Next Article in Journal
Tofacitinib Treatment Suppresses CD4+ T-Cell Activation and Th1 Response, Contributing to Protection against Staphylococcal Toxic Shock
Next Article in Special Issue
Flavonoids-Enriched Vegetal Extract Prevents the Activation of NFκB Downstream Mechanisms in a Bowel Disease In Vitro Model
Previous Article in Journal
Unveiling New Genetic Variants Associated with Age at Onset in Alzheimer’s Disease and Frontotemporal Lobar Degeneration Due to C9orf72 Repeat Expansions
Previous Article in Special Issue
Pseudolaric Acid B Targets CD147 to Selectively Kill Acute Myeloid Leukemia Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Blockage of Autophagy for Cancer Therapy: A Comprehensive Review

by
Ahmed Mostafa Ibrahim Abdelrahman Hassan
1,
Yuxin Zhao
1,
Xiuping Chen
1,2 and
Chengwei He
1,2,*
1
State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China
2
Department of Pharmaceutical Science, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(13), 7459; https://doi.org/10.3390/ijms25137459
Submission received: 1 June 2024 / Revised: 25 June 2024 / Accepted: 3 July 2024 / Published: 7 July 2024
(This article belongs to the Special Issue The Role of Natural Compounds in Cancer and Inflammation)

Abstract

:
The incidence and mortality of cancer are increasing, making it a leading cause of death worldwide. Conventional treatments such as surgery, radiotherapy, and chemotherapy face significant limitations due to therapeutic resistance. Autophagy, a cellular self-degradation mechanism, plays a crucial role in cancer development, drug resistance, and treatment. This review investigates the potential of autophagy inhibition as a therapeutic strategy for cancer. A systematic search was conducted on Embase, PubMed, and Google Scholar databases from 1967 to 2024 to identify studies on autophagy inhibitors and their mechanisms in cancer therapy. The review includes original articles utilizing in vitro and in vivo experimental methods, literature reviews, and clinical trials. Key terms used were “Autophagy”, “Inhibitors”, “Molecular mechanism”, “Cancer therapy”, and “Clinical trials”. Autophagy inhibitors such as chloroquine (CQ) and hydroxychloroquine (HCQ) have shown promise in preclinical studies by inhibiting lysosomal acidification and preventing autophagosome degradation. Other inhibitors like wortmannin and SAR405 target specific components of the autophagy pathway. Combining these inhibitors with chemotherapy has demonstrated enhanced efficacy, making cancer cells more susceptible to cytotoxic agents. Clinical trials involving CQ and HCQ have shown encouraging results, although further investigation is needed to optimize their use in cancer therapy. Autophagy exhibits a dual role in cancer, functioning as both a survival mechanism and a cell death pathway. Targeting autophagy presents a viable strategy for cancer therapy, particularly when integrated with existing treatments. However, the complexity of autophagy regulation and the potential side effects necessitate further research to develop precise and context-specific therapeutic approaches.

1. Introduction

In recent years, the overall incidence and mortality of cancer have continued to increase. Cancer has become one of the leading causes of death, with a reported worldwide mortality rate of nearly 10 million in 2020 [1,2,3]. Currently, surgical interventions, radiotherapy, and chemotherapy are the main treatments for cancers [4,5,6]. However, survival rates in metastatic cancers remain unsatisfactory, attributed to the complex nature of treating cancers and therapeutic resistance [7]. Additionally, many cellular adaptations may affect drug efficacy, one of which is autophagy, the focus of this review.
Autophagy plays a crucial role in the occurrence, drug resistance, and treatment of cancer [8]. It is a multi-step self-degradation mechanism in a wide range of cells, acting as a recycling system within living cells by degrading misfolded proteins and damaged or aging organelles through the lysosomes [9,10]. It plays a vital role in cell proliferation, apoptosis, migration, and the invasion of tumors [11,12]. Moreover, autophagy is usually maintained at basal levels in normal conditions to maintain homeostasis and facilitate adaptation under stress as a cytoprotective process. When cells encounter extrinsic stresses, including reactive oxygen species (ROS), long-lived proteins, infection, and damaged mitochondria, autophagy transports these components to lysosomes for degradation and recycling [13].
The term “autophagy” was coined by Christian de Duve in 1963, referring to “self-eating” and describing the mechanism by which cells break down their components to maintain cellular homeostasis [14]. Since then, there has been increasing research focusing on explaining the relationship between autophagy and various diseases. Dysregulated autophagy has been implicated in a wide range of pathologies, and its modulation by targeting regulatory factors can potentially impact disease progression. In malignancies, for example, the role of autophagy varies among tumor-suppressive, tumor-promoting, or neutral effects depending on the context, highlighting its complexity and the reasons for chemoresistance [15,16,17]. Consequently, many researchers are investigating the potential of autophagy inhibitors as a treatment strategy for various types of cancer, and for understanding the complex interactions in patients [18,19]. By inhibiting autophagy, the deprivation of necessary nutrients and energy sources in cancer cells will cause cell apoptosis and necrosis. This process can be achieved by autophagy inhibitors (either natural or synthetic), which have shown promising results in preclinical studies, with many agents progressing to clinical trials as standalone treatments or in combination with standard-of-care therapeutics. Notably, most current autophagy inhibitors are repurposed agents, previously used for other diseases, such as chloroquine (CQ) and hydroxychloroquine (HCQ) in malaria, and they have progressed to the clinic faster than early-stage development drugs. However, their clinical efficacy is still under investigation [20].

2. The General Biology of Autophagy

As previously discussed, autophagy is a highly conserved cellular process responsible for the degradation of damaged intracellular components and the production of recyclable molecules such as glucose, ATP, amino acids, and fatty acids. This process plays a crucial and indispensable role in cellular degradation, functioning in conjunction with the highly specific proteasomal degradation pathway, known as the ubiquitin–proteasome system [21,22]. Furthermore, autophagy is essential for maintaining cellular homeostasis and can be categorized into three distinct types: chaperone-mediated autophagy, microautophagy, and macroautophagy. These different types of autophagy dispose of cytoplasmic components by transporting them into lysosomes [23,24]. Despite having a common goal, the mechanisms of cytoplasmic sequestration and the pathways used to transport targeted proteins differ among these types. When considering chaperone-mediated autophagy, it specifically requires the presence of certain proteins such as heat shock protein 70 or lysosome-associated membrane protein 2A for lysosomal degradation [25].
Additionally, autophagy can be divided into microautophagy, which captures cargo by utilizing membrane-bound structures and directly enters the lysosome through the invagination of the lysosome membrane without forming an autophagosome [26]; and macroautophagy, which can be divided into five distinct stages: initiation, nucleation, elongation, maturation, fusion, and degradation, which are tightly regulated by a complex network to ensure the sequential advancement of this vital cellular process [11,21]

Molecular Mechanisms and Phases of Autophagy

As shown in Figure 1, the regulation of autophagy at the molecular level primarily occurs through the canonical pathway, which integrates key components like Unc-51-like kinase 1 (ULK1), the phosphoinositide 3-kinase (PI3K) complex, the microtubule-linked protein 1-light chain 3 (LC3) conjugation system, and lysosomal hydrolases. This pathway involves a complex interplay of signaling pathways and proteins that govern the initiation and progression of autophagy [20].
The initiation phase of autophagy begins with the activation of autophagy machinery in response to specific signals, such as cellular stress [27]. This intricate process is orchestrated by a network of signaling pathways, involving the ULK1 protein [20,28,29].
Following initiation, the process advances to the nucleation phase. This phase is characterized by the formation of a double-membrane structure called the isolation membrane, mediated by the class III PI3K complex, specifically targeting VPS34 [30]. The isolation membrane elongates to form a structure known as the phagophore. The elongation and maturation of the phagophore are facilitated by the LC3 conjugation system, which involves autophagy-related genes (ATG), particularly the ATG12–ATG5-ATG16L1 complex. This complex plays a crucial role in promoting LC3 lipidation and autophagosomal membrane formation, ultimately leading to the creation of a completed autophagosome [31].
The final stages of autophagy involve fusion and degradation. During the fusion step, the autophagosome merges with a lysosome to form an autolysosome. Once the autolysosome is formed, lysosomal hydrolases acidify its interior, facilitating the degradation of the autophagic cargo [32].
Consequently, a comprehensive understanding of the molecular mechanisms and phases of autophagy is crucial for developing effective inhibitors that impact this vital cellular process. Surprisingly, many inhibitors targeting different stages of autophagy have been identified and studied for their therapeutic potential. For instance, ULK1 inhibitors disrupt the initiation phase [33], while class III PI3K inhibitors, targeting VPS34, impede the nucleation phase [34,35,36]. Autophagosome maturation can be inhibited by agents that interfering with ATG [37]. Additionally, lysosomotropic agents prevent the autophagosomes’ fusion with lysosomes, thereby preventing the formation of autolysosomes and subsequent cargo degradation [38,39].

3. The Regulation of Autophagy

As autophagy progresses through maturation and degradation, the mechanistic target of rapamycin kinase (mTOR) and AMP-activated kinase (AMPK) signaling pathways play crucial roles as regulators. These nutrient sensors establish complex connections with the autophagy machinery, influencing the dynamics and outcomes of the process. Both mTOR and AMPK are extensively studied and well-understood as the central regulators of autophagy [40,41]. Their coordinated activation and inhibition effectively control the sequestration, transport, fusion, and degradation of cargo in the final stage of autophagy, providing critical insight into the precise coordination required for autophagy completion. While other pathways and molecules are implicated in autophagy regulation, mTOR and AMPK remain the focus of extensive research and comprehension.

3.1. Mammalian Target of Rapamycin (mTOR)

The mTOR pathway functions as the central negative regulator of autophagy [42]. It is a critical serine/threonine kinase that regulates cell growth, metabolism, and autophagy [43]. The activation of mTOR occurs under conditions such nutrient abundance, environmental/cellular stress-free conditions (hypoxia, heat shock, osmotic stress, ROS, DNA damage, and endoplasmic reticulum stress), and sufficient growth factors [41,43,44]. In contrast, nutrient scarcity leads to mTOR inhibition, and initiates autophagy activation [41].

3.2. AMP-Activated Protein Kinase (AMPK)

The AMPK pathway plays a critical role in maintaining ATP levels within the cell and responds to various cellular stresses, such as hypoxia, nutrient deprivation, and mitochondrial stress [45,46]. It triggers autophagy by influencing a variety of proteins, including ULK1 and Beclin-1 [47].
In fact, the role of AMPK in autophagy regulation has been well studied. The studies found that AMPK is required for autophagy induction in response to glucose deprivation and inhibiting its function results in disrupted autophagy flux and the accumulation of autophagosomes. In summary, under conditions of glucose deprivation, AMPK promotes the activation of ULK1 and Beclin-1 via phosphorylation at specific sites, highlighting its pivotal role as a regulator of autophagy initiation [44,48,49,50].

4. Bipolar Nature of Autophagy in Cancer

Many studies have been focused on elucidating the relationship between autophagy and the progression of cancer. The impact of autophagy on the fate of tumor cells is contingent upon factors such as the type of cancer, its stage, and the genetic characteristics involved [51]. The multifaceted role of autophagy in cancer therapy exhibits context-dependent complexity, manifesting in two distinct responses to anticancer drugs [52]. One response is the cytotoxic function known as autophagic cell death, which prevents mutations [53,54]. The other response is the cytoprotective function, which leads to drug resistance. As a result, this mechanism poses a significant clinical challenge for achieving successful cancer treatment and results in poor patient prognosis [55].

4.1. Tumor-Suppressive Role of Autophagy

Autophagic cell death, also known as type II programmed cell death, is a non-apoptotic process induced by an anticancer treatment that promotes the autophagy of cancer cells and culminates in cell death due to overactivated autophagy [56,57,58,59]. Moreover, activating autophagy-related signaling pathways may promote the degradation of potentially oncogenic molecules, thereby contributing to the defense against tumor invasion, angiogenesis, and migration [60,61,62].
Furthermore, autophagic cell death is closely associated with the regulation of pivotal proteins such as Beclin-1, which plays a vital role in tumor suppression. Under normal conditions and early stages of cancer, Beclin-1 safeguards cells against adverse stimuli and stress, thus maintaining cellular function and integrity. Additionally, it counteracts the detrimental effects of ROS, which are implicated in triggering mutations and DNA damage associated with cancer progression. By triggering autophagy in response to increased ROS levels, Beclin-1 facilitates the removal of damaged cellular components, preserving genomic stability and mitigating oxidative stress [63,64]. Nonetheless, disturbances in the regulation of Beclin-1 may compromise autophagy, potentially leading to genome instability and tumorigenesis [65]. Overall, autophagy prevents tumor initiation, and its dysfunction can contribute to tumorigenesis.

4.2. Tumor-Promoting Role of Autophagy

Given that autophagy serves as a survival mechanism and stress response, it also contributes to the endurance of established tumor cells under various stress conditions. Furthermore, autophagy enhances stress tolerance in developed tumors and provides an alternative pathway for cancer cells to sustain their energy requirements and procure vital nutrients [16,17]. This protection is achieved through the breakdown of macromolecules and the recycling of the essential building blocks that fuel the metabolism of tumor cells. Thus, it nullifies the effect of drugs. Therefore, the upregulation of autophagy can offer tumor cells a competitive advantage over normal cells, fostering aggressiveness and resistance to cancer therapy [66].

5. The Rationale for Targeting Autophagy in Cancer Therapy

Autophagy modulation has gained significant attention as a promising strategy for cancer therapy. Based on the above information, autophagy plays a dual role in cancer, working as both a cytoprotective and a cytotoxic factor for cancer cells. Hence, autophagy can function as a tumor promoter or suppressor. This complexity makes it a potential target for cancer therapy using autophagy inhibitors. In detail, cancer cells rely on autophagy for survival in adverse conditions like nutrient deprivation and hypoxia, making it an attractive therapeutic target due to its high energy demands [67].
Moreover, targeting autophagy can enhance the efficacy of existing chemotherapy drugs. When autophagy is inhibited, cancer cells become more susceptible to the cytotoxic effects of anticancer agents, indicating a potential method for improving treatment outcomes in various cancers, including breast, ovarian, and melanoma [68,69,70,71]. As a result, autophagy presents a promising approach to cancer therapy [72,73].
However, the debate on whether to activate or inhibit autophagy remains intense due to the complexity of autophagy physiology, and no consensus has been reached. Drug targets have been identified at almost every step of autophagy, from initiation to vesicle nucleation and maturation, vesicle fusion, and lysosomal degradation. Understanding these regulatory pathways could help develop new cancer treatment strategies [74]. Further research is necessary to explore precise and context-specific strategies for effectively utilizing autophagy inhibitors in cancer treatment.
However, drug resistance poses a significant challenge in cancer therapy, necessitating novel approaches. Increasing research has demonstrated that drug resistance in cancer therapy can be overcome by pharmacologically inhibiting autophagy using inhibitors targeting critical components within the autophagy pathway, or through genomic interference against autophagic genes such as small interfering RNA (siRNA), targeting ATG3, ATG5, and ATG7 [75]. Fortunately, blocking autophagy pharmacologically or genetically has shown promising results in inducing tumor regression in genetic mouse models and pancreatic cancer xenografts, highlighting them as promising therapies for targeting autophagy [76].
Up to now, autophagic inhibitors can be categorized into three classes. The first category comprises Class III PI3K inhibitors that target the VPS34 enzyme involved in autophagy and vesicle dynamics. The other two main types of PI3K inhibitors are Class I inhibitors, which intercept oncogenic PI3Ks, and Class II inhibitors, which regulate membrane trafficking. Collectively, these three PI3K pathways are favored targets for anticancer drug development due to their impact on crucial cellular processes like signaling and cytoskeletal dynamics, resulting in augmented activity of chemotherapies (e.g., taxol and doxorubicin) [34,35,36]. The second category consists of lysosomotropic agents that block autophagic progression by repressing lysosomal acidification [38,39]. The third category includes autophagosome–lysosome fusion inhibitors, which impede the fusion of autophagosomes with lysosomes during the maturation stage, consequently halting the autophagy process [77].
Within the spectrum of autophagy inhibitors, it is worth noting that combining natural products with chemotherapy is a prevalent approach to enhance the anticancer effects while mitigating the dose-dependent adverse effects of cancer treatment. Considering natural products, one of the most prominent components used extensively in healthcare is traditional Chinese medicine (TCM), especially herbal medicine. Recent research findings investigate the potential novel applications of TCMs via the regulation of autophagy, emphasizing their therapeutic effect in cancer treatment [78]. For instance, oxymatrine targets the PI3K pathway, which plays a crucial role in cell growth and survival [79]. Additionally, toosendanin (TSN) and berbamine function as lysosomotropic agents, disrupting the lysosomal activity essential for autophagy [80,81]. This multi-targeted approach of TCM highlights its potential as a valuable compound in cancer therapy.
Furthermore, contemporary scientific investigations have sparked increasing interest in exploring the impact of ULK inhibitors on cellular processes, including autophagy. These inhibitors form complexes with ULK1 regulatory units and operate by impeding kinases in clinical settings, making ULK1 an attractive candidate for autophagy inhibition [33]. Apart from ULK inhibitors, considerable research endeavors have concentrated on comprehending and utilizing the potential of tyrosine kinase and proteasome inhibitors for autophagy suppression as a prospective therapeutic strategy for cancer therapy.
In addition to the main classes mentioned, there are other subordinate classes, such as ATG inhibitors. By utilizing genomic interference techniques, for example, RNA interference (RNAi) and CRISPR, these inhibitors can disrupt ATG3, ATG5, ATG7, and BECN1, impairing the complex formation that is involved in the elongation of phagophores, thereby modulating autophagy [37].

6. The Classes of Pharmacological Inhibitors Targeting Key Components of Autophagy

Given the important roles of autophagy in tumorigenesis and cancer therapy, the inhibition of autophagy is an attractive strategy to enhance the anticancer activity of conventional therapeutic drugs. An increasing number of autophagy inhibitors have been identified. Autophagy can be inhibited at both early (initiation) and late (autophagosome–lysosome fusion and cargo degradation) stages of autophagic flux (Figure 2). The preclinical studies as well as the mechanisms of the major autophagy inhibitors for anticancer therapy are intensively discussed in this section, while others are summarized in Table 1.

6.1. Class III PI3K Inhibitors

PI3Ks are lipid kinases that regulate diverse cellular processes, including proliferation, survival, adhesion, and motility. Among the different classes of PI3Ks, class III holds significant importance in governing autophagy, with VPS34 being a critical constituent in the class III PI3K signaling pathway. The targeted inhibition of VPS34 has led to the development of specific VPS34 inhibitors, which have gained significant attention for their ability to modulate this critical component of PI3K cascades. By disrupting the formation of autophagosomes, VPS34 inhibitors effectively impede the process. Therefore, the linkage between class III PI3K class and autophagy inhibitors supports the elucidation of the intricate mechanisms involved in autophagy regulation, providing valuable insights for manipulating the autophagy pathway. Notable examples of early-stage autophagy inhibitors within this class include the natural products wortmannin [235,236] and viridiol, as well as their respective synthetic compounds, 3-methyladenine (3-MA), LY294002, and SAR405.

6.1.1. Wortmannin

Wortmannin is a metabolite derived from the culture of Penicillium funiculosum, which exhibits a range of pharmacological effects such as weak antifungal properties and highly active anti-inflammatory effects [237,238]. It has gained prominence for its role as an autophagy inhibitor, effectively hindering autophagosome formation by blocking the class III PI3K pathway during the early nucleation stage, with IC50 values ranging from 10 to 50 nM [239,240,241]. Moreover, during the early nucleation stage of autophagy, wortmannin disrupts autophagosome formation by making an irreversible covalent bond with the class III PI3K, consequently activating the cell cycle and apoptosis, and rendering it a non-competitive inhibitor of the PI3K/AKT pathway [239,240,241,242,243,244,245,246,247]. Furthermore, wortmannin can irreversibly inhibit the serine-specific auto-kinase activity of mTOR [239], which contributes to its anticancer properties. One study by Rao et al. revealed that the combination of wortmannin and doxorubicin, using size-adjustable micelles, effectively suppressed the growth of breast cancer and melanoma cells in mice [241].
Additionally, when combined with cisplatin, wortmannin was found to enhance the effectiveness of chemotherapy in overcoming cisplatin resistance in platinum-resistant ovarian cancer [248]. However, wortmannin is not a suitable cancer chemotherapeutic agent for individual use due to stability and toxicity issues. Because of this, there is intense interest in developing new analogs of wortmannin to improve its drug-like properties.

6.1.2. Viridiol

A dihydric derivative called phytotoxic viridiol, derived from the antifungal compound viridian, complements the actions of wortmannin by preventing autophagosome formation. Its 9-epi-viridiol derivative has shown considerable cytotoxic effects on KB and Hela cells [249,250]. In addition, a semi-synthetic viridian derivative, PX-866, has been investigated for its ability to inhibit PI3K activity, resulting in decreased growth and motility of various human cancer cells. PX-866, an improved wortmannin analog, demonstrates greater potency and sustained inhibition of PI3K signaling compared to wortmannin. Notably, PX-866 effectively suppresses cancer cell motility and growth at low nanomolar concentrations, whereas higher concentrations of wortmannin are required for similar effects. Therefore, based on these findings, Howes et al. have suggested PX-866 to be a promising analog of wortmannin, indicating potential improvements in therapeutic applications and PI3K-targeted therapies [251,252].

6.1.3. 3-Methyladenine (3-MA)

As mentioned earlier, natural products have demonstrated the potential to inhibit class III PI3K-mediated autophagy, while synthetic compounds have garnered attention for their ability to modulate autophagy. Known as a well-established autophagy inhibitor, 3-MA effectively blocks autophagy at both the initiation and maturation stages by disrupting the interaction between class III PI3K and various ATG partners. Initially identified for its autophagy inhibitory effects in rat hepatocytes, 3-MA has gained widespread use in research due to its ability to enhance the therapeutic effects of anticancer drugs. Mechanistically, 3-MA inhibits human VPS34 enzymatic activity by binding to a unique hydrophobic pocket of VPS34, a distinct feature from other related kinases like PI3Kα. Moreover, 3-MA has been shown to enhance the therapeutic effect of 5-fluorouracil (5-FU) in gastric carcinoma cells, as well as increasing the sensitivity of paclitaxel-resistant HeLa cervical cancer cells to paclitaxel [253]. However, the in vivo applications of 3-MA are limited due to its poor solubility at room temperature. To overcome this drawback, Wu et al. developed 29 derivatives that not only have improved solubility but also exhibit enhanced activity and selectivity. Among these derivatives, three compounds stand out as promising candidates: [4-(3-methyl-3H-purin-6-yl) thiomorpholine], [3-methyl-6-(3-methylpiperidin-1-yl)-3H-purine], and [6-(4-(3,4-dichlorophenyl) piperazin-1-yl)-3-methyl-3H-purine]. These compounds were identified based on the observation of autophagosome formation upon autophagy induction, using LC3 as an autophagy marker [254]. The development of these novel derivatives with improved solubility presents new opportunities for utilizing 3-MA in vivo and further advancing therapeutic strategies and autophagy research.

6.1.4. LY294002

LY294002, known as 2-(4-morpholinyl)-8-phenylchromone, stands out as one of the earliest PI3K blockers [235,255]. While it has been proven effective in targeting DNA-dependent protein kinase and mTOR, its impact on class III PI3K, particularly VPS34, remains limited [256]. Notably, studies have revealed the potential of LY294002 to enhance the cytotoxicity of temozolomide in cutaneous melanoma cell lines. In vitro research has demonstrated the ability of LY294002 to enhance temozolomide-induced growth arrest and induce G0/G1 cell cycle arrest in Mel Z and Mel IL cell lines [257]. Additionally, in SCC-25 cell lines, LY294002 has been found to reduce GLUT1 expression and influence BAD phosphorylation [258]. Moreover, LY294002 has also demonstrated synergistic cytotoxicity when combined with the natural compound curcumin in breast cancer cell lines, positioning it as a potential enhancer of drug-induced apoptosis [259].

6.1.5. SAR405

In the realm of synthetic compounds, SAR405 is at the forefront as an autophagy inhibitor with low molecular weight, high activity, and remarkable specificity. It selectively targets VPS34 as an ATP-competitive inhibitor. Furthermore, SAR405 demonstrates a distinctive selectivity on class I and II isoforms of PI3K-PtdIns3K, and on mTOR, enabling it to disrupt vesicle trafficking from late endosomes to lysosomes and block PIK3C3 catalytic activity. The potential of SAR405 is evident in Pasquier’s research, which highlights the prospect of combining it with everolimus. This combination inhibits cell proliferation in renal cancer, emphasizing the significant role of SAR405 in advancing cancer therapy [260].
The previous compounds, except for wortmannin, affect related lipids and protein kinases such as mTOR and DNA-dependent protein kinase. However, these compounds have low bioactivity and bioavailability, which is essential to consider within clinical trials [256].

6.2. Lysosomotropic Agents (Repressors of Lysosomal Acidification)

Lysosomotropic agents refer to substances that are taken into lysosomes in vivo or in vitro [261]. Lysosomes are the final cell organelles in the endocytic process, where they break down macromolecules with the help of hydrolytic enzymes. These enzymes are active within an acidic pH range from 4.5 to 5.5 [262,263]. However, lysosomotropic agents hinder lysosomal acidification and inhibit the degradation of autophagosomes, thus blocking autophagy [264]. This activity is achieved by using pharmacological inhibitors, such as the natural product TSN and its synthetic analogues including CQ, HCQ, Lys0569, and Roc-325.

6.2.1. Toosendanin (TSN)

TSN is a TCM extracted from Melia toosendan Sieb et Zucc. It inhibits autophagy by increasing lysosomal pH rather than blocking the fusion of autophagosomes and lysosomes. Moreover, its impact was investigated in both in vitro and in vivo on triple-negative breast cancer (TNBC), when combined with the topoisomerase I inhibitor irinotecan to demonstrate its therapeutic effects on TNBC. The findings revealed that TSN hindered SN-38/irinotecan-induced apoptosis in TNBC cells and significantly induced protective autophagy in tumor xenograft models compared to using SN-38/irinotecan alone [80]. This result highlights the capability of TSN to inhibit autophagy.

6.2.2. Chloroquine (CQ) and Hydroxychloroquine (HCQ)

Upon wrapping up the TSN analysis, the focus shifts to the study of CQ and HCQ medications. CQ is an aminoquinoline that has been certified by the FDA as an anti-malarial remedy since its approval on 31 October 1949 [264]. Additionally, CQ and its derivatives, such as HCQ, are used in the treatment of various diseases including rheumatoid arthritis [265,266], malaria [20], HIV [267], systemic lupus [268,269], and more recently, in the treatment of COVID-19 [270]. The primary mechanism of action for both CQ and HCQ involves the inhibition of lysosomal acidification. CQ interferes with autophagy during the later stages by generating acidic vesicular organelles in the cytoplasm [271,272]. When lysosomes are exposed to an acidic environment, they continuously accumulate CQ and HCQ, leading to an increase in lysosomal pH due to their alkaline properties, thus blocking the activity of hydrolytic enzymes. Consequently, lysosomal digestion and autophagy are suppressed because of the damage of cytoplasmic proteins and endoplasmic reticulum stress, ultimately leading to apoptosis [272].
Interestingly, a study has explored the synergistic potential of combining natural products with CQ. For instance, the combination of CQ with a tetrandrine natural compound exhibited significant antitumor activity, indicating the possibility of enhancing therapeutic outcomes through this combination therapy [135]. Additionally, the combined effect of another natural product called resveratrol with HCQ was investigated on an osteosarcoma cell line (MG-63). This study revealed a synergistic effect and further emphasized the potential of combining autophagy inhibitors with natural products for enhanced therapeutic efficacy [134]. Furthermore, medical progress has prompted investigations into the optimal dosage of CQ and HCQ that balances safety and effectiveness in cancer treatment. Karim et al. conducted a phase I study to evaluate the maximum tolerated dose of CQ and HCQ in combination with carboplatin-gemcitabine in advanced solid tumors. It was observed that the maximum tolerated doses of CQ and HCQ are lower when used concomitantly with the previously reported chemotherapeutic regimes due to the myelosuppressive action of carboplatin–gemcitabine [273]. This study highlights the potential of CQ or HCQ to yield synergistic antitumor effects, pointing towards the possibility of the development of novel therapeutic drugs in cancer treatment.
Although CQ and HCQ have been extensively studied for their potential in various medical applications, a new synthetic drug known as Lys05 is now emerging as a promising competitor. Lys05 is a novel lysosomal autophagy inhibitor currently in development. It is a water-soluble salt derived from its parent compound, Lys01. However, Lys05 tends to accumulate within the lysosome, causing deacidification and leading to a more significant autophagy inhibition. In two models of melanoma xenograft and a model of colon cancer xenograft, lower doses of Lys05 blocked early autophagy obviously and showed effects as a single antitumor agent. Moreover, Lys05 has a higher potency than CQ and HCQ, with 10 times more cytotoxic and antitumor activity [136,137].

6.2.3. ROC-325

ROC-325 has been developed by applying a logical medicinal chemistry approach to drug design. This compound leads to autophagosome accumulation and lysosome deacidification at low doses, and it has been administered orally in some in vivo studies using mice. The analysis of tumor samples from mice treated with ROC-325 indicated its ability to inhibit autophagy and decrease tumor cell proliferation [154]. In another in vivo study, ROC-325 was found to enhance the activity of azacitidine (AZA) as an anti-leukemic agent in acute myeloid leukemia by inducing increased LC3B and p62 levels. It increased the anti-leukemic activity of AZA by antagonizing its effects on p62, while also reducing the suppressive effect of AZA on autophagy induction [153].

6.3. Inhibitors of Autophagosome–Lysosome Fusion

Autophagosome–lysosome fusion is one of the critical steps during autophagy [77]. It occurs at the later stages of autophagy, and it can be inhibited by many agents such as Pulsatilla saponin D (PSD), liensinine, bafilomycin A1(Baf A1), and specific and potent autophagy inhibitor (spautin-1).

6.3.1. Pulsatilla Saponin D (PSD)

PSD is a saponin derivative extracted from Pulsatilla chinensis (Bunge) Regel [274]. It inhibits autophagic flux through three mechanisms: initially, obstructing the fusion of autophagosomes with lysosomes; then elevating lysosomal pH; and finally, inhibiting the activation of lysosomal cathepsins. Notably, a study conducted in vitro explored the effects of PSD in combination with the alkaloid chemotherapeutic agent camptothecin (CPT) as well as its use as a single agent on human breast MCF-7 and MDA-MB-231 cancer cells. The results showed that PSD serves as an autophagy inhibitor, and whether combined with CPT or not, it led to an increase in p62 levels in both MCF-7 and MDA-MB-231 cells, confirming its effectiveness in autophagy inhibition [275,276].

6.3.2. Liensinine

Another specific natural autophagy inhibitor is liensinine, which is an isoquinoline alkaloid extracted from the seed embryo of Nelumbo nucifera Gaertn. It has diverse biological activities, including preventing arrhythmias, reducing hypertension, preventing pulmonary fibrosis, and inducing relaxation in vascular smooth muscle. Studies indicate that liensinine inhibits autophagosome–lysosome fusion, causing the accumulation of autophagosomes and mitophagosomes. More specifically, it hinders autophagic degradation, blocks autophagosome–lysosome fusion, causes the accumulation of autophagy substrates, and delays the maturation of important lysosomal hydrolases. Furthermore, by inhibiting autophagy/mitophagy, liensinine enhances the susceptibility of breast cancer cells to the cell death-inducing effects of doxorubicin via DNM1L-dependent mitochondrial fission, suggesting its potential to synergize with chemotherapy in autophagy inhibition [125].

6.3.3. Bafilomycin A1(Baf A1)

Baf A1 is a macrolide antibiotic isolated from Streptomyces griseus. It has a dual action by inhibiting vacuolar V-ATPase and interrupting the passage of proteins through the lysosomal membrane. It also blocks lysosomal acidification and autophagosome–lysosome fusion [277,278]. In addition, Baf A1 has demonstrated an ability to augment chemotherapy sensitivity in gastric, osteosarcoma, and colon cancer cells [279,280,281]. In the context of colon cancer, Baf A1 specifically targets the aberrant activity of mTOR, which contributes to cancer progression, chemotherapy resistance, and recurrence [279,280]. Moreover, activation of mTORC1/2 is essential to promoting cancer behaviors, such as tumor cell growth, survival, proliferation, and resistance to apoptosis [282,283]. When co-administered with WYE-354, potent inhibitors of mTORC1/2, Baf A1, and 3-MA enhance WYE-354’s anti-survival effect against HT-29 cells [284,285]. Additionally, the combination of Baf A1 and 3-MA increases the apoptotic activity of nedaplatin in cisplatin-resistant nasopharyngeal carcinoma cells by elevating LC3-II, cleaved caspase 3, and cleaved PARP levels in HNE1/DDP cells [286].

6.3.4. Specific and Potent Autophagy Inhibitor Spautin-1

Another potent autophagy inhibitor, sapautin-1, is also worth being focused on. It is a derivative of quinazoline that enhances the downregulation of VPS34 complexes. By primarily targeting and inhibiting ubiquitin-specific peptidases such as USP10 and USP13, sapautin-1 affects the protein Beclin-1, leading to its degeneration during glucose-free conditions [93]. Consequently, autophagy is impeded because of the decrease of VPS34 [94]. Furthermore, the combined application of spautin-1 with imatinib mesylate (IM) could bring a synergistic therapeutic effect on chronic myeloid leukemia (CML) [94]. Acting as a selective agent against BCR-ABL [287,288], Imatinib (IM) helps in the prognostication of CML patients in the chronic phase. However, resistance to IM presents a challenge for patients in the progressive phase [287,289]. A study on synergistic effects revealed that spautin-1 increases the cytotoxic effect of IM in the K562 cell line (an immortalized myelogenous leukemia cell line) as well as in primary cells. It also suppressed IM-induced autophagy in a manner dependent on Beclin-1 and triggered the inactivation of PI3K/AKT while activating GSK3β, subsequently reducing the expression of the anti-apoptotic proteins Mcl-1 and Bcl-2. It is noteworthy that monotherapy of sapautin-1 does not significantly impact apoptosis, but it could increase IM-induced caspase-3 cleavage, indicating the enhancement of apoptosis [94].
Emodin [6-methyl-1,3,8-trihydroxy anthraquinone]. ROC-325 [1-((2-((2-((7-chloroquinolin-4-yl)amino)ethyl)(methyl)amino)ethyl)amino)-4-methyl-9H-thioxanthen-9-one]. Irinotecan [7-ethyl-10-hydroxycamptothecin]. 3-MA derivatives [4-(3-methyl-3H-purin-6-yl) thiomorpholine], [3-methyl-6-(3-methylpiperidin-1-yl)-3H-purine] and [6-(4-(3,4-dichlorophenyl) piperazin-1-yl)-3-methyl-3H-purine]. Spautin-1 [6-Fluoro-N-(4-fluorobenzyl) quinazoline-4 amine]. BAY 80-6946 [2-amino-n-(7-methoxy-8-(3-morpholinopropoxy)-2,3-dihydroimidazo[1,2-c]quinazolin-5-yl)pyrimidine-5-carboxamide]. Compound 31 [1-[4-[4-[4-(2,3-dihydro-1,4-benzodioxin-6-ylsulfanyl)-3-(trifluoromethyl)phenyl]pyridin-2-yl]piperazin-1-yl]ethanone]. CA-5f [3E, 5E0-3-(3,4-dimethoxybenzylidene)-5-[(1H-indol-3—yl)methylene]-1-methylpiperidin-4-one].

7. Gene Therapy Targeting Autophagy

In previous sections, the efficiency of antitumor compounds in regulating autophagy and suppressing cancer progression has been discussed. The next consideration is the availability of efficient genetic tools for suppressing the expression of ATGs at the transcriptional level. Gene silencers are specific DNA or RNA sequences that can hinder the expression of target genes, providing the potential to develop effective molecules with antitumor properties [290]. Studies have identified the upstream regulators of autophagy, and due to the utilization of genetic tools such as the CRISPR system and RNAi, molecular pathways can be targeted in autophagy regulation and affect the progression of cancers.
CRISPR-Cas9 can be used to create gene knockout models by introducing targeted double-strand breaks in the DNA of ATGs such as ATG7, ATG5, or BECN1, leading to the permanent loss of function of these genes and, thus, inhibiting autophagy [291,292,293,294,295]. On the other hand, RNAi utilizes short hairpin RNA (shRNA) or siRNA to silence gene expression post-transcriptionally. Specifically, by designing RNAi molecules to specifically target mRNA transcripts of key autophagy genes like ULK1 or VPS34, the synthesis of these essential autophagy proteins can be effectively reduced [296]. Together, these techniques offer synergistic strategies for autophagy inhibition and potential therapeutic avenues: CRISPR-Cas9 causes permanent gene disruption, while RNAi allows for reversible gene silencing.

8. Clinical Trials Targeting Autophagy for Cancer Therapy

As previously mentioned, CQ and HCQ are the only FDA-approved drugs used as autophagy inhibitors. Notably, these drugs have demonstrated efficacy in the treatment of both COVID-19 and cancer, highlighting their potential therapeutic versatility. In COVID-19, they succeeded in blocking the viral infection through autophagy inhibition, though the clinical evidence is still controversial. Early studies suggested that these drugs could block SARS-CoV-2 infection in vitro. Additionally, an open-label non-randomized trial in France found that HCQ treatment alone significantly reduced COVID-19 patients’ viral load, with an enhanced effect when combined with azithromycin. However, this study had a small sample size. Larger observational studies, such as one involving over 1400 patients, found no benefit of HCQ in reducing the risk of death in hospitalized COVID-19 patients. Consequently, more recent clinical trial results have clearly demonstrated that CQ/HCQ alone or in combination with other agents did not show any benefit, leading the NIH to halt all clinical trials on these drugs and the FDA to strongly advise against their use for COVID-19 treatment [297]. Shifting to cancer treatment, their contribution in cancer therapy is still limited due to the complicated mechanisms of autophagy. Usually, autophagy inhibitors are utilized with cytotoxic agents because antitumor therapy creates intracellular stress and starves the cells. In this case, the addition of autophagy inhibitors could exacerbate the disruption of homeostasis in tumor cells, thus enhancing the efficacy of antitumor agents [298]. For instance, in a preclinical trial, the inhibition of autophagy by CQ or Baf A1 significantly strengthened the cytotoxic effect of pirarubicin on cervical cancer cells and inhibited tumor growth in the xenograft mouse model [299]. However, although autophagy inhibitors are involved in many preclinical studies, there are only limited data about them in clinical studies [300]. CQ and HCQ are both FDA-approved autophagy inhibitors evaluated in many cancer clinical trials [301]. Therefore, we conclude with information about clinical trials of some autophagy inhibitors, mainly focusing on CQ and HCQ in cancer therapy, aiming to gain a broad understanding of the current research status (Table 2).

8.1. CQ Monotherapy

A recent study investigated the potential efficacy of CQ in improving treatment outcomes for glioblastoma combined with chemoradiation [302,303,304,305]. The study is a Phase II randomized controlled clinical trial (ID: NCT02432417), and January 2025 is the final date for primary outcome measures. Two plans of treatment were included in this trial. Patients in the control group received a standard protocol of radiotherapy (30 daily fractions of 2 Gy or 33 fractions of 1.8 Gy) combined with temozolomide (75 mg/m2) daily, followed by six adjuvant cycles of temozolomide (150–200 mg/m2 daily by mouth). The other group used the same protocol except for the addition of CQ (400 mg/day), starting one week before radiotherapy and continuing until the last day. Nausea and fatigue were commonly reported side effects in this study, and most patients could fully recover after the treatment. In total, there were 11 serious adverse effects documented in eight patients, five of which were deemed unrelated to CQ. Two patients demonstrated a significant lengthening of the ECG QT-corrected interval during the study’s final week of treatment (CTCAE grade III). Although neither of these patients experienced any physical complaints due to cardiac conduction disturbances, more information about the drug safety in this experiment has yet to be updated. Despite the current trials and therapies, glioblastoma remains the most lethal type of brain cancer due to its resistance to surgery, radiation, and chemotherapy, with a median survival of only 14.6 months following diagnosis. Although there was a similarity in overall survival between patients taking CQ and those taking a placebo, the survival rate in the placebo group over time was about half that of the patients taking CQ. As a result, increasing the daily dose of CQ may be necessary to increase the efficacy of the combined therapy with temozolomide [302].

8.2. HCQ Monotherapy

The effectiveness of HCQ in treating patients who were diagnosed with metastatic pancreatic cancer was also evaluated. For instance, a phase II study comprised two arms, with the first arm receiving 400 mg (n = 10) and the second arm receiving 600 mg (n = 10) of HCQ orally twice daily. Patients continued taking the doses unless there were any adverse effects. The primary endpoint was two-month progression-free survival. Two (10%) of the 20 patients showed no disease progression at the two-month mark. The overall survival and median progression-free survival were 46.5 days and 69.0 days, respectively. Elevated alanine aminotransferase (n = 1) and lymphopenia (n = 1) are both grade 4 adverse effects of the treatment. The efficacy and tolerability in these two groups were comparable, which suggests that HCQ monotherapy achieves inconsistent autophagy inhibition and negligible therapeutic efficacy [306]. Thus, when similar clinical trials are to be conducted, further optimization is needed to maximize the effectiveness of HCQ. In another trial, HCQ was combined with dabrafenib (DAB) and trametinib (TRA) in a phase I/II study on mutant melanoma. Patients were randomly assigned to either receive DAB, TRA, and HCQ in combination (experimental group) or receive DAB and TRA with the option of adding HCQ when there was proven tumor progression (control group).

8.3. HCQ with Ulixertinib

Here are more examples of HCQ in the combined therapy for various kinds of tumors. In a current trial, HCQ is being combined with ulixertinib for gastrointestinal cancer [307]. Ulixertinib is a novel, potent ERK inhibitor that selectively blocks the MAPK signaling. It has demonstrated a potent effect in patients with tumors having alterations in that pathway. ClinicalTrials.gov has registered a non-randomized open-label phase II study with ID: NCT05221320. This study is an open-label, multi-center, phase II basket study. Ulixertinib is being administered with HCQ to patients with advanced gastrointestinal malignancy mutations in ERK or MAPK kinase, or who are harboring rat sarcoma virus, a member of the rapidly accelerated fibrosarcoma (non-V600 BRAF). The trial consists of five baskets based on the primary disease, including cholangiocarcinoma (intrahepatic, perihilar, or extrahepatic), pancreatic adenocarcinoma, colorectal adenocarcinoma, oesophagal adenoesophagal carcinoma or oesophagal squaoesophagal carcinoma, and gastroesophageal junction adenocarcinoma or gastric adenocarcinoma. The study starts on 26 May 2022, and is expected to be completed on 19 June 2024. It is being conducted in two stages and involves administering the same oral doses of 450 mg twice/day for ulixertinib and 600 mg twice/day for HCQ. Treatment cycles are repeated every 28 days. The objectives of this trial are to evaluate the response rate to the combination of ulixertinib and HCQ and any associated side effects [307].

8.4. HCQ with Sirolimus/Vorinostat

In the next phase of clinical examinations, sirolimus, vorinostat, and HCQ are investigated for their potential use in patients with advanced cancers. Sirolimus is an mTOR inhibitor approved for use in kidney transplant patients. Vorinostat is a histone deacetylase inhibitor approved by the FDA for cutaneous T-cell lymphoma. A phase I clinical trial continues to evaluate this combination’s safety, its antitumor effects, and the highest tolerable dose in patients with advanced cancers. This study, with ID: NCT01266057, is listed on ClinicalTrials.gov and involves 160 participants. There are two experimental groups in the study. Experimental group 1 comprises HCQ with sirolimus, starting with a dose of HCQ (200 mg/day) and sirolimus (2 mg/day). The cycle duration is 21 days. Experimental group 2 includes HCQ with vorinostat, starting with a dose of HCQ (200 mg/day) and vorinostat (200 mg/day). The cycle duration is also 21 days [308].

8.5. HCQ with Atezolizumab/Cobimetinib

An ongoing study aims to test the safety and efficacy of a combination of atezolizumab, cobimetinib, and HCQ for patients with advanced malignancies harboring a KRAS mutation, which contains errors that promote growth. This study is divided into two phases, with phase I utilizing the time-to-event continuous reassessment method to investigate the safety and the maximum tolerated dose of the combination treatment. The three dose levels involve different combinations of the three drugs. Dose 1 consists of HCQ (600 mg twice/day), cobimetinib (40 mg), and no atezolizumab. Dose 2 involves HCQ (600 mg twice/day), cobimetinib (40 mg), and atezolizumab (840 mg). Lastly, dose 3 consists of HCQ (600 mg twice/day), cobimetinib (60 mg), and atezolizumab (840 mg). All doses are administered orally except for atezolizumab, which is administered intravenously. Phase II will use the recommended phase I dose for three cohorts of KRAS mutation: (1) pancreatic adenocarcinoma, (2) colorectal adenocarcinoma, and (3) histology-agnostic adenocarcinoma. A total of 175 participants are enrolled in this active study, but potential participants are still being recruited, with an estimated end date in September 2024. While cobimetinib and atezolizumab are FDA-approved for other cancers, they have not been used to treat gastrointestinal cancer. The preliminary results have shown that the combination of these drugs effectively kills cancer cells and shrinks tumors in several KRAS-mutated cancers in animals. The second phase of the study will be amended after the preliminary safety and efficacy results from phase I [309].

8.6. HCQ with Paricalcitol

Also, HCQ and paricalcitol can be combined with gemcitabine and nab-paclitaxel in pancreatic cancer treatments. Paricalcitol, a form of vitamin D, blocks a signal in cancer cells responsible for the growth and spreading of the tumor cells. A phase II labeled and single-group assignment trial has been established for this drug combination to estimate its safety and the antitumor effect on metastatic pancreatic tumors. The protocol administration is as follows: HCQ (orally, twice/day), paricalcitol (IV, 3 times/week), and gemcitabine and nab-paclitaxel (IV, over 30 min) on days 1, 8, and 15. This trial is still being recruited and is expected to be completed by August 2024. Additionally, the primary outcome measure is the change in tumor size from baseline as measured by cross-sectional imaging at 8 weeks, while the secondary outcome measures are the incidence of adverse events, progression-free survival with a time frame of up to 3 years from the study start, and overall survival with a time frame up to 3 years from the study start [310].

8.7. HCQ with Abemaciclib

In another trial, abemaciclib (CDK4/6 inhibitor) and HCQ were tested in breast cancer patients. Furthermore, a phase II randomized controlled trial included 66 participants. The study assessed whether using HCQ and abemaciclib in combination to target the disseminated tumor cells (DTCs) in bone marrow could decrease or eliminate their number. Experimental arm A used abemaciclib only (150 mg twice/day), while experimental arm B used abemaciclib (100 mg or 150 mg twice/day) with HCQ (600 mg twice/day). Both drugs were taken orally. The primary outcomes for this study were the incidence of treatment-related adverse events during the first cycle of treatment (4 weeks) and the change in bone marrow DTCs after six cycles of therapy (approximately 6 months) compared to baseline. The study also evaluated the frequency of “clearance” of bone marrow DTCs by the arm after six cycles of treatment. A safety cohort of six patients at each dose of abemaciclib were assessed for protocol-defined “severe toxicity” during cycle 1 [311].

8.8. HCQ with Carfilzomib

Multiple myeloma (MM) is a destructive disease characterized by the secretion of large amounts of monoclonal immunoglobulin and the expansion of bone marrow plasma cells. Proteasome inhibitors have been used to target protein degradation in MM, but patients can become resistant to these drugs, and MM utilizes the autophagy mechanism for protein degradation. Preclinical studies have shown that combining carfilzomib and HCQ increases myeloma cell death and reverses MM cell resistance to carfilzomib. A phase I clinical trial has assessed the safety and efficacy of combination therapy with carfilzomib, dexamethasone, and HCQ in patients with relapsed/refractory MM. In this study, 19 patients were enrolled in a single-arm, dose-escalation trial at two centers, using a 3 + 3 design in five dose levels. All patients received a 14-day run-in with monotherapy of HCQ at their assigned dose level, followed by six 28-day HCQ/carfilzomib/dexamethasone cycles. The primary outcome measure was to estimate the maximum tolerated dose of HCQ when added to the standard-dose regimen of carfilzomib/dexamethasone, with secondary outcome measures including toxicity rates and efficacy assessments. This study was completed in December 2021, and no results have yet been posted on ClinicalTrials.gov as of September 2023. This study has important implications for developing new treatments for relapsed/refractory MM, and it could identify a new drug for patients with this disease [312].

8.9. HCQ with LY3214996

According to clinicaltrials.gov (NCT04386057), a clinical trial is assessing the efficacy and safety of the combination of LY3214996, an ERK inhibitor, and HCQ in patients with advanced pancreatic cancer. The trial is a phase II, open-label, randomized, two-arm study with a safety lead-in. The study has enrolled 52 participants randomized 1:1 to receive either the combination of LY3214996 and HCQ or monotherapy with LY3214996. The primary outcome measure is the disease control rate, while the secondary outcome measures include the objective response rate, progression-free survival, overall survival, and dose-limiting toxicity. Participants are given the drugs on an outpatient basis, and treatment is administered continuously throughout each 28-day cycle. This study’s expected primary completion date is 18 November 2023, while the estimated study completion date is 19 December 2023. The trial was initiated on 27 May 2020, and the US FDA has not approved LY3214996 as a treatment for any disease [313].
Currently, only CQ and HCQ have been examined as autophagy inhibitors in clinical trials for cancer treatment, and most studies are still in phase I or II. Additionally, the clinical benefits of single and combinatorial treatments have not been conclusively demonstrated yet, and many clinical trials are still ongoing or planned. Therefore, more studies should be conducted to investigate the potential positive effect of autophagy inhibitors, providing cancer patients with better treatment options in the future.

9. Conclusions and Perspectives

Autophagy is a fundamental cellular process involved in the degradation and recycling of the components of the cell. It plays a complex, microenvironment-dependent role in cancer. Sometimes, this process protects established tumors in the early stages, presenting a convincing rationale for targeting autophagy in cancer therapy. For this reason, various autophagy inhibitors targeting different steps of the autophagy pathway have been developed and evaluated in preclinical studies and early clinical trials. These inhibitors have shown potential efficacy and initial safety, especially when combined with other drugs. However, numerous questions remain regarding optimal dosing, timing, and integration into multi-modal cancer treatment programs, which need further investigations.
Additionally, personalized medicine is an emerging approach to maximize autophagy inhibition therapy by tailoring strategies to specific cancer biochemical profiles and genotypes. Furthermore, the study of autophagy and drug-targeting opportunities is driven by developing new technologies and methodologies like high-throughput drug screening, monitoring autophagy signaling, advanced microscopy, genetic analysis, and computational approaches. While substantial progress has clarified the role of autophagy in cancer and paved the way for therapeutic targeting, significant research gaps remain to be addressed. Continued efforts to improve the integration of autophagy manipulation into oncology, guided by both emerging science and ethical principles, will be key steps for realizing the full potential of this promising new approach and improving cancer treatment and outcomes.

Author Contributions

Conceptualization, C.H.; methodology, A.M.I.A.H.; validation, Y.Z.; formal analysis, A.M.I.A.H. and Y.Z.; investigation, A.M.I.A.H. and Y.Z.; resources, A.M.I.A.H.; writing—original draft preparation, A.M.I.A.H.; writing—review and editing, A.M.I.A.H., Y.Z., X.C. and C.H.; supervision, C.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by The Science and Technology Development Fund (005/2023/SKL, 0081/2021/A2). The APC was funded by 005/2023/SKL and 0081/2021/A2.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ATG, autophagy-related genes; TNBC, triple-negative breast cancer; LC3, the tubule-linked protein 1-light chain 3; AMPK, AMP-activated kinase; mTOR, mechanistic target of rapamycin kinase; 3-MA, 3-methyladenine; spautin-1, specific and potent autophagy inhibitor-1; CQ, chloroquine; HCQ, hydroxychloroquin; PI3K, phosphoinositide 3-kinase; CPT, camptothecin; PSD, Pulsatilla saponin D; Baf A1, bafilomycin A1; RNAi, RNA interference; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; AZA, azacitidine; CML, chronic myeloid leukemia; IM, imatinib mesylate; siRNA, small interfering RNA; MM, multiple myeloma; DAB, dabrafenib; TRA, trametinib; ROS, reactive oxygen species; 5-FU, 5-fluorouracil; DTCs, disseminated tumor cells TCM, traditional chinese medicine; TSN, toosendanin; ULK1, Unc-51-like kinase 1.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  2. Wang, J.; Yu, F.; Shang, Y.; Ping, Z.; Liu, L. Thyroid Cancer: Incidence and Mortality Trends in China, 2005–2015. Endocrine 2020, 68, 163–173. [Google Scholar] [CrossRef]
  3. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  4. Su, Z.; Xu, T.; Wang, Y.; Guo, X.; Tu, J.; Zhang, D.; Kong, X.; Sheng, Y.; Sun, W. Low-intensity Pulsed Ultrasound Promotes Apoptosis and Inhibits Angiogenesis via P38 Signaling-mediated Endoplasmic Reticulum Stress in Human Endothelial Cells. Mol. Med. Rep. 2019, 19, 4645–4654. [Google Scholar] [CrossRef]
  5. Gotwals, P.; Cameron, S.; Cipolletta, D.; Cremasco, V.; Crystal, A.; Hewes, B.; Mueller, B.; Quaratino, S.; Sabatos-Peyton, C.; Petruzzelli, L.; et al. Prospects for Combining Targeted and Conventional Cancer Therapy with Immunotherapy. Nat. Rev. Cancer 2017, 17, 286–301. [Google Scholar] [CrossRef]
  6. Wang, P.; Leung, A.W.; Xu, C. Low-Intensity Ultrasound-Induced Cellular Destruction and Autophagy of Nasopharyngeal Carcinoma Cells. Exp. Ther. Med. 2011, 2, 849–852. [Google Scholar] [CrossRef]
  7. Weiss, F.; Lauffenburger, D.; Friedl, P. Towards Targeting of Shared Mechanisms of Cancer Metastasis and Therapy Resistance. Nat. Rev. Cancer 2022, 22, 157–173. [Google Scholar] [CrossRef]
  8. 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]
  9. Yang, M.H.; Ha, I.J.; Lee, S.-G.; Lee, J.; Um, J.-Y.; Sethi, G.; Ahn, K.S. Brassinin Induces Apoptosis, Autophagy, and Paraptosis via MAPK Signaling Pathway Activation in Chronic Myelogenous Leukemia Cells. Biology 2023, 12, 307. [Google Scholar] [CrossRef]
  10. Kouroumalis, E.; Voumvouraki, A.; Augoustaki, A.; Samonakis, D.N. Autophagy in Liver Diseases. World J. Hepatol. 2021, 13, 6–65. [Google Scholar] [CrossRef]
  11. Onorati, A.V.; Dyczynski, M.; Ojha, R.; Amaravadi, R.K. Targeting Autophagy in Cancer. Cancer 2018, 124, 3307–3318. [Google Scholar] [CrossRef]
  12. Zhou, X.-H.; Kang, J.; Zhong, Z.-D.; Cheng, Y. Osthole Induces Apoptosis of the HT-29 Cells via Endoplasmic Reticulum Stress and Autophagy. Oncol. Lett. 2021, 22, 726. [Google Scholar] [CrossRef]
  13. Yamamoto, H.; Matsui, T. Molecular Mechanisms of Macroautophagy, Microautophagy, and Chaperone-Mediated Autophagy. J. Nippon Med. Sch. 2023, 91, 2–9. [Google Scholar] [CrossRef]
  14. de Duve, C. The Lysosome. Sci. Am. 1963, 208, 64–73. [Google Scholar] [CrossRef]
  15. Levine, B.; Kroemer, G. Autophagy in the Pathogenesis of Disease. Cell 2008, 132, 27–42. [Google Scholar] [CrossRef]
  16. 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]
  17. White, E. Deconvoluting the Context-Dependent Role for Autophagy in Cancer. Nat. Rev. Cancer 2012, 12, 401–410. [Google Scholar] [CrossRef]
  18. Thein, O.S.; Ali, N.A.; Mahida, R.Y.; Dancer, R.C.A.; Ostermann, M.; Amrein, K.; Martucci, G.; Scott, A.; Thickett, D.R.; Parekh, D. Raised FGF23 Correlates to Increased Mortality in Critical Illness, Independent of Vitamin D. Biology 2023, 12, 309. [Google Scholar] [CrossRef]
  19. Comità, S.; Rubeo, C.; Giordano, M.; Penna, C.; Pagliaro, P. Pathways for Cardioprotection in Perspective: Focus on Remote Conditioning and Extracellular Vesicles. Biology 2023, 12, 308. [Google Scholar] [CrossRef]
  20. Amaravadi, R.K.; Lippincott-Schwartz, J.; Yin, X.-M.; Weiss, W.A.; Takebe, N.; Timmer, W.; DiPaola, R.S.; Lotze, M.T.; White, E. Principles and Current Strategies for Targeting Autophagy for Cancer Treatment. Clin. Cancer Res. 2011, 17, 654–666. [Google Scholar] [CrossRef]
  21. Mizushima, N.; Komatsu, M. Autophagy: Renovation of Cells and Tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef]
  22. Kaushik, S.; Singh, R.; Cuervo, A.M. Autophagic Pathways and Metabolic Stress. Diabetes Obes. Metab. 2010, 12, 4–14. [Google Scholar] [CrossRef]
  23. Galluzzi, L.; Bravo-San Pedro, J.M.; Levine, B.; Green, D.R.; Kroemer, G. Pharmacological Modulation of Autophagy: Therapeutic Potential and Persisting Obstacles. Nat. Rev. Drug Discov. 2017, 16, 487–511. [Google Scholar] [CrossRef]
  24. Baginska, J.; Viry, E.; Berchem, G.; Poli, A.; Noman, M.Z.; van Moer, K.; Medves, S.; Zimmer, J.; Oudin, A.; Niclou, S.P.; et al. Granzyme B Degradation by Autophagy Decreases Tumor Cell Susceptibility to Natural Killer-Mediated Lysis under Hypoxia. Proc. Natl. Acad. Sci. USA 2013, 110, 17450–17455. [Google Scholar] [CrossRef]
  25. Kaushik, S.; Cuervo, A.M. The Coming of Age of Chaperone-Mediated Autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 365–381. [Google Scholar] [CrossRef]
  26. 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]
  27. Mizushima, N. Autophagy: Process and Function. Genes. Dev. 2007, 21, 2861–2873. [Google Scholar] [CrossRef]
  28. Russell, R.C.; Tian, Y.; Yuan, H.; Park, H.W.; Chang, Y.-Y.; Kim, J.; Kim, H.; Neufeld, T.P.; Dillin, A.; Guan, K.-L. ULK1 Induces Autophagy by Phosphorylating Beclin-1 and Activating VPS34 Lipid Kinase. Nat. Cell Biol. 2013, 15, 741–750. [Google Scholar] [CrossRef]
  29. Mercer, C.A.; Kaliappan, A.; Dennis, P.B. A Novel, Human Atg13 Binding Protein, Atg101, Interacts with ULK1 and Is Essential for Macroautophagy. Autophagy 2009, 5, 649–662. [Google Scholar] [CrossRef]
  30. Matsubara, Y.; Kawasaki, I.; Urushiyama, S.; Yasuda, T.; Shirakata, M.; Iino, Y.; Shibuya, H.; Yamanashi, Y. The Adaptor-like Protein ROG-1 Is Required for Activation of the Ras-MAP Kinase Pathway and Meiotic Cell Cycle Progression in Caenorhabditis Elegans. Genes Cells 2007, 12, 407–420. [Google Scholar] [CrossRef]
  31. Mizushima, N.; Noda, T.; Yoshimori, T.; Tanaka, Y.; Ishii, T.; George, M.D.; Klionsky, D.J.; Ohsumi, M.; Ohsumi, Y. A Protein Conjugation System Essential for Autophagy. Nature 1998, 395, 395–398. [Google Scholar] [CrossRef]
  32. Eskelinen, E.-L. Maturation of Autophagic Vacuoles in Mammalian Cells. Autophagy 2005, 1, 1–10. [Google Scholar] [CrossRef]
  33. 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]
  34. Lindmo, K.; Stenmark, H. Regulation of Membrane Traffic by Phosphoinositide 3-Kinases. J. Cell Sci. 2006, 119, 605–614. [Google Scholar] [CrossRef]
  35. 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]
  36. 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]
  37. El-Gowily, A.H.; Loutfy, S.A.; Ali, E.M.M.; Mohamed, T.M.; Mansour, M.A. Tioconazole and Chloroquine Act Synergistically to Combat Doxorubicin-Induced Toxicity via Inactivation of PI3K/AKT/MTOR Signaling Mediated ROS-Dependent Apoptosis and Autophagic Flux Inhibition in MCF-7 Breast Cancer Cells. Pharmaceuticals 2021, 14, 254. [Google Scholar] [CrossRef]
  38. 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]
  39. Agarwal, S.; Hartz, A.M.S.; 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]
  40. He, C.; Klionsky, D.J. Regulation Mechanisms and Signaling Pathways of Autophagy. Annu. Rev. Genet. 2009, 43, 67–93. [Google Scholar] [CrossRef]
  41. 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] [PubMed]
  42. Kim, J.; Guan, K.-L. MTOR as a Central Hub of Nutrient Signalling and Cell Growth. Nat. Cell Biol. 2019, 21, 63–71. [Google Scholar] [CrossRef] [PubMed]
  43. Laplante, M.; Sabatini, D.M. MTOR Signaling. Cold Spring Harb. Perspect. Biol. 2012, 4, a011593. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, J.; Kundu, M.; Viollet, B.; Guan, K.-L. AMPK and MTOR Regulate Autophagy through Direct Phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [PubMed]
  45. Herzig, S.; Shaw, R.J. AMPK: Guardian of Metabolism and Mitochondrial Homeostasis. Nat. Rev. Mol. Cell Biol. 2018, 19, 121–135. [Google Scholar] [CrossRef] [PubMed]
  46. Hardie, D.G.; Schaffer, B.E.; Brunet, A. AMPK: An Energy-Sensing Pathway with Multiple Inputs and Outputs. Trends Cell Biol. 2016, 26, 190–201. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, S.; Li, H.; Yuan, M.; Fan, H.; Cai, Z. Role of AMPK in Autophagy. Front. Physiol. 2022, 13, 1015500. [Google Scholar] [CrossRef]
  48. Zhang, D.; Wang, W.; Sun, X.; Xu, D.; Wang, C.; Zhang, Q.; Wang, H.; Luo, W.; Chen, Y.; Chen, H.; et al. AMPK Regulates Autophagy by Phosphorylating BECN1 at Threonine 388. Autophagy 2016, 12, 1447–1459. [Google Scholar] [CrossRef]
  49. Chan, E.Y.W.; Longatti, A.; McKnight, N.C.; Tooze, S.A. Kinase-Inactivated ULK Proteins Inhibit Autophagy via Their Conserved C-Terminal Domains Using an Atg13-Independent Mechanism. Mol. Cell Biol. 2009, 29, 157–171. [Google Scholar] [CrossRef]
  50. 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]
  51. Eisenberg-Lerner, A.; Kimchi, A. The Paradox of Autophagy and Its Implication in Cancer Etiology and Therapy. Apoptosis 2009, 14, 376–391. [Google Scholar] [CrossRef] [PubMed]
  52. Thorburn, A.; Thamm, D.H.; Gustafson, D.L. Autophagy and Cancer Therapy. Mol. Pharmacol. 2014, 85, 830–838. [Google Scholar] [CrossRef] [PubMed]
  53. Denton, D.; Kumar, S. Autophagy-Dependent Cell Death. Cell Death Differ. 2019, 26, 605–616. [Google Scholar] [CrossRef] [PubMed]
  54. Fulda, S.; Kögel, D. Cell Death by Autophagy: Emerging Molecular Mechanisms and Implications for Cancer Therapy. Oncogene 2015, 34, 5105–5113. [Google Scholar] [CrossRef] [PubMed]
  55. Nagelkerke, A.; Bussink, J.; Geurts-Moespot, A.; Sweep, F.C.G.J.; 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]
  56. 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] [PubMed]
  57. 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] [PubMed]
  58. 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]
  59. 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]
  60. Song, P.; Li, Y.; Dong, Y.; Liang, Y.; Qu, H.; Qi, D.; Lu, Y.; Jin, X.; Guo, Y.; Jia, Y.; et al. Estrogen Receptor β Inhibits Breast Cancer Cells Migration and Invasion through CLDN6-Mediated Autophagy. J. Exp. Clin. Cancer Res. 2019, 38, 354. [Google Scholar] [CrossRef]
  61. 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]
  62. 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] [PubMed]
  63. Lorin, S.; Hamaï, A.; Mehrpour, M.; Codogno, P. Autophagy Regulation and Its Role in Cancer. Semin. Cancer Biol. 2013, 23, 361–379. [Google Scholar] [CrossRef] [PubMed]
  64. 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]
  65. Burada, F. Autophagy in Colorectal Cancer: An Important Switch from Physiology to Pathology. World J. Gastrointest. Oncol. 2015, 7, 271. [Google Scholar] [CrossRef] [PubMed]
  66. 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]
  67. Liu, T.; Liu, X.; Li, W. Tetrandrine, a Chinese Plant-Derived Alkaloid, Is a Potential Candidate for Cancer Chemotherapy. Oncotarget 2016, 7, 40800–40815. [Google Scholar] [CrossRef] [PubMed]
  68. Klionsky, D.J.; Emr, S.D. Autophagy as a Regulated Pathway of Cellular Degradation. Science 2000, 290, 1717–1721. [Google Scholar] [CrossRef] [PubMed]
  69. Mizushima, N.; Levine, B. Autophagy in Mammalian Development and Differentiation. Nat. Cell Biol. 2010, 12, 823–830. [Google Scholar] [CrossRef]
  70. White, E.; DiPaola, R.S. The Double-Edged Sword of Autophagy Modulation in Cancer. Clin. Cancer Res. 2009, 15, 5308–5316. [Google Scholar] [CrossRef]
  71. 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]
  72. Liu, T.; Men, Q.; Wu, G.; Yu, C.; Huang, Z.; Liu, X.; Li, W. Tetrandrine Induces Autophagy and Differentiation by Activating ROS and Notch1 Signaling in Leukemia Cells. Oncotarget 2015, 6, 7992–8006. [Google Scholar] [CrossRef] [PubMed]
  73. Huang, A.-C.; Lien, J.-C.; Lin, M.-W.; Yang, J.-S.; Wu, P.-P.; Chang, S.-J.; Lai, T.-Y. Tetrandrine Induces Cell Death in SAS Human Oral Cancer Cells through Caspase Activation-Dependent Apoptosis and LC3-I and LC3-II Activation-Dependent Autophagy. Int. J. Oncol. 2013, 43, 485–494. [Google Scholar] [CrossRef]
  74. 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] [PubMed]
  75. 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]
  76. Singh, S.S.; Vats, S.; Chia, A.Y.-Q.; Tan, T.Z.; Deng, S.; Ong, M.S.; Arfuso, F.; Yap, C.T.; Goh, B.C.; Sethi, G.; et al. Dual Role of Autophagy in Hallmarks of Cancer. Oncogene 2018, 37, 1142–1158. [Google Scholar] [CrossRef]
  77. Hikita, H.; Sakane, S.; Takehara, T. Mechanisms of the Autophagosome-Lysosome Fusion Step and Its Relation to Non-Alcoholic Fatty Liver Disease. Liver Res. 2018, 2, 120–124. [Google Scholar] [CrossRef]
  78. Cui, B.; Yu, J.-M. Autophagy: A New Pathway for Traditional Chinese Medicine. J. Asian Nat. Prod. Res. 2018, 20, 14–26. [Google Scholar] [CrossRef] [PubMed]
  79. Koh, S.-H.; Lo, E.H. The Role of the PI3K Pathway in the Regeneration of the Damaged Brain by Neural Stem Cells after Cerebral Infarction. J. Clin. Neurol. 2015, 11, 297. [Google Scholar] [CrossRef]
  80. 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]
  81. Chen, P.; Dai, C.-H.; Shi, Z.-H.; Wang, Y.; Wu, J.-N.; Chen, K.; Su, J.-Y.; Li, J. Synergistic Inhibitory Effect of Berberine and Icotinib on Non-Small Cell Lung Cancer Cells via Inducing Autophagic Cell Death and Apoptosis. Apoptosis 2021, 26, 639–656. [Google Scholar] [CrossRef]
  82. Farkas, T.; Daugaard, M.; Jäättelä, M. Identification of Small Molecule Inhibitors of Phosphatidylinositol 3-Kinase and Autophagy. J. Biol. Chem. 2011, 286, 38904–38912. [Google Scholar] [CrossRef]
  83. Liu, J.; Liu, Y.; Meng, L.; Ji, B.; Yang, D. Synergistic Antitumor Effect of Sorafenib in Combination with ATM Inhibitor in Hepatocellular Carcinoma Cells. Int. J. Med. Sci. 2017, 14, 523–529. [Google Scholar] [CrossRef]
  84. Misumi, K.; Sun, J.; Kinomura, A.; Miyata, Y.; Okada, M.; Tashiro, S. Enhanced Gefitinib-induced Repression of the Epidermal Growth Factor Receptor Pathway by Ataxia Telangiectasia-mutated Kinase Inhibition in Non-small-cell Lung Cancer Cells. Cancer Sci. 2016, 107, 444–451. [Google Scholar] [CrossRef] [PubMed]
  85. Castro-Guijarro, A.C.; Vanderhoeven, F.; Mondaca, J.M.; Redondo, A.L.; Zoppino, F.C.M.; Fernandez-Muñoz, J.M.; Sanchez, A.M.; Flamini, M.I. Combination Treatment of Retinoic Acid Plus Focal Adhesion Kinase Inhibitor Prevents Tumor Growth and Breast Cancer Cell Metastasis. Cells 2022, 11, 2988. [Google Scholar] [CrossRef]
  86. Merzoug-Larabi, M.; Spasojevic, C.; Eymard, M.; Hugonin, C.; Auclair, C.; Karam, M. Protein Kinase C Inhibitor Gö6976 but Not Gö6983 Induces the Reversion of E- to N-Cadherin Switch and Metastatic Phenotype in Melanoma: Identification of the Role of Protein Kinase D1. BMC Cancer 2017, 17, 12. [Google Scholar] [CrossRef] [PubMed]
  87. Aaltonen, V.; Koivunen, J.; Laato, M.; Peltonen, J. PKC Inhibitor Go6976 Induces Mitosis and Enhances Doxorubicin-Paclitaxel Cytotoxicity in Urinary Bladder Carcinoma Cells. Cancer Lett. 2007, 253, 97–107. [Google Scholar] [CrossRef]
  88. Roy, S.; Eastman, A.; Gribble, G.W. Synthesis of 7-Keto-Goe6976 (ICP-103). ChemInform 2005, 36, 595–601. [Google Scholar] [CrossRef]
  89. Dickstein, R.J.; Nitti, G.; Dinney, C.P.; Davies, B.R.; Kamat, A.M.; McConkey, D.J. Autophagy Limits the Cytotoxic Effects of the AKT Inhibitor AZ7328 in Human Bladder Cancer Cells. Cancer Biol. Ther. 2012, 13, 1325–1338. [Google Scholar] [CrossRef]
  90. Lamoureux, F.; Thomas, C.; Crafter, C.; Kumano, M.; Zhang, F.; Davies, B.R.; Gleave, M.E.; Zoubeidi, A. Blocked Autophagy Using Lysosomotropic Agents Sensitizes Resistant Prostate Tumor Cells to the Novel Akt Inhibitor AZD5363. Clin. Cancer Res. 2013, 19, 833–844. [Google Scholar] [CrossRef]
  91. Tan, J.Y.; Jia, L.Q.; Shi, W.H.; He, Q.; Zhu, L.; Yu, B. Rab5a-Mediated Autophagy Regulates the Phenotype and Behavior of Vascular Smooth Muscle Cells. Mol. Med. Rep. 2016, 14, 4445–4453. [Google Scholar] [CrossRef] [PubMed]
  92. Peng, P.; Zhang, X.; Qi, T.; Cheng, H.; Kong, Q.; Liu, L.; Cao, X.; Ding, Z. Alpha-lipoic Acid Inhibits Lung Cancer Growth via MTOR-mediated Autophagy Inhibition. FEBS Open Bio 2020, 10, 607–618. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, J.; Xia, H.; Kim, M.; Xu, L.; Li, Y.; Zhang, L.; Cai, Y.; Norberg, H.V.; Zhang, T.; Furuya, T.; et al. Beclin1 Controls the Levels of P53 by Regulating the Deubiquitination Activity of USP10 and USP13. Cell 2011, 147, 223–234. [Google Scholar] [CrossRef]
  94. 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] [PubMed]
  95. Liao, Y.; Guo, Z.; Xia, X.; Liu, Y.; Huang, C.; Jiang, L.; Wang, X.; Liu, J.; Huang, H. Inhibition of EGFR Signaling with Spautin-1 Represents a Novel Therapeutics for Prostate Cancer. J. Exp. Clin. Cancer Res. 2019, 38, 157. [Google Scholar] [CrossRef] [PubMed]
  96. Guo, J.; Zhang, J.; Liang, L.; Liu, N.; Qi, M.; Zhao, S.; Su, J.; Liu, J.; Peng, C.; Chen, X.; et al. Potent USP10/13 Antagonist Spautin-1 Suppresses Melanoma Growth via ROS-mediated DNA Damage and Exhibits Synergy with Cisplatin. J. Cell Mol. Med. 2020, 24, 4324–4340. [Google Scholar] [CrossRef] [PubMed]
  97. DeFrances, M.C.; Debelius, D.R.; Cheng, J.; Kane, L.P. Inhibition of T-Cell Activation by PIK3IP1. Eur. J. Immunol. 2012, 42, 2754–2759. [Google Scholar] [CrossRef] [PubMed]
  98. Dowdle, W.E.; Nyfeler, B.; Nagel, J.; Elling, R.A.; Liu, S.; Triantafellow, E.; Menon, S.; Wang, Z.; Honda, A.; Pardee, G.; et al. Selective VPS34 Inhibitor Blocks Autophagy and Uncovers a Role for NCOA4 in Ferritin Degradation and Iron Homeostasis in Vivo. Nat. Cell Biol. 2014, 16, 1069–1079. [Google Scholar] [CrossRef]
  99. Baquero, P.; Dawson, A.; Mukhopadhyay, A.; Kuntz, E.M.; Mitchell, R.; Olivares, O.; Ianniciello, A.; Scott, M.T.; Dunn, K.; Nicastri, M.C.; et al. Targeting Quiescent Leukemic Stem Cells Using Second Generation Autophagy Inhibitors. Leukemia 2019, 33, 981–994. [Google Scholar] [CrossRef]
  100. Pasquier, B.; El-Ahmad, Y.; Filoche-Rommé, B.; Dureuil, C.; Fassy, F.; Abecassis, P.-Y.; Mathieu, M.; Bertrand, T.; Benard, T.; Barrière, C.; et al. Discovery of (2S)-8-[(3R)-3-Methylmorpholin-4-Yl]-1-(3-Methyl-2-Oxobutyl)-2-(Trifluoromethyl)-3,4-Dihydro-2H-Pyrimido[1,2-a]Pyrimidin-6-One: A Novel Potent and Selective Inhibitor of VPS34 for the Treatment of Solid Tumors. J. Med. Chem. 2015, 58, 376–400. [Google Scholar] [CrossRef]
  101. Banti, C.N.; Hadjikakou, S.K.; Sismanoglu, T.; Hadjiliadis, N. Anti-Proliferative and Antitumor Activity of Organotin(IV) Compounds. An Overview of the Last Decade and Future Perspectives. J. Inorg. Biochem. 2019, 194, 114–152. [Google Scholar] [CrossRef] [PubMed]
  102. 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]
  103. Bago, R.; Malik, N.; Munson, M.J.; Prescott, A.R.; Davies, P.; Sommer, E.; Shpiro, N.; Ward, R.; Cross, D.; Ganley, I.G.; et al. Characterization of VPS34-IN1, a Selective Inhibitor of VPS34, Reveals That the Phosphatidylinositol 3-Phosphate-Binding SGK3 Protein Kinase Is a Downstream Target of Class III Phosphoinositide 3-Kinase. Biochem. J. 2014, 463, 413–427. [Google Scholar] [CrossRef]
  104. Ronan, B.; Flamand, O.; Vescovi, L.; Dureuil, C.; Durand, L.; Fassy, F.; Bachelot, M.-F.; Lamberton, A.; Mathieu, M.; Bertrand, T.; et al. A Highly Potent and Selective VPS34 Inhibitor Alters Vesicle Trafficking and Autophagy. Nat. Chem. Biol. 2014, 10, 1013–1019. [Google Scholar] [CrossRef] [PubMed]
  105. Lin, C.; Tsao, Y.; Shu, C. Autophagy Modulation as a Potential Targeted Cancer Therapy: From Drug Repurposing to New Drug Development. Kaohsiung J. Med. Sci. 2021, 37, 166–171. [Google Scholar] [CrossRef]
  106. Carew, J.S.; Espitia, C.M.; Esquivel, J.A.; Mahalingam, D.; Kelly, K.R.; Reddy, G.; Giles, F.J.; Nawrocki, S.T. Lucanthone Is a Novel Inhibitor of Autophagy That Induces Cathepsin D-Mediated Apoptosis. J. Biol. Chem. 2011, 286, 6602–6613. [Google Scholar] [CrossRef] [PubMed]
  107. Radin, D.P.; Smith, G.; Moushiaveshi, V.; Wolf, A.; Bases, R.; Tsirka, S.E. Lucanthone Targets Lysosomes to Perturb Glioma Proliferation, Chemoresistance and Stemness, and Slows Tumor Growth In Vivo. Front. Oncol. 2022, 12, 852940. [Google Scholar] [CrossRef]
  108. Liu, M.; Bamodu, O.A.; Huang, W.-C.; Zucha, M.A.; Lin, Y.-K.; Wu, A.T.H.; Huang, C.-C.; Lee, W.-H.; Yuan, C.-C.; Hsiao, M.; et al. 4-Acetylantroquinonol B Suppresses Autophagic Flux and Improves Cisplatin Sensitivity in Highly Aggressive Epithelial Cancer through the PI3K/Akt/MTOR/P70S6K Signaling Pathway. Toxicol. Appl. Pharmacol. 2017, 325, 48–60. [Google Scholar] [CrossRef]
  109. De Mei, C.; Ercolani, L.; Parodi, C.; Veronesi, M.; Lo Vecchio, C.; Bottegoni, G.; Torrente, E.; Scarpelli, R.; Marotta, R.; Ruffili, R.; et al. Dual Inhibition of REV-ERBβ and Autophagy as a Novel Pharmacological Approach to Induce Cytotoxicity in Cancer Cells. Oncogene 2015, 34, 2597–2608. [Google Scholar] [CrossRef]
  110. Torrente, E.; Parodi, C.; Ercolani, L.; De Mei, C.; Ferrari, A.; Scarpelli, R.; Grimaldi, B. Synthesis and in Vitro Anticancer Activity of the First Class of Dual Inhibitors of REV-ERBβ and Autophagy. J. Med. Chem. 2015, 58, 5900–5915. [Google Scholar] [CrossRef]
  111. Xu, Z.; Chen, X.; Zhong, Z.; Chen, L.; Wang, Y. Ganoderma Lucidum Polysaccharides: Immunomodulation and Potential Anti-Tumor Activities. Am. J. Chin. Med. 2011, 39, 15–27. [Google Scholar] [CrossRef]
  112. Wang, Y.; Fan, X.; Wu, X. Ganoderma lucidum Polysaccharide (GLP) Enhances Antitumor Immune Response by Regulating Differentiation and Inhibition of MDSCs via a CARD9-NF-ΚB-IDO Pathway. Biosci. Rep. 2020, 40, BSR20201170. [Google Scholar] [CrossRef] [PubMed]
  113. Pan, H.; Wang, Y.; Na, K.; Wang, Y.; Wang, L.; Li, Z.; Guo, C.; Guo, D.; Wang, X. Autophagic Flux Disruption Contributes to Ganoderma Lucidum Polysaccharide-Induced Apoptosis in Human Colorectal Cancer Cells via MAPK/ERK Activation. Cell Death Dis. 2019, 10, 456. [Google Scholar] [CrossRef] [PubMed]
  114. Laraia, L.; Garivet, G.; Foley, D.J.; Kaiser, N.; Müller, S.; Zinken, S.; Pinkert, T.; Wilke, J.; Corkery, D.; Pahl, A.; et al. Image-Based Morphological Profiling Identifies a Lysosomotropic, Iron-Sequestering Autophagy Inhibitor. Angew. Chem. Int. Ed. 2020, 59, 5721–5729. [Google Scholar] [CrossRef] [PubMed]
  115. Rodilla, A.M.; Korrodi-Gregório, L.; Hernando, E.; Manuel-Manresa, P.; Quesada, R.; Pérez-Tomás, R.; Soto-Cerrato, V. Synthetic Tambjamine Analogues Induce Mitochondrial Swelling and Lysosomal Dysfunction Leading to Autophagy Blockade and Necrotic Cell Death in Lung Cancer. Biochem. Pharmacol. 2017, 126, 23–33. [Google Scholar] [CrossRef] [PubMed]
  116. Wu, M.-Y.; Wang, S.-F.; Cai, C.-Z.; Tan, J.-Q.; Li, M.; Lu, J.-J.; Chen, X.-P.; Wang, Y.-T.; Zheng, W.; Lu, J.-H. Natural Autophagy Blockers, Dauricine (DAC) and Daurisoline (DAS), Sensitize Cancer Cells to Camptothecin-Induced Toxicity. Oncotarget 2017, 8, 77673–77684. [Google Scholar] [CrossRef]
  117. Fu, R.; Deng, Q.; Zhang, H.; Hu, X.; Li, Y.; Liu, Y.; Hu, J.; Luo, Q.; Zhang, Y.; Jiang, X.; et al. A Novel Autophagy Inhibitor Berbamine Blocks SNARE-Mediated Autophagosome-Lysosome Fusion through Upregulation of BNIP3. Cell Death Dis. 2018, 9, 243. [Google Scholar] [CrossRef]
  118. 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. [Google Scholar] [CrossRef] [PubMed]
  119. Hanson, T.L.; Klopfenstein, T. Monensin, Protein Source and Protein Levels for Growing Steers. J. Anim. Sci. 1979, 48, 474–479. [Google Scholar] [CrossRef]
  120. Markowska, A.; Kaysiewicz, J.; Markowska, J.; Huczyński, A. Doxycycline, Salinomycin, Monensin and Ivermectin Repositioned as Cancer Drugs. Bioorg. Med. Chem. Lett. 2019, 29, 1549–1554. [Google Scholar] [CrossRef]
  121. Kim, S.-H.; Kim, K.-Y.; Yu, S.-N.; Park, S.-G.; Yu, H.-S.; Seo, Y.-K.; Ahn, S.-C. Monensin Induces PC-3 Prostate Cancer Cell Apoptosis via ROS Production and Ca2+ Homeostasis Disruption. Anticancer Res. 2016, 36, 5835–5844. [Google Scholar] [CrossRef] [PubMed]
  122. Miura, K.; Kawano, S.; Suto, T.; Sato, T.; Chida, N.; Simizu, S. Identification of Madangamine A as a Novel Lysosomotropic Agent to Inhibit Autophagy. Bioorg. Med. Chem. 2021, 34, 116041. [Google Scholar] [CrossRef] [PubMed]
  123. Zhao, X.; Fang, Y.; Yang, Y.; Qin, Y.; Wu, P.; Wang, T.; Lai, H.; Meng, L.; Wang, D.; Zheng, Z.; et al. Elaiophylin, a Novel Autophagy Inhibitor, Exerts Antitumor Activity as a Single Agent in Ovarian Cancer Cells. Autophagy 2015, 11, 1849–1863. [Google Scholar] [CrossRef] [PubMed]
  124. Zhou, J.; Huang, X.; Qiu, H.; Gan, R.; Zhou, H.; Zhu, H.; Zhang, X.; Lu, G.; Liang, G. SSPH I, a Novel Anticancer Saponin, Inhibits Autophagy and Induces Apoptosis via ROS Accumulation and ERK1/2 Signaling Pathway in Hepatocellular Carcinoma Cells. Onco Targets Ther. 2020, 13, 5979–5991. [Google Scholar] [CrossRef] [PubMed]
  125. Zhou, J.; Li, G.; Zheng, Y.; Shen, H.-M.; Hu, X.; Ming, Q.-L.; Huang, C.; Li, P.; Gao, N. A Novel Autophagy/Mitophagy Inhibitor Liensinine Sensitizes Breast Cancer Cells to Chemotherapy through DNM1L-Mediated Mitochondrial Fission. Autophagy 2015, 11, 1259–1279. [Google Scholar] [CrossRef] [PubMed]
  126. Qi, H.; Jiang, Z.; Wang, C.; Yang, Y.; Li, L.; He, H.; Yu, Z. Sensitization of Tamoxifen-Resistant Breast Cancer Cells by Z-Ligustilide through Inhibiting Autophagy and Accumulating DNA Damages. Oncotarget 2017, 8, 29300–29317. [Google Scholar] [CrossRef]
  127. Salari, Z.; Khosravi, A.; Pourkhandani, E.; Molaakbari, E.; Salarkia, E.; Keyhani, A.; Sharifi, I.; Tavakkoli, H.; Sohbati, S.; Dabiri, S.; et al. The Inhibitory Effect of 6-Gingerol and Cisplatin on Ovarian Cancer and Antitumor Activity: In Silico, in Vitro, and in Vivo. Front. Oncol. 2023, 13, 1098429. [Google Scholar] [CrossRef] [PubMed]
  128. Nazim, U.; Jeong, J.-K.; Seol, J.-W.; Hur, J.; Eo, S.-K.; Lee, J.-H.; Park, S.-Y. Inhibition of the Autophagy Flux by Gingerol Enhances TRAIL-Induced Tumor Cell Death. Oncol. Rep. 2015, 33, 2331–2336. [Google Scholar] [CrossRef] [PubMed]
  129. Sang, J.; Gan, L.; Zou, M.-F.; Lin, Z.-J.; Fan, R.-Z.; Huang, J.-L.; Li, W.; Tang, G.-H.; Yin, S. Jolkinolide B Sensitizes Bladder Cancer to MTOR Inhibitors via Dual Inhibition of Akt Signaling and Autophagy. Cancer Lett. 2022, 526, 352–362. [Google Scholar] [CrossRef]
  130. Won, G.; Jung, J.H.; Sohn, E.J.; Park, J.E.; Kim, H.; Lee, H.-J.; Shim, B.S.; Kim, S.-H. Misaponin B Induces G2/M Arrest, Cytokinesis Failure and Impairs Autophagy. BioMed Res. Int. 2020, 2020, 5925094. [Google Scholar] [CrossRef]
  131. Bilir, A.; Erguven, M.; Yazihan, N.; Aktas, E.; Oktem, G.; Sabanci, A. Enhancement of Vinorelbine-Induced Cytotoxicity and Apoptosis by Clomipramine and Lithium Chloride in Human Neuroblastoma Cancer Cell Line SH-SY5Y. J. Neurooncol. 2010, 100, 385–395. [Google Scholar] [CrossRef] [PubMed]
  132. Zeki, A.A.; Yeganeh, B.; Kenyon, N.J.; Post, M.; Ghavami, S. Autophagy in Airway Diseases: A New Frontier in Human Asthma? Allergy 2016, 71, 5–14. [Google Scholar] [CrossRef]
  133. Rossi, M.; Munarriz, E.R.; Bartesaghi, S.; Milanese, M.; Dinsdale, D.; Guerra-Martin, M.A.; Bampton, E.T.W.; Glynn, P.; Bonanno, G.; Knight, R.A.; et al. Desmethylclomipramine Induces the Accumulation of Autophagy Markers by Blocking Autophagic Flux. J. Cell Sci. 2009, 122, 3330–3339. [Google Scholar] [CrossRef]
  134. The Synergistic Effect of Resveratrol and Hydroxychloroquine on Osteosarcoma Cell Line (MG-63) by Autophagy Modulating. Available online: https://www.researchgate.net/publication/349641481_The_Synergistic_Effect_of_Resveratrol_and_Hydroxychloroquine_on_Osteosarcoma_Cell_Line_MG-63_by_Autophagy_Modulating (accessed on 28 July 2023).
  135. Mei, L.; Chen, Y.; Wang, Z.; Wang, J.; Wan, J.; Yu, C.; Liu, X.; Li, W. Synergistic Anti-Tumour Effects of Tetrandrine and Chloroquine Combination Therapy in Human Cancer: A Potential Antagonistic Role for P21. Br. J. Pharmacol. 2015, 172, 2232–2245. [Google Scholar] [CrossRef] [PubMed]
  136. McAfee, Q.; Zhang, Z.; Samanta, A.; Levi, S.M.; Ma, X.-H.; Piao, S.; Lynch, J.P.; Uehara, T.; Sepulveda, A.R.; Davis, L.E.; et al. Autophagy Inhibitor Lys05 Has Single-Agent Antitumor Activity and Reproduces the Phenotype of a Genetic Autophagy Deficiency. Proc. Natl. Acad. Sci. USA 2012, 109, 8253–8258. [Google Scholar] [CrossRef] [PubMed]
  137. Amaravadi, R.K.; Winkler, J.D. Lys05: A new lysosomal autophagy inhibitor. Autophagy 2012, 8, 1383–1384. [Google Scholar] [CrossRef]
  138. Cárdenas, M.; Marder, M.; Blank, V.C.; Roguin, L.P. Antitumor Activity of Some Natural Flavonoids and Synthetic Derivatives on Various Human and Murine Cancer Cell Lines. Bioorg. Med. Chem. 2006, 14, 2966–2971. [Google Scholar] [CrossRef]
  139. Jeong, S.B.; Das, R.; Kim, D.-H.; Lee, S.; Oh, H.I.; Jo, S.; Lee, Y.; Kim, J.; Park, S.; Choi, D.K.; et al. Anticancer Effect of Verteporfin on Non-Small Cell Lung Cancer via Downregulation of ANO1. Biomed. Pharmacother. 2022, 153, 113373. [Google Scholar] [CrossRef]
  140. Celli, J.P.; Solban, N.; Liang, A.; Pereira, S.P.; Hasan, T. Verteporfin-Based Photodynamic Therapy Overcomes Gemcitabine Insensitivity in a Panel of Pancreatic Cancer Cell Lines. Lasers Surg. Med. 2011, 43, 565–574. [Google Scholar] [CrossRef]
  141. Scott, L.J.; Goa, K.L. Verteporfin. Drugs Aging 2000, 16, 139–146. [Google Scholar] [CrossRef]
  142. Petroni, G.; Bagni, G.; Iorio, J.; Duranti, C.; Lottini, T.; Stefanini, M.; Kragol, G.; Becchetti, A.; Arcangeli, A. Clarithromycin Inhibits Autophagy in Colorectal Cancer by Regulating the HERG1 Potassium Channel Interaction with PI3K. Cell Death Dis. 2020, 11, 161. [Google Scholar] [CrossRef] [PubMed]
  143. Van Nuffel, A.M. Repurposing Drugs in Oncology (ReDO)—Clarithromycin as an Anticancer Agent. Ecancermedicalscience 2015, 9, 513. [Google Scholar] [CrossRef] [PubMed]
  144. Peters, D.H.; Clissold, S.P. Clarithromycin. Drugs 1992, 44, 117–164. [Google Scholar] [CrossRef] [PubMed]
  145. 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. 2018, 5, e1395504. [Google Scholar] [CrossRef] [PubMed]
  146. Rebecca, V.W.; Nicastri, M.C.; McLaughlin, N.; Fennelly, C.; McAfee, Q.; Ronghe, A.; Nofal, M.; Lim, C.-Y.; Witze, E.; Chude, C.I.; et al. A Unified Approach to Targeting the Lysosome’s Degradative and Growth Signaling Roles. Cancer Discov. 2017, 7, 1266–1283. [Google Scholar] [CrossRef]
  147. Goodall, M.L.; Wang, T.; Martin, K.R.; Kortus, M.G.; Kauffman, A.L.; Trent, J.M.; Gately, S.; MacKeigan, J.P. Development of Potent Autophagy Inhibitors That Sensitize Oncogenic BRAF V600E Mutant Melanoma Tumor Cells to Vemurafenib. Autophagy 2014, 10, 1120–1136. [Google Scholar] [CrossRef]
  148. Xie, Y.; Zhang, J.; Lu, B.; Bao, Z.; Zhao, J.; Lu, X.; Wei, Y.; Yao, K.; Jiang, Y.; Yuan, Q.; et al. Mefloquine Inhibits Esophageal Squamous Cell Carcinoma Tumor Growth by Inducing Mitochondrial Autophagy. Front. Oncol. 2020, 10, 1217. [Google Scholar] [CrossRef]
  149. Sharma, G.; Guardia, C.M.; Roy, A.; Vassilev, A.; Saric, A.; Griner, L.N.; Marugan, J.; Ferrer, M.; Bonifacino, J.S.; DePamphilis, M.L. A Family of PIKFYVE Inhibitors with Therapeutic Potential against Autophagy-Dependent Cancer Cells Disrupt Multiple Events in Lysosome Homeostasis. Autophagy 2019, 15, 1694–1718. [Google Scholar] [CrossRef]
  150. Sharma, G.; Ojha, R.; Noguera-Ortega, E.; Rebecca, V.W.; Attanasio, J.; Liu, S.; Piao, S.; Lee, J.J.; Nicastri, M.C.; Harper, S.L.; et al. PPT1 Inhibition Enhances the Antitumor Activity of Anti–PD-1 Antibody in Melanoma. JCI Insight 2020, 5, e133225. [Google Scholar] [CrossRef]
  151. Chen, C.; Lu, Y.; Siu, H.M.; Guan, J.; Zhu, L.; Zhang, S.; Yue, J.; Zhang, L. Identification of Novel Vacuolin-1 Analogues as Autophagy Inhibitors by Virtual Drug Screening and Chemical Synthesis. Molecules 2017, 22, 891. [Google Scholar] [CrossRef]
  152. Bongiorno-Borbone, L.; Giacobbe, A.; Compagnone, M.; Eramo, A.; De Maria, R.; Peschiaroli, A.; Melino, G. Anti-Tumoral Effect of Desmethylclomipramine in Lung Cancer Stem Cells. Oncotarget 2015, 6, 16926–16938. [Google Scholar] [CrossRef] [PubMed]
  153. Nawrocki, S.T.; Han, Y.; Visconte, V.; Przychodzen, B.; Espitia, C.M.; Phillips, J.; Anwer, F.; Advani, A.; Carraway, H.E.; Kelly, K.R.; et al. The Novel Autophagy Inhibitor ROC-325 Augments the Antileukemic Activity of Azacitidine. Leukemia 2019, 33, 2971–2974. [Google Scholar] [CrossRef] [PubMed]
  154. Buzun, K.; Gornowicz, A.; Lesyk, R.; Bielawski, K.; Bielawska, A. Autophagy Modulators in Cancer Therapy. Int. J. Mol. Sci. 2021, 22, 5804. [Google Scholar] [CrossRef] [PubMed]
  155. Xia, Y.; Jia, C.; Xue, Q.; Jiang, J.; Xie, Y.; Wang, R.; Ran, Z.; Xu, F.; Zhang, Y.; Ye, T. Antipsychotic Drug Trifluoperazine Suppresses Colorectal Cancer by Inducing G0/G1 Arrest and Apoptosis. Front. Pharmacol. 2019, 10, 1029. [Google Scholar] [CrossRef] [PubMed]
  156. Zhang, X.; Xu, R.; Zhang, C.; Xu, Y.; Han, M.; Huang, B.; Chen, A.; Qiu, C.; Thorsen, F.; Prestegarden, L.; et al. Trifluoperazine, a Novel Autophagy Inhibitor, Increases Radiosensitivity in Glioblastoma by Impairing Homologous Recombination. J. Exp. Clin. Cancer Res. 2017, 36, 118. [Google Scholar] [CrossRef]
  157. Zhang, S.; Wang, Y.; Xie, W.; Howe, E.N.W.; Busschaert, N.; Sauvat, A.; Leduc, M.; Gomes-da-Silva, L.C.; Chen, G.; Martins, I.; et al. Squaramide-Based Synthetic Chloride Transporters Activate TFEB but Block Autophagic Flux. Cell Death Dis. 2019, 10, 242. [Google Scholar] [CrossRef] [PubMed]
  158. Quintana, M.; Alegre-Requena, J.V.; Marqués-López, E.; Herrera, R.P.; Triola, G. Squaramides with Cytotoxic Activity against Human Gastric Carcinoma Cells HGC-27: Synthesis and Mechanism of Action. Medchemcomm 2016, 7, 550–561. [Google Scholar] [CrossRef]
  159. Guo, Y.; Yang, L.; Guo, W.; Wei, L.; Zhou, Y. FV-429 Enhances the Efficacy of Paclitaxel in NSCLC by Reprogramming HIF-1α-Modulated FattyAcid Metabolism. Chem. Biol. Interact. 2021, 350, 109702. [Google Scholar] [CrossRef] [PubMed]
  160. Chen, X.; Wei, L.; Yang, L.; Guo, W.; Guo, Q.; Zhou, Y. Glycolysis Inhibition and Apoptosis Induction in Human Prostate Cancer Cells by FV-429-Mediated Regulation of AR-AKT-HK2 Signaling Network. Food Chem. Toxicol. 2020, 143, 111517. [Google Scholar] [CrossRef]
  161. Hu, P.; Wang, J.; Qing, Y.; Li, H.; Sun, W.; Yu, X.; Hui, H.; Guo, Q.; Xu, J. FV-429 Induces Autophagy Blockage and Lysosome-Dependent Cell Death of T-Cell Malignancies via Lysosomal Dysregulation. Cell Death Dis. 2021, 12, 80. [Google Scholar] [CrossRef]
  162. Bestion, E.; Zandi, K.; Belouzard, S.; Andreani, J.; Lepidi, H.; Novello, M.; Rouquairol, C.; Baudoin, J.-P.; Rachid, M.; La Scola, B.; et al. GNS561 Exhibits Potent Antiviral Activity against SARS-CoV-2 through Autophagy Inhibition. Viruses 2022, 14, 132. [Google Scholar] [CrossRef] [PubMed]
  163. Harding, J.J.; Awada, A.; Roth, G.; Decaens, T.; Merle, P.; Kotecki, N.; Dreyer, C.; Ansaldi, C.; Rachid, M.; Mezouar, S.; et al. First-In-Human Effects of PPT1 Inhibition Using the Oral Treatment with GNS561/Ezurpimtrostat in Patients with Primary and Secondary Liver Cancers. Liver Cancer 2022, 11, 268–277. [Google Scholar] [CrossRef] [PubMed]
  164. Tan, Q.; Joshua, A.M.; Saggar, J.K.; Yu, M.; Wang, M.; Kanga, N.; Zhang, J.Y.; Chen, X.; Wouters, B.G.; Tannock, I.F. Effect of Pantoprazole to Enhance Activity of Docetaxel against Human Tumour Xenografts by Inhibiting Autophagy. Br. J. Cancer 2015, 112, 832–840. [Google Scholar] [CrossRef] [PubMed]
  165. Hansen, A.R.; Tannock, I.F.; Templeton, A.; Chen, E.; Evans, A.; Knox, J.; Prawira, A.; Sridhar, S.S.; Tan, S.; Vera-Badillo, F.; et al. Pantoprazole Affecting Docetaxel Resistance Pathways via Autophagy (PANDORA): Phase II Trial of High Dose Pantoprazole (Autophagy Inhibitor) with Docetaxel in Metastatic Castration-Resistant Prostate Cancer (MCRPC). Oncologist 2019, 24, 1188–1194. [Google Scholar] [CrossRef] [PubMed]
  166. Molero-Valenzuela, A.; Fontova, P.; Alonso-Carrillo, D.; Carreira-Barral, I.; Torres, A.A.; García-Valverde, M.; Benítez-García, C.; Pérez-Tomás, R.; Quesada, R.; Soto-Cerrato, V. A Novel Late-Stage Autophagy Inhibitor That Efficiently Targets Lysosomes Inducing Potent Cytotoxic and Sensitizing Effects in Lung Cancer. Cancers 2022, 14, 3387. [Google Scholar] [CrossRef] [PubMed]
  167. Hernando, E.; Soto-Cerrato, V.; Cortés-Arroyo, S.; Pérez-Tomás, R.; Quesada, R. Transmembrane Anion Transport and Cytotoxicity of Synthetic Tambjamine Analogs. Org. Biomol. Chem. 2014, 12, 1771–1778. [Google Scholar] [CrossRef] [PubMed]
  168. Guntuku, L.; Gangasani, J.K.; Thummuri, D.; Borkar, R.M.; Manavathi, B.; Ragampeta, S.; Vaidya, J.R.; Sistla, R.; Vegi, N.G.M. IITZ-01, a Novel Potent Lysosomotropic Autophagy Inhibitor, Has Single-Agent Antitumor Efficacy in Triple-Negative Breast Cancer in Vitro and in Vivo. Oncogene 2019, 38, 581–595. [Google Scholar] [CrossRef]
  169. Chen, J.; Shen, Y.; Wu, B.; Yang, P.; Sun, G.; Liu, X.; Qiang, P.; Gao, Y.; Sha, F.; Li, Z.; et al. CUR5g, a Novel Autophagy Inhibitor, Exhibits Potent Synergistic Anticancer Effects with Cisplatin against Non-Small-Cell Lung Cancer. Cell Death Discov. 2022, 8, 435. [Google Scholar] [CrossRef] [PubMed]
  170. Klionsky, D.J.; Elazar, Z.; Seglen, P.O.; Rubinsztein, D.C. Does Bafilomycin A1 Block the Fusion of Autophagosomes with Lysosomes? Autophagy 2008, 4, 849–850. [Google Scholar] [CrossRef]
  171. Ko, H.; Kim, Y.-J.; Park, J.-S.; Park, J.H.; Yang, H.O. Autophagy Inhibition Enhances Apoptosis Induced by Ginsenoside Rk1 in Hepatocellular Carcinoma Cells. Biosci. Biotechnol. Biochem. 2009, 73, 2183–2189. [Google Scholar] [CrossRef]
  172. Lim, J.-H.; Park, J.-W.; Kim, M.-S.; Park, S.-K.; Johnson, R.S.; Chun, Y.-S. Bafilomycin Induces the P21-Mediated Growth Inhibition of Cancer Cells under Hypoxic Conditions by Expressing Hypoxia-Inducible Factor-1α. Mol. Pharmacol. 2006, 70, 1856–1865. [Google Scholar] [CrossRef] [PubMed]
  173. Redmann, M.; Benavides, G.A.; Berryhill, T.F.; Wani, W.Y.; Ouyang, X.; Johnson, M.S.; Ravi, S.; Barnes, S.; Darley-Usmar, V.M.; Zhang, J. Inhibition of Autophagy with Bafilomycin and Chloroquine Decreases Mitochondrial Quality and Bioenergetic Function in Primary Neurons. Redox Biol. 2017, 11, 73–81. [Google Scholar] [CrossRef] [PubMed]
  174. Yan, Y.; Jiang, K.; Liu, P.; Zhang, X.; Dong, X.; Gao, J.; Liu, Q.; Barr, M.P.; Zhang, Q.; Hou, X.; et al. Bafilomycin A1 Induces Caspase-Independent Cell Death in Hepatocellular Carcinoma Cells via Targeting of Autophagy and MAPK Pathways. Sci. Rep. 2016, 6, 37052. [Google Scholar] [CrossRef] [PubMed]
  175. La Ferla, B.; Airoldi, C.; Zona, C.; Orsato, A.; Cardona, F.; Merlo, S.; Sironi, E.; D’Orazio, G.; Nicotra, F. Natural Glycoconjugates with Antitumor Activity. Nat. Prod. Rep. 2011, 28, 630–648. [Google Scholar] [CrossRef] [PubMed]
  176. Kiyoshima, T.; Yoshida, H.; Wada, H.; Nagata, K.; Fujiwara, H.; Kihara, M.; Hasegawa, K.; Someya, H.; Sakai, H. Chemoresistance to Concanamycin A1 in Human Oral Squamous Cell Carcinoma Is Attenuated by an HDAC Inhibitor Partly via Suppression of Bcl-2 Expression. PLoS ONE 2013, 8, e80998. [Google Scholar] [CrossRef] [PubMed]
  177. 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] [PubMed]
  178. Akin, D.; Wang, S.K.; Habibzadegah-Tari, P.; Law, B.; Ostrov, D.; Li, M.; Yin, X.-M.; Kim, J.-S.; Horenstein, N.; Dunn, W.A. A Novel ATG4B Antagonist Inhibits Autophagy and Has a Negative Impact on Osteosarcoma Tumors. Autophagy 2014, 10, 2021–2035. [Google Scholar] [CrossRef] [PubMed]
  179. Clissold, S.P.; Heel, R.C. Tioconazole. Drugs 1986, 31, 29–51. [Google Scholar] [CrossRef] [PubMed]
  180. Shi, J.; Dong, X.; Li, H.; Wang, H.; Jiang, Q.; Liu, L.; Wang, L.; Dong, J. Nicardipine Sensitizes Temozolomide by Inhibiting Autophagy and Promoting Cell Apoptosis in Glioma Stem Cells. Aging 2021, 13, 6820–6831. [Google Scholar] [CrossRef]
  181. Bosc, D.; Vezenkov, L.; Bortnik, S.; An, J.; Xu, J.; Choutka, C.; Hannigan, A.M.; Kovacic, S.; Loo, S.; Clark, P.G.K.; et al. A New Quinoline-Based Chemical Probe Inhibits the Autophagy-Related Cysteine Protease ATG4B. Sci. Rep. 2018, 8, 11653. [Google Scholar] [CrossRef]
  182. 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] [PubMed]
  183. Wang, Z.L.; Deng, Q.; Chong, T.; Wang, Z.M. Autophagy Suppresses the Proliferation of Renal Carcinoma Cell. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 343–350. [Google Scholar] [CrossRef] [PubMed]
  184. Jones, T.M.; Carew, J.S.; Nawrocki, S.T. Therapeutic Targeting of Autophagy for Renal Cell Carcinoma Therapy. Cancers 2020, 12, 1185. [Google Scholar] [CrossRef] [PubMed]
  185. Chacon-Barahona, J.A.; MacKeigan, J.P.; Lanning, N.J. Unique Metabolic Contexts Sensitize Cancer Cells and Discriminate between Glycolytic Tumor Types. Cancers 2023, 15, 1158. [Google Scholar] [CrossRef] [PubMed]
  186. Petherick, K.J.; Conway, O.J.L.; Mpamhanga, C.; Osborne, S.A.; Kamal, A.; Saxty, B.; Ganley, I.G. Pharmacological Inhibition of ULK1 Kinase Blocks Mammalian Target of Rapamycin (MTOR)-Dependent Autophagy. J. Biol. Chem. 2015, 290, 11376–11383. [Google Scholar] [CrossRef] [PubMed]
  187. 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]
  188. Ahwazi, D.; Neopane, K.; Markby, G.R.; Kopietz, F.; Ovens, A.J.; Dall, M.; Hassing, A.S.; Gräsle, P.; Alshuweishi, Y.; Treebak, J.T.; et al. Investigation of the Specificity and Mechanism of Action of the ULK1/AMPK Inhibitor SBI-0206965. Biochem. J. 2021, 478, 2977–2997. [Google Scholar] [CrossRef] [PubMed]
  189. Egan, D.F.; Chun, M.G.H.; Vamos, M.; Zou, H.; Rong, J.; Miller, C.J.; Lou, H.J.; Raveendra-Panickar, D.; Yang, C.-C.; Sheffler, D.J.; et al. Small Molecule Inhibition of the Autophagy Kinase ULK1 and Identification of ULK1 Substrates. Mol. Cell 2015, 59, 285–297. [Google Scholar] [CrossRef] [PubMed]
  190. Lazarus, M.B.; Novotny, C.J.; Shokat, K.M. Structure of the Human Autophagy Initiating Kinase ULK1 in Complex with Potent Inhibitors. ACS Chem. Biol. 2015, 10, 257–261. [Google Scholar] [CrossRef]
  191. Chen, Y.; Xie, X.; Wang, C.; Hu, Y.; Zhang, H.; Zhang, L.; Tu, S.; He, Y.; Li, Y. Dual Targeting of NUAK1 and ULK1 Using the Multitargeted Inhibitor MRT68921 Exerts Potent Antitumor Activities. Cell Death Dis. 2020, 11, 712. [Google Scholar] [CrossRef]
  192. Bogdan, M.; Timson, M.J.; Al-Hashimi, H.; Smith, B.D.; Flynn, D.L. Abstract 1377: DCC-3116, a First-in-Class Selective Inhibitor of ULK1/2 Kinases and Autophagy, Synergizes with Encorafenib and Cetuximab in BRAF V600E Mutant Colorectal Cancer Models. Cancer Res 2023, 83, 1377. [Google Scholar] [CrossRef]
  193. Smith, B.D.; Vogeti, L.; Gupta, A.; Singh, J.; Al-Ani, G.; Bulfer, S.L.; Caldwell, T.M.; Timson, M.J.; Vogeti, S.; Ahn, Y.M.; et al. Abstract B129: Preclinical Studies with DCC-3116, an ULK Kinase Inhibitor Designed to Inhibit Autophagy as a Potential Strategy to Address Mutant RAS Cancers. Mol. Cancer Ther. 2019, 18, B129. [Google Scholar] [CrossRef]
  194. Xue, S.-T.; Li, K.; Gao, Y.; Zhao, L.-Y.; Gao, Y.; Yi, H.; Jiang, J.-D.; Li, Z.-R. The Role of the Key Autophagy Kinase ULK1 in Hepatocellular Carcinoma and Its Validation as a Treatment Target. Autophagy 2020, 16, 1823–1837. [Google Scholar] [CrossRef] [PubMed]
  195. Huang, T.; Wan, X.; Alvarez, A.A.; James, C.D.; Song, X.; Yang, Y.; Sastry, N.; Nakano, I.; Sulman, E.P.; Hu, B.; et al. MIR93 (MicroRNA-93) Regulates Tumorigenicity and Therapy Response of Glioblastoma by Targeting Autophagy. Autophagy 2019, 15, 1100–1111. [Google Scholar] [CrossRef] [PubMed]
  196. Wood, S.D.; Grant, W.; Adrados, I.; Choi, J.Y.; Alburger, J.M.; Duckett, D.R.; Roush, W.R. In Silico HTS and Structure Based Optimization of Indazole-Derived ULK1 Inhibitors. ACS Med. Chem. Lett. 2017, 8, 1258–1263. [Google Scholar] [CrossRef] [PubMed]
  197. Murray, E.J.B.; Grisanti, M.S.; Bentley, G.V.; Murray, S.S. E64d, a Membrane-Permeable Cysteine Protease Inhibitor, Attenuates the Effects of Parathyroid Hormone on Osteoblasts in Vitro. Metabolism 1997, 46, 1090–1094. [Google Scholar] [CrossRef]
  198. Müller, S.; Dennemärker, J.; Reinheckel, T. Specific Functions of Lysosomal Proteases in Endocytic and Autophagic Pathways. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2012, 1824, 34–43. [Google Scholar] [CrossRef]
  199. Ni, H.-M.; Bockus, A.; Wozniak, A.L.; Jones, K.; Weinman, S.; Yin, X.-M.; Ding, W.-X. Dissecting the Dynamic Turnover of GFP-LC3 in the Autolysosome. Autophagy 2011, 7, 188–204. [Google Scholar] [CrossRef] [PubMed]
  200. Chen, X.; Xu, Z.; Zeng, S.; Wang, X.; Liu, W.; Qian, L.; Wei, J.; Yang, X.; Shen, Q.; Gong, Z.; et al. The Molecular Aspect of Antitumor Effects of Protease Inhibitor Nafamostat Mesylate and Its Role in Potential Clinical Applications. Front. Oncol. 2019, 9, 852. [Google Scholar] [CrossRef]
  201. Hwang, H.-Y.; Cho, Y.S.; Kim, J.Y.; Yun, K.N.; Yoo, J.S.; Lee, E.; Kim, I.; Kwon, H.J. Autophagic Inhibition via Lysosomal Integrity Dysfunction Leads to Antitumor Activity in Glioma Treatment. Cancers 2020, 12, 543. [Google Scholar] [CrossRef]
  202. Yang, Y.; Hu, L.; Zheng, H.; Mao, C.; Hu, W.; Xiong, K.; Wang, F.; Liu, C. Application and Interpretation of Current Autophagy Inhibitors and Activators. Acta Pharmacol. Sin. 2013, 34, 625–635. [Google Scholar] [CrossRef]
  203. Tanida, I.; Minematsu-Ikeguchi, N.; Ueno, T.; Kominami, E. Lysosomal Turnover, but Not a Cellular Level, of Endogenous LC3 Is a Marker for Autophagy. Autophagy 2005, 1, 84–91. [Google Scholar] [CrossRef] [PubMed]
  204. Kominami, E.; Hashida, S.; Khairallah, E.A.; Katunuma, N. Sequestration of Cytoplasmic Enzymes in an Autophagic Vacuole-Lysosomal System Induced by Injection of Leupeptin. J. Biol. Chem. 1983, 258, 6093–6100. [Google Scholar] [CrossRef]
  205. Kakizoe, T.; Takayasu, H.; Kawachi, T.; Sugimura, T.; Takeuchi, T.; Umezawa, H. Effect of Leupeptin, a Protease Inhibitor, on Induction of Bladder Tumors in Rats by N-Butyl-N-(4-Hydroxybutyl) Nitrosamine23. JNCI J. Natl. Cancer Inst. 1976, 56, 433–435. [Google Scholar] [CrossRef] [PubMed]
  206. Kobara, M.; Nessa, N.; Toba, H.; Nakata, T. Induction of Autophagy Has Protective Roles in Imatinib-Induced Cardiotoxicity. Toxicol. Rep. 2021, 8, 1087–1097. [Google Scholar] [CrossRef]
  207. Altman, J.K.; Platanias, L.C. A New Purpose for an Old Drug: Inhibiting Autophagy with Clarithromycin. Leuk. Lymphoma 2012, 53, 1255–1256. [Google Scholar] [CrossRef] [PubMed]
  208. Zeng, X.; Zhao, H.; Li, Y.; Fan, J.; Sun, Y.; Wang, S.; Wang, Z.; Song, P.; Ju, D. Targeting Hedgehog Signaling Pathway and Autophagy Overcomes Drug Resistance of BCR-ABL-Positive Chronic Myeloid Leukemia. Autophagy 2015, 11, 355–372. [Google Scholar] [CrossRef]
  209. Shingu, T.; Fujiwara, K.; Bögler, O.; Akiyama, Y.; Moritake, K.; Shinojima, N.; Tamada, Y.; Yokoyama, T.; Kondo, S. Inhibition of Autophagy at a Late Stage Enhances Imatinib-Induced Cytotoxicity in Human Malignant Glioma Cells. Int. J. Cancer 2009, 124, 1060–1071. [Google Scholar] [CrossRef]
  210. Xie, Y.; Fan, S.; Ni, D.; Wan, W.; Xu, P.; Ding, Y.; Zhang, R.; Lu, J.; Zhang, N.; Zhang, Y.; et al. An ATG4B Inhibitor Blocks Autophagy and Sensitizes Sorafenib Inhibition Activities in HCC Tumor Cells. Bioorg Med. Chem. 2023, 84, 117262. [Google Scholar] [CrossRef]
  211. Yuan, H. Inhibition of Autophagy Significantly Enhances Combination Therapy with Sorafenib and HDAC Inhibitors for Human Hepatoma Cells. World J. Gastroenterol. 2014, 20, 4953. [Google Scholar] [CrossRef]
  212. Liu, X.; Sun, K.; Wang, H.; Dai, Y. Inhibition of Autophagy by Chloroquine Enhances the Antitumor Efficacy of Sorafenib in Glioblastoma. Cell Mol. Neurobiol. 2016, 36, 1197–1208. [Google Scholar] [CrossRef] [PubMed]
  213. Abdel-Aziz, A.K.; Shouman, S.; El-Demerdash, E.; Elgendy, M.; Abdel-Naim, A.B. Chloroquine Synergizes Sunitinib Cytotoxicity via Modulating Autophagic, Apoptotic and Angiogenic Machineries. Chem. Biol. Interact. 2014, 217, 28–40. [Google Scholar] [CrossRef] [PubMed]
  214. Zeng, F.; Li, Y.; Meng, Y.; Sun, H.; He, Y.; Yin, M.; Chen, X.; Deng, G. BET Inhibitors Synergize with Sunitinib in Melanoma through GDF15 Suppression. Exp. Mol. Med. 2023, 55, 364–376. [Google Scholar] [CrossRef] [PubMed]
  215. Li, M.; Xu, Y.; Lu, W.; Li, Y.; Tan, S.; Lin, H.; Wu, T.; Li, Y.; Wang, S.; Zhao, Y. Chloroquine Potentiates the Anticancer Effect of Sunitinib on Renal Cell Carcinoma by Inhibiting Autophagy and Inducing Apoptosis. Oncol. Lett. 2017, 15, 2839–2846. [Google Scholar] [CrossRef]
  216. Pan, H.; Wang, Z.; Jiang, L.; Sui, X.; You, L.; Shou, J.; Jing, Z.; Xie, J.; Ge, W.; Cai, X.; et al. Autophagy Inhibition Sensitizes Hepatocellular Carcinoma to the Multikinase Inhibitor Linifanib. Sci. Rep. 2014, 4, 6683. [Google Scholar] [CrossRef] [PubMed]
  217. Lai, X.; Shi, Y.; Zhou, M. Dihydroartemisinin Enhances Gefitinib Cytotoxicity against Lung Adenocarcinoma Cells by Inducing ROS-dependent Apoptosis and Ferroptosis. Kaohsiung J. Med. Sci. 2023, 39, 699–709. [Google Scholar] [CrossRef] [PubMed]
  218. 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] [PubMed]
  219. 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]
  220. Sugita, S.; Ito, K.; Yamashiro, Y.; Moriya, S.; Che, X.-F.; Yokoyama, T.; Hiramoto, M.; Miyazawa, K. EGFR-Independent Autophagy Induction with Gefitinib and Enhancement of Its Cytotoxic Effect by Targeting Autophagy with Clarithromycin in Non-Small Cell Lung Cancer Cells. Biochem. Biophys. Res. Commun. 2015, 461, 28–34. [Google Scholar] [CrossRef]
  221. Meng, J.; Chang, C.; Chen, Y.; Bi, F.; Ji, C.; Liu, W. EGCG Overcomes Gefitinib Resistance by Inhibiting Autophagy and Augmenting Cell Death through Targeting ERK Phosphorylation in NSCLC. Onco Targets Ther. 2019, 12, 6033–6043. [Google Scholar] [CrossRef]
  222. Hajiahmadi, S.; Lorzadeh, S.; Iranpour, R.; Karima, S.; Rajabibazl, M.; Shahsavari, Z.; Ghavami, S. Temozolomide, Simvastatin and Acetylshikonin Combination Induces Mitochondrial-Dependent Apoptosis in GBM Cells, Which Is Regulated by Autophagy. Biology 2023, 12, 302. [Google Scholar] [CrossRef] [PubMed]
  223. Zou, Y.; Ling, Y.-H.; Sironi, J.; Schwartz, E.L.; Perez-Soler, R.; Piperdi, B. The Autophagy Inhibitor Chloroquine Overcomes the Innate Resistance of Wild-Type EGFR Non-Small-Cell Lung Cancer Cells to Erlotinib. J. Thorac. Oncol. 2013, 8, 693–702. [Google Scholar] [CrossRef] [PubMed]
  224. Lobo, M.R.; Green, S.C.; Schabel, M.C.; Gillespie, G.Y.; Woltjer, R.L.; Pike, M.M. Quinacrine Synergistically Enhances the Antivascular and Antitumor Efficacy of Cediranib in Intracranial Mouse Glioma. Neuro Oncol. 2013, 15, 1673–1683. [Google Scholar] [CrossRef] [PubMed]
  225. Jarauta, V.; Jaime, P.; Gonzalo, O.; de Miguel, D.; Ramírez-Labrada, A.; Martínez-Lostao, L.; Anel, A.; Pardo, J.; Marzo, I.; Naval, J. Inhibition of Autophagy with Chloroquine Potentiates Carfilzomib-Induced Apoptosis in Myeloma Cells in Vitro and in Vivo. Cancer Lett. 2016, 382, 1–10. [Google Scholar] [CrossRef] [PubMed]
  226. Baranowska, K.; Misund, K.; Starheim, K.K.; Holien, T.; Johansson, I.; Darvekar, S.; Buene, G.; Waage, A.; Bjørkøy, G.; Sundan, A. Hydroxychloroquine Potentiates Carfilzomib Toxicity towards Myeloma Cells. Oncotarget 2016, 7, 70845–70856. [Google Scholar] [CrossRef] [PubMed]
  227. Hsu, C.-M.; Yen, C.-H.; Wang, S.-C.; Liu, Y.-C.; Huang, C.-T.; Wang, M.-H.; Chuang, T.-M.; Ke, Y.-L.; Yeh, T.-J.; Gau, Y.-C.; et al. Emodin Ameliorates the Efficacy of Carfilzomib in Multiple Myeloma Cells via Apoptosis and Autophagy. Biomedicines 2022, 10, 1638. [Google Scholar] [CrossRef] [PubMed]
  228. Moriya, S.; Che, X.-F.; Komatsu, S.; Abe, A.; Kawaguchi, T.; Gotoh, A.; Inazu, M.; Tomoda, A.; Miyazawa, K. Macrolide Antibiotics Block Autophagy Flux and Sensitize to Bortezomib via Endoplasmic Reticulum Stress-Mediated CHOP Induction in Myeloma Cells. Int. J. Oncol. 2013, 42, 1541–1550. [Google Scholar] [CrossRef] [PubMed]
  229. 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]
  230. Kawaguchi, T.; Miyazawa, K.; Moriya, S.; Ohtomo, T.; Che, X.F.; Naito, M.; Itoh, M.; Tomoda, A. Combined Treatment with Bortezomib Plus Bafilomycin A1 Enhances the Cytocidal Effect and Induces Endoplasmic Reticulum Stress in U266 Myeloma Cells: Crosstalk among Proteasome, Autophagy-Lysosome and ER Stress. Int. J. Oncol. 2011, 38, 643–654. [Google Scholar] [CrossRef]
  231. Zhang, X.; Li, W.; Wang, C.; Leng, X.; Lian, S.; Feng, J.; Li, J.; Wang, H. Inhibition of Autophagy Enhances Apoptosis Induced by Proteasome Inhibitor Bortezomib in Human Glioblastoma U87 and U251 Cells. Mol. Cell Biochem. 2014, 385, 265–275. [Google Scholar] [CrossRef]
  232. Han, Q.; Bai, H.; Xu, Y.; Zhou, M.; Zhou, H.; Dong, X.; Chen, B. Solamargine Induces Autophagy-mediated Apoptosis and Enhances Bortezomib Activity in Multiple Myeloma. Clin. Exp. Pharmacol. Physiol. 2022, 49, 674–685. [Google Scholar] [CrossRef] [PubMed]
  233. Yang, L. BH3 Mimetic ABT-737 Sensitizes Colorectal Cancer Cells to Ixazomib Through MCL-1 Downregulation and Autophagy Inhibition. Int. J. Radiat. Oncol. Biol. Phys. 2016, 96, E559. [Google Scholar] [CrossRef]
  234. Loh, J.S.; Rahim, N.A.; Tor, Y.S.; Foo, J.B. Simultaneous Proteasome and Autophagy Inhibition Synergistically Enhances Cytotoxicity of Doxorubicin in Breast Cancer Cells. Cell Biochem. Funct. 2022, 40, 403–416. [Google Scholar] [CrossRef] [PubMed]
  235. Petiot, A.; Ogier-Denis, E.; Blommaart, E.F.C.; Meijer, A.J.; Codogno, P. Distinct Classes of Phosphatidylinositol 3′-Kinases Are Involved in Signaling Pathways That Control Macroautophagy in HT-29 Cells. J. Biol. Chem. 2000, 275, 992–998. [Google Scholar] [CrossRef] [PubMed]
  236. Dischler, N.M.; Xu, L.; Li, Y.; Nichols, C.B.; Alspaugh, J.A.; Bills, G.F.; Gloer, J.B. Wortmannin and Wortmannine Analogues from an Undescribed Niesslia sp. J. Nat. Prod. 2019, 82, 532–538. [Google Scholar] [CrossRef] [PubMed]
  237. Wiesinger, D.; Gubler, H.U.; Haefliger, W.; Hauser, D. Antiinflammatory Activity of the New Mould Metabolite 11-Desacetoxy-Wortmannin and of Some of Its Derivatives. Experientia 1974, 30, 135–136. [Google Scholar] [CrossRef]
  238. Teranishi, F.; Takahashi, N.; Gao, N.; Akamo, Y.; Takeyama, H.; Manabe, T.; Okamoto, T. Phosphoinositide 3-Kinase Inhibitor (Wortmannin) Inhibits Pancreatic Cancer Cell Motility and Migration Induced by Hyaluronan in Vitro and Peritoneal Metastasis in Vivo. Cancer Sci. 2009, 100, 770–777. [Google Scholar] [CrossRef] [PubMed]
  239. Powis, G.; Bonjouklian, R.; Berggren, M.M.; Gallegos, A.; Abraham, R.; Ashendel, C.; Zalkow, L.; Matter, W.F.; Dodge, J.; Grindey, G. Wortmannin, a Potent and Selective Inhibitor of Phosphatidylinositol-3-Kinase. Cancer Res. 1994, 54, 2419–2423. [Google Scholar]
  240. Thelen, M.; Wymann, M.P.; Langen, H. Wortmannin Binds Specifically to 1-Phosphatidylinositol 3-Kinase While Inhibiting Guanine Nucleotide-Binding Protein-Coupled Receptor Signaling in Neutrophil Leukocytes. Proc. Natl. Acad. Sci. USA 1994, 91, 4960–4964. [Google Scholar] [CrossRef]
  241. Rao, J.; Mei, L.; Liu, J.; Tang, X.; Yin, S.; Xia, C.; Wei, J.; Wan, D.; Wang, X.; Wang, Y.; et al. Size-Adjustable Micelles Co-Loaded with a Chemotherapeutic Agent and an Autophagy Inhibitor for Enhancing Cancer Treatment via Increased Tumor Retention. Acta Biomater. 2019, 89, 300–312. [Google Scholar] [CrossRef]
  242. Akter, R.; Hossain, M.Z.; Kleve, M.G.; Gealt, M.A. Wortmannin Induces MCF-7 Breast Cancer Cell Death via the Apoptotic Pathway, Involving Chromatin Condensation, Generation of Reactive Oxygen Species, and Membrane Blebbing. Breast Cancer Targets Ther. 2012, 4, 103–113. [Google Scholar] [CrossRef]
  243. Au, K.M.; Min, Y.; Tian, X.; Zhang, L.; Perello, V.; Caster, J.M.; Wang, A.Z. Improving Cancer Chemoradiotherapy Treatment by Dual Controlled Release of Wortmannin and Docetaxel in Polymeric Nanoparticles. ACS Nano 2015, 9, 8976–8996. [Google Scholar] [CrossRef] [PubMed]
  244. Brunn, G.J.; Hudson, C.C.; Sekulić, A.; Williams, J.M.; Hosoi, H.; Houghton, P.J.; Lawrence, J.C.; Abraham, R.T. Phosphorylation of the Translational Repressor PHAS-I by the Mammalian Target of Rapamycin. Science 1997, 277, 99–101. [Google Scholar] [CrossRef] [PubMed]
  245. Sarkaria, J.N.; Tibbetts, R.S.; Busby, E.C.; Kennedy, A.P.; Hill, D.E.; Abraham, R.T. Inhibition of Phosphoinositide 3-Kinase Related Kinases by the Radiosensitizing Agent Wortmannin. Cancer Res. 1998, 58, 4375–4382. [Google Scholar] [PubMed]
  246. Brunn, G.J.; Williams, J.; Sabers, C.; Wiederrecht, G.; Lawrence, J.C.; Abraham, R.T. Direct Inhibition of the Signaling Functions of the Mammalian Target of Rapamycin by the Phosphoinositide 3-Kinase Inhibitors, Wortmannin and LY294002. EMBO J. 1996, 15, 5256–5267. [Google Scholar] [CrossRef]
  247. Feldman, M.E.; Shokat, K.M. New Inhibitors of the PI3K-Akt-MTOR Pathway: Insights into MTOR Signaling from a New Generation of Tor Kinase Domain Inhibitors (TORKinibs). In Phosphoinositide 3-Kinase in Health and Disease; Springer: Berlin/Heidelberg, Germany, 2010; Volume 2, pp. 241–262. [Google Scholar]
  248. Zhang, M.; Hagan, C.T.; Min, Y.; Foley, H.; Tian, X.; Yang, F.; Mi, Y.; Au, K.M.; Medik, Y.; Roche, K.; et al. Nanoparticle Co-Delivery of Wortmannin and Cisplatin Synergistically Enhances Chemoradiotherapy and Reverses Platinum Resistance in Ovarian Cancer Models. Biomaterials 2018, 169, 1–10. [Google Scholar] [CrossRef] [PubMed]
  249. Phuwapraisirisan, P.; Rangsan, J.; Siripong, P.; Tip-Pyang, S. 9-Epi-Viridiol, a Novel Cytotoxic Furanosteroid from Soil Fungus Trichoderma virens. Nat. Prod. Res. 2006, 20, 1321–1325. [Google Scholar] [CrossRef] [PubMed]
  250. Del Bel, M.; Abela, A.R.; Ng, J.D.; Guerrero, C.A. Enantioselective Chemical Syntheses of the Furanosteroids (−)-Viridin and (−)-Viridiol. J. Am. Chem. Soc. 2017, 139, 6819–6822. [Google Scholar] [CrossRef] [PubMed]
  251. Pal, I.; Mandal, M. PI3K and Akt as Molecular Targets for Cancer Therapy: Current Clinical Outcomes. Acta Pharmacol. Sin. 2012, 33, 1441–1458. [Google Scholar] [CrossRef]
  252. Howes, A.L.; Chiang, G.G.; Lang, E.S.; Ho, C.B.; Powis, G.; Vuori, K.; Abraham, R.T. The Phosphatidylinositol 3-Kinase Inhibitor, PX-866, Is a Potent Inhibitor of Cancer Cell Motility and Growth in Three-Dimensional Cultures. Mol. Cancer Ther. 2007, 6, 2505–2514. [Google Scholar] [CrossRef]
  253. Peng, X.; Gong, F.; Chen, Y.; Jiang, Y.; Liu, J.; Yu, M.; Zhang, S.; Wang, M.; Xiao, G.; Liao, H. Autophagy Promotes Paclitaxel Resistance of Cervical Cancer Cells: Involvement of Warburg Effect Activated Hypoxia-Induced Factor 1-α-Mediated Signaling. Cell Death Dis. 2014, 5, e1367. [Google Scholar] [CrossRef] [PubMed]
  254. Wu, Y.; Wang, X.; Guo, H.; Zhang, B.; Zhang, X.-B.; Shi, Z.-J.; Yu, L. Synthesis and Screening of 3-MA Derivatives for Autophagy Inhibitors. Autophagy 2013, 9, 595–603. [Google Scholar] [CrossRef] [PubMed]
  255. Blommaart, E.F.C.; Krause, U.; Schellens, J.P.M.; Vreeling-Sindelarova, H.; Meijer, A.J. The Phosphatidylinositol 3-Kinase Inhibitors Wortmannin and LY294002 Inhibit Autophagy in Isolated Rat Hepatocytes. Eur. J. Biochem. 1997, 243, 240–246. [Google Scholar] [CrossRef] [PubMed]
  256. Knight, Z.A.; Shokat, K.M. Chemically Targeting the PI3K Family. Biochem. Soc. Trans. 2007, 35, 245–249. [Google Scholar] [CrossRef] [PubMed]
  257. Ryabaya, O.O.; Inshakov, A.N.; Egorova, A.V.; Emelyanova, M.A.; Nasedkina, T.V.; Zasedatelev, A.S.; Khochenkov, D.A.; Stepanova, E.V. Autophagy Inhibitors Chloroquine and LY294002 Enhance Temozolomide Cytotoxicity on Cutaneous Melanoma Cell Lines in Vitro. Anticancer. Drugs 2017, 28, 307–315. [Google Scholar] [CrossRef] [PubMed]
  258. Duarte, A.; Silveira, G.G.; Soave, D.F.; Costa, J.P.O.; Silva, A.R. The Role of the LY294002—A Non-Selective Inhibitor of Phosphatidylinositol 3-Kinase (PI3K) Pathway- in Cell Survival and Proliferation in Cell Line SCC-25. Asian Pac. J. Cancer Prev. 2019, 20, 3377–3383. [Google Scholar] [CrossRef] [PubMed]
  259. Akkoç, Y.; Berrak, Ö.; Arısan, E.D.; Obakan, P.; Çoker-Gürkan, A.; Palavan-Ünsal, N. Inhibition of PI3K Signaling Triggered Apoptotic Potential of Curcumin Which Is Hindered by Bcl-2 through Activation of Autophagy in MCF-7 Cells. Biomed. Pharmacother. 2015, 71, 161–171. [Google Scholar] [CrossRef] [PubMed]
  260. 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]
  261. Mindell, J.A. Lysosomal Acidification Mechanisms. Annu. Rev. Physiol. 2012, 74, 69–86. [Google Scholar] [CrossRef]
  262. Piao, S.; Amaravadi, R.K. Targeting the Lysosome in Cancer. Ann. N. Y. Acad. Sci. 2016, 1371, 45–54. [Google Scholar] [CrossRef]
  263. Sharma, R.K.; Chakotiya, A.S. Phytoconstituents of Zingiber Officinale Targeting Host Viral Protein Interaction at Entry Point of SARS CoV 2 A Molecular Docking Study. Def. Life Sci. J. 2020, 5, 268–277. [Google Scholar] [CrossRef]
  264. Homewood, C.A.; Warhurst, D.C.; Peters, W.; Baggaley, V.C. Lysosomes, PH and the Anti-Malarial Action of Chloroquine. Nature 1972, 235, 50–52. [Google Scholar] [CrossRef]
  265. Kremer, J.M. Rational Use of New and Existing Disease-Modifying Agents in Rheumatoid Arthritis. Ann. Intern. Med. 2001, 134, 695. [Google Scholar] [CrossRef] [PubMed]
  266. Sorour, A.A.; Kurmann, R.D.; Shahin, Y.E.; Crowson, C.S.; Achenbach, S.J.; Mankad, R.; Myasoedova, E. Use of Hydroxychloroquine and Risk of Heart Failure in Patients With Rheumatoid Arthritis. J. Rheumatol. 2021, 48, 1508–1511. [Google Scholar] [CrossRef] [PubMed]
  267. Romanelli, F.; Smith, K.; Hoven, A. Chloroquine and Hydroxychloroquine as Inhibitors of Human Immunodeficiency Virus (HIV-1) Activity. Curr. Pharm. Des. 2004, 10, 2643–2648. [Google Scholar] [CrossRef]
  268. Rainsford, K.D.; Parke, A.L.; Clifford-Rashotte, M.; Kean, W.F. Therapy and Pharmacological Properties of Hydroxychloroquine and Chloroquine in Treatment of Systemic Lupus Erythematosus, Rheumatoid Arthritis and Related Diseases. Inflammopharmacology 2015, 23, 231–269. [Google Scholar] [CrossRef] [PubMed]
  269. Jin, Z.; Wang, F.; Pan, W.; Liu, L.; Wu, M.; Hu, H.; Ding, X.; Wei, H.; Zou, Y.; Qian, X.; et al. Association of Antimalarial Drugs with Decreased Overall and Cause Specific Mortality in Systemic Lupus Erythematosus. Rheumatology 2021, 60, 1774–1783. [Google Scholar] [CrossRef]
  270. Liu, J.; Cao, R.; Xu, M.; Wang, X.; Zhang, H.; Hu, H.; Li, Y.; Hu, Z.; Zhong, W.; Wang, M. Hydroxychloroquine, a Less Toxic Derivative of Chloroquine, Is Effective in Inhibiting SARS-CoV-2 Infection in Vitro. Cell Discov. 2020, 6, 16. [Google Scholar] [CrossRef]
  271. O’Neill, P.M.; Bray, P.G.; Hawley, S.R.; Ward, S.A.; Park, B.K. 4-Aminoquinolines—Past, Present, and Future; A Chemical Perspective. Pharmacol. Ther. 1998, 77, 29–58. [Google Scholar] [CrossRef]
  272. Fernández, A.; Ordóñez, R.; Reiter, R.J.; González-Gallego, J.; Mauriz, J.L. Melatonin and Endoplasmic Reticulum Stress: Relation to Autophagy and Apoptosis. J. Pineal Res. 2015, 59, 292–307. [Google Scholar] [CrossRef]
  273. Abdel Karim, N.F.; Ahmad, I.; Gaber, O.; Eldessouki, I.; Olowokure, O.O.; Farooq, M.; Morris, J.C. Phase I Trial of Chloroquine (CQ)/Hydroxychloroquine (HCQ) in Combination with Carboplatin-Gemcitabine (CG) in Patients with Advanced Solid Tumors. J. Clin. Oncol. 2019, 37, 3027. [Google Scholar] [CrossRef]
  274. Kim, Y.; Bang, S.-C.; Lee, J.-H.; Ahn, B.-Z. Pulsatilla Saponin D: The Antitumor Principle FromPulsatilla Koreana. Arch. Pharm. Res. 2004, 27, 915–918. [Google Scholar] [CrossRef] [PubMed]
  275. Wang, K.; Tu, Y.; Wan, J.-B.; Chen, M.; He, C. Synergistic Anti-Breast Cancer Effect of Pulsatilla Saponin D and Camptothecin through Interrupting Autophagic–Lysosomal Function and Promoting P62-Mediated Ubiquitinated Protein Aggregation. Carcinogenesis 2020, 41, 804–816. [Google Scholar] [CrossRef] [PubMed]
  276. Wu, W.K.K.; Coffelt, S.B.; Cho, C.H.; Wang, X.J.; Lee, C.W.; Chan, F.K.L.; Yu, J.; Sung, J.J.Y. The Autophagic Paradox in Cancer Therapy. Oncogene 2012, 31, 939–953. [Google Scholar] [CrossRef] [PubMed]
  277. Zhang, J.; Feng, Y.; Forgac, M. Proton Conduction and Bafilomycin Binding by the V0 Domain of the Coated Vesicle V-ATPase. J. Biol. Chem. 1994, 269, 23518–23523. [Google Scholar] [CrossRef] [PubMed]
  278. Yoshimori, T.; Yamamoto, A.; Moriyama, Y.; Futai, M.; Tashiro, Y. Bafilomycin A1, a Specific Inhibitor of Vacuolar-Type H(+)-ATPase, Inhibits Acidification and Protein Degradation in Lysosomes of Cultured Cells. J. Biol. Chem. 1991, 266, 17707–17712. [Google Scholar] [CrossRef] [PubMed]
  279. Metzger-Filho, O.; Moulin, C.; Awada, A. Molecular Targeted Therapy in Prevalent Tumors: Learning from the Past and Future Perspectives. Curr. Clin. Pharmacol. 2010, 5, 166–177. [Google Scholar] [CrossRef]
  280. Ekstrand, A.I.; Jönsson, M.; Lindblom, A.; Borg, Å.; Nilbert, M. Frequent Alterations of the PI3K/AKT/MTOR Pathways in Hereditary Nonpolyposis Colorectal Cancer. Fam. Cancer 2010, 9, 125–129. [Google Scholar] [CrossRef] [PubMed]
  281. 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]
  282. Shimobayashi, M.; Hall, M.N. Making New Contacts: The MTOR Network in Metabolism and Signalling Crosstalk. Nat. Rev. Mol. Cell Biol. 2014, 15, 155–162. [Google Scholar] [CrossRef]
  283. Dancey, J. MTOR Signaling and Drug Development in Cancer. Nat. Rev. Clin. Oncol. 2010, 7, 209–219. [Google Scholar] [CrossRef] [PubMed]
  284. Wang, L.; Zhu, Y.-R.; Wang, S.; Zhao, S. Autophagy Inhibition Sensitizes WYE-354-Induced Anti-Colon Cancer Activity in Vitro and in Vivo. Tumor Biol. 2016, 37, 11743–11752. [Google Scholar] [CrossRef] [PubMed]
  285. Yu, K.; Toral-Barza, L.; Shi, C.; Zhang, W.-G.; Lucas, J.; Shor, B.; Kim, J.; Verheijen, J.; Curran, K.; Malwitz, D.J.; et al. Biochemical, Cellular, and In Vivo Activity of Novel ATP-Competitive and Selective Inhibitors of the Mammalian Target of Rapamycin. Cancer Res. 2009, 69, 6232–6240. [Google Scholar] [CrossRef] [PubMed]
  286. Liu, Z.; Liu, J.; Li, L.; Nie, D.; Tao, Q.; Wu, J.; Fan, J.; Lin, C.; Zhao, S.; Ju, D. Inhibition of Autophagy Potentiated the Antitumor Effect of Nedaplatin in Cisplatin-Resistant Nasopharyngeal Carcinoma Cells. PLoS ONE 2015, 10, e0135236. [Google Scholar] [CrossRef] [PubMed]
  287. Zhang, Q.-Y.; Mao, J.-H.; Liu, P.; Huang, Q.-H.; Lu, J.; Xie, Y.-Y.; Weng, L.; Zhang, Y.; Chen, Q.; Chen, S.-J.; et al. A Systems Biology Understanding of the Synergistic Effects of Arsenic Sulfide and Imatinib in BCR/ABL-Associated Leukemia. Proc. Natl. Acad. Sci. USA 2009, 106, 3378–3383. [Google Scholar] [CrossRef] [PubMed]
  288. Cleveland, J. Targeting Autophagy: A Novel Anticancer Strategy with Therapeutic Implications for Imatinib Resistance. Biologics 2008, 2, 201–204. [Google Scholar] [CrossRef] [PubMed]
  289. Burchert, A. Roots of Imatinib Resistance: A Question of Self-Renewal? Drug Resist. Updates 2007, 10, 152–161. [Google Scholar] [CrossRef] [PubMed]
  290. Narrandes, S.; Xu, W. Gene Expression Detection Assay for Cancer Clinical Use. J. Cancer 2018, 9, 2249–2265. [Google Scholar] [CrossRef] [PubMed]
  291. Wang, J.; Fang, Y.; Yan, L.; Yuan, N.; Zhang, S.; Xu, L.; Nie, M.; Zhang, X.; Wang, J. Erythroleukemia Cells Acquire an Alternative Mitophagy Capability. Sci. Rep. 2016, 6, 24641. [Google Scholar] [CrossRef]
  292. Cao, Y.; Cai, J.; Li, X.; Yuan, N.; Zhang, S. Autophagy Governs Erythroid Differentiation Both in Vitro and in Vivo. Hematology 2016, 21, 225–233. [Google Scholar] [CrossRef]
  293. 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]
  294. Horne, D.J.; Graustein, A.D.; Shah, J.A.; Peterson, G.; Savlov, M.; Steele, S.; Narita, M.; Hawn, T.R. Human ULK1 Variation and Susceptibility to Mycobacterium tuberculosis Infection. J. Infect. Dis. 2016, 214, 1260–1267. [Google Scholar] [CrossRef] [PubMed]
  295. Ohshima, J.; Lee, Y.; Sasai, M.; Saitoh, T.; Su Ma, J.; Kamiyama, N.; Matsuura, Y.; Pann-Ghill, S.; Hayashi, M.; Ebisu, S.; et al. Role of Mouse and Human Autophagy Proteins in IFN-γ–Induced Cell-Autonomous Responses against Toxoplasma gondii. J. Immunol. 2014, 192, 3328–3335. [Google Scholar] [CrossRef] [PubMed]
  296. Chang, H.; Zou, Z. Targeting Autophagy to Overcome Drug Resistance: Further Developments. J. Hematol. Oncol. 2020, 13, 159. [Google Scholar] [CrossRef] [PubMed]
  297. Sharma, P.; McAlinden, K.D.; Ghavami, S.; Deshpande, D.A. Chloroquine: Autophagy Inhibitor, Antimalarial, Bitter Taste Receptor Agonist in Fight against COVID-19, a Reality Check? Eur. J. Pharmacol. 2021, 897, 173928. [Google Scholar] [CrossRef]
  298. 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] [PubMed]
  299. Wu, Y.; Ni, Z.; Yan, X.; Dai, X.; Hu, C.; Zheng, Y.; He, F.; Lian, J. Targeting the MIR34C-5p-ATG4B-Autophagy Axis Enhances the Sensitivity of Cervical Cancer Cells to Pirarubicin. Autophagy 2016, 12, 1105–1117. [Google Scholar] [CrossRef] [PubMed]
  300. Zhan, L.; Li, J.; Wei, B. Autophagy Therapeutics: Preclinical Basis and Initial Clinical Studies. Cancer Chemother. Pharmacol. 2018, 82, 923–934. [Google Scholar] [CrossRef]
  301. Mizushima, N.; Levine, B. Autophagy in Human Diseases. N. Engl. J. Med. 2020, 383, 1564–1576. [Google Scholar] [CrossRef]
  302. Sotelo, J.; Briceño, E.; López-González, M.A. Adding Chloroquine to Conventional Treatment for Glioblastoma Multiforme: A Randomized, Double-Blind, Placebo-Controlled Trial. Ann. Intern. Med. 2006, 144, 337–343. [Google Scholar] [CrossRef]
  303. Jutten, B.; Keulers, T.G.; Schaaf, M.B.E.; Savelkouls, K.; Theys, J.; Span, P.N.; Vooijs, M.A.; Bussink, J.; Rouschop, K.M.A. EGFR Overexpressing Cells and Tumors Are Dependent on Autophagy for Growth and Survival. Radiother. Oncol. 2013, 108, 479–483. [Google Scholar] [CrossRef] [PubMed]
  304. Jutten, B.; Rouschop, K. EGFR Signaling and Autophagy Dependence for Growth, Survival, and Therapy Resistance. Cell Cycle 2014, 13, 42–51. [Google Scholar] [CrossRef] [PubMed]
  305. Rouschop, K.M.A.; van den Beucken, T.; Dubois, L.; Niessen, H.; Bussink, J.; Savelkouls, K.; Keulers, T.; Mujcic, H.; Landuyt, W.; Voncken, J.W.; et al. The Unfolded Protein Response Protects Human Tumor Cells during Hypoxia through Regulation of the Autophagy Genes MAP1LC3B and ATG5. J. Clin. Investig. 2010, 120, 127–141. [Google Scholar] [CrossRef]
  306. Wolpin, B.M.; Rubinson, D.A.; Wang, X.; Chan, J.A.; Cleary, J.M.; Enzinger, P.C.; Fuchs, C.S.; McCleary, N.J.; Meyerhardt, J.A.; Ng, K.; et al. Phase II and Pharmacodynamic Study of Autophagy Inhibition Using Hydroxychloroquine in Patients With Metastatic Pancreatic Adenocarcinoma. Oncologist 2014, 19, 637–638. [Google Scholar] [CrossRef] [PubMed]
  307. Trial of Ulixertinib in Combination with Hydroxychloroquine in Patients with Advanced Gastrointestinal (GI) Malignancies—Full Text View—ClinicalTrials.Gov. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05221320 (accessed on 6 August 2023).
  308. Sirolimus or Vorinostat and Hydroxychloroquine in Advanced Cancer—Full Text View—ClinicalTrials.Gov. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT01266057 (accessed on 6 August 2023).
  309. Study Record|ClinicalTrials.Gov. Available online: https://www.clinicaltrials.gov/study/NCT04214418?tab=results (accessed on 6 August 2023).
  310. Paricalcitol and Hydroxychloroquine in Combination with Gemcitabine and Nab-Paclitaxel for Advanced Pancreatic Cancer—Full Text View—ClinicalTrials.Gov. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04524702 (accessed on 6 August 2023).
  311. ABemacicliB or Abemaciclib and HydroxYchloroquine to Target Minimal Residual Disease in Breast Cancer—Full Text View—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04523857 (accessed on 13 May 2023).
  312. Combined Carfilzomib and Hydroxychloroquine in Patients with Relapsed/Refractory Multiple Myeloma—Full Text View—ClinicalTrials.Gov. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04163107 (accessed on 6 August 2023).
  313. LY3214996 +/- HCQ in Pancreatic Cancer—Full Text View—ClinicalTrials.Gov. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04386057 (accessed on 6 August 2023).
Figure 1. The mechanism of autophagy.
Figure 1. The mechanism of autophagy.
Ijms 25 07459 g001
Figure 2. Inhibitors targeting key components of autophagy at each stage.
Figure 2. Inhibitors targeting key components of autophagy at each stage.
Ijms 25 07459 g002
Table 1. Subset of autophagy inhibitors for anticancer preclinical research.
Table 1. Subset of autophagy inhibitors for anticancer preclinical research.
Targets Inhibitors The Combination The Molecular Mechanisms of Anticancer ActionReferences
Class III PI3KKU55933Gefitinib
  • Blocking the activity of ABCG2 in colorectal cancer
[82,83,84]
Gö6976Retinoic acid
  • Inhibiting DNA damage-induced G2 arrest and reverting metastatic phenotype in aggressive melanoma by reversing the E- to N-cadherin switch
[85,86,87,88]
AZ7328-
  • Inhibiting proliferation and AKT substrate phosphorylation in bladder cancer cells
[89]
AZD5363CQ
  • Inducing apoptosis and delaying tumor progression in prostate cancer cells
[90]
RAB5A-
  • Blocking autophagy by activating mTOR pathway in gastric cancer cells
[91]
Lipoic Acid -
  • Blocking autophagy by activating mTOR pathway in lung cancer cells
[92]
OxymatrineDoxorubicin
  • Activating PI3K/AKT/ mTOR pathway
[79]
VPS34Spautin-1Imatinib (IM)
  • Inhibiting IM-induced autophagy in a Beclin-1-dependent manner in the K562 cells
[93,94,95,96]
PIK-III-
  • Blocking de novo lipidation of the microtubule-linked protein 1-light chain 3 (LC3)
[97,98,99]
Compound 31-
  • blocking VPS34 and mTORC1 signaling
[100,101]
VPS34-IN1-
  • blocking VPS34 and mTORC1 signaling
[102,103]
SAR405Everolimus
  • Reducing cell proliferation in renal cancer cells
[104,105]
Autophagy flux LucanthoneTemozolomide
  • Inducing lysosomal membrane permeabilization
  • Enhancing TMZ efficacy in glioma stem cells
[106,107]
Toosendanin (TSN) Irinotecan
  • Deacidifying lysosome in triple-negative breast cancer cells (TNBC)
[80]
4-Acetylantroquinon BCisplatin
  • Inhibiting PI3K/AKT/mTOR/p70S6K signaling pathway
[108]
ARN5187-
  • Blocking the late stage of autophagy
[109,110]
Ganoderma Lucidum polysaccharide CQ
  • Inducing autophagosome accumulation and apoptosis
[111,112,113]
Oxautin-1
  • Inhibiting autophagosome biogenesis and maturation
[114]
Tambjamine-
  • Inducing lysosomal deacidification
[115]
Dauricine and
daurisoline
Camptothecin (CPT)
  • Inhibiting the lysosome V-type ATPase activity
  • Inhibiting lysosomal degradation of autophagic vacuoles in HeLa cancer cells
[116]
BerbamineIcotinib
  • Inducing autophagosome accumulation by blocking autophagosome–lysosome fusion in lung cancer cells
[81,117]
MonensinRapamycin
  • Disrupting lysosome acidification
  • Enhancing cell cycle arrest and apoptosis induced by mTOR or EGFR inhibitors in lung cancer cells
[118,119,120,121]
Madangamines-
  • Inhibiting lysosomal function by increasing lysosomal pH
[122]
ElaiophylinCisplatin
  • Inhibiting autophagic flux at the late stage of autophagy in ovarian cancer cells
[123]
Pulsatilla saponin D (PSD)CPT
  • Blocking the autophagosome–lysosome fusion process, elevating lysosomal pH and inhibiting lysosomal cathepsins activation in human breast cancer cells
[72]
Schizocapsa plantaginea Hance 1 -
  • Inhibiting autophagosome–lysosome fusion
[124]
LiensinineDoxorubicin
  • Inhibiting autophagy/mitophagy and sensitizing breast cancer cells to doxorubicin through DNM1L-dependent mitochondrial fission
[125]
Z- LigustilideTamoxifen
  • Inhibiting autophagy and accumulating DNA damage in breast cancer cells
[126]
GinsenosideCisplatin
  • Activating AMPK and attenuating mTOR phosphorylation
  • Inducing apoptosis and enhancing cell cycle alterations in bladder cancer cells
[67,72]
[6]-Gingerol Cisplatin
  • Reducing the expression of VEGF, FLT1, KDR, and Bcl-2 genes in ovarian cancer cells
  • Inhibiting AMPK and AKT/mTOR signaling pathways
[127,128]
Jolkinolide B-
  • Inhibiting mTOR-induced AKT feedback activation
[129]
Misaponin B -
  • Inducing G2/M arrest and cytokinesis failure
[130]
Clomipramine-
  • Blocking autophagolysosomal fluxes
[131,132,133]
Chloroquine
(CQ)
Tetrandrine
  • Blocking autophagosome fusion and degradation in lung and liver cancer cells
[67,72]
Hydroxychloroquine
(HCQ)
Resveratrol
  • Blocking autophagosome fusion and degradation in osteosarcoma cells
[134]
QuinacrineCediranib
  • Accumulating autophagic vacuole and leading to apoptosis in intracranial mouse glioma
[67,72,73,135]
Lys05 -
  • Deacidifying the lysosome and blocking the late stage of autophagy
[136,137]
Compound 30-
  • Blocking autophagosome formation
[110,138]
Verteporfin-
  • Blocking autophagosome formation
[139,140,141]
Clarithromycin5-fluorouracil (5-FU)
  • Modulating the autophagic flux leads to apoptosis in colorectal cancer cells
[142,143,144]
DQ661Gemcitabine
  • Targeting protein-palmitoyl thioesterase 1 and affecting lysosomal function in pancreatic cancer cells
[145,146]
VATG-027/VATG-032-
  • Deacidifying the lysosome and disrupting the autophagosome
[147]
Mefloquine-
  • Disrupting autophagic flux by inducing mitochondrial autophagy
[148]
WX8 family-
  • Disrupting lysosome fission by tubulation and increasing the trafficking of molecules in lysosomes without elevating lysosomal acidity
[149]
Vacuolin-1-
  • Inducing the accumulation of autophagosomes by activating RAB5A
[150,151]
DesmethylclomipramineDoxorubicin
  • Blocking autophagic flux and sensitizing cells to cytotoxic agents
[133,152]
ROC-325Azacitidine (AZA)
  • Increasing autophagosome accumulation in acute myeloid leukemia
[153,154]
Trifluoperazine-
  • Inducing G0/G1 arrest and apoptosis
[155,156]
Squaramides-
  • Arresting the cell cycle at phase G1 and caspase-dependent apoptosis
[157,158]
FV-429-
  • Blocking autophagic flux and induced autophagosome accumulation
[159,160,161]
GNS561/Ezurpimtrostat -
  • Inhibiting the late stage of autophagy by inducing lysosomal dysregulation
[162,163]
Pantoprazole Docetaxel
  • Inhibiting acidification of endosomes or autophagosome–lysosome fusion
[164,165]
LAI-1Cisplatin
  • Inducting lysosomal dysfunction and blocking autophagy–lysosome formation in lung cancer cells
[166]
Tambjamines
(anion-selective ionophores)
-
  • Blocking autophagy by inducting of lysosomal dysfunction
[167]
IITZ-01 and IITZ-02-
  • Blocking autophagy by disrupting lysosomal enzymes and pH in TNBC
[168]
CUR5gCisplatin
  • Blocking the recruitment of STX17 to autophagosomes via a UVRAG-dependent mechanism in NSCLC cells
[166,169]
V-ATPaseBafilomycin A1 (Baf A1)CQ
  • Decreasing mitochondrial quality and bioenergetic function in primary neurons
[170,171,172,173,174]
Concanamycin A Vorinostat
  • Inducing apoptosis in oral squamous cell carcinoma cells
[175,176]
ATGsNSC185058-
  • Inhibiting ATG4B enzymatic activity and blocking autophagic flux
[177,178]
TioconazoleNicardipine
  • Inhibiting autophagy and promoting cell death in glioma stem cells
[37,179,180]
LV-320-
  • Inhibiting ATG4B enzymatic activity and blocking autophagic flux
[181]
S130Caloric restriction
  • Inhibiting ATG4B and inducing cell death in colorectal cancer cells
[182]
ULKULK-101KRAS-targeted drug (AMG-510)
  • Inhibiting autophagy induction and autophagic flux in NSCLC cells
[33,183,184,185]
ULK-100-
  • Inhibiting autophagy induction and autophagic flux
[33,183,184]
MRT67307Amino acid withdrawal
  • Blocking mTOR-dependent autophagy
[186]
SBI-0206965Cisplatin/AZD8055
  • Blocking cisplatin-induced autophagy and promoting cell death
  • Inhibiting AMPK and increasing apoptosis in lung cancer cells
[33,187,188,189]
Compound 6-
  • Inhibiting autophagy by blocking ULK 1/2
[190]
MRT68921WZ4003
  • Inhibiting autophagy by blocking ULK 1 and NUAK1
[191]
DCC-3116Encorafenib and Cetuximab
  • Blocking autophagosome formation and ULK1/2 protein kinase in colorectal cancer cells
[192,193]
XST-14Sorafenib
  • Suppressing the invasion and proliferation of hepatocellular carcinoma cells by blocking ULK1 activity
[194]
MiR-93CQ or NSC185085
  • Downregulating BECN1, ATG5, ATG4B, and SQSTM1 proteins in glioblastoma cells
[195]
SR-17398-
  • Inhibiting autophagy by blocking ULK1
[196]
Acid proteases Pepstatin AE64d
  • Inducing autophagolysosome accumulation
[197,198,199,200,201,202,203]
LeupeptinCycloheximide
  • Inhibiting the autophagic vacuole formation and the sequestrations of cytoplasmic and lysosomal enzymes in pancreatic acinar cells
[203,204,205]
Tyrosine kinase IMCQ/Clarithromycin/BafA1
  • Inhibiting Hedgehog signaling pathway and overcoming drug resistance of BCR-ABL-positive chronic myeloid leukemia (CML) cells
  • Increasing ROS production
[206,207,208,209]
SorafenibCQ/3-MA
  • Blocking autophagy, reducing intracellular energy synthesis, and causing lipid accumulation in hepatocellular carcinoma cells
  • Impairing autophagy via ATG4B inhibition in glioblastoma cells
[210,211,212]
SunitinibCQ/Bromodomain
  • Inducing apoptosis in renal cancer cells
  • Inhibiting autophagy through GDF15 suppression in melanoma cells
[213,214,215]
LinifanibCQ, HCQ or 3-MA
  • Inhibiting autophagy and sensitizing hepatocellular carcinoma cells to linifanib
[216]
GefitiniHCQ/BafA1/3-MA,
Clarithromycin/EGCG/CQ
  • Inducing mitochondrial apoptosis in TNBC cells
  • Inhibiting autophagy through targeting ERK phosphorylation in NSCLC cells
[217,218,219,220,221]
ErlotinibShikonin/CQ
  • Overcoming the innate resistance of wild-type EGFR in NSCLC cells to erlotinib
  • Inhibiting autophagy in lung cancer cells through modulating endoplasmic reticulum stress
[222,223]
CediranibQuinacrine
  • Enhancing anti-vascular and antitumor efficacy of cediranib in intracranial mouse glioma cells
[224]
CarfilzomibEmodin/CQ/HCQ
  • Increasing cellular ROS production and inducing apoptosis in multiple myeloma cells
[225,226,227]
Proteasome BortezomibSolamargine/BafA1/CQ,
3-MA/ATG7 siRNA
  • Inducing autophagy-mediated apoptosis and enhancing bortezomib activity in multiple myeloma cells
  • Inhibiting autophagosome–lysosome fusion by blocking acidification
  • Enhancing apoptosis in human glioblastoma cells and enhancing bortezomib activity in multiple myeloma cells
[228,229,230,231,232]
IxazomibABT-737
Doxorubicin
  • Inhibiting autophagy and MCL-1 expression sensitizes colorectal cancer cells to ixazomib
  • Inhibiting proteasome and autophagy sensitizes breast cancer cells to doxorubicin
[233,234]
Table 2. Overview of clinical trials on autophagy inhibitors for cancer therapy.
Table 2. Overview of clinical trials on autophagy inhibitors for cancer therapy.
Trail IDConditionStatus of StudyTreatmentPhaseOutcome MeasuresMethods
DosesDurationSingle/Combined
NCT03754179MelanomaRecruitingDabrafenib (DAB) Trametinib (TRA) Hydroxychloroquine (HCQ)1, 2Safety, efficacy DAB (150 mg/day)
TRA (2 mg/day)
HCQ (200 mg twice/day)
Started in January 2018 and completed in July 2022Combined
NCT02432417Glioblastoma, astrocytoma
(Grade IV)
WithdrawnCQ
Temozolomide
Chemoradiation
2Overall survival, side effects, tumor hypoxiaCQ (400 mg/day)
Temozolomide (75 mg/m2)
Radiation (30 fractions of 2 Gy)
Started in November 2023, and will complete in November 2023With radiotherapy
NCT05221320Advanced gastrointestinal malignancies (RAS mutation)RecruitingUlixertinib
HCQ
2Overall response rate, side effects Ulixertinib (450 mg BID twice/day)
HCQ (600 mg BID/day)
Started in May 2022, and will complete March in 2025Combined
NCT01266057Advanced cancer
types
CompletedHCQ
Sirolimus
Vorinostat
1Estimated maximum tolerated dose, safety, efficacy HCQ (200 mg/day)
Sirolimus (2 mg/day) Vorinostat (200 mg/day)
Started in April 2011 and completed in February 2021Combined
NCT04214418Gastrointestinal,
pancreatic, and agnostic
cancer (specifically,
KRAS-mutated advanced malignancies)
Active, not recruitingHCQ
Cobimetinib (MEK inhibitor)
Atezolizumab (Immune Checkpoint Blockade)
1, 2Estimated maximum tolerated dose, safety, tolerabilityHCQ (600 mg twice/day)
Cobimetinib (40–60 mg)
Atezolizumab (840 mg/day 1 and 15)
Started in February 2020, and will complete in September 2024Combined
NCT04524702Advanced pancreatic cancerActive, not recruitingParicalcitol HCQ Gemcitabine and Nab-paclitaxel2Antitumor effect, safety Paricalcitol (IV 3 times/week)
HCQ (twice a day/month)
Gemcitabine and Nab-paclitaxel IV (Over 30 min/days 1, 8, and 15)
Started in September 2020, and will complete in August 2024Combined
NCT04386057Pancreatic cancer (metastatic)Active, not recruitingHCQ
LY3214996
2Safety, antitumor activity HCQ (Twice/day by mouth)
LY3214996—not stated
Started in May 2020, and will complete in February 2024Combined
NCT04523857Breast cancerRecruitingAbemaciclib
HCQ
2Safety, efficacy Abemaciclib (100–150 mg twice/day)
HCQ (600 mg twice/day)
Started in November 2021, and will complete in December 2028Combined
NCT00765765Breast cancerTerminatedHCQ
Ixabepilone
1, 2Tumor response rate, survival rate, biomarkers of autophagy inhibitionHCQ (200 mg/day/month to 200 mg twice/day/month) Ixabepilone dose range is 32–40 mg/m2Started in February 2009 and completed in December 2011Combined
NCT03774472ER-positive HER2-negative breast cancerActive, not recruitingHCQ
Palbociclib
Letrozole
1, 2Dose response, cancer cell proliferation, cell cycle HCQ (400 mg versus recommended phase 2 dose)
Palbociclib and Letrozole—not stated
Started in August 2018, and will complete in December 2025Combined
NCT04163107Advanced multiple myeloma CompletedHCQ
Carfilzomib Dexamethasone
1Maximum tolerated dose To be definedStarted in July 2020 and completed in December 2021Combined
NCT00568880MyelomaCompletedHCQ
Bortezomib
1Safety, efficacy HCQ (200–600 mg/day) Bortezomib IV (1.0–1.3 mg/m2)Started in September 2010 and completed in June 2011Combined
NCT04892017Pancreatic ductal adenocarcinoma, melanoma, non-small cell lung cancer, colorectal cancer, and solid metastatic tumorRecruitingDCC-3116 TRA Binimetinib Sotorasib1, 2Tumor response rateDCC-3116 (orally twice/day)
TRA, binimetinib, and sotorasib—not stated
Started in June 2021, and will complete in October 2024Monotherapy
and combination
NCT04911816Pancreatic adenocarcinomaRecruitingmFOLFIRINOX
HCQ
1, 2Maximum tolerated dose of FHQ, tumor response rate mFOLFIRINOX—not stated,
HCQ orally (400–1200 mg)
Started in July 2021, and will complete in June 2028Combined
NCT04566133Bile tract carcinoma (KRAS mutation refractory)CompletedHCQ
TRA
2Progression free survival, response rate, safety HCQ orally (600 mg twice/day)
TRA orally (2 mg/day)
Started in February 2015 and completed in December 2022Combined
NCT02337309NeuroblastomaTerminatedSF11261Safe dose in the pediatric populationSF1126 IV
(3 + 3 dose escalation)
Started in July 2015 and completed in May 2018Monotherapy
NCT03037437Hepatocellular cancerRecruitingSorafenib
HCQ
2Time to tumor progression Sorafenib (400 mg/day)
HCQ (400 mg/day)
Started in February 2017, and will complete in March 2025Combined
NCT05576896Stage IV colorectal
(BRAF V600E)
RecruitingHCQ
Encorafenib
Cetuximab
Panitumumab
2Tumor response, survival, safety HCQ—not stated
Encorafenib (300 mg/day)
Cetuximab (250 mg/m2–400 mg/m2) Panitumumab—not stated
Started in October 2022, and will complete in July 2025Combined
NCT05036226Advanced solid tumors or relapse prostate cancerRecruitingHCQ
Metformin
Sirolimus
Dasatinib and nelfinavir
1, 2Maximum tolerated dose, quality of life, disease control rateHCQ (600mg twice/day)
Metformin (500 mg/day for 7 days, then increase to 1000 mg/daily)
Sirolimus (0.5 mg/day)
Dasatinib and nelfinavir—not stated
Started in March 2022, and will complete in October 2025Combined
NCT05070104Metastatic colorectal cancerWithdrawn (no funding)CPI-613 (Devimistat) Modified FFX Bevacizumab1Safety and tolerability, objective response rate, overall survivalCPI-613 (250–1000 mg/m2)
Irinotecan (50 mg/m2),
Leucovorin (400 mg/m2),
Oxaliplatin (85 mg/m2),
5FU (2400 mg/m2)
Bevacizumab (5 mg/kg)
Started in March 2023, and will complete in November 2024Combined
NCT05708326Chronic lymphocytic leukemia,
small lymphocytic lymphoma
Com-pletedIntermittent FastingA Case Crossover StudyChanges in lymphocytes, metabolites, autophagy, gene expression, inflammation, gut microbiome5:2 Method (intermittent fasting regimen)
16/8 Method (intermittent fast regimen)
Started in June 2023, and completed in March 2024-
NCT04527549Melanoma (Stage IIIC or IV BRAF V600 E/K)Active, not recruitingDAB
TRA with or without HCQ
2Progression-free survival, overall survival, adverse events, treatment durationTo be definedStarted in June 2021, and will complete in November 2025Combined
NCT05763992Triple-negative breast cancerRecruitingChemoimmunotherapy and fasting2Pathologic response, survival, safety, compliance, adverse eventsControl diet or fasting-like approach
Anthracycline–taxane–carboplatin chemotherapy plus pembrolizumab
Started in May 2023, and will complete in May 2026-
NCT00813423Advanced solid tumors (not responded to chemotherapy)CompletedSunitinib Malate
HCQ
1Dose response, survival, efficacy, biomarkers, safety To be definedStarted in February 2010 and completed in July 2023Combined and
monotherapy
NCT04841148Breast cancer ER positive (disseminated tumor cells)RecruitingAvelumab or HCQ with or without palbociclib2Safety, efficacy, recurrenceAvelumab (10 mg/kg)
HCQ (600 mg twice/day)
Palociclib (125 mg/day)
Started in June 2021, and will complete in May 2028Combined
NCT02512926Children with solid tumors (relapsed/refractory) or leukemiaCompletedCarfilzomib Cyclophosphamide Etoposide1Dose response, toxicity, biomarkers, genomic predictors To be definedStarted in February 2016, and completed in January 2024Combined
NCT05448677Hepatocellular carcinoma (unresectable)RecruitingEzurpimtrostat Atezolizumab
Bevacizumab
2Progression-free survival, objective response rate, tumor response Ezurpimtrostat—not stated
Atezolizumab (1200 mg/day)
Bevacizumab (15 mg/kg)
Started in December 2022, and will complete in December 2025Combined
NCT03598595Osteosarcoma (recurrent or refractory)Active, not recruitingGemcitabine
Docetaxel
HCQ
1, 2Maximum tolerated dose, disease control rate, event-free survival, overall responseTo be definedStarted in January 2019, and will complete in September 2024Combined
NCT03529448GlioblastomaRecruitingTN-TC11G (THC + CBD)
Temozolomide
Radiation
1, 2Dose, efficacy, safety, survival, biomarkers TN-TC11G—not stated.
Temozolomide (75 mg/m2, 150 mg/m2, 200 mg/m2)
Radiation (1.8–2.0 Gy/day) (total dose 58–60 Gy)
Started in July 2023, and will complete in December 2025Combined
NCT04201457Glioma of the brain RecruitingDAB
TRA
HCQ
1, 2Dose, efficacy, PK, safety, biomarkers, progressionTo be definedStarted in January 2020, and will complete in June 2029Combined
NCT02339168Hormone-resistant prostate cancerActive, not recruitingEnzalutamide
Metformin Hydrochloride
1Toxicity, efficacy, PSA response, survival, radiographic progression To be definedStarted in June 2016, and will complete in December 2024Combined
NCT01480154Advanced solid tumors (melanoma, prostate or kidney cancers)Active, not recruitingMK2206 (inhibitor) and HCQ1Toxicity, dose, autophagy biomarkers To be definedStarted in November 2011, and its primary completion date was February 2020Combined
NCT04132505Metastatic pancreatic cancer (KRAS mutation)RecruitingBinimetinib
HCQ
1Dose, efficacy, safety, survival, biomarkers, body composition changes To be definedStarted in October 2019, and will complete in December 2023Combined
NCT04873895Metastatic colorectal cancer (liver dominant)RecruitingTACE (transarterial chemoembolization)
Axitinib
HCQ
1Safety, liver response, progression-free overall survival TACE (4–8-week intervals)
Axitinib (5 mg twice/day)
HCQ (600 mg twice/day)
Started in January 2022, and will complete in December 2024Combined
NCT04190433Lymphoma, sarcoma, breast cancerWithdrawnAnthracycline
Lisinopril
Pravastatin Spironolactone
2Cardiac function, recovery rates, time to recovery To be definedStarted in September 2020 and completed in April 2023Combined
NCT02316340Colorectal cancerCompletedVorinostat
HCQ
Regorafenib
2Progression-free survival Vorinostat (400 mg by mouth/day)
HCQ (600 mg by mouth/day)
Regorafenib (160 mg by mouth/day)
Started in February 2015 and completed in April 2018Combined
NCT01206530Rectal cancer, colon cancer metastasisCompletedHCQ
Oxaliplatin
Leucovorin
5-FU
Bevacizumab
1, 2progression-free survival, overall survival, toxicity incidence, autophagy markersHCQ (600 or 800 mg) Oxaliplatin (85 mg/m2) Leucovorin (400 mg/m2) 5-FU (400–2400 mg/m2) Bevacizumab—not statedStarted in September 2010 and completed in September 2017Combined
NCT01978184Pancreatic cancerCompletedGemcitabine
Abraxane
HCQ
2Histopathologic response, survival, toxicityGemcitabine (1000 mg/m2)
Abraxane (125 mg/m2)
HCQ (1200mg)
Started in November 2013 and completed in February 2018Combined
NCT01510119Metastatic clear cell renal cell carcinomaCompletedRAD001
HCQ
1, 2Disease control RAD001 (10 mg/day by mouth)
HCQ (400 mg twice/day by mouth)
Started in September 2011 and completed in January 2017Combined
NCT02257424Advanced BRAF mutant melanomaCompletedHCQ
TRA
DAB
1, 2Maximum tolerated dose, progression-free survival rateHCQ—not stated
TRA (2 mg/day)
DAB (150 mg twice/day by mouth)
Started in October 2014 and completed in October 2021Combined
NCT01023477Breast cancerCompletedCQ1, 2Tumor size, cancer cell proliferation index, treatment-related adverse events CQ (250 mg/week in phase 1)
CQ (500 mg/week in phase 2)
Started in December 2009 and completed in October 2016Monotherapy
NCT01777477Pancreatic cancerCompletedCQ
Gemcitabine
1Maximum tolerated dose CQ (100 mg, 200 mg or 300 mg)
Gemcitabine (1000 mg/m2)
Started in July 2012 and completed in May 2015Combined
NCT01469455MelanomaCompletedDT01CQ1Tolerability, safety, pharmacokineticsDT01 (16–64 mg/3 times a week)
CQ—not stated
Started in October 2011 and completed in July 2015Combined
NCT02378532GlioblastomaCompletedCQ
Temozolomide
Radiotherapy
1Toxicity, pharmacokinetics, maximum tolerated dose, autophagic markers, EGFRvIII statusCQ (200–600 mg)
Temozolomide (75 mg/m2)
Started in August 2016 and completed in July 2019Combined
NCT04397679GlioblastomaRecruitingCQ
Temozolomide
Radiotherapy
2Overall adverse events, incidence of dermatitis toxicityTo be definedStarted in August 2021, and will complete in April 2025Combined
NCT01438177Multiple myelomaTerminatedCQ
Velcade
Cyclophosphamide
2Response rate, adverse events, response relative to autophagyCQ (500 mg/day by mouth)
Velcade (1.3 mg/m2)
Cyclophosphamide (50 mg twice/day by mouth)
Started in October 2011 and completed in February 2014Combined
NCT01006369Colorectal cancerCompletedHCQ2Progression-free survival, overall response rate, safety, disease control rate, response duration, autophagy biomarkers HCQ (200 mg/day)Started in May 2009 and completed in April 2016Combined with
capecitabine,
oxaliplatin, and
bevacizumab
NCT01978184Pancreatic cancerCompletedHCQ
Gemcitabine
Abraxane
2Histopathology, survival, treatment responseHCQ (1200 mg twice/day)
Gemcitabine (1000 mg/m2)
Abraxane (125 mg/m2)
Started in November 2013 and completed in February 2018Combined
NCT00224978GlioblastomaCompletedCQ3Survival after surgery, survival at two yearsTo be definedStarted in January 2005, and its completion date was in August 2005Monotherapy
NCT01446016Breast cancerCompletedCQ
Paclitaxel
Docetaxel
Abraxane
Ixabepilone
2Overall response rate, progression-free survival, overall survivalCQ (250 mg/day)
Paclitaxel-175 mg/m2
Docetaxel (75 mg/m2)
Abraxane (260 mg/m2)
Ixabepilone (40 mg/m2)
Started in September 2011, and its completion date was in March 2019Combined
NCT02496741IDH1/2-mutated solid tumorsCompletedCQ
Metformin
1, 2Maximum tolerated dose, D2HG concentration in serum/urine/bile/ tumor, tumor response, dose response CQ (once/day)
Metformin (twice/day)
Started in November 2015 and completed in November 2019Combined
NCT02232243Solid tumorCompletedHCQ1Number of patients with elevated Par-4 levels, optimal HCQ dose based on Par-4 toxicity and responseHCQ (200 mg twice/day)Started in July 2015 and completed in December 2018Monotherapy
NCT01273805Pancreatic cancerCompletedHCQ2Progression-free survival rate, tumor response rate, overall survival, toxicityHCQ (600 mg twice/day by mouth)Started in January 2011 and completed in February 2014Monotherapy
NCT01649947Lung cancerCompletedHCQ
Paclitaxel
Bevacizumab
Carboplatin
2Tumor response rate, progression-free survival, overall survival, adverse events HCQ (400 mg/day)
Paclitaxel (200 mg/m2)
Bevacizumab (15 mg/kg)
Carboplatin IV (over 15–30 min)
Started in December 2011 and completed in June 2015Combined
NCT01506973Pancreatic cancerCompletedHCQ
Gemcitabine
Abraxane
1, 2Overall and one year of survivalHCQ (1200 mg/day)
Gemcitabine (1000 mg/m2)
Abraxane (125 mg/m2)
Started in December 2011 and completed in March 2022Combined
NCT00486603Central nervous system tumorsCompletedHCQ
Temozolomide
Radiation
1, 2Maximum tolerated dose, pharmacokinetics, overall survival, autophagy inhibition, toxicity, correlations with genetic markersHCQ (200 mg/day) Temozolomide (75 mg/m2)Started in October 2007 and completed in January 2014Combined
NCT02071537Malignant neoplasm CompletedCQ
Gemcitabine
Carboplatin
1Maximum tolerated dose, overall survival, time to disease progressionCQ (50–200 mg/day)
Gemcitabine (1250 mg/m2)
Carboplatin—not stated
Started in December 2014 May and completed in December 2018Combined
NCT03513211Prostate cancerCompletedHCQ
Suba-itraconazole
1, 2Dose, efficacy, safety, disease progressionHCQ (Escalating doses in Rolling 6 Phase I)
Suba-itraconazole (150 mg PO BD)
Started in August 2018, and will complete in October 2023Combined
NCT03344172Pancreatic cancerTerminatedGemcitabine
Nab-Paclitaxel
HCQ
Avelumab
2Safety, histopathologic response, changes in autophagy biomarkers, coagulation index Gemcitabine (1000 mg/m2)
Nab-paclitaxel (125 mg/m2)
HCQ (600 mg/BID/day)
Avelumab (10 mg/kg)
Started in December 2017 and completed in April 2019Combined
NCT00726596Prostate cancerCompletedHCQ2PSA response, safety, autophagy biomarkersHCQ (400–600/day)Started in August 2008 and completed in January 2018Monotherapy
NCT05680662Metastatic breast cancer and TNBCNot yet recruitingQuercetin
EGCG
Metformin
Zinc
1Invasive disease-free survival, adverse eventsQuercetin (500 mg/ day)
EGCG (300 mg/day)
Metformin (850 mg/day)
Zinc (50 mg/day)
Started in January 2023, and completed in January 2024Combined
NCT01128296Pancreatic cancerCompletedHCQ
Gemcitabine
1, 2Efficacy, safety HCQ (200–1200 mg/day) Gemcitabine (10 mg/m2/min)Started in October 2010 and completed in July 2014Combined
NCT04011410Prostate cancerActive, not recruitingHCQ2Cancer progression, ADT-free survival, progression-free survivalHCQ (200 mg twice/day by mouth)Started in December 2019, and will complete in November 2026Monotherapy
NCT01550367Renal cell cancerCompletedHCQ
Interleukin-2
1, 2Efficacy, safety, immune response HCQ (600 mg/day)
Interleukin-2 (600,000 IU/kg)
Started in March 2012 and completed in February 2019Combined
NCT04735068Non-small cell lung cancer,
KRAS mutation-related tumors
Active, not recruitingHCQ
Binimetinib
2Safety, efficacyHCQ (400 mg twice/day)
Binimetinib (45 mg twice/day)
Started in April 2021, and will complete in December 2023Combined
NCT05083780Pancreatic cancerActive, not recruitingHCQ
Chlorphensin carbamate mFOLFIRINOX
1Safety, efficacy HCQ (200 mg twice/day by mouth)
Chlorphenesin carbamate (250 mg twice/day by mouth)
mFOLFIRINOX—not stated
Started in November 2021, and will complete in December 2024Combined
NCT05518110Pancreatic cancerRecruitingHCQ
TRA
2Safety, efficacyHCQ (600 mg twice/day by mouth)
TRA (2 mg/day by mouth)
Started in May 2023, and will complete April 2025Combined
NCT03979651MelanomaCompletedHCQ
TRA
1, 2Safety, efficacy, survival, side effectsHCQ (400 mg once/day by mouth)
TRA (2 mg once/day by mouth)
Started in October 2019, and its completion date was in March 2022Combined
NCT05953350HR+/HER2− breast cancerRecruitingHCQ
Palbociclib
1, 2Safety, efficacy, dose response, survival over 12 monthsHCQ (600 mg)
Palbociclib (100 mg QD, 150 mg QD, and 200 mg QD)
Started in June 2023, and will complete in December 2024Combined
NCT01023737Advanced solid tumorsCompletedHCQ
Vorinostat
1Safe maximum dose, effectiveness in reducing tumor sizeHCQ (400 mg, 600 mg, 800 mg, 1000 mg/day by mouth)
Vorinostat (300 mg/day)
Started in November 2009, and its completion date was in January 2023Combined
NCT02421575Prostate cancerCompletedHCQ1Autophagy, PSA levels, apoptosis markers, circulating tumor cells Not definedStarted in July 2012, and its completion date was in February 2016Monotherapy
NCT05842174Hepatocellular carcinomaNot yet recruitingHCQ
Lipiodol
1, 2Local progression-free survival Not definedStarted in September 2023, and will complete in August 2028Combined
NCT05433402Stage III colon cancerWithdrawnChlorproMAZINE1, 2Overall survival, new colon cancer/ polyp’s occurrence time ChlorproMAZINE (50 mg IV)Started in July 2022, and its completion date was in September 2022Combined
NCT02466802Advanced solid tumorsCompletedRegorafenib
Sildenafil Citrate
1Safety, toxicity, antitumor effects, impact on regorafenib pharmacokineticsNot definedStarted in July 2015, and its completion date was in January 2019Combined
NCT01324596Lymphoma (large B-Cell)CompletedBortezomib3Overall survival, time to progression, response duration, overall response rates, toxicityBortezomib (100 mg by mouth)Started in April 2011, and its completion date was in June 2015Monotherapy
NCT06218524Adult recurrence glioblastomaNot yet recruitingHaloperidol Tablets
Temozolomide
2Relief percentage, overall survival, DRD2 expression, haloperidol adverse reactionsHaloperidol tablet (6 mg triple/day by mouth)
Temozolomide (150 mg/kg once/day by mouth)
Started in January 2024, and will complete in July 2028Combined
NCT06076837Metastatic pancreas cancerNot yet recruitingBotensilimab
Balstilimab
Chloroquine Phosphate
Celecoxib
1Maximum tolerated dose, safety, treatment response, survival rates, biomarker changesBotensilimab (50 mg IV)
Balstilimab (240 mg IV)
Chloroquine Phosphate
(500 mg)
Celecoxib (200 mg twice/day)
Started in December 2023, and will complete in December 2025Combined
NCT01292408Breast cancerUnknown StatusHCQ2Changes in hypoxia and autophagy markers HCQ (400–800 mg)Started in January 2011, and its completion date was in January 2012Monotherapy
NCT01430585Breast cancerTerminatedPF-04691502
Letrozole
2Changes in Ki-67, objective response, pharmacokinetic parameters, genetic alterationsNot definedStarted in March 2012, and its completion date was in December 2012Combined
NCT01697293Stage IIB-IV breast cancerTerminatedCyclophosphamide
Doxorubicin hydrochloride
1, 2Pathologic response rates, safety, biomarker analysis Cyclophosphamide (600 mg) Doxorubicin hydrochloride (60 mg)Started in January 2012, and its completion date was in June 2020Combined
NCT00411788HER-2+ metastatic breast cancerCompletedRapamycin
Trastuzumab
2Objective response rate, cardiac dysfunction incidence, molecular changesRapamycin (6 mg/day)
Trastuzumab (2–4 mg/kg)
Started in December 2006, and its completion date was in April 2010Combined
NCT01628913Pancreatic neuroendocrine tumorsTerminatedBEZ235
Everolimus
2Progression free survival, overall survival, time to treatment failure BEZ235 (400 mg twice/day by mouth)
Everolimus (10 mg/day by mouth)
Started in October 2012, and its completion date was in September 2019Combined
NCT01210911Pancreatic cancerCompletedGemcitabine
Erlotinib
Metformin
2Objective response rate and toxicity profile Gemcitabine (1000 mg/m2)
Erlotinib (100 mg)
Metformin (500–1000 mg twice/day)
Started in August 2010, and its completion date was in April 2014Combined
NCT00786682Metastatic prostate cancerTerminatedHCQ
Docetaxel
2Tumor response rate, time to disease progression, overall survival, safety HCQ (200 mg twice/day)
Docetaxel (75 mg/m2 IV)
Started in December 2008, and its completion date was in October 2010Combined
NCT00003084Prostate cancerCompletedDoxorubicin hydrochloride
Estramustine phosphate sodium
Etoposide
2PSA-based response rate Not definedStarted in December 1997, and its completion date was in November 2011Combined
NCT00657982Intermediate or high-risk prostate cancerUnknow StatusRAD0012Tumor response rate, PSA failure assessed between 3 to 5 yearsRAD001 10 mg/dayStarted in April 2008, and its completion date was in March 2010Monotherapy
NCT01313559Naive prostate cancerTerminatedPasireotide
Everolimus
2Progression-free survival, PSA levels, progression-free survival Not definedStarted in June 2011, and its completion date was in November 2012Combined
NCT00574769Advanced prostate cancerCompletedRAD001
Docetaxel
Bevacizumab
1, 2Efficacy, dose response RAD001 (2.5–5 mg/day by mouth)
Docetaxel (75 mg/m2 every 21 days IV)
Bevacizumab (15 mg/kg every 21 days IV)
Started in February 2010, and its completion date was in February 2016Combined
NCT01433913Prostate cancerCompletedMetformin hydrochloride2Tumor and serum biomarkersNot definedStarted in November 2011, and its completion date was in April 2014Monotherapy
NCT01396200Multiple myelomaCompletedHCQ
Rapamycin
1Number of adverse events, the feasibility of treatmentHCQ (600–800 mg/day by mouth)
Rapamycin (3 mg, 4 mg, 9 mg, 12 mg/day by mouth)
Started in June 2011, and its completion date was in October 2012Combined
NCT02631252Myelogenous leukemiaTerminatedHCQ
Mitoxantrone
Etoposide
1Complete response, overall survival, relapse-free survival, pharmacodynamic endpoints HCQ (600–1400 mg twice/day by mouth)
Mitoxantrone (10 mg/m2 IVPB in 50 mL NS)
Etoposide (100 mg/m2 IV)
Started in August 2016, and its completion date was in October 2017Combined
NCT01227135CMLUnknown StatusHCQ
Imatinib mesylate
2Safety, efficacy, BCR/ABL levels, drug levels, effects on progenitorsNot definedStarted in March 2010, and its primary completion date was in March 2012Monotherapy, and
Combined
NCT01689987Relapsed or refractory multiple myelomaCompletedHCQ
Rapamycin Cyclophosphamide
Dexamethasone
1Maximum tolerated dose, myeloma response, progression-free survivalNoy definedStarted in September 2012, and its primary completion date was in April 2016Combined
NCT01079767Advanced liver cancer and cirrhosisTerminatedTemsirolimus23-month disease-control rate, progression-free survival, response rates, overall survival Not definedStarted in January 2010, and its primary completion date was in December 2010Monotherapy
NCT01035229Advanced hepatocellular carcinomaCompletedEverolimus3Overall survival, time to tumor progression, disease control rate, pharmacokinetics Everolimus (2.5 mg)Started in April 2010, and its completion date was in October 2013Monotherapy
NCT00492752Advanced hepatocellular carcinomaCompletedSorafenib 3Overall survival, time, disease control rate, overall response rate, pharmacokinetics Sorafenib (400 mg twice/day by mouth)Started in October 2005, and its completion date was in July 2009Monotherapy
NCT00522665Colorectal cancerCompletedIrinotecan
Cetuximab
RAD001
1, 2Maximum tolerated dose, response rates Irinotecan (125 mg/m2 IV)
Cetuximab (250 mg/m2 IV)
Not defined
Started in August 2008, and its completion date was in February 2015Combined
NCT01628913Colorectal cancerTerminatedMK-22062Overall objective response rate, overall survival, safety, tolerability, adverse event profilesMK-2206 (200 mg by mouth)Started in August 2010, and its completion date was in August 2011Monotherapy
NCT01941953Refractory colorectal cancerCompletedMetformin
5-FU
2Disease control rate, progression-free survival, overall survivalMetformin (850 mg)
5-FU (425 mg/m2)
Started in November 2012, and its completion date was in March 2015Combined
NCT01460979Ovarian carcinoma or advanced endometrial carcinomaCompletedTemsirolimus2Progression-free survival, safety, stable disease rates, overall survivalTemsirolimus (25 mg)Started in October 2010, and its completion date was in November 2015Monotherapy
NCT01031381Ovarian, peritoneal, and fallopian tube cancer Completed RAD001
Bevacizumab
2Progression-free survival, treatment response RAD001 (10 mg once/day by mouth)
Not defined
Started in September 2010, and its completion date was in December 2014Combined
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

Hassan, A.M.I.A.; Zhao, Y.; Chen, X.; He, C. Blockage of Autophagy for Cancer Therapy: A Comprehensive Review. Int. J. Mol. Sci. 2024, 25, 7459. https://doi.org/10.3390/ijms25137459

AMA Style

Hassan AMIA, Zhao Y, Chen X, He C. Blockage of Autophagy for Cancer Therapy: A Comprehensive Review. International Journal of Molecular Sciences. 2024; 25(13):7459. https://doi.org/10.3390/ijms25137459

Chicago/Turabian Style

Hassan, Ahmed Mostafa Ibrahim Abdelrahman, Yuxin Zhao, Xiuping Chen, and Chengwei He. 2024. "Blockage of Autophagy for Cancer Therapy: A Comprehensive Review" International Journal of Molecular Sciences 25, no. 13: 7459. https://doi.org/10.3390/ijms25137459

APA Style

Hassan, A. M. I. A., Zhao, Y., Chen, X., & He, C. (2024). Blockage of Autophagy for Cancer Therapy: A Comprehensive Review. International Journal of Molecular Sciences, 25(13), 7459. https://doi.org/10.3390/ijms25137459

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