Tumor Biol. (2016) 37:8471–8486
DOI 10.1007/s13277-016-5035-9
REVIEW
Major apoptotic mechanisms and genes involved in apoptosis
Yağmur Kiraz 1,2 & Aysun Adan 1 & Melis Kartal Yandim 2 & Yusuf Baran 1,2
Received: 7 December 2015 / Accepted: 28 March 2016 / Published online: 9 April 2016
# International Society of Oncology and BioMarkers (ISOBM) 2016
Abstract As much as the cellular viability is important for the
living organisms, the elimination of unnecessary or damaged
cells has the opposite necessity for the maintenance of homeostasis in tissues, organs and the whole organism. Apoptosis, a
type of cell death mechanism, is controlled by the interactions
between several molecules and responsible for the elimination
of unwanted cells from the body. Apoptosis can be triggered
by intrinsically or extrinsically through death signals from the
outside of the cell. Any abnormality in apoptosis process can
cause various types of diseases from cancer to auto-immune
diseases. Different gene families such as caspases, inhibitor of
apoptosis proteins, B cell lymphoma (Bcl)-2 family of genes,
tumor necrosis factor (TNF) receptor gene superfamily, or p53
gene are involved and/or collaborate in the process of apoptosis. In this review, we discuss the basic features of apoptosis
and have focused on the gene families playing critical roles,
activation/inactivation mechanisms, upstream/downstream effectors, and signaling pathways in apoptosis on the basis of
cancer studies. In addition, novel apoptotic players such as
miRNAs and sphingolipid family members in various kind
of cancer are discussed.
Keywords Intrinsic/extrinsic pathway . Bcl-2 . Caspase .
TNF . TRAIL . p53
* Yusuf Baran
ybaran@gmail.com
1
Department of Molecular Biology and Genetics, Faculty of Life and
Natural Sciences, Abdullah Gül University, 38080 Kayseri, Turkey
2
Department of Molecular Biology and Genetics, İzmir Institute of
Technology, İzmir 35430, Turkey
Introduction
The main goal for a cell is to stay alive during the lifetime.
Holding the key of proliferation events as much as death
mechanism has vital importance for the cells to keep the balance between living and death cells in the body. As one of the
cellular death mechanisms, apoptosis, also known as programmed cell death, can be defined as the process of a proper
death of any cell under certain or necessary conditions.
Apoptosis is a part of natural homeostatic mechanism to keep
the number of the cells constant in an organism and helps the
tissue to eliminate increasing number of unwanted/unneeded
cells that are damaged or no longer manageable during development, growth or aging [1]. It also plays crucial roles in
early development and differentiation of the embryo in order
to generate a full and decent organism. Although the term of
Bapoptosis^ was firstly used by Kerr et al. in 1972 to identify a
distinguished type of cell death, the first description and understanding of the programmed mechanisms of apoptosis was
derived from the studies on the development of the nematode
Caenorhabtidis elegans in 1999 [2, 3].
Apoptosis is a defense mechanism against damaged,
stressed, or stimulated cells by any agents to prevent accumulation of non-functional cells in the tissues. If apoptosis is not
be mediated properly in unwanted cells, the mutations could
continue to accumulate in the cells that eventually could lead
to generation of cancer and other diseases such as autoimmune diseases, AIDS, or some of neurodegenerative disorders [4]. However, apoptosis is the best defined and wellunderstood Bprogrammed^ type of cell death; there are many
different types of cellular death mechanisms such as
pyroptosis, necrosis, or autophagy, and some others might
not yet be discovered [5, 6]. Apoptosis regulatory pathways,
including a number of gene families, orchestrate the specific
morphological and biochemical changes in the cells during the
8472
process. We touched on these significant chances briefly and
focused on the genes involved in apoptosis with details.
Morphological and biochemical processes of apoptosis
Many biochemical events and a series of morphological
changes occur at the early stage and increasingly continue till
the end of apoptosis process. Some of the changes such as cell
shrinkage, chromatin condensation, or nuclear differences
could be observed by microscopic techniques [1, 7].
Morphological event cascade including cytoplasmic filament
aggregation, nuclear condensation, cellular fragmentation,
and plasma membrane blebbing finally results in the formation of apoptotic bodies. All the morphological hallmarks of
apoptosis can be gathered under three headings; (i) the changes
occur in nucleus; (ii) cell membrane and cytosolic changes; (iii)
those happen in mitochondria [1, 8]. Chromatin condensation,
DNA fragmentation, and nuclear fragmentation are the nuclear changes that could be observed with light and fluorescence
microscopy during apoptosis. The apoptotic cell loses its association with other cells at initiation stage of the process by
different signals breaking the connection. This separation
followed by apoptotic body formation and resulted in
blocking the inflammatory reaction of the cells, since they
package their ingredient and do not release any contaminant
outside the cell. Then, the buddies are rapidly phagocytized by
other neighbor cells and these absorber cells do not produce
any signal that causes any inflammatory response. Also, mitochondria play an important role by interacting with many
different apoptotic/anti-apoptotic proteins and releasing signal
molecules [9, 10].
Chromatin condensation and nuclear fragmentation are the
major modifications observed in the nucleus, eventually
resulting in pyknosis (chromatin condensates irreversibly that
signs cell death) and followed by karyorrexis (nuclear fragmentation, the last event in nucleus during apoptosis) [11].
The fragmentation of double-stranded DNA into 180–200
bp sequences in length by the help of caspase proteins is also
another essential hallmark of nuclear events during apoptosis.
Caspases are responsible for DNA repair during replication as
well as the termination stage of apoptosis. They are also involved in the fragmentation in apoptosis together with DNA
fragmentation factors (DFFs) and endonucleases. Most of the
nuclear changes of apoptosis observed by electron microscopy or even light microscopy make the apoptosis process to be
determined easily [11, 12].
As soon as the apoptosis is initiated in the cells, they lose
the connection between the neighboring cells; membrane
shrinks and the cell packs its cytosolic ingredients into apoptotic buddies. The apoptotic buddies will be eliminated by
phagocytotic cells that recognize phosphatidylserine, which is
normally located in the inner side of the cell membrane and
flips to the outer membrane during apoptosis [13]. At the same
Tumor Biol. (2016) 37:8471–8486
time, the cytoplasmic scaffold proteins and cell junction proteins such as actin, β-catenin, spectrin, or Gas2 are deactivated
by cleavage and the cell loses its integrity by the function of
caspases [14].
Mitochondria have complex and important roles by providing various pro-apoptotic signals, creating a downstream cascade of apoptosis activation. The balance between the proapoptotic and anti-apoptotic molecules keeps the cellular homeostasis stable and determines the cell fate, which is either
apoptosis or proliferation. Mitochondria have a leading role in
releasing a number of important apoptosis inducing molecules
including cytochrome c, SMAC, apoptosis-inducing factor, or
endonuclease G as a result of permeabilization of mitochondrial membrane. Permeabilization is triggered by proapoptotic B cell lymphoma (Bcl)-2 family proteins, while
the integrity of mitochondrial membrane is maintained by
anti-apoptotic members of Bcl-2 family [10, 15, 16].
Several biochemical changes such as protein modifications/degradations, DNA and chromatin deteriorations, and
synthesis of cell surface markers form morphological process
during apoptosis. Caspases are mainly responsible for these
changes with their extensive capabilities to cleave certain molecules from one or more specific points, causing degradation
and inactivation of target protein. Moreover, they can also
inhibit the negative regulatory domains of specific proteins,
which leads to the activation of subjected molecule [17, 18].
They are also involved in DNA fragmentation process.
Intrinsic and extrinsic apoptotic pathways
Apoptosis can be stimulated by two different pathways: (i)
intrinsic pathway (or mitochondria) that mainly occurs via
release of cytochrome c from the mitochondria, which activates different caspases as downstream signals, and (ii) extrinsic pathway when Fas death receptor is activated by a signal
coming from the outside of the cell. After the activation of
different intermediate molecules by signaling cascade, both of
the pathways meet up at the final caspase activation step and
commonly lead to cleavage of different proteins [19] (Fig. 1).
Intrinsic pathway
The intrinsic pathway of apoptosis is independent from a receptor signaling, and mitochondria-associated stimuli create
an intracellular signaling. The inner activation of this pathway
makes the cell undergo apoptosis in either a positive or negative manner. The positive stimuli (e.g., toxic materials, viral
infections, and radiations) directly activate all the mediators
for apoptosis, whereas negative stimuli (loss of growth factors, different cytokines, or certain type of hormones) work in
contrast to positive one and eliminate the factors that suppress
apoptosis in the cells and cause apoptotic activation [1, 20]. In
addition to different infections or cytokine-mediated intrinsic
Tumor Biol. (2016) 37:8471–8486
8473
Fig. 1 Two main apoptotic pathways; intrinsic and extrinsic pathways. In
the extrinsic pathway, interaction between the death receptors and their
ligands initiates the pathway, resulting in caspase 8 activation. This
activation can be inhibited by cFLIP. Caspase 8 can directly induce
apoptosis or activates caspase 3 or Bid, which lead to apoptosis. On the
other hand, intrinsic pathway can be initiated by DNA damage. As a
response, the cells can trigger apoptosis through mitochondrial pathway,
which starts with the activation of the pro-apoptotic member of the Bcl-2
family, Bax. Anti-apoptotic proteins inhibiting the action of Bax are
located on the membrane of the mitochondria such as Bcl-2 and BclXL. Release cytochrome c, APAF-1 complex, and pro-caspase9 can be
gathered in the cytosol, which is called apoptosome. The formation of this
complex will result in the activation of caspase 9 followed by the
transformation of pro-caspase-3 to caspase 3, which is the last step for
apoptosis. The cross talk between extrinsic and intrinsic pathways of
apoptosis is regulated by Bid, a pro-apoptotic member of Bcl-2 family.
The cleavage of Bid is mediated by caspase 8, which induces apoptosis by
releasing cytochrome c release from the mitochondria
apoptosis activation, DNA damage also majorly induces apoptosis as a protection mechanism of the cells that do not let self
to continue proliferation with an imperfect DNA sequence.
DNA damage or any other type of apoptosis stimuli basically
causes the changes in the trans-membrane potential of mitochondria, which result in the release of pro-apoptotic proteins
into the cytoplasm.
Cytochrome c, Smad, or high-temperature requirement
protein A2 (HtrA2)/Omi are a group of pro-apoptotic molecules released from mitochondria and cause the activation of
caspase protein cascade [21, 22]. Cytochrome c interacts with
Apaf-1, resulting in the formation of Bapoptosome^ complex,
which activates pro-caspase-9. After active caspase-9 activates caspase-3, the final cascade is become activated and
nucleus will be fragmented together with the breaking of nuclear membrane [23]. This stage is the initial event for extrinsic and intrinsic pathways of apoptosis, where caspase-3
cleaves the different proteins such as kinases, DNA control
proteins, cytoskeletal proteins, or inhibitor of endonucleases.
DNA condensation, membrane blebbing, and all the morphological changes are regulated by caspases as a common mechanism for both intrinsic and extrinsic trigger [17].
On the other hand, another group of molecules released by
mitochondria including endonuclease G or AIF are also proapoptotic proteins but involved in the process at the later
stages. These molecules are trans-located into the nucleus
where they first cause an elementarily DNA fragmentation
and chromatin condensation which is defined as Bstage 1,^
8474
and an advanced condensation and DNA fragmentation by the
help of caspase-3 at later stage is called as Bstage 2^ [24].
All the intrinsic apoptosis events are primarily controlled by
Bcl-2 family of proteins and p53 tumor suppressor protein
which is majorly involved in the activation of Bcl-2 family
proteins. The members of Bcl-2 protein family can act as either
pro-apoptotic (Bax, Bak, Bid, Bim, Puma, Noxa, Bad, and
Blk) or anti-apoptotic (Bcl-2, Bcl-XL, Bcl-X, and BAG) and
also determine the membrane integrity of mitochondria and are
involved in the process of cytochrome c release [1, 25].
Extrinsic pathway
Apoptosis triggered by extrinsic pathway is primarily mediated by signaling through membrane-bound death receptors that
belong to tumor necrosis factor (TNF) gene superfamily. The
initial signal is provided by the interactions between the ligands and cell membrane death receptors such as Fas ligand/
FasR, TNF/TNF R1, Apo2L/DR4, or TNF-related apoptosisinducing ligand (TRAIL) R1, which is resulted in ligation of
death domains of these receptors [26]. Binding of Fas ligand
to its receptor induces the binding of adaptor protein, Fasassociated death domain (FADD), while TNF/tumor necrosis
factor receptor (TNFR) interaction causes the binding of
TNFR-associated death domain (TRADD), which is resulted
in pro-caspase-8 activation. Pro-caspase-8 is activated autocatalytically by the help of death-inducing signaling complex
(DISC). Active caspase-8 either induce Bid, thus intrinsic
pathway also become involved and activated with an outside
signal, or caspase-3 and caspase-7 and the activation process
of apoptosis is terminated with the same final pathway as
intrinsic stimuli does [1, 19, 27, 28]. Bid is the pro-apoptotic
member of Bcl-2 family, exhibiting a common molecule between intrinsic and extrinsic pathways of apoptosis. Caspase8 causes the cleavage and myristoylation of cytoplasmic Bid
protein, leading to its movement through mitochondria. Then,
apoptosome formation is induced by cytochrome release via
Bak and Bax molecules [29].
The extrinsic activation of apoptosis can also be inhibited
via two different ways. The one is binding of FLICE-like
inhibitory protein (cFLIP) to FADD and pro-capase-8 and
blocking their activity, and the other way is inhibition of
caspase-8 biogenesis by a protein named Toso which is firstly
described in T cells [30, 31].
In the next sessions, we will discuss all the gene families
involved in intrinsic or extrinsic pathways of apoptosis with
their main player and their functions.
Caspase family members
Caspase family comprise conserved cysteine aspartic-specific
proteases, and members of caspase family are considerably
crucial in the regulation of apoptosis [32]. In C. elegans,
Tumor Biol. (2016) 37:8471–8486
Ced-3 was reported to be essential for the cell death, which
is mainly conducted by the caspase Ced-3, Ced-4 activating
Ced-3, and Ced-9 inhibiting apoptosis [33, 34]. There are 14
different caspases in mammals, and they are basically classified as the initiators including caspase-2, -8, -9, and -10; and
the effectors including caspase-3, -6, -7, and -14; and also the
cytokine activators including caspase-1, -4, -5, -11, -12, and 13 [35, 36]. Structurally, while initiator caspases have long Nterminal pro-domain known as caspase recruitment domains
(CARDs) including more than 90 amino acids, effector
caspases have shorter sequences known as death effector domain (DED) including 20–30 amino acids. Since caspases
firstly synthesized as zymogens, they are subsequently activated during the apoptotic process. While initiator caspases
are self-activated, effector caspases are activated by initiator
caspases via internal cleavages [37]. Rather than apoptosis,
most of caspase family members are functional in cellular
proliferation, survival, and inflammation, whereas some of
them are essential for apoptosis [38, 39].
Caspase-1, the first identified caspase, is interleukin-1b
processing enzyme (ICE), and it is known as Ced-3 homologue [40]. Caspase-1 is involved in cytokine activator group
of caspase family since inflammatory cytokines, pro-IL-1b
and pro-IL-18, are the main substrates for caspase-1 [39].
While caspase-1 is not essential for apoptotic signaling, it is
essential in inflammation process [41].
The second identified caspase, caspase-2, containing
CARD, plays important roles in DNA damage-, metabolic
abnormality-, and ER stress-induced apoptosis [42].
Caspase-2 is known as a substrate for both caspase-3 and
caspase-8 [43, 44]. Caspase-2 activation comprises the formation of PIDDosome complex including RIP-associated ICH-1/
ECD3 homologous protein with death domain (RAIDD) that
have CARD and death domain (DD) and p53-induced protein
with death domain (PIDD) [45]. Functional properties of
caspase-2 have still not been clarified thoroughly.
Caspases-3, -6, and -7 are involved in the effector caspase
group, and they act in a similar manner in the apoptotic process [46]. Caspase-3 is activated via both extrinsic and intrinsic apoptotic pathways [47]. Despite there are limited information about caspase-6 and -7 rather than caspase-3, it is
known that while caspase-3 suppression results in the inhibition of apoptosis, suppression of caspase-6 and -7 do not significantly affect the apoptotic process [48]. Furthermore,
caspase-3 was reported to be crucial for PARP cleavage and
DNA fragmentation which are hallmarks of apoptosis [49].
However, some studies showed that under the conditions that
both caspase-3 and caspase-7 were knocked out, the cells
could undergo cell death in an alternative manner via necrosis.
Studies with caspase-3/caspase-7 double-knockout thymocytes and mouse embryonic fibroblasts showed that thymocytes remain sensitive to Fas-mediated apoptosis, whereas fibroblasts become resistant [49, 50].
Tumor Biol. (2016) 37:8471–8486
Functionally well-known member of caspase family, caspase-8, is crucial factor for TNF-induced extrinsic apoptotic
pathway [51]. Pro-caspase-8 is recruited to DISC by FADD,
and dimerization or trimerization triggers pro-caspase-8 activation via reciprocal cleavage. Caspase-8, in turn, cleaves and
activates caspase-3, -7, Bid, and also NF-kB [52, 53].
Caspase-8 activation is regulated by cFLIP that is structurally
homologous to caspase-8 but does not have caspase activity
[54]. FLIPS, the short isoform of cFLIP, controls the DISC
formation in a negative manner. However, the long isoform
of cFLIP, FLIPL, has reciprocal effect on DISC formation and
caspase-8 activity. While some studies reported FLIPL to be
inducer of DISC formation, some reports showed that it could
be an inhibitor [55, 56]. Inhibitor and inducer effects of FLIPL
on caspase-8 activity depend on FLIPL levels. At low concentrations, generation of heterodimers between FLIPL and procaspase-8 or pro-caspase-10 induces their activity. However,
at higher levels of FLIPL, caspase-8 activation diminishes and
NF-kB activation increases [57, 58].
Caspase-9, the initiator caspase, is an important factor for
the generation of apoptosome complex in the mitochondrial
pathway. Once cytochrome c is released from the mitochondria, it binds to Apaf1 which is the receptor for cytochrome c
in the cytoplasm [59]. Cytochrome-c and Apaf-1 generate
apoptosome, and then, pro-caspase-9 binds to Apaf-1.
Afterward, pro-caspase-9 is activated via reciprocal cleavage,
and by this way, apoptosome complex also become activated.
Then, caspase-3 is cleaved and activated via caspase-9 found
in the active apoptosome complex [60].
Inhibitors of apoptosis proteins and inhibitors of apoptosis
protein antagonists
Inhibitors of apoptosis proteins (IAPs) were firstly discovered
in Baculovirus as gene products. All IAPs have baculovirus
IAP repeats (BIRs) that composed of one or more zinc finger
motifs [61]. The first identified IAP, OpIAP, inhibits procaspase cleavage and activation rather than direct inhibition
of caspase activity [62]. NAIP, the first identified mammalian
IAP, was reported to be related to the generation of immune
response against bacterial infection. Hence, it is not directly
correlated with caspase inhibition [63].
Survivin/BIRC5, another identified mammalian IAP, was
firstly reported as caspase inhibitor, but now, it is known that
survivin does not directly inhibit caspase activity. Mechanism
of action of survivin, bearing one BIR domain, is to assemble
with centromeres and p21Waf1 at the beginning of mitosis
[64, 65].
XIAP/BIRC4, the mostly clarified mammalian IAP, has
three BIR domains and a RING domain; it is located on X
chromosome; and also, it is quite effective in apoptosis inhibition via inhibiting caspase activity [66]. Mainly, caspase-3
and caspase-7 can be inhibited by XIAP via inserting a residue
8475
of aspartic acid into the caspase active region [67].
Additionally, XIAP can also inhibit caspase-9 activity via
binding the third BIR to the N-terminus of pro-caspase-9,
resulting in the prevention of caspase-9 dimerization [68].
Similarly, another mammalian IAPs, cIAP1/BIRC2, and
cIAP2/BIRC3 have three BIR domains and a RING domain
in their structures [63]. Rather than inhibiting directly caspase
activity, they inhibit apoptosis indirectly. They can bind IAP
inhibitor, second mitochondrion-derived activator of caspase/
direct inhibitor of apoptosis-binding protein with low pI
(SMAC/DIABLO), trigger NF-kB and MAPK activity, and
also they can trigger proteasomal degradation of caspases
[69].
Bruce/BIRC6, a member of IAP family, is mainly found in
secretory organs, testis, lymphatic cells, and brain. Similar to
survivin, Bruce located at the outer membrane of the transGolgi network bears only one BIR domain. Bruce can inhibit
caspase-3, -6, -7, -8, and -9, and also, it can trigger the
proteasomal degradation of SMAC/DIABLO. Many studies
showed that Bruce is upregulated in ovarian and brain cancer
cell lines, resulting in development of resistance against apoptotic agent [70].
IAP antagonists, SMAC/DIABLO, HtrA2/Omi, and
XIAP-associated factor 1 (XAF1) are known as potent inhibitors of IAPs. SMAC/DIABLO bears a mitochondrial
targeting signal (MTS) at N-teminus, and it become mature
after the MTS cleavage [21]. Once apoptosis is triggered,
SMAC/DIABLO is delivered into the cytosol, and
SMAC/DIABLO homodimers bind to IAPs via its Ala-ValPro-Ile sequence in N-terminal domain [71]. Interaction of
SMAC/DIABLO with XIAP via the second and third BIRs
results in caspase-3 and caspase-9 release [72]. It was reported
that SMAC is overexpressed in several types of solid tumors
such as colon, stomach, prostate, ovary, and lung cancers [73].
Likewise, HtrA2/Omi delivery is triggered via apoptotic induction and then, it binds to IAPs through its IAP-binding
motif (IBM) [74]. XAF1, the other potent IAP antagonist,
binds to BIR domains of IAPs such as XIAP, cIAP1, and
cIAP2 and by this way, promotes apoptosis [75]. A study
suggesting XAF1 as a prognostic marker for colon cancer
showed that XAF1 is overexpressed in colon cancer cells as
compared to adenoma cells which are benign [76].
Bcl-2 family members
Bcl-2 family members, which play important roles in regulating apoptotic signaling, are divided into three subfamilies including (i) pro-survival subfamily members (Bcl-2, BclXL,
BclW, MCL1, and BFL1/A1), (ii) BH3-only subfamily members (Bad, Bim, Noxa, and Puma9), and (iii) pro-apoptotic
mediator subfamily members (Bax and Bak) [77, 78].
Basically, all of the members of Bcl-2 family share typical
characteristic functions; (i) they dimerize with other members
8476
of Bcl-2 family, (ii) they contribute to the regulation of mitochondrial homeostasis by binding proteins, and (iii) they contribute to outer mitochondrial membrane pore formation [79].
The members of BH3-only subfamily members become
activated under stress conditions like growth factor deprivation and DNA damage. Active BH3-only proteins, in turn,
inactivate members of pro-survival subfamily via binding that
promotes the activation of the members of pro-apoptotic subfamily members. Active pro-apoptotic subfamily members,
Bak and Bax, then provide cytochrome c release from the
mitochondria through permeabilizing the outer mitochondrial
membrane [80].
In several types of human malignancies, the balance between the expression levels of Bcl-2 family genes is broken
down, and the equilibrium changes to the pro-survival subfamily member direction. In this case, cancer cells can escape
from apoptotic signals and therefore develop resistance
against therapeutic agents [81, 82]. Additionally, Bcl-2 family
members are also considered in cancer therapy due to their
therapeutic potentials [83]. In the clinical trials, the BH3-only
mimetic agents targeting Bcl-2 are being investigated in order
to find alternative potent therapeutic approaches in several
types of malignancies [84–86].
TNF gene superfamily
One of the most important ways of triggering apoptosis is
mediated through death receptors (DRs), which are classified
in TNF superfamily including also ligands such as TNF,
TRAIL, and Fas ligand (FasL) [87]. The induction of apoptosis by these ligands is initiated by binding to their specific
membrane receptors [88, 89]. TNF superfamily is known to
comprise 19 ligands and 29 receptors that function in highly
different processes in the body including inflammation, apoptosis, proliferation, and invasion [90]. Even though all members of TNFR superfamily are generally trimeric type I transmembrane proteins and possess cysteine-rich extracellular
subdomains, they are actually different in their primary structure, which make them unique to recognize their ligand in a
specific and exclusive manner [91]. These DRs also contain a
homologous cytoplasmic cysteine-rich BDD^ which is responsible for transmission of apoptotic signals from cell surface to
intracellular signaling pathways [92]. Because, adapter molecules such as FADD and TRADD have these death domains
as well to interact with DRs. The ligands included in TNF
superfamily share a common extracellular TNF homology
domain (THD), which is involved in the formation of
homotrimers via non-covalent bonding [93]. The extracellular
domain of most TNF ligands undergoes proteolytic cleavage
to make a soluble ligand, even though they are synthesized as
type II trans-membrane proteins [93].
CD95 (DR2/Fas/APO-1), TNF receptor 1 (DR1/TNFR1),
TRAIL-R1 (DR4), and TRAIL-R2 (DR5) are the best
Tumor Biol. (2016) 37:8471–8486
characterized DRs of which ligands are CD95 ligand
(CD95L/FasL), TNFα, lymphotoxin-α (these two bind to
TNFRI), and TRAIL (these two bind to TRAIL-R1 and
TRAIL-R2), respectively [94]. The genes encoding TNF superfamily receptors and ligands are the scope of this part together with their significant contribution to apoptosis
specifically.
Fas cell surface death receptor and Fas ligand
The proteins encoded by Fas cell surface death receptor (FAS)
and Fas ligand (FASLG) genes are members of the TNF superfamily. Fas also known as CD95/APO-1/DR2 is one of the
best studied DRs with molecular weight of 48 kDa. Fas gene
occupies about 25 kb on human chromosome 10 with nine
exons in which exon 6 codes for trans-membrane domain
[95]. This receptor containing a DD plays a crucial role in
the regulation of programmed cell death and is involved in
the pathogenesis of various malignancies such as cancer. On
the other hand, FasLG gene is located on human chromosome
1, which encodes a type II trans-membrane protein called
FasL (CD95L) present at the surface of activated immune
cells such as T cells and natural killer cells [93]. Therefore,
Fas/FasL interaction results in the elimination of infected and
transformed cells, which is generally used by immune cells to
avoid cancer development.
The interaction of Fas with FasL results in changes in the
conformation and aggregation of receptor on the plasma membrane and triggers an initial signaling event through proteinprotein interactions. The main structural changes take place in
DD of receptor, which recruits FADD through its DD. Then,
FADD interacts with pro-caspase-8 and -10. After activation
of these caspases via auto-cleavage, they are released in the
cytosol as active caspases resulting in the apoptotic cell death.
The complex CD95/FADD/caspase-8/-10 is called DISC
stands for Bdeath-inducing signaling complex^ [96].
In the literature, there are various kinds of studies identifying the roles of Fas/FasL in order to induce apoptosis in several malignancies especially in cancer. Escaping from apoptotic stimuli is a very well known feature of cancer cells which
might develop multiple strategies to inhibit apoptotis mediated
by CD95. In a majority of cancer types from different origins,
somatic mutations in CD95 gene were found to be a common
way to trigger the development of resistance toward apoptosis.
For instance, 5′ region of CD95 gene was analyzed in terms of
somatic mutations in nodal diffuse large B cell lymphoma and
defined mutations were shown to be included in progression
of diseases due to inhibition of apoptosis [97]. Another common way to become resistant to CD95-induced apoptosis is
the regulation of surface expression of receptor [98]. In a study
performed by Ivanow et al. (2006) [98], the treatment of melanoma cells having increased surface expression of Fas receptor with soluble FasL resulted in the induction of apoptosis.
Tumor Biol. (2016) 37:8471–8486
CD95-mediated apoptosis can also be blocked by decreasing
expression of FADD or caspase-8 [99, 100].
In a recent study, the effect of a toxic steroid on human
bladder cancer cells was found to be related to increased expressions of Fas and FasL in vitro and in vivo at both messenger RNA (mRNA) and protein levels [101]. Zhong et al.
(2015) [102] showed that the mechanism of resistance to
Fas-mediated apoptosis in human hepatocellular carcinoma
cells (HCCs) and overexpression of oxysterol-binding protein-related protein 8 (ORP8) which is normally downregulated in HCC was found to induce apoptosis by upregulating
FasL.
TRAIL
TRAIL (also called as APO2 ligand) is a type II transmembrane protein and processed proteolytically at the cell
surface to form a soluble ligand. TRAIL is grouped into the
TNF cytokine family, which was discovered based on its extracellular domain sequence homology with CD95L and TNF
[103]. TRAIL-mediated apoptosis takes place after its binding
to its DD containing receptors, TRAIL receptor 1 (death receptor 4, DR4), and TRAIL receptor 2 (death receptor 5, DR5)
[104]. There are also three other TRAIL receptors, which do
not possess apoptotic ability and function as decoys. Decoy
receptors 1 (DcR1) and 2 (DcR2) are expressed on the cell
surface similar to DR4 and DR5. Even though their extracellular and ligand-binding domains show significant homology
to DR4 and DR5 and are fully functional, they lack of functional intracellular DD [105]. Therefore, increased expression
of either DcR1 or DcR2 provides resistance against TRAILinduced apoptosis [104]. In a recent study, it was shown that
coexpression of DcR1 and DcR2 with DR4/DR5 on the same
cell can block apoptosis. However, TRAIL was engineered in
order to escape from binding to DcRs, which were found to
still exert trans-cellular regulation originating from stromal
cells and affect tumor cells. Therefore, it is important to target
these decoy receptors selectively to gain maximum efficacy
[106]. Another recent study showed that DR4 and DR5 were
upregulated while DcR1 and DcR2 downregulated in colon
cancer cells after their treatment with a non-steroidal anti-inflammatory drug. Therefore, colon cancer cells became sensitive to TRAIL-induced apoptosis [107]. The fifth TRAIL receptor is osteoprotegerin (OPG), which is a secreted receptor
for TRAIL with low affinity. OPG has sequence homology to
TNF receptor superfamily, however but lacks of a transmembrane domain. OPG can bind to TRAIL and prevents
its interaction with death receptors, thus preventing Jurkat
cells from undergoing TRAIL-mediated apoptosis [108].
Therefore, OPG has an anti-apoptotic effect by preventing
interaction between TRAIL and the death receptors [109].
Lane et al. (2012) [110] showed the relationship between
8477
higher OPG expression and resistance of ovarian cancer cells
to TRAIL-induced apoptosis.
Similar to apoptosis induction by Fas/FasL, binding of
TRAIL to its DRs, DR4 and DR5 recruits the adaptor protein
FADD and inactive versions of caspase-8 and -10 to form the
DISC complex in which caspase-8 and -10 become activated
by auto-proteolytic cleavage and released in the cytosol to
activate effector caspases [111].
The size of human TRAIL gene is ~20 kb, containing five
exons and four introns. The first exon encodes for the transmembrane domain and the cytoplasmic domain, whereas
exons 4 and 5 code for the extracellular domain responsible
for the interaction of TRAIL with its receptors. Exon 5 also
encodes for the C-terminal amino acids along with containing
the 3′-untranslated region (3′-UTR) and poly-A tail [112].
There are various variants of TRAIL; however, only one specific isoform of TRAIL (TRAILα) is responsible for its
cancer-selective apoptosis induction potential [113].
TRAIL gene is subjected to strict regulation due to its critical role in the induction apoptosis. Altered expression of
TRAIL gene has been found in various kind of disease.
Multiple studies have been performed to analyze singlenucleotide polymorphisms (SNPs) in various patient populations. In a recent study, peripheral blood samples were analyzed in terms of SNPs in TRAIL promoter and substitution of
C to T at position -723 was found to be significantly associated
with sporadic breast cancer and decreased TRAIL mRNA
levels due to transcriptional repression [114]. Bos et al.
(2009) found that TRAIL mRNA expression decreased
in breast cancer patients with brain metastasis [115].
TRAIL has been considered as a potential therapeutic target due to its selective apoptosis inducing action in cancer
cells as compared normal cells [116]. There are several ongoing and completed clinical phase studies targeting TRAIL
apoptotic pathway by using agents including monoclonal antibodies and recombinant human proteins, and these
studies are giving promising results with no significant toxic
effects [117].
TNF and TNF receptor 1–2 (TNFR1 AND TNFR2)
TNF is expressed as a trans-membrane 26-kDa protein and
then undergoes proteolytic cleavage to form 17-kDa trimeric
soluble cytokine. The resulting TNF functions by binding to
two different receptors, TNRF1 and TNFR2, which are also
trimeric trans-membrane proteins [90]. TNF is produced by
immune cells including activated natural killer cells, T cells,
and activated monocytes/macrophages and a wide range of
non-immune cells such as fibroblasts [118]. TNF/TNFR signaling is well known to be involved in various cellular functions such as apoptosis, cell proliferation, and differentiation
[119].
8478
TNF-mediated apoptosis is generally carried out by
TNFR1 (DR1), since only TNFR1 is known to contain DD
[87]. Binding of TNF to TNFR1 triggers the recruitment of
TRADD protein through its DD. Then, TRADD interacts with
FADD resulting in the recruitment of pro-caspase-8, which is
proteolytically cleaved to active caspase-8. Caspase-8 then
activates caspase-3 responsible for apoptotic cell death
[120]. The activity of caspase-8 is strictly regulated by a negative inhibitor protein cFLIP that contains DED instead of
DD. cFLIP interacts with pro-caspase-8 to prevent its continuous recruitment to the TNFR1 DISC [121]. TNF-induced
cell death occurs only under stress conditions such as altered
cell metabolism, inhibition of cell cycle progression, and protein synthesis [120]. Therefore, it only induces apoptosis in
transformed cells (cancer cells), virus-infected cells, or
stressed cells, not in normal healthy cells. Interestingly,
TNFR2 has been shown to function in cell proliferation unlike
TNFR1 [122].
In a recent study, IL32-α, a novel cytokine, was found to
inhibit colon cancer cell growth in an experimentally generated colon cancer model by increasing TNFR1-induced cell
death signaling, which was evidenced by increased expression of TNFR1. Yu et al. [123, 124] found a new way of
apoptosis induction in adenosine-treated colon cancer cells,
which included increased expression of TNFR1. TNFα secretion increased in acute myeloid leukemia cells after treatment
with SMAC mimetic and IFNα combination [125]. Inhibition
of TNFα and TNFR1 by a pharmacological inhibitor and
genetic silencing, respectively, reduced SMAC mimetic/
IFNα-triggered apoptosis. Tao et al. suggested that survivin,
an inhibitor of apoptosis, inhibitor induced programmed cell
death in Wilms tumor cells by increasing expression of
TNFR1 signaling [126].
As well as TNFα, lymphotoxin α (LTα) which is secreted
as a homotrimer binds to TNRF1 [127]. Even though both
TNFα and LTα bind signal via TNFR1, LTα was found to
possess less ability to induce TNRF-mediated cell death in an
early study [128]. However, a recent research by Etemadi et al.
displayed that LTα has same potential to induce apoptosis via
TNFR1 signaling [129].
Genetic changes in the promoter region of TNFα have
been well studied in several human diseases to find correlation
between apoptosis induction and TNFα expression. De
Oliveria et al. studied the effect of a polymorphism, TNF-α857 C/T, on gastric cancer patients, found that this mutation
decreased mRNA level of TNFα resulting in resistance to
apoptosis [130].
Death receptor 3
Death receptor 3, also known as APO-3, has four characteristic cysteine-rich motifs with molecular weight of 53.5 kDa.
Death receptor 3 (DR3) gene is localized on chromosome 1
Tumor Biol. (2016) 37:8471–8486
and encodes a type II trans-membrane protein. Similar to other
TNFR family members, DR3 has a DD in its cytoplasmic part
and initiates apoptotic signaling [87].
Most of the studies have been related to DR3 involvement
in immune system modulation due to its frequent expression
on lymphoid tissues such as the spleen, thymus, and peripheral blood lymphocytes [131]. The role of DR3 in the development of some human malignancies has also been enlightened. DR3 expression was shown to be increased in human
colon cancer cells treated with cordycepin, a deoxy form of
adenosine, resulting in apoptosis induction [132]. A specific
phenolic compound triggered apoptosis in human non-smallcell lung cancer (NSCLC) cells by increasing the expression
of DR3 [133]. Silencing of DR3 via siRNA approach reversed
its growth inhibitory effect.
p53
p53 is encoded by human TP53 gene localized on the short
arm of chromosome 17 with a molecular mass of 43.7 kDa
[134]. It occupies 19.200 bp including 11 exons. There are
various p53 isoforms based on alternative splicing of TP53
gene; some of which play opposite roles as compared to p53,
while others have similar functions like full-length p53 [135].
Human p53 protein is composed of the following three
different domains with important functions: the DNAbinding domain, the N-terminal trans-activational domain,
and the C-terminal oligomerization domain [136]. DNAbinding domain binds to response elements of target genes,
whereas the N-terminal trans-activation domain forms binding
sites for several negative or positive regulators. The Cterminal domain undergoes alternative splicing and posttranslational modifications [137].
p53 acts as tetrameric transcription factor, which controls
the expression of a large set of genes involved in significant
cellular processes including DNA damage detection, cell cycle arrest, apoptosis, DNA repair, and senescence [134]. It is
both involved in intrinsic and extrinsic pathways of apoptosis
by inducing transcription of several proteins like PUMA, Bid,
Bax, TRAILR2, and CD95, which is called transcriptiondependent apoptotic pathway of p53 taking place in the nucleus [138]. In this pathway of p53-dependent apoptosis, the
trans-activation domain of p53 interacts with the players of
basal transcription machinery such as the transcriptional coactivator p300/CBP [139]. In normal healthy cells, p53 levels
are very low due to its rapid turnover; however, if a damage is
detected in the cells, p53 becomes stabilized resulting increased p53 level [140]. p53 stability is controlled by mouse
double minute 2 (MDM2) gene, which is an E3 ubiquitin
ligase that negatively regulates p53 stability through
ubiquitination, thus proteosomal degradation. MDM2 also inhibits the interaction between the p53 trans-activation domain
and the components of transcription machinery [141]. MDM2
Tumor Biol. (2016) 37:8471–8486
has been found to be overexpressed in many cancer cells,
which leads to neutralization of interaction between p53 and
transcription machinery components, thus impairing
transcription-dependent apoptosis [142, 143].
p53 has the ability to activate intrinsic pathway of apoptosis by inducing the transcription of especially apoptotic Bcl-2
family genes such as PUMA [144]. p53 induces PUMA
mRNA expression immediately in response to DNA damage
by binding the two p53-responsive elements in the PUMA
promoter. p53 binding results in the acetylation of core histones, H3 and H4, which is responsible for chromosome
decondensation and transcriptional activation [145].
Similarly, TRAIL-R2 expression is induced by p53, which
binds to a p53-responsive element in TRAIL-R2 promoter in
order to induce extrinsic pathway of apoptosis [140]. As an
alternative mechanism, p53 functions a transcriptional repressor of certain anti-apoptotic genes including survivin which
promotes caspase activation [146]. p53 is also directly involved in apoptosome formation by activating transcription
of Apaf-1 gene including a p53 response element in its promoter [54]. In response to DNA damage, p53 is also displayed
to activate caspase-6 cleaving nuclear envelope protein lamin
A and various transcription factors through a response element
within the third intron of the gene [147].
As well as transcription-dependent functions of p53, its
transcription-independent functions in terms of apoptosis have
been defined. p53 induces apoptosis by acting directly at mitochondria. p53 trans-locates to the mitochondria in response
to apoptotic signal, where it forms inhibitory complexes with
Bcl-XL and Bcl-2 causing the permeabilization of the mitochondrial membrane and cytochrome c release [148]. The
interaction between p53 and Bcl-XL and Bcl-2 is mediated
by p53 trans-activation domain like p300/CBP binding, even
though these two modes of p53-dependent apoptosis induction occur in different cellular compartments [149]. Moreover,
cytosolic p53 might induce the activation of pro-apoptotic
Bax via direct protein-protein interactions [150]. Leu et al.
displayed that p53 interacts with pro-apoptotic mitochondrial
membrane protein Bak, which makes Bak undergo oligomerization and releases cytochrome c from mitochondria. Binding
of p53 to Bak damages interaction between Bak and antiapoptotic Mcl-1 [151].
Mutations in p53 gene have been considered most common
genetic changes in cancer. Mutant p53 proteins can both lose
their native tumor suppressor activity and provide active tumor development [152]. Missense mutations form the majority of alterations in p53 gene, which commonly occur in the
DNA binding domain of p53, thus preventing p53 from promoting target gene expression [153]. Recently, Saleem et al.
analyzed loss of function mutations in p53 gene in the patients
of oral squamous cell carcinoma and found that AGT to ACT
missense mutation in DNA binding domain may result in
impaired p53 function. In chronic lymphocytic leukemia
8479
patients, missense mutations in DNA binding motif were correlated with poor survival [154, 155].
p53 has been considered as a significant player of apoptosis
in many studies, and there is a growing accumulation of articles revealing its role in many malignancies. In a recent study
by Wang et al., acute pro-myelocytic leukemia cells were subjected to apoptosis when treated with combination of
tetraarsenictetrasulfide and arsenic trioxide through upregulation of p53 and its target gene Bax. Transcription-independent
role of p53 in apoptosis induction was identified in a study in
which p53 trans-located to mitochondria and induced mitochondrial membrane depolarization in HUVEC cells exposed
to heat stress [156, 157].
There is an increasing attention to develop different strategies that can modulate p53-dependent apoptotic pathways
such as inhibition of p53-MDM2 interaction using MDM2
inhibitors, restoring mutated p53 back to its wild-type form
and p53 vaccines [158–160].
MicroRNAs in extrinsic and intrinsic apoptotic pathways
Changes in the apoptotic response in cancer can result in tumor initiation, progression, and treatment resistance [3]. There
are numerous studies including the roles of microRNAs in the
control of apoptosis, and these microRNAs display their effects by directly targeting genes involved in both extrinsic and
intrinsic pathways of apoptosis [161]. These microRNAs
(miRNAs) can be classified as oncogenic and tumor suppressive miRNAs [162]. One of these miRNAs is miR-130a that
was found to reduce drug resistance in non-small-cell lung
cancer by targeting MET proto-oncogene and to sensitize this
cancer cells to TRAIL-induced apoptosis by inhibiting miR221 and miR-222, which are upregulated by MET and involved in TRAIL resistance [163]. Therefore, miR-221 and
miR-222 are oncogenic miRNAs that induce drug resistance
and block apoptosis in several cancer types such as gliomas
[164]. Another oncogenic miRNA exerting its anti-apoptotic
effects by inhibiting FasL directly is miR-21, which is found
to be upregulated in advanced pancreatic cancer patients
[165]. miR-24 regulates apoptosis by binding to coding sequence of Fas-associated factor 1 (FAF1) mRNA and induced
apoptosis of several different types of cancer [166]. miR-21
also negatively regulates PTEN, a tumor suppressor gene,
involved in the apoptotic pathway through the formation of
DISC complex in many tumors such as breast and gastric
cancers [167, 168]. In gastric cells, miR-21 upregulated and
decreased PTEN expression, resulting in significant suppression of trastuzumab-induced apoptosis [168]. miR-200c sensitized cells to apoptosis by directly targeting Fas-associated
phosphatase-1 (FAP-1), which is an inhibitor of Fas-induced
apoptosis [169]. miRNA-886-5p inhibited apoptosis of human cervical cancer cells by downregulating Bax [170].
Zhou et al. (2010) [171] found that miR-125b was upregulated
8480
in taxol-resistant breast cancer cells and suppressed apoptosis.
Bak1, pro-apoptotic Bcl-2 antagonist killer 1, was identified
as the direct target of miR-125b. miR-25 and miR-32 could
directly target the pro-apoptotic function of Bim, therefore
suppressing apoptosis in ovarian and human myeloid leukemia cells, respectively [172, 173]. In addition, miR-483-3p
might target PUMA, whose enforced expression protects cells
from apoptosis [174]. miR-34 decreases the expression of Bcl2, resulting in an increase in apoptosis [175]. In this study,
downregulation of miR-34 induced cell proliferation and invasion in malignant mesothelioma. miR-15b/16 could downregulate Bcl-2, thereby triggering apoptosis [176]. miR-153
induced apoptosis of glioblastoma cells by targeting 3′-UTR
of Bcl-2 and Mcl-1 [177]. miR-491 could induce apoptosis in
colorectal cancer cells by downregulating anti-apoptotic BclXL [178]. There are several studies showing the roles of
miRNAs in the regulation of caspase expression. miR-23a
and miR-24a blocked mitochondrial apoptosis by inhibiting
the expression of caspase-9 [179, 180]. The downregulation of
miR-23a increased the 5-FU-induced apoptosis in colon cancer cells [179]. Wu et al. (2014) [181] displayed that miR-421
upregulated in human gastric cell lines and tissues. miR-421
downregulated the expression of caspase-3 and blocked the
apoptosis of cancer cells. miR-106b-25 was shown to be upregulated in human prostate cancer and functions by partly
inhibiting of caspase-7 expression [182]. Floyd et al. (2014)
[183] demonstrated that miR-582-5p and miR-363 inhibit apoptosis by directly targeting caspase-3 and caspase-9 in glioblastoma. Caspase-3 has been shown to be a target of miR let7a in human squamous carcinoma cells and hepatocellular
carcinoma cells [184]. Curcumin induced apoptosis of
Fig. 2 miRNAs involved in the
apoptotic pathways. Some of the
miRNAs can inhibit apoptosis by
targeting the death-receptor
pathway including miR-21,
miR-24, and miR-200c. In the
mitochondrial pathway, various
miRNAs could target Bcl-2
family proteins and caspases
including miRNA-886-5p,
miR-125b, miR-25, miR-32,
miR-483-3p, miR-34, miR-15b/
16, miR-153, miR-491, miR-23a
and miR-24a, miR-421,
miR-106b-25, miR-582-5p and
miR-363, miR let-7a, and
miR-186
Tumor Biol. (2016) 37:8471–8486
NSCLC by the downregulation of miR-186, whose direct target is caspase-10 [185]. Based on all this evidence discussed
in this particular review, miRNAs play key regulatory roles in
apoptosis and could be important therapeutic targets in cancer
(Fig. 2).
Bioactive sphingolipids in apoptotic pathways
Bioactive sphingolipids are a family of membrane lipids that
have many regulatory roles in several cellular events such as
cell proliferation, senescence, adhesion, migration, and also
apoptosis [186]. Ceramide, the central molecule of bioactive
sphingolipid metabolism, was reported as a regulator of apoptosis in many studies. Treatment of cancer cells with radiation and chemotherapeutics such as vincristine, daunorubicin,
gemcitabine, and etoposide result in ceramide accumulation in
the cells as a secondary effect of these therapeutics [187].
Decreased ceramide levels result in the development of drug
resistance in cancer cells [188]. In vitro studies indicated that
treatment of cancer cells with ceramide triggers the release of
cytochrome-c from the mitochondria [189]. Additionally, it
was reported that anti-apoptotic Bcl-2 blocks ceramide channels in an independent manner from Bak and Bax [190].
Furthermore, ceramide also induces Bax-mediated apoptosis
in several types of cancer including, breast, prostate, and colon
cancers [191]. Ceramide was reported that it activates cathepsin D, an inducer of apoptosis in a form of lysosomal aspartyl
protease, in response to gemcitabine treatment [192]. Unlike
ceramide, the other member of sphingolipid family, sphingosine 1-P (S1P) is related to cell proliferation, survival, and also
inhibition of apoptosis [193]. S1P was reported to cause
Tumor Biol. (2016) 37:8471–8486
angiogenesis by VEGF signaling, which then triggers RAS
and MAPK signaling in cancer cells. By this way, S1P causes
cytoskeleton reconstruction and apoptosis inhibition [194].
While increased ceramide levels cause induction of apoptosis,
increased S1P levels cause inhibition of apoptosis [195]. S1P
leads to drug resistance due to its anti-apoptotic function.
Sphingosine kinase-1 (SK1), which catalyzes the conversion
of apoptotic ceramide to anti-apoptotic S1P, was also reported
to decrease apoptotic effects of chemotherapeutic agents in
prostate cancer [196]. Like S1P, glucosylceramide (GC), the
other important member of sphingolipid family, is also known
as a powerful anti-apoptotic molecule [193].
Glucosylceramide synthase (GCS), the enzyme catalyzing
the conversion of apoptotic ceramide to anti-apoptotic GC,
was found to be increased in drug-resistant cancer cells
[193]. Treatment of pancreatic cancer cells with a ceramide
analogue causes the accumulation of ceramide in the mitochondria, and by this way, ceramide reduces drug resistance
and triggers apoptosis [197]. Additionally, treatment of pancreatic cancer cells with ceramide in combination with a GCS
inhibitor, PDMP, decreases tumor growth in vivo [198].
Moreover, GC degradation increases ceramide generation
resulting in the trigger of apoptosis and also reduced tumor
growth in melanoma xenografts [199]. Reduced tumor growth
was also reported in mouse models with breast cancer treated
with C6:ceramide [200]. Furthermore, a type of sphingosine
kinase inhibitor, SK2, was reported to trigger apoptosis, inhibit cell proliferation, and decrease tumor size in mouse models
bearing hepatoma, mammary adenocarcinoma, and kidney
carcinoma [201, 202]. Safingol, the first agent used in the
clinic due to its ability in sphingosine kinase inhibition, increases apoptotic effects of chemotherapeutics used in cancer
therapy [203]. An FDA-approved drug that has inhibitory effects on SK1 and SK2 activities, FTY720, inhibits cell proliferation and leads to apoptosis in mice bearing breast cancer or
melanoma [204]. In chronic myeloid leukemia (CML) cell
lines, increase in serine palmitoyltransferase levels via BCR/
ABL inhibition result in activation of apoptotic signals [205].
GCS inhibition in CML cell lines bearing T315I mutation
results in apoptosis via activating GSK-3 [206]. Another study
reported that ceramide triggers apoptosis via activating p38,
caspase-8, and c-Jun N-terminal kinase (JNK) in K562 CML
cells [207]. Many studies from our laboratory also showed
effects of bioactive sphingolipids on apoptosis. In K562
CML cell lines resistant to nilotinib, apoptotic ceramide
synthase-1 and Bax genes were found to be downregulated
while anti-apoptotic SK1 and GCS genes were overexpressed,
and inhibition of these overexpressed genes sensitized the
cells against nilotinib treatment [208]. Our studies also
showed that when we overexpress GCS in imatinib-sensitive
K562 cells, the cells developed resistance against the drug via
inhibiting apoptosis. Inhibition of GCS and SK1 triggered
apoptosis via leading to ceramide accumulation in imatinib-
8481
resistant and imatinib-sensitive K562 CML cell lines [209].
Additionally, when we treated K562 human CML cell lines
and HL60 human acute pro-myelocytic leukemia cell lines
with resveratrol in combination with ceramide analog,
GCS inhibitor, or SK1 inhibitor, apoptotic effects of resveratrol increased synergistically in the cells treated with
resveratrol and ceramide combination, while the cells treated with resveratrol and GCS or SK1 combinations resisted
to apoptosis as compared to the control group [210, 211].
Furthermore, our studies also showed that GCS and SK1
inhibition in combination with nilotinib or dasatinib treatment synergistically triggers apoptosis in K562 and Meg01
human CML cell lines [212, 213].
Briefly, while ceramide is a powerful apoptotic molecule,
glucosylceramide and S1P generated from ceramide by GCS
and SK1 activities, respectively, are powerful anti-apoptotic
molecules, and therefore, alterations in intracellular levels of
these sphingolipids could be a novel approach for cancer
therapy.
Summary and perspectives
Apoptosis is highly regulated way of cell death, which is
crucial for all higher-level organisms to balance tissues
homeostasis and control cell proliferation as well as remove damaged or unnecessary cells. Apoptosis has its
own morphological and biochemical properties where
caspases play a central role at the end. Here, we have focused on aspects of apoptosis in terms of critical genes and
their products together with their role in both intrinsic and
extrinsic pathways of apoptosis. In this specific vital process, diverse groups of molecules function compatibly for
strict regulation. Any alterations or abnormalities occurring in apoptotic processes contribute to development of
human diseases and malignancies especially cancer. Deep
understanding of apoptotic signaling mechanisms, individual players, and genes involved in apoptosis have provided
a great opportunity to develop novel agents that make apoptosis deficient cells sensitive to apoptosis.
Compliance with ethical standards
Conflict of interest None
References
1.
2.
Elmore S. Apoptosis: a review of programmed cell death. Toxicol
Pathol. 2013;35:495–516.
Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological
phenomenon with wide-ranging implications in tissue kinetics.
Br J Cancer. 1972;26:239–57.
8482
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
Tumor Biol. (2016) 37:8471–8486
Lowe SW, Lin AW. Apoptosis in cancer. Carcinogenesis.
2000;21:485–95.
Horvitz HR. Genetic control of programmed cell death in the
nematode Caenorhabditis elegans. Cancer Res. 1999;59:1701–6.
Thompson CB. Apoptosis in the pathogenesis and treatment of
disease. Science. 1995;10:1456–62.
Krauss G. Biochemistry of signal transduction and regulation. Ed:
VCH Wiley, 3rd Edition. 2003;511–531.
Power C, Fanning N, Redmond HP. Cellular apoptosis and organ
injury in sepsis: a review. Shoch. 2002;18:197–211.
Savill J, Fadok V. Corpse clearance defines the meaning of cell
death. Nature. 2000;407:784–8.
Rosenblatt J, Raff MC, Cramer LP. An epithelial cell destined for
apoptosis signals its neighbors to extrude it by an actin- and
myosin-dependent mechanism. Curr Biol. 2011;11:1847–57.
Ferri KF, Kroemer G. Organelle-specific initiation of cell death
pathways. Nat Cell Biol. 2001;3:255–63.
Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview
of cell death. Am J Pathol. 1995;146:3–15.
Liu X, Li P, Widlak P, et al. The 40-kDa subunit of DNA fragmentation factor induces DNA fragmentation and chromatin condensation during apoptosis. Proc Natl Acad Sci U S A. 1998;95:
8461–6.
Grimsley C, Ravichandran KS. Cues for apoptotic cell engulfment: eat-me, don’t-eat-me and come-get-me signals. Trends
Cell Biol. 2003;13:648–56.
Mashima T, Naito M, Noguchi K, Miller DK, Nicholson DW,
Tsuruo T. Actin cleavage by CPP-32/apopain during the development of apoptosis. Oncogene. 1997;14:1007–12.
Ziegler U, Groscurth P. Morphological features of cell death.
Physiology. 2004;10:124–8.
Wang X. The expanding role of mitochondria in apoptosis. Genes
Dev. 2001;15:2922–33.
Thornberry NA, Lazebnik Y. Caspases: enemies within. Science.
1998;281:1312–6.
Hengartnere MO. The biochemistry of apoptosis. Nature.
2000;407:770–6.
Ghobrial IM, Witzig TE, Adjei AA. Targeting apoptosis pathways
in cancer therapy. CA Cancer J Clin. 2005;55:178–94.
Saelens X, Festjens N, Vande Walle L, van Gurp M, van Loo G,
Vandenabeele P. Toxic proteins released from mitochondria in cell
death. Oncogene. 2004;23:2861–74.
Du C, Fang M, Li Y, Li L, Wang X. SMAC, a mitochondrial
protein that promotes cytochrome c-dependent caspase activation
by eliminating IAP inhibition. Cell. 2000;102:33–42.
Chinnaiyan AM. The apoptosome: heart and soul of the cell death
machine. Neoplasia. 1999;1:5–15.
Hill MM, Adrain C, Duriez PJ, Creagh EM, Martin SJ. Analysis of
the composition, assembly kinetics and activity of native Apaf-1
apoptosomes. Embo J. 2004;23:2134–45.
Susin SA, Daugas E, Ravagnan L, et al. Two distinct pathways
leading to nuclear apoptosis. J Exp Med. 2000;192:571–80.
Cory S, Adams JM. The Bcl2 family: regulators of the cellular
life-or-death switch. Nat Rev Cancer. 2002;2:647–56.
Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell. 2001;104:
487–501.
Hsu H, Xiong J, Goeddel DV. The TNF receptor 1-associated
protein TRADD signals cell death and NF-kappa B activation.
Cell. 1995;81:495–50.
Wajant H. The Fas signaling pathway: more than a paradigm.
Science. 2002;296:1635–6.
Sax JK, Fei P, Murphy ME, Bernhard E, Korsmeyer SJ, El-Deiry
WS. BID regulation by p53 contributes to chemosensitivity. Nat
Cell Biol. 2002;4:842–9.
30.
Hitoshi Y, Lorens J, Kitada SI, et al. Toso, a cell surface, specific
regulator of Fas-induced apoptosis in T cells. Immunity. 1998;8:
461–71.
31. Scaffidi C, Schmitz I, Krammer PH, Peter ME. The role of c-FLIP
in modulation of CD95-induced apoptosis. J Biol Chem.
1999;274:1541–8.
32. Kuranaga E. Beyond apoptosis: caspase regulatory mechanisms
and functions in vivo. Genes Cells. 2012;17:83–97.
33. Yuan J, Shaham S, Ledoux S, Ellis HM, The HHR. The C. elegans
cell death gene Ced-3 encodes a protein similar to mammalian
interleukin-1 beta-converting enzyme. Cell. 1993;75:641–52.
34. Ellis HM, Horvitz HR. Genetic control of programmed cell death
in the nematode C. elegans. Cell. 1986;44:817–29.
35. Shi Y. Mechanisms of caspase inhibition and activation during
apoptosis. Mol Cell. 2002;9:459–70.
36. Chowdhury I, Tharakan B, Bhat GK. Caspases—an update. Comp
Biochem Physiol. 2008;151:10–27.
37. Yan N, Shi Y. Mechanisms of apoptosis through structural biology. Annu Rev Cell Dev Biol. 2005;21:35–56.
38. Los M, Stroh C, Janicke RU, Schulze-Osthoff K. Caspases: more
than just killers? Trends Immunol. 2001;22:31–4.
39. Martinon F, Tschopp J. Inflammatory caspases: linking an intracellular innate immune system to autoinflammatory diseases. Cell.
2004;117:561–74.
40. Los M, van de Craen M, Penning CL, et al. Requirement of an
ICE/CED-3 protease for Fas/APO-1-1 mediated apoptosis.
Nature. 1995;37:81–3.
41. Fantuzzi G, Puren AJ, Harding MW, Livingston DJ, Dinarello
CA. Interleukin-18 regulation of interferon gamma production
and cell proliferation as shown in interleukin-1beta-converting
enzyme (caspase-1)-deficient mice. Blood. 1998;91:2118–25.
42. Vakifahmetoglu-Norberg H, Zhivotovsky B. The unpredictable
caspase-2: what can it do? Trends Cell Biol. 2010;20:150–9.
43. Paroni G, Henderson C, Schneider C, Brancolini C. Caspase-2induced apoptosis is dependent on caspase-9, but its processing
during UV- or tumor necrosis factor-dependent cell death requires
caspase-3. J Biol Chem. 2001;276:21907–15.
44. Van de Craen M, Declercq W. Van den brande I, Fiers W,
Vandenabeele P. The proteolytic procaspase activation network:
an in vitro analysis. Cell Death Differ. 1999;6:1117–24.
45. Tinel A, Tschopp J. The PIDDosome, a protein complex implicated in activation of caspase-2 in response to genotoxic stress.
Science. 2004;304:843–6.
46. Degterev A, Boyce M, Yuan J. A decade of caspases. Oncogene.
2003;22:8543–67.
47. Porter AG, Janicke RU. Emerging roles of caspase-3 in apoptosis.
Cell Death Differ. 1999;6:99–104.
48. Slee EA, Adrain C, Martin SJ. Executioner caspase-3, -6, and -7
perform distinct, non-redundant roles during the demolition phase
of apoptosis. J Biol Chem. 2001;276:7320–6.
49. Lakhani SA, Masud A, Kuida K, et al. Caspases 3 and 7: key
mediators of mitochondrial events of apoptosis. Science.
2006;311:847–51.
50. Lamkanfi M, Kanneganti TD. Caspase-7: a protease involved
in apoptosis and inflammation. Int J Biochem Cell Biol.
2010;42:21–4.
51. Ghavami S, Eshraghi M, Kadkhoda K, et al. Role of BNIP3 in
TNF-induced cell death—TNF upregulates BNIP3 expression.
Biochim Biophys Acta. 1793;2009:546–60.
52. Carrington PE, Sandu C, Wei Y, et al. The structure of FADD
and its mode of interaction with procaspase-8. Mol Cell.
2006;22:599–610.
53. Ghavami S, Hashemi M, Ande SR, et al. Apoptosis and cancer:
mutations within caspase genes. J Med Genet. 2009;46:497–510.
Tumor Biol. (2016) 37:8471–8486
54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
Micheau O, Thome M, Schneider P, et al. Gr utter MG. The long
form of FLIP is an activator of caspase-8 at the Fas death-inducing
signaling complex. J Biol Chem. 2002;277:45162–71.
Irmler M, Thome M, Hahne M, et al. Inhibition of death receptors
signals by cellular FLIP. Nature. 1997;388:190–5.
Boatright KM, Deis C, Denault JB, Sutherlin DP, Salvesen GC.
Activation of caspases-8 and -10 by FLIP (L). Biochem J.
2004;382:651–7.
Pop C, Oberst A, Drag M, et al. FLIP (L) induces caspase 8
activity in the absence of interdomain caspase 8 cleavage and
alters substrate specificity. Biochem J. 2011;433:447–57.
Chang DW, Xing Z, Pan Y, et al. c-FLIP8(L) is a dual function
regulator for caspase-8 activation and CD95-mediated apoptosis.
EMBO J. 2002;21:3704–14.
Rodriguez J, Lazebnik Y. Caspase-9 and APAF-1 form an active
holoenzyme. Genes Dev. 1999;13:3179–84.
Qin H, Srinivasula SM, Wu G, Fernandes-Alnemri T, Alnemri ES,
Shi Y. Structural basis of procaspase-9 recruitment by the apoptotic protease-activating factor 1. Nature. 1999;399:549–57.
Crook NE, Clem RJ, Miller LK. An apoptosis-inhibiting
baculovirus gene with a zinc finger-like motif. J Virol. 1993;67:
2168–74.
Kaiser WJ, Vucic D, Miller LK. The Drosophila inhibitor of apoptosis D-IAP1 suppresses cell death induced by the caspase
drICE. FEBS Lett. 1998;440:243–8.
Eckelman BP, Salvesen GS. The human anti-apoptotic proteins
cIAP1 and cIAP2 bind but do not inhibit caspases. J Biol Chem.
2006;281:3254–60.
Ambrosini G, Adida C, Altieri DC. A novel anti-apoptosis gene,
survivin, expressed in cancer and lymphoma. Nat Med. 1997;3:
917–22.
Banks DP, Plescia J, Altieri DC, et al. Survivin does not inhibit
caspase-3 activity. Blood. 2000;96:4002–3.
Saleem M, Qadir MI, Perveen N, et al. Inhibitors of apoptotic
proteins: new targets for anticancer therapy. Chem Biol Drug
Des. 2013;82:243–51.
Sun C, Cai M, Gunasekera AH, et al. NMR structure and mutagenesis of the inhibitor-of-apoptosis protein XIAP. Nature.
1999;401:818–21.
Sun CH, Cai ML, Meadows RP, et al. NMR structure and mutagenesis of the third Bir domain of the inhibitor of apoptosis protein
XIAP. J Biol Chem. 2000;275:33777–81.
Vaux DL, Silke J. IAPs, RINGs and ubiquitylation. Nat Rev Mol
Cell Biol. 2005;6:287–97.
Salvesen GS, Duckett CS. IAP proteins: blocking the road to
death’s door. Nat Rev Mol Cell Biol. 2000;3:401–10.
Chai J, Du C, Wu JW, Kyin S, Wang X, Shi Y. Structural and
biochemical basis of apoptotic activation by SMAC/DIABLO.
Nature. 2000;406:855–62.
Srinivasula SM, Hegde R, Saleh A, et al. A conserved XIAPinteraction motif in caspase-9 and SMAC/DIABLO regulates caspase activity and apoptosis. Nature. 2001;410:112–6.
Yoo NJ, Kim HS, Kim SY, et al. Immunohistochemical analysis of
SMAC/DIABLO expression in human carcinomas and sarcomas.
APMIS. 2003;111:382–8.
Yang QH, Church-Hajduk R, Ren J, Newton ML, Du C. Omi/
HtrA2 catalytic cleavage of inhibitor of apoptosis [IAP] irreversibly inactivates IAPs and facilitates caspase activity in apoptosis.
Genes Dev. 2003;17:1487–96.
Liston P, Fong WG, Kelly NL, et al. Identification of XAF1 as an
antagonist of XIAP anti-caspase activity. Nat Cell Biol. 2001;3:
28–133.
Ma TL, Ni PH, Zhong J, Tan JH, Qiao MM, Jiang SH. Low
expression of XIAP- associated factor 1 in human colorectal cancers. Chin J Dig Dis. 2005;6:10–4.
8483
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
97.
98.
99.
Gross A, Mcdonnell JM, Korsmeyer SJ. Bcl-2 family members and the mitochondria in apoptosis. Genes Dev. 1999;13:
1899–911.
Czabotar PE, Lessene G, Strasser A, Adams JM. Control of apoptosis by the Bcl-2 protein family: implications for physiology and
therapy. Nat Rev Mol Cell Biol. 2014;5:49–63.
Pepper C, Bently P. The role of the Bcl-2 family in the modulation
of apoptosis. Symp Soc Exp Biol. 2000;52:43–53.
Green DR, Kroemer G. The pathophysiology of mitochondrial
cell death. Science. 2004;305:626–9.
Frenzel A, Grespi F, Chmelewskij W, Villunger A. Bcl2 family
proteins in carcinogenesis and the treatment of cancer. Apoptosis.
2009;14:584–96.
Camisasca DR, Honorato J, Bernardo V, et al. Expression of Bcl-2
family proteins and associated clinicopathologic factors predict
survival outcome in patients with oral squamous cell carcinoma.
Oral Oncol. 2009;45:225–33.
Kang MH, Reynolds CP. Bcl-2 inhibitors: targeting mitochondrial
apoptotic pathways in cancer therapy. Clin Cancer Res. 2009;15:
126–1132.
Lessene G, Czabotar PE, Colman PM. Bcl-2 family antagonists
for cancer therapy. Nat Rev Drug Discov. 2008;7:989–1000.
Hwang JJ, Kuruvilla J, Mendelson D, et al. Phase I dose finding
studies of obatoclax (GX15-070), a small molecule pan-Bcl-2
family antagonist, in patients with advanced solid tumors or lymphoma. Clin Cancer Res. 2010;16:4038–45.
Anderson MA, Huang D, Roberts A. Targeting Bcl2 for the
treatment of lymphoid malignancies. Semin Hematol.
2014;51:219–27.
Mahmood Z, Shukla Y. Death receptors: targets for cancer therapy.
Exp Cell Res. 2010;316:887–99.
Bhardwaj A, Aggarwal BB. Receptor-mediated choreography of
life and death. J Clin Immunol. 2003;23:317–32.
Ashkenazi A, Dixit VM. Death receptors: signaling and modulation. Sciences. 1998;281:1305–8.
Aggarwal BB, Gupta SC, Kim JH. Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden
journey. Blood. 2012;119:651–65.
Naismith JH, Sprang SR. Modularity in the TNF receptor family.
Trends Biochem Sci. 1998;23:74–9.
Fulda S, Debatin KM. Exploiting death receptor signaling
pathways for tumor therapy. Biochim Biophys Acta.
1705;2004:27–41.
Bremer E. Targeting of the tumor necrosis factor receptor superfamily for cancer immunotherapy. ISRN Oncol. 2013;2013:
371854.
Walczak H, Krammer PH. The CD95 (APO-1/Fas) and the
TRAIL (APO-2L) apoptosis systems. Exp Cell Res. 2000;256:
58–66.
Behrmann I, Walczak H, Krammer PH. Structure of the human
APO-1 gene. Eur J Immunol. 1994;24:3057–62.
Tauzin S, Debure L, Moreau JF, Legembre P. CD95-mediated cell
signaling in cancer: mutations and posttranslational modulations.
Cell Mol Life Sci. 2012;69:1261–77.
Scholl V, Stefanoff CG, Hassan R, Spector N, Renault IZ.
Mutations within the 5′ region of FAS/CD95 gene in nodal diffuse
large B-cell lymphoma. Leuk Lymphoma. 2007;48:957–63.
Ivanov VN, Ronai Z, Hei TK. Opposite roles of FAP-1 and
dynamin in the regulation of Fas (CD95) translocation to the cell
surface and susceptibility to Fas ligand-mediated apoptosis. J Biol
Chem. 2006;281:1840–52.
Tourneur L, Mistou S, Michiels FM, et al. Loss of FADD protein
expression results in a biased Fas-signaling pathway and correlates
with the development of tumoral status in thyroid follicular cells.
Oncogene. 2003;22:2795–280.
8484
100.
101.
102.
103.
104.
105.
106.
107.
108.
109.
110.
111.
112.
113.
114.
115.
116.
117.
118.
119.
Tumor Biol. (2016) 37:8471–8486
Fulda S, Kufer MU, Meyer E, van Valen F, Dockhorn-Dworniczak
B, Debatin KM. Sensitization for death receptor- or drug-induced
apoptosis by re-expression of caspase-8 through demethylation or
gene transfer. Oncogene. 2001;20:5865–77.
Yang T, Shi R, Chang L, et al. Huachansu suppresses human
bladder cancer cell growth through the Fas/Fasl and TNF-alpha/
TNFR1 pathway in vitro and in vivo. J Exp Clin Cancer Res.
2015;34:1–10.
Zhong W, Qin S, Zhu B, et al. Oxysterol-binding protein-related
protein 8 [ORP8] increases sensitivity of hepatocellular carcinoma
cells to Fas-mediated apoptosis. J Biol Chem. 2015;290:8876–87.
Pitti RM, Marsters SA, Ruppert S, Donahue CJ, Moore A,
Ashkenazi A. Induction of apoptosis by Apo-2 ligand, a new
member of the tumor necrosis factor cytokine family. J Biol
Chem. 1996;271:12687–90.
Mahalingam D, Szegezdi E, Keane M, de Jong S, Samali A.
TRAIL receptor signalling and modulation: are we on the right
TRAIL? Cancer Treat Rev. 2009;35:280–8.
Wu GS. TRAIL as a target in anti-cancer therapy. Cancer Lett.
2009;285:1–5.
O’Leary L, van der Sloot AM, Reis CR, et al. Decoy receptors
block TRAIL sensitivity at a supracellular level: the role of stromal
cells in controlling tumour TRAIL sensitivity. Oncogene. 2015.
doi:10.1038/onc.2015.180.
Woo JK, Kang JH, Jang YS, et al. Evaluation of preventive and
therapeutic activity of novel non-steroidal anti-inflammatory drug,
CG100649, in colon cancer: increased expression of TNF-related
apoptosis-inducing ligand receptors enhance the apoptotic response to combination treatment with TRAIL. Oncol Rep.
2015;3:1947–55.
Emery JG, McDonnell P, Burke MB, et al. Osteoprotegerin is a
receptor for the cytotoxic ligand TRAIL. J Biol Chem. 1998;273:
14363–7.
Weichhaus M, Chung ST, Connelly L. Osteoprotegerin in
breast cancer: beyond bone remodeling. Mol Cancer.
2015;14:117.
Lane D, Matte I, Rancourt C, Piché A. Osteoprotegerin [OPG]
protects ovarian cancer cells from TRAIL-induced apoptosis but
does not contribute to malignant ascites-mediated attenuation of
TRAIL-induced apoptosis. J Ovarian Res. 2012;5:34.
Merino D, Lalaoui N, Morizot A, Solary E, Micheau O. TRAIL in
cancer therapy: present and future challenges. Expert Opin Ther
Targets. 2007;11:1299–314.
Gong B, Almasan A. Genomic organization and transcriptional
regulation of human APO2/TRAIL gene. Biochem Biophys Res
Commun. 2000;278:747–52.
Krieg A, Krieg T, Wenzel M, et al. TRAIL-beta and TRAIL-gamma: two novel splice variants of the human TNF-related apoptosisinducing ligand (TRAIL) without apoptotic potential. Br J Cancer.
2003;88:918–27.
Pal R, Gochhait S, Chattopadhyay S, et al. Functional implication
of TRAIL-716 C/T promoter polymorphism on its in vitro and
in vivo expression and the susceptibility to sporadic breast tumor.
Breast Cancer Res Treat. 2012;126:333–43.
Bos PD, Zhang XHF, Nadal C, et al. Genes that mediate breast
cancer metastasis to the brain. Nature. 2009;459:1005–9.
Allen JE, El-Deiry WS. Regulation of the human TRAIL gene.
Cancer Biol Ther. 2012;13:1143–51.
Lim B, Allen JE, Prabhu VV, Talekar MK, Finnberg NK, El-Deiry
WS. Targeting TRAIL in the treatment of cancer: new developments. Expert Opin Ther Targets. 2015;25:1–15.
Falvo JV, Tsytsykova AV, Goldfeld AE. Transcriptional control of
the TNF gene. Curr Dir Autoimmun. 2010;11:27–60.
Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10:45–65.
120.
Sedger LM, McDermott MF. TNF and TNF-receptors: from mediators of cell death and inflammation to therapeutic giants—past,
present and future. Cytokine Growth Factor Rev. 2014;25:453–72.
121. Wachter T, Sprick M, Hausmann D, et al. cFLIPL inhibits tumor
necrosis factor-related apoptosis-inducing ligand-mediated NFkappaB activation at the death-inducing signaling complex in human keratinocytes. J Biol Chem. 2004;279:52824–34.
122. Ebach DR, Riehl TE, Stenson WF. Opposing effects of tumor
necrosis factor receptor 1 and 2 in sepsis due to cecal ligation
and puncture. Shock. 2005;23:311–8.
123. Yun HM, Park KR, Kim EC, Han SB, Yoon do Y, Hong JT. IL32α suppresses colorectal cancer development via TNFR1mediated death signaling. Oncotarget. 2015;6:9061–72.
124. Yu S, Hou D, Chen P, et al. Adenosine induces apoptosis through
TNFR1/RIPK1/P38 axis in colon cancer cells. Biochem Biophys
Res Commun. 2015;460:759–65.
125. Bake V, Roesler S, Eckhardt I, Belz K, Fulda S. Synergistic interaction of SMAC mimetic and IFNα to trigger apoptosis in acute
myeloid leukemia cells. Cancer Lett. 2014;355:224–31.
126. Tao YF, Lu J, Du XJ, et al. Survivin selective inhibitor YM155
induce apoptosis in SK-NEP-1 Wilms tumor cells. BMC Cancer.
2012;26(12):619.
127. Ruddle NH. Lymphotoxin and TNF: how it all began—a tribute to
the travelers. Cytokine Growth Factor Rev. 2014;25:83–9.
128. Chaturvedi MM, LaPushin R, Aggarwal BB. Tumor necrosis factor and lymphotoxin. Qualitative and quantitative differences in
the mediation of early and late cellular response. J Biol Chem.
1994;269:14575–83.
129. Etemadi N, Holien JK, Chau D, et al. Lymphotoxin α induces
apoptosis, necroptosis and inflammatory signals with the same
potency as tumour necrosis factor. FEBS J. 2013;280:5283–97.
130. de Oliveira JG, Rossi AF, Nizato DM, et al. Influence of functional
polymorphisms in TNF-α, IL-8, and IL-10 cytokine genes on
mRNA expression levels and risk of gastric cancer. Tumour
Biol. 2015 (Epub ahead of print).
131. Kang YJ, Kim WJ, Bae HU, et al. Involvement of TL1A and DR3
in induction of proinflammatory cytokines and matrix
metalloproteinase-9 in atherogenesis. Cytokine. 2005;29:229–35.
132. Lee SY, Debnath T, Kim SK, Lim BO. Anti-cancer effect and
apoptosis induction of cordycepin through DR3 pathway in the
human colonic cancer cell HT-29. Food Chem Toxicol. 2013;60:
439–47.
133. Oh SB, Hwang CJ, Song SY, et al. Anti-cancer effect of
tectochrysin in NSCLC cells through overexpression of death
receptor and inactivation of STAT3. Cancer Lett. 2014;353:
95–103.
134. Levine AJ, Oren M. The first 30 years of p53: growing ever more
complex. Nat Rev Cancer. 2009;9:749–58.
135. Surget S, Khoury MP, Bourdon J. Uncovering the role of p53
splice variants in human malignancy: a clinical perspective.
Onco Targets Ther. 2013;7:57–68.
136. Mollereau B, Ma D. The p53 control of apoptosis and proliferation: lessons from Drosophila. Apoptosis. 2014;19:1421–9.
137. Pflaum J, Schlosser S, Müller M. p53 family and cellular stress
responses in cancer. Front Oncol. 2014;4:285.
138. Beckerman R, Prives C. Transcriptional regulation by p53. Cold
Spring Harb Perspect Biol. 2010;2:a000935.
139. Chi SW. Structural insights into the transcription-independent apoptotic pathway of p53. BMB Rep. 2014;47:167–72.
140. Haupt S, Berger M, Goldberg Z, Haupt Y. Apoptosis—the p53
network. J Cell Sci. 2003;116:4077–85.
141. Perry ME. The regulation of the p53-mediated stress response by
MDM2 and MDM4. Cold Spring Harb Perspect Biol. 2010;2:
a000968.
142. Riley MF, You MJ, Multani AS, Lozano G. Mdm2 overexpression
and p73 loss exacerbate genomic instability and dampen
Tumor Biol. (2016) 37:8471–8486
143.
144.
145.
146.
147.
148.
149.
150.
151.
152.
153.
154.
155.
156.
157.
158.
159.
160.
161.
apoptosis, resulting in B-cell lymphoma. Oncogene. 2015. doi:10.
1038/onc.2015.88(Epubaheadofprint).
Jansson MD, Damas ND, Lees M, Jacobsen A, Lund AH. miR339-5p regulates the p53 tumor-suppressor pathway by targeting
MDM2. Oncogene. 2014;34:1908–18.
Yu J, Zhang L. PUMA, a potent killer with or without p53.
Oncogene Suppl. 1998;1:S71–83.
Hikisz P, Kiliańska ZM. PUMA, a critical mediator of cell death—
one decade on from its discovery. Cell Mol Biol Lett. 2012;17:
646–69.
Hoffman WH, Biade S, Zilfou JT, Chen J, Murphy M.
Transcriptional repression of the anti-apoptotic survivin gene by
wild type p53. J Biol Chem. 2002;277:3247–57.
MacLachlan TK, El-Deiry WS. Apoptotic threshold is lowered by
p53 transactivation of caspase-6. Proc Natl Acad Sci U S A.
2002;99:9492–7.
Mihara M, Erster S, Zaika A, et al. p53 has a direct apoptogenic
role at the mitochondria. Mol Cell. 2003;11:577–90.
Ha JH, Shin JS, Yoon MK, et al. Dual-site interactions of p53
protein transactivation domain with anti-apoptotic Bcl-2 family
proteins reveal a highly convergent mechanism of divergent p53
pathways. J Biol Chem. 2013;288:7387–98.
Chipuk JE, Kuwana T, Bouchier-Hayes L, et al. Direct activation
of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science. 2004;303:1010–4.
Leu JI, Dumont P, Hafey M, Murphy ME, George DL.
Mitochondrial p53 activates Bak and causes disruption of a
Bak-Mcl1complex. Nat Cell Biol. 2004;6:443–50.
Olivier M, Hollstein M, Hainaut P. TP53 mutations in human
cancers: origins, consequences, and clinicaluse. Cold Spring
Harb Perspect Biol. 2010;2:a001008.
Brosh R, Rotter V. When mutants gain new powers: news from the
mutant p53 field. Nat Rev Cancer. 2009;9:701–13.
Saleem S, Abbasi ZA, Hameed A, Qureshi NR, Khan MA, Azhar
A. Novel p53 codon 240 Ser > Thr coding region mutation in the
patients of oral squamous cell carcinoma (OSCC). Tumour Biol.
2014;35:7945–50.
Trbusek M, Smardova J, Malcikova J, et al. Missense mutations
located in structural p53 DNA-binding motifs are associated with
extremely poor survival in chronic lymphocytic leukemia. J Clin
Oncol. 2011;29:2703–8.
Wa n g S , Z h o u M , O u y a n g J , G e n g Z , Wa n g Z .
Tetraarsenictetrasulfide and arsenic trioxide exert synergistic effects on induction of apoptosis and differentiation in acute
promyelocytic leukemia cells. PLoS One. 2015;10:e0130343.
Gu ZT, Li L, Wu F, et al. Heat stress induced apoptosis is
triggered by transcription-independent p53, Ca[2+]
dyshomeostasis and the subsequent Bax mitochondrial translocation. Sci Rep. 2015;5:11497.
Sosin AM, Burger AM, Siddiqi A, Abrams J, Mohammad RM,
Al-Katib AM. HDM2 antagonist MI-219 [spiro-oxindole], but not
Nutlin-3 [cis-imidazoline], regulates p53 through enhanced
HDM2 autoubiquitination and degradation in human malignant
B-cell lymphomas. J Hematol Oncol. 2012;5:57.
Boeckler FM, Joerger AC, Jaggi G, Rutherford TJ, Veprintsev
DB, Fersht AR. Targeted rescue of a destabilized mutant of p53
by an in silico screened drug. Proc Natl Acad Sci U S A.
2008;105:10360–5.
Vermeij R, Leffers N, van der Burg SH, Melief CJ, Daemen T,
Nijman HW. Immunological and clinical effects of vaccines
targeting p53-overexpressing malignancies. J Biomed
Biotechnol. 2011;2011:702146.
Lima RT, Busacca S, Almeida GM, Gaudino G, Fennell DA,
Vasconcelos MH. MicroRNA regulation of core apoptosis pathways in cancer. Eur J Cancer. 2011;47:163–74.
8485
162.
Chen Y, Fu LL, Wen X, et al. Oncogenic and tumor suppressive
roles of microRNAs in apoptosis and autophagy. Apoptosis.
2014;19:1177–89.
163. Acunzo M, Visone R, Romano G, et al. Mir-130a targets MET and
induces trail-sensitivity in NSCLC by downregulating mir-221
and 222. Oncogene. 2012;31:634–42.
164. Hao J, Zhang C, Zhang A, et al. miR-221/222 is the regulator of
Cx43 expression in human glioblastoma cells. Oncol Rep.
2012;27:1504–10.
165. Wang P, Zhuang L, Zhang J, et al. The serum miR-21 level serves
as a predictor for the chemosensitivity of advanced pancreatic
cancer, and miR-21 expression confers chemoresistance by
targeting FasL. Mol Oncol. 2013;7:334–45.
166. Qin W, Shi Y, Zhao B, et al. miR-24 regulates apoptosis by
targeting the open reading frame (ORF) region of FAF1 in cancer
cells. PLoS ONE. 2010;5:e9429.
167. Satzger I, Mattern A, Kuettler U, et al. microRNA-21 is upregulated in malignant melanoma and influences apoptosis of
melanocytic cells. Exp Dermatol. 2012;21:509–14.
168. Eto K, Iwatsuki M, Watanabe M, et al. The microRNA-21/PTEN
pathway regulates the sensitivity of HER2-positive gastric cancer
cells to trastuzumab. Ann Surg Oncol. 2013;21:343–50.
169. Schickel R, Park SM, Murmann AE, Peter ME. miR-200c regulates induction of apoptosis through CD95 by targeting FAP-1.
Mol Cell. 2010;38:908–15.
170. Li JH, Xiao X, Zhang YN, et al. MicroRNA miR-886-5p inhibits
apoptosis by down-regulating Bax expression in human cervical
carcinoma cells. Gynecol Oncol. 2011;120:145–51.
171. Zhou M, Liu Z, Zhao Y, et al. MicroRNA-125b confers the resistance of breast cancer cells to paclitaxel through suppression of
pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) expression. J Biol
Chem. 2010;285:21496–507.
172. Zhang H, Zuo Z, Lu X, Wang L, Wang H, Zhu Z. MiR-25 regulates apoptosis by targeting Bim in human ovarian cancer. Oncol
Rep. 2012;27:594–8.
173. Gocek E, Wang X, Liu X, Liu CG, Studzinski GP. MicroRNA-32
upregulation by 1,25-dihydroxyvitamin D3 in human myeloid
leukemia cells leads to Bim targeting and inhibition of AraCinduced apoptosis. Cancer Res. 2011;71:6230–9.
174. Veronese A, Lupini L, Consiglio J, et al. Oncogenic role of miR483-3p at the IGF2/483 locus. Cancer Res. 2010;70:3140–9.
175. Tanaka N, Toyooka S, Soh J, et al. Downregulation of microRNA34 induces cell proliferation and invasion of human mesothelial
cells. Oncol Rep. 2013;29:2169–74.
176. Shen J, Wan R, Hu G, et al. miR-15b and miR-16 induce the
apoptosis of rat activated pancreatic stellate cells by targeting
Bcl-2 in vitro. Pancreatology. 2012;12:91–9.
177. Xu J, Liao X, Wong C. Downregulations of B-cell lymphoma 2
and myeloid cell leukemia sequence 1 by microRNA 153 induce
apoptosis in a glioblastoma cell line DBTRG-05MG. Int J Cancer.
2010;126:1029–35.
178. Nakano H, Miyazawa T, Kinoshita K, Yamada Y, Yoshida T.
Functional screening identifies a microRNA, miR-491 that induces apoptosis by targeting Bcl-X(L) in colorectal cancer cells.
Int J Cancer. 2010;127:1072–80.
179. Shang J, Yang F, Wang Y, et al. Sun S MicroRNA-23a antisense
enhances 5-fluorouracil chemosensitivity through APAF-1/caspase-9 apoptotic pathway in colorectal cancer cells. J Cell
Biochem. 2014;115:772–84.
180. Walker JC, Harland RM. MicroRNA-24a is required to repress
apoptosis in the developing neural retina. Genes Dev. 2009;23:
1046–51.
181. Wu JH, Yao YL, Gu T, et al. MiR-421 regulates apoptosis of
BGC-823 gastric cancer cells by targeting caspase-3. Asian Pac
J Cancer Prev. 2014;15:5463–8.
8486
182.
183.
184.
185.
186.
187.
188.
189.
190.
191.
192.
193.
194.
195.
196.
197.
198.
199.
Tumor Biol. (2016) 37:8471–8486
Hudson RS, Yi M, Esposito D, et al. Microrna-106b-25 cluster
expression is associated with early disease recurrence and targets
caspase-7 and focal adhesion in human prostate cancer. Oncogene.
2013;32:4139–47.
Floyd DH, Zhang Y, Dey BK, et al. Novel anti-apoptotic
microRNAs 582–5p and 363 promote human glioblastoma stem
cell survival via direct inhibition of caspase 3, caspase 9, and Bim.
PLoS One. 2014;9:e96239.
Tsang WP, Kwok TT. Let-7a microRNA suppresses therapeuticsinduced cancer cell death by targeting caspase-3. Apoptosis.
2008;13:1215–22.
Zhang J, Du Y, Wu C, et al. Curcumin promotes apoptosis in
human lung adenocarcinoma cells through miR-186 signaling
pathway. Oncol Rep. 2010;24:1217–23.
Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol.
2008;9:139–50.
Barth BM, Cabot MC, Kester M. Ceramide-based therapeutics for
the treatment of cancer. Anti Cancer Agents Med Chem. 2011;11:
911–9.
Senchenkov A, Litvak DA, Cabot MC. Targeting ceramide metabolism—a strategy for overcoming drug resistance. J Natl
Cancer Inst. 2001;93:347–57.
Siskind LJ, Kolesnick RN, Colombini M. Ceramide channels increase the permeability of the mitochondrial outer membrane to
small proteins. J Biol Chem. 2002;277:26796–803.
Siskind LJ, Feinstein L, Yu TX, et al. Anti-apoptotic Bcl-2 family
proteins disassemble ceramide channels. J Biol Chem. 2008;283:
6622–30.
von Haefen C, Wieder T, Gillissen B, et al. Ceramide induces
mitochondrial activation and apoptosis via a Bax-dependent pathway in human carcinoma cells. Oncogene. 2002;21:4009–19.
Dumitru CA, Sandalcioglu IE, Wagner M, Weller M, Gulbins E.
Lysosomal ceramide mediates gemcitabine-induced death of glioma cells. J Mol Med. 2009;87:1123–32.
Ogretmen B, Hannun YA. Biologically active sphingolipids in
cancer: pathogenesis and treatment. Nat Rev Cancer. 2004;4:
604–16.
Liu F, Verin AD, Wang P, et al. Differential regulation of
sphingosine-1-phosphate- and VEGF-induced endothelial cell
chemotaxis. Involvement of G(ialpha2)-linked Rho kinase activity. Am J Respir Cell Mol Biol. 2001;24:711–9.
Radin NS. The development of aggressive cancer: a possible role
for sphingolipids. Cancer Investig. 2002;20:779–86.
Pchejetski D, Golzio M, Bonhoure E, et al. Sphingosine kinase-1
as a chemotherapy sensor in prostate adenocarcinoma cell and
mouse models. Cancer Res. 2005;65:11667–75.
Beckham TH, Lu P, Jones EE, et al. LCL124, a cationic analog of
ceramide, selectively induces pancreatic cancer cell death by accumulating in mitochondria. J Pharmacol Exp Ther. 2013;344(1):
167–78.
Jiang Y, DiVittore NA, Kaiser JM, et al. Combinatorial therapies
improve the therapeutic efficacy of nanoliposomal ceramide for
pancreatic cancer. Cancer Biol Ther. 2011;12(7):574–85.
Sorli SC, Colié S, Albinet V, et al. The nonlysosomal βglucosidase GBA2 promotes endoplasmic reticulum stress and
200.
201.
202.
203.
204.
205.
206.
207.
208.
209.
210.
211.
212.
213.
impairs tumorigenicity of human melanoma cells. FASEB J.
2013;27(2):489–98.
Stover TC, Sharma A, Robertson GP, Kester M. Systemic delivery
of liposomal short-chain ceramide limits solid tumor growth in
murine models of breast adenocarcinoma. Clin Cancer Res.
2005;11(9):3465–74.
Beljanski V, Lewis CS, Smith CD. Antitumor activity of sphingosine kinase 2 inhibitor ABC294640 and sorafenib in hepatocellular carcinoma xenografts. Cancer Biol Ther. 2011;11(5):524–34.
Adan-Gokbulut A, Kartal-Yandim M, Iskender G, Baran Y. Novel
agents targeting bioactive sphingolipids for the treatment of cancer. Curr Med Chem. 2013;20(1):108–22.
Coward J, Ambrosini G, Musi E, Truman JP, Haimovitz-Friedman
A, Allegood JC. Safingol (L-threo-sphinganine) induces autophagy in solid tumor cells through inhibition of PKC and the PI3kinase pathway. Autophagy. 2009;5:184–93.
Pyne S, Bittman R, Pyne NJ. Sphingosine kinase inhibitors and
cancer: seeking the golden sword of Hercules. Cancer Res.
2011;71(21):6576–82.
Taouji S, Higa A, Delom F, et al. Phosphorylation of serine
palmitoyltransferase long chain-1 (SPTLC1) on tyrosine 164 inhibits its activity and promotes cell survival. J Biol Chem.
2013;288(24):17190–201.
Huang WC, Tsai CC, Chen CL, et al. Glucosylceramide synthase inhibitor PDMP sensitizes chronic myeloid leukemia
T315I mutant to Bcr-Abl inhibitor and cooperatively induces
glycogen synthase kinase-3-regulated apoptosis. FASEB J.
2011;25(10):3661–73.
Nica AF, Tsao CC, Watt JC, et al. Ceramide promotes apoptosis in chronic myelogenous leukemia-derived K562 cells by a
mechanism involving caspase-8 and JNK. Cell Cycle.
2008;7(21):3362–70.
Camgoz A, Gencer EB, Ural AU, Baran Y. Mechanisms responsible for nilotinib resistance in human chronic myeloid leukemia
cells and reversal of resistance. Leukemia Lymphoma. 2013;54:
1279–87.
Baran Y, Bielawski J, Gunduz U, Ogretmen B. Targeting
glucosylceramide synthase sensitizes imatinib-resistant chronic
myeloid leukemia cells via endogenous ceramide accumulation.
J Cancer Res Clin Oncol. 2011;137:1535–44.
Kartal M, Saydam G, Sahin F, Baran Y. Resveratrol triggers apoptosis through regulating ceramide metabolizing genes in human
K562 chronic myeloid leukemia cells. Nutr Cancer. 2011;63(4):
637–44.
Cakir Z, Saydam G, Sahin F, Baran Y. The roles of bioactive
sphingolipids in resveratrol-induced apoptosis in HL60: acute
myeloid leukemia cells. J Cancer Res Clin Oncol.
2011;137(2):279–86.
Camgoz A, Gencer EB, Ural AU, Avcu F, Baran Y. Roles of
ceramide synthase and ceramide clearence genes in nilotinibinduced cell death in chronic myeloid leukemia cells. Leukemia
Lymphoma. 2011;52:1574–84.
Gencer EB, Ural AU, Avcu F, Baran Y. A novel mechanism of
dasatinib-induced apoptosis in chronic myeloid leukemia; ceramide synthase and ceramide clearance genes. Ann Hematol.
2011;90:1265–75.
Downloaded on 8.4.2022 09:03:53
2016 Journal Performance Data for:
TUMOR BIOLOGY
ISSN
EISSN
1010-4283
N/A
JCR ABBREVIATION
ISO ABBREVIATION
TUMOR BIOL
Tumor Biol.
Journal Information
EDITION
CATEGORY
Science Citation Index
Expanded (SCIE)
ONCOLOGY - SCIE
LANGUAGES
REGION
1ST ELECTRONIC JCR YEAR
ENGLISH
SWITZERLAND
1997
PUBLISHER
ADDRESS
PUBLICATION FREQUENCY
SAGE PUBLICATIONS LTD
1 OLIVERS YARD, 55 CITY
ROAD, LONDON EC1Y 1SP,
ENGLAND
12 issues/year
Publisher Information
Journal Citation Reports ™
1-14
© 2022 Clarivate
Journal's Performance
Journal Impact Factor
The Journal Impact Factor (JIF) is a journal-level metric calculated from data indexed in the Web of
Science Core Collection. It should be used with careful attention to the many factors that influence
citation rates, such as the volume of publication and citations characteristics of the subject area and
type of journal. The Journal Impact Factor can complement expert opinion and informed peer review.
In the case of academic evaluation for tenure, it is inappropriate to use a journal-level metric as a
proxy measure for individual researchers, institutions, or articles.
2016 JOURNAL IMPACT FACTOR
2016 JOURNAL IMPACT FACTOR WITHOUT SELF CITATIONS
3.650
3.242
Journal Impact Factor Trend 2016
Journal Citation Reports ™
2-14
© 2022 Clarivate
Journal Impact Factor is calculated using the following metrics
Citations in 2016 to items published in 2014 (4,883) 2015 (4,608)
9,491
=
Number of citable items in 2014 (1,504) + 2015 (1,096)
=
3.650
=
3.242
2,600
Journal Impact Factor without self cites is calculated using the following metrics
Citations in 2016 to items published in 2014 (4,883) +
2015 (4,608) - Self Citations in 2016 to items published
in 2014 (545) + 2015 (516)
9,491 - 1,061
=
Number of citable items in 2014 (1,504) + 2015 (1,096)
Journal Citation Reports ™
3-14
2,600
© 2022 Clarivate
Total Citations
14,054
The total number of times that a journal has been cited by all journals included in the database in the
JCR year. Citations to journals listed in JCR are compiled annually from the JCR years combined
database, regardless of which JCR edition lists the journal.
Journal Citation Reports ™
4-14
© 2022 Clarivate
Rank by Journal Impact factor
Journals within a category are sorted in descending order by Journal Impact Factor (JIF) resulting in
the Category Ranking below. A separate rank is shown for each category in which the journal is listed
in JCR. Data for the most recent year is presented at the top of the list, with other years shown in
reverse chronological order.
EDITION
Science Citation Index Expanded (SCIE)
CATEGORY
ONCOLOGY
81/217
JCR
YEAR
JIF RANK
QUART JIF PERCENTILE
ILE
2020 n/a
n/a
n/a
2019 n/a
n/a
n/a
2018 n/a
n/a
n/a
2017 n/a
n/a
n/a
2016 81/217
Q2
62.90
2015 104/213
Q2
51.41
2014 69/211
Q2
67.54
2013 93/203
Q2
54.43
2012 102/197
Q3
48.48
2011 116/196
Q3
41.07
2010 117/185
Q3
37.03
2009 108/166
Q3
35.24
2008 91/143
Q3
36.71
2007 70/132
Q3
47.35
2006 63/127
Q2
50.79
2005 102/123
Q4
17.48
2004 59/123
Q2
52.44
2003 50/120
Q2
58.75
2002 74/114
Q3
35.53
2001 80/107
Q3
25.70
2000 52/103
Q3
50.00
1999 55/105
Q3
48.10
1998 50/104
Q2
52.40
1997 45/102
Q2
56.37
Journal Citation Reports ™
5-14
© 2022 Clarivate
Citation network
Cited Half-life
2.3 years
The Cited Half-Life is the median age of the items in this journal that were cited in the JCR year. Half
of a journal's cited items were published more recently than the cited half-life.
TOTAL NUMBER OF CITES
NON-SELF CITATIONS
SELF CITATIONS
14,054
12,684
1,370
# OF CITES FROM CUMULATIVE
2016
%
# OF CITING
SOURCES
14,054 citations
100.00%
1,645 sources
1,179 citations
8.39%
334 sources
4,608 citations
41.18%
872 sources
4,883 citations
75.92%
984 sources
1,437 citations
86.14%
495 sources
694 citations
91.08%
317 sources
356 citations
93.61%
209 sources
201 citations
95.04%
139 sources
49 citations
95.39%
38 sources
76 citations
95.93%
54 sources
30 citations
96.14%
27 sources
Previous years:
541 citations
Journal Citation Reports ™
6-14
© 2022 Clarivate
Citing titles in all years
TUMOR BIOLOGY
SOURCE NAME
COUNT
All Others
687
1
TUMOR BIOLOGY
1,370
2
Oncotarget
1,239
3
Scientific Reports
355
4
International Journal of Clinical and Experimental Pathology
351
5
ONCOLOGY REPORTS
323
6
PLoS One
277
7
International Journal of Clinical and Experimental Medicine
273
8
OncoTargets and Therapy
272
9
Oncology Letters
249
10
INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES
194
11
American Journal of Translational Research
181
12
WORLD JOURNAL OF GASTROENTEROLOGY
154
13
Molecular Medicine Reports
151
14
INTERNATIONAL JOURNAL OF ONCOLOGY
150
15
BIOMEDICINE & PHARMACOTHERAPY
132
16
European Review for Medical and Pharmacological Sciences
122
17
BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
110
18
Journal of Cancer
110
19
CELLULAR PHYSIOLOGY AND BIOCHEMISTRY
106
20
CANCER LETTERS
102
Showing 1 - 20 rows of 958 total (use export in the relevant section to download the full table)
Journal Citation Reports ™
7-14
© 2022 Clarivate
Citing Half-life
5.9 years
The Citing Half-Life is the median age of items in other publications cited by this journal in the JCR
year.
TOTAL NUMBER OF CITES
NON-SELF CITATIONS
SELF CITATIONS
67,981
66,611
1,370
# OF CITES FROM CUMULATIVE
2016
%
# OF CITED
SOURCES
67,981 citations
100.00%
4,308 sources
665 citations
0.98%
281 sources
7,424 citations
11.90%
1,265 sources
8,359 citations
24.20%
1,513 sources
6,873 citations
34.31%
1,342 sources
5,918 citations
43.02%
1,184 sources
5,206 citations
50.68%
1,090 sources
4,730 citations
57.64%
1,012 sources
3,997 citations
63.52%
859 sources
3,428 citations
68.56%
796 sources
3,028 citations
73.01%
715 sources
Previous years:
18,353 citations
Journal Citation Reports ™
8-14
© 2022 Clarivate
Cited titles in all years
TUMOR BIOLOGY
SOURCE NAME
COUNT
All Others
2,078
1
CANCER RESEARCH
2,347
2
PLoS One
1,927
3
ONCOGENE
1,434
4
TUMOR BIOLOGY
1,370
5
JOURNAL OF BIOLOGICAL CHEMISTRY
1,252
6
CLINICAL CANCER RESEARCH
1,173
7
CELL
1,066
8
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE
UNITED STATES OF AMERICA
1,061
9
NATURE
1,051
10
INTERNATIONAL JOURNAL OF CANCER
865
11
JOURNAL OF CLINICAL ONCOLOGY
847
12
Oncotarget
786
13
NATURE REVIEWS CANCER
750
14
BRITISH JOURNAL OF CANCER
700
15
CA-A CANCER JOURNAL FOR CLINICIANS
676
16
CANCER LETTERS
674
17
BLOOD
630
18
BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
569
19
BMC CANCER
534
20
ONCOLOGY REPORTS
523
Showing 1 - 20 rows of 2230 total (use export in the relevant section to download the full table)
Journal Citation Reports ™
9-14
© 2022 Clarivate
Content metrics
Source data
This tile shows the breakdown of document types published by the journal. Citable Items are Articles
and Reviews. For the purposes of calculating JIF, a JCR year considers the publications of that
journal in the two prior years.
1,625 total citable items
ARTICLES
REVIEWS
COMBINED (C)
OTHER
DOCUMENT
TYPES (O)
PERCENTAGE
NUMBER IN JCR
YEAR 2016 (A)
1,484
141
1,625
49
97%
NUMBER OF
REFERENCES (B)
52,963
14,941
67,904
77
100%
RATIO (B/A)
35.7
106.0
41.8
1.6
Average JIF Percentile
The Average Journal Impact Factor Percentile takes the sum of the JIF Percentile rank for each
category under consideration, then calculates the average of those values.
ALL CATEGORIES AVERAGE
EDITION
62.90
Science Citation Index Expanded
ONCOLOGY
62.90
Journal Citation Reports ™
10-14
© 2022 Clarivate
Additional metrics
Eigenfactor score
0.03054
The Eigenfactor Score is a reflection of the density of the network of citations around the journal
using 5 years of cited content as cited by the Current Year. It considers both the number of citations
and the source of those citations, so that highly cited sources will influence the network more than
less cited sources. The Eigenfactor calculation does not include journal self-citations.
Normalized Eigenfactor
3.49696
The Normalized Eigenfactor Score is the Eigenfactor score normalized, by rescaling the total number
of journals in the JCR each year, so that the average journal has a score of 1. Journals can then be
compared and influence measured by their score relative to 1.
Journal Citation Reports ™
11-14
© 2022 Clarivate
Article influence score
0.611
The Article Influence Score normalizes the Eigenfactor Score according to the cumulative size of the
cited journal across the prior five years. The mean Article Influence Score for each article is 1.00. A
score greater than 1.00 indicates that each article in the journal has above-average influence.
Journal Citation Reports ™
12-14
© 2022 Clarivate
5 year Impact Factor
3.445
The 5-year Impact Factor is the average number of times articles from the journal published in the
past five years have been cited in the JCR year. It is calculated by dividing the number of citations in
the JCR year by the total number of articles published in the five previous years.
5 year Impact Factor calculation
Citations in 2016 to items published in [2011-2015]
(11,978)
11,978
=
Number of citable items in [2011-2015] (3,477)
Journal Citation Reports ™
13-14
=
3.445
3,477
© 2022 Clarivate
Immediacy Index
0.726
The Immediacy Index is the count of citations in the current year to the journal that reference content
in this same year. Journals that have a consistently high Immediacy Index attract citations rapidly.
Immediacy Index calculation
Cites in 2016 to items published in 2016
1,179
1,179 / 1,625 = 0.726
Number of items published in 2016
Journal Citation Reports ™
1,625
14-14
© 2022 Clarivate