Skip to main content
Cancer Biology & Therapy logoLink to Cancer Biology & Therapy
. 2011 Jan 15;11(2):157–168. doi: 10.4161/cbt.11.2.14622

Autophagy as a therapeutic target in cancer

Ning Chen 2,3, Vassiliki Karantza 1,2,3,
PMCID: PMC3230307  PMID: 21228626

Abstract

Autophagy is a self-catabolic process that maintains intracellular homeostasis and prolongs cell survival under stress via lysosomal degradation of cytoplasmic constituents and recycling of amino acids and energy. Autophagy is intricately involved in many aspects of human health and disease, including cancer. Autophagy is a double-edged sword in tumorigenesis, acting both as a tumor suppressor and a protector of cancer cell survival, and elucidation of its exact role at different stages of cancer progression and in treatment responsiveness is a complex and challenging task. Better understanding of autophagy regulation and its impact on treatment outcome will potentially allow us to identify novel therapeutic targets in cancer. In this review, we summarize current knowledge on the regulation and dual function of autophagy in tumorigenesis, as well as ongoing efforts in modulating autophagy for cancer treatment and prevention. This is a very exciting and highly promising area of cancer research, as pharmacologic modulation of autophagy appears to augment the efficacy of currently available anticancer regimens and opens the way to the development of new combinatorial therapeutic strategies that will hopefully contribute to cancer eradication.

Key words: autophagy, tumorigenesis, chemotherapy, radiation, cancer treatment

Introduction

Macroautophagy, hereafter referred to as autophagy, is an evolutionarily conserved, genetically controlled cell survival pathway involving the degradation of cytoplasmic constituents, and the recycling of ATP and essential building blocks for the maintenance of cellular biosynthesis during nutrient deprivation or metabolic stress.1 This cellular self-consumption process is characterized by sequestration of bulk cytoplasm, long-lived proteins and cellular organelles in double-membrane vesicles, called autophagosomes, which are ultimately delivered to and degraded in the lysosomes.2 Autophagy is intricately implicated in both health and disease.3 Basal autophagy plays a critical role in cellular homeostasis by eliminating excessive, damaged and/or long-lived proteins and organelles, thus preserving the quality of essential cellular components.4,5 Under stress, autophagy is commonly induced as a temporary cell survival mechanism. Autophagy defects play a role in the pathogenesis of diverse diseases, including myopathy,6 neuronal degeneration,7 microbial infection,8 inflammatory bowel disease,9,10 aging11 and cancer.12 Recent studies have shed light on the functional role of autophagy in different cellular processes and the potential of autophagy modulation as a novel therapeutic strategy for different pathologic conditions, including cancer.13,14 Anticancer therapies, such as hormonal agents, chemotherapy and irradiation, frequently induce autophagy, in most cases as a prosurvival response potentially contributing to treatment resistance;1517 however, autophagy activation in particular genetic backgrounds and/or completion of the autophagic process beyond reversibility of cell viability can also lead to cell death, thus enhancing treatment efficacy.18 The complex role of autophagy in tumorigenesis and treatment responsiveness makes it hard to decipher how to universally modulate autophagy for maximum therapeutic benefit, indicating that context- and cell type-specific approaches may be required.

In this review, we present the current knowledge on autophagy regulation, its role as a double-edged sword in tumorigenesis, and the implications of pharmacologic autophagy modulation for cancer treatment. Therapeutic regimens potentially altering the functional status of autophagy in tumors will also be reviewed, hopefully providing a reference for the future development of combinatorial treatments involving autophagy modulation as a cancer therapeutic and/or preventative strategy.

Regulation of Autophagy in Cancer

In normal cells, autophagy is regulated by a cellular network that involves upstream signaling pathways integrated by the mammalian target of rapamycin (mTOR) kinase, a master regulator.12,14,19 Nutrient and/or growth factor availability activates the PI3K/AKT/mTOR axis, which inhibits autophagy thus stimulating cell growth and proliferation; whereas nutrient and/or growth factor limitation, hypoxia and other stressors deactivate this axis, leading to autophagy induction and suppression of cell growth and proliferation.14,20 In tumor cells, autophagy regulation is in principal similar, but the regulatory network is even more complex due to frequently abnormal PI3K/AKT/mTOR and other signaling cascade activation and interactions between these pathways.21,22

PI3K-AKT-mTOR pathway.

Abnormal, and often constitutive, activation of the PI3K-AKT-mTOR axis promoting tumor cell growth, proliferation and survival is a common occurrence in cancer.2325 This may be the result of one or multiple cellular events that are associated with cancer initiation and/or progression, such as phosphatase and tensin-homologue deleted on chromosome 10 (PTEN) mutation or loss, tuberous sclerosis complex (TSC) 1 and/or 2 mutation, tyrosine kinase growth factor receptor and/or type I PI3K mutation, AKT overexpression, mTOR inhibition and carcinogen exposure, leading to the abnormal activation of this signaling cascade and subsequent inhibition of autophagy (Fig. 1).14 PI3K mutations, such as E542K, E545D and E545K, which result in constitutive kinase activation, are frequently detected in human cancers.2629 Activated PI3K results in sequential AKT and mTOR activation, ultimately suppressing autophagy. This is in contrast to the action of a different PI3K (type III) that binds to the Beclin 1 protein forming a complex involved in autophagy induction.30

Figure 1.

Figure 1

Autophagy regulation. Growth factor signaling activates the PI3K/AKT/mTOR axis resulting in autophagy inhibition. In contrast, class III PI3K activates autophagy. Low cellular energy levels with increased AMP/ATP ratio activate the LKB1-AMPK-mTOR pathway to also upregulate autophagy. Furthermore, increased calcium ion levels induce autophagy by two mechanisms, i.e., the Ca2+-CaMKKβ-AMPK pathway and direct Ca2+-induced ER stress. p53 exhibits complex autophagy regulation, as nuclear p53 activated by genotoxic or oncogenic stress positively regulates autophagy by inhibiting mTOR in an activated AMPK- and TSC1/TSC2-dependent manner, whereas cytoplasmic p53 can suppress autophagy.

The tumor suppressor PTEN is frequently mutated or lost in human tumors, also leading to AKT activation and autophagy inhibition.3133 The serine/threonine kinase mTOR, which is a downstream target of AKT, controls a series of autophagyrelated genes and is commonly recognized as the master autophagy regulator.14,34,35 AKT activates the mTOR complex via phosphorylation of TSC2 and PRAS40 (proline-rich AKT substrate of 40 kDa).36 The deregulated PI3K/AKT/mTOR axis not only suppresses autophagy, but also induces protein translation, cell growth and proliferation, which are indisputable driving forces in tumorigenesis.24,37 Pharmacologic inhibition of the PI3K/AKT/mTOR axis for cancer treatment results in reciprocal autophagy induction, raising the critical issue of the potential dual function of autophagy activation in treatment responsiveness. Type I PI3K inhibitors, such as lithium and carbamezapine; type III PI3K inhibitors, such as 3-MA, LY294002 and wortmannin; AKT inhibitors, such as perifostine and API-2; and mTOR inhibitors, such as rapamycin, RAD001 and CCI-779; have all been reported as promising anticancer agents.14,38,39 However, the impact of drug-induced autophagy modulation on antitumor activity and how to best exploit this response for maximum therapeutic benefit are both questions that require further investigation.

LKB1-AMPK-mTOR pathway.

The LKB1-AMPK-mTOR pathway acts as a central sensor regulating lipid and carbohydrate metabolism in metabolically active tissues, such as liver, muscle and fat. Interestingly, AMPK has also been implicated in cancer cell metabolism and tumorigenesis.40,41 As the major upstream regulator, the serine/threonine kinase LKB1 (also known as STK11) activates AMPK by directly phosphorylating its α-subumit at Thr172.42,43 Low energy or metabolic stress conditions, such as nutrient and oxygen deprivation, are associated with reduced cellular ATP levels and an elevated AMP/ATP ratio, which activates the tumor suppressor LBK1, resulting in AMPK activation and, ultimately, mTOR inhibition and autophagy induction (Fig. 1).42 Furthermore, AMPK directly phosphorylates TSC2 and the regulatory associated protein of mTOR (Raptor), thus inhibiting mTOR in both TSC-independent and -dependent ways and activating autophagy as a cell survival mechanism in response to metabolic stress in an LKB1-dependent manner.44,45 Calcium and calmodulin-dependent protein kinase kinase β (CAMKKβ) are also involved in AMPK activation in hypothalamic neurons,46 T cells and endothelial cells,47,48 indicating that calcium metabolism also plays a role in LKB1-AMPK-mTOR-mediated autophagy regulation. In parallel to upregulating autophagy, activation of the LKB1-AMPK-mTOR axis results in the phosphorylation and stabilization of p27kip, a cyclin-dependent kinase inhibitor, to induce cell cycle arrest for energy conservation.49 Finally, the transforming growth factor-β-activating kinase 1 (TAK1) was recently identified as another AMPK activator.50 Further investigation of the LKB1-AMPK-mTOR signaling cascade will advance our understanding of the different mechanisms regulating autophagy and will hopefully contribute to the development of novel treatment regimens that include cancer cell metabolism as a therapeutic target.

p53.

p53 deletion and/or mutation is observed in nearly 50% of human cancers.51 The tumor suppressor p53 is normally activated under genotoxic or oncogenic stress and leads to cell cycle arrest, senescence or apoptosis.52,53 p53 has opposing effects on autophagy regulation based on its cellular localization, and thus the p53-autophagy interaction is highly context-dependent. Transcriptional activation, and thus nuclear localization, of p53 is a positive autophagy regulator by inhibiting mTOR in an activated AMPK- and TSC1/TSC2-dependent manner (Fig. 1).54 p53 also activates autophagy by triggering other downstream targets, such as sestrin and the damage-regulated autophagy modulator (DRAM).55,56 Loss of cytoplasmic p53 function by genetic or pharmacologic manipulation paradoxically activates autophagy as well, suggesting that the non-nuclear p53 pool is a potential autophagy repressor.57 Autophagy induction by p53 loss promotes p53-deficient cell survival by sustaining high ATP levels under conditions of hypoxia and nutrient depletion.58 Interestingly, although cytoplasmic p53 inhibits autophagy, nuclear p53 mutants fail to block autophagy. This dual interplay between p53 and autophagy is still under investigation. A possible explanation is that the role of p53 in autophagy regulation depends on the particular stress and microenvironment that a cell is exposed to, thus likely changing upon cancer progression. In the early stages of tumorigenesis, p53-mediated autophagy induction may act as a gatekeeper in response to genotoxic or oncogenic stress by eliminating malfunctioning cells. In advanced tumors, where p53 is commonly deleted or mutated, p53 deregulation stimulates autophagy as an amino acid and energy recycling mechanism to support tumor cell viability.59

BCL-2.

Beclin 1 (BECN1, also called ATG6) was originally identified as a novel BCL-2-interacting protein in a yeast two-hybrid system.60 The autophagic function of BECN1 1 is evolutionarily conserved from yeast to human,6166 and involves the binding of BCL-2 to other autophagy regulators, such as VPS34, p150, UVRAG, BIF1, ATG14L and Rubicon to form huge protein complexes and initiate double-membrane autophagosome formation.6771 Allelic BECN1 deletion is frequently observed in human breast, ovarian and prostate cancers and leads to tumor development in mice, whereas ectopic BECN1 expression, which restores autophagy, inhibits tumorigenesis; indicating that BECN1 is a tumor suppressor and establishing a connection between defective autophagy and cancer.62,7277

BCL-2 is an anti-apoptotic protein commonly overexpressed in breast cancers, which contributes to treatment resistance by inhibiting chemotherapy and hormonal therapy-induced apoptosis. BCL-2 is also involved in autophagy inhibition by binding to the BH3 domain of BECN1 and negatively regulating the autophagy-promoting BECN1-VPS34 complex.70 Dissociation of BECN1 from BCL-2, and thus autophagy activation, in response to nutrient limitation depends on BCL-2 phosphorylation by the starvation-activated c-JUN N-terminal kinase (JNK).78

BECN1 and BCL-2 complex formation is primarily dependent on the BH3 domain of BECN1 and the BH3-receptor domain of BCL-2.79 Pharmacologic interference with this interaction by BCL-2 knockdown, BH3-mimetics or JNK activation results in autophagy induction and provides a novel therapeutic strategy targeting cancer cells with high treatment resistance due to BCL-2 overexpression.

ER stress and Ca2+ signaling.

The endoplasmic reticulum (ER) is a cellular organelle responsible for secreted and membrane protein folding. Cellular stressors, such as hypoxia, low glucose and deregulation of calcium homeostasis, result in misfolded protein accumulation in the ER, triggering ER stress and cell death, typically via apoptosis.80,81 ER stress activates autophagy as a compensatory mechanism, whereas autophagy inhibition enhances ER stress-induced cell death.82,83

ER stress is accompanied by calcium release into the cytosol, thus activating Ca2+-regulated signaling pathways. Calcium- and CAMKKβ-dependent AMPK activation is one mechanism connecting calcium release from the ER to autophagy, as discussed earlier in this review.84, 85 Recent studies also reported autophagy induction by protein kinase-like endoplasmic reticulum kinase (PERK) and inositol-requiring kinase 1 (IRE1)-dependent mechanisms.86,87 Given the interest in ER stress inducers as potential anticancer agents, it is worthwhile to exploit how to best manipulate the resulting autophagy upregulation for maximizing anticancer benefits.

Other autophagy regulators.

The death-associated protein kinase (DAPK) is a cytokeleton-associated, calmodulin-regulated serine/threonine protein kinase.59 DAPK or DAPK-related protein kinase 1-associated autophagy is observed in many human malignancies, including B and T cell lymphomas and breast, lung, head and neck, gastric, cervical and prostate cancers. Conversely, DAPK inhibition results in autophagy suppression.88,89

IκB kinase (IKK) stimulates autophagy by multiple NFκB-independent mechanisms, including starvation-induced JNK1 and AMPK phosphorylation, and likely plays a pivotal role in autophagy induction by physiological and pharmacological stimuli.90

Role of Autophagy in Cancer: A Double-edged Sword

The dual function of autophagy in cancer, as both a tumor suppressor and a protector of cancer cell survival, has been widely recognized and remains a rigorously investigated topic.14,91 Elucidation of the specific role that autophagy plays at different stages in cancer progression and determination of its cell type and genetic context-dependency will lead to the development of novel, and hopefully more effective, cancer therapeutic and preventative strategies.

Autophagy as a tumor suppressor mechanism.

Morphological evidence of autophagosome accumulation in dying cells linked autophagy to cell death and led to the definition of an autophagosome-associated, non-apoptotic form of programmed cell death (PCD) as autophagic cell death (ACD) or type II PCD, potentially functioning as a tumor suppressor mechanism similar to apoptosis.92 The tumor suppressive role of autophagy was validated by the discovery and characterization of the autophagy-related genes (Atg),93 among which Beclin 1 (Atg6) was the first to be reported as a plausible tumor suppressor, as allelic BECN1 deletion is frequently observed in human breast, ovarian and prostate cancers,62,7377 and aging Becn1+/− mutant mice develop tumors (lymphomas, lung and liver cancers), as well as mammary hyperplasia and hepatitis B virus-induced premalignant lesions.65,66

Genetic alteration of other autophagy-related genes has also been causally associated with tumorigenesis. Atg4C knockout mice exhibit increased susceptibility to chemical carcinogen -induced fibrosarcomas,94 whereas deletion of the essential autophagy regulator Atg5 results in natural killer (NK) cell malignancies.95 Furthermore, loss of Bax-interacting factor-1 (BIF-1), which is a positive apoptosis and autophagy regulator, suppresses programmed cell death and promotes colon adenocarcinomas,96 and nonsense mutations in the BECN1-binding protein UVRAG (ultraviolet radiation resistance-associated gene), which also positively regulates autophagy, are found in colon and gastric cancers.97,98

Both cell autonomous and non-cell autonomous mechanisms have been implicated in autophagy-mediated tumor suppression,12 namely preservation of genome stability and cellular homeostasis and limitation of inflammation, respectively.99102

Autophagy as a protector of cancer cell survival.

Counterintuitive to its tumor suppressive role, autophagy has also been extensively documented to support cancer cell survival under stress.91,103 For example, autophagy induction in immortalized, apoptosis-defective, IL-3-dependent bone marrow cells in response to growth factor withdrawal prolongs cell survival, as supported by the observed cell death acceleration upon autophagy inhibition.104,105 Defective autophagy due to allelic Becn1 deletion or constitutive AKT activation enhances the susceptibility of apoptosis-incompetent immortalized baby mouse kidney (iBMK) cells to metabolic stress.99,102 Similarly, partially autophagy-defective Becn1+/− immortalized mouse mammary epithelial cells (iMMECs) are more sensitive to metabolic stress and show accelerated acinar lumen formation in 3D-culture.100,106

Autophagy-mediated support of tumor cell survival may play a critical role in cancer progression at later stages, such as dissemination and metastasis, which account for most cancer-associated deaths. In favor of this hypothesis, starvation-induced autophagy is accompanied by suppression of protein synthesis, cell division and motility in an energy conservation effort that sustains cells in a dormant state with the capacity to resume cell growth and proliferation upon regular growth condition restoration.12,91,99,107 For example, autophagy induction in mammary epithelial cells upon their detachment from extraceullular matrix (ECM) sustains cell viability in an anoikis-resistant manner, whereas autophagy upregulation in ovarian cancer cells by the tumor suppressor ARHI (aplasia Ras homolog member I) promotes dormant cell survival in vivo.108,109 Finally, many studies have clearly documented that in cancer cells, autophagy is upregulated in response to metabolic and genotoxic stress induced by hormonal deprivation, chemotherapy and radiation as a cell survival mechanism, likely contributing to treatment resistance, but also providing a novel therapeutic target in cancer.12,14,91,110112

Autophagy-mediated cancer cell survival is not unexpected given the functions of autophagy in normal cells: basal autophagy maintains cellular homeostasis by removing protein aggregates and damaged organelles, whereas starvation-induced autophagy prolongs cell survival by recycling amino acid and energy, both important functions for cellular fitness and viability preservation.4,5 Cancer cells are often under higher metabolic stress than normal cells, which in turn increases tumor dependence on autophagy for survival (autophagy addiction), implying a therapeutic window for preferential cancer cell targeting by pharmacologic autophagy modulation. Metabolic stressors uniquely encountered by tumor cells include: (1) oncogene-induced accelerated cell growth and/or proliferation, which increase metabolic demands even in the presence of ample external nutrients,113 (2) recurrent nutrient and oxygen deprivation during rapid tumor growth or anticancer treatment,14,107 and (3) inefficient glucose utilization for energy production due to anaerobic glycolysis (Warburg effect).12,114 Whether cancer cells are addicted to autophagy in an oncogene- and/or tumor type-dependent manner is currently under investigation.

Autophagy Modulation for Cancer Treatment

The goal of anticancer therapy is to effectively compromise tumor cell growth and survival, so as to cause cancer regression and prevent (or at least delay) cancer recurrence, thus improving patient quality of life and survival. Apoptosis is commonly inactivated in cancer, often in association with disease progression, and renders tumors resistant to chemotherapy- and radiation-induced cell death, undisputedly contributing to treatment resistance and earlier patient demise. During the last decade, our understanding of the proteins and molecular mechanisms responsible for apoptosis regulation, initiation and execution has expanded tremendously,14,115 leading to rationally designed approaches for apoptosis reactivation, and thus restoration of cell death potential, in cancer cells.116,117

Despite recent advances in cancer treatment, many tumors still exhibit unsatisfactory responsiveness to biological agents, chemotherapy and/or radiation, either recurring or continuing to grow during or after treatment.85,118,119 Autophagy upregulation is a common occurrence in response to cancer therapies, expected to occur in both tumor and normal cells, but likely playing a more critical role in the survival of the already metabolically stressed cancer cells, as explained above, and thus contributing to treatment resistance. At the same time, however, this prosurvival function of autophagy provides a novel therapeutic opportunity, as concurrent autophagy inhibition may preferentially sensitize tumor cells to anticancer agents by depriving them of an essential survival mechanism that may be dispensable for normal cell viability under similar conditions.91,120 Drug-induced autophagy may also be therapeutically beneficial by itself, limiting tumor cell proliferation and resulting in autophagic cell death in a cell typeand genetic background-specific manner.14,62,85 Thus, contextspecific pharmacologic autophagy modulation holds great promise as a novel therapeutic approach adding another weapon to the currently available armamentarium against cancer.14,121

Autophagy inhibition as a therapeutic strategy in cancer.

In cancer cells, autophagy is generally, and often preferentially as compared to normal cells, induced as a prosurvival function in response to treatment-associated genotoxic and metabolic stress.110,120,122 Thus, concurrent autophagy inhibition is expected to mostly have a synergistic effect with chemotherapy and/or radiation on cancer cell elimination. The impact of autophagy inhibition on anticancer therapy has been evaluated in multiple tumor models, including glioma,123,124 myeloma,125 breast,126,127 colon128,129 and prostate cancers.130 For example, and as described in more detail in Table 1, which summarizes preclinical studies supporting autophagy inhibition as an anticancer strategy, treatment-induced autophagy mediated resistance to the HER2 monoclonal antibody trastuzumab, whereas LC3 knockdown via shRNA resulted in resistant cells being re-sensitized to treatment;127 autophagy inhibition by either pharmacological agents or RNAi targeting essential autophagy regulators potentiated imatinib mesylate-induced cell death in chronic myelogenous leukemia (CML) cells;131 autophagy suppression also enhanced the therapeutic efficacy of cisplatin and 5-FU in esophageal and colon cancers, respectively.132,133

Table 1.

Preclinical studies supporting autophagy inhibition for cancer treatment

Cancer type Primary treatment (target) Autophagy inhibition method (target) References
Breast cancer Trastuzumab (HER2 receptor) 3-MA (PI3K III), BafA (lysosome), RNAi (LC3) 127
Camptothecin (DNA topoisomerase II) 3-MA (PI3K III), BafA (lysosome), RNAi (BECN1, ATG7) 126
Tamoxifen (estrogen receptor) 3-MA (PI3K III), RNAi (BECN1, ATG5, ATG7) 163
Bortezomib (26S proteasome) RNAi (LC3, ATF4, HDAC6) 164
Faslodex (estrogen receptor) RNAi (BECN1) 165
RT (DNA) RNAi (BECN1, ATG3, ATG4B, ATG4C, ATG5, ATG12) 128
Sulforaphane (histone deacetylation) BafA (lysosome) 166
Cervical cancer Bortezomib (26S proteasome) RNAi (BECN1) 167
CML Imatinib mesylate (BCR/ABL) CQ (lysosome), BafA (lysosome), RNAi (ATG5, ATG7) 131
INNO-406 (BCR/ABL) CQ (lysosome) 168
SAHA (histone deacetylation) CQ (lysosome), 3-MA (PI3K III) 169
Imatinib mesylate (BCR/ABL) BafA (lysosome), CQ (lysosome), RNAi (ATG5, ATG7) 131
Imatinib mesylate (BCR/ABL), TPA (differentiation) CQ (lysosome) 170
Colorectal cancer Vorinostat (histone deacetylation) CQ (lysosome), RNAi (ATG7) 129
RT (DNA) RNAi (BECN1, ATG3, ATG4B, ATG5) 128
Sulindac sulfide (cyclooxygenase) 3-MA (PI3K III) 171
TRAIL (TNF receptor) 3-MA (PI3K III), RNAi (BECN1, ATG5, ATG7, UVRAG) 172
5-FU (thymidylate synthase) 3-MA (PI3K III), RNAi (ATG7) 173
TFT, 5-FU (thymidylate synthase) 3-MA (PI3K III) 174
5-FU (thymidylate synthase) 3-MA (PI3K III) 132
Bortezomib (26S proteasome) RNAi (BECN1) 167
Esophageal cancer Cisplatin (DNA) 3-MA (PI3K III) 133
Gastrointestinal stromal tumor Imatinib mesylate (BCR/ABL) CQ (lysosome), RNAi (ATG5, ATG12) 175
Glioma, malignant AKTi-1/2 (AKT) CQ (lysosome) 149
PI-103 (PI3K, mTOR) CQ (lysosome) 149
Imatinib mesylate (BCR/ABL) BafA (lysosome) 176
RT (DNA) 3-MA (PI3K III), BafA (lysosome), RNAi (BECN1, ATG5) 177
RT (DNA) 3-MA (PI3K III), BafA (lysosome) 123
4-HPR (tyrosine kinase) 3-MA (PI3K III), BafA (lysosome) 178
Temozolomide, etoposide (DNA) 3-MA (PI3K III) 178
Temozolomide (DNA) 3-MA (PI3K III), BafA (lysosome) 137
Multiple myeloma 8-Amino-adenosine (AKT/mTOR) CQ (lysosome) 125
Lung cancer, non-small cell RT (DNA) RNAi (BECN1, ATG3, ATG4B, ATG4C, ATG5, ATG12) 128
Pancreatic cancer Gemcitabine, RT (DNA) 3-MA (PI3K III) 185
Pharyngeal cancer RT (DNA) RNAi (BECN1, ATG3, ATG4B, ATG4C, ATG5, ATG12) 128
Prostate cancer ADI-PEG20 (arginine) CQ (lysosome), 3-MA (PI3K III), RNAi (BECN1) 130
TRAIL, FADD (TNF receptor) 3-MA (PI3K III) 179
Androgen deprivation (androgen receptor) 3-MA (PI3K III), RNAi (BECN1) 180
Saracatinib (Src kinase) 3-MA (PI3K III), CQ (lysosome), RNAi (ATG7) 181
Rhabdoid tumor, malignant FK228 (histone deacetylation) CQ (lysosome) 182
Skin cancer, squamous cell Cisplatin (DNA) 3-MA (PI3K III), RNAi (ATG5) 183

In the absence of drugs specifically targeting autophagy regulators, indirect autophagy inhibition by the lysosomotropic antimalaria drugs chloroquine (CQ) and hydroxychloroquine (HCQ), which interfere with lysosomal acidification and thus, block the autophagic process at its final step, are currently under clinical investigation in combination with standard treatment in multiple tumor types. Table 2 summarizes such ongoing clinical trials involving CQ- and HCQ-mediated autophagy modulation for cancer therapy. Treatment with CQ or HCQ is likely not the same as direct autophagy inhibition, given that CQ and HCQ are known to exert additional functions, such as immunomodulation and possibly DNA damage as alkylating agents at higher doses. Nevertheless, the results of the above mentioned clinical trials are eagerly awaited as an initial proof-of-principle that autophagy inhibition has a role in cancer treatment. In the event that these studies fail to show the trends anticipated, before declaring autophagy inhibition as an unsuccessful therapeutic strategy in cancer, it will be necessary to assess whether tumors with particular oncogenic changes are better candidates than others for such treatment, in which case trials should be redesigned to specifically target these malignancies.

Table 2.

Ongoing clinical trials exploring autophagy inhibition for cancer treatment

Cancer type Primary treatment Autophagy inhibitor Study phase Sponsor Clinical trial identifier Title
Advanced solid tumors Sunitinib HCQ I CINJ NCT00813423 Sunitinib and HCQ in advanced solid tumors
Temozolomide HCQ I U Penn NCT00714181 Temozolomde and HCQ in metastatic or unresectable solid tumors
Temsirolimus HCQ I U Penn NCT00909831 Temsirolimus and HCQ in refractory solid tumors
Vorinostat HCQ I U Texas HSC S Ant NCT01023737 Vorinostat and HCQ in advanced solid tumors
Breast cancer Ixabepilone HCQ I/II CINJ NCT00765765 Ixabepilone and HCQ in metastatic breast cancer
CML Imatinib mesylate HCQ II U Glasgow NCT01227135 Imatinib mesylate ± HCQ in CML in major cytogenetic response with residual disease
CLL HCQ II NSLIJHS NCT00771056 HCQ in previously untreated, asymptomatic B-CLL
Colorectal cancer Capecitabine Oxaliplatin Bevacizumab HCQ II CINJ NCT01006369 XELOX, bevacizumab and HCQ in metastatic colorectal cancer
5-FU/leucovorin Oxaliplatin Bevacizumab HCQ I/II U Penn NCT01206530 FOLFOX, bevacizumab and HCQ in colorectal cancer
DCIS Tamoxifen CQ I/IIInova Health Care Serv NCT01023477 Neoadjuvant tamoxifen, tamoxifen + CQ, or CQ in DCIS
Glioblastoma multiforme Radiation Temozolomide HCQ I/II NCI NCT00486603 Adjuvant radiation, temozolomide and HCQ in newly resected GBM
CQ III NINN, Mexico NCT00224978 Adjuvant CQ versus placebo in glioblastoma
Lung cancer, nonsmall cell Carboplatin Paclitaxel Bevacizumab HCQ I/II CINJ NCT00933803 Carboplatin, paclitaxel, bevacizumab and HCQ in advanced or recurrent NSCLC
Erlotinib HCQ II Mass Gen Hosp NCT00977470 Erlotinib ± HCQ in previously untreated metastatic NSCLC with EGFR mutations
Gefitinib HCQ I/II Nat U Hosp, Singapore NCT00809237 Gefitinib and HCQ in metastatic NSCLC
Lung cancer, extensive small cell Cisplatin Etoposide CQ I/II Maastricht Rad Onc NCT00969306 Cisplatin, etoposide and escalating CQ in extensive disease SCLC
Lung cancer, limited small cell RT Cisplatin Etoposide CQ I/II Maastricht Rad Onc NCT00969306 RT, cisplatin, etoposide and escalating CQ in limited disease SCLC
Melanoma HCQ 0 CINJ NCT00962845 Neoadjuvant HCQ in stage III or IV respectable melanoma
Multiple myeloma Bortezomib HCQ I/II U Penn NCT00568880 Bortezomib and HCQ in relapsed or refractory multiple myeloma
Pancreatic cancer Gemcitabine HCQ I/II U Pittsburgh NCT01128296 Neoadjuvant gemcitabine and HCQ in Stage II b or III pancreatic cancer
Prostate cancer HCQ II CINJ NCT00726596 HCQ in patients with rising PSA after local prostate cancer treatment
Docetaxel HCQ II CINJ NCT00786682 Docetaxel and HCQ in metastatic prostate cancer
Renal cell cancer HCQ I U Pittsburgh NCT01144169 Neoadjuvant HCQ in renal cell carcinoma

Autophagy induction as an alternate anticancer strategy.

Although autophagy inhibitors combined with standard treatment are emerging as promising anticancer agents, certain cancer cell lines and xenograft tumors were sensitized to therapeutic regimens involving autophagy induction rather than inhibition. This was commonly observed in an apoptosis-defective background, where cancer cells were committed to non-apoptotic cell death modes, including necrosis, necroptosis and possibly autophagic cell death.122 Alkylating agents, such as actinomycin D and arsenic trioxide; hormonal therapies, including tamoxifen and vitamin D analogues; natural compounds, such as resveratrol; cytokines, such as IFNγ; gene therapies, including p53 and p27kip1; have all been implicated in the induction of autophagic cell death in various cancer cell lines in vitro, as autophagosomes are commonly observed in dying cells.55,134143 However, it is often unclear whether autophagy plays an active role in the cell death process or it is a mere bystander representing the stressed cell's futile attempts to preserve viability by upregulating an energy and amino acid recycling program. Thus, the presence of autophagosomes in dying cells is not necessarily synonymous with cell death by autophagy, unless knockdown of essential autophagy regulators prolongs cell survival under the same stress conditions.144 For example, the autophagy inducer STF-62247 killed VHL-deficient renal cancer cells in association with autophagosome accumulation and reduction of autophagy regulator levels compromised sensitivity to this drug.145

The implication of autophagy as a cell death mechanism in tumors with inactivated apoptosis is not surprising, given that cells with functional apoptosis undergo a rapid and ‘clean’ (not associated with inflammation) apoptotic cell death when severely stressed, making observation, and thus study, of autophagy in an apoptosis-competent background difficult. 99,111 Disabled apoptosis is a frequent occurrence in cancer, thus tumor cells under extreme stress often die by other mechanisms, as already mentioned. However, the conditions-beyond apoptosis inactivation-under which autophagy functions as a primary cell death mechanism remain to be defined; such knowledge will be critical for the rational design and targeted application of therapeutic regimens exploiting autophagy induction for more effective tumor cell killing. Table 3 lists studies where autophagy inducers potentiated the anticancer effect of other treatment modalities, such as chemotherapy and/or radiation.

Table 3.

Preclinical studies supporting autophagy induction for cancer treatment

Cancer type Primary treatment (target) Autophagy induction method (target) References
*Breast cancer Doxorubicin (DNA) RNAi (BCL-2) 184
Nelfinavir (HIV protease) Tamoxifen (estrogen receptor) 185
RT (DNA) Z-VAD (caspases) 186
AML MGCD0103, vorinostat (histone deacetylation) GX15-070 (BCL-2) 187
*Colorectal cancer RT (DNA) BCG (immune system) 188
Glioma, malignant RT (DNA) Arsenic trioxide (thioredoxin reductase) 189
Delta-24-RGD (DNA) RAD001 (mTOR) 190
* OBP-405 RAD001 (mTOR) 191
Lung cancer, non-small cell RT (DNA) ABT-737 (BCL-2) rapamycin (mTOR) 192
RT (DNA) Z-VAD (caspases) 186
RT (DNA) Berberine (angiogenesis, COX-2, TNF) 193
Lymphoma, mantle cell Vorinostat (histone deacetylation) Temsirolimus (mTOR) 194
*Renal cell cancer, VHL-deficient STF-62247 (ER-Golgi trafficking) STF-62247 (ER-Golgi trafficking) 145
*Pancreatic cancer Sorafenib (multiple kinases) + HDACi (histone deacetylation) GX15-070 (BCL-2) 195
Sarcoma, fibro- RT (DNA) Arsenic trioxide (thioredoxin reductase) 196
*Thyroid cancer, papillary Doxorubicin, RT (DNA) RAD001 (mTOR) 197
*

Studies where genetic deletion or knockdown of autophagy regulator(s) prolonged cancer cell survival.

Autophagy Modulation for Cancer Prevention

In the long run, the most effective (and also the least expensive) way to treat cancer is to prevent it from arising in the first place. Cancer prevention strategies must be accessible, easily implemented, well tolerated and safe over time. Accumulation of protein aggregates due to defective autophagy is a well-recognized culprit in neurodegeneration, including Alzheimer's, Parkinson's and polyglutamine diseases, and in hepatic dysfunction, but its contribution to cancer development and progression was not so clear.146148 Recent studies indicated that protein aggregation in autophagy-defective tumor cells is a likely source of genotoxic stress, mostly due to the resultant ER and oxidative stress, in turn contributing to the genomic damage and instability, and thus increased tumorigenicity, associated with autophagy defects.100102,149 Since basal autophagy plays a critical role in cellular homeostasis by degrading aged or malfunctioning organelles and damaged or misfolded proteins, thus maintaining genome integrity, and as such exhibiting a tumor suppressive effect, it is reasonable to target autophagy for cancer prevention. Autophagy stimulators are currently being explored as preventative agents in neurodegeneration;150 their predicted role in cancer prevention is also worthy of investigation and has potentially significant clinical implications, as supported by accumulating scientific evidence.

For example, cancer risk highly correlates with age.151 Caloric restriction (CR) involving limited food intake and physical exercise induces autophagy and delays the aging process, thus extending lifespan and possibly inhibiting tumorigenesis. The longevity resulting from CR was further validated in Tp53−/− mice, rhesus monkeys, yeast, Caenorthabditis elegans and Drosophila.151157 Thus, CR may be one way to modulate autophagy for cancer prevention. However, this hypothesis is far from clinical application, as it still needs to be extensively investigated and ultimately validated in humans. Diet-associated autophagy upregulation may be another approach to cancer prevention. Autophagy induction by carotenoids, lycopene, lutein, polyphenols, resveratrol, curcumin and epigallocatechin-3-gallate has been implicated in the ability of these dietary compounds to inhibit ROS and reactive nitrogen species accumulation.158,159 Pharmacological activation of autophagy by drugs, such as rapamycin analogs, class I PI3K inhibitors, chloroquine and metformin, has also been reported to inhibit malignant transformation, and thus likely prevent cancer, by limiting genomic damage.160162 The possibility of preventing cancer by autophagy modulation is intriguing, but clearly in need of further investigation, as the most effective and safest way to manipulate autophagy for cancer prevention remains to be defined.

Concluding Remarks

The role of autophagy in cancer and treatment responsiveness is undoubtedly complicated. The double-edged sword function of autophagy, as both a tumor suppressor and a protector of cancer cell survival, likely impacts anticancer treatment efficacy in opposing ways. Exploitation of the functional autophagy status in tumors and pharmacologic autophagy modulation for cancer treatment and prevention presents novel opportunities in cancer management. Autophagy defects are associated with susceptibility to metabolic stress, DNA damage accumulation, genomic instability and accelerated tumorigenicity. It is, thus, reasonable to hypothesize that autophagy upregulation may preserve cellular fitness and genome integrity to prevent cancer development and progression; chronically autophagy-deficient tumors may also be particularly sensitive to genotoxic and/or metabolic stressinducing anticancer agents, such as DNA damaging and antiangiogenic drugs. On the other hand, autophagy-competent tumors (even if their autophagy potential is partially compromised due to cancer-associated cellular events, such as constitutive PI3K/AKT/mTOR axis activation) likely utilize, and potentially rely on, this pathway for survival under metabolic stress conditions, such as during rapid tumor growth, metastasis and treatment. In the latter case, concurrent (i.e., acute) autophagy inhibition is expected to increase the efficacy of any anticancer modality, including radiation, chemotherapy, biological agents and combinatorial regimens.

In conclusion, the multifaceted nature of autophagy and its diverse crosstalk with other biological processes, including cell death pathways, must be carefully considered when the autophagic system is targeted for anticancer benefit. Areas of great interest in cancer research and with potentially significant therapeutic implications include autophagy regulation in tumor cells, impact of autophagy functional status in tumors on cancer progression and response to treatment, and elucidation of how to best modulate autophagy for therapeutic benefit and cancer prevention, so as to achieve the ultimate goal of cancer eradication.

Abbreviations

3D

three-dimensional

3-MA

3-methyladenine

4-HPR

N-(4-hydroxyphenyl) retinamide

5-FU

5-fluorouracil

ACD

autophagic cell death

ADI-PEG20

arginine deiminase, pegylated

AML

acute myelogenous leukemia

AMP

adenosine monophosphate

ARHI

aplasia ras homolog member I

ATGs

autophagy related genes

ATP

adenosine triphosphate

BafA

bafilomycin A

BCG

Bacillus Calmette-Guerin

BH3

BCL-2 homology domain 3

BIF-1

Bax-Interacting factor-1

BRCA1

breast cancer susceptibility gene 1

CAMKKβ

calmodulin-dependent protein kinase kinase β

CINJ

Cancer Institute of New Jersey

CLL

chronic lymphocytic leukemia

CML

chronic myelogenous leukemia

CQ

chloroquine

CR

caloric restriction

DAPK

death-associated protein kinase

DCIS

ductal carcinoma in situ

DNA

deoxy-ribonucleic acid

DRAM

damage-regulated autophagy modulator

EGFR

epidermal frowth factor receptor

ER

endoplasmic reticulum

FOLFOX

5-FU/leucovorin and oxaliplatin

HCQ

hydroxychloroquine

HDACi

histone deacetylase inhibitor

HIV

human immunodeficiency virus

iBMK

immortalized baby mouse kidney

IFN

interferon

IKK

IκB kinase

IL

interleukin

iMMECs

immortalized mouse mammary epithelial cells

IRE1

inositol-requiring kinase 1

JNK

c-jun N-terminal kinase

LC3

light chain 3 (also, ATG8)

Maastricht Rad Onc

Maastricht Radiation Oncology

Mass Gen Hosp

Massachusetts General Hospital

mTOR

mammalian target of rapamycin

Nat U Hosp, Singapore

National University Hospital, Singapore

NCI

National Cancer Institute, USA

NK

natural killer

NINN, Mexico

National Institute of Neurology and Neurosurgery, Mexico

NSCLC

non-small cell lung cancer

NSLIJHS

North Shore Long Island Jewish Health System

PERK

protein kinase-like endoplasmic reticulum kinase

PI3K

phosphatidyl inositol-3-kinase

PRAS40

proline-rich AKT substrate of 40 kDa

PTEN

phosphatase and tensin-homologue deleted on chromosome 10

RNAi

RNA interference

ROS

reactive oxygen species

RT

radiation therapy

SAHA

suberoylanilide hydroxamic acid (also, vorinostat)

SCLC

small cell lung cancer

TAK1

transforming growth factor-β-activating kinase 1

TFT

trifluorothymidine

TNF

tumor necrosis factor

TPA

12-O-tetradecanoyl-phorbol-13-acetate

TRAIL

TNF-related apoptosis inducing ligand

TSC

tuberous sclerosis complex

U

university

U Penn

University of Pennsylvania

U Texas HSC S Ant

University of Texas Health Science Center at San Antonio

UVRAG

ultraviolet radiation resistance-associated gene

VHL

Von Hippel-Lindau

XELOX

capecitabine (xeloda) and oxaliplatin

References

  • 1.Codogno P, Meijer AJ. Autophagy and signaling: their role in cell survival and cell death. Cell Death Differ. 2005;12:1509–1518. doi: 10.1038/sj.cdd.4401751. [DOI] [PubMed] [Google Scholar]
  • 2.Klionsky DJ. Autophagy revisited: a conversation with Christian de Duve. Autophagy. 2008;4:740–743. doi: 10.4161/auto.6398. [DOI] [PubMed] [Google Scholar]
  • 3.Shintani T, Klionsky DJ. Autophagy in health and disease: a double-edged sword. Science. 2004;306:990–995. doi: 10.1126/science.1099993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Eskelinen EL, Saftig P. Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta. 2009;1793:664–673. doi: 10.1016/j.bbamcr.2008.07.014. [DOI] [PubMed] [Google Scholar]
  • 5.Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature. 2008;451:1069–1075. doi: 10.1038/nature06639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Malicdan MC, Noguchi S, Nonaka I, Saftig P, Nishino I. Lysosomal myopathies: an excessive build-up in autophagosomes is too much to handle. Neuromuscul Disord. 2008;18:521–529. doi: 10.1016/j.nmd.2008.04.010. [DOI] [PubMed] [Google Scholar]
  • 7.Winslow AR, Rubinsztein DC. Autophagy in neurodegeneration and development. Biochim Biophys Acta. 2008;1782:723–729. doi: 10.1016/j.bbadis.2008.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Orvedahl A, Levine B. Eating the enemy within: autophagy in infectious diseases. Cell Death Differ. 2009;16:57–69. doi: 10.1038/cdd.2008.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cadwell K, Liu JY, Brown SL, Miyoshi H, Loh J, Lennerz JK, et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature. 2008;456:259–263. doi: 10.1038/nature07416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Saitoh T, Fujita N, Jang MH, Uematsu S, Yang BG, Satoh T, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008;456:264–268. doi: 10.1038/nature07383. [DOI] [PubMed] [Google Scholar]
  • 11.Yen WL, Klionsky DJ. How to live long and prosper: autophagy, mitochondria and aging. Physiology (Bethesda) 2008;23:248–262. doi: 10.1152/physiol.00013.2008. [DOI] [PubMed] [Google Scholar]
  • 12.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]
  • 13.Carew JS, Nawrocki ST, Cleveland JL. Modulating autophagy for therapeutic benefit. Autophagy. 2007;3:464–467. doi: 10.4161/auto.4311. [DOI] [PubMed] [Google Scholar]
  • 14.Chen N, Karantza-Wadsworth V. Role and regulation of autophagy in cancer. Biochim Biophys Acta. 2009;1793:1516–1523. doi: 10.1016/j.bbamcr.2008.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Al-Ejeh F, Kumar R, Wiegmans A, Lakhani SR, Brown MP, Khanna KK. Harnessing the complexity of DNA-damage response pathways to improve cancer treatment outcomes. Oncogene. 2010;29:6085–6098. doi: 10.1038/onc.2010.407. [DOI] [PubMed] [Google Scholar]
  • 16.Gewirtz DA. Autophagy as a mechanism of radiation sensitization in breast tumor cells. Autophagy. 2007;3:249–250. doi: 10.4161/auto.3723. [DOI] [PubMed] [Google Scholar]
  • 17.John S, Nayvelt I, Hsu HC, Yang P, Liu W, Das GM, et al. Regulation of estrogenic effects by beclin 1 in breast cancer cells. Cancer Res. 2008;68:7855–7863. doi: 10.1158/0008-5472.CAN-07-5875. [DOI] [PubMed] [Google Scholar]
  • 18.Buytaert E, Callewaert G, Vandenheede JR, Agostinis P. Deficiency in apoptotic effectors Bax and Bak reveals an autophagic cell death pathway initiated by photodamage to the endoplasmic reticulum. Autophagy. 2006;2:238–240. doi: 10.4161/auto.2730. [DOI] [PubMed] [Google Scholar]
  • 19.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]
  • 20.Kroemer G, Marino G, Levine B. Autophagy and the integrated stress response. Mol Cell. 2010;40:280–293. doi: 10.1016/j.molcel.2010.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Easton JB, Houghton PJ. mTOR and cancer therapy. Oncogene. 2006;25:6436–6446. doi: 10.1038/sj.onc.1209886. [DOI] [PubMed] [Google Scholar]
  • 22.Guertin DA, Sabatini DM. An expanding role for mTOR in cancer. Trends Mol Med. 2005;11:353–361. doi: 10.1016/j.molmed.2005.06.007. [DOI] [PubMed] [Google Scholar]
  • 23.Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature. 2001;411:355–365. doi: 10.1038/35077225. [DOI] [PubMed] [Google Scholar]
  • 24.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]
  • 25.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]
  • 26.Hafner C, Lopez-Knowles E, Luis NM, Toll A, Baselga E, Fernandez-Casado A, et al. Oncogenic PIK3CA mutations occur in epidermal nevi and seborrheic keratoses with a characteristic mutation pattern. Proc Natl Acad Sci USA. 2007;104:13450–13454. doi: 10.1073/pnas.0705218104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Horn S, Bergholz U, Jucker M, McCubrey JA, Trumper L, Stocking C, et al. Mutations in the catalytic subunit of class IA PI3K confer leukemogenic potential to hematopoietic cells. Oncogene. 2008;27:4096–4106. doi: 10.1038/onc.2008.40. [DOI] [PubMed] [Google Scholar]
  • 28.Isakoff SJ, Engelman JA, Irie HY, Luo J, Brachmann SM, Pearline RV, et al. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Res. 2005;65:10992–11000. doi: 10.1158/0008-5472.CAN-05-2612. [DOI] [PubMed] [Google Scholar]
  • 29.Zhao JJ, Liu Z, Wang L, Shin E, Loda MF, Roberts TM. The oncogenic properties of mutant p110alpha and p110beta phosphatidylinositol-3-kinases in human mammary epithelial cells. Proc Natl Acad Sci USA. 2005;102:18443–18448. doi: 10.1073/pnas.0508988102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Maiuri MC, Criollo A, Kroemer G. Crosstalk between apoptosis and autophagy within the Beclin 1 interactome. EMBO J. 2010;29:515–516. doi: 10.1038/emboj.2009.377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Maehama T. PTEN: its deregulation and tumorigenesis. Biol Pharm Bull. 2007;30:1624–1627. doi: 10.1248/bpb.30.1624. [DOI] [PubMed] [Google Scholar]
  • 32.Simpson L, Parsons R. PTEN: life as a tumor suppressor. Exp Cell Res. 2001;264:29–41. doi: 10.1006/excr.2000.5130. [DOI] [PubMed] [Google Scholar]
  • 33.Yin Y, Shen WH. PTEN: a new guardian of the genome. Oncogene. 2008;27:5443–5453. doi: 10.1038/onc.2008.241. [DOI] [PubMed] [Google Scholar]
  • 34.Shinojima N, Yokoyama T, Kondo Y, Kondo S. Roles of the Akt/mTOR/p70S6K and ERK1/2 signaling pathways in curcumin-induced autophagy. Autophagy. 2007;3:635–637. doi: 10.4161/auto.4916. [DOI] [PubMed] [Google Scholar]
  • 35.Wu YT, Tan HL, Huang Q, Ong CN, Shen HM. Activation of the PI3K-Akt-mTOR signaling pathway promotes necrotic cell death via suppression of autophagy. Autophagy. 2009;5:824–834. doi: 10.4161/auto.9099. [DOI] [PubMed] [Google Scholar]
  • 36.Vander Haar E, Lee SI, Bandhakavi S, Griffin TJ, Kim DH. Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat Cell Biol. 2007;9:316–323. doi: 10.1038/ncb1547. [DOI] [PubMed] [Google Scholar]
  • 37.Mamane Y, Petroulakis E, LeBacquer O, Sonenberg N. mTOR, translation initiation and cancer. Oncogene. 2006;25:6416–6422. doi: 10.1038/sj.onc.1209888. [DOI] [PubMed] [Google Scholar]
  • 38.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]
  • 39.Tanemura M, Saga A, Kawamoto K, Machida T, Deguchi T, Nishida T, et al. Rapamycin induces autophagy in islets: relevance in islet transplantation. Transplant Proc. 2009;41:334–338. doi: 10.1016/j.transproceed.2008.10.032. [DOI] [PubMed] [Google Scholar]
  • 40.Jansen M, Ten Klooster JP, Offerhaus GJ, Clevers H. LKB1 and AMPK family signaling: the intimate link between cell polarity and energy metabolism. Physiol Rev. 2009;89:777–798. doi: 10.1152/physrev.00026.2008. [DOI] [PubMed] [Google Scholar]
  • 41.Shackelford DB, Shaw RJ. The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer. 2009;9:563–575. doi: 10.1038/nrc2676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci USA. 2004;101:3329–3335. doi: 10.1073/pnas.0308061100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Woods A, Johnstone SR, Dickerson K, Leiper FC, Fryer LG, Neumann D, et al. LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr Biol. 2003;13:2004–2008. doi: 10.1016/j.cub.2003.10.031. [DOI] [PubMed] [Google Scholar]
  • 44.Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, et al. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell. 2008;30:214–226. doi: 10.1016/j.molcel.2008.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival. Cell. 2003;115:577–590. doi: 10.1016/s0092-8674(03)00929-2. [DOI] [PubMed] [Google Scholar]
  • 46.Anderson KA, Ribar TJ, Lin F, Noeldner PK, Green MF, Muehlbauer MJ, et al. Hypothalamic CaMKK2 contributes to the regulation of energy balance. Cell Metab. 2008;7:377–388. doi: 10.1016/j.cmet.2008.02.011. [DOI] [PubMed] [Google Scholar]
  • 47.Stahmann N, Woods A, Carling D, Heller R. Thrombin activates AMP-activated protein kinase in endothelial cells via a pathway involving Ca2+/calmodulin-dependent protein kinase kinase beta. Mol Cell Biol. 2006;26:5933–5945. doi: 10.1128/MCB.00383-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Tamas P, Hawley SA, Clarke RG, Mustard KJ, Green K, Hardie DG, et al. Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes. J Exp Med. 2006;203:1665–1670. doi: 10.1084/jem.20052469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Liang J, Shao SH, Xu ZX, Hennessy B, Ding Z, Larrea M, et al. The energy sensing LKB1-AMPK pathway regulates p27(kip1) phosphorylation mediating the decision to enter autophagy or apoptosis. Nat Cell Biol. 2007;9:218–224. doi: 10.1038/ncb1537. [DOI] [PubMed] [Google Scholar]
  • 50.Herrero-Martin G, Hoyer-Hansen M, Garcia-Garcia C, Fumarola C, Farkas T, Lopez-Rivas A, et al. TAK1 activates AMPK-dependent cytoprotective autophagy in TRAIL-treated epithelial cells. EMBO J. 2009;28:677–685. doi: 10.1038/emboj.2009.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Carson DA, Lois A. Cancer progression and p53. Lancet. 1995;346:1009–1011. doi: 10.1016/s0140-6736(95)91693-8. [DOI] [PubMed] [Google Scholar]
  • 52.Guimaraes DP, Hainaut P. TP53: a key gene in human cancer. Biochimie. 2002;84:83–93. doi: 10.1016/s0300-9084(01)01356-6. [DOI] [PubMed] [Google Scholar]
  • 53.Hainaut P. The tumor suppressor protein p53: a receptor to genotoxic stress that controls cell growth and survival. Curr Opin Oncol. 1995;7:76–82. [PubMed] [Google Scholar]
  • 54.Chen N, Debnath J. Autophagy and tumorigenesis. FEBS Lett. 2010;584:1427–1435. doi: 10.1016/j.febslet.2009.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Crighton D, Wilkinson S, O'Prey J, Syed N, Smith P, Harrison PR, et al. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell. 2006;126:121–134. doi: 10.1016/j.cell.2006.05.034. [DOI] [PubMed] [Google Scholar]
  • 56.Maiuri MC, Malik SA, Morselli E, Kepp O, Criollo A, Mouchel PL, et al. 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]
  • 57.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]
  • 58.Tasdemir E, Chiara Maiuri M, Morselli E, Criollo A, D'Amelio M, Djavaheri-Mergny M, et al. A dual role of p53 in the control of autophagy. Autophagy. 2008;4:810–814. doi: 10.4161/auto.6486. [DOI] [PubMed] [Google Scholar]
  • 59.Eisenberg-Lerner A, Kimchi A. The paradox of autophagy and its implication in cancer etiology and therapy. Apoptosis. 2009;14:376–391. doi: 10.1007/s10495-008-0307-5. [DOI] [PubMed] [Google Scholar]
  • 60.Liang XH, Kleeman LK, Jiang HH, Gordon G, Goldman JE, Berry G, et al. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J Virol. 1998;72:8586–8596. doi: 10.1128/jvi.72.11.8586-8596.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kametaka S, Okano T, Ohsumi M, Ohsumi Y. Apg14p and Apg6/Vps30p form a protein complex essential for autophagy in the yeast, Saccharomyces cerevisiae. J Biol Chem. 1998;273:22284–22291. doi: 10.1074/jbc.273.35.22284. [DOI] [PubMed] [Google Scholar]
  • 62.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]
  • 63.Melendez A, Talloczy Z, Seaman M, Eskelinen EL, Hall DH, Levine B. Autophagy genes are essential for dauer development and life-span extension in C. elegans. Science. 2003;301:1387–1391. doi: 10.1126/science.1087782. [DOI] [PubMed] [Google Scholar]
  • 64.Otto GP, Wu MY, Kazgan N, Anderson OR, Kessin RH. Dictyostelium macroautophagy mutants vary in the severity of their developmental defects. J Biol Chem. 2004;279:15621–15629. doi: 10.1074/jbc.M311139200. [DOI] [PubMed] [Google Scholar]
  • 65.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]
  • 66.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]
  • 67.Cao Y, Klionsky DJ. Physiological functions of Atg6/Beclin 1: a unique autophagy-related protein. Cell Res. 2007;17:839–849. doi: 10.1038/cr.2007.78. [DOI] [PubMed] [Google Scholar]
  • 68.Itakura E, Mizushima N. Atg14 and UVRAG: mutually exclusive subunits of mammalian Beclin 1-PI3K complexes. Autophagy. 2009;5:534–536. doi: 10.4161/auto.5.4.8062. [DOI] [PubMed] [Google Scholar]
  • 69.Matsunaga K, Saitoh T, Tabata K, Omori H, Satoh T, Kurotori N, et al. Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat Cell Biol. 2009;11:385–396. doi: 10.1038/ncb1846. [DOI] [PubMed] [Google Scholar]
  • 70.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]
  • 71.Takahashi Y, Coppola D, Matsushita N, Cualing HD, Sun M, Sato Y, et al. Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis. Nat Cell Biol. 2007;9:1142–1151. doi: 10.1038/ncb1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.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]
  • 73.Eccles DM, Cranston G, Steel CM, Nakamura Y, Leonard RC. Allele losses on chromosome 17 in human epithelial ovarian carcinoma. Oncogene. 1990;5:1599–1601. [PubMed] [Google Scholar]
  • 74.Futreal PA, Soderkvist P, Marks JR, Iglehart JD, Cochran C, Barrett JC, et al. Detection of frequent allelic loss on proximal chromosome 17q in sporadic breast carcinoma using microsatellite length polymorphisms. Cancer Res. 1992;52:2624–2627. [PubMed] [Google Scholar]
  • 75.Gao X, Zacharek A, Salkowski A, Grignon DJ, Sakr W, Porter AT, et al. Loss of heterozygosity of the BRCA1 and other loci on chromosome 17q in human prostate cancer. Cancer Res. 1995;55:1002–1005. [PubMed] [Google Scholar]
  • 76.Russell SE, Hickey GI, Lowry WS, White P, Atkinson RJ. Allele loss from chromosome 17 in ovarian cancer. Oncogene. 1990;5:1581–1583. [PubMed] [Google Scholar]
  • 77.Saito H, Inazawa J, Saito S, Kasumi F, Koi S, Sagae S, et al. Detailed deletion mapping of chromosome 17q in ovarian and breast cancers: 2-cM region on 17q21.3 often and commonly deleted in tumors. Cancer Res. 1993;53:3382–3385. [PubMed] [Google Scholar]
  • 78.Wei Y, Pattingre S, Sinha S, Bassik M, Levine B. JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol Cell. 2008;30:678–688. doi: 10.1016/j.molcel.2008.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Erlich S, Mizrachy L, Segev O, Lindenboim L, Zmira O, Adi-Harel S, et al. Differential interactions between Beclin 1 and Bcl-2 family members. Autophagy. 2007;3:561–568. doi: 10.4161/auto.4713. [DOI] [PubMed] [Google Scholar]
  • 80.Lee AS, Hendershot LM. ER stress and cancer. Cancer Biol Ther. 2006;5:721–722. doi: 10.4161/cbt.5.7.3120. [DOI] [PubMed] [Google Scholar]
  • 81.Ma Y, Hendershot LM. The role of the unfolded protein response in tumour development: friend or foe? Nat Rev Cancer. 2004;4:966–977. doi: 10.1038/nrc1505. [DOI] [PubMed] [Google Scholar]
  • 82.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]
  • 83.Yorimitsu T, Nair U, Yang Z, Klionsky DJ. Endoplasmic reticulum stress triggers autophagy. J Biol Chem. 2006;281:30299–30304. doi: 10.1074/jbc.M607007200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hoyer-Hansen M, Bastholm L, Szyniarowski P, Campanella M, Szabadkai G, Farkas T, et al. Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta and Bcl-2. Mol Cell. 2007;25:193–205. doi: 10.1016/j.molcel.2006.12.009. [DOI] [PubMed] [Google Scholar]
  • 85.Platini F, Perez-Tomas R, Ambrosio S, Tessitore L. Understanding autophagy in cell death control. Curr Pharm Des. 2010;16:101–113. doi: 10.2174/138161210789941810. [DOI] [PubMed] [Google Scholar]
  • 86.Spassieva SD, Mullen TD, Townsend DM, Obeid LM. Disruption of ceramide synthesis by CerS2 downregulation leads to autophagy and the unfolded protein response. Biochem J. 2009;424:273–283. doi: 10.1042/BJ20090699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Younce CW, Kolattukudy PE. MCP-1 causes cardiomyoblast death via autophagy resulting from ER stress caused by oxidative stress generated by inducing a novel zinc-finger protein, MCPIP. Biochem J. 2010;426:43–53. doi: 10.1042/BJ20090976. [DOI] [PubMed] [Google Scholar]
  • 88.Bialik S, Kimchi A. DAP-kinase as a target for drug design in cancer and diseases associated with accelerated cell death. Semin Cancer Biol. 2004;14:283–294. doi: 10.1016/j.semcancer.2004.04.008. [DOI] [PubMed] [Google Scholar]
  • 89.Bialik S, Kimchi A. The death-associated protein kinases: structure, function and beyond. Annu Rev Biochem. 2006;75:189–210. doi: 10.1146/annurev.biochem.75.103004.142615. [DOI] [PubMed] [Google Scholar]
  • 90.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]
  • 91.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]
  • 92.Clarke PG. Developmental cell death: morphological diversity and multiple mechanisms. Anat Embryol. 1990;181:195–213. doi: 10.1007/BF00174615. [DOI] [PubMed] [Google Scholar]
  • 93.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]
  • 94.Marino G, Salvador-Montoliu N, Fueyo A, Knecht E, Mizushima N, Lopez-Otin C. Tissue-specific autophagy alterations and increased tumorigenesis in mice deficient in Atg4C/autophagin-3. J Biol Chem. 2007;282:18573–18583. doi: 10.1074/jbc.M701194200. [DOI] [PubMed] [Google Scholar]
  • 95.Iqbal J, Kucuk C, Deleeuw RJ, Srivastava G, Tam W, Geng H, et al. Genomic analyses reveal global functional alterations that promote tumor growth and novel tumor suppressor genes in natural killer-cell malignancies. Leukemia. 2009;23:1139–1151. doi: 10.1038/leu.2009.3. [DOI] [PubMed] [Google Scholar]
  • 96.Coppola D, Khalil F, Eschrich SA, Boulware D, Yeatman T, Wang HG. Downregulation of Bax-interacting factor-1 in colorectal adenocarcinoma. Cancer. 2008;113:2665–2670. doi: 10.1002/cncr.23892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kim MS, Jeong EG, Ahn CH, Kim SS, Lee SH, Yoo NJ. Frameshift mutation of UVRAG, an autophagy-related gene, in gastric carcinomas with microsatellite instability. Hum Pathol. 2008;39:1059–1063. doi: 10.1016/j.humpath.2007.11.013. [DOI] [PubMed] [Google Scholar]
  • 98.Liang C, Feng P, Ku B, Dotan I, Canaani D, Oh BH, et al. Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG. Nat Cell Biol. 2006;8:688–699. doi: 10.1038/ncb1426. [DOI] [PubMed] [Google Scholar]
  • 99.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]
  • 100.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]
  • 101.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]
  • 102.Mathew R, Kongara S, Beaudoin B, Karp CM, Bray K, Degenhardt K, et al. Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev. 2007;21:1367–1381. doi: 10.1101/gad.1545107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bao XH, Naomoto Y, Hao HF, Watanabe N, Sakurama K, Noma K, et al. Autophagy: can it become a potential therapeutic target? Int J Mol Med. 2010;25:493–503. doi: 10.3892/ijmm_00000369. [DOI] [PubMed] [Google Scholar]
  • 104.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]
  • 105.Lum JJ, DeBerardinis RJ, Thompson CB. Autophagy in metazoans: cell survival in the land of plenty. Nat Rev Mol Cell Biol. 2005;6:439–448. doi: 10.1038/nrm1660. [DOI] [PubMed] [Google Scholar]
  • 106.Karantza-Wadsworth V, White E. A mouse mammary epithelial cell model to identify molecular mechanisms regulating breast cancer progression. Methods Enzymol. 2008;446:61–76. doi: 10.1016/S0076-6879(08)01604-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jin S, DiPaola RS, Mathew R, White E. Metabolic catastrophe as a means to cancer cell death. J Cell Sci. 2007;120:379–383. doi: 10.1242/jcs.03349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Fung C, Lock R, Gao S, Salas E, Debnath J. Induction of autophagy during extracellular matrix detachment promotes cell survival. Mol Biol Cell. 2008;19:797–806. doi: 10.1091/mbc.E07-10-1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.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]
  • 110.Kondo Y, Kanzawa T, Sawaya R, Kondo S. The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 2005;5:726–734. doi: 10.1038/nrc1692. [DOI] [PubMed] [Google Scholar]
  • 111.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]
  • 112.Vousden KH, Ryan KM. p53 and metabolism. Nat Rev Cancer. 2009;9:691–700. doi: 10.1038/nrc2715. [DOI] [PubMed] [Google Scholar]
  • 113.Jones RG, Thompson CB. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev. 2009;23:537–548. doi: 10.1101/gad.1756509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Rabinowitz JD, White E. Autophagy and metabolism. Science. 2010;330:1344–1348. doi: 10.1126/science.1193497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Hoyer-Hansen M, Jaattela M. Autophagy: an emerging target for cancer therapy. Autophagy. 2008;4:574–580. doi: 10.4161/auto.5921. [DOI] [PubMed] [Google Scholar]
  • 116.Reed JC. Drug insight: cancer therapy strategies based on restoration of endogenous cell death mechanisms. Nat Clin Pract Oncol. 2006;3:388–398. doi: 10.1038/ncponc0538. [DOI] [PubMed] [Google Scholar]
  • 117.Tan TT, White E. Therapeutic targeting of death pathways in cancer: mechanisms for activating cell death in cancer cells. Adv Exp Med Biol. 2008;615:81–104. doi: 10.1007/978-1-4020-6554-5_5. [DOI] [PubMed] [Google Scholar]
  • 118.Chen S, Rehman SK, Zhang W, Wen A, Yao L, Zhang J. Autophagy is a therapeutic target in anticancer drug resistance. Biochim Biophys Acta. 2010;1806:220–229. doi: 10.1016/j.bbcan.2010.07.003. [DOI] [PubMed] [Google Scholar]
  • 119.Hippert MM, O'Toole PS, Thorburn A. Autophagy in cancer: good, bad or both? Cancer Res. 2006;66:9349–9351. doi: 10.1158/0008-5472.CAN-06-1597. [DOI] [PubMed] [Google Scholar]
  • 120.Gewirtz DA. Autophagy, senescence and tumor dormancy in cancer therapy. Autophagy. 2009;5:1232–1234. doi: 10.4161/auto.5.8.9896. [DOI] [PubMed] [Google Scholar]
  • 121.Dikic I, Johansen T, Kirkin V. Selective autophagy in cancer development and therapy. Cancer Res. 2010;70:3431–3434. doi: 10.1158/0008-5472.CAN-09-4027. [DOI] [PubMed] [Google Scholar]
  • 122.Kondo Y, Kondo S. Autophagy and cancer therapy. Autophagy. 2006;2:85–90. doi: 10.4161/auto.2.2.2463. [DOI] [PubMed] [Google Scholar]
  • 123.Ito H, Daido S, Kanzawa T, Kondo S, Kondo Y. Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol. 2005;26:1401–1410. [PubMed] [Google Scholar]
  • 124.Shingu T, Fujiwara K, Bogler O, Akiyama Y, Moritake K, Shinojima N, et al. Stage-specific effect of inhibition of autophagy on chemotherapy-induced cytotoxicity. Autophagy. 2009;5:537–539. doi: 10.4161/auto.5.4.8164. [DOI] [PubMed] [Google Scholar]
  • 125.Shanmugam M, McBrayer SK, Qian J, Raikoff K, Avram MJ, Singhal S, et al. Targeting glucose consumption and autophagy in myeloma with the novel nucleoside analogue 8-aminoadenosine. J Biol Chem. 2009;284:26816–26830. doi: 10.1074/jbc.M109.000646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Abedin MJ, Wang D, McDonnell MA, Lehmann U, Kelekar A. Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ. 2007;14:500–510. doi: 10.1038/sj.cdd.4402039. [DOI] [PubMed] [Google Scholar]
  • 127.Vazquez-Martin A, Oliveras-Ferraros C, Menendez JA. Autophagy facilitates the development of breast cancer resistance to the anti-HER2 monoclonal antibody trastuzumab. PLoS One. 2009;4:6251. doi: 10.1371/journal.pone.0006251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008;68:1485–1494. doi: 10.1158/0008-5472.CAN-07-0562. [DOI] [PubMed] [Google Scholar]
  • 129.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]
  • 130.Kim RH, Coates JM, Bowles TL, McNerney GP, Sutcliffe J, Jung JU, et al. Arginine deiminase as a novel therapy for prostate cancer induces autophagy and caspase-independent apoptosis. Cancer Res. 2009;69:700–708. doi: 10.1158/0008-5472.CAN-08-3157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.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]
  • 132.Li J, Hou N, Faried A, Tsutsumi S, Takeuchi T, Kuwano H. Inhibition of autophagy by 3-MA enhances the effect of 5-FU-induced apoptosis in colon cancer cells. Ann Surg Oncol. 2009;16:761–771. doi: 10.1245/s10434-008-0260-0. [DOI] [PubMed] [Google Scholar]
  • 133.Liu D, Yang Y, Liu Q, Wang J. Inhibition of autophagy by 3-MA potentiates cisplatin-induced apoptosis in esophageal squamous cell carcinoma cells. Med Oncol. 2009 doi: 10.1007/s12032-009-9397-3. In press. [DOI] [PubMed] [Google Scholar]
  • 134.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]
  • 135.Buytaert E, Callewaert G, Hendrickx N, Scorrano L, Hartmann D, Missiaen L, et al. Role of endoplasmic reticulum depletion and multidomain proapoptotic BAX and BAK proteins in shaping cell death after hypericin-mediated photodynamic therapy. FASEB J. 2006;20:756–758. doi: 10.1096/fj.05-4305fje. [DOI] [PubMed] [Google Scholar]
  • 136.Hoyer-Hansen M, Bastholm L, Mathiasen IS, Elling F, Jaattela M. Vitamin D analog EB1089 triggers dramatic lysosomal changes and Beclin 1-mediated autophagic cell death. Cell Death Differ. 2005;12:1297–1309. doi: 10.1038/sj.cdd.4401651. [DOI] [PubMed] [Google Scholar]
  • 137.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]
  • 138.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]
  • 139.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]
  • 140.Komata T, Kanzawa T, Takeuchi H, Germano IM, Schreiber M, Kondo Y, et al. Antitumour effect of cyclin-dependent kinase inhibitors (p16(INK4A), p18(INK4C), p19(INK4D), p21(WAF1/CIP1) and p27(KIP1)) on malignant glioma cells. Br J Cancer. 2003;88:1277–1280. doi: 10.1038/sj.bjc.6600862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.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]
  • 142.Pyo JO, Jang MH, Kwon YK, Lee HJ, Jun JI, Woo HN, et al. Essential roles of Atg5 and FADD in autophagic cell death: dissection of autophagic cell death into vacuole formation and cell death. J Biol Chem. 2005;280:20722–20729. doi: 10.1074/jbc.M413934200. [DOI] [PubMed] [Google Scholar]
  • 143.Trincheri NF, Follo C, Nicotra G, Peracchio C, Castino R, Isidoro C. Resveratrol-induced apoptosis depends on the lipid kinase activity of Vps34 and on the formation of autophagolysosomes. Carcinogenesis. 2008;29:381–389. doi: 10.1093/carcin/bgm271. [DOI] [PubMed] [Google Scholar]
  • 144.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]
  • 145.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]
  • 146.Dehay B, Bove J, Rodriguez-Muela N, Perier C, Recasens A, Boya P, et al. Pathogenic lysosomal depletion in Parkinson's disease. J Neurosci. 2010;30:12535–12544. doi: 10.1523/JNEUROSCI.1920-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Lennerz JK, Hurov JB, White LS, Lewandowski KT, Prior JL, Planer GJ, et al. Loss of Par-1a/MARK3/CTAK1 kinase leads to reduced adiposity, resistance to hepatic steatosis and defective gluconeogenesis. Mol Cell Biol. 2010;30:5043–5056. doi: 10.1128/MCB.01472-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. J Cell Sci. 2007;120:4081–4091. doi: 10.1242/jcs.019265. [DOI] [PubMed] [Google Scholar]
  • 149.Degtyarev M, De Maziere A, Orr C, Lin J, Lee BB, Tien JY, et al. Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents. J Cell Biol. 2008;183:101–116. doi: 10.1083/jcb.200801099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Sarkar S, Rubinsztein DC. Small molecule enhancers of autophagy for neurodegenerative diseases. Mol Biosyst. 2008;4:895–901. doi: 10.1039/b804606a. [DOI] [PubMed] [Google Scholar]
  • 151.Cuervo AM, Bergamini E, Brunk UT, Droge W, Ffrench M, Terman A. Autophagy and aging: the importance of maintaining “clean” cells. Autophagy. 2005;1:131–140. doi: 10.4161/auto.1.3.2017. [DOI] [PubMed] [Google Scholar]
  • 152.Alvers AL, Fishwick LK, Wood MS, Hu D, Chung HS, Dunn WA, Jr, et al. Autophagy and amino acid homeostasis are required for chronological longevity in Saccharomyces cerevisiae. Aging Cell. 2009;8:353–369. doi: 10.1111/j.1474-9726.2009.00469.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Bartke A, Wright JC, Mattison JA, Ingram DK, Miller RA, Roth GS. Extending the lifespan of long-lived mice. Nature. 2001;414:412. doi: 10.1038/35106646. [DOI] [PubMed] [Google Scholar]
  • 154.Berrigan D, Perkins SN, Haines DC, Hursting SD. Adult-onset calorie restriction and fasting delay spontaneous tumorigenesis in p53-deficient mice. Carcinogenesis. 2002;23:817–822. doi: 10.1093/carcin/23.5.817. [DOI] [PubMed] [Google Scholar]
  • 155.Colman RJ, Anderson RM, Johnson SC, Kastman EK, Kosmatka KJ, Beasley TM. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science. 2009;325:201–204. doi: 10.1126/science.1173635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Hars ES, Qi H, Ryazanov AG, Jin S, Cai L, Hu C, et al. Autophagy regulates ageing in C. elegans. Autophagy. 2007;3:93–95. doi: 10.4161/auto.3636. [DOI] [PubMed] [Google Scholar]
  • 157.Juhasz G, Erdi B, Sass M, Neufeld TP. Atg7-dependent autophagy promotes neuronal health, stress tolerance and longevity but is dispensable for metamorphosis in Drosophila. Genes Dev. 2007;21:3061–3066. doi: 10.1101/gad.1600707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Davis CD. Nutritional interactions: credentialing of molecular targets for cancer prevention. Exp Biol Med. 2007;232:176–183. [PubMed] [Google Scholar]
  • 159.Pan MH, Ho CT. Chemopreventive effects of natural dietary compounds on cancer development. Chem Soc Rev. 2008;37:2558–2574. doi: 10.1039/b801558a. [DOI] [PubMed] [Google Scholar]
  • 160.Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005;330:1304–1305. doi: 10.1136/bmj.38415.708634.F7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132:27–42. doi: 10.1016/j.cell.2007.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Maclean KH, Dorsey FC, Cleveland JL, Kastan MB. Targeting lysosomal degradation induces p53-dependent cell death and prevents cancer in mouse models of lymphomagenesis. J Clin Invest. 2008;118:79–88. doi: 10.1172/JCI33700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Schoenlein PV, Periyasamy-Thandavan S, Samaddar JS, Jackson WH, Barrett JT. Autophagy facilitates the progression of ERalpha-positive breast cancer cells to antiestrogen resistance. Autophagy. 2009;5:400–403. doi: 10.4161/auto.5.3.7784. [DOI] [PubMed] [Google Scholar]
  • 164.Milani M, Rzymski T, Mellor HR, Pike L, Bottini A, Generali D, et al. The role of ATF4 stabilization and autophagy in resistance of breast cancer cells treated with Bortezomib. Cancer Res. 2009;69:4415–4423. doi: 10.1158/0008-5472.CAN-08-2839. [DOI] [PubMed] [Google Scholar]
  • 165.Clarke R, Shajahan AN, Riggins RB, Cho Y, Crawford A, Xuan J, et al. Gene network signaling in hormone responsiveness modifies apoptosis and autophagy in breast cancer cells. J Steroid Biochem Mol Biol. 2009;114:8–20. doi: 10.1016/j.jsbmb.2008.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Kanematsu S, Uehara N, Miki H, Yoshizawa K, Kawanaka A, Yuri T, et al. Autophagy inhibition enhances sulforaphane-induced apoptosis in human breast cancer cells. Anticancer Res. 2010;30:3381–3390. [PubMed] [Google Scholar]
  • 167.Fels DR, Ye J, Segan AT, Kridel SJ, Spiotto M, Olson M, et al. Preferential cytotoxicity of bortezomib toward hypoxic tumor cells via overactivation of endoplasmic reticulum stress pathways. Cancer Res. 2008;68:9323–9330. doi: 10.1158/0008-5472.CAN-08-2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kamitsuji Y, Kuroda J, Kimura S, Toyokuni S, Watanabe K, Ashihara E, et al. The Bcr-Abl kinase inhibitor INNO-406 induces autophagy and different modes of cell death execution in Bcr-Abl-positive leukemias. Cell Death Differ. 2008;15:1712–1722. doi: 10.1038/cdd.2008.107. [DOI] [PubMed] [Google Scholar]
  • 169.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]
  • 170.Mishima Y, Terui Y, Mishima Y, Taniyama A, Kuniyoshi R, Takizawa T, et al. Autophagy and autophagic cell death are next targets for elimination of the resistance to tyrosine kinase inhibitors. Cancer Sci. 2008;99:2200–2208. doi: 10.1111/j.1349-7006.2008.00932.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Bauvy C, Gane P, Arico S, Codogno P, Ogier-Denis E. Autophagy delays sulindac sulfide-induced apoptosis in the human intestinal colon cancer cell line HT-29. Exp Cell Res. 2001;268:139–149. doi: 10.1006/excr.2001.5285. [DOI] [PubMed] [Google Scholar]
  • 172.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]
  • 173.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]
  • 174.Bijnsdorp IV, Peters GJ, Temmink OH, Fukushima M, Kruyt FA. Differential activation of cell death and autophagy results in an increased cytotoxic potential for trifluorothymidine compared to 5-fluorouracil in colon cancer cells. Int J Cancer. 2010;126:2457–2468. doi: 10.1002/ijc.24943. [DOI] [PubMed] [Google Scholar]
  • 175.Gupta A, Roy S, Lazar AJ, Wang WL, McAuliffe JC, Reynoso D, et al. Autophagy inhibition and antimalarials promote cell death in gastrointestinal stromal tumor (GIST) Proc Natl Acad Sci USA. 2010;107:14333–14338. doi: 10.1073/pnas.1000248107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Shingu T, Fujiwara K, Bogler O, Akiyama Y, Moritake K, Shinojima N, et al. Inhibition of autophagy at a late stage enhances imatinib-induced cytotoxicity in human malignant glioma cells. Int J Cancer. 2009;124:1060–1071. doi: 10.1002/ijc.24030. [DOI] [PubMed] [Google Scholar]
  • 177.Lomonaco SL, Finniss S, Xiang C, Decarvalho A, Umansky F, Kalkanis SN, et al. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. Int J Cancer. 2009;125:717–722. doi: 10.1002/ijc.24402. [DOI] [PubMed] [Google Scholar]
  • 178.Tiwari M, Bajpai VK, Sahasrabuddhe AA, Kumar A, Sinha RA, Behari S, et al. Inhibition of N-(4-hydroxyphenyl)retinamide-induced autophagy at a lower dose enhances cell death in malignant glioma cells. Carcinogenesis. 2008;29:600–609. doi: 10.1093/carcin/bgm264. [DOI] [PubMed] [Google Scholar]
  • 179.Thorburn J, Moore F, Rao A, Barclay WW, Thomas LR, Grant KW, et al. Selective inactivation of a Fasassociated death domain protein (FADD)-dependent apoptosis and autophagy pathway in immortal epithelial cells. Mol Biol Cell. 2005;16:1189–1199. doi: 10.1091/mbc.E04-10-0906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Li M, Jiang X, Liu D, Na Y, Gao GF, Xi Z. Autophagy protects LNCaP cells under androgen deprivation conditions. Autophagy. 2008;4:54–60. doi: 10.4161/auto.5209. [DOI] [PubMed] [Google Scholar]
  • 181.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]
  • 182.Watanabe M, Adachi S, Matsubara H, Imai T, Yui Y, Mizushima Y, et al. Induction of autophagy in malignant rhabdoid tumor cells by the histone deacetylase inhibitor FK228 through AIF translocation. Int J Cancer. 2009;124:55–67. doi: 10.1002/ijc.23897. [DOI] [PubMed] [Google Scholar]
  • 183.Claerhout S, Verschooten L, Van Kelst S, De Vos R, Proby C, Agostinis P, et al. Concomitant inhibition of AKT and autophagy is required for efficient cisplatininduced apoptosis of metastatic skin carcinoma. Int J Cancer. 2010;127:2790–2803. doi: 10.1002/ijc.25300. [DOI] [PubMed] [Google Scholar]
  • 184.Akar U, Chaves-Reyez A, Barria M, Tari A, Sanguino A, Kondo Y, et al. Silencing of Bcl-2 expression by small interfering RNA induces autophagic cell death in MCF-7 breast cancer cells. Autophagy. 2008;4:669–679. doi: 10.4161/auto.6083. [DOI] [PubMed] [Google Scholar]
  • 185.Bruning A, Friese K, Burges A, Mylonas I. Tamoxifen enhances the cytotoxic effects of nelfinavir in breast cancer cells. Breast Cancer Res. 2010;12:45. doi: 10.1186/bcr2602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Moretti L, Kim KW, Jung DK, Willey CD, Lu B. Radiosensitization of solid tumors by Z-VAD, a pancaspase inhibitor. Mol Cancer Ther. 2009 doi: 10.1158/1535-7163.MCT-08-0893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.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]
  • 188.Yuk JM, Shin DM, Song KS, Lim K, Kim KH, Lee SH, et al. Bacillus calmette-guerin cell wall cytoskeleton enhances colon cancer radiosensitivity through autophagy. Autophagy. 2010;6:46–60. doi: 10.4161/auto.6.1.10325. [DOI] [PubMed] [Google Scholar]
  • 189.Chiu HW, Ho SY, Guo HR, Wang YJ. Combination treatment with arsenic trioxide and irradiation enhances autophagic effects in U118-MG cells through increased mitotic arrest and regulation of PI3K/Akt and ERK1/2 signaling pathways. Autophagy. 2009;5:472–483. doi: 10.4161/auto.5.4.7759. [DOI] [PubMed] [Google Scholar]
  • 190.Alonso MM, Jiang H, Yokoyama T, Xu J, Bekele NB, Lang FF, et al. Delta-24-RGD in combination with RAD001 induces enhanced anti-glioma effect via autophagic cell death. Mol Ther. 2008;16:487–493. doi: 10.1038/sj.mt.6300400. [DOI] [PubMed] [Google Scholar]
  • 191.Yokoyama T, Iwado E, Kondo Y, Aoki H, Hayashi Y, Georgescu MM, et al. Autophagy-inducing agents augment the antitumor effect of telerase-selve oncolytic adenovirus OBP-405 on glioblastoma cells. Gene Ther. 2008;15:1233–1239. doi: 10.1038/gt.2008.98. [DOI] [PubMed] [Google Scholar]
  • 192.Kim KW, Moretti L, Mitchell LR, Jung DK, Lu B. Combined Bcl-2/mammalian target of rapamycin inhibition leads to enhanced radiosensitization via induction of apoptosis and autophagy in non-small cell lung tumor xenograft model. Clin Cancer Res. 2009;15:6096–6105. doi: 10.1158/1078-0432.CCR-09-0589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Peng PL, Kuo WH, Tseng HC, Chou FP. Synergistic tumor-killing effect of radiation and berberine combined treatment in lung cancer: the contribution of autophagic cell death. Int J Radiat Oncol Biol Phys. 2008;70:529–542. doi: 10.1016/j.ijrobp.2007.08.034. [DOI] [PubMed] [Google Scholar]
  • 194.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]
  • 195.Martin AP, Park MA, Mitchell C, Walker T, Rahmani M, Thorburn A, et al. BCL-2 family inhibitors enhance histone deacetylase inhibitor and sorafenib lethality via autophagy and overcome blockade of the extrinsic pathway to facilitate killing. Mol Pharmacol. 2009;76:327–341. doi: 10.1124/mol.109.056309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Chiu HW, Lin JH, Chen YA, Ho SY, Wang YJ. Combination treatment with arsenic trioxide and irradiation enhances cell-killing effects in human fibrosarcoma cells in vitro and in vivo through induction of both autophagy and apoptosis. Autophagy. 2010;6:353–365. doi: 10.4161/auto.6.3.11229. [DOI] [PubMed] [Google Scholar]
  • 197.Lin CI, Whang EE, Donner DB, Du J, Lorch J, He F, et al. Autophagy induction with RAD001 enhances chemosensitivity and radiosensitivity through Met inhibition in papillary thyroid cancer. Mol Cancer Res. 2010;8:1217–1226. doi: 10.1158/1541-7786.MCR-10-0162. [DOI] [PubMed] [Google Scholar]

Articles from Cancer Biology & Therapy are provided here courtesy of Taylor & Francis

RESOURCES