Abstract
Autophagy and apoptosis are functionally distinct mechanisms for cytoplasmic and cellular turnover. While these two pathways are distinct, they can also regulate each other, and central components of the apoptosis or autophagy pathway regulate both processes directly. Furthermore, several upstream stress-inducing signaling pathways can influence both autophagy and apoptosis. The crosstalk between autophagy and apoptosis has an integral role in pathological processes, including those related to cancer, homeostasis, and aging. Apoptosis is a form of programmed cell death, tightly regulated by various cellular and biochemical mechanisms, some of which have been the focus of drug discovery efforts targeting cancer therapeutics. Autophagy is a cellular degradation pathway whereby cells recycle macromolecules and organelles to generate energy when subjected to stress. Autophagy can act as either a prodeath or a prosurvival process and is both tissue and microenvironment specific. In this review we describe five groups of proteins that are integral to the apoptosis pathway and discuss their role in regulating autophagy. We highlight several apoptosis-inducing small molecules and biologics that have been developed and advanced into the clinic and discuss their effects on autophagy. For the most part, these apoptosis-inducing compounds appear to elevate autophagy activity. Under certain circumstances autophagy demonstrates cytoprotective functions and is overactivated in response to chemo- or radiotherapy which can lead to drug resistance, representing a clinical obstacle for successful cancer treatment. Thus, targeting the autophagy pathway in combination with apoptosis-inducing compounds may be a promising strategy for cancer therapy.
Keywords: autophagy, apoptosis
Introduction
Autophagy and apoptosis are two self-destructive processes that recycle cytoplasmic organelles and entire cells, respectively. While these pathways are distinct, they can affect each other, and dysregulation of either autophagy or apoptosis influences numerous pathological processes.1
Macroautophagy (hereafter referred to as autophagy) facilitates the proteolytic degradation of cytosolic proteins and organelles in lysosomes to generate energy and remove harmful components from cells.2 The process is set in motion by the formation of double-membrane vesicles, known as autophagosomes, which fuse with lysosomes to form autolysosomes.3 Autophagy is generally considered a cell-protective response that can also act as a prodeath process in a context-dependent manner.4−6 Malignant cancers often have elevated autophagy activity and have a functional role in tumorigenesis, tumor evasion, and resistance.7 The clinically approved antimalarial drug hydroxychloroquine (HCQ) is an autophagy inhibitor that has shown promising results at high doses in combination with standard of care (SOC) cancer chemotherapeutics in clinical trials.8 However, these results were inconsistent and HCQ has significant cardiac and retinal toxicity.9 Thus, there is intense interest in developing novel therapeutic agents that specifically target autophagy and this was recently reviewed by our group.10
Apoptosis is a form of programmed cell death that is a vital component of various processes including cell turnover, proper development and functioning of the immune system, embryonic development, and drug-induced cell death.11 Programmed cell death can be activated in two ways: through the extrinsic or intrinsic pathways. This results in degradation of chromosomal DNA, cytoskeletal and nuclear proteins, the formation of apoptotic bodies, and finally uptake by phagocytic cells. Apoptosis is an essential process during development and for maintaining the homeostasis of cell populations in tissues,11 but it is often dysregulated in malignant cells, and the evasion of apoptosis is a hallmark of cancer.12,13 As cancer cells divide and proliferate, normal control of cell death is impaired and tumor formation occurs. As such, researchers have been working on developing drugs against several targets in the apoptosis pathway. These include the B-cell lymphoma 2 (Bcl-2) family of proteins, the tumor suppressor p53, the inhibitor of apoptosis (IAP) proteins, the tumor necrosis factor receptor (TNFR) superfamily, and the cysteine-dependent aspartate-directed proteases (caspases).
Several therapeutics targeting apoptosis have entered clinical trials for the treatment of solid or hematologic cancers and have shown varying effects in patients. Venetoclax, a Bcl-2 inhibitor, has received FDA approval for hematologic malignancies,14 and Debio 1143, an IAP antagonist, has recently been granted breakthrough therapy designation for head and neck cancer.15 Similar to other apoptosis targeting drugs, however, some cancer patients acquire resistance to these therapeutics. Thus, combination treatment with other anticancer agents may be required to improve the overall outcome for patients with cancer. Apoptosis targeting drugs have been studied extensively in combination with other anticancer agents including chemotherapy, radiation therapy, and targeted small-molecule inhibitors. Moreover, recent studies have suggested that IAP antagonists could enhance the efficacy of immune checkpoint inhibitors (ICIs) for immunotherapy.16,17 Herein, we focus on the crosstalk between the apoptosis and autophagy pathways, highlighting promising combination strategies for these two approaches for the treatment of cancer.
Apoptosis
Apoptosis can be activated through the extrinsic or intrinsic pathways (Figure 1). The extrinsic pathway is initiated by extracellular “death” ligands that bind to the corresponding receptors including TNF/TNFR1, FS-7-associated surface antigen (Fas) ligand (FasL)/Fas receptor (FasR), and TNF-related apoptosis-inducing ligand (TRAIL)/ TRAIL receptor 1/2 (also known as death receptor (DR) 4/5).18 Upon binding to their ligand the TNF receptors form a trimer and recruit cytoplasmic adaptor proteins via the death domains to form the death-induced signaling complex (DISC).19 TNFR1 and DR3/4 recruit the TNFR1-associated death domain (TRADD), which then recruits Fas-associating protein with death domain (FADD) and receptor-interacting protein kinase (RIPK),20,21 while FasR directly recruits FADD.22 FADD associates with procaspase-8 via dimerization, resulting in the activation of caspase-8 and caspase-10.23,24 Once caspase-8 and -10 are activated, apoptosis is carried out by activating the effector caspases-3 and -7 and cleaving BID into truncated BID (tBID).25 By binding to caspase-8 and FADD, c-FLIP (Cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein) can inhibit caspase-8 and -10 activation.25 Caspase-8 activation can be further suppressed by cIAP1 or cIAP2, which associates with TNFR-associated factor (TRAF)1/2 at the DISC.26 The death receptors also activate the JNK and the nuclear factor kappa B (NF-κB) signaling pathways27 and RIPK1/3 mediated necroptotic cell death (Figure 1).28
Figure 1.
Schematic of the cell death pathways. The extrinsic apoptosis pathway is set in motion by the TNF ligands (e.g., TNF, TRAIL, FasL) binding to the TNF receptor superfamily, which recruits TRADD and FADD to form the DISC complex and activates caspase-8. Caspase-8 activation is regulated by TRAF1/2, cIAP1, cIAP2 and c-FLIP. The Bcl-2 family of proteins regulates the intrinsic pathway. The Bcl-2 family of proteins include BH3-only proteins, BIM, BID, BIK, NOXA, and PUMA (purple); prosurvival proteins, Bcl-2 and Bcl-xl (yellow); and prodeath proteins, BAX and BAK (dark blue). Activation of the intrinsic pathway results in the formation of BAX/BAK channels and MOMP, resulting in the release of cyto c (green) and SMAC (red) from the mitochondria. Cytosolic cyto c forms the apoptosome complex with APAF1 and pro-caspase-9, activating caspase-9. Caspase-8 can amplify the intrinsic pathway by cleaving BID into tBID. As illustrated, IAP proteins (light blue) and SMAC also regulate apoptosis. Initiator caspases activate the executioner caspases-3, -6, and -7, which carry out apoptosis. The TNF receptors can also activate the NF-κB signaling pathway and recruit RIPK1 and RIPK3 to activate mixed lineage kinase domain-like (MLKL) and implement the necroptosis pathway in a caspase-independent manner.
The intrinsic pathway senses intracellular damage and is initiated by the Bcl-2 family of proteins.29 The Bcl-2 protein family consists of prosurvival, prodeath, and Bcl-2 homology (BH)3-only proteins. Prosurvival Bcl-2 proteins (Bcl-2, Bcl-XL, Mcl-1) bind to and inhibit death proteins Bcl-2-associated X-protein (BAX) and Bcl-2 homologous antagonist/killer (BAK). To initiate apoptosis, BH3-only proteins (PUMA, BID, BIM, BAD, BIK, NOXA) bind to prosurvival Bcl-2 proteins allowing unbound BAX and BAK to oligomerize and result in mitochondrial outer membrane permeabilization (MOMP). This results in the formation of channels that release cytochrome c (cyto c) and the second mitochondria-derived activator of caspases (SMAC) into the cytosol.30−32 Mitochondrial damage and cyto c release are further mediated by tBID.33 Cytosolic SMAC perpetuates the apoptotic signal by competing with caspases for binding to the IAP proteins.31 Cytoplasmic cyto c associates with apoptotic protease-activating factor 1 (APAF1) and caspase-9 to form the caspase-9-activating apoptosome.34,35 DNA damage can activate proapoptotic BH3-only proteins, p53 upregulated modulator of apoptosis (PUMA), or caspase-2 in a p53-dependent manner.36
The extrinsic and intrinsic apoptosis pathways converge to activate executioner caspases-3, -6, and -7 by initiator caspases-2, -8, -9, and -10.37 The caspases are a group of cysteine proteases that cleave proteins at aspartic acid residues and carry out apoptosis.38,39 Nascent active caspases are regulated by the IAPs.40 There are eight IAP proteins, NAIP, cIAP1, cIAP2, XIAP, survivin, BRUCE (apollon), ML-IAP (livin) and ILP-2, that are ascribed to the IAP family by the presence of one or more baculovirus IAP protein repeat (BIR) domain. XIAP is the best characterized IAP protein and can directly inhibit capsase-3, -7, and -9 via its BIR2 and BIR3 domains. cIAP1 and cIAP2 inhibit caspase-8 activation by binding to TRAF1/2 at the DISC.26 Elevated levels of other IAP proteins (survivin, apollon, ML-IAP) have also been shown to modulate the apoptosis pathway. Survivin has been associated with evasion of apoptosis through stabilizing XIAP,41 and BRUCE has been shown to modulate SMAC and caspase-9 levels. Cytoplasmic SMAC regulates apoptosis by binding to the IAP proteins in a manner that displaces the caspases. In addition, ML-IAP regulates cytoplasmic SMAC levels through its RING domain, facilitating the ubiquitination and degradation of itself and SMAC. Finally, apoptosis is carried out by the effector caspases and results in the degradation of chromosomal DNA and cytoskeletal and nuclear proteins, formation of apoptotic bodies, and lastly uptake by phagocytic cells.
Autophagy
The autophagy pathway is spatially and temporally regulated by multiple molecular components (Figure 2a).3 Autophagy is initiated by a complex comprising UNC51-like kinases 1 or 2 (ULK1/2), the FAK family kinase-interacting protein of 200 kDa (FIP200), and autophagy-related genes (Atg)13 and Atg101.42 The ULK1 kinase initiation complex is negatively regulated by the mechanistic target of rapamycin complex 1 (mTORC1) and positively regulated by AMP-activated protein kinase (AMPK), which are both cellular energy and stress sensors.42,43 Downstream the vacuolar protein sorting 34 (Vps34)-beclin1 complex drives the nucleation of the isolation membrane.44,45 Autophagy is set in motion by the formation of an isolation membrane known as the phagophore. Nucleation facilitates the formation of a double-membrane compartment called the autophagosome that compartmentalizes targeted proteins and organelles.3 Two essential ubiquitin-like frameworks are activated, the Atg5-Atg12 conjugation step and the microtubule-associated protein light chain 3 (LC3) or Atg8 processing step. Mammals have multiple Atg8 homologues (LC3A, LC3B, LC3C, GABARAP, GABARAPL1, and GABARAPL2) involved in the autophagy pathway.46 The protease Atg4 cleaves LC3 into LC3-I and subsequently Atg5 and Atg7 mediate the conjugation of phosphatidylethanolamine (PE) to LC3-I, forming LC3-II, and direct localization to the autophagosome.47 The scaffold protein p62 (or SQSTM1) traffics ubiquitinated protein aggregates to the autophagosomes and directly binds to LC3-II.48 Finally, the autophagosome either may initially fuse with an endosome before depositing its cargo into the lysosome or may directly fuse with the lysosome to form an autolysosome. Autolysosome maturation is propagated by several lysosomal proteases that hydrolyze proteins, lipids, and nucleic acids.3
Figure 2.
Autophagy pathway and crosstalk between autophagy and apoptosis. (a) Components of the autophagy pathway are illustrated. The pathway is initiated by the ULK1/2 complex (red), which consists of ULK1 or ULK2, Atg13, FIP200, and Atg101. AMPK positively and mTORC1 negatively regulates the ULK1/2 complex. The Vps34-beclin1 complex (green) consists of Vps34, beclin1, p150, Atg14, and PI3K-III. The Vps34-beclin1 complex regulates the formation of the isolation membrane. Expansion of the phagophore is modulated by two ubiquitin-like frameworks: the Atg12 system (purple), Atg12, Atg7, Atg5, Atg10, and Atg15, and the Atg8 system (yellow), LC3, Atg4, Atg5, and Atg7. Atg2 (cyan) functions in autophagosome formation. At later stages, the autophagosome fuses with the lysosome to form the autolysosome. Here components are degraded and released back into the cell. (b) Mitophagy regulates mitochondria outer membrane permeabilization (MOMP). Mitophagy removes damaged or unwanted mitochondria, which increases the threshold for MOMP and cyto c release. Cytosolic cytochrome c activates apoptosis and can inhibit mitophagy by activating caspase-8. (c) Formation of intracellular death-induced signaling complexes at the autophagosome membranes modulates in crosstalk between autophagy and apoptosis. Atg5-Atg12-Atg16L bound to the autophagosomal membrane forms a complex with FADD and caspase-8 to induce apoptosis. Similarly, LC3-II and p62 can bind caspase-8 and initial programmed cell death.
Autophagy-Dependent Cell Death
Autophagy-dependent cell death (ADCD) defines a form of regulated cell death that strictly depends on the components of the autophagy machinery rather than the change in autophagy accommodating apoptosis or necroptosis.1,49,50 Autophagy is a cytoprotective process that responds to cell stress, but under certain conditions enhanced autophagic flux results in ADCD and the suppression of autophagy prevents cellular demise.51−53 Much of the evidence for ADCD comes from the study of Drosophila melanogaster. In these studies, suppression of essential Atg genes was shown to prevent midgut degradation, while inactivation of caspases did not affect tissue development.54,55 In the Drosophila salivary glands, both the apoptotic as well as the autophagy machinery are required for cell degradation during development.56,57 In mammals, Shimizu et al. showed that mouse embryonic fibroblasts (MEF) from apoptosis resistant double-knockout Bax/Bak mice were able to undergo cell death when exposed to the DNA damage-inducing chemotherapeutic etoposide.58 This was attributed to an increase in autophagosomes and was further suppressed by genetically or chemically inhibiting autophagy. Further validation of ADCD in mammals was demonstrated using Atg5/Bax/Bak triple-knockout mice.59 Embryos from these mice also showed significantly delayed interdigital loss and brain malformation compared to wild-type mice.58,59 In the A549 lung cancer cell line, resveratrol treatment resulted in ADCD, displaying similar sustained induction of autophagy flux that was attenuated by knocking down certain autophagy genes. GRB2-associated-binding protein 1 (GBA1) was shown to positively regulate resveratrol-dependent autophagic cell death, did not activate the apoptosis or necroptosis pathway, and was not affected by treatment with a pan-caspase inhibitor, z-VAD-FMK.60 This compensatory role of autophagy was also shown to be the main process for cell death by glucocorticoids in hematological malignancies.61,62 Autosis is a specific form of ADCD, whereby elevated autophagy flux and cell death are both dependent on the plasma membrane Na+/K+-ATPase.63 These studies suggest that cytotoxic drugs can induce cell death in an autophagy-dependent manner when the apoptotic or necroptotic pathway is not functional. Additional in vivo mammalian studies are still required to verify these observations and validate the therapeutic potential of ADCD.
Crosstalk between Apoptosis and Autophagy
Autophagy has been shown to regulate apoptosis and vice versa; however, the exact nature of this interaction is unclear. Some connections occur upstream of the autophagic and apoptotic pathways where stress-elicited signaling pathways regulate both processes. p53, a potent activator of apoptosis, can also induce autophagy through increasing the expression of damage-regulated autophagy modulator (DRAM), a target gene for p53.64 Similarly, activation of the PI3 kinase/Akt pathway, which is a well-known pathway that inhibits apoptosis, also inhibits autophagy.65 Furthermore, oncogenes, including myc,66 and major signaling pathways, such as the Ras-PI3K-mTOR axis,67 can simultaneously increase and/or decrease both autophagy and apoptosis.
Proteins that are themselves central components of the apoptosis or autophagy pathway regulate both processes directly.68 Components of the autophagy and apoptosis machinery frequently intersect and cross-regulate one another at the mitochondria (Figure 2b). Intrinsic apoptosis is initiated by MOMP and the release of cytochrome c. Mitophagy is a specific form of autophagy that selectively clears damaged mitochondria.69,70 Autophagy induction (e.g., by nutrient deprivation, sulforaphane treatment, etc.) can result in mitochondria elongation, inhibiting the release of cytochrome c71, which prevents apoptosis and sustains cell viability.72 Apoptosis induction following cytochrome c release activates caspase-8 which cleaves several Atg proteins and inactivates autophagy.73−75 This can enhance apoptosis by further promoting the release of proapoptotic factors from mitochondria.76 Alternatively, in mitochondrial apoptosis deficient cells, autophagy induction can eventually result in stress-induced cell death through caspase-8 activation.77 Furthermore, Atg12 has been shown to interact with apoptosis proteins at the mitochondria; however, the effect of this interaction on cell fate is still unclear.78,79
Another recently identified platform for crosstalk of the two signaling pathways is the autophagosomal membrane. Several groups have demonstrated the formation of intracellular DISCs (iDISC) that recruit procaspase-8 and FADD onto autophagosomes (Figure 2c).80−86 Caspase-8 is recruited to the phagophore by FADD, which interacts with the Atg5-Atg12-Atg16L complex80,83,85, or by binding to p62 and LC3.81,82,86 This complex usually forms independently from death receptor signaling and requires LC3-positive autophagic membranes. Knockdown of Atg5 or Atg7 or treatment with the autophagy inhibitor 3-MA suppresses the formation of the iDISC.80,81,84 Depending on the cellular conditions, recruited caspase-8 will either be activated at the phagophore through oligomerization and enhance cell death,80−86 or the mature autophagosomes will fuse with the lysosome, degrade caspase-8, and block apoptosis.87−89 According to Tang et al. this switch is regulated by c-FLIP and NF-kB signaling on expanding autophagosomal membranes.84 Under nutrient-rich conditions NF-kB signaling upregulates c-FLIP to suppress caspase-8 activation while starvation suppresses NF-kB translocation allowing caspase-8 activation and initiation of apoptosis.84
Several additional mechanisms of cross-regulation between autophagy and apoptosis have been identified, allowing the same protein to participate in these opposing processes. Atg5 has been demonstrated to regulate autophagy and enhance apoptosis.90 Atg5 can be cleaved by the noncaspase protease calpain, allowing truncated Atg5 to associate with Bcl-xl at the mitochondria, resulting in cytochrome c release and caspase activation.90 Betin et al. showed that Atg4D is cleaved by caspase-3 which stimulates the processing of γ-aminobutyric acid receptor-associated protein-like 1 (GABARAP-L1) and autophagosome formation. Interestingly, both genetic knockdown of Atg4D and overexpression of cleaved Atg4D resulted in increased cytotoxicity, suggesting that Atg4D is an important factor at the regulatory interface between autophagy and apoptosis.91 Caspase-10 has also been shown to inhibit autophagy by cleaving Bcl-2-associated transcription factor 1 (BCLAF1) and thus preventing ADCD in multiple myeloma.92 Furthermore, Han et al. demonstrated that caspase-9 forms a complex with Atg7, which inhibits the catalytic activity of caspase-9 and enhances LC3-II processing and autophagosome formation.93 This suggests that linking apoptosis genes to the autophagy process regulates cell death pathways and vice versa.
Necroptosis is a caspase-independent process defined as “a modality of regulated cell death triggered by perturbations of extracellular or intracellular homeostasis that critically depends on MLKL, RIPK3, and RIPK1”.50 Necroptosis has been shown to crosstalk with the apoptosis and autophagy pathways. On autophagosomal membranes, RIPK1 forms a complex with FADD:caspase-8:Atg5:Atg12:Atg16L.80 Under normal cell media conditions, active caspase-8 inhibits autophagy by blocking RIPK1 and cells die through apoptosis. In the absence of caspase-8, active RIPK1 can instruct cells to die through necroptosis rather than through ADCD.80 One study found that RIPK3 directly activated AMPK, which also resulted in phosphorylation of ULK1 and beclin1.94 Furthermore, TNF-induced necroptosis has been shown to block the lysosomal degradation of autophagosomes possibly through dysregulating the SNARE complex.94 In addition, there is evidence that blocking apoptosis with the caspase inhibitor z-VAD-FMK results in cells dying through necroptosis rather than ADCD as z-VAD-FMK also inhibits lysosomal proteases and thereby autophagy.95,96
While many studies show that autophagy is upregulated in response to cancer therapy, which then acts as an adaptive response to promote tumor cell survival, under other circumstances, autophagy induction can further promote cells to undergo cell death. One explanation for this may be that modulation of the autophagy pathway results in changes that are cell type- and context-dependent. Alternatively, commonly used pan-caspase inhibitors used to block the apoptosis pathway, as well as autophagy assays used to measure autophagosome accumulation, have several limitations and this complicates the interpretation of results.
Bcl-2 Proteins and BH3-Mimetics
The Bcl-2 protein family closely regulates the balance between cell survival and cell death.97 These prosurvival proteins (Bcl-2, Bcl-XL, Mcl-1) promote cell survival by inhibiting the proapoptotic proteins BAX and BAK, preventing BAX/BAK from oligomerizing, forming pores, and permeabilizing the outer mitochondrial membrane. The BH3-only proteins (BIM, PUMA, BID, BIK, and NOXA) initiate programmed cell death in response to stress by binding to prosurvival proteins or by activating the effectors of apoptosis.98 Characterization of the functions and structural interactions between proapoptotic and prosurvival Bcl-2 family members resulted in the development of small-molecule mimetics (BH3-mimetics) that kill cancer cells by targeting prosurvival Bcl-2 members (Table 1). To date, six BH3-mimetic drugs have reached the clinic. One of them, the Bcl-2 inhibitor, venetoclax/ABT-199,99 is currently approved for patients with chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL)14 and may become the front-line therapy for acute myeloid leukemia (AML) in the future.100−102 However, leukemic cells can acquire resistance to ABT-199 and other BH3-mimetics,103,104 suggesting that combination therapy with other antileukemic agents may be required to reduce or overcome the resistance.
Table 1. Selected Apoptosis Drugs and Their Effects on Autophagy.
Bcl-2 proteins primarily regulate the autophagy pathway by interacting with a key protein beclin1, which forms the beclin1–Vps34 core complex to induce autophagy (Figure 3a).105 Bcl-2 was initially identified to bind to beclin1 in yeast106 and later in mammalian cells to inhibit beclin1-dependent autophagy.107 Beclin1 has a BH3 domain that mediates the Bcl-2–beclin1 complex formation during low nutrient levels and downregulates autophagy during starvation.107 Beclin1 can also bind to Bcl-XL and Mcl-1.105 Proapoptotic proteins such as BAX decrease autophagy activity by enhancing caspase-mediated cleavage of beclin1 and preventing beclin1 from interacting with Vps34.108 BH3-only proteins also regulate autophagy. For example, BIK displaces nutrient-deprivation autophagy factor-1 (NAF-1) from Bcl-2 at the endoplasmic reticulum and initiates autophagy.109 Beclin1 phosphorylation at the BH3 domain by kinases further regulates its interaction with Bcl-2/Bcl-XL. Death-associated protein kinase (DAPk) phosphorylates beclin1 at T119, promoting beclin1 dissociation from Bcl-XL and autophagy induction.110 Alternatively, serine/threonine kinase 4 (STK4) phosphorylates beclin1 at the Thr108 site and suppresses autophagy by enhancing the interaction between beclin1 and Bcl-1/Bcl-XL.111,112 Additional mechanisms by which beclin1 regulates autophagy and apoptosis have been extensively reviewed by others.105,113
Figure 3.
Components of the apoptosis pathway modulate the autophagy pathway. (a) Bcl-2 proteins module the Vps34–-beclin1 complex. Beclin1 has a BH3 domain allowing it to bind to Bcl-2 proteins. Prosurvival Bcl-2 proteins (Bcl-2, Bcl-XL, and Mcl-1) can form a complex with beclin1, preventing the formation of the Vps34–beclin1 complex and downregulating autophagy. BH3-only proteins (BIK, BID, BIM, and PUMA) indirectly positively regulate autophagy. The proapoptotic protein, BAX, negatively regulates autophagy by enhancing caspase-mediated cleavage of beclin1 and preventing it from interacting with Vps34. (b) p53 can both positively and negatively modulate the autophagy pathway depending on its cellular localization. Cytoplasmic p53 negatively regulates autophagy by binding to FIP200, resulting in the destabilization of the ULK1/2 complex. Nuclear p53 positively regulates autophagy by inducing the transcription of AMPK, mTOR, and DRAM. (c) IAP proteins interact with various components of the autophagy machinery. Survivin downregulates autophagy by inhibiting the Atg12–Atg5–Atg16 complex and Atg7. Apollon positively regulates autophagy initiation through the upstream kinases AMPK and ULK1. Apollon also inhibits the fusion between autophagosomes and lysosomes. Similarly, XIAP can positively or negatively modulate the autophagy pathway via beclin1 and LC3 expression, respectively. (d) The death domain of FADD has been shown to upregulate autophagy under certain cellular circumstances, while under different cellular conditions, elevated autophagy has been shown to degrade DR4 and DR5.
BH3-mimetics have been reported to be implicated in regulating the autophagy pathway (Table 1). Recent studies have focused on ABT-199, a selective inhibitor of Bcl-2, ABT-263, and ABT-737, inhibitors of Bcl-2, Bcl-XL, and Bcl-w, and pan-Bcl-2 inhibitors, obatoclax, and gossypol.114 Most notably, studies in primary follicular lymphoma cells resistant to ABT-199 show an increase in autophagy activity assessed by a decrease in SQSTM1/p62 levels and an increase in LC3 I/II processing.115,116 Treatment of multiple myeloma or colon cancer cells by the ABT-199 analogues ABT-737 and ABT-263, respectively, similarly increased autophagy activity.81,117 BH3-mimetics, ABT-199, ABT-737, and ABT-263, competitively inhibit the interaction between beclin1 and Bcl-2/Bcl-XL, freeing beclin1 from inhibition and activating autophagy.118 Moreover, combination treatment with the autophagy inhibitor chloroquine (CQ) further enhances the ability of ABT-263 to induce apoptosis81 and can be attenuated by knocking down essential autophagy-related proteins. Bortezomib resistant multiple myeloma (MM) cells with low BIM expression were also able to overcome resistance when treated with ABT-737 and CQ.119 These studies suggest that autophagy contributes to mitigating the cytotoxicity of BH3-mimetics.114 The pan-Bcl-2 inhibitor obatoclax (GX15-070) appears to induce autophagy in various cancer cells independently of beclin1 as knockdown of beclin1 did not attenuate obatoclax-induced autophagy.120,121 However, the mechanism by which obatoclax induces autophagy is not fully understood, and questions regarding whether obatoclax-induced autophagy promotes or inhibits cell death are also under discussion.114 On one hand, obatoclax-induced autophagy has been linked to cell death,1 as genetic or pharmacological blockade of autophagy was found to inhibit cell death.114 Alternatively, inhibition of autophagy was demonstrated to enhance obatoclax-induced cell death, supporting a prosurvival function of autophagy.122 Furthermore, obatoclax has also been shown to inhibit late-stage autophagy independently of beclin1, Atg7, and Atg12.123 Gossypol, another pan-Bcl-2 inhibitor, has also been shown to induce autophagy and apoptotic cell death.124,125 In one study, cotreatment of gossypol with autophagy inhibitors potentiated cell death, suggesting that gossypol-induced autophagy is cytoprotective.125 Others have made the antithetical observation that genetic knockdown of beclin1 and/or Atg5 reduced gossypol-induced cell death.124 More recently, genetic deletion of Mcl-1 in the nervous system has been shown to induce autophagy,126 suggesting that Mcl-1 selective inhibitors may also activate autophagy.
BH3-mimetics are promising therapeutics for the treatment of hematologic malignancies, and all small molecules we reviewed appear to upregulate autophagy. Genetic knockdown experiments demonstrate that Bcl-2 proteins intersect with autophagy by interacting with beclin1, suggesting that inhibition of Bcl-2 proteins requires free beclin1 to initiate autophagy. Interestingly, not all BH3-mimetics required beclin1 to increase autophagy as beclin1 knockdown did not always attenuate inhibitor-induced autophagy. This opens two possibilities: (1) Bcl-2 proteins are able to regulate autophagy in a not yet understood beclin1-independent manner, or (2) Bcl-2 inhibitors are binding to off-target proteins involved in regulating autophagy.
p53 and MDM2-p53 Antagonists
Tumor protein p53 is a master regulator of genomic stability, cell death, proliferation, and metabolism and is the most well-characterized tumor suppressor protein.127 It is a stress-responsive transcription factor, and upon activation, p53 can promote cells to undergo apoptosis by inducing proapoptotic genes including PUMA, BAX, and BIM.1 p53 has been extensively studied in the context of cancer, and several compounds that reactivate the tumor suppressor protein have been developed.128 Murine double minute 2 (MDM2) tightly regulates and inhibits the activity of p53. Blocking the MDM2–p53 interaction with small molecules can reactivate the tumor-suppressive functions of p53.129,130 Several small-molecule MDM2 inhibitors have been successfully designed and developed (Table 1), and seven of these compounds have been advanced into human clinical trials as novel anticancer therapeutics.128 It is estimated that p53 is mutated or deleted in ∼50% of human cancers and can vary significantly between cancer types.131 Thus, the ability of these MDM2–p53 antagonists to induce apoptosis is restricted to WT p53 cancers and generally these compounds do not fully eliminate the cancer cells.129 Consequently, MDM2 antagonists are increasingly being tested in combination with other therapeutics in a search for synergy that would enhance the efficiency of these drugs to eliminate cancer.
The role of p53 in the regulation of autophagy appears to be dependent on its cellular localization, and the mechanism is not yet fully understood (Figure 3b).132 Tasdemir et al. found that cytoplasmic, but not nuclear, p53 suppresses autophagy, and knockout, knockdown, or pharmacologic inhibition of p53 induces canonical autophagy signaling.133 This enhanced autophagy helped cancer cells survive under stress. Cytoplasmic p53 inhibits autophagy by coaggregating with FIP200, resulting in the destabilization of ULK1. This blocks the formation of the ULK1–FIP200–Atg13–Atg101 complex and prevents autophagosome formation.134,135 Furthermore, induction of autophagy by starvation or rapamycin treatment stimulates proteasomal degradation of p53 and a further increase in autophagy.133 Alternatively, nuclear p53 has been shown to activate autophagy by regulating expression of the AMPK/mTOR pathway136,137 and is a key regulator of the lysosomal protein DRAM.64
Several groups have looked at the ability of MDM2–p53 antagonists to modulate autophagy in various cancer models. Nutlin-3a138 and MI-63139 have both been shown to induce p53-dependent autophagy in hematologic malignancies.140,141 This p53-dependent increase in autophagy appears to be proapoptotic as genetic or pharmacological inhibition of autophagy impaired apoptosis. CP-31398142 and RITA (reactivation of p53 and induction of tumor cell apoptosis)143 were also shown to induce autophagy; however, in pancreatic cancer cells, cotreatment with autophagy inhibitors further sensitized these cells to apoptotic cell death, suggesting a prosurvival autophagic response in this context.144 Furthermore, nutlin-3a synergizes with several anticancer therapeutics to induce autophagy and apoptosis.145−147 Some studies suggest that nutlin-3a modulates autophagy in a context-dependent manner. Duan et al. compared cancer cells sensitive to, or resistant to, nutlin-3a-induced apoptosis and found that in resistant cells nutlin-3a promoted prosurvival autophagy while in sensitive cells nutlin-3a inhibited autophagy. Additionally, cotreatment with autophagy inhibitors and nutlin-3a could sensitize the nutlin-3a resistant cancer cells to undergo cell death.148
It has been proposed that autophagy suppresses p53 in some cancers, facilitating cell survival and tumorigenesis.149−152 Therefore, cotreatment with MDM2 inhibitors and autophagy inhibitors could hyperactivate WT p53, more effectively induce apoptosis in cancer cells, and impede disease progression. However, WT p53 cancers make up only ∼50% of human cancers, which limits the clinical utility of MDM2–p53 antagonists and would require patient stratification.
IAP Proteins and Antagonists
The family of IAP proteins is involved in blocking and attenuating programmed cell death pathways, predominantly through modulation of the caspase cascade.153,154 IAP proteins cIAP1, cIAP2, XIAP, and ML-IAP are directly involved in the regulation of apoptosis.155 IAPs are often up-regulated in cancers156 and are believed to underlie the resistance of many malignant cells to chemotherapeutics.157,158 IAP antagonists, or SMAC mimetics, have received considerable attention as potential therapeutic agents for the treatment of cancer (Table 1),159,160 with several compounds under clinical evaluation for solid and hematologic cancers.161 Clinical data from these studies showed that SMAC mimetics are well tolerated, display evidence of target engagement, and have a reasonable safety profile.162 However, previous SMAC mimetics have demonstrated limited clinical efficacy as single agents, with the majority of patients showing no response to the drugs.162 More recently, there have been some advances in the clinical efficacy of IAP antagonists, with the breakthrough therapy destination approval for Debio 1143 for front line treatment of head and neck cancer,15 however, there is still a need for safe and effective IAP antagonists that synergize with other anticancer drugs.
Besides their antiapoptotic role, IAP proteins have recently been linked with modulating the autophagy pathway (Figure 3c).163 A study using hepatocellular carcinoma tissue specimens found that XIAP expression was inversely associated with LC3 expression.164 Furthermore, knockdown studies by Huang et al. demonstrated that XIAP negatively regulated the autophagy pathway through MDM2–p53 signaling in vitro and in vivo.165 Alternatively, XIAP and cIAP1 have also been shown to positively regulate beclin1-dependent autophagy expression via the NF-kB pathway.166 Survivin has been suggested to modulate autophagy by interacting with the Atg12–Atg5–Atg16L protein complex to inhibit autophagosome formation and the elongation process.167−169 Recent studies suggest that BRUCE may also play a role in autophagy, potentially at the stage of fusion between autophagosomes and lysosomes to form autolysosomes.163 However, this appears to be unclear as some studies found that BRUCE knockdown negatively regulated autophagy170 by interacting with GABARAP and GABARAPL1, and syntaxin 17171 or through the upstream kinases AMPK-ULK1.172 Alternatively, BRUCE may degrade LC3-I and thus negatively regulate autophagy.173
Recent studies have also evaluated the ability of IAP antagonists to modulate autophagy. APG-1387, a bivalent IAP antagonist that is currently under clinical evaluation for cancer and has been demonstrated to induce autophagy in ovarian cancer cells.174 Furthermore, cotreatment with autophagy inhibitors and APG-1387 further promoted cell death.174 Similarly, WX20120108, a monovalent IAP antagonist, increased autophagy activity in HeLa and MDA-MB-231 cells. Both compounds appear to increase autophagy activity as cells start to undergo cell death with an increase in compound concentration and incubation time.175 Ding et al. suggested that WX20120108 relies on activating the ROS-Foxo3 pathway to induce autophagy.175 The clinical candidate GDC-0152 also induced autophagy in HL-60 cells. However, rather than acting as a cell survival mechanism, autophagy further increased apoptosis, and cotreatment with autophagy inhibitors reduced GDC-0152-induced apoptosis.176 LCL-161 and birinapant were evaluated in an autophagy flux assay and were both shown to inhibit autophagy by blocking the fusion between autophagosomes and lysosomes.177 YM155, a survivin suppressor, was shown to upregulate autophagy and induce cell death and DNA damage in an autophagy-dependent manner.178,179
There is evidence that IAP proteins and antagonists modulate autophagy; however, the mechanism is not fully understood and may be context-dependent. While most studies demonstrate that IAP antagonists elevate both autophagy and apoptosis in certain contexts, in some cells and tissues IAP antagonists have been shown to inhibit autophagy. In the future a better understanding of the functions of IAPs may be necessary to enable us to fine-tune the therapeutic efficacy of IAP antagonists and attenuate possible side effects.
Death Receptor Ligands
The TNFR superfamily constitutes a key component of the extrinsic apoptosis pathway. Death receptor (DR)4 and DR5 are part of the TNFR family and contain “death domains” that can initiate apoptosis.180 The death receptors recruit cytoplasmic adaptor protein TRADD, FADD, and procaspase-8 to form the DISC upon binding to the ligands Apo2 ligand (Apo2L) or TRAIL. This results in the cleavage and activation of initiator caspases-8 and -10, which initiate apoptosis by activating the executioner caspases.180 Apo2L/TRAIL and molecules mimicking their functions have emerged as promising candidates for cancer therapy. This is based on their ability to induce apoptotic cell death in cancerous cells without causing significant toxicity to healthy cells.181 The recombinant human soluble protein Apo2L/TRAIL (dulanermin) and its derivatives were the first attempt to target DR4/5, and while they showed promising efficacy and safety in preclinical models, they did not achieve sufficient overall therapeutic benefit clinical trials.182 Monoclonal antibodies targeting DR4 or DR5 have also been in development, seven of which have entered clinical studies. The DR4/DR5 monoclonal antibodies were well tolerated and relatively safe in patients, but only achieved stable disease progression and no improved response rates were noted as single agents.182
Components of the extrinsic death receptor pathway have also been associated with controlling autophagy (Figure 3d). As mentioned above, TRAIL has a unique ability to induce apoptotic cell death in cancer cells while sparing healthy cells. A study has suggested that this resistance to undergo cell death in normal epithelial cells may be through the upregulation of cytoprotective autophagy.183 This was proposed to be mediated by the death domain of FADD or by AMPK signaling. During malignant transformation, cancer cells may become resistant to the death receptor ligands TNF and TRAIL. This resistance is suggested to be through autophagic degradation of DR4 and DR5,184,185 as TRAIL-resistant cancer cells have shown elevated autophagy activity and pharmacologic or genetic inhibition of autophagy resulted in the death of these cells.186,187 Alternatively, resistant glioma cells have been shown to overcome this resistance and undergo cell death in response to chemotherapeutic agents by activating autophagy-mediated cell death.188
Prostate cancer cells treated with dulanermin were shown to have elevated autophagy flux activity in TRAIL-resistant but not in TRAIL-sensitive cell lines. Cotreatment with the autophagy inhibitor CQ further enhanced dulanermin-induced cell death in TRAIL-resistant prostate cancer cells.186 HW1, a human single-chain fragment variable (scFv) antibody against DR5, was shown to induce autophagy and cell death as a single agent in both TRAIL-resistant and TRAIL-sensitive cells. Furthermore, HW1-induced cell death appeared to be through autophagy-mediated cell death, since knockdown of the autophagy protein beclin1 or Atg7 greatly attenuated HW1-induced cell death.189
Unlike the other apoptosis-inducing drug targets, limited research has been reported on the role of DR antibodies in autophagy. As previously mentioned, death receptors form DISCs in response to TRAIL/TNF-α treatment at the plasma membrane, which results in caspase-8 activation. While it is known that caspase-8 modulates autophagy by cleaving several autophagy proteins, the extent by which DR antibody-dependent caspase activation affects autophagy remains to be elucidated.
Caspase Inhibitors
The caspases are a group of cysteine proteases that are central to initiating and executing the programmed cell death pathway. In mammals, there are seven major caspases that are involved in apoptosis and are classified as initiators (caspase-2, -8, -9, and -10) or executioners (caspase-3, -6, and -7). There is a third group of caspases (caspase-1, -4, and -5) involved in carrying out pyroptosis, which is an inflammatory type of regulated cell death.50 Caspases exist as inactive precursors and are activated by oligomerization, cleavage, and/or dissociation from endogenous inhibitors allowing them to cleave their substrates at specific aspartate-containing recognition sites.190
Unlike the other discussed apoptosis proteins, caspase inhibitors have been used to prevent cell death in various pathologies, including liver disease. In addition, caspase inhibitors serve as useful tool compounds to study the programmed cell death pathways and the crosstalk with the autophagy pathway. It is unsurprising that caspases also recognize and cleave many autophagy proteins.190 A study by Norman et al. showed that in vitro Atg3 is cleaved by caspases-3, -6, and -8, beclin1 is cleaved by caspase-3 and -6, p62 is cleaved by caspase-6 and -8, while Atg7, Atg9, and Atg4 homologues can be cleaved by caspase-3.191 This has been validated by other groups in cellular models, and in most cases, cleavage of autophagy proteins by caspases results in suppression of autophagy, and the homeostatic balance shifts toward apoptosis.190,192,193 Nevertheless, caspase-mediated cleavage of autophagy proteins does not always result in waste products; rather, several Atg-fragments have been shown to acquire new properties, which differ from their initial full-size isoforms and consequently affect apoptosis and autophagy in numerous ways.76,91 On the other hand, caspases have also been found to promote autophagy under certain contexts.91,93 Notably, autophagy is also able to regulate apoptosis by targeting caspases for degradation in the autolysosome.88,190
Several caspase inhibitors have been developed as research tools and as antiapoptotic therapeutics for liver disease.194 The FDA approved pan-caspase inhibitor IDN-6556 has been shown to inhibit autophagy resulting in cell death by necroptosis in mouse dermal fibroblasts.195 Peptide-like pan-caspase inhibitors including z-VAD-FMK appear to modulate autophagy in a context-dependent manner. Caspase-mediated cleavage of beclin1 inactivates autophagy, which can be inhibited by z-VAD-FMK.76,91,196 Similarly, blocking Atg3 cleavage by caspase-8 with z-VAD-FMK has been shown to increase autophagy in cells.75 Under certain cellular contexts z-VAD-FMK appears to induce ROS-accumulation and upregulate autophagy, which results in cell death via ADCD or necroptosis.80,84,197 Alternatively, others have shown that z-VAD-FMK blocks autophagy by inhibiting an off-target lysosomal protease, cathepsin B.96 This would argue that when apoptosis is attenuated by z-VAD-FMK cells undergo necroptosis, refuting published examples of ADCD and highlighting the need for more vigorous experiments when studying autophagosome accumulation.
Implications for Current and Future Cancer Therapeutics
In this review, we discussed the crosstalk between apoptosis and autophagy, while focusing on several small-molecules and biologics that target the apoptosis pathway. Several drugs that modulate apoptosis have been developed and advanced into the clinic in recent years by targeting the Bcl-2 protein family, IAP proteins, death receptors, p53, or caspases. However, these anticancer therapeutics generally lack single-agent efficacy and must be combined with other treatments in clinical trials. In vitro studies using a large panel of cell lines have shown that some cancer cells are more predisposed or “primed” to undergo cell death in response to treatment with apoptosis-inducing compounds while other cancer cells are completely resistant to treatment.198 The majority of studies described in this review demonstrate that autophagy is elevated in response to treatment with apoptosis-inducing compounds. While apoptosis occurs at the same time or is potentially preceded by autophagy, it is unclear to what extent autophagy mediates cell death.
To date, only a handful of autophagy inhibitors (CQ/HCQ, verteporfin, and clarithromycin) have entered clinical trials for the treatment of cancer.199 CQ, and its analogue HCQ, are late-stage autophagy inhibitors that block the fusion between autophagosomes and lysosomes and are being used in several clinical trials in the treatment of various cancers (ClinicalTrials.gov).199 Verteporfin is FDA approved for macular degeneration and was found to inhibit autophagy in a screen of off-patent agents.200 Verteporfin has been shown to increase the efficacy of some anticancer agents when used in combination in various cancer models201−203 and is currently in clinical trials for the treatment of prostate and pancreatic cancer (NCT03067051, NCT03033225). Clarithromycin, an antibiotic used for the treatment of bacterial infections, has also been demonstrated to attenuate autophagy204 and is currently being investigated in ongoing clinical trials for various hematologic malignancies (NCT04302324, NCT04063189). Moreover, several novel small-molecule autophagy inhibitors are in preclinical development, including ULK1/2 inhibitors (SBI-0206965,205 MRT68921206), Vps34 inhibitors (VPS34-IN1,207 SB02024,208 SAR405,209 PIK-III210), Atg4B inhibitors (NSC185058,211 UAMC-2526212), and lysosomal inhibitors (bafilomycin A1,213 Lys05214). Preclinical studies have demonstrated that some of these autophagy inhibitors can induce apoptotic cell death as single agents in various cancer types and could show promise as combination treatments with anticancer therapeutics.205,215−218
In many cases, autophagy mitigates the cytotoxicity induced by proapoptotic drugs, and suppression of autophagy by pharmacological inhibition or by genetic knockdown of Atg genes accelerates apoptotic cell death.215,216 Autophagy inhibition has also been proposed to prime cells for apoptotic cell death in response to anticancer agents by increasing Forkhead box protein O3a (FOXO3a) activity and upregulating the expression of PUMA.219,220 In vivo studies in mice have shown markedly improved survival rates due to tumor burden when combining antiapoptotic compounds with CQ compared with either drug alone.220 This suggests a prominent role for autophagy as a prosurvival response and raises the question of whether autophagy inhibitors could synergize with apoptosis drugs and provide therapeutic benefit to patients with cancer. HCQ and verteporfin have shown promising clinical results with multiple chemotherapies and radiation therapies,199 however, they have not yet been tested in combination with apoptosis targeting compounds in clinical trials. Thus, in principle, it would be feasible to investigate whether administering HCQ or verteporfin in combination with apoptosis-inducing therapeutics would improve treatment outcomes for cancer patients. In addition, novel autophagy inhibitors targeting ULK1/2, Vps34, or Atg4B could also show promise by synergizing with apoptosis therapeutics as they may have fewer off-target effects and could show efficacy at lower doses compared to HCQ.
On the other hand, apoptotic drug-induced autophagy may under different cellular contexts further promote cell death and inhibiting autophagy would suppress cell death. Loss of the essential autophagy gene beclin1 promotes tumorigenesis in mice and this has also been found to occur in human cancers.12,221 Furthermore, p62 is eliminated through autophagy, but in the absence of autophagy, p62 accumulation results in tumor growth.222,223 Paradoxically, this suggests that autophagy is a tumor-suppression mechanism, and inhibition of the pathway in combination with proapoptosis drugs would worsen the outcome. Several studies described in this review have demonstrated in vitro and in vivo that autophagy inhibition reverses the effects of apoptosis-inducing compounds. This would suggest that in certain contexts activating autophagy may be a better therapeutic approach in combination with apoptosis-inducing drugs.
In order to improve future cancer therapy, it is essential to investigate the combined effect of autophagy inhibitors and proapoptosis therapeutics not just in tumors and cancer cells but also in different cellular contexts, such as immune cells. Immunotherapies act on the immune system and enhance its ability to recognize, target, and destroy cancer cells.224,225 Immune checkpoint inhibitors (ICIs) have shown remarkable responses in clinical trials across various cancers.226 There is much focus on novel immunotherapies, such as cancer vaccines and chimeric antigen receptor (CAR) T cell therapies, as well as investigating combination treatments with immunotherapies and other modalities.224 Autophagy and apoptosis exhibit demonstrable crosstalk in T cells to regulate survival and proliferation, and components of both pathways have been shown to regulate the immune system.80,227−229 Moreover, small-molecule proapoptotic drugs have been shown to upregulate major histocompatibility complex (MHC)-I, sensitize cancer cells to T-cell-dependent killing, add to checkpoint inhibitor efficacy in vivo, and are currently being investigated in clinical trials in combination with immunotherapies.16,17 Autophagy inhibitors have been shown to regulate immune cell activation and response and also demonstrate promise as a therapeutic approach for improving cancer immunotherapy.227,230
Clinical trials have demonstrated that targeting the apoptosis pathway can be safe and offer benefit to patients with cancer. Recent clinical studies with the autophagy inhibitor HCQ have also shown safety and efficacy in cancer patients. However, both apoptosis-inducing drugs and autophagy inhibitors generally lack single-agent efficacy, and patients often acquire resistance to the therapy over time. Numerous in vitro and in vivo studies have shown enhanced efficacy at inducing cell death in cancer cells when cotreating proapoptosis drugs with autophagy inhibitors. Further, investigating the combinatorial effect of proapoptosis therapeutics and autophagy inhibitors could provide a novel approach to improve future cancer therapy and offer significant benefit to patients.
Glossary
Abbreviations
- HCQ
hydroxychloroquine
- SOC
standard of care
- Bcl-2
B-cell lymphoma 2
- IAP
inhibitor of apoptosis
- TNF
tumor necrosis factor
- TNFR
tumor necrosis factor receptor
- FASR
FS-7-associated surface antigen receptor
- TRALR
TNF-related apoptosis-inducing ligand receptor
- DISC
death-induced signaling complex
- tBID
truncated BID
- BH3
Bcl-2 homology 3
- BAX
Bcl-2-associated X-protein
- BAK
Bcl-2 homologous antagonist/killer
- MOMP
mitochondrial outer membrane permeabilization
- SMAC
second mitochondria-derived activator of caspases
- TARF
TNFR-associated factor
- APAF1
apoptotic protease-activating factor 1
- BIR
baculovirus IAP protein repeat
- PUMA
p53 upregulated modulator of apoptosis
- MLKL
mixed lineage kinase domain-like
- ULK
UNC51-like kinases
- FIP200
FAK family kinase-interacting protein of 200 kDa
- ATG
autophagy-related genes
- mTORC1
mechanistic target of rapamycin complex 1
- AMPK
AMP-activated protein kinase
- Vps34
vacuolar protein sorting 34
- LC3
microtubule-associated protein light chain 3
- PE
phosphatidylethanolamine
- ADCD
autophagy-dependent cell death
- MEF
mouse embryonic fibroblasts
- GBA1
GRB2-associated-binding protein 1
- RIPK1
receptor interacting serine/threonine kinase 1
- FADD
Fas-associating protein with death domain
- IL
interleukin
- DRAM
damage-regulated autophagy modulator
- iDISC
intracellular DISC
- c-FLIP
cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein
- FADD-like
IL-1β-converting enzyme-inhibitory protein
- NF-kB
nuclear factor kappa B
- GABARAP-L1
gamma-aminobutyric acid receptor-associated protein-like 1
- BCLAF1
Bcl-2-associated transcription factor 1
- CLL
chronic lymphocytic leukemia
- SLL
small lymphocytic lymphoma
- AML
acute myeloid leukemia
- NAF-1
nutrient-deprivation autophagy factor-1
- DAPK
death-associated protein kinase
- STK4
serine/threonine kinase 4
- MM
multiple myeloma
- MDM2
murine double minute 2
- RITA
reactivation of p53 and induction of tumor cell apoptosis
- BIR
baculovirus IAP protein repeat
- TRADD
TNFR1-associated death domain
- Apo2L
Apo2 ligand
- TRAIL
TNF-related apoptosis-inducing ligand
- scFv
single-chain fragment variable
- FOXO3a
forkhead box protein O3a
- CAR
chimeric antigen receptors
- MHC
major histocompatibility complex;
Author Contributions
This manuscript was written through contributions of all authors. All authors have given approval to the finalBcl-2 inhibitors are binding to off-target proteins version of the manuscript.
This work was supported by the Epstein Family Foundation, a Sanford Burnham Prebys National Cancer Institute Cancer Center Support Grant P30 CA030199, a Larry L. Hillblom Foundation Grant 2019-A-005-NET, and a 2019 Pancreatic Cancer Action Network Translational Research Grant 19-65-COSF to NDPC. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
The authors declare no competing financial interest.
References
- Marino G.; Niso-Santano M.; Baehrecke E. H.; Kroemer G. Self-consumption: the interplay of autophagy and apoptosis. Nat. Rev. Mol. Cell Biol. 2014, 15 (2), 81–94. 10.1038/nrm3735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glick D.; Barth S.; Macleod K. F. Autophagy: cellular and molecular mechanisms. J. Pathol 2010, 221 (1), 3–12. 10.1002/path.2697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yorimitsu T.; Klionsky D. J. Autophagy: molecular machinery for self-eating. Cell Death Differ. 2005, 12 (Suppl 2 (2)), 1542–52. 10.1038/sj.cdd.4401765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimmelman A. C.; White E. Autophagy and Tumor Metabolism. Cell Metab. 2017, 25 (5), 1037–1043. 10.1016/j.cmet.2017.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12 (6), 401–10. 10.1038/nrc3262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White E. The role for autophagy in cancer. J. Clin. Invest. 2015, 125 (1), 42–6. 10.1172/JCI73941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kroemer G. Autophagy: a druggable process that is deregulated in aging and human disease. J. Clin. Invest. 2015, 125 (1), 1–4. 10.1172/JCI78652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu R.; Ji Z.; Xu C.; Zhu J. The clinical value of using chloroquine or hydroxychloroquine as autophagy inhibitors in the treatment of cancers: A systematic review and meta-analysis. Medicine (Philadelphia, PA, U. S.) 2018, 97 (46), e12912–e12912. 10.1097/MD.0000000000012912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joyce E.; Fabre A.; Mahon N. Hydroxychloroquine cardiotoxicity presenting as a rapidly evolving biventricular cardiomyopathy: key diagnostic features and literature review. European Heart Journal: Acute Cardiovascular Care 2013, 2 (1), 77–83. 10.1177/2048872612471215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 (12), 1021–1032. 10.1016/j.tips.2018.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elmore S. Apoptosis: a review of programmed cell death. Toxicol. Pathol. 2007, 35 (4), 495–516. 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levine B.; Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008, 132 (1), 27–42. 10.1016/j.cell.2007.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ouyang L.; Shi Z.; Zhao S.; Wang F. T.; Zhou T. T.; Liu B.; Bao J. K. Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis. Cell Proliferation 2012, 45 (6), 487–98. 10.1111/j.1365-2184.2012.00845.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deeks E. D. Venetoclax: first global approval. Drugs 2016, 76 (9), 979–987. 10.1007/s40265-016-0596-x. [DOI] [PubMed] [Google Scholar]
- Sun X.-S.; Tao Y.; Le Tourneau C.; Pointreau Y.; Sire C.; Kaminsky M.-C.; Coutte A.; Alfonsi M.; Boisselier P.; Martin L.; Miroir J.; Ramee J.-F.; Delord J.-P.; Clatot F.; Rolland F.; Villa J.; Magne N.; Elicin O.; Gherga E.; Nguyen F.; Lafond C.; Bera G.; Calugaru V.; Geoffrois L.; Chauffert B.; Zubel A.; Zanna C.; Brienza S.; Crompton P.; Rouits E.; Gollmer K.; Szyldergemajn S.; Bourhis J. Debio 1143 and high-dose cisplatin chemoradiotherapy in high-risk locoregionally advanced squamous cell carcinoma of the head and neck: a double-blind, multicentre, randomised, phase 2 study. Lancet Oncol. 2020, 21 (9), 1173–1187. 10.1016/S1470-2045(20)30327-2. [DOI] [PubMed] [Google Scholar]
- Gu S. S.; Zhang W.; Wang X.; Jiang P.; Traugh N.; Li Z.; Meyer C.; Stewig B.; Xie Y.; Bu X.; Manos M. P.; Font-Tello A.; Gjini E.; Lako A.; Lim K.; Conway J.; Tewari A. K.; Zeng Z.; Das Sahu A.; Tokheim C.; Weirather J. L.; Fu J.; Zhang Y.; Kroger B.; Liang J. H.; Cejas P.; Freeman G. J.; Rodig S.; Long H. W.; Gewurz B. E.; Hodi F. S.; Brown M.; Liu X. S. Therapeutically increasing MHC-I expression potentiates immune checkpoint blockade. Cancer Discovery 2021, 11, 1524. 10.1158/2159-8290.CD-20-0812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michie J.; Kearney C. J.; Hawkins E. D.; Silke J.; Oliaro J. The Immuno-Modulatory Effects of Inhibitor of Apoptosis Protein Antagonists in Cancer Immunotherapy. Cells 2020, 9 (1), 207. 10.3390/cells9010207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Locksley R. M.; Killeen N.; Lenardo M. J. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 2001, 104 (4), 487–501. 10.1016/S0092-8674(01)00237-9. [DOI] [PubMed] [Google Scholar]
- Kischkel F. C.; Hellbardt S.; Behrmann I.; Germer M.; Pawlita M.; Krammer P. H.; Peter M. E. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 1995, 14 (22), 5579–88. 10.1002/j.1460-2075.1995.tb00245.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsu H.; Shu H.-B.; Pan M.-G.; Goeddel D. V. TRADD–TRAF2 and TRADD–FADD Interactions Define Two Distinct TNF Receptor 1 Signal Transduction Pathways. Cell 1996, 84 (2), 299–308. 10.1016/S0092-8674(00)80984-8. [DOI] [PubMed] [Google Scholar]
- Schneider P.; Thome M.; Burns K.; Bodmer J.-L.; Hofmann K.; Kataoka T.; Holler N.; Tschopp J. TRAIL Receptors 1 (DR4) and 2 (DR5) Signal FADD-Dependent Apoptosis and Activate NF-κB. Immunity 1997, 7 (6), 831–836. 10.1016/S1074-7613(00)80401-X. [DOI] [PubMed] [Google Scholar]
- Chinnaiyan A. M.; O’Rourke K.; Tewari M.; Dixit V. M. FADD, a novel death domain-containing protein, interacts with the death domain of fas and initiates apoptosis. Cell 1995, 81 (4), 505–512. 10.1016/0092-8674(95)90071-3. [DOI] [PubMed] [Google Scholar]
- Kischkel F.; Hellbardt S.; Behrmann I.; Germer M.; Pawlita M.; Krammer P. H.; Peter M. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 1995, 14 (22), 5579–5588. 10.1002/j.1460-2075.1995.tb00245.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Micheau O.; Tschopp J. Induction of TNF Receptor I-Mediated Apoptosis via Two Sequential Signaling Complexes. Cell 2003, 114 (2), 181–190. 10.1016/S0092-8674(03)00521-X. [DOI] [PubMed] [Google Scholar]
- Elmore S. Apoptosis: a review of programmed cell death. Toxicol. Pathol. 2007, 35 (4), 495–516. 10.1080/01926230701320337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C. Y.; Mayo M. W.; Korneluk R. G.; Goeddel D. V.; Baldwin A. S. Jr. NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 1998, 281 (5383), 1680–3. 10.1126/science.281.5383.1680. [DOI] [PubMed] [Google Scholar]
- Baker S. J.; Reddy E. P. Modulation of life and death by the TNF receptor superfamily. Oncogene 1998, 17 (25), 3261–3270. 10.1038/sj.onc.1202568. [DOI] [PubMed] [Google Scholar]
- Holler N.; Zaru R.; Micheau O.; Thome M.; Attinger A.; Valitutti S.; Bodmer J.-L.; Schneider P.; Seed B.; Tschopp J. Fas triggers an alternative, caspase-8–independent cell death pathway using the kinase RIP as effector molecule. Nat. Immunol. 2000, 1 (6), 489–495. 10.1038/82732. [DOI] [PubMed] [Google Scholar]
- Jiang X.; Wang X. Cytochrome C-mediated apoptosis. Annu. Rev. Biochem. 2004, 73, 87–106. 10.1146/annurev.biochem.73.011303.073706. [DOI] [PubMed] [Google Scholar]
- Du C.; Fang M.; Li Y.; Li L.; Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000, 102 (1), 33–42. 10.1016/S0092-8674(00)00008-8. [DOI] [PubMed] [Google Scholar]
- Wu G.; Chai J.; Suber T. L.; Wu J. W.; Du C.; Wang X.; Shi Y. Structural basis of IAP recognition by Smac/DIABLO. Nature 2000, 408 (6815), 1008–12. 10.1038/35050012. [DOI] [PubMed] [Google Scholar]
- Garrido C.; Galluzzi L.; Brunet M.; Puig P. E.; Didelot C.; Kroemer G. Mechanisms of cytochrome c release from mitochondria. Cell Death Differ. 2006, 13 (9), 1423–33. 10.1038/sj.cdd.4401950. [DOI] [PubMed] [Google Scholar]
- Li H.; Zhu H.; Xu C.-j.; Yuan J. Cleavage of BID by Caspase 8 Mediates the Mitochondrial Damage in the Fas Pathway of Apoptosis. Cell 1998, 94 (4), 491–501. 10.1016/S0092-8674(00)81590-1. [DOI] [PubMed] [Google Scholar]
- Hill M. M.; Adrain C.; Duriez P. J.; Creagh E. M.; Martin S. J. Analysis of the composition, assembly kinetics and activity of native Apaf-1 apoptosomes. EMBO J. 2004, 23 (10), 2134–45. 10.1038/sj.emboj.7600210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chinnaiyan A. M. The apoptosome: heart and soul of the cell death machine. Neoplasia 1999, 1 (1), 5–15. 10.1038/sj.neo.7900003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villunger A.; Michalak E. M.; Coultas L.; Mullauer F.; Bock G.; Ausserlechner M. J.; Adams J. M.; Strasser A. p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science 2003, 302 (5647), 1036–8. 10.1126/science.1090072. [DOI] [PubMed] [Google Scholar]
- Bouchier-Hayes L. The role of caspase-2 in stress-induced apoptosis. J. Cell Mol. Med. 2010, 14 (6A), 1212–24. 10.1111/j.1582-4934.2010.01037.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eckelman B. P.; Salvesen G. S.; Scott F. L. Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep. 2006, 7 (10), 988–994. 10.1038/sj.embor.7400795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alnemri E. S.; Livingston D. J.; Nicholson D. W.; Salvesen G.; Thornberry N. A.; Wong W. W.; Yuan J. Human ICE/CED-3 protease nomenclature. Cell 1996, 87 (2), 171. 10.1016/S0092-8674(00)81334-3. [DOI] [PubMed] [Google Scholar]
- Salvesen G. S.; Duckett C. S. IAP proteins: blocking the road to death’s door. Nat. Rev. Mol. Cell Biol. 2002, 3 (6), 401–410. 10.1038/nrm830. [DOI] [PubMed] [Google Scholar]
- Dohi T.; Okada K.; Xia F.; Wilford C. E.; Samuel T.; Welsh K.; Marusawa H.; Zou H.; Armstrong R.; Matsuzawa S.; Salvesen G. S.; Reed J. C.; Altieri D. C. An IAP-IAP complex inhibits apoptosis. J. Biol. Chem. 2004, 279 (33), 34087–90. 10.1074/jbc.C400236200. [DOI] [PubMed] [Google Scholar]
- Chan E. Y. mTORC1 phosphorylates the ULK1-mAtg13-FIP200 autophagy regulatory complex. Sci. Signaling 2009, 2 (84), pe51. 10.1126/scisignal.284pe51. [DOI] [PubMed] [Google Scholar]
- Egan D.; Kim J.; Shaw R. J.; Guan K.-L. The autophagy initiating kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR. Autophagy 2011, 7 (6), 643–644. 10.4161/auto.7.6.15123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takahashi Y.; Coppola D.; Matsushita N.; Cualing H. D.; Sun M.; Sato Y.; Liang C.; Jung J. U.; Cheng J. Q.; Mul J. J. Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis. Nature cell biology 2007, 9 (10), 1142–1151. 10.1038/ncb1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Funderburk S. F.; Wang Q. J.; Yue Z. The Beclin 1–VPS34 complex–at the crossroads of autophagy and beyond. Trends Cell Biol. 2010, 20 (6), 355–362. 10.1016/j.tcb.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee Y.-K.; Lee J.-A. Role of the mammalian ATG8/LC3 family in autophagy: differential and compensatory roles in the spatiotemporal regulation of autophagy. BMB Rep 2016, 49 (8), 424–430. 10.5483/BMBRep.2016.49.8.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geng J.; Klionsky D. J. The Atg8 and Atg12 ubiquitin-like conjugation systems in macroautophagy. ’Protein modifications: beyond the usual suspects’ review series. EMBO Rep. 2008, 9 (9), 859–64. 10.1038/embor.2008.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pankiv S.; Clausen T. H.; Lamark T.; Brech A.; Bruun J.-A.; Outzen H.; Øvervatn A.; Bjørkøy G.; Johansen T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 2007, 282 (33), 24131–24145. 10.1074/jbc.M702824200. [DOI] [PubMed] [Google Scholar]
- Denton D.; Kumar S. Autophagy-dependent cell death. Cell Death Differ. 2019, 26 (4), 605–616. 10.1038/s41418-018-0252-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galluzzi L.; Vitale I.; Aaronson S. A.; Abrams J. M.; Adam D.; Agostinis P.; Alnemri E. S.; Altucci L.; Amelio I.; Andrews D. W. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25 (3), 486–541. 10.1038/s41418-017-0012-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kriel J.; Loos B. The good, the bad and the autophagosome: exploring unanswered questions of autophagy-dependent cell death. Cell Death Differ. 2019, 26 (4), 640–652. 10.1038/s41418-018-0267-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denton D.; Nicolson S.; Kumar S. Cell death by autophagy: facts and apparent artefacts. Cell Death Differ. 2012, 19 (1), 87–95. 10.1038/cdd.2011.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anding A. L.; Baehrecke E. H.. Autophagy in Cell Life and Cell Death. In Current Topics in Developmental Biology; Steller H., Ed.; Academic Press: 2015; Vol. 114, Chapter Three, pp 67–91. [DOI] [PubMed] [Google Scholar]
- Denton D.; Shravage B. V.; Simin R.; Baehrecke E. H.; Kumar S. Larval midgut destruction in Drosophila: not dependent on caspases but suppressed by the loss of autophagy. Autophagy 2010, 6 (1), 163–165. 10.4161/auto.6.1.10601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denton D.; Shravage B.; Simin R.; Mills K.; Berry D. L.; Baehrecke E. H.; Kumar S. Autophagy, not apoptosis, is essential for midgut cell death in Drosophila. Curr. Biol. 2009, 19 (20), 1741–1746. 10.1016/j.cub.2009.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daish T. J.; Mills K.; Kumar S. Drosophila caspase DRONC is required for specific developmental cell death pathways and stress-induced apoptosis. Dev. Cell 2004, 7 (6), 909–915. 10.1016/j.devcel.2004.09.018. [DOI] [PubMed] [Google Scholar]
- Berry D. L.; Baehrecke E. H. Growth arrest and autophagy are required for salivary gland cell degradation in Drosophila. Cell 2007, 131 (6), 1137–48. 10.1016/j.cell.2007.10.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimizu S.; Kanaseki T.; Mizushima N.; Mizuta T.; Arakawa-Kobayashi S.; Thompson C. B.; Tsujimoto Y. Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes. Nat. Cell Biol. 2004, 6 (12), 1221–8. 10.1038/ncb1192. [DOI] [PubMed] [Google Scholar]
- Arakawa S.; Tsujioka M.; Yoshida T.; Tajima-Sakurai H.; Nishida Y.; Matsuoka Y.; Yoshino I.; Tsujimoto Y.; Shimizu S. Role of Atg5-dependent cell death in the embryonic development of Bax/Bak double-knockout mice. Cell Death Differ. 2017, 24 (9), 1598–1608. 10.1038/cdd.2017.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dasari S. K.; Bialik S.; Levin-Zaidman S.; Levin-Salomon V.; Merrill A. H. Jr.; Futerman A. H.; Kimchi A. Signalome-wide RNAi screen identifies GBA1 as a positive mediator of autophagic cell death. Cell Death Differ. 2017, 24 (7), 1288–1302. 10.1038/cdd.2017.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grandér D.; Kharaziha P.; Laane E.; Pokrovskaja K.; Panaretakis T. Autophagy as the main means of cytotoxicity by glucocorticoids in hematological malignancies. Autophagy 2009, 5 (8), 1198–1200. 10.4161/auto.5.8.10122. [DOI] [PubMed] [Google Scholar]
- Laane E.; Tamm K. P.; Buentke E.; Ito K.; Kharaziha P.; Oscarsson J.; Corcoran M.; Bjorklund A. C.; Hultenby K.; Lundin J.; Heyman M.; Soderhall S.; Mazur J.; Porwit A.; Pandolfi P. P.; Zhivotovsky B.; Panaretakis T.; Grander D. Cell death induced by dexamethasone in lymphoid leukemia is mediated through initiation of autophagy. Cell Death Differ. 2009, 16 (7), 1018–29. 10.1038/cdd.2009.46. [DOI] [PubMed] [Google Scholar]
- Liu Y.; Shoji-Kawata S.; Sumpter R. M.; Wei Y.; Ginet V.; Zhang L.; Posner B.; Tran K. A.; Green D. R.; Xavier R. J. Autosis is a Na+, K+-ATPase–regulated form of cell death triggered by autophagy-inducing peptides, starvation, and hypoxia–ischemia. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (51), 20364–20371. 10.1073/pnas.1319661110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crighton D.; Wilkinson S.; O’Prey J.; Syed N.; Smith P.; Harrison P. R.; Gasco M.; Garrone O.; Crook T.; Ryan K. M. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 2006, 126 (1), 121–134. 10.1016/j.cell.2006.05.034. [DOI] [PubMed] [Google Scholar]
- Arico S.; Petiot A.; Bauvy C.; Dubbelhuis P. F.; Meijer A. J.; Codogno P.; Ogier-Denis E. The tumor suppressor PTEN positively regulates macroautophagy by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway. J. Biol. Chem. 2001, 276 (38), 35243–35246. 10.1074/jbc.C100319200. [DOI] [PubMed] [Google Scholar]
- Hart L. S.; Cunningham J. T.; Datta T.; Dey S.; Tameire F.; Lehman S. L.; Qiu B.; Zhang H.; Cerniglia G.; Bi M.; Li Y.; Gao Y.; Liu H.; Li C.; Maity A.; Thomas-Tikhonenko A.; Perl A. E.; Koong A.; Fuchs S. Y.; Diehl J. A.; Mills I. G.; Ruggero D.; Koumenis C. ER stress–mediated autophagy promotes Myc-dependent transformation and tumor growth. J. Clin. Invest. 2012, 122 (12), 4621–4634. 10.1172/JCI62973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y.; Wang X. D.; Lapi E.; Sullivan A.; Jia W.; He Y.-W.; Ratnayaka I.; Zhong S.; Goldin R. D.; Goemans C. G.; Tolkovsky A. M.; Lu X. Autophagic activity dictates the cellular response to oncogenic RAS. Proc. Natl. Acad. Sci. U. S. A. 2012, 109 (33), 13325–13330. 10.1073/pnas.1120193109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thorburn A. Apoptosis and autophagy: regulatory connections between two supposedly different processes. Apoptosis 2008, 13 (1), 1–9. 10.1007/s10495-007-0154-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim I.; Rodriguez-Enriquez S.; Lemasters J. J. Selective degradation of mitochondria by mitophagy. Arch. Biochem. Biophys. 2007, 462 (2), 245–253. 10.1016/j.abb.2007.03.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim I.; Lemasters J. J. Mitophagy selectively degrades individual damaged mitochondria after photoirradiation. Antioxid. Redox Signaling 2011, 14 (10), 1919–1928. 10.1089/ars.2010.3768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herman-Antosiewicz A.; Johnson D. E.; Singh S. V. Sulforaphane Causes Autophagy to Inhibit Release of Cytochrome c and Apoptosis in Human Prostate Cancer Cells. Cancer Res. 2006, 66 (11), 5828–5835. 10.1158/0008-5472.Can-06-0139. [DOI] [PubMed] [Google Scholar]
- Gomes L. C.; Benedetto G. D.; Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat. Cell Biol. 2011, 13 (5), 589–598. 10.1038/ncb2220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo S.; Rubinsztein D. C. Apoptosis blocks Beclin 1-dependent autophagosome synthesis: an effect rescued by Bcl-xL. Cell Death Differ. 2010, 17 (2), 268–277. 10.1038/cdd.2009.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H.; Wang P.; Sun Q.; Ding W. X.; Yin X. M.; Sobol R. W.; Stolz D. B.; Yu J.; Zhang L. Following cytochrome c release, autophagy is inhibited during chemotherapy-induced apoptosis by caspase 8-mediated cleavage of Beclin 1. Cancer Res. 2011, 71 (10), 3625–34. 10.1158/0008-5472.CAN-10-4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oral O.; Oz-Arslan D.; Itah Z.; Naghavi A.; Deveci R.; Karacali S.; Gozuacik D. Cleavage of Atg3 protein by caspase-8 regulates autophagy during receptor-activated cell death. Apoptosis 2012, 17 (8), 810–820. 10.1007/s10495-012-0735-0. [DOI] [PubMed] [Google Scholar]
- Wirawan E.; Vande Walle L.; Kersse K.; Cornelis S.; Claerhout S.; Vanoverberghe I.; Roelandt R.; De Rycke R.; Verspurten J.; Declercq W.; Agostinis P.; Vanden Berghe T.; Lippens S.; Vandenabeele P. Caspase-mediated cleavage of Beclin-1 inactivates Beclin-1-induced autophagy and enhances apoptosis by promoting the release of proapoptotic factors from mitochondria. Cell Death Dis. 2010, 1 (1), e18–e18. 10.1038/cddis.2009.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deegan S.; Saveljeva S.; Logue S. E.; Pakos-Zebrucka K.; Gupta S.; Vandenabeele P.; Bertrand M. J. M.; Samali A. Deficiency in the mitochondrial apoptotic pathway reveals the toxic potential of autophagy under ER stress conditions. Autophagy 2014, 10 (11), 1921–1936. 10.4161/15548627.2014.981790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colell A.; Ricci J.-E.; Tait S.; Milasta S.; Maurer U.; Bouchier-Hayes L.; Fitzgerald P.; Guio-Carrion A.; Waterhouse N. J.; Li C. W.; Mari B.; Barbry P.; Newmeyer D. D.; Beere H. M.; Green D. R. GAPDH and Autophagy Preserve Survival after Apoptotic Cytochrome c Release in the Absence of Caspase Activation. Cell 2007, 129 (5), 983–997. 10.1016/j.cell.2007.03.045. [DOI] [PubMed] [Google Scholar]
- Rubinstein A. D.; Eisenstein M.; Ber Y.; Bialik S.; Kimchi A. The Autophagy Protein Atg12 Associates with Antiapoptotic Bcl-2 Family Members to Promote Mitochondrial Apoptosis. Mol. Cell 2011, 44 (5), 698–709. 10.1016/j.molcel.2011.10.014. [DOI] [PubMed] [Google Scholar]
- Bell B. D.; Leverrier S.; Weist B. M.; Newton R. H.; Arechiga A. F.; Luhrs K. A.; Morrissette N. S.; Walsh C. M. FADD and caspase-8 control the outcome of autophagic signaling in proliferating T cells. Proc. Natl. Acad. Sci. U. S. A. 2008, 105 (43), 16677–16682. 10.1073/pnas.0808597105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang S.; Okamoto K.; Yu C.; Sinicrope F. A. p62/sequestosome-1 up-regulation promotes ABT-263-induced caspase-8 aggregation/activation on the autophagosome. J. Biol. Chem. 2013, 288 (47), 33654–33666. 10.1074/jbc.M113.518134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan J.-A.; Ullman E.; Dou Z.; Zong W.-X. Inhibition of protein degradation induces apoptosis through a microtubule-associated protein 1 light chain 3-mediated activation of caspase-8 at intracellular membranes. Mol. Cell. Biol. 2011, 31 (15), 3158–3170. 10.1128/MCB.05460-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pyo J.-O.; Jang M.-H.; Kwon Y.-K.; Lee H.-J.; Jun J.-I.; Woo H.-N.; Cho D.-H.; Choi B.; Lee H.; Kim J.-H. Essential roles of Atg5 and FADD in autophagic cell death dissection of autophagic cell death into vacuole formation and cell death. J. Biol. Chem. 2005, 280 (21), 20722–20729. 10.1074/jbc.M413934200. [DOI] [PubMed] [Google Scholar]
- Tang Z.; Takahashi Y.; Chen C.; Liu Y.; He H.; Tsotakos N.; Serfass J. M.; Gebru M. T.; Chen H.; Young M. M. Atg2A/B deficiency switches cytoprotective autophagy to non-canonical caspase-8 activation and apoptosis. Cell Death Differ. 2017, 24 (12), 2127–2138. 10.1038/cdd.2017.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young M. M.; Takahashi Y.; Khan O.; Park S.; Hori T.; Yun J.; Sharma A. K.; Amin S.; Hu C.-D.; Zhang J. Autophagosomal membrane serves as platform for intracellular death-inducing signaling complex (iDISC)-mediated caspase-8 activation and apoptosis. J. Biol. Chem. 2012, 287 (15), 12455–12468. 10.1074/jbc.M111.309104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y.; Gong J.; Xing T.; Zheng S.; Ding W. Autophagy protein p62/SQSTM1 is involved in HAMLET-induced cell death by modulating apotosis in U87MG cells. Cell Death Dis. 2013, 4 (3), e550–e550. 10.1038/cddis.2013.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amir M.; Zhao E.; Fontana L.; Rosenberg H.; Tanaka K.; Gao G.; Czaja M. J. Inhibition of hepatocyte autophagy increases tumor necrosis factor-dependent liver injury by promoting caspase-8 activation. Cell Death Differ. 2013, 20 (7), 878–887. 10.1038/cdd.2013.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hou W.; Han J.; Lu C.; Goldstein L. A.; Rabinowich H. Autophagic degradation of active caspase-8: a crosstalk mechanism between autophagy and apoptosis. Autophagy 2010, 6 (7), 891–900. 10.4161/auto.6.7.13038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laussmann M. A.; Passante E.; Dussmann H.; Rauen J. A.; Wurstle M. L.; Delgado M. E.; Devocelle M.; Prehn J. H.; Rehm M. Proteasome inhibition can induce an autophagy-dependent apical activation of caspase-8. Cell Death Differ. 2011, 18 (10), 1584–97. 10.1038/cdd.2011.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yousefi S.; Perozzo R.; Schmid I.; Ziemiecki A.; Schaffner T.; Scapozza L.; Brunner T.; Simon H.-U. Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat. Cell Biol. 2006, 8 (10), 1124–1132. 10.1038/ncb1482. [DOI] [PubMed] [Google Scholar]
- Betin V. M.; Lane J. D. Caspase cleavage of Atg4D stimulates GABARAP-L1 processing and triggers mitochondrial targeting and apoptosis. J. Cell Sci. 2009, 122 (Pt 14), 2554–66. 10.1242/jcs.046250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lamy L.; Ngo V. N.; Emre N. C.; Shaffer A. L. 3rd; Yang Y.; Tian E.; Nair V.; Kruhlak M. J.; Zingone A.; Landgren O.; Staudt L. M. Control of autophagic cell death by caspase-10 in multiple myeloma. Cancer Cell 2013, 23 (4), 435–49. 10.1016/j.ccr.2013.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han J.; Hou W.; Goldstein L. A.; Stolz D. B.; Watkins S. C.; Rabinowich H. A Complex between Atg7 and Caspase-9: A NOVEL MECHANISM OF CROSS-REGULATION BETWEEN AUTOPHAGY AND APOPTOSIS. J. Biol. Chem. 2014, 289 (10), 6485–6497. 10.1074/jbc.M113.536854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu W.; Wang X.; Sun Y.; Berleth N.; Deitersen J.; Schlütermann D.; Stuhldreier F.; Wallot-Hieke N.; José Mendiburo M.; Cox J.; Peter C.; Bergmann A. K.; Stork B. TNF-induced necroptosis initiates early autophagy events via RIPK3-dependent AMPK activation, but inhibits late autophagy. Autophagy 2021, 1–18. 10.1080/15548627.2021.1899667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White E. Autophagic cell death unraveled: Pharmacological inhibition of apoptosis and autophagy enables necrosis. Autophagy 2008, 4 (4), 399–401. 10.4161/auto.5907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Y. T.; Tan H. L.; Huang Q.; Kim Y. S.; Pan N.; Ong W. Y.; Liu Z. G.; Ong C. N.; Shen H. M. Autophagy plays a protective role during zVAD-induced necrotic cell death. Autophagy 2008, 4 (4), 457–66. 10.4161/auto.5662. [DOI] [PubMed] [Google Scholar]
- Adams J. M.; Cory S. The BCL-2 arbiters of apoptosis and their growing role as cancer targets. Cell Death Differ. 2018, 25 (1), 27–36. 10.1038/cdd.2017.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalkavan H.; Green D. R. MOMP, cell suicide as a BCL-2 family business. Cell Death Differ. 2018, 25 (1), 46–55. 10.1038/cdd.2017.179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Souers A. J.; Leverson J. D.; Boghaert E. R.; Ackler S. L.; Catron N. D.; Chen J.; Dayton B. D.; Ding H.; Enschede S. H.; Fairbrother W. J. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat. Med. 2013, 19 (2), 202. 10.1038/nm.3048. [DOI] [PubMed] [Google Scholar]
- Konopleva M.; Pollyea D. A.; Potluri J.; Chyla B.; Hogdal L.; Busman T.; McKeegan E.; Salem A. H.; Zhu M.; Ricker J. L. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discovery 2016, 6 (10), 1106–1117. 10.1158/2159-8290.CD-16-0313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DiNardo C. D.; Rausch C. R.; Benton C.; Kadia T.; Jain N.; Pemmaraju N.; Daver N.; Covert W.; Marx K. R.; Mace M. Clinical experience with the BCL 2-inhibitor venetoclax in combination therapy for relapsed and refractory acute myeloid leukemia and related myeloid malignancies. Am. J. Hematol. 2018, 93 (3), 401–407. 10.1002/ajh.25000. [DOI] [PubMed] [Google Scholar]
- DiNardo C. D.; Pratz K.; Pullarkat V.; Jonas B. A.; Arellano M.; Becker P. S.; Frankfurt O.; Konopleva M.; Wei A. H.; Kantarjian H. M. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 2019, 133 (1), 7–17. 10.1182/blood-2018-08-868752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin K. H.; Winter P. S.; Xie A.; Roth C.; Martz C. A.; Stein E. M.; Anderson G. R.; Tingley J. P.; Wood K. C. Targeting MCL-1/BCL-X L forestalls the acquisition of resistance to ABT-199 in acute myeloid leukemia. Sci. Rep. 2016, 6, 27696. 10.1038/srep27696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bose P.; Gandhi V.; Konopleva M. Pathways and mechanisms of venetoclax resistance. Leuk. Lymphoma 2017, 58 (9), 2026–2039. 10.1080/10428194.2017.1283032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang R.; Zeh H.; Lotze M.; Tang D. The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ. 2011, 18 (4), 571–580. 10.1038/cdd.2010.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang X. H.; Kleeman L. K.; Jiang H. H.; Gordon G.; Goldman J. E.; Berry G.; Herman B.; Levine B. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J. Virol. 1998, 72 (11), 8586–8596. 10.1128/JVI.72.11.8586-8596.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pattingre S.; Tassa A.; Qu X.; Garuti R.; Liang X. H.; Mizushima N.; Packer M.; Schneider M. D.; Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 2005, 122 (6), 927–939. 10.1016/j.cell.2005.07.002. [DOI] [PubMed] [Google Scholar]
- Luo S.; Rubinsztein D. Apoptosis blocks Beclin 1-dependent autophagosome synthesis: an effect rescued by Bcl-xL. Cell Death Differ. 2010, 17 (2), 268–277. 10.1038/cdd.2009.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang N. C.; Nguyen M.; Germain M.; Shore G. C. Antagonism of Beclin 1-dependent autophagy by BCL-2 at the endoplasmic reticulum requires NAF-1. EMBO J. 2010, 29 (3), 606–618. 10.1038/emboj.2009.369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zalckvar E.; Berissi H.; Eisenstein M.; Kimchi A. Phosphorylation of Beclin 1 by DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL. Autophagy 2009, 5 (5), 720–722. 10.4161/auto.5.5.8625. [DOI] [PubMed] [Google Scholar]
- Maejima Y.; Kyoi S.; Zhai P.; Liu T.; Li H.; Ivessa A.; Sciarretta S.; Del Re D. P.; Zablocki D. K.; Hsu C.-P.; Lim D.-S.; Isobe M.; Sadoshima J. Mst1 inhibits autophagy by promoting the interaction between Beclin1 and Bcl-2. Nat. Med. 2013, 19 (11), 1478–1488. 10.1038/nm.3322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee E. F.; Smith N. A.; Soares da Costa T. P.; Meftahi N.; Yao S.; Harris T. J.; Tran S.; Pettikiriarachchi A.; Perugini M. A.; Keizer D. W.; Evangelista M.; Smith B. J.; Fairlie W. D. Structural insights into BCL2 pro-survival protein interactions with the key autophagy regulator BECN1 following phosphorylation by STK4/MST1. Autophagy 2019, 15 (5), 785–795. 10.1080/15548627.2018.1564557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukhopadhyay S.; Panda P. K.; Sinha N.; Das D. N.; Bhutia S. K. Autophagy and apoptosis: where do they meet?. Apoptosis 2014, 19 (4), 555–566. 10.1007/s10495-014-0967-2. [DOI] [PubMed] [Google Scholar]
- Yu L.; Liu S.. Autophagy contributes to modulating the cytotoxicities of Bcl-2 homology domain-3 mimetics; Seminars in cancer biology; Elsevier: 2013; pp 553–560. 10.1016/j.semcancer.2013.08.008. [DOI] [PubMed] [Google Scholar]
- Bodo J.; Zhao X.; Smith M. R.; Hsi E. D.. Activity of ABT-199 and acquired resistance in follicular lymphoma cells; American Society of Hematology: Washington, DC, 2014. 10.1182/blood.V124.21.3635.3635. [DOI] [Google Scholar]
- Bodo J.; Zhao X.; Durkin L.; Souers A. J.; Phillips D. C.; Smith M. R.; Hsi E. D.. Acquired resistance to venetoclax (ABT-199) in t(14;18) positive lymphoma cells. Oncotarget 2016, 7 ( (43), ), 70000. 10.18632/oncotarget.12132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen S.; Zhang Y.; Zhou L.; Leng Y.; Lin H.; Kmieciak M.; Pei X.-Y.; Jones R.; Orlowski R. Z.; Dai Y.; Grant S. A Bim-targeting strategy overcomes adaptive bortezomib resistance in myeloma through a novel link between autophagy and apoptosis. Blood 2014, 124 (17), 2687–2697. 10.1182/blood-2014-03-564534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maiuri M. C.; Criollo A.; Tasdemir E.; Vicencio J. M.; Tajeddine N.; Hickman J. A.; Geneste O.; Kroemer G. BH3-Only Proteins and BH3Mimetics Induce Autophagy by Competitively Disrupting the Interaction between Beclin 1 and Bcl-2/Bcl-XL. Autophagy 2007, 3 (4), 374–376. 10.4161/auto.4237. [DOI] [PubMed] [Google Scholar]
- Orlowski R. Z.; Dai Y.; Grant S.; Chen S.; Zhang Y.; Zhou L.; Leng Y.; Lin H.; Kmieciak M.; Pei X.-Y.. A Bim-targeting strategy overcomes adaptive bortezomib-resistance in myeloma through a novel link between autophagy and apoptosis. Blood 2014, 124, 2687–97 10.1182/blood-2014-03-564534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heidari N.; Hicks M.; Harada H. GX15–070 (obatoclax) overcomes glucocorticoid resistance in acute lymphoblastic leukemia through induction of apoptosis and autophagy. Cell Death Dis. 2010, 1 (9), e76–e76. 10.1038/cddis.2010.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCoy F.; Hurwitz J.; McTavish N.; Paul I.; Barnes C.; O’Hagan B.; Odrzywol K.; Murray J.; Longley D.; McKerr G. Obatoclax induces Atg7-dependent autophagy independent of beclin-1 and BAX/BAK. Cell Death Dis. 2010, 1 (12), e108–e108. 10.1038/cddis.2010.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan J.; Cheng C.; Verstovsek S.; Chen Q.; Jin Y.; Cao Q. The BH3-mimetic GX15–070 induces autophagy, potentiates the cytotoxicity of carboplatin and 5-fluorouracil in esophageal carcinoma cells. Cancer Lett. 2010, 293 (2), 167–174. 10.1016/j.canlet.2010.01.006. [DOI] [PubMed] [Google Scholar]
- Koehler B. C.; Jassowicz A.; Scherr A.-L.; Lorenz S.; Radhakrishnan P.; Kautz N.; Elssner C.; Weiss J.; Jaeger D.; Schneider M.; Schulze-Bergkamen H. Pan-Bcl-2 inhibitor Obatoclax is a potent late stage autophagy inhibitor in colorectal cancer cells independent of canonical autophagy signaling. BMC Cancer 2015, 15 (1), 919. 10.1186/s12885-015-1929-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Voss V.; Senft C.; Lang V.; Ronellenfitsch M. W.; Steinbach J. P.; Seifert V.; Kögel D. The pan-Bcl-2 inhibitor (−)-gossypol triggers autophagic cell death in malignant glioma. Mol. Cancer Res. 2010, 8 (7), 1002–1016. 10.1158/1541-7786.MCR-09-0562. [DOI] [PubMed] [Google Scholar]
- Gao P.; Bauvy C.; Souquère S.; Tonelli G.; Liu L.; Zhu Y.; Qiao Z.; Bakula D.; Proikas-Cezanne T.; Pierron G. The Bcl-2 homology domain 3 mimetic gossypol induces both Beclin 1-dependent and Beclin 1-independent cytoprotective autophagy in cancer cells. J. Biol. Chem. 2010, 285 (33), 25570–25581. 10.1074/jbc.M110.118125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Germain M.; Nguyen A. P.; Le Grand J. N.; Arbour N.; Vanderluit J. L.; Park D. S.; Opferman J. T.; Slack R. S. MCL-1 is a stress sensor that regulates autophagy in a developmentally regulated manner. EMBO J. 2011, 30 (2), 395–407. 10.1038/emboj.2010.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vousden K. H.; Lane D. P. p53 in health and disease. Nat. Rev. Mol. Cell Biol. 2007, 8 (4), 275–283. 10.1038/nrm2147. [DOI] [PubMed] [Google Scholar]
- Wang S.; Zhao Y.; Aguilar A.; Bernard D.; Yang C.-Y. Targeting the MDM2–p53 protein–protein interaction for new cancer therapy: progress and challenges. Cold Spring Harbor Perspect. Med. 2017, 7 (5), a026245. 10.1101/cshperspect.a026245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kocik J.; Machula M.; Wisniewska A.; Surmiak E.; Holak T. A.; Skalniak L. Helping the Released Guardian: Drug Combinations for Supporting the Anticancer Activity of HDM2 (MDM2) Antagonists. Cancers 2019, 11 (7), 1014. 10.3390/cancers11071014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vassilev L. T. Small-Molecule Antagonists of p53-MDM2 Binding: Research Tools and Potential Therapeutics. Cell Cycle 2004, 3 (4), 417–419. 10.4161/cc.3.4.801. [DOI] [PubMed] [Google Scholar]
- Feki A.; Irminger-Finger I. Mutational spectrum of p53 mutations in primary breast and ovarian tumors. Critical reviews in oncology/hematology 2004, 52 (2), 103–116. 10.1016/j.critrevonc.2004.07.002. [DOI] [PubMed] [Google Scholar]
- Maiuri M. C.; Galluzzi L.; Morselli E.; Kepp O.; Malik S. A.; Kroemer G. Autophagy regulation by p53. Curr. Opin. Cell Biol. 2010, 22 (2), 181–185. 10.1016/j.ceb.2009.12.001. [DOI] [PubMed] [Google Scholar]
- Tasdemir E.; Maiuri M. C.; Galluzzi L.; Vitale I.; Djavaheri-Mergny M.; D’Amelio M.; Criollo A.; Morselli E.; Zhu C.; Harper F.; Nannmark U.; Samara C.; Pinton P.; Vicencio J. M.; Carnuccio R.; Moll U. M.; Madeo F.; Paterlini-Brechot P.; Rizzuto R.; Szabadkai G.; Pierron G.; Blomgren K.; Tavernarakis N.; Codogno P.; Cecconi F.; Kroemer G. Regulation of autophagy by cytoplasmic p53. Nat. Cell Biol. 2008, 10 (6), 676–687. 10.1038/ncb1730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu X.; Muñoz-Alarcón A.; Ajayi A.; Webling K. E.; Steinhof A.; Langel Ü.; Ström A.-L. Inhibition of autophagy via p53-mediated disruption of ULK1 in a SCA7 polyglutamine disease model. J. Mol. Neurosci. 2013, 50 (3), 586–599. 10.1007/s12031-013-0012-x. [DOI] [PubMed] [Google Scholar]
- Morselli E.; Shen S.; Ruckenstuhl C.; Bauer M. A.; Mariño G.; Galluzzi L.; Criollo A.; Michaud M.; Maiuri M. C.; Chano T. p53 inhibits autophagy by interacting with the human ortholog of yeast Atg17, RB1CC1/FIP200. Cell cycle 2011, 10 (16), 2763–2769. 10.4161/cc.10.16.16868. [DOI] [PubMed] [Google Scholar]
- Feng Z.; Hu W.; De Stanchina E.; Teresky A. K.; Jin S.; Lowe S.; Levine A. J. The regulation of AMPK β1, TSC2, and PTEN expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways. Cancer Res. 2007, 67 (7), 3043–3053. 10.1158/0008-5472.CAN-06-4149. [DOI] [PubMed] [Google Scholar]
- Feng Z.; Zhang H.; Levine A. J.; Jin S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc. Natl. Acad. Sci. U. S. A. 2005, 102 (23), 8204–8209. 10.1073/pnas.0502857102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vassilev L. T.; Vu B. T.; Graves B.; Carvajal D.; Podlaski F.; Filipovic Z.; Kong N.; Kammlott U.; Lukacs C.; Klein C. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004, 303 (5659), 844–848. 10.1126/science.1092472. [DOI] [PubMed] [Google Scholar]
- Ding K.; Lu Y.; Nikolovska-Coleska Z.; Wang G.; Qiu S.; Shangary S.; Gao W.; Qin D.; Stuckey J.; Krajewski K. Structure-based design of spiro-oxindoles as potent, specific small-molecule inhibitors of the MDM2– p53 interaction. J. Med. Chem. 2006, 49 (12), 3432–3435. 10.1021/jm051122a. [DOI] [PubMed] [Google Scholar]
- Gu D.; Wang S.; Kuiatse I.; Wang H.; He J.; Dai Y.; Jones R. J.; Bjorklund C. C.; Yang J.; Grant S. Inhibition of the MDM2 E3 Ligase induces apoptosis and autophagy in wild-type and mutant p53 models of multiple myeloma, and acts synergistically with ABT-737. PloS One 2014, 9 (9), e103015. 10.1371/journal.pone.0103015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borthakur G.; Duvvuri S.; Ruvolo V.; Tripathi D. N.; Piya S.; Burks J.; Jacamo R.; Kojima K.; Ruvolo P.; Fueyo-Margareto J. MDM2 inhibitor, nutlin 3a, induces p53 dependent autophagy in acute leukemia by AMP kinase activation. PLoS One 2015, 10 (10), e0139254. 10.1371/journal.pone.0139254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foster B. A.; Coffey H. A.; Morin M. J.; Rastinejad F. Pharmacological rescue of mutant p53 conformation and function. Science 1999, 286 (5449), 2507–2510. 10.1126/science.286.5449.2507. [DOI] [PubMed] [Google Scholar]
- Issaeva N.; Bozko P.; Enge M.; Protopopova M.; Verhoef L. G.; Masucci M.; Pramanik A.; Selivanova G. Small molecule RITA binds to p53, blocks p53–HDM-2 interaction and activates p53 function in tumors. Nat. Med. 2004, 10 (12), 1321–1328. 10.1038/nm1146. [DOI] [PubMed] [Google Scholar]
- Fiorini C.; Menegazzi M.; Padroni C.; Dando I.; Dalla Pozza E.; Gregorelli A.; Costanzo C.; Palmieri M.; Donadelli M. Autophagy induced by p53-reactivating molecules protects pancreatic cancer cells from apoptosis. Apoptosis 2013, 18 (3), 337–346. 10.1007/s10495-012-0790-6. [DOI] [PubMed] [Google Scholar]
- Zauli G.; Celeghini C.; Melloni E.; Voltan R.; Ongari M.; Tiribelli M.; di Iasio M. G.; Lanza F.; Secchiero P. The sorafenib plus nutlin-3 combination promotes synergistic cytotoxicity in acute myeloid leukemic cells irrespectively of FLT3 and p53 status. haematologica 2012, 97 (11), 1722–1730. 10.3324/haematol.2012.062083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCormack E.; Haaland I.; Venås G.; Forthun R. B.; Huseby S.; Gausdal G.; Knappskog S.; Micklem D. R.; Lorens J. B.; Bruserud Ø.; Gjertsen B. T. Synergistic induction of p53 mediated apoptosis by valproic acid and nutlin-3 in acute myeloid leukemia. Leukemia 2012, 26 (5), 910–917. 10.1038/leu.2011.315. [DOI] [PubMed] [Google Scholar]
- Guo Y.; Li Y.; Wang F.-F.; Xiang B.; Huang X.-O.; Ma H.-B.; Gong Y.-P. The combination of Nutlin-3 and Tanshinone IIA promotes synergistic cytotoxicity in acute leukemic cells expressing wild-type p53 by co-regulating MDM2-P53 and the AKT/mTOR pathway. Int. J. Biochem. Cell Biol. 2019, 106, 8–20. 10.1016/j.biocel.2018.10.008. [DOI] [PubMed] [Google Scholar]
- Duan L.; Perez R. E.; Davaadelger B.; Dedkova E. N.; Blatter L. A.; Maki C. G. p53-regulated autophagy is controlled by glycolysis and determines cell fate. Oncotarget 2015, 6 (27), 23135. 10.18632/oncotarget.5218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huo Y.; Cai H.; Teplova I.; Bowman-Colin C.; Chen G.; Price S.; Barnard N.; Ganesan S.; Karantza V.; White E.; Xia B. Autophagy opposes p53-mediated tumor barrier to facilitate tumorigenesis in a model of PALB2-associated hereditary breast cancer. Cancer Discovery 2013, 3 (8), 894–907. 10.1158/2159-8290.CD-13-0011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo J. Y.; Teng X.; Laddha S. V.; Ma S.; Van Nostrand S. C.; Yang Y.; Khor S.; Chan C. S.; Rabinowitz J. D.; White E. Autophagy provides metabolic substrates to maintain energy charge and nucleotide pools in Ras-driven lung cancer cells. Genes Dev. 2016, 30 (15), 1704–17. 10.1101/gad.283416.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Y.; Karsli-Uzunbas G.; Poillet-Perez L.; Sawant A.; Hu Z. S.; Zhao Y.; Moore D.; Hu W.; White E. Autophagy promotes mammalian survival by suppressing oxidative stress and p53. Genes Dev. 2020, 34 (9–10), 688–700. 10.1101/gad.335570.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang X. D.; Wang Y.; Wang Y.; Zhang X.; Han R.; Wu J. C.; Liang Z. Q.; Gu Z. L.; Han F.; Fukunaga K.; Qin Z. H. p53 mediates mitochondria dysfunction-triggered autophagy activation and cell death in rat striatum. Autophagy 2009, 5 (3), 339–50. 10.4161/auto.5.3.8174. [DOI] [PubMed] [Google Scholar]
- Fan T. J.; Han L. H.; Cong R. S.; Liang J. Caspase family proteases and apoptosis. Acta biochimica et biophysica Sinica 2005, 37 (11), 719–27. 10.1111/j.1745-7270.2005.00108.x. [DOI] [PubMed] [Google Scholar]
- Reubold T. F.; Eschenburg S. A molecular view on signal transduction by the apoptosome. Cell. Signalling 2012, 24 (7), 1420–5. 10.1016/j.cellsig.2012.03.007. [DOI] [PubMed] [Google Scholar]
- Vaux D. L.; Silke J. Mammalian mitochondrial IAP binding proteins. Biochem. Biophys. Res. Commun. 2003, 304 (3), 499–504. 10.1016/S0006-291X(03)00622-3. [DOI] [PubMed] [Google Scholar]
- Deveraux Q. L.; Reed J. C. IAP family proteins--suppressors of apoptosis. Genes Dev. 1999, 13 (3), 239–52. 10.1101/gad.13.3.239. [DOI] [PubMed] [Google Scholar]
- Hong X.; Lei L.; Glas R. Tumors acquire inhibitor of apoptosis protein (IAP)-mediated apoptosis resistance through altered specificity of cytosolic proteolysis. J. Exp. Med. 2003, 197 (12), 1731–43. 10.1084/jem.20020801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holcik M.; Yeh C.; Korneluk R. G.; Chow T. Translational upregulation of X-linked inhibitor of apoptosis (XIAP) increases resistance to radiation induced cell death. Oncogene 2000, 19 (36), 4174–7. 10.1038/sj.onc.1203765. [DOI] [PubMed] [Google Scholar]
- Zhu H.; Li Y.; Liu Y.; Han B. Bivalent SMAC mimetics for treating cancer by antagonizing inhibitor of apoptosis proteins. ChemMedChem 2019, 14 (23), 1951–1962. 10.1002/cmdc.201900410. [DOI] [PubMed] [Google Scholar]
- Vamos M.; Welsh K.; Finlay D.; Lee P. S.; Mace P. D.; Snipas S. J.; Gonzalez M. L.; Ganji S. R.; Ardecky R. J.; Riedl S. J. Expedient synthesis of highly potent antagonists of inhibitor of apoptosis proteins (IAPs) with unique selectivity for ML-IAP. ACS Chem. Biol. 2013, 8 (4), 725–732. 10.1021/cb3005512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai L.; Smith D. C.; Wang S. Small-molecule SMAC mimetics as new cancer therapeutics. Pharmacol. Ther. 2014, 144 (1), 82–95. 10.1016/j.pharmthera.2014.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morrish E.; Brumatti G.; Silke J.. Future Therapeutic Directions for Smac-Mimetics. Cells 2020, 9 ( (2), ), 406. 10.3390/cells9020406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheung C. H. A.; Chang Y.-C.; Lin T.-Y.; Cheng S. M.; Leung E. Anti-apoptotic proteins in the autophagic world: an update on functions of XIAP, Survivin, and BRUCE. J. Biomed. Sci. 2020, 27 (1), 31. 10.1186/s12929-020-0627-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu W.-Y.; Kim H.; Zhang C.-L.; Meng X.-L.; Wu Z.-S. Clinical significance of autophagic protein LC3 levels and its correlation with XIAP expression in hepatocellular carcinoma. Med. Oncol. 2014, 31 (8), 108. 10.1007/s12032-014-0108-3. [DOI] [PubMed] [Google Scholar]
- Huang X.; Wu Z.; Mei Y.; Wu M. XIAP inhibits autophagy via XIAP-Mdm2-p53 signalling. EMBO J. 2013, 32 (16), 2204–2216. 10.1038/emboj.2013.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin F.; Ghislat G.; Luo S.; Renna M.; Siddiqi F.; Rubinsztein D. C. XIAP and cIAP1 amplifications induce Beclin 1-dependent autophagy through NFκB activation. Hum. Mol. Genet. 2015, 24 (10), 2899–2913. 10.1093/hmg/ddv052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Humphry N. J.; Wheatley S. P. Survivin inhibits excessive autophagy in cancer cells but does so independently of its interaction with LC3. Biol. Open 2018, 7 (10), bio037374. 10.1242/bio.037374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maskey D.; Yousefi S.; Schmid I.; Zlobec I.; Perren A.; Friis R.; Simon H.-U. ATG5 is induced by DNA-damaging agents and promotes mitotic catastrophe independent of autophagy. Nat. Commun. 2013, 4 (1), 1–14. 10.1038/ncomms3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin T.-Y.; Chan H.-H.; Chen S.-H.; Sarvagalla S.; Chen P.-S.; Coumar M. S.; Cheng S. M.; Chang Y.-C.; Lin C.-H.; Leung E. BIRC5/Survivin is a novel ATG12–ATG5 conjugate interactor and an autophagy-induced DNA damage suppressor in human cancer and mouse embryonic fibroblast cells. Autophagy 2019, 1–18. 10.1080/15548627.2019.1671643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia R.; Bonifacino J. S. Negative regulation of autophagy by UBA6-BIRC6–mediated ubiquitination of LC3. eLife 2019, 8, e50034. 10.7554/eLife.50034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ebner P.; Poetsch I.; Deszcz L.; Hoffmann T.; Zuber J.; Ikeda F. The IAP family member BRUCE regulates autophagosome–lysosome fusion. Nat. Commun. 2018, 9 (1), 1–15. 10.1038/s41467-018-02823-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Che L.; Yang X.; Ge C.; El-Amouri S. S.; Wang Q.-E.; Pan D.; Herzog T. J.; Du C. Loss of BRUCE reduces cellular energy level and induces autophagy by driving activation of the AMPK-ULK1 autophagic initiating axis. PLoS One 2019, 14 (5), e0216553. 10.1371/journal.pone.0216553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang T.-X.; Zou J.-B.; Zhu Q.-Q.; Liu C. H.; Wang G.-F.; Du T.-T.; Luo Z.-Y.; Guo F.; Zhou L.-M.; Liu J.-J. SIP/CacyBP promotes autophagy by regulating levels of BRUCE/Apollon, which stimulates LC3-I degradation. Proc. Natl. Acad. Sci. U. S. A. 2019, 116 (27), 13404–13413. 10.1073/pnas.1901039116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B.-X.; Wang H.-B.; Qiu M.-Z.; Luo Q.-Y.; Yi H.-J.; Yan X.-L.; Pan W.-T.; Yuan L.-P.; Zhang Y.-X.; Xu J.-H. Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathway. J. Exp. Clin. Cancer Res. 2018, 37 (1), 53. 10.1186/s13046-018-0703-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding R.; Wang X.; Chen W.; Li Z.; Wei A.-l.; Wang Q.-b.; Nie A.-h.; Wang L.-l. WX20120108, a novel IAP antagonist, induces tumor cell autophagy via activating ROS-FOXO pathway. Acta Pharmacol. Sin. 2019, 40 (11), 1466–1479. 10.1038/s41401-019-0253-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peiqi L.; Rong H.; Hongming D.; Zhuogang L.; Wei J.; Miao M. GDC-0152-induced autophagy promotes apoptosis in HL-60 cells. Mol. Cell. Biochem. 2018, 445 (1), 135–143. 10.1007/s11010-017-3259-7. [DOI] [PubMed] [Google Scholar]
- Gradzka S.; Thomas O. S.; Kretz O.; Haimovici A.; Vasilikos L.; Wong W. W.-L.; Häcker G.; Gentle I. E. Inhibitor of apoptosis proteins are required for effective fusion of autophagosomes with lysosomes. Cell Death Dis. 2018, 9 (5), 1–15. 10.1038/s41419-018-0508-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng S.; Chang Y.; Liu C.; Lee J.; Chan H.; Kuo C.; Lin K.; Tsai S.; Chen S.; Li C. YM155 down-regulates survivin and XIAP, modulates autophagy and induces autophagy-dependent DNA damage in breast cancer cells. Br. J. Pharmacol. 2015, 172 (1), 214–234. 10.1111/bph.12935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jane E. P.; Premkumar D. R.; Sutera P. A.; Cavaleri J. M.; Pollack I. F. Survivin inhibitor YM155 induces mitochondrial dysfunction, autophagy, DNA damage and apoptosis in Bcl-xL silenced glioma cell lines. Mol. Carcinog. 2017, 56 (4), 1251–1265. 10.1002/mc.22587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Almasan A.; Ashkenazi A. Apo2L/TRAIL: apoptosis signaling, biology, and potential for cancer therapy. Cytokine Growth Factor Rev. 2003, 14 (3–4), 337–348. 10.1016/S1359-6101(03)00029-7. [DOI] [PubMed] [Google Scholar]
- Gonzalvez F.; Ashkenazi A. New insights into apoptosis signaling by Apo2L/TRAIL. Oncogene 2010, 29 (34), 4752–4765. 10.1038/onc.2010.221. [DOI] [PubMed] [Google Scholar]
- Dubuisson A.; Micheau O. Antibodies and derivatives targeting DR4 and DR5 for cancer therapy. Antibodies 2017, 6 (4), 16. 10.3390/antib6040016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herrero-Martín G.; Høyer-Hansen M.; García-García C.; Fumarola C.; Farkas T.; López-Rivas A.; Jäättelä M. TAK1 activates AMPK-dependent cytoprotective autophagy in TRAIL-treated epithelial cells. EMBO J. 2009, 28 (6), 677–685. 10.1038/emboj.2009.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Twomey J. D.; Zhang B. Circulating tumor cells develop resistance to TRAIL-induced apoptosis through autophagic removal of death receptor 5: Evidence from an in vitro model. Cancers 2019, 11 (1), 94. 10.3390/cancers11010094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di X.; Zhang G.; Zhang Y.; Takeda K.; Rosado L. A. R.; Zhang B. Accumulation of autophagosomes in breast cancer cells induces TRAIL resistance through downregulation of surface expression of death receptors 4 and 5. Oncotarget 2013, 4 (9), 1349. 10.18632/oncotarget.1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh K.; Sharma A.; Mir M. C.; Drazba J. A.; Heston W. D.; Magi-Galluzzi C.; Hansel D.; Rubin B. P.; Klein E. A.; Almasan A. Autophagic flux determines cell death and survival in response to Apo2L/TRAIL (dulanermin). Mol. Cancer 2014, 13 (1), 1–11. 10.1186/1476-4598-13-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Onoe-Takahashi A.; Suzuki-Karasaki M.; Suzuki-Karasaki M.; Ochiai T.; Suzuki-Karasaki Y. Autophagy inhibitors regulate TRAIL sensitivity in human malignant cells by targeting the mitochondrial network and calcium dynamics. Int. J. Oncol. 2019, 54 (5), 1734–1746. 10.3892/ijo.2019.4760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mora R.; Abschuetz A.; Kees T.; Dokic I.; Joschko N.; Kleber S.; Geibig R.; Mosconi E.; Zentgraf H.; Martin-Villalba A. TNF-α-and TRAIL-resistant glioma cells undergo autophagy-dependent cell death induced by activated microglia. Glia 2009, 57 (5), 561–581. 10.1002/glia.20785. [DOI] [PubMed] [Google Scholar]
- Park K.-J.; Lee S.-H.; Kim T.-I.; Lee H.-W.; Lee C.-H.; Kim E.-H.; Jang J.-Y.; Choi K. S.; Kwon M.-H.; Kim Y.-S. A human scFv antibody against TRAIL receptor 2 induces autophagic cell death in both TRAIL-sensitive and TRAIL-resistant cancer cells. Cancer Res. 2007, 67 (15), 7327–7334. 10.1158/0008-5472.CAN-06-4766. [DOI] [PubMed] [Google Scholar]
- Tsapras P.; Nezis I. P. Caspase involvement in autophagy. Cell Death Differ. 2017, 24 (8), 1369–1379. 10.1038/cdd.2017.43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Norman J. M.; Cohen G. M.; Bampton E. T. W. The in vitro cleavage of the hAtg proteins by cell death proteases. Autophagy 2010, 6 (8), 1042–1056. 10.4161/auto.6.8.13337. [DOI] [PubMed] [Google Scholar]
- Yu L.; Alva A.; Su H.; Dutt P.; Freundt E.; Welsh S.; Baehrecke E. H.; Lenardo M. J. Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8. Science 2004, 304 (5676), 1500–2. 10.1126/science.1096645. [DOI] [PubMed] [Google Scholar]
- Pagliarini V.; Wirawan E.; Romagnoli A.; Ciccosanti F.; Lisi G.; Lippens S.; Cecconi F.; Fimia G. M.; Vandenabeele P.; Corazzari M.; Piacentini M. Proteolysis of Ambra1 during apoptosis has a role in the inhibition of the autophagic pro-survival response. Cell Death Differ. 2012, 19 (9), 1495–504. 10.1038/cdd.2012.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Callus B. A.; Vaux D. L. Caspase inhibitors: viral, cellular and chemical. Cell Death Differ. 2007, 14 (1), 73–8. 10.1038/sj.cdd.4402034. [DOI] [PubMed] [Google Scholar]
- Frank D.; Vaux D. L.; Murphy J. M.; Vince J. E.; Lindqvist L. M.. Activated MLKL attenuates autophagy following its translocation to intracellular membranes. J. Cell Sci. 2019, 132 ( (5), ), 10.1242/jcs.220996. [DOI] [PubMed] [Google Scholar]
- Cho D.-H.; Jo Y. K.; Hwang J. J.; Lee Y. M.; Roh S. A.; Kim J. C. Caspase-mediated cleavage of ATG6/Beclin-1 links apoptosis to autophagy in HeLa cells. Cancer Lett. 2009, 274 (1), 95–100. 10.1016/j.canlet.2008.09.004. [DOI] [PubMed] [Google Scholar]
- Yu L.; Wan F.; Dutta S.; Welsh S.; Liu Z.; Freundt E.; Baehrecke E. H.; Lenardo M. Autophagic programmed cell death by selective catalase degradation. Proc. Natl. Acad. Sci. U. S. A. 2006, 103 (13), 4952–7. 10.1073/pnas.0511288103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Del Gaizo Moore V.; Letai A. BH3 profiling--measuring integrated function of the mitochondrial apoptotic pathway to predict cell fate decisions. Cancer Lett. 2013, 332 (2), 202–205. 10.1016/j.canlet.2011.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buzun K.; Gornowicz A.; Lesyk R.; Bielawski K.; Bielawska A. Autophagy Modulators in Cancer Therapy. Int. J. Mol. Sci. 2021, 22 (11), 5804. 10.3390/ijms22115804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donohue E.; Tovey A.; Vogl A. W.; Arns S.; Sternberg E.; Young R. N.; Roberge M. Inhibition of Autophagosome Formation by the Benzoporphyrin Derivative Verteporfin*. J. Biol. Chem. 2011, 286 (9), 7290–7300. 10.1074/jbc.M110.139915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donohue E.; Thomas A.; Maurer N.; Manisali I.; Zeisser-Labouebe M.; Zisman N.; Anderson H. J.; Ng S. S. W.; Webb M.; Bally M.; Roberge M. The Autophagy Inhibitor Verteporfin Moderately Enhances the Antitumor Activity of Gemcitabine in a Pancreatic Ductal Adenocarcinoma Model. J. Cancer 2013, 4 (7), 585–596. 10.7150/jca.7030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gavini J.; Dommann N.; Jakob M. O.; Keogh A.; Bouchez L. C.; Karkampouna S.; Julio M. K.-d.; Medova M.; Zimmer Y.; Schläfli A. M.; Tschan M. P.; Candinas D.; Stroka D.; Banz V. Verteporfin-induced lysosomal compartment dysregulation potentiates the effect of sorafenib in hepatocellular carcinoma. Cell Death Dis. 2019, 10 (10), 749. 10.1038/s41419-019-1989-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saini H.; Sharma H.; Mukherjee S.; Chowdhury S.; Chowdhury R. Verteporfin disrupts multiple steps of autophagy and regulates p53 to sensitize osteosarcoma cells. Cancer Cell Int. 2021, 21 (1), 52. 10.1186/s12935-020-01720-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakamura M.; Kikukawa Y.; Takeya M.; Mitsuya H.; Hata H. Clarithromycin attenuates autophagy in myeloma cells. Int. J. Oncol. 2010, 37 (4), 815–20. 10.3892/ijo_00000731. [DOI] [PubMed] [Google Scholar]
- Egan D. F.; Chun M. G.; Vamos M.; Zou H.; Rong J.; Miller C. J.; Lou H. J.; Raveendra-Panickar D.; Yang C.-C.; Sheffler D. J. Small molecule inhibition of the autophagy kinase ULK1 and identification of ULK1 substrates. Mol. Cell 2015, 59 (2), 285–297. 10.1016/j.molcel.2015.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petherick K. J.; Conway O. J.; 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 (18), 11376–11383. 10.1074/jbc.C114.627778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bago R.; Malik N.; Munson M. J.; Prescott A. R.; Davies P.; Sommer E.; Shpiro N.; Ward R.; Cross D.; Ganley I. G.; Alessi D. R. 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 (3), 413–427. 10.1042/bj20140889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dyczynski M.; Yu Y.; Otrocka M.; Parpal S.; Braga T.; Henley A. B.; Zazzi H.; Lerner M.; Wennerberg K.; Viklund J. Targeting autophagy by small molecule inhibitors of vacuolar protein sorting 34 (Vps34) improves the sensitivity of breast cancer cells to Sunitinib. Cancer Lett. 2018, 435, 32–43. 10.1016/j.canlet.2018.07.028. [DOI] [PubMed] [Google Scholar]
- Ronan B.; Flamand O.; Vescovi L.; Dureuil C.; Durand L.; Fassy F.; Bachelot M.-F.; Lamberton A.; Mathieu M.; Bertrand T. A highly potent and selective Vps34 inhibitor alters vesicle trafficking and autophagy. Nat. Chem. Biol. 2014, 10 (12), 1013–1019. 10.1038/nchembio.1681. [DOI] [PubMed] [Google Scholar]
- Honda A.; Harrington E.; Cornella-Taracido I.; Furet P.; Knapp M. S.; Glick M.; Triantafellow E.; Dowdle W. E.; Wiedershain D.; Maniara W. Potent, selective, and orally bioavailable inhibitors of VPS34 provide chemical tools to modulate autophagy in vivo. ACS Med. Chem. Lett. 2016, 7 (1), 72–76. 10.1021/acsmedchemlett.5b00335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Jr A novel ATG4B antagonist inhibits autophagy and has a negative impact on osteosarcoma tumors. Autophagy 2014, 10 (11), 2021–2035. 10.4161/auto.32229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurdi A.; Cleenewerck M.; Vangestel C.; Lyssens S.; Declercq W.; Timmermans J.-P.; Stroobants S.; Augustyns K.; De Meyer G. R.; Van Der Veken P. ATG4B inhibitors with a benzotropolone core structure block autophagy and augment efficiency of chemotherapy in mice. Biochem. Pharmacol. 2017, 138, 150–162. 10.1016/j.bcp.2017.06.119. [DOI] [PubMed] [Google Scholar]
- Bowman E. J.; Siebers A.; Altendorf K. Bafilomycins: a class of inhibitors of membrane ATPases from microorganisms, animal cells, and plant cells. Proc. Natl. Acad. Sci. U. S. A. 1988, 85 (21), 7972–7976. 10.1073/pnas.85.21.7972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amaravadi R. K.; Winkler J. D. Lys05: a new lysosomal autophagy inhibitor. Autophagy 2012, 8 (9), 1383–1384. 10.4161/auto.20958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stamenkovic M.; Janjetovic K.; Paunovic V.; Ciric D.; Kravic-Stevovic T.; Trajkovic V. Comparative analysis of cell death mechanisms induced by lysosomal autophagy inhibitors. Eur. J. Pharmacol. 2019, 859, 172540. 10.1016/j.ejphar.2019.172540. [DOI] [PubMed] [Google Scholar]
- Meunier G.; Birsen R.; Cazelles C.; Belhadj M.; Cantero-Aguilar L.; Kosmider O.; Fontenay M.; Azar N.; Mayeux P.; Chapuis N.; Tamburini J.; Bouscary D. Antileukemic activity of the VPS34-IN1 inhibitor in acute myeloid leukemia. Oncogenesis 2020, 9 (10), 94. 10.1038/s41389-020-00278-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young C. D.; Arteaga C. L.; Cook R. S. Dual inhibition of Type I and Type III PI3 kinases increases tumor cell apoptosis in HER2+ breast cancers. Breast Cancer Res. 2015, 17 (1), 1–11. 10.1186/s13058-015-0656-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan N.; Song L.; Zhang S.; Lin W.; Cao Y.; Xu F.; Fang Y.; Wang Z.; Zhang H.; Li X. Bafilomycin A1 targets both autophagy and apoptosis pathways in pediatric B-cell acute lymphoblastic leukemia. Haematologica 2015, 100 (3), 345. 10.3324/haematol.2014.113324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thorburn J.; Andrysik Z.; Staskiewicz L.; Gump J.; Maycotte P.; Oberst A.; Green D. R.; Espinosa J. M.; Thorburn A. Autophagy controls the kinetics and extent of mitochondrial apoptosis by regulating PUMA levels. Cell Rep. 2014, 7 (1), 45–52. 10.1016/j.celrep.2014.02.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levy J. M. M.; Thorburn A. Autophagy in cancer: Moving from understanding mechanism to improving therapy responses in patients. Cell Death Differ. 2020, 27, 843. 10.1038/s41418-019-0474-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aita V. M.; Liang X. H.; Murty V. V.; Pincus D. L.; Yu W.; Cayanis E.; Kalachikov S.; Gilliam T. C.; Levine B. Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics 1999, 59 (1), 59–65. 10.1006/geno.1999.5851. [DOI] [PubMed] [Google Scholar]
- Moscat J.; Diaz-Meco M. T. p62 at the crossroads of autophagy, apoptosis, and cancer. Cell 2009, 137 (6), 1001–4. 10.1016/j.cell.2009.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mathew R.; Karp C. M.; Beaudoin B.; Vuong N.; Chen G.; Chen H. Y.; Bray K.; Reddy A.; Bhanot G.; Gelinas C.; Dipaola R. S.; Karantza-Wadsworth V.; White E. Autophagy suppresses tumorigenesis through elimination of p62. Cell 2009, 137 (6), 1062–75. 10.1016/j.cell.2009.03.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barbari C.; Fontaine T.; Parajuli P.; Lamichhane N.; Jakubski S.; Lamichhane P.; Deshmukh R. R. Immunotherapies and Combination Strategies for Immuno-Oncology. Int. J. Mol. Sci. 2020, 21 (14), 5009. 10.3390/ijms21145009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mellman I.; Coukos G.; Dranoff G. Cancer immunotherapy comes of age. Nature 2011, 480 (7378), 480–9. 10.1038/nature10673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Topalian S. L.; Hodi F. S.; Brahmer J. R.; Gettinger S. N.; Smith D. C.; McDermott D. F.; Powderly J. D.; Carvajal R. D.; Sosman J. A.; Atkins M. B.; Leming P. D.; Spigel D. R.; Antonia S. J.; Horn L.; Drake C. G.; Pardoll D. M.; Chen L.; Sharfman W. H.; Anders R. A.; Taube J. M.; McMiller T. L.; Xu H.; Korman A. J.; Jure-Kunkel M.; Agrawal S.; McDonald D.; Kollia G. D.; Gupta A.; Wigginton J. M.; Sznol M. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366 (26), 2443–54. 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dengjel J.; Schoor O.; Fischer R.; Reich M.; Kraus M.; Muller M.; Kreymborg K.; Altenberend F.; Brandenburg J.; Kalbacher H.; Brock R.; Driessen C.; Rammensee H. G.; Stevanovic S. Autophagy promotes MHC class II presentation of peptides from intracellular source proteins. Proc. Natl. Acad. Sci. U. S. A. 2005, 102 (22), 7922–7. 10.1073/pnas.0501190102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pua H. H.; Dzhagalov I.; Chuck M.; Mizushima N.; He Y. W. A critical role for the autophagy gene Atg5 in T cell survival and proliferation. J. Exp. Med. 2007, 204 (1), 25–31. 10.1084/jem.20061303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walsh C. M.; Edinger A. L. The complex interplay between autophagy, apoptosis, and necrotic signals promotes T-cell homeostasis. Immunol. Rev. 2010, 236 (1), 95–109. 10.1111/j.1600-065X.2010.00919.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishimwe N.; Zhang W.; Qian J.; Zhang Y.; Wen L. Autophagy regulation as a promising approach for improving cancer immunotherapy. Cancer Lett. 2020, 475, 34–42. 10.1016/j.canlet.2020.01.034. [DOI] [PubMed] [Google Scholar]