Abstract
Autophagy is a homeostatic and catabolic process that enables the sequestration and lysosomal degradation of cytoplasmic organelles and proteins that is important for the maintenance of genomic stability and cell survival. Beclin 1+/− gene knockout mice are tumor prone, indicating a tumor suppressor role for autophagy. Autophagy is also a mechanism of stress tolerance that maintains cell viability and can lead to tumor dormancy, progression and therapeutic resistance. Many anticancer drugs induce cytotoxic stress that can activate pro-survival autophagy. In some contexts, excessive or prolonged autophagy can lead to tumor cell death. Inhibition of cytoprotective autophagy by genetic or pharmacological means has been shown to enhance anticancer drug-induced cell death, suggesting a novel therapeutic strategy. Studies are ongoing to define optimal strategies to modulate autophagy for cancer prevention and therapy, and to exploit it as a target for anticancer drug discovery.
Key words: autophagy, beclin 1, cell death, cancer treatment
Introduction
Macroautophagy (hereafter referred to as autophagy) is an evolutionarily conserved, homeostatic process that functions to dispose of damaged cellular organelles and proteins. Autophagy involves the formation of double membrane vesicles, known as autophagosomes, that engulf intracellular contents and fuse with lysosomes for their degradation.1 Monoallelic loss of the essential autophagy gene Beclin1 in mice promotes tumorigenesis, and this abnormality is also found in human cancers, indicating a role for autophagy in tumor suppression.2,3 Autophagy defective tumor cells accumulate the adaptor protein p62/sequestosome1 (p62/SQSTM1) that can lead to genome damage and promote tumor formation.4 Autophagy is induced in cancer cells as a mechanism of stress tolerance when cells are exposed to nutrient deprivation, hypoxia, reactive oxygen species (ROS) or anticancer treatments.1 Autophagy generates energy from the recycling of cellular proteins and organelles.5,6 In established tumors, stress-induced autophagy enables tumor cell survival which can facilitate tumor progression and confer resistance to anticancer treatments. Accordingly, targeting cytoprotective autophagy in cancers has the potential to enhance treatment efficacy. Autophagy inhibition has been shown to promote apoptotic cell death.7 Inducing excessive autophagy can be pro-death in certain contexts, and should be considered as an alternative tumor killing strategy, particularly in apoptosis-defective cells.8–10 A complex interplay exists between autophagy and apoptosis and strategies to exploit this cross-talk may also hold therapeutic potential. In this review, the multiple roles of autophagy in tumor biology will be outlined including its emergence as a target for both cancer prevention and cancer treatment.
Regulation of Autophagy
Core machinery.
Autophagy is regulated by Beclin 1 that forms a complex with vacuolar sorting protein 34 (Vps34), a class III phosphoinositide 3-kinase (PI3K), and serves as a platform for recruitment of other autophagy-related proteins that are critical for autophagosome formation.11 During the initiation phase, Atg12 conjugates Atg5 which in turn promotes the recruitment and conversion of cytosolic-associated protein light chain 3 (LC3-I) to the membrane-bound LC3-II form.12 Upon completion of autophagosome formation and with the exception of a proportion of LC3-II bound to the luminal membrane, the Atg proteins are then recycled in the cytosol.11 Biochemical or microscopic detection of the conversion of LC3-I to LC3-II is a biochemical hallmark of autophagy since the lipidated form (LC3-II) is stably associated with the autophagosomal membrane.13 LC3-II binds to p62/SQSTM1 that is involved in trafficking proteins to the proteasome and serves to facilitate the autophagic degradation of ubiquitinated protein aggregates.14 p62/SQSTM1 is normally degraded with autophagosomes and accumulates when autophagy is impaired.4 The late events in autophagy involve the final maturation and fusion of autophagosomes with lysosomes, a step requiring the small GTPase Rab7 and lysosome-associated membrane protein2 (Lamp2).7,15
Regulation of stress-induced autophagy (Fig. 1).
Figure 1.
Autophagy pathway and relationship to mTOR signaling. Cellular stress (hypoxia, genotoxic, cytotoxic) activates autophagy as a stress response and cell survival mechanism. Inhibition of cytoprotective autophagy using 3-MA or CQ/HCQ can augment tumor cell death. Inhibition of mTOR induces autophagy, and in certain cellular contexts, excessive or sustained autophagy may promote cell death. mTOR inhibits autophagy by blocking the formation of ULK-Atg13-FIP200 trimeric complex.
The mammalian target of rapamycin (mTOR) serves as a central regulator of autophagy and forms two distinct signaling complexes, known as mTORC1 and mTORC2.16 A number of upstream signaling pathways are involved in autophagy regulation that converge on mTOR, and include the class I PI3K-AKT pathway which is frequently dysregulated in human cancer.17 Activation of mTOR can occur due to loss of tumor suppressors (LKB1, PML, PTEN, TSC1/2) or through gain-of-function mutations in receptor tyrosine kinases.18 Under stress stimuli that include metabolic stress, hypoxia and mitochondrial damage, suppression of mTOR triggers the autophagic cascade and inhibits cell proliferation.16 Nutrient deprivation results in activation of adenosine monophosphate kinase (AMPK) which is a central metabolic sensor with important functions in regulating lipid and glucose metabolism. Activation of AMPK serves to repress mTOR and to initiate autophagy.19 mTOR negatively regulates autophagy by phosphorylation of Atg13 and Unc51-like kinase (ULK) [human homolog of Atg1], and inhibits formation of a trimeric complex required for autophagosome formation.20,21 Therefore, the trimeric complex ULK-Atg13-FIP200 is a direct target of mTOR.
A major ER stress pathway is the unfolded protein response (UPR) that can potently induce autophagy. The binding of misfolded proteins to the ER chaperone Bip/GRP78 leads to release of three ER membrane-associated proteins, PKR-like eIF2α kinase (PERK), activating transcription factor-6 (ATF6) and inositol-requiring enzyme 1 (IRE1).22,23 While PERK and ATF6 are autophagy inducers, IRE1 negatively regulates autophagy. Other factors that link cellular stress with autophagy is the transcription factor NF-kappaB(NF-kB) and its upstream regulators, IKK complex and TAK1, that integrate diverse stress signals including starvation, rapamycin or ER stress with the autophagy pathway.24 Autophagy induction by hypoxia is regulated through various mechanisms including HIF (hypoxia-inducible factor).25
The tumor suppressor p53 protein can modulate autophagy depending upon its cellular localization. Nuclear p53 acts as a transcription factor that transactivates several autophagy inducers including DRAM1 and Sestrin2 to activate autophagy,26 whereas cytoplasmic p53 inhibits autophagy by an unknown mechanism. Inducers of autophagy can stimulate proteosomemediated degradation of p53.27 Recently, some novel regulators of autophagy have been found. Ataxia-telangiectasia mutated (ATM) is a cellular damage sensor that coordinates the cell cycle with DNA damage-response checkpoints and DNA repair, and engages the TSC2/mTORC1 signaling axis to regulate autophagy.28 Additionally, the high mobility group box 1 (HMGB1) is an immune modulator and regulator of stress-induced autophagy5 that directly interacts with Beclin 1.29
Interplay between autophagy and apoptosis.
Evidence suggests a complex interplay between autophagy and apoptosis pathways.30 Autophagy can be an alternative mode of cell death in apoptosis-resistant cells.8–10 Conversely, apoptosis is promoted when autophagy is inhibited.7,31,32 Non-apoptotic cell death accompanied by autophagy is also well described in cancer cells.33–35 Cross-talk between autophagy and apoptosis exists at many levels where both processes share mediators ranging from the core machinery (Atg5, Beclin 1-Bcl-2/Bcl-xL interaction) to upstream regulating pathways (AKT, DAPK, ARF, E2F1, HMGB1 and p53). Furthermore, recent findings link p62/SQSTM1 activity to caspase-8 activation and the extrinsic apoptosis pathway.36 HMGB1 release has been shown to regulate both autophagy and apoptosis, depending upon its redox state.37 Cytoplasmic translocation of HMGB1 was shown to mediate stress-induced autophagy through the MEK-ERK pathway and limited apoptosis.29
Autophagy in Cancer
The tumor suppressor role of autophagy.
Autophagy has been shown to play a role in tumor suppression. Beclin 1 is a haploinsufficient tumor suppressor gene and Beclin 1+/− mice are tumorprone.2,3 Monoallelic loss of Beclin 1 has been detected in human breast, ovarian and prostate cancers.38 Conversely, Beclin 1 over-expression can inhibit tumor development.39 A potential link between defective autophagy and tumorigenesis is the accumulation of p62/SQSTM1 protein aggregates, damaged mitochondria and misfolded proteins that lead to the production of ROS to cause DNA damage.4 The accumulation of p62/SQSTM1 is frequently observed in human tumors with defective autophagy.40 Suppressing p62/SQSTM1 accumulation or ROS production was shown to prevent damage resulting from defective autophagy.4 p62/SQSTM1 has a TRAF6-binding domain and promotes the oligomerization of TRAF6 leading to the activation of NFκB.41 Autophagy may also protect against tumorigenesis through limiting necrosis and chronic inflammation that is associated with the release of pro-inflammatory HMGB1.5,42
Autophagy in cytoprotection and as a therapeutic target.
A cytoprotective or pro-survival function of autophagy in cancer cells is supported by substantial evidence.43 Cancer cells have increased metabolic demands due to high levels of cellular proliferation and cellular stress activates autophagy to maintain energy and to prevent death in these cells. Autophagy confers stress tolerance, limits damage and confers a survival advantage to cancer cells. Furthermore, autophagy may enable a state of dormancy in residual cancer cells post treatment that may contribute to cancer recurrence and metastasis.44 Autophagy has been shown to localize to hypoxic regions of tumors most distal to blood vessels, where it contributes to the maintenance of cell viability.5,45 The upregulation of autophagy in regions of hypoxia5,45 is enhanced by HIF-1α expression,46 although it can also be independent of HIF-1.18,47 Given the pro-survival role of autophagy, its inhibition has been shown to enhance the efficacy of anticancer therapy.43 Understanding the role of autophagy in cancer treatment is critical since many anticancer therapies activate autophagy that may limit their therapeutic efficacy. Along with conventional cytotoxic drugs, other anti-neoplastic and pharmacological agents such as kinase inhibitors,48–51 proteosome inhibitors,52 arsenic trioxide,53 tamoxifen,9 lucanthone,54 cyclooxygenase inhibitors55 and the HIV protease inhibitor, nelfinavir56 have been shown to induce autophagy in tumor cells (Table 1).
Table 1.
Modulation of autophagy in cancer therapy
| Compound | Target | FDA approved indication in cancer | Tumor type/drug development status |
| Autophagy inducers | |||
| mTOR inhibitors | mTORC1 | ||
| - rapamycin | - | Malignant glioma/in vitro94 | |
| - everolimus | Renal cell carcinoma, subependymal giant cell astrocytoma | ALL/in vivo95 | |
| - temsirolimus | Renal cell carcinoma | Mantle cell lymphoma/in vitro96 | |
| Akt inhibitors | Akt | ||
| - perifosine | - | Lung cancer/in vitro97 | |
| - triciribine | - | T-cell ALL/in vitro98 | |
| Radiation | DNA | Different tumor types | Breast, colon and prostate cancer/in vitro77 |
| Proteosome inhibitor | Proteosome | ||
| - bortezomib | Multiple myeloma, mantle cell lymphoma | Prostate cancer/in vitro52 | |
| EGFR antibody | EGFR | ||
| - cetuximab | Head and neck cancer, metastatic colorectal cancer | Vulvar, colorectal and lung cancer/in vitro51 | |
| HDAC inhibitors | Histone deacetylase | ||
| - vorinostat - suberoylanilide hydroxamic acid (SAHA) |
Primary cutaneous T-cell lymphoma | Multiple cancers/in vitro35, Leukemia/in vitro100 | |
| - osu-HDAC42 | - | Hepatocellular carcinoma/in vitro99 | |
| Phase I in solid tumors www.clinicaltrials.gov | |||
| Tamoxifen | Estrogen receptor | Breast and endometrial cancer | Breast cancer/in vitro9 |
| Temozolomide (alkylating agent) | DNA | Anaplastic astrocytoma, glioblastoma multiforme, metastatic melanoma | Malignant glioma/in vitro75 |
| Glioblastoma/Phase I101 | |||
| Arsenic trioxide | - | Acute promyelocytic leukemia | Malignant glioma/in vitro53 |
| Tyrosine kinase inhibitor | Multiple tyrosine kinases | ||
| - imatinib | *CML, MdS, MPN | Multiple cancers/in vitro48 | |
| CML*/in vitro102 | |||
| Hiv protease inhibitors | Protease | ||
| - nelfinavir | - | Multiple cancers/in vitro56 | |
| Phase I www.clinicaltrials.gov | |||
| Cyclooxygenase-2 inhibitor | Cyclooxygenase-2 | ||
| - celecoxib | - | Colon cancer/in vitro55 | |
| Antidepressants | |||
| - selective serotonin reuptake inhibitor (fluoxetine) | serotonin | - | Burkitt's lymphoma/in vitro68 |
| - norepinephrine reuptake inhibitor (maprotiline) | norepinephrine | - | Burkitt's lymphoma/in vitro68 |
| Autophagy inhibitors | |||
| Chloroquine | Lysosomotropic agent | - | Lymphoma/in vitro and in vivo82 |
| Prostate cancer/in vivo50 | |||
| Several cancers/Phase I/II www.clinicaltrials.gov | |||
| Lucanthone | Topoisomerase II and AP endonuclease | - | Breast cancer/in vitro54 |
| ?Metformin | AMPK | - | Colon cancer/in vitro and in vivo71 |
| Antidepressants | |||
| - Tricyclic (Clomipramine) | Serotonin, norepinephrine, dopamine | - | Cervical cancer/in vitro80 |
CML, chronic myelogenous leukemia; MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasm.
An increased understanding of the autophagy pathway and its modulation holds much promise for cancer therapy and prevention. In contrast to the cytoprotective function of autophagy, induction of autophagic cell death has also been proposed as a cell death mechanism given that features of autophagy were observed in dying cells.57 Specifically, accumulation of autophagosomes has been observed in response to chemotherapy or molecularly targeted therapeutics, suggesting that autophagy induction may contribute to or accompany cell death.58 Prolonged stress and sustained autophagy can eventually lead to cell death when protein and organelle turnover overwhelm the capacity of the cell.58 A small molecule, STF-62247, was shown to promote cell death in VHL-deficient renal cell carcinomas in vitro and in vivo through chronic induction of autophagy.59 However, in vivo evidence is limited and whether induction of autophagic death in tumor cells can be achieved for cancer therapy remains unknown.
Autophagy inducers in cancer therapy.
Excessive or sustained autophagy has the potential to induce tumor cell death and may, therefore, be a potential strategy for cancer treatment. Autophagy is regulated by the mTOR pathway and mTOR inhibitors can activate autophagy. Rapamycin is a naturally occurring mTOR inhibitor and its analogs temsirolimus (CCI-779), everolimus (RAD-001) and deforolimus (AP-23573) selectively target mTORC1 to stimulate autophagy (Table 1). With the exception of renal cell and neuroendocrine carcinomas and lymphoma, rapamycin and its analogs have had limited success in the clinical setting.60,61 A potential explanation for their modest antitumor activity is the inability of rapamycin to inhibit mTORC2, and its ability to abrogate the S6K1-mediated negative feedback loop to the PI3K-AKT pathway that results in rebound AKT activation.62 The goal of achieving a more complete blockade of the mTOR pathway has led to the development of ATP-competitive mTOR inhibitors of both mTORC1 and mTORC2 (e.g., PP242, AZD8055, WYE132) and the dual PI3K-mTOR inhibitor NVP-BEZ235 that also inhibits PI3K. While there is preclinical evidence of antitumor activity,63–65 their effectiveness in the clinical setting has yet to be demonstrated. The limited activity of mTOR inhibitors as monotherapy has led to the evaluation of drug combinations. Rapamycin combined with cytotoxic chemotherapy was shown to enhance apoptosis in vitro and to enhance antitumor efficacy in vivo.60,61 Furthermore, rapamycin was found to potentiate radiation-induced clonogenic cell death.66 Another kinase inhibitor that promotes autophagy is imatinib, a Bcr-Abl tyrosine kinase inhibitor (TKI) that is an effective agent against chronic myelogenous leukemia (CML). Imatinib has been shown to modulate autophagy through the regulation of lysosomal components.67 Other autophagy modulators include antidepressants such as the selective serotonin reuptake inhibitor, fluoxetine and the norepinephrine reuptake inhibitor, maprotiline. Both drugs were shown to induce autophagy in chemoresistant Burkitt's lymphoma cell lines68 (Table 1).
The anti-diabetic, biguanide drug metformin has been shown to inhibit mTOR signaling through its upstream mediator, AMPK69 (Table 1). Metformin has a cytostatic effect on a variety of cancer cell types.69–71 In prostate cancer cells, metformin inhibited 2-deoxyglucose-induced autophagy, decreased Beclin 1 expression and triggered a switch from cell survival to cell death.72 This role contradicts the effect of metformin which is an activator of AMPK and an inhibitor of mTOR that is expected to induce autophagy. However, only one study shows that metformin induces autophagy in cancer cells,71 suggesting that depending upon the cell type, activation of AMPK does not always lead to the induction of autophagy.72
Autophagy can be activated by pro-angiogenic HIF-1α in the tumor microenvironment in the presence or absence of hypoxia.18,46 HIF-1α can increase expression of vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF) and nitric oxide synthase (NOS).18 The mTOR inhibitor, WYE132, was shown to disrupt the accumulation of HIF-1α and HIF-2α, and its combination with the anti-VEGF antibody, bevacizumab, was shown to be highly effective in promoting tumor regression in renal cell carcinoma xenografts.73 Since mTOR inhibitors have been successfully used in combination with epidermal growth factor receptor (EGFR) TKIs or bevacizumab,74 the triple combination of an mTOR inhibitor together with a TKI and bevacizumab may potentially lead to an even greater antitumor effect.
Autophagy inhibitors in cancer therapy.
Preclinical studies have shown that inhibition of cytoprotective autophagy by genetic or pharmacological means can overcome treatment resistance.75–77 Autophagy inhibitors can be broadly classified as early versus late stage inhibitors of the pathway. Early stage inhibitors include 3-methyadenine (3-MA), wortmannnin and LY294002 that target the class III PI3K (Vps34), while late stage inhibitors include chloroquine (CQ) or hydrochloroquine (HCQ), bafilomycin A1 and monensin that prevent fusion of autophagosomes with the lysosomes (Table 1). Bafilomycin A1 is a specific inhibitor of vacuolar-ATPase while monensin and CQ/HCQ are lysosomotropic drugs that prevent the acidification of lysosomal compartment. In CML cells, combined treatment with CQ and the histone deacetylase (HDAC) inhibitor vorinostat or suberoylanilide hydroxamic acid (SAHA) induced cell death in association with an increase of cathepsin D.78 Recently, a small molecule targeting HSP70, a multifunctional and stress-induced molecular chaperone, was found to disrupt lysosomal function and autophagy. This HSP70 inhibitor induced caspase-independent cell death and suppressed lymphomagenesis in a murine model.79 The tricyclic antidepressant drug, clomipramine has been shown inhibit autophagy by blocking autophagosome degradation.80
The inhibition of autophagy combined with chemotherapy may enhance treatment efficacy by inhibiting stress adaptation and increasing cell death. Of the known autophagy inhibitors, only CQ/HCQ have been evaluated in humans given their common usage as anti-malarial drugs and in autoimmune disorders. HCQ is preferred to CQ in humans given its more favorable side effect profile.81 In preclinical studies, CQ/HCQ have been shown to effectively inhibit autophagy and to augment cell death and/or tumor regression.50,78,82,83 In a Myc-induced murine lymphoma model, inhibition of autophagy by CQ was shown to enhance cyclophosphamide-induced tumor cell death to a similar degree as did shRNA knockdown of Atg5, and delayed the time-to-tumor recurrence.82 In a p53 mutated colon cancer xenograft model, 5-fluorouracil (5-FU) in combination with the autophagy inhibitor 3-MA increased caspase-3 cleavage and reduced overall tumor weight and volume as compared to 5-FU alone.84 The combination of CQ and the HDAC inhibitor vorinostat was also shown to significantly reduce tumor burden and to induce apoptosis in a colon cancer xenograft model.83 Similarly, CQ in combination with saracatinib, a Src inhibitor, produced a 2-fold increase in apoptotic tumor cells compared to saracatinib alone.50 Together, these data indicate that autophagy inhibition can enhance tumor cell death by anticancer drugs that utilizing diverse cellular mechanisms. Given these data, there are several ongoing phase I/II trials evaluating the combination of HCQ with cytotoxic drugs in patients with brain, lung, breast, colorectal, pancreas, kidney and prostate cancers43 (www.clinicaltrials.gov).
Protein degradation occurs through autophagy in lysosomes, but also within the proteasome for ubiquitinated proteins.85 Since autophagy and the proteosome are the two main protein degradation pathways, it has been postulated that their combined blockade may lead to ER stress-induced cytotoxicity through the accumulation of unfolded protein aggregates.43 Unfolded proteins can stimulate the unfolded protein response (UPR) to activate autophagy through JNK86 or PERK/eIF2α.87,88 The combination of bortezomib and CQ was shown to suppress tumor growth to a greater extent than did either drug alone in a colon cancer xenograft model.89 Phase I/II clinical trials evaluating this combination are ongoing in patients with relapsed/refractory multiple myeloma.
Evidence indicates that exploiting the interplay between apoptosis and autophagy may represent a promising therapeutic strategy. TNF-related apoptosis-inducing ligand (TRAIL) is a cytokine known to induce apoptosis in multiple cancer cell lines, and is being evaluated in phase I/II studies.90 TRAIL induced autophagy in apoptosis-defective leukemic and colon cancer cell lines, and an autophagy inhibitor sensitized resistant cells to TRAIL-mediated apoptosis.91 Anti-apoptotic Bcl-2/Bcl-xL proteins are capable of binding to and disrupting the autophagic function of Beclin 1 which contains a functional BH3 domain.92 Bcl-2/Bcl-xL can inhibit both apoptosis and autophagy and small molecule inhibitors of Bcl-2/Bcl-xL, i.e., BH3 mimetics, can competitively disrupt the Beclin 1-Bcl-2/Bcl-xL interaction to induce autophagy.93 These data suggest that inhibition of cytoprotective autophagy by BH3 mimetics may further enhance apoptosis. In this regard, the selective cyclooxygenase-2 inhibitor, celecoxib, was shown to induce apoptosis that was enhanced by the BH3 mimetic, ABT-737, and was further augmented by inhibition of autophagy in colon cancer cells.55
Conclusion
Strategies to exploit autophagy for therapeutic advantage against cancer is an area of intense investigation. Autophagy plays a dual role as both a mechanism of tumor suppression and as an adaptive stress response that can facilitate tumor cell survival and progression. Defective autophagy may contribute to cancer development in that heterozygous deletion of the Beclin 1 gene has been found in human breast, ovarian and prostate cancers. Given its role in tumor suppression, activation of autophagy may also be an important strategy for cancer prevention. In established cancers, autophagy enables tumor cells to survive increased metabolic demands, a hypoxic microenvironment and cancer therapy. Modulation of autophagy may represent a new paradigm for cancer treatment that is relevant to both conventional cytotoxic drugs and to targeted agents. Considerable evidence supports a cytoprotective role for autophagy in cancer cells, and autophagy inhibition has been shown to enhance chemosensitivity and antitumor efficacy in multiple tumor cell types both in vitro and in vivo. While multiple drugs can inhibit autophagy, most lack specificity and antitumor activity. Autophagy inhibition by hydroxychloroquine in combination with chemotherapy is currently being evaluated in multiple ongoing clinical trials in patients with solid tumors. Certain anticancer drugs can induce autophagy, and excessive or sustained induction of autophagy may lead to tumor cell death in some contexts. While tumor cell susceptibility to autophagy may depend on tumor genotype and the therapeutic agents utilized, data are very limited and it remains unclear as to whether such a strategy will be beneficial clinically. Elucidation of the mechanisms of autophagy activation, its interplay with apoptosis and the specific mechanisms by which autophagy confers treatment resistance await further study. The outcome of studies examining autophagy inhibition in combination with cytotoxic agents is early awaited. However, more selective agents targeting autophagy are needed and represent a high priority for cancer drug development.
References
- 1.Yang Z, Klionsky DJ. Eaten alive: a history of macroautophagy. Nat Cell Biol. 2010;12:814–822. doi: 10.1038/ncb0910-814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. 2003;112:1809–1820. doi: 10.1172/JCI20039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Yue Z, Jin S, Yang C, Levine AJ, Heintz N. Beclin 1. An autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci USA. 2003;100:15077–15082. doi: 10.1073/pnas.2436255100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, et al. Autophagy suppresses tumorigenesis through elimination of p62. Cell. 2009;137:1062–1075. doi: 10.1016/j.cell.2009.03.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation and tumorigenesis. Cancer Cell. 2006;10:51–64. doi: 10.1016/j.ccr.2006.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, et al. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell. 2005;120:237–248. doi: 10.1016/j.cell.2004.11.046. [DOI] [PubMed] [Google Scholar]
- 7.Gonzalez-Polo RA, Boya P, Pauleau AL, Jalil A, Larochette N, Souquere S, et al. The apoptosis/autophagy paradox: autophagic vacuolization before apoptotic death. J Cell Sci. 2005;118:3091–3102. doi: 10.1242/jcs.02447. [DOI] [PubMed] [Google Scholar]
- 8.Shimizu S, Kanaseki T, Mizushima N, Mizuta T, Arakawa-Kobayashi S, Thompson CB, et al. Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes. Nat Cell Biol. 2004;6:1221–1228. doi: 10.1038/ncb1192. [DOI] [PubMed] [Google Scholar]
- 9.Bursch W, Ellinger A, Kienzl H, Torok L, Pandey S, Sikorska M, et al. Active cell death induced by the anti-estrogens tamoxifen and ICI 164 384 in human mammary carcinoma cells (MCF-7) in culture: the role of autophagy. Carcinogenesis. 1996;17:1595–1607. doi: 10.1093/carcin/17.8.1595. [DOI] [PubMed] [Google Scholar]
- 10.Yu L, Alva A, Su H, Dutt P, Freundt E, Welsh S, et al. Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8. Science. 2004;304:1500–1502. doi: 10.1126/science.1096645. [DOI] [PubMed] [Google Scholar]
- 11.Pattingre S, Espert L, Biard-Piechaczyk M, Codogno P. Regulation of macroautophagy by mTOR and Beclin 1 complexes. Biochimie. 2008;90:313–323. doi: 10.1016/j.biochi.2007.08.014. [DOI] [PubMed] [Google Scholar]
- 12.Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000;19:5720–5728. doi: 10.1093/emboj/19.21.5720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research. Cell. 2010;140:313–326. doi: 10.1016/j.cell.2010.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, et al. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 2007;282:24131–24145. doi: 10.1074/jbc.M702824200. [DOI] [PubMed] [Google Scholar]
- 15.Eskelinen EL. Maturation of autophagic vacuoles in Mammalian cells. Autophagy. 2005;1:1–10. doi: 10.4161/auto.1.1.1270. [DOI] [PubMed] [Google Scholar]
- 16.Jung CH, Ro SH, Cao J, Otto NM, Kim DH. mTOR regulation of autophagy. FEBS Lett. 2010;584:1287–1295. doi: 10.1016/j.febslet.2010.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2002;2:489–501. doi: 10.1038/nrc839. [DOI] [PubMed] [Google Scholar]
- 18.Semenza GL. HIF-1: upstream and downstream of cancer metabolism. Curr Opin Genet Dev. 2010;20:51–56. doi: 10.1016/j.gde.2009.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sabatini DM. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer. 2006;6:729–734. doi: 10.1038/nrc1974. [DOI] [PubMed] [Google Scholar]
- 20.Chan EY, Longatti A, McKnight NC, Tooze SA. Kinase-inactivated ULK proteins inhibit autophagy via their conserved C-terminal domains using an Atg13-independent mechanism. Mol Cell Biol. 2009;29:157–171. doi: 10.1128/MCB.01082-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Jung CH, Jun CB, Ro SH, Kim YM, Otto NM, Cao J, et al. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol Biol Cell. 2009;20:1992–2003. doi: 10.1091/mbc.E08-12-1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Shen J, Chen X, Hendershot L, Prywes R. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Dev Cell. 2002;3:99–111. doi: 10.1016/s1534-5807(02)00203-4. [DOI] [PubMed] [Google Scholar]
- 23.Okada T, Yoshida H, Akazawa R, Negishi M, Mori K. Distinct roles of activating transcription factor 6 (ATF6) and double-stranded RNA-activated protein kinase-like endoplasmic reticulum kinase (PERK) in transcription during the mammalian unfolded protein response. Biochem J. 2002;366:585–594. doi: 10.1042/BJ20020391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Criollo A, Senovilla L, Authier H, Maiuri MC, Morselli E, Vitale I, et al. The IKK complex contributes to the induction of autophagy. EMBO J. 2010;29:619–631. doi: 10.1038/emboj.2009.364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mazure NM, Pouyssegur J. Hypoxia-induced autophagy: cell death or cell survival? Curr Opin Cell Biol. 2010;22:177–180. doi: 10.1016/j.ceb.2009.11.015. [DOI] [PubMed] [Google Scholar]
- 26.Maiuri MC, Malik SA, Morselli E, Kepp O, Criollo A, Mouchel PL, Carnuccio R, Kroemer G. Stimulation of autophagy by the p53 target gene Sestrin2. Cell Cycle. 2009;8:1571–1576. doi: 10.4161/cc.8.10.8498. [DOI] [PubMed] [Google Scholar]
- 27.Tasdemir E, Maiuri MC, Galluzzi L, Vitale I, Djavaheri-Mergny M, D'Amelio M, et al. Regulation of autophagy by cytoplasmic p53. Nat Cell Biol. 2008;10:676–687. doi: 10.1038/ncb1730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Alexander A, Cai SL, Kim J, Nanez A, Sahin M, MacLean KH, 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. doi: 10.1073/pnas.0913860107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tang D, Kang R, Livesey KM, Cheh CW, Farkas A, Loughran P, et al. Endogenous HMGB1 regulates autophagy. J Cell Biol. 2010;190:881–892. doi: 10.1083/jcb.200911078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 2007;8:741–752. doi: 10.1038/nrm2239. [DOI] [PubMed] [Google Scholar]
- 31.Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature. 2006;441:885–889. doi: 10.1038/nature04724. [DOI] [PubMed] [Google Scholar]
- 32.Pua HH, Dzhagalov I, Chuck M, Mizushima N, He YW. A critical role for the autophagy gene Atg5 in T cell survival and proliferation. J Exp Med. 2007;204:25–31. doi: 10.1084/jem.20061303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kanzawa T, Zhang L, Xiao L, Germano IM, Kondo Y, Kondo S. Arsenic trioxide induces autophagic cell death in malignant glioma cells by upregulation of mitochondrial cell death protein BNIP3. Oncogene. 2005;24:980–991. doi: 10.1038/sj.onc.1208095. [DOI] [PubMed] [Google Scholar]
- 34.Opipari AW, Jr, Tan L, Boitano AE, Sorenson DR, Aurora A, Liu JR. Resveratrol-induced autophagocytosis in ovarian cancer cells. Cancer Res. 2004;64:696–703. doi: 10.1158/0008-5472.can-03-2404. [DOI] [PubMed] [Google Scholar]
- 35.Shao Y, Gao Z, Marks PA, Jiang X. Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci USA. 2004;101:18030–18035. doi: 10.1073/pnas.0408345102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Jin Z, Li Y, Pitti R, Lawrence D, Pham VC, Lill JR, et al. Cullin3-based polyubiquitination and p62-dependent aggregation of caspase-8 mediate extrinsic apoptosis signaling. Cell. 2009;137:721–735. doi: 10.1016/j.cell.2009.03.015. [DOI] [PubMed] [Google Scholar]
- 37.Tang D, Kang R, Cheh CW, Livesey KM, Liang X, Schapiro NE, et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene. 2010 doi: 10.1038/onc.2010.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Aita VM, Liang XH, Murty VV, Pincus DL, Yu W, Cayanis E, et al. Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics. 1999;59:59–65. doi: 10.1006/geno.1999.5851. [DOI] [PubMed] [Google Scholar]
- 39.Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H, et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature. 1999;402:672–676. doi: 10.1038/45257. [DOI] [PubMed] [Google Scholar]
- 40.Zatloukal K, French SW, Stumptner C, Strnad P, Harada M, Toivola DM, et al. From Mallory to Mallory-Denk bodies: what, how and why? Exp Cell Res. 2007;313:2033–2049. doi: 10.1016/j.yexcr.2007.04.024. [DOI] [PubMed] [Google Scholar]
- 41.Moscat J, Diaz-Meco MT. p62 at the crossroads of autophagy, apoptosis and cancer. Cell. 2009;137:1001–1004. doi: 10.1016/j.cell.2009.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–195. doi: 10.1038/nature00858. [DOI] [PubMed] [Google Scholar]
- 43.White E, DiPaola RS. The double-edged sword of autophagy modulation in cancer. Clin Cancer Res. 2009;15:5308–5316. doi: 10.1158/1078-0432.CCR-07-5023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lu Z, Luo RZ, Lu Y, Zhang X, Yu Q, Khare S, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J Clin Invest. 2008;118:3917–3929. doi: 10.1172/JCI35512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Karantza-Wadsworth V, Patel S, Kravchuk O, Chen G, Mathew R, Jin S, et al. Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev. 2007;21:1621–1635. doi: 10.1101/gad.1565707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhang H, Bosch-Marce M, Shimoda LA, Tan YS, Baek JH, Wesley JB, et al. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem. 2008;283:10892–10903. doi: 10.1074/jbc.M800102200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 47.Papandreou I, Lim AL, Laderoute K, Denko NC. Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3 and BNIP3L. Cell Death Differ. 2008;15:1572–1581. doi: 10.1038/cdd.2008.84. [DOI] [PubMed] [Google Scholar]
- 48.Ertmer A, Huber V, Gilch S, Yoshimori T, Erfle V, Duyster J, et al. The anticancer drug imatinib induces cellular autophagy. Leukemia. 2007;21:936–942. doi: 10.1038/sj.leu.2404606. [DOI] [PubMed] [Google Scholar]
- 49.Takeuchi H, Kanzawa T, Kondo Y, Kondo S. Inhibition of platelet-derived growth factor signalling induces autophagy in malignant glioma cells. Br J Cancer. 2004;90:1069–1075. doi: 10.1038/sj.bjc.6601605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wu Z, Chang PC, Yang JC, Chu CY, Wang LY, Chen NT, et al. Autophagy blockade sensitizes prostate cancer cells towards Src family kinase inhibitors. Genes Cancer. 2010;1:40–49. doi: 10.1177/1947601909358324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Li X, Fan Z. The epidermal growth factor receptor antibody cetuximab induces autophagy in cancer cells by downregulating HIF-1alpha and Bcl-2 and activating the beclin 1/hVps34 complex. Cancer Res. 2010;70:5942–5952. doi: 10.1158/0008-5472.CAN-10-0157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zhu K, Dunner K, Jr, McConkey DJ. Proteasome inhibitors activate autophagy as a cytoprotective response in human prostate cancer cells. Oncogene. 2010;29:451–462. doi: 10.1038/onc.2009.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kanzawa T, Kondo Y, Ito H, Kondo S, Germano I. Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res. 2003;63:2103–2108. [PubMed] [Google Scholar]
- 54.Carew JS, Espitia CM, Esquivel JA, 2nd, Mahalingam D, Kelly KR, Reddy G, et al. Lucanthone: A novel inhibitor of autophagy that induces cathepsin D-mediated apoptosis. J Biol Chem. 2010 doi: 10.1074/jbc.M110.151324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Huang S, Sinicrope FA. Celecoxib-induced apoptosis is enhanced by ABT-737 and by inhibition of autophagy in human colorectal cancer cells. Autophagy. 2010;6:256–269. doi: 10.4161/auto.6.2.11124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gills JJ, Lopiccolo J, Dennis PA. Nelfinavir, a new anticancer drug with pleiotropic effects and many paths to autophagy. Autophagy. 2008;4:107–109. doi: 10.4161/auto.5224. [DOI] [PubMed] [Google Scholar]
- 57.Kroemer G, Levine B. Autophagic cell death: the story of a misnomer. Nat Rev Mol Cell Biol. 2008;9:1004–1010. doi: 10.1038/nrm2527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Mathew R, Karantza-Wadsworth V, White E. Role of autophagy in cancer. Nat Rev Cancer. 2007;7:961–967. doi: 10.1038/nrc2254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Turcotte S, Chan DA, Sutphin PD, Hay MP, Denny WA, Giaccia AJ. A molecule targeting VHL-deficient renal cell carcinoma that induces autophagy. Cancer Cell. 2008;14:90–102. doi: 10.1016/j.ccr.2008.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Meric-Bernstam F, Gonzalez-Angulo AM. Targeting the mTOR signaling network for cancer therapy. J Clin Oncol. 2009;27:2278–2287. doi: 10.1200/JCO.2008.20.0766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Abraham RT, Gibbons JJ. The mammalian target of rapamycin signaling pathway: twists and turns in the road to cancer therapy. Clin Cancer Res. 2007;13:3109–3114. doi: 10.1158/1078-0432.CCR-06-2798. [DOI] [PubMed] [Google Scholar]
- 62.O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66:1500–1508. doi: 10.1158/0008-5472.CAN-05-2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yu K, Shi C, Toral-Barza L, Lucas J, Shor B, Kim JE, et al. Beyond rapalog therapy: preclinical pharmacology and antitumor activity of WYE-125132, an ATP-competitive and specific inhibitor of mTORC1 and mTORC2. Cancer Res. 2010;70:621–631. doi: 10.1158/0008-5472.CAN-09-2340. [DOI] [PubMed] [Google Scholar]
- 64.Chresta CM, Davies BR, Hickson I, Harding T, Cosulich S, Critchlow SE, et al. AZD8055 is a potent, selective and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res. 2010;70:288–298. doi: 10.1158/0008-5472.CAN-09-1751. [DOI] [PubMed] [Google Scholar]
- 65.Feldman ME, Apsel B, Uotila A, Loewith R, Knight ZA, Ruggero D, et al. Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol. 2009;7:38. doi: 10.1371/journal.pbio.1000038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Paglin S, Lee NY, Nakar C, Fitzgerald M, Plotkin J, Deuel B, et al. Rapamycin-sensitive pathway regulates mitochondrial membrane potential, autophagy and survival in irradiated MCF-7 cells. Cancer Res. 2005;65:11061–11070. doi: 10.1158/0008-5472.CAN-05-1083. [DOI] [PubMed] [Google Scholar]
- 67.Yogalingam G, Pendergast AM. Abl kinases regulate autophagy by promoting the trafficking and function of lysosomal components. J Biol Chem. 2008;283:35941–35953. doi: 10.1074/jbc.M804543200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Cloonan SM, Williams DC. The antidepressants maprotiline and fluoxetine induce Type II autophagic cell death in drug-resistant Burkitt's lymphoma. Int J Cancer. 2010 doi: 10.1002/ijc.25477. In press. [DOI] [PubMed] [Google Scholar]
- 69.Dowling RJ, Zakikhani M, Fantus IG, Pollak M, Sonenberg N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 2007;67:10804–10812. doi: 10.1158/0008-5472.CAN-07-2310. [DOI] [PubMed] [Google Scholar]
- 70.Ben Sahra I, Laurent K, Loubat A, Giorgetti-Peraldi S, Colosetti P, Auberger P, et al. The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene. 2008;27:3576–3586. doi: 10.1038/sj.onc.1211024. [DOI] [PubMed] [Google Scholar]
- 71.Buzzai M, Jones RG, Amaravadi RK, Lum JJ, DeBerardinis RJ, Zhao F, et al. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res. 2007;67:6745–6752. doi: 10.1158/0008-5472.CAN-06-4447. [DOI] [PubMed] [Google Scholar]
- 72.Ben Sahra I, Laurent K, Giuliano S, Larbret F, Ponzio G, Gounon P, et al. Targeting cancer cell metabolism: the combination of metformin and 2-deoxyglucose induces p53-dependent apoptosis in prostate cancer cells. Cancer Res. 2010;70:2465–2475. doi: 10.1158/0008-5472.CAN-09-2782. [DOI] [PubMed] [Google Scholar]
- 73.Yu K, Shi C, Toral-Barza L, Lucas J, Shor B, Kim JE, et al. Beyond rapalog therapy: preclinical pharmacology and antitumor activity of WYE-125132, an ATP-competitive and specific inhibitor of mTORC1 and mTORC2. Cancer Res. 2010;70:621–631. doi: 10.1158/0008-5472.CAN-09-2340. [DOI] [PubMed] [Google Scholar]
- 74.LoPiccolo J, Blumenthal GM, Bernstein WB, Dennis PA. Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat. 2008;11:32–50. doi: 10.1016/j.drup.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kanzawa T, Germano IM, Komata T, Ito H, Kondo Y, Kondo S. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ. 2004;11:448–457. doi: 10.1038/sj.cdd.4401359. [DOI] [PubMed] [Google Scholar]
- 76.Boya P, Gonzalez-Polo RA, Casares N, Perfettini JL, Dessen P, Larochette N, et al. Inhibition of macroautophagy triggers apoptosis. Mol Cell Biol. 2005;25:1025–1040. doi: 10.1128/MCB.25.3.1025-1040.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Paglin S, Hollister T, Delohery T, Hackett N, McMahill M, Sphicas E, et al. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 2001;61:439–444. [PubMed] [Google Scholar]
- 78.Carew JS, Nawrocki ST, Kahue CN, Zhang H, Yang C, Chung L, et al. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood. 2007;110:313–322. doi: 10.1182/blood-2006-10-050260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Leu JI, Pimkina J, Frank A, Murphy ME, George DL. A small molecule inhibitor of inducible heat shock protein 70. Mol Cell. 2009;36:15–27. doi: 10.1016/j.molcel.2009.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Rossi M, Munarriz ER, Bartesaghi S, Milanese M, Dinsdale D, Guerra-Martin MA, et al. Desmethylclomipramine induces the accumulation of autophagy markers by blocking autophagic flux. J Cell Sci. 2009;122:3330–3339. doi: 10.1242/jcs.048181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ruiz-Irastorza G, Ramos-Casals M, Brito-Zeron P, Khamashta MA. Clinical efficacy and side effects of antimalarials in systemic lupus erythematosus: a systematic review. Ann Rheum Dis. 2010;69:20–28. doi: 10.1136/ard.2008.101766. [DOI] [PubMed] [Google Scholar]
- 82.Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, et al. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest. 2007;117:326–336. doi: 10.1172/JCI28833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Carew JS, Medina EC, Esquivel JA, 2nd, Mahalingam D, Swords R, Kelly K, et al. Autophagy inhibition enhances vorinostat-induced apoptosis via ubiquitinated protein accumulation. J Cell Mol Med. 2010;14:2448–2459. doi: 10.1111/j.1582-4934.2009.00832.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Li J, Hou N, Faried A, Tsutsumi S, Kuwano H. Inhibition of autophagy augments 5-fluorouracil chemotherapy in human colon cancer in vitro and in vivo model. Eur J Cancer. 2010;46:1900–1909. doi: 10.1016/j.ejca.2010.02.021. [DOI] [PubMed] [Google Scholar]
- 85.Korolchuk VI, Mansilla A, Menzies FM, Rubinsztein DC. Autophagy inhibition compromises degradation of ubiquitin-proteasome pathway substrates. Mol Cell. 2009;33:517–527. doi: 10.1016/j.molcel.2009.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ogata M, Hino S, Saito A, Morikawa K, Kondo S, Kanemoto S, et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol. 2006;26:9220–9231. doi: 10.1128/MCB.01453-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, et al. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 2007;14:230–239. doi: 10.1038/sj.cdd.4401984. [DOI] [PubMed] [Google Scholar]
- 88.Travers KJ, Patil CK, Wodicka L, Lockhart DJ, Weissman JS, Walter P. Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ER-associated degradation. Cell. 2000;101:249–258. doi: 10.1016/s0092-8674(00)80835-1. [DOI] [PubMed] [Google Scholar]
- 89.Ding WX, Ni HM, Gao W, Chen X, Kang JH, Stolz DB, et al. Oncogenic transformation confers a selective susceptibility to the combined suppression of the proteasome and autophagy. Mol Cancer Ther. 2009;8:2036–2045. doi: 10.1158/1535-7163.MCT-08-1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.de Vries EG, Gietema JA, de Jong S. Tumor necrosis factor-related apoptosis-inducing ligand pathway and its therapeutic implications. Clin Cancer Res. 2006;12:2390–2393. doi: 10.1158/1078-0432.CCR-06-0352. [DOI] [PubMed] [Google Scholar]
- 91.Han J, Hou W, Goldstein LA, Lu C, Stolz DB, Yin XM, et al. Involvement of protective autophagy in TRAIL resistance of apoptosis-defective tumor cells. J Biol Chem. 2008;283:19665–19677. doi: 10.1074/jbc.M710169200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, et al. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122:927–939. doi: 10.1016/j.cell.2005.07.002. [DOI] [PubMed] [Google Scholar]
- 93.Kessel D, Reiners JJ., Jr Initiation of apoptosis and autophagy by the Bcl-2 antagonist HA14-1. Cancer Lett. 2007;249:294–299. doi: 10.1016/j.canlet.2006.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Takeuchi H, Kondo Y, Fujiwara K, Kanzawa T, Aoki H, Mills GB, et al. Synergistic augmentation of rapamycin-induced autophagy in malignant glioma cells by phosphatidylinositol-3-kinase/protein kinase B inhibitors. Cancer Res. 2005;65:3336–3346. doi: 10.1158/0008-5472.CAN-04-3640. [DOI] [PubMed] [Google Scholar]
- 95.Crazzolara R, Bradstock KF, Bendall LJ. RAD001 (Everolimus) induces autophagy in acute lymphoblastic leukemia. Autophagy. 2009;5:727–728. doi: 10.4161/auto.5.5.8507. [DOI] [PubMed] [Google Scholar]
- 96.Yazbeck VY, Buglio D, Georgakis GV, Li Y, Iwado E, Romaguera JE, et al. Temsirolimus downregulates p21 without altering cyclin D1 expression and induces autophagy and synergizes with vorinostat in mantle cell lymphoma. Exp Hematol. 2008;36:443–450. doi: 10.1016/j.exphem.2007.12.008. [DOI] [PubMed] [Google Scholar]
- 97.Fu L, Kim YA, Wang X, Wu X, Yue P, Lonial S, et al. Perifosine inhibits mammalian target of rapamycin signaling through facilitating degradation of major components in the mTOR axis and induces autophagy. Cancer Res. 2009;69:8967–8976. doi: 10.1158/0008-5472.CAN-09-2190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Evangelisti C, Ricci F, Tazzari P, Chiarini F, Battistelli M, Falcieri E, et al. Preclinical testing of the AKT inhibitor triciribine in T-cell acute lymphoblastic leukemia. J Cell Physiol. 2011;226:822–831. doi: 10.1002/jcp.22407. [DOI] [PubMed] [Google Scholar]
- 99.Liu YL, Yang PM, Shun CT, Wu MS, Weng JR, Chen CC. Autophagy potentiates the anti-cancer effects of the histone deacetylase inhibitors in hepatocellular carcinoma. Autophagy. 2010;6:1057–1065. doi: 10.4161/auto.6.8.13365. [DOI] [PubMed] [Google Scholar]
- 100.Wei Y, Kadia T, Tong W, Zhang M, Jia Y, Yang H, et al. The combination of a histone deacetylase inhibitor with the Bcl-2 homology domain-3 mimetic GX15-070 has synergistic antileukemia activity by activating both apoptosis and autophagy. Clin Cancer Res. 2010;16:3923–3932. doi: 10.1158/1078-0432.CCR-10-0032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Rosenfeld MR, Brem S, Mikkelsen T, Wang D, Piao S, Davis LE, et al. Pharmacokinetic analysis and pharmacodynamic evidence of autophagy inhibition in patients with newly diagnosed glioblastoma treated on a phase I trial of hydroxychloroquine in combination with adjuvant temozolomide and radiation (ABTC 0603) J Clin Oncol. 2010:28. [Google Scholar]
- 102.Bellodi C, Lidonnici MR, Hamilton A, Helgason GV, Soliera AR, Ronchetti M, et al. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin Invest. 2009;119:1109–1123. doi: 10.1172/JCI35660. [DOI] [PMC free article] [PubMed] [Google Scholar]

