Next Article in Journal
TMEM43 Protects against Sepsis-Induced Cardiac Injury via Inhibiting Ferroptosis in Mice
Next Article in Special Issue
High Fat-High Fructose Diet Elicits Hypogonadotropism Culminating in Autophagy-Mediated Defective Differentiation of Ovarian Follicles
Previous Article in Journal
The Effect of Germanium-Loaded Hydroxyapatite Biomaterials on Bone Marrow Mesenchymal Stem Cells Growth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Autophagy in Cancer Immunotherapy

1
Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518032, China
2
Bao′an Authentic TCM Therapy Hospital, Shenzhen 518126, China
3
School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
4
Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(19), 2996; https://doi.org/10.3390/cells11192996
Submission received: 16 August 2022 / Revised: 19 September 2022 / Accepted: 23 September 2022 / Published: 26 September 2022
(This article belongs to the Special Issue Molecular Targets for Autophagy in Cancer Treatment)

Abstract

:
Autophagy is a stress-induced process that eliminates damaged organelles and dysfunctional cargos in cytoplasm, including unfolded proteins. Autophagy is involved in constructing the immunosuppressive microenvironment during tumor initiation and progression. It appears to be one of the most common processes involved in cancer immunotherapy, playing bidirectional roles in immunotherapy. Accumulating evidence suggests that inducing or inhibiting autophagy contributes to immunotherapy efficacy. Hence, exploring autophagy targets and their modifiers to control autophagy in the tumor microenvironment is an emerging strategy to facilitate cancer immunotherapy. This review summarizes recent studies on the role of autophagy in cancer immunotherapy, as well as the molecular targets of autophagy that could wake up the immune response in the tumor microenvironment, aiming to shed light on its immense potential as a therapeutic target to improve immunotherapy.

1. Introduction

The rise of tumor immunotherapy has brought new hope for humankind to conquer cancer completely. Immune checkpoint blockade (ICB) therapy, with PD-1/PD-L1 blockade therapy as the most typical representative, has become one of the most popular immunotherapies today, and has significantly changed the current cancer treatment landscape [1]. However, many patients do not respond to these therapies for multiple reasons [2]. Therefore, further insight into the molecular mechanisms underlying the resistance of cancer cells to T-cell killing is needed. As an evolutionarily conserved process, autophagy captures and degrades dysfunctional cellular components under the circumstance of various cellular stresses, including hypoxia and nutrient deprivation [3]. Tumor cells always maintain a higher level of basal autophagy than normal cells. Autophagy plays a significant role in tumor cell survival [4]. Accumulating evidence has demonstrated that the autophagic pathway is also involved in the survival and apoptosis of immune cell subpopulations that affect the differentiation, activation, effector function, and translocation to tumors. At the same time, autophagy affects tumor growth by modulating immune responses. In addition, the pro-tumor effects of autophagy can be eliminated by a combination treatment of ICB and autophagy inhibitors [5]. Thus, autophagy is a complex but promising target in cancer immunotherapy. Understanding the mechanisms of autophagy in immunotherapy may prevent tumor recurrence and benefit patients from immunotherapy.

2. The Induction and Regulation of Autophagy

In response to cellular starvation or stresses, autophagy is activated to degrade unfolded proteins, damaged organelles, or intracellular pathogens in lysosomes. The digested products are recycled to support metabolic adaptation and maintain energy homeostasis [6]. Macro-autophagy is referred to as autophagy, and its mechanism has been extensively studied. Autophagy is a complex multistep process including induction, vesicle nucleation and elongation, docking and fusion, and degradation and recycling [7]. Each step of autophagy is tightly regulated by conserved autophagy-related genes (ATG) or other genes in the endosomal/lysosomal system [8]. The initiation of autophagic vesicle formation is followed by Unc-51-like kinase (ULK) complex activation. The ULK complex, consisting of ULK family kinase, Atg13, and focal adhesion kinase interacting protein 200 kDa (FIP200), is inhibited by the master cell growth regulator, the target of rapamycin (mTOR), and activated by a major sensor of energy stress, AMP-activated protein kinase (AMPK) [9]. At the stage of initiation, the ULK complex localizes to the phagophore and phosphorylates the Beclin 1-VPS34 complex composed of Beclin 1, Atg14, AMBRA1, VPS34, and VPS15 [10]. The VPS34 lipid kinase converts endoplasmic reticulum (ER) and Golgi complex-derived phosphoinositides to phosphatidylinositol-3-phosphate (PI3P) [11]. The PI3P is then bound with WIPI2B to recruit two protein conjugation systems, Atg7-Atg3 and the Atg5-Atg12-Atg6L1 complex [12]. These two complexes function to conjugate lipidated microtubule-associated protein 1A/1B light chain 3 (LC3) family members to phosphatidylethanolamine (PE) [13]. Atg4 is necessary for transforming pro-LC3 into the active cytosolic isoform LC3-I [14]. LC3-I is then conjugated to PE to produce LC3-II, the lipidated form of LC3. The insertion of LC3-II on the surface of the emerging AV is crucial for specific substrate recognition in the selective degradation process [15]. Additionally, an increasing number of autophagy cargo receptors have been found to use LC3 as a docking site to deliver autophagic cargo to the AVs. It is reported that cargo receptors, including SQSTM1 (p62) and neighbor of BRCA1 (NBR1), directly bind to marked autophagic cargo for degradation through NIX and FAM134B [16]. Curvature and sealing of the AV are facilitated by the cooperation of the LC3-PE system and the Atg5-Atg12 system [17]. AV maturation needs Atg9 to recruit the lipid membrane, which is originated from mitochondria, ER, or Golgi, to close the vesicle with trapped cargo [18]. Subsequently, the AVs fuse with lysosomes to create AV-lysosomes where the cargo loaded by the LC3 binding proteins is degraded by lysosomal enzymes. Then, amino acids or other macromolecules are recycled to support cell growth [19]. This step is mediated by Rab GTPases, SNARE, and HOPS complex [20].

3. The Role of Autophagy in Cancer Progression

The main function of autophagy is to deal with a range of intracellular and extracellular stresses through recycling metabolites [21]. Multiple stresses induce autophagy in cancer cells, such as metabolic stress, hypoxia, oxidative stress, and immune signals. In response to nutrient deprivation, autophagy is activated to rewire metabolic pathways under the regulation of AMPK and mTORC1 [22]. AMPK is an energy sensor, whereas mTORC1 is a nutrient sensor. AMPK activation or mTORC1 inhibition results in the phosphorylation of targets in the autophagy pre-initiation complex [9]. Moreover, AMPK may inhibit mTORC1 activity by targeting Raptor, a subunit of mTORC1 [23]. Hypoxia was confirmed to be another factor triggering autophagy since the inhibition of oxidative phosphorylation in mitochondria by hypoxia contributes to the activation of AMPK [24]. Additionally, hypoxia suppresses mTOR signaling to increase the stabilization of hypoxia-induced factor 1α (HIF1α) [25]. HIF1α is a transcription factor that upregulates autophagy promoting genes [26]. Furthermore, hypoxia activates activating transcription factor 4 (ATF4) to maintain high levels of autophagic flux by upregulating LC3B and Atg5 [27]. Accumulating reactive oxygen species (ROS) is an important characteristic of oxidative stress in cancer cells, which facilitates DNA damage and tumor progression [28]. The elevated ROS induces autophagy through mTORC1 suppression or nuclear factor-κB (NF-κB)-mediated p62/SQSTM1 upregulation [29,30].
The role of autophagy in cancer progression has been well established recently. It is widely accepted that autophagy is an endogenous defense and nutrient-scavenging mechanism in response to cellular stresses. Recent findings indicated that selective forms of autophagy represent a quality-control mechanism by regulating some specific organelles and proteins [31]. In the dynamics of tumor development, autophagy plays a tumor-suppressive or tumor-promoting role depending on type, stage, and genetic context of the cancers. The dual effects of autophagy in tumor progression are closely correlated to microenvironment stress and the conditions of immune system [32]. In the earliest stages of tumorigenesis, autonomous autophagy is a protective mechanism to limit tumor initiation by eliminating damaged proteins and organelles that are toxic to cells [33]. Hence, mutation or deletion of ATGs contributes to tumor initiation. For example, a mouse model with BECN1 deletion had a higher percentage of spontaneous tumor formation compared with BECN1 wild-type mouse [34]. However, autophagy is upregulated in established cancers to cope with cellular stresses such as hypoxia and nutrient deprivation, thus supporting cancer growth, survival, and malignancy to mediate cancer development, drug resistance, and metastasis [8]. Moreover, the autophagic level in aggressive tumors is higher than non-aggressive tumors. At the initial stage of metastasis, autophagy may play a suppressive role by preventing tumor necrosis and restricting inflammatory cell infiltration [35]. However, autophagy may act as a promoter during cancer metastasis by accelerating dissemination of cancer cells and enhancing their colonization in the destination organs [36,37]. The dual effects of autophagy in suppression or promotion of cancer remains controversial and has not been fully elucidated [38].

4. Autophagy in Tumor Immune System

Autophagy in cancer cells often occurs in response to cytokines and damage-associated molecular patterns (DAMPs) derived from the tumor microenvironment (TME). Several pattern recognition receptors have been verified as autophagy inducers when they receive extracellular DAMPs, including ATP, DNA complexes, and high-mobility group box 1 protein (HMGB1). [39]. For example, polyinosinic:polycytidylic acid and lipopolysaccharide (LPS) activate Toll-like receptor 3 (TLR3) and TLR4, thus triggering autophagy in non-small cell lung cancer (NSCLC) cells [40]. Another study demonstrated that advanced glycosylation end product-specific receptor (AGER) is crucial for HMGB1-induced autophagy in pancreatic and colon cancer cells [41]. As for cytokines, they participate in autophagy initiation in a context-dependent way. Accumulated evidence suggests that transforming growth factor-β (TGF-β) and interferons (type I and II) induce autophagy in various kinds of cancer cells [42]. For instance, TGF-β activates autophagy in human hepatocellular carcinoma cells by upregulating the levels of Beclin 1, Atg5 and Atg7 [43]. In addition, tumor necrosis factor (TNF) and interleukin-6 (IL-6) were found to be autophagy inducers that are associated with carcinogenesis and tumor progression [44]. As a major mechanism of autophagy-associated tumor progression, the immunosuppressive microenvironment shaped by autophagy is our focus. The immune system is essential for preventing the development and metastasis of tumors and for shaping the tumor response to treatment. The immune system recognizes and eliminates tumor cells by immune surveillance. However, some tumor cells survive because of immune evasion, a process where the immunogenicity of tumor cells is reduced, and the immunosuppressive networks are formed [45]. In TME, changes in the autophagy pathway are observed in cancer cells and immune cells, which shape tumor immunity and affect immunotherapy [17].
The role of autophagy in immune system activation and immunosuppressive TME formation is complex. On the one hand, autophagy may participate in the process of antigen presentation by APCs through proteasome degradation and the vacuolar pathway [46]. Autophagic degradation can promote APC efficiency by accelerating antigen processing [47]. Autophagy deficiency in pancreatic ductal adenocarcinoma stimulates PD-L1 expression, which may aid in the formation of immunosuppressive TME [48]. Similarly, autophagy inhibition can upregulate PD-L1 expression by accumulating p62/SQSTM1 and activating NF-κB in gastric cancer [49] (Figure 1a). Autophagy contributes to immune evasion by suppressing the innate and adaptive immune response. The inducers of the innate immune response, such as damaged proteins, organelles, and bacteria, are cleared by autophagy. DAMPs and pathogen-associated molecular patterns (PAMPs) are both inducers of innate immunity, which can be captured and degraded in autolysosomes [50]. These facts indicate that autophagy seems to be a key negative regulator of innate immune responses when activated [51]. As a consequence, inhibition of autophagy facilitates the production and secretion of proinflammatory cytokines such as IFNs (type I, II) and TNF-α in vitro and in vivo [52]. Inhibition of autophagy can also trigger programmed cell death of cancer cells, producing DAMPs to activate the adaptive immune system [53]. Moreover, autophagy can suppress the adaptive immune response by weakening T cells’ ability to kill tumor cells [54]. It has been shown that antigenic tumors are recognized and eliminated by T cells when autophagy is inhibited in mice [55]. There are several mechanisms by which autophagy restrains the antitumor T cell response. Autophagy suppresses the functions of NK cells, CD4+ T cells, and CD8+ T cells to help tumor cells combat immunosurveillance, acting as a protective mechanism in tumors [56]. For example, hypoxia-induced autophagy can impair NK cell-mediated killing of breast cancer cells by degrading NK-derived granzyme B in autophagosomes [57] (Figure 1b). Hypoxia-induced autophagy is also reported to attenuate cancer cell sensitivity to CTL by regulating the STAT3 pathway in IGR-Heu lung carcinoma cells [58]. Hence, autophagy in tumors may be a potential target to block immune evasion in the TME. Meanwhile, the initiation of immune response often needs a major histocompatibility complex (MHC) to present antigens to T cells. MHC includes class I and II, that are recognized by CD4+ and CD8+ T cells, respectively [59]. MHC-I presents peptides that prime and activate CD8+ CTLs. Then, CTLs clear the targeted tumor cells by detecting a matching antigen by the MHC-I [60]. The expression of MHC-I is downregulated in pancreatic cancer, resulting in defective antigen presentation, which limits the ability of tumor killing by T cells and immunotherapy. Mechanistically, MHC-I is recognized by NBR1 and degraded in lysosomes through autophagy [55] (Figure 1c). Additionally, in dendritic cells (DCs), autophagy has been shown to accelerate the internalization and degradation of MHC-I. DCs with autophagy inhibition enhance the presentation of viral antigens to CD8+ T cells [61,62]. Recently, hepatic autophagy immune tolerance (HAIT) has been studies and is regarded as another immune evasion mechanism. A study by Laura et al. revealed that autophagy in the liver induces tumor immune tolerance by promoting Treg function and inhibiting T-cell response and interferon-γ production, resulting in the growth of tumors with high tumor mutational burden (TMB). Therefore, autophagy may be a potential target to overcome HAIT [52].

4.1. Autophagy in Immune Cells

Tumor development and therapeutic response are influenced by tumor-infiltrating immune cells in TME [63]. A growing number of studies have revealed that autophagy is involved in the differentiation, homeostasis, and development of immune cells [64]. Hence, the alterations of autophagy pathway in immune cells may shape their phenotypes and functions in the TME [65].

4.1.1. Autophagy in T cells

Autophagy plays a vital role in the survival and proliferation of T cells. In TME of tumor-bearing mice and cancer patients, T cells undergo apoptosis. Xia et al. found that tumor-infiltrating T cells exhibit defective autophagy and a decreased level of FIP200, which is necessary for autophagosome formation. This process is mediated by tumor-derived lactate, which lowers FIP200 expression and causes T cell death by disrupting the balance between pro- and anti-apoptotic factors in the Bcl-2 family to boost immune evasion of cancer [66] (Figure 2). The interactions of T cell receptor (TCR) with stromal cells and IL-7 signaling are essential for naive T cells to survive in the periphery, and it appears that Atg3-dependent autophagy is intrinsically required for these processes [67]. Atg7-deficient T cells have poor proliferative capacity and cannot enter the S phase when TCR is activated. Mechanistically, this is attributed to the accumulation of CDKN1B, which cannot be degraded in autophagy-deficient naive T cells, and negatively regulates cell cycle [68]. Defective autophagy can also disturb T cell activation, differentiation, and stemness. It has been reported that the differentiation from T cells to invariant natural killer T (iNKT) and Treg is facilitated by autophagy [69]. Yang et al. found that deletion of autophagy-related protein PIK3C3/VPS34 reduced the activity of mitochondria upon T cell activation, so that CD4+ T cells failed to differentiate into T helper 1 cells [70]. Similarly, the quality of mitochondria declines when the mitochondrial components cannot be degraded adequately, resulting in ROS accumulation and T cell damage [71]. Depolarized mitochondria were also detected in IL-15-induced resident memory CD8+ T cells with autophagy inhibited by MRT68921 dihydrochloride and 3-methyladenine (3MA), causing T cells exhaustion [72]. CD4+ Treg cells in TME confer another primary mechanism of immune evasion and immunotherapy resistance. Autophagy was shown to participate in maintaining Treg cell function [73]. Inhibition of autophagy by Atg5, Atg7 and AMBRA1 deletion induces Treg cell apoptosis and dysfunction in vivo [74,75,76] (Figure 2).
Autophagy also serves a crucial function in other types of immune cells that interact with T cells. Lysosomal proteolysis of autophagy is necessary for the antigen presentation in dendritic cells [77]. Deleting PIK3C3 (a key player in autophagy) in dendritic cells contributes to reduced CD8α+ dendritic cells and B16 melanoma-specific CTL activity [62]. Myeloid-derived suppressor cells (MDSCs) dampen antitumor immune responses and treatment effectiveness by directly suppressing T cell activation in TME [78]. An elevated level of autophagy has been detected in MDSCs [79]. Blocking autophagy in MDSCs leads to increased MHC-II expression and impaired tumor growth in vivo [80].

4.1.2. Autophagy in B cells

The survival and development of B cells depends on autophagy. For instance, the development of B cells depends on Atg5 [81]. After ligand LPS activation, basal levels of autophagy are required to preserve the survival of a normal number of peripheral B cells [82]. Intrinsic autophagy in B cells is necessary to sustain the normal function of alloreactive B memory cells. Atg7 deletion in B cell blocks B cell autophagy, inhibiting secondary alloantibody responses and decreasing the frequencies of alloreactive B memory cells [83].

4.1.3. Autophagy in Natural Killer (NK) Cells

The survival of iNKT cells requires intrinsic autophagy. Facing viral infection, autophagy is activated with the aid of phosphorylated FoxO1 and Atg7, which promotes the development and function of NKT cells against viral infection [84]. Autophagy-deficient iNKT cells undergo apoptosis in the mitochondrial pathway. Absence of autophagy not only hinders the maturation of iNKT cells by reducing NKT cell proliferation, but also limits the transition of iNKT cells to a quiescent state [85]. Moreover, the secretion of iNKT can be affected by autophagy. Autophagy-deficient iNKT cells release low levels of IL-4 and IFN-γ compared with normal iNKT cells [86].

4.1.4. Autophagy in Dendritic Cells (DCs)

Autophagy is indispensable for cytokine secretion and antigen presentation in dendritic cells (DCs). It has been reported that Atg5 is essential for DCs to activate CD4+ T cells through antigen presentation [84,85]. Atg5 deficiency in DCs reduces the secretion of IL-2 and IFN-γ by CD4+ T cells, but the levels of IL-12, IL-6, and TNF-α are not affected [87]. Moreover, antigen presentation was hampered by Atg5 deficiency through the MHC-II pathway, which may be resulted from the delay of lysosome-phagosome fusion [84,85,86,87,88,89].

4.1.5. Autophagy in Macrophages

The generation of macrophages requires autophagy at different stages. It is well accepted that tumor-associated macrophages (TAMs) originate from monocytes. The recruitment of monocytes to tumors requires chemokines and cytokines derived from tumor cells and stromal cells. One of the chemokines, [C-C motif] ligand 2 (CCL2), protects monocytes from apoptosis and induces a high level of autophagy in TAMs [90]. The transition from monocytes into macrophages requires colony-stimulating factor 1 (CSF1), which upregulates and activates ULK1 to trigger autophagy [91]. CSF2 also facilitates the differentiation of monocytes. Autophagy in monocytes maintains a high-level during differentiation, since JNK signaling is activated to release Beclin 1 and inhibits Atg5 cleavage. Autophagy inhibition restrains the differentiation process [92]. In addition, autophagy is involved in macrophage polarization. Downregulated autophagy promotes M1 polarization and upregulated autophagy promotes M2 polarization in macrophages [93]. M1 macrophages stimulate immune responses, whereas M2 macrophages play an immunosuppressive role, indicating that autophagy is a potential target to modulate macrophage polarization toward the M1 phenotype [94].

4.2. Autophagy in Regulating Immune Checkpoint Molecules

Immune checkpoint molecules, including PD-1 and CTLA-4, play crucial roles in tumor immune tolerance, which is the main reason for the poor clinical outcomes of immunotherapy. These immune checkpoint molecules regulate tumor immune tolerance through autophagy. For example, activation of PD-1 by PD-L1 promotes immunologic tolerance, suppresses effector T cells and maintains tumor Tregs, boosting tumor survival [95]. Hence, elucidating the regulation of immune checkpoints by autophagy is of great significance in immunotherapy.
As a T cell checkpoint molecule, PD-1 inhibits T cell proliferation and impedes the recognition of tumor cells once activated by PD-L1. Clark et al. found that tumor cells initiate autophagy in response to anti-PD1 or anti-PD-L1 antibody treatment. Engagement of PD-1 to PD-L1 induces autophagy in nearby T cells [96]. A study indicated that inhibition of autophagy by the Sigma1 inhibitor leads to degradation of PD-L1 and impaired PD-1/PD-L1 interaction, suggesting the Sigma1 inhibitor as a promising tool to block PD-1/PD-L1 [97]. Therefore, combining antibodies with autophagy inhibitors may be an attractive therapeutic strategy [98].
CTLA-4 was confirmed to be another immune checkpoint molecule that mediates tumor immune tolerance. Shukla et al. identified a subcluster of MAGE-A cancer-germline antigens that confer resistance to the CTLA-4 blockade. Autophagy activation combats this resistance by decreasing MAGE-A protein levels in human melanomas [99]. However, another study reported that mTORC1 inhibitor-induced autophagy restores CTLA-4 expression and corrects Treg cell function in systemic lupus erythematosus (SLE) [100]. CTLA4 engagement inhibits autophagy by activating PI3K/Akt/mTOR signaling pathway [101].
IDO promotes immunologic tolerance by suppressing responses of CTLs and maturation of DCs, magnifying tolerogenic APCs, and inducing Tregs generation. Autophagy can prevent inflammation-induced IDO synthesis by reducing inflammation [102]. IDO triggers the activation of general control nonderepressible 2 (GCN2) and inhibition of eukaryotic translation initiation factor 2α (eIF2α), which participate in inflammatory carcinogenesis [103]. Autophagy induced by IDO1-GCN2 plays a protective role against fatal inflammation disease [104]. Another mechanism of IDO-induced autophagy has been reported. The inhibition of tryptophan sufficiency signaling by IDO causes mTOR inactivation, thus triggering autophagy in an LC3-dependent way [103].

4.3. Autophagy in Immune Cytokines

Autophagy can increase or inhibit cytokine production and affect tumor progression. Autophagy and cytokines can regulate each other. Numerous cytokines have been demonstrated to govern or be governed by autophagy.

4.3.1. Interleukins

IL-1 is a class of pro-inflammatory cytokines that promotes cancer progression by inhibiting COX-1, IkB, and JNK signaling pathways. Inhibition of IL-1 in tumor cells restrains tumor development [105]. IL-1β is negatively regulated by autophagy in most situations, whereas IL-1α is only negatively regulated by autophagy [106]. However, both IL-1α and IL-1β triggers autophagy, indicating that autophagy is a negative feedback mechanism in regulating IL-1 [107]. Autophagy activation in Atg5-deficient macrophages causes decreased IL-1β secretion, suppressing T cell activation and cytokine production [87]. Conversely, IL-1 level is elevated in autophagy-deficient macrophages to promote tumorigenesis through ROS-NF-κB signaling pathway [108]. IL-2 induces autophagy in an ATG5, HMGB1, and Beclin1-dependent way [109]. High-dose interleukin-2 (HDIL-2) inhibits the growth of metastatic liver tumors in vivo, accompanied by elevated levels of HMGB1, IFN-γ, IL-6, and IL-18 in serum. HMGB1 is translocated from the nucleus to the cytosol to trigger autophagy, and suppression of autophagy by CQ may enhance the proliferation and infiltration of immune cells in the liver and spleen [110]. IL-6 negatively regulates autophagy by activating STAT3 and inhibiting LC3-II and Beclin 1 expression in vitro [111]. However, IL-6 trans-signaling stimulates autophagy when IL-6 interacts with soluble IL-6R in vivo, which is mediated by AMPK and AKT activation [112]. In addition, IL-10 is also an autophagy suppressor by activating the JAK/STAT3 and PI3K/Akt/mTORC1 pathway [113]. As Th2 cytokines, IL-4, IL-13, and IL-10 can activate the PI3K/mTORC1 pathway to inhibit autophagy in most environments [114]. In turn, autophagy may exert dual effects on the production of IL-10, which needs further investigation [115].

4.3.2. Interferons

Autophagy is needed in the synthesis of IFN-γ. IFN can be categorized into type I, II, and III. Type I IFN directly induces autophagy by activating JAK/STAT pathway [116]. IFN-α is a class of type I IFN, and the effect of IFN-α varies on different cell types [117]. As a type II IFN, IFN-γ induces autophagy in various types of immune cells and tumor cells, which is mediated by the acceleration of autophagosome formation and maturation [118]. In addition, IFN-γ can facilitate the MHCI expression and induce autophagy [119]. It has been reported that effector CD4+ T cells lacking Atg7 express low amounts of IFN-γ in the absence of autophagy [120].

4.3.3. Transforming Growth Factor Beta (TGF-β)

TGF-β contributes to immune evasion by directly suppressing effector cells and indirectly facilitating the differentiation of Treg cells. TGF-β can also inhibit NK cells and IFN-γ to form immunosuppressive TME [121]. Inhibition of autophagy upregulates TGF-β expression by impairing its degradation [122]. In turn, TGF-β is responsible for autophagy induction in cancer cells [117]. For example, TGF-β induces autophagy in HCC and breast cancer cells by increasing the levels of ATGs, including Beclin1, Atg5, and Atg7. Autophagy promotes the expression of proapoptotic genes such as Bim and Bmf in the Bcl-2 family to mediate apoptosis, suggesting the correlations between autophagy and apoptosis [123,124].

4.3.4. Tumor Necrosis Factor Alpha (TNF-α)

TNF-α is an apoptosis and necrosis inducer in multiple cells and can impair autophagy by decreasing lysosomal acidification [125]. Inhibition of autophagy by Bafilomycin A1 contributes to TNF-α-induced cell death by increasing oxidative stress and toxicity, indicating that autophagy is a negative regulator of TNF-α [126]. A mechanism study revealed that autophagy inhibits TNF-α expression through p38MAPK dephosphorylation and TRAF6 downregulation [127].

5. The Bidirectional Role of Autophagy in Immunotherapy

Whether autophagy plays a protective or destructive role in tumor cells or other cells depends on the degree of autophagy. Autophagy is a key player in regulating tumor immunity by affecting cells and cytokine release in tumor immunotherapy. Autophagy maintains this characteristic in TME. Although the direct connection between autophagy and immunotherapy has not been excessively explored, accumulating evidence indicates that autophagy may exert divergent effects on the tumor response to immunotherapy. The efficiency of immunotherapy may be enhanced or attenuated by autophagy, making autophagy a key player and a potential target for improving immunotherapy efficiency. In this section, we discuss the bidirectional role of autophagy in response to immunotherapy and demonstrate that modulation of autophagy can directly or indirectly enhance immunotherapy efficacy [81].

5.1. Autophagy Enhances the Effects of Immunotherapy

In immunotherapy, ferroptosis is one of the key mechanisms of tumor cell death mediated by CD8+ T cells [128]. A study demonstrated that knockout or knockdown of Atg5 and Atg7 prevents cancer cells from ferroptosis by decreasing intracellular ferrous iron levels and lipid peroxidation, indicating that autophagy may activate ferroptosis to enhance tumor immunotherapy [129,130]. Triple-negative breast cancer (TNBC) is usually characterized by autophagy defects, which inhibit T cell-mediated tumor killing in vitro and in vivo by blocking Tenascin-C degradation. Consequently, Tenascin-C inhibition sensitizes autophagy-defective TNBC cells to T cell killing and enhances the therapeutic effect of a single anti-PD1/PDL1 treatment. Based on these results, the researchers proposed a combination of Tenascin-C blockade and immune checkpoint inhibitors as a promising TNBC treatment [131].
As a supplement to immunotherapy, immunogenic cell death (ICD) is induced by many antitumor therapies to trigger antitumor immune responses. Autophagy is activated when ICD inducers are applied. In colon tumors, mitoxantrone or oxaliplatin treatment leads to autophagy in tumor cells, contributing to the infiltration of dendritic cells and T cells [132]. Mechanistically, T cell immunity is activated through antigen-presenting cells (APCs)-provided tumor antigens and DAMPs harbored in autophagosomes and released from dying cells [39]. Additionally, mitoxantrone-induced autophagy is also associated with the secretion of ATP in CT26 colon cancer cells. Extracellular ATP then recruits APCs and facilitates the synthesis of IL-1β to trigger an antitumor adaptive immune response [133]. Besides chemotherapy, autophagy may also contribute to the ICD to amplify the efficacy of radiotherapy. Ko et al. found that autophagy-induced ATP release from stressed or dying tumor cells triggers an anticancer immune response during radiotherapy [134]. The involvement of autophagy in targeted therapy has also been emphasized. As a tyrosine kinase inhibitor, sunitinib activates p62-dependent selective autophagy, thus inhibiting tumor PDL1 expression and facilitating anticancer immunity in metastatic breast cancer [135].

5.2. Autophagy Attenuates the Effects of Immunotherapy

Accumulating evidence indicate that hypoxia-induced autophagy is a mechanism related to immunotherapy failure that impairs CTLs-mediated tumor killing linked to STAT3 phosphorylation [136]. Additionally, hypoxia-induced autophagy eliminates NK cell-derived GrzB and weakens NK-mediated lysis in vitro and in vivo, indicating that autophagy may counteract immunotherapies that stimulate NK cells [57]. Tara et al. found that autophagy confers the resistance of cancer cells to immunotherapy. They discovered that deletion of Rb1cc1, an autophagy-related gene, sensitizes tumor cells to T cell killing, improving immune checkpoint blockade efficacy in mouse models [137]. Another study demonstrated the pleiotropic effects of autophagy in immunotherapy. It reported that knockout of ATG12 sensitizes cancer cells to CTLs, whereas knockout of ATG5 or ATG16L1 together with ATG12 results in resistance of cancer cells to CTLs [138].
Autophagy also participates in the indirect modulation of tumor immunity. When cancer cells undergo programmed cell death in response to various cell death inducers, dying cells release DAMPs into TME to activate immune response [53]. Inhibiting autophagy can facilitate this process, suggesting that a combination of autophagy inhibitors and cell death inducers can promote the anti-tumor effect of indirect immunotherapy, which has been verified in some animal models [139,140].

6. The Strategies of Targeting Autophagy to Facilitate Cancer Immunotherapy

Immunotherapy has demonstrated its great potential in cancer treatment and entered a new era after decades of development. Since autophagy plays a vital role in regulating tumor immune response, targeting autophagy appears to be a promising strategy to enhance immunotherapy efficacy and overcome immunotherapy resistance.
It has been reported that combining immunotherapy with radiotherapy or chemotherapy displays better efficacy than treatment alone. Radiotherapy or chemotherapy-induced autophagy contributes to augmenting mannose-6-phopsphate receptors (MPRs) in the tumor cell surface by autophagosome transportation, which increases the effectiveness of immunotherapy by making tumor cells more sensitive to CTLs [141,142]. Other research found that a semisynthetic vitamin E derivative, alpha-tocopheryloxyacetic acid (α-TEA), is an autophagy inducer that can aid in the presentation of tumor antigens to CD8+ T cells, thus improving anti-tumor immunity and immunotherapy efficacy [143]. Shen et al. designed a new probe, lactosylated N-Alkyl polyethylenimine-coated superparamagnetic iron oxide (SPIO) nanoparticles. They induced autophagy to promote DC maturation, indicating that SPIO nanoparticles may be a promising tool to enhance the vaccine functions of DCs [144]. Additionally, shikonin-induced autophagy can lead to DAMPs upregulation and activation of DCs [145]. Furthermore, the efficacy of DNA vaccines can be enhanced by synthesizing intracellular vaccine-encoded tumor antigens [146]. SQSTM1/p62 is a new cancer antigen connected to autophagy. The anti-tumor and anti-metastatic effects of p62-encoding DNA vaccines have been detected, and may be a potential method for tumor immunotherapy [147].
In melanoma and colorectal cancer, vacuolar protein sorting 34 (VPS34) inhibition by SB02024 or SAR405 upregulates the level of CCL5, CXCL10, and IFNγ in the TME, resulting in increased tumor infiltration of NK cells and T cells. Combining treatment of SB02024 or SAR405 with either anti-PD-1 or anti-PD-L1 prolonged survival of mice nearly 9 days in ananti-PD-1/PD-L1 treatment alone, indicating that the therapeutic efficacy of anti-PD-L1/PD-1 is improved by VPS34 inhibitor, which terminates autophagosome formation [148] (Table 1). Lawson et al. also reported that a VPS34 inhibitor, autophinib, sensitizes various tumor cell lines to TNFα [138] (Table 1). Sharma et al. found that anti-PD-1 Ab and HCQ decreased the tumor volume and increased the survival of mice compared with anti-PD-1 Ab alone. They proposed that targeting palmitoyl-protein thioesterase 1 (PPT1) by hydroxychloroquine (HCQ) or genetic Ppt1 inhibition can enhance the anti-PD-1 Ab efficacy in melanoma, which may be mediated by lysosome dysfunction [149] (Table 1). Other research demonstrated that autophagy inhibition by CQ improves antigen presentation and anti-tumor T cell responses by restoring surface levels of MHC-I [55] (Figure 3, Table 1). Yu et al. proposed an osteosarcoma treatment strategy by combination of the autophagy inhibitor 3-MA and photodynamic therapy (PDT), which induces an immunological response by downregulating PD-L1 expression, leading to tumor growth block in vitro and in vivo [150]. HDIL-2 is often applied for melanoma and renal cell carcinoma treatment, but this treatment easily causes side effects. Autophagy inhibitor CQ strengthens the effectiveness of HDIL-2 immunotherapy and reduces the toxicity in metastatic liver cancer [110] (Table 1). Similarly, the efficacy of HDIL-2 immunotherapy in treating renal cell carcinoma is improved by CQ by enhancing the functions of DCs, T cells, and NK cells, limiting ATP production, and increasing apoptosis [151] (Table 1). Other research discovered that the autophagy inhibitor 3-MA significantly promotes IL-24-induced apoptosis in oral squamous cell carcinomas (OSCC), suggesting that combining 3-MA and IL-24 may be a potential method in immunotherapy. Inhibition of autophagy can recover deficient ICD-based cancer immunotherapy. However, autophagy inhibition resulted in a loss of ATP and hampered the antitumor immune response. Hence, Li et al. proposed a novel coping strategy by introducing ATP as a remote loading gradient of the liposome to encapsulate HCQ (LipHCQa), which displayed a potent anti-colon cancer effect without restraining the immune response [152] (Table 1). The combination of HCQ and radiation therapy can enhance ICD in patients with glioblastoma multiforme [153]. Meanwhile, in combination with other therapies, autophagy inhibition can improve immune activity. For example, the results of a clinical trial indicated that gemcitabine and nab-paclitaxel chemotherapy in combination with HCQ treatment increased the infiltration of immune cells in patients with resectable pancreatic adenocarcinoma [154]. However, the lack of specificity in autophagy inhibitors needs to be addressed. CQ and HCQ may exert suppressive effects on immune responses to IDO1 and PD-L1 [155].
In addition to targeting autophagy itself, targeting the signaling that regulates autophagy has the potential to improve immunotherapy. For example, Wu et al. found that combination of the PD-1 blockade and endostar remarkably inhibited Lewis lung carcinoma (LLC) growth by activating PI3K/AKT/mTOR-mediated autophagy [156]. Conversely, inhibition of mTOR triggers autophagy, which mediates the resistance of tumor cells to T cell killing [137]. These facts suggest that mTOR is a promising autophagy-related target that may aid in immune system activation. Moreover, RAS, RAF, AMPK, and STAT3 are regarded as autophagy modulators to affect immunotherapy efficacy [4]. However, the highly context-dependent roles of these targets must be taken into consideration.
As a type of immunotherapy, CAR T cell therapy is clinically effective in treating hematologic cancers but is ineffective in treating solid tumors [157]. This may result from the TME of solid tumors, which creates a barrier to infiltration and the function of CAR T cells. Autophagy regulation is a recently proposed strategy to improve the efficacy of CAR T cell therapy since autophagy is closely related to TME and chemokine synthesis [158]. On the one hand, autophagy inhibition has been confirmed to promote CAR T cell tumor trafficking. CAR T cells eliminate tumor cells by recognizing cell surface antigens which can be degraded by autophagy [159]. Hence, inhibiting autophagy in tumors may enhance CAR T therapy efficacy by increasing the expression of antigens. On the other hand, the survival and normal function of T cells requires a high level of autophagy to deal with metabolic stress in TME [160]; therefore, strengthened autophagy in CAR T cells before transfusion to patients may improve the therapeutic outcome.

7. Perspectives

Currently, immunotherapies have produced favorable clinical outcomes. However, many patients do not respond, or develop resistance, to immunotherapy. Autophagy is a key regulator of immune responses in TME. The bidirectional roles of autophagy in cancer progression have been well established, which is context-dependent and tumor type-dependent. Autophagy in cancer cells or immune cells may improve or attenuate the effects of immunotherapy. Due to the insufficient understanding of the complexity of autophagy, the development of autophagy-base monotherapy or combined therapy is challenging. Hence, whether autophagy exerts pro-survival or pro-death effects, and how to enhance or inhibit it at different stages and in different cells, warrants further study. Some autophagy activators and inhibitors are being tested in preclinical and clinical studies. The combinations of autophagy modulators and various therapies have been confirmed to be effective, but only a few examples are immunotherapies. Since targeting autophagy appears to be an option in combinatorial therapy from which patients my benefit, further efforts should be taken to explore how to enhance tumor immunogenicity, improve T cell function, and weaken immunosuppressive TME by targeting autophagy.

Author Contributions

Conceptualization and modification: Y.L. (Yingjie Li). Writing review: Y.L. (Yuhe Lei) and L.B. Editing: E.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (31900505, Y.L.), (82074382, E.Z.), and the Natural Science Foundation of Guangdong Province (2021A1515011232, E.Z.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef]
  2. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
  3. Lei, Y.; Chen, L.; Liu, J.; Zhong, Y.; Deng, L. The MicroRNA-Based Strategies to Combat Cancer Chemoresistance via Regulating Autophagy. Front. Oncol. 2022, 12, 841625. [Google Scholar] [CrossRef]
  4. 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. [Google Scholar] [CrossRef] [PubMed]
  5. Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef] [PubMed]
  6. White, E.; Lattime, E.C.; Guo, J.Y. Autophagy Regulates Stress Responses, Metabolism, and Anticancer Immunity. Trends Cancer 2021, 7, 778–789. [Google Scholar] [CrossRef] [PubMed]
  7. Jiang, G.M.; Tan, Y.; Wang, H.; Peng, L.; Chen, H.T.; Meng, X.J.; Li, L.L.; Liu, Y.; Li, W.F.; Shan, H. The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol. Cancer 2019, 18, 17. [Google Scholar] [CrossRef] [PubMed]
  8. Amaravadi, R.K.; Kimmelman, A.C.; Debnath, J. Targeting Autophagy in Cancer: Recent Advances and Future Directions. Cancer Discov. 2019, 9, 1167–1181. [Google Scholar] [CrossRef] [PubMed]
  9. Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef]
  10. Behrends, C.; Sowa, M.E.; Gygi, S.P.; Harper, J.W. Network organization of the human autophagy system. Nature 2010, 466, 68–76. [Google Scholar] [CrossRef] [Green Version]
  11. Ge, L.; Melville, D.; Zhang, M.; Schekman, R. The ER-Golgi intermediate compartment is a key membrane source for the LC3 lipidation step of autophagosome biogenesis. Elife 2013, 2, e00947. [Google Scholar] [CrossRef] [PubMed]
  12. Dooley, H.C.; Razi, M.; Polson, H.E.; Girardin, S.E.; Wilson, M.I.; Tooze, S.A. WIPI2 links LC3 conjugation with PI3P, autophagosome formation, and pathogen clearance by recruiting Atg12-5-16L1. Mol. Cell 2014, 55, 238–252. [Google Scholar] [CrossRef] [PubMed]
  13. Walczak, M.; Martens, S. Dissecting the role of the Atg12-Atg5-Atg16 complex during autophagosome formation. Autophagy 2013, 9, 424–425. [Google Scholar] [CrossRef] [PubMed]
  14. Li, M.; Hou, Y.; Wang, J.; Chen, X.; Shao, Z.M.; Yin, X.M. Kinetics comparisons of mammalian Atg4 homologues indicate selective preferences toward diverse Atg8 substrates. J. Biol. Chem. 2011, 286, 7327–7338. [Google Scholar] [CrossRef] [PubMed]
  15. Romanov, J.; Walczak, M.; Ibiricu, I.; Schuchner, S.; Ogris, E.; Kraft, C.; Martens, S. Mechanism and functions of membrane binding by the Atg5-Atg12/Atg16 complex during autophagosome formation. EMBO J. 2012, 31, 4304–4317. [Google Scholar] [CrossRef] [PubMed]
  16. Moosavi, M.A.; Djavaheri-Mergny, M. Autophagy: New Insights into Mechanisms of Action and Resistance of Treatment in Acute Promyelocytic leukemia. Int. J. Mol. Sci. 2019, 20, 3559. [Google Scholar] [CrossRef]
  17. Xia, H.; Green, D.R.; Zou, W. Autophagy in tumour immunity and therapy. Nat. Rev. Cancer 2021, 21, 281–297. [Google Scholar] [CrossRef] [PubMed]
  18. Shibutani, S.T.; Yoshimori, T. A current perspective of autophagosome biogenesis. Cell Res. 2014, 24, 58–68. [Google Scholar] [CrossRef] [PubMed]
  19. Kimmelman, A.C.; White, E. Autophagy and Tumor Metabolism. Cell Metab. 2017, 25, 1037–1043. [Google Scholar] [CrossRef] [PubMed]
  20. Nakamura, S.; Yoshimori, T. New insights into autophagosome-lysosome fusion. J. Cell Sci. 2017, 130, 1209–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Mizushima, N.; Levine, B. Autophagy in Human Diseases. N. Engl. J. Med. 2020, 383, 1564–1576. [Google Scholar] [CrossRef] [PubMed]
  22. Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466. [Google Scholar] [CrossRef] [PubMed]
  23. Herzig, S.; Shaw, R.J. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. Cell Biol. 2018, 19, 121–135. [Google Scholar] [CrossRef] [PubMed]
  24. Hu, Y.L.; DeLay, M.; Jahangiri, A.; Molinaro, A.M.; Rose, S.D.; Carbonell, W.S.; Aghi, M.K. Hypoxia-induced autophagy promotes tumor cell survival and adaptation to antiangiogenic treatment in glioblastoma. Cancer Res. 2012, 72, 1773–1783. [Google Scholar] [CrossRef] [PubMed]
  25. Di Conza, G.; Trusso Cafarello, S.; Loroch, S.; Mennerich, D.; Deschoemaeker, S.; Di Matteo, M.; Ehling, M.; Gevaert, K.; Prenen, H.; Zahedi, R.P.; et al. The mTOR and PP2A Pathways Regulate PHD2 Phosphorylation to Fine-Tune HIF1alpha Levels and Colorectal Cancer Cell Survival under Hypoxia. Cell Rep. 2017, 18, 1699–1712. [Google Scholar] [CrossRef] [PubMed]
  26. Mazure, N.M.; Pouyssegur, J. Atypical BH3-domains of BNIP3 and BNIP3L lead to autophagy in hypoxia. Autophagy 2009, 5, 868–869. [Google Scholar] [CrossRef] [PubMed]
  27. Rouschop, K.M.; van den Beucken, T.; Dubois, L.; Niessen, H.; Bussink, J.; Savelkouls, K.; Keulers, T.; Mujcic, H.; Landuyt, W.; Voncken, J.W.; et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J. Clin. Investig. 2010, 120, 127–141. [Google Scholar] [CrossRef] [PubMed]
  28. Poillet-Perez, L.; Despouy, G.; Delage-Mourroux, R.; Boyer-Guittaut, M. Interplay between ROS and autophagy in cancer cells, from tumor initiation to cancer therapy. Redox Biol. 2015, 4, 184–192. [Google Scholar] [CrossRef]
  29. Alexander, A.; Cai, S.L.; Kim, J.; Nanez, A.; Sahin, M.; MacLean, K.H.; Inoki, K.; Guan, K.L.; Shen, J.; Person, M.D.; et al. ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS. Proc. Natl. Acad. Sci. USA 2010, 107, 4153–4158. [Google Scholar] [CrossRef] [PubMed]
  30. Song, C.; Mitter, S.K.; Qi, X.; Beli, E.; Rao, H.V.; Ding, J.; Ip, C.S.; Gu, H.; Akin, D.; Dunn, W.A., Jr.; et al. Oxidative stress-mediated NFkappaB phosphorylation upregulates p62/SQSTM1 and promotes retinal pigmented epithelial cell survival through increased autophagy. PLoS ONE 2017, 12, e0171940. [Google Scholar] [CrossRef] [Green Version]
  31. Morishita, H.; Mizushima, N. Diverse Cellular Roles of Autophagy. Annu. Rev. Cell Dev. Biol. 2019, 35, 453–475. [Google Scholar] [CrossRef] [PubMed]
  32. Li, X.; He, S.; Ma, B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 2020, 19, 12. [Google Scholar] [CrossRef] [PubMed]
  33. Mizushima, N. The ATG conjugation systems in autophagy. Curr. Opin. Cell Biol. 2020, 63, 1–10. [Google Scholar] [CrossRef] [PubMed]
  34. Qu, X.; Yu, J.; Bhagat, G.; Furuya, N.; Hibshoosh, H.; Troxel, A.; Rosen, J.; Eskelinen, E.L.; Mizushima, N.; Ohsumi, Y.; et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Investig. 2003, 112, 1809–1820. [Google Scholar] [CrossRef]
  35. Kenific, C.M.; Thorburn, A.; Debnath, J. Autophagy and metastasis: Another double-edged sword. Curr. Opin. Cell Biol. 2010, 22, 241–245. [Google Scholar] [CrossRef]
  36. Sosa, M.S.; Bragado, P.; Aguirre-Ghiso, J.A. Mechanisms of disseminated cancer cell dormancy: An awakening field. Nat. Rev. Cancer 2014, 14, 611–622. [Google Scholar] [CrossRef] [PubMed]
  37. Peng, Y.F.; Shi, Y.H.; Shen, Y.H.; Ding, Z.B.; Ke, A.W.; Zhou, J.; Qiu, S.J.; Fan, J. Promoting colonization in metastatic HCC cells by modulation of autophagy. PLoS ONE 2013, 8, e74407. [Google Scholar] [CrossRef]
  38. Dikic, I.; Elazar, Z. Mechanism and medical implications of mammalian autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 349–364. [Google Scholar] [CrossRef]
  39. Ma, Y.; Galluzzi, L.; Zitvogel, L.; Kroemer, G. Autophagy and cellular immune responses. Immunity 2013, 39, 211–227. [Google Scholar] [CrossRef]
  40. Zhan, Z.; Xie, X.; Cao, H.; Zhou, X.; Zhang, X.D.; Fan, H.; Liu, Z. Autophagy facilitates TLR4- and TLR3-triggered migration and invasion of lung cancer cells through the promotion of TRAF6 ubiquitination. Autophagy 2014, 10, 257–268. [Google Scholar] [CrossRef] [Green Version]
  41. Tang, D.; Kang, R.; Cheh, C.W.; Livesey, K.M.; Liang, X.; Schapiro, N.E.; Benschop, R.; Sparvero, L.J.; Amoscato, A.A.; Tracey, K.J.; et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene 2010, 29, 5299–5310. [Google Scholar] [CrossRef] [PubMed]
  42. Schmeisser, H.; Fey, S.B.; Horowitz, J.; Fischer, E.R.; Balinsky, C.A.; Miyake, K.; Bekisz, J.; Snow, A.L.; Zoon, K.C. Type I interferons induce autophagy in certain human cancer cell lines. Autophagy 2013, 9, 683–696. [Google Scholar] [CrossRef] [PubMed]
  43. Kiyono, K.; Suzuki, H.I.; Matsuyama, H.; Morishita, Y.; Komuro, A.; Kano, M.R.; Sugimoto, K.; Miyazono, K. Autophagy is activated by TGF-beta and potentiates TGF-beta-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res. 2009, 69, 8844–8852. [Google Scholar] [CrossRef] [PubMed]
  44. Katheder, N.S.; Khezri, R.; O’Farrell, F.; Schultz, S.W.; Jain, A.; Rahman, M.M.; Schink, K.O.; Theodossiou, T.A.; Johansen, T.; Juhasz, G.; et al. Microenvironmental autophagy promotes tumour growth. Nature 2017, 541, 417–420. [Google Scholar] [CrossRef]
  45. Zou, W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat. Rev. Cancer 2005, 5, 263–274. [Google Scholar] [CrossRef]
  46. Semmling, V.; Lukacs-Kornek, V.; Thaiss, C.A.; Quast, T.; Hochheiser, K.; Panzer, U.; Rossjohn, J.; Perlmutter, P.; Cao, J.; Godfrey, D.I.; et al. Alternative cross-priming through CCL17-CCR4-mediated attraction of CTLs toward NKT cell-licensed DCs. Nat. Immunol. 2010, 11, 313–320. [Google Scholar] [CrossRef]
  47. Li, H.; Li, Y.; Jiao, J.; Hu, H.M. Alpha-alumina nanoparticles induce efficient autophagy-dependent cross-presentation and potent antitumour response. Nat. Nanotechnol. 2011, 6, 645–650. [Google Scholar] [CrossRef]
  48. Yang, S.; Imamura, Y.; Jenkins, R.W.; Canadas, I.; Kitajima, S.; Aref, A.; Brannon, A.; Oki, E.; Castoreno, A.; Zhu, Z.; et al. Autophagy Inhibition Dysregulates TBK1 Signaling and Promotes Pancreatic Inflammation. Cancer Immunol. Res. 2016, 4, 520–530. [Google Scholar] [CrossRef]
  49. Wang, X.; Wu, W.K.K.; Gao, J.; Li, Z.; Dong, B.; Lin, X.; Li, Y.; Li, Y.; Gong, J.; Qi, C.; et al. Autophagy inhibition enhances PD-L1 expression in gastric cancer. J. Exp. Clin. Cancer Res. 2019, 38, 140. [Google Scholar] [CrossRef]
  50. Deretic, V.; Levine, B. Autophagy balances inflammation in innate immunity. Autophagy 2018, 14, 243–251. [Google Scholar] [CrossRef] [Green Version]
  51. Mathew, R.; Khor, S.; Hackett, S.R.; Rabinowitz, J.D.; Perlman, D.H.; White, E. Functional role of autophagy-mediated proteome remodeling in cell survival signaling and innate immunity. Mol. Cell 2014, 55, 916–930. [Google Scholar] [CrossRef] [PubMed]
  52. Poillet-Perez, L.; Sharp, D.W.; Yang, Y.; Laddha, S.V.; Ibrahim, M.; Bommareddy, P.K.; Hu, Z.S.; Vieth, J.; Haas, M.; Bosenberg, M.W.; et al. Autophagy promotes growth of tumors with high mutational burden by inhibiting a T-cell immune response. Nat. Cancer 2020, 1, 923–934. [Google Scholar] [CrossRef] [PubMed]
  53. Yatim, N.; Cullen, S.; Albert, M.L. Dying cells actively regulate adaptive immune responses. Nat. Rev. Immunol. 2017, 17, 262–275. [Google Scholar] [CrossRef] [PubMed]
  54. DeVorkin, L.; Pavey, N.; Carleton, G.; Comber, A.; Ho, C.; Lim, J.; McNamara, E.; Huang, H.; Kim, P.; Zacharias, L.G.; et al. Autophagy Regulation of Metabolism Is Required for CD8(+) T Cell Anti-tumor Immunity. Cell Rep. 2019, 27, 502–513.e5. [Google Scholar] [CrossRef] [PubMed]
  55. Yamamoto, K.; Venida, A.; Yano, J.; Biancur, D.E.; Kakiuchi, M.; Gupta, S.; Sohn, A.S.W.; Mukhopadhyay, S.; Lin, E.Y.; Parker, S.J.; et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 2020, 581, 100–105. [Google Scholar] [CrossRef]
  56. Garg, A.D.; Dudek, A.M.; Agostinis, P. Autophagy-dependent suppression of cancer immunogenicity and effector mechanisms of innate and adaptive immunity. Oncoimmunology 2013, 2, e26260. [Google Scholar] [CrossRef]
  57. Baginska, J.; Viry, E.; Berchem, G.; Poli, A.; Noman, M.Z.; van Moer, K.; Medves, S.; Zimmer, J.; Oudin, A.; Niclou, S.P.; et al. Granzyme B degradation by autophagy decreases tumor cell susceptibility to natural killer-mediated lysis under hypoxia. Proc. Natl. Acad. Sci. USA 2013, 110, 17450–17455. [Google Scholar] [CrossRef]
  58. Noman, M.Z.; Janji, B.; Kaminska, B.; Van Moer, K.; Pierson, S.; Przanowski, P.; Buart, S.; Berchem, G.; Romero, P.; Mami-Chouaib, F.; et al. Blocking hypoxia-induced autophagy in tumors restores cytotoxic T-cell activity and promotes regression. Cancer Res. 2011, 71, 5976–5986. [Google Scholar] [CrossRef]
  59. Zhong, Z.; Sanchez-Lopez, E.; Karin, M. Autophagy, Inflammation, and Immunity: A Troika Governing Cancer and Its Treatment. Cell 2016, 166, 288–298. [Google Scholar] [CrossRef]
  60. Blum, J.S.; Wearsch, P.A.; Cresswell, P. Pathways of antigen processing. Annu. Rev. Immunol. 2013, 31, 443–473. [Google Scholar] [CrossRef] [Green Version]
  61. Loi, M.; Muller, A.; Steinbach, K.; Niven, J.; Barreira da Silva, R.; Paul, P.; Ligeon, L.A.; Caruso, A.; Albrecht, R.A.; Becker, A.C.; et al. Macroautophagy Proteins Control MHC Class I Levels on Dendritic Cells and Shape Anti-viral CD8(+) T Cell Responses. Cell Rep. 2016, 15, 1076–1087. [Google Scholar] [CrossRef] [PubMed]
  62. Parekh, V.V.; Pabbisetty, S.K.; Wu, L.; Sebzda, E.; Martinez, J.; Zhang, J.; Van Kaer, L. Autophagy-related protein Vps34 controls the homeostasis and function of antigen cross-presenting CD8alpha(+) dendritic cells. Proc. Natl. Acad. Sci. USA 2017, 114, E6371–E6380. [Google Scholar] [CrossRef] [PubMed]
  63. Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, 328rv324. [Google Scholar] [CrossRef] [PubMed]
  64. Jang, Y.J.; Kim, J.H.; Byun, S. Modulation of Autophagy for Controlling Immunity. Cells 2019, 8, 138. [Google Scholar] [CrossRef] [PubMed]
  65. Clarke, A.J.; Simon, A.K. Autophagy in the renewal, differentiation and homeostasis of immune cells. Nat. Rev. Immunol. 2019, 19, 170–183. [Google Scholar] [CrossRef] [PubMed]
  66. Xia, H.; Wang, W.; Crespo, J.; Kryczek, I.; Li, W.; Wei, S.; Bian, Z.; Maj, T.; He, M.; Liu, R.J.; et al. Suppression of FIP200 and autophagy by tumor-derived lactate promotes naive T cell apoptosis and affects tumor immunity. Sci. Immunol. 2017, 2, eaan4631. [Google Scholar] [CrossRef] [PubMed]
  67. Sena, L.A.; Li, S.; Jairaman, A.; Prakriya, M.; Ezponda, T.; Hildeman, D.A.; Wang, C.R.; Schumacker, P.T.; Licht, J.D.; Perlman, H.; et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 2013, 38, 225–236. [Google Scholar] [CrossRef]
  68. Jia, W.; He, M.X.; McLeod, I.X.; Guo, J.; Ji, D.; He, Y.W. Autophagy regulates T lymphocyte proliferation through selective degradation of the cell-cycle inhibitor CDKN1B/p27Kip1. Autophagy 2015, 11, 2335–2345. [Google Scholar] [CrossRef] [PubMed]
  69. Bronietzki, A.W.; Schuster, M.; Schmitz, I. Autophagy in T-cell development, activation and differentiation. Immunol. Cell Biol. 2015, 93, 25–34. [Google Scholar] [CrossRef] [PubMed]
  70. Yang, G.; Song, W.; Postoak, J.L.; Chen, J.; Martinez, J.; Zhang, J.; Wu, L.; Van Kaer, L. Autophagy-related protein PIK3C3/VPS34 controls T cell metabolism and function. Autophagy 2021, 17, 1193–1204. [Google Scholar] [CrossRef]
  71. Pua, H.H.; Guo, J.; Komatsu, M.; He, Y.W. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J. Immunol. 2009, 182, 4046–4055. [Google Scholar] [CrossRef] [PubMed]
  72. Swadling, L.; Pallett, L.J.; Diniz, M.O.; Baker, J.M.; Amin, O.E.; Stegmann, K.A.; Burton, A.R.; Schmidt, N.M.; Jeffery-Smith, A.; Zakeri, N.; et al. Human Liver Memory CD8(+) T Cells Use Autophagy for Tissue Residence. Cell Rep. 2020, 30, 687–698.e686. [Google Scholar] [CrossRef] [PubMed]
  73. Delgoffe, G.M.; Kole, T.P.; Zheng, Y.; Zarek, P.E.; Matthews, K.L.; Xiao, B.; Worley, P.F.; Kozma, S.C.; Powell, J.D. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 2009, 30, 832–844. [Google Scholar] [CrossRef] [PubMed]
  74. Le Texier, L.; Lineburg, K.E.; Cao, B.; McDonald-Hyman, C.; Leveque-El Mouttie, L.; Nicholls, J.; Melino, M.; Nalkurthi, B.C.; Alexander, K.A.; Teal, B.; et al. Autophagy-dependent regulatory T cells are critical for the control of graft-versus-host disease. JCI Insight 2016, 1, e86850. [Google Scholar] [CrossRef]
  75. Wei, J.; Long, L.; Yang, K.; Guy, C.; Shrestha, S.; Chen, Z.; Wu, C.; Vogel, P.; Neale, G.; Green, D.R.; et al. Autophagy enforces functional integrity of regulatory T cells by coupling environmental cues and metabolic homeostasis. Nat. Immunol. 2016, 17, 277–285. [Google Scholar] [CrossRef]
  76. Becher, J.; Simula, L.; Volpe, E.; Procaccini, C.; La Rocca, C.; D’Acunzo, P.; Cianfanelli, V.; Strappazzon, F.; Caruana, I.; Nazio, F.; et al. AMBRA1 Controls Regulatory T-Cell Differentiation and Homeostasis Upstream of the FOXO3-FOXP3 Axis. Dev. Cell 2018, 47, 592–607.e596. [Google Scholar] [CrossRef]
  77. Munz, C. Autophagy proteins in antigen processing for presentation on MHC molecules. Immunol. Rev. 2016, 272, 17–27. [Google Scholar] [CrossRef]
  78. Li, W.; Tanikawa, T.; Kryczek, I.; Xia, H.; Li, G.; Wu, K.; Wei, S.; Zhao, L.; Vatan, L.; Wen, B.; et al. Aerobic Glycolysis Controls Myeloid-Derived Suppressor Cells and Tumor Immunity via a Specific CEBPB Isoform in Triple-Negative Breast Cancer. Cell Metab. 2018, 28, 87–103.e106. [Google Scholar] [CrossRef]
  79. Parker, K.H.; Horn, L.A.; Ostrand-Rosenberg, S. High-mobility group box protein 1 promotes the survival of myeloid-derived suppressor cells by inducing autophagy. J. Leukoc. Biol. 2016, 100, 463–470. [Google Scholar] [CrossRef]
  80. Alissafi, T.; Hatzioannou, A.; Mintzas, K.; Barouni, R.M.; Banos, A.; Sormendi, S.; Polyzos, A.; Xilouri, M.; Wielockx, B.; Gogas, H.; et al. Autophagy orchestrates the regulatory program of tumor-associated myeloid-derived suppressor cells. J. Clin. Investig. 2018, 128, 3840–3852. [Google Scholar] [CrossRef]
  81. Pan, H.; Chen, L.; Xu, Y.; Han, W.; Lou, F.; Fei, W.; Liu, S.; Jing, Z.; Sui, X. Autophagy-associated immune responses and cancer immunotherapy. Oncotarget 2016, 7, 21235–21246. [Google Scholar] [CrossRef] [PubMed]
  82. Arnold, J.; Murera, D.; Arbogast, F.; Fauny, J.D.; Muller, S.; Gros, F. Autophagy is dispensable for B-cell development but essential for humoral autoimmune responses. Cell Death Differ. 2016, 23, 853–864. [Google Scholar] [CrossRef] [PubMed]
  83. Fribourg, M.; Ni, J.; Nina Papavasiliou, F.; Yue, Z.; Heeger, P.S.; Leventhal, J.S. Allospecific Memory B Cell Responses Are Dependent on Autophagy. Am. J. Transpl. 2018, 18, 102–112. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, S.; Xia, P.; Huang, G.; Zhu, P.; Liu, J.; Ye, B.; Du, Y.; Fan, Z. FoxO1-mediated autophagy is required for NK cell development and innate immunity. Nat. Commun. 2016, 7, 11023. [Google Scholar] [CrossRef]
  85. Salio, M.; Puleston, D.J.; Mathan, T.S.; Shepherd, D.; Stranks, A.J.; Adamopoulou, E.; Veerapen, N.; Besra, G.S.; Hollander, G.A.; Simon, A.K.; et al. Essential role for autophagy during invariant NKT cell development. Proc. Natl. Acad. Sci. USA 2014, 111, E5678–E5687. [Google Scholar] [CrossRef]
  86. Pei, B.; Zhao, M.; Miller, B.C.; Vela, J.L.; Bruinsma, M.W.; Virgin, H.W.; Kronenberg, M. Invariant NKT cells require autophagy to coordinate proliferation and survival signals during differentiation. J. Immunol. 2015, 194, 5872–5884. [Google Scholar] [CrossRef]
  87. Liu, E.; Van Grol, J.; Subauste, C.S. Atg5 but not Atg7 in dendritic cells enhances IL-2 and IFN-gamma production by Toxoplasma gondii-reactive CD4+ T cells. Microbes Infect. 2015, 17, 275–284. [Google Scholar] [CrossRef]
  88. Seto, S.; Tsujimura, K.; Horii, T.; Koide, Y. Autophagy adaptor protein p62/SQSTM1 and autophagy-related gene Atg5 mediate autophagosome formation in response to Mycobacterium tuberculosis infection in dendritic cells. PLoS ONE 2013, 8, e86017. [Google Scholar] [CrossRef]
  89. Lee, H.K.; Mattei, L.M.; Steinberg, B.E.; Alberts, P.; Lee, Y.H.; Chervonsky, A.; Mizushima, N.; Grinstein, S.; Iwasaki, A. In vivo requirement for Atg5 in antigen presentation by dendritic cells. Immunity 2010, 32, 227–239. [Google Scholar] [CrossRef]
  90. Chen, P.; Cescon, M.; Bonaldo, P. Autophagy-mediated regulation of macrophages and its applications for cancer. Autophagy 2014, 10, 192–200. [Google Scholar] [CrossRef]
  91. Jacquel, A.; Obba, S.; Solary, E.; Auberger, P. Proper macrophagic differentiation requires both autophagy and caspase activation. Autophagy 2012, 8, 1141–1143. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, Y.; Morgan, M.J.; Chen, K.; Choksi, S.; Liu, Z.G. Induction of autophagy is essential for monocyte-macrophage differentiation. Blood 2012, 119, 2895–2905. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, K.; Zhao, E.; Ilyas, G.; Lalazar, G.; Lin, Y.; Haseeb, M.; Tanaka, K.E.; Czaja, M.J. Impaired macrophage autophagy increases the immune response in obese mice by promoting proinflammatory macrophage polarization. Autophagy 2015, 11, 271–284. [Google Scholar] [CrossRef] [PubMed]
  94. Li, N.; Qin, J.; Lan, L.; Zhang, H.; Liu, F.; Wu, Z.; Ni, H.; Wang, Y. PTEN inhibits macrophage polarization from M1 to M2 through CCL2 and VEGF-A reduction and NHERF-1 synergism. Cancer Biol. Ther. 2015, 16, 297–306. [Google Scholar] [CrossRef] [PubMed]
  95. Khatoon, E.; Parama, D.; Kumar, A.; Alqahtani, M.S.; Abbas, M.; Girisa, S.; Sethi, G.; Kunnumakkara, A.B. Targeting PD-1/PD-L1 axis as new horizon for ovarian cancer therapy. Life Sci. 2022, 306, 120827. [Google Scholar] [CrossRef]
  96. Robainas, M.; Otano, R.; Bueno, S.; Ait-Oudhia, S. Understanding the role of PD-L1/PD1 pathway blockade and autophagy in cancer therapy. OncoTargets Ther. 2017, 10, 1803–1807. [Google Scholar] [CrossRef]
  97. Maher, C.M.; Thomas, J.D.; Haas, D.A.; Longen, C.G.; Oyer, H.M.; Tong, J.Y.; Kim, F.J. Small-Molecule Sigma1 Modulator Induces Autophagic Degradation of PD-L1. Mol. Cancer Res. 2018, 16, 243–255. [Google Scholar] [CrossRef]
  98. Clark, C.A.; Gupta, H.B.; Curiel, T.J. Tumor cell-intrinsic CD274/PD-L1: A novel metabolic balancing act with clinical potential. Autophagy 2017, 13, 987–988. [Google Scholar] [CrossRef]
  99. Shukla, S.A.; Bachireddy, P.; Schilling, B.; Galonska, C.; Zhan, Q.; Bango, C.; Langer, R.; Lee, P.C.; Gusenleitner, D.; Keskin, D.B.; et al. Cancer-Germline Antigen Expression Discriminates Clinical Outcome to CTLA-4 Blockade. Cell 2018, 173, 624–633.e628. [Google Scholar] [CrossRef]
  100. Kato, H.; Perl, A. Blockade of Treg Cell Differentiation and Function by the Interleukin-21-Mechanistic Target of Rapamycin Axis Via Suppression of Autophagy in Patients With Systemic Lupus Erythematosus. Arthritis Rheumatol. 2018, 70, 427–438. [Google Scholar] [CrossRef] [Green Version]
  101. Alissafi, T.; Banos, A.; Boon, L.; Sparwasser, T.; Ghigo, A.; Wing, K.; Vassilopoulos, D.; Boumpas, D.; Chavakis, T.; Cadwell, K.; et al. Tregs restrain dendritic cell autophagy to ameliorate autoimmunity. J. Clin. Investig. 2017, 127, 2789–2804. [Google Scholar] [CrossRef] [PubMed]
  102. Folgiero, V.; Miele, E.; Carai, A.; Ferretti, E.; Alfano, V.; Po, A.; Bertaina, V.; Goffredo, B.M.; Benedetti, M.C.; Camassei, F.D.; et al. IDO1 involvement in mTOR pathway: A molecular mechanism of resistance to mTOR targeting in medulloblastoma. Oncotarget 2016, 7, 52900–52911. [Google Scholar] [CrossRef] [PubMed]
  103. Metz, R.; Rust, S.; Duhadaway, J.B.; Mautino, M.R.; Munn, D.H.; Vahanian, N.N.; Link, C.J.; Prendergast, G.C. IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: A novel IDO effector pathway targeted by D-1-methyl-tryptophan. Oncoimmunology 2012, 1, 1460–1468. [Google Scholar] [CrossRef] [PubMed]
  104. McGaha, T.L. IDO-GCN2 and autophagy in inflammation. Oncotarget 2015, 6, 21771–21772. [Google Scholar] [CrossRef]
  105. Schauer, I.G.; Zhang, J.; Xing, Z.; Guo, X.; Mercado-Uribe, I.; Sood, A.K.; Huang, P.; Liu, J. Interleukin-1beta promotes ovarian tumorigenesis through a p53/NF-kappaB-mediated inflammatory response in stromal fibroblasts. Neoplasia 2013, 15, 409–420. [Google Scholar] [CrossRef]
  106. Jiang, S.; Dupont, N.; Castillo, E.F.; Deretic, V. Secretory versus degradative autophagy: Unconventional secretion of inflammatory mediators. J. Innate Immun. 2013, 5, 471–479. [Google Scholar] [CrossRef]
  107. Peral de Castro, C.; Jones, S.A.; Ni Cheallaigh, C.; Hearnden, C.A.; Williams, L.; Winter, J.; Lavelle, E.C.; Mills, K.H.; Harris, J. Autophagy regulates IL-23 secretion and innate T cell responses through effects on IL-1 secretion. J. Immunol. 2012, 189, 4144–4153. [Google Scholar] [CrossRef]
  108. Sun, K.; Xu, L.; Jing, Y.; Han, Z.; Chen, X.; Cai, C.; Zhao, P.; Zhao, X.; Yang, L.; Wei, L. Autophagy-deficient Kupffer cells promote tumorigenesis by enhancing mtROS-NF-kappaB-IL1alpha/beta-dependent inflammation and fibrosis during the preneoplastic stage of hepatocarcinogenesis. Cancer Lett. 2017, 388, 198–207. [Google Scholar] [CrossRef]
  109. Kang, R.; Tang, D.; Lotze, M.T.; Zeh Iii, H.J. Autophagy is required for IL-2-mediated fibroblast growth. Exp. Cell Res. 2013, 319, 556–565. [Google Scholar] [CrossRef]
  110. Liang, X.; De Vera, M.E.; Buchser, W.J.; Romo de Vivar Chavez, A.; Loughran, P.; Beer Stolz, D.; Basse, P.; Wang, T.; Van Houten, B.; Zeh, H.J., 3rd; et al. Inhibiting systemic autophagy during interleukin 2 immunotherapy promotes long-term tumor regression. Cancer Res. 2012, 72, 2791–2801. [Google Scholar] [CrossRef] [Green Version]
  111. Qin, B.; Zhou, Z.; He, J.; Yan, C.; Ding, S. IL-6 Inhibits Starvation-induced Autophagy via the STAT3/Bcl-2 Signaling Pathway. Sci. Rep. 2015, 5, 15701. [Google Scholar] [CrossRef] [PubMed]
  112. Linnemann, A.K.; Blumer, J.; Marasco, M.R.; Battiola, T.J.; Umhoefer, H.M.; Han, J.Y.; Lamming, D.W.; Davis, D.B. Interleukin 6 protects pancreatic beta cells from apoptosis by stimulation of autophagy. FASEB J. 2017, 31, 4140–4152. [Google Scholar] [CrossRef]
  113. Santarelli, R.; Gonnella, R.; Di Giovenale, G.; Cuomo, L.; Capobianchi, A.; Granato, M.; Gentile, G.; Faggioni, A.; Cirone, M. STAT3 activation by KSHV correlates with IL-10, IL-6 and IL-23 release and an autophagic block in dendritic cells. Sci. Rep. 2014, 4, 4241. [Google Scholar] [CrossRef] [PubMed]
  114. Cho, S.H.; Oh, S.Y.; Lane, A.P.; Lee, J.; Oh, M.H.; Lee, S.; Zheng, T.; Zhu, Z. Regulation of nasal airway homeostasis and inflammation in mice by SHP-1 and Th2/Th1 signaling pathways. PLoS ONE 2014, 9, e103685. [Google Scholar] [CrossRef] [PubMed]
  115. Qi, G.M.; Jia, L.X.; Li, Y.L.; Li, H.H.; Du, J. Adiponectin suppresses angiotensin II-induced inflammation and cardiac fibrosis through activation of macrophage autophagy. Endocrinology 2014, 155, 2254–2265. [Google Scholar] [CrossRef]
  116. Schmeisser, H.; Bekisz, J.; Zoon, K.C. New function of type I IFN: Induction of autophagy. J. Interf. Cytokine Res. 2014, 34, 71–78. [Google Scholar] [CrossRef]
  117. Buchser, W.J.; Laskow, T.C.; Pavlik, P.J.; Lin, H.M.; Lotze, M.T. Cell-mediated autophagy promotes cancer cell survival. Cancer Res. 2012, 72, 2970–2979. [Google Scholar] [CrossRef]
  118. Matsuzawa, T.; Kim, B.H.; Shenoy, A.R.; Kamitani, S.; Miyake, M.; Macmicking, J.D. IFN-gamma elicits macrophage autophagy via the p38 MAPK signaling pathway. J. Immunol. 2012, 189, 813–818. [Google Scholar] [CrossRef]
  119. Tu, S.P.; Quante, M.; Bhagat, G.; Takaishi, S.; Cui, G.; Yang, X.D.; Muthuplani, S.; Shibata, W.; Fox, J.G.; Pritchard, D.M.; et al. IFN-gamma inhibits gastric carcinogenesis by inducing epithelial cell autophagy and T-cell apoptosis. Cancer Res. 2011, 71, 4247–4259. [Google Scholar] [CrossRef]
  120. Hubbard, V.M.; Valdor, R.; Patel, B.; Singh, R.; Cuervo, A.M.; Macian, F. Macroautophagy regulates energy metabolism during effector T cell activation. J. Immunol. 2010, 185, 7349–7357. [Google Scholar] [CrossRef] [Green Version]
  121. Wilson, E.B.; El-Jawhari, J.J.; Neilson, A.L.; Hall, G.D.; Melcher, A.A.; Meade, J.L.; Cook, G.P. Human tumour immune evasion via TGF-beta blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity. PLoS ONE 2011, 6, e22842. [Google Scholar] [CrossRef] [PubMed]
  122. Ding, Y.; Kim, S.; Lee, S.Y.; Koo, J.K.; Wang, Z.; Choi, M.E. Autophagy regulates TGF-beta expression and suppresses kidney fibrosis induced by unilateral ureteral obstruction. J. Am. Soc. Nephrol. 2014, 25, 2835–2846. [Google Scholar] [CrossRef]
  123. Zhang, C.; Zhang, X.; Xu, R.; Huang, B.; Chen, A.J.; Li, C.; Wang, J.; Li, X.G. TGF-beta2 initiates autophagy via Smad and non-Smad pathway to promote glioma cells’ invasion. J. Exp. Clin. Cancer Res. 2017, 36, 162. [Google Scholar] [CrossRef]
  124. Suzuki, H.I.; Kiyono, K.; Miyazono, K. Regulation of autophagy by transforming growth factor-beta (TGF-beta) signaling. Autophagy 2010, 6, 645–647. [Google Scholar] [CrossRef]
  125. Wang, M.X.; Cheng, X.Y.; Jin, M.; Cao, Y.L.; Yang, Y.P.; Wang, J.D.; Li, Q.; Wang, F.; Hu, L.F.; Liu, C.F. TNF compromises lysosome acidification and reduces alpha-synuclein degradation via autophagy in dopaminergic cells. Exp. Neurol. 2015, 271, 112–121. [Google Scholar] [CrossRef] [PubMed]
  126. Ullio, C.; Brunk, U.T.; Urani, C.; Melchioretto, P.; Bonelli, G.; Baccino, F.M.; Autelli, R. Autophagy of metallothioneins prevents TNF-induced oxidative stress and toxicity in hepatoma cells. Autophagy 2015, 11, 2184–2198. [Google Scholar] [CrossRef] [PubMed]
  127. Pun, N.T.; Subedi, A.; Kim, M.J.; Park, P.H. Globular Adiponectin Causes Tolerance to LPS-Induced TNF-alpha Expression via Autophagy Induction in RAW 264.7 Macrophages: Involvement of SIRT1/FoxO3A Axis. PLoS ONE 2015, 10, e0124636. [Google Scholar] [CrossRef]
  128. Wang, W.; Green, M.; Choi, J.E.; Gijon, M.; Kennedy, P.D.; Johnson, J.K.; Liao, P.; Lang, X.; Kryczek, I.; Sell, A.; et al. CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019, 569, 270–274. [Google Scholar] [CrossRef]
  129. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J., 3rd; Kang, R.; Tang, D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016, 12, 1425–1428. [Google Scholar] [CrossRef]
  130. Gao, M.; Monian, P.; Pan, Q.; Zhang, W.; Xiang, J.; Jiang, X. Ferroptosis is an autophagic cell death process. Cell Res. 2016, 26, 1021–1032. [Google Scholar] [CrossRef] [Green Version]
  131. Li, Z.L.; Zhang, H.L.; Huang, Y.; Huang, J.H.; Sun, P.; Zhou, N.N.; Chen, Y.H.; Mai, J.; Wang, Y.; Yu, Y.; et al. Autophagy deficiency promotes triple-negative breast cancer resistance to T cell-mediated cytotoxicity by blocking tenascin-C degradation. Nat. Commun. 2020, 11, 3806. [Google Scholar] [CrossRef] [PubMed]
  132. Michaud, M.; Martins, I.; Sukkurwala, A.Q.; Adjemian, S.; Ma, Y.; Pellegatti, P.; Shen, S.; Kepp, O.; Scoazec, M.; Mignot, G.; et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 2011, 334, 1573–1577. [Google Scholar] [CrossRef] [PubMed]
  133. Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef] [PubMed]
  134. Ko, A.; Kanehisa, A.; Martins, I.; Senovilla, L.; Chargari, C.; Dugue, D.; Marino, G.; Kepp, O.; Michaud, M.; Perfettini, J.L.; et al. Autophagy inhibition radiosensitizes in vitro, yet reduces radioresponses in vivo due to deficient immunogenic signalling. Cell Death Differ. 2014, 21, 92–99. [Google Scholar] [CrossRef] [PubMed]
  135. Li, H.; Kuang, X.; Liang, L.; Ye, Y.; Zhang, Y.; Li, J.; Ma, F.; Tao, J.; Lei, G.; Zhao, S.; et al. The Beneficial Role of Sunitinib in Tumor Immune Surveillance by Regulating Tumor PD-L1. Adv. Sci. 2021, 8, 2001596. [Google Scholar] [CrossRef]
  136. Noman, M.Z.; Janji, B.; Berchem, G.; Mami-Chouaib, F.; Chouaib, S. Hypoxia-induced autophagy: A new player in cancer immunotherapy? Autophagy 2012, 8, 704–706. [Google Scholar] [CrossRef]
  137. Young, T.M.; Reyes, C.; Pasnikowski, E.; Castanaro, C.; Wong, C.; Decker, C.E.; Chiu, J.; Song, H.; Wei, Y.; Bai, Y.; et al. Autophagy protects tumors from T cell-mediated cytotoxicity via inhibition of TNFalpha-induced apoptosis. Sci. Immunol. 2020, 5, eabb9561. [Google Scholar] [CrossRef]
  138. Lawson, K.A.; Sousa, C.M.; Zhang, X.; Kim, E.; Akthar, R.; Caumanns, J.J.; Yao, Y.; Mikolajewicz, N.; Ross, C.; Brown, K.R.; et al. Functional genomic landscape of cancer-intrinsic evasion of killing by T cells. Nature 2020, 586, 120–126. [Google Scholar] [CrossRef]
  139. Mulcahy Levy, J.M.; Zahedi, S.; Griesinger, A.M.; Morin, A.; Davies, K.D.; Aisner, D.L.; Kleinschmidt-DeMasters, B.K.; Fitzwalter, B.E.; Goodall, M.L.; Thorburn, J.; et al. Autophagy inhibition overcomes multiple mechanisms of resistance to BRAF inhibition in brain tumors. Elife 2017, 6, e19671. [Google Scholar] [CrossRef]
  140. Bryant, K.L.; Stalnecker, C.A.; Zeitouni, D.; Klomp, J.E.; Peng, S.; Tikunov, A.P.; Gunda, V.; Pierobon, M.; Waters, A.M.; George, S.D.; et al. Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat. Med. 2019, 25, 628–640. [Google Scholar] [CrossRef]
  141. Kim, S.; Ramakrishnan, R.; Lavilla-Alonso, S.; Chinnaiyan, P.; Rao, N.; Fowler, E.; Heine, J.; Gabrilovich, D.I. Radiation-induced autophagy potentiates immunotherapy of cancer via up-regulation of mannose 6-phosphate receptor on tumor cells in mice. Cancer Immunol. Immunother. 2014, 63, 1009–1021. [Google Scholar] [CrossRef] [PubMed]
  142. Ramakrishnan, R.; Huang, C.; Cho, H.I.; Lloyd, M.; Johnson, J.; Ren, X.; Altiok, S.; Sullivan, D.; Weber, J.; Celis, E.; et al. Autophagy induced by conventional chemotherapy mediates tumor cell sensitivity to immunotherapy. Cancer Res. 2012, 72, 5483–5493. [Google Scholar] [CrossRef] [PubMed]
  143. Hahn, T.; Akporiaye, E.T. alpha-TEA as a stimulator of tumor autophagy and enhancer of antigen cross-presentation. Autophagy 2013, 9, 429–431. [Google Scholar] [CrossRef] [PubMed]
  144. Shen, T.; Zhu, W.; Yang, L.; Liu, L.; Jin, R.; Duan, J.; Anderson, J.M.; Ai, H. Lactosylated N-Alkyl polyethylenimine coated iron oxide nanoparticles induced autophagy in mouse dendritic cells. Regen. Biomater. 2018, 5, 141–149. [Google Scholar] [CrossRef]
  145. Lin, S.Y.; Hsieh, S.Y.; Fan, Y.T.; Wei, W.C.; Hsiao, P.W.; Tsai, D.H.; Wu, T.S.; Yang, N.S. Necroptosis promotes autophagy-dependent upregulation of DAMP and results in immunosurveillance. Autophagy 2018, 14, 778–795. [Google Scholar] [CrossRef]
  146. Dai, Z.; Huang, J.; Lei, X.; Yan, Y.; Lu, P.; Zhang, H.; Lin, W.; Chen, W.; Ma, J.; Xie, Q. Efficacy of an autophagy-targeted DNA vaccine against avian leukosis virus subgroup J. Vaccine 2017, 35, 808–813. [Google Scholar] [CrossRef]
  147. Gabai, V.L.; Shifrin, V.I. Feasibility analysis of p62 (SQSTM1)-encoding DNA vaccine as a novel cancer immunotherapy. Int. Rev. Immunol. 2014, 33, 375–382. [Google Scholar] [CrossRef]
  148. Noman, M.Z.; Parpal, S.; Van Moer, K.; Xiao, M.; Yu, Y.; Viklund, J.; De Milito, A.; Hasmim, M.; Andersson, M.; Amaravadi, R.K.; et al. Inhibition of Vps34 reprograms cold into hot inflamed tumors and improves anti-PD-1/PD-L1 immunotherapy. Sci. Adv. 2020, 6, eaax7881. [Google Scholar] [CrossRef]
  149. Sharma, G.; Ojha, R.; Noguera-Ortega, E.; Rebecca, V.W.; Attanasio, J.; Liu, S.; Piao, S.; Lee, J.J.; Nicastri, M.C.; Harper, S.L.; et al. PPT1 inhibition enhances the antitumor activity of anti-PD-1 antibody in melanoma. JCI Insight 2020, 5, e133225. [Google Scholar] [CrossRef]
  150. Yu, W.; Wang, Y.; Zhu, J.; Jin, L.; Liu, B.; Xia, K.; Wang, J.; Gao, J.; Liang, C.; Tao, H. Autophagy inhibitor enhance ZnPc/BSA nanoparticle induced photodynamic therapy by suppressing PD-L1 expression in osteosarcoma immunotherapy. Biomaterials 2019, 192, 128–139. [Google Scholar] [CrossRef]
  151. Lotze, M.T.; Buchser, W.J.; Liang, X. Blocking the interleukin 2 (IL2)-induced systemic autophagic syndrome promotes profound antitumor effects and limits toxicity. Autophagy 2012, 8, 1264–1266. [Google Scholar] [CrossRef] [PubMed]
  152. Li, J.; Cai, W.; Yu, J.; Zhou, S.; Li, X.; He, Z.; Ouyang, D.; Liu, H.; Wang, Y. Autophagy inhibition recovers deficient ICD-based cancer immunotherapy. Biomaterials 2022, 287, 121651. [Google Scholar] [CrossRef] [PubMed]
  153. Rosenfeld, M.R.; Ye, X.; Supko, J.G.; Desideri, S.; Grossman, S.A.; Brem, S.; Mikkelson, T.; Wang, D.; Chang, Y.C.; Hu, J.; et al. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy 2014, 10, 1359–1368. [Google Scholar] [CrossRef] [PubMed]
  154. Zeh, H.J.; Bahary, N.; Boone, B.A.; Singhi, A.D.; Miller-Ocuin, J.L.; Normolle, D.P.; Zureikat, A.H.; Hogg, M.E.; Bartlett, D.L.; Lee, K.K.; et al. A Randomized Phase II Preoperative Study of Autophagy Inhibition with High-Dose Hydroxychloroquine and Gemcitabine/Nab-Paclitaxel in Pancreatic Cancer Patients. Clin. Cancer Res. 2020, 26, 3126–3134. [Google Scholar] [CrossRef] [PubMed]
  155. Gostner, J.M.; Schrocksnadel, S.; Becker, K.; Jenny, M.; Schennach, H.; Uberall, F.; Fuchs, D. Antimalarial drug chloroquine counteracts activation of indoleamine (2,3)-dioxygenase activity in human PBMC. FEBS Open Biol. 2012, 2, 241–245. [Google Scholar] [CrossRef]
  156. Wu, J.; Zhao, X.; Sun, Q.; Jiang, Y.; Zhang, W.; Luo, J.; Li, Y. Synergic effect of PD-1 blockade and endostar on the PI3K/AKT/mTOR-mediated autophagy and angiogenesis in Lewis lung carcinoma mouse model. Biomed. Pharmacother. 2020, 125, 109746. [Google Scholar] [CrossRef]
  157. Newick, K.; O’Brien, S.; Moon, E.; Albelda, S.M. CAR T Cell Therapy for Solid Tumors. Annu. Rev. Med. 2017, 68, 139–152. [Google Scholar] [CrossRef]
  158. Levy, J.; Cacheux, W.; Bara, M.A.; L’Hermitte, A.; Lepage, P.; Fraudeau, M.; Trentesaux, C.; Lemarchand, J.; Durand, A.; Crain, A.M.; et al. Intestinal inhibition of Atg7 prevents tumour initiation through a microbiome-influenced immune response and suppresses tumour growth. Nat. Cell Biol. 2015, 17, 1062–1073. [Google Scholar] [CrossRef]
  159. Li, Y.; Wang, L.X.; Yang, G.; Hao, F.; Urba, W.J.; Hu, H.M. Efficient cross-presentation depends on autophagy in tumor cells. Cancer Res. 2008, 68, 6889–6895. [Google Scholar] [CrossRef]
  160. Bian, Y.; Li, W.; Kremer, D.M.; Sajjakulnukit, P.; Li, S.; Crespo, J.; Nwosu, Z.C.; Zhang, L.; Czerwonka, A.; Pawlowska, A.; et al. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature 2020, 585, 277–282. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of autophagy-mediated immune evasion. (a) Autophagy deficiency in gastric cancer enhances PD-L1 expression by activating NF-κB, therefore promoting PD1/PD-L1-induced immune evasion. (b) Hypoxia-induced autophagy impairs killing of breast cancer by degrading NK and CD8+ T cell-derived granzyme B in autophagosomes. In addition, autophagy inhibits chemokine CCL5 production. As a result, the recruitment of NK cells to the TME is blocked. (c) MHC-I is recognized by NBR1 and degraded in lysosomes by autophagy in pancreatic cancer cells, which impairs antigen presentation and tumor killing by T cells.
Figure 1. Mechanisms of autophagy-mediated immune evasion. (a) Autophagy deficiency in gastric cancer enhances PD-L1 expression by activating NF-κB, therefore promoting PD1/PD-L1-induced immune evasion. (b) Hypoxia-induced autophagy impairs killing of breast cancer by degrading NK and CD8+ T cell-derived granzyme B in autophagosomes. In addition, autophagy inhibits chemokine CCL5 production. As a result, the recruitment of NK cells to the TME is blocked. (c) MHC-I is recognized by NBR1 and degraded in lysosomes by autophagy in pancreatic cancer cells, which impairs antigen presentation and tumor killing by T cells.
Cells 11 02996 g001
Figure 2. The formation of immunosuppressive TME. Cancer cell-derived lactate lowers FIP200 expression and attenuates autophagy, which is pivotal for energy homeostasis of effector T cells, thus facilitating T cell exhaustion and formation of immunosuppressive TME. In Treg cells, decreased intracellular arginine suppresses mTOR, leading to autophagy activation that is responsible for Treg cell survival. As a consequence, the function of Treg cell is improved to form the immunosuppressive TME.
Figure 2. The formation of immunosuppressive TME. Cancer cell-derived lactate lowers FIP200 expression and attenuates autophagy, which is pivotal for energy homeostasis of effector T cells, thus facilitating T cell exhaustion and formation of immunosuppressive TME. In Treg cells, decreased intracellular arginine suppresses mTOR, leading to autophagy activation that is responsible for Treg cell survival. As a consequence, the function of Treg cell is improved to form the immunosuppressive TME.
Cells 11 02996 g002
Figure 3. Autophagy blockage sensitizes cancer cells to immunotherapy. (a) In tumor cells, surface MHC-I is degraded by the autophagy-lysosome traffic, facilitating malignant cells to evade immunologic surveillance. (b) Blockage of autophagosome or lysosome intercepts MHC-I degradation, enhancing efficacy of checkpoint blockage inhibitors.
Figure 3. Autophagy blockage sensitizes cancer cells to immunotherapy. (a) In tumor cells, surface MHC-I is degraded by the autophagy-lysosome traffic, facilitating malignant cells to evade immunologic surveillance. (b) Blockage of autophagosome or lysosome intercepts MHC-I degradation, enhancing efficacy of checkpoint blockage inhibitors.
Cells 11 02996 g003
Table 1. Autophagy inhibitors enhance the efficacy of immunotherapy.
Table 1. Autophagy inhibitors enhance the efficacy of immunotherapy.
Autophagy InhibitorTargetsImmunotherapyTumor TypesRefs.
SB02024Vps34Anti-PD-L1 and Anti-PD-1Melanoma, CRC[148]
SAR405Vps34Anti-PD-L1 and Anti-PD-1Melanoma, CRC[148]
AutophinibVps34TNFαVarious cancers[138]
HydroxychloroquineLysosomes, PPT1anti-PD-1Melanoma[149]
LipHCQaLysosomesShikonin-induced ICDColon cancer[152]
ChloroquineLysosomesanti-PD1/CTLA4
HDIL-2
Pancreatic cancer, metastatic liver cancer, renal cell carcinoma[55,110,151]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lei, Y.; Zhang, E.; Bai, L.; Li, Y. Autophagy in Cancer Immunotherapy. Cells 2022, 11, 2996. https://doi.org/10.3390/cells11192996

AMA Style

Lei Y, Zhang E, Bai L, Li Y. Autophagy in Cancer Immunotherapy. Cells. 2022; 11(19):2996. https://doi.org/10.3390/cells11192996

Chicago/Turabian Style

Lei, Yuhe, Enxin Zhang, Liangliang Bai, and Yingjie Li. 2022. "Autophagy in Cancer Immunotherapy" Cells 11, no. 19: 2996. https://doi.org/10.3390/cells11192996

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

Article Metrics

Back to TopTop