Next Article in Journal
The Degree of Branching of Serum IgG N-glycans as a Marker of Advanced Endometriosis
Next Article in Special Issue
Effects of Biguanide-PROTACs in Pancreatic Cancer Cells
Previous Article in Journal
Practical Remediation of Hg-Contaminated Groundwater by MoS2: Batch and Column Tests
Previous Article in Special Issue
Thiazolidinedione-Conjugated Lupeol Derivatives as Potent Anticancer Agents Through a Mitochondria-Mediated Apoptotic Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Cancer Strategy Based on Changes in the Role of Autophagy Depending on the Survival Environment and Tumorigenesis Stages

1
Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
2
Institute for New Drug Development, Incheon National University, Incheon 22012, Republic of Korea
*
Author to whom correspondence should be addressed.
Molecules 2024, 29(21), 5134; https://doi.org/10.3390/molecules29215134
Submission received: 2 October 2024 / Revised: 21 October 2024 / Accepted: 29 October 2024 / Published: 30 October 2024
(This article belongs to the Special Issue Anticancer Drug Discovery and Development II)

Abstract

:
Autophagy is a crucial mechanism for recycling intracellular materials, and under normal metabolic conditions, it is maintained at low levels in cells. However, when nutrients are deficient or under hypoxic conditions, the level of autophagy significantly increases. Particularly in cancer cells, which grow more rapidly than normal cells and tend to grow in a three-dimensional manner, cells inside the cell mass often face limited oxygen supply, leading to inherently higher levels of autophagy. Therefore, the initial development of anticancer drugs targeting autophagy was based on a strategy to suppress these high levels of autophagy. However, anticancer drugs that inhibit autophagy have not shown promising results in clinical trials, as it has been revealed that autophagy does not always play a role that favors cancer cell survival. Hence, this review aims to suggest anticancer strategies based on the changes in the role of autophagy according to survival conditions and tumorigenesis stage.

1. Introduction

Autophagy is a cellular process in which intracellular materials are delivered to the lysosome for degradation [1], and refers to the concept that cells can survive through the recycling of materials under nutrient-deprived conditions. Yoshinori Ohsumi, awarded the Nobel Prize in Physiology or Medicine in 2016 for his discovery of autophagy mechanisms, identified several autophagy-related genes that are known as APG through yeast genetic screening [2]. Since autophagy plays a crucial role in degrading unnecessary proteins and cellular organelles, its dysregulation is associated with several diseases, including neurodegenerative disorders [3,4,5], as well as malignant tumors such as those of the liver, colon, stomach, and breast [6]. In particular, there has been significant research on the role of autophagy in tumorigenesis, with many studies reporting the development of liver tumors in autophagy-deficient mouse models [7,8,9]. Our laboratory has also confirmed that the loss of ATG5 induces cellular transformation [10].
It has been reported that a type of autophagy called alternative autophagy exists, which shares similarities with canonical autophagy but operates independently of ATG5/ATG7 [11,12]. Although the exact role and mechanism of alternative autophagy remain unclear, there is a key difference in the membrane source. Canonical autophagy originates from the endoplasmic reticulum (ER) and mitochondria-associated ER membranes, whereas alternative autophagy originates from the trans-Golgi membrane [11]. In addition, in canonical autophagy, ATG5, ATG7, and LC3 are involved in autophagosome formation. However, in alternative autophagy, the Rab9 protein, which participates in late endosome-trans-Golgi transport, attaches to the autophagic membrane, leading to the generation of autophagosomes and autolysosomes [13,14]. Studies have shown that alternative autophagy plays an important role in the pathophysiological processes of various diseases, including cardiac diseases [15], neurodegenerative diseases [14], and tumorigenesis [16,17,18]. Notably, alternative autophagy has been reported to be primarily involved in mitochondrial clearance [19]. Alternative autophagy appears to be induced only under cellular stress conditions rather than under basal conditions. Our laboratory has reported that the higher viability of Ras-transformed cells compared to that of normal cell lines during long-term culture is due to relatively higher levels of alternative autophagy [20,21]. Furthermore, when treated with paclitaxel, a natural anticancer drug commonly used to treat several types of cancer, Ras-transformed cells showed a significant reduction in alternative autophagy and increased cell death, suggesting that alternative autophagy plays a crucial role in the survival of Ras-transformed cells [21].
Increasing evidence has linked autophagy closely to drug resistance and immune evasion in tumor cells [22]. Canonical autophagy has been demonstrated to function at each maturation stage of dendritic cells, including antigen presentation and cytokine secretion [23]. Additionally, alternative autophagy has also been shown to take over the role of canonical autophagy in regulating bacterial phagocytosis, as well as cell differentiation and maturation [24].
Autophagy is divided into four major steps: initiation, nucleation, elongation, and fusion/degradation (Figure 1). Any protein involved in these four steps could be a target for anticancer drugs. In the initiation step, signaling proteins respond to external factors such as stress stimuli. When the cellular energy levels are low, AMPK directly activates ULK1 to induce autophagy, whereas the type I PI3K/AKT pathway inhibits autophagy via the mTOR pathway. In the nucleation step, BECN1 acts as a platform to form a core complex with proteins, such as VPS34 (type III PI3K), AMBRA, and ATG14L. Two ubiquitin-like conjugation systems are involved in the elongation step. ATG12 plays an important regulatory role in initial autophagosome formation by forming a complex with ATG5 and interacting with ATG16L to form a larger multiprotein complex. LC3 binds to the lipid PE through the ATG12–ATG5 conjugate to generate LC3-II, a key marker of autophagy. Once the autophagosome is fully formed, it fuses with lysosomes to create autolysosomes, a process that requires SNARE proteins, such as STX17, SNAP29, and VAMP8. To date, several autophagy regulators have been approved by the U.S. Food and Drug Administration (FDA) for cancer treatment and many others are currently under research and development [25,26,27,28,29,30].

2. The Role of Autophagy in Different Stages of Cellular Transformation

As the impact of autophagy on tumor formation and development has been increasingly recognized, strategies targeting autophagy have been continuously explored as efficient approaches for cancer therapy. Since autophagy is a mechanism that continuously degrades proteins and organelles to maintain normal function and protect against adverse stress, it was initially considered that autophagy inhibition might be effective in treating cancer [31]. However, it has been revealed that depending on the type and developmental stage of the tumor, autophagy activation or inhibition can contribute differently to tumor formation [31,32,33,34,35]. In particular, the role of autophagy varies according to the stage of tumorigenesis [36]. In normal cells, autophagy has a tumor-suppressive effect, and its absence may exacerbate the early stages of tumor development [10]. In RAS-induced cancers, autophagy blocks further progression to malignancy [37]. According to Sun et al. [38], autophagy suppresses liver cancer formation during the dysplastic stage but promotes it during the tumor-forming stage.

2.1. The Tumor-Suppressive Role of Autophagy

Many reports have indicated that autophagy prevents cancer development in the early stages of tumor formation [31,32,39,40]. Autophagy is a highly adaptive process for maintaining cellular homeostasis and preventing tumorigenesis in response to various forms of cellular stress [41,42,43,44]. Mitochondrial damage leads to the accumulation of reactive oxygen species (ROS), which can cause DNA damage and promote tumorigenesis [45,46,47,48]. Autophagy helps prevent ROS accumulation, which can cause DNA damage associated with cancer progression by removing damaged mitochondria [6,49,50]. Additionally, Beclin-1, an essential mediator of autophagy, acts as a tumor suppressor, protecting cells from unfavorable stimuli and stress in early cancer stages [51,52]. In fact, in various tumors, such as breast and prostate cancers, monoallelic deletion of Beclin-1 and inactivation of autophagy have been observed [53,54,55,56]. Furthermore, decreased expression of autophagy-related genes such as ATG5, ATG7, and Beclin-1 has been observed in hepatocellular carcinoma (HCC) cells [57], and our laboratory has confirmed that autophagy deficiency due to ATG5 knockout can induce malignant cellular transformation [10].

2.2. The Tumor-Promoting Role of Autophagy

Autophagy is a survival mechanism that occurs in response to stress. In established tumors, autophagy provides an alternative route to supply essential nutrients, supporting the increased metabolic activity associated with explosive cell growth and thereby meeting the energy demands of cancer cells [58,59,60]. Autophagy has also been shown to promote immune evasion in tumor cells [61,62]. Furthermore, autophagy upregulation may induce resistance to cancer therapy [26]. Using a mouse model of cancer induced by oncogenic Ras, Ying et al. [63] demonstrated that autophagy is required for tumor development. In our laboratory, we have shown that Ras-transformed cells maintain higher levels of basal autophagy than parental cells [20,64].

3. Changes in Autophagy Levels Based on the Survival Environment of Transformed Cells

When the tumor microenvironment is subjected to stress conditions such as starvation, hypoxia, and ROS accumulation, tumor cells induce autophagy to maintain metabolic homeostasis [65,66]. In highly proliferative cancer cells, the increased metabolic demands and poor vascularization of solid tumors often lead to hypoxic conditions in the tumor microenvironment. As a result, adaptive metabolic responses are promoted because cells cannot meet the high demands for amino acids, oxygen, and growth factors [67,68,69]. The relationship between hypoxia and autophagy as a survival mechanism has been demonstrated in various cancer and normal cells [70]. However, our recent laboratory studies have shown that increased cell density due to long-term culture results in a decrease in canonical autophagy and a relatively greater role of alternative autophagy in transformed cells [20]. Furthermore, the importance of mitochondrial clearance in tumor suppression is evidenced by the accumulation of damaged mitochondria and the subsequent accumulation of ROS and DNA damage in cells with the deletion of key autophagy genes [71]. Our laboratory observed that Ras-transformed cell lines generated higher ROS levels than normal cell lines [21]. In particular, under unfavorable culture conditions due to long-term culture, parental NIH 3T3 cells continuously reduced their mitochondrial mass, whereas Ras-transformed NIH 3T3 cells maintained a high mitochondrial mass. Recent studies confirmed the functional significance of alternative autophagy in the removal of damaged mitochondria [19]. Therefore, while tumor cells generally rely on increased canonical autophagy to supply nutrients, such as metabolites, the role of alternative autophagy in removing damaged mitochondria may become more critical in unfavorable survival conditions.

4. The Relationship Between Alternative Autophagy and Cancer

Nishida et al. [11] reported the existence of Rab9 protein-dependent alternative autophagy, which is involved in transport between endosomes and the trans-Golgi network, even in the absence of major ATG factors during etoposide-induced autophagy. Subsequently, it was found that almost all cells possess both canonical and alternative autophagy mechanisms [72] and that different types of autophagy can be induced depending on the type of substrate or cellular stress. For instance, starvation primarily induces canonical autophagy, whereas genotoxic stress activates both types of autophagy [11]. Alternative autophagy has been reported to be associated with age-related diseases such as aging and neurodegenerative disorders; however, it has also been noted that tumor cells have adapted to utilize non-canonical alternative autophagy pathways to support tumor growth and progression [73]. To meet the increased metabolic demand of established tumor cells, inhibition of hyperactivated autophagy tends to promote alternative autophagy [74]. Recently, our laboratory showed that Ras-transformed cells exhibited reduced canonical autophagy upon long-term culture, resulting in increased resistance to the autophagy inhibitor chloroquine [20]. This suggests that as tumor cell confluency increases, canonical autophagy may no longer be essential for tumor cell survival. Furthermore, in Ras-transformed cells, increasing cell density enhances alternative autophagy and increases resistance to the chemotherapeutic drug paclitaxel [21,75]. In addition, inhibition of alternative autophagy with Brefeldin A or knockdown of Rab9 significantly reduced the viability of Ras-NIH 3T3 cells. These results suggest that alternative autophagy is more important than canonical autophagy in maintaining cell survival under adverse conditions such as high cell density and anticancer drug exposure [21]. Therefore, the physiological relevance and molecular mechanisms mediating alternative autophagy in cancer may be attractive therapeutic targets, alongside canonical autophagy.
In fact, substances specifically regulating alternative autophagy have not yet been reported. However, anticancer drugs such as etoposide [76], camptothecin [77], and staurosporine [78] are known to activate alternative autophagy. Meanwhile, canonical autophagy modulators such as bafilomycin A1 [11], 3-methyladenine [11], wortmannin [78], and chloroquine [79] have also been reported to inhibit alternative autophagy. Brefeldin A [11], a widely used selective inhibitor of alternative autophagy, is known to increase the cytotoxicity of imatinib, a targeted cancer drug for chronic myeloid leukemia [80]; however, the relationship between this mechanism of action and the inhibition of alternative autophagy has not been reported. Consequently, to elucidate the role of alternative autophagy in tumorigenesis, it is necessary to develop new autophagy modulators that selectively target the autophagy signaling molecules essential for the alternative autophagy pathway.

5. Current Status of Anticancer Drug Development Using Autophagy Regulators

Autophagy regulation has emerged as a promising approach for cancer treatment and has become a significant target in the field. However, the dual role of autophagy in tumor growth and development poses a major challenge for the development of anticancer drugs that target autophagy. Therefore, anticancer drugs targeting autophagy should be differentiated using inhibitors or inducers depending on the stage of tumor development (Table 1). In the excessive growth stage, cancer cells rely on autophagy to survive under unfavorable conditions, such as nutrient deprivation and hypoxia, making it an attractive therapeutic target because of its high energy requirements. Consequently, many anticancer drugs target autophagy through inhibitory strategies. Moreover, the inhibition of autophagy renders cancer cells more susceptible to the cytotoxic effects of conventional anticancer drugs, thereby improving therapeutic outcomes in a variety of cancers, including breast and ovarian cancers and melanoma [81,82,83,84]. However, given the developmental nature of tumors, they are often discovered in patients only after they have progressed beyond their initial developmental stages. Considering the dual nature of autophagy, some studies have focused on activating autophagy along with other conventional treatments to directly induce cell death. Uncontrolled and continuous activation of autophagy can ultimately lead to cell death [85]. Excessive autophagy is sometimes induced via pharmacological or genetic treatments to prompt tumor cell death. However, owing to the complexity of autophagic physiology, there is still an intense debate about whether autophagy should be activated or inhibited, and no consensus has been reached.
As shown in Table 1, anticancer drugs developed to induce autophagy often target proteins that regulate the initiation stages, particularly mTOR. On the other hand, anticancer drugs developed as autophagy inhibitors target almost every stage of autophagy, from vesicle nucleation and maturation to vesicle fusion and lysosomal degradation. In the development of autophagy inhibitors, the target protein at the initiation stage is primarily ULK1. These inhibitors function by forming complexes with the ULK1 regulatory unit and interfere with kinases in clinical settings, making ULK1 an attractive candidate for autophagy inhibition [86]. Inhibitors targeting type III PI3K (VPS34), at the nucleation stage, which is involved in autophagy and vesicle dynamics, have been developed [87,88,89]. Inhibitors that target the maturation stage mainly include autophagosome–lysosome fusion inhibitors, which inhibit lysosomal acidification and consequently disrupt autophagy [90,91]. Additionally, the potential of proteasome inhibitors to suppress autophagy has been explored as a promising therapeutic strategy for cancer [92]. Figure 2 also shows autophagy modulators targeting each step of the autophagy pathway.
Table 1. Pharmacological inducers and inhibitors targeting autophagy for anticancer therapy.
Table 1. Pharmacological inducers and inhibitors targeting autophagy for anticancer therapy.
Autophagy StageTargetDrugTarget TumorRefs.
Inducer
InitiationAkt inhibitorMK-2206Glioma[93]
Akt inhibitorPerifosineColorectal cancer[94]
AMPK activatorAICARRenal cancer[95]
AMPK activatorMetforminHepatocellular carcinoma,
colon cancer
[96,97]
ULK1 activatorLYN-1604Breast cancer[98]
PI3K/Akt/mTORGefitinibLung cancer[99]
dual class I PI3K/mTOR inhibitorNVP-BEZ235Multiple myeloma[100]
PI3K/mTOR inhibitorPI-103Acute myelogenous leukemia[101]
PI3K/mTOR inhibitorPKI-587Hepatocellular carcinoma[102]
PI3K/mTOR inhibitorNVP-BGT226Hepatocarcinoma[103]
PI3K/mTOR inhibitorOmipalisib (GSK2126458)Esophageal squamous cell carcinoma[104]
Akt/mTOR inhibitorSalvianolic acid BColorectal cancer[105]
Akt/mTOR inhibitorABTL0812Advanced solid tumor[106]
Akt/mTOR inhibitorGefitinibNon-small cell lung cancer[99]
PAK1/Akt/mTOR signaling inhibitorIpomoea batatas polysaccharides (IBPs)Lung cancer[107]
PI3K inhibitorPF-04691502Solid tumor[108]
mTOR inhibitorEverolimusAcute lymphoblastic leukemia[109]
mTOR inhibitorRidaforolimus (Deforolimus)Soft-tissue sarcoma[110]
mTOR inhibitorTemsirolimusRenal cell carcinoma[111]
mTOR inhibitorSorafenibRenal cancer, hepatocellular cancer[112]
mTOR inhibitorResveratrolOvarian cancer[113,114]
mTOR inhibitorAZD-8055Advanced solid tumor[115]
mTOR inhibitorRapamycinBreast cancer, bladder cancer[116]
mTOR inhibitorTorin 2Hepatocarcinoma[117]
mTOR inhibitorAZD8055Acute myeloid leukemia[118]
mTOR inhibitorWYE-354Colon cancer[119]
MEK1/2 inhibitorSelumetinib (AZD6244)Colorectal cancer[120]
NucleationBcl-2 inhibitorObatoclaxHead and neck squamous cell carcinoma[121]
Bcl-2 inhibitorQuercetinOvarian cancer[122]
BH3 mimeticGossypolHepatocellular carcinoma[123]
Initiation/
Nucleation
TKI inhibitorErlotinibNon-small cell lung cancer[124]
TKI inhibitorSunitinibClear cell ovarian carcinoma[125]
Microtubule-stabilizing agentsDocetaxelProstate cancer[126]
Phosphatidylinositol transfer protein alpha/betaMicrocolin HGastric cancer[127]
mAB against the folate receptor alphaMORAB-003Ovarian cancer[128]
Antiparkinsonian drugMetixeneMetastatic brain cancer[129]
Alkylating agentTemozolomideGlioblastoma multiforme[130]
Disruption of androgen receptor signalingSulforaphaneProstate cancer[131]
Proteasome inhibitorBortezomibMultiple myeloma[92]
Inhibitor
InitiationULK1 inhibitorSBI-0206965Neuroblastoma, non-small cell lung cancer[132,133]
ULK1/2 inhibitorDCC3116Lung cancer[134]
NucleationPIK3C3/Vps34 inhibitorSAR405Renal cell carcinoma[135]
Vps34 inhibitorVPS34-IN1Acute myeloid leukemia[136]
Dual inhibitor of Type I and III PI3K inhibitorWortmanninLung cancer, breast cancer[137]
ElongationAtg4 inhibitorTioconazoleColorectal cancer, breast cancer, glioma[138]
Atg4 inhibitorS130Colorectal cancer[139]
Atg4 inhibitorFMK-9a-[140]
Degradationv-ATPase inhibitorBafilomycin A1Breast cancer[141]
Phagosome–lysosome fusion inhibitorMonensinLung cancer[142]
Lysosome inhibitorHydroxychloroquineProstate cancer, melanoma [143]
Lysosomal autophagy inhibitorLys05Thyroid cancer[144]
Lysosomal autophagy inhibitorROC-325Renal cell carcinoma[145]
Multi-step inhibitor of autophagyVerteporfinOsteosarcoma[146]
USP10 and 13 inhibitorSpautin-1Chronic myeloid leukemia[147]
NEDD8 inhibitorPevonedistat(MLN4924)Mantle cell lymphoma[148]
Protein-palmitoyl thioesterase 1 (PPT1) inhibitorDQ661Melanoma[149]
Several studies have reported the role of autophagy in increasing drug resistance in tumor cells [150,151,152]. Autophagy can induce drug resistance to protect cells, posing significant clinical challenges to achieving successful cancer treatment [153]. Resistance to anticancer drugs can be overcome by the pharmacological inhibition of key components of the autophagy pathway [154]. Our laboratory has also reported that autophagy deficiency through ATG5 knockout can increase sensitivity to the anticancer drugs paclitaxel and gossypol [155,156]. Therefore, cotreatment with autophagy inhibitors and other conventional cancer therapies could aid in developing new cancer treatment strategies to overcome drug resistance [157].

6. Challenges in Developing Anticancer Drugs Targeting Autophagy

The effect of autophagy on tumors is complex and multifaceted [158,159,160]. Chronic pan-autophagy inhibition can lead to significant limitations, including toxicity to multiple organs and potential tumor formation in mouse models where key autophagy genes are inactivated [161]. Conversely, the overactivation of autophagy pathways can make certain cell types more susceptible to cell death [162]. Therefore, regulation of autophagy is a critical and complex therapeutic strategy for improving cancer treatment [163,164,165].
However, the role of alternative autophagy in tumor development has not been fully elucidated. Our laboratory has reported that under unfavorable survival conditions, mitochondrial clearance through alternative autophagy rather than canonical autophagy could be a new survival strategy for tumor cells [20]. We found that Ras-transformed cells maintained their survival by downregulating canonical autophagy at high cell confluency. This suggests that, in such cases, autophagy inhibitors may be less effective in achieving anticancer effects. Therefore, strategies to regulate alternative autophagy should be considered. Canonical autophagy and alternative autophagy proceed through different mechanisms in the initial stages but follow similar processes when forming the autolysosome. Therefore, drugs that inhibit the fusion of autophagosomes and lysosomes could potentially inhibit not only canonical autophagy, but also alternative autophagy.
Figure 3 shows a proposed model for a potential anticancer drug development strategy that considers the roles of canonical and alternative autophagy in tumorigenesis. Canonical autophagy suppresses tumor formation in the early stages of tumorigenesis but plays an important role in cell survival in the advanced stages of cancer. Therefore, it is important to use canonical autophagy inducers at the initiation stage of tumorigenesis and inhibitors at the progression stage for effective treatment. In addition, our study showed that alternative autophagy is more important than canonical autophagy in maintaining cell survival in a microenvironment that is difficult for tumor cell survival. Thus, it is important to develop an anticancer strategy that focuses on using inhibitors for alternative autophagy rather than canonical autophagy depending on the tumor microenvironment as a treatment for advanced tumors.
Another major unresolved issue in the development of drugs targeting autophagy is the lack of robust autophagy assays and biomarkers [29]. The LC3 protein, the most widely used marker in autophagy measurements, is increasingly reported to be associated with novel pathways, such as LC3-associated phagocytosis and LC3-associated endocytosis [166,167]. More importantly, LC3 accumulation cannot distinguish between the induction of autophagic flux and suppression of lysosomal activity. Therefore, current overreliance on LC3 may lead to errors in the measurement of canonical autophagy. The issue of measuring autophagic processes can confuse whether a drug developed to target autophagy acts as an inducer or an inhibitor. Moreover, it was recently discovered that LC3 lipidation does not occur during alternative autophagy [12]. Rab9 and Syntaxin 7 appear to be more appropriate potential markers [19]. To date, the most appealing method for quantifying alternative autophagy is to measure autolysosomes, which are dually stained with antibodies against the autophagosome marker Rab9 and the lysosome marker Lamp2, using fluorescence microscopy or flow cytometry.

7. Conclusions

The role of autophagy in cancer is complex, and our current knowledge in this field is very limited. To date, the two main roles of autophagy identified in tumorigenesis stages are as follows: basal autophagy generally suppresses tumor formation by inhibiting DNA damage and genomic instability in the early stages of tumor development, and conversely, autophagy promotes tumor progression in most cancers. Therefore, it is essential to investigate whether protective and cytotoxic autophagy can be simultaneously induced within cells, and how cells balance and regulate these processes. Based on the dual role of autophagy, cancer treatment strategies that combine autophagy inhibitors and inducers have been proposed [168]. In particular, inhibition of the late stages of autophagy suggests that the cytotoxic effects of autophagy inducers may be enhanced, contributing to tumor chemotherapy [169]. Therefore, a better understanding of the role of autophagy at different stages of tumor development is crucial for developing novel and effective therapeutic strategies for cancer treatment.

Author Contributions

Writing—original draft, M.L. and H.-G.K.; investigation, M.L. and H.-G.K.; writing—review and editing, M.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Ministry of Science and ICT (MIST) (NRF-2021R1F1A1046654).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This is a review and the majority of the article’s references are cited appropriately in the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Klionsky, D.J. Autophagy revisited: A conversation with Christian de Duve. Autophagy 2008, 4, 740–743. [Google Scholar] [CrossRef] [PubMed]
  2. Tsukada, M.; Ohsumi, Y. Isolation and characterization of autophagy-defective mutants of Saccharomyces cerevisiae. FEBS Lett. 1993, 333, 169–174. [Google Scholar] [CrossRef] [PubMed]
  3. Corti, O.; Blomgren, K.; Poletti, A.; Beart, P.M. Autophagy in neurodegeneration: New insights underpinning therapy for neurological diseases. J. Neurochem. 2020, 154, 354–371. [Google Scholar] [CrossRef] [PubMed]
  4. Nguyen, T.N.; Padman, B.S.; Lazarou, M. Deciphering the Molecular Signals of PINK1/Parkin Mitophagy. Trends Cell Biol. 2016, 26, 733–744. [Google Scholar] [CrossRef] [PubMed]
  5. Wong, E.; Cuervo, A.M. Autophagy gone awry in neurodegenerative diseases. Nat. Neurosci. 2010, 13, 805–811. [Google Scholar] [CrossRef] [PubMed]
  6. Lorin, S.; Hamai, A.; Mehrpour, M.; Codogno, P. Autophagy regulation and its role in cancer. Semin. Cancer Biol. 2013, 23, 361–379. [Google Scholar] [CrossRef] [PubMed]
  7. Huda, N.; Khambu, B.; Liu, G.; Nakatsumi, H.; Yan, S.; Chen, X.; Ma, M.; Dong, Z.; Nakayama, K.I.; Yin, X.M. Senescence Connects Autophagy Deficiency to Inflammation and Tumor Progression in the Liver. Cell. Mol. Gastroenterol. Hepatol. 2022, 14, 333–355. [Google Scholar] [CrossRef]
  8. Yang, H.; Ni, H.M.; Ding, W.X. Emerging Players in Autophagy Deficiency-Induced Liver Injury and Tumorigenesis. Gene Expr. 2019, 19, 229–234. [Google Scholar] [CrossRef]
  9. Takamura, A.; Komatsu, M.; Hara, T.; Sakamoto, A.; Kishi, C.; Waguri, S.; Eishi, Y.; Hino, O.; Tanaka, K.; Mizushima, N. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 2011, 25, 795–800. [Google Scholar] [CrossRef]
  10. Hwang, S.H.; Han, B.I.; Lee, M. Knockout of ATG5 leads to malignant cell transformation and resistance to Src family kinase inhibitor PP2. J. Cell. Physiol. 2018, 233, 506–515. [Google Scholar] [CrossRef]
  11. Nishida, Y.; Arakawa, S.; Fujitani, K.; Yamaguchi, H.; Mizuta, T.; Kanaseki, T.; Komatsu, M.; Otsu, K.; Tsujimoto, Y.; Shimizu, S. Discovery of Atg5/Atg7-independent alternative macroautophagy. Nature 2009, 461, 654–658. [Google Scholar] [CrossRef] [PubMed]
  12. Shimizu, S. Biological Roles of Alternative Autophagy. Mol. Cells 2018, 41, 50–54. [Google Scholar] [PubMed]
  13. Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef] [PubMed]
  14. Honda, S.; Arakawa, S.; Yamaguchi, H.; Torii, S.; Tajima Sakurai, H.; Tsujioka, M.; Murohashi, M.; Shimizu, S. Association Between Atg5-independent Alternative Autophagy and Neurodegenerative Diseases. J. Mol. Biol. 2020, 432, 2622–2632. [Google Scholar] [CrossRef]
  15. Saito, T.; Nah, J.; Oka, S.I.; Mukai, R.; Monden, Y.; Maejima, Y.; Ikeda, Y.; Sciarretta, S.; Liu, T.; Li, H.; et al. An alternative mitophagy pathway mediated by Rab9 protects the heart against ischemia. J. Clin. Investig. 2019, 129, 802–819. [Google Scholar] [CrossRef]
  16. Torii, S.; Honda, S.; Murohashi, M.; Yamaguchi, H.; Shimizu, S. Autophagy involvement in oncogenesis. Cancer Sci. 2020, 111, 3993–3999. [Google Scholar] [CrossRef]
  17. Feng, H.; Wang, N.; Zhang, N.; Liao, H.H. Alternative autophagy: Mechanisms and roles in different diseases. Cell Commun. Signal. 2022, 20, 43. [Google Scholar] [CrossRef]
  18. Urbanska, K.; Orzechowski, A. The Secrets of Alternative Autophagy. Cells 2021, 10, 3241. [Google Scholar] [CrossRef]
  19. Ma, T.; Li, J.; Xu, Y.; Yu, C.; Xu, T.; Wang, H.; Liu, K.; Cao, N.; Nie, B.M.; Zhu, S.Y.; et al. Atg5-independent autophagy regulates mitochondrial clearance and is essential for iPSC reprogramming. Nat. Cell Biol. 2015, 17, 1379–1387. [Google Scholar] [CrossRef]
  20. Kim, H.G.; Ro, M.H.; Hwang, S.H.; Lee, M. Transformed cells maintain survival by downregulating autophagy at a high cell confluency. J. Cell. Physiol. 2023, 238, 2468–2480. [Google Scholar] [CrossRef]
  21. Kim, H.-G.; Ro, M.-H.; Lee, M. The role of alternative autophagy in cell viability and response to paclitaxel treatment in v-Ha-ras-transformed NIH 3T3 cells. Biochem. Biophys. Res. Commun. 2024, 736, 150506. [Google Scholar] [CrossRef] [PubMed]
  22. Yamazaki, T.; Bravo-San Pedro, J.M.; Galluzzi, L.; Kroemer, G.; Pietrocola, F. Autophagy in the cancer-immunity dialogue. Adv. Drug Deliv. Rev. 2020, 169, 40–50. [Google Scholar] [CrossRef] [PubMed]
  23. Germic, N.; Frangez, Z.; Yousefi, S.; Simon, H.U. Regulation of the innate immune system by autophagy: Monocytes, macrophages, dendritic cells and antigen presentation. Cell Death Differ. 2019, 26, 715–727. [Google Scholar] [CrossRef]
  24. Toh, H.; Nozawa, T.; Minowa-Nozawa, A.; Hikichi, M.; Nakajima, S.; Aikawa, C.; Nakagawa, I. Group A Streptococcus modulates RAB1- and PIK3C3 complex-dependent autophagy. Autophagy 2020, 16, 334–346. [Google Scholar] [CrossRef] [PubMed]
  25. Marinkovic, M.; Sprung, 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]
  26. Salimi-Jeda, A.; Ghabeshi, S.; Gol Mohammad Pour, Z.; Jazaeri, E.O.; Araiinejad, M.; Sheikholeslami, F.; Abdoli, M.; Edalat, M.; Abdoli, A. Autophagy Modulation and Cancer Combination Therapy: A Smart Approach in Cancer Therapy. Cancer Treat. Res. Commun. 2022, 30, 100512. [Google Scholar] [CrossRef]
  27. Mohsen, S.; Sobash, P.T.; Algwaiz, G.F.; Nasef, N.; Al-Zeidaneen, S.A.; Karim, N.A. Autophagy Agents in Clinical Trials for Cancer Therapy: A Brief Review. Curr. Oncol. 2022, 29, 1695–1708. [Google Scholar] [CrossRef]
  28. Kondapuram, S.K.; Sarvagalla, S.; Coumar, M.S. Targeting autophagy with small molecules for cancer therapy. J. Cancer Metastasis Treat. 2019, 5, 32. [Google Scholar] [CrossRef]
  29. Kocak, M.; Ezazi Erdi, S.; Jorba, G.; Maestro, I.; Farres, J.; Kirkin, V.; Martinez, A.; Pless, O. Targeting autophagy in disease: Established and new strategies. Autophagy 2022, 18, 473–495. [Google Scholar] [CrossRef]
  30. Limpert, A.S.; Lambert, L.J.; Bakas, N.A.; Bata, N.; Brun, S.N.; Shaw, R.J.; Cosford, N.D.P. Autophagy in Cancer: Regulation by Small Molecules. Trends Pharmacol. Sci. 2018, 39, 1021–1032. [Google Scholar] [CrossRef]
  31. 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]
  32. Amaravadi, R.K.; Kimmelman, A.C.; Debnath, J. Targeting Autophagy in Cancer: Recent Advances and Future Directions. Cancer Discov. 2019, 9, 1167–1181. [Google Scholar] [CrossRef] [PubMed]
  33. Kenific, C.M.; Debnath, J. Cellular and metabolic functions for autophagy in cancer cells. Trends Cell Biol. 2015, 25, 37–45. [Google Scholar] [CrossRef] [PubMed]
  34. Nassour, J.; Radford, R.; Correia, A.; Fuste, J.M.; Schoell, B.; Jauch, A.; Shaw, R.J.; Karlseder, J. Autophagic cell death restricts chromosomal instability during replicative crisis. Nature 2019, 565, 659–663. [Google Scholar] [CrossRef]
  35. Kocaturk, N.M.; Akkoc, Y.; Kig, C.; Bayraktar, O.; Gozuacik, D.; Kutlu, O. Autophagy as a molecular target for cancer treatment. Eur. J. Pharm. Sci. 2019, 134, 116–137. [Google Scholar] [CrossRef]
  36. Debnath, J.; Gammoh, N.; Ryan, K.M. Autophagy and autophagy-related pathways in cancer. Nat. Rev. Mol. Cell Biol. 2023, 24, 560–575. [Google Scholar] [CrossRef]
  37. Guo, J.Y.; Chen, H.Y.; Mathew, R.; Fan, J.; Strohecker, A.M.; Karsli-Uzunbas, G.; Kamphorst, J.J.; Chen, G.; Lemons, J.M.; Karantza, V.; et al. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 2011, 25, 460–470. [Google Scholar] [CrossRef]
  38. Sun, K.; Guo, X.L.; Zhao, Q.D.; Jing, Y.Y.; Kou, X.R.; Xie, X.Q.; Zhou, Y.; Cai, N.; Gao, L.; Zhao, X.; et al. Paradoxical role of autophagy in the dysplastic and tumor-forming stages of hepatocarcinoma development in rats. Cell Death Dis. 2013, 4, e501. [Google Scholar] [CrossRef]
  39. Levy, J.M.M.; Towers, C.G.; Thorburn, A. Targeting autophagy in cancer. Nat. Rev. Cancer 2017, 17, 528–542. [Google Scholar] [CrossRef]
  40. Russell, R.C.; Guan, K.L. The multifaceted role of autophagy in cancer. EMBO J. 2022, 41, e110031. [Google Scholar] [CrossRef]
  41. 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] [PubMed]
  42. Song, P.; Li, Y.; Dong, Y.; Liang, Y.; Qu, H.; Qi, D.; Lu, Y.; Jin, X.; Guo, Y.; Jia, Y.; et al. Estrogen receptor beta inhibits breast cancer cells migration and invasion through CLDN6-mediated autophagy. J. Exp. Clin. Cancer Res. 2019, 38, 354. [Google Scholar] [CrossRef] [PubMed]
  43. Jiang, H.; Wang, H.; Zou, W.; Hu, Y.; Chen, C.; Wang, C. Sufentanil impairs autophagic degradation and inhibits cell migration in NCI-H460 in vitro. Oncol. Lett. 2019, 18, 6829–6835. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, D.; Guo, H.; Yang, H.; Wang, D.; Gao, P.; Wei, W. Pterostilbene, An Active Constituent of Blueberries, Suppresses Proliferation Potential of Human Cholangiocarcinoma via Enhancing the Autophagic Flux. Front. Pharmacol. 2019, 10, 1238. [Google Scholar] [CrossRef]
  45. Ganesher, A.; Chaturvedi, P.; Sahai, R.; Meena, S.; Mitra, K.; Datta, D.; Panda, G. New Spisulosine Derivative promotes robust autophagic response to cancer cells. Eur. J. Med. Chem. 2020, 188, 112011. [Google Scholar] [CrossRef]
  46. Simonet, S.; Rodriguez-Lafrasse, C.; Beal, D.; Gerbaud, S.; Malesys, C.; Tillement, O.; Lux, F.; Fayyad-Kazan, H.; Rachidi, W.; Ardail, D. Gadolinium-Based Nanoparticles Can Overcome the Radioresistance of Head and Neck Squamous Cell Carcinoma Through the Induction of Autophagy. J. Biomed. Nanotechnol. 2020, 16, 111–124. [Google Scholar] [CrossRef]
  47. Zhu, Y.X.; Jia, H.R.; Gao, G.; Pan, G.Y.; Jiang, Y.W.; Li, P.; Zhou, N.; Li, C.; She, C.; Ulrich, N.W.; et al. Mitochondria-acting nanomicelles for destruction of cancer cells via excessive mitophagy/autophagy-driven lethal energy depletion and phototherapy. Biomaterials 2020, 232, 119668. [Google Scholar] [CrossRef]
  48. Booth, L.A.; Roberts, J.L.; Dent, P. The role of cell signaling in the crosstalk between autophagy and apoptosis in the regulation of tumor cell survival in response to sorafenib and neratinib. Semin. Cancer Biol. 2020, 66, 129–139. [Google Scholar] [CrossRef]
  49. Kenific, C.M.; Thorburn, A.; Debnath, J. Autophagy and metastasis: Another double-edged sword. Curr. Opin. Cell Biol. 2010, 22, 241–245. [Google Scholar] [CrossRef]
  50. Mathew, R.; Kongara, S.; Beaudoin, B.; Karp, C.M.; Bray, K.; Degenhardt, K.; Chen, G.; Jin, S.; White, E. Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev. 2007, 21, 1367–1381. [Google Scholar] [CrossRef]
  51. Ye, J.; Zhang, J.; Zhu, Y.; Wang, L.; Jiang, X.; Liu, B.; He, G. Targeting autophagy and beyond: Deconvoluting the complexity of Beclin-1 from biological function to cancer therapy. Acta Pharm. Sin. B 2023, 13, 4688–4714. [Google Scholar] [CrossRef]
  52. Burada, F.; Nicoli, E.R.; Ciurea, M.E.; Uscatu, D.C.; Ioana, M.; Gheonea, D.I. Autophagy in colorectal cancer: An important switch from physiology to pathology. World J. Gastrointest. Oncol. 2015, 7, 271–284. [Google Scholar] [CrossRef] [PubMed]
  53. Qu, X.; Yu, J.; Bhagat, G.; Furuya, N.; Hibshoosh, H.; Troxel, A.; Rosen, J.; Eskelinen, E.L.; Mizushima, N.; Ohsumi, Y.; et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Investig. 2003, 112, 1809–1820. [Google Scholar] [CrossRef] [PubMed]
  54. Yue, Z.; Jin, S.; Yang, C.; Levine, A.J.; Heintz, N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. USA 2003, 100, 15077–15082. [Google Scholar] [CrossRef] [PubMed]
  55. 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]
  56. Valente, G.; Morani, F.; Nicotra, G.; Fusco, N.; Peracchio, C.; Titone, R.; Alabiso, O.; Arisio, R.; Katsaros, D.; Benedetto, C.; et al. Expression and clinical significance of the autophagy proteins BECLIN 1 and LC3 in ovarian cancer. Biomed. Res. Int. 2014, 2014, 462658. [Google Scholar] [CrossRef]
  57. Yang, S.; Yang, L.; Li, X.; Li, B.; Li, Y.; Zhang, X.; Ma, Y.; Peng, X.; Jin, H.; Li, H. New insights into autophagy in hepatocellular carcinoma: Mechanisms and therapeutic strategies. Am. J. Cancer Res. 2019, 9, 1329–1353. [Google Scholar]
  58. 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]
  59. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12, 401–410. [Google Scholar] [CrossRef]
  60. Fujii, S.; Mitsunaga, S.; Yamazaki, M.; Hasebe, T.; Ishii, G.; Kojima, M.; Kinoshita, T.; Ueno, T.; Esumi, H.; Ochiai, A. Autophagy is activated in pancreatic cancer cells and correlates with poor patient outcome. Cancer Sci. 2008, 99, 1813–1819. [Google Scholar] [CrossRef]
  61. Young, T.M.; Reyes, C.; Pasnikowski, E.; Castanaro, C.; Wong, C.; Decker, C.E.; Chiu, J.; Song, H.; Wei, Y.; Bai, Y.; et al. Autophagy protects tumors from T cell-mediated cytotoxicity via inhibition of TNFalpha-induced apoptosis. Sci. Immunol. 2020, 5, eabb9561. [Google Scholar] [CrossRef]
  62. Lawson, K.A.; Sousa, C.M.; Zhang, X.; Kim, E.; Akthar, R.; Caumanns, J.J.; Yao, Y.; Mikolajewicz, N.; Ross, C.; Brown, K.R.; et al. Functional genomic landscape of cancer-intrinsic evasion of killing by T cells. Nature 2020, 586, 120–126. [Google Scholar] [CrossRef]
  63. Ying, H.; Dey, P.; Yao, W.; Kimmelman, A.C.; Draetta, G.F.; Maitra, A.; DePinho, R.A. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2016, 30, 355–385. [Google Scholar] [CrossRef]
  64. Hwang, S.H.; Kim, H.G.; Lee, M. Paradoxical downregulation of LPAR3 exerts tumor-promoting activity through autophagy induction in Ras-transformed cells. BMC Cancer 2022, 22, 969. [Google Scholar] [CrossRef]
  65. Wang, W.M.; Shen, H.; Liu, Z.N.; Chen, Y.Y.; Hou, L.J.; Ding, Y. Interaction between tumor microenvironment, autophagy, and epithelial-mesenchymal transition in tumor progression. Cancer Treat. Res. Commun. 2022, 32, 100592. [Google Scholar] [CrossRef]
  66. Yang, X.; Yu, D.D.; Yan, F.; Jing, Y.Y.; Han, Z.P.; Sun, K.; Liang, L.; Hou, J.; Wei, L.X. The role of autophagy induced by tumor microenvironment in different cells and stages of cancer. Cell Biosci. 2015, 5, 14. [Google Scholar] [CrossRef]
  67. Degenhardt, K.; Mathew, R.; Beaudoin, B.; Bray, K.; Anderson, D.; Chen, G.; Mukherjee, C.; Shi, Y.; Gelinas, C.; Fan, Y.; et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006, 10, 51–64. [Google Scholar] [CrossRef]
  68. Petrova, V.; Annicchiarico-Petruzzelli, M.; Melino, G.; Amelio, I. The hypoxic tumour microenvironment. Oncogenesis 2018, 7, 10. [Google Scholar] [CrossRef]
  69. Chavez-Dominguez, R.; Perez-Medina, M.; Lopez-Gonzalez, J.S.; Galicia-Velasco, M.; Aguilar-Cazares, D. The Double-Edge Sword of Autophagy in Cancer: From Tumor Suppression to Pro-tumor Activity. Front. Oncol. 2020, 10, 578418. [Google Scholar] [CrossRef]
  70. Bellot, G.; Garcia-Medina, R.; Gounon, P.; Chiche, J.; Roux, D.; Pouyssegur, J.; Mazure, N.M. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol. Cell. Biol. 2009, 29, 2570–2581. [Google Scholar] [CrossRef]
  71. Mathew, R.; White, E. Autophagy, stress, and cancer metabolism: What doesn’t kill you makes you stronger. Cold Spring Harb. Symp. Quant. Biol. 2011, 76, 389–396. [Google Scholar] [CrossRef]
  72. Yamaguchi, H.; Arakawa, S.; Kanaseki, T.; Miyatsuka, T.; Fujitani, Y.; Watada, H.; Tsujimoto, Y.; Shimizu, S. Golgi membrane-associated degradation pathway in yeast and mammals. EMBO J. 2016, 35, 1991–2007. [Google Scholar] [CrossRef]
  73. Zhang, P.; Ling, L.; Zheng, Z.; Zhang, Y.; Wang, R.; Wu, M.; Zhang, N.; Hu, M.; Yang, X. ATG7-dependent and independent autophagy determine the type of treatment in lung cancer. Pharmacol. Res. 2021, 163, 105324. [Google Scholar] [CrossRef]
  74. Kimmelman, A.C.; White, E. Autophagy and Tumor Metabolism. Cell Metab. 2017, 25, 1037–1043. [Google Scholar] [CrossRef]
  75. Kim, H.-G.; Ro, M.-H.; Lee, M. Atg5 knockout induces alternative autophagy via the downregulation of Akt expression. Toxicol. Res. 2023, 39, 637–647. [Google Scholar] [CrossRef]
  76. Arakawa, S.; Honda, S.; Yamaguchi, H.; Shimizu, S. Molecular mechanisms and physiological roles of Atg5/Atg7-independent alternative autophagy. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2017, 93, 378–385. [Google Scholar] [CrossRef]
  77. Nagata, M.; Arakawa, S.; Yamaguchi, H.; Torii, S.; Endo, H.; Tsujioka, M.; Honda, S.; Nishida, Y.; Konishi, A.; Shimizu, S. Dram1 regulates DNA damage-induced alternative autophagy. Cell Stress 2018, 2, 55–65. [Google Scholar] [CrossRef]
  78. Honda, S.; Arakawa, S.; Nishida, Y.; Yamaguchi, H.; Ishii, E.; Shimizu, S. Ulk1-mediated Atg5-independent macroautophagy mediates elimination of mitochondria from embryonic reticulocytes. Nat. Commun. 2014, 5, 4004. [Google Scholar] [CrossRef]
  79. Chen, X.; Clark, J.; Guan, J.; Kumar, A.R.; Zheng, Y. Susceptibility of AML to chloroquine therapy Is independent of autophagy. Blood 2015, 126, 1262. [Google Scholar] [CrossRef]
  80. Crowley, L.C.; O’Donovan, T.R.; Nyhan, M.J.; McKenna, S.L. Pharmacological agents with inherent anti-autophagic activity improve the cytotoxicity of imatinib. Oncol. Rep. 2013, 29, 2261–2268. [Google Scholar] [CrossRef]
  81. Klionsky, D.J.; Emr, S.D. Autophagy as a regulated pathway of cellular degradation. Science 2000, 290, 1717–1721. [Google Scholar] [CrossRef]
  82. Mizushima, N.; Levine, B. Autophagy in mammalian development and differentiation. Nat. Cell Biol. 2010, 12, 823–830. [Google Scholar] [CrossRef]
  83. White, E.; DiPaola, R.S. The double-edged sword of autophagy modulation in cancer. Clin. Cancer Res. 2009, 15, 5308–5316. [Google Scholar] [CrossRef]
  84. Amaravadi, R.K.; Yu, D.; Lum, J.J.; Bui, T.; Christophorou, M.A.; Evan, G.I.; Thomas-Tikhonenko, A.; Thompson, C.B. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J. Clin. Investig. 2007, 117, 326–336. [Google Scholar] [CrossRef]
  85. Dasari, S.K.; Bialik, S.; Levin-Zaidman, S.; Levin-Salomon, V.; Merrill, A.H., Jr.; Futerman, A.H.; Kimchi, A. Signalome-wide RNAi screen identifies GBA1 as a positive mediator of autophagic cell death. Cell Death Differ. 2017, 24, 1288–1302. [Google Scholar] [CrossRef]
  86. Martin, K.R.; Celano, S.L.; Solitro, A.R.; Gunaydin, H.; Scott, M.; O’Hagan, R.C.; Shumway, S.D.; Fuller, P.; MacKeigan, J.P. A Potent and Selective ULK1 Inhibitor Suppresses Autophagy and Sensitizes Cancer Cells to Nutrient Stress. iScience 2018, 8, 74–84. [Google Scholar] [CrossRef]
  87. Lindmo, K.; Stenmark, H. Regulation of membrane traffic by phosphoinositide 3-kinases. J. Cell Sci. 2006, 119, 605–614. [Google Scholar] [CrossRef]
  88. Lan, Y.T.; Jen-Kou, L.; Lin, C.H.; Yang, S.H.; Lin, C.C.; Wang, H.S.; Chen, W.S.; Lin, T.C.; Jiang, J.K.; Chang, S.C. Mutations in the RAS and PI3K pathways are associated with metastatic location in colorectal cancers. J. Surg. Oncol. 2015, 111, 905–910. [Google Scholar] [CrossRef]
  89. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef]
  90. Zou, Z.; Zhang, J.; Zhang, H.; Liu, H.; Li, Z.; Cheng, D.; Chen, J.; Liu, L.; Ni, M.; Zhang, Y.; et al. 3-Methyladenine can depress drug efflux transporters via blocking the PI3K-AKT-mTOR pathway thus sensitizing MDR cancer to chemotherapy. J. Drug Target. 2014, 22, 839–848. [Google Scholar] [CrossRef]
  91. Agarwal, S.; Hartz, A.M.; Elmquist, W.F.; Bauer, B. Breast cancer resistance protein and P-glycoprotein in brain cancer: Two gatekeepers team up. Curr. Pharm. Des. 2011, 17, 2793–2802. [Google Scholar] [CrossRef] [PubMed]
  92. Di Lernia, G.; Leone, P.; Solimando, A.G.; Buonavoglia, A.; Saltarella, I.; Ria, R.; Ditonno, P.; Silvestris, N.; Crudele, L.; Vacca, A.; et al. Bortezomib Treatment Modulates Autophagy in Multiple Myeloma. J. Clin. Med. 2020, 9, 552. [Google Scholar] [CrossRef] [PubMed]
  93. Cheng, Y.; Zhang, Y.; Zhang, L.; Ren, X.; Huber-Keener, K.J.; Liu, X.; Zhou, L.; Liao, J.; Keihack, H.; Yan, L.; et al. MK-2206, a novel allosteric inhibitor of Akt, synergizes with gefitinib against malignant glioma via modulating both autophagy and apoptosis. Mol. Cancer Ther. 2012, 11, 154–164. [Google Scholar] [CrossRef]
  94. Richardson, P.G.; Eng, C.; Kolesar, J.; Hideshima, T.; Anderson, K.C. Perifosine, an oral, anti-cancer agent and inhibitor of the Akt pathway: Mechanistic actions, pharmacodynamics, pharmacokinetics, and clinical activity. Expert Opin. Drug Metab. Toxicol. 2012, 8, 623–633. [Google Scholar] [CrossRef]
  95. Meley, D.; Bauvy, C.; Houben-Weerts, J.H.; Dubbelhuis, P.F.; Helmond, M.T.; Codogno, P.; Meijer, A.J. AMP-activated protein kinase and the regulation of autophagic proteolysis. J. Biol. Chem. 2006, 281, 34870–34879. [Google Scholar] [CrossRef]
  96. Ding, Z.B.; Shi, Y.H.; Zhou, J.; Qiu, S.J.; Xu, Y.; Dai, Z.; Shi, G.M.; Wang, X.Y.; Ke, A.W.; Wu, B.; et al. Association of autophagy defect with a malignant phenotype and poor prognosis of hepatocellular carcinoma. Cancer Res. 2008, 68, 9167–9175. [Google Scholar] [CrossRef]
  97. Sena, P.; Mancini, S.; Benincasa, M.; Mariani, F.; Palumbo, C.; Roncucci, L. Metformin Induces Apoptosis and Alters Cellular Responses to Oxidative Stress in Ht29 Colon Cancer Cells: Preliminary Findings. Int. J. Mol. Sci. 2018, 19, 1478. [Google Scholar] [CrossRef]
  98. Zhang, L.; Fu, L.; Zhang, S.; Zhang, J.; Zhao, Y.; Zheng, Y.; He, G.; Yang, S.; Ouyang, L.; Liu, B. Discovery of a small molecule targeting ULK1-modulated cell death of triple negative breast cancer in vitro and in vivo. Chem. Sci. 2017, 8, 2687–2701. [Google Scholar] [CrossRef]
  99. Zhao, Z.Q.; Yu, Z.Y.; Li, J.; Ouyang, X.N. Gefitinib induces lung cancer cell autophagy and apoptosis via blockade of the PI3K/AKT/mTOR pathway. Oncol. Lett. 2016, 12, 63–68. [Google Scholar] [CrossRef]
  100. Ma, Y.; Jin, Z.; Yu, K.; Liu, Q. NVP-BEZ235-induced autophagy as a potential therapeutic approach for multiple myeloma. Am. J. Transl. Res. 2019, 11, 87–105. [Google Scholar] [PubMed]
  101. Park, S.; Chapuis, N.; Bardet, V.; Tamburini, J.; Gallay, N.; Willems, L.; Knight, Z.A.; Shokat, K.M.; Azar, N.; Viguie, F.; et al. PI-103, a dual inhibitor of Class IA phosphatidylinositide 3-kinase and mTOR, has antileukemic activity in AML. Leukemia 2008, 22, 1698–1706. [Google Scholar] [CrossRef]
  102. Mallon, R.; Feldberg, L.R.; Lucas, J.; Chaudhary, I.; Dehnhardt, C.; Santos, E.D.; Chen, Z.; dos Santos, O.; Ayral-Kaloustian, S.; Venkatesan, A.; et al. Antitumor efficacy of PKI-587, a highly potent dual PI3K/mTOR kinase inhibitor. Clin. Cancer Res. 2011, 17, 3193–3203. [Google Scholar] [CrossRef]
  103. Simioni, C.; Cani, A.; Martelli, A.M.; Zauli, G.; Alameen, A.A.; Ultimo, S.; Tabellini, G.; McCubrey, J.A.; Capitani, S.; Neri, L.M. The novel dual PI3K/mTOR inhibitor NVP-BGT226 displays cytotoxic activity in both normoxic and hypoxic hepatocarcinoma cells. Oncotarget 2015, 6, 17147–17160. [Google Scholar] [CrossRef]
  104. Zhu, D.S.; Dong, J.Y.; Xu, Y.Y.; Zhang, X.T.; Fu, S.B.; Liu, W. Omipalisib Inhibits Esophageal Squamous Cell Carcinoma Growth Through Inactivation of Phosphoinositide 3-Kinase (PI3K)/AKT/Mammalian Target of Rapamycin (mTOR) and ERK Signaling. Med. Sci. Monit. 2020, 26, e927106. [Google Scholar] [CrossRef]
  105. Jing, Z.; Fei, W.; Zhou, J.; Zhang, L.; Chen, L.; Zhang, X.; Liang, X.; Xie, J.; Fang, Y.; Sui, X.; et al. Salvianolic acid B, a novel autophagy inducer, exerts antitumor activity as a single agent in colorectal cancer cells. Oncotarget 2016, 7, 61509–61519. [Google Scholar] [CrossRef]
  106. Vidal, L.; Victoria, I.; Gaba, L.; Martin, M.G.; Brunet, M.; Colom, H.; Cortal, M.; Gomez-Ferreria, M.; Yeste-Velasco, M.; Perez, A.; et al. A first-in-human phase I/Ib dose-escalation clinical trial of the autophagy inducer ABTL0812 in patients with advanced solid tumours. Eur. J. Cancer 2021, 146, 87–94. [Google Scholar] [CrossRef]
  107. Bu, H.; Tan, S.; Yuan, B.; Huang, X.; Jiang, J.; Wu, Y.; Jiang, J.; Li, R. Therapeutic potential of IBP as an autophagy inducer for treating lung cancer via blocking PAK1/Akt/mTOR signaling. Mol. Ther. Oncolytics 2021, 20, 82–93. [Google Scholar] [CrossRef]
  108. Britten, C.D.; Adjei, A.A.; Millham, R.; Houk, B.E.; Borzillo, G.; Pierce, K.; Wainberg, Z.A.; LoRusso, P.M. Phase I study of PF-04691502, a small-molecule, oral, dual inhibitor of PI3K and mTOR, in patients with advanced cancer. Investig. New Drugs 2014, 32, 510–517. [Google Scholar] [CrossRef]
  109. Crazzolara, R.; Bradstock, K.F.; Bendall, L.J. RAD001 (Everolimus) induces autophagy in acute lymphoblastic leukemia. Autophagy 2009, 5, 727–728. [Google Scholar] [CrossRef]
  110. Dancey, J.E.; Monzon, J. Ridaforolimus: A promising drug in the treatment of soft-tissue sarcoma and other malignancies. Future Oncol. 2011, 7, 827–839. [Google Scholar] [CrossRef]
  111. Benjamin, D.; Colombi, M.; Moroni, C.; Hall, M.N. Rapamycin passes the torch: A new generation of mTOR inhibitors. Nat. Rev. Drug Discov. 2011, 10, 868–880. [Google Scholar] [CrossRef] [PubMed]
  112. Tai, W.T.; Shiau, C.W.; Chen, H.L.; Liu, C.Y.; Lin, C.S.; Cheng, A.L.; Chen, P.J.; Chen, K.F. Mcl-1-dependent activation of Beclin 1 mediates autophagic cell death induced by sorafenib and SC-59 in hepatocellular carcinoma cells. Cell Death Dis. 2013, 4, e485. [Google Scholar] [CrossRef]
  113. Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: The in vivo evidence. Nat. Rev. Drug Discov. 2006, 5, 493–506. [Google Scholar] [CrossRef]
  114. Lang, F.; Qin, Z.; Li, F.; Zhang, H.; Fang, Z.; Hao, E. Apoptotic Cell Death Induced by Resveratrol Is Partially Mediated by the Autophagy Pathway in Human Ovarian Cancer Cells. PLoS ONE 2015, 10, e0129196. [Google Scholar] [CrossRef]
  115. Liu, M.; Gu, P.; Guo, W.; Fan, X. C6 ceramide sensitizes the anti-hepatocellular carcinoma (HCC) activity by AZD-8055, a novel mTORC1/2 dual inhibitor. Tumour Biol. 2016, 37, 11039–11048. [Google Scholar] [CrossRef]
  116. Galluzzi, L.; Morselli, E.; Kepp, O.; Vitale, I.; Younes, A.B.; Maiuri, M.C.; Kroemer, G. Evaluation of rapamycin-induced cell death. Methods Mol. Biol. 2012, 821, 125–169. [Google Scholar]
  117. Wang, C.; Wang, X.; Su, Z.; Fei, H.; Liu, X.; Pan, Q. The novel mTOR inhibitor Torin-2 induces autophagy and downregulates the expression of UHRF1 to suppress hepatocarcinoma cell growth. Oncol. Rep. 2015, 34, 1708–1716. [Google Scholar] [CrossRef]
  118. Chresta, C.M.; Davies, B.R.; Hickson, I.; Harding, T.; Cosulich, S.; Critchlow, S.E.; Vincent, J.P.; Ellston, R.; Jones, D.; Sini, P.; et al. AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res. 2010, 70, 288–298. [Google Scholar] [CrossRef]
  119. Wang, L.; Zhu, Y.R.; Wang, S.; Zhao, S. Autophagy inhibition sensitizes WYE-354-induced anti-colon cancer activity in vitro and in vivo. Tumour Biol. 2016, 37, 11743–11752. [Google Scholar] [CrossRef] [PubMed]
  120. Grasso, S.; Pereira, G.J.S.; Palmeira-Dos-Santos, C.; Calgarotto, A.K.; Martinez-Lacaci, I.; Ferragut, J.A.; Smaili, S.S.; Bincoletto, C. Autophagy regulates Selumetinib (AZD6244) induced-apoptosis in colorectal cancer cells. Eur. J. Med. Chem. 2016, 122, 611–618. [Google Scholar] [CrossRef]
  121. Yazbeck, V.Y.; Li, C.; Grandis, J.R.; Zang, Y.; Johnson, D.E. Single-agent obatoclax (GX15-070) potently induces apoptosis and pro-survival autophagy in head and neck squamous cell carcinoma cells. Oral Oncol. 2014, 50, 120–127. [Google Scholar] [CrossRef] [PubMed]
  122. Liu, Y.; Gong, W.; Yang, Z.Y.; Zhou, X.S.; Gong, C.; Zhang, T.R.; Wei, X.; Ma, D.; Ye, F.; Gao, Q.L. Quercetin induces protective autophagy and apoptosis through ER stress via the p-STAT3/Bcl-2 axis in ovarian cancer. Apoptosis 2017, 22, 544–557. [Google Scholar] [CrossRef] [PubMed]
  123. Zhang, G.; Wang, Z.; Chen, W.; Cao, Y.; Wu, J.; Qiang, G.; Ji, A.; Wu, J.; Jiang, C. Dual effects of gossypol on human hepatocellular carcinoma via endoplasmic reticulum stress and autophagy. Int. J. Biochem. Cell Biol. 2019, 113, 48–57. [Google Scholar] [CrossRef] [PubMed]
  124. Li, Y.Y.; Lam, S.K.; Mak, J.C.; Zheng, C.Y.; Ho, J.C. Erlotinib-induced autophagy in epidermal growth factor receptor mutated non-small cell lung cancer. Lung Cancer 2013, 81, 354–361. [Google Scholar] [CrossRef] [PubMed]
  125. DeVorkin, L.; Hattersley, M.; Kim, P.; Ries, J.; Spowart, J.; Anglesio, M.S.; Levi, S.M.; Huntsman, D.G.; Amaravadi, R.K.; Winkler, J.D.; et al. Autophagy Inhibition Enhances Sunitinib Efficacy in Clear Cell Ovarian Carcinoma. Mol. Cancer Res. 2017, 15, 250–258. [Google Scholar] [CrossRef]
  126. Hu, F.; Zhao, Y.; Yu, Y.; Fang, J.M.; Cui, R.; Liu, Z.Q.; Guo, X.L.; Xu, Q. Docetaxel-mediated autophagy promotes chemoresistance in castration-resistant prostate cancer cells by inhibiting STAT3. Cancer Lett. 2018, 416, 24–30. [Google Scholar] [CrossRef]
  127. Yang, H.; Zhang, X.; Wang, C.; Zhang, H.; Yi, J.; Wang, K.; Hou, Y.; Ji, P.; Jin, X.; Li, C.; et al. Microcolin H, a novel autophagy inducer, exerts potent antitumour activity by targeting PITPalpha/beta. Signal Transduct. Target. Ther. 2023, 8, 428. [Google Scholar] [CrossRef]
  128. Wen, Y.; Graybill, W.S.; Previs, R.A.; Hu, W.; Ivan, C.; Mangala, L.S.; Zand, B.; Nick, A.M.; Jennings, N.B.; Dalton, H.J.; et al. Immunotherapy targeting folate receptor induces cell death associated with autophagy in ovarian cancer. Clin. Cancer Res. 2015, 21, 448–459. [Google Scholar] [CrossRef]
  129. Fares, J.; Petrosyan, E.; Kanojia, D.; Dmello, C.; Cordero, A.; Duffy, J.T.; Yeeravalli, R.; Sahani, M.H.; Zhang, P.; Rashidi, A.; et al. Metixene is an incomplete autophagy inducer in preclinical models of metastatic cancer and brain metastases. J. Clin. Investig. 2023, 133, e161142. [Google Scholar] [CrossRef]
  130. Wurstle, S.; Schneider, F.; Ringel, F.; Gempt, J.; Lammer, F.; Delbridge, C.; Wu, W.; Schlegel, J. Temozolomide induces autophagy in primary and established glioblastoma cells in an EGFR independent manner. Oncol. Lett. 2017, 14, 322–328. [Google Scholar] [CrossRef]
  131. Mordecai, J.; Ullah, S.; Ahmad, I. Sulforaphane and Its Protective Role in Prostate Cancer: A Mechanistic Approach. Int. J. Mol. Sci. 2023, 24, 6979. [Google Scholar] [CrossRef] [PubMed]
  132. Dower, C.M.; Bhat, N.; Gebru, M.T.; Chen, L.; Wills, C.A.; Miller, B.A.; Wang, H.G. Targeted Inhibition of ULK1 Promotes Apoptosis and Suppresses Tumor Growth and Metastasis in Neuroblastoma. Mol. Cancer Ther. 2018, 17, 2365–2376. [Google Scholar] [CrossRef] [PubMed]
  133. Tang, F.; Hu, P.; Yang, Z.; Xue, C.; Gong, J.; Sun, S.; Shi, L.; Zhang, S.; Li, Z.; Yang, C.; et al. SBI0206965, a novel inhibitor of Ulk1, suppresses non-small cell lung cancer cell growth by modulating both autophagy and apoptosis pathways. Oncol. Rep. 2017, 37, 3449–3458. [Google Scholar] [CrossRef] [PubMed]
  134. Ghazi, P.C.; O’Toole, K.T.; Srinivas Boggaram, S.; Scherzer, M.T.; Silvis, M.R.; Zhang, Y.; Bogdan, M.; Smith, B.D.; Lozano, G.; Flynn, D.L.; et al. Inhibition of ULK1/2 and KRAS(G12C) controls tumor growth in preclinical models of lung cancer. Elife 2024, 13, RP96992. [Google Scholar] [CrossRef] [PubMed]
  135. Pasquier, B. SAR405, a PIK3C3/Vps34 inhibitor that prevents autophagy and synergizes with MTOR inhibition in tumor cells. Autophagy 2015, 11, 725–726. [Google Scholar] [CrossRef]
  136. Meunier, G.; Birsen, R.; Cazelles, C.; Belhadj, M.; Cantero-Aguilar, L.; Kosmider, O.; Fontenay, M.; Azar, N.; Mayeux, P.; Chapuis, N.; et al. Antileukemic activity of the VPS34-IN1 inhibitor in acute myeloid leukemia. Oncogenesis 2020, 9, 94. [Google Scholar] [CrossRef]
  137. Vakifahmetoglu-Norberg, H.; Xia, H.G.; Yuan, J. Pharmacologic agents targeting autophagy. J. Clin. Investig. 2015, 125, 5–13. [Google Scholar] [CrossRef]
  138. Liu, P.F.; Tsai, K.L.; Hsu, C.J.; Tsai, W.L.; Cheng, J.S.; Chang, H.W.; Shiau, C.W.; Goan, Y.G.; Tseng, H.H.; Wu, C.H.; et al. Drug Repurposing Screening Identifies Tioconazole as an ATG4 Inhibitor that Suppresses Autophagy and Sensitizes Cancer Cells to Chemotherapy. Theranostics 2018, 8, 830–845. [Google Scholar] [CrossRef]
  139. Fu, Y.; Hong, L.; Xu, J.; Zhong, G.; Gu, Q.; Gu, Q.; Guan, Y.; Zheng, X.; Dai, Q.; Luo, X.; et al. Discovery of a small molecule targeting autophagy via ATG4B inhibition and cell death of colorectal cancer cells in vitro and in vivo. Autophagy 2019, 15, 295–311. [Google Scholar] [CrossRef]
  140. Chu, J.; Fu, Y.; Xu, J.; Zheng, X.; Gu, Q.; Luo, X.; Dai, Q.; Zhang, S.; Liu, P.; Hong, L.; et al. ATG4B inhibitor FMK-9a induces autophagy independent on its enzyme inhibition. Arch. Biochem. Biophys. 2018, 644, 29–36. [Google Scholar] [CrossRef]
  141. Graham, R.M.; Thompson, J.W.; Webster, K.A. Inhibition of the vacuolar ATPase induces Bnip3-dependent death of cancer cells and a reduction in tumor burden and metastasis. Oncotarget 2014, 5, 1162–1173. [Google Scholar] [CrossRef] [PubMed]
  142. Choi, H.S.; Jeong, E.H.; Lee, T.G.; Kim, S.Y.; Kim, H.R.; Kim, C.H. Autophagy Inhibition with Monensin Enhances Cell Cycle Arrest and Apoptosis Induced by mTOR or Epidermal Growth Factor Receptor Inhibitors in Lung Cancer Cells. Tuberc. Respir. Dis. 2013, 75, 9–17. [Google Scholar] [CrossRef] [PubMed]
  143. Verbaanderd, C.; Maes, H.; Schaaf, M.B.; Sukhatme, V.P.; Pantziarka, P.; Sukhatme, V.; Agostinis, P.; Bouche, G. Repurposing Drugs in Oncology (ReDO)-chloroquine and hydroxychloroquine as anti-cancer agents. Ecancermedicalscience 2017, 11, 781. [Google Scholar] [CrossRef]
  144. Amaravadi, R.K.; Winkler, J.D. Lys05: A new lysosomal autophagy inhibitor. Autophagy 2012, 8, 1383–1384. [Google Scholar] [CrossRef]
  145. Carew, J.S.; Espitia, C.M.; Zhao, W.; Han, Y.; Visconte, V.; Phillips, J.; Nawrocki, S.T. Disruption of Autophagic Degradation with ROC-325 Antagonizes Renal Cell Carcinoma Pathogenesis. Clin. Cancer Res. 2017, 23, 2869–2879. [Google Scholar] [CrossRef]
  146. Saini, H.; Sharma, H.; Mukherjee, S.; Chowdhury, S.; Chowdhury, R. Verteporfin disrupts multiple steps of autophagy and regulates p53 to sensitize osteosarcoma cells. Cancer Cell Int. 2021, 21, 52. [Google Scholar] [CrossRef]
  147. Shao, S.; Li, S.; Qin, Y.; Wang, X.; Yang, Y.; Bai, H.; Zhou, L.; Zhao, C.; Wang, C. Spautin-1, a novel autophagy inhibitor, enhances imatinib-induced apoptosis in chronic myeloid leukemia. Int. J. Oncol. 2014, 44, 1661–1668. [Google Scholar] [CrossRef]
  148. Czuczman, N.M.; Barth, M.J.; Gu, J.; Neppalli, V.; Mavis, C.; Frys, S.E.; Hu, Q.; Liu, S.; Klener, P.; Vockova, P.; et al. Pevonedistat, a NEDD8-activating enzyme inhibitor, is active in mantle cell lymphoma and enhances rituximab activity in vivo. Blood 2016, 127, 1128–1137. [Google Scholar] [CrossRef]
  149. Nicastri, M.C.; Rebecca, V.W.; Amaravadi, R.K.; Winkler, J.D. Dimeric quinacrines as chemical tools to identify PPT1, a new regulator of autophagy in cancer cells. Mol. Cell. Oncol. 2017, 5, e1395504. [Google Scholar] [CrossRef]
  150. Paglin, S.; Hollister, T.; Delohery, T.; Hackett, N.; McMahill, M.; Sphicas, E.; Domingo, D.; Yahalom, J. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 2001, 61, 439–444. [Google Scholar]
  151. Levy, J.M.; Thompson, J.C.; Griesinger, A.M.; Amani, V.; Donson, A.M.; Birks, D.K.; Morgan, M.J.; Mirsky, D.M.; Handler, M.H.; Foreman, N.K.; et al. Autophagy inhibition improves chemosensitivity in BRAF(V600E) brain tumors. Cancer Discov. 2014, 4, 773–780. [Google Scholar] [CrossRef] [PubMed]
  152. Liao, Y.X.; Yu, H.Y.; Lv, J.Y.; Cai, Y.R.; Liu, F.; He, Z.M.; He, S.S. Targeting autophagy is a promising therapeutic strategy to overcome chemoresistance and reduce metastasis in osteosarcoma. Int. J. Oncol. 2019, 55, 1213–1222. [Google Scholar] [CrossRef] [PubMed]
  153. Nagelkerke, A.; Bussink, J.; Geurts-Moespot, A.; Sweep, F.C.; Span, P.N. Therapeutic targeting of autophagy in cancer. Part II: Pharmacological modulation of treatment-induced autophagy. Semin. Cancer Biol 2015, 31, 99–105. [Google Scholar] [CrossRef] [PubMed]
  154. Kumar, A.; Singh, U.K.; Chaudhary, A. Targeting autophagy to overcome drug resistance in cancer therapy. Future Med. Chem. 2015, 7, 1535–1542. [Google Scholar] [CrossRef]
  155. Kim, N.Y.; Han, B.I.; Lee, M. Cytoprotective role of autophagy against BH3 mimetic gossypol in ATG5 knockout cells generated by CRISPR-Cas9 endonuclease. Cancer Lett. 2016, 370, 19–26. [Google Scholar] [CrossRef]
  156. Hwang, S.H.; Yeom, H.; Lee, M. ATG5 knockout promotes paclitaxel sensitivity in drug-resistant cells via induction of necrotic cell death. Korean J. Physiol. Pharmacol. 2020, 24, 233–240. [Google Scholar] [CrossRef]
  157. Li, Y.; Gao, S.; Du, X.; Ji, J.; Xi, Y.; Zhai, G. Advances in autophagy as a target in the treatment of tumours. J. Drug Target. 2022, 30, 166–187. [Google Scholar] [CrossRef]
  158. Ho, T.T.; Warr, M.R.; Adelman, E.R.; Lansinger, O.M.; Flach, J.; Verovskaya, E.V.; Figueroa, M.E.; Passegue, E. Autophagy maintains the metabolism and function of young and old stem cells. Nature 2017, 543, 205–210. [Google Scholar] [CrossRef]
  159. Ferro, F.; Servais, S.; Besson, P.; Roger, S.; Dumas, J.F.; Brisson, L. Autophagy and mitophagy in cancer metabolic remodelling. Semin. Cell Dev. Biol. 2020, 98, 129–138. [Google Scholar] [CrossRef]
  160. Li, X.; He, S.; Ma, B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 2020, 19, 12. [Google Scholar] [CrossRef]
  161. Kim, K.W.; Hwang, M.; Moretti, L.; Jaboin, J.J.; Cha, Y.I.; Lu, B. Autophagy upregulation by inhibitors of caspase-3 and mTOR enhances radiotherapy in a mouse model of lung cancer. Autophagy 2008, 4, 659–668. [Google Scholar] [CrossRef] [PubMed]
  162. Button, R.W.; Luo, S.; Rubinsztein, D.C. Autophagic activity in neuronal cell death. Neurosci. Bull. 2015, 31, 382–394. [Google Scholar] [CrossRef] [PubMed]
  163. Shintani, T.; Klionsky, D.J. Autophagy in health and disease: A double-edged sword. Science 2004, 306, 990–995. [Google Scholar] [CrossRef] [PubMed]
  164. Dunlop, E.A.; Tee, A.R. mTOR and autophagy: A dynamic relationship governed by nutrients and energy. Semin. Cell Dev. Biol. 2014, 36, 121–129. [Google Scholar] [CrossRef] [PubMed]
  165. Ishaq, M.; Ojha, R.; Sharma, A.P.; Singh, S.K. Autophagy in cancer: Recent advances and future directions. Semin. Cancer Biol. 2020, 66, 171–181. [Google Scholar] [CrossRef]
  166. Heckmann, B.L.; Green, D.R. LC3-associated phagocytosis at a glance. J. Cell Sci. 2019, 132, jcs222984. [Google Scholar] [CrossRef]
  167. Heckmann, B.L.; Teubner, B.J.W.; Tummers, B.; Boada-Romero, E.; Harris, L.; Yang, M.; Guy, C.S.; Zakharenko, S.S.; Green, D.R. LC3-Associated Endocytosis Facilitates beta-Amyloid Clearance and Mitigates Neurodegeneration in Murine Alzheimer’s Disease. Cell 2019, 178, 536–551.e14. [Google Scholar] [CrossRef]
  168. Shalhoub, H.; Gonzalez, P.; Dos Santos, A.; Guillermet-Guibert, J.; Moniaux, N.; Dupint, N.; Faivre, J. Simultaneous activation and blockade of autophagy to fight hepatocellular carcinoma. Autophgy Rep. 2024, 3, 2326241. [Google Scholar] [CrossRef]
  169. Li, C.; Liu, Y.; Liu, H.; Zhang, W.; Shen, C.; Cho, K.; Chen, X.; Peng, F.; Bi, Y.; Hou, X.; et al. Impact of autophagy inhibition at different stages on cytotoxic effect of autophagy inducer in glioblastoma cells. Cell. Physiol. Biochem. 2015, 35, 1303–1316. [Google Scholar] [CrossRef]
Figure 1. The four stages of autophagy. Autophagy is a process divided into four main stages: initiation, phagophore nucleation, elongation, and fusion/degradation. During the initiation stage, signaling proteins respond to external factors such as stress stimuli to regulate autophagy. When cellular energy levels are low, AMPK directly activates ULK1 to induce autophagy, while the type I PI3K/AKT pathway inhibits it via mTOR. In the nucleation stage, BECN1 serves as a platform, forming core complexes with proteins like VPS34 (a type III PI3K), AMBRA, and ATG14L. The elongation stage involves two ubiquitin-like conjugation systems: LC3 and ATG12. ATG12 forms a complex with ATG5, playing a crucial regulatory role in early autophagosome formation and interacting with ATG16L to create larger multiprotein complexes. LC3 is activated by ATG7 and transferred to ATG3, where it binds to the lipid PE through the ATG12–ATG5 conjugate, producing LC3-II, a key marker of autophagy. Once the autophagosome is fully formed, it fuses with lysosomes to create autolysosomes, a process requiring SNARE proteins such as STX17, SNAP29, and VAMP8. In the autolysosome’s acidic environment, the inner membrane is degraded, and the contents are broken down by hydrolytic enzymes.
Figure 1. The four stages of autophagy. Autophagy is a process divided into four main stages: initiation, phagophore nucleation, elongation, and fusion/degradation. During the initiation stage, signaling proteins respond to external factors such as stress stimuli to regulate autophagy. When cellular energy levels are low, AMPK directly activates ULK1 to induce autophagy, while the type I PI3K/AKT pathway inhibits it via mTOR. In the nucleation stage, BECN1 serves as a platform, forming core complexes with proteins like VPS34 (a type III PI3K), AMBRA, and ATG14L. The elongation stage involves two ubiquitin-like conjugation systems: LC3 and ATG12. ATG12 forms a complex with ATG5, playing a crucial regulatory role in early autophagosome formation and interacting with ATG16L to create larger multiprotein complexes. LC3 is activated by ATG7 and transferred to ATG3, where it binds to the lipid PE through the ATG12–ATG5 conjugate, producing LC3-II, a key marker of autophagy. Once the autophagosome is fully formed, it fuses with lysosomes to create autolysosomes, a process requiring SNARE proteins such as STX17, SNAP29, and VAMP8. In the autolysosome’s acidic environment, the inner membrane is degraded, and the contents are broken down by hydrolytic enzymes.
Molecules 29 05134 g001
Figure 2. Autophagy modulators targeting each step of the autophagy pathway.
Figure 2. Autophagy modulators targeting each step of the autophagy pathway.
Molecules 29 05134 g002
Figure 3. Anticancer drug development strategy based on the changing role of autophagy in tumorigenesis stages. Canonical ATG5/LC3-dependent autophagy suppresses tumor formation in the early stages of tumorigenesis but plays a crucial role in cell survival during the advanced stages of cancer. Therefore, for effective tumor therapy, it is important to use inducers of canonical autophagy during the initiation stage of tumor formation and inhibitors during the progression stage. Our research indicates that in challenging microenvironments for tumor cell survival, such as high cell confluency and exposure to anticancer drugs, alternative autophagy is more critical for maintaining cell survival than canonical autophagy. Thus, from a therapeutic perspective for advanced tumors, it is vital to develop anticancer strategies that focus on using inhibitors of alternative autophagy rather than canonical autophagy, depending on the tumor microenvironment.
Figure 3. Anticancer drug development strategy based on the changing role of autophagy in tumorigenesis stages. Canonical ATG5/LC3-dependent autophagy suppresses tumor formation in the early stages of tumorigenesis but plays a crucial role in cell survival during the advanced stages of cancer. Therefore, for effective tumor therapy, it is important to use inducers of canonical autophagy during the initiation stage of tumor formation and inhibitors during the progression stage. Our research indicates that in challenging microenvironments for tumor cell survival, such as high cell confluency and exposure to anticancer drugs, alternative autophagy is more critical for maintaining cell survival than canonical autophagy. Thus, from a therapeutic perspective for advanced tumors, it is vital to develop anticancer strategies that focus on using inhibitors of alternative autophagy rather than canonical autophagy, depending on the tumor microenvironment.
Molecules 29 05134 g003
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

Lee, M.; Kim, H.-G. Anti-Cancer Strategy Based on Changes in the Role of Autophagy Depending on the Survival Environment and Tumorigenesis Stages. Molecules 2024, 29, 5134. https://doi.org/10.3390/molecules29215134

AMA Style

Lee M, Kim H-G. Anti-Cancer Strategy Based on Changes in the Role of Autophagy Depending on the Survival Environment and Tumorigenesis Stages. Molecules. 2024; 29(21):5134. https://doi.org/10.3390/molecules29215134

Chicago/Turabian Style

Lee, Michael, and Hye-Gyo Kim. 2024. "Anti-Cancer Strategy Based on Changes in the Role of Autophagy Depending on the Survival Environment and Tumorigenesis Stages" Molecules 29, no. 21: 5134. https://doi.org/10.3390/molecules29215134

APA Style

Lee, M., & Kim, H. -G. (2024). Anti-Cancer Strategy Based on Changes in the Role of Autophagy Depending on the Survival Environment and Tumorigenesis Stages. Molecules, 29(21), 5134. https://doi.org/10.3390/molecules29215134

Article Metrics

Back to TopTop