Proteins substrates of caspases (Blank and Shiloh 2007; Cohen et al 1997)
1. Introduction
1.1. History, definition and classification
Life and death are essential parts of the natural cycle of all multicellular organisms. In metazoans, somatic cells divide normally during the process known as mitosis. Cell proliferation is tightly controlled, according to the organism needs. An increase in the number of cells takes place during growth and when one of these cells finishes its physiological function or detects DNA or cell damage, it undergoes a physiological process known as apoptosis that induces its own death. In humans about a hundred thousand cells are formed every second through mitosis, while a similar number is destroyed by apoptosis [1]. This dynamic balance between proliferation and cell death is known as homeostasis. If altered, different pathologic processes such as carcinogenesis can take place. Besides its role in embryonic development, homeostasis maintenance and aging, apoptosis is also a defence mechanism by which infected, injured or mutated cells as a result of irradiation or chemotherapeutic drugs are eliminated. This type of cell death involves the activation of an evolutionary conserved and tightly regulated intracellular machinery that requires energy consumption [2]. An important feature of apoptosis is that the cell is eliminated without triggering an immune response, avoiding thus tissue damage [3].
The term

Figure 1.
Classification of different types of cell death according to the NCCD (modified from Galluzzi et al. 2012)
It is important to mention that a single stimulus can trigger more than one mechanism of cell death simultaneously within a cell. However, only the most efficacious mechanism will be prevalent.
2. Apoptosis
2.1. Morphological and biochemical features
Morphological features that share both caspase-dependent and independent apoptotic pathways are: a) loss of plasma membrane symmetry and loss of cell-to-cell contact and cell round-up, b) chromatin condensation (picnosis) and fragmentation and nuclear breakdown, c) overall cell shrinkage and cytoskeleton alterations although the majority of the cell organelles remain intact, d) membrane blebbing and formation of the membrane-enclosed particles called apoptotic bodies that contain nuclear or cytoplasmic material and will be engulfed by phagocytes or neighbouring cells [6].
Because apoptotic cells are eaten so quickly, there are few dead cells left on tissue sections. This is the reason why apoptosis was neglected by pathologists for a long time, even though apoptosis is the main mechanism for discarding of harmful or unwanted cells in multicellular organisms.
Along with the morphological transformation described, there are several biochemical alterations that take place. For instance, activation of endonucleases, some of them dependent on Ca2+ and Mg2+ channels, that cleaves genomic DNA. In the apoptotic process, this event gives rise to internucleosomal DNA double-strand breaks with fragments of multiples of 180 bp, resulting in the typical pattern of

Figure 2.
Features of Apoptosis, Oncosis and Necrosis (taken from Hail et al., 2006)
The most distinguishable feature of apoptosis is the formation of apoptotic bodies. On the other hand, oncosis is characterized by cytoplasmic swelling, dilation of organelles and vacuolization and plasma membrane blebbing. The cell will finally die by cytolysis, which is a typical hallmark of necrosis [9]. Apoptotic cells can also lose their plasma membrane and eventually undergo a secondary necrosis. However, this phenomenon has only been observed
Apoptosis can be divided into three stages: initiation, integration/decision and execution/degradation [12]. The initiation phase mainly depends upon cell type and apoptotic stimulus. The integration/decision phase consists of the activation of proteases, nucleases and other effector molecules. The execution/degradation phase involves morphological and biochemical changes that are common to all apoptotic mechanisms, regardless of the stimulus that initiated the process. The classical apoptosis is defined as a type of programmed cell death characterized by the activation of zymogens known as caspases, which are
3. Caspases: Executioners of the apoptotic process
Caspases are the effector molecules of apoptosis in mammals. They were first discovered mediating programmed cell death during development in the nematode
DNA damage repair | |
mRNA processing | |
Cellular structure | |
Apoptosis and cell cycle | |
Cell cycle | |
Apoptosis regulation | |
Cytokine | |
Rho GTPases regulator | |
Nuclear shape maintenance | |
Regulation of chromatin condensation | |
DNA fragmentation | |
Huntington disease | |
Sterol regulatory element binding proteins | |
Transcription factor |
Table 1.
Caspase activity can be regulated both in a positive and in a negative manner. Negative caspase regulators are called Inhibitors of Apoptosis Proteins (IAPs) that bind to the catalytic site of caspases neutralizing its activity [19], or targeting them to degradation by ubiquinitation [20,21]. Some examples of IAPs are: XIAP, c-IAP1, c-IAP2, NAIP and survivin. On the other hand, Smac/DIABLO and Omi/HtrA2 act as positive regulators by inactivating IAPs [22].
4. Extrinsic and intrinsic apoptotic pathways
There are at least two main well characterized apoptotic routes: the death receptor or extrinsic pathway and the mitochondrial or intrinsic pathway. The extrinsic pathway plays a major role in tissue homeostasis and responds to external cues, coming especially from the immune system, whereas the intrinsic pathway is triggered as a response to various internal insults such as DNA damage, cytosolic calcium overload, starvation, oxidative stress, radiation, cytotoxic agents, etc., and involves mitochondrial destabilization [23,24].
The death receptor pathway is initiated by extracellular stimuli that are recognized by a subgroup of the tumour necrosis factor receptor (TNF-R) family named dead receptors (Fas/CD95/APO-1, TNFR1, TRAIL R1/DR4 and TRAIL R2/DR5). Upon binding of their ligands (FAS, TNFα and TRAIL) these receptors become activated and interact via their death domain with the protein motif
The intrinsic mitochondrial pathway is characterized by the action of B-cell lymphoma (Bcl-2) proteins. This family consists of proapoptotic and antiapoptotic proteins. The proapoptotic members promote mitochondrial outer membrane permeabilization (MOMP) and the antiapoptotic members counteract this action, so that the balance between these two groups of proteins determines the final outcome [27,28]. If the balance is in favour of the proapoptotic members, the outer mitochondrial membrane is permeabilized through pore formation and cytchrome c and other proteins such as Smac/DIABLO and Omi/HtrA2 are released to the cytosol. Then, cytochrome c binds to the adaptor protein Apaf-1 and dATP, forming the apoptosome, a catalytic complex that activates caspase-9 which in turn activates the executioner caspases.
The extrinsic and intrinsic pathways are interconnected through Bid. In some cases, when DISC formation is low, caspase-8 activation can induce MOMP through Bid cleavage, which translocates to the mitochondria and induces cytchrome c release, apoptosome formation and engagement of the caspase cascade [29].

Figure 3.
The extrinsic and intrinsic apoptotic pathways.
5. Death receptors
Death receptors belong to the TNF-R superfamily of receptors. They participate in proliferation, differentiation, immune response, gene expression, survival and cell death. In fact, TNF-R1 and Fas (CD95/APO-1) are involved in apoptotic processes [30]. Death receptors are stimulated by death ligands: TNF; Fas ligand (FasL) and
6. Role of mitochondria in cell death
MMP, a crucial event of the intrinsic mitochondrial apoptotic pathway, is considered a “point of no return” in the sequence of events leading to apoptosis. This phenomenon is associated with mitochondrial membrane potential loss (∆Ψm) that occurs as a result of assymetrical distribution of protons on both sides of the inner mitochondrial membrane. This irreversible process can take place before, during or after MOMP. The pore formation caused by Bcl-2 proteins induces MOMP, which leads to ∆Ψm dissipation, inhibition of ATP synthesis and ∆Ψm-dependent transport activities. Consequently, the respiratory change ceases, causing reactive oxygen species (ROS) generation and release of proteins confined within the inner mitochondrial space [35]. The contribution of the inner mitochondrial permeabilization, however, is controversial. MOMP can also result from the phenomenon called mitochondrial permeability transition that implies the opening of a non-selective pore in the inner mitochondrial membrane known as the mitochondrial permeability transition pore complex [12].
6.1. Mechanisms of MOMP
The interplay between antiapoptotic and proapoptotic proteins decides the final fate of the damaged cell. Antiapoptotic proteins can be found in the outer mitochondrial membrane, in the cytoplasm or in the Endoplasmic Reticulum (ER). Some propapoptotic members such as Bax or Bid reside in the cytosol, but translocate to the outer membrane upon triggering of a death stimulus, where they oligomerize to form a channel, either with themselves or with membrane anchored-Bak or tBid. The importance of the interactions between proteins and lipids is becoming more evident. In fact, it is now thought that Bak or Bax destabilize lipid bilayers instead of forming pores. BH3-only proteins activity is transcriptionally and posttranslationally regulated. Bid, for instance, is regulated by caspase-8, cathepsins, granzyme B or calpain [37-39]. The proapototic protein Bad in inhibited through phosphorylation by Akt and activated through dephosphorylation by calcineurin [40].

Figure 4.
Proposed model of mitochondria membrane permeabilization.

Figure 5.
Proposed model of mitochondrial membrane permeabilization.
The rise of mitochondrial matrix Ca2+ induces oxidative metabolism. However, upon apoptotic stimulus, Ca2+ can cause PTP opening, which can be transient and provide a fast calcium release mechanism, or persistent, giving rise to outer membrane rupture and release of apoptotic factors. Additionally, a different channel has been identified in the mitochondrion, composed of ceramide, a lipid that can form hydrogen bonds giving rise to ceramide structures that form channels which allow the efflux of proteins up to 60 kDa [42].
6.2. Cell death effectors released from mitochondria
The release of proteins from the intermembrane space and the loss of membrane-associated mitochondrial functions lead to cell death. Albeit is not clear the mechanism, mitochondrial destabilization provokes release of factors that mediate in caspase-dependent and independent pathways.
In addition, to the caspase-dependent effectors already mentioned, other caspase-independent proteins can be activated:
Besides its function as IAP inhibitor,
AIF and Endo G activity together with Omi/HtrA2 serine protease activity are considered responsible for the recently called
6.3. Afferent signals from other organelles
Mitochondria play a central role in programmed cell death pathways and integrate different signals coming from other organelles, being MMP, a point of no return, as already mentioned.
6.3.1. Nuclear DNA damage
The tumour suppressor p53 mediates DNA damage response, either by stimulating DNA damage response or by inducing apoptosis. As a transcription factor, p53 transactivates Bcl-2 proteins (Bad, Bid, Puma and Noxa) [52], which induce MMP and release of proteins from the intermembrane space [12]. After DNA damage, p53 can also induce the expression of
6.3.2. Endoplasmic reticulum: Unfolded protein response
The ER has primordial roles in normal physiological and survival processes. These include intracellular calcium homeostasis, protein secretion and lipid biosynthesis [58].Apoptosis can be initiated as a consequence of stress in the ER. This stress condition can be caused by calcium homeostasis alteration, glucose deprivation, hypoxia, low redox potential, excessive protein synthesis or defective protein secretion. These insults can cause accumulation of unfolded proteins in the lumen of the ER, triggering an evolutionary conserved signalling pathway known as the Unfolded Protein Response (UPR) which can culminate in cell death. UPR consists of global protein synthesis reduction, synthesis induction of chaperones and other proteins related to protein folding and retro-translocation of misfolded or unfolded proteins from the ER to the cytosol, where they will be degraded by the proteasome [59]. When the ER stress is sustained and ER function cannot be restored, UPR activates a specific apoptotic pathway. Caspase-12, which is localized in the ER, is activated by calcium-dependent proteases known as calpains [60].Once activated, caspase-12 activates caspase-9 without apoptosome intervention [61,62]. It has been postulated that ER stress can also activate caspase-8, which induces cytochrome c release through Bid processing [29,63]. In addition, ER stress can also initiate
6.3.3. Lysosomes
Lysosomes are organelles that contain acidic hydrolases such as cathepsins. Rupture of lysosomes release cathepsins to the cytosol, where they can trigger apoptosis or necrosis. Cystatins, on the other hand, are cytosolic proteins that act as negative regulators of cathepsins when they are translocated from lysosomes to the cytosol. Apoptosis initiated in lysosomes follows a mitochondrion-dependent pathway associated to caspase activation. However, it has been shown that cathepsin D activates Bax and AIF release, triggering a caspase-independent apoptotic pathway [37]. Furthermore, some cathepsins induce Bid cleavage [38], interaction with Bcl-2 proteins and permeabilization of the mitochondrial membrane [64].In addition, cathepsins can alter mitochondria functions by cleaving subunits of the oxidative phosphorylation complexes, inducing ROS generation [12].Lysosomes are essentially involved in
6.3.4. Cytosol
Several signals coming from the cytosol can induce MMP. These include: metabolites such as glucose 6-phospate and palmitate, ROS and activation of certain kinases: GSK3β, protein kinase C (PKC) δ, [65] and members of the JNK signalling pathway [38,66]. On the other hand, other molecules inactivate PTP and protect mitochondrial membrane from permeabilization, inhibiting apoptosis. These include: metabolites (ATP, glucose, NADH, UTP, etc.), antiapoptotic Bcl-2 family members, antioxidant enzymes such as glutathiones S transferase and prosurvival kinases such as Akt. In this sense, Akt can inhibit apoptosis by several mechanisms: activation of NFkB [67], inactivation of GSK3β and caspases and through hexokinase II-dependent mechanisms [12].
6.3.5. Cytoskeleton
The cytoskeleton is composed of microtubules, microfilaments and intermediate filaments that play important roles in cell motility, polarity, attachment, shape maintenance, etc. Adherent cells can undergo a specific type of caspase-dependent cell death called
7. Parp proteolysis as an indicator of cell death
Poly-ADP-Ribose Polymerase (PARP) is a family of 16 nuclear enzymes, among which, the best characterized is PARP-1. PARPs have several functions in cell proliferation, cell death, DNA recombination and DNA repair. PARP-1 is a 116 kDa nuclear protein involved in DNA repair mechanisms. PARP synthesis is activated when DNA is fragmented in the presence of nuclear poly-ADP ribosylated proteins. In an early apoptotic stage, caspases cleave PARP resulting in an 89 kDa and a 24 kDa fragments [73]. The smaller fragment irreversibly binds DNA fragment ends, impeding the access of DNA repair enzymes. Hence, PARP proteolysis facilitates nuclear disorganization and ensures irreversibility of the apoptotic process [74]. PARP cleaves also takes place during necrosis. However, the fragments obtained are of different size [75]. A role of PARP cleavage in autophagy induced by DNA damage has been recently suggested [76].
8. Autophagy
Macroautophagy (hereafter referred to as autophagy) is the most studied and prevalent form of autophagy in cells. This process begins with the formation of a “C” shaped double-membrane structure in the cytosol, called “omegasome”, which is formed from the ER (Initiation phase). Following this, the omegasome grows to form the “isolation membrane”, which elongates to engulf cytoplasmic components (Elongation phase). Then, the “isolation membrane” curves and closes to form a vacuole called the autophagosome (Maturation phase). As a result, portions of the cell cytoplasm and some organelles are sequestered in this vacuole. Finally, the outer membrane of the autophagosome fuses with the lysosomal membrane and the inner membrane (the autophagic body) carrying the cytosolic constituents enters the lysosome. The autophagic body is degraded in the lysosome by hydrolases and the resulting free amino acids and macromolecules are transported back into the cytosol for reuse [84]. In this way, autophagy contributes to the maintenance of the cellular energy homeostasis, to the clearance of damaged organelles and to adaptation to environmental stresses [85]. Accordingly, autophagy defects have been linked to a wide range of human pathologies, including cancer.

Figure 6.
9. Mitotic catastrophe
The cell death process that takes place when mitosis cannot be completed is called mitotic catastrophe. This phenomenon is triggered as a consequence of perturbations of the mitotic machinery that governs appropriate chromosome segregation. The main hallmark of mitotic catastrophe is the enlarged cell size accompanied by multinucleation [5].Other features are chromatin condensation, DNA degradation, MMP, cyrochrome c release from the mitochondria and caspase activation [86].Some types of mitotic catastrophe, however, take place without intervention of caspases, what has been named caspase-independent mitotic death [87].Mitotic catastrophe results form the combination of deficient checkpoints (DNA and mitotic spindle) and DNA damage. Cells that evade the mitotic checkpoint and do not undergo apoptosis are prone to generate aneuploidy. Therefore, mitotic catastrophe is conceived as a device to avoid genomic instability. The players that take part in mitotic catastrophe are: cell cycle-dependent kinases (Cdk1, Aurora, Plk), cell cycle-check points proteins (Chk2, p53, p73), survivin, MCl-2, Blc-2 proteins, caspase-2, etc. [86]. Mitotic catastrophe is a poorly defined molecular signalling pathway that precedes apoptosis, necrosis or senescence [88].

Figure 7.
10. Necroptosis
Necrosis is characterized by plasma membrane permeabilization, swelling and rupture. Necrosis can be accompanied in many instances by release of lysosomal hydrolases. Recently, a novel form of regulated necrosis has emerged and has been named

Figure 8.
TNFR1-mediated apoptosis and necroptosis (taken from Long and Ryan 2012).
11. Apoptosis, chemoresistance and cancer
So far, we have reviewed generalities of different modalities of cell death that can take place after various pathologies: inflammation, stroke, ischemic injury, neurodegenerative disorders, viral infection, neoplasia, etc. The implication of apoptosis in cancer was initially observed as the type of cell death occurring in untreated tumours and in tumour regression after radiotherapy [4]. The oncogenic process requires accumulation of diverse alterations within a cell that disrupt its normal homeostasis of cell death and growth. It is well established that excessive proliferation is not only due to oncogene activation but also to failure of the pathways controlling programmed cell death mechanisms [94]. A malignant cell can be protected from cell demise though expression and/or activation of antiapoptotic factors (acting as oncogenes) or through inactivation of antiapoptotic factors (acting as tumour suppressor genes). Evasion of apoptosis is a hallmark of cancer [95]. Dysregulation of apoptotic pathways renders cells resistant to antitumour strategies since the final outcome of radio and chemotherapy is frequently apoptosis of cancer cells. Therefore, resistance to cell death- in particular apoptotic cell death- is an important aspect of carcinogenesis, as it confers resistance to anticancer agents [96].In many tumours, chemoresistance acquisition is due to upregulation or modification of key elements of apoptosis control, such as Bcl-2, Bcl-XL and IAP family members [36]. Other mechanisms are characterized by inactivating mutations in proapoptotic proteins, such as p53.

Figure 9.
Transcription-dependent and independent effects of p53 activation
11.1. Defective checkpoints

Figure 10.
DNA damage and cell cycle checkpoints

Figure 11.
Akt signalling pathway
11.2. Immunogenecity of cancer cell death
It is becoming more evident that the immune response facilitates the effects of chemotherapy. Physiological death avoids autoimmunity. However cancer cell death triggered by radiotherapy or some chemotherapeutic agents such as anthracyclines can be immunogenic [128].Immunogenic death involves changes in the composition of the cell membrane and the release of molecules called Damage Associated Molecular Patterns or alarmins. In particular, calreticulin has been shown to be crucial for immunogenic cancer cell death [129].The immune system determines the long-term success of antitumor therapies. It seems that mitochondrial events as well as the ER response in conjunction with autophagy can establish whether cancer cells die in response to chemotherapy [130]. It has been found that calreticulin is the dominant pro-phagocytic signal on several cancers including neuroblastoma, non Hodgkin’s lymphoma and bladder cancer. However, calreticulin is counterbalanced by the “don´t eat me” signal CD47, which prevents cancer cell phagocytosis and is also highly expressed in these tumours [131]. Moreover, since CD47 is expressed on the surface of all human cancer cells but not in normal cells, blocking CD47 function with antibodies is emerging as a novel potential cancer strategy [132].
12. Senescence and cancer
We have already mentioned that strong p53 activation induces apoptosis. However, Leontieva and colleagues have shown that a weak and sustained p53 activation during cell cycle arrest can promote a different type of cell demise known as
13. Autophagy and cancer
Regarding the role played by autophagy in cancer, mounting evidences suggest that autophagic cell death functions as a tumour suppressor mechanism. Several tumour suppressor proteins have been shown to induce autophagy. Supporting this idea, there are also several works showing that certain oncogenic proteins inhibit autophagy. Most of this oncogene products form part of the classic PI3K-Akt-mTOR pathway. In the following section, we summarize the literature on the role played by key tumour suppressors and oncogene products in the regulation of autophagy and the involvement of autophagic genes in cancer.
13.1. Tumour suppressor genes that regulate autophagy
To date, several tumour suppressor proteins that regulate autophagy have been described. These include: Beclin1, UV irradiation-resistance-associated gene (UVRAG), PTEN, Bcl-2, and p53. The majority of them act as autophagy inducers with the exception of p53 that is able to activate or inhibit autophagy.
13.2. Oncogenic genes that regulate autophagy
Most of the oncogenic genes that regulate autophagy described to date are key proteins of the PI3K-Akt-mTOR pathway. These proteins inhibit autophagy and promote carcinogenesis.
13.3. How does autophagy suppress tumourigenesis?
The combined data presented above strongly support the idea that autophagy functions as a tumour suppressor process. Consistent with this, oncogenes that have a role in autophagy are potent inhibitors of this process. Although the molecular mechanisms by which autophagy functions in tumour suppression are poorly defined, at least two mechanisms have been described:
Autophagy maintains the integrity of the genome
The first hypothesis is that autophagy may function as a housekeeping pathway to exert the quality control of organelles, proteins and DNA. Mathews and colleague observed that in autophagy deficient tumour cells, metabolic stress promotes the accumulation of p62, damaged mitochondria and ROS generation, promoting genomic instability and leading to oncogene activation and tumour progression [157,158]. Furthermore, immortalized mouse epithelial cells with a defect in ATG genes (loss of Beclin 1 or Atg 5) display increased DNA damage, centrosome abnormalities, numerical and structural chromosomal abnormalities and gene amplification, especially after ischemic stress.
However, the mechanisms through which autophagy preserves the integrity of the genome remains elusive. One possibility could be that autophagy may contribute to cell cycle regulation, for example by degrading organelles and/or proteins involved in the cell cycle checkpoints [159]. Another possibility is that autophagy might simply function at a more general level to ensure the minimal amount of ATP and other metabolites required for DNA repair. Finally, autophagy may act by removing old and/or damaged organelles (for example, uncoupled mitochondria) which may act as a source of genotoxic chemical species such as ROS.
Autophagy limits necrosis-mediated inflammation
Necrosis normally results from physical injury in which the cell lyses and releases its intracellular contents, which activate the innate immune system and a wound-healing response [160]. As a result, inflammatory cells are recruited and cytokines are released to promote cell growth to replace the damaged tissue [161,162].
In contrast, apoptosis may be the preferred means of cell demise for cells upon metabolic stress, as cells are eliminated without inflammation. However, in cancer cells with a defect in apoptosis, autophagy is induced for cell survival. Through autophagy, cells eliminate damaged organelles and may maintain their normal cellular function under adverse conditions of fluctuating oxygen and nutrient supply. However, this beneficial effect of autophagy functions during short term interruptions in nutrient availability, as in the long term (excess of autophagy) it can possibly lead to cell death.
A high proportion of tumours have been seen to present a defect both in autophagy and apoptosis. Degenhardt and colleagues have shown that the inhibition of both processes under conditions of metabolic stress generates a necrotic cell-death, suggesting that apoptosis and autophagy function to limit necrosis [163]. In these necrotic tumours a persistent inflammatory infiltration and cytokine production exists, which is thought to promote tumour growth and thus, is associated with poor prognosis.
13.4. Role of autophagy in tumour survival
The data presented above strongly supports the idea that autophagy functions as a tumour suppressor process and that inhibition of autophagy leads to carcinogenesis. However, there are some circumstances where autophagy contributes to tumour survival promoting carcinogenesis:
Autophagy is induced by nutrient starvation
The survival role of autophagy during nutrient limitation is well established. When cells encounter environmental stresses such as nutrient starvation, autophagy can be activated and protects cells by preventing them from undergoing apoptosis. Through autophagy, starving cells degrade cytoplasmic material to generate both nutrients and energy [85]. Consistent with this, during nutrient starvation, inhibition of autophagy promotes apoptosis [164].
Autophagy is induced by hypoxia
Hypoxia in tumours results from inadequate tumour vasculature and is associated with a more malignant phenotype, higher predisposition for metastasis, and poor prognosis. Hypoxic stress selects for cells that are resistant to apoptosis as well as poses a major barrier to chemotherapy and radiotherapy.
White and colleagues first showed that autophagy is induced specifically in the hypoxic core of tumours, where it promotes survival [163]. Further studies have unveiled the molecular connections between hypoxia and the activation of autophagy. For example, it has been reported that when oxygen concentration falls below 5% hypoxia inducible factor 1 (HIF1) is activated and this transcription factor activates key autophagy inducers (BNIP3), which in turn activate the key autophagy complex formed by PI3K III [165,166]. Further mechanistic studies have revealed that induction of BNIP3 and BNIP3L in hypoxic cells disrupts the Becn1-Bcl-2 complex, thereby releasing Becn1 to induce autophagy [167].
The role of autophagy as a key mediator of survival of hypoxic cells is emerging so that the exact mechanisms underlying this phenotype remain unclear. Because chronic hypoxia leads to major metabolic perturbations in tumour tissues, one can postulate that by recycling basic cellular components, autophagy helps stressed cells cope with the increased metabolic demand [168]. However, further studies are needed to validate this hypothesis.
Autophagy is induced in metastatic cells
Epithelial cells critically depend on cell adhesion to extracellular matrix (ECM) for proper growth and survival. Upon detachment of cells from the ECM, cells undergo anoikis, a type of apoptotic cell death that serves the homeostatic function of killing cells that have lost contact with the basement membrane [68]. It has been shown that autophagy is induced in oncogene-transformed cells following matrix detachment [169]. Similarly, in three dimensional (3D) epithelial cell culture models, autophagy is significantly increased in the detached luminal cells. Furthermore, when autophagy is inhibited accelerated luminal clearance occurs [158,169]. Altogether, these results suggest that autophagy is fundamental in anoikis resistance, a process exploited by tumour cells to survive after detachment from the primary site, as well as while migrating to distant metastatic sites [170].
Debnath and colleagues have shown that detachment induces autophagy in both nontumourigenic epithelial cell lines and in primary epithelial cells. Autophagy inhibition through siRNA for ATG genes inhibits detachment-induced autophagy and increases apoptosis. Remarkably, even when apoptosis is inhibited matrix-detached cells still exhibit autophagy. Moreover, inhibition of autophagy in MCF-10 acini enhances luminal apoptosis during morphogenesis and fails to elicit long-term luminal filling [171]. Altogether, these results indicate that autophagy promotes epithelial cell survival during anoikis, including in detached cells harbouring antiapoptotic lesions.
14. Current therapeutic advances
In addition to inactivation of proliferative and prosurvival oncogenic pathways, current anticancer strategies deal with reactivation of cancer death cell signalling routes in order to induce tumour regression. The most promising cancer therapies that specifically target apoptosis, necrosis/necroptosis and autophagy are described in the following section.
14.1. Extrinsic apoptotic pathway
14.2. Intrinsic apoptotic pathway
Other IAP-targeted therapies such as XIAP and survivin antisense oligonucleotides have been developed [102]. Antisense oligonucleotides against survivin synergize with etoposide in non small lung cancer cells [117]. LY2181308 is a second generation antisense oligonucleotide being evaluated in clinical trials. In addition, a small molecule inhibitor of survivin, YM155, has shown potency in preclinical models and has entered phase II clinical trails [110].
14.3. DNA damage and cell cycle
14.4. Necroptosis
14.5. Autophagy
Autophagy as a protective mechanism
A large series of anticancer drugs (both clinically approved and experimental) are able to induce a significant accumulation of autophagosomes in tumour cells both
Autophagy as a death mechanism
Although there is robust evidence indicating that autophagy has a protective role in cancer therapeutics, in certain cancer treatments, autophagy can kill cells by inducing
14.6. What determines if autophagy is cytoprotective or cytotoxic?
Autophagy is a process that allows cells to escape death or paradoxically leads to cell death. It is not yet understood what factors determine whether autophagy is cytoprotective or cytotoxic. It has been suggested that autophagy induced under pathological conditions functions as an adaptive cell response, allowing the cell to survive bioenergetic stress. However, autophagy is a process that destroys cellular content and organelles. In this way, it has been suggested that deregulated, excessive or persistent autophagy may lead to autophagic cell death. That is, the destruction of proteins and organelles may pass a threshold, leading to cell death. However, the point at which autophagy becomes autophagic cell death remains unclear. In this perspective, the dissection of the transition from autophagy to autophagic cell death and the cross-talking between apoptosis and autophagy may help to understand this process, leading to more efficacious treatments in cancer. In contrast, a different study has reported that when cells are subjected to prolonged growth factor deprivation or shortage of glucose and oxygen they can lose the majority of their mass via autophagy. However, when these cells are placed in optimal culture conditions, they are able to fully recover [163,213].This result suggests that cell death via autophagy may not be simply a matter of crossing a quantitative threshold of self-digestion.
14.7. Interplay among apoptosis, necrosis/necroptosis and autophagy
Cell death process
15. Concluding remarks
Programmed cell death mechanisms are intricate and usually interconnected processes. Evasion of cell death is a common feature of cancer cells leading to chemoresistance. Apoptosis, necrosis/necroptosis and autophagy are the main explored pathways that had gained interest among cancer biologists, as means to develop novel cancer therapeutics. Deeping our knowledge on the nexus between cell death and cancer will enable us to predict in a more refined manner the carcinogenic process and therefore, pave the way for a personalized approach to the disease.
References
- 1.
Vaux DL, Korsmeyer SJ 1999 Cell death in development. Cell ;96 245 254 - 2.
Vermeulen K. Van Bockstaele D. R. Berneman Z. N. 2005 Apoptosis: mechanisms and relevance in cancer. Ann.Hematol.84 627 639 - 3.
deCathelineau AM, Henson PM 2003 The final step in programmed cell death: phagocytes carry apoptotic cells to the grave. Essays Biochem.39 105 117 - 4.
Kerr JF, Wyllie AH, Currie AR 1972 Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br.J.Cancer ;26 239 257 - 5.
Kroemer G. Galluzzi L. Vandenabeele P. Abrams J. Alnemri E. S. Baehrecke E. H. et al. 2009 Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death.Differ.16 3 11 - 6.
Wyllie AH, Kerr JF, Currie AR 1980 Cell death: the significance of apoptosis. Int.Rev.Cytol.68 251 306 - 7.
Zamzami N. Marchetti P. Castedo M. Zanin C. Vayssiere J. L. Petit P. X. et al. 1995 Reduction in mitochondrial potential constitutes an early irreversible step of programmed lymphocyte death in vivo. J.Exp.Med.181 1661 1672 - 8.
Martin SJ, Reutelingsperger CP, McGahon AJ, Rader JA, van Schie RC, LaFace DM, et al. 1995 Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of Bcl-2 and Abl. J.Exp.Med.182 1545 1556 - 9.
Majno G. Joris I. 1995 Apoptosis, oncosis, and necrosis. An overview of cell death. Am.J.Pathol.146 3 15 - 10.
Hail N. Jr Carter B. Z. Konopleva M. Andreeff M. 2006 Apoptosis effector mechanisms: a requiem performed in different keys. Apoptosis.11 889 904 - 11.
Galluzzi L. Vitale I. Abrams J. M. Alnemri E. S. Baehrecke E. H. Blagosklonny M. V. et al. 2012 Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death.Differ.19 107 120 - 12.
Kroemer G. Galluzzi L. Brenner C. 2007 Mitochondrial membrane permeabilization in cell death. Physiol Rev.87 99 163 - 13.
Ellis HM, Horvitz HR 1986 Genetic control of programmed cell death in the nematode C. elegans. Cell ;44 817 829 - 14.
Thornberry N. A. Lazebnik Y. 1998 Caspases: enemies within. Science ;281 1312 1316 - 15.
Pop C. Salvesen G. S. 2009 Human caspases: activation, specificity, and regulation. J.Biol.Chem.284 21777 21781 - 16.
Kumar S. 2007 Caspase function in programmed cell death. Cell Death.Differ.14 32 43 - 17.
Blank M. Shiloh Y. 2007 Programs for cell death: apoptosis is only one way to go. Cell Cycle ;6 686 695 - 18.
Cohen GM 1 EOF 16 EOF - 19.
Tenev T. Zachariou A. Wilson R. Ditzel M. Meier P. 2005 IAPs are functionally non-equivalent and regulate effector caspases through distinct mechanisms. Nat.Cell Biol.7 70 77 - 20.
Huang H. CA Joazeiro Bonfoco. E. Kamada S. JD Leverson Hunter. T. 2000 The inhibitor of apoptosis, cIAP2, functions as a ubiquitin-protein ligase and promotes in vitro monoubiquitination of caspases 3 and 7. J.Biol.Chem.275 26661 26664 - 21.
Suzuki Y. Nakabayashi Y. Takahashi R. 2001 Ubiquitin-protein ligase activity of X-linked inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3 and enhances its anti-apoptotic effect in Fas-induced cell death. Proc.Natl.Acad.Sci.U.S.A ;98 8662 8667 - 22.
Yang Q. H. Church-Hajduk R. Ren J. Newton M. L. Du C. 2003 Omi/HtrA2 catalytic cleavage of inhibitor of apoptosis (IAP) irreversibly inactivates IAPs and facilitates caspase activity in apoptosis. Genes Dev.17 1487 1496 - 23.
Danial NN, Korsmeyer SJ 2004 Cell death: critical control points. Cell ;116 205 219 - 24.
Meier P. Vousden K. H. 2007 Lucifer’s labyrinth--ten years of path finding in cell death. Mol.Cell ;28 746 754 - 25.
ME Peter Budd. R. C. Desbarats J. Hedrick S. M. Hueber A. O. Newell M. K. et al. 2007 The CD95 receptor: apoptosis revisited. Cell ;129 447 450 - 26.
Lavrik I. Golks A. Krammer P. H. 2005 Death receptor signaling. J.Cell Sci.118 265 267 - 27.
Green D. R. Kroemer G. 2004 The pathophysiology of mitochondrial cell death. Science ;305 626 629 - 28.
Tait SW, Green DR 2010 Mitochondria and cell death: outer membrane permeabilization and beyond. Nat.Rev.Mol.Cell Biol.11 621 632 - 29.
Luo X. Budihardjo I. Zou H. Slaughter C. Wang X. 1998 Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell ;94 481 490 - 30.
Chinnaiyan A. M. Tepper C. G. Seldin M. F. O’Rourke K. Kischkel F. C. Hellbardt S. et al. 1996 FADD/MORT1 is a common mediator of CD95 (Fas/APO-1) and tumor necrosis factor receptor-induced apoptosis. J.Biol.Chem.271 4961 4965 - 31.
Carswell E. A. Old L. J. Kassel R. L. Green S. Fiore N. Williamson B. 1975 An endotoxin-induced serum factor that causes necrosis of tumors. Proc.Natl.Acad.Sci.U.S.A ;72 3666 3670 - 32.
Schievella AR, Chen JH, Graham JR, Lin LL 1997 MADD, a novel death domain protein that interacts with the type 1 tumor necrosis factor receptor and activates mitogen-activated protein kinase. J.Biol.Chem.272 12069 12075 - 33.
Yang J. Liu X. Bhalla K. Kim C. N. Ibrado A. M. Cai J. et al. 1997 Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science ;275 1129 1132 - 34.
Vandenabeele P. Galluzzi L. Vanden Berghe. T. Kroemer G. 2010 Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat.Rev.Mol.Cell Biol.11 700 714 - 35.
Galluzzi L. Zamzami N. de La Motte R. T. Lemaire C. Brenner C. Kroemer G. 2007 Methods for the assessment of mitochondrial membrane permeabilization in apoptosis. Apoptosis.12 803 813 - 36.
Adams J. M. Cory S. 2007 The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene ;26 1324 1337 - 37.
Bidere N. Lorenzo H. K. Carmona S. Laforge M. Harper F. Dumont C. et al. 2003 Cathepsin D triggers Bax activation, resulting in selective apoptosis-inducing factor (AIF) relocation in T lymphocytes entering the early commitment phase to apoptosis. J.Biol.Chem.278 31401 31411 - 38.
Cirman T. Oresic K. Mazovec G. D. Turk V. Reed J. C. Myers R. M. et al. 2004 Selective disruption of lysosomes in HeLa cells triggers apoptosis mediated by cleavage of Bid by multiple papain-like lysosomal cathepsins. J.Biol.Chem.279 3578 3587 - 39.
Thomas D. A. Scorrano L. Putcha G. V. Korsmeyer S. J. Ley T. J. 2001 Granzyme B can cause mitochondrial depolarization and cell death in the absence of BID, BAX, and BAK. Proc.Natl.Acad.Sci.U.S.A ;98 14985 14990 - 40.
Datta S. R. Dudek H. Tao X. Masters S. Fu H. Gotoh Y. et al. 1997 Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell ;91 231 241 - 41.
Donovan M. Cotter T. G. 2004 Control of mitochondrial integrity by Bcl-2 family members and caspase-independent cell death. Biochim.Biophys.Acta ;1644 133 147 - 42.
Belizario J. E. Alves J. Occhiucci J. M. Garay-Malpartida M. Sesso A. 2007 A mechanistic view of mitochondrial death decision pores. Braz.J.Med.Biol.Res.40 1011 1024 - 43.
Szabadkai G. Rizzuto R. 2004 Participation of endoplasmic reticulum and mitochondrial calcium handling in apoptosis: more than just neighborhood? FEBS Lett.567 111 115 - 44.
MJ Thomenius Wang. N. S. Reineks E. Z. Wang Z. Distelhorst C. W. 2003 Bcl-2 on the endoplasmic reticulum regulates Bax activity by binding to BH3-only proteins. J.Biol.Chem.278 6243 6250 - 45.
Liu X. Kim C. N. Yang J. Jemmerson R. Wang X. 1996 Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell ;86 147 157 - 46.
Wang D. Liang J. Zhang Y. Gui B. Wang F. Yi X. et al. 2012 Steroid Receptor Coactivator-interacting Protein (SIP) Inhibits Caspase-independent Apoptosis by Preventing Apoptosis-inducing Factor (AIF) from Being Released from Mitochondria. J.Biol.Chem.287 12612 12621 - 47.
Norberg E. Gogvadze V. Ott M. Horn M. Uhlen P. Orrenius S. et al. 2008 An increase in intracellular Ca2+ is required for the activation of mitochondrial calpain to release AIF during cell death. Cell Death.Differ.15 1857 1864 - 48.
Yu S. W. Wang H. Poitras M. F. Coombs C. Bowers W. J. Federoff H. J. et al. 2002 Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science ;297 259 263 - 49.
Trencia A. Fiory F. MA Maitan Vito. P. Barbagallo A. P. Perfetti A. et al. 2004 Omi/HtrA2 promotes cell death by binding and degrading the anti-apoptotic protein ped/pea-15. J.Biol.Chem.279 46566 46572 - 50.
Cilenti L. MM Soundarapandian Kyriazis. G. A. Stratico V. Singh S. Gupta S. et al. 2004 Regulation of HAX-1 anti-apoptotic protein by Omi/HtrA2 protease during cell death. J.Biol.Chem.279 50295 50301 - 51.
Vande W. L. Wirawan E. Lamkanfi M. Festjens N. Verspurten J. Saelens X. et al. 2010 The mitochondrial serine protease HtrA2/Omi cleaves RIP1 during apoptosis of Ba/F3 cells induced by growth factor withdrawal. Cell Res.20 421 433 - 52.
Vousden K. H. Lu X. 2002 Live or let die: the cell’s response to53 Nat.Rev.Cancer ;2: 594-604. - 53.
Berube C. Boucher L. M. Ma Wakeham W. Salmena A. Hakem L. et R. al 2005 Apoptosis caused by53 protein with death domain (PIDD) depends on the death adapter protein RAIDD. Proc.Natl.Acad.Sci.U.S.A ;102: 14314-14320. - 54.
Janssens S. Tinel A. 2012 The PIDDosome, DNA-damage-induced apoptosis and beyond. Cell Death.Differ.19 13 20 - 55.
Bouchier-Hayes L. Lartigue L. Newmeyer D. D. 2005 Mitochondria: pharmacological manipulation of cell death. J.Clin.Invest ;115 2640 2647 - 56.
Yu Q. 2006 Restoring53 apoptosis in cancer cells: new opportunities for cancer therapy. Drug Resist.Updat.9: 19-25. - 57.
Watcharasit P. Bijur G. N. Song L. Zhu J. Chen X. Jope R. S. 2003 Glycogen synthase kinase-3beta (GSK3beta) binds to and promotes the actions of53 J.Biol.Chem.278: 48872-48879. - 58.
Kim I. Xu W. Reed J. C. 2008 Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities. Nat.Rev.Drug Discov.7 1013 1030 - 59.
Hoyer-Hansen M. Jaattela M. 2007 Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium. Cell Death.Differ.14 1576 1582 - 60.
Nakagawa T. Yuan J. 2000 Cross-talk between two cysteine protease families. Activation of caspase-12 by calpain in apoptosis. J.Cell Biol.150 887 894 - 61.
Morishima N. Nakanishi K. Takenouchi H. Shibata T. Yasuhiko Y. 2002 An endoplasmic reticulum stress-specific caspase cascade in apoptosis. Cytochrome c-independent activation of caspase-9 by caspase-12. J.Biol.Chem.277 34287 34294 - 62.
Rao R. V. Castro-Obregon S. Frankowski H. Schuler M. Stoka V. del et R. G. al 2002 Coupling endoplasmic reticulum stress to the cell death program. An Apaf-1-independent intrinsic pathway. J.Biol.Chem.277 21836 21842 - 63.
Jimbo A. Fujita E. Kouroku Y. Ohnishi J. Inohara N. Kuida K. et al. 2003 ER stress induces caspase-8 activation, stimulating cytochrome c release and caspase-9 activation. Exp.Cell Res.283 156 166 - 64.
Fehrenbacher N. Jaattela M. 2005 Lysosomes as targets for cancer therapy. Cancer Res.65 2993 2995 - 65.
Deacon E. M. Pongracz J. Griffiths G. Lord J. M. 1997 Isoenzymes of protein kinase C: differential involvement in apoptosis and pathogenesis. Mol.Pathol.50 124 131 - 66.
Kharbanda S. Saxena S. Yoshida K. Pandey P. Kaneki M. Wang Q. et al. 2000 Translocation of SAPK/JNK to mitochondria and interaction with Bcl-x(L) in response to DNA damage. J.Biol.Chem.275 322 327 - 67.
Ozes ON, Mayo LD, Gustin JA, Pfeffer SR, Pfeffer LM, Donner DB 1999 NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature ;401 82 85 - 68.
Frisch S. M. Francis H. 1994 Disruption of epithelial cell-matrix interactions induces apoptosis. J.Cell Biol.124 619 626 - 69.
Gourlay CW, Ayscough KR 2005 The actin cytoskeleton: a key regulator of apoptosis and ageing? Nat.Rev.Mol.Cell Biol.6 583 589 - 70.
Day C. L. Puthalakath H. Skea G. Strasser A. Barsukov I. Lian L. Y. et al. 2004 Localization of dynein light chains 1 and 2 and their pro-apoptotic ligands. Biochem.J.377 597 605 - 71.
Kusano H. Shimizu S. Koya R. C. Fujita H. Kamada S. Kuzumaki N. et al. 2000 Human gelsolin prevents apoptosis by inhibiting apoptotic mitochondrial changes via closing VDAC. Oncogene ;19 4807 4814 - 72.
Tang HL, Le AH, Lung HL 2006 The increase in mitochondrial association with actin precedes Bax translocation in apoptosis. Biochem.J.396 1 5 - 73.
Kaufmann S. H. Desnoyers S. Ottaviano Y. Davidson N. E. Poirier G. G. 1993 Specific proteolytic cleavage of poly(ADP-ribose) polymerase: an early marker of chemotherapy-induced apoptosis. Cancer Res.53 3976 3985 - 74.
Oliver FJ, Menissier-de MJ, de MG 1999 Poly(ADP-ribose) polymerase in the cellular response to DNA damage, apoptosis, and disease. Am.J.Hum.Genet.64 1282 1288 - 75.
Guillouf C. Wang T. S. Liu J. CE Walsh Poirier. G. G. Moustacchi E. et al. 1999 Fanconi anemia C protein acts at a switch between apoptosis and necrosis in mitomycin C-induced cell death. Exp.Cell Res.246 384 394 - 76.
Munoz-Gamez J. A. Rodriguez-Vargas J. M. Quiles-Perez R. Aguilar-Quesada R. Martin-Oliva D. de et M. G. al 2009 PARP-1 is involved in autophagy induced by DNA damage. Autophagy.5 61 74 - 77.
Wang Y. Dawson V. L. Dawson T. M. 2009 Poly(ADP-ribose) signals to mitochondrial AIF: a key event in parthanatos. Exp.Neurol.218 193 202 - 78.
Long JS, Ryan KM 2012 New frontiers in promoting tumour cell death: targeting apoptosis, necroptosis and autophagy. Oncogene. - 79.
Klionsky DJ 2007 Autophagy: from phenomenology to molecular understanding in less than a decade. Nat.Rev.Mol.Cell Biol.8 931 937 - 80.
Nakatogawa H. Suzuki K. Kamada Y. Ohsumi Y. 2009 Dynamics and diversity in autophagy mechanisms: lessons from yeast. Nat.Rev.Mol.Cell Biol.10 458 467 - 81.
Tanida I. Ueno T. Kominami E. 2004 LC3 conjugation system in mammalian autophagy. Int.J.Biochem.Cell Biol.36 2503 2518 - 82.
Mijaljica D. Prescott M. Devenish R. J. 2011 Microautophagy in mammalian cells: revisiting a 40-year-old conundrum. Autophagy.7 673 682 - 83.
Majeski AE, Dice JF 2004 Mechanisms of chaperone-mediated autophagy. Int.J.Biochem.Cell Biol.36 2435 2444 - 84.
Tanida I. 2011 Autophagosome formation and molecular mechanism of autophagy. Antioxid.Redox.Signal.14 2201 2214 - 85.
Yen WL, Klionsky DJ 2008 How to live long and prosper: autophagy, mitochondria, and aging. Physiology. (Bethesda.) ;23 248 262 - 86.
Castedo M. Perfettini J. L. Roumier T. Andreau K. Medema R. Kroemer G. 2004 Cell death by mitotic catastrophe: a molecular definition. Oncogene ;23 2825 2837 - 87.
Niikura Y. Dixit A. Scott R. Perkins G. Kitagawa K. 2007 BUB1 mediation of caspase-independent mitotic death determines cell fate. J.Cell Biol.178 283 296 - 88.
Vitale I. Galluzzi L. Castedo M. Kroemer G. 2011 Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat.Rev.Mol.Cell Biol.12 385 392 - 89.
Vanlangenakker N. Vanden Berghe. T. Vandenabeele P. 2012 Many stimuli pull the necrotic trigger, an overview. Cell Death.Differ.19 75 86 - 90.
Feoktistova M. Geserick P. Kellert B. Dimitrova D. P. Langlais C. Hupe M. et al. 2011 cIAPs block Ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol.Cell ;43 449 463 - 91.
Tenev T. Bianchi K. Darding M. Broemer M. Langlais C. Wallberg F. et al. 2011 The Ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Mol.Cell ;43 432 448 - 92.
Cho Y. S. Challa S. Moquin D. Genga R. Ray T. D. Guildford M. et al. 2009 Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell ;137 1112 1123 - 93.
Dunai Z. Bauer P. I. Mihalik R. 2011 Necroptosis: biochemical, physiological and pathological aspects. Pathol.Oncol.Res.17 791 800 - 94.
Martinez-Lacaci I. Garcia M. P. Soto J. L. Saceda M. 2007 Tumour cells resistance in cancer therapy. Clin.Transl.Oncol.9 13 20 - 95.
Hanahan D. Weinberg R. A. 2000 The hallmarks of cancer. Cell ;100 57 70 - 96.
Hersey P. Zhang X. D. 2003 Overcoming resistance of cancer cells to apoptosis. J.Cell Physiol ;196 9 18 - 97.
Hockenbery D. Nunez G. Milliman C. Schreiber R. D. Korsmeyer S. J. 1990 Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature ;348 334 336 - 98.
Tsujimoto Y. Finger L. R. Yunis J. Nowell P. C. Croce C. M. 1984 Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science ;226 1097 1099 - 99.
Vaux D. L. Cory S. Adams J. M. 1988 Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature ;335 440 442 - 100.
Strasser A. Harris A. W. Bath M. L. Cory S. 1990 Novel primitive lymphoid tumours induced in transgenic mice by cooperation between myc and bcl-2. Nature ;348 331 333 - 101.
Strasser A. Cory S. Adams J. M. 2011 Deciphering the rules of programmed cell death to improve therapy of cancer and other diseases. EMBO J.30 3667 3683 - 102.
Plati J. Bucur O. Khosravi-Far R. 2011 Apoptotic cell signaling in cancer progression and therapy. Integr.Biol. (Camb.) ;3 279 296 - 103.
Rampino N. Yamamoto H. Ionov Y. Li Y. Sawai H. Reed J. C. et al. 1997 Somatic frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype. Science ;275 967 969 - 104.
Tagawa H. Karnan S. Suzuki R. Matsuo K. Zhang X. Ota A. et al. 2005 Genome-wide array-based CGH for mantle cell lymphoma: identification of homozygous deletions of the proapoptotic gene BIM. Oncogene ;24 1348 1358 - 105.
Beroukhim R. Mermel C. H. Porter D. Wei G. Raychaudhuri S. Donovan J. et al. 2010 The landscape of somatic copy-number alteration across human cancers. Nature ;463 899 905 - 106.
Garrison S. P. Jeffers J. R. Yang C. Nilsson J. A. MA Hall Rehg. J. E. et al. 2008 Selection against PUMA gene expression in Myc-driven B-cell lymphomagenesis. Mol.Cell Biol.28 5391 5402 - 107.
Richter-Larrea J. A. Robles E. F. Fresquet V. Beltran E. Rullan A. J. Agirre X. et al. 2010 Reversion of epigenetically mediated BIM silencing overcomes chemoresistance in Burkitt lymphoma. Blood ;116 2531 2542 - 108.
Kashkar H. 2010 X-linked inhibitor of apoptosis: a chemoresistance factor or a hollow promise. Clin.Cancer Res.16 4496 4502 - 109.
Kaufmann T. Strasser A. Jost P. J. 2012 Fas death receptor signalling: roles of Bid and XIAP. Cell Death.Differ.19 42 50 - 110.
Mita AC, Mita MM, Nawrocki ST, Giles FJ 2008 Survivin: key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin.Cancer Res.14 5000 5005 - 111.
Reichert S. Rodel C. Mirsch J. Harter P. N. Tomicic M. T. Mittelbronn M. et al. 2011 Survivin inhibition and DNA double-strand break repair: a molecular mechanism to overcome radioresistance in glioblastoma. Radiother.Oncol.101 51 58 - 112.
Islam A. Kageyama H. Takada N. Kawamoto T. Takayasu H. Isogai E. et al. 2000 High expression of Survivin, mapped to 17q25, is significantly associated with poor prognostic factors and promotes cell survival in human neuroblastoma. Oncogene ;19 617 623 - 113.
Hattori M. Sakamoto H. Satoh K. Yamamoto T. 2001 DNA demethylase is expressed in ovarian cancers and the expression correlates with demethylation of CpG sites in the promoter region of c-erbB-2 and survivin genes. Cancer Lett.169 155 164 - 114.
Li F. Altieri D. C. 1999 The cancer antiapoptosis mouse survivin gene: characterization of locus and transcriptional requirements of basal and cell cycle-dependent expression. Cancer Res.59 3143 3151 - 115.
Mesri M. Morales-Ruiz M. Ackermann E. J. Bennett C. F. Pober J. S. Sessa W. C. et al. 2001 Suppression of vascular endothelial growth factor-mediated endothelial cell protection by survivin targeting. Am.J.Pathol.158 1757 1765 - 116.
Zaffaroni N. Daidone M. G. 2002 Survivin expression and resistance to anticancer treatments: perspectives for new therapeutic interventions. Drug Resist.Updat.5 65 72 - 117.
Rufini A. Melino G. 2011 Cell death pathology: the war against cancer. Biochem.Biophys.Res.Commun.414 445 450 - 118.
Li G. Ho V. C. 1998 53 DNA repair and apoptosis respond differently to high- and low-dose ultraviolet radiation. Br.J.Dermatol.139: 3-10. - 119.
Gabrielli B. Brooks K. Pavey S. 2012 Defective cell cycle checkpoints as targets for anti-cancer therapies. Front Pharmacol.3:9 EOF - 120.
Roos W. P. Kaina B. 2012 DNA damage-induced apoptosis: From specific DNA lesions to the DNA damage response and apoptosis. Cancer Lett. - 121.
Hanahan D. Weinberg R. A. 2011 Hallmarks of cancer: the next generation. Cell ;144 646 674 - 122.
Hers I. EE Vincent Tavare. J. M. 2011 Akt signalling in health and disease. Cell Signal.23 1515 1527 - 123.
Sarbassov DD, Guertin DA, Ali SM, Sabatini DM 2005 Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science ;307 1098 1101 - 124.
Liu P. Cheng H. Roberts T. M. Zhao J. J. 2009 Targeting the phosphoinositide 3-kinase pathway in cancer. Nat.Rev.Drug Discov.8 627 644 - 125.
Vivanco I. Sawyers C. L. 2002 The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat.Rev.Cancer ;2 489 501 - 126.
Haddad JJ, Abdel-Karim NE 2011 NF-kappaB cellular and molecular regulatory mechanisms and pathways: therapeutic pattern or pseudoregulation? Cell Immunol.271 5 14 - 127.
Hafsi S. Pezzino F. M. Candido S. Ligresti G. Spandidos D. A. Soua Z. et al. 2012 Gene alterations in the PI3K/PTEN/AKT pathway as a mechanism of drug-resistance (review). Int.J.Oncol.40 639 644 - 128.
Zitvogel L. Apetoh L. Ghiringhelli F. Andre F. Tesniere A. Kroemer G. 2008 The anticancer immune response: indispensable for therapeutic success? J.Clin.Invest ;118 1991 2001 - 129.
Garg A. D. Krysko D. V. Verfaillie T. Kaczmarek A. Ferreira G. B. Marysael T. et al. 2012 A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J.31 1062 1079 - 130.
Tesniere A. Panaretakis T. Kepp O. Apetoh L. Ghiringhelli F. Zitvogel L. et al. 2008 Molecular characteristics of immunogenic cancer cell death. Cell Death.Differ.15 3 12 - 131.
Chao M. P. Jaiswal S. Weissman-Tsukamoto R. AA Alizadeh Gentles. A. J. Volkmer J. et al. 2010 Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci.Transl.Med.2:63ra94 EOF - 132.
Chao M. P. Weissman I. L. Majeti R. 2012 The CD47-SIRPalpha pathway in cancer immune evasion and potential therapeutic implications. Curr.Opin.Immunol.24 225 232 - 133.
Leontieva OV, Gudkov AV, Blagosklonny MV 2010 Weak53 permits senescence during cell cycle arrest. Cell Cycle ;9: 4323-4327. - 134.
Rodier F. Campisi J. 2011 Four faces of cellular senescence. J.Cell Biol.192 547 556 - 135.
Schmitt CA 2007 Cellular senescence and cancer treatment. Biochim.Biophys.Acta ;1775 5 20 - 136.
Fimia G. M. Stoykova A. Romagnoli A. Giunta L. BS Di Nardacci R. et al. 2007 Ambra1 regulates autophagy and development of the nervous system. Nature ;447 1121 1125 - 137.
Liang C. Feng P. Ku B. Dotan I. Canaani D. Oh B. H. et al. 2006 Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG. Nat.Cell Biol.8 688 699 - 138.
Mehrpour M. Esclatine A. Beau I. Codogno P. 2010 Overview of macroautophagy regulation in mammalian cells. Cell Res.20 748 762 - 139.
Ku B. Woo J. S. Liang C. Lee K. H. Jung J. U. Oh B. H. 2008 An insight into the mechanistic role of Beclin 1 and its inhibition by prosurvival Bcl-2 family proteins. Autophagy.4 519 520 - 140.
Liang X. H. Jackson S. Seaman M. Brown K. Kempkes B. Hibshoosh H. et al. 1999 Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature ;402 672 676 - 141.
Yue Z. Jin S. Yang C. Levine A. J. Heintz N. 2003 Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc.Natl.Acad.Sci.U.S.A ;100 15077 15082 - 142.
Qu X. Yu J. Bhagat G. Furuya N. Hibshoosh H. Troxel A. et al. 2003 Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J.Clin.Invest ;112 1809 1820 - 143.
Matsui Y. Takagi H. Qu X. Abdellatif M. Sakoda H. Asano T. et al. 2007 Distinct roles of autophagy in the heart during ischemia and reperfusion: roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ.Res.100 914 922 - 144.
Takahashi Y. Coppola D. Matsushita N. Cualing H. D. Sun M. Sato Y. et al. 2007 Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis. Nat.Cell Biol.9 1142 1151 - 145.
Ionov Y. Nowak N. Perucho M. Markowitz S. Cowell J. K. 2004 Manipulation of nonsense mediated decay identifies gene mutations in colon cancer Cells with microsatellite instability. Oncogene ;23 639 645 - 146.
Kim MS, Jeong EG, Ahn CH, Kim SS, Lee SH, Yoo NJ 2008 Frameshift mutation of UVRAG, an autophagy-related gene, in gastric carcinomas with microsatellite instability. Hum.Pathol.39 1059 1063 - 147.
Bekri S. Adelaide J. Merscher S. Grosgeorge J. Caroli-Bosc F. Perucca-Lostanlen D. et al. 1997 Detailed map of a region commonly amplified at 11q13-->q14 in human breast carcinoma. Cytogenet.Cell Genet.79 125 131 - 148.
Lee JW, Jeong EG, Soung YH, Nam SW, Lee JY, Yoo NJ, et al. 2006 Decreased expression of tumour suppressor Bax-interacting factor-1 (Bif-1), a Bax activator, in gastric carcinomas. Pathology ;38 312 315 - 149.
Coppola D. Khalil F. Eschrich S. A. Boulware D. Yeatman T. Wang H. G. 2008 Down-regulation of Bax-interacting factor-1 in colorectal adenocarcinoma. Cancer ;113 2665 2670 - 150.
Cuddeback S. M. Yamaguchi H. Komatsu K. Miyashita T. Yamada M. Wu C. et al. 2001 Molecular cloning and characterization of Bif-1. A novel Src homology 3 domain-containing protein that associates with Bax. J.Biol.Chem.276 20559 20565 - 151.
Yang YP, Liang ZQ, Gu ZL, Qin ZH 2005 Molecular mechanism and regulation of autophagy. Acta Pharmacol.Sin.26 1421 1434 - 152.
Gozuacik D. Kimchi A. 2004 Autophagy as a cell death and tumor suppressor mechanism. Oncogene ;23 2891 2906 - 153.
Samuels Y. Velculescu V. E. 2004 Oncogenic mutations of PIK3CA in human cancers. Cell Cycle ;3 1221 1224 - 154.
Saeki K. Yuo A. Okuma E. Yazaki Y. Susin S. A. Kroemer G. et al. 2000 Bcl-2 down-regulation causes autophagy in a caspase-independent manner in human leukemic HL60 cells. Cell Death.Differ.7 1263 1269 - 155.
Pattingre S. Tassa A. Qu X. Garuti R. Liang X. H. Mizushima N. et al. 2005 Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell ;122 927 939 - 156.
Maiuri M. C. Le Criollo T. G. Rain A. Gautier J. C. Juin F. et P. al 2007 Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J.26 2527 2539 - 157.
Mathew R. Kongara S. Beaudoin B. Karp C. M. Bray K. Degenhardt K. et al. 2007 Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev.21 1367 1381 - 158.
Karantza-Wadsworth V. Patel S. Kravchuk O. Chen G. Mathew R. Jin S. et al. 2007 Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev.21 1621 1635 - 159.
Tasdemir E. Maiuri M. C. Galluzzi L. Vitale I. Djavaheri-Mergny M. D’Amelio M. et al. 2008 Regulation of autophagy by cytoplasmic53 Nat.Cell Biol.10: 676-687. - 160.
Zong WX, Thompson CB 2006 Necrotic death as a cell fate. Genes Dev.20 1 15 - 161.
Balkwill F. Charles K. A. Mantovani A. 2005 Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell ;7 211 217 - 162.
Vakkila J. Lotze M. T. 2004 Inflammation and necrosis promote tumour growth. Nat.Rev.Immunol.4 641 648 - 163.
Degenhardt K. Mathew R. Beaudoin B. Bray K. Anderson D. Chen G. et al. 2006 Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell ;10 51 64 - 164.
Boya P. Gonzalez-Polo R. A. Casares N. Perfettini J. L. Dessen P. Larochette N. et al. 2005 Inhibition of macroautophagy triggers apoptosis. Mol.Cell Biol.25 1025 1040 - 165.
Majmundar AJ, Wong WJ, Simon MC 2010 Hypoxia-inducible factors and the response to hypoxic stress. Mol.Cell ;40 294 309 - 166.
Mazure N. M. Pouyssegur J. 2010 Hypoxia-induced autophagy: cell death or cell survival? Curr.Opin.Cell Biol.22 177 180 - 167.
Bellot G. Garcia-Medina R. Gounon P. Chiche J. Roux D. Pouyssegur J. et al. 2009 Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol.Cell Biol.29 2570 2581 - 168.
JD Rabinowitz White. E. 2010 Autophagy and metabolism. Science ;330 1344 1348 - 169.
Fung C. Lock R. Gao S. Salas E. Debnath J. 2008 Induction of autophagy during extracellular matrix detachment promotes cell survival. Mol.Biol.Cell ;19 797 806 - 170.
Kenific C. M. Thorburn A. Debnath J. 2010 Autophagy and metastasis: another double-edged sword. Curr.Opin.Cell Biol.22 241 245 - 171.
Debnath J. 2008 Detachment-induced autophagy during anoikis and lumen formation in epithelial acini. Autophagy.4 351 353 - 172.
Yerbes R. Palacios C. Lopez-Rivas A. 2011 The therapeutic potential of TRAIL receptor signalling in cancer cells. Clin.Transl.Oncol.13 839 847 - 173.
Ashkenazi A. Dixit V. M. 1998 Death receptors: signaling and modulation. Science ;281 1305 1308 - 174.
Nguyen T. Zhang X. D. Hersey P. 2001 Relative resistance of fresh isolates of melanoma to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. Clin.Cancer Res.7: 966s-973s. - 175.
Pavet V. MM Portal Moulin. J. C. Herbrecht R. Gronemeyer H. 2011 Towards novel paradigms for cancer therapy. Oncogene ;30 1 20 - 176.
Hetschko H. Voss V. Seifert V. Prehn J. H. Kogel D. 2008 Upregulation of DR5 by proteasome inhibitors potently sensitizes glioma cells to TRAIL-induced apoptosis. FEBS J.275 1925 1936 - 177.
Johnstone RW, Frew AJ, Smyth MJ 2008 The TRAIL apoptotic pathway in cancer onset, progression and therapy. Nat.Rev.Cancer ;8 782 798 - 178.
Speirs C. K. Hwang M. Kim S. Li W. Chang S. Varki V. et al. 2011 Harnessing the cell death pathway for targeted cancer treatment. Am.J.Cancer Res.1 43 61 - 179.
Martinez-Lostao L. Marzo I. Anel A. Naval J. 2012 Targeting the Apo2L/TRAIL system for the therapy of autoimmune diseases and cancer. Biochem.Pharmacol.83 1475 1483 - 180.
Aggarwal BB 2003 Signalling pathways of the TNF superfamily: a double-edged sword. Nat.Rev.Immunol.3 745 756 - 181.
Holler N. Zaru R. Micheau O. Thome M. Attinger A. Valitutti S. et al. 2000 Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat.Immunol.1 489 495 - 182.
Vanden Berghe. T. van Saelens L. G. Van Brouckaert X. G. M. Kalai G. et M. al 2004 Differential signaling to apoptotic and necrotic cell death by Fas-associated death domain protein FADD. J.Biol.Chem.279 7925 7933 - 183.
Tarhini AA, Kirkwood JM 2007 Oblimersen in the treatment of metastatic melanoma. Future.Oncol.3 263 271 - 184.
Heidari N. MA Hicks Harada. H. 2010 GX15-070 (obatoclax) overcomes glucocorticoid resistance in acute lymphoblastic leukemia through induction of apoptosis and autophagy. Cell Death.Dis.1:e76 EOF - 185.
Mc Coy F. Hurwitz J. Mc Tavish N. Paul I. Barnes C. O’Hagan B. et al. 2010 Obatoclax induces Atg7 dependent autophagy independent of beclin-1 and BAX/BAK. Cell Death.Dis.1: e108. - 186.
Oltersdorf T. Elmore S. W. Shoemaker A. R. Armstrong R. C. Augeri D. J. BA Belli et. al 2005 An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature ;435 677 681 - 187.
van Delft M. F. Wei A. H. Mason K. D. Vandenberg C. J. Chen L. Czabotar P. E. et al. 2006 The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer Cell ;10 389 399 - 188.
MS Cragg Harris. C. Strasser A. Scott C. L. 2009 Unleashing the power of inhibitors of oncogenic kinases through BH3 mimetics. Nat.Rev.Cancer ;9 321 326 - 189.
Carrington E. M. Vikstrom I. B. Light A. Sutherland R. M. Londrigan S. L. Mason K. D. et al. 2010 BH3 mimetics antagonizing restricted prosurvival Bcl-2 proteins represent another class of selective immune modulatory drugs. Proc.Natl.Acad.Sci.U.S.A ;107 10967 10971 - 190.
Chen D. J. Huerta S. 2009 Smac mimetics as new cancer therapeutics. Anticancer Drugs ;20 646 658 - 191.
Straub CS 2011 Targeting IAPs as an approach to anti-cancer therapy. Curr.Top.Med.Chem.11 291 316 - 192.
Gyrd-Hansen M. Meier P. 2010 IAPs: from caspase inhibitors to modulators of NF-kappaB, inflammation and cancer. Nat.Rev.Cancer ;10 561 574 - 193.
Kaelin W. G. Jr 2009 Synthetic lethality: a framework for the development of wiser cancer therapeutics. Genome Med.1:99 EOF - 194.
Chan DA, Giaccia AJ 2011 Harnessing synthetic lethal interactions in anticancer drug discovery. Nat.Rev.Drug Discov.10 351 364 - 195.
Bolderson E. Richard D. J. Zhou B. B. Khanna K. K. 2009 Recent advances in cancer therapy targeting proteins involved in DNA double-strand break repair. Clin.Cancer Res.15 6314 6320 - 196.
Dai Y. Grant S. 2010 New insights into checkpoint kinase 1 in the DNA damage response signaling network. Clin.Cancer Res.16 376 383 - 197.
Calvert H. Azzariti A. 2011 The clinical development of inhibitors of poly(ADP-ribose) polymerase. Ann.Oncol.22 Suppl 1: i53 i59. - 198.
Javle M. Curtin N. J. 2011 The potential for poly (ADP-ribose) polymerase inhibitors in cancer therapy. Ther.Adv.Med.Oncol.3 257 267 - 199.
Imre G. Larisch S. Rajalingam K. 2011 Ripoptosome: a novel IAP-regulated cell death-signalling platform. J.Mol.Cell Biol.3 324 326 - 200.
Safa AR, Pollok KE 2011 Targeting the Anti-Apoptotic Protein c-FLIP for Cancer Therapy. Cancers. (Basel) ;3 1639 1671 - 201.
Maiuri M. C. Zalckvar E. Kimchi A. Kroemer G. 2007 Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat.Rev.Mol.Cell Biol.8 741 752 - 202.
Chen N. Karantza-Wadsworth V. 2009 Role and regulation of autophagy in cancer. Biochim.Biophys.Acta ;1793 1516 1523 - 203.
Carew J. S. Nawrocki S. T. Kahue C. N. Zhang H. Yang C. Chung L. et al. 2007 Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood ;110 313 322 - 204.
Apel A. Herr I. Schwarz H. Rodemann H. P. Mayer A. 2008 Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res.68 1485 1494 - 205.
Nahta R. Yuan L. X. Zhang B. Kobayashi R. Esteva F. J. 2005 Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res.65 11118 11128 - 206.
Esparis-Ogando A. Ocana A. Rodriguez-Barrueco R. Ferreira L. Borges J. Pandiella A. 2008 Synergic antitumoral effect of an IGF-IR inhibitor and trastuzumab on HER2-overexpressing breast cancer cells. Ann.Oncol.19 1860 1869 - 207.
Ropero S. Menendez J. A. Vazquez-Martin A. Montero S. Cortes-Funes H. Colomer R. 2004 Trastuzumab plus tamoxifen: anti-proliferative and molecular interactions in breast carcinoma. Breast Cancer Res.Treat.86 125 137 - 208.
Yang S. Wang X. Contino G. Liesa M. Sahin E. Ying H. et al. 2011 Pancreatic cancers require autophagy for tumor growth. Genes Dev.25 717 729 - 209.
Guo J. Y. Chen H. Y. Mathew R. Fan J. Strohecker A. M. Karsli-Uzunbas G. et al. 2011 Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev.25 460 470 - 210.
Abe A. Yamada H. Moriya S. Miyazawa K. 2011 The beta-carboline alkaloid harmol induces cell death via autophagy but not apoptosis in human non-small cell lung cancer A549 cells. Biol.Pharm.Bull.34 1264 1272 - 211.
Yu L. Alva A. Su H. Dutt P. Freundt E. Welsh S. et al. 2004 Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8. Science ;304 1500 1502 - 212.
Yu L. Wan F. Dutta S. Welsh S. Liu Z. Freundt E. et al. 2006 Autophagic programmed cell death by selective catalase degradation. Proc.Natl.Acad.Sci.U.S.A ;103 4952 4957 - 213.
Lum JJ, DeBerardinis RJ, Thompson CB 2005 Autophagy in metazoans: cell survival in the land of plenty. Nat.Rev.Mol.Cell Biol.6 439 448 - 214.
Wu Y. T. Tan H. L. Huang Q. Kim Y. S. Pan N. Ong W. Y. et al. 2008 Autophagy plays a protective role during zVAD-induced necrotic cell death. Autophagy.4 457 466 - 215.
Amaravadi RK, Thompson CB 2007 The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin.Cancer Res.13 7271 7279 - 216.
Lieberthal W. Triaca V. Levine J. 1996 Mechanisms of death induced by cisplatin in proximal tubular epithelial cells: apoptosis vs. necrosis. Am.J.Physiol ;270: F700 F708. - 217.
Healy E. Dempsey M. Lally C. Ryan M. P. 1998 Apoptosis and necrosis: mechanisms of cell death induced by cyclosporine A in a renal proximal tubular cell line. Kidney Int.54 1955 1966 - 218.
Chan F. K. Shisler J. Bixby J. G. Felices M. Zheng L. Appel M. et al. 2003 A role for tumor necrosis factor receptor-2 and receptor-interacting protein in programmed necrosis and antiviral responses. J.Biol.Chem.278 51613 51621 - 219.
Cho DH, Jo YK, Hwang JJ, Lee YM, Roh SA, Kim JC 2009 Caspase-mediated cleavage of ATG6/Beclin-1 links apoptosis to autophagy in HeLa cells. Cancer Lett.274 95 100 - 220.
Djavaheri-Mergny M. Maiuri M. C. Kroemer G. 2010 Cross talk between apoptosis and autophagy by caspase-mediated cleavage of Beclin 1. Oncogene ;29 1717 1719 - 221.
Hou W. Han J. Lu C. Goldstein L. A. Rabinowich H. 2010 Autophagic degradation of active caspase-8: a crosstalk mechanism between autophagy and apoptosis. Autophagy.6 891 900