International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Radiotherapy remains a major treatment option for numerous patients, serving both as a radical therapy and as palliative care (1,2). Since the discovery of X-rays by Wilhelm Conrad Röntgen in 1895, the field of radiotherapy has undergone marked evolution (3). This advancement has been driven by the pioneering contributions of renowned scientists such as Nikola Tesla, Mihajlo Idvorski Pupin and Maria Sklodowska-Curie (4). Their innovative efforts have established a solid foundation for modern clinical applications of radiotherapy, which can be employed individually or in combination with other therapies for both curative and palliative purposes at all stages of cancer (5,6). More than half of all patients with cancer undergo radiotherapy; however, its effectiveness is often diminished by the emergence of radioresistance, which can profoundly impact the quality of life of patients (7,8). Radiation therapy exerts its effects by destroying cancer cells either directly or indirectly through the induction of DNA damage. Ionizing radiation (IR) can also activate several prosurvival signaling pathways, including the p53, ataxia telangiectasia mutated and NF-κB pathways, which enhance DNA damage checkpoint activation, promote DNA repair and stimulate autophagy (9–11). Together, these signaling pathways provide protection to cancer cells against radiation-induced damage, thereby contributing to their resistance to treatment (12).
Autophagy is a vital protein degradation pathway in cells, and is essential for maintaining cellular homeostasis and overall health (13). Autophagy, which was first identified in human liver cells, is categorized into three forms in mammalian cells: Macroautophagy, microautophagy and chaperone-mediated autophagy (14). The autophagy process typically includes four main stages: Initiation, formation and maturation of the autophagosome, precise fusion of the autophagosome with the lysosome, and finally content degradation (15) (Fig. 1). During the autophagy process activated by radiotherapy, a cellular mechanism directs old, damaged or malfunctioning biomolecules and organelles to lysosomes for degradation (16,17). This process is preserved through evolution and is triggered by different internal and external stressors, including nutrient shortage, absence of growth factors, IR or low oxygen levels (18). Under such circumstances, autophagy functions primarily as a survival tactic by clearing out dysfunctional organelles or toxic aggregates that could otherwise lead to apoptosis (19). Furthermore, the lysosomal decomposition of excess cellular parts supplies crucial raw materials and nutrients that can be reused to create important biomolecules (20). Therefore, regulating tumor autophagy levels to reduce radiotherapy resistance represents a novel approach.
Long non-coding RNAs (lncRNAs) are defined as transcripts longer than 200 nucleotides that do not encode proteins (21). Despite their low conservation across species, lncRNAs function as molecular switches that influence cell survival, inflammation and lipid metabolism in the vasculature by regulating autophagy (22). Research has established a relationship between lncRNAs and resistance to tumor radiotherapy (23,24). Additionally, lncRNAs serve various roles, including acting as molecular signals, decoys, guides and scaffolds (25). Therefore, reviewing the role of autophagy-modulating lncRNAs in tumor radioresistance can enhance the understanding of these regulatory mechanisms and may lead to the identification of novel therapeutic strategies aimed at increasing tumor radiosensitivity through the modulation of autophagy.
The present review aims to present the current understanding of the role of autophagy in cancer and its impact on tumor radioresistance through interactions with lncRNAs. By elucidating the function of autophagy-modulating lncRNAs in tumor radioresistance, the present review aims to provide valuable insights into the context-dependent applications of therapeutic strategies.
The role of autophagy in cancer is complex and can be viewed as a double-edged sword; it has the potential to inhibit tumor development, while also being implicated in tumor initiation (26). Autophagy operates at a basal level in all cell types and actively contributes to tumor prevention (27). As a critical intracellular mechanism, autophagy effectively responds to genotoxic stress through pathways involving reactive oxygen species (ROS), helping to prevent the accumulation of mutations and maintain genetic stability (28). Furthermore, autophagy is responsible for the removal of damaged organelles, ensuring that cellular damage does not accumulate, thereby preserving homeostasis and normal cellular function (29). This underscores its precise, standardized and essential role in maintaining cellular health and stability (30).
Among the numerous regulators of autophagy that have been identified, beclin 1 (BECN1) is recognized as one of the most crucial (31). As a vital regulatory protein, BECN1 is essential for initiating autophagy and serves a role in maintaining the delicate balance between cell survival and death (32). Research has indicated that heterozygous loss of the BECN1 gene enhances tumor development in mice, highlighting its importance in understanding the mechanisms underlying tumorigenesis (33). Notably, the deletion of the BECN1 gene is frequently observed in various malignant tumors, including breast, ovarian and prostate cancer, underscoring its critical role in cancer pathology (34–36). Furthermore, a study has demonstrated that overexpression of BECN1 in breast cancer cells effectively inhibited cell proliferation and colony formation, and reduced tumor growth in nude mice (37). In addition to BECN1, research has shown that the knockout of the autophagy-related gene (ATG)5 and ATG7 genes in mice disrupts the autophagy process, leading to exacerbated oxidative stress responses and an increased risk of developing liver cancer (38). These findings emphasize the crucial role of autophagy in tumor prevention.
Autophagy can promote the occurrence and development of tumors (39). Tumor cells can be likened to ‘small strongholds’ existing in a challenging environment, with autophagy serving as their ‘super housekeeper’. This process efficiently recycles large biomolecules, enabling the cells to thrive while also helping to clear cellular damage and prevent serious injuries. Thus, autophagy may function as a ‘behind-the-scenes hero’ in sustaining and promoting cancer cell proliferation (40). Notably, in some cancer cells, the level of autophagy is higher than that in normal cells, likely as an adaptive mechanism to meet their elevated metabolic demands (41). This suggests that autophagy serves a diverse and intricate role in tumor survival. Additionally, autophagy has been linked to enhanced migration and invasion of cancer cells, particularly in hepatocellular carcinoma (HCC) (42). Furthermore, it serves a role in maintaining the characteristics of cancer stem cells (CSCs) in breast cancer cell lines (43). In general, autophagy serves a crucial role in maintaining genomic integrity, supporting cellular metabolism and ensuring cell survival.
Although radiotherapy is a main approach for the treatment of cancer, resistance frequently arises, resulting in unsuccessful treatments (44). This radioresistance arises from the capacity of tumors to disrupt stress responses, such as DNA damage and repair processes, which either promote or inhibit autophagy and aid in nutrient recycling (45) (Fig. 2). For example, by modulating Unc-51 like autophagy activating kinase 1 (ULK1)-mediated autophagy, butyrophilin subfamily 3 member A1 contributes to tumor progression and radiation resistance in esophageal squamous cell carcinoma (46). Xu et al (47) reported that blocking autophagic flux and DNA repair in tumor cells could enhance the effectiveness of radiotherapy for orthotopic glioblastoma. In addition to damaged proteins, various intermediate molecules, their complexes, and organelles such as mitochondria and micronuclei can serve as cargo for the autophagy process (48). Previous studies have revealed both pro-survival and anti-survival characteristics of autophagic pathways (49,50). Cancer cells frequently take advantage of the dual role of autophagy to survive in metabolically difficult conditions, escape immune system attacks, prevent cell death and induce metastasis (51,52). Research has revealed that using both radiation and berberine together suppresses autophagy and alternative end-joining DNA repair, which are linked to radioresistance, thereby enhancing sensitivity to radiation (53).
The sensitivity of tumor cells to radiation is frequently affected by autophagic processes (54). BECN1 moves to the nucleus after IR exposure, resulting in G2/M cell cycle arrest (55). Furthermore, autophagy driven by ATG5 has been demonstrated to increase the radiosensitivity of prostate cancer cells, especially when nutrients are scarce or glutamine is depleted, and also when MYC is silenced (56). Chen et al (57) proposed that inhibiting the nuclear factor erythroid 2-related factor 2 (NRF2) antioxidative pathway could make U-2 osteosarcoma cells more sensitive to radiation, with the NRF2 antioxidative response being controlled by autophagy-driven activation of ERK 1/2 kinases. Consequently, reducing the expression levels of BECN1, ATG5 or NRF2 has been shown to decrease the IR sensitivity of cancer cells. Furthermore, the inhibition of ATG7 by lncRNA HOTAIR, which is notably upregulated in prostate cancer cell lines exposed to radiation, has been associated with radioresistance in these cells (58). These results emphasize the essential function of autophagy in influencing radiosensitivity and suggest the possibility of targeting autophagic pathways to enhance radiotherapy effectiveness (58).
Research on breast cancer cells, which naturally resist apoptosis, further underscores the link between IR and autophagic pathways (59). After IR exposure, these cells show increased autophagic characteristics, resulting in more iron accumulation. This accumulation, in conjunction with subsequent ROS generation, oxidative stress and DNA damage, may trigger cell death through a mechanism known as ferroptosis (60,61). These insights have sparked a rising interest in pairing autophagic inducers with IR to improve the radiosensitivity of cancer cells. For instance, in a similar approach, the treatment of non-small cell lung carcinoma (NSCLC) cells with rapamycin alongside a histone deacetylase inhibitor has been found to promote radiosensitization (62). This combined treatment has two effects: It boosts autophagy and at the same time hinders DNA damage repair processes. Observations in cultured cells and tumor xenograft mouse models have demonstrated the effectiveness of the strategy, pointing to a potential improvement in radiotherapy outcomes (63).
Autophagy affects cancer cell survival in complex ways, acting both as a supporter and an opponent of radiosensitization (62,64). Various cancer cell types have been shown to exploit enhanced autophagy as a survival strategy. For instance, the application of autophagy inhibitors renders otherwise radio-resistant bladder cancer (BLCA) cells more sensitive to chemotherapy (65). Similarly, silencing of ATG5, a key component of the autophagy process, has been found to increase IR-induced cell death in nasopharyngeal carcinoma (66). Furthermore, the use of autophagy inhibitors in combination with IR has emerged as a factor influencing the bystander and abscopal effects associated with chemotherapy and radiotherapy (67). These findings underscore the importance of context in the role of autophagy in cancer treatment and highlight the potential of targeting autophagy pathways to enhance treatment efficacy.
Autophagy serves a vital role in preserving the stemness of CSCs (68). A number of tumors activate the epithelial-mesenchymal transition (EMT) program to acquire stem cell-like properties, facilitating their growth, invasion and metastasis (69). A study involving cervical cancer cells has shown that ATGs, especially ATG5, are crucial in the EMT process (70). Autophagy is crucial for enhancing tumor growth, invasiveness and maintaining stem cell-like traits in radio-resistant cancer cells, including pancreatic ductal adenocarcinoma and NSCLC stem cells (71,72). As a result, autophagy inhibitors, either on their own or alongside conventional cancer treatments, have been successful in preventing cell proliferation, colony formation and spheroid formation in pancreatic CSC populations (73). Additionally, the suppression of crucial autophagic genes, including Atg5, has been identified to enhance radiosensitivity in radio-resistant CSCs, such as those found in prostate cancer (56). These results emphasize the complex function of autophagy in maintaining CSC characteristics and its potential as a therapeutic target to improve treatment results.
Radiotherapy is a common cancer treatment that induces different forms of cell death, including autophagy (74). Besides directly causing DNA strand breaks, IR can lead to the production of significant amounts of ROS in cells, resulting in oxidative stress damage (75). According to Yang et al (76), mitophagy triggered by IR boosts ferroptosis by raising the levels of free fatty acids inside cells. Lysosomes serve a crucial role in autophagy, a cellular degradation process (77). A conserved protein associated with autophagy, Atgs/P62/LC3II, controls the process (78). In cancer cells, autophagy is a double-edged sword. In the initial phases, it might restrict the development of tumors. Nevertheless, it might also offer a survival advantage for adaptation and detoxification in challenging conditions such as starvation, low oxygen levels, and chemotherapy or radiotherapy (79). Investigators have reported that autophagy activity rises following IR. This acts as a mechanism for cells to survive when faced with cytotoxic agents, including IR and temozolomide, in the treatment of glioblastoma (80,81). Furthermore, researchers have revealed that IR promotes autophagy via the Wnt/β-Catenin pathway (82). In conclusion, IR has an important effect on tumor autophagy.
Numerous cancer types have been found to exhibit abnormal expression of lncRNAs, and the relationship between lncRNAs and autophagy has garnered interest across various cancer types (Table I), including lung, gastric, breast and prostate cancer (83). The majority of research suggests that lncRNAs regulate autophagy mainly through mechanisms such as microRNA (miRNA/miR) sponging (functioning as competing endogenous RNAs), interactions between RNAs, RNA-protein regulation and various other pathways (84,85). Studies have indicated that lncRNAs are involved in various phases of the autophagy process from the beginning to the maturation stage (86,87). They assist in starting autophagic phagocytosis by controlling essential proteins such as ULK1, mTOR and BECN1, and they also affect the extension of autophagic vesicles by regulating ATG3, ATG5, ATG4, ATG12 and ATG7 (88). The present review highlights the complex and diverse functions of lncRNAs in regulating autophagy, and their possible impact on cancer biology and treatment.
Proliferation is one of the most critical malignant phenotypes observed in cancer, while apoptosis is equally significant in the context of neoplastic development (89). Although their functions are fundamentally opposite (proliferation promotes cell growth and survival, while apoptosis leads to programmed cell death), their interactions work synergistically to regulate cancer development. This delicate balance between cell proliferation and apoptosis serves a crucial role in tumor progression and resistance to therapy (90). In digestive system cancers, various lncRNAs have been identified to serve roles in tumor progression and autophagy regulation (91). For instance, lncRNA SNHG11 is upregulated in gastric cancer and is associated with poor patient prognosis. lncRNA SNHG11 post-transcriptionally enhances ATG12 expression through miR-1276, thereby promoting autophagy, cell proliferation and activation of the Wnt/β-catenin signaling pathway (92). Similarly, in HCC, lncRNA MALAT1 exhibits elevated expression levels compared with those in normal tissues. Silencing MALAT1 has been shown to increase HCC autophagy by promoting the conversion of LC3-I to LC3-II and simultaneously suppressing HCC cell proliferation (93). Conversely, lncRNA NBR2 acts as a tumor suppressor in HCC by inhibiting BECN1 and autophagy through the ERK/JNK pathways, thereby restraining HCC cell proliferation (94). In colorectal cancer (CRC), lncRNA SLCO4A1-AS1 acts as an oncogenic element, exhibiting a positive association with par-3 family cell polarity regulator (PARD3) and sponging miR-508-3p. This lncRNA enhances the proliferation of CRC cells and initiates autophagy through the miR-508-3p/PARD3 pathway. Nonetheless, the regulatory effect is interrupted by the application of the autophagy inhibitor 3-methyladenine (95). Another lncRNA in CRC, FIRRE, directly interacts with polypyrimidine tract binding protein 1 to enhance BECN1 mRNA stability, thus reducing autophagy and promoting CRC cell proliferation (96). In the context of oral squamous cell carcinoma (OSCC) and cholangiocarcinoma (CCA), several upregulated lncRNAs have been shown to suppress autophagy. For instance, LINC01207 is highly expressed in OSCC, where its upregulation promotes cell proliferation but inhibits both apoptosis and autophagy via the miR-1301-3p/lactate dehydrogenase A axis (97). By contrast, overexpression of LINC00958 reduces apoptosis while promoting autophagy by upregulating autophagy-related proteins such as BECN1 and ATG5, and increasing the LC3-II/LC3-I ratio, driven by p53 mediated by sirtuin 1 (SIRT1) (98). In CCA, lncRNA HOTAIR inhibits both apoptotic and autophagic processes, thereby promoting the proliferation of CCA cells by targeting the miR-204-5p/high mobility group box 1 axis (99). Pancreatic cancer (PANC), known for its poor prognosis, remains a complex malignancy with unclear progression mechanisms (100). A study by Wu et al (101) revealed that LZTS1-AS1, a highly expressed lncRNA in PANC cells and tissues, promoted PANC cell proliferation while inhibiting apoptosis and autophagy via the miR-532/twist family bHLH transcription factor 1 axis. Overall, these findings highlight the diverse and critical roles of lncRNAs in modulating autophagy and influencing the malignant phenotypes of various cancer types.
Lv et al (102) found that silencing TUG1 suppressed cell proliferation while promoting apoptosis and autophagy in ccRCC cells. This effect is considered to be mediated via the miR-31-5p/flotillin 1 axis (102). In addition, lncRNA SCAMP1 has been observed to be upregulated in renal cell carcinoma (RCC) cells and tumors, where it regulates zinc finger E-box binding homeobox 1 and JUN as well as autophagy. This regulation is considered to contribute to oxidative stress-induced RCC in pediatric patients via miR-429 (103). In the context of BLCA, the lncRNA autophagy network serves a crucial role in disease progression. Zhang et al (104) demonstrated that ADAMTS9-AS2 inhibited proliferation and enhanced both autophagy and apoptosis through the PI3K/AKT/mTOR signaling pathway.
The downregulation of lncRNA CASC2 enhances apoptosis in NSCLC and diminishes ATG5-mediated autophagy by regulating the miR-214/tripartite motif containing 16 axis in A549 and H1299 NSCLC cell lines (105). Conversely, a reversed autophagic state has been noted in papillary thyroid carcinoma (PTC), where overexpression of lncRNA SLC26A4-AS1 inhibited the proliferation of PTC cells and stimulated autophagy by recruiting the transcription factor ETS1 and elevating inositol 1,4,5-trisphosphate receptor type 1 expression (106). Additionally, Qin et al (107) revealed that lncRNA SNHG5 was stabilized by RNA binding motif protein 47 (RBM47) and directed to FOXO3, resulting in decreased proliferation and the activation of autophagy in PTC cells through the RBM47/small nucleolar RNA host gene 5/FOXO3 pathway.
Current studies (108,109) have highlighted the role of several lncRNAs, which primarily regulate autophagy, as significant contributors to tumor radioresistance (Fig. 3), and these lncRNAs are discussed subsequently.
CASC19, a newly identified lncRNA located on chromosome 8q24.21, consists of 324 nucleotides (110). A study has demonstrated that CASC19 expression is upregulated in a variety of human cancer types, including NSCLC, gastric cancer, CRC, PANC, ccRCC, glioma, cervical cancer and nasopharyngeal carcinoma (111). Notably, some research has indicated that CASC19 can enhance radioresistance in nasopharyngeal carcinoma by modulating autophagy signaling pathways (112,113). Furthermore, the dysregulation of CASC19 has been closely linked to numerous clinicopathological characteristics (prognosis and pathological staging) and cancer progression, particularly in HCC and gastric cancer (114,115). In summary, CASC19 influences various cellular phenotypes, including cell proliferation, apoptosis, cell cycle regulation, migration, invasion, EMT, autophagy and therapeutic resistance (116).
NEAT1 is predominantly located in the nucleus, although a smaller fraction is present in the cytoplasm (117). NEAT1 functions as a structural component of nuclear paraspeckles and is involved in regulating gene expression at both transcriptional and post-transcriptional levels (118). Research has demonstrated that NEAT1 contributes to radioresistance in HCC cells by inducing autophagy through GABA type A receptor-associated protein (119). The role of NEAT1 in carcinogenesis is multifaceted, encompassing interactions with miRNAs, modulation of gene expression, regulation of epigenetic factors and participation in various signaling pathways, such as the PI3K-AKT and TGF-β1 pathways (120). Additionally, the involvement of NEAT1 in cancer is complicated by its impact on CSCs and the tumor microenvironment (121). The interaction of NEAT1 with autophagy further adds to the complexity of its functions in cancer biology (122). Understanding the interactions between NEAT1, autophagy and radioresistance is crucial for researchers as it may lead to the identification of novel therapeutic targets and strategies aimed at disrupting oncogenic processes, reducing treatment resistance and improving patient survival (123). Consequently, this understanding could facilitate the development of specialized treatment approaches and regimens.
lncRNA TP53TG1 (National Center for Biotechnology Information reference sequence, NR_015381.1), located on chromosome 12, is transcribed as an ~0.7-kilobase lncRNA molecule (124). Its expression can be induced under conditions of cellular stress in a wild-type TP53-dependent manner (125). As a relatively recent discovery in the field of lncRNAs, TP53TG1 has been implicated to serve oncogenic and anti-oncogenic roles across various cancer types, such as gastric and breast cancer (126). For example, overexpression of TP53TG1 promotes proliferation of colon cancer cells (127). In addition, Cheng et al (128) found that lncRNA TP53TG1 exerted anticancer effects in cervical cancer by comprehensively regulating the transcriptome profile of HeLa cells. Research has indicated a close association between TP53TG1 and autophagy (129). Furthermore, it has been demonstrated that lncRNA TP53TG1 contributes to the radioresistance of glioma cells through the miR-524-5p/RAB5A axis (130). Given these findings, TP53TG1 represents a promising target to enhance the sensitivity of tumors to radiotherapy.
HULC, found on chromosome 6p24.3, has been identified to be upregulated in HCC and is linked to cancer advancement (131). The expression of this transcript is regulated by miRNAs and transcriptionally modulated by Sp1 family factors (132). HULC has been identified as a novel biomarker in different types of cancer, such as prostate cancer and CRC, serving a role as an oncogenic factor (133). Research conducted previously indicated that inhibiting HULC could reduce angiogenesis in human gliomas (134). Another study demonstrated that HULC facilitated the advancement of CRC (135). A study has shown that lncRNA HULC serves a role in radioresistance by reducing autophagy in prostate cancer cells (136). Thus, lncRNA HULC could serve as a therapeutic target to combat tumor resistance to radiotherapy.
FIRRE is a lncRNA that is conserved and found on the X chromosome (137). FIRRE is associated with reduced overall survival in neuroblastoma and has been found to promote cell proliferation in lymphoma (138). According to Shi et al (139), lncRNA FIRRE is triggered by MYC and facilitates the progression of diffuse large B-cell lymphoma through the Wnt/β-catenin signaling pathway. FIRRE is a lncRNA associated with autophagy and serves as a prognostic biomarker for esophageal cancer, which was identified using a bioinformatics approach (140). Cai et al (141) revealed that lncRNA FIRRE influenced the sensitivity of endometrial cancer to radiotherapy through autophagy mediated by the miR-199b-5p/SIRT1/BECN1 axis.
PVT1, a lncRNA, was initially identified as a driver of variant translocations in mouse plasmacytomas (142). The human lncPVT1 gene is situated on chromosome 8q24.21, sharing the same location as the MYC oncogene (143). lncPVT1 has been identified as a factor that accelerates the development of several cancer types, such as nasopharyngeal cancer and lung cancer (144,145). Radioresistance poses a major challenge to tumor treatment success, and is connected to the abnormal regulation of physiological functions in cancer cells, including apoptosis, autophagy, stemness (for CSCs), hypoxia, EMT and DNA damage repair (146,147). Recently, lncPVT1 has been linked to the modulation of radioresistance in certain cancer types, including nasopharyngeal cancer and lung cancer (148).
Research interest in lncRNA therapeutics has only emerged in the past decade, and so far, no treatments targeting lncRNAs have reached clinical development (149). SP100-AS1, a lncRNA that targets the antisense sequence of the SP100 gene, is upregulated in the tissues of patients with CRC who are resistant to radiation, according to RNA-sequencing analysis (150). Notably, reducing SP100-AS1 expression lowered radioresistance and decreased cell proliferation and tumor growth in both laboratory and live models. To identify the proteins and miRNAs interacting with SP100-AS1, mass spectrometry and bioinformatics analyses were utilized. SP100-AS1 was shown to interact with and stabilize the ATG3 protein via the ubiquitination-dependent proteasome mechanism (150). This interaction highlights the possible involvement of SP100-AS1 in regulating autophagy-related mechanisms and enhancing radioresistance in CRC. SP100-AS1 holds potential as a therapeutic target to enhance the response of CRC to radiation therapy (150). As aforementioned, autophagy affects tumor cell proliferation, survival and the response to treatment in intricate ways across different types of cancer, such as nasopharyngeal cancer and CRC. Methods to target autophagy might involve aiming at specific proteins or pathways associated with autophagy, as well as using agents that promote or block autophagy.
Scientists seek to improve treatment effectiveness by utilizing the interaction between autophagy and processes regulated by lncRNAs. They aim to develop more effective treatments by integrating autophagy modulation with lncRNA-targeted methods (151). Furthermore, these combination therapies provide a comprehensive strategy to boost patient outcomes (152).
There are both obstacles and prospects in the future research of radiation oncology and lncRNAomics (153). For precise tumor targeting, technological advancements such as hybrid MRI and positron emission tomography scans are vital, although they entail significant costs and demand specialized expertise (154). One major obstacle in lncRNA research is the restricted knowledge gained from examining just a small portion of lncRNAs, which limits the understanding of their mechanisms, roles and structures (155). Despite significant progress in the field, delivering oligonucleotides to solid tumors continues to be a major challenge. Advancements in artificial intelligence and sophisticated statistical modeling have enabled molecular-guided treatment plans customized for each patient, representing a significant achievement in precision medicine. Autophagy-modulating lncRNAs represent a vast yet largely unexplored reservoir of oncogenes, tumor suppressors and radioresponse modifiers. Integrating this knowledge into treatment strategies, from diagnosis and response prediction to the design of targeted therapies, could greatly enhance management and outcomes for patients with cancer (156). In conclusion, the present review summarizes the effects of autophagy-modulating lncRNAs on tumor radioresistance, providing novel ideas for tumor radiosensitization.
lncRNAs that modulate autophagy are crucial elements in the radioresistance of tumors. These lncRNAs are crucial in different stages of cancer development and advancement, such as tumor expansion, spread and resistance to treatment. Studies are concentrating on using autophagy-based treatment methods to tackle radioresistance in patients with cancer (157,158). Modifying autophagy can influence the survival of tumor cells, their response to therapy and the overall success of treatment. Creating targeted cancer treatments requires a deep comprehension of the interaction between lncRNAs and autophagy. Targeting specific lncRNAs could potentially adjust autophagy and increase tumor radiosensitivity. Current clinical trials and preclinical research are investigating the potential of targeting autophagy and lncRNAs to enhance treatment outcomes for patients with cancers resistant to radiation (159,160).
Not applicable.
The present study was supported by The Wings Scientific and Technological Foundation of The First People's Hospital of Changde City (grant no. Z2025ZC04) and Interdisciplinary Research Program in Medicine and Engineering, The First Affiliated Hospital of University of South China (grant no. IRP-M&E-2025-05)
Not applicable.
HL wrote and reviewed the drafts of the paper, while ZH created the figures and tables, and revised the paper. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Schaue D and McBride WH: Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol. 12:527–540. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Yerolatsite M, Torounidou N, Amylidi AL, Rapti IC, Zarkavelis G, Kampletsas E and Voulgari PV: A Systematic review of pneumonitis following treatment with immune checkpoint inhibitors and radiotherapy. Biomedicines. 13:9462025. View Article : Google Scholar : PubMed/NCBI | |
|
Tubiana M: Wilhelm conrad röntgen and the discovery of X-rays. Bull Acad Natl Med. 180:97–108. 1996.(In French). PubMed/NCBI | |
|
Babic RR, Stankovic Babic G, Babic SR and Babic NR: 120 years since the discovery of X-Rays. Med Pregl. 69:323–330. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Klucznik KA, Ravkilde T, Skouboe S, Møller DS, Hokland S, Keall P, Buus S, Bentzen L and Poulsen PR: Cone-beam CT-based estimations of prostate motion and dose distortion during radiotherapy. Phys Imaging Radiat Oncol. 35:1007982025. View Article : Google Scholar : PubMed/NCBI | |
|
Lam MB, Landrum MB, McWilliams JM, Buzzee B, Wright AA, Keating NL and Landon BE: Practice-level spending variation for radiation treatment episodes among older adults with cancer. JAMA Health Forum. 6:e2519522025. View Article : Google Scholar : PubMed/NCBI | |
|
Hill RM, Fok M, Grundy G, Parsons JL and Rocha S: The role of autophagy in hypoxia-induced radioresistance. Radiother Oncol. 189:1099512023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Song R, Yuan J, Hou G, Chu AL, Huang Y, Xiao C, Chai T, Sun C and Liu Z: Exosome-Shuttled METTL14 From AML-Derived mesenchymal stem cells promotes the proliferation and radioresistance in AML cells by stabilizing ROCK1 expression via an m6A-IGF2BP3-dependent mechanism. Drug Dev Res. 86:e700252025. View Article : Google Scholar : PubMed/NCBI | |
|
Luo H, Huang MF, Xu A, Wang D, Gingold JA, Tu J, Wang R, Huo Z, Chiang YT, Tsai KL, et al: Mutant p53 confers chemoresistance by activating KMT5B-mediated DNA repair pathway in nasopharyngeal carcinoma. Cancer Lett. 625:2177362025. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Y, Zhang L, Zhang Y, Xu Y, Zhou K, Yang Z, Zhu W, Zhang Q, Cao J, Wang L and Jiao Y: NEDD4-mediated endothelial-mesenchymal transition participates in radiation-induced lung injury through the ATM signaling pathway. Dose Response. 23:155932582513527262025. View Article : Google Scholar : PubMed/NCBI | |
|
Qu Z, Shi L, Wang P, Zhao A, Zheng X and Yin Q: Dual targeting of inflammatory and immune checkpoint pathways to overcome radiotherapy resistance in esophageal squamous cell carcinoma. J Inflamm Res. 18:9091–9106. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Porrazzo A, Cassandri M, D'Alessandro A, Morciano P, Rota R, Marampon F and Cenci G: DNA repair in tumor radioresistance: Insights from fruit flies genetics. Cell Oncol. 47:717–732. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Mizushima N and Komatsu M: Autophagy: Renovation of cells and tissues. Cell. 147:728–741. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Cursaro I, Rossi S, Butini S, Gemma S, Carullo G and Campiani G: A focus on natural autophagy modulators as potential host-directed weapons against emerging and re-emerging viruses. Med Res Rev. Jul 16–2025.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI | |
|
Parzych KR and Klionsky DJ: An overview of autophagy: Morphology, mechanism, and regulation. Antioxid Redox Signal. 20:460–473. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Debnath J, Gammoh N and Ryan KM: Autophagy and autophagy-related pathways in cancer. Nat Rev Mol Cell Biol. 24:560–575. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Vafadar A, Tajbakhsh A, Hosseinpour-Soleimani F, Savardshtaki A and Hashempur MH: Phytochemical-mediated efferocytosis and autophagy in inflammation control. Cell Death Discov. 10:4932024. View Article : Google Scholar : PubMed/NCBI | |
|
Feng X, Zhang H, Meng L, Song H, Zhou Q, Qu C, Zhao P, Li Q, Zou C, Liu X and Zhang Z: Hypoxia-induced acetylation of PAK1 enhances autophagy and promotes brain tumorigenesis via phosphorylating ATG5. Autophagy. 17:723–742. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu S, Yao S, Yang H, Liu S and Wang Y: Autophagy: Regulator of cell death. Cell Death Dis. 14:6482023. View Article : Google Scholar : PubMed/NCBI | |
|
Martini-Stoica H, Xu Y, Ballabio A and Zheng H: The autophagy-lysosomal pathway in neurodegeneration: A TFEB perspective. Trends Neurosci. 39:221–234. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lin W, Zhou Q, Wang CQ, Zhu L, Bi C, Zhang S, Wang X and Jin H: LncRNAs regulate metabolism in cancer. Int J Biol Sci. 16:1194–1206. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Y, Liu L, Wu H, Zheng Y, Zhan H and Li L: LncRNA GAS5 regulated by FTO-mediated m6A demethylation promotes autophagic cell death in NSCLC by targeting UPF1/BRD4 axis. Mol Cell Biochem. 479:553–566. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Tong Y, Yu C, Pu J, Zhu W, Zhou Y, Wang Y, Xiong Y and Sun X: FAP positive cancer-associated fibroblasts promote tumor progression and radioresistance in esophageal squamous cell carcinoma by transferring exosomal lncRNA AFAP1-AS1. Mol Carcinog. 63:1922–1937. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Hjazi A, Jasim SA, Altalbawy FMA, Kaur H, Hamzah HF, Kaur I, Deorari M, Kumar A, Elawady A and Fenjan MN: Relationship between lncRNA MALAT1 and Chemo-radiotherapy resistance of cancer cells: Uncovered truths. Cell Biochem Biophys. 82:1613–1627. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Huang H, Jin H, Lei R, He Z, He S, Chen J, Saw PE, Qiu Z, Ren G and Nie Y: lncRNA-WAL promotes triple-negative breast cancer aggression by inducing β-catenin nuclear translocation. Mol Cancer Res. 22:1036–1050. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Russell RC and Guan KL: The multifaceted role of autophagy in cancer. EMBO J. 41:e1100312022. View Article : Google Scholar : PubMed/NCBI | |
|
Bhol CS, Senapati PK, Kar RK, Chew G, Mahapatra KK, Lee EHC, Kumar AP, Bhutia SK and Sethi G: Autophagy paradox: Genetic and epigenetic control of autophagy in cancer progression. Cancer Lett. 630:2179092025. View Article : Google Scholar : PubMed/NCBI | |
|
Gao L, Loveless J, Shay C and Teng Y: Targeting ROS-Mediated crosstalk between autophagy and apoptosis in cancer. Adv Exp Med Biol. 1260:1–12. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Carretero-Fernández M, Cabrera-Serrano AJ, Sánchez-Maldonado JM, Ruiz-Durán L, Jiménez-Romera F, García-Verdejo FJ, González-Olmedo C, Cardús A, Díaz-Beltrán L, Gutiérrez-Bautista JF, et al: Autophagy and oxidative stress in solid tumors: Mechanisms and therapeutic opportunities. Crit Rev Oncol Hematol. 212:1048202025. View Article : Google Scholar : PubMed/NCBI | |
|
Hama Y, Ogasawara Y and Noda NN: Autophagy and cancer: Basic mechanisms and inhibitor development. Cancer Sci. 114:2699–2708. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Liu F, Li F, Zeng G, Wen X, Ding J and Zhou J: DHX9-mediated epigenetic silencing of BECN1 contributes to impaired autophagy and tumor progression in breast cancer via recruitment of HDAC5. Cell Death Dis. 16:5242025. View Article : Google Scholar : PubMed/NCBI | |
|
Cao Z, Tian K, Ran Y, Zhou H, Zhou L, Ding Y and Tang X: Beclin-1: A therapeutic target at the intersection of autophagy, immunotherapy, and cancer treatment. Front Immunol. 15:15064262024. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Yang KB, Chen W, Mai J, Wu XQ, Sun T, Wu RY, Jiao L, Li DD, Ji J, et al: CUL3 (cullin 3)-mediated ubiquitination and degradation of BECN1 (beclin 1) inhibit autophagy and promote tumor progression. Autophagy. 17:4323–4340. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Sun Y, Wang B and Wang H: Prognostic significance of autophagy-related genes BECN1 and LC3 in ovarian cancer: A meta-analysis. J Int Med Res. 48:3000605209682992020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Xu P, Chen D, Fan X, Xu Y, Li M, Yang X and Wang C: Roles of autophagy-related genes Beclin-1 and LC3 in the development and progression of prostate cancer and benign prostatic hyperplasia. Biomed Rep. 1:855–860. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Yao Q, Chen J, Lv Y, Wang T, Zhang J, Fan J and Wang L: The significance of expression of autophagy-related gene Beclin, Bcl-2, and Bax in breast cancer tissues. Tumour Biol. 32:1163–1171. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Wu CL, Liu JF, Liu Y, Wang YX, Fu KF, Yu XJ, Pu Q, Chen XX and Zhou LJ: BECN1 inhibition enhances paclitaxel-mediated cytotoxicity in breast cancer in vitro and in vivo. Int J Mol Med. 43:1866–1878. 2019.PubMed/NCBI | |
|
Tran S, Juliani J, Harris TJ, Evangelista M, Ratcliffe J, Ellis SL, Baloyan D, Reehorst CM, Nightingale R, Luk IY, et al: BECN1 is essential for intestinal homeostasis involving autophagy-independent mechanisms through its function in endocytic trafficking. Commun Biol. 7:2092024. View Article : Google Scholar : PubMed/NCBI | |
|
Ishaq M, Ojha R, Sharma AP and Singh SK: Autophagy in cancer: Recent advances and future directions. Semin Cancer Biol Semin Cancer Biol. 66:171–181. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Senapati PK, Mahapatra KK, Singh A and Bhutia SK: mTOR inhibitors in targeting autophagy and autophagy-associated signaling for cancer cell death and therapy. Biochim Biophys Acta Rev Cancer. 1880:1893422025. View Article : Google Scholar : PubMed/NCBI | |
|
Ferro F, Servais S, Besson P, Roger S, Dumas JF and Brisson L: Autophagy and mitophagy in cancer metabolic remodelling. Semin Cell Dev Biol. 98:129–138. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M, Liu S, Chua MS, Li H, Luo D, Wang S, Zhang S, Han B and Sun C: SOCS5 inhibition induces autophagy to impair metastasis in hepatocellular carcinoma cells via the PI3K/Akt/mTOR pathway. Cell Death Dis. 10:6122019. View Article : Google Scholar : PubMed/NCBI | |
|
Maycotte P, Jones KL, Goodall ML, Thorburn J and Thorburn A: Autophagy supports breast cancer stem cell maintenance by regulating IL6 secretion. Mol Cancer Res. 13:651–658. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Song J, Yang P, Chen C, Ding W, Tillement O, Bai H and Zhang S: Targeting epigenetic regulators as a promising avenue to overcome cancer therapy resistance. Signal Transduct Target Ther. 10:2192025. View Article : Google Scholar : PubMed/NCBI | |
|
ALKhemeiri N, Eljack S and Saber-Ayad MM: Perspectives of targeting autophagy as an adjuvant to anti-PD-1/PD-L1 therapy for colorectal cancer treatment. Cells. 14:7452025. View Article : Google Scholar : PubMed/NCBI | |
|
Yang W, Cheng B, Chen P, Sun X, Wen Z and Cheng Y: BTN3A1 promotes tumor progression and radiation resistance in esophageal squamous cell carcinoma by regulating ULK1-mediated autophagy. Cell Death Dis. 13:9842022. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Q, Zhang H, Liu H, Han Y, Qiu W and Li Z: Inhibiting autophagy flux and DNA repair of tumor cells to boost radiotherapy of orthotopic glioblastoma. Biomaterials. 280:1212872022. View Article : Google Scholar : PubMed/NCBI | |
|
Chaurasia M, Bhatt AN, Das A, Dwarakanath BS and Sharma K: Radiation-induced autophagy: Mechanisms and consequences. Free Radic Res. 50:273–290. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Yue T, Zheng D, Yang J, He J and Hou J: Potential value and cardiovascular risks of programmed cell death in cancer treatment. Front Pharmacol. 16:16159742025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu S, Wang L, Zhu L, Zhao T, Han P, Yan F, Wang X, Li C, Wang Z and Yang BF: Mechanism and regulation of mitophagy in liver diseases: A review. Front Cell Dev Biol. 13:16149402025. View Article : Google Scholar : PubMed/NCBI | |
|
Yun CW and Lee SH: The roles of autophagy in cancer. Int J Mol Sci. 19:34662018. View Article : Google Scholar : PubMed/NCBI | |
|
Singh SS, Vats S, Chia AY, Tan TZ, Deng S, Ong MS, Arfuso F, Yap CT, Goh BC, Sethi G, et al: Dual role of autophagy in hallmarks of cancer. Oncogene. 37:1142–1158. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Clark A, Villarreal MR, Huang SB, Jayamohan S, Rivas P, Hussain SS, Ybarra M, Osmulski P, Gaczynska ME, Shim EY, et al: Targeting S6K/NFκB/SQSTM1/Polθ signaling to suppress radiation resistance in prostate cancer. Cancer Lett. 597:2170632024. View Article : Google Scholar : PubMed/NCBI | |
|
Sezen Us A, Dagsuyu E, Us H, Cöremen M, Karabulut Bulan O and Yanardag R: Apocynin may alleviate side effects of autophagy-blocked radiotherapy through antioxidant effects. Biotech Histochem. Jun 30–2025.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI | |
|
Ma S, Fu X, Liu L, Liu Y, Feng H, Jiang H, Liu X, Liu R, Liang Z, Li M, et al: Iron-dependent autophagic cell death induced by radiation in MDA-MB-231 breast cancer cells. Front Cell Dev Biol. 9:7238012021. View Article : Google Scholar : PubMed/NCBI | |
|
Mukha A, Kahya U, Linge A, Chen O, Löck S, Lukiyanchuk V, Richter S, Alves TC, Peitzsch M, Telychko V, et al: GLS-driven glutamine catabolism contributes to prostate cancer radiosensitivity by regulating the redox state, stemness and ATG5-mediated autophagy. Theranostics. 11:7844–7868. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen N, Zhang R, Konishi T and Wang J: Upregulation of NRF2 through autophagy/ERK 1/2 ameliorates ionizing radiation induced cell death of human osteosarcoma U-2 OS. Mutat Res Genet Toxicol Environ Mutagen. 813:10–17. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wu C, Yang L, Qi X, Wang T, Li M and Xu K: Inhibition of long non-coding RNA HOTAIR enhances radiosensitivity via regulating autophagy in pancreatic cancer. Cancer Manag Res. 10:5261–5271. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Cui L, Song Z, Liang B, Jia L, Ma S and Liu X: Radiation induces autophagic cell death via the p53/DRAM signaling pathway in breast cancer cells. Oncol Rep. 35:3639–3647. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lei G, Zhang Y, Koppula P, Liu X, Zhang J, Lin SH, Ajani JA, Xiao Q, Liao Z, Wang H and Gan B: The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30:146–162. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Kang R, Kroemer G and Tang D: Broadening horizons: The role of ferroptosis in cancer. Nat Rev Clin Oncol. 18:280–296. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Liu F, Fang C, Xu L, Chen L, Xu Z, Chen J, Peng W, Fu B and Li Y: Combination of rapamycin and SAHA enhanced radiosensitization by inducing autophagy and acetylation in NSCLC. Aging (Albany NY). 13:18223–18237. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Luo H, Wu X, Dong M, Wang D, Ou Y, Wang Y, Sun S, Liu Z, Yang Z, et al: Inhibition of Phosphoglycerate Kinase 1 Enhances Radiosensitivity of Esophageal Squamous Cell Carcinoma to X-rays and Carbon Ion Irradiation. Front Biosci (Landmark Ed). 30:364302025. View Article : Google Scholar : PubMed/NCBI | |
|
Mukha A, Kahya U and Dubrovska A: Targeting glutamine metabolism and autophagy: The combination for prostate cancer radiosensitization. Autophagy. 17:3879–3881. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ma X, Mao G, Chang R, Wang F, Zhang X and Kong Z: Down-regulation of autophagy-associated protein increased acquired radio-resistance bladder cancer cells sensitivity to taxol. Int J Radiat Biol. 97:507–516. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mo N, Lu YK, Xie WM, Liu Y, Zhou WX, Wang HX, Nong L, Jia YX, Tan AH, Chen Y, et al: Inhibition of autophagy enhances the radiosensitivity of nasopharyngeal carcinoma by reducing Rad51 expression. Oncol Rep. 32:1905–1912. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Prise KM and O'Sullivan JM: Radiation-induced bystander signalling in cancer therapy. Nat Rev Cancer. 9:351–360. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Jia H, Wei J, Zheng W and Li Z: The dual role of autophagy in cancer stem cells: Implications for tumor progression and therapy resistance. J Transl Med. 23:5832025. View Article : Google Scholar : PubMed/NCBI | |
|
Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, et al: The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 133:704–715. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou S, Wang X, Ding J, Yang H and Xie Y: Increased ATG5 expression predicts poor prognosis and promotes EMT in cervical carcinoma. Front Cell Dev Biol. 9:7571842021. View Article : Google Scholar : PubMed/NCBI | |
|
Pustovalova M, Alhaddad L, Blokhina T, Smetanina N, Chigasova A, Chuprov-Netochin R, Eremin P, Gilmutdinova I, Osipov AN and Leonov S: The CD44high subpopulation of multifraction irradiation-surviving NSCLC cells exhibits partial EMT-program activation and DNA damage response depending on their p53 Status. Int J Mol Sci. 22:23692021. View Article : Google Scholar : PubMed/NCBI | |
|
Yazal T, Bailleul J, Ruan Y, Sung D, Chu FI, Palomera D, Dao A, Sehgal A, Gurunathan V, Aryan L, et al: Radiosensitizing pancreatic cancer via effective autophagy inhibition. Mol Cancer Ther. 21:79–88. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Gajate C, Gayet O, Fraunhoffer NA, Iovanna J, Dusetti N and Mollinedo F: Induction of apoptosis in human pancreatic cancer stem cells by the endoplasmic reticulum-targeted alkylphospholipid analog edelfosine and potentiation by autophagy inhibition. Cancers (Basel). 13:61242021. View Article : Google Scholar : PubMed/NCBI | |
|
Fischer P, Schmid M, Ohradanova-Repic A, Schneeweiss R, Hadatsch J, Grünert O, Benedum J, Röhrer A, Staudinger F, Schatzlmaier P, et al: Molecular features of TNBC govern heterogeneity in the response to radiation and autophagy inhibition. Cell Death Dis. 16:5402025. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao S, Wang Y, Pan S, Mu M, Chen B, Li H, Feng C, Fan R, Yu W, Han B, et al: Bismuth-functionalized probiotics for enhanced antitumor radiotherapy and immune activation. J Mater Chem B. Jul 14–2025.(Epub ahead of print). View Article : Google Scholar | |
|
Yang P, Li J, Zhang T, Ren Y, Zhang Q, Liu R, Li H, Hua J, Wang WA, Wang J and Zhou H: Ionizing radiation-induced mitophagy promotes ferroptosis by increasing intracellular free fatty acids. Cell Death Differ. 30:2432–2445. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Liu X, Li Z, Wei Y, Liu B, Zhu P, Liu Y and Zhao R: VPS37A activates the autophagy-lysosomal pathway for TNFR1 degradation and induces NF-κB-Regulated cell death under metabolic stress in colorectal cancer. Oncol Res. 33:2085–2105. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Azmat MA, Zaheer M, Shaban M, Arshad S, Hasan M, Ashraf A, Naeem M, Ahmad A and Munawar N: Autophagy: A new avenue and biochemical mechanisms to mitigate the climate change. Scientifica (Cairo). 2024:99083232024. View Article : Google Scholar : PubMed/NCBI | |
|
Choi Y, Bowman JW and Jung JU: Autophagy during viral infection-a double-edged sword. Nat Rev Microbiol. 16:341–354. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Daniel P, Sabri S, Chaddad A, Meehan B, Jean-Claude B, Rak J and Abdulkarim BS: Temozolomide induced hypermutation in glioma: Evolutionary mechanisms and therapeutic opportunities. Front Oncol. 9:412019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu EK, Sulman EP, Wen PY and Kurz SC: Novel therapies for glioblastoma. Curr Neurol Neurosci Rep. 20:192020. View Article : Google Scholar : PubMed/NCBI | |
|
Tsai CY, Ko HJ, Huang CF, Lin CY, Chiou SJ, Su YF, Lieu AS, Loh JK, Kwan AL, Chuang TH and Hong YR: Ionizing radiation induces resistant glioblastoma stem-like cells by promoting autophagy via the Wnt/β-catenin pathway. Life (Basel). 11:4512021.PubMed/NCBI | |
|
Wang Y, Fu Y, Lu Y, Chen S, Zhang J, Liu B and Yuan Y: Unravelling the complexity of lncRNAs in autophagy to improve potential cancer therapy. Biochim Biophys Acta Rev Cancer. 1878:1889322023. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng J, Mat Ludin AF, Rajab NF, Shaolong L and Jufri NF: The roles of lncMALAT1 in coronary artery disease regulation and therapeutic perspective: A systematic review. iScience. 28:1129452025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Shi Y, Wang C and Zhang K: LncRNA MALAT1 knockdown inhibits apoptosis of mouse hippocampus neuron cells with high glucose by Silencing autophagy. BMC Endocr Disord. 25:1732025. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L, Wang H, Shen Q, Feng L and Jin H: Long non-coding RNAs involved in autophagy regulation. Cell Death Dis. 8:e30732017. View Article : Google Scholar : PubMed/NCBI | |
|
Xia N, Zhang P, Yang L, Yin X, Wu SQ, Yao Y, Shang MY and Weng L: The lncRNA MIR503HG/miR-16-5p/FOSL1 pathway mediates autophagy to promote esophageal epithelial cells proliferation and EMT in esophageal restenosis. Arch Biochem Biophys. 772:1105362025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu X, Sun Y, Yu Q, Wang X, Wang Y and Zhao Y: Exosomal lncRNA GAS5 promotes M1 macrophage polarization in allergic rhinitis via restraining mTORC1/ULK1/ATG13-mediated autophagy and subsequently activating NF-кB signaling. Int Immunopharmacol. 121:1104502023. View Article : Google Scholar : PubMed/NCBI | |
|
DeBerardinis RJ, Lum JJ, Hatzivassiliou G and Thompson CB: The biology of cancer: Metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7:11–20. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Song Y, Wang Z, Fu L, Xu L, Feng X, Zhang Z and Yuan K: Dietary sugar intervention: A promising approach for cancer therapy. Biochim Biophys Acta Rev Cancer. 1880:1894022025. View Article : Google Scholar : PubMed/NCBI | |
|
Li C, Wang K, Zhao L, Liu J, Jin Y, Zhang C, Xu M, Wang M, Kuang Y, Liu J, et al: LINC00622 transcriptionally promotes RRAGD to repress mTORC1-modulated autophagic cell death by associating with BTF3 in cutaneous melanoma. Cell Death Dis. 16:5152025. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Q, Ma J, Wei J, Meng W, Wang Y and Shi M: lncRNA SNHG11 promotes gastric cancer progression by activating the Wnt/β-Catenin pathway and oncogenic autophagy. Mol Ther. 29:1258–1278. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen F, Zhong Z, Tan HY, Guo W, Zhang C, Cheng CS, Wang N, Ren J and Feng Y: Suppression of lncRNA MALAT1 by betulinic acid inhibits hepatocellular carcinoma progression by targeting IAPs via miR-22-3p. Clin Transl Med. 10:e1902020. View Article : Google Scholar : PubMed/NCBI | |
|
Sheng JQ, Wang MR, Fang D, Liu L, Huang WJ, Tian DA, He XX and Li PY: LncRNA NBR2 inhibits tumorigenesis by regulating autophagy in hepatocellular carcinoma. Biomed Pharmacother. 133:1110232021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z and Jin J: LncRNA SLCO4A1-AS1 promotes colorectal cancer cell proliferation by enhancing autophagy via miR-508-3p/PARD3 axis. Aging (Albany NY). 11:4876–4889. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Li Z, Xu S, Li W, Chen M, Jiang M and Fan X: LncRNA FIRRE functions as a tumor promoter by interaction with PTBP1 to stabilize BECN1 mRNA and facilitate autophagy. Cell Death Dis. 13:982022. View Article : Google Scholar : PubMed/NCBI | |
|
Lu X, Chen L, Li Y, Huang R, Meng X and Sun F: Long non-coding RNA LINC01207 promotes cell proliferation and migration but suppresses apoptosis and autophagy in oral squamous cell carcinoma by the microRNA-1301-3p/lactate dehydrogenase isoform A axis. Bioengineered. 12:7780–7793. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang N, Zhang X, Gu X, Li X and Shang L: Progress in understanding the role of lncRNA in programmed cell death. Cell Death Discov. 7:302021. View Article : Google Scholar : PubMed/NCBI | |
|
Lu M, Qin X, Zhou Y, Li G, Liu Z, Yue H and Geng X: LncRNA HOTAIR suppresses cell apoptosis, autophagy and induces cell proliferation in cholangiocarcinoma by modulating the miR-204-5p/HMGB1 axis. Biomed Pharmacother. 130:1105662020. View Article : Google Scholar : PubMed/NCBI | |
|
Murakami K, Yokoi Y, Hirane N, Otaki M and Nishimura SI: Nanosomal irinotecan targeting pancreatic cancer cell surface neuraminidase-1 sialidase. Adv Healthc Mater. Jul 22–2025.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI | |
|
Wu H, Li A, Zheng Q, Gu J and Zhou W: LncRNA LZTS1-AS1 induces proliferation, metastasis and inhibits autophagy of pancreatic cancer cells through the miR-532/TWIST1 signaling pathway. Cancer Cell Int. 23:1302023. View Article : Google Scholar : PubMed/NCBI | |
|
Lv D, Xiang Y, Yang Q, Yao J and Dong Q: Long Non-Coding RNA TUG1 promotes cell proliferation and inhibits cell apoptosis, autophagy in clear cell renal cell carcinoma via MiR-31-5p/FLOT1 axis. Onco Targets Ther. 13:5857–5868. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Shao Q, Wang Q and Wang J: LncRNA SCAMP1 regulates ZEB1/JUN and autophagy to promote pediatric renal cell carcinoma under oxidative stress via miR-429. Biomed Pharmacother. 120:1094602019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Jia JP, Zhang YJ, Liu G, Zhou F and Zhang BC: Long Noncoding RNA ADAMTS9-AS2 inhibits the proliferation, migration, and invasion in bladder tumor cells. Onco Targets Ther. 13:7089–7100. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Li Q, Chen K, Dong R and Lu H: LncRNA CASC2 inhibits autophagy and promotes apoptosis in non-small cell lung cancer cells via regulating the miR-214/TRIM16 axis. RSC Adv. 8:40846–40855. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Peng D, Li W, Zhang B and Liu X: Overexpression of lncRNA SLC26A4-AS1 inhibits papillary thyroid carcinoma progression through recruiting ETS1 to promote ITPR1-mediated autophagy. J Cell Mol Med. 25:8148–8158. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Qin Y, Sun W, Wang Z, Dong W, He L, Zhang T, Lv C and Zhang H: RBM47/SNHG5/FOXO3 axis activates autophagy and inhibits cell proliferation in papillary thyroid carcinoma. Cell Death Dis. 13:2702022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Liu Z, Xu Z, Shao W, Hu D, Zhong H and Zhang J: Introduction of long non-coding RNAs to regulate autophagy-associated therapy resistance in cancer. Mol Biol Rep. 49:10761–10773. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Shaw A and Gullerova M: Home and away: The role of non-coding RNA in intracellular and intercellular DNA damage response. Genes (Basel). 12:14752021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Qiao C, Fang R, Yang S, Zhao G, Liu S and Li P: LncRNA CASC19: A novel oncogene involved in human cancer. Clin Transl Oncol. 25:2841–2851. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Mou J, Zhou G and Yuan C: CASC19: An oncogenic long non-coding RNA in different cancers. Curr Pharm Des. 30:1157–1166. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu H, Zheng W, Chen Q, Zhou Y, Pan Y, Zhang J, Bai Y and Shao C: lncRNA CASC19 contributes to radioresistance of nasopharyngeal carcinoma by promoting autophagy via AMPK-mTOR pathway. Int J Mol Sci. 22:14072021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu H, Chen Q, Zheng W, Zhou Y, Bai Y, Pan Y, Zhang J and Shao C: LncRNA CASC19 enhances the radioresistance of nasopharyngeal carcinoma by regulating the miR-340-3p/FKBP5 axis. Int J Mol Sci. 24:30472023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang WJ, Guo CA, Li R, Xu ZP, Yu JP, Ye Y, Zhao J, Wang J, Wang WA, Zhang A, et al: Long non-coding RNA CASC19 is associated with the progression and prognosis of advanced gastric cancer. Aging (Albany NY). 11:5829–5847. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hou Y, Tang Y, Ma C, Yu J and Jia Y: Overexpression of CASC19 contributes to tumor progression and predicts poor prognosis after radical resection in hepatocellular carcinoma. Dig Liver Dis. 55:799–806. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Teng S, Ge J, Yang Y, Cui Z, Min L, Li W, Yang G, Liu K and Wu J: M1 macrophages deliver CASC19 via exosomes to inhibit the proliferation and migration of colon cancer cells. Med Oncol. 41:2862024. View Article : Google Scholar : PubMed/NCBI | |
|
Chiu HS, Somvanshi S, Patel E, Chen TW, Singh VP, Zorman B, Patil SL, Pan Y, Chatterjee SS; Cancer Genome Atlas Research Network;, ; et al: Pan-cancer analysis of lncRNA regulation supports their targeting of cancer genes in each tumor context. Cell Rep. 23:297–312.e12. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Z, Li X, Wang X, Wu J, Gong Z, Kõks S and Wang M: Nuclear paraspeckle assembly transcript 1 promotes photophobia behavior in mice via miR-196a-5p/Trpm3 coupling. J Headache Pain. 26:1182025. View Article : Google Scholar : PubMed/NCBI | |
|
Sakaguchi H, Tsuchiya H, Kitagawa Y, Tanino T, Yoshida K, Uchida N and Shiota G: NEAT1 Confers radioresistance to hepatocellular carcinoma cells by inducing autophagy through GABARAP. Int J Mol Sci. 23:7112022. View Article : Google Scholar : PubMed/NCBI | |
|
Mahmoud HS, Eldesoky NA, Shaker OG and El-Husseiny AA: The diagnostic value of LncRNA NEAT1 targeting miR-129-5p in pancreatic cancer patients. Sci Rep. 15:276382025. View Article : Google Scholar : PubMed/NCBI | |
|
Dong P, Xiong Y, Yue J, Xu D, Ihira K, Konno Y, Kobayashi N, Todo Y and Watari H: Long noncoding RNA NEAT1 drives aggressive endometrial cancer progression via miR-361-regulated networks involving STAT3 and tumor microenvironment-related genes. J Exp Clin Cancer Res. 38:2952019. View Article : Google Scholar : PubMed/NCBI | |
|
Almujri SS and Almalki WH: The paradox of autophagy in cancer: NEAT1′s role in tumorigenesis and therapeutic resistance. Pathol Res Pract. 262:1555232024. View Article : Google Scholar : PubMed/NCBI | |
|
Wen J, Zheng W, Zeng L, Wang B, Chen D, Chen Y, Lu X, Shao C, Chen J and Fan M: LTF Induces Radioresistance by Promoting Autophagy and Forms an AMPK/SP2/NEAT1/miR-214-5p feedback loop in lung squamous cell carcinoma. Int J Biol Sci. 19:1509–1527. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
He H: Targeting TP53TG1: A promising prognostic biomarker and therapeutic target for personalized cancer therapy. Discov Oncol. 16:12712025. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Liu R, Hu H, Huang Y, Shi Y, Li H, Chen H, Cai M, Wang N, Yan T, et al: Methionine deprivation inhibits glioma proliferation and EMT via the TP53TG1/miR-96-5p/STK17B ceRNA pathway. NPJ Precis Oncol. 8:2702024. View Article : Google Scholar : PubMed/NCBI | |
|
Liao D, Liu X, Yuan X, Feng P, Ouyang Z, Liu Y and Li C: Long non-coding RNA tumor protein 53 target gene 1 promotes cervical cancer development via regulating microRNA-33a-5p to target forkhead box K2. Cell Cycle. 21:572–584. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ji W, Wang W and Wei Y: TP53TG1/STAT axis is involved in the development of colon cancer through affecting PD-L1 expression and immune escape mechanism of tumor cells. Am J Cancer Res. 13:5218–5235. 2023.PubMed/NCBI | |
|
Cheng Y, Huang N, Yin Q, Cheng C, Chen D, Gong C, Xiong H, Zhao J, Wang J, Li X, et al: LncRNA TP53TG1 plays an anti-oncogenic role in cervical cancer by synthetically regulating transcriptome profile in HeLa cells. Front Genet. 13:9810302022. View Article : Google Scholar : PubMed/NCBI | |
|
Luan F, Chen W, Chen M, Yan J, Chen H, Yu H, Liu T and Mo L: An autophagy-related long non-coding RNA signature for glioma. FEBS Open Bio. 9:653–667. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gao W, Qiao M and Luo K: Long noncoding RNA TP53TG1 contributes to radioresistance of glioma cells Via miR-524-5p/RAB5A Axis. Cancer Biother Radiopharm. 36:600–612. 2021.PubMed/NCBI | |
|
Wang C, Li Y, Yan S, Wang H, Shao X, Xiao M, Yang B, Qin G, Kong R, Chen R and Zhang N: Interactome analysis reveals that lncRNA HULC promotes aerobic glycolysis through LDHA and PKM2. Nat Commun. 11:31622020. View Article : Google Scholar : PubMed/NCBI | |
|
Gandhy SU, Imanirad P, Jin UH, Nair V, Hedrick E, Cheng Y, Corton JC, Kim K and Safe S: Specificity protein (Sp) transcription factors and metformin regulate expression of the long non-coding RNA HULC. Oncotarget. 6:26359–26372. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Lin J, Liu Y, Peng J, Cao Y, Su Z, Wang T, Cheng J and Hu D: AlncRNA HULC as an effective biomarker for surveillance of the outcome of cancer: A meta-analysis. PLoS One. 12:e01712102017. View Article : Google Scholar : PubMed/NCBI | |
|
Li YP, Liu Y, Xiao LM, Chen LK, Tao EX, Zeng EM and Xu CH: Induction of cancer cell stemness in glioma through glycolysis and the long noncoding RNA HULC-activated FOXM1/AGR2/HIF-1α axis. Lab Invest. 102:691–701. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Chang M, Tong L, Wang Y, Wang M and Wang F: Screening potential lncRNA biomarkers for breast cancer and colorectal cancer combining random walk and logistic matrix factorization. Front Genet. 13:10236152023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Wang K, Wang Q and Wang X: LncRNA HULC mediates radioresistance via autophagy in prostate cancer cells. Braz J Med Biol Res. 51:e70802018. View Article : Google Scholar : PubMed/NCBI | |
|
Hacisuleyman E, Goff LA, Trapnell C, Williams A, Henao-Mejia J, Sun L, McClanahan P, Hendrickson DG, Sauvageau M, Kelley DR, et al: Topological organization of multichromosomal regions by the long intergenic noncoding RNA Firre. Nat Struct Mol Biol. 21:198–206. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Meng X, Fang E, Zhao X and Feng J: Identification of prognostic long noncoding RNAs associated with spontaneous regression of neuroblastoma. Cancer Med. 9:3800–3815. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Shi X, Cui Z, Liu X, Wu S, Wu Y, Fang F and Zhao H: LncRNA FIRRE is activated by MYC and promotes the development of diffuse large B-cell lymphoma via Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun. 510:594–600. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Zhang J, Liu Y, Zhao R, Zhou X and Wang H: An integrated autophagy-related long noncoding RNA signature as a prognostic biomarker for human endometrial cancer: A bioinformatics-based approach. Biomed Res Int. 2020:57174982020. View Article : Google Scholar : PubMed/NCBI | |
|
Cai J, Wang R, Chen Y, Zhang C, Fu L and Fan C: LncRNA FIRRE regulated endometrial cancer radiotherapy sensitivity via the miR-199b-5p/SIRT1/BECN1 axis-mediated autophagy. Genomics. 116:1107502024. View Article : Google Scholar : PubMed/NCBI | |
|
Cory S, Graham M, Webb E, Corcoran L and Adams JM: Variant (6;15) translocations in murine plasmacytomas involve a chromosome 15 locus at least 72 kb from the c-myc oncogene. EMBO J. 4:675–681. 1985. View Article : Google Scholar : PubMed/NCBI | |
|
Tseng YY, Moriarity BS, Gong W, Akiyama R, Tiwari A, Kawakami H, Ronning P, Reuland B, Guenther K, Beadnell TC, et al: PVT1 dependence in cancer with MYC copy-number increase. Nature. 512:82–86. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Boloix A, Masanas M, Jiménez C, Antonelli R, Soriano A, Roma J, Sánchez de Toledo J, Gallego S and Segura MF: Long Non-coding RNA PVT1 as a prognostic and therapeutic target in pediatric cancer. Front Oncol. 9:11732019. View Article : Google Scholar : PubMed/NCBI | |
|
Ogunwobi OO and Segura MF: Editorial: PVT1 in cancer. Front Oncol. 10:5887862020. View Article : Google Scholar : PubMed/NCBI | |
|
Lai SW, Chen MY, Bamodu OA, Hsieh MS, Huang TY, Yeh CT, Lee WH and Cherng YG: Exosomal lncRNA PVT1/VEGFA axis promotes colon cancer metastasis and stemness by downregulation of tumor suppressor miR-152-3p. Oxid Med Cell Longev. 2021:99598072021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Y, Wu F, Gui W, Zhang N, Matro E, Zhu L, Eserberg DT and Lin X: A positive feedback regulatory loop involving the lncRNA PVT1 and HIF-1α in pancreatic cancer. J Mol Cell Biol. 13:676–689. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yao W, Li S, Liu R, Jiang M, Gao L, Lu Y, Liang X and Zhang H: Long non-coding RNA PVT1: A promising chemotherapy and radiotherapy sensitizer. Front Oncol. 12:9592082022. View Article : Google Scholar : PubMed/NCBI | |
|
Tan YT, Lin JF, Li T, Li JJ, Xu RH and Ju HQ: LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun (Lond). 41:109–120. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Y, Shao Y, Hu W, Zhang J, Shi Y, Kong X and Jiang J: A novel long noncoding RNA SP100-AS1 induces radioresistance of colorectal cancer via sponging miR-622 and stabilizing ATG3. Cell Death Differ. 30:111–124. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Guo Z, Zhang X, Zhu H, Zhong N, Luo X, Zhang Y, Tu F, Zhong J, Wang X, He J and Huang L: TELO2 induced progression of colorectal cancer by binding with RICTOR through mTORC2. Oncol Rep. 45:523–534. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chang S, Zhang M, Liu C, Li M, Lou Y and Tan H: Redox mechanism of glycerophospholipids and relevant targeted therapy in ferroptosis. Cell Death Discov. 11:3582025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Yu S, Wang C, Jia C, Lu Z and Chen J: Establishment of a non-coding RNAomics screening platform for the regulation of KRAS in pancreatic cancer by RNA sequencing. Int J Oncol. 53:2659–2670. 2018.PubMed/NCBI | |
|
Muthoni A, Anyango A and V Wanjala H: Mucilage-based nanocarriers for targeted cancer therapy-design, functionalization, and therapeutic potential. Drug Dev Ind Pharm. Aug 4–2025.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI | |
|
Wu T and Du Y: LncRNAs: From basic research to medical application. Int J Biol Sci. 13:295–307. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Chen H, Yang Y, Zhao L, Xie H, Li P, Lv X, He L, Liu N and Liu B: Targeting LINC02320 prevents colorectal cancer growth via GRB7-dependent inhibition of MAPK signaling pathway. Cell Mol Biol Lett. 30:862025. View Article : Google Scholar : PubMed/NCBI | |
|
Taylor MA, Das BC and Ray SK: Targeting autophagy for combating chemoresistance and radioresistance in glioblastoma. Apoptosis. 23:563–575. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Han F, Chen S, Zhang K, Zhang K, Wang M and Wang P: Targeting the HNRNPA2B1/HDGF/PTN axis to overcome radioresistance in non-small cell lung cancer. Antioxid Redox Signal. 43:189–214. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Chen X, Chen X, Liu J, Gu H, Fan R and Ge H: Long non-coding RNA HOTAIR knockdown enhances radiosensitivity through regulating microRNA-93/ATG12 axis in colorectal cancer. Cell Death Dis. 11:1752020. View Article : Google Scholar : PubMed/NCBI | |
|
Yan Y, Chen X, Wang X, Zhao Z, Hu W, Zeng S, Wei J, Yang X, Qian L, Zhou S, et al: The effects and the mechanisms of autophagy on the cancer-associated fibroblasts in cancer. J Exp Clin Cancer Res. 38:1712019. View Article : Google Scholar : PubMed/NCBI |