears to be related to a weakened functionality of T-cells when confronting HPV16. This may occur due to alterations induced by stress in the production of Th1/Th2 cytokines, ultimately fostering viral persistence and facilitating the progression of cancer [227]. Similarly, in a research study, 426 men underwent semi-annual HPV testing, while their stress levels were classified as either high or low based on the Perceived Stress Scale (PSS-4). The prevalence of HR-HPV infection is 1.53 times higher [PR = 1.53 (95% CI: 1.06–2.20)] in the group with high-life stress events. Additionally, high stress is linked to a lower rate of clearing HPV infections in men over 50 years of age [228].

A cohort study conducted on 4.6 million individuals revealed that psychological stress resulting from the loss of a child was connected to an increased risk of HPV-related cancers, including cervical cancer (RR: 1.46; 95% CI: 1.17–1.80) [229]. Kuebler et al. investigated the links between chronic stress, diurnal cortisol patterns, and HPV infection at the beginning and after a one-year follow-up. Higher chronic stress, specifically from work pressure and anxiety, along with a higher cortisol awakening response (CAR), were associated with HR-HPV positivity at the beginning of the test [230]. Additionally, glucocorticoids induce HPV-E6 expression and reduce miR-145 in cervical cancer cells, resulting in p53 dysfunction and promoting chemotherapy resistance. MiR-145 enhances p53's tumor-suppressing effects and inhibits cancer cell motility [231]. In a study involving 1,696 women who had experienced the loss of a family member, this stress was correlated with a 62 percent higher likelihood of contracting HPV-16 infection, along with a heightened viral load and the occurrence of repeat HPV infections. Psychological stress was also linked to a 4–9% heightened risk of initial abnormal cervix cytology [232]. Stressful life also plays a role in the progression of HPV viral infections towards carcinogenesis. A study's findings revealed that women co-infected with HR-HPV and HIV, who also experienced 6 months of stress, had a seven-fold increased risk of developing persistent or progressive squamous intraepithelial lesions (SIL) in the cervix a year later [233]. Additionally, exposure to marital life stress was linked to the occurrence of SIL in the cervix, especially in the presence of HR-HPV [234]. The likelihood of developing cervical cancer was higher in women with high-stress occupations. Apart from the influence of life stress, conditions like depression and bipolar disorder are also associated with cervix cancer in a cohort study [235].

In summary, lifestyle choices like diet, alcohol intake, smoking, and psychological stress, as well as genetic and epigenetic changes and viral oncoproteins, can affect OS, influencing the development of HPV-related cancers (Fig. 3). Understanding and controlling OS is crucial for preventing and treating cancer. This study proposes investigating dietary antioxidants and targeting cancer cells using reactive oxygen species mechanisms as potential therapeutic strategies to reduce the impact of OS on HPV-linked malignancies.

Fig. 3
figure 3

Cervical cancer and oxidative stress factors

Conclusion

HPV infection is a significant risk factor for cancer, but it operates in conjunction with other complex mechanisms, most notably OS. OS arises from various sources, including lifestyle choices like smoking and alcohol consumption and even psychological stress. OS plays a crucial role in DNA damage and tumorigenesis in HPV-related cancers. HPV promotes carcinogenesis through OS by various mechanisms, such as altering mitochondrial cristae, raising ROS levels and DNA damage by lowering the SOD 2 and GPx 1/2 antioxidant, activating the EGFR/MEK/ERK cell signaling pathway, and upregulating the OS sensor Pirin, and also causing LDHA translocation due to elevated ROS caused by HPV oncoprotein and subsequent methylation of histone H3K79. Furthermore, HPV-infected cells adapt to OS situations by inhibiting OS-induced apoptosis and modulating antioxidant activity via its oncogenes, which bypasses DNA damage response pathways. On a positive note, dietary antioxidants such as vitamins offer a potential defense against HPV infection by neutralizing free radicals and safeguarding DNA. Understanding the relationship between OS, HPV, and cancer is essential for developing strategies to prevent and treat HPV-related malignancies. Current HPV-related cancer treatments involve surgery and chemotherapy, but ongoing research may yield more effective strategies targeting OS for improved outcomes.

Availability of data and materials

Not applicable.

References

  1. de Sanjose S, Brotons M, Pavon MA. The natural history of human papillomavirus infection. Best Pract Res Clin Obstet Gynaecol. 2018;47:2–13.

    Article  PubMed  Google Scholar 

  2. De Martel C, Plummer M, Vignat J, Franceschi S. Worldwide burden of cancer attributable to HPV by site, country and HPV type. Int J Cancer. 2017;141(4):664–70.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Ren S, Gaykalova DA, Guo T, Favorov AV, Fertig EJ, Tamayo P, et al. HPV E2, E4, E5 drive alternative carcinogenic pathways in HPV positive cancers. Oncogene. 2020;39(40):6327–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sallam M, Al-Mahzoum K, Eid H, Assaf AM, Abdaljaleel M, Al-Abbadi M, et al. Attitude towards HPV vaccination and the intention to get vaccinated among female university students in health schools in Jordan. Vaccines. 2021;9(12):1432.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Saraiya M, Unger ER, Thompson TD, Lynch CF, Hernandez BY, Lyu CW, et al. US assessment of HPV types in cancers: implications for current and 9-valent HPV vaccines. J Natl Cancer Inst. 2015;107(6):1086.

    Article  Google Scholar 

  6. World Health Organization. health-topics/cervical-cancer: World Health Organization; 2023 https://www.who.int/health-topics/cervical-cancer#tab=tab_1.

  7. Diori Karidio I, Sanlier SH. Reviewing cancer’s biology: an eclectic approach. J Egypt Natl Canc Inst. 2021;33:1–17.

    Article  Google Scholar 

  8. Alpay M, Backman LR, Cheng X, Dukel M, Kim W-J, Ai L, et al. Oxidative stress shapes breast cancer phenotype through chronic activation of ATM-dependent signaling. Breast Cancer Res Treat. 2015;151:75–87.

    Article  CAS  PubMed  Google Scholar 

  9. Veskoukis AS, Tsatsakis AM, Kouretas D. Dietary oxidative stress and antioxidant defense with an emphasis on plant extract administration. Cell Stress Chaperones. 2012;17:11–21.

    Article  CAS  PubMed  Google Scholar 

  10. Forcados GE, James DB, Sallau AB, Muhammad A, Mabeta P. Oxidative stress and carcinogenesis: potential of phytochemicals in breast cancer therapy. Nutr Cancer. 2017;69(3):365–74.

    Article  CAS  PubMed  Google Scholar 

  11. Klaunig JE. Oxidative stress and cancer. Curr Pharm Des. 2018;24(40):4771–8.

    Article  CAS  PubMed  Google Scholar 

  12. Williams VM, Filippova M, Filippov V, Payne KJ, Duerksen-Hughes P. Human papillomavirus type 16 E6* induces oxidative stress and DNA damage. J Virol. 2014;88(12):6751–61.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Wongworawat YC, Filippova M, Williams VM, Filippov V, Duerksen-Hughes PJ. Chronic oxidative stress increases the integration frequency of foreign DNA and human papillomavirus 16 in human keratinocytes. Am J Cancer Res. 2016;6(4):764.

    CAS  Google Scholar 

  14. Chow LT, Broker TR. Human papillomavirus infections: warts or cancer? Cold Spring Harb Perspect Biol. 2013;5(7):a012997.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Nicolae I, Tampa M, Mitran C, Ene CD, Mitran M, Matei C, et al. Gamma-glutamyl transpeptidase alteration as a biomarker of oxidative stress in patients with human papillomavirus lesions following topical treatment with sinecatechins. Farmacia. 2017;65(4):617–23.

  16. Baedyananda F, Sasivimolrattana T, Chaiwongkot A, Varadarajan S, Bhattarakosol P. Role of HPV16 E1 in cervical carcinogenesis. Front Cell Infect Microbiol. 2022;12: 955847.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nelson CW, Mirabello L. Human papillomavirus genomics: Understanding carcinogenicity. Tumour Virus Res. 2023;15: 200258.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hu Z, Ma D. The precision prevention and therapy of HPV-related cervical cancer: new concepts and clinical implications. Cancer Med. 2018;7(10):5217–36.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Doorbar J, Quint W, Banks L, Bravo IG, Stoler M, Broker TR, et al. The biology and life-cycle of human papillomaviruses. Vaccine. 2012;30:F55–70.

    Article  CAS  PubMed  Google Scholar 

  20. Munger K, Gwin TK, McLaughlin-Drubin ME. p16 in HPV-associated cancers. Oncotarget. 2013;4(11):1864–5.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Sano D, Oridate N. The molecular mechanism of human papillomavirus-induced carcinogenesis in head and neck squamous cell carcinoma. Int J Clin Oncol. 2016;21:819–26.

    Article  CAS  PubMed  Google Scholar 

  22. Duensing S, Münger K. Mechanisms of genomic instability in human cancer: insights from studies with human papillomavirus oncoproteins. Int J Cancer. 2004;109(2):157–62.

    Article  CAS  PubMed  Google Scholar 

  23. Mantovani F, Banks L. The human papillomavirus E6 protein and its contribution to malignant progression. Oncogene. 2001;20(54):7874–87.

    Article  CAS  PubMed  Google Scholar 

  24. Katzenellenbogen RA. Activation of telomerase by HPVs. Virus Res. 2017;231:50–5.

    Article  CAS  PubMed  Google Scholar 

  25. Tomaić V. Functional roles of E6 and E7 oncoproteins in HPV-induced malignancies at diverse anatomical sites. Cancers. 2016;8(10):95.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Pützer B. E2F1 death pathways as targets for cancer therapy. J Cell Mol Med. 2007;11(2):239–51.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Korzeniewski N, Treat B, Duensing S. The HPV-16 E7 oncoprotein induces centriole multiplication through deregulation of Polo-like kinase 4 expression. Mol Cancer. 2011;10:1–5.

    Article  Google Scholar 

  28. Sen P, Ganguly P, Ganguly N. Modulation of DNA methylation by human papillomavirus E6 and E7 oncoproteins in cervical cancer. Oncol Lett. 2018;15(1):11–22.

    PubMed  Google Scholar 

  29. Chen B, Zhao L, Yang R, Xu T. Advances in molecular mechanism of HPV16 E5 oncoprotein carcinogenesis. Arch Biochem Biophys. 2023;745:109716.

  30. Przybylski M, Pruski D, Millert-Kalińska S, Krzyżaniak M, de Mezer M, Frydrychowicz M, et al. Expression of E4 protein and HPV Major Capsid Protein (L1) as a novel combination in squamous intraepithelial lesions. Biomedicines. 2023;11(1):225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sullivan LB, Chandel NS. Mitochondrial reactive oxygen species and cancer. Cancer Metabol. 2014;2:1–12.

    Article  Google Scholar 

  32. Tafani M, Sansone L, Limana F, Arcangeli T, De Santis E, Polese M, et al. The interplay of reactive oxygen species, hypoxia, inflammation, and sirtuins in cancer initiation and progression. Oxidat Med Cell Longev. 2016;2016.

  33. Aranda-Rivera AK, Cruz-Gregorio A, Arancibia-Hernández YL, Hernández-Cruz EY, Pedraza-Chaverri J. RONS and oxidative stress: an overview of basic concepts. Oxygen. 2022;2(4):437–78.

    Article  CAS  Google Scholar 

  34. Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O. Oxidative stress and antioxidant defense. World Allergy Organ J. 2012;5:9–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Persson T, Popescu BO, Cedazo-Minguez A. Oxidative stress in Alzheimer’s disease: why did antioxidant therapy fail? Oxidative Med Cell Longev. 2014;2014:427318.

  36. Choudhari SK, Chaudhary M, Gadbail AR, Sharma A, Tekade S. Oxidative and antioxidative mechanisms in oral cancer and precancer: a review. Oral Oncol. 2014;50(1):10–8.

    Article  CAS  PubMed  Google Scholar 

  37. Katakwar P, Metgud R, Naik S, Mittal R. Oxidative stress marker in oral cancer: a review. J Cancer Res Ther. 2016;12(2):438–46.

    Article  CAS  PubMed  Google Scholar 

  38. Trueba GP, Sánchez GM, Giuliani A. Oxygen free radical and antioxidant defense mechanism in cancer. Front Biosci-Landmark. 2004;9(3):2029–44.

    Article  Google Scholar 

  39. Kulbacka J, Saczko J, Chwiłkowska A. Oxidative stress in cells damage processes. Polski merkuriusz lekarski: organ Polskiego Towarzystwa Lekarskiego. 2009;27(157):44–7.

    CAS  PubMed  Google Scholar 

  40. Williams VM, Filippova M, Soto U, Duerksen-Hughes PJ. HPV-DNA integration and carcinogenesis: putative roles for inflammation and oxidative stress. Futur Virol. 2011;6(1):45–57.

    Article  Google Scholar 

  41. Cruz-Gregorio A, Aranda-Rivera AK, Pedraza-Chaverri J. Human papillomavirus-related cancers and mitochondria. Virus Res. 2020;286: 198016.

    Article  CAS  PubMed  Google Scholar 

  42. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cruz-Gregorio A, Manzo-Merino J, Gonzaléz-García 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gao L, Gu P, Zhao W, Ding W, Zhao X, Guo S, et al. The role of globular heads of the C1q receptor in HPV 16 E2-induced human cervical squamous carcinoma cell apoptosis is associated with p38 MAPK/JNK activation. J Trans Med. 2013;11(1):1–11.

    Article  Google Scholar 

  45. Villota C, Campos A, Vidaurre S, Oliveira-Cruz L, Boccardo E, Burzio VA, et al. Expression of mitochondrial non-coding RNAs (ncRNAs) is modulated by high risk human papillomavirus (HPV) oncogenes. J Biol Chem. 2012;287(25):21303–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Villota C, Varas-Godoy M, Jeldes E, Campos A, Villegas J, Borgna V, et al. HPV-18 E2 protein downregulates antisense noncoding mitochondrial RNA-2, delaying replicative senescence of human keratinocytes. Aging (Albany NY). 2019;11(1):33.

    Article  CAS  Google Scholar 

  47. Cruz-Gregorio A, Martínez-Ramírez I, Pedraza-Chaverri J, Lizano M. Reprogramming of energy metabolism in response to radiotherapy in head and neck squamous cell carcinoma. Cancers. 2019;11(2):182.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Evans W, Filippova M, Filippov V, Bashkirova S, Zhang G, Reeves ME, et al. Overexpression of HPV16 E6* alters β-integrin and mitochondrial dysfunction pathways in cervical cancer cells. Cancer Genomics Proteom. 2016;13(4):259–73.

    CAS  Google Scholar 

  49. Seo M-D, Kang TJ, Lee CH, Lee A-Y, Noh M. HaCaT keratinocytes and primary epidermal keratinocytes have different transcriptional profiles of cornified envelope-associated genes to T helper cell cytokines. Biomol Therapeut. 2012;20(2):171.

    Article  CAS  Google Scholar 

  50. Shim J-H, Kim K-H, Cho Y-S, Choi H-S, Song EY, Myung P-K, et al. Protective effect of oxidative stress in HaCaT keratinocytes expressing E7 oncogene. Amino Acids. 2008;34:135–41.

    Article  CAS  PubMed  Google Scholar 

  51. Marullo R, Werner E, Zhang H, Chen GZ, Shin DM, Doetsch PW. HPV16 E6 and E7 proteins induce a chronic oxidative stress response via NOX2 that causes genomic instability and increased susceptibility to DNA damage in head and neck cancer cells. Carcinogenesis. 2015;36(11):1397–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Narisawa-Saito M, Kiyono T. Basic mechanisms of high-risk human papillomavirus-induced carcinogenesis: roles of E6 and E7 proteins. Cancer Sci. 2007;98(10):1505–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cruz-Gregorio A, Aranda-Rivera AK. Redox-sensitive signalling pathways regulated by human papillomavirus in HPV-related cancers. Rev Med Virol. 2021;31(6): e2230.

    Article  CAS  PubMed  Google Scholar 

  54. Godinho S, Pellman D. Causes and consequences of centrosome abnormalities in cancer. Philos Trans R Soc B: Biol Sci. 2014;369(1650):20130467.

    Article  Google Scholar 

  55. Hannoodee S, Nasuruddin DN. Acute inflammatory response. 2020.

  56. Sameer AS, Nissar S. Toll-like receptors (TLRs): structure, functions, signaling, and role of their polymorphisms in colorectal cancer susceptibility. BioMed Res Int. 2021;2021:1157023.

  57. Behzadi P, García-Perdomo HA, Karpiński TM. Toll-like receptors: general molecular and structural biology. J Immunol Res. 2021;2021:1–21.

    Article  Google Scholar 

  58. Kavathas PB, Krause PJ, Ruddle NH. Innate immunity: recognition and effector functions. Immunoepidemiology. 2019:39–53.

  59. Fekete T, Ágics B, Bencze D, Bene K, Szántó A, Tarr T, et al. Regulation of RLR-mediated antiviral responses of human dendritic cells by mTOR. Front Immunol. 2020;11: 572960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ahmed D, Al-Daraawi M, Cassol E. Innate sensing and cellular metabolism: role in fine tuning antiviral immune responses. J Leukoc Biol. 2023;113(2):164–90.

    Article  PubMed  Google Scholar 

  61. Zhang T, Ma C, Zhang Z, Zhang H, Hu H. NF-κB signaling in inflammation and cancer. MedComm. 2021;2(4):618–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Noh J-Y, Yoon SR, Kim T-D, Choi I, Jung H. Toll-like receptors in natural killer cells and their application for immunotherapy. J Immunol Res. 2020;2020:2045860.

  63. Ghezzi P, Floridi L, Boraschi D, Cuadrado A, Manda G, Levic S, et al. Oxidative stress and inflammation induced by environmental and psychological stressors: a biomarker perspective. Antioxid Redox Signal. 2018;28(9):852–72.

    Article  CAS  PubMed  Google Scholar 

  64. Silva GÁF, Nunes RAL, Morale MG, Boccardo E, Aguayo F, Termini L. Oxidative stress: therapeutic approaches for cervical cancer treatment. Clinics (Sao Paulo). 2018;73(suppl 1):e548s.

    Article  PubMed  Google Scholar 

  65. Fishbein A, Hammock BD, Serhan CN, Panigrahy D. Carcinogenesis: failure of resolution of inflammation? Pharmacol Ther. 2021;218: 107670.

    Article  CAS  PubMed  Google Scholar 

  66. Payazdan M, Khatami S, Galehdari H, Delfan N, Shafiei M, Heydaran S. The anti-inflammatory effects of sialic acid on the human glia cells by the upregulation of IL-4 and IL-10 genes’ expressions. Gene Reports. 2021;24: 101218.

    Article  CAS  Google Scholar 

  67. Hemmat N, Bannazadeh BH. Association of human papillomavirus infection and inflammation in cervical cancer. Pathogens Disease. 2019;77(5):ftz048.

    Article  CAS  PubMed  Google Scholar 

  68. Alhamlan FS, Alfageeh MB, Al Mushait MA, Al-Badawi IA, Al-Ahdal MN. Human papillomavirus-associated cancers. Microbial Pathogenesis: Infection and Immunity. 2021:1–14.

  69. Gorvel L, Olive D, editors. Tumor associated macrophage in HPV+ tumors: between immunosuppression and inflammation. Seminars in Immunology; 2023: Elsevier.

  70. Lukhele S, Boukhaled GM, Brooks DG, editors. Type I interferon signaling, regulation and gene stimulation in chronic virus infection. Seminars in Immunology; 2019: Elsevier.

  71. Ferreira AR, Ramalho AC, Marques M, Ribeiro D. The interplay between antiviral signalling and carcinogenesis in human papillomavirus infections. Cancers. 2020;12(3):646.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Jee B, Yadav R, Pankaj S, Shahi SK. Immunology of HPV-mediated cervical cancer: current understanding. Int Rev Immunol. 2021;40(5):359–78.

    Article  CAS  PubMed  Google Scholar 

  73. Moody CA. Regulation of the innate immune response during the human papillomavirus life cycle. Viruses. 2022;14(8):1797.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Nowicka H. The role of phosphorylation of ISGF3 components in the regulation of ISG expression and viral protection: institute of molecular biology; 2022.

  75. Bussey KA, Brinkmann MM. Strategies for immune evasion by human tumor viruses. Curr Opin Virol. 2018;32:30–9.

    Article  CAS  PubMed  Google Scholar 

  76. Wakabayashi R, Nakahama Y, Nguyen V, Espinoza JL. The host-microbe interplay in human papillomavirus-induced carcinogenesis. Microorganisms. 2019;7(7):199.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Fernandez-Avila L, Castro-Amaya AM, Molina-Pineda A, Hernández-Gutiérrez R, Jave-Suarez LF, Aguilar-Lemarroy A. The value of CXCL1, CXCL2, CXCL3, and CXCL8 as potential prognosis markers in cervical cancer: evidence of E6/E7 from HPV16 and 18 in chemokines regulation. Biomedicines. 2023;11(10):2655.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Mukherjee AG, Ramesh Wanjari U, Valsala Gopalakrishnan A, Jayaraj R, Katturajan R, Kannampuzha S, et al. HPV-associated cancers: insights into the mechanistic scenario and latest updates. Med Oncol. 2023;40(8):212.

    Article  CAS  PubMed  Google Scholar 

  79. Pérez-Soto E, Fernández-Martínez E, Oros-Pantoja R, Medel-Flores O, Miranda-Covarrubias JC, Sánchez-Monroy V. Proinflammatory and oxidative stress states induced by human papillomavirus and chlamydia trachomatis coinfection affect sperm quality in asymptomatic infertile men. Medicina. 2021;57(9):862.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Pérez-Soto E, Medel-Flores MO, Fernández-Martínez E, Oros-Pantoja R, Miranda-Covarrubias JC, Sánchez-Monroy V. High-risk HPV with multiple infections promotes CYP2E1, lipoperoxidation and pro-inflammatory cytokines in semen of asymptomatic infertile men. Antioxidants. 2022;11(6):1051.

    Article  PubMed  PubMed Central  Google Scholar 

  81. 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.

    Article  PubMed  Google Scholar 

  82. Hochmann J, Parietti F, Martínez J, Lopez AC, Carreño M, Quijano C, et al. Human papillomavirus type 18 E5 oncoprotein cooperates with E6 and E7 in promoting cell viability and invasion and in modulating the cellular redox state. Mem Inst Oswaldo Cruz. 2020;115: e190405.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Aguayo F, Perez-Dominguez F, Osorio JC, Oliva C, Calaf GM. PI3K/AKT/mTOR signaling pathway in HPV-driven head and neck carcinogenesis: therapeutic implications. Biology. 2023;12(5):672.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Ghafouri-Fard S, Noie Alamdari A, Noee Alamdari Y, Abak A, Hussen BM, Taheri M, et al. Role of PI3K/AKT pathway in squamous cell carcinoma with an especial focus on head and neck cancers. Cancer Cell Int. 2022;22(1):1–27.

    Article  Google Scholar 

  85. Perez-Dominguez F, Carrillo-Beltrán D, Blanco R, Muñoz JP, León-Cruz G, Corvalan AH, et al. Role of pirin, an oxidative stress sensor protein, in epithelial carcinogenesis. Biology. 2021;10(2):116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Soleimani A, Rahmani F, Ferns GA, Ryzhikov M, Avan A, Hassanian SM. Role of the NF-κB signaling pathway in the pathogenesis of colorectal cancer. Gene. 2020;726: 144132.

    Article  CAS  PubMed  Google Scholar 

  87. Bossler F, Hoppe-Seyler K, Hoppe-Seyler F. PI3K/AKT/mTOR signaling regulates the virus/host cell crosstalk in HPV-positive cervical cancer cells. Int J Mol Sci. 2019;20(9):2188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Baumeister P, Zhou J, Canis M, Gires O. Epithelial-to-mesenchymal transition-derived heterogeneity in head and neck squamous cell carcinomas. Cancers. 2021;13(21):5355.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Ling Z, Cheng B, Tao X. Epithelial-to-mesenchymal transition in oral squamous cell carcinoma: challenges and opportunities. Int J Cancer. 2021;148(7):1548–61.

    Article  CAS  PubMed  Google Scholar 

  90. Chen H-F, Chuang H-C, Tan T-H. Regulation of dual-specificity phosphatase (DUSP) ubiquitination and protein stability. Int J Mol Sci. 2019;20(11):2668.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Rahmani F, Hashemian P, Tabrizi AT, Ghorbani Z, Ziaeemehr A, Alijannejad S, et al. Regulatory role of miRNAs on Wnt/β-catenin signaling in tumorigenesis of glioblastoma. Indian J Cancer. 2023;60(3):295–302.

  92. Zhu G-X, Gao D, Shao Z-Z, Chen L, Ding W-J, Yu Q-F. Wnt/β-catenin signaling: causes and treatment targets of drug resistance in colorectal cancer. Mol Med Rep. 2021;23(2):1.

    Google Scholar 

  93. Rahmani F, Safavi P, Fathollahpour A, Sabz FTK, Tajzadeh P, Arefnezhad M, et al. The interplay between non-coding RNAs and Wnt/ß-catenin signaling pathway in urinary tract cancers: from tumorigenesis to metastasis. EXCLI J. 2022;21:1273.

    PubMed  PubMed Central  Google Scholar 

  94. Scarth JA, Patterson MR, Morgan EL, Macdonald A. The human papillomavirus oncoproteins: a review of the host pathways targeted on the road to transformation. j General Virol. 2021;102(3):001540.

    Article  CAS  Google Scholar 

  95. Wang B, Li X, Liu L, Wang M. β-Catenin: oncogenic role and therapeutic target in cervical cancer. Biol Res. 2020;53:33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Chakraborti S, Karmakar A, Guha R, Ngan C, Das RK, Whitaker N. Induction of epithelial to mesenchymal transition in HPV16 E6/E7 oncogene transfected C33A cell line. Tissue Cell. 2023;82: 102041.

    Article  CAS  PubMed  Google Scholar 

  97. Aghbash PS, Hemmat N, Baradaran B, Mokhtarzadeh A, Poortahmasebi V, Oskuee MA, et al. The effect of Wnt/β-catenin signaling on PD-1/PDL-1 axis in HPV-related cervical cancer. Oncol Res. 2022;30(3):99.

    Article  PubMed  Google Scholar 

  98. De Marco F. Oxidative stress and HPV carcinogenesis. Viruses. 2013;5(2):708–31.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Visalli G, Riso R, Facciolà A, Mondello P, Caruso C, Picerno I, et al. Higher levels of oxidative DNA damage in cervical cells are correlated with the grade of dysplasia and HPV infection. J Med Virol. 2016;88(2):336–44.

    Article  CAS  PubMed  Google Scholar 

  100. Cadet J, Wagner JR. DNA base damage by reactive oxygen species, oxidizing agents, and UV radiation. Cold Spring Harb Perspect Biol. 2013;5(2): a012559.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Voulgaridou G-P, Anestopoulos I, Franco R, Panayiotidis MI, Pappa A. DNA damage induced by endogenous aldehydes: current state of knowledge. Mutat Res Fundam Mol Mech Mutagen. 2011;711(1–2):13–27.

    Article  CAS  Google Scholar 

  102. Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature. 2009;461(7267):1071–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Giglia-Mari G, Zotter A, Vermeulen W. DNA damage response. Cold Spring Harb Perspect Biol. 2011;3(1): a000745.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Georgescu SR, Mitran CI, Mitran MI, Caruntu C, Sarbu MI, Matei C, et al. New insights in the pathogenesis of HPV infection and the associated carcinogenic processes: the role of chronic inflammation and oxidative stress. J Immunol Res. 2018;2018:5315816.

  105. Massimi P, Shai A, Lambert P, Banks L. HPV E6 degradation of p53 and PDZ containing substrates in an E6AP null background. Oncogene. 2008;27(12):1800–4.

    Article  CAS  PubMed  Google Scholar 

  106. Achanta G, Huang P. Role of p53 in sensing oxidative DNA damage in response to reactive oxygen species-generating agents. Can Res. 2004;64(17):6233–9.

    Article  CAS  Google Scholar 

  107. Chen Y, Williams V, Filippova M, Filippov V, Duerksen-Hughes P. Viral carcinogenesis: factors inducing DNA damage and virus integration. Cancers. 2014;6(4):2155–86.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Spardy N, Covella K, Cha E, Hoskins EE, Wells SI, Duensing A, et al. Human papillomavirus 16 E7 oncoprotein attenuates DNA damage checkpoint control by increasing the proteolytic turnover of claspin. Can Res. 2009;69(17):7022–9.

    Article  CAS  Google Scholar 

  109. Liu Q, Lopez K, Murnane J, Humphrey T, Barcellos-Hoff MH. Misrepair in context: TGFβ regulation of DNA repair. Front Oncol. 2019;9:799.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Senapati R, Senapati NN, Dwibedi B. Molecular mechanisms of HPV mediated neoplastic progression. Infect Agent Cancer. 2016;11:59.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Basukala O, Banks L. The not-so-good, the bad and the ugly: HPV E5, E6 and E7 oncoproteins in the orchestration of carcinogenesis. Viruses. 2021;13(10):1892.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Ramesh PS, Bovilla VR, Swamy V, Manoli NN, Dasegowda K, Siddegowda S, et al. NRF2 signaling is repressed in HPV-driven head and neck cancer and correlates with better prognosis: HPV-E6/E7 confers chemosensitivity by inhibiting NRF2. 2022.

  113. Thomas M, Pim D, Banks L. The role of the E6–p53 interaction in the molecular pathogenesis of HPV. Oncogene. 1999;18(53):7690–700.

    Article  CAS  PubMed  Google Scholar 

  114. Xu M, Katzenellenbogen RA, Grandori C, Galloway DA. NFX1 plays a role in human papillomavirus type 16 E6 activation of NFκB activity. J Virol. 2010;84(21):11461–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Boyer SN, Wazer DE, Band V. E7 protein of human papilloma virus-16 induces degradation of retinoblastoma protein through the ubiquitin-proteasome pathway. Can Res. 1996;56(20):4620–4.

    CAS  Google Scholar 

  116. Oh J-M, Kim S-H, Cho E-A, Song Y-S, Kim W-H, Juhnn Y-S. Human papillomavirus type 16 E5 protein inhibits hydrogen peroxide-induced apoptosis by stimulating ubiquitin–proteasome-mediated degradation of Bax in human cervical cancer cells. Carcinogenesis. 2010;31(3):402–10.

    Article  CAS  PubMed  Google Scholar 

  117. Hamilton JP. Epigenetics: principles and practice. Dig Dis. 2011;29(2):130–5.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Guo D, Yang M, Li S, Zhu W, Chen M, Pan J, et al. Expression and molecular regulation of non-coding RNAs in HPV-positive head and neck squamous cell carcinoma. Front Oncol. 2023;13:1122982.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Ghiani L, Chiocca S. High risk-human papillomavirus in HNSCC: present and future challenges for epigenetic therapies. Int J Mol Sci. 2022;23(7):3483.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Di Domenico M, Giovane G, Kouidhi S, Iorio R, Romano M, De Francesco F, et al. HPV epigenetic mechanisms related to oropharyngeal and cervix cancers. Cancer Biol Ther. 2018;19(10):850–7.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Torres-Rojas FI, Alarcón-Romero LdC, Leyva-Vázquez MA, Ortiz-Ortiz J, Mendoza-Catalán MÁ, Hernández-Sotelo D, et al. Methylation of the L1 gene and integration of human papillomavirus 16 and 18 in cervical carcinoma and premalignant lesions. Oncol Lett. 2018;15(2):2278–86.

    PubMed  Google Scholar 

  122. Gautam D, Johnson BA, Mac M, Moody CA. SETD2-dependent H3K36me3 plays a critical role in epigenetic regulation of the HPV31 life cycle. PLoS Pathog. 2018;14(10): e1007367.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Castro-Oropeza R, Piña-Sánchez P. Epigenetic and transcriptomic regulation landscape in HPV+ cancers: biological and clinical implications. Front Genet. 2022;13: 886613.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Lourenço de Freitas N, Deberaldini MG, Gomes D, Pavan AR, Sousa Â, Dos Santos JL, et al. Histone deacetylase inhibitors as therapeutic interventions on cervical cancer induced by human papillomavirus. Front Cell Dev Biol. 2021;8: 592868.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Liu Y, Guo J-Z, Liu Y, Wang K, Ding W, Wang H, et al. Nuclear lactate dehydrogenase A senses ROS to produce α-hydroxybutyrate for HPV-induced cervical tumor growth. Nat Commun. 2018;9(1):4429.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Zhang L, Tian S, Pei M, Zhao M, Wang L, Jiang Y, et al. Crosstalk between histone modification and DNA methylation orchestrates the epigenetic regulation of the costimulatory factors, Tim-3 and galectin-9, in cervical cancer. Oncol Rep. 2019;42(6):2655–69.

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Gulati N, Béguelin W, Giulino-Roth L. Enhancer of zeste homolog 2 (EZH2) inhibitors. Leuk Lymphoma. 2018;59(7):1574–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Topchu I, Bychkov I, Makhov P, Izumchenko E, Karanicolas J, Yu J, et al. NSD1/2 histone methyltransferases regulate cell growth in HPV-negative head and neck squamous cell carcinoma (HNSCC). Cancer Res. 2023;83(7_supplement):4756.

    Article  Google Scholar 

  129. Sun Z, Zhang Y, Jia J, Fang Y, Tang Y, Wu H, et al. H3K36me3, message from chromatin to DNA damage repair. Cell Biosci. 2020;10(1):1–9.

    Article  CAS  Google Scholar 

  130. Lai Y, He Z, Zhang A, Yan Z, Zhang X, Hu S, et al. Tip60 and p300 function antagonistically in the epigenetic regulation of HPV18 E6/E7 genes in cervical cancer HeLa cells. Genes Genom. 2020;42:691–8.

    Article  CAS  Google Scholar 

  131. Kgatle MM, Spearman CW, Kalla AA, Hairwadzi HN. DNA oncogenic virus-induced oxidative stress, genomic damage, and aberrant epigenetic alterations. Oxidative Med Cell Longev. 2017;2017:3179421.

  132. Boscolo-Rizzo P, Furlan C, Lupato V, Polesel J, Fratta E. Novel insights into epigenetic drivers of oropharyngeal squamous cell carcinoma: role of HPV and lifestyle factors. Clin Epigenetics. 2017;9(1):1–19.

    Article  Google Scholar 

  133. Da Silva MLR, De Albuquerque BHDR, Allyrio TADMF, De Almeida VD, Cobucci RN, Bezerra FL, et al. The role of HPV-induced epigenetic changes in cervical carcinogenesis. Biomed Rep. 2021;15(1):1–20.

    Article  Google Scholar 

  134. Khan MA, Tiwari D, Dongre A, Sadaf, Mustafa S, Das CR, et al. Exploring the p53 connection of cervical cancer pathogenesis involving north-east Indian patients. PLoS ONE. 2020;15(9):10238500.

    Article  Google Scholar 

  135. Verlaat W, Snijders PJF, Novianti PW, Wilting SM, De Strooper LMA, Trooskens G, et al. Genome-wide DNA methylation profiling reveals methylation markers associated with 3q gain for detection of cervical precancer and cancer. Clin Cancer Res. 2017;23(14):3813–22.

    Article  CAS  PubMed  Google Scholar 

  136. Verlaat W, Snoek BC, Heideman DAM, Wilting SM, Snijders PJF, Novianti PW, et al. Identification and validation of a 3-gene methylation classifier for HPV-based cervical screening on self-samples. Clin Cancer Res. 2018;24(14):3456–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Van Keer S, van Splunter AP, Pattyn J, De Smet A, Herzog SA, Van Ostade X, et al. Triage of human papillomavirus infected women by methylation analysis in first-void urine. Sci Rep. 2021;11(1):7862.

    Article  PubMed  PubMed Central  Google Scholar 

  138. He J, Jiang B-H. Interplay between reactive oxygen species and microRNAs in cancer. Curr Pharmacol Rep. 2016;2(2):82–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Wang X, Huang X, Zhang Y. Involvement of human papillomaviruses in cervical cancer. Front Microbiol. 2018;9:2896.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Foppoli C, De Marco F, Cini C, Perluigi M. Redox control of viral carcinogenesis: The human papillomavirus paradigm. Biochimica et Biophysica Acta (BBA). 2015;1850(8):1622–32.

    Article  CAS  PubMed  Google Scholar 

  141. Cui J, Pan Y, Wang J, Liu Y, Wang H, Li H. MicroRNA-206 suppresses proliferation and predicts poor prognosis of HR-HPV-positive cervical cancer cells by targeting G6PD. Oncol Lett. 2018;16(5):5946–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Ju H, Lu Y, Wu Q, Liu J, Zeng Z, Mo H, et al. Disrupting G6PD-mediated Redox homeostasis enhances chemosensitivity in colorectal cancer. Oncogene. 2017;36(45):6282–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Caliri AW, Tommasi S, Besaratinia A. Relationships among smoking, oxidative stress, inflammation, macromolecular damage, and cancer. Mutat Res Rev Mutat Res. 2021;787: 108365.

    Article  CAS  PubMed  Google Scholar 

  144. Nagelhout G, Ebisch RM, Van Der Hel O, Meerkerk G-J, Magnée T, De Bruijn T, et al. Is smoking an independent risk factor for developing cervical intra-epithelial neoplasia and cervical cancer? A systematic review and meta-analysis. Expert Rev Anticancer Ther. 2021;21(7):781–94.

    Article  CAS  PubMed  Google Scholar 

  145. Krishnan AR, Zheng H, Kwok JG, Qu Y, Zou AE, Korrapati A, et al. A comprehensive study of smoking-specific microRNA alterations in head and neck squamous cell carcinoma. Oral Oncol. 2017;72:56–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Chatzistamatiou K, Moysiadis T, Vryzas D, Chatzaki E, Kaufmann AM, Koch I, et al. Cigarette smoking promotes infection of cervical cells by high-risk human papillomaviruses, but not subsequent E7 oncoprotein expression. Int J Mol Sci. 2018;19(2):422.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Ma K, Li S, Wu S, Zhu J, Yang Y. Impact of smoking exposure on human papillomavirus clearance among Chinese women: A follow-up propensity score matching study. Tobacco Induced Diseases. 2023;21:42.

  148. Aguayo F, Muñoz JP, Perez-Dominguez F, Carrillo-Beltrán D, Oliva C, Calaf GM, et al. High-risk human papillomavirus and tobacco smoke interactions in epithelial carcinogenesis. Cancers. 2020;12(8):2201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Banerjee NS, Moore D, Parker CJ, Broker TR, Chow LT. Targeting DNA damage response as a strategy to treat HPV infections. Int J Mol Sci. 2019;20(21):5455.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Moktar A, Ravoori S, Vadhanam MV, Gairola CG, Gupta RC. Cigarette smoke-induced DNA damage and repair detected by the comet assay in HPV-transformed cervical cells. Int J Oncol. 2009;35(6):1297–304.

    CAS  PubMed  Google Scholar 

  151. Carrillo-Beltrán D, Osorio JC, Blanco R, Oliva C, Boccardo E, Aguayo F. Interaction between cigarette smoke and human papillomavirus 16 E6/E7 oncoproteins to induce SOD2 expression and DNA damage in head and neck cancer. Int J Mol Sci. 2023;24(8):6907.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Ighodaro O, Akinloye O. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): their fundamental role in the entire antioxidant defence grid. Alexandria J Med. 2018;54(4):287–93.

    Article  Google Scholar 

  153. Lee DJ, Lee HM, Kim JH, Park I, Rho YS. Heavy alcohol drinking downregulates ALDH2 gene expression but heavy smoking up-regulates SOD2 gene expression in head and neck squamous cell carcinoma. World J Surg Oncol. 2017;15(1):1–8.

    Article  Google Scholar 

  154. Guo L, Wang T, Wu Y, Yuan Z, Dong J, An J, et al. WNT/β-catenin signaling regulates cigarette smoke-induced airway inflammation via the PPARδ/p38 pathway. Lab Invest. 2016;96(2):218–29.

    Article  CAS  PubMed  Google Scholar 

  155. Shishodia G, Toledo RRG, Rong X, Zimmerman E, Xiao AY, Harrison L, et al. 4NQO enhances differential activation of DNA repair proteins in HPV positive and HPV negative HNSCC cells. Oral Oncol. 2021;122: 105578.

    Article  CAS  PubMed  Google Scholar 

  156. Janaydeh M, Ismail A, Zulkifli SZ, Omar H. Toxic heavy metal (Pb and Cd) content in tobacco cigarette brands in Selangor state. Penins Malays Environ Monitor Assessm. 2019;191:1–8.

    CAS  Google Scholar 

  157. Banihani SA, Jaradat SA, Khader YS. Serum chromium level is increased in Jordanian smokers, decreased in Jordanians with prediabetes and type 2 diabetes, but not altered in Jordanians with hypertension, with obesity, or with family history of diabetes. Int J Prevent Med. 2019;10:145.

    Article  Google Scholar 

  158. Kashyap VK, Nagesh PK, Singh AK, Massey A, Darkwah GP, Yallapu MM, et al. Smoking and Drinking Activates NF-κB/IL-6 axis to promote inflammation during cervical carcinogenesis. 2023.

  159. Sarmiento-Salinas FL, Perez-Gonzalez A, Acosta-Casique A, Ix-Ballote A, Diaz A, Treviño S, et al. Reactive oxygen species: Role in carcinogenesis, cancer cell signaling and tumor progression. Life Sci. 2021;284: 119942.

    Article  CAS  PubMed  Google Scholar 

  160. Moktar A, Singh R, Vadhanam MV, Ravoori S, Lillard JW, Gairola CG, et al. Cigarette smoke condensate-induced oxidative DNA damage and its removal in human cervical cancer cells. Int J Oncol. 2011;39(4):941–7.

    CAS  PubMed  Google Scholar 

  161. Prokopczyk B, Sinha I, Trushin N, Freeman WM, El-Bayoumy K. Gene expression profiles in HPV-immortalized human cervical cells treated with the nicotine-derived carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Chem Biol Interact. 2009;177(3):173–80.

    Article  CAS  PubMed  Google Scholar 

  162. Zhuang Z, Li J, Sun G, Cui X, Zhang N, Zhao L, et al. Synergistic effect between human papillomavirus 18 and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone on malignant transformation of immortalized SHEE cells. Chem Res Toxicol. 2019;33(2):470–81.

    Article  Google Scholar 

  163. Utami TW, Kusuma F, Winarto H, Anggraeni TD, Peters AAW, Spaans V, et al. Tobacco use and its association with HPV infection in normal uterine cervix: a study from a sustainable development goals perspective. Tob Induc Dis. 2021;19:64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Wiley DJ, Wiesmeier E, Masongsong E, Gylys KH, Koutsky LA, Ferris DG, et al. Smokers at higher risk for undetected antibody for oncogenic human papillomavirus type 16 infection. Cancer Epidemiol Biomark Prev. 2006;15(5):915–20.

    Article  CAS  Google Scholar 

  165. Jiang L, Ma S, Zhang G, Jiang L, Yan L. Analysis of tobacco exposures and high-risk HPV infection in american women: national health and nutrition examination survey. Environ Sci Pollut Res. 2023;30(51):110489–98.

  166. Umutoni V, Schabath MB, Nyitray AG, Wilkin TJ, Villa LL, Lazcano-Ponce E, et al. The association between smoking and anal human papillomavirus in the HPV infection in men study. Cancer Epidemiol Biomarkers Prev. 2022;31(8):1546–53.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Schostag K, Lynch PT, Leavitt T, Sumer BD, Yang A, Shah A, et al. Smoking and other patient factors in HPV-mediated oropharynx cancer: a retrospective cohort study. Am J Otolaryngol. 2022;43(5): 103555.

    Article  PubMed  Google Scholar 

  168. Skoulakis A, Tsea M, Koltsidopoulos P, Lachanas V, Hajiioannou J, Petinaki E, et al. Do smoking and human papilloma virus have a synergistic role in the development of head and neck cancer? A systematic review and meta-analysis. J Buon. 2020;25(2):1107–15.

    PubMed  Google Scholar 

  169. Salaspuro M. Local acetaldehyde: its key role in alcohol-related oropharyngeal cancer. Visceral Med. 2020;36(3):167–74.

    Article  Google Scholar 

  170. Rumgay H, Shield K, Charvat H, Ferrari P, Sornpaisarn B, Obot I, et al. Global burden of cancer in 2020 attributable to alcohol consumption: a population-based study. Lancet Oncol. 2021;22(8):1071–80.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Gormley M, Dudding T, Sanderson E, Martin RM, Thomas S, Tyrrell J, et al. A multivariable Mendelian randomization analysis investigating smoking and alcohol consumption in oral and oropharyngeal cancer. Nat Commun. 2020;11(1):6071.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Radojicic J, Zaravinos A, Spandidos DA. HPV, KRAS mutations, alcohol consumption and tobacco smoking effects on esophageal squamous-cell carcinoma carcinogenesis. Int J Biol Markers. 2012;27(1):1–12.

    Article  CAS  PubMed  Google Scholar 

  173. Novoplansky O, Jagadeeshan S, Regev O, Menashe I, Elkabets M. Worldwide prevalence and clinical characteristics of RAS mutations in head and neck cancer: a systematic review and meta-analysis. Front Oncol. 2022;12: 838911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Wang H, Zhao Q, Zhang Y, Zhang Q, Zheng Z, Liu S, et al. Immunotherapy advances in locally advanced and recurrent/metastatic head and neck squamous cell carcinoma and its relationship with human papillomavirus. Front Immunol. 2021;12: 652054.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Kumar R, Rai AK, Das D, Das R, Kumar RS, Sarma A, et al. Alcohol and tobacco increases risk of high risk HPV infection in head and neck cancer patients: Study from North-East Region of India. PLoS ONE. 2015;10(10): e0140700.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Oh HY, Seo S-S, Kim MK, Lee DO, Chung YK, Lim MC, et al. Synergistic effect of viral load and alcohol consumption on the risk of persistent high-risk human papillomavirus infection. PLoS ONE. 2014;9(8): e104374.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Min K-J, Lee J-K, Lee S, Kim MK. Alcohol consumption and viral load are synergistically associated with CIN1. PLoS ONE. 2013;8(8): e72142.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Lai YH, Su CC, Wu SY, Hsueh WT, Wu YH, Chen HHW, et al. Impact of alcohol and smoking on outcomes of HPV-related oropharyngeal cancer. J Clin Med. 2022;11(21):6510.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Oh H, Kim M, Seo S, Lee D, Chung Y, Lim M, et al. Alcohol consumption and persistent infection of high-risk human papillomavirus. Epidemiol Infect. 2015;143(7):1442–50.

    Article  CAS  PubMed  Google Scholar 

  180. Ferraguti G, Terracina S, Petrella C, Greco A, Minni A, Lucarelli M, et al. Alcohol and head and neck cancer: updates on the role of oxidative stress, genetic, epigenetics, oral microbiota, antioxidants, and alkylating agents. Antioxidants. 2022;11(1):145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Orywal K, Jelski W, Zdrodowski M, Szmitkowski M. The diagnostic value of alcohol dehydrogenase isoenzymes and aldehyde dehydrogenase measurement in the sera of patients with endometrial cancer. Anticancer Res. 2013;33(9):3725–30.

    CAS  PubMed  Google Scholar 

  182. Teschke R. Microsomal ethanol-oxidizing system: success over 50 years and an encouraging future. Alcohol Clin Exp Res. 2019;43(3):386–400.

    Article  CAS  PubMed  Google Scholar 

  183. Chu J, Tong M, de la Monte SM. Chronic ethanol exposure causes mitochondrial dysfunction and oxidative stress in immature central nervous system neurons. Acta Neuropathol. 2007;113:659–73.

    Article  CAS  PubMed  Google Scholar 

  184. Schwartz JL, Munaretto A, Bagchi S, Crowe D, Izaguirre G. Inhibition of EGFR suppresses ethyl alcohol and tobacco cell effects on growth of human oral keratinocytes and human papillomavirus 16 entry as a function of furin. J Cancer Ther. 2014;6(01):90.

    Article  Google Scholar 

  185. Yu W, Zhang L, Wei Q, Shao A. O6-methylguanine-DNA methyltransferase (MGMT): challenges and new opportunities in glioma chemotherapy. Front Oncol. 2020;9:1547.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Jiang Y, Zhang T, Kusumanchi P, Han S, Yang Z, Liangpunsakul S. Alcohol metabolizing enzymes, microsomal ethanol oxidizing system, cytochrome P450 2E1, catalase, and aldehyde dehydrogenase in alcohol-associated liver disease. Biomedicines. 2020;8(3):50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Zhang H, Fu L. The role of ALDH2 in tumorigenesis and tumor progression: Targeting ALDH2 as a potential cancer treatment. Acta Pharmaceutica Sinica B. 2021;11(6):1400–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Gui S, Xie X, O’Neill WQ, Chatfield-Reed K, Yu J-G, Teknos TN, et al. p53 functional states are associated with distinct aldehyde dehydrogenase transcriptomic signatures. Sci Rep. 2020;10(1):1097.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Auguste A, Deloumeaux J, Joachim C, Gaete S, Michineau L, Herrmann-Storck C, et al. Joint effect of tobacco, alcohol, and oral HPV infection on head and neck cancer risk in the French West Indies. Cancer Med. 2020;9(18):6854–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Arif RT, Mogaddam MA, Merdad LA, Farsi NJ. Does human papillomavirus modify the risk of oropharyngeal cancer related to smoking and alcohol drinking? A systematic review and meta-analysis. Laryngoscope Invest Otolaryngol. 2022;7(5):1391–401.

    Article  Google Scholar 

  191. Koshiyama M. The effects of the dietary and nutrient intake on gynecologic cancers. Healthcare (Basel). 2019;7(3).

  192. Naidu MSK, Suryakar A, Swami SC, Katkam R, Kumbar K. Oxidative stress and antioxidant status in cervical cancer patients. Indian J Clin Biochem. 2007;22:140–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Zhou Q, Fan M, Wang Y, Ma Y, Si H, Dai G. Association between dietary Vitamin E intake and human papillomavirus infection among US adults: a cross-sectional study from national health and nutrition examination survey. Nutrients. 2023;15(17):3825.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Lin H-Y, Fu Q, Kao Y-H, Tseng T, Reiss K, Cameron JE, et al. Antioxidants Associated With Oncogenic Human Papillomavirus Infection in Women. J Infect Dis. 2021;224(9):1520–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Grandi G, Botticelli L, Fraia PD, Babalini C, Masini M, Unfer V. The association of four natural molecules—EGCG, folic acid, Vitamin B12, and HA—to counteract HPV cervical lesions: a case report. J Person Med. 2023;13(3):567.

    Article  Google Scholar 

  196. Chatterjee K, Mukherjee S, Vanmanen J, Banerjee P, Fata JE. Dietary polyphenols, resveratrol and pterostilbene exhibit antitumor activity on an HPV E6-positive cervical cancer model: an in vitro and in vivo analysis. Front Oncol. 2019;9:352.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Markowska A, Antoszczak M, Markowska J, Huczyński A. Role of vitamin E in selected malignant neoplasms in women. Nutr Cancer. 2022;74(4):1163–70.

    Article  CAS  PubMed  Google Scholar 

  198. Preci DP, Almeida A, Weiler AL, Franciosi MLM, Cardoso AM. Oxidative damage and antioxidants in cervical cancer. Int J Gynecol Cancer. 2021;31(2):265–71.

  199. García-Closas R, Castellsagué X, Bosch X, González CA. The role of diet and nutrition in cervical carcinogenesis: a review of recent evidence. Int J Cancer. 2005;117(4):629–37.

    Article  PubMed  Google Scholar 

  200. Letafati A, Sakhavarz T, Khosravinia MM, Ardekani OS, Sadeghifar S, Norouzi M, et al. Exploring the correlation between progression of human papillomavirus infection towards carcinogenesis and nutrition. Microbial Pathogenesis. 2023:106302.

  201. Zheng C, Zheng Z, Chen W. Association between serum vitamin C and HPV infection in American women: a cross-sectional study. BMC Womens Health. 2022;22(1):404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Barchitta M, Maugeri A, La-Mastra C, La-Rosa MC, Favara G, Magnano San Lio R, et al. Dietary antioxidant intake and human papillomavirus infection: evidence from a cross-sectional study in Italy. Nutrients. 2020;12(5):1384.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Piyathilake CJ, Henao OL, Macaluso M, Cornwell PE, Meleth S, Heimburger DC, et al. Folate is associated with the natural history of high-risk human papillomaviruses. Can Res. 2004;64(23):8788–93.

    Article  CAS  Google Scholar 

  204. Yokoyama M, Noguchi M, Nakao Y, Pater A, Iwasaka T. The tea polyphenol,(−)-epigallocatechin gallate effects on growth, apoptosis, and telomerase activity in cervical cell lines. Gynecol Oncol. 2004;92(1):197–204.

    Article  CAS  PubMed  Google Scholar 

  205. Yap JK, Kehoe ST, Woodman CB, Dawson CW. The major constituent of green tea, epigallocatechin-3-Gallate (EGCG), inhibits the growth of HPV18-infected keratinocytes by stimulating proteasomal turnover of the E6 and E7 oncoproteins. Pathogens. 2021;10(4):459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Song JY, Han JH, Song Y, Lee JH, Choi SY, Park YM. Epigallocatechin-3-gallate can prevent type 2 human papillomavirus E7 from suppressing interferon-stimulated genes. Int J Mol Sci. 2021;22(5):2418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Savini C, Yang R, Savelyeva L, Göckel-Krzikalla E, Hotz-Wagenblatt A, Westermann F, et al. Folate repletion after deficiency induces irreversible genomic and transcriptional changes in human papillomavirus type 16 (HPV16)-immortalized human keratinocytes. Int J Mol Sci. 2019;20(5):1100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Xiao S, Tang Y-S, Kusumanchi P, Stabler SP, Zhang Y, Antony AC. Folate deficiency facilitates genomic integration of human papillomavirus type 16 DNA in vivo in a novel mouse model for rapid oncogenic transformation of human keratinocytes. J Nutr. 2018;148(3):389–400.

    Article  PubMed  Google Scholar 

  209. Aragona C, Bezerra Espinola MS, Bilotta G, Porcaro G, Calcagno M. Evaluating the efficacy of pervistop®, a new combination based on EGCG, folic acid, Vitamin B12 and hyaluronic acid on patients with human papilloma virus (HPV) persistent infections and cervical lesions: a pilot study. J Clin Med. 2023;12(6):2171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Giuliano AR, Siegel EM, Roe DJ, Ferreira S, Luiza Baggio M, Galan L, et al. Dietary intake and risk of persistent human papillomavirus (HPV) infection: the Ludwig-McGill HPV Natural History Study. J Infect Dis. 2003;188(10):1508–16.

    Article  PubMed  Google Scholar 

  211. Cao D, Shen K, Li Z, Xu Y, Wu D. Association between vitamin C Intake and the risk of cervical neoplasia: a meta-analysis. Nutr Cancer. 2016;68(1):48–57.

    Article  CAS  PubMed  Google Scholar 

  212. Reddy VG, Khanna N, Singh N. Vitamin C augments chemotherapeutic response of cervical carcinoma HeLa cells by stabilizing P53. Biochem Biophys Res Commun. 2001;282(2):409–15.

    Article  CAS  PubMed  Google Scholar 

  213. Kim J, Kim MK, Lee JK, Kim J-H, Son SK, Song E-S, et al. Intakes of vitamin A, C, and E, and β-carotene are associated with risk of cervical cancer: a case-control study in Korea. Nutr Cancer. 2010;62(2):181–9.

    Article  PubMed  Google Scholar 

  214. Huang X, Chen C, Zhu F, Zhang Y, Feng Q, Li J, et al. Association between dietary vitamin A and HPV infection in American women: data from NHANES 2003–2016. BioMed Res Int 2020;2020.

  215. Piyathilake CJ, Macaluso M, Chambers MM, Badiga S, Siddiqui NR, Bell WC, et al. Folate and vitamin B12 may play a critical role in lowering the HPV 16 methylation–associated risk of developing higher grades of CIN. Cancer Prev Res. 2014;7(11):1128–37.

    Article  CAS  Google Scholar 

  216. Piyathilake CJ, Badiga S, Paul P, Vijayaraghavan K, Vedantham H, Sudula M, et al. Indian women with higher serum concentrations of folate and vitamin B12 are significantly less likely to be infected with carcinogenic or high-risk (HR) types of human papillomaviruses (HPVs). Int J Women's Health. 2010;2:7–12.

  217. Lopes RVC, Teixeira JA, Marchioni D, Villa LL, Giuliano AR, Luiza-Baggio M, et al. Dietary intake of selected nutrients and persistence of HPV infection in men. Int J Cancer. 2017;141(4):757–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Jiang Y, Xu S, Lan J, Zhang J, Chen T. Dietary Vitamin K Intake and HPV-infection status among American women: a secondary analysis from national health and nutrition examination survey data from 2003 to 2016. Int J Public Health. 2022;67:1604616.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Smith SM, Vale WW. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin Neurosci. 2022.

  220. Flaherty RL, Owen M, Fagan-Murphy A, Intabli H, Healy D, Patel A, et al. Glucocorticoids induce production of reactive oxygen species/reactive nitrogen species and DNA damage through an iNOS mediated pathway in breast cancer. Breast Cancer Res. 2017;19(1):35.

    Article  PubMed  PubMed Central  Google Scholar 

  221. Falcinelli M, Thaker PH, Lutgendorf SK, Conzen SD, Flaherty RL, Flint MS. The role of psychologic stress in cancer initiation: Clinical relevance and potential molecular mechanisms. Can Res. 2021;81(20):5131–40.

    Article  CAS  Google Scholar 

  222. Salim S. Oxidative stress and psychological disorders. Curr Neuropharmacol. 2014;12(2):140–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Wang L, Muxin G, Nishida H, Shirakawa C, Sato S, Konishi T. Psychological stress-induced oxidative stress as a model of sub-healthy condition and the effect of TCM. Eviden-Based Complem Altern Med. 2007;4:195–202.

    Article  Google Scholar 

  224. Lempesis IG, Georgakopoulou VE, Papalexis P, Chrousos GP, Spandidos DA. Role of stress in the pathogenesis of cancer. Int J Oncol. 2023;63(5):1–14.

    Article  Google Scholar 

  225. Lugović-Mihić L, Cvitanović H, Djaković I, Kuna M, Šešerko A. The influence of psychological stress on HPV infection manifestations and carcinogenesis. Cell Physiol Biochem. 2021;55(S2):71–88.

    Article  PubMed  Google Scholar 

  226. Cvitanović H, Milošević M, Bukvić-Bešlić I, Lugović-Mihić L. Determination of psychological stress, serum immune parameters, and cortisol levels in patients with human papilloma virus. Clin Ther. 2020;42(5):783–99.

    Article  PubMed  Google Scholar 

  227. Fang CY, Miller SM, Bovbjerg DH, Bergman C, Edelson MI, Rosenblum NG, et al. Perceived stress is associated with impaired T-cell response to HPV16 in women with cervical dysplasia. Ann Behav Med. 2008;35(1):87–96.

    Article  PubMed  Google Scholar 

  228. Kolar SK. Associations of perceived stress, sleep, and human papillomavirus in a prospective cohort of men: University of South Florida; 2013.

  229. Fang F, Fall K, Sparén P, Adami HO, Valdimarsdóttir HB, Lambe M, et al. Risk of infection-related cancers after the loss of a child: a follow-up study in Sweden. Cancer Res. 2011;71(1):116–22.

    Article  CAS  PubMed  Google Scholar 

  230. Kuebler U, Fischer S, Mernone L, Breymann C, Abbruzzese E, Ehlert U. Is stress related to the presence and persistence of oncogenic human papillomavirus infection in young women? BMC Cancer. 2021;21(1):419.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Shi M, Du L, Liu D, Qian L, Hu M, Yu M, et al. Glucocorticoid regulation of a novel HPV-E6-p53-miR-145 pathway modulates invasion and therapy resistance of cervical cancer cells. J Pathol. 2012;228(2):148–57.

    Article  CAS  PubMed  Google Scholar 

  232. Lu D, Sundström K, Sparén P, Fall K, Sjölander A, Dillner J, et al. Bereavement is associated with an increased risk of HPV infection and cervical cancer: an epidemiological study in Sweden. Cancer Res. 2016;76(3):643–51.

    Article  CAS  PubMed  Google Scholar 

  233. Pereira DB, Antoni MH, Danielson A, Simon T, Efantis-Potter J, Carver CS, et al. Life stress and cervical squamous intraepithelial lesions in women with human papillomavirus and human immunodeficiency virus. Psychosom Med. 2003;65(3):427–34.

    Article  PubMed  Google Scholar 

  234. Coker AL, Bond S, Madeleine MM, Luchok K, Pirisi L. Psychosocial stress and cervical neoplasia risk. Psychosom Med. 2003;65(4):644–51.

    Article  PubMed  Google Scholar 

  235. Wu CT, Chiu LT. The Impact of Psychological Distress on Cervical Cancer. Cancers (Basel). 2023;15(4):1100.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The figures include parts that were created using images from Servier Medical Art, which is licensed under a Creative Commons Attribution 3.0 Unported License and also www.freepik.com.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

A.L and Z.T: writing original draft, tables, investigation. M.M, A.A, S.C, N.Z: Investigation. L.S, S.M.J: Conceptualization, validation, Review and editing.

Corresponding authors

Correspondence to Luciano Saso or Seyed Mohammad Jazayeri.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent to participate

Not applicable.

Competing interests

Authors declare there is no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Letafati, A., Taghiabadi, Z., Zafarian, N. et al. Emerging paradigms: unmasking the role of oxidative stress in HPV-induced carcinogenesis. Infect Agents Cancer 19, 30 (2024). https://doi.org/10.1186/s13027-024-00581-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13027-024-00581-8

Keywords