Autophagy is a therapeutic target in anticancer drug resistance

Autophagy is a therapeutic target in anticancer drug resistance

Biochimica et Biophysica Acta 1806 (2010) 220–229 Contents lists available at ScienceDirect Biochimica et Biophysica Acta j o u r n a l h o m e p a ...

472KB Sizes 25 Downloads 182 Views

Biochimica et Biophysica Acta 1806 (2010) 220–229

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / b b a c a n

Review

Autophagy is a therapeutic target in anticancer drug resistance Suning Chen a,b, Sumaiyah K. Rehman c, Wei Zhang d, Aidong Wen a, Libo Yao b,⁎, Jian Zhang a,b,⁎ a

Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, People's Republic of China The Department of Biochemistry and Molecular Biology and The State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, People's Republic of China c Department of Molecular and Cellular Oncology, The University of Texas, MD Anderson Cancer Center and the University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, 77030, USA d Department of Biopharmaceutics, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, People's Republic of China b

a r t i c l e

i n f o

Article history: Received 29 April 2010 Received in revised form 5 July 2010 Accepted 7 July 2010 Available online 13 July 2010 Keywords: Autophagy Cancer Drug resistance

a b s t r a c t Autophagy is a type of cellular catabolic degradation response to nutrient starvation or metabolic stress. The main function of autophagy is to maintain intracellular metabolic homeostasis through degradation of unfolded or aggregated proteins and organelles. Although autophagic regulation is a complicated process, solid evidence demonstrates that the PI3K-Akt-mTOR, LKB1-AMPK-mTOR and p53 are the main upstream regulators of the autophagic pathway. Currently, there is a bulk of data indicating the important function of autophagy in cancer. It is noteworthy that autophagy facilitates the cancer cells' resistance to chemotherapy and radiation treatment. The abrogation of autophagy potentiates the re-sensitization of therapeutic resistant cancer cells to the anticancer treatment via autophagy inhibitors, such as 3-MA, CQ and BA, or knockdown of the autophagy related molecules. In this review, we summarize the accumulation of evidence for autophagy's involvement in mediating resistance of cancer cells to anticancer therapy and suggest that autophagy might be a potential therapeutic target in anticancer drug resistance in the future. © 2010 Elsevier B.V. All rights reserved.

Contents 1. 2.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Molecular regulation mechanism of autophagy in cancer . . . . . . . . . . . . 2.1. PI3K-Akt-mTOR . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. LKB1-AMPK-mTOR . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. p53 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4. Beclin1 and Bcl-2 . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.5. Other mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . 3. Double face of autophagy's function in cancer . . . . . . . . . . . . . . . . . 3.1. Autophagy inhibits tumorigenesis . . . . . . . . . . . . . . . . . . . . 3.2. Autophagy promotes cancer cells survival . . . . . . . . . . . . . . . 4. Autophagy inhibition facilitates anticancer drug resistance . . . . . . . . . . . 4.1. Autophagy confers the anticancer drug resistance . . . . . . . . . . . . 4.2. Autophagy inhibition sensitizes the tumor cells to anticancer therapy . . . 4.3. Autophagy inhibitors as the potential drug to overcome anticancer therapy 5. Concluding remarks. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . resistance . . . . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . .

220 221 221 221 221 222 222 222 222 222 223 223 224 224 225 226 226

1. Introduction ⁎ Corresponding authors. J. Zhang is to be contacted at Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, People's Republic of China. Tel./fax: + 86 29 84774513. L. Yao, Tel./fax: + 86 29 84774513. E-mail addresses: [email protected] (L. Yao), [email protected] (J. Zhang). 0304-419X/$ – see front matter © 2010 Elsevier B.V. All rights reserved. doi:10.1016/j.bbcan.2010.07.003

Autophagy, also called macroautophagy, is a kind of cellular catabolic degradation response to nutrient starvation or metabolic stress [1–3]. During the initial stages of autophagic process, cellular proteins, organelles and cytoplasm are sequestered and engulfed by the intracellular double-membrane-bound structures, called

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229

autophagosomes (early autophagic vesicles) [1,2,4,5]. These autophagosomes mature by fusing with lysosomes to form the autolysosomes (late autophagic vesicles) [2,4–8], in which the sequestered proteins and organelles are digested by lysosomal hydrolases and recycled to sustain cellular metabolism [9–13]. Whenever we talked about autophagy, we had to refer to apoptosis (process of programmed cell death), which shared several common regulatory elements with autophagy [14–16]. Autophagy was believed as a non-apoptotic programme of cell death or “type-II” cell death to distinguish with apoptosis [17]. Under most circumstances, autophagy promoted cell survival by adapting cells to the stress conditions, which was functionally paradoxical to apoptosis. However, it is still fundamentally important to clarify whether autophagy is a main strategy for cell survival, or if it also serves as a trigger for cell death [17]. Autophagy is an evolutionarily conserved process from yeast to mammals [3,18,19]. The main function of autophagy is to maintain intracellular metabolic homeostasis. In parallel with the ubiquitin proteasome degradation pathway, the dynamic autophagic process supervises and maintains the protein and organelle quality control to prevent the accumulation of unfolded and aggregated proteins [6– 8,11,13]. In addition to this key function, autophagy was also found to be responsible for other important functions, especially under stressful situation [18,20,21]. Whenever faced with environmental stressors, such as metabolic deprivation DNA damage caused by chemotherapy or radiation and hypoxia, the process of autophagy is activated which leads to cell survival or death [2,18,22–27]. There is an accumulation of evidence that highlights the important function of autophagy in cancer [2,5,22–25,28]. Although it is still controversial about whether autophagy kills cancer cells or sustains their survival under stressful conditions, more and more reports provide data to support that autophagy promotes cancer cell survival after chemotherapy or radiation therapy [22,29,30]. For example, autophagy facilitates resistance chronic myeloid leukemia (CML) to Imatinib mesylate (IM) [31], and also potentiates resistance of HER2 positive breast cancer cells to anti-HER2 monoclonal antibody trastuzumab [32]. Intriguingly, abrogation of autophagy by autophagy inhibitors, such as 3MA, CQ and BA, or by shRNA knockdown of autophagy related molecules, re-sensitizes the resistant cancer cells to the chemotherapy or radiation [30,31]. It is also noteworthy that the autophagic inhibitor hydroxychloroquine (HCQ) has already been applied in a clinical trial. So, it is possible to expect autophagic inhibitors to be the next generation of drugs to overcome anticancer therapeutic resistance. Herein, in this review, we will discuss the state-of-art of the molecular regulation mechanisms of autophagy, autophagy function in cancer, and emphasize the facilitation of autophagic inhibition in anticancer drug resistance.

221

constitutive PI3K activation, which are detected in many human cancers [48–53]. As the major downstream of class I PI3K, Akt can activate mTOR and lead to inhibition of autophagy [39,54]. Contrary to class I PI3K, class III PI3K can induce autophagy. So, the class III PI3K inhibitor, such as 3-MA, LY294002 and wortmannin, can repress the autophagy process [55–57]. In addition, the tumor suppressor gene PTEN can activate autophagy through inhibition of Akt activity [58]. Hence, the loss or mutation of PTEN will cause Akt activation and inhibit autophagy. Similar to PTEN, another tumor suppressor ARHI also can induce autophagy through inhibition of the PI3K-Akt signaling pathway [59]. As the downstream target of Akt, the serine/threonine kinase mTOR controls most of the autophagy-related genes, both transcriptionally and translationally. It is speculated that the inhibition of autophagy by mTOR is mainly dependent on p70S6 kinase and the eukaryotic initiation factor 4EBP1 activity [60,61]. Thus, the mTOR inhibitors rapamycin and RAD001 could induce autophagy via inhibition of mTOR activity and demonstrate their intriguing potential in cancer treatment [62–64]. 2.2. LKB1-AMPK-mTOR As the central metabolic sensor, AMP-activated protein kinase (AMPK) regulates lipid, cholesterol and glucose metabolism and is also closely related with cell metabolism and cancer [65,66]. AMPK activates under the energy stress conditions with increased AMP/ATP ratio to repress mTOR and initiate autophagy [67]. So far, the serine/ threonine kinase LKB1 (also known as STK11) is believed to be the major upstream regulator of AMPK, which directly phosphorylates and activates AMPKα at Thr172 site [68,69]. It is noteworthy that AMPK can also be activated by other upstream kinases, such as calcium and calmodulin-dependent protein kinase kinase 2 (CAMKK2), in hypothalamic neurons [70], T cells and endothelial cells [71,72]. It is suggested that these group of cells are more sensitive to response to the cellular calcium concentration, which signifies the link between calcium concentration mediated regulation and autophagy. Currently, Griselda et al. identified Transforming growth factor-β-activating kinase 1 (TAK1) as another new AMPK upregulator [73]. Whenever treated with the tumour necrosis factor-related apoptosis inducing ligand (TRAIL), the untransformed human breast epithelial MCF10A cells generated cytoprotective autophagy via AMPK-mTOR pathway, which was dependent on TAK1 and TAK1-binding subunit 2, but not the LKB1 or CAMKK2. Taken together, it is possible to identify more AMPK upregulators, but it is beyond doubt that the AMPK-mTOR-autophagy pathway already displays the well-recognized contribution to metabolic remodeling, cancer development and anticancer drug resistance.

2. Molecular regulation mechanism of autophagy in cancer 2.3. p53 Autophagy is a complicated regulatory process, which involves a great number of upstream regulating signaling pathways [2,22,28]. Nevertheless, no matter it is normal or cancer cells, the mammalian target of rapamycin (mTOR) serves as the main regulator of autophagy [22,33–43]. In response to nutrient availability, mTOR activation suppresses autophagy and stimulates cell proliferation. However, under nutrient deprivation, hypoxia, genomic instability or other stress conditions, the suppression of mTOR triggers the autophagic cascade and inhibits the cell proliferation [21,22,34,35,44]. Herein, we mainly introduce PI3K-Akt-mTOR, LKB1-AMPK-mTOR, P53, Beclin1 and Bcl-2 pathway in the regulation of autophagy. 2.1. PI3K-Akt-mTOR Increasing evidence shows that the over-active PI3K-Akt pathway promotes tumor cell survival [45–47]. The somatic mutations of PI3K, such as E542K, E545D and E545K, are common mutations that lead to

As a well-known tumor suppressor gene, p53 also involves in genotoxic stress response and DNA damage repair [74–76]. Therefore, it is no surprise that p53 can also regulate autophagy [77–79], though p53's function in the control of autophagy is a little complicated [80,81] mainly due to the intricate cellular microenvironment. On one hand, p53 can activate AMPK and then inhibit mTOR to induce the autophagy process [67,82]. Simultaneously, p53 promotes autophagy through triggering other downstream targets, such as Sestrin2 [83,84], damage-regulated autophagy modulator (DRAM) [78,85], Bcl-2-associated X protein (Bax) and p53-upregulated modulator of apoptosis (PUMA) [86], which contribute to the induction of apoptosis. Under nutrient deprivation or other stressful conditions, p53 activates autophagy in adaptation to the low ATP level, which facilitates the apoptosis course. Therefore, it is concordant with the tumor suppressor function of p53 in the positive regulatory process of autophagy [78].

222

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229

On the other hand, in addition to the autophagy inducer, p53 also can repress autophagy [87]. Tasdemir et al. [88,89] showed that depletion of p53 either in vitro by RNA interference or pharmacological inhibition in cell lines or in vivo by genetic deletion in mice could induce autophagy. And the autophagy induced by p53 loss promotes the survival of p53deficient cells to sustain high ATP levels under conditions of hypoxia and nutrient depletion [89]. Therefore, this dual interplay between p53 and regulation of autophagy is still indefinable due to possible key underlying aspects of metabolism and cancer biology. Actually, it is difficult to reconcile both positive and negative function of p53 in autophagy regulation. Taken together, it is important to note that these two distinct effects take place under different stress conditions, which reflects the ability of p53 to promote cell survival during low levels of stress or damage and induce cell death when the damage is more serious [90,91]. 2.4. Beclin1 and Bcl-2 Beclin1 (also called Atg6) is essential for the double-membrane autophagosome formation, which is required during the initial steps of autophagy [92]. The autophagic induction of Beclin1 facilitates the inhibition of tumorigenesis [93]. The allelic loss of Beclin1 is frequently seen in human breast, ovarian and prostate cancers [94]. Beclin1 promotes the initiation of autophagy mainly through binding with other autophagy-related proteins such as Bcl-2, Vps34, p150, UVRAG, Bif1, Atg14L and Rubicon to form huge protein complexes [95–99]. Bcl-2 proto-oncogene, overexpressed in 50–70% of breast cancers, is one of the major causes that induces resistance to chemotherapy and hormone therapy-induced apoptosis. In addition to the wellestablished anti-apoptosis function, Bcl-2 is also well-documented to inhibit autophagy via the physical interaction with Beclin1 [98]. And the autophagy inhibition might be the accumulative effect on the oncogenic properties of Bcl-2 family. Akar et al. [100] reported that RNAi knockdown of Bcl-2 induced the autophagy and apoptosis in MCF-7 breast cancer cells. Simultaneously, Saeki et al. [101] found that Bcl-2 silencing by antisense mRNA induced the autophagy-dependent cell death in human leukemia HL60 cells. The interaction of Beclin1 with Bcl-2 is mainly dependent on its BH3 domain with BH3-receptor domain on Bcl-2 proteins [102]. Therefore, in addition to the direct knockdown of Bcl-2, it is also feasible to abrogate some of their interactions by some BH3 mimics to induce autophagy. As one BH3-mimetic compound, ABT737 could induce autophagy through competitively disrupting the inhibitory interaction between Beclin1 and Bcl-2/Bcl-X [103,104]. At least, silencing Bcl-2 or disrupting the Beclin1 and Bcl-2 interaction to induce autophagic cell death might be used as a novel therapeutic strategy in cancer cells overexpressing Bcl-2. 2.5. Other mechanisms Under hypoxia and nutrient deprivation condition, a great number of unfolded or misfolded proteins can trigger endoplasmic reticulum (ER) stress. There are evidences showing that the accumulation of ER stress could induce autophagy through protein kinase-like endoplasmic reticulum kinase (PERK) and inositol-requiring kinase 1 (IRE1) dependent mechanisms in the heart [105,106]. Although there is no documented report of ER stress inducing autophagy currently in cancer cells, it is worthwhile to exploit the adaptive mechanism and biological function for therapeutic benefit of cancer treatment. Recently, Criollo et al. [107] reported that IKK (IκB kinase) stimulated autophagy in an NF-κB-independent manner. They found that the constitutively active IKK subunits stimulated autophagy through activating multiple signals, including the phosphorylation of AMPK and JNK1 in starvation-induced condition. These results indicate that IKK has a pivotal role in the stimulation of autophagy by physiological and pharmacological stimuli.

3. Double face of autophagy's function in cancer Although the regulatory mechanism of autophagy is partially manifested, however, the function of autophagy in cancer is still in an undetermined debate. Whether autophagy is a death-induced mechanism or a protective effort for cellular survival is still a controversy [1,2]. There are a number of evidences to support the double-faced function of autophagy in cancer: tumor suppression and also promotion. 3.1. Autophagy inhibits tumorigenesis Distinct from apoptosis, autophagy is termed the type II programmed cell death (PCD) [108]. The basal level of autophagy in normal tissues provides a very important homeostatic and housekeeping function. Autophagy and ubiquitin proteasome degradation system monitor and clean the aggregated unfolded or misfolded proteins, and damaged cellular organelles to maintain intracellular homeostasis [6–8,11,13]. Therefore, abrogation of the dynamic autophagy pathway will result in disease or tumorigenesis. The autophagy related genes knockout mice with Atg5−/− or Atg7−/− developed the progressive deficits in motor function and behavioral defects [9,10]. The major reason is due to the loss of functional autophagy to induce the accumulation of cytoplasmic inclusion bodies. These results suggest that the functional clearance of aggregated cytosolic proteins through basal autophagy is crucial for preventing the accumulation of abnormal proteins. The tumor suppressor role of autophagy was firstly documented through genetic approaches of Beclin1. In 1999, Liang et al. [93] reported Beclin1 as a potential tumor suppressor gene, the expression of which is frequently decreased in human breast epithelial carcinoma cell lines and tissue compared to the higher level in normal tissue. These findings suggested that decreased expression of autophagy proteins might contribute to the initiation or progression of breast cancer. This notion was further confirmed by others Yue et al. discovered that Beclin1−/− mice died early in embryogenesis and Beclin1+/− mutant mice suffered from a high incidence of spontaneous tumors [109]. Also, the heterozygous disruption of Beclin1 increases the spontaneous malignancies frequency and accelerates the development of hepatitis B virusinduced premalignant lesions [110]. These data provided the direct proof of the function of Beclin 1 as a tumor suppressor. Actually, the tumor suppressor function is not only subjected to Beclin1. Alteration of other autophagic genes has also been reported in other tumor types. Marino et al. [111] found that Atg4C knockout mice show an increased susceptibility to the development of fibrosarcomas induced by chemical carcinogens. Deletion of Atg5 conferred natural killer (NK) cells malignancy [112]. Bax-interacting factor-1 (Bif-1) protein, a positive regulator of apoptosis and autophagy, plays a critical role in tumorigenesis. Loss of Bif-1 suppresses programmed cell death and promotes colon adenocarcinomas [113]. Another important autophagy-related molecule, Ultraviolet radiation resistance-associated gene (UVRAG) could bind with Beclin1 and induce autophagy [114]. The frame shift mutations of UVRAG gene exon 8 resulted in decreased autophagy activity and induced the colorectal and gastric carcinomas occurrence [114,115]. All these data support the tumor suppressive function of autophagy. The inhibition of tumorigenesis by autophagy is a common effect, and not limited to one unique autophagic molecule. However, the detailed mechanism by which autophagy functions as a tumor suppressor has not been fully elucidated. Nonetheless, it is believed that the failure of DNA repair to maintain genome integrity from genotoxic stress might be the major reason [2,4]. 3.2. Autophagy promotes cancer cells survival Although many research results display the tumor suppressive function of autophagy, there is growing evidence that supports the

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229

autophagic role in tumor survival. During progression, cancer cells require plenty of nutrients and oxygen to sustain the rapid cell proliferation. Unfortunately, many tumors undergo the hypoxia and metabolic stress during the progression and invasion, especially solid tumors with their poor vascularization. But how do these cells survive under the metabolic stress conditions? Part of the answer is that autophagy can maintain and promote autophagic tumor cells’ survival. Metabolic stress can robustly induce autophagy in cancer cells with defective apoptosis [116]. Thus, autophagy inhibition by constitutively active Akt or allelic disruption of Beclin1 confers sensitivity to metabolic stress by inhibiting the autophagy-dependent survival pathway in vitro and in vivo. Karantza-Wadsworth [23] found that defective autophagy induced by the allelic loss of Beclin1 sensitized mammary epithelial cells to metabolic stress and accelerated lumen formation in mammary acini. In the growth factordependent Bax/Bak-deficient cells, autophagy can prevent the cell death following growth factor withdrawal [117]. Cancer cells can initiate, proliferate and propagate in many kinds of serious stress conditions, such as glucose starvation, hypoxia, growth-factor withdrawal and even the cytotoxic cellular damage of chemotherapy or radiation [2,4,91,117]. In general, autophagy activation follows the rule of the “Survival of the Fittest” for the cancerous cells. Autophagy facilitates the removal of damaged proteins and organelles, and functions as a back-up energy reserve to provide extra energy to sustain the tumor cells' survival against anticancer therapy. Thus, autophagy may function in tumor promotion by mitigating and limiting metabolic stress to protect the genome’s integrity and confer resistance to anticancer therapy. On the contrary, defective autophagy facilitates DNA damage and genomic instability which ultimately confers apoptosis or death upon cancer cells.

4. Autophagy inhibition facilitates anticancer drug resistance 4.1. Autophagy confers the anticancer drug resistance Adjuvant cancer treatment, such as chemotherapy or radiotherapy, etc., is very important to prevent or postpone the cancer’s relapse and prolong the patients' survival. However, one of the most daunting clinical problems is the frequent relapse after the treatment even with longer dormancy [2,118]. Most of the therapeutic failures are due to the

Table 1 Known sensitizing function of autophagy inhibition in breast cancer. Cancer cell types

Inhibition targets of autophagy

Treatment

Ref.

Breast cancer cells lines with HER2 overexpression SKBR3 Breast cancer cells lines with ER overexpression T47D and MCF-7 Breast cancer cells lines with ER overexpression T47D and MCF-7 Breast cancer cells lines MCF7 and MDA-MB-231 Breast cancer cells lines MCF-7/LCC9 Breast cancer cell lines MCF-7/MCF-7. Beclin1 Breast cancer cell lines MDA-MB-231

Knockdown of LC3

anti-HER2 monoclonal antibody Trastuzumab

[32]

Pharmacological inhibitor 3-MA

Estrogen receptor antagonist Tamoxifen

[119–121]

Knockdown of Beclin1 Knockdown of Atg5, Beclin1 and Atg7

Estrogen receptor antagonist Tamoxifen

[122]

Knockdown of LC3B, ATF4 and HDAC6

Inhibitor of the 26 S proteasome Bortezomib

[123]

Knockdown of Beclin 1 Estrogen receptor antagonist Faslodex (FAS) Knockdown of Beclin 1 Estrogen receptor antagonist Faslodex (FAS) and Tamoxifen Knockdown of Beclin 1 Atg3 Atg4b, Atg4c, Atg5 and atg12

γ-radiation

[124]

[121]

[30]

intrinsic or acquired resistance towards the therapy. Presumably, autophagy is one of the most important mechanisms to enable cancer cells' survival and eventual recurrence after long-term cytotoxic treatment. Especially during the acquired resistance process, therapeutic drugs can kill most of the cancer cells. But a few remaining cells tolerate the dramatically harsh and stressful conditions (Fig. 1) due to the cell’s protective mechanism of autophagy. The recovery of this group of cells results in the tumor to relapse once growth conditions are

Table 2 Known sensitizing function of autophagy inhibition in colorectal cancer. Cancer cell types

Inhibition targets of autophagy

Colon cancer cell lines HT29 and HCT116

Vorinostat Pharmacological inhibitors chloroquine (CQ), Knockdown of Atg7 Pharmacological TFT and 5-FU inhibitors 3-MA

[125]

Pharmacological inhibitors 3-MA Knockdown of Beclin1 and UVRAG Pharmacological inhibitors PepA, Knockdown of Atg7 Pharmacological inhibitors 3-MA

5-FU

[56]

UVRAG

[114]

Colorectal cancer cell lines WiDR, Lovo92 and Colo320 Colorectal cancer cell lines colon26 and HT29 Colorectal cancer cell lines HCT116 Colorectal cancer cell lines SW480, DLD-1, WiDr, SW620 and LoVo Colon cancer cell lines HT-29 Colon cancer cell lines HCT116

Fig. 1. Autophagy confers the anticancer therapy resistance. Cancer cells can tolerate the metabolic stress caused by anticancer therapy, including chemotherapy, radiation, tyrosine kinase inhibitors, target therapy antibody, etc. The induced autophagy facilitates to reserve the energy and promote the cancer cells survival, which finally confer the anticancer therapy resistance.

223

Colorectal cancer cell lines DLD1 and HT29 Rectum carcinoma cell lines SW707

Pharmacological nhibitors 3-MA, E64D and PepA Knockdown of atg5, atg7, Beclin1 and UVRAG Knockdown of Beclin1 Knockdown of Beclin 1 Atg3 Atg4b, Atg4c, Atg5 and atg12

Treatment

Ref.

[126]

Amino acid and [127] glucose deprivation Cyclooxygenase inhibition with Sulindac sulfide TRAIL

[128]

Hypoxia and Bortezomib γ-radiation

[130]

[129]

[30]

224

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229

Table 3 Known sensitizing function of autophagy inhibition in glioblastoma and lung cancer. Cancer cell types

Inhibition targets of autophagy

Treatment

Ref.

CD133+

Pharmacological inhibitor 3-MA and BA Knockdown of Beclin 1 and Atg5 Pharmacological inhibitor 3-MA and BA

γ-radiation

[26,131]

Glioma stem cells Glioma cell lines U87-MG and U373-MG Glioma cell lines LN229, LZN308, U87-MG and U373-MG Astrocytoma, Glioblastoma cells Glioma cell lines

U251, SF188 and U373 Glioma cell lines A172, T98G, U87-MG and U373-MG Lung cancer cell lines A549

Pharmacological inhibitors 3-MA and BA Knockdown of Atg12-Atg5

Cancer cell types

Inhibition targets of autophagy

Pharmacological inhibitors BA and CQ, Knockdown of Atg5 and Atg7 K562, BaF3/E255K cells and Pharmacological BaF3/T315I CML cells inhibitors CQ CML cells Pharmacological inhibitors CQ Chronic myeloid leukemia (CML) cells K562, BV173 and 32D-p210BCR/ABL

N-(4-hydroxyphenyl) [132] retinamide (4-HRP) induced autophagy tyrosine kinase [133] inhibitor (TKI) Imatinib

AGT inhibitor O6-benzylguanine (BG) Pharmacological inhibitors 3-MA and oligomycin Knockdown of Beclin1

Temozolomide (TMZ)

[134]

Temozolomide (TMZ) and Etoposide

[135]

Pharmacological inhibitors 3-MA and BA

Temozolomide (TMZ) [136]

Knockdown of Beclin 1 Atg3 Atg4b, Atg4c, Atg5 and atg12

γ-radiation

[30]

favourable. Since autophagy confers anticancer drug resistance mainly through allowing the residual or metastasized tumor cells to tolerate the cytotoxic stress, the potent inhibition of autophagy in resistant cancer cells might be an efficient approach for cancer eradication. 4.2. Autophagy inhibition sensitizes the tumor cells to anticancer therapy It is remarkable to notice that a great number of studies show that inhibition of autophagy confers cancer cells sensitive to a wide spectrum of therapies. Currently, there are several potential strategies to inhibit autophagy, including autophagy inhibitors and shRNA approach, to re-sensitize the resistant cancer cells to anticancer therapy. The accumulated data show that autophagy is a shared mechanism to induce the tumor cells resistant to different cancer therapies, such as chemotherapy, radiotherapy and targeted therapy

Table 4 Known sensitizing function of autophagy inhibition in cervical and prostate cancer. Cancer cell types

Inhibition targets of autophagy Treatment

Cervical cancer cell lines HeLa

Knockdown of Beclin1

Cervical cancer cell lines Hela

Pharmacological inhibitors 3-MA, HCQ, bafilomycin A1 and monensin; shRNA Knockdown of Atg5, Atg6, Beclin1, Atg10, and Atg12 Inhibition of Bif-1 Nutrient [97] (Endophilin B1) deprivation Knockdown of Beclin 1 Atg3 γ-radiation [30] Atg4b, Atg4c, Atg5 and atg12

Cervical cancer cells Hela HTB35 cervical squamous cell carcinoma cells Normal human prostate epithelial cells and DU145 prostate cancer cells Human prostate epithelial cells (PrEC) LNCap prostate cancer cell line

Table 5 Known sensitizing function of autophagy inhibition in leukemia, esophageal squamous carcinoma and pharyngeal cancer cells.

Ref.

[130] Hypoxia and Bortezomib Nutrient [137] depletion

Pharmacological inhibitors 3-MA

TRAIL and FADD

[138]

Myc overexpression abrogating autophagy Pharmacological inhibitors 3-MA Knockdown of Beclin1

Rapamycin [139]

Ba/F3, K562 and LAMA 84 CML cells and primary human CML cells Esophageal squamous carcinoma (ESCC) EC9706 cell line Pharyngeal HTB43 cancer cells

Knockdown of Beclin 1 Atg3 Atg4b, Atg4c, Atg5 and atg12

Ref.

Imatinib

[31,141]

Imatinib and TPA Imatinib and HDAC inhibitor SAHA Imatinib and HDAC inhibitor SAHA Cisplatin (DDP)

[142]

γ-radiation

[30]

[143]

[29]

[55]

All cell lines are human origin. Abbreviation: 3-MA, 3-methyladenine; Baf-A1, bafilomycin A1; CQ, chloroquine, HCQ, hydroxychloroquine; PepA, pepstatin A; UVRAG, ultraviolet radiation resistanceassociated gene; shRNA, short hairpin RNA.

etc. For example, increased autophagy induced the resistance to the HER2 monoclonal antibody Trastuzumab which is a cytostatic agent used for HER2 positive breast cancers and mediates its effects by targeting HER2 [32]. Interestingly, knockdown of LC3 expression via shRNA resulted in resistant cells being re-sensitized to Trastuzumab treatment [32]. Also, inhibition of autophagy using either pharmacological inhibitors or RNAi of essential autophagy genes potentiates cell death induced by Imatinib mesylate in CML cells [31]. Repression of autophagy enhances the therapeutic efficacy of cisplatin and 5-FU in esophageal and colon cancer, respectively [55,56]. In the following section, we summarize the updated research data according to the different tumor classification, such as breast cancer (Table 1), colorectal cancer (Table 2), glioblastoma (Table 3), cervical and prostate cancer (Table 4), leukemia and esophageal squamous carcinoma (Table 5). In each table, we clearly list out the cancer cell types or cell lines, the inhibited targets of autophagy and the treatment regimen according to the references. 4.3. Autophagy inhibitors as the potential drug to overcome anticancer therapy resistance So far, different inhibitors of autophagy which affect different stages of the autophagy pathway have been developed and used in the studies Table 6 Autophagy inhibitors. Compound

Mechanism & target

Effect

Ref.

3-methyladenine (3-MA) Chloroquine (CQ) Bafilomycin A1 (BA) Hydroxychloroquine (HCQ) Wortmannin

Class III PI3K inhibitor Lysosomal pH

Autophagy inhibition

Vacuolar-ATPase

Autophagy inhibition

Lysosomal pH

Autophagy inhibition

[118,126, 144–156] [29,59,125, 143,157–166] [58,136, 167–169] [158,170]

Class III PI3K inhibitor Change Endocytic and Lysosomal pH Class III PI3K inhibitor

Autophagy inhibition

[171–176]

Autophagy inhibition

[171,177,178]

Autophagy inhibition

[179–182]

Monensin Androgen [140] deprivation

Pharmacological inhibitors 3-MA and CQ, Pharmacological inhibitors 3-MA

Treatment

LY294002

Autophagy inhibition

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229

225

via different mechanisms, which might be used to re-sensitize the resistant cancer cells to anticancer therapy. Since autophagy might be one of the main reasons to induce the cancer cells survival to anticancer therapy, the investigators want to know whether blocking autophagy while giving standard treatment will improve the treatment of cancer. It is noteworthy that some autophagy inhibitors have already been applied in clinical trials (http://clinicaltrials.gov/ct2/results?term=autophagy). Currently, investigators have mainly explored the potential clinical application of the autophagy inhibitor hydroxychloroquine (HCQ), in addition to the standard regimen in the treatment of cancer, such as non-small cell lung cancer (NSCLC), breast cancer, metastatic prostate cancer, colon cancer, refractory multiple myeloma, melanoma and other solid cancer types (Table 7). It is promising to see inhibitors of autophagy becoming the new therapy in anticancer therapy.

5. Concluding remarks Fig. 2. Autophagy inhibitors facilitate to re-sensitize the resistant cancer cells to antitumor therapies. Autophagy inhibitors such as Hydroxychloroquine (HCQ), 3-methyladenine (3-MA) and Bafilomycin A1 (BA) can facilitate the re-sensitization of the resistant cancer cells to antitumor therapies via abrogating the autophagic cells survival function. It is promising to expect the autophagy inhibitors to be the next generation drugs to overcome the resistant cancer cells to antitumor therapies.

of autophagy in tumorigenesis and cancer therapy. As shown in Table 1 to Table 5, much evidence demonstrates that the autophagy inhibitors facilitate the re-sensitization of resistant tumor cells to anticancer treatment. Herein, the current available autophagy inhibitors are summarized in Table 6. Different autophagy inhibitors block the autophagy effect through different mechanisms. For example, 3-MA, LY294002 and Wortmannin can inhibit the PI3K activity [55,56], one of the major upstream regulators of autophagy. Hydroxychloroquine (HCQ) or chloroquine (CQ) can inhibit autophagosome fusion with lysosomes and autophagosome degradation [31]. As shown in Fig. 2, autophagy inhibitors can abrogate the cell survival effect of autophagy

One of the most daunting clinical issues is the frequent tumors progression after standard treatment, mainly due to therapeutic resistance. It is urgent to elucidate the mechanisms that induce anticancer drug resistance. As we discussed in this review, although the controversy about the pro- or anticancer effect of autophagy is still heated, the in vitro and in vivo data fully support that autophagy can facilitate the tumor cells' survival to anticancer treatment [2,4,21,55,56,183]. The data suggest that cancer cells may take advantage of autophagy to survive despite of treatment with anticancer therapy regimens, such as chemotherapy or radiation. So, it is possible to inhibit autophagy through shRNA knockdown or inhibitors to promote the conversion from autophagic survival to apoptotic process. It is noteworthy that the autophagy inhibitors such as Hydroxychloroquine (HCQ), 3methyladenine (3-MA) and Bafilomycin A1 (BA) can effectively sensitize the cancer cells to therapeutic agents. Therefore, in the near future, it is promising to expect autophagy inhibitors, such as HCQ, in clinical trials, to be the next generation drugs that re-sensitize the resistant cancer cells to anticancer therapies.

Table 7 Clinical trials of autophagy inhibitors in cancer therapy. Cancer types

Treatment regimen

Sponsor

Starting date

Estimated Primary Completion Date

Clinical Trials. gov ID

Study phase

Non-Small Cell Lung Cancer (NSCLC) Metastatic Breast Cancer Ductal Carcinoma in Situ (breast) Refractory Solid Tumors Metastatic Prostate Cancer Metastatic Colorectal Cancer Advanced Non-Small Cell Lung Cancer (NSCLC) Relapsed or Refractory Multiple Myeloma Stage III or IV Resectable Melanoma Small Cell Lung Cancer Advanced Solid Tumors

Hydroxychloroquine, Carboplatin, Paclitaxel and Bevacizumb Ixabepilone and Hydroxychloroquine Tamoxifen and Chloroquine

University of Medicine and Dentistry New Jersey Cancer Institute of New Jersey

June 2008

June 2012

NCT00933803

Phase I/II

February 2009

August 2011

NCT00765765

Phase I/II

Inova Health Care Services

December 2009

December 2011

NCT01023477

Phase I/II

Hydroxychloroquine and Temsirolimus Docetaxel and Hydroxychloroquine Hydroxychloroquine, Capecitabine, Oxaliplatin, and Bevacizumab Hydroxychloroquine, Carboplatin, Paclitaxel, and Bevacizumab Hydroxychloroquine and Bortezomib Hydroxychloroquine

University of Pennsylvania

October 2008

May 2012

NCT00909831

Phase I

Cancer Institute of New Jersey

December 2008

October 2011

NCT00786682

Phase II

Cancer Institute of New Jersey

May 2009

July 2011

NCT01006369

Phase II

Cancer Institute of New Jersey

June 2008

August 2011

NCT00728845

Phase I/II

University of Pennsylvania

November 2007

November 2009

NCT00568880

Phase I/II

Cancer Institute of New Jersey

March 2009

March 2010

NCT00962845

Phase I

Chloroquine and A-CQ 100 Hydroxychloroquine and Vorinostat

June 2010 November 2009

December 2015 November 2010

NCT00969306 NCT01023737

Phase I/II Phase I

Breast Cancer

Ritonavir

October 2009

October 2013

NCT01009437

Phase I/II

Renal Cell Carcinoma Pancreatic Cancer

Hydroxychloroquine Hydroxychloroquine and Gemcitabine

Maastricht Radiation Oncology University of Texas Health Science Center at San Antonio Masonic Cancer Center, University of Minnesota University of Pittsburgh University of Pittsburgh

July 2010 June 2010

July 2012 June 2012

NCT01144169 NCT01128296

Phase I Phase I/II

226

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229

Acknowledgments The authors would like to acknowledge support from Chinese National Key Basic Research & Development Program (2009CB521704). References [1] J.J. Lum, R.J. DeBerardinis, C.B. Thompson, Autophagy in metazoans: cell survival in the land of plenty, Nat. Rev. Mol. Cell Biol. 6 (2005) 439–448. [2] R. Mathew, V. Karantza-Wadsworth, E. White, Role of autophagy in cancer, Nat. Rev. Cancer 7 (2007) 961–967. [3] B. Levine, D.J. Klionsky, Development by self-digestion: molecular mechanisms and biological functions of autophagy, Dev. Cell 6 (2004) 463–477. [4] Y. Kondo, T. Kanzawa, R. Sawaya, S. Kondo, The role of autophagy in cancer development and response to therapy, Nat. Rev. Cancer 5 (2005) 726–734. [5] S. Jin, E. White, Role of autophagy in cancer: management of metabolic stress, Autophagy 3 (2007) 28–31. [6] Z. Yang, D.J. Klionsky, An overview of the molecular mechanism of autophagy, Curr. Top. Microbiol. Immunol. 335 (2009) 1–32. [7] Y.P. Yang, Z.Q. Liang, Z.L. Gu, Z.H. Qin, Molecular mechanism and regulation of autophagy, Acta Pharmacol. Sin. 26 (2005) 1421–1434. [8] C.W. Wang, D.J. Klionsky, The molecular mechanism of autophagy, Mol. Med. 9 (2003) 65–76. [9] T. Hara, K. Nakamura, M. Matsui, A. Yamamoto, Y. Nakahara, R. Suzuki-Migishima, M. Yokoyama, K. Mishima, I. Saito, H. Okano, N. Mizushima, Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice, Nature 441 (2006) 885–889. [10] M. Komatsu, S. Waguri, T. Chiba, S. Murata, J. Iwata, I. Tanida, T. Ueno, M. Koike, Y. Uchiyama, E. Kominami, K. Tanaka, Loss of autophagy in the central nervous system causes neurodegeneration in mice, Nature 441 (2006) 880–884. [11] M. Komatsu, S. Waguri, T. Ueno, J. Iwata, S. Murata, I. Tanida, J. Ezaki, N. Mizushima, Y. Ohsumi, Y. Uchiyama, E. Kominami, K. Tanaka, T. Chiba, Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice, J. Cell Biol. 169 (2005) 425–434. [12] B. Ravikumar, R. Duden, D.C. Rubinsztein, Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy, Hum. Mol. Genet. 11 (2002) 1107–1117. [13] A. Williams, L. Jahreiss, S. Sarkar, S. Saiki, F.M. Menzies, B. Ravikumar, D.C. Rubinsztein, Aggregate-prone proteins are cleared from the cytosol by autophagy: therapeutic implications, Curr. Top. Dev. Biol. 76 (2006) 89–101. [14] M.C. Maiuri, E. Zalckvar, A. Kimchi, G. Kroemer, Self-eating and self-killing: crosstalk between autophagy and apoptosis, Nat. Rev. Mol. Cell Biol. 8 (2007) 741–752. [15] J.M. Vicencio, L. Galluzzi, N. Tajeddine, C. Ortiz, A. Criollo, E. Tasdemir, E. Morselli, A. Ben Younes, M.C. Maiuri, S. Lavandero, G. Kroemer, Senescence, apoptosis or autophagy? When a damaged cell must decide its path—a mini-review, Gerontology 54 (2008) 92–99. [16] J. Moscat, M.T. Diaz-Meco, p62 at the crossroads of autophagy, apoptosis, and cancer, Cell 137 (2009) 1001–1004. [17] A. Rami, Review: autophagy in neurodegeneration: firefighter and/or incendiarist? Neuropathol. Appl. Neurobiol. 35 (2009) 449–461. [18] D.J. Klionsky, S.D. Emr, Autophagy as a regulated pathway of cellular degradation, Science 290 (2000) 1717–1721. [19] A.J. Meijer, P. Codogno, Regulation and role of autophagy in mammalian cells, Int. J. Biochem. Cell Biol. 36 (2004) 2445–2462. [20] R.G. Jones, D.R. Plas, S. Kubek, M. Buzzai, J. Mu, Y. Xu, M.J. Birnbaum, C.B. Thompson, AMP-activated protein kinase induces a p53-dependent metabolic checkpoint, Mol. Cell 18 (2005) 283–293. [21] S. Yousefi, H.U. Simon, Autophagy in cancer and chemotherapy, Results Probl. Cell Differ. (2009). [22] N. Chen, V. Karantza-Wadsworth, Role and regulation of autophagy in cancer, Biochim. Biophys. Acta 1793 (2009) 1516–1523. [23] V. Karantza-Wadsworth, S. Patel, O. Kravchuk, G. Chen, R. Mathew, S. Jin, E. White, Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis, Genes Dev. 21 (2007) 1621–1635. [24] V. Karantza-Wadsworth, E. White, Role of autophagy in breast cancer, Autophagy 3 (2007) 610–613. [25] B. Levine, Unraveling the role of autophagy in cancer, Autophagy 2 (2006) 65–66. [26] S.L. Lomonaco, S. Finniss, C. Xiang, A. Decarvalho, F. Umansky, S.N. Kalkanis, T. Mikkelsen, C. Brodie, The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells, Int. J. Cancer 125 (2009) 717–722. [27] I. Papandreou, A.L. Lim, K. Laderoute, N.C. Denko, Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L, Cell Death Differ. 15 (2008) 1572–1581. [28] N. Chen, J. Debnath, Autophagy and tumorigenesis, FEBS Lett. (2009). [29] J.S. Carew, S.T. Nawrocki, C.N. Kahue, H. Zhang, C. Yang, L. Chung, J.A. Houghton, P. Huang, F.J. Giles, J.L. Cleveland, Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance, Blood 110 (2007) 313–322. [30] A. Apel, I. Herr, H. Schwarz, H.P. Rodemann, A. Mayer, Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy, Cancer Res. 68 (2008) 1485–1494. [31] C. Bellodi, M.R. Lidonnici, A. Hamilton, G.V. Helgason, A.R. Soliera, M. Ronchetti, S. Galavotti, K.W. Young, T. Selmi, R. Yacobi, R.A. Van Etten, N. Donato, A. Hunter, D. Dinsdale, E. Tirro, P. Vigneri, P. Nicotera, M.J. Dyer, T. Holyoake, P. Salomoni, B. Calabretta, Targeting autophagy potentiates tyrosine kinase inhibitor-induced

[32]

[33] [34] [35]

[36]

[37] [38]

[39]

[40]

[41]

[42]

[43]

[44] [45] [46] [47]

[48]

[49]

[50] [51]

[52]

[53]

[54]

[55]

[56]

[57]

[58]

[59]

cell death in Philadelphia chromosome-positive cells, including primary CML stem cells, J. Clin. Invest. 119 (2009) 1109–1123. A. Vazquez-Martin, C. Oliveras-Ferraros, J.A. Menendez, Autophagy facilitates the development of breast cancer resistance to the anti-HER2 monoclonal antibody trastuzumab, PLoS ONE 4 (2009) e6251. L. Qin, Z. Wang, L. Tao, Y. Wang, ER stress negatively regulates AKT/TSC/mTOR pathway to enhance autophagy, Autophagy 6 (2010). C.H. Jung, S.H. Ro, J. Cao, N.M. Otto, D.H. Kim, mTOR regulation of autophagy, FEBS Lett. (2010). K.C. Choi, S.H. Kim, J.Y. Ha, S.T. Kim, J.H. Son, A novel mTOR activating protein protects dopamine neurons against oxidative stress by repressing autophagy related cell death, J. Neurochem. 112 (2010) 366–376. A. Sobolewska, M. Gajewska, J. Zarzynska, B. Gajkowska, T. Motyl, IGF-I, EGF, and sex steroids regulate autophagy in bovine mammary epithelial cells via the mTOR pathway, Eur. J. Cell Biol. 88 (2009) 117–130. J.M. Rosenbluth, J.A. Pietenpol, mTOR regulates autophagy-associated genes downstream of p73, Autophagy 5 (2009) 114–116. L. Annovazzi, M. Mellai, V. Caldera, G. Valente, L. Tessitore, D. Schiffer, mTOR, S6 and AKT expression in relation to proliferation and apoptosis/autophagy in glioma, Anticancer Res. 29 (2009) 3087–3094. N. Shinojima, T. Yokoyama, Y. Kondo, S. Kondo, Roles of the Akt/mTOR/p70S6K and ERK1/2 signaling pathways in curcumin-induced autophagy, Autophagy 3 (2007) 635–637. S. Sarkar, J.E. Davies, Z. Huang, A. Tunnacliffe, D.C. Rubinsztein, Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein, J. Biol. Chem. 282 (2007) 5641–5652. W. Martinet, S. Verheye, G.R. De Meyer, Everolimus-induced mTOR inhibition selectively depletes macrophages in atherosclerotic plaques by autophagy, Autophagy 3 (2007) 241–244. J.J. Jaboin, E.T. Shinohara, L. Moretti, E.S. Yang, J.M. Kaminski, B. Lu, The role of mTOR inhibition in augmenting radiation induced autophagy, Technol. Cancer Res. Treat. 6 (2007) 443–447. B. Ravikumar, C. Vacher, Z. Berger, J.E. Davies, S. Luo, L.G. Oroz, F. Scaravilli, D.F. Easton, R. Duden, C.J. O'Kane, D.C. Rubinsztein, Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease, Nat. Genet. 36 (2004) 585–595. P.M. Jaakkola, J.P. Pursiheimo, p62 degradation by autophagy: another way for cancer cells to survive under hypoxia, Autophagy 5 (2009) 410–412. P. Blume-Jensen, T. Hunter, Oncogenic kinase signalling, Nature 411 (2001) 355–365. I. Vivanco, C.L. Sawyers, The phosphatidylinositol 3-Kinase AKT pathway in human cancer, Nat. Rev. Cancer 2 (2002) 489–501. J. LoPiccolo, G.M. Blumenthal, W.B. Bernstein, P.A. Dennis, Targeting the PI3K/Akt/ mTOR pathway: effective combinations and clinical considerations, Drug Resist. Updat. 11 (2008) 32–50. J.A. Engelman, L. Chen, X. Tan, K. Crosby, A.R. Guimaraes, R. Upadhyay, M. Maira, K. McNamara, S.A. Perera, Y. Song, L.R. Chirieac, R. Kaur, A. Lightbown, J. Simendinger, T. Li, R.F. Padera, C. Garcia-Echeverria, R. Weissleder, U. Mahmood, L.C. Cantley, K.K. Wong, Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers, Nat. Med. 14 (2008) 1351–1356. S. Horn, U. Bergholz, M. Jucker, J.A. McCubrey, L. Trumper, C. Stocking, J. Basecke, Mutations in the catalytic subunit of class IA PI3K confer leukemogenic potential to hematopoietic cells, Oncogene 27 (2008) 4096–4106. A. Denley, S. Kang, U. Karst, P.K. Vogt, Oncogenic signaling of class I PI3K isoforms, Oncogene 27 (2008) 2561–2574. C. Hafner, E. Lopez-Knowles, N.M. Luis, A. Toll, E. Baselga, A. Fernandez-Casado, S. Hernandez, A. Ribe, T. Mentzel, R. Stoehr, F. Hofstaedter, M. Landthaler, T. Vogt, R. M. Pujol, A. Hartmann, F.X. Real, Oncogenic PIK3CA mutations occur in epidermal nevi and seborrheic keratoses with a characteristic mutation pattern, Proc. Natl Acad. Sci. USA 104 (2007) 13450–13454. J.J. Zhao, Z. Liu, L. Wang, E. Shin, M.F. Loda, T.M. Roberts, The oncogenic properties of mutant p110alpha and p110beta phosphatidylinositol 3-kinases in human mammary epithelial cells, Proc. Natl Acad. Sci. USA 102 (2005) 18443–18448. S.J. Isakoff, J.A. Engelman, H.Y. Irie, J. Luo, S.M. Brachmann, R.V. Pearline, L.C. Cantley, J.S. Brugge, Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells, Cancer Res. 65 (2005) 10992–11000. Y.T. Wu, H.L. Tan, Q. Huang, C.N. Ong, H.M. Shen, Activation of the PI3K-AktmTOR signaling pathway promotes necrotic cell death via suppression of autophagy, Autophagy 5 (2009) 824–834. D. Liu, Y. Yang, Q. Liu, J. Wang, Inhibition of autophagy by 3-MA potentiates cisplatin-induced apoptosis in esophageal squamous cell carcinoma cells, Med. Oncol. (2009). J. Li, N. Hou, A. Faried, S. Tsutsumi, T. Takeuchi, H. Kuwano, Inhibition of autophagy by 3-MA enhances the effect of 5-FU-induced apoptosis in colon cancer cells, Ann. Surg. Oncol. 16 (2009) 761–771. E.F. Blommaart, U. Krause, J.P. Schellens, H. Vreeling-Sindelarova, A.J. Meijer, The phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002 inhibit autophagy in isolated rat hepatocytes, Eur. J. Biochem. 243 (1997) 240–246. M. Degtyarev, A. De Maziere, C. Orr, J. Lin, B.B. Lee, J.Y. Tien, W.W. Prior, S. van Dijk, H. Wu, D.C. Gray, D.P. Davis, H.M. Stern, L.J. Murray, K.P. Hoeflich, J. Klumperman, L.S. Friedman, K. Lin, Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents, J. Cell Biol. 183 (2008) 101–116. Z. Lu, R.Z. Luo, Y. Lu, X. Zhang, Q. Yu, S. Khare, S. Kondo, Y. Kondo, Y. Yu, G.B. Mills, W.S. Liao, R.C. Bast Jr., The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells, J. Clin. Invest. 118 (2008) 3917–3929.

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229 [60] D.J. Klionsky, A.J. Meijer, P. Codogno, Autophagy and p70S6 kinase, Autophagy 1 (2005) 59–60 discussion 60–51. [61] B.S. Balakumaran, J.T. Herbert, P.G. Febbo, MYC activity mitigates response to rapamycin in prostate cancer through 4EBP1-mediated inhibition of autophagy, Autophagy 6 (2010). [62] R. Crazzolara, K.F. Bradstock, L.J. Bendall, RAD001 (Everolimus) induces autophagy in acute lymphoblastic leukemia, Autophagy 5 (2009) 727–728. [63] M. Tanemura, A. Saga, K. Kawamoto, T. Machida, T. Deguchi, T. Nishida, Y. Sawa, Y. Doki, M. Mori, T. Ito, Rapamycin induces autophagy in islets: relevance in islet transplantation, Transplant. Proc. 41 (2009) 334–338. [64] S. Sarkar, B. Ravikumar, R.A. Floto, D.C. Rubinsztein, Rapamycin and mTORindependent autophagy inducers ameliorate toxicity of polyglutamine-expanded huntingtin and related proteinopathies, Cell Death Differ. 16 (2009) 46–56. [65] D.B. Shackelford, R.J. Shaw, The LKB1-AMPK pathway: metabolism and growth control in tumour suppression, Nat. Rev. Cancer 9 (2009) 563–575. [66] Z. Luo, A.K. Saha, X. Xiang, N.B. Ruderman, AMPK, the metabolic syndrome and cancer, Trends Pharmacol. Sci. 26 (2005) 69–76. [67] A.V. Budanov, M. Karin, p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling, Cell 134 (2008) 451–460. [68] R.J. Shaw, M. Kosmatka, N. Bardeesy, R.L. Hurley, L.A. Witters, R.A. DePinho, L.C. Cantley, The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress, Proc. Natl Acad. Sci. USA 101 (2004) 3329–3335. [69] A. Woods, S.R. Johnstone, K. Dickerson, F.C. Leiper, L.G. Fryer, D. Neumann, U. Schlattner, T. Wallimann, M. Carlson, D. Carling, LKB1 is the upstream kinase in the AMP-activated protein kinase cascade, Curr. Biol. 13 (2003) 2004–2008. [70] K.A. Anderson, T.J. Ribar, F. Lin, P.K. Noeldner, M.F. Green, M.J. Muehlbauer, L.A. Witters, B.E. Kemp, A.R. Means, Hypothalamic CaMKK2 contributes to the regulation of energy balance, Cell Metab. 7 (2008) 377–388. [71] P. Tamas, S.A. Hawley, R.G. Clarke, K.J. Mustard, K. Green, D.G. Hardie, D.A. Cantrell, Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes, J. Exp. Med. 203 (2006) 1665–1670. [72] N. Stahmann, A. Woods, D. Carling, R. Heller, Thrombin activates AMP-activated protein kinase in endothelial cells via a pathway involving Ca2+/calmodulindependent protein kinase kinase beta, Mol. Cell. Biol. 26 (2006) 5933–5945. [73] G. Herrero-Martin, M. Hoyer-Hansen, C. Garcia-Garcia, C. Fumarola, T. Farkas, A. Lopez-Rivas, M. Jaattela, TAK1 activates AMPK-dependent cytoprotective autophagy in TRAIL-treated epithelial cells, EMBO J. 28 (2009) 677–685. [74] O. Krejci, M. Wunderlich, H. Geiger, F.S. Chou, D. Schleimer, M. Jansen, P.R. Andreassen, J.C. Mulloy, p53 signaling in response to increased DNA damage sensitizes AML1-ETO cells to stress-induced death, Blood 111 (2008) 2190–2199. [75] S. Cuenin, A. Tinel, S. Janssens, J. Tschopp, p53-induced protein with a death domain (PIDD) isoforms differentially activate nuclear factor-kappaB and caspase-2 in response to genotoxic stress, Oncogene 27 (2008) 387–396. [76] T. Yamaguchi, Y. Miki, K. Yoshida, Protein kinase C delta activates IkappaB-kinase alpha to induce the p53 tumor suppressor in response to oxidative stress, Cell. Signal. 19 (2007) 2088–2097. [77] S. Jin, p53, Autophagy and tumor suppression, Autophagy 1 (2005) 171–173. [78] D. Crighton, S. Wilkinson, J. O'Prey, N. Syed, P. Smith, P.R. Harrison, M. Gasco, O. Garrone, T. Crook, K.M. Ryan, DRAM, a p53-induced modulator of autophagy, is critical for apoptosis, Cell 126 (2006) 121–134. [79] W.X. Zong, U. Moll, p53 in autophagy control, Cell Cycle 7 (2008) 2947. [80] E. Tasdemir, M. Chiara Maiuri, E. Morselli, A. Criollo, M. D'Amelio, M. DjavaheriMergny, F. Cecconi, N. Tavernarakis, G. Kroemer, A dual role of p53 in the control of autophagy, Autophagy 4 (2008) 810–814. [81] B. Levine, J. Abrams, p53: the Janus of autophagy? Nat. Cell Biol. 10 (2008) 637–639. [82] Z. Feng, H. Zhang, A.J. Levine, S. Jin, The coordinate regulation of the p53 and mTOR pathways in cells, Proc. Natl Acad. Sci. USA 102 (2005) 8204–8209. [83] M. D'Amelio, F. Cecconi, A novel player in the p53-mediated autophagy: Sestrin2, Cell Cycle 8 (2009) 1467. [84] M.C. Maiuri, S.A. Malik, E. Morselli, O. Kepp, A. Criollo, P.L. Mouchel, R. Carnuccio, G. Kroemer, Stimulation of autophagy by the p53 target gene Sestrin2, Cell Cycle 8 (2009) 1571–1576. [85] D. Crighton, S. Wilkinson, K.M. Ryan, DRAM links autophagy to p53 and programmed cell death, Autophagy 3 (2007) 72–74. [86] K.S. Yee, S. Wilkinson, J. James, K.M. Ryan, K.H. Vousden, PUMA- and Baxinduced autophagy contributes to apoptosis, Cell Death Differ. 16 (2009) 1135–1145. [87] E. Tasdemir, M.C. Maiuri, I. Orhon, O. Kepp, E. Morselli, A. Criollo, G. Kroemer, p53 represses autophagy in a cell cycle-dependent fashion, Cell Cycle 7 (2008) 3006–3011. [88] A.R. Young, M. Narita, M. Ferreira, K. Kirschner, M. Sadaie, J.F. Darot, S. Tavare, S. Arakawa, S. Shimizu, F.M. Watt, Autophagy mediates the mitotic senescence transition, Genes Dev. 23 (2009) 798–803. [89] E. Tasdemir, M.C. Maiuri, L. Galluzzi, I. Vitale, M. Djavaheri-Mergny, M. D'Amelio, A. Criollo, E. Morselli, C. Zhu, F. Harper, U. Nannmark, C. Samara, P. Pinton, J.M. Vicencio, R. Carnuccio, U.M. Moll, F. Madeo, P. Paterlini-Brechot, R. Rizzuto, G. Szabadkai, G. Pierron, K. Blomgren, N. Tavernarakis, P. Codogno, F. Cecconi, G. Kroemer, Regulation of autophagy by cytoplasmic p53, Nat. Cell Biol. 10 (2008) 676–687. [90] D.R. Green, J.E. Chipuk, p53 and metabolism: Inside the TIGAR, Cell 126 (2006) 30–32. [91] K.H. Vousden, K.M. Ryan, p53 and metabolism, Nat. Rev. Cancer 9 (2009) 691–700. [92] Y. Cao, D.J. Klionsky, Physiological functions of Atg6/Beclin 1: a unique autophagyrelated protein, Cell Res. 17 (2007) 839–849.

227

[93] X.H. Liang, S. Jackson, M. Seaman, K. Brown, B. Kempkes, H. Hibshoosh, B. Levine, Induction of autophagy and inhibition of tumorigenesis by beclin 1, Nature 402 (1999) 672–676. [94] V.M. Aita, X.H. Liang, V.V. Murty, D.L. Pincus, W. Yu, E. Cayanis, S. Kalachikov, T.C. Gilliam, B. Levine, Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21, Genomics 59 (1999) 59–65. [95] E. Itakura, N. Mizushima, Atg14 and UVRAG: mutually exclusive subunits of mammalian Beclin 1-PI3K complexes, Autophagy 5 (2009) 534–536. [96] E. Itakura, C. Kishi, K. Inoue, N. Mizushima, Beclin 1 forms two distinct phosphatidylinositol 3-kinase complexes with mammalian Atg14 and UVRAG, Mol. Biol. Cell 19 (2008) 5360–5372. [97] Y. Takahashi, D. Coppola, N. Matsushita, H.D. Cualing, M. Sun, Y. Sato, C. Liang, J.U. Jung, J.Q. Cheng, J.J. Mule, W.J. Pledger, H.G. Wang, Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis, Nat. Cell Biol. 9 (2007) 1142–1151. [98] S. Pattingre, A. Tassa, X. Qu, R. Garuti, X.H. Liang, N. Mizushima, M. Packer, M.D. Schneider, B. Levine, Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy, Cell 122 (2005) 927–939. [99] K. Matsunaga, T. Saitoh, K. Tabata, H. Omori, T. Satoh, N. Kurotori, I. Maejima, K. Shirahama-Noda, T. Ichimura, T. Isobe, S. Akira, T. Noda, T. Yoshimori, Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages, Nat. Cell Biol. 11 (2009) 385–396. [100] U. Akar, A. Chaves-Reyez, M. Barria, A. Tari, A. Sanguino, Y. Kondo, S. Kondo, B. Arun, G. Lopez-Berestein, B. Ozpolat, Silencing of Bcl-2 expression by small interfering RNA induces autophagic cell death in MCF-7 breast cancer cells, Autophagy 4 (2008) 669–679. [101] K. Saeki, A. Yuo, E. Okuma, Y. Yazaki, S.A. Susin, G. Kroemer, F. Takaku, Bcl-2 down-regulation causes autophagy in a caspase-independent manner in human leukemic HL60 cells, Cell Death Differ. 7 (2000) 1263–1269. [102] S. Erlich, L. Mizrachy, O. Segev, L. Lindenboim, O. Zmira, S. Adi-Harel, J.A. Hirsch, R. Stein, R. Pinkas-Kramarski, Differential interactions between Beclin 1 and Bcl-2 family members, Autophagy 3 (2007) 561–568. [103] M.C. Maiuri, A. Criollo, E. Tasdemir, J.M. Vicencio, N. Tajeddine, J.A. Hickman, O. Geneste, G. Kroemer, BH3-only proteins and BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin 1 and Bcl-2/Bcl-X(L), Autophagy 3 (2007) 374–376. [104] M.C. Maiuri, G. Le Toumelin, A. Criollo, J.C. Rain, F. Gautier, P. Juin, E. Tasdemir, G. Pierron, K. Troulinaki, N. Tavernarakis, J.A. Hickman, O. Geneste, G. Kroemer, Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1, EMBO J. 26 (2007) 2527–2539. [105] C.W. Younce, P.E. Kolattukudy, 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. 426 (2010) 43–53. [106] S.D. Spassieva, T.D. Mullen, D.M. Townsend, L.M. Obeid, Disruption of ceramide synthesis by CerS2 down-regulation leads to autophagy and the unfolded protein response, Biochem. J. 424 (2009) 273–283. [107] A. Criollo, L. Senovilla, H. Authier, M.C. Maiuri, E. Morselli, I. Vitale, O. Kepp, E. Tasdemir, L. Galluzzi, S. Shen, M. Tailler, N. Delahaye, A. Tesniere, D. De Stefano, A.B. Younes, F. Harper, G. Pierron, S. Lavandero, L. Zitvogel, A. Israel, V. Baud, G. Kroemer, The IKK complex contributes to the induction of autophagy, EMBO J. (2009). [108] J.M. Coates, J.M. Galante, R.J. Bold, Cancer therapy beyond apoptosis: autophagy and anoikis as mechanisms of cell death, J. Surg. Res. (2009). [109] Z. Yue, S. Jin, C. Yang, A.J. Levine, N. Heintz, Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor, Proc. Natl Acad. Sci. USA 100 (2003) 15077–15082. [110] X. Qu, J. Yu, G. Bhagat, N. Furuya, H. Hibshoosh, A. Troxel, J. Rosen, E.L. Eskelinen, N. Mizushima, Y. Ohsumi, G. Cattoretti, B. Levine, Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene, J. Clin. Invest. 112 (2003) 1809–1820. [111] G. Marino, N. Salvador-Montoliu, A. Fueyo, E. Knecht, N. Mizushima, C. LopezOtin, Tissue-specific autophagy alterations and increased tumorigenesis in mice deficient in Atg4C/autophagin-3, J. Biol. Chem. 282 (2007) 18573–18583. [112] J. Iqbal, C. Kucuk, R.J. Deleeuw, G. Srivastava, W. Tam, H. Geng, D. Klinkebiel, J.K. Christman, K. Patel, K. Cao, L. Shen, K. Dybkaer, I.F. Tsui, H. Ali, N. Shimizu, W.Y. Au, W.L. Lam, W.C. Chan, Genomic analyses reveal global functional alterations that promote tumor growth and novel tumor suppressor genes in natural killercell malignancies, Leukemia 23 (2009) 1139–1151. [113] D. Coppola, F. Khalil, S.A. Eschrich, D. Boulware, T. Yeatman, H.G. Wang, Downregulation of Bax-interacting factor-1 in colorectal adenocarcinoma, Cancer 113 (2008) 2665–2670. [114] C. Liang, P. Feng, B. Ku, I. Dotan, D. Canaani, B.H. Oh, J.U. Jung, Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG, Nat. Cell Biol. 8 (2006) 688–699. [115] M.S. Kim, E.G. Jeong, C.H. Ahn, S.S. Kim, S.H. Lee, N.J. Yoo, Frameshift mutation of UVRAG, an autophagy-related gene, in gastric carcinomas with microsatellite instability, Hum. Pathol. 39 (2008) 1059–1063. [116] K. Degenhardt, R. Mathew, B. Beaudoin, K. Bray, D. Anderson, G. Chen, C. Mukherjee, Y. Shi, C. Gelinas, Y. Fan, D.A. Nelson, S. Jin, E. White, Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis, Cancer Cell 10 (2006) 51–64. [117] J.J. Lum, D.E. Bauer, M. Kong, M.H. Harris, C. Li, T. Lindsten, C.B. Thompson, Growth factor regulation of autophagy and cell survival in the absence of apoptosis, Cell 120 (2005) 237–248. [118] D.C. Rubinsztein, J.E. Gestwicki, L.O. Murphy, D.J. Klionsky, Potential therapeutic applications of autophagy, Nat. Rev. Drug Discov. 6 (2007) 304–312.

228

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229

[119] J.S. Samaddar, V.T. Gaddy, J. Duplantier, S.P. Thandavan, M. Shah, M.J. Smith, D. Browning, J. Rawson, S.B. Smith, J.T. Barrett, P.V. Schoenlein, A role for macroautophagy in protection against 4-hydroxytamoxifen-induced cell death and the development of antiestrogen resistance, Mol. Cancer Ther. 7 (2008) 2977–2987. [120] P.V. Schoenlein, S. Periyasamy-Thandavan, J.S. Samaddar, W.H. Jackson, J.T. Barrett, Autophagy facilitates the progression of ERalpha-positive breast cancer cells to antiestrogen resistance, Autophagy 5 (2009) 400–403. [121] S. John, I. Nayvelt, H.C. Hsu, P. Yang, W. Liu, G.M. Das, T. Thomas, T.J. Thomas, Regulation of estrogenic effects by beclin 1 in breast cancer cells, Cancer Res. 68 (2008) 7855–7863. [122] M.A. Qadir, B. Kwok, W.H. Dragowska, K.H. To, D. Le, M.B. Bally, S.M. Gorski, Macroautophagy inhibition sensitizes tamoxifen-resistant breast cancer cells and enhances mitochondrial depolarization, Breast Cancer Res. Treat. 112 (2008) 389–403. [123] M. Milani, T. Rzymski, H.R. Mellor, L. Pike, A. Bottini, D. Generali, A.L. Harris, The role of ATF4 stabilization and autophagy in resistance of breast cancer cells treated with Bortezomib, Cancer Res. 69 (2009) 4415–4423. [124] R. Clarke, A.N. Shajahan, R.B. Riggins, Y. Cho, A. Crawford, J. Xuan, Y. Wang, A. Zwart, R. Nehra, M.C. Liu, Gene network signaling in hormone responsiveness modifies apoptosis and autophagy in breast cancer cells, J. Steroid Biochem. Mol. Biol. 114 (2009) 8–20. [125] J.S. Carew, E.C. Medina, J.A. Esquivel II, D. Mahalingam, R. Swords, K. Kelly, H. Zhang, P. Huang, A.C. Mita, M.M. Mita, F.J. Giles, S.T. Nawrocki, Autophagy inhibition enhances vorinostat-induced apoptosis via ubiquitinated protein accumulation, J. Cell. Mol. Med. (2009) [Epub ahead of print]. [126] I.V. Bijnsdorp, G.J. Peters, O.H. Temmink, M. Fukushima, F.A. Kruyt, 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 (2009). [127] K. Sato, K. Tsuchihara, S. Fujii, M. Sugiyama, T. Goya, Y. Atomi, T. Ueno, A. Ochiai, H. Esumi, Autophagy is activated in colorectal cancer cells and contributes to the tolerance to nutrient deprivation, Cancer Res. 67 (2007) 9677–9684. [128] C. Bauvy, P. Gane, S. Arico, P. Codogno, E. Ogier-Denis, Autophagy delays sulindac sulfide-induced apoptosis in the human intestinal colon cancer cell line HT-29, Exp. Cell Res. 268 (2001) 139–149. [129] J. Han, W. Hou, L.A. Goldstein, C. Lu, D.B. Stolz, X.M. Yin, H. Rabinowich, Involvement of protective autophagy in TRAIL resistance of apoptosis-defective tumor cells, J. Biol. Chem. 283 (2008) 19665–19677. [130] D.R. Fels, J. Ye, A.T. Segan, S.J. Kridel, M. Spiotto, M. Olson, A.C. Koong, C. Koumenis, Preferential cytotoxicity of bortezomib toward hypoxic tumor cells via overactivation of endoplasmic reticulum stress pathways, Cancer Res. 68 (2008) 9323–9330. [131] W. Zhuang, Z. Qin, Z. Liang, The role of autophagy in sensitizing malignant glioma cells to radiation therapy, Acta Biochim. Biophys. Sin. (Shanghai) 41 (2009) 341–351. [132] M. Tiwari, V.K. Bajpai, A.A. Sahasrabuddhe, A. Kumar, R.A. Sinha, S. Behari, M.M. Godbole, Inhibition of N-(4-hydroxyphenyl)retinamide-induced autophagy at a lower dose enhances cell death in malignant glioma cells, Carcinogenesis 29 (2008) 600–609. [133] T. Shingu, K. Fujiwara, O. Bogler, Y. Akiyama, K. Moritake, N. Shinojima, Y. Tamada, T. Yokoyama, S. Kondo, Inhibition of autophagy at a late stage enhances imatinib-induced cytotoxicity in human malignant glioma cells, Int. J. Cancer 124 (2009) 1060–1071. [134] T. Kanzawa, J. Bedwell, Y. Kondo, S. Kondo, I.M. Germano, Inhibition of DNA repair for sensitizing resistant glioma cells to temozolomide, J. Neurosurg. 99 (2003) 1047–1052. [135] M. Katayama, T. Kawaguchi, M.S. Berger, R.O. Pieper, DNA damaging agent-induced autophagy produces a cytoprotective adenosine triphosphate surge in malignant glioma cells, Cell Death Differ. 14 (2007) 548–558. [136] T. Kanzawa, I.M. Germano, T. Komata, H. Ito, Y. Kondo, S. Kondo, Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells, Cell Death Differ. 11 (2004) 448–457. [137] P. Boya, R.A. Gonzalez-Polo, N. Casares, J.L. Perfettini, P. Dessen, N. Larochette, D. Metivier, D. Meley, S. Souquere, T. Yoshimori, G. Pierron, P. Codogno, G. Kroemer, Inhibition of macroautophagy triggers apoptosis, Mol. Cell. Biol. 25 (2005) 1025–1040. [138] J. Thorburn, F. Moore, A. Rao, W.W. Barclay, L.R. Thomas, K.W. Grant, S.D. Cramer, A. Thorburn, Selective inactivation of a Fas-associated death domain protein (FADD)-dependent apoptosis and autophagy pathway in immortal epithelial cells, Mol. Biol. Cell 16 (2005) 1189–1199. [139] B.S. Balakumaran, A. Porrello, D.S. Hsu, W. Glover, A. Foye, J.Y. Leung, B.A. Sullivan, W.C. Hahn, M. Loda, P.G. Febbo, MYC activity mitigates response to rapamycin in prostate cancer through eukaryotic initiation factor 4E-binding protein 1-mediated inhibition of autophagy, Cancer Res. 69 (2009) 7803–7810. [140] M. Li, X. Jiang, D. Liu, Y. Na, G.F. Gao, Z. Xi, Autophagy protects LNCaP cells under androgen deprivation conditions, Autophagy 4 (2008) 54–60. [141] P. Salomoni, B. Calabretta, Targeted therapies and autophagy: new insights from chronic myeloid leukemia, Autophagy 5 (2009) 1050–1051. [142] Y. Mishima, Y. Terui, A. Taniyama, R. Kuniyoshi, T. Takizawa, S. Kimura, K. Ozawa, K. Hatake, Autophagy and autophagic cell death are next targets for elimination of the resistance to tyrosine kinase inhibitors, Cancer Sci. 99 (2008) 2200–2208. [143] J.S. Carew, S.T. Nawrocki, F.J. Giles, J.L. Cleveland, Targeting autophagy: a novel anticancer strategy with therapeutic implications for imatinib resistance, Biologics 2 (2008) 201–204.

[144] C.I. Lin, E.E. Whang, M.A. Abramson, X. Jiang, B.D. Price, D.B. Donner, F.D. Moore Jr., D.T. Ruan, Autophagy: a new target for advanced papillary thyroid cancer therapy, Surgery 146 (2009) 1208–1214. [145] J.Y. Hung, Y.L. Hsu, C.T. Li, Y.C. Ko, W.C. Ni, M.S. Huang, P.L. Kuo, 6-Shogaol, an Active Constituent of Dietary Ginger, Induces Autophagy by Inhibiting the AKT/mTOR Pathway in Human Non-Small Cell Lung Cancer A549 Cells, J. Agric. Food Chem. 57 (2009) 9809–9816. [146] W.J. Guo, S.S. Ye, N. Cao, J. Huang, J. Gao, Q.Y. Chen, ROS-mediated autophagy was involved in cancer cell death induced by novel copper(II) complex, Exp. Toxicol. Pathol. (2009) [Epub ahead of print]. [147] H. Kawakita, A. Tsuchida, K. Miyazawa, M. Naito, M. Shigoka, B. Kyo, M. Enomoto, T. Wada, K. Katsumata, K. Ohyashiki, M. Itoh, A. Tomoda, T. Aoki, Growth inhibitory effects of vitamin K2 on colon cancer cell lines via different types of cell death including autophagy and apoptosis, Int. J. Mol. Med. 23 (2009) 709–716. [148] Y. Cheng, F. Qiu, T. Ikejima, Molecular mechanisms of oridonin-induced apoptosis and autophagy in murine fibrosarcoma L929 cells, Autophagy 5 (2009) 430–431. [149] J. Yang, L.J. Wu, S. Tashino, S. Onodera, T. Ikejima, Reactive oxygen species and nitric oxide regulate mitochondria-dependent apoptosis and autophagy in evodiaminetreated human cervix carcinoma HeLa cells, Free Radic. Res. 42 (2008) 492–504. [150] K.W. Kim, M. Hwang, L. Moretti, J.J. Jaboin, Y.I. Cha, B. Lu, Autophagy upregulation by inhibitors of caspase-3 and mTOR enhances radiotherapy in a mouse model of lung cancer, Autophagy 4 (2008) 659–668. [151] W.L. Lai, N.S. Wong, The PERK/eIF2 alpha signaling pathway of Unfolded Protein Response is essential for N-(4-hydroxyphenyl)retinamide (4HPR)-induced cytotoxicity in cancer cells, Exp. Cell Res. 314 (2008) 1667–1682. [152] H.B. Li, X. Yi, J.M. Gao, X.X. Ying, H.Q. Guan, J.C. Li, Magnolol-induced H460 cells death via autophagy but not apoptosis, Arch. Pharm. Res. 30 (2007) 1566–1574. [153] Y. Chen, E. McMillan-Ward, J. Kong, S.J. Israels, S.B. Gibson, Mitochondrial electron-transport-chain inhibitors of complexes I and II induce autophagic cell death mediated by reactive oxygen species, J. Cell Sci. 120 (2007) 4155–4166. [154] C.H. Yan, Y.P. Yang, Z.H. Qin, Z.L. Gu, P. Reid, Z.Q. Liang, Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells, Acta Pharmacol. Sin. 28 (2007) 540–548. [155] Y. Chen, L. Yang, C. Feng, L.P. Wen, Nano neodymium oxide induces massive vacuolization and autophagic cell death in non-small cell lung cancer NCI-H460 cells, Biochem. Biophys. Res. Commun. 337 (2005) 52–60. [156] E. Ogier-Denis, C. Bauvy, J.J. Houri, P. Codogno, Evidence for a dual control of macroautophagic sequestration and intracellular trafficking of N-linked glycoproteins by the trimeric G(i3) protein in HT-29 cells, Biochem. Biophys. Res. Commun. 235 (1997) 166–170. [157] K.M. Rouschop, T. van den Beucken, L. Dubois, H. Niessen, J. Bussink, K. Savelkouls, T. Keulers, H. Mujcic, W. Landuyt, J.W. Voncken, P. Lambin, A.J. van der Kogel, M. Koritzinsky, B.G. Wouters, The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5, J. Clin. Invest. 120 (2010) 127–141. [158] K.M. Livesey, D. Tang, H.J. Zeh, M.T. Lotze, Autophagy inhibition in combination cancer treatment, Curr. Opin. Investig. Drugs 10 (2009) 1269–1279. [159] M. Shanmugam, S.K. McBrayer, J. Qian, K. Raikoff, M.J. Avram, S. Singhal, V. Gandhi, P.T. Schumacker, N.L. Krett, S.T. Rosen, Targeting glucose consumption and autophagy in myeloma with the novel nucleoside analogue 8-aminoadenosine, J. Biol. Chem. 284 (2009) 26816–26830. [160] R.H. Kim, R.J. Bold, H.J. Kung, ADI, autophagy and apoptosis: metabolic stress as a therapeutic option for prostate cancer, Autophagy 5 (2009) 567–568. [161] J. Pimkina, M.E. Murphy, ARF, autophagy and tumor suppression, Autophagy 5 (2009) 397–399. [162] R.H. Kim, J.M. Coates, T.L. Bowles, G.P. McNerney, J. Sutcliffe, J.U. Jung, R. Gandour-Edwards, F.Y. Chuang, R.J. Bold, H.J. Kung, Arginine deiminase as a novel therapy for prostate cancer induces autophagy and caspase-independent apoptosis, Cancer Res. 69 (2009) 700–708. [163] R.K. Amaravadi, Autophagy-induced tumor dormancy in ovarian cancer, J. Clin. Invest. 118 (2008) 3837–3840. [164] M. Watanabe, S. Adachi, H. Matsubara, T. Imai, Y. Yui, Y. Mizushima, Y. Hiraumi, K. Watanabe, Y. Kamitsuji, S.Y. Toyokuni, H. Hosoi, T. Sugimoto, J. Toguchida, T. Nakahata, Induction of autophagy in malignant rhabdoid tumor cells by the histone deacetylase inhibitor FK228 through AIF translocation, Int. J. Cancer 124 (2009) 55–67. [165] J.S. Carew, S.T. Nawrocki, J.L. Cleveland, Modulating autophagy for therapeutic benefit, Autophagy 3 (2007) 464–467. [166] R.K. Amaravadi, D. Yu, J.J. Lum, T. Bui, M.A. Christophorou, G.I. Evan, A. ThomasTikhonenko, C.B. Thompson, Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma, J. Clin. Invest. 117 (2007) 326–336. [167] H. Ko, Y.J. Kim, J.S. Park, J.H. Park, H.O. Yang, Autophagy inhibition enhances apoptosis induced by ginsenoside Rk1 in hepatocellular carcinoma cells, Biosci. Biotechnol. Biochem. 73 (2009) 2183–2189. [168] T. Kawakami, R. Inagi, H. Takano, S. Sato, J.R. Ingelfinger, T. Fujita, M. Nangaku, Endoplasmic reticulum stress induces autophagy in renal proximal tubular cells, Nephrol. Dial. Transplant. 24 (2009) 2665–2672. [169] H. Ito, S. Daido, T. Kanzawa, S. Kondo, Y. Kondo, Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells, Int. J. Oncol. 26 (2005) 1401–1410. [170] R. Rahim, J.S. Strobl, Hydroxychloroquine, chloroquine, and all-trans retinoic acid regulate growth, survival, and histone acetylation in breast cancer cells, Anticancer Drugs 20 (2009) 736–745. [171] G. Filomeni, E. Desideri, S. Cardaci, I. Graziani, S. Piccirillo, G. Rotilio, M.R. Ciriolo, Carcinoma cells activate AMP-activated protein kinase-dependent

S. Chen et al. / Biochimica et Biophysica Acta 1806 (2010) 220–229

[172]

[173]

[174]

[175]

[176]

autophagy as survival response to kaempferol-mediated energetic impairment, Autophagy 6 (2010). K.F. Hsu, C.L. Wu, S.C. Huang, C.M. Wu, J.R. Hsiao, Y.T. Yo, Y.H. Chen, A.L. Shiau, C. Y. Chou, Cathepsin L mediates resveratrol-induced autophagy and apoptotic cell death in cervical cancer cells, Autophagy 5 (2009) 451–460. M. Tafani, L. Schito, T. Anwar, M. Indelicato, P. Sale, M. Di Vito, E. Morgante, R. Beraldi, F. Makovec, O. Letari, G. Caselli, C. Spadafora, B. Pucci, M.A. Russo, Induction of autophagic cell death by a novel molecule is increased by hypoxia, Autophagy 4 (2008) 1042–1053. L.Y. Xue, S.M. Chiu, K. Azizuddin, S. Joseph, N.L. Oleinick, The death of human cancer cells following photodynamic therapy: apoptosis competence is necessary for Bcl-2 protection but not for induction of autophagy, Photochem. Photobiol. 83 (2007) 1016–1023. T. Proikas-Cezanne, S. Waddell, A. Gaugel, T. Frickey, A. Lupas, A. Nordheim, WIPI1alpha (WIPI49), a member of the novel 7-bladed WIPI protein family, is aberrantly expressed in human cancer and is linked to starvation-induced autophagy, Oncogene 23 (2004) 9314–9325. A. Petiot, E. Ogier-Denis, E.F. Blommaart, A.J. Meijer, P. Codogno, Distinct classes of phosphatidylinositol 3'-kinases are involved in signaling pathways that control macroautophagy in HT-29 cells, J. Biol. Chem. 275 (2000) 992–998.

229

[177] G. Griffiths, P. Quinn, G. Warren, Dissection of the Golgi complex. I. Monensin inhibits the transport of viral membrane proteins from medial to trans Golgi cisternae in baby hamster kidney cells infected with Semliki Forest virus, J. Cell Biol. 96 (1983) 835–850. [178] M. Marsh, J. Wellsteed, H. Kern, E. Harms, A. Helenius, Monensin inhibits Semliki Forest virus penetration into culture cells, Proc. Natl Acad. Sci. USA 79 (1982) 5297–5301. [179] M. No, E.J. Choi, I.A. Kim, Targeting HER2 signaling pathway for radiosensitization: Alternative strategy for therapeutic resistance, Cancer Biol. Ther. 8 (2009). [180] C.Y. Zou, K.D. Smith, Q.S. Zhu, J. Liu, I.E. McCutcheon, J.M. Slopis, F. Meric-Bernstam, Z. Peng, W.G. Bornmann, G.B. Mills, A.J. Lazar, R.E. Pollock, D. Lev, Dual targeting of AKT and mammalian target of rapamycin: A potential therapeutic approach for malignant peripheral nerve sheath tumor, Mol. Cancer Ther. 8 (2009) 1157–1168. [181] H. Roca, Z. Varsos, K.J. Pienta, CCL2 protects prostate cancer PC3 cells from autophagic death via phosphatidylinositol 3-kinase/AKT-dependent survivin up-regulation, J. Biol. Chem. 283 (2008) 25057–25073. [182] C. Xing, B. Zhu, H. Liu, H. Yao, L. Zhang, Class I phosphatidylinositol 3-kinase inhibitor LY294002 activates autophagy and induces apoptosis through p53 pathway in gastric cancer cell line SGC7901, Acta Biochim. Biophys. Sin. (Shanghai) 40 (2008) 194–201. [183] Q. Sun, W. Fan, Q. Zhong, Regulation of Beclin 1 in autophagy, Autophagy 5 (2009) 713–716.