Skip to main content
American Journal of Clinical and Experimental Urology logoLink to American Journal of Clinical and Experimental Urology
. 2025 Apr 25;13(2):92–117. doi: 10.62347/QCPV6064

Dual role of autophagy in bone metastasis: mechanistic insights and therapeutic targeting

Xinyi Gu 1,*, Dejian Xiang 2,*, Haozhong Zhu 3, Xiaoqian He 1, Chenhui Yang 4,5, Rongjin Chen 4,5
PMCID: PMC12089224  PMID: 40400996

Abstract

Autophagy, a cellular degradation mechanism, plays a dual role in the progression and therapy of bone metastases in cancers, including prostate cancer. This review delves into the intricate roles of autophagy in both tumor suppression and progression, with a focus on its impact on bone metastasis and osteosarcoma (OS). Initially, autophagy acts as a tumor suppressor by eliminating damaged organelles and proteins, thus preventing tumor initiation. However, as cancer progresses, autophagy supports cancer cell survival under stress conditions, such as nutrient deprivation and hypoxia, and contributes to drug resistance. Specifically, in bone metastasis from breast and prostate cancers, autophagy facilitates tumor cell migration, invasion, and survival. The review also highlights the therapeutic potential of targeting autophagy in cancer treatment, especially in overcoming drug resistance in osteosarcoma, where autophagy modulation could reduce chemoresistance. By understanding the dual roles of autophagy in cancer and bone metastasis, new therapeutic strategies can be developed to target this process, offering hope for improved treatment outcomes in cancers prone to bone metastasis, including prostate cancer.

Keywords: Autophagy, bone neoplasms, osteosarcoma, neoplasm metastasis, drug resistance, prostate cancer

Introduction

Autophagy, an intracellular degradation process, is pivotal for maintaining cellular homeostasis and responding to various stressors [1,2]. The complex role of autophagy in both the development and progression of cancer has become increasingly clear. It serves to prevent tumor initiation by eliminating damaged organelles and proteins, yet paradoxically supports cancer cell survival and fosters drug resistance as the tumor progresses [3-5]. Hence, modulating autophagy mechanisms presents a potential target in cancer therapy.

An epidemiological study on cancer bone metastasis reveals significant disparities in incidence rates across different cancer types, with breast and prostate cancers being the most common sources of bone metastasis [6,7]. The ramifications of bone metastasis transcend survival implications, critically undermining patients’ quality of life through a spectrum of bone-related complications, including pathological fractures and spinal cord compression [8,9]. Emerging evidence suggests autophagy’s substantial influence on the genesis and progression of bone metastases, potentially through its modulation of tumor cell migration, invasion, and survival mechanisms [10].

Osteosarcoma (OS), the most prevalent type of bone sarcoma, represents a group of malignant neoplasms characterized by osteoid matrix production [11]. OS predominantly spreads through the hematogenous route, with the lungs being the primary site for metastasis [12-14]. The inclusion of chemotherapy in the treatment regimen for localized OS has significantly improved prognoses, increasing survival rates up to 70% [15]. However, drug resistance, both inherent and acquired, poses a significant clinical challenge, with patients exhibiting poor response and refractory disease facing limited effective options in second- or third-line chemotherapy [16-18]. Chemotherapy primarily works by triggering apoptosis in cancer cells, yet it can also stimulate autophagy within these cells [19,20]. Therefore, modulating autophagy in osteosarcoma (OS) cells to diminish their chemoresistance is a critical aspect of researching and developing innovative therapeutic approaches for OS.

Given the complex mechanisms of autophagy in cancer bone metastasis and osteosarcoma, involving various intracellular signaling pathways, a thorough understanding of these mechanisms is crucial for developing new therapeutic strategies. With advancing research into autophagy mechanisms, clinical exploration of autophagy-based therapeutic strategies has begun, which could potentially offer new directions for the treatment of cancer bone metastasis and osteosarcoma. This review will focus on the role and mechanisms of autophagy in cancer bone metastasis and osteosarcoma, exploring potential therapeutic strategies and providing a reference for future research and clinical applications.

Processes of autophagy

Molecular mechanisms of autophagy: insights into cellular self-digestion processes

Autophagy is a self-digestive process that plays a crucial role in cellular survival under conditions of external stress, hypoxia, and starvation [21-23]. It functions by clearing damaged organelles and long-lived proteins, thereby maintaining cellular homeostasis. The autophagic process encompasses macroautophagy, microautophagy, and chaperone-mediated autophagy, with macroautophagy being predominantly observed in the context of cancer [24-26]. Autophagosomes, double-membraned vesicles in the cytoplasm, are named based on their morphological characteristics and are regulated by a set of autophagy-related proteins (ATGs) [27,28].

The autophagic process unfolds in four distinct steps. Initially, the formation of the phagophore is triggered by a complex composed of autophagy-related genes, including ATG1, ATG13, and ATG17, necessitating the involvement of the membrane protein ATG9 [29,30]. The complex formation of Class III phosphatidylinositol 3-kinase (PI3K-III) and vacuolar protein sorting 34 (Vps34) with Beclin-1 (ATG6) further facilitates the development of the phagophore. Mitophagy, the selective autophagic degradation of mitochondria, begins with the interaction between B-cell lymphoma 2 (Bcl-2) and adenovirus E1B 19-kDa interacting protein 3 (BNIP3), which upon binding to Bcl-2, releases Beclin-1, leading to the formation of a Beclin-1-ATG14-PI3K-III complex [31,32].

Subsequently, the assembly process within the phagophore is completed. ATG12 is covalently bonded to ATG5 through the action of ATG10, which is transported from ATG12 after being activated by ATG7 [33-35]. This ATG12-ATG5 conjugate promotes the elongation and closure of the phagophore to form the autophagosome [36-38]. The formation of the autophagosome also involves the conversion of LC3-I to LC3-II, mediated by ATG4. LC3, a mammalian homolog of yeast ATG8, is conjugated to phosphatidylethanolamine (PE) through the action of ATG7 and ATG3, resulting in LC3-II [38-40]. LC3-II becomes an integral part of the autophagosomal membrane. Finally, the autophagosome fuses with lysosomes to form a mature autolysosome, where the engulfed cytoplasmic proteins and organelles are degraded by lysosomal hydrolases.

This intricate process of autophagy illustrates the cell’s sophisticated regulatory mechanisms in response to environmental changes, highlighting its vital role in cellular function and survival. Understanding the molecular intricacies of autophagy not only sheds light on basic cellular physiology but also opens avenues for therapeutic interventions in diseases where autophagy is dysregulated, such as cancer.

Regulatory molecular mechanisms of autophagy: deciphering the control of cellular self-eating processes

Autophagy is a crucial intracellular clearance mechanism that aids cells in coping with external stresses, such as starvation, ischemia-hypoxia, oxidative stress, calcium homeostasis imbalance, and mitochondrial dysfunction, by removing damaged organelles and long-lived proteins. The autophagic process is regulated by multiple signaling pathways, including key pathways influenced by mTOR (mammalian target of rapamycin), Beclin-1, and p53.

mTOR signaling pathway

mTOR is a pivotal regulator of cell growth and metabolism, belonging to the phosphatidylinositol kinase-related kinase (PIKK) family [41,42]. It regulates cell growth, metabolism, and autophagy by sensing ATP, hormones, and amino acid levels. In the regulation of autophagy, mTOR acts as a negative regulator [43]. Studies have shown that the downstream ribosomal protein S6 (p70S6) of the mTOR signaling pathway inhibits autophagy, while rapamycin induces autophagy by inhibiting mTOR activity, thereby reducing the activity of p70S6 [44,45]. There are two forms of mTOR: mTORC1 and mTORC2, with mTORC1 being most closely related to autophagy regulation and the focus of current research.

Beclin-1 signaling pathway

Beclin-1 is a key regulatory factor in the autophagic process, homologous to Atg6/Vps30 in yeast [46-48]. By forming a complex with Class III PI3K, Beclin-1 regulates the activity of other autophagy-related genes, promoting the formation and maturation of autophagosomes. The expression level of Beclin-1 is closely related to the survival and therapeutic response of cancer cells, making the Beclin-1-regulated autophagic process a potential target for cancer treatment [49-51].

p53 signaling pathway

p53 is a critical tumor suppressor gene that regulates cellular autophagy in multiple ways [52-54]. In the nucleus, p53 can upregulate the level of cellular autophagy; in the cytoplasm, however, p53 may inhibit autophagy. Thus, p53 plays a dual regulatory role in cellular autophagy, with its specific mechanisms requiring further investigation [55,56].

Other signaling pathways

In addition to the aforementioned pathways, the PI3K signaling pathway, hypoxia-inducible factors (HIF), AMPK, REDD1, and Tsc1/Tsc2 also participate in regulating cellular autophagy. For example, hypoxic conditions can induce autophagy through HIF-dependent or -independent mTOR mechanisms to adapt to the stress caused by low oxygen on the endoplasmic reticulum and mitochondria.

The molecular mechanisms regulating cellular autophagy involve multiple signaling pathways and regulatory factors. These mechanisms work together, enabling cells to adapt to continuously changing internal and external environments. By delving deeper into these molecular mechanisms, not only can our understanding of the cellular autophagy process be enhanced, but new strategies for treating diseases such as cancer may also be developed (Figure 1).

Figure 1.

Figure 1

The occurrence and regulation process of autophagy. ATGs, autophagy-related proteins; Bcl-2, B-cell lymphoma 2; BNIP3, adenovirus E1B 19-kDa interacting protein 3; mTOR, mammalian target of rapamycin; PI3K-III, Class III phosphatidylinositol 3-kinase; PE, phosphatidylethanolamine; Vps34, vacuolar protein sorting 34. Created by Figdraw.com (https://www.figdraw.com).

The dual role of autophagy in cancer

Autophagy in tumor suppression

Autophagy, a self-degradative process within cells, plays a crucial role in suppressing tumorigenesis by eliminating misfolded proteins, damaged organelles, and reactive oxygen species (ROS), thereby mitigating chronic tissue damage and preventing tumor development, particularly in early tumorigenesis.

Maintenance of genomic integrity

Beclin-1, a tumor suppressor, induces autophagy by binding to and activating Vps34, exerting antitumor effects [57]. Studies have observed a monoallelic deletion of Beclin-1 in 40-75% of breast, ovarian, and prostate cancers, with an increased tumor incidence in Beclin-1+/- mouse models [48,58]. Furthermore, frame-shift mutations in certain autophagy-related genes have been identified in gastric and colorectal cancers, contributing to tumorigenesis by deregulating autophagy [59]. The phosphatase gene PTEN, the first identified tumor suppressor gene with lipid phosphatase activity, upregulates autophagy by inhibiting the PI3K/Akt pathway [60,61]. Consequently, mutations in the PTEN gene or activation of Akt suppress autophagy. Clinical studies have found PTEN gene mutations in samples from 135 patients with gastric, colorectal, and hepatocellular carcinomas, highlighting autophagy’s role in limiting genomic damage and mutations, thus inhibiting early tumor development [62,63].

Suppression of necrotic inflammation

Chronic inflammation is a significant factor in early tumor development. Inflammatory cytokines regulate autophagic responses by binding to specific receptors on the cytoplasmic membrane [64,65]. Activation of oncogenes, due to increased oxidative stress, creates a pro-tumorigenic inflammatory microenvironment [66,67]. Similarly, inhibiting autophagy in defective cancer cells increases cell death and pro-inflammatory cytokine secretion secretion (e.g., HMGB1), exacerbating inflammation [68]. As a central inflammasome regulator, autophagy prevents tissue necrosis, chronic inflammation, and genomic instability, thereby reducing cancer susceptibility [69,70].

Reduction of p62/SQSTM1 accumulation

The multifunctional ubiquitin-binding protein p62/SQSTM1, involved in both the ubiquitin-proteasome and autophagy-lysosome degradation pathways, selectively degrades misfolded proteins [71,72]. p62 directly interacts with LC3, inducing its autophagic degradation [73]. Lack of Atg expression or impaired autophagosome-lysosome fusion leads to significant p62 protein accumulation, causing cytotoxicity, oxidative stress, and DNA damage closely associated with tumor progression [74,75].

Autophagy in tumor progression

In the later stages of tumor progression, environmental stresses such as limited angiogenesis, nutrient deprivation, and hypoxia induce autophagy. This process supports established tumors survive and grow by degrading cellular contents and macromolecules for recycling, thereby enhancing tumor invasiveness and metastasis. Additionally, as a cellular defense mechanism, autophagy reduces the clinical effectiveness of most anticancer treatments.

Prevention of cellular damage

Under stress conditions, autophagy protects cells by clearing damaged mitochondria, inhibiting DNA damage, maintaining genomic stability, and limiting inflammatory responses [76,77]. Mitochondria are a primary source of ROS. A deficiency in ATG proteins leads to accumulation of damaged mitochondria, excessive ROS production, DNA damage, and genomic instability [78,79]. Autophagy suppresses ROS-induced DNA damage and provides nucleotides for DNA replication and repair through its recycling function.

Promotion of cell metastasis

During advanced tumor progression, autophagy facilitates dissemination of malignant cells in the circulatory system, enhances colonization in target organs/tissues, induces dormancy, and enables survival in new microenvironments, with upregulated autophagy levels observed during metastasis [80,81]. Elevated autophagy levels have been confirmed in the metastasis of breast cancer, melanoma, hepatocellular carcinoma, and glioblastoma [82].

Resistance to radio- and chemotherapy

Most anticancer therapies, including radiotherapy, chemotherapy, histone deacetylase inhibitors (colon cancer), temozolomide/rapamycin (malignant glioma), γ-ray irradiation (breast/prostate/colon cancers), resveratrol (ovarian cancer), and tamoxifen (breast cancer/glioblastoma), can induce autophagy [83,84]. As a pro-survival mechanism, autophagy drives treatment-resistant cancer cells into dormancy, promotes tumor relapse/metastasis, and diminishes therapeutic efficacy [85,86]. Given autophagy’s protective role, combining autophagy inhibitors with conventional therapies may improve clinical outcomes [87,88].

The relationship between autophagy and bone metastasis in cancer

The bone microenvironment’s influence on cancer cell autophagy

The bone microenvironment plays a pivotal role in the regulation of autophagy in cancer cells. Tumor cells within the bone microenvironment exhibit aberrant expression of bone development-related genes, altering cellular activities and promoting tumor progression [89]. Cytokines and growth factors in the bone microenvironment enhance cancer cell survival and drug resistance by activating autophagy pathways (e.g., upregulating LC3-II and p62/SQSTM1), thereby increasing treatment tolerance [90]. The bone matrix, acting as a fertile niche for cancer cells, provides physical support via the extracellular matrix (ECM) and regulates autophagy through bidirectional interactions [91]. Osteoblasts and osteoclasts critically modulate autophagy in the bone microenvironment. Osteoblast-derived RANKL (Receptor Activator of Nuclear Factor κB Ligand) promotes osteoclast formation and activity, indirectly influencing cancer cell autophagy [92,93]. Osteoclasts release bioactive molecules (e.g., TGF-β, IGF-1) during bone resorption, which regulate autophagy pathways to support cancer cell survival and proliferation [94]. Intercellular communication within the bone microenvironment is essential for autophagy regulation. Osteoblasts, osteoclasts, and bone marrow stromal cells exchange materials and signals with cancer cells via exosomes and microvesicles, containing microRNAs (e.g., miR-21, miR-30a) and proteins (e.g., HSP90) that modulate autophagy-related gene expression and signaling [95-97].

Autophagy’s regulatory role in cancer cell proliferation and migration during bone metastasis

During the process of bone metastasis, autophagy influences cancer cell proliferation by regulating intracellular energy metabolism and levels of reactive oxygen species (ROS), and tumor cells remove damaged mitochondria through autophagy within the bone microenvironment, thereby maintaining cellular energy balance and viability [98,99]. However, excessive activation of autophagy might enhance the migratory capabilities of tumor cells, facilitating bone metastasis [100]. Moreover, the interaction between osteoblasts and osteoclasts within the bone microenvironment can also affect cancer cell migration by modulating autophagy. Secretions from osteoblasts, for example, can activate autophagy and promote tumor cell metastasis [101]. In some cancer types, the activation of autophagy may lead to cytoskeletal reorganization and enhanced cell adhesion, limiting tumor cell migration [102]. Furthermore, autophagy could suppress tumor invasion and bone metastasis through mechanisms such as inducing apoptosis or inhibiting epithelial-mesenchymal transition (EMT) [103]. This dual role of autophagy makes it a significant target in cancer treatment research.

Current clinical targeted therapies affecting the skeletal system

In the realm of oncological therapeutics, a diverse array of drugs has been developed targeting specific molecular pathways to inhibit cancer progression and metastasis, particularly in bone. These drugs, while primarily focused on their direct anticancer effects, also exhibit potential implications for autophagy. Denosumab [104-107], a drug used in clinical routines, targets RANKL to inhibit osteoclast formation and activity. This action not only reduces bone resorption but also potentially impacts metastatic tumor growth within the bone. Although its direct effect on autophagy is not specified, the mechanism suggests a possible influence on cellular processes related to cancer progression.

Vemurafenib and Dabrafenib [108], both targeting BRAF in melanoma, are in clinical application and share a similar mechanism that affects multiple pathways in cancer cells, which could potentially alter autophagy. Similarly, Erlotinib [109] and Crizotinib [108], targeting EGFR and ALK in NSCLC respectively, are noted for their roles in reducing skeletal-related events and exhibiting antitumor efficacy, which may indirectly influence autophagy through effects on bone remodeling. Cabozantinib [110-112], with its targets including VEGFR2, AXL, and MET, is recognized for its capacity to reduce tumor-induced osteolysis. Moreover, Everolimus [113,114], currently in clinical trials and targeting mTOR, is one of the few drugs explicitly noted for its autophagy inhibition activity. By inducing osteoprotegerin expression and promoting apoptosis of osteoclasts, it offers a clear link between its primary mechanism and potential effects on autophagy. Imatinib [115,116], targeting C-KIT and PDGFR, exhibits dual activity that affects osteoblast activity and, consequently, may influence autophagy through its impact on bone remodeling. This dual activity provides an intriguing insight into the complex interplay between cancer therapeutics, bone metabolism, and autophagy (Table 1).

Table 1.

Current clinical targeted therapies affecting the skeletal system

Drug Status of Application Target Autophagy Activity Mechanism Reference
Denosumab Clinical Routine RANKL Not Specified Inhibiting osteoclast formation and activity, reducing bone resorption and metastatic tumor growth in bone [104-107]
Vemurafenib Clinical Application BRAF in Melanoma Not Specified Affecting multiple pathways in cancer cells, potentially altering autophagy [108]
Dabrafenib Clinical Application BRAF in Melanoma Not Specified Similar to Vemurafenib, affecting cancer cell pathways, potentially impacting autophagy [108]
Erlotinib Clinical Application EGFR in NSCLC Not Specified Reducing SREs in bone metastasis, suggesting an impact on bone remodeling and possibly autophagy [109]
Crizotinib Clinical Application ALK in NSCLC Not Specified May influence bone remodeling through its antitumor efficacy, indirectly affecting autophagy [108]
Cabozantinib Clinical Application VEGFR2, AXL, MET Not Specified Reducing tumor-induced osteolysis, suggesting an effect on bone remodeling and autophagy [110-112]
Everolimus Clinical Trials mTOR Inhibition Inducing osteoprotegerin expression and apoptosis of osteoclasts, potentially affecting autophagy [113,114]
Imatinib Clinical Application C-KIT, PDGFR Dual Activity Affecting osteoblast activity, potentially influencing autophagy through bone remodeling [115,116]

Autophagy-related medications for common cancers with bone metastasis

Bone metastases are predominantly linked with certain cancers, such as breast cancer (70%), prostate cancer (85%), lung cancer (40%), renal cancer (40%), and multiple myeloma (95%) [117]. The prevalent incidence of breast, lung, and prostate cancers contributes to more than 80% of metastatic bone disease cases [118]. This section reviews autophagy-modulating therapies for these cancers.

Breast cancer

Autophagy modulation has emerged as a strategy to enhance conventional breast cancer therapies. Autophagy inhibitors such as bafilomycin A1 (BafA1) and chloroquine (CQ) [119,120] has demonstrated increased radiation sensitivity in breast cancer cells, highlighting the potential of targeting autophagy to overcome radioresistance. The PI3K/AKT pathway, known for its role in promoting tumor growth and survival, has been targeted by inhibitors such as ipatasertib and taselisib [121]. Hormonal therapies like tamoxifen [122], which have been the mainstay for treating estrogen receptor-positive (ER+) breast cancer, stimulate autophagy, contributing to both cell survival and resistance mechanisms. Similarly, Lapatinib [123], a HER2/EGFR inhibitor, exhibits a dual role in autophagy regulation, where short-term treatment induces apoptosis through autophagy, whereas long-term treatment fosters protective autophagy, leading to resistance. Trastuzumab and its conjugate, trastuzumab emtansine (T-DM1) [124,125], widely used in HER2+ breast cancer treatment, also interact with autophagy pathways, contributing to their cytotoxic effects. Overall, the strategic modulation of autophagy in breast cancer therapy offers a promising avenue for enhancing the efficacy of existing treatments and overcoming resistance. Further research and clinical trials are warranted to fully elucidate the mechanisms and optimize the therapeutic strategies involving autophagy modulators.

The exploration of phytochemicals as potential therapeutic agents in breast cancer treatment has unveiled a diverse array of natural compounds capable of modulating autophagy. These phytochemicals, derived from various plants, exhibit a wide range of molecular effects on breast cancer cells, including the induction or inhibition of autophagy, apoptosis induction, cell cycle arrest, and sensitization to traditional chemotherapy. Artemisinin and its derivatives [126-129], for instance, have been shown to up-regulate autophagy markers like Beclin-1 and LC3-II, enhancing the sensitivity of breast cancer cells to chemotherapy drugs such as epirubicin. Baicalein [130-133], another phytochemical, suppresses cell proliferation and triggers both apoptosis and autophagy by modulating the PI3K/AKT/mTOR pathway, demonstrating its potential as a chemosensitizer in breast cancer therapy. Britannin [134,135] and gaillardin [136], by down-regulating autophagy markers, induce apoptosis, suggesting a role in suppressing autophagy to enhance cancer cell death. Celastrol [137-139], known for its antiproliferative activity, elevates the expression of autophagy markers, indicating its potential to induce autophagy as a mechanism of action against breast cancer cells. Cucurbitacin B [140-142] and curcumin [143-145] have been noted for their ability to induce autophagy through various mechanisms, including ROS-mediated DNA damage and inactivation of the PI3K/Akt/mTOR pathway, respectively. These mechanisms highlight the multifaceted roles of phytochemicals in targeting autophagy pathways to combat breast cancer. Icariin [146-148], paclitaxel [149-151], and resveratrol [152-154] exhibit dual activities in modulating autophagy, demonstrating the complexity of autophagy as a therapeutic target. Tetrandrine [155-157], thymoquinone [158-160], and tocotrienols [161-163] activate autophagy, contributing to decreased cancer cell viability and enhanced sensitivity to chemotherapy. Ursolic acid [164-166] emerges as a potent inducer of cytotoxic autophagy and apoptosis by affecting AKT signaling and glycolysis, underscoring the potential of natural compounds in overcoming resistance to conventional therapies (Table 2).

Table 2.

Autophagy-related medications for breast cancer

Drug Application Status Target Tumor or cell Type Autophagy Activity Mechanism Reference
BafA1 Research Autophagosome Breast cancer Inhibition Enhancing radiation-induced death in breast cancer cells by likely modulating endoplasmic stress and mTOR pathway [119]
Chloroquine (CQ) Clinical Application Autophagosome Breast cancer Inhibition Boosting radiation sensitivity by suppressing TAK1; improving cytotoxicity of chemotherapy in resistant tumors [119,120]
Ipatasertib Preclinical & Clinical Trials PI3K/AKT Breast cancer Induction Induces autophagy in TNBC, enhancing antitumor activity of PI3K/AKT suppressors in combination with CQ [121]
Taselisib Preclinical & Clinical Trials PI3K/AKT Breast cancer Induction Similar to Ipatasertib, inducing autophagy in TNBC, potentiating antitumor effects of chemotherapy with CQ [121]
Tamoxifen Clinical Application ER Breast cancer Induction Induces autophagy in ER+ breast cancer cells, potentially contributing to cell survival and resistance [122]
Lapatinib Clinical Application HER2/EGFR Breast cancer Dual Activity Short-term treatment induces apoptosis via autophagy; long-term treatment leads to protective autophagy [123]
Trastuzumab Clinical Application HER2 Breast cancer Not Specified Autophagy plays a significant role in the cytotoxic effects of trastuzumab and T-DM1 on HER2+ breast cancer cells [124,125]
Artemisinin Research Beclin-1, P21 MCF-7, MDA-MB-231 Up-regulation Inducing autophagy by up-regulating Beclin-1 and P21, leading to increased sensitivity to chemotherapy and apoptosis [126-129]
Baicalein Research PI3K/AKT, mTOR MCF-7, MDA-MB-231 Activation Suppressing cell proliferation and triggers apoptosis and Inducing autophagy by modulating the PI3K/AKT/mTOR pathway [130-133]
Britannin Research ATG1, ATG4, ATG5 MCF-7 Inhibition Inducing apoptosis and inhibits autophagy by down-regulating autophagy markers such as ATG4, ATG5, Beclin1, and LC-III [134,135]
Celastrol Research LC3 A/B, p62 MCF-7 Activation Exhibits antiproliferative activity and inhibits colony formation by elevating expression of autophagy markers LC3 A/B, p62, and Beclin-1 [137-139]
Cucurbitacin B Research LC3 II, p-mTOR MCF-7 Activation Decreasing cell viability and inducing DNA damage and autophagy by up-regulating LC3 II and inhibiting p-mTOR, p-Akt, and p62 expressions [140-142]
Curcumin Research PI3K/Akt/mTOR MCF7, MDA-MB-231 Activation Sensitizing MDR breast cancer cells to cisplatin and inducing apoptosis and autophagy by down-regulating CCAT1 and inactivating the PI3K/Akt/mTOR pathway [143-145]
Gaillardin Research ATG1, ATG4, ATG5 MCF-7 Inhibition Inducing apoptosis and inhibiting autophagy by down-regulating autophagy markers such as ATG1, ATG4, ATG5, Beclin1, and LC-III [136]
Icariin Research CDK2, CDK4 MCF-7, MDA-MB-231 Dual Activity Inducing cell cycle arrest and apoptosis through suppression of autophagy in TAM-resistant cells and enhances autophagy in other contexts [146-148]
Paclitaxel Research Beclin1 BT474 Inhibition Inhibiting tumor growth by reducing the expression of Beclin1 [149-151]
Resveratrol Research AKT, mTORC1 MDA-MB-231, MCF-7 Dual Activity Sensitizing breast cancer cells to talazoparib and inducing apoptosis by suppressing AKT signaling and inhibiting autophagy flux [152-154]
Tetrandrine Research PI3K/AKT/mTOR MDA-MB-231, MCF-7 Activation Blocks cell proliferation and stimulating autophagy and apoptosis by elevating Beclin1, LC3-II/LC3-I and reducing p62/SQSTM1 [155-157]
Thymoquinone Research Beclin-1, LC3 MCF-7, MDA-MB-231 Dual Activity Inhibiting autophagy and cell migration in some contexts, while in others, it increases autophagic vesicles and sensitizes cells to PTX [158-160]
Tocotrienols Research Beclin-1, LC3B MCF-7, MDA-MB-231 Activation Decreasing cancer cell viability by increasing markers of both early and late phase autophagy [161-163]
Ursolic acid Research PI3K, AKT MCF-7, MDA-MB-231 Activation Inducing cytotoxic autophagy and apoptosis by diminishing AKT signaling and affecting glycolysis [164-166]

These findings underscore the potential of phytochemicals as adjunctive or alternative therapies in breast cancer treatment, offering new avenues for overcoming drug resistance and enhancing therapeutic outcomes through the modulation of autophagy.

Prostate cancer

In the realm of urological system cancer therapies, particularly focusing on prostate cancer (PC), the modulation of autophagy through small-molecule compounds represents a promising therapeutic strategy. BML-275 [167,168], also known as Dorsomorphin, serves a dual role by inhibiting AMPK and mTOR, leading to autophagy inhibition and activation, respectively, and demonstrating antitumor activities in PC models. Metformin [169], a well-known AMPK activator, has been shown to induce autophagy in PC, highlighting its potential beyond its traditional use in treating diabetes. The classical autophagy inhibitor 3-MA [170,171] and the ULK1 inhibitor SBI-0206965 [172] demonstrate the therapeutic potential of autophagy inhibition in PC by disrupting autophagy-dependent processes and inducing apoptosis. The ATG4B inhibitors such as 21f [173] and DHA [174], show efficacy in inhibiting autophagy, leading to cell death in PC.

Marchantin M [175] display the inhibition of autophagy-mediated epithelial-mesenchymal transition (EMT) and activation of PERK/eIF2α-associated autophagy in PC. NCL1 [176], an LSD1 inhibitor, activates autophagy in PC, offering a novel approach to targeting autophagy regulation. Further research and clinical trials are essential to fully understand the therapeutic potential and mechanisms of these small-molecule compounds in modulating autophagy for cancer therapy (Table 3).

Table 3.

Autophagy-related medications for prostate and lung cancer

Drug Application Status Target/Pathway Tumor Type Autophagy Activity Mechanism Reference
BML-275 (Dorsomorphin) Preclinical AMPK inhibitor, mTOR inhibitor PC Inhibition/Activation Inhibiting autophagy via AMPK; Induces mitochondria-mediated apoptosis and autophagy via mTOR [167,168]
Metformin Clinical AMPK activator PC Activation Activating AMPK-induced autophagy by binding to PEN2 [169]
3-MA Preclinical PI3K inhibitor PC Inhibition Suppressing ATG5-dependent autophagy and autophagy-dependent exocytosis by inhibiting PI3K [170,171]
SBI-0206965 Preclinical ULK1 inhibitor PC Inhibition Inhibiting autophagy, PPP flux, and ROS clearance, inducing apoptosis [172]
21f Preclinical ATG4B inhibitor PC Inhibition Reducing LC3II/I levels and p62 degradation, inhibiting cellular autophagy [173]
Docosahexaenoic acid (DHA) Preclinical ATG4B inhibitor PC Inhibition Inhibiting ARSI-induced autophagy through mitochondrial dysfunction [174]
Marchantin M Preclinical 20S proteasome inhibitor PC Inhibition Activating PERK/eIF2 associated autophagy by inhibiting the 20S proteasome [175]
Sunitinib Clinical RTK inhibitor RCC Activation Inducing autophagy by activating ERK1/2 and inhibiting mTOR/p70S6K [231]
NCL1 Preclinical LSD1 inhibitor PC Activation Activating autophagy by inducing H3K4me2 accumulation at the P21 promoter [176]
Dihydroartemisinin (DHA) Research MAPK, mTOR/HIF-1 NSCLC Activation/Inhibition Inducing excessive autophagic cell death via HMGB1/MAPK signaling; Inhibits mitophagy to decrease radiation resistance; Induces ROS-dependent apoptosis [177,178]
Curcumin Research PI3K/Akt/mTOR, IP3R, miR-192-5p NSCLC Activation Initiating autophagy by causing mitochondrial damage; Triggers apoptosis via calcium overload and caspase-3 activation [179-181]
Baicalein Research MAP4K3/mTORC1/TFEB, PI3K/Akt/NF-kB NSCLC Activation Regulating autophagy via MAP4K3 inhibition; Induces apoptosis by downregulating Bcl-2 and caspase-3 proform; Enhances chemosensitivity to cisplatin [182-184]
Ginsenosides Research ATF4-CHOP-AKT1-mTOR, PI3K/Akt, PINK1-Parkin NSCLC Activation Enhancing autophagy flux and induces autophagic cell death; Promotes apoptosis and mitophagy via ROS and PINK1-Parkin activation [185-188]

Lung cancers

In the quest to combat non-small cell lung cancer (NSCLC), researchers have explored various natural compounds for their potential therapeutic effects. Dihydroartemisinin (DHA) [177,178], a derivative of artemisinin, has demonstrated a multifaceted approach in NSCLC treatment. It has been shown to initiate excessive autophagic cell death via HMGB1/MAPK signaling and inhibit mitophagy to decrease radiation resistance, thereby enhancing the therapeutic effect against NSCLC. Moreover, DHA induces ROS-dependent apoptosis, showcasing its potential as a comprehensive anticancer agent. Curcumin [179-181], derived from Curcuma longa, is recognized for its anti-inflammatory and antioxidant properties. In NSCLC, Curcumin has been found to initiate autophagy by causing mitochondrial damage and triggering apoptosis through calcium overload and caspase-3 activation. Its ability to modulate the PI3K/Akt/mTOR pathway further underscores its therapeutic potential in NSCLC by enhancing autophagy and promoting apoptosis. Baicalein [182-184], a flavonoid from Scutellaria baicalensis roots, exhibits anti-tumor activity by regulating autophagy and apoptosis. It directly binds to and inactivates MAP4K3, leading to TFEB-dependent autophagy and tumor growth inhibition. Baicalein also induces apoptosis by downregulating Bcl-2 and caspase-3 proform, and it enhances NSCLC chemosensitivity to cisplatin through the PI3K/Akt/NF-kB pathway. Ginsenosides [185-188], key constituents of ginseng, have been studied for their anticancer effects, which include cell cycle arrest, apoptosis, and autophagy induction. They enhance autophagy flux and induce autophagic cell death in NSCLC cells through ATF4-CHOP-AKT1-mTOR axis activation. Ginsenosides also promote apoptosis and mitophagy via ROS and the PINK1-Parkin signaling pathway, highlighting their role in NSCLC therapy.

By targeting various signaling pathways, these compounds not only inhibit tumor progression but also enhance the sensitivity of cancer cells to conventional treatments, offering a promising avenue for future therapeutic strategies against NSCLC. Further clinical research and trials are essential to validate these preclinical findings and to determine the efficacy and safety of these compounds in human subjects (Table 3).

The interaction between autophagy and osteosarcoma

Autophagic activity in osteosarcoma cells

Autophagy, a cellular self-degradation process, plays a crucial role in osteosarcoma cells. Studies have shown that autophagic activity is significantly enhanced in osteosarcoma cells, closely associated with tumor initiation and progression. Specifically, autophagy facilitates tumor cell survival and proliferation by maintaining intracellular homeostasis through the clearance of damaged organelles and proteins. For instance, autophagy-related genes such as Beclin1 and LC3B are found to be highly expressed in osteosarcoma patients, correlating with poor prognosis [189]. Moreover, research has identified that Sirt1 promotes autophagic activity in osteosarcoma cells by regulating the phosphorylation of histone H3, potentially influencing tumor progression [190]. Therefore, autophagy serves not only as a mechanism for osteosarcoma cells to adapt to microenvironmental stress but also as a vital support for tumor cell survival.

The role of autophagy in osteosarcoma drug resistance

Autophagy plays a dual role in the drug resistance of osteosarcoma. While autophagy can protect cells by eliminating intracellular toxic substances, it may also promote resistance to chemotherapy drugs in certain contexts. For example, chemotherapeutic agents like Doxorubicin can induce autophagy in osteosarcoma cells, a response closely linked to the development of drug resistance [191]. Furthermore, miR-331-3p has been found to promote chemotherapeutic resistance by regulating the autophagy process, highlighting the significant regulatory role of autophagy in the drug tolerance of osteosarcoma cells [192]. Therefore, therapeutic strategies targeting autophagy, such as the application of autophagy inhibitors, may offer new insights and approaches to overcoming osteosarcoma resistance. By modulating autophagic activity, it is hoped that the sensitivity of osteosarcoma patients to chemotherapy can be enhanced, thereby improving treatment outcomes.

Adjusting autophagy in osteosarcoma treatment strategies

The modulation of autophagy in osteosarcoma therapy involves a diverse range of pharmacological inhibitors and inducers targeting various components of the autophagy pathway. Inhibitors like LY294002 [193], 3-MA [193,194], and Wortmannin [193] act on early stages of autophagy by targeting the PI3K pathway, thereby improving the chemosensitivity of OS cells. Late-stage inhibitors such as Chloroquine and Hydroxychloroquine [195,196] target the fusion of autophagosomes with lysosomes, leading to cell death independent of autophagy. The use of specific VPS34 inhibitors like Spautin-1 [197] and SAR405 [198], as well as ATG4B inhibitors NSC185058 [196] and NSC377071 [196], demonstrates the potential of targeting specific autophagy-related proteins for therapeutic benefits in OS. Verteporfin is noted for its ability to disrupt early autophagic processes, induce lysosomal instability, and inhibit autophagic flux, presenting a multifaceted approach to inhibiting autophagy at multiple levels [199]. Bafilomycin A1 targets lysosomal acidification, a crucial step for autophagosome maturation, and has shown cytotoxic effects in OS cells when combined with other inhibitors, highlighting its role in hindering the autophagy process [200]. NBDHEX and MC3181 [201], both acting through JNK activation, lead to autophagic impairment and subsequent cell death, underscoring the potential of targeting the JNK pathway as a means to disrupt autophagy and promote cancer cell death in OS.

Conversely, inducers of autophagy, such as EGCG [202,203], Rapamycin [204], and its analogs Temsirolimus [205], aim to increase tumor autophagy, thereby reducing tumor growth and inducing cell death. Metformin [206] and PF-06409577 [207], which activate AMPK, represent another strategy for inducing autophagy, with the latter showing more potent effects than traditional activators.

This comprehensive approach to modulating autophagy in osteosarcoma therapy highlights the complexity of targeting autophagy for therapeutic benefits. Given the dual role of autophagy in cancer - both suppressive and promotive - it is imperative to carefully consider the context in which autophagy modulation is applied. Future research should focus on elucidating the precise mechanisms of action of these drugs, optimizing their delivery and efficacy, and evaluating their safety and therapeutic potential in clinical trials.

It’s evident that natural products offer a diverse arsenal against osteosarcoma, targeting various pathways to inhibit tumor growth, induce apoptosis, and modulate autophagy. Oridonin [208-210] stands out for its ability to induce mitochondria-mediated apoptosis and modulate several signaling pathways, including p38 MAPK, JNK, and PPAR-γ, while also inhibiting Akt and Nrf2 pathways, highlighting its potential as a multifaceted anti-cancer agent. Wogonin [211-213] and Oleuropein [214-216] also demonstrate significant anti-cancer properties through ROS production and anti-proliferative effects, respectively, though the specific role of autophagy in their mechanisms remains to be further elucidated.

Evodiamine [217-219], with its capacity to induce mitochondrial apoptosis and arrest cell-cycle progression via the inactivation of key signaling pathways like PTEN/PI3K/Akt and Wnt/β-catenin, offers another promising avenue for osteosarcoma treatment. Parthenolide’s [220-222] ability to suppress NF-κB activity and induce autophagy through ROS generation makes it a valuable candidate for further research, especially considering its potential to prevent metastasis and enhance radiosensitivity.

Shikonin [223,224] and Berberine [225-227] both exhibit potent anti-cancer effects, with Shikonin inducing apoptosis and necroptosis and Berberine inhibiting cell migration and enhancing the effects of cisplatin. Triptolide’s [228-230] multifunctional approach, affecting apoptosis, autophagy, and angiogenesis through a variety of pathways, underscores the complex interplay between these processes in cancer treatment and the potential of natural products to offer comprehensive therapeutic strategies (Table 4).

Table 4.

Autophagy-related medications for osteosarcoma

Drug Application Status Target Tumor Type Autophagy Activity Mechanism Reference
LY294002 Preclinical PI3K OS Inhibitor Inhibiting PI3K, increasing chemosensitivity in combination with cisplatin [193]
3-MA Preclinical PI3K-III, PI3K-I OS Inhibitor Transient inhibition of PI3K-III and permanent inhibition of PI3K-I, improving OS cell sensitivity to chemotherapy [193,194]
Wortmannin Preclinical PI3KIII, PI3K-I OS Inhibitor Permanently inhibiting PI3KIII and transiently inhibits PI3K-I, also affects mTOR [193]
NSC185058 Preclinical ATG4B OS Inhibitor Suppressing LC3 lipidation, inhibits autophagy in OS cells in vivo, suppresses tumor growth [196]
NSC377071 Preclinical ATG4B OS Inhibitor May suppress ATG4B activity by regulating the mTOR pathway or PI3K pathway down-regulation [196]
Chloroquine (CQ) Preclinical Autophagosomes and lysosomes OS Inhibitor Deacidifies and blocks the fusion of autophagosomes with lysosomes, triggering lysosomal lysis and cell death [195,196]
Hydroxychloroquine (HCQ) Preclinical Autophagosomes and lysosomes OS Inhibitor Similar to CQ, interferes with critical biological processes independent of autophagy [195,196]
Spautin-1 Preclinical VPS34 OS Inhibitor Enhancing degradation of VPS34 complexes, synergistic anti-tumor effects with rapamycin [197]
SAR405 Preclinical VPS34 OS Inhibitor Potently inhibiting VPS34, enhances celecoxib-mediated suppression of cell viability [198]
Verteporfin Preclinical Multiple levels of autophagy OS Inhibitor Disrupting early autophagic processes, induces lysosomal instability, inhibits autophagic flux [199]
Bafilomycin A1 Preclinical Lysosomal acidification OS Inhibitor Inhibiting lysosomal acidification, combined with other inhibitors shows cytotoxic effects in OS cells [200]
NBDHEX Preclinical JNK activation OS Inhibitor Triggering autophagic impairment and cell death by JNK activation [201]
MC3181 Preclinical JNK activation OS Inhibitor Similar to NBDHEX, triggers autophagic impairment and cell death [201]
EGCG Preclinical p38/MAPK, PI3K/AKT OS Inducer Inhibiting DOX-induced pro-survival autophagy, partially inhibits self-renewal capacity of OSCs [202,203]
Rapamycin Preclinical mTORC1 OS Inducer Forms complex with FKBP12, resulting in RAPTOR dissociation and mTORC1 inactivation [204]
Temsirolimus Preclinical mTORC1 OS Inducer Water-soluble analog of rapamycin, increases tumor autophagy and reduces tumor growth [205]
Metformin Preclinical AMPK activation OS Inducer Indirectly activating AMPK, promoting inactivation of mTORC1 Complex [206]
PF-06409577 Preclinical AMPK subunits OS Inducer Binds selectively to AMPK subunits, more potent effects than traditional activators [207]
Oridonin Preclinical p38 MAPK, JNK, PPAR-γ, Akt, Nrf2 pathways OS Inducer Inducing mitochondria-mediated apoptosis, ROS production, inhibits Akt and Nrf2, activates p38 MAPK, JNK, and PPAR-γ [208-210]
Wogonin Preclinical ROS, STAT3, PRX5 OS Inducer Inducing ROS production leading to apoptosis, inhibits STAT3 and PRX5, affects MMP-9 expression in CSCs [211-213]
Oleuropein Preclinical Not specified OS Implied Anti-proliferative properties, role in autophagy induction unclear, used in combination therapy [214-216]
Evodiamine Preclinical PTEN/PI3K/Akt, Wnt/β-catenin OS Not specified Inducing mitochondrial apoptosis, cell-cycle arrest, inactivates PTEN/PI3K/Akt and Wnt/β-catenin pathways [217-219]
Parthenolide Preclinical NF-κB, JNK, ROS OS Inducer Suppressing NF-κB, activates JNK, induces caspase-independent cell death and autophagy via ROS production [220-222]
Shikonin Preclinical ROS, ERK, RIP1, RIP3 OS Not specified Inducing apoptosis and necroptosis, increases ROS and activates ERK, suppresses MMP13, enhances effects of doxorubicin [223,224]
Berberine Preclinical MMP2, p53, MAPK signaling OS Not specified Inhibiting cell proliferation, migration, and colony formation, induces cell-cycle arrest and apoptosis, enhances effects of cisplatin [225-227]
Triptolide Preclinical Fas/FasL, caspases, Wnt/β-catenin, DUSP1 OS Inducer Reducing cell viability, induces apoptosis and autophagy, suppresses angiogenesis, enhances sensitivity to chemotherapy [228-230]

These findings underscore the potential of natural products as valuable candidates for osteosarcoma treatment, either as standalone therapies or in combination with conventional treatments, to overcome resistance and reduce side effects. Further research is needed to fully understand their mechanisms of action, optimize their delivery, and evaluate their efficacy and safety in clinical settings.

Future perspectives and directions in autophagy-related cancer therapies

Given the critical role of autophagy in both promoting and suppressing cancer, particularly in the context of bone metastasis and osteosarcoma, future research should concentrate on several key areas to advance our understanding and therapeutic capabilities.

(1) Precision targeting of autophagy: The dual role of autophagy in cancer highlights the need for precision targeting, where autophagy can be modulated based on the cancer stage, type, and specific patient genetics. Developing biomarkers that can accurately predict the autophagic flux in tumor cells and its impact on patient prognosis is crucial. This approach will enable clinicians to tailor autophagy-modulating therapies to individual patients, maximizing efficacy while minimizing potential adverse effects. (2) Combination therapies: The interplay between autophagy and various signaling pathways, such as mTOR, Beclin-1, and p53, suggests that combination therapies targeting these pathways alongside autophagy could be particularly effective. Future research should explore the synergistic effects of combining autophagy modulators with existing chemotherapeutics, targeted therapies, and immunotherapies. This strategy could potentially overcome drug resistance and improve treatment outcomes in bone metastasis and osteosarcoma patients. (3) Novel autophagy inhibitors and activators: The development of new autophagy-modulating agents is imperative. The current review highlights several preclinical compounds with promising effects on autophagy modulation. Future efforts should focus on optimizing these compounds for better specificity, potency, and safety profiles. Additionally, the discovery of novel autophagy regulators through high-throughput screening and computational modeling could provide new therapeutic candidates. (4) Clinical trials: There is a pressing need to translate preclinical findings into clinical applications. Designing clinical trials that specifically target autophagy in cancer patients, particularly those with bone metastasis and osteosarcoma, will be essential. These trials should aim to assess not only the efficacy and safety of autophagy-modulating therapies but also their impact on quality of life and long-term outcomes. (5) Mechanistic studies: Despite significant advances, the precise mechanisms by which autophagy influences cancer progression and treatment response remain incompletely understood. Future research should aim to elucidate these mechanisms in greater detail, exploring the role of autophagy in cancer stem cells, the tumor microenvironment, and metastatic niches. Understanding these aspects could reveal novel therapeutic targets and strategies to inhibit cancer progression. (6) Interdisciplinary approaches: Combining insights from cell biology, oncology, pharmacology, and computational biology will be crucial in advancing our understanding of autophagy in cancer. Interdisciplinary research efforts can accelerate the identification of biomarkers, the development of novel therapeutics, and the design of effective treatment strategies.

In summary, the complex role of autophagy in cancer, especially in the context of bone metastasis and osteosarcoma, presents both challenges and opportunities for therapeutic intervention. By focusing on precision targeting, combination therapies, novel autophagy modulators, clinical translation, mechanistic studies, and interdisciplinary approaches, future research can pave the way for more effective and personalized cancer treatments.

Conclusion

In conclusion, the dual nature of autophagy in cancer, particularly in bone metastasis and osteosarcoma, highlights its potential as a therapeutic target. Despite its complex role in both suppressing and promoting tumor growth, strategies that modulate autophagy offer promising avenues for enhancing treatment efficacy and overcoming drug resistance. Future research should focus on the selective targeting of autophagy pathways to leverage its beneficial effects against cancer, paving the way for more effective and personalized treatment approaches.

Acknowledgements

This work was supported by The Second Hospital of Lanzhou University for their support in facilitating this research.

Disclosure of conflict of interest

None.

References

  • 1.Su T, Li X, Yang M, Shao Q, Zhao Y, Ma C, Wang P. Autophagy: an intracellular degradation pathway regulating plant survival and stress response. Front Plant Sci. 2020;11:164. doi: 10.3389/fpls.2020.00164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wesselborg S, Stork B. Autophagy signal transduction by ATG proteins: from hierarchies to networks. Cell Mol Life Sci. 2015;72:4721–4757. doi: 10.1007/s00018-015-2034-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, Cañadas I, Sethi G, Tergaonkar V, Wang L, Lin Y, Kang D, Klionsky DJ. Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat. 2025;78:101170. doi: 10.1016/j.drup.2024.101170. [DOI] [PubMed] [Google Scholar]
  • 4.Wang Y, Qin C, Yang G, Zhao B, Wang W. The role of autophagy in pancreatic cancer progression. Biochim Biophys Acta Rev Cancer. 2021;1876:188592. doi: 10.1016/j.bbcan.2021.188592. [DOI] [PubMed] [Google Scholar]
  • 5.Zhang Y, Tang J, Wang C, Zhang Q, Zeng A, Song L. Autophagy-related lncRNAs in tumor progression and drug resistance: a double-edged sword. Genes Dis. 2023;11:367–381. doi: 10.1016/j.gendis.2023.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Coleman RE. Bone cancer in 2011: prevention and treatment of bone metastases. Nat Rev Clin Oncol. 2011;9:76–78. doi: 10.1038/nrclinonc.2011.198. [DOI] [PubMed] [Google Scholar]
  • 7.Ahmadi-Dehlaghi F, Mohammadi P, Valipour E, Pournaghi P, Kiani S, Mansouri K. Autophagy: a challengeable paradox in cancer treatment. Cancer Med. 2023;12:11542–11569. doi: 10.1002/cam4.5577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Takei D, Tagami K. Management of cancer pain due to bone metastasis. J Bone Miner Metab. 2023;41:327–336. doi: 10.1007/s00774-022-01382-y. [DOI] [PubMed] [Google Scholar]
  • 9.Fitch M, Maxwell C, Ryan C, Löthman H, Drudge-Coates L, Costa L. Bone metastases from advanced cancers: clinical implications and treatment options. Clin J Oncol Nurs. 2009;13:701–710. doi: 10.1188/09.CJON.701-710. [DOI] [PubMed] [Google Scholar]
  • 10.Mutlu H, Mutlu S, Bostancıklıoğlu M. Profiling of autophagy-associated microRNAs in the osteosarcoma cell line of U2OS. Anticancer Agents Med Chem. 2021;21:1732–1737. doi: 10.2174/1871520621666201202090128. [DOI] [PubMed] [Google Scholar]
  • 11.Mavrogenis AF, Angelini A, Vottis C, Palmerini E, Rimondi E, Rossi G, Papagelopoulos PJ, Ruggieri P. State-of-the-art approach for bone sarcomas. Eur J Orthop Surg Traumatol. 2015;25:5–15. doi: 10.1007/s00590-014-1468-2. [DOI] [PubMed] [Google Scholar]
  • 12.Chen C, Zhang H, Yu Y, Huang Q, Wang W, Niu J, Lou J, Ren T, Huang Y, Guo W. Chloroquine suppresses proliferation and invasion and induces apoptosis of osteosarcoma cells associated with inhibition of phosphorylation of STAT3. Aging (Albany NY) 2021;13:17901–17913. doi: 10.18632/aging.203196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kiany S, Huang G, Kleinerman ES. Effect of entinostat on NK cell-mediated cytotoxicity against osteosarcoma cells and osteosarcoma lung metastasis. Oncoimmunology. 2017;6:e1333214. doi: 10.1080/2162402X.2017.1333214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Weiss A, Khoury JD, Hoffer FA, Wu J, Billups CA, Heck RK, Quintana J, Poe D, Rao BN, Daw NC. Telangiectatic osteosarcoma: the St. Jude Children’s Research Hospital’s experience. Cancer. 2007;109:1627–1637. doi: 10.1002/cncr.22574. [DOI] [PubMed] [Google Scholar]
  • 15.Klein MJ, Siegal GP. Osteosarcoma: anatomic and histologic variants. Am J Clin Pathol. 2006;125:555–581. doi: 10.1309/UC6K-QHLD-9LV2-KENN. [DOI] [PubMed] [Google Scholar]
  • 16.Gill J, Gorlick R. Advancing therapy for osteosarcoma. Nat Rev Clin Oncol. 2021;18:609–624. doi: 10.1038/s41571-021-00519-8. [DOI] [PubMed] [Google Scholar]
  • 17.Thebault E, Piperno-Neumann S, Tran D, Pacquement H, Marec-Berard P, Lervat C, Castex MP, Cleirec M, Bompas E, Vannier JP, Plantaz D, Saumet L, Verite C, Collard O, Pluchart C, Briandet C, Monard L, Brugieres L, Le Deley MC, Gaspar N. Successive osteosarcoma relapses after the first line O2006/sarcome-09 trial: what can we learn for further phase-II trials? Cancers (Basel) 2021;13:1683. doi: 10.3390/cancers13071683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Smeland S, Bielack SS, Whelan J, Bernstein M, Hogendoorn P, Krailo MD, Gorlick R, Janeway KA, Ingleby FC, Anninga J, Antal I, Arndt C, Brown KLB, Butterfass-Bahloul T, Calaminus G, Capra M, Dhooge C, Eriksson M, Flanagan AM, Friedel G, Gebhardt MC, Gelderblom H, Goldsby R, Grier HE, Grimer R, Hawkins DS, Hecker-Nolting S, Sundby Hall K, Isakoff MS, Jovic G, Kühne T, Kager L, von Kalle T, Kabickova E, Lang S, Lau CC, Leavey PJ, Lessnick SL, Mascarenhas L, Mayer-Steinacker R, Meyers PA, Nagarajan R, Randall RL, Reichardt P, Renard M, Rechnitzer C, Schwartz CL, Strauss S, Teot L, Timmermann B, Sydes MR, Marina N. Survival and prognosis with osteosarcoma: outcomes in more than 2000 patients in the EURAMOS-1 (European and American osteosarcoma study) cohort. Eur J Cancer. 2019;109:36–50. doi: 10.1016/j.ejca.2018.11.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Liu F, Liu D, Yang Y, Zhao S. Effect of autophagy inhibition on chemotherapy-induced apoptosis in A549 lung cancer cells. Oncol Lett. 2013;5:1261–1265. doi: 10.3892/ol.2013.1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sun WL. Ambra1 in autophagy and apoptosis: implications for cell survival and chemotherapy resistance. Oncol Lett. 2016;12:367–374. doi: 10.3892/ol.2016.4644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Azad MB, Chen Y, Gibson SB. Regulation of autophagy by reactive oxygen species (ROS): implications for cancer progression and treatment. Antioxid Redox Signal. 2009;11:777–790. doi: 10.1089/ars.2008.2270. [DOI] [PubMed] [Google Scholar]
  • 22.Werling K. Role of autophagy in the pathogenesis of liver diseases. Orv Hetil. 2011;152:1955–1961. doi: 10.1556/OH.2011.29269. [DOI] [PubMed] [Google Scholar]
  • 23.Gao C, Cao W, Bao L, Zuo W, Xie G, Cai T, Fu W, Zhang J, Wu W, Zhang X, Chen YG. Autophagy negatively regulates Wnt signalling by promoting Dishevelled degradation. Nat Cell Biol. 2010;12:781–790. doi: 10.1038/ncb2082. [DOI] [PubMed] [Google Scholar]
  • 24.Wang L, Klionsky DJ, Shen HM. The emerging mechanisms and functions of microautophagy. Nat Rev Mol Cell Biol. 2023;24:186–203. doi: 10.1038/s41580-022-00529-z. [DOI] [PubMed] [Google Scholar]
  • 25.Yamamoto H, Matsui T. Molecular mechanisms of macroautophagy, microautophagy, and chaperone-mediated autophagy. J Nippon Med Sch. 2024;91:2–9. doi: 10.1272/jnms.JNMS.2024_91-102. [DOI] [PubMed] [Google Scholar]
  • 26.Zhang G, Xiang M, Gu L, Zhou J, Zhang B, Tian W, Deng D. The essential role of TTC28 in maintaining chromosomal stability via HSPA8 chaperone-mediated autophagy. Proc Natl Acad Sci U S A. 2024;121:e2409447121. doi: 10.1073/pnas.2409447121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Sakamoto H, Nakada-Tsukui K, Besteiro S. The autophagy machinery in human-parasitic protists; diverse functions for universally conserved proteins. Cells. 2021;10:1528. doi: 10.3390/cells10051258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Reggiori F, Ungermann C. Autophagosome maturation and fusion. J Mol Biol. 2017;429:486–496. doi: 10.1016/j.jmb.2017.01.002. [DOI] [PubMed] [Google Scholar]
  • 29.Yamamoto H, Fujioka Y, Suzuki SW, Noshiro D, Suzuki H, Kondo-Kakuta C, Kimura Y, Hirano H, Ando T, Noda NN, Ohsumi Y. The intrinsically disordered protein Atg13 mediates supramolecular assembly of autophagy initiation complexes. Dev Cell. 2016;38:86–99. doi: 10.1016/j.devcel.2016.06.015. [DOI] [PubMed] [Google Scholar]
  • 30.Rao Y, Perna MG, Hofmann B, Beier V, Wollert T. The Atg1-kinase complex tethers Atg9-vesicles to initiate autophagy. Nat Commun. 2016;7:10338. doi: 10.1038/ncomms10338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhou X, Zhao X, Zhou W, Qi H, Zhang H, Han TL, Baker P. Impaired placental mitophagy and oxidative stress are associated with dysregulated BNIP3 in preeclampsia. Sci Rep. 2021;11:20469. doi: 10.1038/s41598-021-99837-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Huang Z, Ye B, Dai Z, Wu X, Lu Z, Shan P, Huang W. Curcumin inhibits autophagy and apoptosis in hypoxia/reoxygenation-induced myocytes. Mol Med Rep. 2015;11:4678–4684. doi: 10.3892/mmr.2015.3322. [DOI] [PubMed] [Google Scholar]
  • 33.Phillips AR, Suttangkakul A, Vierstra RD. The ATG12-conjugating enzyme ATG10 Is essential for autophagic vesicle formation in Arabidopsis thaliana. Genetics. 2008;178:1339–1353. doi: 10.1534/genetics.107.086199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Noda NN, Fujioka Y, Ohsumi Y, Inagaki F. Crystallization of the Atg12-Atg5 conjugate bound to Atg16 by the free-interface diffusion method. J Synchrotron Radiat. 2008;15:266–268. doi: 10.1107/S0909049507054799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Shao Y, Gao Z, Feldman T, Jiang X. Stimulation of ATG12-ATG5 conjugation by ribonucleic acid. Autophagy. 2007;3:10–16. doi: 10.4161/auto.3270. [DOI] [PubMed] [Google Scholar]
  • 36.Walczak M, Martens S. Dissecting the role of the Atg12-Atg5-Atg16 complex during autophagosome formation. Autophagy. 2013;9:424–425. doi: 10.4161/auto.22931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ishibashi K, Fujita N, Kanno E, Omori H, Yoshimori T, Itoh T, Fukuda M. Atg16L2, a novel isoform of mammalian Atg16L that is not essential for canonical autophagy despite forming an Atg12-5-16L2 complex. Autophagy. 2011;7:1500–1513. doi: 10.4161/auto.7.12.18025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kaufmann A, Wollert T. Scaffolding the expansion of autophagosomes. Autophagy. 2014;10:1343–1345. doi: 10.4161/auto.28980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Vujić N, Bradić I, Goeritzer M, Kuentzel KB, Rainer S, Kratky D, Radović B. ATG7 is dispensable for LC3-PE conjugation in thioglycolate-elicited mouse peritoneal macrophages. Autophagy. 2021;17:3402–3407. doi: 10.1080/15548627.2021.1874132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wu J, Dang Y, Su W, Liu C, Ma H, Shan Y, Pei Y, Wan B, Guo J, Yu L. Molecular cloning and characterization of rat LC3A and LC3B--two novel markers of autophagosome. Biochem Biophys Res Commun. 2006;339:437–442. doi: 10.1016/j.bbrc.2005.10.211. [DOI] [PubMed] [Google Scholar]
  • 41.Soliman GA. The role of mechanistic target of rapamycin (mTOR) complexes signaling in the immune responses. Nutrients. 2013;5:2231–2257. doi: 10.3390/nu5062231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lamming DW. Inhibition of the mechanistic target of rapamycin (mTOR)-rapamycin and beyond. Cold Spring Harb Perspect Med. 2016;6:a025924. doi: 10.1101/cshperspect.a025924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Parkhitko AA, Favorova OO, Khabibullin DI, Anisimov VN, Henske EP. Kinase mTOR: regulation and role in maintenance of cellular homeostasis, tumor development, and aging. Biochemistry (Mosc) 2014;79:88–101. doi: 10.1134/S0006297914020023. [DOI] [PubMed] [Google Scholar]
  • 44.Mir SA, Dar A, Alshehri SA, Wahab S, Hamid L, Almoyad MAA, Ali T, Bader GN. Exploring the mTOR signalling pathway and its inhibitory scope in cancer. Pharmaceuticals (Basel) 2023;16:1004. doi: 10.3390/ph16071004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nyfeler B, Bergman P, Triantafellow E, Wilson CJ, Zhu Y, Radetich B, Finan PM, Klionsky DJ, Murphy LO. Relieving autophagy and 4EBP1 from rapamycin resistance. Mol Cell Biol. 2011;31:2867–2876. doi: 10.1128/MCB.05430-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fogel AI, Dlouhy BJ, Wang C, Ryu SW, Neutzner A, Hasson SA, Sideris DP, Abeliovich H, Youle RJ. Role of membrane association and Atg14-dependent phosphorylation in beclin-1-mediated autophagy. Mol Cell Biol. 2013;33:3675–3688. doi: 10.1128/MCB.00079-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.He C, Levine B. The Beclin 1 interactome. Curr Opin Cell Biol. 2010;22:140–149. doi: 10.1016/j.ceb.2010.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kaur S, Changotra H. The beclin 1 interactome: modification and roles in the pathology of autophagy-related disorders. Biochimie. 2020;175:34–49. doi: 10.1016/j.biochi.2020.04.025. [DOI] [PubMed] [Google Scholar]
  • 49.Huang T, Wan X, Alvarez AA, James CD, Song X, Yang Y, Sastry N, Nakano I, Sulman EP, Hu B, Cheng SY. MIR93 (microRNA -93) regulates tumorigenicity and therapy response of glioblastoma by targeting autophagy. Autophagy. 2019;15:1100–1111. doi: 10.1080/15548627.2019.1569947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Vicente GP, Della Salda L, Strefezzi RF. Beclin-1 and LC3B expression in canine mast cell tumours: an immuno-ultrastructural and immunohistochemical study of autophagy. Vet Q. 2024;44:1–15. doi: 10.1080/01652176.2024.2419585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cj P, Hv E, Vijayakurup V, R Menon G, Nair S, Gopala S. High LC3/beclin expression correlates with poor survival in glioma: a definitive role for autophagy as evidenced by in vitro autophagic flux. Pathol Oncol Res. 2019;25:137–148. doi: 10.1007/s12253-017-0310-7. [DOI] [PubMed] [Google Scholar]
  • 52.Maiuri MC, Tasdemir E, Criollo A, Morselli E, Vicencio JM, Carnuccio R, Kroemer G. Control of autophagy by oncogenes and tumor suppressor genes. Cell Death Differ. 2009;16:87–93. doi: 10.1038/cdd.2008.131. [DOI] [PubMed] [Google Scholar]
  • 53.Xu-Monette ZY, Medeiros LJ, Li Y, Orlowski RZ, Andreeff M, Bueso-Ramos CE, Greiner TC, McDonnell TJ, Young KH. Dysfunction of the TP53 tumor suppressor gene in lymphoid malignancies. Blood. 2012;119:3668–3683. doi: 10.1182/blood-2011-11-366062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Shi Y, Norberg E, Vakifahmetoglu-Norberg H. Mutant p53 as a regulator and target of autophagy. Front Oncol. 2021;10:607149. doi: 10.3389/fonc.2020.607149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tasdemir E, Chiara Maiuri M, Morselli E, Criollo A, D’Amelio M, Djavaheri-Mergny M, Cecconi F, Tavernarakis N, Kroemer G. A dual role of p53 in the control of autophagy. Autophagy. 2008;4:810–814. doi: 10.4161/auto.6486. [DOI] [PubMed] [Google Scholar]
  • 56.White E. Autophagy and p53. Cold Spring Harb Perspect Med. 2016;6:a026120. doi: 10.1101/cshperspect.a026120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Furuya N, Yu J, Byfield M, Pattingre S, Levine B. The evolutionarily conserved domain of Beclin 1 is required for Vps34 binding, autophagy and tumor suppressor function. Autophagy. 2005;1:46–52. doi: 10.4161/auto.1.1.1542. [DOI] [PubMed] [Google Scholar]
  • 58.Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, Cattoretti G, Levine B. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. 2003;112:1809–1820. doi: 10.1172/JCI20039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.An CH, Kim MS, Yoo NJ, Park SW, Lee SH. Mutational and expressional analyses of ATG5, an autophagy-related gene, in gastrointestinal cancers. Pathol Res Pract. 2011;207:433–437. doi: 10.1016/j.prp.2011.05.002. [DOI] [PubMed] [Google Scholar]
  • 60.Zhang KK, Burns CM, Skinner ME, Lombard DB, Miller RA, Endicott SJ. PTEN is both an activator and a substrate of chaperone-mediated autophagy. J Cell Biol. 2023;222:e202208150. doi: 10.1083/jcb.202208150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Uner AH, Sağlam A, Han U, Hayran M, Sungur A, Ruacan S. PTEN and p27 expression in mature T-cell and NK-cell neoplasms. Leuk Lymphoma. 2005;46:1463–1470. doi: 10.1080/10428190500144813. [DOI] [PubMed] [Google Scholar]
  • 62.Singh SS, Vats S, Chia AY, Tan TZ, Deng S, Ong MS, Arfuso F, Yap CT, Goh BC, Sethi G, Huang RY, Shen HM, Manjithaya R, Kumar AP. Dual role of autophagy in hallmarks of cancer. Oncogene. 2018;37:1142–1158. doi: 10.1038/s41388-017-0046-6. [DOI] [PubMed] [Google Scholar]
  • 63.Yang H, Ni HM, Ding WX. The double-edged sword of MTOR in autophagy deficiency induced-liver injury and tumorigenesis. Autophagy. 2019;15:1671–1673. doi: 10.1080/15548627.2019.1634445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Muthusami S, Ramachandran IK, Babu KN, Krishnamoorthy S, Guruswamy A, Queimado L, Chaudhuri G, Ramachandran I. Role of inflammation in the development of colorectal cancer. Endocr Metab Immune Disord Drug Targets. 2021;21:77–90. doi: 10.2174/1871530320666200909092908. [DOI] [PubMed] [Google Scholar]
  • 65.Liu H, Cao MM, Wang Y, Li LC, Zhu LB, Xie GY, Li YB. Endoplasmic reticulum stress is involved in the connection between inflammation and autophagy in type 2 diabetes. Gen Comp Endocrinol. 2015;210:124–129. doi: 10.1016/j.ygcen.2014.09.006. [DOI] [PubMed] [Google Scholar]
  • 66.Li N, Grivennikov SI, Karin M. The unholy trinity: inflammation, cytokines, and STAT3 shape the cancer microenvironment. Cancer Cell. 2011;19:429–431. doi: 10.1016/j.ccr.2011.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Bu W, Sun X, Xue X, Geng S, Yang T, Zhang J, Li Y, Feng C, Liu Q, Zhang X, Li P, Liu Z, Shi Y, Shao C. Early onset of pathological polyploidization and cellular senescence in hepatocytes lacking RAD51 creates a pro-fibrotic and pro-tumorigenic inflammatory microenvironment. Hepatology. 2025;81:491–508. doi: 10.1097/HEP.0000000000000821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Tang D, Kang R, Zeh HJ, Lotze MT. The multifunctional protein HMGB1: 50 years of discovery. Nat Rev Immunol. 2023;23:824–841. doi: 10.1038/s41577-023-00894-6. [DOI] [PubMed] [Google Scholar]
  • 69.Quach C, Song Y, Guo H, Li S, Maazi H, Fung M, Sands N, O’Connell D, Restrepo-Vassalli S, Chai B, Nemecio D, Punj V, Akbari O, Idos GE, Mumenthaler SM, Wu N, Martin SE, Hagiya A, Hicks J, Cui H, Liang C. A truncating mutation in the autophagy gene UVRAG drives inflammation and tumorigenesis in mice. Nat Commun. 2019;10:5681. doi: 10.1038/s41467-019-13475-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sumpter R Jr, Sirasanagandla S, Fernández ÁF, Wei Y, Dong X, Franco L, Zou Z, Marchal C, Lee MY, Clapp DW, Hanenberg H, Levine B. Fanconi anemia proteins function in mitophagy and immunity. Cell. 2016;165:867–881. doi: 10.1016/j.cell.2016.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Cha-Molstad H, Kwon YT, Kim BY. Amino-terminal arginylation as a degradation signal for selective autophagy. BMB Rep. 2015;48:487–488. doi: 10.5483/BMBRep.2015.48.9.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Cohen-Kaplan V, Ciechanover A, Livneh I. Stress-induced polyubiquitination of proteasomal ubiquitin receptors targets the proteolytic complex for autophagic degradation. Autophagy. 2017;13:759–760. doi: 10.1080/15548627.2016.1278327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Huang X, Yao J, Liu L, Chen J, Mei L, Huangfu J, Luo D, Wang X, Lin C, Chen X, Yang Y, Ouyang S, Wei F, Wang Z, Zhang S, Xiang T, Neculai D, Sun Q, Kong E, Tate EW, Yang A. S-acylation of p62 promotes p62 droplet recruitment into autophagosomes in mammalian autophagy. Mol Cell. 2023;83:3485–3501. e11. doi: 10.1016/j.molcel.2023.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ohnishi Y, Tsuji D, Itoh K. Oxidative stress impairs autophagy via inhibition of lysosomal transport of VAMP8. Biol Pharm Bull. 2022;45:1609–1615. doi: 10.1248/bpb.b22-00131. [DOI] [PubMed] [Google Scholar]
  • 75.Son S, Baek A, Lee JH, Kim DE. Autophagosome-lysosome fusion is facilitated by plectin-stabilized actin and keratin 8 during macroautophagic process. Cell Mol Life Sci. 2022;79:95. doi: 10.1007/s00018-022-04144-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zhang Y, Guo R, Wang SS, Jiang XY, Cui HY, Guo Y, Song XY, Guo QQ, Cao L. Autophagy-related proteins in genome stability: autophagy-dependent and independent actions. Int J Biol Sci. 2022;18:5329–5344. doi: 10.7150/ijbs.76134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cheng SM, Shieh MC, Lin TY, Cheung CHA. The “dark side” of autophagy on the maintenance of genome stability: does it really exist during excessive activation? J Cell Physiol. 2022;237:178–188. doi: 10.1002/jcp.30555. [DOI] [PubMed] [Google Scholar]
  • 78.Burhans WC, Weinberger M. DNA replication stress, genome instability and aging. Nucleic Acids Res. 2007;35:7545–7556. doi: 10.1093/nar/gkm1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kawamura K, Qi F, Kobayashi J. Potential relationship between the biological effects of low-dose irradiation and mitochondrial ROS production. J Radiat Res. 2018;59:ii91–ii97. doi: 10.1093/jrr/rrx091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Leonardi L, Siberil S, Alifano M, Cremer I, Joubert PE. Autophagy-related gene signature highlights metabolic and immunogenic status of malignant cells in non-small cell lung cancer adenocarcinoma. Cancers (Basel) 2022;14:3462. doi: 10.3390/cancers14143462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ferraresi A, Esposito A, Girone C, Vallino L, Salwa A, Ghezzi I, Thongchot S, Vidoni C, Dhanasekaran DN, Isidoro C. Resveratrol contrasts LPA-induced ovarian cancer cell migration and platinum resistance by rescuing hedgehog-mediated autophagy. Cells. 2021;10:3213. doi: 10.3390/cells10113213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wu D, Liang J. Activating transcription factor 4: a regulator of stress response in human cancers. Front Cell Dev Biol. 2024;12:1370012. doi: 10.3389/fcell.2024.1370012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Josset E, Burckel H, Noël G, Bischoff P. The mTOR inhibitor RAD001 potentiates autophagic cell death induced by temozolomide in a glioblastoma cell line. Anticancer Res. 2013;33:1845–1851. [PubMed] [Google Scholar]
  • 84.Lefranc F, Kiss R. Autophagy, the Trojan horse to combat glioblastomas. Neurosurg Focus. 2006;20:E7. doi: 10.3171/foc.2006.20.4.4. [DOI] [PubMed] [Google Scholar]
  • 85.Vera-Ramirez L. Cell-intrinsic survival signals. The role of autophagy in metastatic dissemination and tumor cell dormancy. Semin Cancer Biol. 2020;60:28–40. doi: 10.1016/j.semcancer.2019.07.027. [DOI] [PubMed] [Google Scholar]
  • 86.Finnegan RM, Elshazly AM, Schoenlein PV, Gewirtz DA. Therapeutic potential for targeting autophagy in ER+ breast cancer. Cancers (Basel) 2022;14:4289. doi: 10.3390/cancers14174289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Marsh T, Debnath J. Autophagy suppresses breast cancer metastasis by degrading NBR1. Autophagy. 2020;16:1164–1165. doi: 10.1080/15548627.2020.1753001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Fitzwalter BE, Towers CG, Sullivan KD, Andrysik Z, Hoh M, Ludwig M, O’Prey J, Ryan KM, Espinosa JM, Morgan MJ, Thorburn A. Autophagy inhibition mediates apoptosis sensitization in cancer therapy by relieving FOXO3a turnover. Dev Cell. 2018;44:555–565. e3. doi: 10.1016/j.devcel.2018.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Othman A, Winogradzki M, Lee L, Tandon M, Blank A, Pratap J. Bone metastatic breast cancer: advances in cell signaling and autophagy related mechanisms. Cancers (Basel) 2021;13:4310. doi: 10.3390/cancers13174310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Anlaş AA, Nelson CM. Soft microenvironments induce chemoresistance by increasing autophagy downstream of integrin-linked kinase. Cancer Res. 2020;80:4103–4113. doi: 10.1158/0008-5472.CAN-19-4021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kolb AD, Bussard KM. The bone extracellular matrix as an ideal milieu for cancer cell metastases. Cancers (Basel) 2019;11:1020. doi: 10.3390/cancers11071020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Chen YC, Sosnoski DM, Mastro AM. Breast cancer metastasis to the bone: mechanisms of bone loss. Breast Cancer Res. 2010;12:215. doi: 10.1186/bcr2781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chen X, Wang Z, Duan N, Zhu G, Schwarz EM, Xie C. Osteoblast-osteoclast interactions. Connect Tissue Res. 2018;59:99–107. doi: 10.1080/03008207.2017.1290085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Győri DS, Mócsai A. Osteoclast signal transduction during bone metastasis formation. Front Cell Dev Biol. 2020;8:507. doi: 10.3389/fcell.2020.00507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Liu M, Sun Y, Zhang Q. Emerging role of extracellular vesicles in bone remodeling. J Dent Res. 2018;97:859–868. doi: 10.1177/0022034518764411. [DOI] [PubMed] [Google Scholar]
  • 96.Lyu H, Xiao Y, Guo Q, Huang Y, Luo X. The role of bone-derived exosomes in regulating skeletal metabolism and extraosseous diseases. Front Cell Dev Biol. 2020;8:89. doi: 10.3389/fcell.2020.00089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ji L, Li X, He S, Chen S. Regulation of osteoclast-mediated bone resorption by microRNA. Cell Mol Life Sci. 2022;79:287. doi: 10.1007/s00018-022-04298-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Chen X, Tian P, Chai W, Zhang L, Qin M, Fan M, Liang N, Kim J, Wang Y, Lu WW, Wang D, Cui X, Pan H. A multisynergistic strategy for bone tumor treatment: orchestrating oxidative stress and autophagic flux inhibition by environmental-response nanoparticle. Adv Healthc Mater. 2025;14:e2402872. doi: 10.1002/adhm.202402872. [DOI] [PubMed] [Google Scholar]
  • 99.Verma AK, Bharti PS, Rafat S, Bhatt D, Goyal Y, Pandey KK, Ranjan S, Almatroodi SA, Alsahli MA, Rahmani AH, Almatroudi A, Dev K. Autophagy paradox of cancer: role, regulation, and duality. Oxid Med Cell Longev. 2021;2021:8832541. doi: 10.1155/2021/8832541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ria R, Vacca A. Bone marrow stromal cells-induced drug resistance in multiple myeloma. Int J Mol Sci. 2020;21:613. doi: 10.3390/ijms21020613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zeng Y, Pan Z, Yuan J, Song Y, Feng Z, Chen Z, Ye Z, Li Y, Bao Y, Ran Z, Li X, Ye H, Zhang K, Liu X, He Y. Inhibiting osteolytic breast cancer bone metastasis by bone-targeted nanoagent via remodeling the bone tumor microenvironment combined with NIR-II photothermal therapy. Small. 2023;19:e2301003. doi: 10.1002/smll.202301003. [DOI] [PubMed] [Google Scholar]
  • 102.Tuloup-Minguez V, Hamaï A, Greffard A, Nicolas V, Codogno P, Botti J. Autophagy modulates cell migration and β1 integrin membrane recycling. Cell Cycle. 2013;12:3317–3328. doi: 10.4161/cc.26298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Mohammed WH, Sulaiman GM, Abomughaid MM, Klionsky DJ, Abu-Alghayth MH. The dual role of autophagy in suppressing and promoting hepatocellular carcinoma. Front Cell Dev Biol. 2024;12:1472574. doi: 10.3389/fcell.2024.1472574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Henry DH, Costa L, Goldwasser F, Hirsh V, Hungria V, Prausova J, Scagliotti GV, Sleeboom H, Spencer A, Vadhan-Raj S, von Moos R, Willenbacher W, Woll PJ, Wang J, Jiang Q, Jun S, Dansey R, Yeh H. Randomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma. J. Clin. Oncol. 2011;29:1125–1132. doi: 10.1200/JCO.2010.31.3304. [DOI] [PubMed] [Google Scholar]
  • 105.Henry D, Vadhan-Raj S, Hirsh V, von Moos R, Hungria V, Costa L, Woll PJ, Scagliotti G, Smith G, Feng A, Jun S, Dansey R, Yeh H. Delaying skeletal-related events in a randomized phase 3 study of denosumab versus zoledronic acid in patients with advanced cancer: an analysis of data from patients with solid tumors. Support Care Cancer. 2014;22:679–687. doi: 10.1007/s00520-013-2022-1. [DOI] [PubMed] [Google Scholar]
  • 106.Fizazi K, Carducci M, Smith M, Damião R, Brown J, Karsh L, Milecki P, Shore N, Rader M, Wang H, Jiang Q, Tadros S, Dansey R, Goessl C. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377:813–822. doi: 10.1016/S0140-6736(10)62344-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Scagliotti GV, Hirsh V, Siena S, Henry DH, Woll PJ, Manegold C, Solal-Celigny P, Rodriguez G, Krzakowski M, Mehta ND, Lipton L, García-Sáenz JA, Pereira JR, Prabhash K, Ciuleanu TE, Kanarev V, Wang H, Balakumaran A, Jacobs I. Overall survival improvement in patients with lung cancer and bone metastases treated with denosumab versus zoledronic acid: subgroup analysis from a randomized phase 3 study. J Thorac Oncol. 2012;7:1823–1829. doi: 10.1097/JTO.0b013e31826aec2b. [DOI] [PubMed] [Google Scholar]
  • 108.Obenauf AC, Zou Y, Ji AL, Vanharanta S, Shu W, Shi H, Kong X, Bosenberg MC, Wiesner T, Rosen N, Lo RS, Massagué J. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature. 2015;520:368–372. doi: 10.1038/nature14336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Sun JM, Ahn JS, Lee S, Kim JA, Lee J, Park YH, Park HC, Ahn MJ, Ahn YC, Park K. Predictors of skeletal-related events in non-small cell lung cancer patients with bone metastases. Lung Cancer. 2011;71:89–93. doi: 10.1016/j.lungcan.2010.04.003. [DOI] [PubMed] [Google Scholar]
  • 110.Smith DC, Smith MR, Sweeney C, Elfiky AA, Logothetis C, Corn PG, Vogelzang NJ, Small EJ, Harzstark AL, Gordon MS, Vaishampayan UN, Haas NB, Spira AI, Lara PN Jr, Lin CC, Srinivas S, Sella A, Schöffski P, Scheffold C, Weitzman AL, Hussain M. Cabozantinib in patients with advanced prostate cancer: results of a phase ii randomized discontinuation trial. J. Clin. Oncol. 2013;31:412–419. doi: 10.1200/JCO.2012.45.0494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Smith M, De Bono J, Sternberg C, Le Moulec S, Oudard S, De Giorgi U, Krainer M, Bergman A, Hoelzer W, De Wit R, Bögemann M, Saad F, Cruciani G, Thiery-Vuillemin A, Feyerabend S, Miller K, Houédé N, Hussain S, Lam E, Polikoff J, Stenzl A, Mainwaring P, Ramies D, Hessel C, Weitzman A, Fizazi K. Phase III study of cabozantinib in previously treated metastatic castration-resistant prostate cancer: COMET-1. J. Clin. Oncol. 2016;34:3005–3013. doi: 10.1200/JCO.2015.65.5597. [DOI] [PubMed] [Google Scholar]
  • 112.Varkaris A, Corn PG, Parikh NU, Efstathiou E, Song JH, Lee YC, Aparicio A, Hoang AG, Gaur S, Thorpe L, Maity SN, Bar Eli M, Czerniak BA, Shao Y, Alauddin M, Lin SH, Logothetis CJ, Gallick GE. Integrating murine and clinical trials with cabozantinib to understand roles of MET and VEGFR2 as targets for growth inhibition of prostate cancer. Clin Cancer Res. 2016;22:107–121. doi: 10.1158/1078-0432.CCR-15-0235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Sousa S, Clézardin P. Bone-targeted therapies in cancer-induced bone disease. Calcif Tissue Int. 2018;102:227–250. doi: 10.1007/s00223-017-0353-5. [DOI] [PubMed] [Google Scholar]
  • 114.Browne AJ, Kubasch ML, Göbel A, Hadji P, Chen D, Rauner M, Stölzel F, Hofbauer LC, Rachner TD. Concurrent antitumor and bone-protective effects of everolimus in osteotropic breast cancer. Breast Cancer Res. 2017;19:92. doi: 10.1186/s13058-017-0885-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Vandyke K, Fitter S, Dewar AL, Hughes TP, Zannettino AC. Dysregulation of bone remodeling by imatinib mesylate. Blood. 2010;115:766–774. doi: 10.1182/blood-2009-08-237404. [DOI] [PubMed] [Google Scholar]
  • 116.Gobin B, Moriceau G, Ory B, Charrier C, Brion R, Blanchard F, Redini F, Heymann D. Imatinib mesylate exerts anti-proliferative effects on osteosarcoma cells and inhibits the tumour growth in immunocompetent murine models. PLoS One. 2014;9:e90795. doi: 10.1371/journal.pone.0090795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Coleman R, Hadji P, Body JJ, Santini D, Chow E, Terpos E, Oudard S, Bruland Ø, Flamen P, Kurth A, Van Poznak C, Aapro M, Jordan K ESMO Guidelines Committee. Bone health in cancer: ESMO clinical practice guidelines. Ann Oncol. 2020;31:1650–1663. doi: 10.1016/j.annonc.2020.07.019. [DOI] [PubMed] [Google Scholar]
  • 118.Coleman RE, Croucher PI, Padhani AR, Clézardin P, Chow E, Fallon M, Guise T, Colangeli S, Capanna R, Costa L. Bone metastases. Nat Rev Dis Primers. 2020;6:83. doi: 10.1038/s41572-020-00216-3. [DOI] [PubMed] [Google Scholar]
  • 119.Han MW, Lee JC, Choi JY, Kim GC, Chang HW, Nam HY, Kim SW, Kim SY. Autophagy inhibition can overcome radioresistance in breast cancer cells through suppression of TAK1 activation. Anticancer Res. 2014;34:1449–1455. [PubMed] [Google Scholar]
  • 120.Gonçalves RM, Agnes JP, Delgobo M, de Souza PO, Thomé MP, Heimfarth L, Lenz G, Moreira JCF, Zanotto-Filho A. Late autophagy inhibitor chloroquine improves efficacy of the histone deacetylase inhibitor SAHA and temozolomide in gliomas. Biochem Pharmacol. 2019;163:440–450. doi: 10.1016/j.bcp.2019.03.015. [DOI] [PubMed] [Google Scholar]
  • 121.Cocco S, Leone A, Roca MS, Lombardi R, Piezzo M, Caputo R, Ciardiello C, Costantini S, Bruzzese F, Sisalli MJ, Budillon A, De Laurentiis M. Inhibition of autophagy by chloroquine prevents resistance to PI3K/AKT inhibitors and potentiates their antitumor effect in combination with paclitaxel in triple negative breast cancer models. J Transl Med. 2022;20:290. doi: 10.1186/s12967-022-03462-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nagelkerke A, Sieuwerts AM, Bussink J, Sweep FC, Look MP, Foekens JA, Martens JW, Span PN. LAMP3 is involved in tamoxifen resistance in breast cancer cells through the modulation of autophagy. Endocr Relat Cancer. 2014;21:101–112. doi: 10.1530/ERC-13-0183. [DOI] [PubMed] [Google Scholar]
  • 123.Chen S, Zhu X, Qiao H, Ye M, Lai X, Yu S, Ding L, Wen A, Zhang J. Protective autophagy promotes the resistance of HER2-positive breast cancer cells to lapatinib. Tumour Biol. 2016;37:2321–2331. doi: 10.1007/s13277-015-3800-9. [DOI] [PubMed] [Google Scholar]
  • 124.Liu P, Fan J, Wang Z, Zai W, Song P, Li Y, Ju D. The role of autophagy in the cytotoxicity induced by trastuzumab emtansine (T-DM1) in HER2-positive breast cancer cells. AMB Express. 2020;10:107. doi: 10.1186/s13568-020-01044-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Wang J, Xu B. Targeted therapeutic options and future perspectives for HER2-positive breast cancer. Signal Transduct Target Ther. 2019;4:34. doi: 10.1038/s41392-019-0069-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Yang X, Zheng Y, Liu L, Huang J, Wang F, Zhang J. Progress on the study of the anticancer effects of artesunate. Oncol Lett. 2021;22:750. doi: 10.3892/ol.2021.13011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Guan X, Guan Y. Artemisinin induces selective and potent anticancer effects in drug resistant breast cancer cells by inducing cellular apoptosis and autophagy and G2/M cell cycle arrest. J BUON. 2020;25:1330–1336. [PubMed] [Google Scholar]
  • 128.Hamacher-Brady A, Stein HA, Turschner S, Toegel I, Mora R, Jennewein N, Efferth T, Eils R, Brady NR. Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production. J Biol Chem. 2011;286:6587–6601. doi: 10.1074/jbc.M110.210047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Gu L, Zhang J, Liu D, Chen J, Liu S, Peng Q, Tian Y, Du M, Zhang J, Xiao W, Shen S, Wang J. Development of artesunate intelligent prodrug liposomes based on mitochondrial targeting strategy. J Nanobiotechnology. 2022;20:376. doi: 10.1186/s12951-022-01569-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Wang M, Qiu S, Qin J. Baicalein induced apoptosis and autophagy of undifferentiated thyroid cancer cells by the ERK/PI3K/Akt pathway. Am J Transl Res. 2019;11:3341–3352. [PMC free article] [PubMed] [Google Scholar]
  • 131.Hua F, Xiao YY, Qu XH, Li SS, Zhang K, Zhou C, He JL, Zhu Y, Wan YY, Jiang LP, Tou FF, Han XJ. Baicalein sensitizes triple negative breast cancer MDA-MB-231 cells to doxorubicin via autophagy-mediated down-regulation of CDK1. Mol Cell Biochem. 2023;478:1519–1531. doi: 10.1007/s11010-022-04597-9. [DOI] [PubMed] [Google Scholar]
  • 132.Liu B, Ding L, Zhang L, Wang S, Wang Y, Wang B, Li L. Baicalein induces autophagy and apoptosis through AMPK pathway in human glioma cells. Am J Chin Med. 2019;47:1405–1418. doi: 10.1142/S0192415X19500721. [DOI] [PubMed] [Google Scholar]
  • 133.Yan W, Ma X, Zhao X, Zhang S. Baicalein induces apoptosis and autophagy of breast cancer cells via inhibiting PI3K/AKT pathway in vivo and vitro. Drug Des Devel Ther. 2018;12:3961–3972. doi: 10.2147/DDDT.S181939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Rajabi S, Irani M, Moeinifard M, Hamzeloo-Moghadam M. Britannin suppresses MCF-7 breast cancer cell growth by inducing apoptosis and inhibiting autophagy. Avicenna J Phytomed. 2024;14:90–99. doi: 10.22038/AJP.2023.22995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Hsu CY, Rajabi S, Hamzeloo-Moghadam M, Kumar A, Maresca M, Ghildiyal P. Sesquiterpene lactones as emerging biomolecules to cease cancer by targeting apoptosis. Front Pharmacol. 2024;15:1371002. doi: 10.3389/fphar.2024.1371002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Rajabi S, Tahmasvand Z, Maresca M, Hamzeloo-Moghadam M. Gaillardin inhibits autophagy and induces apoptosis in MCF-7 breast cancer cells by regulating JAK/STAT pathway. Mol Biol Rep. 2024;51:158. doi: 10.1007/s11033-023-09131-8. [DOI] [PubMed] [Google Scholar]
  • 137.Shi J, Li J, Xu Z, Chen L, Luo R, Zhang C, Gao F, Zhang J, Fu C. Celastrol: a review of useful strategies overcoming its limitation in anticancer application. Front Pharmacol. 2020;11:558741. doi: 10.3389/fphar.2020.558741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Feng Y, Zhang B, Lv J, Zhang P, Mao Q, Lin F, Zhao J, Fu X, Yang Y, Li Z, Zhang L, Mou Y, Wang S. Scaffold hopping of celastrol provides derivatives containing pepper ring, pyrazine and oxazole substructures as potent autophagy inducers against breast cancer cell line MCF-7. Eur J Med Chem. 2022;234:114254. doi: 10.1016/j.ejmech.2022.114254. [DOI] [PubMed] [Google Scholar]
  • 139.Wang L, Tang L, Yao C, Liu C, Shu Y. The synergistic effects of celastrol in combination with tamoxifen on apoptosis and autophagy in MCF-7 cells. J Immunol Res. 2021;2021:5532269. doi: 10.1155/2021/5532269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Duangmano S, Dakeng S, Jiratchariyakul W, Suksamrarn A, Smith DR, Patmasiriwat P. Antiproliferative effects of cucurbitacin B in breast cancer cells: down-regulation of the c-Myc/hTERT/telomerase pathway and obstruction of the cell cycle. Int J Mol Sci. 2010;11:5323–5338. doi: 10.3390/ijms11125323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ren G, Sha T, Guo J, Li W, Lu J, Chen X. Cucurbitacin B induces DNA damage and autophagy mediated by reactive oxygen species (ROS) in MCF-7 breast cancer cells. J Nat Med. 2015;69:522–530. doi: 10.1007/s11418-015-0918-4. [DOI] [PubMed] [Google Scholar]
  • 142.Chan KT, Meng FY, Li Q, Ho CY, Lam TS, To Y, Lee WH, Li M, Chu KH, Toh M. Cucurbitacin B induces apoptosis and S phase cell cycle arrest in BEL-7402 human hepatocellular carcinoma cells and is effective via oral administration. Cancer Lett. 2010;294:118–124. doi: 10.1016/j.canlet.2010.01.029. [DOI] [PubMed] [Google Scholar]
  • 143.Hewlings SJ, Kalman DS. Curcumin: a review of its effects on human health. Foods. 2017;6:92. doi: 10.3390/foods6100092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Khorsandi K, Hosseinzadeh R, Shahidi FK. Photodynamic treatment with anionic nanoclays containing curcumin on human triple-negative breast cancer cells: cellular and biochemical studies. J Cell Biochem. 2019;120:4998–5009. doi: 10.1002/jcb.27775. [DOI] [PubMed] [Google Scholar]
  • 145.Wang K, Zhang C, Bao J, Jia X, Liang Y, Wang X, Chen M, Su H, Li P, Wan JB, He C. Synergistic chemopreventive effects of curcumin and berberine on human breast cancer cells through induction of apoptosis and autophagic cell death. Sci Rep. 2016;6:26064. doi: 10.1038/srep26064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Jiang S, Chang H, Deng S, Fan D. Icariin enhances the chemosensitivity of cisplatinresistant ovarian cancer cells by suppressing autophagy via activation of the AKT/mTOR/ATG5 pathway. Int J Oncol. 2019;54:1933–1942. doi: 10.3892/ijo.2019.4785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wang YX, Jin YY, Wang J, Zhao ZC, Xue KW, Xiong H, Che HL, Ge YJ, Wu GS. Icaritin derivative ic2 induces cytoprotective autophagy of breast cancer cells via SCD1 inhibition. Molecules. 2023;28:1109. doi: 10.3390/molecules28031109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Cheng X, Tan S, Duan F, Yuan Q, Li Q, Deng G. Icariin induces apoptosis by suppressing autophagy in tamoxifen-resistant breast cancer cell line MCF-7/TAM. Breast Cancer. 2019;26:766–775. doi: 10.1007/s12282-019-00980-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Wu CL, Liu JF, Liu Y, Wang YX, Fu KF, Yu XJ, Pu Q, Chen XX, Zhou LJ. Beclin1 inhibition enhances paclitaxelmediated cytotoxicity in breast cancer in vitro and in vivo. Int J Mol Med. 2019;43:1866–1878. doi: 10.3892/ijmm.2019.4089. [DOI] [PubMed] [Google Scholar]
  • 150.Škubník J, Svobodová Pavlíčková V, Ruml T, Rimpelová S. Autophagy in cancer resistance to paclitaxel: development of combination strategies. Biomed Pharmacother. 2023;161:114458. doi: 10.1016/j.biopha.2023.114458. [DOI] [PubMed] [Google Scholar]
  • 151.Hu W, Zheng W, Du J, Tian Z, Zhao Y, Zhao P, Li J. TIPE2 sensitizes breast cancer cells to paclitaxel by suppressing drug-induced autophagy and cancer stem cell properties. Hum Cell. 2023;36:1485–1500. doi: 10.1007/s13577-023-00900-y. [DOI] [PubMed] [Google Scholar]
  • 152.Pai Bellare G, Sankar Patro B. Resveratrol sensitizes breast cancer to PARP inhibitor, talazoparib through dual inhibition of AKT and autophagy flux. Biochem Pharmacol. 2022;199:115024. doi: 10.1016/j.bcp.2022.115024. [DOI] [PubMed] [Google Scholar]
  • 153.Wang J, Huang P, Pan X, Xia C, Zhang H, Zhao H, Yuan Z, Liu J, Meng C, Liu F. Resveratrol reverses TGF-β1-mediated invasion and metastasis of breast cancer cells via the SIRT3/AMPK/autophagy signal axis. Phytother Res. 2023;37:211–230. doi: 10.1002/ptr.7608. [DOI] [PubMed] [Google Scholar]
  • 154.Alayev A, Berger SM, Kramer MY, Schwartz NS, Holz MK. The combination of rapamycin and resveratrol blocks autophagy and induces apoptosis in breast cancer cells. J Cell Biochem. 2015;116:450–457. doi: 10.1002/jcb.24997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Luan F, He X, Zeng N. Tetrandrine: a review of its anticancer potentials, clinical settings, pharmacokinetics and drug delivery systems. J Pharm Pharmacol. 2020;72:1491–1512. doi: 10.1111/jphp.13339. [DOI] [PubMed] [Google Scholar]
  • 156.Wang Y, Yue W, Lang H, Ding X, Chen X, Chen H. Resuming sensitivity of tamoxifen-resistant breast cancer cells to tamoxifen by tetrandrine. Integr Cancer Ther. 2021;20:1534735421996822. doi: 10.1177/1534735421996822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Wong VKW, Zeng W, Chen J, Yao XJ, Leung ELH, Wang QQ, Chiu P, Ko BCB, Law BYK. Tetrandrine, an activator of autophagy, induces autophagic cell death via PKC-α inhibition and mTOR-dependent mechanisms. Front Pharmacol. 2017;8:351. doi: 10.3389/fphar.2017.00351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Xu H, Liu B, Xiao Z, Zhou M, Ge L, Jia F, Liu Y, Jin H, Zhu X, Gao J, Akhtar J, Xiang B, Tan K, Wang G. Computational and experimental studies reveal that thymoquinone blocks the entry of coronaviruses into in vitro cells. Infect Dis Ther. 2021;10:483–494. doi: 10.1007/s40121-021-00400-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Ünal TD, Hamurcu Z, Delibaşı N, Çınar V, Güler A, Gökçe S, Nurdinov N, Ozpolat B. Thymoquinone inhibits proliferation and migration of MDA-MB-231 triple negative breast cancer cells by suppressing autophagy, Beclin-1 and LC3. Anticancer Agents Med Chem. 2021;21:355–364. doi: 10.2174/1871520620666200807221047. [DOI] [PubMed] [Google Scholar]
  • 160.Alkhatib MH, Bawadud RS, Gashlan HM. Incorporation of docetaxel and thymoquinone in borage nanoemulsion potentiates their antineoplastic activity in breast cancer cells. Sci Rep. 2020;10:18124. doi: 10.1038/s41598-020-75017-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Ahsan H, Ahad A, Iqbal J, Siddiqui WA. Pharmacological potential of tocotrienols: a review. Nutr Metab (Lond) 2014;11:52. doi: 10.1186/1743-7075-11-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Tiwari RV, Parajuli P, Sylvester PW. Synergistic anticancer effects of combined γ-tocotrienol and oridonin treatment is associated with the induction of autophagy. Mol Cell Biochem. 2015;408:123–137. doi: 10.1007/s11010-015-2488-x. [DOI] [PubMed] [Google Scholar]
  • 163.Tiwari RV, Parajuli P, Sylvester PW. γ-Tocotrienol-induced endoplasmic reticulum stress and autophagy act concurrently to promote breast cancer cell death. Biochem Cell Biol. 2015;93:306–320. doi: 10.1139/bcb-2014-0123. [DOI] [PubMed] [Google Scholar]
  • 164.Wang Z, Zhang P, Jiang H, Sun B, Luo H, Jia A. Ursolic acid enhances the sensitivity of MCF-7 and MDA-MB-231 cells to epirubicin by modulating the autophagy pathway. Molecules. 2022;27:3399. doi: 10.3390/molecules27113399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Luo J, Hu YL, Wang H. Ursolic acid inhibits breast cancer growth by inhibiting proliferation, inducing autophagy and apoptosis, and suppressing inflammatory responses via the PI3K/AKT and NF-κB signaling pathways in vitro. Exp Ther Med. 2017;14:3623–3631. doi: 10.3892/etm.2017.4965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Lewinska A, Adamczyk-Grochala J, Kwasniewicz E, Deregowska A, Wnuk M. Ursolic acid-mediated changes in glycolytic pathway promote cytotoxic autophagy and apoptosis in phenotypically different breast cancer cells. Apoptosis. 2017;22:800–815. doi: 10.1007/s10495-017-1353-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Ojha R, Singh SK, Bhattacharyya S, Dhanda RS, Rakha A, Mandal AK, Jha V. Inhibition of grade dependent autophagy in urothelial carcinoma increases cell death under nutritional limiting condition and potentiates the cytotoxicity of chemotherapeutic agent. J Urol. 2014;191:1889–1898. doi: 10.1016/j.juro.2014.01.006. [DOI] [PubMed] [Google Scholar]
  • 168.Li W, Li D, Ma Q, Chen Y, Hu Z, Bai Y, Xie L. Targeted inhibition of mTOR by BML-275 induces mitochondrial-mediated apoptosis and autophagy in prostate cancer. Eur J Pharmacol. 2023;957:176035. doi: 10.1016/j.ejphar.2023.176035. [DOI] [PubMed] [Google Scholar]
  • 169.Chen C, Wang H, Geng X, Zhang D, Zhu Z, Zhang G, Hou J. Metformin exerts anti-AR-negative prostate cancer activity via AMPK/autophagy signaling pathway. Cancer Cell Int. 2021;21:404. doi: 10.1186/s12935-021-02043-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Ariotti N, Wu Y, Okano S, Gambin Y, Follett J, Rae J, Ferguson C, Teasdale RD, Alexandrov K, Meunier FA, Hill MM, Parton RG. An inverted CAV1 (caveolin 1) topology defines novel autophagy-dependent exosome secretion from prostate cancer cells. Autophagy. 2021;17:2200–2216. doi: 10.1080/15548627.2020.1820787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Chai D, Shi SY, Sobhani N, Ding J, Zhang Z, Jiang N, Wang G, Li M, Li H, Zheng J, Bai J. IFI35 promotes renal cancer progression by inhibiting pSTAT1/pSTAT6-dependent autophagy. Cancers (Basel) 2022;14:2861. doi: 10.3390/cancers14122861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Lu J, Zhu L, Zheng LP, Cui Q, Zhu HH, Zhao H, Shen ZJ, Dong HY, Chen SS, Wu WZ, Tan JM. Overexpression of ulk1 represents a potential diagnostic marker for clear cell renal carcinoma and the antitumor effects of SBI-0206965. EBioMedicine. 2018;34:85–93. doi: 10.1016/j.ebiom.2018.07.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Kudo Y, Endo S, Fujita M, Ota A, Kamatari YO, Tanaka Y, Ishikawa T, Ikeda H, Okada T, Toyooka N, Fujimoto N, Matsunaga T, Ikari A. Discovery and structure-based optimization of novel Atg4B inhibitors for the treatment of castration-resistant prostate cancer. J Med Chem. 2022;65:4878–4892. doi: 10.1021/acs.jmedchem.1c02113. [DOI] [PubMed] [Google Scholar]
  • 174.Kudo Y, Nakamura K, Tsuzuki H, Hirota K, Kawai M, Takaya D, Fukuzawa K, Honma T, Yoshino Y, Nakamura M, Shiota M, Fujimoto N, Ikari A, Endo S. Docosahexaenoic acid enhances the treatment efficacy for castration-resistant prostate cancer by inhibiting autophagy through Atg4B inhibition. Arch Biochem Biophys. 2024;760:110135. doi: 10.1016/j.abb.2024.110135. [DOI] [PubMed] [Google Scholar]
  • 175.Jiang H, Sun J, Xu Q, Liu Y, Wei J, Young CY, Yuan H, Lou H. Marchantin M: a novel inhibitor of proteasome induces autophagic cell death in prostate cancer cells. Cell Death Dis. 2013;4:e761. doi: 10.1038/cddis.2013.285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Etani T, Naiki T, Naiki-Ito A, Suzuki T, Iida K, Nozaki S, Kato H, Nagayasu Y, Suzuki S, Kawai N, Yasui T, Takahashi S. NCL1, a highly selective lysine-specific demethylase 1 inhibitor, suppresses castration-resistant prostate cancer growth via regulation of apoptosis and autophagy. J Clin Med. 2019;8:442. doi: 10.3390/jcm8040442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Liu X, Wu J, Fan M, Shen C, Dai W, Bao Y, Liu JH, Yu BY. Novel dihydroartemisinin derivative DHA-37 induces autophagic cell death through upregulation of HMGB1 in A549 cells. Cell Death Dis. 2018;9:1048. doi: 10.1038/s41419-018-1006-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Lai XY, Shi YM, Zhou MM. Dihydroartemisinin enhances gefitinib cytotoxicity against lung adenocarcinoma cells by inducing ROS-dependent apoptosis and ferroptosis. Kaohsiung J Med Sci. 2023;39:699–709. doi: 10.1002/kjm2.12684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Tang X, Ding H, Liang M, Chen X, Yan Y, Wan N, Chen Q, Zhang J, Cao J. Curcumin induces ferroptosis in non-small-cell lung cancer via activating autophagy. Thorac Cancer. 2021;12:1219–1230. doi: 10.1111/1759-7714.13904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Liu F, Gao S, Yang Y, Zhao X, Fan Y, Ma W, Yang D, Yang A, Yu Y. Antitumor activity of curcumin by modulation of apoptosis and autophagy in human lung cancer A549 cells through inhibiting PI3K/Akt/mTOR pathway. Oncol Rep. 2018;39:1523–1531. doi: 10.3892/or.2018.6188. [DOI] [PubMed] [Google Scholar]
  • 181.Wang A, Wang J, Zhang S, Zhang H, Xu Z, Li X. Curcumin inhibits the development of non-small cell lung cancer by inhibiting autophagy and apoptosis. Exp Ther Med. 2017;14:5075–5080. doi: 10.3892/etm.2017.5172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Kim HJ, Park C, Han MH, Hong SH, Kim GY, Hong SH, Kim ND, Choi YH. Baicalein induces caspase-dependent apoptosis associated with the generation of ROS and the activation of AMPK in human lung carcinoma A549 cells. Drug Dev Res. 2016;77:73–86. doi: 10.1002/ddr.21298. [DOI] [PubMed] [Google Scholar]
  • 183.Deng X, Liu J, Liu L, Sun X, Huang J, Dong J. Drp1-mediated mitochondrial fission contributes to baicalein-induced apoptosis and autophagy in lung cancer via activation of AMPK signaling pathway. Int J Biol Sci. 2020;16:1403–1416. doi: 10.7150/ijbs.41768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Li J, Yan L, Luo J, Tong L, Gao Y, Feng W, Wang F, Cui W, Li S, Sun Z. Baicalein suppresses growth of non-small cell lung carcinoma by targeting MAP4K3. Biomed Pharmacother. 2021;133:110965. doi: 10.1016/j.biopha.2020.110965. [DOI] [PubMed] [Google Scholar]
  • 185.Hu M, Yang J, Qu L, Deng X, Duan Z, Fu R, Liang L, Fan D. Ginsenoside Rk1 induces apoptosis and downregulates the expression of PD-L1 by targeting the NF-κB pathway in lung adenocarcinoma. Food Funct. 2020;11:456–471. doi: 10.1039/c9fo02166c. [DOI] [PubMed] [Google Scholar]
  • 186.Chen Y, Zhang Y, Song W, Zhang Y, Dong X, Tan M. Ginsenoside Rh2 improves the cisplatin anti-tumor effect in lung adenocarcinoma A549 cells via superoxide and PD-L1. Anticancer Agents Med Chem. 2020;20:495–503. doi: 10.2174/1871520619666191209091230. [DOI] [PubMed] [Google Scholar]
  • 187.Li C, Dong Y, Wang L, Xu G, Yang Q, Tang X, Qiao Y, Cong Z. Ginsenoside metabolite compound K induces apoptosis and autophagy in non-small cell lung cancer cells via AMPK-mTOR and JNK pathways. Biochem Cell Biol. 2019;97:406–414. doi: 10.1139/bcb-2018-0226. [DOI] [PubMed] [Google Scholar]
  • 188.Xie Q, Wen H, Zhang Q, Zhou W, Lin X, Xie D, Liu Y. Inhibiting PI3K-AKt signaling pathway is involved in antitumor effects of ginsenoside Rg3 in lung cancer cell. Biomed Pharmacother. 2017;85:16–21. doi: 10.1016/j.biopha.2016.11.096. [DOI] [PubMed] [Google Scholar]
  • 189.Warburton D, Bellusci S. Normal lung development needs self-eating. J Clin Invest. 2019;129:2658–2659. doi: 10.1172/JCI129442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Ying H, Ying B, Zhang J, Kong D. Sirt1 modulates H3 phosphorylation and facilitates osteosarcoma cell autophagy. Artif Cells Nanomed Biotechnol. 2019;47:3374–3381. doi: 10.1080/21691401.2019.1648280. [DOI] [PubMed] [Google Scholar]
  • 191.Zhang J, Rao D, Ma H, Kong D, Xu X, Lu H. LncRNA SNHG15 contributes to doxorubicin resistance of osteosarcoma cells through targeting the miR-381-3p/GFRA1 axis. Open Life Sci. 2020;15:871–883. doi: 10.1515/biol-2020-0086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Meng C, Yang Y, Feng W, Ma P, Bai R. Exosomal miR-331-3p derived from chemoresistant osteosarcoma cells induces chemoresistance through autophagy. J Orthop Surg Res. 2023;18:892. doi: 10.1186/s13018-023-04338-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Pasquier B. Autophagy inhibitors. Cell Mol Life Sci. 2016;73:985–1001. doi: 10.1007/s00018-015-2104-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Miao XD, Cao L, Zhang Q, Hu XY, Zhang Y. Effect of PI3K-mediated autophagy in human osteosarcoma MG63 cells on sensitivity to chemotherapy with cisplatin. Asian Pac J Trop Med. 2015;8:731–738. doi: 10.1016/j.apjtm.2015.07.024. [DOI] [PubMed] [Google Scholar]
  • 195.Wen X, Klionsky DJ. At a glance: a history of autophagy and cancer. Semin Cancer Biol. 2020;66:3–11. doi: 10.1016/j.semcancer.2019.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Akin D, Wang SK, Habibzadegah-Tari P, Law B, Ostrov D, Li M, Yin XM, Kim JS, Horenstein N, Dunn WA Jr. A novel ATG4B antagonist inhibits autophagy and has a negative impact on osteosarcoma tumors. Autophagy. 2014;10:2021–2035. doi: 10.4161/auto.32229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Horie R, Nakamura O, Yamagami Y, Mori M, Nishimura H, Fukuoka N, Yamamoto T. Apoptosis and antitumor effects induced by the combination of an mTOR inhibitor and an autophagy inhibitor in human osteosarcoma MG63 cells. Int J Oncol. 2016;48:37–44. doi: 10.3892/ijo.2015.3227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Zhou P, Li Y, Li B, Zhang M, Xu C, Liu F, Bian L, Liu Y, Yao Y, Li D. Autophagy inhibition enhances celecoxib-induced apoptosis in osteosarcoma. Cell Cycle. 2018;17:997–1006. doi: 10.1080/15384101.2018.1467677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Saini H, Sharma H, Mukherjee S, Chowdhury S, Chowdhury R. Verteporfin disrupts multiple steps of autophagy and regulates p53 to sensitize osteosarcoma cells. Cancer Cell Int. 2021;21:52. doi: 10.1186/s12935-020-01720-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Liao YX, Yu HY, Lv JY, Cai YR, Liu F, He ZM, He SS. Targeting autophagy is a promising therapeutic strategy to overcome chemoresistance and reduce metastasis in osteosarcoma. Int J Oncol. 2019;55:1213–1222. doi: 10.3892/ijo.2019.4902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Palumbo C, De Luca A, Rosato N, Forgione M, Rotili D, Caccuri AM. c-Jun N-terminal kinase activation by nitrobenzoxadiazoles leads to late-stage autophagy inhibition. J Transl Med. 2016;14:37. doi: 10.1186/s12967-016-0796-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Wang W, Chen D, Zhu K. SOX2OT variant 7 contributes to the synergistic interaction between EGCG and Doxorubicin to kill osteosarcoma via autophagy and stemness inhibition. J Exp Clin Cancer Res. 2018;37:37. doi: 10.1186/s13046-018-0689-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Dong C, Wang Z, Shen P, Chen Y, Wang J, Wang H. Epigallocatechin-3-gallate suppresses the growth of human osteosarcoma by inhibiting the Wnt/β-catenin signalling pathway. Bioengineered. 2022;13:8490–8502. doi: 10.1080/21655979.2022.2051805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Chiarini F, Evangelisti C, McCubrey JA, Martelli AM. Current treatment strategies for inhibiting mTOR in cancer. Trends Pharmacol Sci. 2015;36:124–135. doi: 10.1016/j.tips.2014.11.004. [DOI] [PubMed] [Google Scholar]
  • 205.Buzun K, Gornowicz A, Lesyk R, Bielawski K, Bielawska A. Autophagy modulators in cancer therapy. Int J Mol Sci. 2021;22:5804. doi: 10.3390/ijms22115804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Lin CJ, Tsao YN, Shu CW. Autophagy modulation as a potential targeted cancer therapy: from drug repurposing to new drug development. Kaohsiung J Med Sci. 2021;37:166–171. doi: 10.1002/kjm2.12361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Zhu YR, Zhang XY, Wu QP, Yu CJ, Liu YY, Zhang YQ. PF-06409577 activates AMPK signaling and inhibits osteosarcoma cell growth. Front Oncol. 2021;11:659181. doi: 10.3389/fonc.2021.659181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Du Y, Zhang J, Yan S, Tao Z, Wang C, Huang M, Zhang X. Oridonin inhibits the proliferation, migration and invasion of human osteosarcoma cells via suppression of matrix metalloproteinase expression and STAT3 signalling pathway. J BUON. 2019;24:1175–1180. [PubMed] [Google Scholar]
  • 209.Lu Y, Sun Y, Zhu J, Yu L, Jiang X, Zhang J, Dong X, Ma B, Zhang Q. Oridonin exerts anticancer effect on osteosarcoma by activating PPAR-γ and inhibiting Nrf2 pathway. Cell Death Dis. 2018;9:15. doi: 10.1038/s41419-017-0031-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Jin S, Shen JN, Wang J, Huang G, Zhou JG. Oridonin induced apoptosis through Akt and MAPKs signaling pathways in human osteosarcoma cells. Cancer Biol Ther. 2007;6:261–268. doi: 10.4161/cbt.6.2.3621. [DOI] [PubMed] [Google Scholar]
  • 211.Kimura Y, Sumiyoshi M. Anti-tumor and anti-metastatic actions of wogonin isolated from Scutellaria baicalensis roots through anti-lymphangiogenesis. Phytomedicine. 2013;20:328–336. doi: 10.1016/j.phymed.2012.10.016. [DOI] [PubMed] [Google Scholar]
  • 212.Lin CC, Kuo CL, Lee MH, Lai KC, Lin JP, Yang JS, Yu CS, Lu CC, Chiang JH, Chueh FS, Chung JG. Wogonin triggers apoptosis in human osteosarcoma U-2 OS cells through the endoplasmic reticulum stress, mitochondrial dysfunction and caspase-3-dependent signaling pathways. Int J Oncol. 2011;39:217–224. doi: 10.3892/ijo.2011.1027. [DOI] [PubMed] [Google Scholar]
  • 213.Koh H, Sun HN, Xing Z, Liu R, Chandimali N, Kwon T, Lee DS. Wogonin influences osteosarcoma stem cell stemness through ROS-dependent signaling. In Vivo. 2020;34:1077–1084. doi: 10.21873/invivo.11878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Gioti K, Papachristodoulou A, Benaki D, Aligiannis N, Skaltsounis AL, Mikros E, Tenta R. Assessment of the nutraceutical effects of oleuropein and the cytotoxic effects of adriamycin, when administered alone and in combination, in MG-63 human osteosarcoma cells. Nutrients. 2021;13:354. doi: 10.3390/nu13020354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Morana JM, Leal-Hernande O, Canal-Macías ML, Roncero-Martin R, Guerrero-Bonmatty R, Aliaga I, Zamorano JD. Antiproliferative properties of oleuropein in human osteosarcoma cells. Nat Prod Commun. 2016;11:491–492. [PubMed] [Google Scholar]
  • 216.Przychodzen P, Wyszkowska R, Gorzynik-Debicka M, Kostrzewa T, Kuban-Jankowska A, Gorska-Ponikowska M. Anticancer potential of oleuropein, the polyphenol of olive oil, with 2-methoxyestradiol, separately or in combination, in human osteosarcoma cells. Anticancer Res. 2019;39:1243–1251. doi: 10.21873/anticanres.13234. [DOI] [PubMed] [Google Scholar]
  • 217.Yang S, Chen J, Tan T, Wang N, Huang Y, Wang Y, Yuan X, Zhang P, Luo J, Luo X. Evodiamine exerts anticancer effects against 143B and MG63 cells through the Wnt/β-catenin signaling pathway. Cancer Manag Res. 2020;12:2875–2888. doi: 10.2147/CMAR.S238093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Bai X, Meng H, Ma L, Guo A. Inhibitory effects of evodiamine on human osteosarcoma cell proliferation and apoptosis. Oncol Lett. 2015;9:801–805. doi: 10.3892/ol.2014.2791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Meng ZJ, Wu N, Liu Y, Shu KJ, Zou X, Zhang RX, Pi CJ, He BC, Ke ZY, Chen L, Deng ZL, Yin LJ. Evodiamine inhibits the proliferation of human osteosarcoma cells by blocking PI3K/Akt signaling. Oncol Rep. 2015;34:1388–1396. doi: 10.3892/or.2015.4084. [DOI] [PubMed] [Google Scholar]
  • 220.Kishida Y, Yoshikawa H, Myoui A. Parthenolide, a natural inhibitor of Nuclear Factor-kappaB, inhibits lung colonization of murine osteosarcoma cells. Clin Cancer Res. 2007;13:59–67. doi: 10.1158/1078-0432.CCR-06-1559. [DOI] [PubMed] [Google Scholar]
  • 221.Yang C, Yang QO, Kong QJ, Yuan W, Ou Yang YP. Parthenolide induces reactive oxygen species-mediated autophagic cell death in human osteosarcoma cells. Cell Physiol Biochem. 2016;40:146–154. doi: 10.1159/000452532. [DOI] [PubMed] [Google Scholar]
  • 222.D’Anneo A, Carlisi D, Lauricella M, Emanuele S, Di Fiore R, Vento R, Tesoriere G. Parthenolide induces caspase-independent and AIF-mediated cell death in human osteosarcoma and melanoma cells. J Cell Physiol. 2013;228:952–967. doi: 10.1002/jcp.24131. [DOI] [PubMed] [Google Scholar]
  • 223.Chang IC, Huang YJ, Chiang TI, Yeh CW, Hsu LS. Shikonin induces apoptosis through reactive oxygen species/extracellular signal-regulated kinase pathway in osteosarcoma cells. Biol Pharm Bull. 2010;33:816–824. doi: 10.1248/bpb.33.816. [DOI] [PubMed] [Google Scholar]
  • 224.Yang Q, Li S, Fu Z, Lin B, Zhou Z, Wang Z, Hua Y, Cai Z. Shikonin promotes adriamycininduced apoptosis by upregulating caspase3 and caspase8 in osteosarcoma. Mol Med Rep. 2017;16:1347–1352. doi: 10.3892/mmr.2017.6729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Mishra R, Nathani S, Varshney R, Sircar D, Roy P. Berberine reverses epithelial-mesenchymal transition and modulates histone methylation in osteosarcoma cells. Mol Biol Rep. 2020;47:8499–8511. doi: 10.1007/s11033-020-05892-8. [DOI] [PubMed] [Google Scholar]
  • 226.Liu Z, Liu Q, Xu B, Wu J, Guo C, Zhu F, Yang Q, Gao G, Gong Y, Shao C. Berberine induces p53-dependent cell cycle arrest and apoptosis of human osteosarcoma cells by inflicting DNA damage. Mutat Res. 2009;662:75–83. doi: 10.1016/j.mrfmmm.2008.12.009. [DOI] [PubMed] [Google Scholar]
  • 227.Gao X, Zhang C, Wang Y, Zhang P, Zhang J, Hong T. Berberine and cisplatin exhibit synergistic anticancer effects on osteosarcoma MG-63 cells by inhibiting the MAPK pathway. Molecules. 2021;26:1666. doi: 10.3390/molecules26061666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Zhao X, Zhang Q, Chen L. Triptolide induces the cell apoptosis of osteosarcoma cells through the TRAIL pathway. Oncol Rep. 2016;36:1499–1505. doi: 10.3892/or.2016.4957. [DOI] [PubMed] [Google Scholar]
  • 229.Li X, Lu Q, Xie W, Wang Y, Wang G. Anti-tumor effects of triptolide on angiogenesis and cell apoptosis in osteosarcoma cells by inducing autophagy via repressing Wnt/β-Catenin signaling. Biochem Biophys Res Commun. 2018;496:443–449. doi: 10.1016/j.bbrc.2018.01.052. [DOI] [PubMed] [Google Scholar]
  • 230.Qin W, Li S, Miao Y, Shi Q, Wang Y, Li J, Chen Z, Wang J, Ling C. Triptolide induces mitochondrial apoptosis through modulating dual specificity phosphatase 1/mitogen-activated protein kinases cascade in osteosarcoma cells. Neoplasma. 2018;65:21–33. doi: 10.4149/neo_2018_170109N16. [DOI] [PubMed] [Google Scholar]
  • 231.Wang B, Lu D, Xuan M, Hu W. Antitumor effect of sunitinib in human prostate cancer cells functions via autophagy. Exp Ther Med. 2017;13:1285–1294. doi: 10.3892/etm.2017.4134. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Clinical and Experimental Urology are provided here courtesy of e-Century Publishing Corporation

RESOURCES