You are on page 1of 14

Oncogene (2006) 25, 47984811

& 2006 Nature Publishing Group All rights reserved 0950-9232/06 $30.00
www.nature.com/onc

REVIEW

Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy

S Fulda and K-M Debatin


University Childrens Hospital, Ulm, Germany

Apoptosis or programmed cell death is a key regulator 2000). Multiple stress-inducible molecules, for example,
of physiological growth control and regulation of tissue c-Jun N-terminal kinase (JNK), mitogen-activated
homeostasis. One of the most important advances in protein kinase (MAPK)/extracellular signal-regulated
cancer research in recent years is the recognition that protein kinase (ERK), nuclear factor kappa B (NF-kB)
cell death mostly by apoptosis is crucially involved in the or ceramide, have been implied in transmitting the
regulation of tumor formation and also critically deter- apoptotic signal (Davis, 2000; Karin et al., 2002).
mines treatment response. Killing of tumor cells by most Proteolytic enzymes such as caspases are important
anticancer strategies currently used in clinical oncology, effector molecules in apoptosis (Degterev et al., 2003).
for example, chemotherapy, c-irradiation, suicide gene Activation of caspases in response to anticancer
therapy or immunotherapy, has been linked to activation of chemotherapy can be initiated through activation of
apoptosis signal transduction pathways in cancer cells such the extrinsic (receptor) pathway or at the mitochondria
as the intrinsic and/or extrinsic pathway. Thus, failure to by stimulating the intrinsic pathway (Fulda and
undergo apoptosis may result in treatment resistance. Debatin, 2004). Because of the potential detrimental
Understanding the molecular events that regulate apop- effects on cell survival in case of inappropriate activa-
tosis in response to anticancer chemotherapy, and how tion of apoptosis programs, apoptosis pathways have to
cancer cells evade apoptotic death, provides novel oppor- be tightly controlled. The antiapoptotic mechanisms
tunities for a more rational approach to develop molecular- regulating cell death have also been implicated in
targeted therapies for combating cancer. conferring drug resistance to tumor cells (Fulda and
Oncogene (2006) 25, 47984811. doi:10.1038/sj.onc.1209608 Debatin, 2004). However, the concept that apoptosis
represents the major mechanism by which cancer cells
Keywords: apoptosis; death receptor; mitochondria; are eliminated may not universally apply and caspase-
cancer; chemotherapy independent apoptosis or other modes of cell death have
also to be considered as cellular response to anticancer
therapy (Brown and Wilson, 2003; Brown and Attardi,
2005). Thus, a better understanding of these diverse
modes of cell death in cancer therapy will provide a
Introduction molecular basis for new strategies targeting cell death
pathways in resistant forms of cancer.
Current cancer therapies, for example, chemotherapy,
g-irradiation, immunotherapy or suicide gene therapy,
primarily exert their antitumor effect by triggering Apoptosis signaling pathways
apoptosis in cancer cells (Makin and Dive, 2001; Fulda
and Debatin, 2004). Apoptosis is an evolutionary In most cases, anticancer therapies eventually result in
conserved, intrinsic program of cell death that occurs activation of caspases, a family of cysteine proteases
in various physiological and pathological situations that act as common death effector molecules in various
(Hengartner, 2000). Apoptosis is characterized by forms of cell death (Degterev et al., 2003). Caspases are
typical morphological and biochemical hallmarks, synthesized as inactive proforms and upon activation,
including cell shrinkage, nuclear DNA fragmentation they cleave next to aspartate residues (Degterev et al.,
and membrane blebbing (Hengartner, 2000). The under- 2003). The fact that caspases can activate each other by
lying mechanisms for initiation of an apoptosis response cleavage at identical sequences results in amplication
upon cytotoxic therapy may depend on the individual of caspase activity through a protease cascade (Degterev
stimulus and have often not exactly been identied. et al., 2003). Caspases cleave a number of different
However, damage to DNA or to other critical molecules substrates in the cytoplasm or nucleus leading to many
is considered to be a common initial event which is then of the morphologic features of apoptotic cell death
propagated by the cellular stress response (Rich et al., (Degterev et al., 2003). For example, polynucleosomal
DNA fragmentation is mediated by cleavage of ICAD
Correspondence: Dr S Fulda, University Childrens Hospital, Eythstr. (inhibitor of caspase-activated DNase), the inhibitor of
24, Ulm D-89075, Germany. the endonuclease CAD (caspase-activated DNase) that
E-mail: simone.fulda@uniklinik-ulm.de cleaves DNA into the characteristic oligomeric frag-
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4799
ments (Nagata, 2000). Likewise, proteolysis of several cing ligand (TRAIL) receptors results in activation of the
cytoskeletal proteins such as actin or fodrin leads to loss initiator caspase-8 which can propagate the apoptosis
of overall cell shape, whereas degradation of lamin signal by direct cleavage of downstream effector caspases
results in nuclear shrinking (Degterev et al., 2003). such as caspase-3 (Walczak and Krammer, 2000). The
Activation of caspases can be initiated from different mitochondrial pathway is initiated by the release of
entry points, for example, at the plasma membrane apoptogenic factors such as cytochrome c, apoptosis-
upon ligation of death receptor (receptor pathway) or at inducing factor (AIF), Smac (second mitochondria-
the mitochondria (mitochondrial pathway) (Hengartner, derived activator of caspase)/DIABLO (direct inhibitor
2000) (Figure 1). Stimulation of death receptors of the of apoptosis protein (IAP)-binding protein with low PI),
tumor necrosis factor (TNF) receptor superfamily such Omi/HtrA2 or endonuclease G from the mitochondrial
as CD95 (APO-1/Fas) or TNF-related apoptosis-indu- intermembrane space (Cande et al., 2002; Saelens et al.,
2004). The release of cytochrome c into the cytosol
triggers caspase-3 activation through formation of the
cytochrome c/Apaf-1/caspase-9-containing apoptosome
complex, whereas Smac/DIABLO and Omi/HtrA2
promote caspase activation through neutralizing the
inhibitory effects to the IAPs (Saelens et al., 2004).
Links between the receptor and the mitochondrial
pathway exist at different levels. Upon death receptor
triggering, activation of caspase-8 may result in cleavage
of Bid, a Bcl-2 family protein with a BH3 domain only,
which in turn translocates to mitochondria to release
cytochrome c thereby initiating a mitochondrial ampli-
cation loop (Cory and Adams, 2002). In addition,
cleavage of caspase-6 downstream of mitochondria may
feed back to the receptor pathway by cleaving caspase-8
(Cowling and Downward, 2002).

Nonapoptotic forms of cell death

Although apoptosis pathways and signal-transducing


molecules have been shown to play a crucial role for
killing of tumor cells in response to cytotoxic agents used
in the treatment of cancer patients, the signicance of
apoptosis as the main mechanism of cancer cell death
in response to anticancer chemotherapy has also been
challenged. So far, no clear pattern has emerged between
the level of apoptosis or proteins that regulate apoptosis
Figure 1 Apoptosis signaling pathways. Apoptosis pathways can and treatment response in most solid tumors. In addition
be initiated through different entry sites, for example, at the plasma to apoposis, cancer cells have also been shown to be
membrane by death receptor ligation (receptor pathway) or at the effectively eliminated after DNA damage by necrosis,
mitochondria (mitochondrial pathway). Stimulation of death mitotic catastrophe and autophagy (Brown and Wilson,
receptors of the tumor necrosis factor (TNF) receptor superfamily
such as CD95 (APO-1/Fas) or TNF-related apoptosis-inducing 2003; Brown and Attardi, 2005). Thus, nonapoptotic
ligand (TRAIL) receptors by CD95 ligand (CD95-L) or TRAIL modes of cell death have also been taken into consi-
results in receptor aggregation and recruitment of the adaptor deration as response to cytotoxic therapy. Although the
molecule Fas-associated death domain (FADD) and caspase-8. signaling pathways and molecules involved in these
Upon recruitment, caspase-8 becomes activated and initiates
apoptosis by direct cleavage of downstream effector caspases.
alternative forms of cell death have not yet exactly been
The mitochondrial pathway is initiated by stress signals through dened, non-caspase proteases such as calpains or
the release of apoptogenic factors such as cytochrome c, apoptosis cathepsins, lysosomal enzymes or PARP-1 may be
inducing factor (AIF), or Smac/DIABLO from the mitochondrial involved (Okada and Mak, 2004). The form of cell
intermembrane space. The release of cytochrome c into the cytosol death may depend on the context, including the cell type,
triggers caspase-3 activation through formation of the cytochrome
c/Apaf-1/caspase-9-containing apoptosome complex. The Smac/ the genotype of the cell, the type of DNA damage and
DIABLO promotes caspase activation through neutralizing the the dose of the agent used. Thus, the relative contribu-
inhibitory effects to IAPs, whereas AIF causes DNA condensation. tion of these different modes of cell death for chemo-
The receptor and the mitochondrial pathway can be interconnected responses in vitro and in vivo remains to be dened.
at different levels, for example, by Bid, a BH3 domain-containing
protein of the Bcl-2 family which assumes cytochrome-c-releasing
activity upon cleavage by caspase-8. Activation of caspases is Extrinsic pathway
negatively regulated at the receptor level by FLIP, which block
caspase-8 activation, at the mitochondria by Bcl-2 family proteins
and by inhibitor of apoptosis proteins (IAPs). See text for more Death receptors are members of the tumor necrosis
details. factor (TNF) receptor gene superfamily that consists of
Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4800
more than 20 proteins with a broad range of biological Signaling through CD95 or TRAIL receptors
functions, including regulation of cell death and Ligation of death receptors such as CD95 or the
survival, differentiation or immune regulation (Walczak agonistic TRAIL receptors TRAIL-R1 and TRAIL-
and Krammer, 2000; Ashkenazi, 2002). Members of R2 by their cognate ligands or agonistic antibodies
the TNF receptor family share similar, cysteine-rich results in receptor trimerization, clustering of the
extracellular domains. In addition, death receptors are receptors death domains and recruitment of adaptor
dened by a cytoplasmic domain of about 80 amino molecules such as Fas-associated death domain
acids called death domain, which plays a crucial role in (FADD) through homophilic interaction mediated by
transmitting the death signal from the cells surface to the death domain (Walczak and Krammer, 2000). Fas-
intracellular signaling pathways. The best-characterized associated death domain in turn recruits caspase-8 to the
death receptors include CD95 (APO-1/Fas), TNF activated CD95 receptor to form the CD95 death-
receptor 1 (TNFRI), TNF-related apoptosis-inducing inducing signaling complex (DISC) (Kischkel et al.,
ligand-receptor 1 (TRAIL-R1) and TRAIL-R2, whereas 1995). Oligomerization of caspase-8 upon DISC forma-
the role of DR3 (TRAMP/Apo-3/WSL-1/LARD) or tion drives its activation through self-cleavage. Caspase-
DR6 has not exactly been dened (Walczak and 8 then activates downstream effector caspases such as
Krammer, 2000). The corresponding ligands of the caspase-3. For the CD95 signaling pathway, two distinct
TNF superfamily comprise death receptor ligands such prototypic cell types have been identied (Scafdi et al.,
as CD95 ligand (CD95L), TNFa, lymphotoxin-a (the 1998). In type I cells, caspase-8 is activated upon death
later two bind to TNFRI), TRAIL and TWEAK, a receptor ligation at the DISC in quantities sufcient to
ligand for DR3 (Walczak and Krammer, 2000). directly activate downstream effector caspases such as
caspase-3 (Scafdi et al., 1998). In type II cells, however,
the amount of active caspase-8 generated at the DISC is
CD95 insufcient to activate caspase-3 and a mitochondrial
The CD95 receptor/CD95L system is a key signal amplication loop is required for full activation of
pathway involved in the regulation of apoptosis in caspases (Scafdi et al., 1998). Also, a similar cell type-
several different cell types, for example, in the immune dependent organization (type I and type II) of the
system (Krammer, 2000). CD95, a 48 kDa type I trans- TRAIL signaling pathway has been described (Fulda
membrane receptor, is expressed on activated lympho- et al., 2002a).
cytes, on a variety of tissues of lymphoid or non-
lymphoid origin, as well as on tumor cells (Krammer, Disruption of the extrinsic pathway in cancers
2000). CD95 ligand is produced by activated T cells Cancer cells have evolved numerous strategies to resist
and plays a crucial role in the regulation of the immune cell death induction via the extrinsic pathway. Signaling
system by triggering autocrine suicide or paracrine to cell death in response to death receptor ligation can
death in lymphocytes or other target cells (Krammer, principally be inhibited by an increase in antiapoptotic
2000). Furthermore, CD95L expression on cancer molecules or by a decrease or defective function in
cells has been implicated in immune escape of tumors proapoptotic proteins.
(Krammer, 2000). By constitutive expression of death For example, surface expression of death receptors
receptor ligands such as CD95L, tumors may adopt may vary between different cell types and can be
a killing mechanism from cytotoxic lymphocytes to downregulated or absent in resistant forms of tumors.
delete the attacking antitumor T cells through induction To this end, drug-resistant leukemia or neuroblastoma
of apoptosis via CD95/CD95L interaction. However, cells showed strong downregulation of CD95 expression
this model of tumor counterattack has also been suggesting that critical levels of CD95 expression may
challenged, since no study has so far conclusively have an impact on drug sensitivity (Friesen et al., 1997;
demonstrated that tumor counterattack is a relevant Fulda et al., 1998b). Also, decient transport of the
immune escape mechanism in vivo (Igney and Krammer, agonistic TRAIL receptors TRAIL-R1 and TRAIL-R2
2002). from intracellular stores, for example, endoplasmatic
reticulum, to the cell surface was identied as a
mechanism conferring TRAIL resistance in colon cancer
TNF-related apoptosis-inducing ligand and its receptors cells (Jin et al., 2004). Mutations of the CD95 gene have
TNF-related apoptosis-inducing ligand/Apo-2L was been identied in a variety of hematological malignan-
identied in 1995 based on its sequence homology to cies and solid tumors (Debatin et al., 2003). The
other members of the TNF superfamily and is consti- incidence of CD95 mutations found in human tumors
tutively expressed in a wide range of tissues (LeBlanc has been taken as evidence that the CD95 system exerts
and Ashkenazi, 2003). The two agonistic TRAIL recep- a tumor suppressor function. Moreover, decoy receptor
tors, TRAIL-R1 and TRAIL-R2, contain a conserved 3 (DcR3), which act as decoy receptors by competitively
cytoplasmic death domain motif, which enables them to binding CD95L, can interfere with CD95-triggered
engage the cells apoptotic machinery upon ligand apoptosis and was found to be genetically amplied or
binding, whereas TRAIL-R3 to R5 are antagonistic overexpressed in lung carcinoma, colon carcinoma or
decoy receptors, which bind TRAIL, but do not glioblastoma (Pitti et al., 1998; Roth et al., 2001). A
transmit a death signal (LeBlanc and Ashkenazi, 2003). decoy receptor for TRAIL, TRAIL-R3, was found to be

Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4801
overexpressed in gastric carcinoma (Sheikh et al., 1999). Signaling through the extrinsic pathway in cancer therapy
Moreover, loss of expression of the agonistic TRAIL The CD95 system has been implicated in chemotherapy-
receptors TRAIL-R1 and -R2 may account for TRAIL induced tumor cell death in a number of studies. To t
resistance. Both receptors are located on chromosome his end, treatment with anticancer drugs triggered an
8p, a region of frequent loss of heterozygosity (LOH) in increase in CD95L expression, which stimulated the
tumors (LeBlanc and Ashkenazi, 2003). In a small receptor pathway in an autocrine or paracrine manner
percentage of cancers, for example, non-Hodgkins by binding to its receptor CD95 (Friesen et al., 1996;
lymphoma, colorectal, breast, head and neck cancer, Fulda et al., 1997, 1998a; Houghton et al., 1997; Muller
osteosarcoma or lung carcinoma, deletions or muta- et al., 1997). Various anticancer agents with different
tions were found, which resulted in loss of both copies primary intracellular targets have been used in these
of TRAIL-R1 or R2 (Pai et al., 1998; Dechant et al., studies including DNA-damaging agents such as dox-
2004). orubicin, etoposide, cisplatin or bleomycin (Friesen
Impaired surface expression of CD95 or TRAIL et al., 1996; Fulda et al., 1997, 1998a; Muller et al.,
receptors may also be caused by epigenetic changes such 1997). The CD95 receptor/ligand system has also been
as CpG-island hypermethylation of gene promoters, for implicated in thymine-less death in colon carcinoma
example, in neuroblastoma or colon carcinoma cells cells following treatment with 5-uorouracil (Houghton
(van Noesel et al., 2002; Petak et al., 2003). Also, et al., 1997). Activation of the transcription factors
alterations in chromatin structure, for example, chro- activator protein-1 (AP-1) and NF-kB was shown to
matin condensation because of histone deacetylation, mediate the increase in CD95L transcription and
may block transcription by preventing the access of mRNA levels in response to chemotherapy and AP-1-
transcription factors to the DNA (Marks et al., 2001). and NF-kB-binding sites were identied in the human
Interestingly, epigenetic changes in CD95 expression CD95L promotor (Kasibhatla et al., 1998). In addition
have been reported to determine immune escape and to CD95L, CD95 expression on the cells surface
response to therapy (Maecker et al., 2002). In tumors increased upon drug treatment, in particular in cells
with epigentically silenced CD95, restoration of CD95 harboring wild-type p53 (Muller et al., 1997). p53-
expression by histone deacetylase inhibitors resulted in responsive elements have been identied in the rst
suppression of tumor growth and restoration of intron of the CD95 gene, as well as three putative p53-
chemosensitivity in an natural killer (NK) cell-depen- binding sites within the CD95 promotor, which showed
dent manner (Maecker et al., 2002). limited homology with the p53 consensus-binding site
Signaling by death receptors can also be negatively (Muller et al., 1998). Moreover, antagonistic CD95L
regulated by proteins that associate with their cytoplas- antibodies, soluble antagonistic CD95 receptors or DN-
matic domains, for example, FLIP or phosphoprotein FADD were found to reduce drug-induced apoptosis
enriched in diabetes/phosphoprotein enriched in astro- under certain circumstances. In addition to upregulation
cytes-15 kDa (PED/PEA-15) (Hao et al., 2001; Krueger of CD95L and CD95, anticancer agents have been
et al., 2001). Two splice variants of FLIP, a long form reported to activate the CD95 pathway by modulating
(FLIPL) and a short form (FLIPS), have been identied expression and recruitment of pro- or antiapoptotic
in human cells, which have sequence homology to components of the CD95 DISC to activated receptors.
caspase-8 and caspase-10, but lack its catalytic site Upregulation of FADD and procaspase-8 was found
(Krueger et al., 2001). Consequently, the recruitment of upon treatment with doxorubicin, cisplatin or mitomy-
FLIP to the DISC instead of procaspase-8 or -10 can cin C in colon carcinoma cells (Micheau et al., 1999).
block caspase activation (Krueger et al., 2001). High Also, increased recruitment of FADD and procaspase-8
FLIP expression has been found in many tumor cells to the CD95 receptor to form the CD95 DISC was
and has been correlated with resistance to CD95- or observed in certain tumor cells upon drug treatment
TRAIL- and also to chemotherapy-induced apoptosis in a CD95L-dependent or CD95L-independent manner
(Fulda et al., 2000; Longley et al., 2006). However, the (Fulda et al., 2001). These ndings indicate that in cells
impact of FLIP on apoptosis sensitivity towards with an inducible CD95 receptor/ligand system, drug-
cytotoxic drugs may vary between cell types, since induced apoptosis may involve CD95L-initiated DISC
overexpression of FLIP did not confer protection formation and activation of downstream effector
against cytotoxic drugs in T-cell leukemia cells (Kataoka programs similar to activation-induced cell death
et al., 1998), whereas FLIP antisense oligonucleotides or (AICD) in T cells.
short interference RNA sensitized osteosarcoma or Despite the reproducibility of these ndings in
colorectal cancer cells for chemotherapeutic drugs different model systems, other reports challenged the
(Kinoshita et al., 2000; Longley et al., 2006). The model that death receptor signaling is involved in drug-
PEA-15 is another death effector domain (DED)- mediated cell death (Eischen et al., 1997; Villunger et al.,
containing protein, which blocks CD95-, TRAIL- or 1997). To that end, antagonistic antibodies against
TNFa-triggered apoptosis in a receptor-proximal man- CD95L or CD95 did not confer protection against
ner by disrupting FADD and caspase-8 inter- apoptosis induced by cytotoxic drugs in other cell lines.
actions (Hao et al., 2001). Recently, PEA-15 has also Enforced expression of FLIP, DN-FADD or the serpin
been implicated in mediating AKT-dependent chemo- crmA did not inhibit drug-induced apoptosis, although
resistance in human breast cancer cells (Stassi et al., it inhibited caspase-8 activation (Kataoka et al., 1998).
2005). Also, targeted disruption of genes involved in death
Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4802
receptor signaling conferred no protection against appears to be a relatively safe and promising candidate
cytotoxic drug treatment, at least in nontransformed for clinical application, particularly in its non-tagged,
cells. The FADD / and caspase-8 / broblasts were zinc-bound homotrimeric form (LeBlanc and Ashkenazi,
sensitive to cytotoxic drugs, whereas they remained 2003). Studies in non-human primates such as chim-
resistant to death receptor stimulation (Varfolomeev panzees and cynomolgus monkeys showed no toxicity
et al., 1998; Yeh et al., 1998). In contrast, caspase-9 / upon intravenous infusion, even at high doses (Ashke-
embryonic stem cells and Apaf-1 / thymocytes remain nazi et al., 1999). In addition, no cytotoxic activity of
sensitive to death receptor triggering, however they are TRAIL was reported on a variety of normal human cells
resistant to cytotoxic drugs (Hakem et al., 1998; of different lineages including broblasts, endothelial
Yoshida et al., 1998) cells, smooth muscle cells, epithelial cells or astro-
The discrepancies in data may be explained by the cytes (Ashkenazi et al., 1999; Lawrence et al., 2001;
relative contribution of the extrinsic versus the intrinsic Pollack et al., 2001). However, some concerns about
pathway depending on the cytotoxic drug, dose or potential toxic side effects on human hepatocytes or
kinetics or on the differences between certain cell types. brain tissue have also been raised (Jo et al., 2000; Nitsch
The discrepancies in data may also be explained by et al., 2000). The loss of tumor selectivity may be related
differences in the inhibitory reagents used to block to the TRAIL preparations used in these studies, since
CD95/CD95L interaction. The quality of CD95/ TRAIL preparations, which are antibody-crosslinked or
CD95L-blocking agents including anti-CD95 antibody, not optimized for Zn content, have been reported to
anti-CD95L antibody or soluble decoy CD95-Fc re- form multimeric aggregates thereby overpassing the
ceptor constructs may vary depending on their origin threshold of sensitivity of normal cells (Lawrence et al.,
and preparation. Also, the lack of efcacy of these 2001).
CD95/CD95L-neutralizing agents may be owing to
inaccessibility of their proposed targets. Experiments
Antitumor activity of TNF-related apoptosis-inducing
with adenoviral delivery of a CD95L-green uorescent
ligand. Recombinant soluble TRAIL induced apopto-
protein (GFP) construct showed that CD95 and CD95L
sis in a broad spectrum of cancer cell lines, including
are stored intracellularly and colocalize to the same
colon carcinoma, breast carcinoma, lung carcinoma,
intracellular compartments, for example, the Golgi and/
pancreas carcinoma, prostate carcinoma, renal carcino-
or endoplasmatic reticulum (Hyer et al., 2000). An anti-
ma, thyroid carcinoma, malignant brain tumors, Ewing
CD95-blocking antibody did not inhibit CD95L-in-
tumor, osteasarcoma, neuroblastoma, leukemia and
duced cell death suggesting that CD95L may trigger
lymphoma and also exhibited potent tumoricidal
CD95 within the same intracellular compartment and
activity in vivo in several xenograft models of colon
that these two molecules may already interact before
carcinoma, breast carcinoma, malignant glioma or
surface presentation (Hyer et al., 2000). Thus, CD95/
multiple myeloma (Chinnaiyan et al., 2000; Nagane
CD95L-neutralizing agents may under certain circum-
et al., 2000; Walczak et al., 2000; Pollack et al., 2001;
stances not even gain access to their targets prior to
Rohn et al., 2001). Furthermore, monoclonal antibodies
triggering of the CD95 pathway. Moreover, some
that engage the TRAIL receptors DR4 or DR5 also
studies that challenge an involvement of the CD95
demonstrated potent antitumor activity against tumor
system in chemotherapy of tumor cells are based on
cell lines and in preclinical cancer models (Chuntharapai
experiments performed in nontransformed cells, for
et al., 2001; Ichikawa et al., 2001). Currently, recombi-
example, embryonic broblasts, but not in cancer cells.
nant soluble TRAIL or agonistic TRAIL receptors are
However, the mechanisms regulating apoptosis in non-
being evaluated in phase I clinical trials.
malignant cells may vary considerably from those in
malignant tumor cells, which is highlighted by the
differential sensitivity of these cell types to various death Combination therapy with TNF-related apoptosis-indu-
stimuli. cing ligand. Although these studies provided ample
evidence of the potential of TRAIL for cancer therapy,
many tumors remain refractory towards treatment
Exploiting death receptor signaling pathways for tumor with TRAIL, which has been related to the dominance
therapy of antiapoptotic signals. Importantly, numerous studies
Death receptor as targets. The idea to specically have shown that TRAIL synergized together with
target the extrinsic pathway to trigger apoptosis in cytotoxic drugs or g-irradiation to achieve antitumor
tumor cells, for example by ligation of death receptors, activity in various cancer cell lines including malignant
is attractive for cancer therapy since death receptors glioma, melanoma, leukemia, breast, colon or prostate
have a direct link to the cells death machinery carcinoma and also cooperated to suppress tumor
(Ashkenazi, 2002). In addition, apoptosis upon death growth in different mouse models of human cancers
receptor triggering has been reported to occur indepen- (Gliniak and Le, 1999; Chinnaiyan et al., 2000; Keane
dent of the p53 tumor suppressor gene, which is deleted et al., 2000; Nagane et al., 2000; Walczak et al., 2000;
or inactivated in more than half of human tumors Belka et al., 2001; Rohn et al., 2001). Dependent on the
(El-Deiry, 2001). However, the clinical application of therapeutic combinations and cell type used, different
CD95Lor TNFa is hampered by severe toxic side effects mechanisms of sensitization to TRAIL-induced apop-
(Walczak and Krammer, 2000). In contrast, TRAIL tosis have been identied. For example, the molecular
Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4803
mechanisms, which account for the synergistic interac- 2004). Once in the cytosol, these apoptogenic proteins
tion of TRAIL and conventional chemotherapeutics, trigger the execution of cell death by promoting caspase
may include transcriptional upregulation of the agonis- activation or by acting as caspase-independent death
tic TRAIL receptors TRAIL-R1 and -R2, which effectors (Saelens et al., 2004).
occurred in a p53-dependent or p53-independent man-
ner (Takimoto and El-Deiry, 2000; Meng and El-Deiry,
2001). Of note, p53 has also been shown to transcrip- Mitochondrial mediators of caspase-dependent apoptosis
tionally activate the antagonistic TRAIL receptors Cytochrome c. The release of cytochrome c from
TRAIL-R3 and -R4 (Meng et al., 2000). Recent mitochondria directly triggers caspase-3 activation
evidence suggest that p53 is crucial for sensitization to through formation of the cytochrome c/Apaf-1/cas-
TRAIL by chemotherapy through transcriptional upre- pase-9-containing apoptosome complex (Cain et al.,
gulation of TRAIL-R2 in some tumors, for example, 2000). Once in the cytosol, cytochrome c binds to the
mismatch repair-decient colorectal cancer cells harbor- C-terminal region of Apaf-1, a cytosolic protein with
ing Bax mutations (Wang and El-Deiry, 2003). Intrigu- an N-terminal caspase-recruitment domain (CARD), a
ingly, pre-exposure to chemotherapy restored TRAIL nucleotide-binding domain with homology to Caenor-
sensitivity through p53-mediated increase of TRAIL-R2 habditis elegans CED-4 and a C-terminal domain
expression even in resistant colorectal carcinoma cells containing 1213 WD-40 repeats (Zou et al., 1997).
lacking Bax expression indicating that sequential com- Binding of cytochrome c to Apaf-1 facilitates the
bination of anticancer agents with TRAIL may over- association of dATP with Apaf-1 and exposes its
come some forms of resistance (Wang and El-Deiry, N-terminal CARD, which can now oligomerize and
2003). In addition, chemotherapy has been reported become a platform on which the initiator caspase-9
to enhance receptor assembly of CD95 or TRAIL is recruited and activated through a CARDCARD
receptors, for example, in colon carcinoma cells (Lacour interaction (Adrain et al., 1999). Consecutively, the
et al., 2004). The synergistic interaction between executioner caspase-3 is recruited to the apoptosome,
cytotoxic drugs and TRAIL may also be mediated by where it is activated by the resident caspase-9 (Bratton
downregulation of antiapoptotic proteins such as Bcl-2, et al., 2001). Caspase-3 then cleaves key substrates in the
Bcl-XL or FLIP upon drug treatment (Olsson et al., cell to produce many of the cellular and biochemical
2001). In addition, anticancer agents may sensitize events of apoptosis.
tumor cells for TRAIL treatment by upregulating In certain cases, caspase activity instigated by
proapoptotic molecules incuding caspases or FADD cytosolic cytochrome c contributes to the drop of matrix
(Micheau et al., 1999). The cooperative interaction of metalloproteinase (MMP), as was shown by the use of
chemotherapeutics and TRAIL has also been explained synthetic caspase inhibitors and Apaf1 / cells
by the simultaneous activation of the intrinsic and (Waterhouse et al., 2001). In addition, caspase activity
extrinsic pathway leading to enhanced cell death further damages the function of permeabilized mito-
through functional complementation. As TRAIL de- chondria by affecting the activity of complexes I and II,
monstrated synergistic interaction with anticancer inevitably leading to the loss of MMP and the
agents or irradiation, TRAIL may be most effective in generation of reactive oxygen species (Ricci et al.,
combination with conventional cancer treatments. 2004). Thus, secondary events resulting from cytosolic
changes, caused by the release of cytochrome c and
other mitochondrial IMS proteins, can feed back on
permeabilized mitochondria and affect their function.
Intrinsic pathway of apoptosis Importantly, this mitochondrial amplication loop of
caspase activity may critically determine the response of
Signaling through the intrinsic pathway of apoptosis cancer cells to cytotoxic treatments.
In the mitochondrial pathway of apoptosis, caspase Moreover, there is recent evidence for the existence of
activation is closely linked to permeabilization of the a cytochrome c- and apoptosome-independent, but
outer mitochondrial membrane by proapoptotic mem- Apaf-1-dependent mechanism(s) for caspase activation.
bers of the Bcl family (Green and Kroemer, 2004). A targeted knock-in of a variant of cytochrome c that
Numerous cytotoxic stimuli and proapoptotic signal- lacks apoptogenic properties, but has electron transfer
transducing molecules converge on mitochondria to and antioxidant activity (K72A), allowed the evaluation
induce outer mitochondrial membrane permeabilization of the contribution of cytochrome c to apoptosis
(Decaudin et al., 1998; Green and Kroemer, 2004). This without affecting its function in oxidative phosphoryla-
permeabilization is regulated by proteins from the Bcl-2 tion (Hao et al., 2005). Interestingly, thymocytes from
family, mitochondrial lipids, proteins that regulate the KA/KA mice were markedly more sensitive to death
bioenergetic metabolite ux and components of the stimuli including etoposide and g-irradiation than Apaf-
permeability transition pore (Green and Kroemer, 1( / ) thymocytes (Hao et al., 2005). Upon treatment
2004). Upon disruption of the outer mitochondrial with g-irradiation, procaspases were efciently activated
membrane, a set of proteins normally found in the space in apoptotic KA/KA thymocytes, but Apaf-1 oligomer-
between the inner and outer mitochondrial membranes ization was not observed (Hao et al., 2005) pointing to a
is released, including cytochrome c, Smac/DIABLO, differential requirement for cytochrome c and Apaf-1 in
Omi/HtrA2, AIF and endonuclease G (Saelens et al., apoptosis.
Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4804
Smac/DIABLO. Other proteins released from mito- liberating an IBM at its N-terminus (Suzuki et al., 2001;
chondria, such as Smac/DIABLO and Omi/HtrA2, Martins et al., 2002; van Loo et al., 2002) (Table 1).
facilitate caspase activation through neutralizing endo- Although recombinant Omi/HtrA2 can catalyse its own
genous inhibitors of caspases, the inhibitor of apoptosis maturation in vitro, the protease responsible for its
proteins (IAPs). Smac and its murine homolog DIA- maturation in cells remains unknown (Martins et al.,
BLO are nuclear encoded mitochondrial proteins, which 2002). Omi/HtrA2 plays an essential role in regulating
contain a mitochondrial localization signal, which is mitochondrial homeostasis requiring its proteolytic
proteolytically removed upon mitochondrial import activity, although the molecular targets and interaction
to yield the mature 23 kDa protein (Du et al., 2000; partners of Omi/HtrA2 in the mitochondrion have not
Verhagen et al., 2000). This maturation step exposes the yet been dened (Saelens et al., 2004). Once Omi/HtrA2
IAP-binding motif (IBM) at the N-terminus of Smac/ is released from mitochondria into the cytosol, it
DIABLO (Du et al., 2000) (Table 1). Second mitochon- promotes cell death in a caspase-dependent way by
dria-derived activator of caspase/DIABLO was shown antagonizing IAPs, and in a caspase-independent way as
to bind to XIAP, cIAP1, cIAP2, survivin and Apollon a protease (Saelens et al., 2004). Similar to Smac/
in a BIR-dependent way (Vaux and Silke, 2003) DIABLO, Omi/HtrA2 blocks IAPs through its N-
(Figure 2),and acts as homodimer with the IBM present terminal IAP-binding motif, presented in a trimeric
in a bivalent conguration (Chai et al., 2000). One conguration (Li et al., 2002)
Smac/DIABLO dimer binds one XIAP molecule by Although the release of cytochrome c into the cytosol
both IAP-binding motifs, one interacting with BIR2 and directly triggers caspase-3 activation through formation
the other one with BIR3 (Huang et al., 2003). of the cytochrome c/Apaf-1/caspase-9-containing apop-
Intriguingly, the same BIR3 groove binds the IBM tosome complex, Smac/DIABLO and Omi/HtrA2
exposed at the N-terminus (Ala-Thr-Pro-Phe) of the indirectly promote caspase activation through antago-
small subunit of caspase-9 following its autocatalytic nizing the inhibitory effects to IAPs (Saelens et al.,
processing after Asp315, allowing Smac/DIABLO to 2004). Thus, a dynamic equilibrium exists between pro-
displace caspase-9 from XIAP (Srinivasula et al., 2001) and antiapoptotic effector molecules, which allows the
(Table 1). cell to cope with limited mitochondrial damage, in
The physiological mitochondrial function of Smac/ which case IAPs can adequately block caspase activa-
DIABLO is unknown, and DIABLO / mice appear tion initiated by a small amount of released cytochrome
normal (Okada et al., 2002). Although the in vitro c. However, under circumstances where mitochondrial
cleavage of procaspase-3 upon addition of cytochrome c damage proceeds or simultaneously affects multiple
was inhibited in lysates of Smac / cells, Smac / mitochondria, the antiapoptotic hurdle imposed by
mice and cells responded normally to apoptotic stimuli IAPs can be overcome by the higher cytosolic concen-
such as UV irradiation, staurosporin, etoposide and tration of their antagonists Smac/DIABLO and HtrA2/
TNF/cyclohexamide (Okada et al., 2002). These ob- OMI, which neutralize IAPs by direct binding.
servations suggest the existence of redundant factors There is mounting evidence that cancer cells have an
compensating for the loss of Smac/DIABLO, possibly intrinsic drive to apoptosis that is held in check by IAPs.
HtrA2/OMI. To this end, high basal levels of caspase-3 and caspase-8
Omi/HtrA2 is a nuclear-encoded, 49 kDa protein with activities and active caspase-3 fragments in the absence
an N-terminal mitochondrial localization signal that of apoptosis were detected in various tumor cell lines
mediates its translocation into the mitochondrial inter- and cancer tissues, but not in normal cells (Yang et al.,
membrane space (Suzuki et al., 2001; Martins et al., 2003a). Tumor cells, but not normal cells also expressed
2002; van Loo et al., 2002). Omi/HtrA2 is processed in high levels of IAPs, suggesting that upregulated IAP
the intermembrane space into the 37 kDa mature form, expression counteracted the high basal caspase activity
selectively in tumor cells (Yang et al., 2003a). Therefore,

Table 1 Alignment of the N-terminal sequences of proteins with an


IBM
Reaper MAVAF
Grim MAIAY
Hid MAVPF
Smac/DIABLO AVPI
Omi/HtrA2 AVPS
Caspase-9 ATPF

Drosophila proteins Reaper, Grim and Hid have an IAP-binding motif


(IBM) following the initiator methione, Smac/DIABLO and Omi/
HtrA2 have their IBM exposed after removal of the N-terminal
mitochondrial localization sequence, whereas the small subunit of
caspase-9 contains the IBM after autoprocessing. IBM, inhibitor of
Figure 2 Schematic diagram of domain structure of human IAP apoptosis protein (IAP)-binding motif; Smac/DIABLO, second
proteins. BIR, Baculovirus IAP repeats; CARD, caspase-activating mitochondria-derived activator of caspase/direct inhibitor of apoptosis
and recruitment domain; RING, ring zinc-nger. protein (IAP)-binding protein with low isoelectric point (PI).

Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4805
strategies targeting IAPs are considered as a promising the immunglobulin heavy chain gene locus, is associated
approach to enhance the efcacy of cytotoxic therapies with approximately 85% of human follicular lymphoma
selectively in cancer cells. To this end, transfection- (Tsujimoto et al., 1984). Experiments with transgenic
enforced expression of Smac/DIABLO lowered the mice have demonstrated that Bcl-2 overexpression can
threshold for TRAIL-induced killing in different tumors promote neoplastic transformation of B and T lympho-
(Ng and Bonavida, 2002; Okano et al., 2003) and also cytes and myeloid cells (McDonnell and Korsmeyer,
sensitized cancer cells for chemotherapy (McNeish et al., 1991; Traver et al., 1998).
2003; Zhao et al., 2006). However, translocation of Given the fact that overexpression of antiapoptotic
endogenous Smac/DIABLO into the cytosol during Bcl-2 family members promotes oncogenesis, it follows
anticancer drug-induced apoptosis does not appear to that multi BH domain proapoptotic members of this
play a major role under certain conditions, for example, family act as tumor suppressors. However, since Bax
in human lung carcinoma cells upon treatment with and Bak have largely overlapping roles in apoptosis, it
etoposide (Bartling et al., 2004). Downregulation of has been difcult to determine whether this hypothesis
Smac/DIABLO by small interfering RNA (siRNA) did holds true and indeed bax / mice are not markedly
not inuence killing by etoposide in these cells, although predisposed to neoplasia (Knudson et al., 2001).
an IAP-binding peptide Smac-N7 enhanced etoposide- However, somatic mutations that inactivate the bax
induced apoptosis (Bartling et al., 2004). These data gene have been found in certain solid tumors and
suggest that Smac/DIABLO deciency may be compen- hematological malignancies. To this end, single nucleo-
sated for by action of redundant determinants in certain tide substitution or frameshift mutations, which inacti-
cancer cells. vate the Bax gene in mismatch repair-decient (MMR)
colon cancer or hematopoetic malignancies, have been
described (Rampino et al., 1997; Kitada et al., 2002).
Mitochondrial mediators of caspase-independent
Moreover, there is mounting evidence that BH3-only
apoptosis
proteins may contribute to the suppression of malignant
Upon its release from the mitochondrial intermembrane
transformation, indicating that they can function as
space, HtrA2/OMI contributes to cell death also in a
bona de tumor suppressors. For example, loss of a
caspase-independent way as a protease in addition to
single allele of Bim was shown to accelerate B-cell
promoting apoptosis in a caspase-dependent way by
lymphomagenesis induced by expression of a c-myc
antagonizing IAPs (Suzuki et al., 2001; Martins et al.,
transgene (Egle et al., 2004), in line with the key role of
2002; van Loo et al., 2002). In vitro data demonstrated
Bim as a regulator of lymphoid homeostasis (Strasser,
the degradation of XIAP, cIAP1, cIAP2 and Apollon by
2005). Interestingly, homozygous deletions in the
the protease activity of HtrA2/OMI (Suzuki et al.,
chromosomal region harboring the bim gene have
2004). In addition, Trencia et al. (2004) demonstrated
recently been identied in patients with mantle cell
the interaction of antiapoptotic PED/PEA-15 with, and
lymphoma (Tagawa et al., 2005). Mice lacking bid
its degradation by, cytosolic HtrA2/OMI. Decreasing
spontaneously develop a myeloproliferative disorder
the levels of HtrA2/OMI in cells by antisense or RNA
that can progress to a malignancy resembling chronic
interference lowers the sensitivity of different cancer cell
myelomonocytic leukemia (CMML) (Zinkel et al.,
lines to cell death induced by staurosporin, Fas, UV or
2003). Moreover, RNAi-mediated suppression of Puma
cisplatin (Martins et al., 2002).
was reported to accelerate Myc-induced lymphomagen-
Furthermore, AIF and endonuclease G are released
esis (Hemann et al., 2004).
from mitochondria upon outer mitochondrial mem-
In addition to genetic alterations, aberrant expression
brane permeabilization and translocate into the nucleus
of Bcl-2 family proteins is mostly regulated at the
to contribute to nuclear chromatin condensation and
transcriptional or post-transcriptional level. For exam-
large-scale DNA fragmentation (Cande et al., 2004;
ple, expression of several antiapoptotic Bcl-2 family
Saelens et al., 2004). Whether these two proteins are
proteins, for example, Bcl-2, Bcl-XL, Mcl-1 or B-1, is
released before, together or after cytochrome c has been
transcriptionally regulated by NF-kB (Cory and Adams,
controversially discussed (Arnoult et al., 2002). Also, it
2002).
is still not exactly clear how AIF contributes to nuclear
Besides Bcl-2 family proteins, decreased or absent
DNA fragmentation as it lacks intrinsic DNase activity.
activity of Apaf-1 was found in ovarian cancer,
In mammalian cells, cyclophilin A, a peptidyl-prolyl cis
melanoma and leukemia. Furthermore, mutations in
trans isomerase, cooperates with AIF to induce break-
the tumor suppressor gene p53, the most common
down of DNA (Cande et al., 2004).
genetic defect in human cancers, impinge on the intrinsic
pathway. For example, activation of p53 upregulates a
Disruption of the intrinsic pathway in cancer number of genes possessing p53-responsive elements
Mutations in genes involved in the regulation of the present in their promoters, such as the proapoptotic
mitochondrial pathway are very common in cancer cells. BH3-only proteins Puma, Noxa and Bid (Oda et al.,
As the majority of anticancer therapies induce apoptosis 2000; Yu et al., 2001; Sax et al., 2002). As a result, cells
in cancer cells by triggering the intrinsic pathway, such in which p53 is stabilized are sensitized for activation of
mutations are usually associated with treatment resis- the mitochondrial cell death pathway. In addition, p53
tance. For example, overexpression of Bcl-2, as a result may directly affect mitochondrial integrity without the
of chromosomal translocation of the bcl-2 oncogene into need for gene activation. Indeed, it has been reported
Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4806
that p53 can bind to Bcl-2 and Bcl-XL at the of several Bcl-2 family proteins, including Bax, Bak and
mitochondria, thereby promoting mitochondrial desta- Bcl-2 (Robertson et al., 2004). Further, caspase-2 was
bilization (Mihara et al., 2003). also shown to have the surprising ability to disrupt the
association between cytochrome c and anionic phos-
pholipids, notably cardiolipin, thereby making addi-
Signaling through the intrinsic pathway in cancer therapy tional cytochrome c available for release into the cytosol
Most conventional chemotherapeutic agents, for exam- (Enoksson et al., 2004). Upon DNA damage, caspase-2
ple, etoposide, doxorubicin, cisplatin or paclitaxel, elicit has recently been reported to become activated in the so-
mitochondrial permeabilization in an indirect fashion by called PIDDosome, a complex of the p53-inducible,
triggering perturbations of intermediate metabolism or death domain-containing protein PIDD, caspase-2 and
by increasing the concentration of proapoptotic second the adaptor protein RAIDD (Tinel and Tschopp, 2004)
messengers, for example, by inducing p53 expression, pointing to the existence of a nucleo-mitochondrial
by inducing the ceramide/GD3 pathway, by inducing apoptotic pathway.
the CD95/CD95L ligand system, affecting Bcl-2-like Additionally, histone H1.2 has been reported to play
proteins and/or by compromising the redox or energy an important role in transmitting apoptotic signals from
balance. the nucleus to the mitochondria following DNA double-
There is mounting evidence for the existence of a strand breaks (Konishi et al., 2003). Nuclear histone
nucleo-mitochondrial cross-talk following DNA da- H1.2 is released into the cytoplasm through a p53-
mage. For example, apoptotic signals arising from dependent mechanism after DNA double-strand breaks
DNA damage can be transmitted via the tumor and induced cytochrome c release from isolated
suppressor p53 to mitochondria, which in turn release mitochondria in a Bak-dependent manner (Konishi
apoptogenic factors into the cytoplasm that activate et al., 2003). Reducing H1.2 expression enhanced
downstream destruction programs (Moll et al., 2005). cellular resistance to apoptosis induced by X-ray
p53 can indirectly engage the mitochondrial pathway by irradiation or etoposide (Konishi et al., 2003).
transcriptionally activating the expression of proapop- Moreover, the orphan nuclear receptor Nur77 (also
totic Bcl-2 proteins, for example, Bid, Puma or Noxa known as TR3) has recently been coupled with the Bcl-2
(Oda et al., 2000; Yu et al., 2001; Sax et al., 2002). apoptotic machinery at the mitochondria (Lin et al.,
Additionally, p53 can directly trigger permeabilization 2004). Nur77 binding to the Bcl-2 N-terminal loop
of the outer mitochondrial membrane in a transcription- region, located between its BH4 and BH3 domains,
independent fashion through direct activation of proa- induces a Bcl-2 conformational change that exposes its
poptotic Bcl-2 proteins Bax or Bak or through binding BH3 domain, resulting in conversion of Bcl-2 from a
and inactivating of antiapoptotic Bcl-2 proteins such as protector to a killer (Lin et al., 2004). Interestingly,
Bcl-2 or Bcl-XL (Mihara et al., 2003; Chipuk et al., elevated levels of an Nur77-family member have been
2004; Moll et al., 2005). Importantly, mitochondrially associated with favorable responses to chemotherapeu-
targeted p53 has recently been shown to possess tumor tic agents in patients (Shipp et al., 2002).
suppressor activities also in vivo (Talos et al., 2005). Furthermore, upon cellular stress including che-
Moreover, caspase-2 possesses the ability to engage motherapeutic drugs, specic proapoptotic Bcl-2 family
the mitochondrial apoptotic pathway in response to members are activated, derepressed or induced, and
DNA damage by permeabilizing the outer mitochon- thereby act as sensors. The activity of BH3-only proteins
drial membrane and/or by breaching the association of is kept in check by several mechanisms, keeping these
cytochrome c with the inner mitochondrial membrane. proteins away from the multidomain Bcl-2 counterparts
Hence, cells stably transfected with procaspase-2 anti- under normal circumstances, yet allowing their rapid
sense, or transiently expressing siRNA, were refractory activation under stress conditions (Bouillet and Strasser,
to cytochrome c release and various downstream events, 2002). As mentioned above, Bcl-2 family protein such as
such as caspase activation and DNA fragmentation, Bid, Puma or Noxa are under the transcriptional control
induced by DNA damage (Lassus et al., 2002; Robert- of the tumor suppressor p53 and hence, upregulated in
son et al., 2002). Caspase-2 can act indirectly on the response to DNA-damaging agents (Oda et al., 2000; Yu
mitochondria, for example, by cleaving the proapoptotic et al., 2001; Sax et al., 2002). The BH3-only protein Bim,
protein Bid, followed by its translocation to the which is associated with the cytoskeleton by binding to
mitochondria to induce cytochrome c release (Guo microtubules, is set free and activates the intrinsic
et al., 2002). Additionally, caspase-2 can directly pathway following treatment with taxol, which acts on
permeabilize the outer mitochondria membrane and microtubule assembly (Sunters et al., 2003). Recently,
stimulate the release of cytochrome c and Smac/ paclitaxel has been demonstrated to induce Bim
DIABLO, possibly as a result of direct interaction of accumulation and Bim-dependent apoptosis in epithelial
processed caspase-2 with putative proteins and/or tumors in vitro and also in vivo (Tan et al., 2005). Active
phospholipids located in the outer mitochondrial BH3-only proteins bind and counteract antiapoptotic
membrane, or at contact sites between the outer and and in some cases activate multidomain proapoptotic
inner membranes (Robertson et al., 2004). Permeabili- Bcl-2 family members, leading to the loss of mitochon-
zation of the outer mitochondria membrane requires drial membrane permeability (Bouillet and Strasser,
processing of the caspase-2 zymogen but not the 2002). How Bcl-2 proteins induce disturbance of the
associated proteolytic activity and occurs independently mitochondrial outer membrane is still a matter of debate
Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4807
and may involve the pore-forming and self-oligomeriz- gets the rst six codons (i.e., 18 bases) of the mRNA
ing capacity of some Bcl-2 family proteins, modulation open reading frame encoding the Bcl-2 protein (Cotter,
of the mitochondrial permeability transition pore by 2004). Treatment with genasense markedly enhanced the
Bcl-2 family proteins and/or lipid changes and lipid antitumor activity of many chemotherapeutic drugs, for
protein interactions within the mitochondrial mem- example, taxanes, anthracyclines, alkylators or plati-
branes. Moreover, chemotherapeutic agents such as num-containing agents (Cotter, 2004). In a preclinical
paclitaxel cause hyperphosphorylation and inactivation model of melanoma, pretreatment with genasense
of Bcl-2, and, simultaneously, favor opening of the increased the chemosensitivity of human melanoma
permeability transition (PT) pore (Ruvolo et al., 2001). (Jansen et al., 1998). Also in a clinical trial, genasense
Chemotherapeutic drugs may also induce or facilitate was reported to act as chemosensitizer for dacarbazine
permeabilization of the outer mitochondrial membrane in patients with malignant melanoma (Jansen et al.,
through changes in cellular redox potentials owing to an 2000). To optimize antisense-based therapy, bispecic
enhanced generation of reactive oxygen species (or a antisense oligonucleotides directed against a sequence,
decrease in their detoxication), depletion of reduced which is highly homologous in Bcl-2 and Bcl-xL, but
glutathione or depletion of NADPH, as the mitochon- missing in Bcl-xS mRNA, were subsequently designed
drial megachannel possesses several redox-sensitive sites (Zangemeister-Wittke et al., 2000). Simultaneous down-
(Debatin et al., 2002). In addition, changes in energy regulation of both Bcl-2 and Bcl-xL induced apoptosis
metabolism, for example depletion in ADP and ATP, and enhanced chemosensitivity in various cancer cells
might facilitate permeability transition pore complex (Gautschi et al., 2001; Tortora et al., 2003; Milella et al.,
(PTPC) opening, since ADP and ATP are the physiologic 2004; Yamanaka et al., 2005).
ligands of the adenine nucleotide translocator, which Furthermore, BH3-domain peptides or synthetic small
function as endogenous inhibitors of the PTPC (Costan- molecule inhibitors were developed to target anti-
tini et al., 2000). Also, uncoupling or inhibition of the apoptotic Bcl-2-like proteins. The BH3 domain com-
respiratory chain or matrix alkalinization may favor prises a nine-amino-acid amphipathic a-helix that binds
mitochondrial membrane permeabilization. In addition, to a hydrophobic pocket of Bcl-2-like proteins (Cory and
lipid messengers such as ceramide, which is generated in Adams, 2002). Similarly, BH3-domain peptides aim at
cells exposed to several apoptosis-inducing stimuli, disrupting this complex, thereby sensitizing cancer cells
including cytotoxic drugs, can contribute to mitochon- to apoptosi (Letai et al., 2002). Additionally, substitution
drial membrane permeabilization (Susin et al., 1997). At of the Bid BH3 domain with non-natural amino acids on
high concentrations. some chemotherapeutic drugs, for the surface opposite to the interacting region by
example, etoposide or paclitaxel, can also induce mito- hydrocarbon stapling resulted in stabilized BH3 peptides
chondrial outer membrane permeabilization in isolated termed SAHBs (stabilized a-helix of Bcl-2 domains),
mitochondria (Robertson et al., 2000; Kidd et al., 2002). with improved pharmacological properties (Walensky
In addition, an increasing number of experimental et al., 2004). These stabilized BH3 peptides triggered
anticancer drugs, including arsenite, lonidamide, the apoptosis in a variety of leukemic cell lines and also
synthetic retinoid CD437 or the natural product inhibited the growth of leukemia xenografts in mice
betulinic acid, have been shown to act directly on without adverse side effects (Walensky et al., 2004).
mitochondria (Debatin et al., 2002). For example, Also, several small molecule compounds interfering
betulinic acid has been reported to trigger apoptosis with Bcl-2/Bcl-xL function have been identied. Screen-
by directly inducing loss of mitochondrial membrane ing of a chemical library for compounds able to bind to
potential in isolated mitochondria in a way that is not the BH3 pocket of Bcl-2 proteins resulted in the
affected by the caspase inhibitor Z-VAD-fmk and yet is identication of HA14-1, a compound that competes
inhibited by BA, an inhibitor of the PTPC, or by Bcl-2 with Bak for the binding to Bcl-2 (Wang et al., 2000). By
and Bcl-XL (Fulda et al., 1998b). screening a library of 16 320 preselected compounds for
the ability to displace a uorescent Bak BH3 peptide
from Bcl-xL in a uorescent polarization assay, Degterev
Strategies targeting the intrinsic pathway et al. (2003) identied two classes of agents called BH3
Bcl-2 family proteins. Since antiapoptotic Bcl-2 pro- inhibitors (BH3Is), which also disrupt the Bcl-xL
teins, which potently block the intrinsic apoptosis complex with Bax and Bad in intact cells.
pathway, are found at elevated levels in human cancers Using nuclear magnetic resonance (NMR)-based
of both hematological and nonhematological origin screening, parallel synthesis and structure-based design,
(Cotter, 2004), they represent promising targets for a small-molecule inhibitor of the antiapoptotic proteins
therapeutic interventions. Consequently, several strate- Bcl-2, Bcl-X(L) and Bcl-w, ABT-737, was recently
gies have been developed to target Bcl-2 proteins, for discovered. ABT-737 was effective as a single agent
example, antisense techniques, BH3-domain peptides or against certain lymphomas and solid tumors and
synthetic small molecule drugs interfering with Bcl-2- displayed synergistic cytotoxicity with chemotherapeu-
like protein function. To downregulate Bcl-2 expression, tics and radiation (Oltersdorf et al., 2005).
Bcl-2 antisense oligonucleotidex (genasense) were devel-
oped by Genta Incorporated (Berkeley Heights, NJ,
USA). Genasense is a synthetic, 18-base, single-stranded Smac/DIABLO agonists. For the design of potentially
phosphorothioate oligonucleotide that selectively tar- therapeutic small molecules to target XIAP, the binding
Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4808
groove of the BIR3 domain of XIAP, to which Smac/ Conclusions
DIABLO binds to after its release from mitochondria,
has attracted most attention. Structural analysis has Continued efforts over many years to understand the
provided a clear rationale for the synthesis of small mechanisms controlling tumor cell proliferation and cell
compounds that can mimic the caspase-9 displacing death have delineated key molecular pathways involved
activity of Smac/DIABLO from XIAP BIR3 (Chai in cancer formation, progression and treatment resis-
et al., 2000; Wu et al., 2000). To enhance intracellular tance. One of the key discoveries in cancer research has
delivery, Smac peptides were linked to a carrier, for been the recognition that anticancer chemotherapy kills
example, the protein transduction motif of the HIV Tat cancer cells by activating the intrinsic and/or extrinsic
protein (Fulda et al., 2002), the Drosophila antenna- apoptosis pathway. Although a considerable amount of
paedia penetratin sequence (Arnt et al., 2002) or a data support a role of the CD95 system in anticancer
polyarginine stretch (Yang et al., 2003b). A heptapep- drug-induced apoptosis, at least under certain circum-
tide representing the N-terminus of mature Smac/ stances, most cytotoxic drugs are considered to primar-
DIABLO, which is essential for binding to XIAP, was ily initiate cell death by triggering a cytochrome c/Apaf-
reported to promote caspase activation and to sensitize 1/caspase-9-dependent pathway linked to mitochondria.
various tumor cell lines and also primary patients Collectively, these data point to a key role of the
derived tumor cells for apoptosis induced by death- mitochondrial pathway in chemotherapy-induced apop-
receptor ligation or cytotoxic drugs (Fulda et al., 2002). tosis, whereas the CD95 system may amplify killing by
Importantly, Smac peptides even enhanced the anti- cytotoxic drugs under certain conditions. Importantly,
tumor activity of TRAIL in vivo in an intracranial this amplication of the chemoresponse may have
malignant glioma xenograft model (Fulda et al., 2002b). important clinical implications, since it may critically
Likewise, an 8-mer peptide (AVPIAQKS) fused to the affect the time required for execution of the death
protein transduction domain of Drosophila antennapae- program. Thus, defects in the intrinsic or extrinsic
dia penetratin was able to enter breast cancer cells, bind pathway may promote tumor progression and therapy
XIAP and cIAP1, and potentiate caspase activity resistance. Accordingly, targeting defective apoptosis
induced by a number of anticancer drugs, including programs may restore sensitivity in resistant forms of
paclitaxel, etoposide, 7-ethyl-10-hydroxycamptothecin cancer. However, apoptosis does not represent the sole
(SN-38) and doxorubicin (Arnt et al., 2002). Moreover, killing mechanism by which cancers are eradicated, and
on the basis of the three-dimensional structure of Smac/ other modes of cell death, for example, necrosis,
DIABLO in complex with XIAP BIR3, Smac peptido- autophagy, mitotic catastrophe or some forms of cell
mimetics were designed, which cooperated with TRAIL, death that cannot be easily classied at present, have
TNFa, cisplatin or etoposide to trigger apoptosis in also to be considered. Further insight into the complex
tumor cells (Li et al., 2004; Sun et al., 2004a, b, 2005). signaling network activated in response to anticancer
Subsequently, non-peptidic small molecule antago- therapy using cancer cell lines, primary tumor cells and
nists of XIAP, which were derived by screening of a animal models are necessary to see to what extent the
phage library or polyphenylurea library, were developed current knowledge can be exploited for the design of
for targeting IAPs (Schimmer et al., 2004; Wang et al., apoptosis-based cancer therapies. Also, further studies
2004). In addition, the natural product embelin from the on the role of apoptosis signaling molecules in clinical
Japanese Ardisia herb was recently discovered as a cell- samples using DNA or proteomic arrays are warranted
permeable, non-peptidic, small-molecular weight inhi- to assess the impact of these molecular parameters
bitor of XIAP through structure-based computational on clinical outcome. Hopefully, these studies may
screening of a traditional herbal medicine three-dimen- eventually allow the identication of novel therapeutic
sional structure database (Nikolovska-Coleska et al., targets thereby providing the basis for tailored, indivi-
2004). Embelin was shown to effectively overcome the dual tumor therapy.
protective effect of XIAP in prostate cancer cells with
high endogenous levels of XIAP or in Jurkat cells
transfected with XIAP through binding to the XIAP Acknowledgements
BIR3 domain (Nikolovska-Coleska et al., 2004). Thus,
Smac agonists or low molecular weight XIAP antago- Work in the authors laboratory was supported by grants from
nists may be promising candidates for cancer therapy by the Deutsche Forschungsgemeinschaft, the Deutsche Kreb-
potentiating the efcacy of cytotoxic therapies selec- shilfe, Wilhelm-Sander-Stiftung, Kind-Phillipp-Stiftung and
tively in cancer cells. Else-Kroner-Stiftung.

References

Adrain C, Slee EA, Harte MT, Martin SJ. (1999). J Biol Chem Ashkenazi A. (2002). Nat Rev Cancer 2: 420430.
274: 2085520860. Ashkenazi A, Pai RC, Fong S, Leung S, Lawrence
Arnoult D, Parone P, Martinou J-C, Antonsson B, Estaquier DA, Marsters SA et al. 1999)). J Clin Invest 104:
J, Ameisen JC. (2002). J Cell Biol 159: 923929. 155162.
Arnt CR, Chiorean MV, Heldebrant MP, Gores GJ, Bartling B, Lewensohn R, Zhivotovsky B. (2004). Exp Cell Res
Kaufmann SH. (2002). J Biol Chem 277: 4423644243. 298: 8395.

Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4809
Belka C, Schmid B, Marini P, Durand E, Rudner J, Faltin H Fulda S, Sieverts H, Friesen C, Herr I, Debatin KM. (1997).
et al. (2001). Oncogene 20: 21902196. Cancer Res 57: 38233829.
Bouillet P, Strasser A. (2002). J Cell Sci 115: 15671574. Fulda S, Wick W, Weller M, Debatin KM. (2002b). Nat Med
Bratton SB, Walker G, Srinivasula SM, Sun XM, Butterworth 8: 808815.
M, Alnemri ES et al. (2001). EMBO J 20: 9981009. Gautschi O, Tschopp S, Olie RA, Leech SH, Simoes-Wust A
Brown JM, Attardi LD. (2005). Nat Rev Cancer 5: 231237. P, Ziegler A et al. (2001). J Natl Cancer Inst 93: 463471.
Brown JM, Wilson G. (2003). Cancer Biol Ther 2: 477490. Gliniak B, Le T. (1999). Cancer Res 59: 61536158.
Cain K, Bratton SB, Langlais C, Walker G, Brown DG, Sun Green DR, Kroemer G. (2004). Science 305: 626629.
XM. (2000). J Biol Chem 275: 60676070. Guo Y, Srinivasula SM, Druilhe A, Fernandes-Alnemri T,
Cande C, Cecconi F, Dessen P, Kroemer G. (2002). J Cell Sci Alnemri ES. (2002). J Biol Chem 277: 1343013437.
15: 47274734. Hakem R, Hakem A, Duncan GS, Henderson JT, Woo M,
Cande C, Vahsen N, Kouranti I, Schmitt E, Daugas E, Spahr Soengas MS et al. (1998). Cell 94: 339352.
C et al. (2004). Oncogene 23: 15141521. Hao C, Beguinot F, Condorelli G, Trencia A, Van Meir EG,
Chai J, Du C, Wu JW, Kyin S, Wang X, Shi Y. (2000). Nature Yong V et al. (2001). Cancer Res 61: 11621170.
406: 855862. Hao Z, Duncan GS, Chang CC, Elia A, Fang M, Wakeham A
Chinnaiyan AM, Prasad U, Shankar S, Hamstra DA, et al. (2005). Cell 121: 579591.
Shanaiah M, Chenevert TL et al. (2000). Proc Natl Acad Hemann M, Zilfou JT, Zhao Z, Burgess DJ, Hannon GJ,
Sci USA 97: 17541759. Lowe SW. (2004). Proc Natl Acad Sci USA 101: 93339338.
Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Hengartner MO. (2000). Nature 407: 770776.
Newmeyer DD, Schuler M et al. (2004). Science 303: Houghton JA, Harwood FG, Tillman DM. (1997). Proc Natl
10101014. Acad Sci USA 94: 81448149.
Chuntharapai A, Dodge K, Grimmer K, Schroeder K, Huang Y, Rich RL, Myszka DG, Wu H. (2003). J Biol Chem
Marsters SA, Koeppen H et al. (2001). J Immunol 166: 278: 4951749522.
48914898. Hyer ML, Voelkel-Johnson C, Rubinchik S, Dong J, Norris
Cory S, Adams JM. (2002). Nat Rev Cancer 2: 647656. JS. (2000). Mol Ther 2: 348358.
Costantini P, Jacotot E, Decaudin D, Kroemer G. (2000). Ichikawa K, Liu W, Zhao L, Wang Z, Liu D, Ohtsuka T et al.
J Natl Cancer Inst 92: 10421053. (2001). Nat Med 7: 954960.
Cotter FE. (2004). Semin Oncol 31: 1821. Igney FH, Krammer PH. (2002). J Leuk Biol 71: 907920.
Cowling V, Downward J. (2002). Cell Death Differ 9: Jansen B, Schlagbauer-Wadl H, Brown BD, Bryan RN, van
10461056. Elsas A, Muller M et al. (1998). Nat Med 4: 232234.
Davis RJ. (2000). Cell 103: 239252. Jansen B, Wacheck V, Heere-Ress E, Schlagbauer-Wadl H,
Debatin KM, Poncet D, Kroemer G. (2002). Oncogene 21: Hoeller C, Lucas T et al. (2000). The Lancet 356: 17281733.
87868803. Jin Z, McDonald ER, Dicker DT, El-Deiry WS. (2004). J Biol
Debatin KM, Stahnke K, Fulda S. (2003). Semin Cancer Biol Chem 279: 3582935839.
13: 149158. Jo M, Kim TH, Seol DW, Esplen JE, Dorko K, Billiar TR
Decaudin D, Marzo I, Brenner C, Kroemer G. (1998). Int J et al. (2000). Nat Med 6: 564567.
Oncol 12: 141152. Karin M, Cao Y, Greten FR, Li ZW. (2002). Nat Rev Cancer
Dechant MJ, Fellenberg J, Scheuerpug CG, Ewerbeck V, 2: 301310.
Debatin KM. (2004). Int J Cancer 109: 661667. Kasibhatla S, Brunner T, Genestier L, Echeverri F, Mahboubi
Degterev A, Boyce M, Yuan J. (2003). Oncogene 22: A, Green DR. (1998). Mol Cell 1: 543551.
85438567. Kataoka T, Schroter M, Hahne M, Schneider P, Irmler M,
Du C, Fang M, Li Y, Li L, Wang X. (2000). Cell 102: 3342. Thome M et al. (1998). J Immunol 161: 39363942.
Egle A, Harris AW, Bouillet P, Cory S. (2004). Proc Natl Acad Keane MM, Rubinstein Y, Cuello M, Ettenberg SA, Banerjee
Sci USA 101: 61646169. P, Nau MM et al. (2000). Breast Cancer Res Treat 64:
Eischen CM, Kottke TJ, Martins LM, Basi GS, Tung JS, 211219.
Earnshaw WC et al. (1997). Blood 90: 935943. Kidd JF, Pilkington MF, Schell MJ, Fogarty KE, Skepper JN,
El-Deiry WS. (2001). Cell Death Differ 8: 10661075. Taylor CW et al. (2002). J Biol Chem 2: 65046510.
Enoksson M, Robertson JD, Gogvadze V, Bu P, Kropotov Kinoshita H, Yoshikawa H, Shiiki K, Hamada Y, Nakajima
A, Zhivotovsky B et al. (2004). J Biol Chem 279: Y, Tasaka K. (2000). Int J Cancer 88: 986991.
4957549578. Kischkel FC, Hellbardt S, Behrmann I, Germer M, Pawlita M,
Friesen C, Fulda S, Debatin KM. (1997). Leukemia 11: Krammer PH et al. (1995). EMBO J 14: 55795588.
18331841. Kitada S, Pedersen IM, Schimmer AD, Reed JC. (2002).
Friesen C, Herr I, Krammer PH, Debatin KM. (1996). Nat Oncogene 21: 34593474.
Med 2: 574577. Knudson C, Johnson GM, Lin Y, Korsmeyer SJ. (2001).
Fulda S, Debatin KM. (2004). Curr Cancer Drug Targets 4: Cancer Res 61: 659665.
569576. Konishi A, Shimizu S, Hirota J, Takao T, Fan Y, Matsuoka Y
Fulda S, Los M, Friesen C, Debatin KM. (1998a). Int J Cancer et al. (2003). Cell 114: 673688.
76: 105114. Krammer PH. (2000). Nature 407: 789795.
Fulda S, Meyer E, Debatin KM. (2000). Cancer Res 60: Krueger A, Baumann S, Krammer PH, Kirchhoff S. (2001).
39473956. Mol Cell Biol 21: 82478254.
Fulda S, Meyer E, Debatin KM. (2002a). Oncogene 21: Lacour S, Hammann A, Grazide S, Lagadic-Gossmann D,
22832294. Athias A, Sergent O et al. (2004). Cancer Res 64: 35933598.
Fulda S, Meyer E, Friesen C, Susin SA, Kroemer G, Debatin Lassus P, Opitz-Araya X, Lazebnik Y. (2002). Science 297:
KM. (2001). Oncogene 20: 10631075. 13521354.
Fulda S, Scafdi C, Susin SA, Krammer PH, Kroemer G, Lawrence D, Shahrokh Z, Marsters S, Achilles K, Shih D,
Peter ME et al. (1998b). J Biol Chem 273: 3394233948. Mounho B et al. (2001). Nat Med 7: 383385.

Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4810
LeBlanc HN, Ashkenazi A. (2003). Cell Death Differ 10: Ricci JE, Munoz-Pinedo C, Fitzgerald P, Bailly-Maitre B,
6675. Perkins GA, Yadava N et al. (2004). Cell 117: 773786.
Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Rich T, Allen RL, Wyllie AH. (2000). Nature 407: 777783.
Korsmeyer SJ. (2002). Cancer Cell 2: 183192. Robertson JD, Enoksson M, Suomela M, Zhivotovsky B,
Li L, Thomas RM, Suzuki H, De Brabander JK, Wang X, Orrenius S. (2002). J Biol Chem 277: 2980329809.
Harran PG. (2004). Science 305: 14711474. Robertson JD, Gogvadze V, Kropotov A, Vakifahmetoglu H,
Li W, Srinivasula SM, Chai J, Li P, Wu JW, Zhang Z et al. Zhivotovsky B, Orrenius S. (2004). EMBO Rep 5: 643648.
(2002). Nature 9: 436441. Robertson JD, Gogvadze V, Zhivotovsky B, Orrenius S.
Lin B, Kolluri SK, Lin F, Liu W, Han YH, Cao X et al. (2000). J Biol Chem 275: 3243832443.
(2004). Cell 116: 527540. Rohn TA, Wagenknecht B, Roth W, Naumann U, Gulbins E,
Longley DB, Wilson TR, McEwan M, Allen WL, McDermott Krammer PH et al. (2001). Oncogene 20: 41284137.
U, Galligan L et al. (2006). Oncogene 25: 838848. Roth W, Isenmann S, Nakamura M, Platten M, Wick W,
Maecker HL, Yun Z, Maecker HT, Giaccia AJ. (2002). Cancer Kleihues P et al. (2001). Cancer Res 61: 27592765.
Cell 2: 139148. Ruvolo PP, Deng X, May WS. (2001). Leukemia 15: 515522.
Makin G, Dive C. (2001). Trends Cell Biol 11: S22S26. Saelens X, Festjens N, Walle LV, van Gurp M, van Loo G,
Marks P, Rifkind RA, Richon VM, Breslow R, Miller T, Kelly Vandenabeele P. (2004). Oncogene 23: 28612874.
WK. (2001). Nat Rev Cancer 1: 194202. Sax JK, Fei P, Murphy ME, Bernhard E, Korsmeyer SJ,
Martins LM, Iaccarino I, Tenev T, Gschmeissner S, Totty NF, El-Deiry WS. (2002). Nat Cell Biol 4: 842849.
Lemoine NR et al. (2002). J Biol Chem 277: 439444. Scafdi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli
McDonnell TJ, Korsmeyer SJ. (1991). Nature 349: 254256. KJ et al. (1998). EMBO J 17: 16751687.
McNeish IA, Bell S, McKay T, Tenev T, Marani M, Lemoine Schimmer AD, Welsh K, Pinilla C, Wang Z, Krajewska M,
NR. (2003). Exp Cell Res 286: 186198. Bonneau MJ et al. (2004). Cancer Cell 5: 2535.
Meng RD, El-Deiry WS. (2001). Exp Cell Res 262: 154169. Sheikh MS, Huang Y, Fernandez-Salas EA, El-Deiry
Meng RD, McDonald ER, Sheikh MS, Fornace AJ, El-Deiry WS, Friess H, Amundson S et al. (1999). Oncogene 18:
WS. (2000). Mol Ther 1: 130144. 41534159.
Micheau O, Hammann A, Solary E, Dimanche-boitrel MT. Shipp MA, Ross KN, Tamayo P, Weng AP, Kutok JL, Aguiar
(1999). Biophys Res Commun 256: 603611. RCT et al. (2002). Nat Med 8: 6874.
Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Srinivasula SM, Hegde R, Saleh A, Datta P, Shiozaki E, Chai
Pancoska P et al. (2003). Mol Cell 11: 577590. J et al. (2001). Nature 410): 112116.
Milella M, Trisciuoglio D, Bruno T, Ciuffreda L, Mottolese Stassi G, Garofalo M, Zerilli M, Ricci-Vitiani L, Zanca C,
M, Cianciulli A et al. (2004). Clin Cancer Res 10: 77477756. Todaro M et al. (2005). Cancer Res 65: 66686675.
Moll UM, Wolff S, Speidel D, Deppert W. (2005). Curr Opin Strasser A. (2005). Nat Rev Immunol 5: 189200.
Cell Biol 17: 631636. Sun H, Nikolovska-Coleska Z, Chen J, Yang CY, Tomita Y,
Muller M, Strand S, Hug H, Heinemann EM, Walczak H, Pan H et al. (2005). Bioorg Med Chem Lett 15: 793797.
Hofmann WJ et al. (1997). J Clin Invest 99: 403413. Sun H, Nikolovska-Coleska Z, Yang CY, Xu L, Liu M,
Muller M, Wilder S, Bannasch D, Israeli D, Lehlbach K, Tomita Y et al. (2004a). J Am Chem Soc 126: 1668616687.
Li-Weber M et al. (1998). J Exp Med 188: 20332045. Sun H, Nikolovska-Coleska Z, Yang CY, Xu L, Tomita Y,
Nagane M, Pan G, Weddle JJ, Dixit VM, Cavenee WK, Krajewski K et al. (2004b). J Med Chem 47: 41474150.
Huang HJ. (2000). Cancer Res 60: 847853. Sunters A, Fernandez de Mattos S, Stahl M, Brosens JJ,
Nagata S. (2000). Exp Cell Res 256: 1218. Zoumpoulidou G, Saunders CA et al. (2003). J Biol Chem
Ng CP, Bonavida B. (2002). Mol Cancer Ther 1: 10511058. 278: 4979549805.
Nikolovska-Coleska Z, Xu L, Hu Z, Tomita Y, Li P, Roller Susin SA, Zamzami N, Castedo M, Daugas E, Wang HG,
PP et al. (2004). J Med Chem 47: 24302440. Geley S et al. (1997). J Exp Med 186: 2537.
Nitsch R, Bechmann I, Deisz RA, Haas D, Lehmann TN, Suzuki Y, Imai Y, Nakayama H, Takahashi K, Takio K,
Wendling U et al. (2000). Lancet 356: 827828. Takahashi R. (2001). Mol Cell 8: 613621.
Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T, Yamashita Suzuki Y, Takahashi-Niki K, Akagi T, Hashikawa T,
T et al. (2000). Science 288: 10531058. Takahashi R. (2004). Cell Death Differ 11: 208216.
Okada H, Mak TW. (2004). Nat Rev Cancer 4: 592603. Tagawa H, Karnan S, Suzuki R, Matsuo K, Zhang X, Ota A
Okada H, Suh WK, Jin J, Woo M, Du C, Elia A et al. (2002). et al. (2005). Oncogene 24: 13481358.
Mol Cell Biol 22: 35093517. Takimoto R, El-Deiry WS. (2000). Oncogene 19: 17351743.
Okano H, Shiraki K, Inoue H, Kawakita T, Saitou Y, Talos F, Petrenko O, Mena P, Moll UM. (2005). Cancer Res
Enokimura N et al. (2003). Int J Mol Med 12: 2528. 65: 99719981.
Olsson A, Diaz T, Aguilar-Santelises M. (2001). Leukemia 15: Tan TT, Degenhardt K, Nelson DA, Beaudoin B, Nieves-
18681877. Neira W, Bouillet P et al. (2005). Cancer Cell 7: 227238.
Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Tinel A, Tschopp J. (2004). Science 304: 843846.
Augeri DJ, Belli BA et al. (2005). Nature 435: 677681. Tortora G, Caputo R, Damiano V, Caputo R, Troiani T,
Pai SI, Wu GS, Ozoren N, Wu L, Jen J, Sidransky D et al. Veneziani BM et al. (2003). Clin Cancer Res 9: 866871.
(1998). Cancer Res 58: 35133518. Traver D, Akashi K, Weissman IL, Lagasse E. (1998).
Petak I, Danam RP, Tillman DM, Vernes R, Howell SR, Immunity 9: 4757.
Berczi L et al. (2003). Cell Death Differ 10: 211217. Trencia A, Fiory F, Maitan MA, Vito P, Barbagallo APM,
Pitti RM, Marsters SA, Lawrence DA, Roy M, Kischkel FC, Perfetti A et al. (2004). J Biol Chem 279: 4656646572.
Dowd P et al. (1998). Nature 396: 699703. Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce CM.
Pollack IF, Erff M, Ashkenazi A. (2001). Clin Cancer Res 7: (1984). Science 226: 10971099.
13621369. van Loo G, van Gurp M, Depuydt B, Srinivasula SM,
Rampino N, Yamamoto H, Ionov Y, Li Y, Sawai H, Reed JC Rodriguez I, Alnemri ES et al. (2002). Cell Death Differ 9:
et al. (1997). Science 275: 967969. 2026.

Oncogene
Apoptosis pathways in anticancer chemotherapy
S Fulda and K-M Debatin
4811
van Noesel MM, van Bezouw S, Salomons GS, Voute PA, Wu G, Chai J, Suber TL, Wu JW, Du C, Wang X et al. (2000).
Pieters R, Baylin SB et al. (2002). Cancer Res 62: 21572161. Nature 408: 10081012.
Varfolomeev EE, Schuchmann M, Luria V, Chiannilkulchai Yamanaka K, Rocchi P, Miyake H, Fazli L, Vessella B,
N, Beckmann JS, Mett IL et al. (1998). Immunity 9: Zangemeister-Wittke U et al. (2005). Mol Cancer Ther 4:
267276. 16891698.
Vaux DL, Silke J. (2003). Biochem Biophys Res Commun 304: Yang L, Cao Z, Yan H, Wood WC. (2003a). Cancer Res 63:
499504. 68156824.
Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, Yang L, Mashima T, Sato S, Mochizuki M, Sakamoto H,
Reid GE et al. (2000). Cell 102: 4353. Yamori T et al. (2003b). Cancer Res 63: 831837.
Villunger A, Egle A, Kos M, Hartmann BL, Geley S, Koer R Yeh WC, Pompa JL, McCurrach ME, Shu HB, Elia AJ,
et al. (1997). Cancer Res 57: 33313334. Shahinian A et al. (1998). Science 279: 19541958.
Walczak H, Bouchon A, Stahl H, Krammer PH. (2000). Yoshida H, Kong YY, Yoshida R, Elia AJ, Hakem A, Hakem
Cancer Res 60: 30513057. R et al. (1998). Cell 94: 739750.
Walczak H, Krammer PH. (2000). Exp Cell Res 256: 5866. Yu J, Zhang L, Hwang PM, Kinzler KW, Vogelstein B. (2001).
Walensky LD, Kung AL, Escher I, Malia TJ, Barbuto S, Mol Cell 7: 673682.
Wright RD et al. (2004). Science 305: 14661470. Zangemeister-Wittke U, Leech SH, Olie RA, Simoes-Wust
Wang JL, Liu D, Zhang ZJ, Shan S, Han X, Srinivasula SM AP, Gautschi O, Luedke GH et al. (2000). Clin Cancer Res
et al. (2000). Proc Natl Acad Sci USA 97: 71247129. 6: 25472555.
Wang S, El-Deiry WS. (2003). Proc Natl Acad Sci USA 100: Zhao J, Jin J, Zhang X, Shi M, Dai J, Wu MY et al. (2006).
1509515100. Cancer Gene Ther 13: 420427.
Wang Z, Cuddy M, Samuel T, Welsh K, Schimmer A, Hanaii Zinkel SS, Ong CC, Ferguson DO, Iwasaki H, Akashi K,
F et al. (2004). J Biol Chem 279: 4816848176. Bronson RT et al. (2003). Genes Dev 17: 229239.
Waterhouse NJ, Goldstein JC, von Ahsen O, Schuler M, Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. (1997). Cell
Newmeyer DD, Green DR. (2001). J Cell Biol 153: 319328. 90: 405413.

Oncogene

You might also like