Next Article in Journal
Multiple Roles of cAMP in Vertebrate Retina
Next Article in Special Issue
Dissecting the Role of Autophagy-Related Proteins in Cancer Metabolism and Plasticity
Previous Article in Journal
Pathophysiological Impact of the MEK5/ERK5 Pathway in Oxidative Stress
Previous Article in Special Issue
Curcumin-Mediated Resistance to Lenvatinib via EGFR Signaling Pathway in Hepatocellular Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Autophagy and Breast Cancer: Connected in Growth, Progression, and Therapy

Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287-0013, USA
*
Author to whom correspondence should be addressed.
Cells 2023, 12(8), 1156; https://doi.org/10.3390/cells12081156
Submission received: 6 March 2023 / Revised: 29 March 2023 / Accepted: 5 April 2023 / Published: 14 April 2023
(This article belongs to the Special Issue Autophagy and Human Cancers)

Abstract

:
Despite an increase in the incidence of breast cancer worldwide, overall prognosis has been consistently improving owing to the development of multiple targeted therapies and novel combination regimens including endocrine therapies, aromatase inhibitors, Her2-targeted therapies, and cdk4/6 inhibitors. Immunotherapy is also being actively examined for some breast cancer subtypes. This overall positive outlook is marred by the development of resistance or reduced efficacy of the drug combinations, but the underlying mechanisms are somewhat unclear. It is interesting to note that cancer cells quickly adapt and evade most therapies by activating autophagy, a catabolic process designed to recycle damaged cellular components and provide energy. In this review, we discuss the role of autophagy and autophagy-associated proteins in breast cancer growth, drug sensitivity, tumor dormancy, stemness, and recurrence. We further explore how autophagy intersects and reduces the efficacy of endocrine therapies, targeted therapies, radiotherapy, chemotherapies as well as immunotherapy via modulating various intermediate proteins, miRs, and lncRNAs. Lastly, the potential application of autophagy inhibitors and bioactive molecules to improve the anticancer effects of drugs by circumventing the cytoprotective autophagy is discussed.

Graphical Abstract

1. Autophagy, a Complex Process Designed to Support Cell Death as Well as Survival via Recycling

Autophagy is a highly conserved process that functions to transport cargos to the lysosome for recycling and cellular degradation in eukaryotes [1]. Autophagy not only serves to remove defective or damaged organelles and cellular components by self-digestion, as a catabolic mechanism, it recycles substrates required to sustain homeostasis when nutrients are scarce [1,2]. The significance of proper autophagy extends to the soundness of immune cell function [3], intercellular communication [4], regulation of tissue-resident stem cells [5,6], and the integrity of the tissue barrier [7]. It can be triggered by tissue remodeling, long-term nutritional deprivation, quality control of organelles, cellular stress, and immune reaction [8]. In ideal circumstances, autophagy can be cytoprotective or destructive because an immoderate self-degradation process can be damaging [9]. As the key cellular process that regulates the stress response and thus takes part in the quality control in the cells [10,11], autophagy has a recondite impact on human lifespan and health [4]. Consequently, numerous human illnesses, including neurodegeneration, myopathies, cancer, aging, and lung, liver and heart diseases, as well as metabolic complications such as diabetes are linked to autophagic dysfunction [12].
Autophagy can be distinguished into four categories based on how the protein is transported to the lysosome [2]—microautophagy, chaperone-mediated autophagy, macroautophagy, and selective autophagy [2]. In microautophagy, the lysosomal membrane will undergo invagination or protrusion for cargo uptake [13]. Instead of manipulating membrane structures, chaperone-mediated autophagy uses chaperones for cargo identification that carry a specific pentapeptide motif. Subsequently, each of these components are then unfolded and individually translocated through the lysosomal membrane [14]. In contrast, macroautophagy generates double-membrane vehicles (autophagosomes) for cargo sequestration [15]. Guided by specific autophagy-related genes (ATGs) and BECN1 (Beclin-1), the initiation step of macroautophagy precedes phagophore elongation, autophagosome maturation, and fusion of lysosome and autophagosome. This process is concluded by proteolytic degradation of the cargo [2]. On the other hand, the macroautophagy of a particular cellular component is known as selective autophagy. Different from macroautophagy, the key regulator of selective autophagy is PINK1 (phosphatase and tensin homolog-induced putative kinase 1) [2]. The specificity of selective autophagy is preserved by ubiquitination or labeling of each cargo. In this process, p62 is an autophagy substrate that serves as a reporter [16]. Subsequently, autophagy receptors selectively bind to the tagged cargo and proceed to the formation of autophagosome [17,18]. These types of autophagy are mechanically varied, but they all culminate in lysosomal degradation of unwanted substances in the cell [1]. Adding to this complexity, several key proteins of autophagy machinery are known to be regulated by long non-coding RNAs (lncRNAs). LncRNA H19 promotes autophagy via modulating the Let-7–Lin28 axis [19], whereas LC3 and beclin1 are targeted by lncRNA ROR leading to autophagy promotion [20]. ATG10 is activated by direct binding of the lncRNA AGAP2-AS1–ELAVL1 complex to its promoter region [21]. LncRNAs, including HOTAIR, TALNEC2, EGOT, ZNF649, GAS5, DANCR, OTUD6B, and NAMPT, have been reported to regulate the expression of autophagy-associated proteins and impact cancer progression [22,23].
Interestingly, in cancer cells autophagy contributes to both death and survival [24]. The impact of autophagy in homeostasis serves to guard the genomic integrity of quiescent and growing cells in tissues [25]. Since genome instability is one of the cancer hallmarks, fidelity of autophagy has the leverage to prevent healthy cells from becoming cancerous [4,26]. It has been reported that autophagy in healthy cells prevents tumorigenesis via counteracting pro-oncogene stimuli [27]. Autophagy also activates the oncogene-induced senescence program, which keeps proliferative events at bay [28]. Nevertheless, many factors, such as the stage of disease, type of cancer, and condition of the patient can interfere with the real impact of autophagy in the progression of cancer [29].

2. Complex Relationship between Key Autophagic Proteins and Various Aspects of Breast Cancer Growth

Healthy cells commonly face intrinsic and extrinsic stress that can potentially result in genomic instability and mutations, which will aid neoplasia and hyperproliferation [30]. Autophagy serves to prevent such complications by eliminating tumorigenic stressors such as oncoproteins, protein aggregates, reactive oxygen species (ROS) production, and dysfunctional mitochondria [26,31,32,33,34,35,36,37,38,39]. Besides that, autophagy has roles in immune responses and inflammation [40,41,42,43]. As such, the maintenance of cellular integrity and defense against neoplastic transformation are both facilitated by autophagy [40]. Owing to the cytoprotective function of autophagy, it serves to suppress tumorigenesis in terms of cancer initiation. Indeed, an elevated gene signature for autophagy is observed in healthy mammary glands, which is found to be decreased as breast cancer progresses [44]. Indeed, autophagy is a complex multistep process involving multiple proteins that participate in breast cancer initiation, growth, and metastatic progression as well as recurrence.

2.1. BECN1 Negatively Associates with Breast Cancer

Macroautophagy is the most well-studied subtype of autophagy [9]. When cells are under stress, ULK1 is activated directly or indirectly leading to the recruitment of BECN1 and ATGs, thus allowing the assembly of molecular complexes, which subsequently lead to the initiation of phagophore formation [45,46]. BECN1 deficiency is observed in breast cancer [47]. Consistently, monoallelic loss of BECN1 is often observed in human breast cancer cells [47,48]. Further, progression of ex vivo HER2-enriched breast tumor is hindered upon overexpression of BECN1 [49]. In addition, monoallelic deletion of Becn1 in FVB/N mice results in the development of mammary tumors after parity [50]. In fact, mammary tumorigenesis in MMTV-Wnt1 mice with monoallelic Becn1 deletion is more aggressive compared with those in mice with homozygous Becn1 [50]. Among human breast cancer samples excluding HER2-enriched tumors, tumors with overexpression of WNT-related genes and a low mRNA level of BECN1 present a poorer prognosis, and they are primarily TNBC [50,51]. On the other hand, it has been recognized that the etiology and aggressive phenotype of TNBC is related to the activation of the Notch1 pathway [52]. Macroautophagy has roles in such oncogenic signaling as well. Of note, BECN1 can induce autophagic degradation of Notch1, which leads to a phenotype that diminishes Notch1-signaling-dependent tumorigenesis. Indeed, silencing BECN1 in TNBC cell lines leads to an enhanced clonogenicity, migration, and anchorage-independent growth [53]. These studies suggest that the expression of the autophagic indicator, BECN1, hampers the progression of breast cancer.

2.2. Autophagy-Related Genes (ATGs) Have a Dual Impact on Breast Cancer

There are approximately twenty evolutionarily conserved ATGs that actively participate in the autophagic process. Depending on the context, some ATGs may contribute to the prevention of tumorigenesis [43]. For example, enrichment of ATG7 has a negative impact on growth and glycolysis in TNBC cells. Similarly, TNBC tumors that bear a higher level of ATG7 present better prognoses [54]. On the other hand, autophagy in human breast epithelial cells with mutationally active oncogenic Ras can be pro-tumorigenic [55]. In such mutated cells, an enhanced glycolysis capacity and proliferation is observed in autophagy-competent cells compared with autophagy-deficient cells. Additionally, more autophagy-competent cells undergo Ras-mediated adhesion-independent transformation, which suggest that autophagy has the potential to stimulate Ras-mediated tumorigenesis under certain metabolic conditions [55]. Of note, given that autophagy can stimulate Signal Transducer and Activator of Transcription 3 (STAT3), and that STAT3 is frequently activated in TNBC, modulation of autophagy influences the TNBC subtype the most [56].

2.3. FOXO Can Modulate Breast Cancer via Autophagy

As a putative tumor suppressor, Forkhead Box O (FOXO) is a transcription factor that takes part in regulating cellular homeostasis, the maintenance of stemness, and aging [43,57]. Downregulation of FOXO1 in breast cancer is related to a worse prognosis in breast cancer, especially in HER2-positive subtypes [58]. Similarly, nuclear localization of FOXO3 is found to be related to a reduced metastatic event in luminal-like breast cancer [59]. Interestingly, FOXO3 has the leverage to induce the expression of proteins that participate in the initiation and autophagosome formation in macroautophagy. In agreement with this, the loss of FOXO3 leads to diminished expression of several ATGs, resulting in a declined activity of autophagy [60,61,62]. Indeed, tumorigenesis can be stimulated by the absence of FOXO3, which implies suppression of FOXO3-mediated autophagy contributes to mammary carcinogenesis [60,61,62]. To add another level of complexity to this matter, FOXO3 also induces autophagy in cancer stem cells (CSCs) to preserve their well-being, and thus contributes to recurrence and metastasis [63,64]. Altogether, FOXO3 has the leverage to suppress tumorigenesis in healthy cells but may induce cytoprotective autophagy in cancer stem cells.

2.4. Autophagy Contributes to Reduced Drug Sensitivity in Breast Cancer

Breast-cancer-related mortality has been increasing in the past two decades [43,65]. Following preliminary diagnosis, metastatic relapse accounts for 90% of breast-cancer-related deaths, which is ascribed to the resurgence of dormant breast cancer cells [66,67]. Many therapeutics for breast cancer, namely chemotherapy, target actively dividing cells by destructing DNA and key proteins [68,69,70,71]. In such circumstances, autophagy serves to breakdown long-lived proteins, macromolecular waste, and damaged organelles. Residual cancer cells that survive the therapeutic assault may result in dormancy transformation. Since autophagy can be used as nutritional support, the cells will have time to repair and thus contribute to chemoresistance, relapse, and disease progression [72]. Autophagy thereby decreases drug sensitivity to breast cancer cells while protecting them. Hence, autophagy can be cytoprotective to breast cancer cells [72].

2.5. Autophagy Influences Tumor Dormancy in Breast Cancer

When the environment becomes unfavorable for growth, tumor cells can become quiescent, which is termed tumor dormancy [73]. It has been discussed that tumor dormancy largely contributes to metastasis, disease recurrence, and therapy resistance [73,74]. Dormant cancer cells can remain latent for decades before being activated to a proliferative state [75]. Autophagy not only supports the growth of dormant cells within the tumor microenvironment (TME) [76,77] but also participates in distant colonization and extravasation of dormant cells under environmental stress [66,78]. Upon inhibition of autophagy in vivo and in human preclinical models of dormant breast cancer cells with hydroxychloroquine (HCQ), a substantial decline of metastasis burden and cell viability have been observed [66]. This phenomenon is ascribed to the accumulation of damaged mitochondria and ROS, which in turns leads to cell death [72]. Further, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) acts to promote cell cycle progression while inhibiting apoptosis [43]. PFKFB3 can also be a substrate for autophagosomal degradation by interacting with the autophagy receptor p62 [79]. Interestingly, metastatic breast cancer cells bear higher levels of PFKFB3 compared with dormant cells but have less autophagic activity. Of note, low levels of autophagy stabilize PFKFB3, which leads to activation of dormant cells to a metastatic state [79]. These findings suggest that autophagy promotes dormancy in breast cancer cells [43]. The AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) pathways are essential for autophagic regulation in tumor cells [80]. Canonically, activation of the mTOR complex 1 (mTORC1) directly phosphorylates and thus sequesters transcription factor EB (TFEB) in the cytoplasm [81,82]. Since TFEB is a chief transcriptional regulator of lysosomal and autophagy genes, activation of mTOR suppresses induction of autophagy at the transcriptional level [82]. For AMPK-mediated regulation of autophagy, AMPK is activated upon energy depletion, which in turn inhibits the autophagy regulatory complex, thus resulting in disruption of autophagosome biogenesis [83,84]. Additionally, activated AMPK can phosphorylate Unc-51-like kinase (ULK1) and the TSC1/TSC2 complex, thereby inducing autophagy via suppressing the activation of mTORC1 [80,85]. In breast cancer, environmental stress stimulates the secretion of auto- and paracrine signaling factors, which block phosphoinositide 3-kinase (PI3K) activation and lead to the inactivation of AKT and mTOR, thereby resulting in the activation of autophagy [43,75]. The PI3K/AKT/mTOR pathways can be inhibited by Diras Family GTPase 3 (DIRAS3), which is found to be enriched in dormant breast cancer cells [75,86,87], suggesting that autophagy may contribute to tumor dormancy in breast cancer and thus plays a role in chemoresistance.

2.6. Autophagy Influences Hypoxia, Chemoresistance, and Stem-like Phenotype in Breast Cancer

It is known that cancer stem cells are a significant contributor to the development of chemoresistance in breast cancer [88,89,90,91]. Autophagy enables the survival of CSCs under hypoxia in the tumor microenvironment [92]. In fact, a subpopulation of TNBC cancer stem cells stay in an autophagic state in relation to hypoxia [93]. Indeed, environmental stress such as nutrient deprivation and hypoxia can lead to the activation of autophagy for cellular component recycling in order to sustain survival [94,95,96]. In cancer cells, hypoxia-inducible factor-1 (HIF-1) is the main regulator of hypoxic conditions [97,98]. Upon activation of HIF-1, stemness can be triggered via several pathways such as activation of NANOG, SOX2, SOX17, etc. [98,99,100]. Importantly, hypoxia stimulates autophagy via HIF-1α [101], a subunit of HIF-1. HIF-1α is involved in the generation, differentiation, invasion, plasticity, and therapeutic resistance of CSCs [92]. In two stem-like breast cancer cell lines, induction of stemness can be performed by autophagy via the EGFR/Stat3 and TGFβ/Smad pathways in a murine model [102]. It is also reported that inhibition of autophagy in certain breast cancer cell lines results in a decreased stemness phenotype [56,103]. Other than that, dormant stem-cell-like breast cancer cells express autophagy markers, and upon inhibition of autophagy using 3-methyladenine (3-MA), these cells are transformed to active state [104]. Moreover, doxycycline not only inhibits EMT (epithelial–mesenchymal transition) and stemness markers in breast cancer stem cells but also causes a down-regulation of autophagy activity [105], suggesting the possibility that autophagy may play a role in stemness [106].
In patient-derived xenografts, suppression of autophagy via inhibition of BECN1 leads to re-sensitization of chemoresistant cells to therapy [93], which emphasizes the role of autophagy in the development of chemoresistance. In luminal and HER2-enriched subtypes of breast cancer, similar results are observed, as chemoresistant cells not only have an elevated autophagic activity compared with their drug-sensitive counterparts but inhibition of autophagy also results in the restoration of chemosensitivity [107,108,109,110]. Further, expression of mesenchymal markers, vimentin, and the stem cell marker CD44 is increased upon autophagic activity in CSCs [106]. Additionally, self-renewal of a hormone-independent murine breast cancer cell line LM38-LP requires autophagy [111]. Interestingly, disruption of circadian rhythm is related to the acquirement of chemoresistance [112]. By itself, melatonin suppresses the development of chemoresistance in breast cancer by interfering with tumor metabolism [112,113]. However, the combination of dim light at night (dLAN) results in the activation of STAT3, which is often overexpressed in paclitaxel-resistant breast cancer [112]. Under the synergetic influence of dLAN and melatonin, the activated STAT3 inhibits DIRAS3 in an epigenetic manner, resulting in decreased autophagic activity and increased resistance of breast cancer to paclitaxel [112]. This finding implies that DIRAS3 can be a regulator for the development of chemoresistance via autophagy. On the other hand, it has been demonstrated that chemotherapeutics can trigger autophagy, which enhances the survival of CSCs [114]. In TNBC, inhibiting autophagy with chloroquine (CQ) causes the accumulation of dysfunctional mitochondria and ROS in CSCs, which results in cell death [115]. Interference of autophagy also disrupts the preservation of breast CSCs. When combining autophagy inhibitors and chemotherapy, a decreased expression of stemness markers is observed along with an increased sensitivity to chemotherapeutics and a decreased cancer cell viability and metastasis [105,111,115,116]. Indeed, autophagy can be cytoprotective by contributing to induced chemosensitivity in breast cancer cells [72].

2.7. Intermediate Steps in the Autophagic Process Play an Important Role in Breast Cancer

Autophagy flux can impact how breast cancer cells respond to treatment. Unsurprisingly, in-depth inquiries about autophagy have discovered that intermediate regulation of autophagy flux can impact the influence of autophagy on therapeutic resistance [72]. For example, the reporter in selective autophagy, p62, also has roles in the proteolytic system [16]. It not only functions to deliver ubiquitinated proteins to the proteasome for breakdown but also governs protein quality by binding with ubiquitinated cargoes while shuttling between the nucleus and cytoplasm [16]. Upon administration of bortezomib (a proteasome inhibitor), an elevated p62 expression is observed, which implies the failure in the turnover of autolysosomal protein, demonstrating the interplay between proteasomal degradation and selective autophagy [117]. Breast cancer cells fail to restore metabolic homeostasis via autophagy upon inhibition of autophagosomal degradation using obatoclax [118]. Further, the combined treatment of bortezomib and obatoclax in antiestrogen-resistant breast cancer cells results in a hindered autolysosomal function without preventing the formation of autophagosomes [117,118]. Interestingly, this sensitizes antiestrogen-resistant breast cancer cells to tamoxifen [76,118], which indicates that the indirect influence of the proteasome pathway can contribute to drug resistance. On the other hand, lysosomal-associated protein transmembrane 4β (LAPTM4B) is an essential maturation step in autophagy but also plays a significant role in lysosomal activities [119]. Expression of LAPTM4B is positively related to chemoresistance in breast cancer [119]. From the mechanistic standpoint, deficiency of LAPTM4B leads to an enhanced permeability of the lysosomal and the autolysosomal membranes [120]. Consequently, drugs can enter the nucleus more readily. The increased permeability of cathepsin also causes cathepsin to be released, which triggers lysosomal-mediated programmed cell death [120]. LAPTM4B deficiency significantly hinders the fusion of lysosomes and autophagic bodies, which begets the accumulation of autophagosomes, resulting in cell death [121] and thus diminishing therapeutic resistance. In summary, simply increasing or decreasing autophagic activity is not a wise route to regulate autophagy-mediated therapeutic resistance in breast cancer. It is also important to inquire how autophagy regulates the sensitivity of breast cancer to therapeutics [72].

3. A multifaceted Involvement of Autophagic Processes Modulate the Efficacy of Breast Cancer Therapeutics

Several studies suggest the involvement of autophagy in endocrine therapy, chemotherapy as well as immunotherapy. As autophagy can be cytoprotective as well as cytotoxic, its impact on various therapies can be context dependent, leading to either the development of resistance or increased efficacy [122,123,124,125].

3.1. Autophagy and Endocrine Therapy—Response and Resistance

Estrogen-receptor-positive breast cancer accounts for the majority of breast cancer cases, and they are predominantly treated with endocrine therapy [126]. The estrogen receptor antagonist tamoxifen has been successfully used for over 40 years [127]; however, some cancers do not respond to tamoxifen [128], owing to intrinsic or acquired resistance [129]. Several mechanisms associated with tamoxifen resistance including alteration of the estrogen receptor 1 (ESR1) gene, modulation of coregulator proteins, and modification of proliferative signaling pathways have been put forth [129,130,131]. Recent studies have explored the role of autophagy in tamoxifen resistance [132,133]. G protein coupled estrogen receptor (GPR30), a transmembrane estrogen receptor, contributes to tamoxifen resistance [134] via transactivating epidermal growth factor receptor (EGFR) [134,135,136], leading to the activation of several pathways that regulate autophagy, including the NCK/PAK/JNK, JAK/STAT, PI3K/AKT/mTOR, PLC/PAG/PKC, and MAPK pathways [137,138]. GPR30 is highly expressed in breast-cancer-associated fibroblasts [139,140], and GPR30-induced gene activation results in an enrichment of high mobility group box 1 (HMGB1) [132], which induces tamoxifen resistance through the MEK–ERK signaling pathway while increasing autophagic activities. A higher level of MTA1 (metastasis associated antigen 1) and autophagic activity has been observed in tamoxifen-resistant cells compared with their non-resistant counterparts [141]. Interestingly, knocking down MTA1 in tamoxifen-resistant cells not only sensitizes them to tamoxifen but also decreases autophagic activity [141,142]. In fact, the tamoxifen resistance conferred by MTA1 is associated with modification of autophagic activity [141]. Mechanistically, as evidenced by an increased AMP:ATP ratio, stable expression of MTA1 leads to the activation of AMPK and increased availability of p-PRKAA (phospho-protein kinase AMPK-activated catalytic subunit alpha), which increases autophagy in tamoxifen-resistant cells [141]. Interestingly, both autophagic activity and tamoxifen resistance were substantially decreased when PRKAA is knocked down in MCF7/TAMR-1 cells, even when MTA1 is overexpressed, implying that the AMPK pathway is essential to regulate autophagy, which is important for tamoxifen resistance [141]. When it comes to endocrine resistance, estrogen withdrawal can activate UPR (unfolded protein response) [143], leading to the release of glucose-regulated protein 78 (GRP78) [144], which balances pro-survival autophagy and pro-death apoptosis while bestowing endocrine resistance in ER-positive breast cancer [145,146]. Knockdown of GRP78 in MCF7/LCC9 (resistant to fulvestrant and tamoxifen) and MCF7-RR (tamoxifen-resistant) results in re-sensitization to the respective therapeutics [145]. Mechanistically, GRP78-mediated signaling may suppress apoptosis and stimulate autophagy to help cells endure the stress [147] via inhibition of mTOR [145,148]. In the breast tumor microenvironment, it has been reported that GRP78 also promotes autophagy by binding to insulin-like growth factor binding protein-3 (IGFBP-3), which is associated with poor prognosis [149]. Additionally, GRP78 indirectly activates BECN1 to promote autophagy in breast cancer cell lines (Figure 1) [148,150,151].
In addition to the genes/proteins associated with tamoxifen resistance and autophagy, recent studies have pointed out that long noncoding RNAs (lncRNAs) are involved in tamoxifen resistance [133,152,153]. In line with this, lncRNA H19 is found to be highly enriched in breast cancer cell lines and tumor tissues that are resistant to tamoxifen, and H19 knockdown in tamoxifen-resistant models results in a resensitization to tamoxifen [133]. Importantly, the expression of H19 is positively related to autophagic activity in MCF7/TAMR cells. Increased methylation in the promoter region of BECN1 is observed upon H19 silencing in tamoxifen-resistant cells. It is evident that H19 promotes autophagy via epigenetic modification of BECN1 [133]. As another type of non-coding molecule, microRNAs (miRs) also regulate a number of cellular processes including autophagy [72]. Endocrine resistance is negatively related to the expression of miR-214, which directly targets uncoupling protein 2 (UCP2) [154]. Interestingly, while it is well-recognized that the activation of mTOR leads to the inhibition of autophagy [82,155,156], UCP2 overexpression contradictorily leads to the enhanced activity of both the PI3K–Akt–mTOR signaling pathway and autophagic activity [154]. A previous study has speculated that the influence of UCP2 in autophagy and endocrine resistance is through the regulation of the PI3K–Akt–mTOR signaling pathway [154,157]. However, more research is needed to better understand the miR-214–UCP2-mediated autophagy and tamoxifen resistance in breast cancer [154]. Similarly, miR-23b-3p inhibits SLC6A14 (Solute Carrier Family 6 Member 14), a basic amino acid transporter, and results in an altered level of amino acids [158]. Such disruption may stimulate autophagy, and it has been reported that inhibition of SLC6A14 enhances autophagic activity in colon cancer [159,160]. Consistent with this, it has been reported that tumors resistant to endocrine therapy have a reduced SLC6A14 activity and upregulation of miR-23b-3p, leading to overactivation of cytoprotective autophagy that contributes to the development of therapeutic resistance [158]. Collectively, these findings reveal the importance of autophagy in the development of endocrine resistance in breast cancer (Figure 1).

3.2. Autophagy and Targeted Therapy—Response and Resistance

Trastuzumab (Herceptin)—The Her2-positive breast cancer subtype accounts for ~20% of all breast cancers [161], and it is associated with poor prognosis [162]. Trastuzumab is a monoclonal antibody that targets the extracellular region of HER2 serving as a targeted therapy for Her2-positive breast cancer; however, many patients develop resistance to trastuzumab following an initial response [163]. Multiple modulators of trastuzumab resistance have been uncovered. Sphingosine kinase 1 is a protooncogene associated with therapeutic resistance in breast cancer [164,165] and can be inhibited with the synthetic structural analog of sphingosine, Fingolimod (FTY720) [166,167]. Interestingly, FTY720 inhibits autophagy and triggers apoptosis in trastuzumab-resistant breast cancer cells [168]. In fact, co-treatment of FTY720 and trastuzumab results in a substantial growth inhibition compared with monotherapy [168], and the results are comparable to autophagy inhibitors [168,169], implying that FTY720 restrains autophagy to reverse therapeutic resistance while activating apoptosis [168,170]. Autophagy can be regulated by miRNA via posttranscriptional regulation [72,171]. As an important regulator of many biological processes, miRNAs have been demonstrated to participate in tumor progression and treatment response [172]. Unsurprisingly, miRNA can regulate autophagy via posttranscriptional modulation of autophagy-related protein expression [173]. Interestingly, miR-567 promotes the efficacy of trastuzumab in trastuzumab-resistant breast cancer cells by targeting ATG5, which results in the suppression of autophagy [173]. An enhanced expression of a lncRNA, ZNF649-AS1 (ZNF649 Antisense RNA 1), is detected in trastuzumab-resistant breast cells compared with trastuzumab-sensitive cells [174]. Inhibition of ZNF649-AS1 in trastuzumab-resistant cells results in suppression of autophagy and cells are re-sensitized to trastuzumab [174]. Further investigation presents that ZNF649-AS1 binds to polypyrimidine tract binding protein 1 (PTBP1), which stimulates the translation of ATG5 and thus results in enhanced autophagic activity, thereby contributing to the acquisition of trastuzumab resistance (Figure 2) [174].
EGFR inhibitors—As an important therapeutic target in cancer, the epidermal growth factor receptor (EGFR) family is often abnormally activated in many types of cancers and plays an important role in their development and progression [175,176]. Lapatinib is a small molecule tyrosine kinase inhibitor of HER2 and EGFR [177]. Mechanically, lapatinib binds to the cytoplasmic ATP-binding sites of HER2 and EGFR, thus preventing the binding of ATP and resulting in the inhibition of tyrosine kinase phosphorylation and downstream events [177]. Lapatinib-resistant breast cancer cells have higher levels of autophagic activity and they can be sensitized to treatment upon inhibition of autophagy [178]. Lapatinib-induced autophagy requires p62, which can be inhibited by a triterpenoid extracted from Schisandra plants (P3–15) to resensitize TNBC to lapatinib treatment in vitro and in vivo [179]. It is clear that autophagy aids in the development of lapatinib resistance, but the underlying mechanisms are yet to be elucidated [178]. Similar to lapatinib, gefitinib is also a tyrosine kinase inhibitor that targets EGFR and HER2 in breast cancer [180,181,182,183]. Among TNBC patients, 40% of them have enrichment of EGFR [184,185], but inhibiting EFGR is generally not efficient for the treatment of TNBC patients owing to drug resistance. Increasing evidence has pointed to the contribution of autophagy in resistance to EGFR inhibitors [186]. Interestingly, TNBC is sensitized to gefitinib therapy when autophagy is suppressed, which implies that the combination of autophagy inhibitor and gefitinib can be a good therapeutic strategy in treating TNBC patients who have enriched EGFR [187]. Silencing EGFR in SKBR3 (HER2+ breast cancer cell line) and MCF7 cells results in an elevated autophagy response. It is speculated that inhibition of EGFR in both gefitinib-sensitive or -insensitive cell lines can lead to disruption of basal intracellular glucose levels via inhibition of tyrosine kinase, and thus autophagy is triggered to offset the energy depletion [188,189]. Additionally, gefitinib-mediated autophagy activation can be a consequence of altered downstream signaling of EGFR and HER2 [188]. Yet again, more studies are needed to explore how autophagy contributes to therapeutic resistance.
CDK4/6 inhibitors—Cyclin-dependent kinases are a group of proteins that govern the cell cycle progression [190]. Cyclin-dependent kinase 4 (CDK4) and CDK6 are dysregulated in cancer cells and many preclinical studies have demonstrated the hyperactivity of the cyclin D–CDK4/6 axis, rendering CDK inhibitors as desirable therapeutic approaches [191,192,193]. CDK4/6 activation results in the phosphorylation of Serine/threonine kinase 11 (STK11) at S325 by activated cyclin D1 (CCND1, a regulatory subunit of CDK4/6). Activation of CDK4/6 leads to inactivation of AMPK, which in turn relieves its suppression on mTOR; activated mTOR can then lead to inhibition of autophagy [194,195]. The current approved CDK4/6 inhibitors are Abemaciclib, Palbociclib, and Ribociclib [196,197]. It is recognized that CDK4/6 inhibitors promote autophagy in various cancer models, including breast cancer [198,199,200]. Mechanistically, CDK4/6 inhibition renders CCND1 inactive, leading to AMPK activation and increased autophagy [194,201]. Indeed, when autophagy is inhibited, the therapeutic response of breast cancer to palbociclib is greatly increased [200], which suggests that inhibition of autophagy can be an attractive avenue for overcoming drug resistance [200,202]. Consistently, transcriptomic profiling results of palbociclib-sensitive and -resistant breast cancer cells reveal that resistant cells have upregulation of many autophagy-related genes [203]. Additionally, autophagy assists cells in managing stress from CDK4/6 inhibition by preventing apoptosis [203]. Combining CDK4/6 inhibitors and lysosomal destabilizers results in improved therapeutic efficacy in CDK4/6-inhibitor-resistant TNBC cells [204]. This demonstrates that lysosomes can be a therapeutic target to cope with resistance to CDK4/6 inhibitors (Figure 2) [205].

3.3. Autophagy and Radiotherapy—Response and Resistance

In radiotherapy, cancer cells are subjected to high physical energy of radiation that causes enormous DNA damage resulting in cell death [206]. Autophagy may be stimulated to govern cancer cell survival post-radiotherapy [207]. Damaged-regulated autophagy modulator 1 (DRAM1) encourages autophagy, possibly by suppression of the AKT signaling pathway or augmentation of lysosomal acidification [208]. Unsurprisingly, miRNA may regulate the effect of radiotherapy via modulation of autophagy. As reported by a recent study, overexpressing miR-26b can lead to downregulation of DRAM1, which in turn leads to decreased autophagic activity and increased radiation sensitivity in breast cancer cells [209]. Similarly, miR-200C has been demonstrated to downregulate Ubqln1 (ubiquitin 1) when expressed ectopically in breast cancer [210]. In autophagy-mediated degradation, Ubqln1 is important in autophagosome maturation [211]. In a combined clinical and in vitro study, expression of miR-200C results in enhanced sensitivity to radiotherapy, which is ascribed to downregulation of Ubqln1 and thus reduction of autophagy (Figure 3) [210].

3.4. Autophagy and Chemotherapy—Response and Resistance

Anthracycline—Doxorubicin and epirubicin are both anthracyclines that are widely used chemotherapeutic agents that interfere with DNA replication and transcription, thus stopping cell proliferation [212]. As a member of the high mobility group protein superfamily, HMGB1 has secretory and intracellular activity and participates in breast cancer tumorigenesis [210,213,214]. The role of HMGB1 in the cytoplasm is associated with BECN1-mediated autophagy [215], metastasis, and chemo- and radiotherapy resistance in breast cancer [216,217,218,219,220]. Its contribution to therapeutic resistance is ascribed to the induction of autophagy [221,222,223]. The therapeutic efficacy of doxorubicin can be improved by inhibiting Med19 (Mediator Complex Subunit 19), which downregulates HMGB1 and results in the suppression of autophagy [219]. HMGB1 is positively related to autophagy level, NFκB/p65 activity, and doxorubicin-resistance in breast cancer cells [219]. The silencing of HMGB1 also reduces radiotherapy resistance in breast cancer, which is known to be related to the diminished autophagic activity [223]. Emerging studies have also suggested that ATG5 can promote chemoresistance in different types of cancers [224,225]. Indeed, an increased epirubicin response is observed in both anthracycline-sensitive and -resistant TNBC cells upon knockdown of ATG5 [226]. Combination of autophagy inhibitor and anthracycline has the potential to combat chemoresistance in TNBC (Figure 4) [226].
Paclitaxel—As one of the most widely used chemotherapies in metastatic breast cancer, taxane inhibits microtubule depolarization, thus arresting cells at prometaphase and resulting in cell death [227,228,229]. A recent study reported that APRIL, a proliferation-inducing ligand, is related to chemoresistance in TNBC cells [230]; its inhibition sensitizes paclitaxel-resistant TNBC to treatment, whereas overexpression of APRIL promotes chemoresistance [230]. APRIL modulates AKT-mTOR activity [230] to induce autophagy leading to chemoresistance. Indeed, autophagy inhibitors sensitize breast cancer cells to paclitaxel treatment, suggesting that APRIL stimulates the development of chemoresistance by activating autophagy [230]. Interestingly, miR-18a also promotes paclitaxel resistance in TNBC cells by hindering paclitaxel-induced apoptosis and suppressing the expression of Dicer, which is a processer for miRNA [231,232]. DICER promotes autophagy by inhibiting the activation of the PI3K/AKT/mTOR pathway and thus leads to cisplatin resistance non-small cell lung cancer [233]. It is, however, unclear how DICER impacts autophagy in breast cancer paclitaxel resistance. The role of miR-18a and increased autophagy is corroborated by another study that shows how paclitaxel-resistant MDAMB231 cells have higher basal levels of both miR-18a and autophagic activity compared with parental cells [234]. Mechanistically, miR-18a suppresses the mTOR signaling pathway, which in turn leads to an elevated autophagy activity and thus contributes to paclitaxel resistance [234] Interestingly, administration of an autophagy inhibitor yielded a similar, yet stronger effect compared with miR-18a inhibition in paclitaxel-resistant TNBC cells [234]. Even though the mechanistic story of how autophagy contributes to paclitaxel resistance is unclear, it is evident that autophagy is beneficial to chemoresistance in TNBC (Figure 4).
Platinum Agents—The use of platinum agents in treating breast cancer can be dated back to the 1970s [227]. Carboplatin and cisplatin are both platinum agents that function by interfering with DNA strand separation, thus inducing cell death in the middle of DNA replication and transcription [235,236,237]. It is observed that co-administration of carboplatin and autophagy inhibitors results in a drastic reduction in TNBC tumors in vivo [115]. Although there is no other research that directly uncovers the relationship between autophagy and resistance to platinum agents in breast cancer, it has been reported that autophagy is one of the big contributors to the resistance to platinum agents [238]. Upon administration of platinum agents, increased autophagy is reported in esophageal cancer [239], colon cancer [240], liver cancer [241], and neuroblastoma [242]. Accordingly, it has been demonstrated that inhibition of autophagy can reduce platinum resistance in endometrial cancer [243] and ovarian cancer [244].

3.5. Autophagy and Immunotherapy—Response and Resistance

In recent decades, immunotherapy consisting of monoclonal antibodies (anti-PD-1 or anti-PD-L1) has been widely used in breast cancer patients [245,246]. Sigma 1 receptor (S1R) is a unique drug-binding site that is commonly expressed in malignant breast epithelial cells and breast cancer cells [247]. Of interest, inhibition of S1R promotes autophagic flux, resulting in reduced levels of cell surface PD-L1, which succumbs to autophagic degradation [248]. Inversely, treatment with an S1R activator leads to an elevated expression of cell surface PD-L1. This phenomenon is consistent in both TNBC cells and prostate cancer cells [248]. The role of autophagy, in this case, is neither cytoprotective nor cytotoxic; instead, it solely functions as a degradation tool. When S1R is expressed, autophagic flux is inhibited; therefore, PD-L1 is overexpressed on the cancer cell surface, modulating the efficacy of immunotherapy [249]. As aforementioned, HMGB1 may promote autophagy in breast cancer, thus contributing to therapeutic resistance [219]. As a multifunctional redox-sensitive protein, it participates in both intracellular processes such as autophagy, chromatin remodeling, and regulation of transcription [250,251,252,253] and also has roles in extracellular processes such as the regulation of autoimmunity and inflammation [254,255,256]. Recently, a role for HMGB1 blockage in breast cancer and immunotherapy has been uncovered [257]. In immunocompetent mice, co-administration of HMGB1 inhibitor and anti-PD-1 immunotherapy results in a substantial reduction of tumor growth compared with mice who received anti-PD-1 therapy alone [257], suggesting that combination treatment of immunotherapy and an HMGB1 inhibitor can be an optimal strategy. Similarly, autophagy inhibitors combined with immunotherapy lead to an enhanced therapeutic response in pancreatic cancer [258]. There are a couple of reasons that may explain the synergistic effect of autophagy inhibition alongside immunotherapy. Inhibition of autophagy may increase the amount of surface MHC-I on dendritic cells, which strengthens the immune response mediated by CD8+ T cells [259]. Additionally, suppression of autophagy can also directly augment the tumor-suppressing ability of CD8+ T cells [258,260]. At this point, very limited studies have investigated how autophagy contributes to resistance to immunotherapy in breast cancer. As immunotherapy becomes more prominent, autophagy can be a worthy topic of exploration to increase therapeutic efficacy (Figure 5).

4. Inhibition of Autophagy Using Various Inhibitors May Result in Improved Therapeutic Outcomes

4.1. Hydroxychloroquine (HCQ)

Hydroxychloroquine is a weak base that inhibits autophagy by accumulating in the lysosomal compartment and thereby inhibiting the formation of autolysosomes [261]. Many preclinical studies that utilize autophagy inhibitors indicate that HCQ can lead to cancer cell death alone or can strengthen the efficacy of various chemotherapies and targeted therapies [262,263,264]. Some clinical trials that attempted to investigate the combination of HCQ and other therapeutics such as Ixabepilone (NCT00765765) or hormonal therapy (NCT02414776) have been terminated due to slow accrual and the departure of PI, respectively, whereas another study that wished to examine the combination of Gedatolisib (NCT03400254) has been withdrawn due to a business decision. HCQ has also been associated with irreversible retinal toxicity with long-term use [265]. However, there are four actively recruiting clinical trials that aim to explore the benefits of HCQ in combination with other therapies in breast cancer, as stated in Table 1.

4.2. Chloroquine (CQ)

Compared with HCQ, CQ is less popular in the clinical setting due to ocular toxicity [266]. However, a clinical study explored the efficacy of oral CQ in breast ductal carcinoma in situ (DCIS) (NCT01023477). Upon the completion of this trial, many of the patients had a reduction in autophagy activity and PCNA proliferation index in DCIS lesions [267], implying a decreased rate of invasion and metastasis [268]. Among 12 patients in this clinical trial, 7 of them indicated a shrinkage of the DCIS lesion. Compared with untreated controls, there was an increase in tumor-infiltrating macrophages in the DCIS ducts [267]. Collectively, these results suggested that oral chloroquine led to a significant reduction in DCIS progression along with increased migration of immune cells into the duct [267]. On the other hand, the outcome from a recent clinical trial indicated that chloroquine combined with taxane and related chemotherapy resulted in a 45.16% objective response rate (ORR) [269].

4.3. Parthenolide

Parthenolide (PTL) is a sesquiterpene lactone isolated from feverfew [270]. In vitro studies report that PTL hinders proliferation while eliciting apoptosis in breast cancer, cholangiocarcinoma, colorectal cancer, and hepatoma pancreatic cancer [271,272,273]. PTL-induced apoptosis is related to NF-kB inhibition, mitochondrial dysfunction, and an increase in ROS [274,275,276,277,278]. However, treatment with PTL leads to an elevation of autophagic activity in breast cancer cells [279]. Even though PTL increases autophagic activity, autophagy inhibition on top of PTL treatment promotes apoptosis, which proves that PTL-induced autophagy is an attempt to overcome the chemical stress brought by PTL [279]. Hence, PTL supplementation along with autophagy inhibition may suppress the progression of breast cancer.

4.4. Honokiol

Honokiol (HNK) is a lignan extracted from Magnolia, which is a common eastern herbal medicine with a long history [280,281]. It has been reported that HNK treatment leads to significant growth reduction and apoptosis in several subtypes of breast cancer [282,283]. Similar to PTL, HNK is a substance extracted from plants; it also induces autophagy in breast cancer as an attempt to subside the stress brought by HNK [284]. The combined treatment with autophagy inhibitors and HNK results in a substantial reduction in breast tumorigenesis and lung metastasis [284].

4.5. Withaferin A

Ashwagandha is one of the most wildly used ancient medicine in India [285]. Withaferin A (WFA) is one of the bioactive extracts derived from Ashwagandha, and its anti-neoplastic tendency has been well documented [286,287]. WFA can stimulate autophagy [288], possibly owing to its activation effect on the AMPK pathway [289]. Even though the treatment of WFA activates [286] autophagy in breast cancer cells, it also leads to hyperacetylation of tubulin, thus hindering the fusion of lysosomes and autophagosomes, which results in the build-up of autophagosomes that confer cellular toxicity [288,290]. In addition, treatment with WFA also interferes with Unfolded Protein System (UPS)-mediated proteolysis, which potentiates proteotoxicity and results in cell death [288]. Consistently, our group has shown that WFA is a lysosomal inhibitor [289]. Further, as a lysosomal inhibitor, WFA hinders proteolytic lysosomal activity, causing a deficiency of substrates to feed the citric acid cycle, a vital process of aerobic respiration, resulting in growth inhibition in breast cancer cells [199,289]. These studies imply that WFA inhibits the growth of breast cancer cells and manipulates various aspects of the autophagic pathway. Additional preclinical and clinical studies are required to assess the potential of WFA as a modulator of autophagy that can be beneficial for improving the efficacy of standard therapies for breast cancer.

4.6. Toosendanin

Melia toosendan Sieb et Zucc was used as an anthelmintic vermifuge in ancient China [291,292]. The antitumor effect of toosendanin (TSN), a triterpenoid derivative from the bark of Melia roosendan Sieb et Zucc [291,292], has been investigated in recent years. In breast cancer, TSN has the leverage to counter adriamycin resistance in vitro and in vivo via inhibition of the PI2K/Akt signaling pathway and downregulation of ABCB1 [293]. Recently, a study reported that TSN can act as a potent autophagy inhibitor to reverse irinotecan resistance [294]. Unlike widely used autophagy inhibitors such as CQ and HCQ, which disrupt the fusion of lysosome and autophagosome, TSN impairs autophagy by elevating the pH of lysosomes in TNBC cells. Importantly, the effective dose of CQ in vitro is 30 times more than TSN [294], indicating that TSN is a more potent autophagy inhibitor than CQ. To determine whether TSN is a good additive to current treatments of breast cancer, additional preclinical research is needed.

5. Conclusions and Future Perspectives

Based on the functional outcomes, autophagy can be cytoprotective (enhances cell survival), cytostatic (leads to growth arrest), cytotoxic (causes cell death), or nonprotective (no impact on cell death or cell survival). However, in terms of interfering with drug efficacy, it is mostly considered cytoprotective. In a bidirectional manner, several drugs modulate the autophagic process and various autophagy-associated proteins directly or indirectly modulate key signaling pathways, miRs and lncRNAs to impact therapeutic efficacy. In last few years, several new targeted therapies or new drug combinations have been put forth especially for metastatic breast cancer, and it is intriguing to note that cancer cells elicit the autophagic response to evade therapy, promote stemness, and induce dormancy in response to many therapeutic strategies. Although, there is ample preclinical evidence to support the strategies combining autophagy inhibitors with anti-cancer drugs and underlying mechanisms have also been explored, clinical studies are few and far between. A major caveat with most autophagy inhibitors is the lack of selectivity towards cancer cells, which ensues overall toxicity since autophagy is important for maintaining homeostasis. Several endeavors are currently underway to develop novel compounds to selectively target autophagy in cancer cells. Focusing on ATG12–ATG3 interaction, a high throughput screen identified a lead compound that selectively inhibits growth in autophagy-addicted cancer cells [295]. A hydrocarbon-stapled peptide directly targeting ATG5–ATG16L1 interaction presents another new strategy for autophagy inhibition [296]. Although discovering and developing new compounds for efficient and selective autophagy inhibition in cancer cells is important, selective delivery of existing autophagy inhibitors is also important. Nanophotosensitizers loaded with chloroquine show increased tumor accumulation and can be sensitized with photodynamic therapy to yield autophagy inhibition [297]. In an interesting strategy, a fasting mimicking diet (FMD) is used to enhance autophagy in cancer cells that are selectively targeted with insulin-like growth factor 2 receptor (IGF2R)-targeted liposomes containing hydroxychloroquine (iLipo-H) [298]. Additional well-designed pre-clinical and clinical studies are required to examine the benefits of adding autophagy inhibitors in combination regimens. Moreover, new autophagy inhibitors with lower toxicity, a better safety profile, and higher efficacy need to be developed to propel this field forward and reap the benefits of preclinical findings.

Author Contributions

Q.W.: Investigation, Data Curation, Writing—Original Draft Preparation; D.S.: Investigation, Conceptualization, Analysis, Supervision, Writing—Reviewing and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by NCI NIH R01CA204555, Breast Cancer Research Foundation (BCRF) 90047965, CDMRP DOD BCRP (BC191572, BC210668), and The Fetting Fund to DS.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Yang, Z.; Klionsky, D.J. Mammalian autophagy: Core molecular machinery and signaling regulation. Curr. Opin. Cell Biol. 2010, 22, 124–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Bednarczyk, M.; Zmarzły, N.; Grabarek, B.; Mazurek, U.; Muc-Wierzgoń, M. Genes involved in the regulation of different types of autophagy and their participation in cancer pathogenesis. Oncotarget 2018, 9, 34413–34428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Deretic, V. Autophagy in inflammation, infection, and immunometabolism. Immunity 2021, 54, 437–453. [Google Scholar] [CrossRef]
  4. Klionsky, D.J.; Petroni, G.; Amaravadi, R.K.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cadwell, K.; Cecconi, F.; Choi, A.M.K.; et al. Autophagy in major human diseases. EMBO J. 2021, 40, e108863. [Google Scholar] [CrossRef]
  5. Guan, J.-L.; Simon, A.K.; Prescott, M.; Menendez, J.A.; Liu, F.; Wang, F.; Wang, C.; Wolvetang, E.; Vazquez-Martin, A.; Zhang, J. Autophagy in stem cells. Autophagy 2013, 9, 830–849. [Google Scholar] [CrossRef]
  6. Chang, N.C. Autophagy and Stem Cells: Self-Eating for Self-Renewal. Front. Cell Dev. Biol. 2020, 8, 138. [Google Scholar] [CrossRef] [Green Version]
  7. Galluzzi, L.; Green, D.R. Autophagy-Independent Functions of the Autophagy Machinery. Cell 2019, 177, 1682–1699. [Google Scholar] [CrossRef]
  8. Jia, S.-N.; Lin, C.; Chen, D.-F.; Li, A.-Q.; Dai, L.; Zhang, L.; Zhao, L.-L.; Yang, J.-S.; Yang, F.; Yang, W.-J. The Transcription Factor p8 Regulates Autophagy in Response to Palmitic Acid Stress via a Mammalian Target of Rapamycin (mTOR)-independent Signaling Pathway. J. Biol. Chem. 2016, 291, 4462–4472. [Google Scholar] [CrossRef] [Green Version]
  9. Parzych, K.R.; Klionsky, D.J. An Overview of Autophagy: Morphology, Mechanism, and Regulation. Antioxid. Redox Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef] [Green Version]
  10. Mancias, J.D.; Kimmelman, A.C. Mechanisms of Selective Autophagy in Normal Physiology and Cancer. J. Mol. Biol. 2016, 428, 1659–1680. [Google Scholar] [CrossRef] [Green Version]
  11. Conway, O.; Akpinar, H.A.; Rogov, V.V.; Kirkin, V. Selective Autophagy Receptors in Neuronal Health and Disease. J. Mol. Biol. 2020, 432, 2483–2509. [Google Scholar] [CrossRef] [PubMed]
  12. Wirawan, E.; Vanden Berghe, T.; Lippens, S.; Agostinis, P.; Vandenabeele, P. Autophagy: For better or for worse. Cell Res. 2012, 22, 43–61. [Google Scholar] [CrossRef] [Green Version]
  13. Mijaljica, D.; Prescott, M.; Devenish, R.J. Microautophagy in mammalian cells: Revisiting a 40-year-old conundrum. Autophagy 2011, 7, 673–682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Massey, A.; Kiffin, R.; Cuervo, A.M. Pathophysiology of chaperone-mediated autophagy. Int. J. Biochem. Cell Biol. 2004, 36, 2420–2434. [Google Scholar] [CrossRef] [PubMed]
  15. Yorimitsu, T.; Klionsky, D.J. Autophagy: Molecular machinery for self-eating. Cell Death Differ. 2005, 12 (Suppl. S2), 1542–1552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Liu, W.J.; Ye, L.; Huang, W.F.; Guo, L.J.; Xu, Z.G.; Wu, H.L.; Yang, C.; Liu, H.F. p62 links the autophagy pathway and the ubiqutin–proteasome system upon ubiquitinated protein degradation. Cell. Mol. Biol. Lett. 2016, 21, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Faruk, M.O.; Ichimura, Y.; Komatsu, M. Selective autophagy. Cancer Sci. 2021, 112, 3972–3978. [Google Scholar] [CrossRef]
  18. Münch, C.; Dikic, I. Hitchhiking on selective autophagy. Nat. Cell Biol. 2018, 20, 122–124. [Google Scholar] [CrossRef]
  19. Xiong, H.; Shen, J.; Chen, Z.; Yang, J.; Xie, B.; Jia, Y.; Jayasinghe, U.; Wang, J.; Zhao, W.; Xie, S.; et al. H19/let-7/Lin28 ceRNA network mediates autophagy inhibiting epithelial-mesenchymal transition in breast cancer. Int. J. Oncol. 2020, 56, 794–806. [Google Scholar] [CrossRef]
  20. Li, Y.; Jiang, B.; Zhu, H.; Qu, X.; Zhao, L.; Tan, Y.; Jiang, Y.; Liao, M.; Wu, X. Inhibition of long non-coding RNA ROR reverses resistance to Tamoxifen by inducing autophagy in breast cancer. Tumor Biol. 2017, 39, 1010428317705790. [Google Scholar] [CrossRef] [Green Version]
  21. Qian, X.; Qu, H.; Zhang, F.; Peng, S.; Dou, D.; Yang, Y.; Ding, Y.; Xie, M.; Dong, H.; Liao, Y.; et al. Exosomal long noncoding RNA AGAP2-AS1 regulates trastuzumab resistance via inducing autophagy in breast cancer. Am. J. Cancer Res. 2021, 11, 1962–1981. [Google Scholar] [PubMed]
  22. Han, X.; Mo, J.; Yang, Y.; Wang, Y.; Lu, H. Crucial Roles of LncRNAs-Mediated Autophagy in Breast Cancer. Int. J. Med. Sci. 2022, 19, 1082–1092. [Google Scholar] [CrossRef] [PubMed]
  23. Li, X.; Jin, F.; Li, Y. A novel autophagy-related lncRNA prognostic risk model for breast cancer. J. Cell. Mol. Med. 2021, 25, 4–14. [Google Scholar] [CrossRef] [PubMed]
  24. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  25. Hewitt, G.; Korolchuk, V.I. Repair, Reuse, Recycle: The Expanding Role of Autophagy in Genome Maintenance. Trends Cell Biol. 2017, 27, 340–351. [Google Scholar] [CrossRef] [Green Version]
  26. Galluzzi, L.; Pietrocola, F.; Bravo-San Pedro, J.M.; Amaravadi, R.K.; Baehrecke, E.H.; Cecconi, F.; Codogno, P.; Debnath, J.; Gewirtz, D.A.; Karantza, V.; et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015, 34, 856–880. [Google Scholar] [CrossRef] [PubMed]
  27. Rybstein, M.D.; Bravo-San Pedro, J.M.; Kroemer, G.; Galluzzi, L. The autophagic network and cancer. Nat. Cell Biol. 2018, 20, 243–251. [Google Scholar] [CrossRef]
  28. Young, A.R.; Narita, M.; Ferreira, M.; Kirschner, K.; Sadaie, M.; Darot, J.F.; Tavaré, S.; Arakawa, S.; Shimizu, S.; Watt, F.M.; et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 2009, 23, 798–803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Santana-Codina, N.; Mancias, J.D.; Kimmelman, A.C. The Role of Autophagy in Cancer. Annu. Rev. Cancer Biol. 2017, 1, 19–39. [Google Scholar] [CrossRef]
  30. Maslov, A.Y.; Vijg, J. Genome instability, cancer and aging. Biochim. Biophys. Acta 2009, 1790, 963–969. [Google Scholar] [CrossRef] [Green Version]
  31. Garufi, A.; Pucci, D.; D’Orazi, V.; Cirone, M.; Bossi, G.; Avantaggiati, M.L.; D’Orazi, G. Degradation of mutant p53H175 protein by Zn(II) through autophagy. Cell Death Dis. 2014, 5, e1271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Choundhury, S.; Kolukula, V.K.; Preet, A.; Albanese, C.; Avantaggiati, M.L. Dissecting the pathways that destabilize mutant p53: The proteasome or autophagy? Cell Cycle 2013, 12, 1022–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rodriguez, O.C.; Choudhury, S.; Kolukula, V.; Vietsch, E.E.; Catania, J.; Preet, A.; Reynoso, K.; Bargonetti, J.; Wellstein, A.; Albanese, C.; et al. Dietary downregulation of mutant p53 levels via glucose restriction. Cell Cycle 2012, 11, 4436–4446. [Google Scholar] [CrossRef] [Green Version]
  34. Guo, H.; Chitiprolu, M.; Gagnon, D.; Meng, L.; Perez-Iratxeta, C.; Lagace, D.; Gibbings, D. Autophagy supports genomic stability by degrading retrotransposon RNA. Nat. Commun. 2014, 5, 5276. [Google Scholar] [CrossRef] [Green Version]
  35. Rello-Varona, S.; Lissa, D.; Shen, S.; Niso-Santano, M.; Senovilla, L.; Mariño, G.; Vitale, I.; Jemaa, M.; Harper, F.; Pierron, G.; et al. Autophagic removal of micronuclei. Cell Cycle 2012, 11, 170–176. [Google Scholar] [CrossRef] [PubMed]
  36. Mathew, R.; Karp, C.M.; Beaudoin, B.; Vuong, N.; Chen, G.; Chen, H.-Y.; Bray, K.; Reddy, A.; Bhanot, G.; Gelinas, C.; et al. Autophagy Suppresses Tumorigenesis through Elimination of p62. Cell 2009, 137, 1062–1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Komatsu, M.; Waguri, S.; Koike, M.; Sou, Y.-S.; Ueno, T.; Hara, T.; Mizushima, N.; Iwata, J.-I.; Ezaki, J.; Murata, S.; et al. Homeostatic Levels of p62 Control Cytoplasmic Inclusion Body Formation in Autophagy-Deficient Mice. Cell 2007, 131, 1149–1163. [Google Scholar] [CrossRef] [Green Version]
  38. Takahashi, Y.; Hori, T.; Cooper, T.K.; Liao, J.; Desai, N.; Serfass, J.M.; Young, M.M.; Park, S.; Izu, Y.; Wang, H.-G. Bif-1 haploinsufficiency promotes chromosomal instability and accelerates Myc-driven lymphomagenesis via suppression of mitophagy. Blood 2013, 121, 1622–1632. [Google Scholar] [CrossRef] [Green Version]
  39. Green, D.R.; Galluzzi, L.; Kroemer, G. Mitochondria and the Autophagy-Inflammation-Cell Death Axis in Organismal Aging. Science 2011, 333, 1109–1112. [Google Scholar] [CrossRef] [Green Version]
  40. Greim, H.; Kaden, D.A.; Larson, R.A.; Palermo, C.M.; Rice, J.M.; Ross, D.; Snyder, R. The bone marrow niche, stem cells, and leukemia: Impact of drugs, chemicals, and the environment. Ann. N. Y. Acad. Sci. 2014, 1310, 7–31. [Google Scholar] [CrossRef] [Green Version]
  41. Deretic, V.; Saitoh, T.; Akira, S. Autophagy in infection, inflammation and immunity. Nat. Rev. Immunol. 2013, 13, 722–737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Ma, Y.; Galluzzi, L.; Zitvogel, L.; Kroemer, G. Autophagy and Cellular Immune Responses. Immunity 2013, 39, 211–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Niklaus, N.J.; Tokarchuk, I.; Zbinden, M.; Schläfli, A.M.; Maycotte, P.; Tschan, M.P. The Multifaceted Functions of Autophagy in Breast Cancer Development and Treatment. Cells 2021, 10, 1447. [Google Scholar] [CrossRef] [PubMed]
  44. Tavera-Mendoza, L.E.; Westerling, T.; Libby, E.; Marusyk, A.; Cato, L.; Cassani, R.; Cameron, L.A.; Ficarro, S.B.; Marto, J.A.; Klawitter, J.; et al. Vitamin D receptor regulates autophagy in the normal mammary gland and in luminal breast cancer cells. Proc. Natl. Acad. Sci. USA 2017, 114, E2186–E2194. [Google Scholar] [CrossRef] [Green Version]
  45. He, C.; Klionsky, D.J. Regulation mechanisms and signaling pathways of autophagy. Annu. Rev. Genet. 2009, 43, 67–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Mizushima, N.; Komatsu, M. Autophagy: Renovation of Cells and Tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Liang, X.H.; Jackson, S.; Seaman, M.; Brown, K.; Kempkes, B.; Hibshoosh, H.; Levine, B. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999, 402, 672–676. [Google Scholar] [CrossRef]
  48. Miracco, C.; Cosci, E.; Oliveri, G.; Luzi, P.; Pacenti, L.; Monciatti, I.; Mannucci, S.; De Nisi, M.C.; Toscano, M.; Malagnino, V.; et al. Protein and mRNA expression of autophagy gene Beclin 1 in human brain tumours. Int. J. Oncol. 2007, 30, 429–436. [Google Scholar]
  49. Vega-Rubin-de-Celis, S.; Zou, Z.; Fernandez, A.F.; Ci, B.; Kim, M.; Xiao, G.; Xie, Y.; Levine, B. Increased autophagy blocks HER2-mediated breast tumorigenesis. Proc. Natl. Acad. Sci. USA 2018, 115, 4176–4181. [Google Scholar] [CrossRef] [Green Version]
  50. Cicchini, M.; Chakrabarti, R.; Kongara, S.; Price, S.; Nahar, R.; Lozy, F.; Zhong, H.; Vazquez, A.; Kang, Y.; Karantza, V. Autophagy regulator BECN1 suppresses mammary tumorigenesis driven by WNT1 activation and following parity. Autophagy 2014, 10, 2036–2052. [Google Scholar] [CrossRef] [Green Version]
  51. Hatzis, C.; Pusztai, L.; Valero, V.; Booser, D.J.; Esserman, L.; Lluch, A.; Vidaurre, T.; Holmes, F.; Souchon, E.; Wang, H.; et al. A Genomic Predictor of Response and Survival Following Taxane-Anthracycline Chemotherapy for Invasive Breast Cancer. JAMA 2011, 305, 1873–1881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Giuli, M.V.; Giuliani, E.; Screpanti, I.; Bellavia, D.; Checquolo, S. Notch Signaling Activation as a Hallmark for Triple-Negative Breast Cancer Subtype. J. Oncol. 2019, 2019, 8707053. [Google Scholar] [CrossRef] [PubMed]
  53. Ahn, J.-S.; Ann, E.-J.; Kim, M.-Y.; Yoon, J.-H.; Lee, H.-J.; Jo, E.-H.; Lee, K.; Lee, J.S.; Park, H.-S. Autophagy negatively regulates tumor cell proliferation through phosphorylation dependent degradation of the Notch1 intracellular domain. Oncotarget 2016, 7, 79047–79063. [Google Scholar] [CrossRef] [Green Version]
  54. Li, M.; Liu, J.; Li, S.; Feng, Y.; Yi, F.; Wang, L.; Wei, S.; Cao, L. Autophagy-related 7 modulates tumor progression in triple-negative breast cancer. Lab. Investig. 2019, 99, 1266–1274. [Google Scholar] [CrossRef] [PubMed]
  55. Lock, R.; Roy, S.; Kenific, C.M.; Su, J.S.; Salas, E.; Ronen, S.M.; Debnath, J. Autophagy facilitates glycolysis during Ras-mediated oncogenic transformation. Mol. Biol. Cell 2011, 22, 165–178. [Google Scholar] [CrossRef] [PubMed]
  56. Maycotte, P.; Gearheart, C.M.; Barnard, R.; Aryal, S.; Mulcahy Levy, J.M.; Fosmire, S.P.; Hansen, R.J.; Morgan, M.J.; Porter, C.C.; Gustafson, D.L.; et al. STAT3-Mediated Autophagy Dependence Identifies Subtypes of Breast Cancer Where Autophagy Inhibition Can Be Efficacious. Cancer Res. 2014, 74, 2579–2590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Hornsveld, M.; Smits, L.M.M.; Meerlo, M.; van Amersfoort, M.; Groot Koerkamp, M.J.A.; van Leenen, D.; Kloet, D.E.A.; Holstege, F.C.P.; Derksen, P.W.B.; Burgering, B.M.T.; et al. FOXO Transcription Factors Both Suppress and Support Breast Cancer Progression. Cancer Res. 2018, 78, 2356–2369. [Google Scholar] [CrossRef] [Green Version]
  58. Wu, Y.; Elshimali, Y.; Sarkissyan, M.; Mohamed, H.; Clayton, S.; Vadgama, J.V. Expression of FOXO1 is associated with GATA3 and Annexin-1 and predicts disease-free survival in breast cancer. Am. J. Cancer Res. 2012, 2, 104–115. [Google Scholar] [CrossRef]
  59. Habashy, H.O.; Rakha, E.A.; Aleskandarany, M.; Ahmed, M.A.; Green, A.; Ellis, I.O.; Powe, D.G. FOXO3a nuclear localisation is associated with good prognosis in luminal-like breast cancer. Breast Cancer Res. Treat. 2011, 129, 11–21. [Google Scholar] [CrossRef]
  60. Smit, L.; Berns, K.; Spence, K.; Ryder, W.D.; Zeps, N.; Madiredjo, M.; Beijersbergen, R.; Bernards, R.; Clarke, R.B. An integrated genomic approach identifies that the PI3K/AKT/FOXO pathway is involved in breast cancer tumor initiation. Oncotarget 2016, 7, 2596–2610. [Google Scholar] [CrossRef] [Green Version]
  61. Schäffner, I.; Minakaki, G.; Khan, M.A.; Balta, E.-A.; Schlötzer-Schrehardt, U.; Schwarz, T.J.; Beckervordersandforth, R.; Winner, B.; Webb, A.E.; DePinho, R.A.; et al. FoxO Function Is Essential for Maintenance of Autophagic Flux and Neuronal Morphogenesis in Adult Neurogenesis. Neuron 2018, 99, 1188–1203.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Audesse, A.J.; Dhakal, S.; Hassell, L.-A.; Gardell, Z.; Nemtsova, Y.; Webb, A.E. FOXO3 directly regulates an autophagy network to functionally regulate proteostasis in adult neural stem cells. PLoS Genet. 2019, 15, e1008097. [Google Scholar] [CrossRef] [PubMed]
  63. Sharif, T.; Martell, E.; Dai, C.; Kennedy, B.E.; Murphy, P.; Clements, D.R.; Kim, Y.; Lee, P.W.K.; Gujar, S.A. Autophagic homeostasis is required for the pluripotency of cancer stem cells. Autophagy 2017, 13, 264–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Boya, P.; Codogno, P.; Rodriguez-Muela, N. Autophagy in stem cells: Repair, remodelling and metabolic reprogramming. Development 2018, 145, dev146506. [Google Scholar] [CrossRef] [Green Version]
  65. Azamjah, N.; Soltan-Zadeh, Y.; Zayeri, F. Global Trend of Breast Cancer Mortality Rate: A 25-Year Study. Asian Pac. J. Cancer Prev. 2019, 20, 2015–2020. [Google Scholar] [CrossRef]
  66. Vera-Ramirez, L.; Vodnala, S.K.; Nini, R.; Hunter, K.W.; Green, J.E. Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence. Nat. Commun. 2018, 9, 1944. [Google Scholar] [CrossRef] [Green Version]
  67. Maishman, T.; Cutress, R.I.; Hernandez, A.; Gerty, S.; Copson, E.R.; Durcan, L.; Eccles, D.M. Local Recurrence and Breast Oncological Surgery in Young Women With Breast Cancer: The POSH Observational Cohort Study. Ann. Surg. 2017, 266, 165–172. [Google Scholar] [CrossRef] [Green Version]
  68. Clarke, S.J.; Rivory, L. Clinical Pharmacokinetics of Docetaxel. Clin. Pharmacokinet. 1999, 36, 99–114. [Google Scholar] [CrossRef]
  69. Trudeau, M.; Charbonneau, F.; Gelmon, K.; Laing, K.; Latreille, J.; Mackey, J.; McLeod, D.; Pritchard, K.; Provencher, L.; Verma, S. Selection of adjuvant chemotherapy for treatment of node-positive breast cancer. Lancet Oncol. 2005, 6, 886–898. [Google Scholar] [CrossRef]
  70. Jamieson, E.R.; Lippard, S.J. Structure, recognition, and processing of cisplatin—DNA adducts. Chem. Rev. 1999, 99, 2467–2498. [Google Scholar] [CrossRef]
  71. Thomas, S.S.; Clements, P.J.; Furst, D.E. Immunosuppressives: Cyclosporine, cyclophosphamide, azathioprine, mycophenolate mofetil. In Rheumatology; Elsevier: Amsterdam, The Netherlands, 2015; pp. 459–467. [Google Scholar]
  72. Wen, N.; Lv, Q.; Du, Z.-G. MicroRNAs involved in drug resistance of breast cancer by regulating autophagy. J. Zhejiang Univ. Sci. B 2020, 21, 690–702. [Google Scholar] [CrossRef] [PubMed]
  73. Rossari, F.; Zucchinetti, C.; Buda, G.; Orciuolo, E. Tumor dormancy as an alternative step in the development of chemoresistance and metastasis-clinical implications. Cell. Oncol. 2020, 43, 155–176. [Google Scholar] [CrossRef] [PubMed]
  74. Bushnell, G.G.; Deshmukh, A.P.; den Hollander, P.; Luo, M.; Soundararajan, R.; Jia, D.; Levine, H.; Mani, S.A.; Wicha, M.S. Breast cancer dormancy: Need for clinically relevant models to address current gaps in knowledge. NPJ Breast Cancer 2021, 7, 66. [Google Scholar] [CrossRef]
  75. Sosa, M.S.; Bragado, P.; Aguirre-Ghiso, J.A. Mechanisms of disseminated cancer cell dormancy: An awakening field. Nat. Rev. Cancer 2014, 14, 611–622. [Google Scholar] [CrossRef] [PubMed]
  76. Katheder, N.S.; Khezri, R.; O’Farrell, F.; Schultz, S.W.; Jain, A.; Rahman, M.M.; Schink, K.O.; Theodossiou, T.A.; Johansen, T.; Juhász, G.; et al. Microenvironmental autophagy promotes tumour growth. Nature 2017, 541, 417–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Amaravadi, R.; Kimmelman, A.C.; White, E. Recent insights into the function of autophagy in cancer. Genes Dev. 2016, 30, 1913–1930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Smith, A.G.; MacLeod, K.F. Autophagy, cancer stem cells and drug resistance. J. Pathol. 2019, 247, 708–718. [Google Scholar] [CrossRef] [Green Version]
  79. La Belle Flynn, A.; Calhoun, B.C.; Sharma, A.; Chang, J.C.; Almasan, A.; Schiemann, W.P. Autophagy inhibition elicits emergence from metastatic dormancy by inducing and stabilizing Pfkfb3 expression. Nat. Commun. 2019, 10, 3668. [Google Scholar] [CrossRef] [Green Version]
  80. Gwinn, D.M.; Shackelford, D.B.; Egan, D.F.; Mihaylova, M.M.; Mery, A.; Vasquez, D.S.; Turk, B.E.; Shaw, R.J. AMPK Phosphorylation of Raptor Mediates a Metabolic Checkpoint. Mol. Cell 2008, 30, 214–226. [Google Scholar] [CrossRef] [Green Version]
  81. Settembre, C.; Fraldi, A.; Medina, D.L.; Ballabio, A. Signals from the lysosome: A control centre for cellular clearance and energy metabolism. Nat. Rev. Mol. Cell Biol. 2013, 14, 283–296. [Google Scholar] [CrossRef] [Green Version]
  82. Kim, Y.C.; Guan, K.-L. mTOR: A pharmacologic target for autophagy regulation. J. Clin. Investig. 2015, 125, 25–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Feldman, M.E.; Apsel, B.; Uotila, A.; Loewith, R.; Knight, Z.A.; Ruggero, D.; Shokat, K.M. Active-Site Inhibitors of mTOR Target Rapamycin-Resistant Outputs of mTORC1 and mTORC2. PLoS Biol. 2009, 7, e38. [Google Scholar] [CrossRef]
  84. Marinković, M.; Šprung, M.; Buljubasic, M.; Novak, I. Autophagy Modulation in Cancer: Current Knowledge on Action and Therapy. Oxidative Med. Cell. Longev. 2018, 2018, 8023821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Egan, D.F.; Shackelford, D.B.; Mihaylova, M.M.; Gelino, S.; Kohnz, R.A.; Mair, W.; Vasquez, D.S.; Joshi, A.; Gwinn, D.M.; Taylor, R.; et al. Phosphorylation of ULK1 (hATG1) by AMP-Activated Protein Kinase Connects Energy Sensing to Mitophagy. Science 2011, 331, 456–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lu, Z.; Luo, R.Z.; Lu, Y.; Zhang, X.; Yu, Q.; Khare, S.; Kondo, S.; Kondo, Y.; Yu, Y.; Mills, G.B.; et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J. Clin. Investig. 2008, 118, 3917–3929. [Google Scholar] [CrossRef] [Green Version]
  87. Zou, C.-F.; Jia, L.; Jin, H.; Yao, M.; Zhao, N.; Huan, J.; Lu, Z.; Bast, R.C., Jr.; Feng, Y.; Yu, Y. Re-expression of ARHI (DIRAS3) induces autophagy in breast cancer cells and enhances the inhibitory effect of paclitaxel. BMC Cancer 2011, 11, 22. [Google Scholar] [CrossRef] [Green Version]
  88. Bai, X.; Ni, J.; Beretov, J.; Graham, P.; Li, Y. Triple-negative breast cancer therapeutic resistance: Where is the Achilles’ heel? Cancer Lett. 2021, 497, 100–111. [Google Scholar] [CrossRef] [PubMed]
  89. Bai, X.; Ni, J.; Beretov, J.; Graham, P.; Li, Y. Cancer stem cell in breast cancer therapeutic resistance. Cancer Treat. Rev. 2018, 69, 152–163. [Google Scholar] [CrossRef]
  90. Lee, K.-M.; Giltnane, J.M.; Balko, J.M.; Schwarz, L.J.; Guerrero-Zotano, A.L.; Hutchinson, K.E.; Nixon, M.J.; Estrada, M.V.; Sánchez, V.; Sanders, M.E.; et al. MYC and MCL1 Cooperatively Promote Chemotherapy-Resistant Breast Cancer Stem Cells via Regulation of Mitochondrial Oxidative Phosphorylation. Cell Metab. 2017, 26, 633–647.e7. [Google Scholar] [CrossRef] [Green Version]
  91. Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 2010, 29, 4741–4751. [Google Scholar] [CrossRef] [Green Version]
  92. Nazio, F.; Bordi, M.; Cianfanelli, V.; Locatelli, F.; Cecconi, F. Autophagy and cancer stem cells: Molecular mechanisms and therapeutic applications. Cell Death Differ. 2019, 26, 690–702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bousquet, G.; El Bouchtaoui, M.; Sophie, T.; Leboeuf, C.; de Bazelaire, C.; Ratajczak, P.; Giacchetti, S.; de Roquancourt, A.; Bertheau, P.; Verneuil, L.; et al. Targeting autophagic cancer stem-cells to reverse chemoresistance in human triple negative breast cancer. Oncotarget 2017, 8, 35205–35221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Mathew, R.; Karantza-Wadsworth, V.; White, E. Role of autophagy in cancer. Nat. Rev. Cancer 2007, 7, 961–967. [Google Scholar] [CrossRef]
  95. Thomas, M.; Davis, T.; Loos, B.; Sishi, B.; Huisamen, B.; Strijdom, H.; Engelbrecht, A.-M. Autophagy is essential for the maintenance of amino acids and ATP levels during acute amino acid starvation in MDAMB231 cells. Cell Biochem. Funct. 2018, 36, 65–79. [Google Scholar] [CrossRef] [PubMed]
  96. Martin, S.; Dudek-Peric, A.M.; Garg, A.D.; Roose, H.; Demirsoy, S.; Van Eygen, S.; Mertens, F.; Vangheluwe, P.; Vankelecom, H.; Agostinis, P. An autophagy-driven pathway of ATP secretion supports the aggressive phenotype of BRAF(V600E) inhibitor-resistant metastatic melanoma cells. Autophagy 2017, 13, 1512–1527. [Google Scholar] [CrossRef] [Green Version]
  97. Schito, L.; Semenza, G.L. Hypoxia-Inducible Factors: Master Regulators of Cancer Progression. Trends Cancer 2016, 2, 758–770. [Google Scholar] [CrossRef] [Green Version]
  98. Michiels, C. Physiological and Pathological Responses to Hypoxia. Am. J. Pathol. 2004, 164, 1875–1882. [Google Scholar] [CrossRef] [Green Version]
  99. Liu, L.; Salnikov, A.V.; Bauer, N.; Aleksandrowicz, E.; Labsch, S.; Nwaeburu, C.; Mattern, J.; Gladkich, J.; Schemmer, P.; Werner, J.; et al. Triptolide reverses hypoxia-induced epithelial–mesenchymal transition and stem-like features in pancreatic cancer by NF-κB downregulation. Int. J. Cancer 2014, 134, 2489–2503. [Google Scholar] [CrossRef] [Green Version]
  100. Samanta, D.; Gilkes, D.M.; Chaturvedi, P.; Xiang, L.; Semenza, G.L. Hypoxia-inducible factors are required for chemotherapy resistance of breast cancer stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, E5429–E5438. [Google Scholar] [CrossRef] [Green Version]
  101. Daskalaki, I.; Gkikas, I.; Tavernarakis, N. Hypoxia and Selective Autophagy in Cancer Development and Therapy. Front. Cell Dev. Biol. 2018, 6, 104. [Google Scholar] [CrossRef] [Green Version]
  102. Yeo, S.K.; Wen, J.; Chen, S.; Guan, J.-L. Autophagy Differentially Regulates Distinct Breast Cancer Stem-like Cells in Murine Models via EGFR/Stat3 and Tgfβ/Smad Signaling. Cancer Res. 2016, 76, 3397–3410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Golden, E.B.; Cho, H.-Y.; Jahanian, A.; Hofman, F.M.; Louie, S.G.; Schönthal, A.H.; Chen, T.C. Chloroquine enhances temozolomide cytotoxicity in malignant gliomas by blocking autophagy. Neurosurg. Focus 2014, 37, E12. [Google Scholar] [CrossRef] [Green Version]
  104. Chaterjee, M.; Van Golen, K.L. Breast Cancer Stem Cells Survive Periods of Farnesyl-Transferase Inhibitor-Induced Dormancy by Undergoing Autophagy. Bone Marrow Res. 2011, 2011, 362938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Zhang, L.; Xu, L.; Zhang, F.; Vlashi, E. Doxycycline inhibits the cancer stem cell phenotype and epithelial-to-mesenchymal transition in breast cancer. Cell Cycle 2017, 16, 737–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Ibragimova, M.; Tsyganov, M.; Litviakov, N. Tumour Stem Cells in Breast Cancer. Int. J. Mol. Sci. 2022, 23, 5058. [Google Scholar] [CrossRef] [PubMed]
  107. Vazquez-Martin, A.; Oliveras-Ferraros, C.; Menendez, J.A. Autophagy Facilitates the Development of Breast Cancer Resistance to the Anti-HER2 Monoclonal Antibody Trastuzumab. PLoS ONE 2009, 4, e6251. [Google Scholar] [CrossRef] [Green Version]
  108. Sun, W.-L.; Chen, J.; Wang, Y.-P.; Zheng, H. Autophagy protects breast cancer cells from epirubicin-induced apoptosis and facilitates epirubicin-resistance development. Autophagy 2011, 7, 1035–1044. [Google Scholar] [CrossRef] [Green Version]
  109. Kim, H.S.; Tian, L.; Jung, M.; Choi, S.K.; Sun, Y.; Kim, H.; Moon, W.K. Downregulation of Choline Kinase-Alpha Enhances Autophagy in Tamoxifen-Resistant Breast Cancer Cells. PLoS ONE 2015, 10, e0141110. [Google Scholar] [CrossRef] [Green Version]
  110. Gu, Y.; Chen, T.; Li, G.; Xu, C.; Xu, Z.; Zhang, J.; He, K.; Zheng, L.; Guan, Z.; Su, X.; et al. Lower Beclin 1 downregulates HER2 expression to enhance tamoxifen sensitivity and predicts a favorable outcome for ER positive breast cancer. Oncotarget 2017, 8, 52156–52177. [Google Scholar] [CrossRef] [Green Version]
  111. Berardi, D.E.; Flumian, C.; Rodriguez, C.E.; Bessone, M.I.; Cirigliano, S.M.; Joffé, E.D.; Fiszman, G.L.; Urtreger, A.J.; Todaro, L.B. PKCδ Inhibition Impairs Mammary Cancer Proliferative Capacity But Selects Cancer Stem Cells, Involving Autophagy. J. Cell. Biochem. 2016, 117, 730–740. [Google Scholar] [CrossRef]
  112. Xiang, S.; Dauchy, R.T.; Hoffman, A.E.; Pointer, D.; Frasch, T.; Blask, D.E.; Hill, S.M. Epigenetic inhibition of the tumor suppressor ARHI by light at night-induced circadian melatonin disruption mediates STAT3-driven paclitaxel resistance in breast cancer. J. Pineal Res. 2019, 67, e12586. [Google Scholar] [CrossRef] [PubMed]
  113. Hill, S.M.; Blask, D.E.; Xiang, S.; Yuan, L.; Mao, L.; Dauchy, R.T.; Dauchy, E.M.; Frasch, T.; Duplesis, T. Melatonin and Associated Signaling Pathways that Control Normal Breast Epithelium and Breast Cancer. J. Mammary Gland. Biol. Neoplasia 2011, 16, 235–245. [Google Scholar] [CrossRef] [PubMed]
  114. Sun, R.; Shen, S.; Zhang, Y.J.; Xu, C.-F.; Cao, Z.-T.; Wen, L.-P.; Wang, J. Nanoparticle-facilitated autophagy inhibition promotes the efficacy of chemotherapeutics against breast cancer stem cells. Biomaterials 2016, 103, 44–55. [Google Scholar] [CrossRef]
  115. Liang, D.H.; Choi, D.S.; Ensor, J.E.; Kaipparettu, B.A.; Bass, B.L.; Chang, J.C. The autophagy inhibitor chloroquine targets cancer stem cells in triple negative breast cancer by inducing mitochondrial damage and impairing DNA break repair. Cancer Lett. 2016, 376, 249–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Yue, W.; Hamaï, A.; Tonelli, G.; Bauvy, C.; Nicolas, V.; Tharinger, H.; Codogno, P.; Mehrpour, M. Inhibition of the autophagic flux by salinomycin in breast cancer stem-like/progenitor cells interferes with their maintenance. Autophagy 2013, 9, 714–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Periyasamy-Thandavan, S.; Jackson, W.H.; Samaddar, J.S.; Erickson, B.; Barrett, J.R.; Raney, L.; Gopal, E.; Ganapathy, V.; Hill, W.D.; Bhalla, K.N.; et al. Bortezomib blocks the catabolic process of autophagy via a cathepsin-dependent mechanism, affects endoplasmic reticulum stress, and induces caspase-dependent cell death in antiestrogen–sensitive and resistant ER+ breast cancer cells. Autophagy 2010, 6, 19–35. [Google Scholar] [CrossRef] [Green Version]
  118. Schwartz-Roberts, J.L.; Shajahan, A.N.; Cook, K.L.; Wärri, A.; Abu-Asab, M.; Clarke, R. GX15-070 (Obatoclax) Induces Apoptosis and Inhibits Cathepsin D- and L–Mediated Autophagosomal Lysis in Antiestrogen-Resistant Breast Cancer Cells. Mol. Cancer Ther. 2013, 12, 448–459. [Google Scholar] [CrossRef] [Green Version]
  119. Li, Y.; Zou, L.; Li, Q.; Haibe-Kains, B.; Tian, R.; Li, Y.; Desmedt, C.; Sotiriou, C.; Szallasi, Z.; Iglehart, J.D.; et al. Amplification of LAPTM4B and YWHAZ contributes to chemotherapy resistance and recurrence of breast cancer. Nat. Med. 2010, 16, 214–218. [Google Scholar] [CrossRef] [Green Version]
  120. Li, Y.; Zhang, Q.; Tian, R.; Wang, Q.; Zhao, J.J.; Iglehart, J.D.; Wang, Z.C.; Richardson, A.L. Lysosomal Transmembrane Protein LAPTM4B Promotes Autophagy and Tolerance to Metabolic Stress in Cancer Cells. Cancer Res. 2011, 71, 7481–7489. [Google Scholar] [CrossRef] [Green Version]
  121. Li, Y.; Iglehart, J.D.; Richardson, A.L.; Wang, Z.C. The amplified cancer gene LAPTM4B promotes tumor growth and tolerance to stress through the induction of autophagy. Autophagy 2012, 8, 273–274. [Google Scholar] [CrossRef] [Green Version]
  122. Heng, M.Y.; Detloff, P.J.; Paulson, H.L.; Albin, R.L. Early alterations of autophagy in Huntington disease-like mice. Autophagy 2010, 6, 1206–1208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Decressac, M.; Mattsson, B.; Weikop, P.; Lundblad, M.; Jakobsson, J.; Bjorklund, A. TFEB-mediated autophagy rescues midbrain dopamine neurons from α-synuclein toxicity. Proc. Natl. Acad. Sci. USA 2013, 110, E1817–E1826. [Google Scholar] [CrossRef] [Green Version]
  124. Kimura, T.; Takabatake, Y.; Takahashi, A.; Isaka, Y. Chloroquine in Cancer Therapy: A Double-Edged Sword of Autophagy. Cancer Res. 2013, 73, 3–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Li, Y.-J.; Lei, Y.-H.; Yao, N.; Wang, C.-R.; Hu, N.; Ye, W.-C.; Zhang, D.-M.; Chen, Z.-S. Autophagy and multidrug resistance in cancer. Chin. J. Cancer 2017, 36, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Yeo, B.; Turner, N.C.; Jones, A. An update on the medical management of breast cancer. BMJ 2014, 348, g3608. [Google Scholar] [CrossRef]
  127. Davies, C.; Godwin, J.; Gray, R.; Clarke, M.; Cutter, D.; Darby, S.; McGale, P.; Pan, H.C.; Taylor, C.; Wang, Y.C.; et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: Patient-level meta-analysis of randomised trials. Lancet 2011, 378, 771–784. [Google Scholar] [CrossRef] [Green Version]
  128. Nass, N.; Kalinski, T. Tamoxifen resistance: From cell culture experiments towards novel biomarkers. Pathol. Res. Pract. 2015, 211, 189–197. [Google Scholar] [CrossRef]
  129. Dixon, J.M. Endocrine Resistance in Breast Cancer. New J. Sci. 2014, 2014, 390618. [Google Scholar] [CrossRef] [Green Version]
  130. Sharma, D.; Blum, J.; Yang, X.; Beaulieu, N.; MacLeod, A.R.; Davidson, N.E. Release of Methyl CpG Binding Proteins and Histone Deacetylase 1 from the Estrogen Receptor α (ER) Promoter upon Reactivation in ER-Negative Human Breast Cancer Cells. Mol. Endocrinol. 2005, 19, 1740–1751. [Google Scholar] [CrossRef] [Green Version]
  131. Jeselsohn, R.; Buchwalter, G.; DE Angelis, C.; Brown, M.; Schiff, R. ESR1 mutations—A mechanism for acquired endocrine resistance in breast cancer. Nat. Rev. Clin. Oncol. 2015, 12, 573–583. [Google Scholar] [CrossRef] [Green Version]
  132. Liu, L.; Liu, S.; Luo, H.; Chen, C.; Zhang, X.; He, L.; Tu, G. GPR30-mediated HMGB1 upregulation in CAFs induces autophagy and tamoxifen resistance in ERα-positive breast cancer cells. Aging 2021, 13, 16178–16197. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, J.; Xie, S.; Yang, J.; Xiong, H.; Jia, Y.; Zhou, Y.; Chen, Y.; Ying, X.; Chen, C.; Ye, C.; et al. The long noncoding RNA H19 promotes tamoxifen resistance in breast cancer via autophagy. J. Hematol. Oncol. 2019, 12, 81. [Google Scholar] [CrossRef] [PubMed]
  134. Mo, Z.; Liu, M.; Yang, F.; Luo, H.; Li, Z.; Tu, G.; Yang, G. GPR30 as an initiator of tamoxifen resistance in hormone-dependent breast cancer. Breast Cancer Res. 2013, 15, R114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Lappano, R.; De Marco, P.; De Francesco, E.M.; Chimento, A.; Pezzi, V.; Maggiolini, M. Cross-talk between GPER and growth factor signaling. J. Steroid Biochem. Mol. Biol. 2013, 137, 50–56. [Google Scholar] [CrossRef] [PubMed]
  136. Yu, T.; Yang, G.; Hou, Y.; Tang, X.; Wu, C.; Wu, X.-A.; Guo, L.; Zhu, Q.; Luo, H.; Du, Y.-E.; et al. Cytoplasmic GPER translocation in cancer-associated fibroblasts mediates cAMP/PKA/CREB/glycolytic axis to confer tumor cells with multidrug resistance. Oncogene 2017, 36, 2131–2145. [Google Scholar] [CrossRef] [PubMed]
  137. Henson, E.S.; Gibson, S.B. Surviving cell death through epidermal growth factor (EGF) signal transduction pathways: Implications for cancer therapy. Cell. Signal. 2006, 18, 2089–2097. [Google Scholar] [CrossRef] [PubMed]
  138. Yan, J.; Yang, H.; Wang, G.; Sun, L.; Zhou, Y.; Guo, Y.; Xi, Z.; Jiang, X. Autophagy augmented by troglitazone is independent of EGFR transactivation and correlated with AMP-activated protein kinase signaling. Autophagy 2010, 6, 67–73. [Google Scholar] [CrossRef] [Green Version]
  139. Madeo, A.; Maggiolini, M. Nuclear Alternate Estrogen Receptor GPR30 Mediates 17β-Estradiol–Induced Gene Expression and Migration in Breast Cancer–Associated Fibroblasts. Cancer Res. 2010, 70, 6036–6046. [Google Scholar] [CrossRef] [Green Version]
  140. Luo, H.; Yang, G.; Yu, T.; Luo, S.; Wu, C.; Sun, Y.; Liu, M.; Tu, G. GPER-mediated proliferation and estradiol production in breast cancer-associated fibroblasts. Endocr. Relat. Cancer 2014, 21, 355–369. [Google Scholar] [CrossRef] [Green Version]
  141. Lee, M.-H.; Koh, D.; Na, H.; Ka, N.-L.; Kim, S.; Kim, H.-J.; Hong, S.; Shin, Y.K.; Seong, J.K.; Lee, M.-O. MTA1 is a novel regulator of autophagy that induces tamoxifen resistance in breast cancer cells. Autophagy 2018, 14, 812–824. [Google Scholar] [CrossRef]
  142. Desta, Z.; Ward, B.A.; Soukhova, N.V.; Flockhart, D.A. Comprehensive Evaluation of Tamoxifen Sequential Biotransformation by the Human Cytochrome P450 System in Vitro: Prominent Roles for CYP3A and CYP2D6. J. Pharmacol. Exp. Ther. 2004, 310, 1062–1075. [Google Scholar] [CrossRef]
  143. Shajahan-Haq, A.N.; Cook, K.L.; Schwartz-Roberts, J.L.; Eltayeb, A.E.; Demas, D.M.; Warri, A.M.; Facey, C.O.; Hilakivi-Clarke, L.A.; Clarke, R. MYC regulates the unfolded protein response and glucose and glutamine uptake in endocrine resistant breast cancer. Mol. Cancer 2014, 13, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Cook, K.L.; Clarke, R. Role of GRP78 in promoting therapeutic-resistant breast cancer. Future Med. Chem. 2015, 7, 1529–1534. [Google Scholar] [CrossRef] [PubMed]
  145. Cook, K.L.; Shajahan, A.N.; Wärri, A.; Jin, L.; Hilakivi-Clarke, L.A.; Clarke, R. Glucose-Regulated Protein 78 Controls Cross-talk between Apoptosis and Autophagy to Determine Antiestrogen Responsiveness. Cancer Res. 2012, 72, 3337–3349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Cook, K.L.; Clarke, P.A.G.; Parmar, J.; Hu, R.; Schwartz-Roberts, J.L.; Abu-Asab, M.; Wärri, A.; Baumann, W.T.; Clarke, R. Knockdown of estrogen receptor-α induces autophagy and inhibits antiestrogen-mediated unfolded protein response activation, promoting ROS-induced breast cancer cell death. FASEB J. 2014, 28, 3891–3905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Clarke, R.; Tyson, J.J.; Dixon, J.M. Endocrine resistance in breast cancer—An overview and update. Mol. Cell. Endocrinol. 2015, 418, 220–234. [Google Scholar] [CrossRef] [Green Version]
  148. Clarke, R.; Cook, K.L.; Hu, R.; Facey, C.O.; Tavassoly, I.; Schwartz, J.L.; Baumann, W.T.; Tyson, J.J.; Xuan, J.; Wang, Y.; et al. Endoplasmic Reticulum Stress, the Unfolded Protein Response, Autophagy, and the Integrated Regulation of Breast Cancer Cell Fate. Cancer Res. 2012, 72, 1321–1331. [Google Scholar] [CrossRef] [Green Version]
  149. Grkovic, S.; O’Reilly, V.C.; Han, S.; Hong, M.; Baxter, R.C.; Firth, S.M. IGFBP-3 binds GRP78, stimulates autophagy and promotes the survival of breast cancer cells exposed to adverse microenvironments. Oncogene 2013, 32, 2412–2420. [Google Scholar] [CrossRef] [Green Version]
  150. Gomez, B.P.; Riggins, R.B.; Shajahan, A.N.; Klimach, U.; Wang, A.; Crawford, A.C.; Zhu, Y.; Zwart, A.; Wang, M.; Clarke, R. Human X-Box binding protein-1 confers both estrogen independence and antiestrogen resistance in breast cancer cell lines. FASEB J. 2007, 21, 4013–4027. [Google Scholar] [CrossRef]
  151. Crawford, A.C.; Riggins, R.B.; Shajahan, A.N.; Zwart, A.; Clarke, R. Co-Inhibition of BCL-W and BCL2 Restores Antiestrogen Sensitivity through BECN1 and Promotes an Autophagy-Associated Necrosis. PLoS ONE 2010, 5, e8604. [Google Scholar] [CrossRef] [Green Version]
  152. Prensner, J.R.; Chinnaiyan, A.M. The Emergence of lncRNAs in Cancer Biology. Cancer Discov. 2011, 1, 391–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Huarte, M.; Guttman, M.; Feldser, D.; Garber, M.; Koziol, M.J.; Kenzelmann-Broz, D.; Khalil, A.M.; Zuk, O.; Amit, I.; Rabani, M.; et al. A Large Intergenic Noncoding RNA Induced by p53 Mediates Global Gene Repression in the p53 Response. Cell 2010, 142, 409–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Yu, X.; Luo, A.; Liu, Y.; Wang, S.; Li, Y.; Shi, W.; Liu, Z.; Qu, X. MiR-214 increases the sensitivity of breast cancer cells to tamoxifen and fulvestrant through inhibition of autophagy. Mol. Cancer 2015, 14, 208. [Google Scholar] [CrossRef] [Green Version]
  155. Jung, C.H.; Ro, S.-H.; Cao, J.; Otto, N.M.; Kim, D.-H. mTOR regulation of autophagy. FEBS Lett. 2010, 584, 1287–1295. [Google Scholar] [CrossRef] [Green Version]
  156. Zhao, G.-X.; Pan, H.; Ouyang, D.-Y.; He, X.-H. The critical molecular interconnections in regulating apoptosis and autophagy. Ann. Med. 2015, 47, 305–315. [Google Scholar] [CrossRef]
  157. Schumacker, P.T. Reactive oxygen species in cancer cells: Live by the sword, die by the sword. Cancer Cell 2006, 10, 175–176. [Google Scholar] [CrossRef] [Green Version]
  158. Bacci, M.; Lorito, N.; Ippolito, L.; Ramazzotti, M.; Luti, S.; Romagnoli, S.; Parri, M.; Bianchini, F.; Cappellesso, F.; Virga, F.; et al. Reprogramming of Amino Acid Transporters to Support Aspartate and Glutamate Dependency Sustains Endocrine Resistance in Breast Cancer. Cell Rep. 2019, 28, 104–118.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Meijer, A.J.; Lorin, S.; Blommaart, E.F.; Codogno, P. Regulation of autophagy by amino acids and MTOR-dependent signal transduction. Amino Acids 2015, 47, 2037–2063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Sikder, M.O.F.; Sivaprakasam, S.; Brown, T.P.; Thangaraju, M.; Bhutia, Y.D.; Ganapathy, V. SLC6A14, a Na+/Cl−-coupled amino acid transporter, functions as a tumor promoter in colon and is a target for Wnt signaling. Biochem. J. 2020, 477, 1409–1425. [Google Scholar] [CrossRef] [Green Version]
  161. Ziaeemehr, A.; Shahidsales, S.; Ghosi, Z.; Avan, A.; Aldavood, A.S.; Anvari, K.; Makhdoomi, Y.; Asadi, M. Association of the involvement of axillary lymph nodes in HER-2/neu overexpression in patients with breast cancer. Breast J. 2019, 25, 537–538. [Google Scholar] [CrossRef]
  162. Giannone, G.; Milani, A.; Geuna, E.; Galizia, D.; Biello, F.; Montemurro, F. What is the best pharmacotherapeutic strategy for HER-2 positive breast cancer? Expert Opin. Pharmacother. 2019, 20, 5–9. [Google Scholar] [CrossRef] [PubMed]
  163. Adamczyk, A.; Kruczak, A.; Harazin-Lechowska, A.; Ambicka, A.; Grela-Wojewoda, A.; Domagała-Haduch, M.; Janecka-Widła, A.; Majchrzyk, K.; Cichocka, A.; Ryś, J.; et al. Relationship between HER2 gene status and selected potential biological features related to trastuzumab resistance and its influence on survival of breast cancer patients undergoing trastuzumab adjuvant treatment. OncoTargets Ther. 2018, 11, 4525–4535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Datta, A.; Loo, S.Y.; Huang, B.; Wong, L.; Tan, S.S.; Tan, T.Z.; Lee, S.-C.; Thiery, J.P.; Lim, Y.C.; Yong, W.P.; et al. SPHK1 regulates proliferation and survival responses in triple-negative breast cancer. Oncotarget 2014, 5, 5920–5933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Maczis, M.A.; Maceyka, M.; Waters, M.R.; Newton, J.; Singh, M.; Rigsby, M.F.; Turner, T.H.; Alzubi, M.A.; Harrell, J.C.; Milstien, S.; et al. Sphingosine kinase 1 activation by estrogen receptor α36 contributes to tamoxifen resistance in breast cancer. J. Lipid Res. 2018, 59, 2297–2307. [Google Scholar] [CrossRef] [Green Version]
  166. Carreras, I.; Aytan, N.; Choi, J.-K.; Tognoni, C.M.; Kowall, N.W.; Jenkins, B.G.; Dedeoglu, A. Dual dose-dependent effects of fingolimod in a mouse model of Alzheimer’s disease. Sci. Rep. 2019, 9, 10972. [Google Scholar] [CrossRef] [Green Version]
  167. White, C.; Alshaker, H.; Cooper, C.; Winkler, M.; Pchejetski, D. The emerging role of FTY720 (Fingolimod) in cancer treatment. Oncotarget 2016, 7, 23106–23127. [Google Scholar] [CrossRef] [Green Version]
  168. Chung, W.P.; Huang, W.L.; Liao, W.A.; Hung, C.H.; Chiang, C.W.; Cheung, C.H.A.; Su, W.C. FTY720 in resistant human epidermal growth factor receptor 2-positive breast cancer. Sci. Rep. 2022, 12, 241. [Google Scholar] [CrossRef]
  169. Kataoka, Y.; Mukohara, T.; Shimada, H.; Saijo, N.; Hirai, M.; Minami, H. Association between gain-of-function mutations in PIK3CA and resistance to HER2-targeted agents in HER2-amplified breast cancer cell lines. Ann. Oncol. 2010, 21, 255–262. [Google Scholar] [CrossRef]
  170. Bjørkøy, G.; Lamark, T.; Brech, A.; Outzen, H.; Perander, M.; Øvervatn, A.; Stenmark, H.; Johansen, T. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J. Cell Biol. 2005, 171, 603–614. [Google Scholar] [CrossRef] [Green Version]
  171. Liu, C.-H.; Ampuero, J.; Gil-Gómez, A.; Montero-Vallejo, R.; Rojas, Á.; Muñoz-Hernández, R.; Gallego-Durán, R.; Romero-Gómez, M. miRNAs in patients with non-alcoholic fatty liver disease: A systematic review and meta-analysis. J. Hepatol. 2018, 69, 1335–1348. [Google Scholar] [CrossRef]
  172. Yu, Y.; Chai, J. The function of miRNAs and their potential as therapeutic targets in burn-induced insulin resistance (Review). Int. J. Mol. Med. 2015, 35, 305–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Han, M.; Hu, J.; Lu, P.; Cao, H.; Yu, C.; Li, X.; Qian, X.; Yang, X.; Yang, Y.; Han, N.; et al. Exosome-transmitted miR-567 reverses trastuzumab resistance by inhibiting ATG5 in breast cancer. Cell Death Dis. 2020, 11, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Han, M.; Qian, X.; Cao, H.; Wang, F.; Li, X.; Han, N.; Yang, X.; Yang, Y.; Dou, D.; Hu, J.; et al. lncRNA ZNF649-AS1 Induces Trastuzumab Resistance by Promoting ATG5 Expression and Autophagy. Mol. Ther. 2020, 28, 2488–2502. [Google Scholar] [CrossRef] [PubMed]
  175. Yarden, Y.; Sliwkowski, M.X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2001, 2, 127–137. [Google Scholar] [CrossRef]
  176. Appert-Collin, A.; Hubert, P.; Crémel, G.; Bennasroune, A. Role of ErbB Receptors in Cancer Cell Migration and Invasion. Front. Pharmacol. 2015, 6, 283. [Google Scholar] [CrossRef] [Green Version]
  177. Tsang, R.Y.; Sadeghi, S.; Finn, R.S. Lapatinib, a Dual-Targeted Small Molecule Inhibitor of EGFR and HER2, in HER2-Amplified Breast Cancer: From Bench to Bedside. Clin. Med. Insights Ther. 2011, 3, CMT.S3783. [Google Scholar] [CrossRef] [Green Version]
  178. Chen, S.; Li, X.; Feng, J.; Chang, Y.; Wang, Z.; Wen, A. Autophagy facilitates the Lapatinib resistance of HER2 positive breast cancer cells. Med. Hypotheses 2011, 77, 206–208. [Google Scholar] [CrossRef]
  179. Huang, M.; Zhou, Y.; Duan, D.; Yang, C.; Zhou, Z.; Li, F.; Kong, Y.; Hsieh, Y.-C.; Zhang, R.; Ding, W.; et al. Targeting ubiquitin conjugating enzyme UbcH5b by a triterpenoid PC3-15 from Schisandra plants sensitizes triple-negative breast cancer cells to lapatinib. Cancer Lett. 2021, 504, 125–136. [Google Scholar] [CrossRef]
  180. Moulder, S.L.; Yakes, F.M.; Muthuswamy, S.K.; Bianco, R.; Simpson, J.F.; Arteaga, C.L. Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Res. 2001, 61, 8887–8895. [Google Scholar]
  181. Anido, J.; Matar, P.; Albanell, J.; Guzmán, M.; Rojo, F.; Arribas, J.; Averbuch, S.; Baselga, J. ZD1839, a specific epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, induces the formation of inactive EGFR/HER2 and EGFR/HER3 heterodimers and prevents heregulin signaling in HER2-overexpressing breast cancer cells. Clin. Cancer Res. 2003, 9, 1274–1283. [Google Scholar]
  182. Campiglio, M.; Locatelli, A.; Olgiati, C.; Normanno, N.; Somenzi, G.; Viganò, L.; Fumagalli, M.; Ménard, S.; Gianni, L. Inhibition of proliferation and induction of apoptosis in breast cancer cells by the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor ZD1839 (‘Iressa’) is independent of EGFR expression level. J. Cell. Physiol. 2004, 198, 259–268. [Google Scholar] [CrossRef] [PubMed]
  183. Baselga, J.; Albanell, J.; Ruiz, A.; Lluch, A.; Gascón, P.; Guillém, V.; González, S.; Sauleda, S.; Marimón, I.; Tabernero, J.M.; et al. Phase II and Tumor Pharmacodynamic Study of Gefitinib in Patients with Advanced Breast Cancer. J. Clin. Oncol. 2005, 23, 5323–5333. [Google Scholar] [CrossRef] [PubMed]
  184. Nielsen, T.O.; Hsu, F.D.; Jensen, K.; Cheang, M.; Karaca, G.; Hu, Z.; Hernandez-Boussard, T.; Livasy, C.; Cowan, D.; Dressler, L.; et al. Immunohistochemical and Clinical Characterization of the Basal-Like Subtype of Invasive Breast Carcinoma. Clin. Cancer Res. 2004, 10, 5367–5374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Fu, W.; Sun, H.; Zhao, Y.; Chen, M.; Yang, X.; Liu, Y.; Jin, W. BCAP31 drives TNBC development by modulating ligand-independent EGFR trafficking and spontaneous EGFR phosphorylation. Theranostics 2019, 9, 6468–6484. [Google Scholar] [CrossRef]
  186. Liao, M.; Qin, R.; Huang, W.; Zhu, H.-P.; Peng, F.; Han, B.; Liu, B. Targeting regulated cell death (RCD) with small-molecule compounds in triple-negative breast cancer: A revisited perspective from molecular mechanisms to targeted therapies. J. Hematol. Oncol. 2022, 15, 44. [Google Scholar] [CrossRef]
  187. Liu, Z.; He, K.; Ma, Q.; Yu, Q.; Liu, C.; Ndege, I.; Wang, X.; Yu, Z. Autophagy inhibitor facilitates gefitinib sensitivity in vitro and in vivo by activating mitochondrial apoptosis in triple negative breast cancer. PLoS ONE 2017, 12, e0177694. [Google Scholar] [CrossRef] [Green Version]
  188. Dragowska, W.H.; Weppler, S.A.; Wang, J.C.; Wong, L.Y.; Kapanen, A.I.; Rawji, J.S.; Warburton, C.; Qadir, M.A.; Donohue, E.; Roberge, M.; et al. Induction of Autophagy Is an Early Response to Gefitinib and a Potential Therapeutic Target in Breast Cancer. PLoS ONE 2013, 8, e76503. [Google Scholar] [CrossRef] [Green Version]
  189. Weihua, Z.; Tsan, R.; Huang, W.-C.; Wu, Q.; Chiu, C.-H.; Fidler, I.J.; Hung, M.-C. Survival of Cancer Cells Is Maintained by EGFR Independent of Its Kinase Activity. Cancer Cell 2008, 13, 385–393. [Google Scholar] [CrossRef] [Green Version]
  190. Pernas, S.; Tolaney, S.M.; Winer, E.P.; Goel, S. CDK4/6 inhibition in breast cancer: Current practice and future directions. Ther. Adv. Med. Oncol. 2018, 10, 1758835918786451. [Google Scholar] [CrossRef] [Green Version]
  191. Yu, Q.; Geng, Y.; Sicinski, P. Specific protection against breast cancers by cyclin D1 ablation. Nature 2001, 411, 1017–1021. [Google Scholar] [CrossRef]
  192. Arnold, A.; Papanikolaou, A. Cyclin D1 in Breast Cancer Pathogenesis. J. Clin. Oncol. 2005, 23, 4215–4224. [Google Scholar] [CrossRef] [PubMed]
  193. Yu, Q.; Sicinska, E.; Geng, Y.; Ahnström, M.; Zagozdzon, A.; Kong, Y.; Gardner, H.; Kiyokawa, H.; Harris, L.N.; Stål, O.; et al. Requirement for CDK4 kinase function in breast cancer. Cancer Cell 2006, 9, 23–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Casimiro, M.C.; Di Sante, G.; Di Rocco, A.; Loro, E.; Pupo, C.; Pestell, T.G.; Bisetto, S.; Velasco-Velázquez, M.A.; Jiao, X.; Li, Z.; et al. Cyclin D1 Restrains Oncogene-Induced Autophagy by Regulating the AMPK–LKB1 Signaling Axis. Cancer Res. 2017, 77, 3391–3405. [Google Scholar] [CrossRef] [Green Version]
  195. Li, Y.; Chen, Y. AMPK and Autophagy. Adv. Exp. Med. Biol. 2019, 1206, 85–108. [Google Scholar] [CrossRef] [PubMed]
  196. Piezzo, M.; Chiodini, P.; Riemma, M.; Cocco, S.; Caputo, R.; Cianniello, D.; Di Gioia, G.; Di Lauro, V.; Rella, F.D.; Fusco, G.; et al. Progression-Free Survival and Overall Survival of CDK 4/6 Inhibitors Plus Endocrine Therapy in Metastatic Breast Cancer: A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2020, 21, 6400. [Google Scholar] [CrossRef]
  197. Piezzo, M.; Cocco, S.; Caputo, R.; Cianniello, D.; Gioia, G.D.; Lauro, V.D.; Fusco, G.; Martinelli, C.; Nuzzo, F.; Pensabene, M.; et al. Targeting Cell Cycle in Breast Cancer: CDK4/6 Inhibitors. Int. J. Mol. Sci. 2020, 21, 6479. [Google Scholar] [CrossRef]
  198. Wang, R.; Zhang, Q.; Peng, X.; Zhou, C.; Zhong, Y.; Chen, X.; Qiu, Y.; Jin, M.; Gong, M.; Kong, D. Stellettin B Induces G1 Arrest, Apoptosis and Autophagy in Human Non-small Cell Lung Cancer A549 Cells via Blocking PI3K/Akt/mTOR Pathway. Sci. Rep. 2016, 6, 27071. [Google Scholar] [CrossRef] [Green Version]
  199. Yang, Z.J.; Chee, C.E.; Huang, S.; Sinicrope, F.A. The Role of Autophagy in Cancer: Therapeutic Implications. Mol. Cancer Ther. 2011, 10, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
  200. Vijayaraghavan, S.; Karakas, C.; Doostan, I.; Chen, X.; Bui, T.; Yi, M.; Raghavendra, A.S.; Zhao, Y.; Bashour, S.I.; Ibrahim, N.K.; et al. CDK4/6 and autophagy inhibitors synergistically induce senescence in Rb positive cytoplasmic cyclin E negative cancers. Nat. Commun. 2017, 8, 15916. [Google Scholar] [CrossRef] [Green Version]
  201. Nardone, V.; Barbarino, M.; Angrisani, A.; Correale, P.; Pastina, P.; Cappabianca, S.; Reginelli, A.; Mutti, L.; Miracco, C.; Giannicola, R.; et al. CDK4, CDK6/cyclin-D1 Complex Inhibition and Radiotherapy for Cancer Control: A Role for Autophagy. Int. J. Mol. Sci. 2021, 22, 8391. [Google Scholar] [CrossRef]
  202. Cocco, S.; Leone, A.; Piezzo, M.; Caputo, R.; Di Lauro, V.; Di Rella, F.; Fusco, G.; Capozzi, M.; Gioia, G.D.; Budillon, A.; et al. Targeting Autophagy in Breast Cancer. Int. J. Mol. Sci. 2020, 21, 7836. [Google Scholar] [CrossRef] [PubMed]
  203. Lanceta, L.; O’Neill, C.; Lypova, N.; Li, X.; Rouchka, E.; Waigel, S.; Gomez-Gutierrez, J.G.; Chesney, J.; Imbert-Fernandez, Y. Transcriptomic Profiling Identifies Differentially Expressed Genes in Palbociclib-Resistant ER+ MCF7 Breast Cancer Cells. Genes 2020, 11, 467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Fassl, A.; Brain, C.; Abu-Remaileh, M.; Stukan, I.; Butter, D.; Stepien, P.; Feit, A.S.; Bergholz, J.; Michowski, W.; Otto, T.; et al. Increased lysosomal biomass is responsible for the resistance of triple-negative breast cancers to CDK4/6 inhibition. Sci. Adv. 2020, 6, eabb2210. [Google Scholar] [CrossRef] [PubMed]
  205. Scheidemann, E.R.; Shajahan-Haq, A.N. Resistance to CDK4/6 Inhibitors in Estrogen Receptor-Positive Breast Cancer. Int. J. Mol. Sci. 2021, 22, 12292. [Google Scholar] [CrossRef]
  206. Connell, P.P.; Hellman, S. Advances in Radiotherapy and Implications for the Next Century: A Historical Perspective. Cancer Res. 2009, 69, 383–392. [Google Scholar] [CrossRef] [Green Version]
  207. Tam, S.Y.; Wu, V.W.C.; Law, H.K.W. Influence of autophagy on the efficacy of radiotherapy. Radiat. Oncol. 2017, 12, 57. [Google Scholar] [CrossRef] [Green Version]
  208. Lu, T.; Zhu, Z.; Wu, J.; She, H.; Han, R.; Xu, H.; Qin, Z.-H. DRAM1 regulates autophagy and cell proliferation via inhibition of the phosphoinositide 3-kinase-Akt-mTOR-ribosomal protein S6 pathway. Cell Commun. Signal. 2019, 17, 28. [Google Scholar] [CrossRef] [Green Version]
  209. Meng, C.; Liu, Y.; Shen, Y.; Liu, S.; Wang, Z.; Ye, Q.; Liu, H.; Liu, X.; Jia, L. MicroRNA-26b suppresses autophagy in breast cancer cells by targeting DRAM1 mRNA, and is downregulated by irradiation. Oncol. Lett. 2018, 15, 1435–1440. [Google Scholar] [CrossRef] [Green Version]
  210. Sun, S.; Zhang, W.; Cui, Z.; Chen, Q.; Xie, P.; Zhou, C.; Liu, B.; Peng, X.; Zhang, Y. High mobility group box-1 and its clinical value in breast cancer. OncoTargets Ther. 2015, 8, 413–419. [Google Scholar] [CrossRef] [Green Version]
  211. Lee, D.Y.; Arnott, D.; Brown, E.J. Ubiquilin4 is an adaptor protein that recruits Ubiquilin1 to the autophagy machinery. EMBO Rep. 2013, 14, 373–381. [Google Scholar] [CrossRef]
  212. Ahmad, A. Introduction to Cancer Metastasis; Academic Press: Cambridge, MA, USA, 2016. [Google Scholar]
  213. Tang, D.; Kang, R.; Zeh, H.J., III; Lotze, M.T. High-mobility group box 1 and cancer. Biochim. Biophys. Acta 2010, 1799, 131–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Wang, C.Q.; Huang, B.F.; Wang, Y.; Hu, G.R.; Wang, Q.; Shao, J.K. Expression of HMGB1 protein in breast cancer and its clinicopathological significance. Zhonghua Bing Li Xue Za Zhi 2020, 49, 57–61. [Google Scholar] [PubMed]
  215. Kang, R.; Livesey, K.M.; Zeh, I.H.J.; Loze, M.T.; Tang, D. HMGB1: A novel Beclin 1-binding protein active in autophagy. Autophagy 2010, 6, 1209–1211. [Google Scholar] [CrossRef] [PubMed]
  216. Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Criollo, A.; Ortiz, C.; Lidereau, R.; Mariette, C.; Chaput, N.; Mira, J.-P.; Delaloge, S.; et al. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol. Rev. 2007, 220, 47–59. [Google Scholar] [CrossRef] [PubMed]
  217. Ni, P.; Zhang, Y.; Liu, Y.; Lin, X.; Su, X.; Lu, H.; Shen, H.; Xu, W.; Xu, H.; Su, Z. HMGB1 silence could promote MCF-7 cell apoptosis and inhibit invasion and metastasis. Int. J. Clin. Exp. Pathol. 2015, 8, 15940–15946. [Google Scholar] [PubMed]
  218. Wang, L.-L.; Meng, Q.-H.; Jiao, Y.; Xu, J.-Y.; Ge, C.-M.; Zhou, J.-Y.; Rosen, E.M.; Wang, H.-C.; Fan, S.-J. High-Mobility Group Boxes Mediate Cell Proliferation and Radiosensitivity via Retinoblastoma-Interaction-Dependent and -Independent Mechanisms. Cancer Biother. Radiopharm. 2012, 27, 329–335. [Google Scholar] [CrossRef] [Green Version]
  219. Liu, B.; Qi, X.; Zhang, X.; Gao, D.; Fang, K.; Guo, Z.; Li, L. Med19 is involved in chemoresistance by mediating autophagy through HMGB1 in breast cancer. J. Cell. Biochem. 2019, 120, 507–518. [Google Scholar] [CrossRef] [Green Version]
  220. Jiao, Y.; Wang, H.-C.; Fan, S.-J. Growth suppression and radiosensitivity increase by HMGB1 in breast cancer. Acta Pharmacol. Sin. 2007, 28, 1957–1967. [Google Scholar] [CrossRef] [Green Version]
  221. Ke, S.; Zhou, F.; Yang, H.; Wei, Y.; Gong, J.; Mei, Z.; Wu, L.; Yu, H.; Zhou, Y. Downregulation of high mobility group box 1 modulates telomere homeostasis and increases the radiosensitivity of human breast cancer cells. Int. J. Oncol. 2015, 46, 1051–1058. [Google Scholar] [CrossRef] [Green Version]
  222. Ladoire, S.; Enot, D.; Senovilla, L.; Ghiringhelli, F.; Poirier-Colame, V.; Chaba, K.; Semeraro, M.; Chaix, M.; Penault-Llorca, F.; Arnould, L.; et al. The presence of LC3B puncta and HMGB1 expression in malignant cells correlate with the immune infiltrate in breast cancer. Autophagy 2016, 12, 864–875. [Google Scholar] [CrossRef] [Green Version]
  223. Luo, J.; Chen, J.; He, L. mir-129-5p Attenuates Irradiation-Induced Autophagy and Decreases Radioresistance of Breast Cancer Cells by Targeting HMGB1. Med. Sci. Monit. 2015, 21, 4122–4129. [Google Scholar] [CrossRef] [Green Version]
  224. Ge, J.; Chen, Z.; Huang, J.; Chen, J.; Yuan, W.; Deng, Z.; Chen, Z. Upregulation of Autophagy-Related Gene-5 (ATG-5) Is Associated with Chemoresistance in Human Gastric Cancer. PLoS ONE 2014, 9, e110293. [Google Scholar] [CrossRef] [PubMed]
  225. Zheng, K.; Li, Y.; Wang, S.; Wang, X.; Liao, C.; Hu, X.; Fan, L.; Kang, Q.; Zeng, Y.; Wu, X.; et al. Inhibition of autophagosome-lysosome fusion by ginsenoside Ro via the ESR2-NCF1-ROS pathway sensitizes esophageal cancer cells to 5-fluorouracil-induced cell death via the CHEK1-mediated DNA damage checkpoint. Autophagy 2016, 12, 1593–1613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Chittaranjan, S.; Bortnik, S.; Dragowska, W.H.; Xu, J.; Abeysundara, N.; Leung, A.; Go, N.E.; DeVorkin, L.; Weppler, S.A.; Gelmon, K.; et al. Autophagy Inhibition Augments the Anticancer Effects of Epirubicin Treatment in Anthracycline-Sensitive and -Resistant Triple-Negative Breast Cancer. Clin. Cancer Res. 2014, 20, 3159–3173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Yin, L.; Duan, J.-J.; Bian, X.-W.; Yu, S.-C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020, 22, 61. [Google Scholar] [CrossRef] [PubMed]
  228. Ghersi, D.; Wilcken, N.; Simes, R.J. A systematic review of taxane-containing regimens for metastatic breast cancer. Br. J. Cancer 2005, 93, 293–301. [Google Scholar] [CrossRef] [Green Version]
  229. Wu, Q.; Siddharth, S.; Sharma, D. Triple Negative Breast Cancer: A Mountain Yet to Be Scaled Despite the Triumphs. Cancers 2021, 13, 3697. [Google Scholar] [CrossRef]
  230. Lin, H.-Y.; Kuei, C.-H.; Lee, H.-H.; Lin, C.-H.; Chen, Y.-L.; Chen, C.-L.; Lin, Y.-F. TNFSF13 upregulation confers chemotherapeutic resistance via triggering autophagy initiation in triple-negative breast cancer. J. Mol. Med. 2020, 98, 1255–1267. [Google Scholar] [CrossRef]
  231. Koscianska, E.; Starega-Roslan, J.; Krzyzosiak, W.J. The Role of Dicer Protein Partners in the Processing of MicroRNA Precursors. PLoS ONE 2011, 6, e28548. [Google Scholar] [CrossRef] [Green Version]
  232. Sha, L.-Y.; Zhang, Y.; Wang, W.; Sui, X.; Liu, S.-K.; Wang, T.; Zhang, H. MiR-18a upregulation decreases Dicer expression and confers paclitaxel resistance in triple negative breast cancer. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2201–2208. [Google Scholar]
  233. Li, C.; Chen, L.; Song, W.; Peng, B.; Zhu, J.; Fang, L. DICER activates autophagy and promotes cisplatin resistance in non-small cell lung cancer by binding with let-7i-5p. Acta Histochem. 2021, 123, 151788. [Google Scholar] [CrossRef]
  234. Fan, Y.-X.; Dai, Y.-Z.; Wang, X.-L.; Ren, Y.-Q.; Han, J.-J.; Zhang, H. MiR-18a upregulation enhances autophagy in triple negative cancer cells via inhibiting mTOR signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2194–2200. [Google Scholar] [PubMed]
  235. Tanida, S.; Mizoshita, T.; Ozeki, K.; Tsukamoto, H.; Kamiya, T.; Kataoka, H.; Sakamuro, D.; Joh, T. Mechanisms of Cisplatin-Induced Apoptosis and of Cisplatin Sensitivity: Potential of BIN1 to Act as a Potent Predictor of Cisplatin Sensitivity in Gastric Cancer Treatment. Int. J. Surg. Oncol. 2012, 2012, 862879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Rabik, C.A.; Dolan, M.E. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev. 2007, 33, 9–23. [Google Scholar] [CrossRef] [Green Version]
  237. Basourakos, S.P.; Li, L.; Aparicio, A.M.; Corn, P.G.; Kim, J.; Thompson, T.C. Combination Platinum-based and DNA Damage Response-targeting Cancer Therapy: Evolution and Future Directions. Curr. Med. Chem. 2017, 24, 1586–1606. [Google Scholar] [CrossRef] [PubMed]
  238. Zhou, J.; Kang, Y.; Chen, L.; Wang, H.; Liu, J.; Zeng, S.; Yu, L. The Drug-Resistance Mechanisms of Five Platinum-Based Anticancer Agents. Front. Pharmacol. 2020, 11, 343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Yu, L.; Gu, C.; Zhong, D.; Shi, L.; Kong, Y.; Zhou, Z.; Liu, S. Induction of autophagy counteracts the anticancer effect of cisplatin in human esophageal cancer cells with acquired drug resistance. Cancer Lett. 2014, 355, 34–45. [Google Scholar] [CrossRef] [PubMed]
  240. Liu, W.; Zhang, Z.; Zhang, Y.; Chen, X.; Guo, S.; Lei, Y.; Xu, Y.; Ji, C.; Bi, Z.; Wang, K. HMGB1-mediated autophagy modulates sensitivity of colorectal cancer cells to oxaliplatin via MEK/ERK signaling pathway. Cancer Biol. Ther. 2015, 16, 511–517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Wu, B.; Cui, J.; Yang, X.-M.; Liu, Z.-Y.; Song, F.; Li, L.; Jiang, J.-L.; Chen, Z.-N. Cytoplasmic fragment of CD147 generated by regulated intramembrane proteolysis contributes to HCC by promoting autophagy. Cell Death Dis. 2017, 8, e2925. [Google Scholar] [CrossRef] [Green Version]
  242. Gao, J.; Wang, W. Knockdown of galectin-1 facilitated cisplatin sensitivity by inhibiting autophagy in neuroblastoma cells. Chem. Interactions 2019, 297, 50–56. [Google Scholar] [CrossRef]
  243. Fukuda, T.; Oda, K.; Wada-Hiraike, O.; Sone, K.; Inaba, K.; Ikeda, Y.; Miyasaka, A.; Kashiyama, T.; Tanikawa, M.; Arimoto, T.; et al. The anti-malarial chloroquine suppresses proliferation and overcomes cisplatin resistance of endometrial cancer cells via autophagy inhibition. Gynecol. Oncol. 2015, 137, 538–545. [Google Scholar] [CrossRef] [PubMed]
  244. Ma, L.; Xu, Y.; Su, J.; Yu, H.; Kang, J.; Li, H.; Li, X.; Xie, Q.; Yu, C.; Sun, L.; et al. Autophagic flux promotes cisplatin resistance in human ovarian carcinoma cells through ATP-mediated lysosomal function. Int. J. Oncol. 2015, 47, 1890–1900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Shukla, A.A.; Thömmes, J. Recent advances in large-scale production of monoclonal antibodies and related proteins. Trends Biotechnol. 2010, 28, 253–261. [Google Scholar] [CrossRef]
  246. Cimino-Mathews, A.; Foote, J.B.; Emens, L.A. Immune targeting in breast cancer. Oncology 2015, 29, 375–385. [Google Scholar] [PubMed]
  247. Wang, B.; Rouzier, R.; Albarracin, C.T.; Şahin, A.; Wagner, P.; Yang, Y.; Smith, T.L.; Meric-Bernstam, F.; Marcelo, A.C.; Hortobagyi, G.N.; et al. Expression of sigma 1 receptor in human breast cancer. Breast Cancer Res. Treat. 2004, 87, 205–214. [Google Scholar] [CrossRef] [PubMed]
  248. Maher, C.M.; Thomas, J.D.; Haas, D.A.; Longen, C.G.; Oyer, H.M.; Tong, J.Y.; Kim, F.J. Small-Molecule Sigma1 Modulator Induces Autophagic Degradation of PD-L1. Mol. Cancer Res. 2018, 16, 243–255. [Google Scholar] [CrossRef] [Green Version]
  249. Gao, L.; Chen, Y. Autophagy controls programmed death-ligand 1 expression on cancer cells (Review). Biomed. Rep. 2021, 15, 84. [Google Scholar] [CrossRef]
  250. Wang, H.; Bloom, O.; Zhang, M.; Vishnubhakat, J.M.; Ombrellino, M.; Che, J.; Frazier, A.; Yang, H.; Ivanova, S.; Borovikova, L.; et al. HMG-1 as a Late Mediator of Endotoxin Lethality in Mice. Science 1999, 285, 248–251. [Google Scholar] [CrossRef]
  251. Pullerits, R.; Jonsson, I.-M.; Verdrengh, M.; Bokarewa, M.; Andersson, U.; Erlandsson-Harris, H.; Tarkowski, A. High mobility group box chromosomal protein 1, a DNA binding cytokine, induces arthritis. Arthritis Rheum. 2003, 48, 1693–1700. [Google Scholar] [CrossRef]
  252. Popovic, K.; Ek, M.; Espinosa, A.; Padyukov, L.; Harris, H.E.; Wahren-Herlenius, M.; Nyberg, F. Increased expression of the novel proinflammatory cytokine high mobility group box chromosomal protein 1 in skin lesions of patients with lupus erythematosus. Arthritis Rheum. 2005, 52, 3639–3645. [Google Scholar] [CrossRef]
  253. Zong, M.; Bruton, J.D.; Grundtman, C.; Yang, H.; Li, J.H.; Alexanderson, H.; Palmblad, K.; Andersson, U.; Harris, H.E.; Lundberg, I.E.; et al. TLR4 as receptor for HMGB1 induced muscle dysfunction in myositis. Ann. Rheum. Dis. 2013, 72, 1390–1399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Park, J.S.; Svetkauskaite, D.; He, Q.; Kim, J.Y.; Strassheim, D.; Ishizaka, A.; Abraham, E. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J. Biol. Chem. 2004, 279, 7370–7377. [Google Scholar] [CrossRef] [Green Version]
  255. Andersson, U.; Wang, H.; Palmblad, K.; Aveberger, A.-C.; Bloom, O.; Erlandsson-Harris, H.; Janson, A.; Kokkola, R.; Zhang, M.; Yang, H.; et al. High Mobility Group 1 Protein (Hmg-1) Stimulates Proinflammatory Cytokine Synthesis in Human Monocytes. J. Exp. Med. 2000, 192, 565–570. [Google Scholar] [CrossRef] [PubMed]
  256. Yang, D.; Chen, Q.; Yang, H.; Tracey, K.J.; Bustin, M.; Oppenheim, J.J. High mobility group box-1 protein induces the migration and activation of human dendritic cells and acts as an alarmin. J. Leukoc. Biol. 2007, 81, 59–66. [Google Scholar] [CrossRef]
  257. Hubert, P.; Roncarati, P.; Demoulin, S.; Pilard, C.; Ancion, M.; Reynders, C.; Lerho, T.; Bruyere, D.; Lebeau, A.; Radermecker, C.; et al. Extracellular HMGB1 blockade inhibits tumor growth through profoundly remodeling immune microenvironment and enhances checkpoint inhibitor-based immunotherapy. J. Immunother. Cancer 2021, 9, e001966. [Google Scholar] [CrossRef] [PubMed]
  258. Yamamoto, K.; Venida, A.; Yano, J.; Biancur, D.E.; Kakiuchi, M.; Gupta, S.; Sohn, A.S.W.; Mukhopadhyay, S.; Lin, E.Y.; Parker, S.J.; et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 2020, 581, 100–105. [Google Scholar] [CrossRef]
  259. Loi, M.; Müller, A.; Steinbach, K.; Niven, J.; Barreira da Silva, R.; Paul, P.; Ligeon, L.-A.; Caruso, A.; Albrecht, R.A.; Becker, A.C.; et al. Macroautophagy Proteins Control MHC Class I Levels on Dendritic Cells and Shape Anti-viral CD8(+) T Cell Responses. Cell Rep. 2016, 15, 1076–1087. [Google Scholar] [CrossRef] [Green Version]
  260. DeVorkin, L.; Pavey, N.; Carleton, G.; Comber, A.; Ho, C.; Lim, J.; McNamara, E.; Huang, H.; Kim, P.; Zacharias, L.G.; et al. Autophagy Regulation of Metabolism Is Required for CD8(+) T Cell Anti-tumor Immunity. Cell Rep. 2019, 27, 502–513.e5. [Google Scholar] [CrossRef] [Green Version]
  261. Schrezenmeier, E.; Dörner, T. Mechanisms of action of hydroxychloroquine and chloroquine: Implications for rheumatology. Nat. Rev. Rheumatol. 2020, 16, 155–166. [Google Scholar] [CrossRef]
  262. Cook, K.L.; Wärri, A.; Soto-Pantoja, D.R.; Clarke, P.A.; Cruz, M.I.; Zwart, A.; Clarke, R. Hydroxychloroquine inhibits autophagy to potentiate antiestrogen responsiveness in ER+ breast cancer. Clin. Cancer Res. 2014, 20, 3222–3232. [Google Scholar] [CrossRef] [Green Version]
  263. Lee, H.-O.; Mustafa, A.; Hudes, G.R.; Kruger, W.D. Hydroxychloroquine Destabilizes Phospho-S6 in Human Renal Carcinoma Cells. PLoS ONE 2015, 10, e0131464. [Google Scholar] [CrossRef] [PubMed]
  264. Shi, T.-T.; Yu, X.-X.; Yan, L.-J.; Xiao, H.-T. Research progress of hydroxychloroquine and autophagy inhibitors on cancer. Cancer Chemother. Pharmacol. 2017, 79, 287–294. [Google Scholar] [CrossRef]
  265. Leung, L.-S.B.; Neal, J.W.; Wakelee, H.A.; Sequist, L.V.; Marmor, M.F. Rapid Onset of Retinal Toxicity from High-Dose Hydroxychloroquine Given for Cancer Therapy. Am. J. Ophthalmol. 2015, 160, 799–805.e1. [Google Scholar] [CrossRef]
  266. Carmichael, S.J.; Charles, B.; Tett, S.E. Population Pharmacokinetics of Hydroxychloroquine in Patients with Rheumatoid Arthritis. Ther. Drug Monit. 2003, 25, 671–681. [Google Scholar] [CrossRef] [PubMed]
  267. Espina, V.A.; Liotta, L.; Rassulova, S.; Gallimore, H.; Grant-Wisdom, T.; Menezes, G.; Nayer, H.; Edmiston, K. Abstract CT140: PINC trial: Preventing invasive breast neoplasia with chloroquine. Cancer Res. 2017, 77, CT140. [Google Scholar] [CrossRef]
  268. Qiu, X.; Mei, J.; Yin, J.; Wang, H.; Wang, J.; Xie, M. Correlation analysis between expression of PCNA, Ki-67 and COX-2 and X-ray features in mammography in breast cancer. Oncol. Lett. 2017, 14, 2912–2918. [Google Scholar] [CrossRef] [Green Version]
  269. Anand, K.; Niravath, P.; Patel, T.; Ensor, J.; Rodriguez, A.; Boone, T.; Wong, S.T.; Chang, J.C. A Phase II Study of the Efficacy and Safety of Chloroquine in Combination with Taxanes in the Treatment of Patients With Advanced or Metastatic Anthracycline-refractory Breast Cancer. Clin. Breast Cancer 2021, 21, 199–204. [Google Scholar] [CrossRef]
  270. Pareek, A.; Suthar, M.; Rathore, G.S.; Bansal, V. Feverfew (Tanacetum parthenium L.): A systematic review. Pharmacogn. Rev. 2011, 5, 103–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  271. Zhang, S.; Ong, C.-N.; Shen, H.-M. Critical roles of intracellular thiols and calcium in parthenolide-induced apoptosis in human colorectal cancer cells. Cancer Lett. 2004, 208, 143–153. [Google Scholar] [CrossRef] [PubMed]
  272. Kim, J.-H.; Liu, L.; Lee, S.-O.; Kim, Y.-T.; You, K.-R.; Kim, D.-G. Susceptibility of Cholangiocarcinoma Cells to Parthenolide-Induced Apoptosis. Cancer Res. 2005, 65, 6312–6320. [Google Scholar] [CrossRef] [Green Version]
  273. Liu, J.-W.; Cai, M.-X.; Xin, Y.; Wu, Q.-S.; Ma, J.; Yang, P.; Xie, H.-Y.; Huang, D.-S. Parthenolide induces proliferation inhibition and apoptosis of pancreatic cancer cells in vitro. J. Exp. Clin. Cancer Res. 2010, 29, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Wen, J.; You, K.R.; Lee, S.Y.; Song, C.H.; Kim, D.G. Oxidative stress-mediated apoptosis. The anticancer effect of the sesquiterpene lactone parthenolide. J. Biol. Chem. 2002, 277, 38954–38964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Guzman, M.L.; Rossi, R.M.; Karnischky, L.; Li, X.; Peterson, D.R.; Howard, D.S.; Jordan, C.T. The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cells. Blood 2005, 105, 4163–4169. [Google Scholar] [CrossRef] [PubMed]
  276. Yip-Schneider, M.T.; Nakshatri, H.; Sweeney, C.J.; Marshall, M.S.; Wiebke, E.A.; Schmidt, C.M. Parthenolide and sulindac cooperate to mediate growth suppression and inhibit the nuclear factor-κB pathway in pancreatic carcinoma cells. Mol. Cancer Ther. 2005, 4, 587–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Wang, W.; Adachi, M.; Kawamura, R.; Sakamoto, H.; Hayashi, T.; Ishida, T.; Imai, K.; Shinomura, Y. Parthenolide-induced apoptosis in multiple myeloma cells involves reactive oxygen species generation and cell sensitivity depends on catalase activity. Apoptosis 2006, 11, 2225–2235. [Google Scholar] [CrossRef]
  278. Zunino, S.J.; Ducore, J.M.; Storms, D.H. Parthenolide induces significant apoptosis and production of reactive oxygen species in high-risk pre-B leukemia cells. Cancer Lett. 2007, 254, 119–127. [Google Scholar] [CrossRef]
  279. Lu, C.; Wang, W.; Jia, Y.; Liu, X.; Tong, Z.; Li, B. Inhibition of AMPK/Autophagy Potentiates Parthenolide-Induced Apoptosis in Human Breast Cancer Cells. J. Cell. Biochem. 2014, 115, 1458–1466. [Google Scholar] [CrossRef]
  280. Luo, L.; Wang, J.N.; Kong, L.D.; Jiang, Q.G.; Tan, R.X. Antidepressant effects of Banxia Houpu decoction, a traditional Chinese medicinal empirical formula. J. Ethnopharmacol. 2000, 73, 277–281. [Google Scholar] [CrossRef]
  281. Lee, Y.-J.; Lee, Y.M.; Lee, C.-K.; Jung, J.K.; Han, S.B.; Hong, J.T. Therapeutic applications of compounds in the Magnolia family. Pharmacol. Ther. 2011, 130, 157–176. [Google Scholar] [CrossRef]
  282. Liu, H.; Zang, C.; Emde, A.; Planas-Silva, M.D.; Rosche, M.; Kühnl, A.; Schulz, C.-O.; Elstner, E.; Possinger, K.; Eucker, J. Anti-tumor effect of honokiol alone and in combination with other anti-cancer agents in breast cancer. Eur. J. Pharmacol. 2008, 591, 43–51. [Google Scholar] [CrossRef]
  283. Wolf, I.; O’Kelly, J.; Wakimoto, N.; Nguyen, A.; Amblard, F.; Karlan, B.Y.; Arbiser, J.L.; Koeffler, H.P. Honokiol, a natural biphenyl, inhibits in vitro and in vivo growth of breast cancer through induction of apoptosis and cell cycle arrest. Int. J. Oncol. 2007, 30, 1529–1537. [Google Scholar] [CrossRef] [PubMed]
  284. Muniraj, N.; Siddharth, S.; Shriver, M.; Nagalingam, A.; Parida, S.; Woo, J.; Elsey, J.; Gabrielson, K.; Gabrielson, E.; Arbiser, J.L.; et al. Induction of STK11-dependent cytoprotective autophagy in breast cancer cells upon honokiol treatment. Cell Death Discov. 2020, 6, 81. [Google Scholar] [CrossRef]
  285. Singh, N.; Bhalla, M.; De Jager, P.; Gilca, M. An Overview on Ashwagandha: A Rasayana (Rejuvenator) of Ayurveda. Afr. J. Tradit. Complement. Altern. Med. 2011, 8, 208–213. [Google Scholar] [CrossRef] [PubMed]
  286. Vanden Berghe, W.; Sabbe, L.; Kaileh, M.; Haegeman, G.; Heyninck, K. Molecular insight in the multifunctional activities of Withaferin A. Biochem. Pharmacol. 2012, 84, 1282–1291. [Google Scholar] [CrossRef] [PubMed]
  287. Vyas, A.R.; Singh, S.V. Molecular Targets and Mechanisms of Cancer Prevention and Treatment by Withaferin A, A Naturally Occurring Steroidal Lactone. AAPS J. 2014, 16, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Ghosh, K.; De, S.; Mukherjee, S.; Das, S.; Ghosh, A.N.; Sengupta, S. Withaferin A induced impaired autophagy and unfolded protein response in human breast cancer cell-lines MCF-7 and MDA-MB-231. Toxicol. Vitr. 2017, 44, 330–338. [Google Scholar] [CrossRef]
  289. Muniraj, N.; Siddharth, S.; Nagalingam, A.; Walker, A.; Woo, J.; Győrffy, B.; Gabrielson, E.; Saxena, N.K.; Sharma, D. Withaferin A inhibits lysosomal activity to block autophagic flux and induces apoptosis via energetic impairment in breast cancer cells. Carcinogenesis 2019, 40, 1110–1120. [Google Scholar] [CrossRef]
  290. Button, R.W.; Roberts, S.L.; Willis, T.L.; Hanemann, C.O.; Luo, S. Accumulation of autophagosomes confers cytotoxicity. J. Biol. Chem. 2017, 292, 13599–13614. [Google Scholar] [CrossRef] [Green Version]
  291. He, Y.; Wang, J.; Liu, X.; Zhang, L.; Yi, G.; Li, C.; He, X.; Wang, P.; Jiang, H. Toosendanin Inhibits Hepatocellular Carcinoma Cells by Inducing Mitochondria-dependent Apoptosis. Planta Med. 2010, 76, 1447–1453. [Google Scholar] [CrossRef] [Green Version]
  292. Shi, Y.-L.; Li, M.-F. Biological effects of toosendanin, a triterpenoid extracted from Chinese traditional medicine. Prog. Neurobiol. 2007, 82, 1–10. [Google Scholar] [CrossRef]
  293. Kai, W.; Yating, S.; Lin, M.; Kaiyong, Y.; Baojin, H.; Wu, Y.; Fangzhou, Y.; Yan, C. Natural product toosendanin reverses the resistance of human breast cancer cells to adriamycin as a novel PI3K inhibitor. Biochem. Pharmacol. 2018, 152, 153–164. [Google Scholar] [CrossRef] [PubMed]
  294. Zhang, S.; Dong, Y.; Chen, X.; Tan, C.S.H.; Li, M.; Miao, K.; Lu, J.-H. Toosendanin, a late-stage autophagy inhibitor, sensitizes triple-negative breast cancer to irinotecan chemotherapy. Chin. Med. 2022, 17, 55. [Google Scholar] [CrossRef] [PubMed]
  295. Nuta, G.C.; Gilad, Y.; Goldberg, N.; Meril, S.; Bahlsen, M.; Carvalho, S.; Kozer, N.; Barr, H.; Fridmann Sirkis, Y.; Hercík, K.; et al. Identifying a selective inhibitor of autophagy that targets ATG12-ATG3 protein-protein interaction. Autophagy 2023, 1–14. [Google Scholar] [CrossRef]
  296. Cui, J.; Ogasawara, Y.; Kurata, I.; Matoba, K.; Fujioka, Y.; Noda, N.N.; Shibasaki, M.; Watanabe, T. Targeting the ATG5-ATG16L1 Protein–Protein Interaction with a Hydrocarbon-Stapled Peptide Derived from ATG16L1 for Autophagy Inhibition. J. Am. Chem. Soc. 2022, 144, 17671–17679. [Google Scholar] [CrossRef]
  297. Sun, Q.; Hou, X.; Yang, J.; Zhang, M.; Yang, Y.; Liu, Y.; Shen, W.; Yin, D. Heparin-Coated Photosensitive Metal–Organic Frameworks as Drug Delivery Nanoplatforms of Autophagy Inhibitors for Sensitized Photodynamic Therapy against Breast Cancer. ACS Appl. Mater. Interfaces 2021, 13, 55577–55590. [Google Scholar] [CrossRef]
  298. Qian, R.; Cao, G.; Su, W.; Zhang, J.; Jiang, Y.; Song, H.; Jia, F.; Wang, H. Enhanced Sensitivity of Tumor Cells to Autophagy Inhibitors Using Fasting-Mimicking Diet and Targeted Lysosomal Delivery Nanoplatform. Nano Lett. 2022, 22, 9154–9162. [Google Scholar] [CrossRef]
Figure 1. Cytoprotective autophagy is activated in cancer cells resistant to endocrine therapy (ATP = adenosine triphosphate; AKT = protein kinase B; AMP = adenosine monophosphate; AMPK = AMP-activated protein kinase; BECN1 = Beclin-1; DNMT1 = DNA-methyltransferase 1; EGFR = epidermal growth factor receptor; ERK = extracellular signal-regulated kinase; GRP78 = glucose-regulated protein 78; GPR30 = G protein coupled estrogen receptor; H19 = long noncoding RNA H19; HMGB1 = high mobility group box 1; IGFBP-3 = insulin-like growth factor binding protein 3; JAK = Janus kinase; JNK = c-Jun N-terminal kinase; MAPK = mitogen-activated protein kinase; MEK = mitogen-activated protein kinase kinase; miR-214 = microRNA 214; miR-214-3p = microRNA 214-3p; MTA1 = metastasis-associated antigen 1; mTOR = mammalian target of rapamycin; NCK = non-catalytic region of tyrosine kinase; PAG = glycosphingolipid-enriched microdomains; PAK = p-21-activated kinase; PI3K = phosphoinositide 3-kinase; PKC = protein kinase C; PLC = phosphoinositide-specific phospholipase C; SLC6A14 (solute carrier family 6 member 14); STAT = signal transducer and activators of transcription; TAM = tamoxifen; UCP2 = mitochondrial uncoupling protein 2; ULK1 = Unc-51 like autophagy activating kinase 1; UPR = unfolded protein response).
Figure 1. Cytoprotective autophagy is activated in cancer cells resistant to endocrine therapy (ATP = adenosine triphosphate; AKT = protein kinase B; AMP = adenosine monophosphate; AMPK = AMP-activated protein kinase; BECN1 = Beclin-1; DNMT1 = DNA-methyltransferase 1; EGFR = epidermal growth factor receptor; ERK = extracellular signal-regulated kinase; GRP78 = glucose-regulated protein 78; GPR30 = G protein coupled estrogen receptor; H19 = long noncoding RNA H19; HMGB1 = high mobility group box 1; IGFBP-3 = insulin-like growth factor binding protein 3; JAK = Janus kinase; JNK = c-Jun N-terminal kinase; MAPK = mitogen-activated protein kinase; MEK = mitogen-activated protein kinase kinase; miR-214 = microRNA 214; miR-214-3p = microRNA 214-3p; MTA1 = metastasis-associated antigen 1; mTOR = mammalian target of rapamycin; NCK = non-catalytic region of tyrosine kinase; PAG = glycosphingolipid-enriched microdomains; PAK = p-21-activated kinase; PI3K = phosphoinositide 3-kinase; PKC = protein kinase C; PLC = phosphoinositide-specific phospholipase C; SLC6A14 (solute carrier family 6 member 14); STAT = signal transducer and activators of transcription; TAM = tamoxifen; UCP2 = mitochondrial uncoupling protein 2; ULK1 = Unc-51 like autophagy activating kinase 1; UPR = unfolded protein response).
Cells 12 01156 g001
Figure 2. In cancer cells resistant to targeted therapies, cytoprotective autophagy is promoted (AMPK = AMP-activated protein kinase; ATG5 = autophagy-related gene-5; CCND1 = cyclin D1; CDK4/6 = cyclin-dependent kinase 4 and 6; EGFR = epidermal growth factor receptor; miR-567 = microRNA 567; mTOR = mammalian target of rapamycin; ULK1 = Unc-51 like autophagy activating kinase 1; PTBP1 = polypyrimidine tract binding protein 1; ZNF649-AS1 = ZNF649 Antisense RNA 1).
Figure 2. In cancer cells resistant to targeted therapies, cytoprotective autophagy is promoted (AMPK = AMP-activated protein kinase; ATG5 = autophagy-related gene-5; CCND1 = cyclin D1; CDK4/6 = cyclin-dependent kinase 4 and 6; EGFR = epidermal growth factor receptor; miR-567 = microRNA 567; mTOR = mammalian target of rapamycin; ULK1 = Unc-51 like autophagy activating kinase 1; PTBP1 = polypyrimidine tract binding protein 1; ZNF649-AS1 = ZNF649 Antisense RNA 1).
Cells 12 01156 g002
Figure 3. Cytoprotective autophagy is activated in cancer cells resistant to radiotherapy (AKT = protein kinase B; DRAM1 = damaged-regulated autophagy modulator 1; miR-200C = microRNA 200c; miR-26b = microRNA 26b; mTOR = mammalian target of rapamycin; PI3K = phosphoinositide 3-kinase; Ubqln1 = Ubiquilin-1).
Figure 3. Cytoprotective autophagy is activated in cancer cells resistant to radiotherapy (AKT = protein kinase B; DRAM1 = damaged-regulated autophagy modulator 1; miR-200C = microRNA 200c; miR-26b = microRNA 26b; mTOR = mammalian target of rapamycin; PI3K = phosphoinositide 3-kinase; Ubqln1 = Ubiquilin-1).
Cells 12 01156 g003
Figure 4. Cytoprotective autophagy is activated in chemoresistant cancer cells (AKT = protein kinase B; BECN1 = Beclin-1; ATG5 = autophagy-related gene-5; EGFR = epidermal growth factor receptor; ERK = extracellular signal-regulated kinase; HMGB1 = high mobility group box 1; MAPK = mitogen-activated protein kinase; MEK = mitogen-activated protein kinase kinase; miR-18a = microRNA 18a; mTOR = mammalian target of rapamycin; PI3K = phosphoinositide 3-kinase).
Figure 4. Cytoprotective autophagy is activated in chemoresistant cancer cells (AKT = protein kinase B; BECN1 = Beclin-1; ATG5 = autophagy-related gene-5; EGFR = epidermal growth factor receptor; ERK = extracellular signal-regulated kinase; HMGB1 = high mobility group box 1; MAPK = mitogen-activated protein kinase; MEK = mitogen-activated protein kinase kinase; miR-18a = microRNA 18a; mTOR = mammalian target of rapamycin; PI3K = phosphoinositide 3-kinase).
Cells 12 01156 g004
Figure 5. Roles of autophagy in cancer cells resistant to immunotherapy. Besides the activation of cytoprotective autophagy, cancer cells’ acquired immunotherapy resistance inhibits degradatory autophagy, thus increasing the amount of surface PD-L1 to interfere with the efficacy of immunotherapy. (HMGB1 = high mobility group box 1; PD-1 = programmed cell death protein 1; PD-L1 = programmed death-ligand 1; S1R = Sigma 1 receptor).
Figure 5. Roles of autophagy in cancer cells resistant to immunotherapy. Besides the activation of cytoprotective autophagy, cancer cells’ acquired immunotherapy resistance inhibits degradatory autophagy, thus increasing the amount of surface PD-L1 to interfere with the efficacy of immunotherapy. (HMGB1 = high mobility group box 1; PD-1 = programmed cell death protein 1; PD-L1 = programmed death-ligand 1; S1R = Sigma 1 receptor).
Cells 12 01156 g005
Table 1. List of all clinical trials of autophagy inhibitors in breast cancer.
Table 1. List of all clinical trials of autophagy inhibitors in breast cancer.
Phase Clinical TrialTreatmentIdentifier
Unknown or Terminated or Withdrawn or Completed
2 Autophagy Inhibition Using Hydrochloroquine in Breast Cancer Patients: a Pilot Study Hydroxychloroquine NCT01292408
(Unknown status)
2012
1/2 Phase I/II Study of Ixabepilone in Combination With the Autophagy Inhibitor Hydroxychloroquine for the Treatment of Patients with Metastatic Breast Cancer Ixabepilone and Hydroxychloroquine NCT00765765
(Terminated)
2013
2 Phase Ib/II Study of Hydroxychloroquine in Metastatic ER-Positive Breast Cancer Progressing on Hormonal Therapy Hydroxychloroquine in combination with hormonal therapy NCT02414776
(Terminated)
2015
1/2 Preventing Invasive Breast Neoplasia with Chloroquine (PINC) Trial High or low dose of Chloroquine NCT01023477
(Completed)
2016
2 A Phase 2 Randomized, Double-blind, Window of Opportunity Trial Evaluating Trial Clinical and Correlative Effects of Chloroquine as a Novel Therapeutic Strategy in Breast Cancer Chloroquine and placebo NCT02333890
(Unknown)
2016
1/2 A Phase Ib/II Trial of Gedatolisib, Hydroxychloroquine or the Combination for Prevention of Recurrent Breast Cancer (“GLACIER”) Gedatolisib or Hydroxychloroquine alone or in combination NCT03400254
(Withdrawn)
2020
2 Phase II Study of The Efficacy and Safety of Chloroquine (C) in CombinAtion With Taxane or taxane-like (T) Chemo Agents in The Treatment of Patients With Advanced or Metastatic Breast Cancer Who Have Failed Anthracycline Chemo Base Therapy Chloroquine with taxane or taxane-like chemotherapy NCT01446016
(Completed)
2022
Recruiting or Active
1 Hydroxychloroquine (HCQ) in Combination with Abemaciclib and Endocrine Therapy in HR+/Her2- Advanced Breast Cancer After a Lead in Dose Escalation Cohort of HCQ and Abemaciclib in Advanced Solid Tumors Abemaciclib with alternative dose of HCQ or Abemaciclib with both HCQ and endocrine therapy NCT04316169
(Recruiting)
2028
2 A Phase II Pilot Trial of ABmacocliB or Abemaciclin and HydroxYchloroquine to Target Minimal Residual Disease in Brease Cancer Patients Abemaciclib with or without Hydroxychloroquine NCT04523857
(Recruiting)
2028
2 A Phase II Trial of Avelumab or Hydrochloroquine With or Without Palbociclib to Eliminate Dormant Breast Cancer (PALAVY) HCQ alone, or Avelumab alone, or Avelumab with Palbociclib, or HCQ with Palbociclib NCT04841148
(Recruiting)
2028
1/2 Phase I/II Safety and Efficacy Study of Autophagy Inhibition With Hydroxychloroquine to Augment the Antiproliferative and Biological Effects of Pre-Operative Palbociclib Plus Letrozole for Estrogen Receptor-Positive and HER2-Negative Breast Cancer Hydroxycholoroquine, Letrozole, and Palbociclib NCT03774472
(Active)
2024
2 CLEVER Pilot Trial: A Phase II Pilot Trial of HydroxyChLoroquine, EVErolimus or the Combination for Prevention of Recurrent Breast Cancer Hydroxychloroquine alone, or Everolimus alone, or the combination of hydroxychloroquine and Everolimus NCT03032406
(Recruiting)
2025
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wu, Q.; Sharma, D. Autophagy and Breast Cancer: Connected in Growth, Progression, and Therapy. Cells 2023, 12, 1156. https://doi.org/10.3390/cells12081156

AMA Style

Wu Q, Sharma D. Autophagy and Breast Cancer: Connected in Growth, Progression, and Therapy. Cells. 2023; 12(8):1156. https://doi.org/10.3390/cells12081156

Chicago/Turabian Style

Wu, Qitong, and Dipali Sharma. 2023. "Autophagy and Breast Cancer: Connected in Growth, Progression, and Therapy" Cells 12, no. 8: 1156. https://doi.org/10.3390/cells12081156

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop