![Open Access](/resources/images/iconopenaccess.png)
Role of APE1 in hepatocellular carcinoma and its prospects as a target in clinical settings (Review)
- Authors:
- Published online on: September 6, 2024 https://doi.org/10.3892/mco.2024.2780
- Article Number: 82
-
Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
1. Introduction
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related mortality measured in a total of 185 countries in 2020, following only colorectal cancer among digestive system tumors, and its fatality rate is increasing (1,2). A global review suggested that there are hundreds of thousands of new HCC cases annually, which demonstrates a progressive rise in the prevalence of this disease (3,4). Numerous factors can contribute to the development of HCC. Viral hepatitis B and C infection, alcohol abuse, non-alcoholic fatty liver disease and other metabolic liver disorders may induce mutations in hepatocytes, thereby precipitating the onset of liver cancer (5). Due to the asymptomatic or insidious onset of HCC, detection is often challenging, with the majority of patients being diagnosed at advanced stages of the disease (6). Currently, the treatment options for HCC are relatively limited, mainly including surgical resection, liver transplantation, radiofrequency ablation and arterial embolization (7-9). However, these treatment modalities each come with their own set of limitations, and due to the varying physical conditions of patients, there are also constraints on the selection of appropriate therapies (10). Therefore, targeted biological therapies for HCC have increasingly garnered attention from researchers (11). As a multi-kinase inhibitor, sorafenib is the first oral medication approved for the treatment of unresectable HCC (12,13). Previous studies have reported that sorafenib exerts inhibitory effects on the proliferation and migration of tumor cells (14,15). Additionally, it regulates vascular endothelial growth factors, suppresses tumor angiogenesis and promotes apoptosis in cancer cells by inhibiting related signaling pathways (16-18). However, sorafenib is associated with serious side effects and comes with a high cost, which limits its scope of application (19,20). Lenvatinib exerts its therapeutic effects on HCC by targeting multiple kinase receptors, such as VEGFR1-3, fibroblast growth factor receptor 1-4 and c-KIT, albeit with the limitation of potential resistance development due to various mechanisms, which include tumor microenvironment alterations and drug transport activation (21,22). Combination therapy with atezolizumab and bevacizumab is associated with limited improvements in survival and may lead to adverse reactions, including hypertension, cardiac dysfunction and thyroid function changes (23,24). Therefore, it is necessary to develop a more efficacious and targeted drug for the treatment of HCC.
The protein encoded by the human apurinic/apyrimidinic endonuclease 1 (APE1) gene is also referred to as APEX1, APE, APE1, APEX, APX, HAP1 and REF1. The APE1 protein possesses two distinct domains [DNA repair and reduction-oxidation (redox)], serving a crucial role in the DNA base excision repair (BER) pathway, which is closely associated with tumor cell proliferation (25). Additionally, APE1 is critically involved in the repair of DNA damage induced by a wide array of carcinogens, encompassing both those generated internally through cellular metabolism and externally through environmental exposure, thereby safeguarding the integrity of the genetic material and mitigating the risk of carcinogenesis (25,26). In the tumor microenvironment, APE1 may serve a role in inflammation and stromal cells, affecting tumor development and therapy efficacy due to its key role in regulating oxidative stress responses and inflammatory processes (27,28). Furthermore, APE1 is involved in the regulation of multiple transcription factors associated with cancer-related signaling pathways, including p53, NF-κB, activator protein 1 (AP-1), paired box (Pax)-5, Pax-8, hypoxia-inducible factor 1 (HIF-1), cAMP response element binding protein, activating transcription factor and HIF-1α. Previous studies have reported that, following the malignant transformation of tissues, the expression levels of APE1 are elevated in lung cancer (29), prostate cancer (30), breast cancer (31), HCC (32) and pancreatic cancer (33), which suggests that APE1 is a tumor-associated factor.
In normal hepatocytes, APE1 is expressed at lower levels and is primarily localized in the nuclei (34). However, in liver cancer cells, the expression level of APE1 is increased, is ectopically localized and can also be detected in the cytoplasm (35,36). Therefore, APE1 has been considered to be an important diagnostic indicator for hepatocellular carcinogenesis.
2. Functions of APE1
APE1 and DNA damage repair
When cells are exposed to ionizing radiation, assault from oxygen free radicals or improper cleavage by DNA endonucleases, apurinic/apyrimidinic sites (AP sites) can be readily formed (37). AP sites, a common form of DNA damage, can result from exposure to X-ray and ultraviolet radiation. The absence of a nucleotide base at an AP site can disrupt the action of DNA/RNA polymerases during transcription and synthesis, which leads to interruptions in nucleotide substitution and insertion. Furthermore, the chemical reactivity of AP sites can cause the breakage of cross-links between DNA molecules, as well as DNA-protein and DNA-DNA cross-links (38,39). These factors contribute to the high mutagenicity and cytotoxicity within radiation exposed cells. Consequently, the repair of AP sites is a crucial mechanism for maintaining genomic stability. The BER pathway is the primary pathway for repairing DNA damage, including AP sites, and APE1 is a key rate-limiting enzyme in this pathway (40,41). During the DNA repair process, APE1 interacts with proteins involved in the BER pathway, such as poly(ADP-ribose) polymerase 1(42), X-ray repair cross complementing 1(43), DNA polymerase β (44), DNA ligase III, proliferating cell nuclear antigen and flap structure-specific endonuclease 1, and exerts certain stimulatory effects on these proteins, allowing it to participate in and regulate BER (37) (Fig. 1).
APE1 and RNA damage repair
APE1 is also capable of cleaving AP sites present in single-stranded RNA molecules (45). Previous studies have reported that numerous non-coding RNAs and a number of specific microRNAs (miRNAs) directly bind to APE1 in cancer cells in specific ways (46,47). Early in vitro evidence and indirect observations have demonstrated the ability of APE1 to bind to and cleave RNA, as well as the relationship between the downregulation of APE1 and miRNA expression (25,47). Malfatti et al (48) demonstrated that APE1 can bind to the drosha ribonuclease III (DROSHA)-processing complex, which is associated with the regulation of primary miRNAs (pri-miRs) in cervical cancer in response to oxidative stress. The deletion of APE1 leads to increased oxidation of pri-miR-221/222 and enhances its interaction with DROSHA. The endoribonuclease activity of APE1 towards pri-miR-221/222 affects PTEN expression and is directly related to cancer progression (48).
APE1 and redox regulation
APE1 can also act as a redox factor, stimulating the binding of multiple redox factors, including AP-1, NF-κB, HIF-1α and p53, to DNA, thereby participating in processes such as cell proliferation, migration and inflammatory reactions (49,50). For instance, APE1 reduces the binding of the transcription factor Oct1 to the lectin-like oxidized low-density lipoprotein receptor-1 (LOX1) promoter, which leads to the downregulation of LOX1. Consequently, this suppression inhibits the activation of macrophages and the formation of foam cells induced by oxidized low-density lipoprotein in THP-1 cells (51). Of the seven cysteine residues present in the APE1 protein (Cys65, Cys93, Cys99, Cys138, Cys208, Cys296 and Cys310), Cys65, Cys93 and Cys99 are associated with the redox activity of APE1 (52,53) (Fig. 2). Furthermore, since only Cys65 is present in mammalian cells, mutations in the structure of Cys65 may affect the redox activity of APE1, whereas its DNA repair function remains unaffected.
3. Involvement of APE1 in signaling pathways
Research into the functions of APE1 initially focused on its repair activity as an endonuclease for DNA damage (38,48,54). However, as research progresses, an increasing number of studies have indicated that APE1 not only functions as a DNA repair enzyme but also acts as a redox protein to regulate the activation of various transcription factors, and it is increasingly recognized as a key factor in multiple signaling pathways (25,55) (Fig. 3). The APE1 pathway is a signaling pathway that is highly conserved from prokaryotes to humans, whereas its redox regulation of other signaling pathways is unique to mammals (56). APE1 inhibits adipocyte differentiation by suppressing the expression of transcription factors such as CCAAT-enhancer binding protein α, peroxisome proliferator-activated receptor γ and adaptor protein complex 2 in 3T3-L1 cells (57). APE1 has been reported to be associated with a large number of signaling pathways, including the STAT3, HIF-1α, NF-κB, AP-1 and P53 signaling pathways, which are typically closely related to the occurrence and development of tumor cells (26,33,58,59). APE1 has increased activity levels in pediatric acute lymphoblastic leukemia when compared with normal cells, and testing on patient samples has shown that signaling pathways associated with APE1, such as the NF-κB signaling pathway, are activated under conditions of oxidative and DNA damage (60,61). E3330 is an inhibitor that specifically targets the redox activity of APE1 and has been demonstrated in cell experiments to suppress the survival of lymphoma cells by interfering with the redox activity of APE1, downregulating the expression levels of the downstream target genes of APE1 and promoting lymphoma cell apoptosis (60,61). However, in vivo experiments using xenogeneic cell transplantation in nude mice have shown that the growth rate of tumor cells in E3330-treated mice is decreased, the expression of APE1-associated downstream target genes is downregulated (62) and E3330 has an inhibitory effect on tumors (63,64).
STAT3
STAT3 serves a vital role in both normal and cancerous cells. During the processes of cell proliferation and survival, STAT3 can be phosphorylated by Janus kinase to form dimers, which are then translocated into the nucleus to regulate gene transcription (65-67). In tumor cells, the STAT3 signaling pathway is often activated, and APE1 is also active, regulating gene transcription by affecting the sensitivity of STAT3 to DNA binding sites (68). Studies have shown that inhibition of the redox activity of APE1 by inhibitors against REF1/APE1 can reduce the expression levels of downstream target genes of STAT3 and suppress cancer cell proliferation (68,69). Therefore, the combined use of the STAT3 signaling pathway and APE1 inhibitors to inhibit tumor cell proliferation may represent a novel direction for clinical cancer treatment.
HIF-1
HIFs are a class of cellular factors produced by cells in a specific hypoxic environment, serving an essential role in various tissues and organs, and are abnormally expressed during the process of cellular carcinogenesis. Research has shown that HIF-1 is closely associated with the APE1 signaling pathway (33,70). The redox function of APE1 can promote the activation of HIF-1. Under hypoxic conditions, inhibiting the activity of APE1 can reduce the expression levels of downstream genes of HIF-1 and decrease the survival of tumor cells (33). HIF-1 and STAT3 also exhibit a synergistic effect against cell carcinogenesis (70-72). Furthermore, inhibitors of APE1 can be used to simultaneously suppress the transcription of genes downstream of both signaling pathways (73).
NF-κB
NF-κB is an intracellular transcription factor generated in response to both intracellular and extracellular signals, serving a vital role in various physiological processes of cells, including proliferation, migration, regeneration and the immune response (74). Research has demonstrated that the NF-κB signaling pathway also serves an important role in tumor formation (74). Once activated, it can lead to carcinogenesis through a series of changes, including inhibiting apoptosis or altering the tumor cell microenvironment. During the cell repair process following DNA damage, the NF-κB signaling pathway is also activated, promoting the generation of reactive oxygen species (ROS). Subsequently, ROS cause DNA damage in cells (75), which further activates the NF-κB signaling pathway and increases the expression levels of related anti-apoptotic and pro-proliferation genes, promoting the malignant transformation of cells (76).
APE1 is essential for the activation of the NF-κB signaling pathway, and the DNA binding of NF-κB depends on the redox activity of APE1. Research has demonstrated that inhibition of APE1 leads to decreased transcription of downstream genes regulated by NF-κB (62,77), and that APE1 can reduce the proliferative capacity of tumor cells and promote tumor cell apoptosis (34,69).
4. Impact of APE1 on cell carcinogenesis
During cell carcinogenesis, APE1 expression in the nuclei and cytoplasm is elevated, and there are changes to the DNA repair and redox functions involving APE1(78). Furthermore, high intracellular APE1 expression levels have been associated with poor outcomes of anticancer treatment, poor response to chemotherapy, low survival rates and shorter relapse-free intervals (79,80). Plasma APE1 levels are elevated in patients with colorectal, kidney, liver and pancreatic cancers and cutaneous squamous cell carcinoma (cSCC). APE1 is upregulated in human pancreatic cancer cells, and modulating its redox activity using APE1 inhibitors blocks the proliferation and migration of cancer cells (62), indicating that the redox activity of APE1 is closely related to cell proliferation and migration. A similar phenomenon has been observed in ovarian tumors, where ovarian tumor cells exhibited increased APE1 expression, and its knockdown inhibited tumor cell proliferation (81). Additionally, APE1 promotes the proliferation of cSCC cells. The deletion of APE1 can inhibit the viability of cSCC cells, while the upregulation of APE1 promotes cell proliferation (82). This has also been reported in HCC (62). Conventionally, the notion persists that APE1 is exclusively located within the nucleus, but emerging research has demonstrated that APE1 is expressed in the cytoplasm of both lung tumor and HCC cells (36,74). In HCC, all signaling pathways involving APE1 can stimulate cells to enhance their proliferation, metastasis and anti-apoptotic capabilities (83). Therefore, the downregulation of APE1 expression in tumor cells is likely to become a treatment for tumors in the future, and it may be ideal for patients who cannot tolerate chemotherapy or radiotherapy.
The development and progression of tumors involves the abnormal expression of multiple related genes (84). APE1 is a transcription factor that serves a vital role in DNA repair and redox functions, regulating cancer-related pathways (78,85), and is closely associated with carcinogenesis and proliferation of tumor cells. Research has shown that APE1 expression is associated with the staging and classification, degree of invasion and recurrence of tumors. Therefore, plasma APE1 expression levels can be used as biomarkers for the detection of bladder cancer (34). However, to date, there have been few reports on the relationship between APE1 and HCC. Current research suggests that, compared with that in non-cancerous tissues, APE1 expression in HCC tissues is increased, and it serves a crucial role in the carcinogenesis and progression of HCC (35). The downregulation of APE1 expression can effectively reduce the proliferation of Hep3B cells and promote tumor cell apoptosis (34). This suggests that APE1 may be able to promote tumor growth by inhibiting cell apoptosis. Tumor cells exhibit an enhanced proliferative capacity and a low apoptosis rate compared with non-tumor cells. The cytoplasmic localization of APE1 has been implicated in the carcinogenesis of various cancer types, including ovarian, lung and colorectal cancer, suggesting its potential as a prognostic marker and therapeutic target (86,87). Enhanced cytoplasmic APE1 expression, often associated with p53 aberrations, may predict survival and relapse in patients with cancer, highlighting the importance of the subcellular distribution of APE1 in tumor progression and aggression (74,88,89). Therefore, for cancer treatment, inhibiting cell proliferation and differentiation by promoting tumor cell apoptosis can be an effective method for tumor suppression.
5. Prospects of APE1-targeted drugs in clinical settings
APE1 serves an important role in the development and progression of cancer, and its expression is closely associated with the prognosis of patients (90). In HCC, APE1 expression is higher in cancerous tissue cells compared with that in para-neoplastic tissues (32). Elevated APE1 levels dysregulate homologous recombination and the cell cycle, contributing to chemoresistance (91). Research has indicated that APE1 inhibitors can enhance the efficacy of cisplatin chemotherapy, photodynamic therapy and radiotherapy (69,92-94). High APE1 expression is inversely associated with CD4+ naive T cell infiltration, which is a predictor of recurrence-free survival in patients with non-small cell lung cancer, with improved survival in patients with high APE1 expression levels (95). High APE1 expression in breast cancer nuclei is associated with poor disease-free survival, and is associated with the luminal A subtype and estrogen receptor positivity, while low APE1 expression in patients with low Ki-67 cases predicts worse overall survival rates (96). Combining APE1/REF1 redox inhibitors with the standard-of-care chemotherapy drug cisplatin in vitro more effectively inhibits bladder cancer cell proliferation when compared with cisplatin alone (69). Therefore a number of studies have proposed methods to treat HCC by inhibiting the action of APE1. By administering APE1 inhibitors, the functions of APE1 are inhibited, which further suppresses tumor cell proliferation and promotes tumor cell apoptosis (97-100) (Table I).
![]() | Table IComparison of APE1 blockers. Resveratrol, isoflavones, tanshinone and E3330 are clinical drugs that inhibit tumor growth by blocking APE1 expression and inhibiting the redox function of APE1. |
Resveratrol is also known as 3,4',5-trihydroxy-trans-stilbene and is a type of natural polyphenol compound. Experiments have shown that resveratrol pretreatment enhances human melanoma cell sensitivity to the chemotherapeutic drug dacarbazine (97,101). However, it has been shown in vitro that resveratrol effectively reduces the activities of AP-1 and NF-κB by inhibiting the redox function of APE1, and such an effect has been observed in a wide range of cancer types (102). Furthermore, resveratrol can be used as a selective inhibitor of APE1, laying the foundation for its clinical application.
Isoflavones are a class of natural compounds that have an important protective effect against cancer (98,103). It has been reported in vitro that isoflavones can effectively inhibit tumor cell proliferation and potentiate cell death by radiation (104). In non-small cell lung cancer, isoflavones sensitize tumor cells to radiation by inhibiting the DNA repair function of APE1(105). In addition, in prostate cancer, soy isoflavones can downregulate NF-κB and HIF-1 simultaneously by inhibiting APE1, potentiating tumor cell apoptosis, inhibiting tumor tissue angiogenesis and sensitizing tumor cells to radiation (102). From this perspective, isoflavones, as inhibitors of APE1, have an inhibitory effect on tumor progression. This also suggests that inhibiting APE1 activity could be a potential effective treatment strategy for cancer.
Tanshinone is a Traditional Chinese Medicine that can inhibit the redox activity of APE1(99). It can effectively inhibit the proliferation of human cervical and colon cancer cells. Furthermore, tanshinone pre-treatment can enhance the sensitivity of certain tumor cell lines, such as HeLa and HCT116 cells, to ionizing radiation and chemotherapy drugs. As an inhibitor of APE1, tanshinone may have a promising future in cancer treatment.
E3330 is also known as (2E)-3-[5-(2,3-dimethoxy-6-methyl-1,4-benzoquinoyl)]-2-nonyl-2-propenoic acid and is a compound that can selectively inhibit the redox activity of APE1 without having any impact on its DNA repair function (100). Therefore, E3330 has no inhibitory effect on the BER pathway. By increasing disulfide bond formation involving Cys65 and/or Cys93, E3330 effectively decreases the redox-active population of APE1 molecules (100,106). A previous study reported that E3330 can effectively inhibit the proliferation of tumor cells in ovarian, colon, lung, breast, brain, pancreatic and prostate cancers and multiple myeloma but does not significantly inhibit the proliferation of normal cells (107). By inhibiting the activity of APE1, the activities of some transcriptional regulators, including NF-κB, activator protein and HIF-1, are blocked, which have marked effects on the proliferation, invasion and metabolism of tumor cells, thereby inhibiting tumor progression (62). In addition, E3330 can effectively inhibit tumor cell proliferation and migration in pancreatic cancer (64).
A number of studies have reported that the redox domain of APE1 is indispensable for tumor-associated epithelial cell differentiation, function and angiogenesis following tumor cell migration (77,108). In liver cancer, APE1 can facilitate the development of HCC both in vitro and in vivo (35). APE1 overexpression and the increase in enzyme activity are related to the survival and drug resistance of cancer cells (109). Furthermore, the inhibition of APE1 leads to the accumulation of lipid peroxidation and enhanced ferroptosis in HCC (110). Western blotting analysis has demonstrated that diethylnitrosamine (DEN) treatment enhanced APE1 protein expression (111). The antioxidant effect of Licochalcone B and fullerene C60 may be the mechanism by which these compounds reduce the expression of APE1, which is predominantly activated by oxidative stress (111,112). This has a protective effect against DEN-induced HCC.
6. Conclusions
Inhibition of APE1 has the potential to be an effective treatment approach for tumors. It can be concluded from the aforementioned findings that APE1 serves a vital role in the development and progression of tumors. The inhibition of APE1 activity can effectively control tumor cell proliferation and spread, suggesting that targeting of APE1 may be a novel direction for the treatment of HCC, especially for patients with poor response to surgical therapy and chemotherapy.
Acknowledgements
Not applicable.
Funding
Funding: The present study was supported by the National Natural Science Foundation of China (grant nos. 81930119, 82090050 and 82090053) and the CAMS Innovation Fund for Medical Sciences (grant no. 2019-I2M-5-056).
Availability of data and materials
Not applicable.
Authors' contributions
LY and ZS equally contributed to the present manuscript, the conception and design of the study, literature review and analysis, and drafting, critical revision and editing of the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar | |
Singal AG, Kanwal F and Llovet JM: Global trends in hepatocellular carcinoma epidemiology: Implications for screening, prevention and therapy. Nat Rev Clin Oncol. 20:864–884. 2023.PubMed/NCBI View Article : Google Scholar | |
Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A and Roberts LR: A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 16:589–604. 2019.PubMed/NCBI View Article : Google Scholar | |
Clark T, Maximin S, Meier J, Pokharel S and Bhargava P: Hepatocellular carcinoma: Review of epidemiology, screening, imaging diagnosis, response assessment, and treatment. Curr Probl Diagn Radiol. 44:479–486. 2015.PubMed/NCBI View Article : Google Scholar | |
Sia D, Villanueva A, Friedman SL and Llovet JM: Liver cancer cell of origin, molecular class, and effects on patient prognosis. Gastroenterology. 152:745–761. 2017.PubMed/NCBI View Article : Google Scholar | |
Zhang H, Su X, Burley SK and Zheng XFS: mTOR regulates aerobic glycolysis through NEAT1 and nuclear paraspeckle-mediated mechanism in hepatocellular carcinoma. Theranostics. 12:3518–3533. 2022.PubMed/NCBI View Article : Google Scholar | |
Vogel A, Meyer T, Sapisochin G, Salem R and Saborowski A: Hepatocellular carcinoma. Lancet. 400:1345–1362. 2022.PubMed/NCBI View Article : Google Scholar | |
Chang Y, Jeong SW, Young Jang J and Jae Kim Y: Recent updates of transarterial chemoembolilzation in hepatocellular carcinoma. Int J Mol Sci. 21(8165)2020.PubMed/NCBI View Article : Google Scholar | |
Galle PP, Dufour JF, Peck-Radosavljevic M, Trojan J and Vogel A: Systemic therapy of advanced hepatocellular carcinoma. Future Oncol. 17:1237–1251. 2021.PubMed/NCBI View Article : Google Scholar | |
Llovet JM, De Baere T, Kulik L, Haber PK, Greten TF, Meyer T and Lencioni R: Locoregional therapies in the era of molecular and immune treatments for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 18:293–313. 2021.PubMed/NCBI View Article : Google Scholar | |
Llovet JM, Montal R, Sia D and Finn RS: Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol. 15:599–616. 2018.PubMed/NCBI View Article : Google Scholar | |
Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, Luo R, Feng J, Ye S, Yang TS, et al: Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: A phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 10:25–34. 2009.PubMed/NCBI View Article : Google Scholar | |
Llovet JM, Castet F, Heikenwalder M, Maini MK, Mazzaferro V, Pinato DJ, Pikarsky E, Zhu AX and Finn RS: Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol. 19:151–172. 2022.PubMed/NCBI View Article : Google Scholar | |
Kong FH, Ye QF, Miao XY, Liu X, Huang SQ, Xiong L, Wen Y and Zhang ZJ: Current status of sorafenib nanoparticle delivery systems in the treatment of hepatocellular carcinoma. Theranostics. 11:5464–5490. 2021.PubMed/NCBI View Article : Google Scholar | |
Ladd AD, Duarte S, Sahin I and Zarrinpar A: Mechanisms of drug resistance in HCC. Hepatology. 79:926–940. 2024.PubMed/NCBI View Article : Google Scholar | |
Dattachoudhury S, Sharma R, Kumar A and Jaganathan BG: Sorafenib inhibits proliferation, migration and invasion of breast cancer cells. Oncology. 98:478–486. 2020.PubMed/NCBI View Article : Google Scholar | |
Tang W, Chen Z, Zhang W, Cheng Y, Zhang B, Wu F, Wang Q, Wang S, Rong D, Reiter FP, et al: The mechanisms of sorafenib resistance in hepatocellular carcinoma: Theoretical basis and therapeutic aspects. Signal Transduct Target Ther. 5(87)2020.PubMed/NCBI View Article : Google Scholar | |
Tian C, Liu Y, Xue L, Zhang D, Zhang X, Su J, Chen J, Li X, Wang L and Jiao S: Sorafenib inhibits ovarian cancer cell proliferation and mobility and induces radiosensitivity by targeting the tumor cell epithelial-mesenchymal transition. Open Life Sci. 17:616–625. 2022.PubMed/NCBI View Article : Google Scholar | |
Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, et al: Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 359:378–390. 2008.PubMed/NCBI View Article : Google Scholar | |
Gupta N, Verma RK, Prinja S and Dhiman RK: Cost-effectiveness of sorafenib for treatment of advanced hepatocellular carcinoma in India. J Clin Exp Hepatol. 9:468–475. 2019.PubMed/NCBI View Article : Google Scholar | |
Zschäbitz S and Grüllich C: Lenvantinib: A tyrosine kinase inhibitor of VEGFR 1-3, FGFR 1-4, PDGFRα, KIT and RET. Recent Results Cancer Res. 211:187–198. 2018.PubMed/NCBI View Article : Google Scholar | |
Bo W and Chen Y: Lenvatinib resistance mechanism and potential ways to conquer. Front Pharmacol. 14(1153991)2023.PubMed/NCBI View Article : Google Scholar | |
Liu X, Lu Y and Qin S: Atezolizumab and bevacizumab for hepatocellular carcinoma: Mechanism, pharmacokinetics and future treatment strategies. Future Oncol. 17:2243–2256. 2021.PubMed/NCBI View Article : Google Scholar | |
Gao X, Zhao R, Ma H and Zuo S: Efficacy and safety of atezolizumab plus bevacizumab treatment for advanced hepatocellular carcinoma in the real world: A single-arm meta-analysis. BMC Cancer. 23(635)2023.PubMed/NCBI View Article : Google Scholar | |
López DJ, Rodríguez JA and Bañuelos S: Molecular mechanisms regulating the DNA repair protein APE1: A focus on its flexible N-terminal tail domain. Int J Mol Sci. 22(6308)2021.PubMed/NCBI View Article : Google Scholar | |
Caston RA, Gampala S, Armstrong L, Messmann RA, Fishel ML and Kelley MR: The multifunctional APE1 DNA repair-redox signaling protein as a drug target in human disease. Drug Discov Today. 26:218–228. 2021.PubMed/NCBI View Article : Google Scholar | |
He H, Liu X, Wu Y, Qi L, Huang J, Zhou Y, Zeng J, Wang K and He X: DNA nanotechnology-empowered fluorescence imaging of APE1 Activity. Chemistry. 5:1815–1831. 2023. | |
An SY, Jin SA, Seo HJ, Lee YR, Kim S, Jeon BH and Jeong JO: Protective effect of secretory APE1/Ref-1 on doxorubicin-induced cardiotoxicity via suppression of ROS and p53 pathway. ESC Heart Fail. 11:1182–1193. 2024.PubMed/NCBI View Article : Google Scholar | |
Zhang S, He L, Dai N, Guan W, Shan J, Yang X, Zhong Z, Qing Y, Jin F, Chen C, et al: Serum APE1 as a predictive marker for platinum-based chemotherapy of non-small cell lung cancer patients. Oncotarget. 7:77482–77494. 2016.PubMed/NCBI View Article : Google Scholar | |
McIlwain DW, Fishel ML, Boos A, Kelley MR and Jerde TJ: APE1/Ref-1 redox-specific inhibition decreases survivin protein levels and induces cell cycle arrest in prostate cancer cells. Oncotarget. 9:10962–10977. 2018.PubMed/NCBI View Article : Google Scholar | |
Lee YR, Park MS, Joo HK, Kim KM, Kim J, Jeon BH and Choi S: Therapeutic positioning of secretory acetylated APE1/Ref-1 requirement for suppression of tumor growth in triple-negative breast cancer in vivo. Sci Rep. 8(8701)2018.PubMed/NCBI View Article : Google Scholar | |
Di Maso V, Mediavilla MG, Vascotto C, Lupo F, Baccarani U, Avellini C, Tell G, Tiribelli C and Crocè LS: Transcriptional Up-Regulation of APE1/Ref-1 in hepatic tumor: Role in hepatocytes resistance to oxidative stress and apoptosis. PLoS One. 10(e0143289)2015.PubMed/NCBI View Article : Google Scholar | |
Logsdon DP, Grimard M, Luo M, Shahda S, Jiang Y, Tong Y, Yu Z, Zyromski N, Schipani E, Carta F, et al: Regulation of HIF1α under Hypoxia by APE1/Ref-1 Impacts CA9 expression: Dual targeting in patient-derived 3D pancreatic cancer models. Mol Cancer Ther. 15:2722–2732. 2016.PubMed/NCBI View Article : Google Scholar | |
Sun Z, Zhu Y, Aminbuhe Fan Q, Peng J and Zhang N: Differential expression of APE1 in hepatocellular carcinoma and the effects on proliferation and apoptosis of cancer cells. Biosci Trends. 12:456–462. 2018.PubMed/NCBI View Article : Google Scholar | |
Lu X, Zhao H, Yuan H, Chu Y and Zhu X: High nuclear expression of APE1 correlates with unfavorable prognosis and promotes tumor growth in hepatocellular carcinoma. J Mol Histol. 52:219–231. 2021.PubMed/NCBI View Article : Google Scholar | |
Di Maso V, Avellini C, Crocè LS, Rosso N, Quadrifoglio F, Cesaratto L, Codarin E, Bedogni G, Beltrami CA, Tell G and Tiribelli C: Subcellular localization of APE1/Ref-1 in human hepatocellular carcinoma: Possible prognostic significance. Mol Med. 13:89–96. 2007.PubMed/NCBI View Article : Google Scholar | |
Hegde ML, Hazra TK and Mitra S: Early steps in the DNA base excision/single-strand interruption repair pathway in mammalian cells. Cell Res. 18:27–47. 2008.PubMed/NCBI View Article : Google Scholar | |
Demple B, Herman T and Chen DS: Cloning and expression of APE, the cDNA encoding the major human apurinic endonuclease: Definition of a family of DNA repair enzymes. Proc Natl Acad Sci U S A. 88:11450–11454. 1991.PubMed/NCBI View Article : Google Scholar | |
Kciuk M, Marciniak B, Mojzych M and Kontek R: Focus on UV-Induced DNA damage and repair-disease relevance and protective strategies. Int J Mol Sci. 21(7264)2020.PubMed/NCBI View Article : Google Scholar | |
Krokan HE and Bjørås M: Base excision repair. Cold Spring Harb Perspect Biol. 5(a012583)2013.PubMed/NCBI View Article : Google Scholar | |
Hindi NN, Elsakrmy N and Ramotar D: The base excision repair process: Comparison between higher and lower eukaryotes. Cell Mol Life Sci. 78:7943–7965. 2021.PubMed/NCBI View Article : Google Scholar | |
Khodyreva SN, Prasad R, Ilina ES, Sukhanova MV, Kutuzov MM, Liu Y, Hou EW, Wilson SH and Lavrik OI: Apurinic/apyrimidinic (AP) site recognition by the 5'-dRP/AP lyase in poly(ADP-ribose) polymerase-1 (PARP-1). Proc Natl Acad Sci USA. 107:22090–22095. 2010.PubMed/NCBI View Article : Google Scholar | |
Vidal AE, Boiteux S, Hickson ID and Radicella JP: XRCC1 coordinates the initial and late stages of DNA abasic site repair through protein–protein interactions. EMBO J. 20(6530-6539-6539)2001.PubMed/NCBI View Article : Google Scholar | |
Bennett RA, Wilson DM III, Wong D and Demple B: Interaction of human apurinic endonuclease and DNA polymerase beta in the base excision repair pathway. Proc Natl Acad Sci USA. 94:7166–7169. 1997.PubMed/NCBI View Article : Google Scholar | |
Antoniali G, Serra F, Lirussi L, Tanaka M, D'Ambrosio C, Zhang S, Radovic S, Dalla E, Ciani Y, Scaloni A, et al: Mammalian APE1 controls miRNA processing and its interactome is linked to cancer RNA metabolism. Nat Commun. 8(797)2017.PubMed/NCBI View Article : Google Scholar | |
Berquist BR, McNeill DR and Wilson DM III: Characterization of abasic endonuclease activity of human Ape1 on alternative substrates, as well as effects of ATP and sequence context on AP site incision. J Mol Biol. 379:17–27. 2008.PubMed/NCBI View Article : Google Scholar | |
Antoniali G, Dalla E, Mangiapane G, Zhao X, Jing X, Cheng Y, De Sanctis V, Ayyildiz D, Piazza S, Li M and Tell G: APE1 controls DICER1 expression in NSCLC through miR-33a and miR-130b. Cell Mol Life Sci. 79(446)2022.PubMed/NCBI View Article : Google Scholar | |
Malfatti MC, Antoniali G, Codrich M and Tell G: Coping with RNA damage with a focus on APE1, a BER enzyme at the crossroad between DNA damage repair and RNA processing/decay. DNA Repair (Amst). 104(103133)2021.PubMed/NCBI View Article : Google Scholar | |
Kladova OA, Bazlekowa-Karaban M, Baconnais S, Piétrement O, Ishchenko AA, Matkarimov BT, Iakovlev DA, Vasenko A, Fedorova OS, Le Cam E, et al: The role of the N-terminal domain of human apurinic/apyrimidinic endonuclease 1, APE1, in DNA glycosylase stimulation. DNA Repair (Amst). 64:10–25. 2018.PubMed/NCBI View Article : Google Scholar | |
Oliveira TT, Coutinho LG, de Oliveira LOA, Timoteo ARS, Farias GC and Agnez-Lima LF: APE1/Ref-1 role in inflammation and immune response. Front Immunol. 13(793096)2022.PubMed/NCBI View Article : Google Scholar | |
Hu Z, Hui B, Hou X, Liu R, Sukhanov S and Liu X: APE1 inhibits foam cell formation from macrophages via LOX1 suppression. Am J Transl Res. 12:6559–6568. 2020.PubMed/NCBI | |
Luo M, Zhang J, He H, Su D, Chen Q, Gross ML, Kelley MR and Georgiadis MM: Characterization of the Redox activity and disulfide bond formation in apurinic/apyrimidinic endonuclease. Biochemistry. 51:695–705. 2012.PubMed/NCBI View Article : Google Scholar | |
Pekhale K, Haval G, Perween N, Antoniali G, Tell G and Ghaskadbi S and Ghaskadbi S: DNA repair enzyme APE1 from evolutionarily ancient Hydra reveals redox activity exclusively found in mammalian APE1. DNA Repair (Amst). 59:44–56. 2017.PubMed/NCBI View Article : Google Scholar | |
Kelley MR, Logsdon D and Fishel ML: Targeting DNA repair pathways for cancer treatment: What's new? Future Oncol. 10:1215–1237. 2014.PubMed/NCBI View Article : Google Scholar | |
Kelley MR, Georgiadis MM and Fishel ML: APE1/Ref-1 role in redox signaling: Translational applications of targeting the redox function of the DNA repair/redox protein APE1/Ref-1. Curr Mol Pharmacol. 5:36–53. 2012.PubMed/NCBI View Article : Google Scholar | |
Georgiadis MM, Luo M, Gaur RK, Delaplane S, Li X and Kelley MR: Evolution of the redox function in mammalian Apurinic/apyrimidinic endonuclease. Mutat Res. 643:54–63. 2008.PubMed/NCBI View Article : Google Scholar | |
Lee EO, Joo HK, Lee YR, Kim S, Lee KH, Lee SD and Jeon BH: APE1/Ref-1 inhibits adipogenic transcription factors during adipocyte differentiation in 3T3-L1 cells. Int J Mol Sci. 24(3251)2023.PubMed/NCBI View Article : Google Scholar | |
Shah F, Logsdon D, Messmann RA, Fehrenbacher JC, Fishel ML and Kelley MR: Exploiting the Ref-1-APE1 node in cancer signaling and other diseases: From bench to clinic. NPJ Precis Oncol. 1(19)2017.PubMed/NCBI View Article : Google Scholar | |
Garcia-Bailo B, El-Sohemy A, Haddad PS, Arora P, Benzaied F, Karmali M and Badawi A: Vitamins D, C, and E in the prevention of type 2 diabetes mellitus: Modulation of inflammation and oxidative stress. Biologics. 5:7–19. 2011.PubMed/NCBI View Article : Google Scholar | |
Biswas A, Khanna S, Roy S, Pan X, Sen CK and Gordillo GM: Endothelial cell tumor growth is Ape/ref-1 dependent. Am J Physiol Cell Physiol. 309:C296–C307. 2015.PubMed/NCBI View Article : Google Scholar | |
Ding J, Fishel ML, Reed AM, McAdams E, Czader MB, Cardoso AA and Kelley MR: Ref-1/APE1 as a transcriptional regulator and novel therapeutic target in pediatric T-cell Leukemia. Mol Cancer Ther. 16:1401–1411. 2017.PubMed/NCBI View Article : Google Scholar | |
Fishel ML, Jiang Y, Rajeshkumar NV, Scandura G, Sinn AL, He Y, Shen C, Jones DR, Pollok KE, Ivan M, et al: Impact of APE1/Ref-1 redox inhibition on pancreatic tumor growth. Mol Cancer Ther. 10:1698–1708. 2011.PubMed/NCBI View Article : Google Scholar | |
Vasko MR, Guo C, Thompson EL and Kelley MR: The repair function of the multifunctional DNA repair/redox protein APE1 is neuroprotective after ionizing radiation. DNA Repair (Amst). 10:942–952. 2011.PubMed/NCBI View Article : Google Scholar | |
Zou GM and Maitra A: Small-molecule inhibitor of the AP endonuclease 1/REF-1 E3330 inhibits pancreatic cancer cell growth and migration. Mol Cancer Ther. 7:2012–2021. 2008.PubMed/NCBI View Article : Google Scholar | |
Huynh J, Chand A, Gough D and Ernst M: Therapeutically exploiting STAT3 activity in cancer-using tissue repair as a road map. Nat Rev Cancer. 19:82–96. 2019.PubMed/NCBI View Article : Google Scholar | |
Hu X, li J, Fu M, Zhao X and Wang W: The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct Target Ther. 6(402)2021.PubMed/NCBI View Article : Google Scholar | |
Seif F, Khoshmirsafa M, Aazami H, Mohsenzadegan M, Sedighi G and Bahar M: The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun Signal. 15(23)2017.PubMed/NCBI View Article : Google Scholar | |
Cardoso AA, Jiang Y, Luo M, Reed AM, Shahda S, He Y, Maitra A, Kelley MR and Fishel ML: APE1/Ref-1 regulates STAT3 transcriptional activity and APE1/Ref-1-STAT3 dual-targeting effectively inhibits pancreatic cancer cell survival. PLoS One. 7(e47462)2012.PubMed/NCBI View Article : Google Scholar | |
Fishel ML, Xia H, McGeown J, McIlwain DW, Elbanna M, Craft AA, Kaimakliotis HZ, Sandusky GE, Zhang C, Pili R, et al: Antitumor activity and mechanistic characterization of APE1/Ref-1 inhibitors in bladder cancer. Mol Cancer Ther. 18:1947–1960. 2019.PubMed/NCBI View Article : Google Scholar | |
Pawlus MR, Wang L and Hu CJ: STAT3 and HIF1α cooperatively activate HIF1 target genes in MDA-MB-231 and RCC4 cells. Oncogene. 33:1670–1679. 2014.PubMed/NCBI View Article : Google Scholar | |
Dinarello A, Betto RM, Diamante L, Tesoriere A, Ghirardo R, Cioccarelli C, Meneghetti G, Peron M, Laquatra C, Tiso N, et al: STAT3 and HIF1α cooperatively mediate the transcriptional and physiological responses to hypoxia. Cell Death Discov. 9(226)2023.PubMed/NCBI View Article : Google Scholar | |
Rad E, Dodd K, Thomas L, Upadhyaya M and Tee A: STAT3 and HIF1α signaling drives oncogenic cellular phenotypes in malignant peripheral nerve sheath tumors. Mol Cancer Res. 13:1149–1160. 2015.PubMed/NCBI View Article : Google Scholar | |
Bhakat KK, Mantha AK and Mitra S: Transcriptional regulatory functions of mammalian AP-endonuclease (APE1/Ref-1), an essential multifunctional protein. Antioxid Redox Signal. 11:621–638. 2009.PubMed/NCBI View Article : Google Scholar | |
Wu HH, Cheng YW, Chang JT, Wu TC, Liu WS, Chen CY and Lee H: Subcellular localization of apurinic endonuclease 1 promotes lung tumor aggressiveness via NF-kappaB activation. Oncogene. 29:4330–4340. 2010.PubMed/NCBI View Article : Google Scholar | |
Huang TT, Wuerzberger-Davis SM, Wu ZH and Miyamoto S: Sequential modification of NEMO/IKKgamma by SUMO-1 and ubiquitin mediates NF-kappaB activation by genotoxic stress. Cell. 115:565–576. 2003.PubMed/NCBI View Article : Google Scholar | |
Xia L, Tan S, Zhou Y, Lin J, Wang H, Oyang L, Tian Y, Liu L, Su M, Wang H, et al: Role of the NFκB-signaling pathway in cancer. Onco Targets Ther. 11:2063–2073. 2018.PubMed/NCBI View Article : Google Scholar | |
Siqueira PB, de Sousa Rodrigues MM, de Amorim ÍSS, da Silva TG, da Silva Oliveira M, Rodrigues JA, de Souza da Fonseca A and Mencalha AL: The APE1/REF-1 and the hallmarks of cancer. Mol Biol Rep. 51(47)2024.PubMed/NCBI View Article : Google Scholar | |
Shin JH, Choi S, Lee YR, Park MS, Na YG, Irani K, Lee SD, Park JB, Kim JM, Lim JS and Jeon BH: APE1/Ref-1 as a serological biomarker for the detection of bladder cancer. Cancer Res Treat. 47:823–833. 2015.PubMed/NCBI View Article : Google Scholar | |
Luo M and Kelley MR: Inhibition of the human apurinic/apyrimidinic endonuclease (APE1) repair activity and sensitization of breast cancer cells to DNA alkylating agents with lucanthone. Anticancer Res. 24:2127–2134. 2004.PubMed/NCBI | |
Long K, Gu L, Li L, Zhang Z, Li E, Zhang Y, He L, Pan F, Guo Z and Hu Z: Small-molecule inhibition of APE1 induces apoptosis, pyroptosis, and necroptosis in non-small cell lung cancer. Cell Death Dis. 12(503)2021.PubMed/NCBI View Article : Google Scholar | |
Fishel ML, He Y, Reed AM, Chin-Sinex H, Hutchins GD, Mendonca MS and Kelley MR: Knockdown of the DNA repair and redox signaling protein Ape1/Ref-1 blocks ovarian cancer cell and tumor growth. DNA Repair (Amst). 7:177–186. 2008.PubMed/NCBI View Article : Google Scholar | |
Deng X, Zhen P, Niu X, Dai Y, Wang Y and Zhou M: APE1 promotes proliferation and migration of cutaneous squamous cell carcinoma. J Dermatol Sci. 100:67–74. 2020.PubMed/NCBI View Article : Google Scholar | |
Yang Z, Yang S, Misner BJ, Liu-Smith F and Meyskens FL: The role of APE/Ref-1 signaling pathway in hepatocellular carcinoma progression. Int J Oncol. 45:1820–1828. 2014.PubMed/NCBI View Article : Google Scholar | |
Singh AK, Kumar R and Pandey AK: Hepatocellular carcinoma: Causes, mechanism of progression and biomarkers. Curr Chem Genom Transl Med. 12:9–26. 2018.PubMed/NCBI View Article : Google Scholar | |
Tell G, Quadrifoglio F, Tiribelli C and Kelley MR: The many functions of APE1/Ref-1: Not only a DNA repair enzyme. Antioxid Redox Signal. 11:601–620. 2009.PubMed/NCBI View Article : Google Scholar | |
Sheng Q, Zhang Y, Wang R, Zhang J, Chen B, Wang J, Zhang W and Xin X: Prognostic significance of APE1 cytoplasmic localization in human epithelial ovarian cancer. Med Oncol. 29:1265–1271. 2012.PubMed/NCBI View Article : Google Scholar | |
Bazzani V, Barchiesi A, Radecka D, Pravisani R, Guadagno A, Di Loreto C, Baccarani U and Vascotto C: Mitochondrial apurinic/apyrimidinic endonuclease 1 enhances mtDNA repair contributing to cell proliferation and mitochondrial integrity in early stages of hepatocellular carcinoma. BMC Cancer. 20(969)2020.PubMed/NCBI View Article : Google Scholar | |
Wu HH, Chu YC, Wang L, Tsai LH, Lee MC, Chen CY, Shieh SH, Cheng YW and Lee H: Cytoplasmic Ape1 Expression Elevated by p53 aberration may predict survival and relapse in resected non-small cell lung cancer. Ann Surg Oncol. 20 (Suppl 3):S336–S347. 2013.PubMed/NCBI View Article : Google Scholar | |
Abbotts R and Madhusudan S: Human AP endonuclease 1 (APE1): From mechanistic insights to druggable target in cancer. Cancer Treat Rev. 36:425–435. 2010.PubMed/NCBI View Article : Google Scholar | |
Malfatti MC, Bellina A, Antoniali G and Tell G: Revisiting two decades of research focused on targeting APE1 for cancer therapy: The pros and cons. Cells. 12(1895)2023.PubMed/NCBI View Article : Google Scholar | |
Kumar S, Zhao J, Talluri S, Buon L, Mu S, Potluri LB, Liao C, Shi J, Chakraborty C, Gonzalez GB, et al: Elevated APE1 dysregulates homologous recombination and cell cycle driving genomic evolution, tumorigenesis, and chemoresistance in esophageal adenocarcinoma. Gastroenterology. 165:357–373. 2023.PubMed/NCBI View Article : Google Scholar | |
Wang D, Xiang DB, Yang XQ, Chen LS, Li MX, Zhong ZY and Zhang YS: APE1 overexpression is associated with cisplatin resistance in non-small cell lung cancer and targeted inhibition of APE1 enhances the activity of cisplatin in A549 cells. Lung Cancer. 66:298–304. 2009.PubMed/NCBI View Article : Google Scholar | |
Franchi LP, de Freitas Lima JEB, Piva HL and Tedesco AC: The redox function of apurinic/apyrimidinic endonuclease 1 as key modulator in photodynamic therapy. J Photochem Photobiol B. 211(111992)2020.PubMed/NCBI View Article : Google Scholar | |
Zhou J, Wei Z, Yang C, Jia D, Pan B, Zeng Y, Sun D and Yu Y: APE1 promotes radiation resistance against radiation-induced pyroptosis by inhibiting the STING pathway in lung adenocarcinoma. Transl Oncol. 36(101749)2023.PubMed/NCBI View Article : Google Scholar | |
Li Y, Zhao X, Xiao H, Yang B, Liu J, Rao W, Dai X, Li M, Dai N, Yang Y and Wang D: APE1 may influence CD4+ naïve T cells on recurrence free survival in early stage NSCLC. BMC Cancer. 21(233)2021.PubMed/NCBI View Article : Google Scholar | |
Woo J, Park H, Sung SH, Moon BI, Suh H and Lim W: Prognostic Value of Human Apurinic/Apyrimidinic Endonuclease 1 (APE1) Expression in Breast Cancer. PLoS One. 9(e99528)2014.PubMed/NCBI View Article : Google Scholar | |
Lee SG, Lee DG, Joo YH and Chung N: Synergistic inhibitory effects of the oxyresveratrol and dacarbazine combination against melanoma cells. Oncol Lett. 22(667)2021.PubMed/NCBI View Article : Google Scholar | |
Gómez-Zorita S, González-Arceo M, Fernández-Quintela A, Eseberri I, Trepiana J and Portillo MP: Scientific evidence supporting the beneficial effects of isoflavones on human health. Nutrients. 12(3853)2020.PubMed/NCBI View Article : Google Scholar | |
Sui J, Li M, Qian C, Wang S, Cheng Y, Chen BP and Wang D: Functional analysis of tanshinone IIA that blocks the redox function of human apurinic/apyrimidinic endonuclease 1/redox factor-1. Drug Des Devel Ther. 8:2147–2160. 2014.PubMed/NCBI View Article : Google Scholar | |
Cesaratto L, Codarin E, Vascotto C, Leonardi A, Kelley MR, Tiribelli C and Tell G: Specific inhibition of the redox activity of ape1/ref-1 by e3330 blocks tnf-α-induced activation of IL-8 production in liver cancer cell lines. PLoS One. 8(e70909)2013.PubMed/NCBI View Article : Google Scholar | |
Kang S, Wang Z, Li B, Gao X, He W, Cao S, Cai Y and Chen H: Anti-tumor effects of resveratrol on malignant melanoma is associated with promoter demethylation of RUNX3 gene. Pharmazie. 74:163–167. 2019.PubMed/NCBI View Article : Google Scholar | |
Laev SS, Salakhutdinov NF and Lavrik OI: Inhibitors of nuclease and redox activity of apurinic/apyrimidinic endonuclease 1/redox effector factor 1 (APE1/Ref-1). Bioorg Med Chem. 25:2531–2544. 2017.PubMed/NCBI View Article : Google Scholar | |
Kim IS: Current perspectives on the beneficial effects of soybean isoflavones and their metabolites for humans. Antioxidants (Basel). 10(1064)2021.PubMed/NCBI View Article : Google Scholar | |
Hillman GG: Soy isoflavones protect normal tissues while enhancing radiation responses. Semin Radiat Oncol. 29:62–71. 2019.PubMed/NCBI View Article : Google Scholar | |
Singh-Gupta V, Joiner MC, Runyan L, Yunker CK, Sarkar FH, Miller S, Gadgeel SM, Konski AA and Hillman GG: Soy isoflavones augment radiation effect by inhibiting APE1/Ref-1 DNA repair activity in non-small cell lung cancer. J Thorac Oncol. 6:688–698. 2011.PubMed/NCBI View Article : Google Scholar | |
Su D, Delaplane S, Luo M, Rempel DL, Vu B, Kelley MR, Gross ML and Georgiadis MM: Interactions of apurinic/apyrimidinic endonuclease with a redox inhibitor: Evidence for an alternate conformation of the enzyme. Biochemistry. 50:82–92. 2011.PubMed/NCBI View Article : Google Scholar | |
Luo M, Delaplane S, Jiang A, Reed A, He Y, Fishel M, Nyland RL II, Borch RF, Qiao X, Georgiadis MM and Kelley MR: Role of the multifunctional DNA repair and redox signaling protein Ape1/Ref-1 in cancer and endothelial cells: Small-molecule inhibition of the redox function of Ape1. Antioxid Redox Signal. 10:1853–1867. 2008.PubMed/NCBI View Article : Google Scholar | |
Zou GM, Karikari C, Kabe Y, Handa H, Anders RA and Maitra A: The Ape-1/Ref-1 redox antagonist E3330 inhibits the growth of tumor endothelium and endothelial progenitor cells: Therapeutic implications in tumor angiogenesis. J Cell Physiol. 219:209–218. 2009.PubMed/NCBI View Article : Google Scholar | |
Sengupta S, Mantha AK, Mitra S and Bhakat KK: Human AP endonuclease (APE1/Ref-1) and its acetylation regulate YB-1-p300 recruitment and RNA polymerase II loading in the drug-induced activation of multidrug resistance gene MDR1. Oncogene. 30:482–493. 2011.PubMed/NCBI View Article : Google Scholar | |
Du Y, Zhou Y, Yan X, Pan F, He L, Guo Z and Hu Z: APE1 inhibition enhances ferroptotic cell death and contributes to hepatocellular carcinoma therapy. Cell Death Differ. 31:431–446. 2024.PubMed/NCBI View Article : Google Scholar | |
Sadek K, Abouzed T, Nasr S and Shoukry M: Licochalcone B ameliorates liver cancer via targeting of apoptotic genes, DNA repair systems, and cell cycle control. Iran J Pharm Res. 19:372–386. 2020.PubMed/NCBI View Article : Google Scholar | |
Sadek K, Abouzeid T, Nasr S and Shukry M: Role and potential targeting of hepatic apurinic/apyrimidinic endonuclease-1 and cyclin-dependent kinase-4 in hepatocellular carcinoma. Can J Physiol Pharmacol. 96(X)2018.PubMed/NCBI View Article : Google Scholar |