Abstract
Macro-autophagy is an ancient and highly-conserved self-degradative process that plays a homeostatic role in normal cells by eliminating organelles, pathogens and protein aggregates. Autophagy, as it is routinely referred to, also allows cells to maintain metabolic sufficiency and survive under conditions of nutrient stress by recycling the by-products of autophagic degradation, such as fatty acids, amino acids and nucleotides. Tumor cells are more reliant than normal cells on autophagy for survival in part due to their rapid growth rate, altered metabolism and nutrient deprived growth environment. How this dependence of tumor cells on autophagy affects their progression to malignancy and metastatic disease is an area of increasing research focus. Here, we review recent work identifying critical functions for autophagy in tumor cell migration and invasion, tumor stem cell maintenance and therapy resistance and cross-talk between tumor cells and their microenvironment.
Keywords: autophagy, mitophagy, tumor microenvironment, tumor stem cells, tumor cell migration, invasion, metastasis
Graphical abstract
Autophagy is upregulated as tumors progress to become malignant and this review addresses how autophagy modulates cancer metastasis. Specifically, we examine the critical role played by autophagy in tumor cell migration and invasion, in maintaining the cancer stem cell phenotype and tumor cell dormancy as well as how autophagy affects interactions of tumor cells with components of the tumor microenvironment. Finally, we discuss how these functions make autophagy inhibition an attractive therapeutic option for metastatic cancers.
Autophagy & Cancer
The process of macro-autophagy involves the formation of double-membraned vesicles called autophagosomes, around the cellular content that is to be degraded, known as autophagic cargo [1] [2]. Such cargo typically includes organelles of all types, intra-cellular pathogens and protein aggregates. Induction of autophagy is controlled at the transcriptional and post-translational level (figure 1). Autophagy (Atg) genes are transcriptionally regulated by the ATF4 and MIT/TFE transcription factors [3, 4], and as such are induced by cellular stresses including amino acid deprivation and ER stress (ATF4) known to promote autophagy, as well as by other signals that inhibit mTOR, a negative regulator of MIT/TFE factors (figure 1)[4]. However, autophagy is primarily regulated in a post-translational manner to permit a rapid response to nutrient stress. The formation of autophagosomes is regulated by a pre-initiation complex involving the Ulk1/Ulk2 serine/threonine kinase that is sensitive to amino acid supply and cellular energy status, as a result of being regulated negatively by mTOR and positively by AMPK (figure 1) [5, 6]. As part of the pre-initiation complex with ATG13 and FIP200, Ulk1/2 phosphorylates Beclin1 to activate the lipid kinase activity of Vps34 (a class III PI3K), the catalytic component of the initiation complex, increasing phospho-inositol-3-phosphate (PIP3) production and recruiting further components of the autophagy machinery to drive the process forward (figure 1) [1]. A key consequence of increased initiation complex activity is the recruitment and activation of the conjugation complexes, including ATG5-ATG12/ATG16L that in turn recruits processed LC3 and related molecules to the expanding phagophore (figure 1) [1, 6]. Processed LC3 at nascent phagophores is key to selecting cargo for degradation and proteins, or their cargo adaptors, at cellular structures destined for degradation contain specific motifs known as LC3-interacting region (LIR) motifs that promote their interaction with LC3 (figure 1) [7, 8]. Closure of the phagophore to form a mature autophagosome requires LC3-related proteins leading to fusion with the lysosome resulting in degradation of autophagosomal cargo and recycling of constituent amino acids, nucleotides and fatty acids (figure 1) [1, 2, 9]. Novel non-canonical functions for autophagy proteins are emerging, including roles in anti-viral immunity, secretory processes and endocytic trafficking that are distinct from autophagy but rely on a select cadre of Atg gene products [10, 11]. Together with the increasing variety of cargo that autophagy seems capable of turning over, this has complicated our ability to dissect at a mechanistic level how autophagy influences disease processes, such as cancer. In particular, the pleiotropic nature of autophagy has made it challenging to determine whether autophagy is tumor-promoting, and therefore a valid therapeutic target, or indeed tumor-suppressive which might limit its value as a target in cancer treatment [12].
Figure 1. Autophagy overview.
Autophagy is exquisitely sensitive to amino acid deprivation in addition to other nutrient stresses. Amino acid deprivation induces AMPK and suppresses mTORC1 to promote Ulk1 kinase activity as part of the pre-initiation complex (Ulk1/FIP200/Atg13) that in turn activates the lipid kinase activity of the initiation complex (Beclin1/VPS34 and associated proteins). This is turn recruits the Atg12/Atg5/Atg16L conjugation complex that promotes processing and insertion of lipid conjugated LC3 into nascent phagophore membranes. LC3 and related proteins interact with cargo to be turned over via autophagy and also promotes closure of the autophagosome around the cargo. Fusion of these double-membraned autophagosomes with the lysosome results in proteolytic degradation of the cargo and release from the autophagolysosome of amino acids, nucleotides and lipids derived from degraded cargo. In addition to induction of autophagy by these post-translational mechanisms, autophagy is also stimulated by transcriptional control of Atg genes, notably by ATF4 and MiT/TFE transcription factors that are activated by amino acid deprivation amongst other stresses.
Much of the focus on the role of autophagy in cancer has been aimed at its role in tumor metabolism [9] where its ability to modulate mitochondrial turnover has been proposed as the main explanation for the altered metabolic phenotype in tumors forming in autophagy-deficient GEM models. Such models, including Ras-driven lung and pancreatic cancers [13–16], showed reduced fatty acid oxidation, a switch to Warburg metabolism and increased ROS, consistent with failure to degrade mitochondria in an appropriate manner [12, 17]. More recently, the role of autophagy in modulating how the tumor microenvironment sustains growing tumors has also come to the fore [18, 19]. Beyond its role in modulating tumor metabolism, autophagy performs other functions that likely contribute to its role in cancer and response to cancer therapy [12].
Metastasis is a multi-step process in which tumor cells at the primary tumor site undergo epithelial-to-mesenchymal transition (EMT) [20], acquire invasive and other properties that allow them to escape through the basement membrane into the vasculature, in a part of the metastatic cascade known as intravasation [21]. This is followed by selective pressure on escaped tumor cells to survive in the circulation where they sometimes become lodged, or if they are able to survive at all, use their invasive properties to extravasate at a distant site [21]. Frequently, such tumor cells alight in an environment that does not favor their growth and where they are again subject to evolutionary pressures to survive [22]. However, even if tumor cells survive such seemingly unfavorable conditions, they may not be able to expand and can remain dormant at such sites for years [22, 23]. Recently, it has emerged that primary tumors can release signals that mobilize other cell types to establish a pre-metastatic niche ahead of disseminating tumor cells landing there to promote the survival and outgrowth of metastatic lesions [24–26]. Throughout all these steps in the metastatic cascade, tumor cells also need to evolve the means to evade immune surveillance and T-cell mediated killing [27]. Intriguingly, many features of the metastatic tumor cell, whether that be mesenchymal properties or ability to escape immune surveillance, are associated with a stem-like phenotype and linked to drug resistance [20, 28]. Autophagy is up-regulated in primary human breast, glioblastoma, melanoma, esophageal cancer and hepatocellular carcinoma upon progression to advanced metastatic disease and expression of autophagy markers in these cancers is associated with poor prognosis [29–31] [32], highlighting novel and important roles for autophagy at different points in the metastatic cascade. Here, we review recent work defining new functions of autophagy in cancer metastasis including how autophagy promotes acquisition of pro-migratory and invasive properties, maintains tumor cell stemness and drug resistant phenotypes, and molds the co-evolution of tumors with their microenvironment.
Autophagic control of tumor cell migration and invasion
Increased motility is required for tumor cells to escape the primary tumor site and to successfully colonize secondary sites during metastasis [23, 33, 34]. This involves protrusion of the plasma membrane in the forward direction, adhesion of cellular integrins to the extra-cellular matrix (ECM) and induced signaling through focal adhesions [35, 36], polymerization of the actin cytoskeleton and contraction of the cell body [37], and finally detachment of the cell from the ECM behind the direction of movement [36]. Cells vary in how they migrate (amoeboid versus mesenchymal, for example) depending on cell type and microenvironment [36, 38], but all cell migration involves exquisite coordination of the processes described above, in some fashion. Over the past several years, increasing evidence has emerged to demonstrate a direct role for autophagy in tumor cell motility and invasion during metastasis [39–42], including through turnover of components of the cell migration machinery [43, 44] and ECM proteins [45], as well as other roles such as modulation of the tumor cell secretome [46].
Various studies have demonstrated a key role for autophagy in focal adhesion dynamics during cell migration and invasion [42–44] [47–49]. Inhibition of focal adhesion kinase (FAK), such as occurs during cell detachment from the sub-stratum, resulted in autophagic degradation of active SRC kinase. This required c-CBL, a SRC binding protein and E3 ubiquitin ligase that contains a LIR motif and interacts directly with processed LC3B to promote autophagic turnover of SRC [42]. SRC turnover by autophagy also required Ambra1, a multi-functional protein known as an inhibitor of the Beclin1-VPS34 initiation complex but that alternatively can interact with FAK to limit active SRC at focal adhesions and promote SRC degradation by autophagy [49]. Inhibition of autophagy restored SRC expression at focal adhesions in adhesion-stressed cells but was associated with cell death [42] indicating that autophagic turnover of SRC was required in response to cell detachment or FAK inactivation (figure 2). Interestingly, FAK inhibition increased autophagic flux suggesting that FAK activity inhibits autophagy, possibly by sequestering or otherwise interfering with FAK-interacting protein of 200 kD (FIP200) that although first identified as a protein that interacts with and inhibits FAK kinase activity [50], has subsequently been identified as the mammalian homologue of yeast autophagy gene Atg17 (Autophagy related gene 17) and a key component of the pre-initiation complex in mammals that interacts with ULK1 and ATG13 (figure 1) [51]. FAK may also inhibit autophagy through its inhibitory interaction with Tuberous Schlerosis Complex 2 (TSC2) resulting in activation of mTOR (figure 2)[52, 53]. It will be important in future studies to determine whether FAK requires FIP200, TSC2 or other signaling mechanisms to modulate autophagy and to assess the extent to which FIP200 mediates cross-talk between autophagy and cell migration.
Figure 2. Coordinate control of autophagy and cell migration.
Autophagy and cellular motility are coordinated at the molecular level through reciprocal control of both processes by common modulators including Focal adhesion kinase (FAK), FAK-Interacting Protein 200 kD (FIP200) and Paxillin (PXN). Autophagy promotes cell motility through turnover of focal adhesion complexes and specifically via degradation of focal adhesion component, Paxillin (PXN). Autophagy also promotes turnover of SRC in response to FAK inhibition to prevent cell death in response to cell detachment. Both FAK (through TSC2) and PXN (through ill-defined mechanisms) feed back to modulate autophagic flux. Similarly, there is a reciprocal interaction between autophagy and members of the Rho family of small G proteins involved in cell migration. Autophagy promotes Rho turnover via p62/Sqstm1 while ROCK1 that acts downstream of Rho stimulates autophagy through phosphorylation of Beclin1.
Recent work from our laboratory demonstrated a critical role for autophagy in the motility and invasion of metastatic tumor cells in vitro and for tumor metastasis in vivo [44]. Inhibition of autophagy reduced tumor cell motility due to decreased focal adhesion disassembly. This was attributed to accumulation of Paxillin (PXN), a core component of focal adhesions [44, 48] and PXN was identified as a LC3-interacting protein that contains a conserved LIR motif (figure 2) [44]. The interaction between PXN and LC3B was promoted by oncogenic SRC and required the Y40 residue at position +1 of the LIR motif in PXN [44], a site previously identified as a target of SRC phosphorylation [54]. Consistently, the ability of oncogenic SRC to promote cell motility and invasion was dependent on phosphorylation of Y40, interaction of PXN with LC3 and functional autophagy (figure 2) [44]. The targeting of PXN for autophagic degradation in the highly metastatic tumor cells studied did not require either of the cargo adaptors p62/Sqstm1 or Near BRCA1 (NBR1) [44] but a different mechanism may be at play in other cell types since in Ras-transformed MCF10A breast epithelial cells, focal adhesion turnover by autophagy was specifically dependent on NBR1 (figure 2) [43]. In addition, c-CBL has also been reported to be required for targeting PXN to autophagosomes for degradation [48], in addition to its role in promoting SRC turnover [42]. Similar to FAK that is both a regulator of autophagy and regulated by autophagy, PXN is required for efficient autophagosome formation in MEFs [55], is phosphorylated by Ulk1 and along with vinculin relocates from focal adhesions to autophagosomes in response to nutrient deprivation [55]. These studies highlight a critical role for autophagy in focal adhesion dynamics in tumor cells and a reciprocal role for focal adhesion components in modulating autophagy.
An intriguing reciprocal relationship also exists between control of the Rho family of small GTPases and autophagy during cell migration. RhoA, Rac1 and CDC42 GTPases modulate cell motility by promoting formation of membrane protrusions, lamellopodia and filopodia respectively [36, 56, 57]. The ability of rho1 to induce hemocyte migration during wound healing in Drosophila was dependent on atg1 (autophagy related gene-1) and ref(2)P the fly homologue of cargo adaptor p62/sqstm1 [40]. Chemical inhibition of autophagy prevented blood cell migration to larval wound sites in flies while knockdown of beclin1 or ulk1 prevented mouse macrophages spreading in response to inflammatory signals [40]. p62/Sqstm1 has since been shown to target mammalian RhoA to the autophagosome for degradation [58] with the failure to turn over RhoA in cells knocked down for ATG5 resulting in RhoA build-up at the midbody during mitosis, cytokinesis defects and aneuploidy [58]. Conversely, Rho signaling has been implicated in the regulation of autophagy [59, 60] with Rho-associated kinase 1 (ROCK1) identified as a regulator of starvation-induced but not basal autophagy [59]. Inhibition of ROCK1 resulted in the formation of enlarged, immature autophagosomes leading the authors to suggest that ROCK1 promotes autophagy by limiting time spent in early phagophore elongation phases of autophagy [60]. ROCK1 is also activated by amino acid deprivation leading to direct phosphorylation of Beclin1 by ROCK1 on Thr119 causing disruption of the Beclin1/Bcl-2 complex resulting in derepression of autophagy (figure 2) [61, 62]. Meanwhile, Rac1 plays a role in modulating Rab7, a different small GTPase that promotes maturation of late stage autophagosomes and lysosomal fusion [63]. The efficient cycling of Rab7 GTPase activity as required for autophagolysosome maturation in response to amino acid starvation is controlled by armus, a RabGAP that localizes to autophagosomes through direct interaction with LC3 [64, 65]. Rac1 also interacts with armus and promotes autophagic recycling of E-Cadherin during EGF-stimulated cell scattering [64]. Failure to inactivate Rac1 during amino acid starvation blocked autophagy due to Rac1 binding to LC3 interfering with the interaction of armus with LC3, thereby preventing Rab7 localization to maturing autophagolysosomes [65]. Finally, RhoA, Rac1 and CDC42 were also identified within an autophagy-centered human gene interaction network built on known autophagic responses to microenvironmental cues (figure 2) [66]. These and other studies highlight yet again how cell migration is molecularly coordinated with control of autophagy and vice versa
Autophagy in Tumor Stem cells and Metastatic Dormancy
Autophagy is required for normal tissue stem cell maintenance and differentiation [67–69] with hematopoietic stem cells dependent on autophagy for survival [70, 71] and muscle stem cells dependent on autophagy to prevent senescence [72]. More specifically, mitophagy has been directly implicated in preventing stem cell aging and promoting stem cell self-renewal [73–75] such that defective mitophagy in mammary stem cells inhibited preferential segregation of younger mitochondria to daughter stem cells and older mitochondria to daughter non-stem cells [73]. The requirement for mitophagy in the self-renewal of hematopoietic stem cells has been linked to elimination of metabolically active mitochondria thereby maintaining HSCs in a glycolytic state with low levels of oxidative metabolism [76–78]. The balance between glycolysis and oxidative metabolism is key to determining whether stem cells remain quiescent or undergo differentiation [74, 79–81]. By limiting mitochondrial mass through mitophagy and stimulating glycolysis while limiting oxidative metabolism, stem cells can maintain their slow cycling, self-renewing state (figure 3) [80]. By contrast, inhibition of glycolysis and enhancement of mitochondrial respiration promotes differentiation that involves mitochondrial remodeling, reduced mitophagy, dispersed cytoplasmic localization, and increased expression of enzymes involved in the TCA cycle and the electron transport chain [79, 81–84].
Figure 3. Requirement for autophagy in stem cell maintenance.
Autophagy is required for maintenance of a stem-like phenotype both in normal tissue stem cells but also in certain types of cancer stem cells. Stresses (hypoxia, TGF-β) that induce stem-like properties, including self-renewal capability, EMT, up-regulation of CD44, also induce autophagy that is required for many of these stem-like features, including up-regulation of CD44. Recent work has highlighted a specific role for mitophagy in promoting a stem-like state in cancers, possibly by limiting ROS production and promoting glycolytic metabolism that may limit differentiation.
Cancer stem cells (CSCs) are defined functionally as those relatively rare cells in a tumor that have the capacity to self-renew and to differentiate to regenerate all aspects of tumor heterogeneity [23, 85, 86]. Various reports have correlated the expression of CSC markers, such as CD44, with increased invasiveness and metastasis [87–91]. Indeed, stresses such as hypoxia and TGF-β that promote epithelial to mesenchymal transition (EMT) and a more motile tumor cell phenotype, also induce features of CSCs, including increased self-renewal and upregulation of CD44 (figure 3) [34, 85, 86, 92–94]. Significantly, stresses such as hypoxia and TGF-β also induce autophagy, alongside EMT and stemness [32, 95] and numerous studies have now linked increased autophagy to maintenance of CSCs (figure 3) [29–32, 96]. In human breast ductal carcinoma in situ (DCIS), increased autophagy was detected in those rare tumor cells that showed tumor-initiating capacity and migratory ability and CD44+ CSCs were dependent on autophagic flux for their ability to form mammospheres in vitro and tumors in vivo [29, 96–98]. In esophageal squamous cell carcinoma (ESCC), autophagy was required for the upregulation of CD44 that accompanied induction of EMT (figure 3) [30]. Here again, it was mitophagy that was specifically up-regulated in ESCC cells undergoing EMT [30], possibly in response to increased ROS production and membrane depolarization that is known to activate Parkin-dependent mitophagy [99]. Inhibition of Parkin-mediated mitophagy attenuated up-regulation of CD44 and led to cell death [30]. Similar to what has been described for pluripotent stem cells, reduced mitochondrial mass has also been reported to distinguish CSCs from non-CSCs (figure 3) [100, 101]. However, quiescent PDAC cells with CSC properties were recently described as relying on oxidative respiration to maintain their stem cell state and were less dependent on glycolysis than proliferating cells in the bulk of the tumor [102]. Clearly, the role of mitophagy in CSCs needs to be addressed more directly to determine whether reduced mitochondrial mass arising from increased mitophagy in response to EMT, cancer therapy or other challenges, is indeed required to establish and/or maintain tumor stem cell properties.
The “re-awakening” of tumor cells at distant sites leading to outgrowth of macrometastatic disease many years after primary tumors were “successfully” treated has led to the concept of metastatic dormancy [22, 23, 103, 104]. Various studies have shown a role for autophagy in promoting tumor cell survival during tumor dormancy [105]. For example, inhibition of autophagy blocked re-emergence of ovarian tumors after dormancy induced by the ARHI tumor suppressor [105]. Pre-clinical studies also showed that autophagy inhibition in the Eμ-Myc mouse model of B-cell lymphoma following treatment with alkylating agents blocked tumor recurrence validating a role for autophagy in promoting both tumor dormancy and drug resistance [106]. Autophagy may promote the dormancy of disseminated tumor cells simply by supplying key amino acids and other nutrients or alternatively, as discussed above, autophagy may play a more instructive role by eliminating mitochondria, modulating redox balance and actively promoting the stem cell state [22, 107, 108]. Indeed, CSC markers are up-regulated on disseminated tumor cells in the bone marrow of breast cancer patients [109] and such observations have led to the suggestion that dormant tumor cells are in fact CSCs that depend on autophagy to survive at secondary sites over extended periods of time and expand later as metastatic lesions made up of both CSCs and non-stem tumor cells representing the full heterogeneity of rapidly growing tumors [22, 105]. If as suggested, dormant tumor cells are preferentially reliant on autophagy for survival, then this provides a rationale for combining autophagy inhibition with conventional therapeutic responses to eliminate dormant tumor cells and prevent subsequent metastatic outgrowth.
Autophagy contributes to the therapy resistance of numerous cancers, including resistance to conventional genotoxic therapies [110, 111], prostate cancer to androgen ablation therapy [112], breast cancer to SERMs [113–115], GIST to Imatinib [116, 117], lung cancer to tyrosine kinase inhibitors [118–120], glioblastoma to temozolomide [121], myeloma to bortezomib [122, 123], melanoma and brain cancers to B-Raf inhibitors [124–126], as well as resistance of various cancers to PI3K inhibitors [127]. Thus, there is a compelling rationale to combine these therapeutic approaches with agents that inhibit autophagy, such as chloroquine, an anti-malarial drug that inhibits autophagy by increasing the pH of the lysosome and blocking lysosomal proteases [128, 129]. Early clinical trials combining chloroquine with more conventional therapies, such as temozolomide for glioblastoma treatment showed promise with autophagy inhibition more than doubling survival times [128, 130, 131]. One particularly successful trial applying chloroquine to conventional doxorubicin treatment of non-Hodgkins lymphoma in dogs demonstrated both that effective chloroquine doses were well-tolerated but also improved overall drug response and progression-free survival [132]. More recent studies have reported efficacy of adjuvant chloroquine treatment in clinical trials for an extended range of human cancers, as reviewed in more detail elsewhere [128, 129].
However, the ability to target autophagy in cancer has also been limited by the high doses of agents such as chloroquine required to elicit an effect in humans [128, 129, 133], the unpredictable effects of lysosomal inhibitors used to block autophagy on the activity of mTORC1 that is regulated at the lysosome also [134, 135], the unknown off-target effects of compounds such as chloroquine [128] and the possible adverse consequences of systemic autophagy inhibition [136]. An additional hurdle to assessing the efficacy of autophagy inhibition in cancer treatment is the lack of reliable markers of autophagy in human tumors [129]. Nevertheless, the development of improved derivatives of chloroquine that are efficacious at lower doses [133] and simultaneously target both autophagy and mTORC1 activity at the lysosome [137], in addition to development of targeted inhibitors of ULK1 [138, 139], VPS34 [140] and other enzymes required for autophagy [141] holds significant promise for our ability to overcome the tumor-promoting effects of autophagy in cancer therapy resistance and metastatic dormancy [12].
Autophagy in the tumor microenvironment
Cancer cells co-evolve with their tumor microenvironment and the role of autophagy in modulating how the cancer cell interacts with other cell types in the surrounding milieu is emerging as a key topic in determining whether autophagy inhibition is likely to be effective in cancer treatment [129]. How autophagy modulates the interaction of tumor cells with components of both the innate and adaptive immune systems is particularly complex, as has been reviewed extensively elsewhere [142, 143]. Autophagy promotes the release of damage-associated molecular patterns (DAMPs) and ATP from dying tumor cells thereby recruiting CD8+ cytotoxic T lymphocytes (CTLs) that synergize with conventional therapeutics to eliminate cancers [144, 145]. Autophagy also promotes tumor immune surveillance through trafficking of tumor-antigens through the lysosome for cross-presentation on dendritic cells and by suppressing infiltration of tumor-promoting FoxP3+ T-regulatory cells [145, 146]. These anti-tumor activities of autophagy in eliciting an immune response would argue that inhibiting autophagy is not justified in in combination with conventional cancer therapy. However, the role of autophagy in modulating tumor immunity may be context dependent with other studies using less immunogenic tumor models indicating that anti-tumor immunity is not adversely affected by autophagy inhibition [147] and that in fact autophagy promotes escape from immune surveillance in some models [148, 149] possibly through indirect mechanisms relying on the role of autophagy in promoting EMT and secretion of immune-modulatory cytokines [150]. A recent study has identified a novel role for autophagy in suppressing the anti-tumor activity of certain types macrophages in a mouse model of pancreatic ductal carcinoma (PDAC) [151]. Inhibiting autophagy through inducible expression of a dominant negative Atg4B allele (Atg4BCA) decreased PDAC tumor growth and induced tumor regression of already formed tumors in an autochtonous model of PDAC [151]. However, when autophagy was similarly inhibited in an orthotopic transplant model using primary tumor cells from the autochtonous model, tumor growth was inhibited but tumor regression of existing tumors was not observed [151]. Inhibiting autophagy by expression of Atg4BCA caused macrophage accumulation in autochtonous PDAC tumors while depletion of macrophages prevented tumor regression induced by autophagy inhibition without affecting rates of tumor cell growth. These findings indicate strongly that autophagy plays both a cell intrinsic role promoting tumor cell growth but also has a non-cell autonomous effect suppressing the anti-tumor activities of macrophages [151]. It was not determined how autophagy inhibition resulted in macrophage recruitment to the tumor although production of DAMPs by dying tumor cells as a result of autophagy inhibition could provide an explanation. As reviewed elsewhere [142] [143], further investigation is required to clarify when and how autophagy modulates the interaction of tumor cells with immune cells in the tumor microenvironment.
Several recent reports have illuminated how autophagy can be induced in certain other non-tumor cells that make up the tumor microenvironment to promote tumor cell growth and invasion [18, 152]. Examination of the interaction between pancreatic ductal adenocarcinoma (PDAC) cells and pancreatic stellate cells (PSCs), a specialized type of fibroblast that makes up a large part of the desmoplastic microenvironment in PDAC, revealed that autophagy was up-regulated specifically in PSCs when grown with pancreatic tumor cells but not when co-cultured with normal ductal epithelia. This in turn promoted protein catabolism in PSCs generating alanine to fuel the TCA cycle, oxygen consumption and lipid synthesis in the adjacent tumor cells (figure 4) [18]. The effects of increased alanine uptake by tumor cells and its conversion to pyruvate to fuel TCA cycle and lipid synthesis allowed glucose-derived carbon to be more efficiently used to promote serine and glycine biosynthesis with knock-on effects for nucleotide production. These effects of co-culture of tumor cells with PSCs or PSC-conditioned media was blocked by inhibiting autophagy in PSCs or by knocking down the GPT1 alanine transaminase (that converts alanine to pyruvate) in tumor cells. The growth stimulatory effects of PSCs on PDAC growth were observed both in vitro and in vivo in both subcutaneous and orthotopic mouse models using human PDAC cells and both human and mouse PSCs [18]. Consistent with a role in promoting tumor growth, there was selection against expression of the autophagy inhibitory activity of Atg4BCA in PSCs in an autochtonous mouse model of PDAC [151].
Figure 4. Autophagy in the tumor microenvironment supports tumor cell growth through supply of nutrients and other factors.
The growth of pancreatic ductal adenocarcinoma (PDAC) is supported by a highly complex and dense desmoplastic stroma that includes pancreatic stellate cells (PSCs). Autophagy plays a critical role in the tumor-promoting activity of PSCs that supply associated PDAC tumor cells with the breakdown products of autophagy, including amino acids such as alanine. Inhibition of alanine metabolism in tumor cells neutralized the pro-tumor activity of PSCs. Autophagy is induced in PSCs by their association with PDAC cells although how tumor cells signal to PSCs to induce autophagy is not yet clear. Autophagy in PSCs was also required for production of pro-inflammatory cytokines such as IL-6, that may be feeding back to both promote tumor cell motility and mobilization of metabolites systemically in the animal, but also may also contribute to the signal that induces autophagy in PSCs in the first place. Additional metabolites and factors may also contribute to additional aspects of how autophagy in the tumor microenvironment promotes tumor growth and metastasis.
Complementary work highlighted the role played by autophagy in activating PSCs to produce pro-migratory and invasive cytokines such as IL-6, in addition to extracellular matrix proteins [153]. Conversely, inhibition of autophagy in PSCs resulted in adoption of a quiescent state, reduced expression of IL-6 and ECM proteins, attenuated migration/invasion properties and reduced metastasis to the liver of co-injected PDAC cells in an orthotopic mouse model (figure 4) [153]. Autophagy is known to promote secretion of IL-6 and other factors [46, 154, 155] and thus inhibiting autophagy as a means to limit tumor cell migration and invasion, in addition to tumor cell growth is well-justified.
In pancreatic cancer, the desmoplastic reaction that is dependent on PSC activation is a barrier to therapy both by inducing hypoxia and stem-like phenotypes in the tumor cells, and by physically preventing access of the vasculature and drugs to the tumor [156]. Efforts to improve delivery of drugs to PDAC tumors, by degrading the desmoplastic “fortress” around these tumors [157], have failed and this has been attributed to selection for more aggressive tumor cells and increased vasculature leading to easier egress of Tregs, reduced tumor immune surveillance and easier escape of more aggressive mesenchymal tumor cells into the periphery [158, 159]. Thus efforts to limit autophagy in PSCs may provide a more promising alternative in PDAC treatment since it would both limit the ability of PSCs to supply tumor cells with nutrients such as alanine [18], as well as pro-invasive factors, such as IL-6 [153] but importantly autophagy inhibition would also be predicted to limit acquisition of stem-like, drug-resistant features and other malignant properties by the tumor itself, as described above (figure 3).
Genetic analysis of the role of autophagy in tumor growth in Drosophila has further emphasized the significance of autophagy in the tumor microenvironment [152]. Here, it was shown that autophagy is up-regulated systemically in flies bearing malignant RasV12;scrib−/− tumors in their eye-antennal imaginal discs but not with benign RasV12 tumors [152]. Ablation of autophagy in the tumor had only a modest effect on tumor growth but inhibition of autophagy in either the non-tumor epithelia surrounding the tumor or in the entire animal had a marked effect on both tumor growth and invasiveness [152]. Indeed, transplant of quiescent RasV12;scrib−/−;Atg13−/− autophagy deficient tumors into autophagy competent host flies could rescue tumor growth while transplant into autophagy deficient host flies could not [152]. In this model, induction of autophagy in the tumor microenvironment, which was referred to as non-cell-autonomous autophagy (NAA) was independent of tumor growth since dominant negative PI3K blocked tumor cell growth but the tumor was still able to induce autophagy in neighboring non-tumor cells. Intriguingly, the ability of the tumor to induce NAA was dependent on tumor-specific activation of TNF, JAK/STAT, JNK and Hippo pathway signaling via Yorkie [152] with Yorkie transcriptional targets upd1 and upd3 (both IL-6 like cytokines in flies) able to induce NAA [152]. The Yorkie target ImpL2, an insulin/IGF antagonist secreted by over-proliferating tissues was previously shown to cause systemic tissue wasting in flies [160, 161] but was not required to induce NAA in flies with RasV12;scrib−/− tumors [152]. In line with the work from Kimmelman and colleagues [18], silencing of the amino acid transporter slimfast in tumors, reduced tumor growth although uptake of a specific amino acid or other possible mediators of the non-cell-autonomous/autophagy-dependent signal feeding tumor cells was not identified [152].
These various studies collectively demonstrate that malignant tumors induce autophagy in non-tumor cells making up their microenvironment in a manner that further promotes tumor growth and progression (figure 4). Intriguingly, the fly studies suggest that tumor growth can induce autophagy systemically in tissues throughout the animal and it remains to be determined which factors drive this phenomenon but likely signals that circulate, again perhaps IL-6. Critically, these reports demonstrate a key role for autophagy in non-tumor cells in producing metabolites that fuel tumor cell growth. In the case of mammalian pancreas cancer studies, this appears to be driven by autophagy-dependent production of alanine from associated non-tumor cells that is taken up by the tumor and fed into the TCA cycle and lipid biosynthetic pathways amongst other [18] with a similar mechanism identified in flies where slimfast was required for the positive effects of NAA on tumor growth [152]. Production of alanine by cancer-associated fibroblasts in PDAC predominantly affected tumor growth rate [18] but autophagy in non-tumor cells also drove tumor cell invasion and metastasis [152, 153], perhaps through production of pro-migratory cytokines, such as IL-6.
These studies open up new perspectives on the role of autophagy in cancer progression and metastasis. Particularly intriguing is the possibility that diet and metabolic stresses inducing autophagy in distant organs could influence tumor growth and metastasis at another site or indeed that tumor growth may influence systemic metabolism by promoting autophagy in key organs, such as the liver. Also, there is the possibility that other metabolites in addition to alanine, produced by protein catabolism during autophagy in non-tumor tissue may play a signaling or metabolic role in promoting tumor progression and metastasis. These are all avenues of future investigation.
Acknowledgments
This work was supported by NIH/NCI RO1 CA162405 (KFM), NIH/NCI T32 CA009594 (MNS), NIH/GM T32 GM007281 (EEM).
Abbreviations
- AMPK
adenosine monophosphate activated kinase
- ATG/Atg
human/mouse autophagy gene
- ATF4
activating transcription factor 4
- ATP
adenosine triphosphate
- CBL
Casitas B-lineage lymphoma gene
- CD44
cell differentiation antigen 44
- CSC
cancer stem cell
- CTL
cytotoxic T lymphocyte
- DAMP
damage associated molecular pattern
- DCIS
ductal carcinoma in situ
- ECM
extracellular matrix
- EMT
epithelial-mesenchymal transition
- ER
endoplasmic reticulum
- FAK
focal adhesion kinase
- FIP200
FAK interacting protein 200 kD
- GPT1
glutamate-pyruvate transaminase-1
- HSC
hematopoietic stem cell
- IL-6
interleukin 6
- JAK
janus kinase
- JNK
c-Jun N-terminal kinase
- LC3
microtubule associated light chain 3
- MIT/TFE
microphthalmia-associated transcription factor/transcription factor E3
- PDAC
pancreatic ductal carcinoma
- PSC
pancreatic stellate cell
- PXN
paxillin
- ROS
reactive oxygen species
- SERM
selective estrogen receptor modulator
- SRC
sarcoma oncoprotein encoded by c-SRC
- STAT
signal transducers and activators of transcription
- TCA
tricarboxylic acid
- TGF-β
transforming growth factor beta
- TNF
tumor necrosis factor
- mTOR
mammalian target of rapamycin
- mTORC1
mammalian target of rapamycin complex 1
- ULK1
Unc51-like kinase-1
- VPS34
Vacuolar protein sorting-associated protein 34
Footnotes
Author Contributions
All authors reviewed the literature, discussed the key topics to include and contributed to all sections of the manuscript.
References
- 1.Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147:728–41. doi: 10.1016/j.cell.2011.10.026. [DOI] [PubMed] [Google Scholar]
- 2.Kroemer G, Marino G, Levine B. Autophagy and the integrated stress response. Molecular cell. 2010;40:280–93. doi: 10.1016/j.molcel.2010.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rouschop KM, van den Beucken T, Dubois L, Niessen H, Bussink J, Savelkouls K, Keulers T, Mujcic H, Landuyt W, Voncken JW, Lambin P, van der Kogel AJ, Koritzinsky M, Wouters BG. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. The Journal of clinical investigation. 2010;120:127–41. doi: 10.1172/JCI40027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, Lengrand J, Deshpande V, Selig MK, Ferrone CR, Settleman J, Stephanopoulos G, Dyson NJ, Zoncu R, Ramaswamy S, Haas W, Bardeesy N. Transcriptional control of the autophagylysosome system in pancreatic cancer. Nature. 2015;524:361–5. doi: 10.1038/nature14587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor RC, Asara JM, Fitzpatrick J, Dillin A, Viollet B, Kundu M, Hansen M, Shaw RJ. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. 2011;331:456–61. doi: 10.1126/science.1196371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Carlsson SR, Simonsen A. Membrane dynamics in autophagosome biogenesis. J Cell Sci. 2015;128:193–205. doi: 10.1242/jcs.141036. [DOI] [PubMed] [Google Scholar]
- 7.Weidberg H, Shvets E, Elazar Z. Biogenesis and Cargo Selectivity of Autophagosomes. Ann Rev Biochem. 2011;80:125–56. doi: 10.1146/annurev-biochem-052709-094552. [DOI] [PubMed] [Google Scholar]
- 8.Rogov V, Dotsch V, Johansen T, Kirkin V. Interactions between autophagy receptors and ubiquitin-like proteins form the molecular basis for selective autophagy. Mol Cell. 2014;53:167–78. doi: 10.1016/j.molcel.2013.12.014. [DOI] [PubMed] [Google Scholar]
- 9.Kimmelman AC, White E. Autophagy and Tumor Metabolism. Cell Metab. 2017;25:1037–1043. doi: 10.1016/j.cmet.2017.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Green DR, Levine B. To Be or Not to Be? How Selective Autophagy and Cell Death Govern Cell Fate. Cell. 2014;157:65–75. doi: 10.1016/j.cell.2014.02.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature. 2011;469:323–35. doi: 10.1038/nature09782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Amaravadi R, Kimmelman AC, White E. Recent insights into the function of autophagy in cancer. Genes & development. 2016;30:1913–30. doi: 10.1101/gad.287524.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, Karsli-Uzumbas G, Kamphorst JJ, Chen G, Lemmons JMS, Karantza V, Coller HA, DiPaola RS, Gelinas C, Rabinowitz JD, White E. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes & Dev. 2011;25:460–70. doi: 10.1101/gad.2016311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Guo JY, Karsli-Uzunbas G, Mathew R, Aisner SC, Kamphorst JJ, Strohecker AM, Chen G, Price S, Lu W, Teng X, Snyder E, Santanam U, Dipaola RS, Jacks T, Rabinowitz JD, White E. Autophagy suppresses progression of K-ras-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes & Dev. 2013;27:1447–61. doi: 10.1101/gad.219642.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Strohecker AM, Guo JY, Karsli-Uzunbas G, Price SM, Chen GJ, Mathew R, McMahon M, White E. Autophagy sustains mitochondrial glutamine metabolism and growth of BrafV600E-driven lung tumors. Cancer Discov. 2013;3:1272–85. doi: 10.1158/2159-8290.CD-13-0397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Rosenfeldt MT, O'Prey J, Morton JP, Nixon C, MacKay G, Mrowinska A, Au A, Rai TS, Zheng L, Ridgway R, Adams PD, Anderson KI, Gottlieb E, Sansom OJ, Ryan KM. p53 status determines the role of autophagy in pancreatic tumour development. Nature. 2013;504:296–300. doi: 10.1038/nature12865. [DOI] [PubMed] [Google Scholar]
- 17.Zong WX, Rabinowitz JD, White E. Mitochondria and Cancer. Molecular cell. 2016;61:667–76. doi: 10.1016/j.molcel.2016.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Sousa CM, Biancur DE, Wang X, Halbrook CJ, Sherman MH, Zhang L, Kremer D, Hwang RF, Witkiewicz AK, Ying H, Asara JM, Evans RM, Cantley LC, Lyssiotis CA, Kimmelman AC. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature. 2016;536:479–83. doi: 10.1038/nature19084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Maes H, Rubio N, Garg AD, Agostinis P. Autophagy: shaping the tumor microenvironment and therapeutic resposne. Trends Mol Med. 2013;19:428–46. doi: 10.1016/j.molmed.2013.04.005. [DOI] [PubMed] [Google Scholar]
- 20.Chaffer CL, San Juan BP, Lim E, Weinberg RA. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 2016;35:645–654. doi: 10.1007/s10555-016-9648-7. [DOI] [PubMed] [Google Scholar]
- 21.Lambert AW, Pattabiraman DR, Weinberg RA. Emerging Biological Principles of Metastasis. Cell. 2017;168:670–691. doi: 10.1016/j.cell.2016.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sosa MS, Bragado P, Aguirre-Ghiso JA. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat Rev Cancer. 2014;14:611–22. doi: 10.1038/nrc3793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.McGowan PM, Kirstein JM, Chambers AF. Micrometastatic disease and metastatic outgrowth: clinical issues and experimental approaches. Future Oncol. 2009;5:1083–98. doi: 10.2217/fon.09.73. [DOI] [PubMed] [Google Scholar]
- 24.Kaplan RN, Rafii S, Lyden D. Preparing the "Soil": the pre-metastatic niche. Cancer Res. 2006;66:11089–93. doi: 10.1158/0008-5472.CAN-06-2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Sceneay J, Smyth MJ, Moller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64. doi: 10.1007/s10555-013-9420-1. [DOI] [PubMed] [Google Scholar]
- 26.Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, Soplop N, Uryu K, Pharmer L, King T, Bojmar L, Davies AE, Ararso Y, Zhang T, Zhang H, Hernandez J, Weiss JM, Dumont-Cole VD, Kramer K, Wexler LH, Narendran A, Schwartz GK, Healey JH, Sandstrom P, Labori KJ, Kure EH, Grandgenett PM, Hollingsworth MA, de Sousa M, Kaur S, Jain M, Mallya K, Batra SK, Jarnagin WR, Brady MS, Fodstad O, Muller V, Pantel K, Minn AJ, Bissell MJ, Garcia BA, Kang Y, Rajasekhar VK, Ghajar CM, Matei I, Peinado H, Bromberg J, Lyden D. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–35. doi: 10.1038/nature15756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–330. doi: 10.1038/nature21349. [DOI] [PubMed] [Google Scholar]
- 28.Mowers EE, Sharifi MN, Macleod KF. Autophagy in cancer metastasis. Oncogene. 2017;36:1619–1630. doi: 10.1038/onc.2016.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Espina V, Mariani BD, Gallagher RI, Tran K, Banks S, Wiedemann J, Huryk H, Mueller C, Adamo L, Deng J, Petricoin EF, Pastore L, Zaman S, Menezes G, Mize J, Johal J, Edmiston K, Liotta LA. Malignant precursor cells pre-exist in human breast DCIS and require autophagy for survival. PLoS One. 2010;5:e10240. doi: 10.1371/journal.pone.0010240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Whelan KA, Chandramouleeswaran PM, Tanaka K, Natsuizaka M, Guha M, Srinivasan S, Darling DS, Kita Y, Natsugoe S, Winkler JD, Klein-Szanto AJ, Amaravadi RK, Avadhani NG, Rustgi AK, Nakagawa H. Autophagy supports generation of cells with high CD44 expression via modulation of oxidative stress and Parkin-mediated mitochondrial clearance. Oncogene. 2017;36:4843–4858. doi: 10.1038/onc.2017.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Galavotti S, Bartesaghi S, Faccenda D, Shaked-Rabi M, Sanzone S, McEvoy A, Dinsdale D, Condorelli F, Brandner S, Campanella M, Grose R, Jones C, Salomoni P. The autophagy-associated factors DRAM1 and p62 regulate cell migration and invasion in glioblastoma stem cells. Oncogene. 2013;32:699–712. doi: 10.1038/onc.2012.111. [DOI] [PubMed] [Google Scholar]
- 32.Li J, Yang B, Zhou Q, Wu Y, Shang D, Guo Y, Song Z, Zheng Q, Xiong J. Autophagy promotes hepatocellular carcinoma cell invasion through activation of epithelial-mesenchymal transition. Carcinogenesis. 2013;34:1343–51. doi: 10.1093/carcin/bgt063. [DOI] [PubMed] [Google Scholar]
- 33.Langley RR, Fidler IJ. The seed and soil hypothesis revisited--the role of tumor-stroma interactions in metastasis to different organs. Int J Cancer. 2011;128:2527–35. doi: 10.1002/ijc.26031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.May CD, Sphyris N, Evans KW, Werden SJ, Guo W, Mani SA. Epithelial-mesenchymal transition and cancer stem cells: a dangerously dynamic duo in breast cancer progression. Breast cancer research : BCR. 2011;13:202. doi: 10.1186/bcr2789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Parsons JT, Horwitz AF, Schwartz MA. Cell adhesion: integrating cytoskeletal dynamics and cellular tension. Nat Rev Mol Biol. 2010;11:633–43. doi: 10.1038/nrm2957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ridley AJ. Life at the leading edge. Cell. 2011;145:1012–22. doi: 10.1016/j.cell.2011.06.010. [DOI] [PubMed] [Google Scholar]
- 37.Olson MF, Sahai E. The actin cytoskeleton in cancer cell motility. Clin & Exp Metastasis. 2009;26:273–87. doi: 10.1007/s10585-008-9174-2. [DOI] [PubMed] [Google Scholar]
- 38.Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147:992–1009. doi: 10.1016/j.cell.2011.11.016. [DOI] [PubMed] [Google Scholar]
- 39.Kenific CM, Thorburn A, Debnath J. Autophagy and metastasis: another double-edged sword. Curr Op Cell Biol. 2010;22:1–5. doi: 10.1016/j.ceb.2009.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kadandale P, Stender JD, Glass CK, Kiger AA. Conserved role for autophagy in Rho1-mediated coritcal remodeling and blood cell recruitment. Proc Natl Acad Sci U S A. 2010;107:10502–07. doi: 10.1073/pnas.0914168107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mackintosh RL, Timpson P, Thorburn J, Anderson KI, Thornburn A, Ryan KM. Inhibition of autophagy impairs tumor cell invasion in an organotypic model. Cell Cycle. 2012;11:2022–29. doi: 10.4161/cc.20424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Sandilands E, Serrels B, McEwan DG, Morton JP, Macagno JP, McLeod K, Stevens C, Brunton VG, Langdon WY, Vidal M, Sansom OJ, Dikic I, Wilkinson S, Frame MC. Autophagic targeting of Src promotes cancer cell survival following reduced FAK signalling. Nat Cell Biol. 2011;14:51–60. doi: 10.1038/ncb2386. [DOI] [PubMed] [Google Scholar]
- 43.Kenific CM, Stehbens SJ, Goldsmith J, Leidal AM, Faure N, Ye J, Wittmann T, Debnath J. NBR1 enables autophagy-dependent focal adhesion turnover. J Cell Biol. 2016;212:577–90. doi: 10.1083/jcb.201503075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sharifi MN, Mowers EE, Drake LE, Collier C, Chen H, Zamora M, Mui S, Macleod KF. Autophagy Promotes Focal Adhesion Disassembly and Cell Motility of Metastatic Tumor Cells through the Direct Interaction of Paxillin with LC3. Cell reports. 2016;15:1660–72. doi: 10.1016/j.celrep.2016.04.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kim SI, Na HJ, Ding Y, Wang Z, Lee SJ, Choi ME. Autophagy promotes intracellular degradation of type I collagen induced by transforming growth factor (TGF)-beta1. J Biol Chem. 2012;287:11677–88. doi: 10.1074/jbc.M111.308460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lock R, Kenific CM, Leidal AM, Salas E, Debnath J. Autophagy dependent production of secreted factors facilitates oncogenic RAS-driven invasion. Cancer Discov. 2014;4:466–79. doi: 10.1158/2159-8290.CD-13-0841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Sandilands E, Serrels B, Wilkinson S, Frame MC. Src-dependent autophagic degradation of Ret in FAK-signalling-defective cancer cells. EMBO Rep. 2012;13:733–40. doi: 10.1038/embor.2012.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chang CH, Bijian K, Qiu D, Su J, Saad A, Dahabieh MS, Miller WH, Jr, Alaoui-Jamali MA. Endosomal sorting and c-Cbl targeting of paxillin to autophagosomes regulate cell-matrix adhesion turnover in human breast cancer cells. Oncotarget. 2017;8:31199–31214. doi: 10.18632/oncotarget.16105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Schoenherr C, Byron A, Sandilands E, Paliashvili K, Baillie GS, Garcia-Munoz A, Valacca C, Cecconi F, Serrels B, Frame MC. Ambra1 spatially regulates Src activity and Src/FAK-mediated cancer cell invasion via trafficking networks. eLife. 2017;6 doi: 10.7554/eLife.23172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Abbi S, Ueda H, Zheng C, Cooper LA, Zhao J, Christopher R, Guan JL. Regulation of focal adhesion kinase by a novel protein inhibitor FIP200. Mol Biol Cell. 2002;13:3178–91. doi: 10.1091/mbc.E02-05-0295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hara T, Takamura A, Kishi C, Iemura S, Natsume T, Guan JL, Mizushima N. FIP200, a ULK-interacting protein, is required for autophagosome formation in mammalian cells. J Cell Biol. 2008;181:497–510. doi: 10.1083/jcb.200712064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gan B, Melkoumian ZK, Wu X, Guan KL, Guan JL. Identification of FIP200 interaction with the TSC1-TSC2 complex and its role in regulation of cell size control. J Cell Biol. 2005;170:379–89. doi: 10.1083/jcb.200411106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gan B, Yoo Y, Guan JL. Association of focal adhesion kinase with tuberous sclerosis complex 2 in the regulation of s6 kinase activation and cell growth. J Biol Chem. 2006;281:37321–9. doi: 10.1074/jbc.M605241200. [DOI] [PubMed] [Google Scholar]
- 54.Deakin NO, Turner CE. Paxillin comes of age. Journal of cell science. 2008;121:2435–44. doi: 10.1242/jcs.018044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Chen GC, Lee JY, Tang HW, Debnath J, Thomas SM, Settleman J. Genetic interactions between Drosophila Atg1 and paxillin reveal a role for paxillin in autophagosome formation. Autophagy. 2008;4:37–45. doi: 10.4161/auto.5141. [DOI] [PubMed] [Google Scholar]
- 56.Sahai E, Marshall CJ. RHO-GTPases and cancer. Nat Rev Cancer. 2002;2:133–42. doi: 10.1038/nrc725. [DOI] [PubMed] [Google Scholar]
- 57.Sahai E, Marshall CJ. Differing modes of tumor cell invasion have distinct requirements for Rho/ROCK signaling and extracellular proteolysis. Nat Cell Biol. 2003;5:711–9. doi: 10.1038/ncb1019. [DOI] [PubMed] [Google Scholar]
- 58.Belaid A, Cerezo M, Chargui A, Corcelle-Termeau E, Pedeutour F, Giuliano S, Ilie M, Rubera I, Tauc M, Barale S, Bertolotto C, Brest P, Vouret-Craviari V, Klionsky DJ, Carle GF, Hofman P, Mograbi B. Autophagy plays a critical role in the degradation of active RHOA, the control of cell cytokinesis, and genomic stability. Cancer research. 2013;73:4311–22. doi: 10.1158/0008-5472.CAN-12-4142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Chan EY, Kir S, Tooze SA. siRNA screening of the kinome identifies ULK1 as a multidomain modulator of autophagy. J Biol Chem. 2007;282:25464–74. doi: 10.1074/jbc.M703663200. [DOI] [PubMed] [Google Scholar]
- 60.Mleczak A, Millar S, Tooze SA, Olson MF, Chan EY. Regulation of autophagosome formation by Rho kinase. Cellular signalling. 2013;25:1–11. doi: 10.1016/j.cellsig.2012.09.010. [DOI] [PubMed] [Google Scholar]
- 61.Pattingre S, Tassa A, Qu X, Garuti R, Linag XH, Mizushima N, Packer M, Schneider MD, Levine B. Bcl-2 anti-apoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122:927–39. doi: 10.1016/j.cell.2005.07.002. [DOI] [PubMed] [Google Scholar]
- 62.Gurkar AU, Chu K, Raj L, Bouley R, Lee SH, Kim YB, Dunn SE, Mandinova A, Lee SW. Identification of ROCK1 kinase as a critical regulator of Beclin1-mediated autophagy during metabolic stress. Nature communications. 2013;4:2189. doi: 10.1038/ncomms3189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gutierrez MG, Munafo DB, Beron W, Colombo MI. Rab7 is required for the normal progression of the autophagic pathway in mammalian cells. Journal of cell science. 2004;117:2687–97. doi: 10.1242/jcs.01114. [DOI] [PubMed] [Google Scholar]
- 64.Frasa MA, Maximiano FC, Smolarczyk K, Francis RE, Betson ME, Lozano E, Goldenring J, Seabra MC, Rak A, Ahmadian MR, Braga VM. Armus is a Rac1 effector that inactivates Rab7 and regulates E-cadherin degradation. Current biology : CB. 2010;20:198–208. doi: 10.1016/j.cub.2009.12.053. [DOI] [PubMed] [Google Scholar]
- 65.Carroll B, Mohd-Naim N, Maximiano F, Frasa MA, McCormack J, Finelli M, Thoresen SB, Perdios L, Daigaku R, Francis RE, Futter C, Dikic I, Braga VM. The TBC/RabGAP Armus coordinates Rac1 and Rab7 functions during autophagy. Dev Cell. 2013;25:15–28. doi: 10.1016/j.devcel.2013.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Till A, Saito R, Merkurjev D, Liu JJ, Syed GH, Kolnik M, Siddiqui A, Glas M, Scheffler B, Ideker T, Subramani S. Evolutionary trends and functional anatomy of the human expanded autophagy network. Autophagy. 2015;11:1652–67. doi: 10.1080/15548627.2015.1059558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Oliver L, Hue E, Priault M, Vallette FM. Basal autophagy decreased during the differentiation of human adult mesenchymal stem cells. Stem cells and development. 2012;21:2779–88. doi: 10.1089/scd.2012.0124. [DOI] [PubMed] [Google Scholar]
- 68.Tra T, Gong L, Kao LP, Li XL, Grandela C, Devenish RJ, Wolvetang E, Prescott M. Autophagy in human embryonic stem cells. PLoS One. 2011;6:e27485. doi: 10.1371/journal.pone.0027485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Vazquez P, Arroba AI, Cecconi F, de la Rosa EJ, Boya P, de Pablo F. Atg5 and Ambra1 differentially modulate neurogenesis in neural stem cells. Autophagy. 2012;8:187–99. doi: 10.4161/auto.8.2.18535. [DOI] [PubMed] [Google Scholar]
- 70.Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nature reviews Drug discovery. 2011;10:417–27. doi: 10.1038/nrd3455. [DOI] [PubMed] [Google Scholar]
- 71.Warr MR, Binnewies M, Flach J, Reynaud D, Garg T, Malhotra R, Debnath J, Passegue E. FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature. 2013;494:323–7. doi: 10.1038/nature11895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Garcia-Prat L, Martinez-Vicente M, Perdiguero E, Ortet L, Rodriguez-Ubreva J, Rebollo E, Ruiz-Bonilla V, Gutarra S, Ballestar E, Serrano AL, Sandri M, Munoz-Canoves P. Autophagy maintains stemness by preventing senescence. Nature. 2016;529:37–42. doi: 10.1038/nature16187. [DOI] [PubMed] [Google Scholar]
- 73.Katajisto P, Dohla J, Chaffer CL, Pentinmikko N, Marjanovic N, Iqbal S, Zoncu R, Chen W, Weinberg RA, Sabatini DM. Stem cells. Asymmetric apportioning of aged mitochondria between daughter cells is required for stemness. Science. 2015;348:340–3. doi: 10.1126/science.1260384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Adams WC, Chen YH, Kratchmarov R, Yen B, Nish SA, Lin WW, Rothman NJ, Luchsinger LL, Klein U, Busslinger M, Rathmell JC, Snoeck HW, Reiner SL. Anabolism-Associated Mitochondrial Stasis Driving Lymphocyte Differentiation over Self-Renewal. Cell reports. 2016;17:3142–3152. doi: 10.1016/j.celrep.2016.11.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sin J, Andres AM, Taylor DJ, Weston T, Hiraumi Y, Stotland A, Kim BJ, Huang C, Doran KS, Gottlieb RA. Mitophagy is required for mitochondrial biogenesis and myogenic differentiation of C2C12 myoblasts. Autophagy. 2016;12:369–80. doi: 10.1080/15548627.2015.1115172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ito K, Turcotte R, Cui J, Zimmerman SE, Pinho S, Mizoguchi T, Arai F, Runnels JM, Alt C, Teruya-Feldstein J, Mar JC, Singh R, Suda T, Lin CP, Frenette PS, Ito K. Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science. 2016;354:1156–1160. doi: 10.1126/science.aaf5530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ho TT, Warr MR, Adelman ER, Lansinger OM, Flach J, Verovskaya EV, Figueroa ME, Passegue E. Autophagy maintains the metabolism and function of young and old stem cells. Nature. 2017;543:205–210. doi: 10.1038/nature21388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Vannini N, Girotra M, Naveiras O, Nikitin G, Campos V, Giger S, Roch A, Auwerx J, Lutolf MP. Specification of haematopoietic stem cell fate via modulation of mitochondrial activity. Nature communications. 2016;7:13125. doi: 10.1038/ncomms13125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gu W, Gaeta X, Sahakyan A, Chan AB, Hong CS, Kim R, Braas D, Plath K, Lowry WE, Christofk HR. Glycolytic Metabolism Plays a Functional Role in Regulating Human Pluripotent Stem Cell State. Cell Stem Cell. 2016;19:476–490. doi: 10.1016/j.stem.2016.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ito K, Suda T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat Rev Mol Cell Biol. 2014;15:243–56. doi: 10.1038/nrm3772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Xu X, Duan S, Yi F, Ocampo A, Liu GH, Izpisua Belmonte JC. Mitochondrial regulation in pluripotent stem cells. Cell Metab. 2013;18:325–32. doi: 10.1016/j.cmet.2013.06.005. [DOI] [PubMed] [Google Scholar]
- 82.Chung S, Dzeja PP, Faustino RS, Perez-Terzic C, Behfar A, Terzic A. Mitochondrial oxidative metabolism is required for the cardiac differentiation of stem cells. Nature clinical practice Cardiovascular medicine. 2007;4(Suppl 1):S60–7. doi: 10.1038/ncpcardio0766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Varum S, Rodrigues AS, Moura MB, Momcilovic O, Easley CAt, Ramalho-Santos J, Van Houten B, Schatten G. Energy metabolism in human pluripotent stem cells and their differentiated counterparts. PLoS One. 2011;6:e20914. doi: 10.1371/journal.pone.0020914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Folmes CD, Nelson TJ, Martinez-Fernandez A, Arrell DK, Lindor JZ, Dzeja PP, Ikeda Y, Perez-Terzic C, Terzic A. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. Cell Metab. 2011;14:264–71. doi: 10.1016/j.cmet.2011.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Chaffer CL, Brueckmann I, Scheel C, Kaestli AJ, Wiggins PA, Rodrigues LO, Brooks M, Reinhardt F, Su Y, Polyak K, Arendt LM, Kuperwasser C, Bierie B, Weinberg RA. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci U S A. 2011;108:7950–5. doi: 10.1073/pnas.1102454108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331:1559–64. doi: 10.1126/science.1203543. [DOI] [PubMed] [Google Scholar]
- 87.Comen E, Norton L, Massague J. Clinical implications of cancer self-seeding. Nature reviews Clinical oncology. 2011;8:369–77. doi: 10.1038/nrclinonc.2011.64. [DOI] [PubMed] [Google Scholar]
- 88.Charafe-Jauffret E, Ginestier C, Iovino F, Wicinski J, Cervera N, Finetti P, Hur MH, Diebel ME, Monville F, Dutcher J, Brown M, Viens P, Xerri L, Bertucci F, Stassi G, Dontu G, Birnbaum D, Wicha MS. Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Cancer research. 2009;69:1302–13. doi: 10.1158/0008-5472.CAN-08-2741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Pang R, Law WL, Chu AC, Poon JT, Lam CS, Chow AK, Ng L, Cheung LW, Lan XR, Lan HY, Tan VP, Yau TC, Poon RT, Wong BC. A subpopulation of CD26+ cancer stem cells with metastatic capacity in human colorectal cancer. Cell Stem Cell. 2010;6:603–15. doi: 10.1016/j.stem.2010.04.001. [DOI] [PubMed] [Google Scholar]
- 90.Chen C, Wei Y, Hummel M, Hoffmann TK, Gross M, Kaufmann AM, Albers AE. Evidence for epithelial-mesenchymal transition in cancer stem cells of head and neck squamous cell carcinoma. PLoS One. 2011;6:e16466. doi: 10.1371/journal.pone.0016466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Nieto MA, Huang RY, Jackson RA, Thiery JP. EMT: 2016. Cell. 2016;166:21–45. doi: 10.1016/j.cell.2016.06.028. [DOI] [PubMed] [Google Scholar]
- 92.Guo W, Keckesova Z, Donaher JL, Shibue T, Tischler V, Reinhardt F, Itzkovitz S, Noske A, Zurrer-Hardi U, Bell G, Tam WL, Mani SA, van Oudenaarden A, Weinberg RA. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell. 2012;148:1015–28. doi: 10.1016/j.cell.2012.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks MW, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–15. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ye X, Tam WL, Shibue T, Kaygusuz Y, Reinhardt F, Ng Eaton E, Weinberg RA. Distinct EMT programs control normal mammary stem cells and tumour-initiating cells. Nature. 2015;525:256–60. doi: 10.1038/nature14897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kiyono K, Suzuki HI, Matsuyama H, Morishita Y, Komuro A, Kano MR, Sugimoto K, Miyazono K. Autophagy is activated by TGF-b and potentiates TGF-b-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res. 2009;69:8844–52. doi: 10.1158/0008-5472.CAN-08-4401. [DOI] [PubMed] [Google Scholar]
- 96.Cufi S, Vazquez-Martin A, Oliveras-Ferraros C, Martin-Castillo B, Vellon L, Menendez JA. Autophagy positively regulates the CD44(+) CD24(−/low) breast cancer stem-like phenotype. Cell Cycle. 2011;10:3871–85. doi: 10.4161/cc.10.22.17976. [DOI] [PubMed] [Google Scholar]
- 97.Paget S. THE DISTRIBUTION OF SECONDARY GROWTHS IN CANCER OF THE BREAST. The Lancet. 1889;133:571–73. [PubMed] [Google Scholar]
- 98.Wolf J, Dewi DL, Fredebohm J, Muller-Decker K, Flechtenmacher C, Hoheisel JD, Boettcher M. A mammosphere formation RNAi screen reveals that ATG4A promotes a breast cancer stem-like phenotype. Breast cancer research : BCR. 2013;15:R109. doi: 10.1186/bcr3576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Springer MZ, Macleod KF. In Brief: Mitophagy: mechanisms and role in human disease. The Journal of pathology. 2016;240:253–255. doi: 10.1002/path.4774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ye XQ, Li Q, Wang GH, Sun FF, Huang GJ, Bian XW, Yu SC, Qian GS. Mitochondrial and energy metabolism-related properties as novel indicators of lung cancer stem cells. Int J Cancer. 2011;129:820–31. doi: 10.1002/ijc.25944. [DOI] [PubMed] [Google Scholar]
- 101.Shen YA, Wang CY, Hsieh YT, Chen YJ, Wei YH. Metabolic reprogramming orchestrates cancer stem cell properties in nasopharyngeal carcinoma. Cell Cycle. 2015;14:86–98. doi: 10.4161/15384101.2014.974419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Viale A, Pettazzoni P, Lyssiotis CA, Ying H, Sánchez N, Marchesini M, Carugo A, Green T, Seth S, Giuliani V, Kost-Alimova M, Muller F, Colla S, Nezi L, Genovese G, Deem AK, Kapoor A, Yao W, Brunetto E, Kang Y, Yuan M, Asara JM, Wang YA, Heffernan TP, Kimmelman AC, Wang H, Fleming JB, Cantley LC, DePinho RA, Draetta GF. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature. 2014;514:628–32. doi: 10.1038/nature13611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Karrison TG, Ferguson DJ, Meier P. Dormancy of mammary carcinoma after mastectomy. Journal of the National Cancer Institute. 1999;91:80–5. doi: 10.1093/jnci/91.1.80. [DOI] [PubMed] [Google Scholar]
- 104.Giancotti FG. Mechanisms governing metastatic dormancy and reactivation. Cell. 2013;155:750–64. doi: 10.1016/j.cell.2013.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lu Z, Luo RZ, Lu Y, Zhang X, Yu Q, Khare S, Kondo S, Kondo Y, Yu Y, Mills GB, Liao WSL, Bast J, R C. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cells. J Clin Invest. 2008;118:3917–29. doi: 10.1172/JCI35512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Amaravadi RK, Yu DS, Lum JJ, Bui T, Christophorou MA, Evan GI, Thomas-Tikhonenko A, Thompson CB. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. JClinInvest. 2007;117:326–36. doi: 10.1172/JCI28833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lum JJ, Bauer DE, Kong M, Harris MH, Li CY, Lindsten T, Thompson CB. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120:237–49. doi: 10.1016/j.cell.2004.11.046. [DOI] [PubMed] [Google Scholar]
- 108.Galluzzi L, Pietrocola F, Levine B, Kroemer G. Metabolic control of autophagy. Cell. 2014;159:1263–76. doi: 10.1016/j.cell.2014.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Balic M, Lin H, Young L, Hawes D, Giuliano A, McNamara G, Datar RH, Cote RJ. Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12:5615–21. doi: 10.1158/1078-0432.CCR-06-0169. [DOI] [PubMed] [Google Scholar]
- 110.Wang J, Wu GS. Role of autophagy in cisplatin resistance in ovarian cancer cells. J Biol Chem. 2014;289:17163–73. doi: 10.1074/jbc.M114.558288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.O'Donovan TR, O'Sullivan GC, McKenna SL. Induction of autophagy by drug-resistant esophageal cancer cells promotes their survival and recovery following treatment with chemotherapeutics. Autophagy. 2011;7:509–24. doi: 10.4161/auto.7.6.15066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Nguyen HG, Yang JC, Kung HJ, Shi XB, Tilki D, Lara PN, Jr, DeVere White RW, Gao AC, Evans CP. Targeting autophagy overcomes Enzalutamide resistance in castration-resistant prostate cancer cells and improves therapeutic response in a xenograft model. Oncogene. 2014;33:4521–30. doi: 10.1038/onc.2014.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Samaddar JS, Gaddy VT, Duplantier J, Thandavan SP, Shah M, Smith MJ, Browning D, Rawson J, Smith SB, Barrett JT, Schoenlein PV. A role for macroautophagy in protection against 4-hydroxytamoxifen-induced cell death and the development of antiestrogen resistance. Molecular cancer therapeutics. 2008;7:2977–87. doi: 10.1158/1535-7163.MCT-08-0447. [DOI] [PubMed] [Google Scholar]
- 114.Qadir MA, Kwok B, Dragowska WH, To KH, Le D, Bally MB, Gorski SM. Macroautophagy inhibition sensitizes tamoxifen-resistant breast cancer cells and enhances mitochondrial depolarization. Breast cancer research and treatment. 2008;112:389–403. doi: 10.1007/s10549-007-9873-4. [DOI] [PubMed] [Google Scholar]
- 115.Cook KL, Warri A, Soto-Pantoja DR, Clarke PA, Cruz MI, Zwart A, Clarke R. Hydroxychloroquine inhibits autophagy to potentiate antiestrogen responsiveness in ER+ breast cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2014;20:3222–32. doi: 10.1158/1078-0432.CCR-13-3227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Gupta A, Roy S, Lazar AJF, Wang WL, McAuliffe JC, Reynoso D, McMahon J, Taguchi T, Floris G, Debiec-Rychter M, Schoffski P, Trent JA, Debnath J, Rubin BP. Autophagy inhibition and antimalarials promote cell death in gastrointestinal stromal tumor (GIST) Proc Natl Acad Sci U S A. 2010;107:14333–8. doi: 10.1073/pnas.1000248107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Farrow JM, Yang JC, Evans CP. Autophagy as a modulator and target in prostate cancer. Nature reviews Urology. 2014;11:508–16. doi: 10.1038/nrurol.2014.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Sui X, Kong N, Zhu M, Wang X, Lou F, Han W, Pan H. Cotargeting EGFR and autophagy signaling: A novel therapeutic strategy for non-small-cell lung cancer. Molecular and clinical oncology. 2014;2:8–12. doi: 10.3892/mco.2013.187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Han W, Pan H, Chen Y, Sun J, Wang Y, Li J, Ge W, Feng L, Lin X, Wang X, Wang X, Jin H. EGFR tyrosine kinase inhibitors activate autophagy as a cytoprotective response in human lung cancer cells. PLoS One. 2011;6:e18691. doi: 10.1371/journal.pone.0018691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wei Y, Zou Z, Becker N, Anderson M, Sumpter R, Xiao G, Kinch L, Koduru P, Christudass CS, Veltri RW, Grishin NV, Peyton M, Minna J, Bhagat G, Levine B. EGFR-mediated Beclin 1 phosphorylation in autophagy suppression, tumor progression, and tumor chemoresistance. Cell. 2013;154:1269–84. doi: 10.1016/j.cell.2013.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Yan Y, Xu Z, Dai S, Qian L, Sun L, Gong Z. Targeting autophagy to sensitive glioma to temozolomide treatment. Journal of experimental & clinical cancer research : CR. 2016;35:23. doi: 10.1186/s13046-016-0303-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Frassanito MA, De Veirman K, Desantis V, Di Marzo L, Vergara D, Ruggieri S, Annese T, Nico B, Menu E, Catacchio I, Ria R, Racanelli V, Maffia M, Angelucci E, Derudas D, Fumarulo R, Dammacco F, Ribatti D, Vanderkerken K, Vacca A. Halting pro-survival autophagy by TGFbeta inhibition in bone marrow fibroblasts overcomes bortezomib resistance in multiple myeloma patients. Leukemia. 2016;30:640–8. doi: 10.1038/leu.2015.289. [DOI] [PubMed] [Google Scholar]
- 123.Milan E, Perini T, Resnati M, Orfanelli U, Oliva L, Raimondi A, Cascio P, Bachi A, Marcatti M, Ciceri F, Cenci S. A plastic SQSTM1/p62-dependent autophagic reserve maintains proteostasis and determines proteasome inhibitor susceptibility in multiple myeloma cells. Autophagy. 2015;11:1161–78. doi: 10.1080/15548627.2015.1052928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ma XH, Piao SF, Dey S, McAfee Q, Karakousis G, Villanueva J, Hart LS, Levi S, Hu J, Zhang G, Lazova R, Klump V, Pawelek JM, Xu X, Xu W, Schuchter LM, Davies MA, Herlyn M, Winkler J, Koumenis C, Amaravadi RK. Targeting ER stress-induced autophagy overcomes BRAF inhibitor resistance in melanoma. The Journal of clinical investigation. 2014;124:1406–17. doi: 10.1172/JCI70454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Mulcahy Levy E, Thomspson J, Greisinger AM, Amani V, Donson AM, Birks DK, Morgan MJ, Mirsky DM, Handler MH, Foreman NK, Thorburn A. Autophagy Inhibition Improves Chemosensitivity in BRAFV600E Brain Tumors. Cancer Discovery. 2014;4:773–80. doi: 10.1158/2159-8290.CD-14-0049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Mulcahy Levy JM, Zahedi S, Griesinger AM, Morin A, Davies KD, Aisner DL, Kleinschmidt-DeMasters BK, Fitzwalter BE, Goodall ML, Thorburn J, Amani V, Donson AM, Birks DK, Mirsky DM, Hankinson TC, Handler MH, Green AL, Vibhakar R, Foreman NK, Thorburn A. Autophagy inhibition overcomes multiple mechanisms of resistance to BRAF inhibition in brain tumors. eLife. 2017;6 doi: 10.7554/eLife.19671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Kuo WL, Sharifi MN, Lingen MW, Ahmed O, Liu J, Nagilla M, Macleod KF, Cohen EE. p62/SQSTM1 accumulation in squamous cell carcinoma of head and neck predicts sensitivity to phosphatidylinositol 3-kinase pathway inhibitors. PLoS One. 2014;9:e90171. doi: 10.1371/journal.pone.0090171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Pascolo S. Time to use a dose of Chloroquine as an adjuvant to anti-cancer chemotherapies. European journal of pharmacology. 2016;771:139–44. doi: 10.1016/j.ejphar.2015.12.017. [DOI] [PubMed] [Google Scholar]
- 129.Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17:528–542. doi: 10.1038/nrc.2017.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Briceno E, Reyes S, Sotelo J. Therapy of glioblastoma multiforme improved by the antimutagenic chloroquine. Neurosurgical focus. 2003;14:e3. doi: 10.3171/foc.2003.14.2.4. [DOI] [PubMed] [Google Scholar]
- 131.Sotelo J, Briceno E, Lopez-Gonzalez MA. Adding chloroquine to conventional treatment for glioblastoma multiforme: a randomized, double-blind, placebo-controlled trial. Annals of internal medicine. 2006;144:337–43. doi: 10.7326/0003-4819-144-5-200603070-00008. [DOI] [PubMed] [Google Scholar]
- 132.Barnard RA, Wittenburg LA, Amaravadi RK, Gustafson DL, Thorburn A, Thamm DH. Phase I clinical trial and pharmacodynamic evaluation of combination hydroxychloroquine and doxorubicin treatment in pet dogs treated for spontaneously occurring lymphoma. Autophagy. 2014;10:1415–25. doi: 10.4161/auto.29165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.McAfee Q, Zhang Z, Samanta A, Levi SM, Ma XH, Piao S, Lynch JP, Uehara T, Sepulveda AR, Davis LE, Winkler JD, Amaravadi RK. Autophagy inhibitor Lys05 has single-agent antitumor activity and reproduces the phenotype of a genetic autophagy deficiency. Proc Natl Acad Sci U S A. 2012;109:8253–8. doi: 10.1073/pnas.1118193109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Zoncu R, Bar-Peled L, Efeyan A, Wang S, Sancak Y, Sabatini DM. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H(+)-ATPase. Science. 2011;334:678–83. doi: 10.1126/science.1207056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wyant GA, Abu-Remaileh M, Wolfson RL, Chen WW, Freinkman E, Danai LV, Vander Heiden MG, Sabatini DM. mTORC1 Activator SLC38A9 Is Required to Efflux Essential Amino Acids from Lysosomes and Use Protein as a Nutrient. Cell. 2017;171:642–654.e12. doi: 10.1016/j.cell.2017.09.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Karsli-Uzunbas G, Guo JY, Price S, Teng X, Laddha SV, Khor S, Kalaany NY, Jacks T, Chan CS, Rabinowitz JD, White E. Autophagy Is Required for Glucose Homeostasis and Lung Tumor Maintenance. Cancer Discov. 2014;4:914–27. doi: 10.1158/2159-8290.CD-14-0363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Rebecca VW, Nicastri MC, McLaughlin N, Fennelly C, McAfee Q, Ronghe A, Nofal M, Lim CY, Witze E, Chude CI, Zhang G, Alicea GM, Piao S, Murugan S, Ojha R, Levi SM, Wei Z, Barber-Rotenberg JS, Murphy ME, Mills GB, Lu Y, Rabinowitz J, Marmorstein R, Liu Q, Liu S, Xu X, Herlyn M, Zoncu R, Brady DC, Speicher DW, Winkler JD, Amaravadi RK. A Unified Approach to Targeting the Lysosome's Degradative and Growth Signaling Roles. Cancer Discov. 2017;7:1266–1283. doi: 10.1158/2159-8290.CD-17-0741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Egan DF, Chun MG, Vamos M, Zou H, Rong J, Miller CJ, Lou HJ, Raveendra-Panickar D, Yang CC, Sheffler DJ, Teriete P, Asara JM, Turk BE, Cosford ND, Shaw RJ. Small Molecule Inhibition of the Autophagy Kinase ULK1 and Identification of ULK1 Substrates. Molecular cell. 2015;59:285–97. doi: 10.1016/j.molcel.2015.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Petherick KJ, Conway OJ, Mpamhanga C, Osborne SA, Kamal A, Saxty B, Ganley IG. Pharmacological inhibition of ULK1 kinase blocks mammalian target of rapamycin (mTOR)-dependent autophagy. J Biol Chem. 2015;290:11376–83. doi: 10.1074/jbc.C114.627778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Ronan B, Flamand O, Vescovi L, Dureuil C, Durand L, Fassy F, Bachelot MF, Lamberton A, Mathieu M, Bertrand T, Marquette JP, El-Ahmad Y, Filoche-Romme B, Schio L, Garcia-Echeverria C, Goulaouic H, Pasquier B. A highly potent and selective Vps34 inhibitor alters vesicle trafficking and autophagy. Nature chemical biology. 2014;10:1013–9. doi: 10.1038/nchembio.1681. [DOI] [PubMed] [Google Scholar]
- 141.Akin D, Wang SK, Habibzadegah-Tari P, Law B, Ostrov D, Li M, Yin XM, Kim JS, Horenstein N, Dunn WA., Jr A novel ATG4B antagonist inhibits autophagy and has a negative impact on osteosarcoma tumors. Autophagy. 2014;10:2021–35. doi: 10.4161/auto.32229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Ma Y, Galluzzi L, Zitvogel L, Kroemer G. Autophagy and cellular immune responses. Immunity. 2013;39:211–27. doi: 10.1016/j.immuni.2013.07.017. [DOI] [PubMed] [Google Scholar]
- 143.Zhong Z, Sanchez-Lopez E, Karin M. Autophagy, Inflammation, and Immunity: A Troika Governing Cancer and Its Treatment. Cell. 2016;166:288–98. doi: 10.1016/j.cell.2016.05.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, Shen S, Kepp O, Scoazec M, Mignot G, Rello-Varona S, Tailler M, Menger L, Vacchelli E, Galluzzi L, Ghiringhelli F, di Virgilio F, Zitvogel L, Kroemer G. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–7. doi: 10.1126/science.1208347. [DOI] [PubMed] [Google Scholar]
- 145.Ladoire S, Enot D, Senovilla L, Ghiringhelli F, Poirier-Colame V, Chaba K, Semeraro M, Chaix M, Penault-Llorca F, Arnould L, Poillot ML, Arveux P, Delaloge S, Andre F, Zitvogel L, Kroemer G. The presence of LC3B puncta and HMGB1 expression in malignant cells correlate with the immune infiltrate in breast cancer. Autophagy. 2016;12:864–75. doi: 10.1080/15548627.2016.1154244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Rao S, Tortola L, Perlot T, Wirnsberger G, Novatchkova M, Nitsch R, Sykacek P, Frank L, Schramek D, Komnenovic V, Sigl V, Aumayr K, Schmauss G, Fellner N, Handschuh S, Glösmann M, Pasierbek P, Schlederer M, Resch GP, Ma Y, Yang H, Popper H, Kenner L, Kroemer G, Penninger JM. A dual role for autophagy in a murine model of lung cancer. Nat Comm. 2014;5:3056–70. doi: 10.1038/ncomms4056. [DOI] [PubMed] [Google Scholar]
- 147.Starobinets H, Ye J, Broz M, Barry K, Goldsmith J, Marsh T, Rostker F, Krummel M, Debnath J. Antitumor adaptive immunity remains intact following inhibition of autophagy and antimalarial treatment. The Journal of clinical investigation. 2016;126:4417–4429. doi: 10.1172/JCI85705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Wei H, Wei S, Gan B, Peng X, Zou W, Guan JL. Suppression of autophagy by FIP200 deletion inhibits mammary tumorigenesis. Genes & Dev. 2011;25:1510–27. doi: 10.1101/gad.2051011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Noman MZ, Janji B, Kaminska B, Van Moer K, Pierson S, Przanowski P, Buart S, Berchem G, Romero P, Mami-Chouaib F, Chouaib S. Blocking hypoxia-induced autophagy in tumors restores cytotoxic T-cell activity and promotes regression. Cancer research. 2011;71:5976–86. doi: 10.1158/0008-5472.CAN-11-1094. [DOI] [PubMed] [Google Scholar]
- 150.Akalay I, Janji B, Hasmim M, Noman MZ, Andre F, De Cremoux P, Bertheau P, Badoual C, Vielh P, Larsen AK, Sabbah M, Tan TZ, Keira JH, Hung NT, Thiery JP, Mami-Chouaib F, Chouaib S. Epithelial-to-mesenchymal transition and autophagy induction in breast carcinoma promote escape from T-cell-mediated lysis. Cancer research. 2013;73:2418–27. doi: 10.1158/0008-5472.CAN-12-2432. [DOI] [PubMed] [Google Scholar]
- 151.Yang A, Herter-Sprie G, Zhang H, Lin EY, Biancur D, Wang X, Deng J, Hai J, Yang S, Wong KK, Kimmelman AC. Autophagy sustains pancreatic cancer growth through both cell autonomous and non-autonomous mechanisms. Cancer Discov. 2018 doi: 10.1158/2159-8290.CD-17-0952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Katheder NS, Khezri R, O'Farrell F, Schultz SW, Jain A, Rahman MM, Schink KO, Theodossiou TA, Johansen T, Juhasz G, Bilder D, Brech A, Stenmark H, Rusten TE. Microenvironmental autophagy promotes tumour growth. Nature. 2017;541:417–420. doi: 10.1038/nature20815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Endo S, Nakata K, Ohuchida K, Takesue S, Nakayama H, Abe T, Koikawa K, Okumura T, Sada M, Horioka K, Zheng B, Mizuuchi Y, Iwamoto C, Murata M, Moriyama T, Miyasaka Y, Ohtsuka T, Mizumoto K, Oda Y, Hashizume M, Nakamura M. Autophagy Is Required for Activation of Pancreatic Stellate Cells, Associated With Pancreatic Cancer Progression and Promotes Growth of Pancreatic Tumors in Mice. Gastroenterology. 2017;152:1492–1506.e24. doi: 10.1053/j.gastro.2017.01.010. [DOI] [PubMed] [Google Scholar]
- 154.Dupont N, Jiang S, Pilli M, Ornatowski W, Bhattacharya D, Deretic V. Autophagy-based unconventional secretory pathway for extracellular delivery of IL-1beta. Embo j. 2011;30:4701–11. doi: 10.1038/emboj.2011.398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Jiang S, Dupont N, Castillo EF, Deretic V. Secretory versus degradative autophagy: unconventional secretion of inflammatory mediators. Journal of innate immunity. 2013;5:471–9. doi: 10.1159/000346707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Gore J, Korc M. Pancreatic cancer stroma: friend or foe? Cancer Cell. 2014;25:711–2. doi: 10.1016/j.ccr.2014.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, Denicola G, Feig C, Combs C, Winter SP, Ireland-Zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Ruckert F, Grutzmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Davies SE, Plunkett W, Egorin M, Hruban RH, Whitebread N, McGovern K, Adams J, Iacobuzio-Donahue C, Griffiths J, Tuveson DA. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–61. doi: 10.1126/science.1171362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, Westphalen CB, Kitajewski J, Fernandez-Barrena MG, Fernandez-Zapico ME, Iacobuzio-Donahue C, Olive KP, Stanger BZ. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–47. doi: 10.1016/j.ccr.2014.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, De Jesus-Acosta A, Sharma P, Heidari P, Mahmood U, Chin L, Moses HL, Weaver VM, Maitra A, Allison JP, LeBleu VS, Kalluri R. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719–34. doi: 10.1016/j.ccr.2014.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Kwon Y, Song W, Droujinine IA, Hu Y, Asara JM, Perrimon N. Systemic organ wasting induced by localized expression of the secreted insulin/IGF antagonist ImpL2. Dev Cell. 2015;33:36–46. doi: 10.1016/j.devcel.2015.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Figueroa-Clarevega A, Bilder D. Malignant Drosophila tumors interrupt insulin signaling to induce cachexia-like wasting. Dev Cell. 2015;33:47–55. doi: 10.1016/j.devcel.2015.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]