Cell Death in Biology and Diseases: Series Editors
Cell Death in Biology and Diseases: Series Editors
Cell Death in Biology and Diseases: Series Editors
Series Editors
Xiao-Ming Yin
Zheng Dong
vii
viii Series Preface
creatively from the initiation and continue this on the development and produc-
tion of each volume of the series. Finally, we hope the readers will enjoy the read-
ing, find the content helpful to their work, and consider this series an invaluable
resource.
Over the past few decades, it has become widely appreciated that genetically
programmed, active cell death processes are critically important for immune-
mediated removal of infected or transformed cells, as well as self-elimination of
damaged cells. This includes removal via cellular suicide of cells that have been
damaged by conventional anti-cancer therapies such as chemotherapy and radia-
tion. Concurrently, it has been determined that defects in cell death pathways can
promote tumor development and progression, including the development of resist-
ance to chemotherapy, radiation, immunotherapies, and biologic agents. These
resistance mechanisms have devastating consequences for the successful treatment
of cancer patients. A key goal in the development of new therapeutic strategies
and agents for the treatment of cancer is to achieve selective and efficient killing
of tumor cells. Assisting this goal are numerous ongoing studies and discoveries
that have elucidated the molecular mechanisms of cell death pathways, including
the extrinsic and intrinsic apoptosis pathways, and the proteins that act to regulate
these pathways. Understanding of these cell death pathways is now making it pos-
sible to identify the specific cell death defects that occur in cancer cells, and is
leading to the development of novel strategies and agents to overcome or correct
these defects in cancer patients.
This volume summarizes current understanding of the molecular mechanisms
that govern cell death in normal and malignant cells, and introduces cutting-edge
opportunities for achieving selective killing of cancer cells by targeting these
mechanisms in patient’s tumors. A particular emphasis is placed on data emerging
from the translation of basic research findings to clinical trials. The book begins
by describing many of the common cell death defects that have been identified
in primary patient specimens, including aberrant overexpression of anti-apoptotic
proteins and oncoproteins, and mutation or loss of expression of pro-apoptotic
proteins and tumor suppressors. The unique bioenergetics of cancer cells and
unique characteristics of the tumor microenvironment are then discussed, along
with opportunities for novel therapeutic interventions that are afforded by these
distinctive features. The role of autophagy in regulating cancer cell death and
current progress in targeting autophagy to improve responsiveness to conven-
tional anti-cancer therapies is also presented. Additionally, a rapidly expanding
field implicates microRNAs in the regulation of cell death proteins and pathways.
ix
x Book Preface
xi
xii Contents
Index. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 403
Contributors
xiii
xiv Contributors
Daniel E. Johnson
The elimination of normal or neoplastic cells via induction of a cell death program
has been recognized since the 1960s, with the term “apoptosis” first being used
to describe this process in 1972 [1]. Early studies defined a number of ordered
morphologic changes in cells undergoing apoptosis, including cell shrinkage,
chromatin condensation, membrane blebbing, and eventual breakup of the cell
into membrane encapsulated apoptotic bodies [1, 2]. In vivo, these apoptotic bod-
ies were observed to be engulfed and degraded by macrophages or neighboring
cells [1, 3, 4]. The early characterization of apoptotic cells also identified a few
D. E. Johnson (*)
Departments of Medicine and Pharmacology and Chemical Biology, University of Pittsburgh
and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
e-mail: [email protected]
Activation of “initiator”
caspases
Activation of
“executioner”
caspases
from a variety of apoptotic stimuli, underscoring the importance of ICE and ICE-
like proteases in apoptosis execution [32–37]. Subsequently, more than a dozen
proteases related to ICE have been cloned and have come to be known as caspases
(reviewed in [38]). The term caspases refers to the fact that these proteases are
“c-asp-ases”. The caspases have conveniently been assigned names of caspase-1
through caspase-14 [38]. Normally, caspases exist as inactive zymogens in cells.
However, in response to an apoptotic stimulus, a subset of caspases, called the initi-
ator caspases (e.g., caspase-8 and caspase-9), undergo processing to active enzyme
forms (Fig. 1.1). The activated initiator caspases then cleave and activate execu-
tioner caspases, of which the most common are caspase-3 and caspase-7 [39, 40].
Activated executioner caspases cleave specific cellular substrate proteins promoting
the eventual destruction of the cell.
Following the identification of caspases as executioners of apoptosis, it was
soon determined that overexpression of Bcl-2 could act to prevent caspase pro-
tease activation in the cell. But how was Bcl-2 preventing caspase activation,
and what are the mechanisms for caspase activation in the absence of Bcl-2?
To address these questions, Wang and colleagues performed cellular fractiona-
tion studies to identify proteins that could promote activation of a procaspase in
cell-free extracts [41]. Remarkably, cytochrome c, a mitochondrial protein, was
found to be capable of promoting caspase activation in conjunction with a sec-
ond protein that was given the name Apaf-1 (apoptotic protease activating factor
1) [41, 42]. Interestingly, Apaf-1 was found to bear close homology with Ced-4
protein from C. elegans [43]. Further studies revealed that during the course of
apoptosis caused by chemotherapy, radiation, or cytokine withdrawal, cytochrome
c is released into the cytosol, and this release is prevented in cells overexpressing
Bcl-2 [41, 42, 44, 45].
A further foundational development in the understanding of apoptosis signal-
ing has come with the identification of the tumor necrosis factor (TNF) family of
death ligands. This family includes proteins such as TNF, Fas ligand, and TRAIL
(TNF-related apoptosis-inducing ligand), which bind to cognate plasma membrane
4 D. E. Johnson
procasp-8/-10 caspase-8/-10
(intrinsic apoptosis pathway)
Chemotherapy/Radiation
Damage
Cleavage of caspase
substrate proteins
cytochrome c
release DNA fragmentation
Cell destruction
caspase-9
Apaf-1
procasp-9
“Apoptosome”
receptors, promoting caspase activation and cell death (see Chap. 11). The identifi-
cation of this receptor-mediated pathway of apoptosis, in conjuction with the early
discoveries regarding Bcl-2, caspases, Apaf-1, and cytochrome c, has provided the
foundation for detailed delineation of the extrinsic and intrinsic pathways of apop-
tosis described in the section below.
Fig. 1.3 DISC formation
and caspase-8 activation
Fas ligand (death ligand)
FADD
Procaspase-8
FADD also contains a domain called a death-effector domain (DED) which then
recruits the DED found in the prodomain of the zymogen form of the initiator cas-
pase, caspase-8 (or the initiator caspase-10). Recruitment of procaspase-8 to the
DISC results in a slight conformational change in the zymogen protein, resulting
in modest activation of the enzyme activity and proximity-induced proteolytic pro-
cessing of procaspase-8 proteins present in the DISC (Fig. 1.3) [46–48]. This pro-
cessing removes the inhibitory prodomain and produces large and small caspase-8
subunits. A heterotetrameric complex consisting of two large subunits and two
small subunits comprises the fully activated caspase enzyme (Fig. 1.3) [38]. Once
the initiator caspases, caspase-8 or caspase-10, are activated, they then cleave
and activate executioner caspases, including the primary executioner, caspase-3
(Fig. 1.2) [38]. The activated executioners cleave specific substrate proteins,
resulting in activation of cellular DNAses and proteolytic destruction of the cell.
The intrinsic apoptosis pathway is activated by a variety of cellular insults,
including withdrawal of essential cytokines or neurotrophic factors, or treat-
ment with chemotherapy or radiation. In the case of agents causing cellular
damage (e.g., chemotherapy drugs), the damage is detected by the cell, and a
signal (described in Sect. 1.4.3) is relayed to the mitochondria, causing release
of cytochrome c into the cytosol (Fig. 1.2) [41]. Once released, the cytosolic
cytochrome c associates with the cytoplasmic adapter protein Apaf-1 and the
zymogen form of the initiator caspase, caspase-9, forming a complex referred
to as the apoptosome [42]. Formation of the apoptosome results in a slight con-
formational change in procaspase-9, causing sufficient activation to promote
autoprocessing to active caspase-9 [48]. Active caspase-9 cleaves and activates
downstream executioner caspases, and at this point, the intrinsic and extrinsic
apoptosis pathways converge. In some instances, earlier cross-talk between the
intrinsic and extrinsic pathways can occur. Active caspase-8 from the extrinsic
pathway is known to cleave Bid protein, and the Bid cleavage product (tBid) can
6 D. E. Johnson
procasp-8/-10 caspase-8/-10
Chemotherapy/Radiation
Damage
Cleavage of caspase
substrate proteins
cytochrome c
release X DNA fragmentation
Cell destruction
caspase-9
Apaf-1
procasp-9
“Apoptosome”
In view of the critical importance of caspases to both the extrinsic and intrinsic
apoptosis pathways, one might predict that the genes encoding these proteases
would be targets for genetic alterations impacting the function or expression of
caspases in human tumors. Indeed, a considerable body of evidence indicates that
this is likely the case, particularly for the genes encoding the initiator caspases,
caspase-8 and caspase-10. A smaller number of publications also point to muta-
tions or reduced expression of the initiator, caspase-9, and the executioners, cas-
pase-3 and caspase-7.
1 Defective Apoptosis Signaling in Cancer 7
Fig. 1.5 XIAP inhibition of
caspases in healthy cells
XIAP
procasp-3 caspase-3
X
Cleavage of caspase
X substrate proteins
DNA fragmentation
Cell destruction
caspase-9
XIAP
detected somatic mutations of the caspase-3 gene in 4/98 (4.1 %) colon carcino-
mas, 4/181 (2.2 %) non-small-cell lung cancers (NSCLCs), 2/129 (1.6 %) non-
Hodgkin’s lymphomas, 1/28 (3.6 %) multiple myelomas, 1/80 (1.3 %) HCCs, and
2/165 (1.2 %) gastric carcinomas [76]. Inactivating mutations of caspase-7 have
been reported in 1/33 (3.0 %) head and neck carcinomas, 1/50 (2.0 %) esophageal
carcinomas, and 2/98 (2.0 %) colon carcinomas [77].
The intracellular activities of the initiator, caspase-9, and the executioners, caspase-3
and caspase-7, are negatively regulated by certain members of the inhibitor of
apoptosis (IAP) protein family (reviewed in [78, 79] and Chap. 10). The IAP fam-
ily is comprised of eight members (XIAP, cIAP1, cIAP2, NAIP, survivin, MLIAP,
BRUCE, and ILP2), with each characterized by the presence of 1 or 3 baculovi-
rus IAP repeat (BIR) domains (see Chap. 10). Four members of this family (XIAP,
cIAP1, cIAP2, MLIAP2, and ILP2) also contain carboxy-terminal RING domains,
which possess E3 ubiquitin ligase activity. Among the IAPs, XIAP has potent and
direct inhibitory activity against caspase-9, caspase-3, and caspase-7, while cIAP1
and cIAP2 can act to indirectly inhibit the activities of these caspases [80].
XIAP directly binds active caspase-3 and caspase-7 with high affinity via two
interaction sites. The BIR2 domain of XIAP binds to an IBM (IAP-binding motif)
that becomes exposed in the processed/active proteases [81, 82]. In addition, a
linker region between the BIR1 and BIR2 domains binds to the active site of cas-
pase-3 and caspase-7 and prevents the access of caspase substrate proteins [81–84].
In the case of caspase-9, the BIR3 domain of XIAP binds to the monomeric form
of caspase-9, preventing dimerization and full activation of the enzyme [85, 86].
As shown in Fig. 1.5, XIAP-mediated inhibition of any caspase-9, caspase-3,
or caspase-7 activities present in healthy growing cells prevents undesirable
1 Defective Apoptosis Signaling in Cancer 9
Damage
Cleavage of caspase
substrate proteins
cytochrome c &
SMAC release DNA fragmentation
Cell destruction
caspase-9
Apaf-1
procasp-9
XIAP
“Apoptosome”
activation of the caspase protease cascade and death of the cells. However, dur-
ing activation of the intrinsic apoptosis pathway, the mitochondrial protein SMAC
(second mitochondria-derived activator of caspases), is released from the mito-
chondria in conjunction with cytochrome c. The amino-terminal domain of the
processed SMAC protein contains a tetrapeptide sequence (AVPI) that binds
tightly to XIAP and displaces XIAP from bound caspases, leading to the activa-
tion of caspase-9, as well as caspase-3 and caspase-7 (see Fig. 1.6; reviewed in
Chap. 10). While this is the scenario in cells that efficiently undergo apoptosis via
the intrinsic apoptosis pathway, it is now evident that many tumors overexpress
XIAP (discussed below), thwarting the ability of limiting levels of SMAC to over-
come XIAP inhibition of the caspase cascade (see Fig. 1.7).
cIAP1 and cIAP2 act to indirectly inhibit caspase-9, caspase-3, and caspase-7
via two distinct mechanisms. First, the cIAP1 and cIAP2 proteins are capable of
binding to SMAC and can thereby sequester SMAC protein and prevent displace-
ment of XIAP from bound caspases [80]. Second, acting via their RING domains,
cIAP1, cIAP2, as well as XIAP, can promote ubiquitination of caspase-3 and cas-
pase-7 [87–90]. The ubiquitination of these executioner caspases results in inhibi-
tion of their activities and their eventual degradation via the proteasome.
Aberrant overexpression of XIAP, as well as other IAPs, has been observed in
multiple solid tumor and hematopoietic malignancies. In several cases, overex-
pression of XIAP has been shown to correlate with treatment resistance or poor
prognosis. XIAP is overexpressed in HCC, where its expression has been linked
to metastasis, recurrence, and poor prognosis [91–93]. In advanced head and neck
cancer, XIAP correlates with resistance to cisplatin-based regimens and poor
prognosis [94]. Similarly, XIAP overexpression correlates with chemoresistance
10 D. E. Johnson
X
Damage
Cleavage of caspase
X substrate proteins
cytochrome c &
SMAC release DNA fragmentation
Cell destruction
caspase-9
Apaf-1
procasp-9 IAPs
IAPs
IAPs IAPs
XIAP
“Apoptosome” IAPs
XIAP
Early efforts to uncover the molecular mechanism whereby Bcl-2 acts to inhibit
apoptosis were stymied by the failure to identify any enzymatic activity associ-
ated with the Bcl-2 protein. The first clue regarding the mechanism of Bcl-2 action
came with the discovery that Bcl-2 binds tightly to the closely related proapop-
totic protein Bax [184]. Subsequent studies have revealed a complex network of
physical interactions between anti-apoptotic Bcl-2 family members and proapop-
totic members of the Bcl-2 protein family [114, 115, 126, 185]. Additional stud-
ies determined that heterodimerization between pro- and anti-apoptotic Bcl-2
family members served to neutralize the apoptosis-inducing activity of the proa-
poptotic protein, and led to the proposal of the “rheostat model” by Korsmeyer
and colleagues [186]. The rheostat model proposed that the relative levels of pro-
versus anti-apoptotic Bcl-2 family members expressed in a cell would determine
whether that cell is prone to undergo apoptosis (i.e., chemotherapy sensitive), or
1 Defective Apoptosis Signaling in Cancer 13
prone to be apoptosis resistant. The second major clue regarding the mechanism
of Bcl-2 action came with the discovery discussed previously that Bcl-2 (and Bcl-
XL/Mcl-1) acts to prevent cytochrome c release from the mitochondria [44, 45].
The third major clue came from biophysical and biochemical studies of proapop-
totic Bax and Bak. These studies revealed that Bax and Bak can homooligomer-
ize and form small pores in artificial lipid membranes, as well as mitochondrial
membranes in intact cells [187–191]. The pores formed by Bax and Bak were
found to be sufficient in size to allow the release of cytochrome c from mitochon-
dria. The binding of anti-apoptotic Bcl-2 proteins to Bax or Bak prevents their
homooligomerization, explaining how these anti-apoptotic proteins can prevent
cytochrome c release and activation of the intrinsic apoptosis pathway. A large
number of additional studies have shown that certain “BH3 domain-only” proap-
optotic proteins, such as PUMA, tBid, and Bim, can bind directly to Bax or Bak
and induce pore formation. These “BH3 domain-only” proteins are commonly
referred to as “activators” [117, 192–195]. The other “BH3 domain-only” proap-
optotic proteins are called “derepressors” (or “sensitizers”) and exert their effects
by binding to the anti-apoptotic Bcl-2 family members (e.g., Bcl-2, Bcl-XL, and
Mcl-1) [117, 192–195]. In binding to anti-apoptotic proteins, the “derepressors,”
as well as “activators,” cause release of sequestered Bax and Bak, freeing these
proteins up to form pores. In addition, “derepressor” binding can cause the release
of bound “activators,” freeing them up to directly bind and activate Bax and Bak
[196–198].
Just as anti-apoptotic Bcl-2 family members are found to be frequently over-
expressed in human cancers, it is reasonable to predict that the proapoptotic Bcl-2
proteins might be underexpressed or functionally inactivated in tumors. Indeed,
this appears to be the case [117, 199]. Frameshift and inactivating mutations in
the bax gene have been detected in colorectal and hematopoietic malignancies
[200–202]. Mutation of noxa gene has been reported in DLBCL [203], while
bak gene mutations are present in gastric and colon tumors [204]. In mantle cell
lymphomas (MCL), homozygous deletion of bim gene has been observed [205].
Loss of heterozygosity in bik gene occurs in renal cell carcinoma [206].
Promoter methylation also appears to play an important role in reducing the
expression of proapoptotic proteins in cancer. Hypermethylation of bim gene pro-
moter has been reported in Burkitt’s lymphoma (BL) and DLBCL [203, 207];
hypermethylation of puma gene occurs in BL [208]; and hypermethylation of noxa
gene is found in DLBCL [203]. Hypermethylation of bik gene is present in renal
cell carcinomas [206], while colorectal tumors exhibit hypermethylation of hrk
gene [209].
Emerging evidence indicates that expression of proapoptotic proteins is likely
regulated by the expression of specific microRNAs (see also Chap. 5). Expression
of Bim has been found to be downregulated in several types of cancer via
expression of microRNAs encoded by the miR-17-92 and miR-106b-25 clusters
[210–213]. It appears highly likely that regulation of the expression of pro- and
anti-apoptotic proteins by microRNAs will remain an active area of research in the
coming years.
14 D. E. Johnson
Bak
Bak
cytochrome c
release
Bax
Bax
Fig. 1.8 p53 induces PUMA and NOXA to activate the intrinsic apoptosis pathway
1.4.3 Mutation/Deletion of p53
The tumor suppressor protein p53 is often cited as the most commonly mutated
protein in human cancers [214]. Mutations in the p53 gene lead to loss of p53
function as a transcription factor, with frequent conversion to a dominant-negative
inhibitor, or loss of expression of the p53 protein [214]. The prevalence of p53
mutation or loss of expression in cancer is likely due to the critical roles that p53
plays in the regulation of cell cycle arrest, DNA repair, apoptosis, and senescence.
p53 actively promotes apoptosis induction via the intrinsic apoptosis pathway,
particularly in response to DNA damage. In damaged cells, activation of ATM and
ATR kinases leads to phosphorylation of p53 and MDM2, disrupting p53/MDM2
interactions and abrogating MDM2-mediated degradation of p53 via the proteas-
ome [215–221]. This leads to upregulation of the p53 protein, with the majority of
the protein being expressed in the nucleus, but some also appearing in the cytosol
and at the mitochondria.
Upregulated p53 protein in damaged cells activates the intrinsic apoptosis path-
way via both transcription-dependent and transcription-independent mechanisms.
The transcription-dependent mechanism requires functional DNA binding and
transcription factor function of the p53 protein. The genes encoding Bax and Apaf-
1 have been shown to be p53 target genes, and their transcription is moderately
induced in p53-expressing cells [222, 223]. However, although Bax and Apaf-1
are central factors in the intrinsic pathway, p53 induction of these proteins may
play only a modest role in the proapoptotic activity of p53. By contrast, p53 induc-
tion of select proapoptotic “BH3 domain-only” proteins appears to play a major
role in the transcription-dependent mechanism of apoptosis induction by p53.
p53 markedly induces the expression of the “BH3 domain-only” proteins PUMA
[224, 225], NOXA [226], and BID [227]. As illustrated in Fig. 1.8, p53-mediated
1 Defective Apoptosis Signaling in Cancer 15
induction of PUMA and NOXA results in migration of these proteins to the mito-
chondria, where PUMA can directly activate Bax or Bak, or indirectly activate
these proteins via inhibitory interaction with anti-apoptotic Bcl-2 family members,
including Bcl-2 and Bcl-XL [197, 198, 228–230]. Induced NOXA demonstrates
specificity for binding to anti-apoptotic Mcl-1 and functionally inactivates Mcl-1
by promoting the release of bound proapoptotic proteins [193, 196–198, 229–231].
Transcription-independent activation of the intrinsic apoptosis pathway by p53
occurs via several different mechanisms. Cytosolic p53 directly activates proapop-
totic Bax, leading to mitochondrial membrane permeabilization and cytochrome
c release [232, 233]. p53 present at the mitochondria physically associates with
proapoptotic Bak, disrupting Bak sequestration by Mcl-1 and resulting in Bak acti-
vation [234]. In addition, mitochondrial-localized p53 can bind to Bcl-2 or Bcl-
XL, promoting the release of sequestered proapoptotic proteins [233, 235].
Collectively, the combination of transcription-dependent mechanisms mediated
by nuclear p53 and transcription-independent mechanisms mediated by cytosolic/
mitochondrial p53 confers a potent impact of p53 upregulation on cellular apop-
tosis. Moreover, it is readily apparent why loss of p53 can provide an important
survival advantage for tumors and may enhance resistance to chemotherapy and
radiation. However, it is important to note that activation of the intrinsic apoptosis
pathway is not entirely dependent on p53, as activation of this pathway can also
be observed in some cell types lacking p53. The mechanisms of p53-independ-
ent activation of the intrinsic apoptosis pathway are less well understood, and this
remains an area of active investigation.
Similar to what is seen with the intrinsic apoptosis pathway, multiple defects in
the extrinsic (or death receptor-mediated) pathway have been observed in human
tumors. In healthy individuals, the extrinsic apoptosis pathway plays a central
role in immune-mediated elimination of infected or transformed cells. Therefore,
defects in the extrinsic pathway most likely contribute to tumorigenesis primar-
ily by negatively impacting the efficiency of immune surveillance. However, it
is also important to note that in certain cell types, cross-talk occurs between the
extrinsic and intrinsic pathways, meaning defects in one pathway may affect
killing initiated by the other pathway. For example, in cells referred to as type I
cells, stimulation with a death ligand leads to efficient killing via a linear pathway
involving caspase-8 activation, followed by caspase-3 activation. By contrast, in
cells referred to as type II cells, efficient killing by a death ligand also requires
caspase-8-mediated cleavage of Bid, followed by tBid induction of cytochrome c
release and activation of the intrinsic pathway [49, 50, 236]. Thus, in type II cells,
defects in the intrinsic pathway can impair killing initiated by the extrinsic path-
way. In addition, other reports have indicated that defects in the extrinsic pathway
may reduce the sensitivity of leukemic cells to chemotherapy [237–240].
16 D. E. Johnson
Similar to what is seen for caspase-8 and caspase-10, the expression and function
of Fas death receptor is commonly altered in autoimmune disease and malignant
tumors (reviewed in [241]; see also Chap. 11). In the lpr strain of mice, sequences
from the 3′ LTR of a transposable element are inserted into the second exon of
the fas gene, resulting in expression of defective Fas protein [242–245]. The lpr
mice are characterized by ineffective deletion of self-reactive T cells, lymphopro-
liferation, and development of autoimmune disease [242]. In humans, somatic Fas
mutations are also commonly detected in patients with autoimmune lymphoprolif-
erative syndrome (ALPS) [241, 246, 247]. Loss of heterozygosity in fas gene has
also been reported in human ALPS [248, 249]. Mutations in Fas, typically lead-
ing to an inactive protein, have been detected in both hematologic and solid tumor
malignancies. In hematologic malignancies, Fas mutations have been reported in
cutaneous T cell [250, 251], MALT-type [252, 253], nasal NK/T cell [254], non-
Hodgkin’s [255], DLBC [256] and thyroid lymphomas [257], as well as multiple
myeloma [258]. Solid tumors harboring Fas mutations include bladder carcinoma
[259], malignant melanoma [260], NSCLC [261, 262], squamous cell carcinoma
[263], and cancers of the stomach [264] and testis [265]. Methylation of the fas
gene may be an important mechanism for downregulation of Fas expression in
human tumors, with hypermethylation having been reported in Sezary syndrome
[266] and cancers of the bladder [267], colon [268], and prostate [269].
Apoptosis induced by the death ligand TRAIL is mediated by the death
receptors TRAIL-R1 (also called DR4) and TRAIL-R2 (also called DR5)
(reviewed in [270–272]; see also Chap. 11). As might be expected, reduced
expression and mutation of TRAIL-R1 and TRAIL-R2 has been reported in a
variety of cancers. Mutations in TRAIL-R1 have been detected in non-Hodg-
kin’s lymphoma [273], metastatic breast cancer [274], gastric carcinoma [275],
head and neck cancer [275], NSCLC [275], and osteosarcoma [276]. Mutations
in TRAIL-R2 occur in non-Hodgkin’s lymphoma [273], metastatic breast can-
cer [274], gastric carcinoma [277], and NSCLC [278]. Loss of heterozygosity
in the TRAIL-R2 has been reported in colorectal carcinoma [279], gastric car-
cinoma [277], and NSCLC [278]. Hypermethylation of the TRAIL-R1 gene has
been observed in gastric carcinoma [280], glioma [281], SCLC [282], and ovar-
ian cancer [283].
1 Defective Apoptosis Signaling in Cancer 17
1.5.3 FADD Mutation
The FADD adaptor protein plays an important role in caspase-8 activation fol-
lowing stimulation of the receptors for Fas ligand, TNF, and TRAIL. During Fas-
and TRAIL receptor-mediated activation of caspase-8, FADD is the first protein
recruited to the DISC. During TNF receptor-mediated activation of caspase-8,
FADD recruitment to the DISC follows the recruitment of RIP (receptor-inter-
acting protein), TRADD (TNF receptor-associated death domain protein), and
TRAF-1/-2 (TNF receptor-associated factor). Mutation of FADD protein has been
occasionally observed. Bolze et al. identified a missense mutation in FADD in an
ALPS patient that leads to reduced FADD protein levels and impaired apopto-
sis induction following Fas stimulation [294]. In addition, FADD mutations have
been reported in 4/80 NSCLC patients [262] and 1/98 colon cancer patients [295].
18 D. E. Johnson
Just as anti-apoptotic Bcl-2 family members and IAPs act as endogenous negative
regulators of cytochrome c release and caspase activities, respectively, the c-FLIP
(cellular FLICE-inhibitory protein) proteins are endogenous inhibitors of death
receptor-mediated caspase-8/caspase-10 activation. Three major c-FLIP pro-
tein isoforms have been reported, c-FLIPL, c-FLIPs, and c-FLIPr (reviewed in
[116, 296, 297]). Similar to procaspase-8 and procaspase-10, all 3 c-FLIP iso-
forms contain two amino-terminal DED domains. The c-FLIPs and c-FLIPr con-
tain only a short carboxyl-terminal sequence following the DED domains. By
contrast, the DED domains of c-FLIPL are followed by sequences closely homol-
ogous to the caspase-8 enzyme, although c-FLIPL is catalytically inactive. A
large number of publications have shown that high levels of c-FLIPL, c-FLIPs, or
c-FLIPr effectively compete with procaspase-8 and procaspase-10 for binding to
FADD or TRADD proteins present in the forming DISC of activated death recep-
tors [296, 297]. In so doing, high levels of the c-FLIP proteins effectively prevent
caspase-8/caspase-10 activation and execution of the extrinsic apoptosis pathway.
Conversely, when expressed at only low levels, c-FLIPL has been shown to heter-
odimerize with procaspase-8, promoting caspase-8 activation [298, 299].
Overexpression of c-FLIP proteins, particularly c-FLIPL, has been detected
in a variety of human tumors, rendering the cancer cells more resistant to apop-
tosis induction by death ligands. Specifically, c-FLIP is overexpressed in breast,
colon, endometrial, liver, ovarian, and prostate cancers, as well as melanomas,
glioblastomas, and NSCLCs (reviewed in [116, 296, 297]). Importantly, high lev-
els of c-FLIPL expression have been determined to correlate with tumor progres-
sion or poor prognosis in colon, endometrial, liver, ovarian, and prostate cancers,
as well as BL [116, 296, 297, 300–302]. The prevalence of c-FLIP overexpres-
sion in human tumors, and the impact of these proteins on the extrinsic apop-
tosis pathway, has made the development of c-FLIP inhibitors an area of active
investigation.
1.6 Conclusions
multiple components have been identified that act to negatively regulate apoptosis
(e.g., anti-apoptotic Bcl-2 family members, IAPs, c-FLIP, etc.). Detailed analyses
have determined that several of the proapoptotic proteins involved in the extrin-
sic and intrinsic pathways commonly exhibit reduced expression in a wide vari-
ety of human tumors via loss of gene heterozygosity or promoter methylation.
Additionally, the proapoptotic proteins also exhibit frequent functional inactivation
via mutation. By contrast, many malignancies overexpress the anti-apoptotic pro-
teins that negatively regulate the extrinsic and intrinsic pathways. These findings
are stimulating a flurry of drug discovery efforts (described in subsequent Chapters)
aimed at restoring the expression or activities of proapoptotic proteins, or reduc-
ing the expression or inhibiting the activities of anti-apoptotic proteins in patient
tumors. Moreover, by identifying the location of defects within the apoptosis path-
ways, treatment options can focus on either correcting the defect, or activating the
pathway downstream from the molecular block. Ultimately, the rapid determination
of the apoptosis defects present in a patient’s tumor should allow the development
of effective personalized treatments to restore sensitivity to cell death–inducing
therapies such as chemotherapy, radiation, and therapeutic death ligands.
Acknowledgments This work was supported by National Institutes of Health grants R01
CA137260 and P50 CA097190. A vast number of researchers have made important contributions
to the work described in this review. We apologize to those authors whose work we have not cited.
References
1. Kerr JF, Wyllie AH, Currie AR (1972) Apoptosis: a basic biological phenomenon with
wide-ranging implications in tissue kinetics. Br J Cancer 26(4):239–257
2. Wyllie AH, Kerr JF, Currie AR (1980) Cell death: the significance of apoptosis. Int Rev
Cytol 68:251–306
3. Savill J, Fadok V, Henson P, Haslett C (1993) Phagocyte recognition of cells undergoing
apoptosis. Immunol Today 14(3):131–136
4. Savill J, Fadok V (2000) Corpse clearance defines the meaning of cell death. Nature
407(6805):784–788
5. Wyllie AH, Morris RG, Smith AL, Dunlop D (1984) Chromatin cleavage in apoptosis:
association with condensed chromatin morphology and dependence on macromolecular
synthesis. J Pathol 142(1):67–77
6. Fukuhara S, Rowley JD, Variakojis D, Golomb HM (1979) Chromosome abnormalities in
poorly differentiated lymphocytic lymphoma. Cancer Res 39(8):3119–3128
7. Yunis JJ, Frizzera G, Oken MM, McKenna J, Theologides A, Arnesen M (1987) Multiple
recurrent genomic defects in follicular lymphoma. A possible model for cancer. N Engl J
Med 316(2):79–84
8. Cleary ML, Sklar J (1985) Nucleotide sequence of a t(14;18) chromosomal breakpoint in
follicular lymphoma and demonstration of a breakpoint-cluster region near a transcription-
ally active locus on chromosome 18. Proc Natl Acad Sci U S A 82(21):7439–7443
9. Tsujimoto Y, Cossman J, Jaffe E, Croce CM (1985) Involvement of the bcl-2 gene in
human follicular lymphoma. Science 228(4706):1440–1443
10. Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce CM (1984) Cloning of the chromo-
some breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science
226(4678):1097–1099
20 D. E. Johnson
11. Cleary ML, Smith SD, Sklar J (1986) Cloning and structural analysis of cDNAs for bcl-2
and a hybrid bcl-2/immunoglobulin transcript resulting from the t(14;18) translocation.
Cell 47(1):19–28
12. Vaux DL, Cory S, Adams JM (1988) Bcl-2 gene promotes haemopoietic cell survival and
cooperates with c-myc to immortalize pre-B cells. Nature 335(6189):440–442
13. Nunez G, London L, Hockenbery D, Alexander M, McKearn JP, Korsmeyer SJ (1990)
Deregulated Bcl-2 gene expression selectively prolongs survival of growth factor-deprived
hemopoietic cell lines. J Immunol 144(9):3602–3610
14. Hockenbery D, Nunez G, Milliman C, Schreiber RD, Korsmeyer SJ (1990) Bcl-2 is
an inner mitochondrial membrane protein that blocks programmed cell death. Nature
348(6299):334–336
15. Garcia I, Martinou I, Tsujimoto Y, Martinou JC (1992) Prevention of programmed cell
death of sympathetic neurons by the bcl-2 proto-oncogene. Science 258(5080):302–304
16. Allsopp TE, Wyatt S, Paterson HF, Davies AM (1993) The proto-oncogene bcl-2 can
selectively rescue neurotrophic factor-dependent neurons from apoptosis. Cell 73(2):
295–307
17. Mah SP, Zhong LT, Liu Y, Roghani A, Edwards RH, Bredesen DE (1993) The protoonco-
gene bcl-2 inhibits apoptosis in PC12 cells. J Neurochem 60(3):1183–1186
18. Sentman CL, Shutter JR, Hockenbery D, Kanagawa O, Korsmeyer SJ (1991) bcl-2 inhibits
multiple forms of apoptosis but not negative selection in thymocytes. Cell 67(5):879–888
19. Strasser A, Harris AW, Cory S (1991) bcl-2 transgene inhibits T cell death and perturbs
thymic self-censorship. Cell 67(5):889–899
20. Alnemri ES, Fernandes TF, Haldar S, Croce CM, Litwack G (1992) Involvement of BCL-2
in glucocorticoid-induced apoptosis of human pre-B-leukemias. Cancer Res 52(2):491–495
21. Walton MI, Whysong D, O’Connor PM, Hockenbery D, Korsmeyer SJ, Kohn KW (1993)
Constitutive expression of human Bcl-2 modulates nitrogen mustard and camptothecin
induced apoptosis. Cancer Res 53(8):1853–1861
22. Miyashita T, Reed JC (1993) Bcl-2 oncoprotein blocks chemotherapy-induced apoptosis in
a human leukemia cell line. Blood 81(1):151–157
23. McDonnell TJ, Deane N, Platt FM, Nunez G, Jaeger U, McKearn JP et al (1989) bcl-2-im-
munoglobulin transgenic mice demonstrate extended B cell survival and follicular lym-
phoproliferation. Cell 57(1):79–88
24. McDonnell TJ, Korsmeyer SJ (1991) Progression from lymphoid hyperplasia to high-grade
malignant lymphoma in mice transgenic for the t(14; 18). Nature 349(6306):254–256
25. Hengartner MO, Ellis RE, Horvitz HR (1992) Caenorhabditis elegans gene ced-9 protects
cells from programmed cell death. Nature 356(6369):494–499
26. Vaux DL, Weissman IL, Kim SK (1992) Prevention of programmed cell death in
Caenorhabditis elegans by human bcl-2. Science 258(5090):1955–1957
27. Ellis HM, Horvitz HR (1986) Genetic control of programmed cell death in the nematode C.
elegans. Cell 44(6):817–829
28. Yuan J, Shaham S, Ledoux S, Ellis HM, Horvitz HR (1993) The C. elegans cell death gene
ced-3 encodes a protein similar to mammalian interleukin-1 beta-converting enzyme. Cell
75(4):641–652
29. Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ et al
(1992) A novel heterodimeric cysteine protease is required for interleukin-1 beta
processing in monocytes. Nature 356(6372):768–774
30. Cerretti DP, Kozlosky CJ, Mosley B, Nelson N, Van Ness K, Greenstreet TA et al (1992)
Molecular cloning of the interleukin-1 beta converting enzyme. Science 256(5053):97–100
31. Miura M, Zhu H, Rotello R, Hartwieg EA, Yuan J (1993) Induction of apoptosis in fibro-
blasts by IL-1 beta-converting enzyme, a mammalian homolog of the C. elegans cell death
gene ced-3. Cell 75(4):653–660
32. Nicholson DW, Ali A, Thornberry NA, Vaillancourt JP, Ding CK, Gallant M et al
(1995) Identification and inhibition of the ICE/CED-3 protease necessary for mammalian
apoptosis. Nature 376(6535):37–43
1 Defective Apoptosis Signaling in Cancer 21
33. Datta R, Banach D, Kojima H, Talanian RV, Alnemri ES, Wong WW et al (1996)
Activation of the CPP32 protease in apoptosis induced by 1-beta-D-arabinofuranosylcyto-
sine and other DNA-damaging agents. Blood 88(6):1936–1943
34. Kondo S, Barna BP, Morimura T, Takeuchi J, Yuan J, Akbasak A et al (1995) Interleukin-1
beta-converting enzyme mediates cisplatin-induced apoptosis in malignant glioma cells.
Cancer Res 55(24):6166–6171
35. Zhu H, Fearnhead HO, Cohen GM (1995) An ICE-like protease is a common media-
tor of apoptosis induced by diverse stimuli in human monocytic THP.1 cells. FEBS Lett
374(2):303–308
36. An B, Dou QP (1996) Cleavage of retinoblastoma protein during apoptosis: an interleukin
1 beta-converting enzyme-like protease as candidate. Cancer Res 56(3):438–442
37. Dou QP, An B, Antoku K, Johnson DE (1997) Fas stimulation induces RB dephospho-
rylation and proteolysis that is blocked by inhibitors of the ICE protease family. J Cell
Biochem 64(4):586–594
38. Riedl SJ, Shi Y (2004) Molecular mechanisms of caspase regulation during apoptosis. Nat
Rev Mol Cell Biol 5(11):897–907
39. Stennicke HR, Jurgensmeier JM, Shin H, Deveraux Q, Wolf BB, Yang X et al (1998) Pro-
caspase-3 is a major physiologic target of caspase-8. J Biol Chem 273(42):27084–27090
40. Slee EA, Harte MT, Kluck RM, Wolf BB, Casiano CA, Newmeyer DD et al
(1999) Ordering the cytochrome c-initiated caspase cascade: hierarchical activa-
tion of caspases-2, -3, -6, -7, -8, and -10 in a caspase-9-dependent manner. J Cell Biol
144(2):281–292
41. Liu X, Kim CN, Yang J, Jemmerson R, Wang X (1996) Induction of apoptotic program in
cell-free extracts: requirement for dATP and cytochrome c. Cell 86(1):147–157
42. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES et al (1997)
Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an
apoptotic protease cascade. Cell 91(4):479–489
43. Zou H, Henzel WJ, Liu X, Lutschg A, Wang X (1997) Apaf-1, a human protein homolo-
gous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3.
Cell 90(3):405–413
44. Yang J, Liu X, Bhalla K, Kim CN, Ibrado AM, Cai J et al (1997) Prevention of
apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science
275(5303):1129–1132
45. Kluck RM, Bossy-Wetzel E, Green DR, Newmeyer DD (1997) The release of cytochrome
c from mitochondria: a primary site for Bcl-2 regulation of apoptosis. Science
275(5303):1132–1136
46. Muzio M, Stockwell BR, Stennicke HR, Salvesen GS, Dixit VM (1998) An induced prox-
imity model for caspase-8 activation. J Biol Chem 273(5):2926–2930
47. Martin DA, Siegel RM, Zheng L, Lenardo MJ (1998) Membrane oligomerization and
cleavage activates the caspase-8 (FLICE/MACHalpha1) death signal. J Biol Chem
273(8):4345–4349
48. Yang X, Chang HY, Baltimore D (1998) Autoproteolytic activation of pro-caspases by oli-
gomerization. Mol Cell 1(2):319–325
49. Li H, Zhu H, Xu CJ, Yuan J (1998) Cleavage of BID by caspase 8 mediates the mitochon-
drial damage in the Fas pathway of apoptosis. Cell 94(4):491–501
50. Luo X, Budihardjo I, Zou H, Slaughter C, Wang X (1998) Bid, a Bcl2 interacting protein,
mediates cytochrome c release from mitochondria in response to activation of cell surface
death receptors. Cell 94(4):481–490
51. Mandruzzato S, Brasseur F, Andry G, Boon T, van der Bruggen P (1997) A CASP-8 muta-
tion recognized by cytolytic T lymphocytes on a human head and neck carcinoma. J Exp
Med 186(5):785–793
52. Soung YH, Lee JW, Kim SY, Sung YJ, Park WS, Nam SW et al (2005) Caspase-8 gene is
frequently inactivated by the frameshift somatic mutation 1225_1226delTG in hepatocel-
lular carcinomas. Oncogene 24(1):141–147
22 D. E. Johnson
53. Kim HS, Lee JW, Soung YH, Park WS, Kim SY, Lee JH et al (2003) Inactivating mutations
of caspase-8 gene in colorectal carcinomas. Gastroenterology 125(3):708–715
54. Soung YH, Lee JW, Kim SY, Jang J, Park YG, Park WS et al (2005) CASPASE-8 gene is
inactivated by somatic mutations in gastric carcinomas. Cancer Res 65(3):815–821
55. Teitz T, Wei T, Valentine MB, Vanin EF, Grenet J, Valentine VA et al (2000) Caspase 8
is deleted or silenced preferentially in childhood neuroblastomas with amplification of
MYCN. Nat Med 6(5):529–535
56. Banelli B, Casciano I, Croce M, Di Vinci A, Gelvi I, Pagnan G et al (2002) Expression
and methylation of CASP8 in neuroblastoma: identification of a promoter region. Nat Med
8(12):1333–1335, author reply 5
57. Lazcoz P, Munoz J, Nistal M, Pestana A, Encio I, Castresana JS (2006) Frequent promoter
hypermethylation of RASSF1A and CASP8 in neuroblastoma. BMC Cancer 6:254
58. Fulda S, Poremba C, Berwanger B, Hacker S, Eilers M, Christiansen H et al (2006) Loss of
caspase-8 expression does not correlate with MYCN amplification, aggressive disease, or
prognosis in neuroblastoma. Cancer Res 66(20):10016–10023
59. Yang Q, Kiernan CM, Tian Y, Salwen HR, Chlenski A, Brumback BA et al (2007)
Methylation of CASP8, DCR2, and HIN-1 in neuroblastoma is associated with poor out-
come. Clin Cancer Res 13(11):3191–3197
60. Kamimatsuse A, Matsuura K, Moriya S, Fukuba I, Yamaoka H, Fukuda E et al (2009)
Detection of CpG island hypermethylation of caspase-8 in neuroblastoma using an oligo-
nucleotide array. Pediatr Blood Cancer 52(7):777–783
61. Harada K, Toyooka S, Shivapurkar N, Maitra A, Reddy JL, Matta H et al (2002)
Deregulation of caspase 8 and 10 expression in pediatric tumors and cell lines. Cancer Res
62(20):5897–5901
62. Gonzalez-Gomez P, Bello MJ, Inda MM, Alonso ME, Arjona D, Aminoso C et al (2004)
Deletion and aberrant CpG island methylation of Caspase 8 gene in medulloblastoma.
Oncol Rep 12(3):663–666
63. Pingoud-Meier C, Lang D, Janss AJ, Rorke LB, Phillips PC, Shalaby T et al (2003) Loss
of caspase-8 protein expression correlates with unfavorable survival outcome in childhood
medulloblastoma. Clin Cancer Res 9(17):6401–6409
64. Bello MJ, De Campos JM, Isla A, Casartelli C, Rey JA (2006) Promoter CpG methylation
of multiple genes in pituitary adenomas: frequent involvement of caspase-8. Oncol Rep
15(2):443–448
65. Malekzadeh K, Sobti RC, Nikbakht M, Shekari M, Hosseini SA, Tamandani DK et al
(2009) Methylation patterns of Rb1 and Casp-8 promoters and their impact on their expres-
sion in bladder cancer. Cancer Invest 27(1):70–80
66. Liedtke C, Zschemisch NH, Cohrs A, Roskams T, Borlak J, Manns MP et al (2005)
Silencing of caspase-8 in murine hepatocellular carcinomas is mediated via methylation of
an essential promoter element. Gastroenterology 129(5):1602–1615
67. Cho S, Lee JH, Cho SB, Yoon KW, Park SY, Lee WS et al (2010) Epigenetic methylation and
expression of caspase 8 and survivin in hepatocellular carcinoma. Pathol Int 60(3):203–211
68. Shivapurkar N, Toyooka S, Eby MT, Huang CX, Sathyanarayana UG, Cunningham HT et al
(2002) Differential inactivation of caspase-8 in lung cancers. Cancer Biol Ther 1(1):65–69
69. Wang J, Zheng L, Lobito A, Chan FK, Dale J, Sneller M et al (1999) Inherited human
Caspase 10 mutations underlie defective lymphocyte and dendritic cell apoptosis in auto-
immune lymphoproliferative syndrome type II. Cell 98(1):47–58
70. Tadaki H, Saitsu H, Kanegane H, Miyake N, Imagawa T, Kikuchi M et al (2011) Exonic
deletion of CASP10 in a patient presenting with systemic juvenile idiopathic arthri-
tis, but not with autoimmune lymphoproliferative syndrome type IIa. Int J Immunogenet
38(4):287–293
71. Park WS, Lee JH, Shin MS, Park JY, Kim HS, Kim YS et al (2002) Inactivating mutations
of the caspase-10 gene in gastric cancer. Oncogene 21(18):2919–2925
72. Oh JE, Kim MS, Ahn CH, Kim SS, Han JY, Lee SH et al (2010) Mutational analysis of
CASP10 gene in colon, breast, lung and hepatocellular carcinomas. Pathology 42(1):73–76
1 Defective Apoptosis Signaling in Cancer 23
73. Shin MS, Kim HS, Kang CS, Park WS, Kim SY, Lee SN et al (2002) Inactivating muta-
tions of CASP10 gene in non-Hodgkin lymphomas. Blood 99(11):4094–4099
74. Kim MS, Oh JE, Min CK, Lee S, Chung NG, Yoo NJ et al (2009) Mutational analysis of
CASP10 gene in acute leukaemias and multiple myelomas. Pathology 41(5):484–487
75. Soung YH, Lee JW, Kim SY, Park WS, Nam SW, Lee JY et al (2006) Mutational analysis
of proapoptotic caspase-9 gene in common human carcinomas. APMIS 114(4):292–297
76. Soung YH, Lee JW, Kim SY, Park WS, Nam SW, Lee JY et al (2004) Somatic mutations of
CASP3 gene in human cancers. Hum Genet 115(2):112–115
77. Soung YH, Lee JW, Kim HS, Park WS, Kim SY, Lee JH et al (2003) Inactivating mutations
of CASPASE-7 gene in human cancers. Oncogene 22(39):8048–8052
78. Gyrd-Hansen M, Meier P (2010) IAPs: from caspase inhibitors to modulators of NF-
kappaB, inflammation and cancer. Nat Rev Cancer 10(8):561–574
79. Fulda S, Vucic D (2012) Targeting IAP proteins for therapeutic intervention in cancer. Nat
Rev Drug Discov 11(2):109–124
80. Eckelman BP, Salvesen GS, Scott FL (2006) Human inhibitor of apoptosis proteins: why
XIAP is the black sheep of the family. EMBO Rep 7(10):988–994
81. Scott FL, Denault JB, Riedl SJ, Shin H, Renatus M, Salvesen GS (2005) XIAP inhibits cas-
pase-3 and -7 using two binding sites: evolutionarily conserved mechanism of IAPs. EMBO
J 24(3):645–655
82. Riedl SJ, Renatus M, Schwarzenbacher R, Zhou Q, Sun C, Fesik SW et al (2001)
Structural basis for the inhibition of caspase-3 by XIAP. Cell 104(5):791–800
83. Huang Y, Park YC, Rich RL, Segal D, Myszka DG, Wu H (2001) Structural basis of
caspase inhibition by XIAP: differential roles of the linker versus the BIR domain. Cell
104(5):781–790
84. Chai J, Shiozaki E, Srinivasula SM, Wu Q, Datta P, Alnemri ES et al (2001) Structural
basis of caspase-7 inhibition by XIAP. Cell 104(5):769–780
85. Srinivasula SM, Hegde R, Saleh A, Datta P, Shiozaki E, Chai J et al (2001) A conserved
XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and
apoptosis. Nature 410(6824):112–116
86. Shiozaki EN, Chai J, Rigotti DJ, Riedl SJ, Li P, Srinivasula SM et al (2003) Mechanism of
XIAP-mediated inhibition of caspase-9. Mol Cell 11(2):519–527
87. Huang H, Joazeiro CA, Bonfoco E, Kamada S, Leverson JD, Hunter T (2000) The inhibi-
tor of apoptosis, cIAP2, functions as a ubiquitin-protein ligase and promotes in vitro mon-
oubiquitination of caspases 3 and 7. J Biol Chem 275(35):26661–26664
88. Suzuki Y, Nakabayashi Y, Takahashi R (2001) Ubiquitin-protein ligase activity of X-linked
inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3 and enhances its
anti-apoptotic effect in Fas-induced cell death. Proc Natl Acad Sci U S A 98(15):8662–8667
89. Schile AJ, Garcia-Fernandez M, Steller H (2008) Regulation of apoptosis by XIAP ubiqui-
tin-ligase activity. Genes Dev 22(16):2256–2266
90. Choi YE, Butterworth M, Malladi S, Duckett CS, Cohen GM, Bratton SB (2009) The E3
ubiquitin ligase cIAP1 binds and ubiquitinates caspase-3 and -7 via unique mechanisms at
distinct steps in their processing. J Biol Chem 284(19):12772–12782
91. Shiraki K, Sugimoto K, Yamanaka Y, Yamaguchi Y, Saitou Y, Ito K et al (2003)
Overexpression of X-linked inhibitor of apoptosis in human hepatocellular carcinoma. Int J
Mol Med 12(5):705–708
92. Shi YH, Ding WX, Zhou J, He JY, Xu Y, Gambotto AA et al (2008) Expression of X-linked
inhibitor-of-apoptosis protein in hepatocellular carcinoma promotes metastasis and tumor
recurrence. Hepatology 48(2):497–507
93. Augello C, Caruso L, Maggioni M, Donadon M, Montorsi M, Santambrogio R et al (2009)
Inhibitors of apoptosis proteins (IAPs) expression and their prognostic significance in
hepatocellular carcinoma. BMC Cancer 9:125
94. Yang XH, Feng ZE, Yan M, Hanada S, Zuo H, Yang CZ et al (2012) XIAP is a predictor of
cisplatin-based chemotherapy response and prognosis for patients with advanced head and
neck cancer. PLoS One 7(3):e31601
24 D. E. Johnson
95. Lopes RB, Gangeswaran R, McNeish IA, Wang Y, Lemoine NR (2007) Expression of the
IAP protein family is dysregulated in pancreatic cancer cells and is important for resistance
to chemotherapy. Int J Cancer 120(11):2344–2352
96. Xiang G, Wen X, Wang H, Chen K, Liu H (2009) Expression of X-linked inhibitor of apop-
tosis protein in human colorectal cancer and its correlation with prognosis. J Surg Oncol
100(8):708–712
97. Ramp U, Krieg T, Caliskan E, Mahotka C, Ebert T, Willers R et al (2004) XIAP expres-
sion is an independent prognostic marker in clear-cell renal carcinomas. Hum Pathol
35(8):1022–1028
98. Yan Y, Mahotka C, Heikaus S, Shibata T, Wethkamp N, Liebmann J et al (2004) Disturbed
balance of expression between XIAP and Smac/DIABLO during tumour progression in
renal cell carcinomas. Br J Cancer 91(7):1349–1357
99. Mizutani Y, Nakanishi H, Li YN, Matsubara H, Yamamoto K, Sato N et al (2007)
Overexpression of XIAP expression in renal cell carcinoma predicts a worse prognosis. Int
J Oncol 30(4):919–925
100. Ferreira CG, van der Valk P, Span SW, Ludwig I, Smit EF, Kruyt FA et al (2001)
Expression of X-linked inhibitor of apoptosis as a novel prognostic marker in radically
resected non-small cell lung cancer patients. Clin Cancer Res 7(8):2468–2474
101. Ferreira CG, van der Valk P, Span SW, Jonker JM, Postmus PE, Kruyt FA et al (2001)
Assessment of IAP (inhibitor of apoptosis) proteins as predictors of response to chemo-
therapy in advanced non-small-cell lung cancer patients. Ann Oncol 12(6):799–805
102. Krepela E, Dankova P, Moravcikova E, Krepelova A, Prochazka J, Cermak J et al (2009)
Increased expression of inhibitor of apoptosis proteins, survivin and XIAP, in non-small
cell lung carcinoma. Int J Oncol 35(6):1449–1462
103. Kempkensteffen C, Jager T, Bub J, Weikert S, Hinz S, Christoph F et al (2007)
The equilibrium of XIAP and Smac/DIABLO expression is gradually deranged during
the development and progression of testicular germ cell tumours. Int J Androl
30(5):476–483
104. Gu LQ, Li FY, Zhao L, Liu Y, Zang XX, Wang TX et al (2009) BRAFV600E mutation
and X-linked inhibitor of apoptosis expression in papillary thyroid carcinoma. Thyroid
19(4):347–354
105. Hussain AR, Uddin S, Ahmed M, Bu R, Ahmed SO, Abubaker J et al (2010) Prognostic
significance of XIAP expression in DLBCL and effect of its inhibition on AKT signalling.
J Pathol 222(2):180–190
106. Yamamoto K, Abe S, Nakagawa Y, Suzuki K, Hasegawa M, Inoue M et al (2004)
Expression of IAP family proteins in myelodysplastic syndromes transforming to overt leu-
kemia. Leuk Res 28(11):1203–1211
107. Tamm I, Richter S, Scholz F, Schmelz K, Oltersdorf D, Karawajew L et al (2004) XIAP
expression correlates with monocytic differentiation in adult de novo AML: impact on
prognosis. Hematol J 5(6):489–495
108. Chen GH, Lin FR, Ren JH, Chen J, Zhang JN, Wang Y et al (2006) Expression and signifi-
cance of X-linked inhibitor of apoptosis protein and its antagonized proteins in acute leuke-
mia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 14(4):639–643
109. Ibrahim AM, Mansour IM, Wilson MM, Mokhtar DA, Helal AM, Al Wakeel HM (2012)
Study of survivin and X-linked inhibitor of apoptosis protein (XIAP) genes in acute mye-
loid leukemia (AML). Lab Hematol 18(1):1–10
110. Tamm I, Richter S, Oltersdorf D, Creutzig U, Harbott J, Scholz F et al (2004) High
expression levels of x-linked inhibitor of apoptosis protein and survivin correlate with
poor overall survival in childhood de novo acute myeloid leukemia. Clin Cancer Res
10(11):3737–3744
111. Sung KW, Choi J, Hwang YK, Lee SJ, Kim HJ, Kim JY et al (2009) Overexpression
of X-linked inhibitor of apoptosis protein (XIAP) is an independent unfavorable
prognostic factor in childhood de novo acute myeloid leukemia. J Korean Med Sci
24(4):605–613
1 Defective Apoptosis Signaling in Cancer 25
153. Bauer JJ, Sesterhenn IA, Mostofi FK, McLeod DG, Srivastava S, Moul JW (1996) Elevated
levels of apoptosis regulator proteins p53 and bcl-2 are independent prognostic biomarkers
in surgically treated clinically localized prostate cancer. J Urol 156(4):1511–1516
154. Pallis M, Zhu YM, Russell NH (1997) Bcl-x(L) is heterogenously expressed by acute
myeloblastic leukaemia cells and is associated with autonomous growth in vitro and with
P-glycoprotein expression. Leukemia 11(7):945–949
155. Deng G, Lane C, Kornblau S, Goodacre A, Snell V, Andreeff M et al (1998) Ratio of bcl-
xshort to bcl-xlong is different in good- and poor-prognosis subsets of acute myeloid leuke-
mia. Mol Med 4(3):158–164
156. Tu Y, Renner S, Xu F, Fleishman A, Taylor J, Weisz J et al (1998) BCL-X expression in
multiple myeloma: possible indicator of chemoresistance. Cancer Res 58(2):256–262
157. Tang L, Tron VA, Reed JC, Mah KJ, Krajewska M, Li G et al (1998) Expression of apopto-
sis regulators in cutaneous malignant melanoma. Clin Cancer Res 4(8):1865–1871
158. Leiter U, Schmid RM, Kaskel P, Peter RU, Krahn G (2000) Antiapoptotic bcl-2 and bcl-xL
in advanced malignant melanoma. Arch Dermatol Res 292(5):225–232
159. Aebersold DM, Kollar A, Beer KT, Laissue J, Greiner RH, Djonov V (2001) Involvement
of the hepatocyte growth factor/scatter factor receptor c-met and of Bcl-xL in the resistance
of oropharyngeal cancer to ionizing radiation. Int J Cancer 96(1):41–54
160. Trask DK, Wolf GT, Bradford CR, Fisher SG, Devaney K, Johnson M et al (2002)
Expression of Bcl-2 family proteins in advanced laryngeal squamous cell carcinoma: corre-
lation with response to chemotherapy and organ preservation. Laryngoscope 112(4):638–644
161. Watanabe J, Kushihata F, Honda K, Mominoki K, Matsuda S, Kobayashi N (2002) Bcl-xL
overexpression in human hepatocellular carcinoma. Int J Oncol 21(3):515–519
162. Olopade OI, Adeyanju MO, Safa AR, Hagos F, Mick R, Thompson CB et al (1997)
Overexpression of BCL-x protein in primary breast cancer is associated with high tumor
grade and nodal metastases. Cancer J Sci Am 3(4):230–237
163. Kirsh EJ, Baunoch DA, Stadler WM (1998) Expression of bcl-2 and bcl-X in bladder can-
cer. J Urol 159(4):1348–1353
164. Krajewska M, Moss SF, Krajewski S, Song K, Holt PR, Reed JC (1996) Elevated expres-
sion of Bcl-X and reduced Bak in primary colorectal adenocarcinomas. Cancer Res
56(10):2422–2427
165. Friess H, Lu Z, Andren-Sandberg A, Berberat P, Zimmermann A, Adler G et al (1998)
Moderate activation of the apoptosis inhibitor bcl-xL worsens the prognosis in pancreatic
cancer. Ann Surg 228(6):780–787
166. Miyamoto Y, Hosotani R, Wada M, Lee JU, Koshiba T, Fujimoto K et al (1999)
Immunohistochemical analysis of Bcl-2, Bax, Bcl-X, and Mcl-1 expression in pancreatic
cancers. Oncology 56(1):73–82
167. Kaufmann SH, Karp JE, Svingen PA, Krajewski S, Burke PJ, Gore SD et al (1998)
Elevated expression of the apoptotic regulator Mcl-1 at the time of leukemic relapse. Blood
91(3):991–1000
168. Zhang B, Gojo I, Fenton RG (2002) Myeloid cell factor-1 is a critical survival factor for
multiple myeloma. Blood 99(6):1885–1893
169. Le Gouill S, Podar K, Amiot M, Hideshima T, Chauhan D, Ishitsuka K et al (2004) VEGF
induces Mcl-1 up-regulation and protects multiple myeloma cells against apoptosis. Blood
104(9):2886–2892
170. Kitada S, Andersen J, Akar S, Zapata JM, Takayama S, Krajewski S et al (1998)
Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations
with In vitro and In vivo chemoresponses. Blood 91(9):3379–3389
171. Shigemasa K, Katoh O, Shiroyama Y, Mihara S, Mukai K, Nagai N et al (2002) Increased
MCL-1 expression is associated with poor prognosis in ovarian carcinomas. Jpn J Cancer
Res 93(5):542–550
172. Li C, Li R, Grandis JR, Johnson DE (2008) Bortezomib induces apoptosis via Bim and Bik
up-regulation and synergizes with cisplatin in the killing of head and neck squamous cell
carcinoma cells. Mol Cancer Ther 7(6):1647–1655
28 D. E. Johnson
173. Feuerhake F, Kutok JL, Monti S, Chen W, LaCasce AS, Cattoretti G et al (2005) NFkappaB
activity, function, and target-gene signatures in primary mediastinal large B-cell lymphoma
and diffuse large B-cell lymphoma subtypes. Blood 106(4):1392–1399
174. Brien G, Trescol-Biemont MC, Bonnefoy-Berard N (2007) Downregulation of Bfl-1 pro-
tein expression sensitizes malignant B cells to apoptosis. Oncogene 26(39):5828–5832
175. Wilson JW, Nostro MC, Balzi M, Faraoni P, Cianchi F, Becciolini A et al (2000) Bcl-w
expression in colorectal adenocarcinoma. Br J Cancer 82(1):178–185
176. Monni O, Joensuu H, Franssila K, Klefstrom J, Alitalo K, Knuutila S (1997) BCL2 over-
expression associated with chromosomal amplification in diffuse large B-cell lymphoma.
Blood 90(3):1168–1174
177. Olejniczak ET, Van Sant C, Anderson MG, Wang G, Tahir SK, Sauter G et al (2007)
Integrative genomic analysis of small-cell lung carcinoma reveals correlates of sensitivity
to bcl-2 antagonists and uncovers novel chromosomal gains. Mol Cancer Res 5(4):331–339
178. Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J et al (2010)
The landscape of somatic copy-number alteration across human cancers. Nature
463(7283):899–905
179. Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M et al (2005)
miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A
102(39):13944–13949
180. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E et al (2002) Frequent dele-
tions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic
lymphocytic leukemia. Proc Natl Acad Sci U S A 99(24):15524–15529
181. Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu M, Wojcik SE et al (2005) A
MicroRNA signature associated with prognosis and progression in chronic lymphocytic
leukemia. N Engl J Med 353(17):1793–1801
182. Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M et al (2008) MiR-
15a and miR-16-1 cluster functions in human leukemia. Proc Natl Acad Sci U S A
105(13):5166–5171
183. Aqeilan RI, Calin GA, Croce CM (2010) miR-15a and miR-16-1 in cancer: discovery,
function and future perspectives. Cell Death Differ 17(2):215–220
184. Oltvai ZN, Milliman CL, Korsmeyer SJ (1993) Bcl-2 heterodimerizes in vivo with a con-
served homolog, Bax, that accelerates programmed cell death. Cell 74(4):609–619
185. Cory S, Adams JM (2002) The Bcl2 family: regulators of the cellular life-or-death switch.
Nat Rev Cancer 2(9):647–656
186. Korsmeyer SJ, Shutter JR, Veis DJ, Merry DE, Oltvai ZN (1993) Bcl-2/Bax: a rheostat that
regulates an anti-oxidant pathway and cell death. Semin Cancer Biol 4(6):327–332
187. Annis MG, Soucie EL, Dlugosz PJ, Cruz-Aguado JA, Penn LZ, Leber B et al (2005) Bax
forms multispanning monomers that oligomerize to permeabilize membranes during apop-
tosis. EMBO J 24(12):2096–2103
188. Korsmeyer SJ, Wei MC, Saito M, Weiler S, Oh KJ, Schlesinger PH (2000) Pro-apoptotic
cascade activates BID, which oligomerizes BAK or BAX into pores that result in the
release of cytochrome c. Cell Death Differ 7(12):1166–1173
189. Wei MC, Lindsten T, Mootha VK, Weiler S, Gross A, Ashiya M et al (2000) tBID, a
membrane-targeted death ligand, oligomerizes BAK to release cytochrome c. Genes Dev
14(16):2060–2071
190. Sharpe JC, Arnoult D, Youle RJ (2004) Control of mitochondrial permeability by Bcl-2
family members. Biochim Biophys Acta 1644(2–3):107–113
191. Kuwana T, Mackey MR, Perkins G, Ellisman MH, Latterich M, Schneiter R et al (2002)
Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial
membrane. Cell 111(3):331–342
192. Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ (2002) Distinct
BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype can-
cer therapeutics. Cancer Cell 2(3):183–192
1 Defective Apoptosis Signaling in Cancer 29
193. Kuwana T, Bouchier-Hayes L, Chipuk JE, Bonzon C, Sullivan BA, Green DR et al (2005)
BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial
membrane permeabilization both directly and indirectly. Mol Cell 17(4):525–535
194. Certo M, Del Gaizo Moore V, Nishino M, Wei G, Korsmeyer S, Armstrong SA et al (2006)
Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2
family members. Cancer Cell 9(5):351–365
195. Cartron PF, Gallenne T, Bougras G, Gautier F, Manero F, Vusio P et al (2004) The first
alpha helix of Bax plays a necessary role in its ligand-induced activation by the BH3-only
proteins Bid and PUMA. Mol Cell 16(5):807–818
196. Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG et al (2005) Differential target-
ing of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apop-
totic function. Mol Cell 17(3):393–403
197. Willis SN, Chen L, Dewson G, Wei A, Naik E, Fletcher JI et al (2005) Proapoptotic Bak
is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins.
Genes Dev 19(11):1294–1305
198. Willis SN, Fletcher JI, Kaufmann T, van Delft MF, Chen L, Czabotar PE et al (2007)
Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak.
Science 315(5813):856–859
199. Fernandez-Luna JL (2008) Regulation of pro-apoptotic BH3-only proteins and its contribu-
tion to cancer progression and chemoresistance. Cell Signal 20(11):1921–1926
200. Rampino N, Yamamoto H, Ionov Y, Li Y, Sawai H, Reed JC et al (1997) Somatic frameshift
mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype.
Science 275(5302):967–969
201. Meijerink JP, Mensink EJ, Wang K, Sedlak TW, Sloetjes AW, de Witte T et al (1998)
Hematopoietic malignancies demonstrate loss-of-function mutations of BAX. Blood
91(8):2991–2997
202. Miquel C, Borrini F, Grandjouan S, Auperin A, Viguier J, Velasco V et al (2005) Role of
bax mutations in apoptosis in colorectal cancers with microsatellite instability. Am J Clin
Pathol 123(4):562–570
203. Mestre-Escorihuela C, Rubio-Moscardo F, Richter JA, Siebert R, Climent J, Fresquet V et
al (2007) Homozygous deletions localize novel tumor suppressor genes in B-cell lympho-
mas. Blood 109(1):271–280
204. Kondo S, Shinomura Y, Miyazaki Y, Kiyohara T, Tsutsui S, Kitamura S et al (2000)
Mutations of the bak gene in human gastric and colorectal cancers. Cancer Res
60(16):4328–4330
205. Tagawa H, Karnan S, Suzuki R, Matsuo K, Zhang X, Ota A et al (2005) Genome-wide
array-based CGH for mantle cell lymphoma: identification of homozygous deletions of the
proapoptotic gene BIM. Oncogene 24(8):1348–1358
206. Sturm I, Stephan C, Gillissen B, Siebert R, Janz M, Radetzki S et al (2006) Loss of the
tissue-specific proapoptotic BH3-only protein Nbk/Bik is a unifying feature of renal cell
carcinoma. Cell Death Differ 13(4):619–627
207. Richter-Larrea JA, Robles EF, Fresquet V, Beltran E, Rullan AJ, Agirre X et al (2010)
Reversion of epigenetically mediated BIM silencing overcomes chemoresistance in Burkitt
lymphoma. Blood 116(14):2531–2542
208. Garrison SP, Jeffers JR, Yang C, Nilsson JA, Hall MA, Rehg JE et al (2008) Selection
against PUMA gene expression in Myc-driven B-cell lymphomagenesis. Mol Cell Biol
28(17):5391–5402
209. Obata T, Toyota M, Satoh A, Sasaki Y, Ogi K, Akino K et al (2003) Identification of HRK
as a target of epigenetic inactivation in colorectal and gastric cancer. Clin Cancer Res
9(17):6410–6418
210. Fontana L, Fiori ME, Albini S, Cifaldi L, Giovinazzi S, Forloni M et al (2008) Antagomir-
17-5p abolishes the growth of therapy-resistant neuroblastoma through p21 and BIM. PLoS
ONE 3(5):e2236
30 D. E. Johnson
234. Leu JI, Dumont P, Hafey M, Murphy ME, George DL (2004) Mitochondrial p53 activates
Bak and causes disruption of a Bak-Mcl1 complex. Nat Cell Biol 6(5):443–450
235. Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P et al (2003) p53 has a
direct apoptogenic role at the mitochondria. Mol Cell 11(3):577–590
236. Yin XM, Wang K, Gross A, Zhao Y, Zinkel S, Klocke B et al (1999) Bid-deficient mice are
resistant to Fas-induced hepatocellular apoptosis. Nature 400(6747):886–891
237. Friesen C, Fulda S, Debatin KM (1997) Deficient activation of the CD95 (APO-1/Fas) sys-
tem in drug-resistant cells. Leukemia 11(11):1833–1841
238. Antoku K, Liu Z, Johnson DE (1997) Inhibition of caspase proteases by CrmA enhances
the resistance of human leukemic cells to multiple chemotherapeutic agents. Leukemia
11(10):1665–1672
239. Fulda S, Los M, Friesen C, Debatin KM (1998) Chemosensitivity of solid tumor cells in
vitro is related to activation of the CD95 system. Int J Cancer 76(1):105–114
240. Fulda S, Meyer E, Friesen C, Susin SA, Kroemer G, Debatin KM (2001) Cell type spe-
cific involvement of death receptor and mitochondrial pathways in drug-induced apoptosis.
Oncogene 20(9):1063–1075
241. Tauzin S, Debure L, Moreau JF, Legembre P (2010) CD95-mediated cell signal-
ing in cancer: mutations and post-translational modulations. Cell Mol Life Sci
69(8):1261–1277
242. Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA, Nagata S (1992)
Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates
apoptosis. Nature 356(6367):314–317
243. Chu JL, Drappa J, Parnassa A, Elkon KB (1993) The defect in Fas mRNA expression
in MRL/lpr mice is associated with insertion of the retrotransposon, ETn. J Exp Med
178(2):723–730
244. Wu J, Zhou T, He J, Mountz JD (1993) Autoimmune disease in mice due to integration of
an endogenous retrovirus in an apoptosis gene. J Exp Med 178(2):461–468
245. Adachi M, Watanabe-Fukunaga R, Nagata S (1993) Aberrant transcription caused by the
insertion of an early transposable element in an intron of the Fas antigen gene of lpr mice.
Proc Natl Acad Sci U S A 90(5):1756–1760
246. Dowdell KC, Niemela JE, Price S, Davis J, Hornung RL, Oliveira JB et al (2010) Somatic
FAS mutations are common in patients with genetically undefined autoimmune lymphopro-
liferative syndrome. Blood 115(25):5164–5169
247. Hsu AP, Dowdell KC, Davis J, Niemela JE, Anderson SM, Shaw PA et al (2012)
Autoimmune lymphoproliferative syndrome due to FAS mutations outside the signal-
transducing death domain: molecular mechanisms and clinical penetrance. Genet Med
14(1):81–89
248. Kuehn HS, Caminha I, Niemela JE, Rao VK, Davis J, Fleisher TA et al (2011) FAS haplo-
insufficiency is a common disease mechanism in the human autoimmune lymphoprolifera-
tive syndrome. J Immunol 186(10):6035–6043
249. Magerus-Chatinet A, Neven B, Stolzenberg MC, Daussy C, Arkwright PD, Lanzarotti N et
al (2011) Onset of autoimmune lymphoproliferative syndrome (ALPS) in humans as a con-
sequence of genetic defect accumulation. J Clin Invest 121(1):106–112
250. Wu J, Siddiqui J, Nihal M, Vonderheid EC, Wood GS (2011) Structural alterations of the
FAS gene in cutaneous T-cell lymphoma (CTCL). Arch Biochem Biophys 508(2):185–191
251. Dereure O, Levi E, Vonderheid EC, Kadin ME (2002) Infrequent Fas mutations but no Bax
or p53 mutations in early mycosis fungoides: a possible mechanism for the accumulation
of malignant T lymphocytes in the skin. J Invest Dermatol 118(6):949–956
252. Seeberger H, Starostik P, Schwarz S, Knorr C, Kalla J, Ott G et al (2001) Loss of Fas
(CD95/APO-1) regulatory function is an important step in early MALT-type lymphoma
development. Lab Invest 81(7):977–986
253. Wohlfart S, Sebinger D, Gruber P, Buch J, Polgar D, Krupitza G et al (2004) FAS (CD95)
mutations are rare in gastric MALT lymphoma but occur more frequently in primary gas-
tric diffuse large B-cell lymphoma. Am J Pathol 164(3):1081–1089
32 D. E. Johnson
276. Dechant MJ, Fellenberg J, Scheuerpflug CG, Ewerbeck V, Debatin KM (2004) Mutation
analysis of the apoptotic “death-receptors” and the adaptors TRADD and FADD/MORT-1
in osteosarcoma tumor samples and osteosarcoma cell lines. Int J Cancer 109(5):661–667
277. Park WS, Lee JH, Shin MS, Park JY, Kim HS, Kim YS et al (2001) Inactivating mutations
of KILLER/DR5 gene in gastric cancers. Gastroenterology 121(5):1219–1225
278. Lee SH, Shin MS, Kim HS, Lee HK, Park WS, Kim SY et al (1999) Alterations
of the DR5/TRAIL receptor 2 gene in non-small cell lung cancers. Cancer Res
59(22):5683–5686
279. Arai T, Akiyama Y, Okabe S, Saito K, Iwai T, Yuasa Y (1998) Genomic organization and
mutation analyses of the DR5/TRAIL receptor 2 gene in colorectal carcinomas. Cancer
Lett 133(2):197–204
280. Lee KH, Lim SW, Kim HG, Kim DY, Ryu SY, Joo JK et al (2009) Lack of death receptor
4 (DR4) expression through gene promoter methylation in gastric carcinoma. Langenbecks
Arch Surg 394(4):661–670
281. Elias A, Siegelin MD, Steinmuller A, von Deimling A, Lass U, Korn B et al (2009)
Epigenetic silencing of death receptor 4 mediates tumor necrosis factor-related apoptosis-
inducing ligand resistance in gliomas. Clin Cancer Res 15(17):5457–5465
282. Hopkins-Donaldson S, Ziegler A, Kurtz S, Bigosch C, Kandioler D, Ludwig C et al (2003)
Silencing of death receptor and caspase-8 expression in small cell lung carcinoma cell lines
and tumors by DNA methylation. Cell Death Differ 10(3):356–364
283. Horak P, Pils D, Haller G, Pribill I, Roessler M, Tomek S et al (2005) Contribution of
epigenetic silencing of tumor necrosis factor-related apoptosis inducing ligand receptor 1
(DR4) to TRAIL resistance and ovarian cancer. Mol Cancer Res 3(6):335–343
284. Zhang Y, Zhang B (2008) TRAIL resistance of breast cancer cells is associated with consti-
tutive endocytosis of death receptors 4 and 5. Mol Cancer Res 6(12):1861–1871
285. Jin Z, McDonald ER 3rd, Dicker DT, El-Deiry WS (2004) Deficient tumor necrosis factor-
related apoptosis-inducing ligand (TRAIL) death receptor transport to the cell surface in
human colon cancer cells selected for resistance to TRAIL-induced apoptosis. J Biol Chem
279(34):35829–35839
286. Wu GS, Burns TF, McDonald ER 3rd, Jiang W, Meng R, Krantz ID et al (1997)
KILLER/DR5 is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet
17(2):141–143
287. Sheikh MS, Burns TF, Huang Y, Wu GS, Amundson S, Brooks KS et al (1998)
p53-dependent and -independent regulation of the death receptor KILLER/DR5 gene
expression in response to genotoxic stress and tumor necrosis factor alpha. Cancer Res
58(8):1593–1598
288. Wu GS, Burns TF, McDonald ER 3rd, Meng RD, Kao G, Muschel R et al (1999) Induction
of the TRAIL receptor KILLER/DR5 in p53-dependent apoptosis but not growth arrest.
Oncogene 18(47):6411–6418
289. Takimoto R, El-Deiry WS (2000) Wild-type p53 transactivates the KILLER/DR5 gene
through an intronic sequence-specific DNA-binding site. Oncogene 19(14):1735–1743
290. Guan B, Yue P, Clayman GL, Sun SY (2001) Evidence that the death receptor DR4 is a
DNA damage-inducible, p53-regulated gene. J Cell Physiol 188(1):98–105
291. Liu X, Yue P, Khuri FR, Sun SY (2004) p53 upregulates death receptor 4 expression
through an intronic p53 binding site. Cancer Res 64(15):5078–5083
292. Sheikh MS, Huang Y, Fernandez-Salas EA, El-Deiry WS, Friess H, Amundson S et al
(1999) The antiapoptotic decoy receptor TRID/TRAIL-R3 is a p53-regulated DNA dam-
age-inducible gene that is overexpressed in primary tumors of the gastrointestinal tract.
Oncogene 18(28):4153–4159
293. Liu X, Yue P, Khuri FR, Sun SY (2005) Decoy receptor 2 (DcR2) is a p53 target gene and
regulates chemosensitivity. Cancer Res 65(20):9169–9175
294. Bolze A, Byun M, McDonald D, Morgan NV, Abhyankar A, Premkumar L et al (2010)
Whole-exome-sequencing-based discovery of human FADD deficiency. Am J Hum Genet
87(6):873–881
34 D. E. Johnson
295. Soung YH, Lee JW, Kim SY, Nam SW, Park WS, Kim SH et al (2004) Mutation of FADD
gene is rare in human colon and stomach cancers. APMIS 112(9):595–597
296. Safa AR, Day TW, Wu CH (2008) Cellular FLICE-like inhibitory protein (C-FLIP): a novel
target for cancer therapy. Curr Cancer Drug Targets 8(1):37–46
297. Bagnoli M, Canevari S, Mezzanzanica D (2010) Cellular FLICE-inhibitory protein
(c-FLIP) signalling: a key regulator of receptor-mediated apoptosis in physiologic context
and in cancer. Int J Biochem Cell Biol 42(2):210–213
298. Chang DW, Xing Z, Pan Y, Algeciras-Schimnich A, Barnhart BC, Yaish-Ohad S et al
(2002) c-FLIP(L) is a dual function regulator for caspase-8 activation and CD95-mediated
apoptosis. EMBO J 21(14):3704–3714
299. Yu JW, Jeffrey PD, Shi Y (2009) Mechanism of procaspase-8 activation by c-FLIPL. Proc
Natl Acad Sci U S A 106(20):8169–8174
300. Korkolopoulou P, Saetta AA, Levidou G, Gigelou F, Lazaris A, Thymara I et al (2007)
c-FLIP expression in colorectal carcinomas: association with Fas/FasL expression and
prognostic implications. Histopathology 51(2):150–156
301. Du X, Bao G, He X, Zhao H, Yu F, Qiao Q et al (2009) Expression and biological signifi-
cance of c-FLIP in human hepatocellular carcinomas. J Exp Clin Cancer Res 28:24
302. Bagnoli M, Ambrogi F, Pilotti S, Alberti P, Ditto A, Barbareschi M et al (2009) c-FLIPL
expression defines two ovarian cancer patient subsets and is a prognostic factor of adverse
outcome. Endocr Relat Cancer 16(2):443–453
Chapter 2
The Warburg Effect and Beyond:
Metabolic Dependencies for Cancer Cells
Abstract Current definitions of cancer are best realized as a list of traits or hall-
marks, such as tissue invasion, metastasis, cell-autonomous growth, and resist-
ance to apoptosis. A recent update included deregulated cellular energetics as an
emerging hallmark. However, debate about tumor cell metabolism occupied center
stage in the pre-oncogene era. Over the last 15 years, direct links of oncogenes and
tumor suppressor genes to cell metabolism have brought cancer metabolism to the
forefront once again. Current tools provide much greater opportunities for prob-
ing metabolic differences between normal and cancer cells, in some cases reveal-
ing flux through unexpected metabolic pathways. Metabolic networks may also be
truncated, presenting opportunities for selective growth inhibition or death by tar-
geting non-redundant pathways in cancer cells. These “metabolic dependencies”
are not likely to be associated with classically defined oncogenes or computation-
ally derived drivers and thus may require novel strategies for discovery.
Two giants of biochemistry, Otto Warburg and Herbert Crabtree, developed novel
insights into cancer metabolism in the 1920s. Warburg improved on previous man-
ometric techniques to study respiratory quotients in a variety of cell types and tis-
sue slices. Based on his demonstration of a sixfold increase of oxygen uptake in
sea urchin eggs following fertilization, he entertained the notion that tumor growth
could be explained by increased bioenergetic metabolism. In opposition to his
original predictions, the respiratory rate in Flexner rat carcinomas was similar to
normal rat tissues. However, the rate of glycolysis was increased up to 30-fold in
the carcinoma compared with rat liver.
Comparing glycolysis in air and nitrogen, Warburg observed that both tissues
had higher rates of lactate generation in the absence of oxygen. In normoxic
samples, glycolytic rates were at or below the limit of detection in most normal
tissues. In contrast, glycolysis in normoxic cancer tissues still accounted for the
majority of glucose turnover. The ratio of aerobic glycolysis to respiration was
substantially elevated in cancers, expressed in various ways as mole percent glu-
cose metabolized to lactate (~90 %), ATP generated by glycolysis (35–50 %), or
mass of lactate produced (10 % of tissue weight per hour) [1].
Warburg extended these results to human cancer tissues and also demonstrated
that although many adult tissues did not produce lactate even under hypoxic condi-
tions, normal growing tissue (e.g., embryonic samples) had intermediate rates of aer-
obic glycolysis. According to the Pasteur effect, oxygen should inhibit fermentation
(lactate production). Warburg interpreted his findings as demonstrating that respira-
tion was insufficient to suppress glycolysis, either due to limitations in oxygen sup-
ply or due to mitochondrial activity. Since his aerobic experiments were conducted in
thin tissue slices in which oxygen diffusion was not rate limiting, the problem seemed
to Warburg to be intrinsic to mitochondria. Furthermore, if respiration was insuffi-
cient for the cellular energy demand, selection of cells with high glycolytic rates for
survival was possible. Several normal tissues, including retina, kidney medulla, car-
tilage, bone marrow, skin, fibroblasts, intestinal mucosa, placenta, and proliferating
thymocytes, have been demonstrated to have high rates of aerobic glycolysis. In fact,
Warburg argued against using aerobic glycolysis as a specific test for cancer cells [2].
The central observation that cancer cells have high rates of aerobic glycolysis,
now known as the Warburg effect , has been repeated numerous times and is the
basis for the use of 2-deoxy-2-(18F) fluoro-D-glucose positron emission tomogra-
phy (PET) scans for tumor staging. However, mitochondrial dysfunction as a basis
for the Warburg effect is still debatable. The Warburg effect is inhibited by treat-
ment with mitochondrial uncouplers, indicating a well functioning Krebs cycle
and electron transport chain. Anaerobic glycolysis occurs at higher rates in cancer
cells than aerobic glycolysis, while the ratio of the decrease in lactic acid produc-
tion to oxygen consumption in normoxia is similar in cancer and normal tissues.
Thus, if energy demand remains constant, the efficiency of oxidative phosphoryla-
tion at suppressing glycolysis is similar in tumors and normal tissues.
However, it has been suggested that cancer cells may have reduced mitochon-
drial reserve in response to glycolytic inhibition [3]. Isolated mitochondria from
cancer cell lines with pronounced Warburg effects have been shown to exhibit
specific defects in substrate utilization, respiratory control ratios, and mitochon-
drial content [4, 5]. Reduced expression of the β subunit of the mitochondrial
F1FO ATPase has been linked to breast, lung, and colon adenocarcinomas with
poor prognosis [6–8]. Upregulated expression of the ATPase inhibitor IF1 has also
been reported in cancers [9, 10]. Aside from intrinsic differences in mitochondrial
function, several regulatory mechanisms restricting mitochondrial oxidative phos-
phorylation in cancer cells have been identified.
The Crabtree effect refers to the ability of glucose to inhibit respiration
in cancer cells, as opposed to the stimulation of respiration noted in normal tis-
sues [11]. This dynamic regulation provides another perspective on the glycolytic
shift in cancer cells identified by Warburg. Typically observed in cells in which
2 The Warburg Effect and Beyond: Metabolic Dependencies for Cancer Cells 37
adaptations that diminish ATP generation. Pyruvate kinase catalyzes the transfer of
phosphate from phosphoenolpyruvate (PEP) to ADP, generating pyruvate and ATP.
Originally described by Sato in 1978 [19], the pyruvate kinase splicing isoform
M2 (PK-M2) is highly expressed in cancers, embryonic tissues, and several adult
tissues, including adipocytes, retina, and lung [20]. PK-M2 is active as a tetramer,
but mostly occurs as inactive dimer in cancer cells. Tetramer formation is induced
allosterically by the proximal glycolytic intermediate, fructose-1,6-bisphosphate
(F-1,6-BP), but opposed by tyrosine phosphorylation of PK-M2 or interactions
with phosphotyrosine-bearing proteins at the F-1,6-BP-binding site [21–24].
Reduced activity of PK-M2 increases the diversion of upstream glycolytic inter-
mediates into anabolic pathways, including the pentose phosphate pathway [25],
as well as slowing ATP generation. Interestingly, substitution of the PK-M1 splic-
ing isoform increases glucose oxidation and reduces lactate production by an
unexplained mechanism [23].
Reduction in the PK-M2 activity in cancer cells raises questions as to the
source of pyruvate utilized for lactate generation in aerobic glycolysis. Analogous
to bacterial pathways, the substrate for PK-M2, PEP can be used as a phosphate
donor in protein kinase reactions [26, 27]. One of the protein substrates for PEP-
dependent phosphorylation is the glycolytic enzyme phosphoglycerate mutase
(PGAM1), which is phosphorylated on the catalytic histidine (H11) involved in
transferring phosphate to the C-2 position of 3-phosphoglycerate. This reaction
produces pyruvate as a product in the absence of ATP generation. Thus, cancer
cells can switch to an alternative glycolytic pathway that does not produce net ATP.
Finally, the concept that the predominant function of glycolysis is ATP produc-
tion can be questioned by a study of glucose deprivation in IL-3-dependent cells
[28]. Glucose is necessary for cell growth and proliferation in many cell types.
In this example, glucose was shown to be required for N-linked glycosylation of
the IL-3 receptor, required for its cell surface localization and in turn, uptake of
an alternative substrate fuel, glutamine. Glucose could be replaced by the specific
product of the hexosamine biosynthetic pathway, N-acetylglucosamine, enabling
glutamine uptake and cell growth.
Three major pathways are involved in deregulating glycolysis in cancer, the
transcription factors c-Myc and HIF-1, and the serine/threonine kinase, Akt . The
c-Myc oncoprotein is overexpressed in more than 50 % of human cancers and
binds to 15 % of all gene promoters in the genome. Among the pathways con-
trolled by c-Myc is glycolysis, with 9/10 glycolytic enzymes and glucose trans-
porters type 1, 2, and 4 identified as Myc targets. Myc-dependent cell cycle entry
and transformation are associated with increased lactic acid production [29, 30].
Stable isotope labeling studies indicate that Myc directs glucose carbons to mul-
tiple anabolic pathways, including nucleotide, amino acid, and lipid biosynthesis
[31, 32]. N-Myc appears to share similar targets in glycolysis [33].
The hypoxia-inducible transcription factor, HIF-1α, is degraded by the ubiq-
uitin–proteasome system following oxygen-dependent proline hydroxylation.
HIF-1α is overexpressed in 13/19 common tumor types [34]. HIF-1 targets include
glycolytic enzymes, glucose transporters and the pyruvate dehydrogenase kinase
2 The Warburg Effect and Beyond: Metabolic Dependencies for Cancer Cells 39
Inhibition of glycolysis is often lethal in tumor cell lines in vitro, prompting both
early and renewed interest in this strategy for cancer treatment. The glucose analog,
2-deoxy-D-glucose (2-DG), is phosphorylated by hexokinase, but in the absence
of further glycolytic transformations, 2-DG-6-P accumulates in cells and inhibits
hexokinase. 2-DG was first administered to human cancer patients [48] with a vari-
ety of cancers and has since been studied in early stage clinical trials in patients
with glioblastomas [49] and advanced solid malignancies [50]. Novel LDH-A inhib-
itors have been reported [51, 52]. A peptide inhibitor of PK-M2 is in clinical tri-
als for melanoma and renal cell carcinoma (www.thallion.com). Several classes of
inhibitors of the PI3K–Akt signaling pathway are in clinical trials, including PI3K
inhibitors, Akt inhibitors, and dual PI3K–mTOR inhibitors [53]. A common theme
in cell death following glucose deprivation or glycolytic inhibition is an increase in
mitochondrial generation of reactive oxygen species, with decreased generation of
NADPH reducing equivalents in the pentose phosphate pathway [54, 55].
Fig. 2.1 Truncated and reversed TCA cycles (red) with glutaminolysis, involving malic enzyme
producing pyruvate and NADPH, and reductive carboxylation of α-ketoglutarate to citrate
and hexosamine synthesis and its α-nitrogen to alanine and aspartate in transami-
nation reactions. Glutamate is utilized in glutathione synthesis, and glutamine
exchange with leucine and other essential amino acids is essential for mTOR
activity [58]. However, a significant portion of glutamine nitrogen and carbon is
transported extracellularly, as glutamate, alanine, and lactate, indicating that, like
glycolysis, glutaminolysis is an inefficient process in cancer cells [59].
c-Myc has emerged as a major driver of glutamine metabolism. Glutamine
deprivation is sufficient to kill Myc-overexpressing cells [60, 61]. Surprisingly,
glutamine-deprivation-induced cell death involves depletion of TCA cycle
intermediates, but not ATP deficiency, glutathione depletion, or DNA dam-
age, and can be partially rescued with antioxidants [60, 62]. The anaplerotic
requirement for glutamine is highlighted by the glutamine-independent growth
of tumor cells expressing pyruvate carboxylase, an anaplerotic enzyme associ-
ated with glucose metabolism [63]. Glutaminase translation is suppressed by
two microRNAs, miR-23a and miR-23b, that are in turn targets for Myc tran-
scriptional repression [62]. Myc also increases the expression of glutamine
transporters [61].
Although there are differences in the regulation of glucose and glutamine
metabolism, one mechanism for dual control is the basic helix-loop-helix leu-
cine zipper transcription factor, MondoA (also known as MLXIP), related to Myc
[64]. In glucose-containing growth media, MondoA functions as a transcrip-
tional activator for TXNIP, which suppresses glucose uptake and aerobic glyco-
lysis. However, the addition of glutamine converts MondoA to a transcriptional
repressor, increasing glucose uptake. The ability of a cell-permeable analog of
α-ketoglutarate to similarly affect MondoA transcriptional activity suggests
that the levels of TCA cycle intermediates may be monitored to adjust glucose
uptake and metabolism. Recent data also indicate that Nrf2, a transcription factor
responding to oxidative stress, directs both glucose and glutamine utilization in
anabolic pathways, such as the pentose phosphate pathway and nucleotide syn-
thesis [65].
Tumor cells engage in de novo fatty acid synthesis, in contrast to importation of
circulating lipids by most normal cells in the body [66, 67]. Several recent reports
have indicated that cancer cells may also exhibit high rates of peroxisomal and
mitochondrial β-oxidation [68, 69]. Prostate cancers with low 2-deoxy-2-fluoro-
D-glucose (FDG) avidity on PET scans may engage in fatty acid β-oxidation as a
principal bioenergetic pathway [69]. Respiration in glioblastoma cells is inhibited
30–40 % by treatment with the carnitine palmitoyltransferase-1 (CPT1) inhibitor,
etomoxir, with a 50 % decrease in ATP levels [70]. Fatty acid β-oxidation has been
associated with uncoupling protein-2 expression and chemoresistance [71, 72].
The brain CPT1C isoform is upregulated in non-small-cell lung cancers and con-
fers resistance to hypoxia, glucose deprivation, and the mTOR inhibitor, rapamy-
cin [73]. Adipocytes promote the growth of ovarian cancer cells by mobilizing free
fatty acids for β-oxidation by tumor cells [74]. Inhibition of fatty acid β-oxidation
(etomoxir, ranolazine) or fatty acid biosynthesis (cerulenin, C75, orlistat) is selec-
tively cytotoxic to some cancer cells [70, 75].
42 D. M. Hockenbery et al.
fumarate hydratase (Fh1), cause hereditary leiomyomatosis and renal cell can-
cer (HLRCC). The accumulation of fumarate inhibits prolyl hydroxylases, lead-
ing to an increase in HIF expression under normoxic conditions. Incorporation
of cellular growth as consumption of biosynthetic precursors in the in silico
model allowed testing of specific gene knockouts for effects on growth. Frezza
et al. predicted 24 reactions to be synthetically lethal with Fh1 deletion, 18 of
which involved heme metabolism. Heme biosynthesis utilizes TCA-derived suc-
cinate, providing an alternative route to fumarate synthesis. Subsequent analysis
of Fh1−/− cells demonstrated increased excretion of bilirubin, the degradation
product of heme. Only three enzymes involved in heme degradation were overex-
pressed in Fh1−/− cells, highlighting the power of the computational approach to
identify significant pathways in the absence of expression criteria. Several strate-
gies to inhibit heme pathway flux reduced growth of Fh−/− cells.
Toward an unbiased screening strategy, we have generated a chemical library
with inhibitors for enzymes of intermediary metabolism, as validated in the litera-
ture by direct enzymatic assay. Inhibitors were chosen from the BRENDA enzyme
database (www.brenda-enzymes.org) and review of published literature, purchased
from Sigma-Aldrich or other chemical supply houses, or donated by academic
labs. Inhibitors of 585 enzymes have been identified. Each inhibitor is plated
at 100X EC50 concentration in DMSO. Transcriptome analysis in mouse and
human databases indicates ~750 enzymes of intermediary metabolism encoded
in the genome with unique EC numbers [84]. Such a library has several applica-
tions relevant to cancer research. Screening cancer cell lines versus normal cel-
lular counterparts for loss of viability or cell growth can reveal critical metabolic
dependencies associated with the transformed phenotype. Altered expressions
of metabolic enzymes, in particular, as revealed by Gene Set Enrichment analy-
ses, are frequently observed in RNA microarray profiles. As yet, there is no rapid
method to determine the essentiality of a metabolic pathway for cancer cell viabil-
ity. Metabolomic studies of cancer cells are increasingly being reported. A chemi-
cal inhibitor library can rapidly ascertain which pathways connecting to a given
metabolite are casually linked to cell growth or viability.
We have tested 217 causally compounds out of a total of 585 in our enzyme
inhibitor compound library in the MCF10A cell line expressing Myc-ERTAM. As
shown in Fig. 2.2, inhibitors are available for a high proportion of enzymes in
major metabolic pathways. MCF10A-MycERTAM cells were treated with tamoxifen
to activate Myc for 24 h and compared with comparably treated vehicle controls.
Cell metabolism was evaluated using a Seahorse analyzer, which demonstrated a
shift toward glycolytic metabolism in tamoxifen-treated, but not control, cells.
Cells were tested with each compound in triplicate at a single dose, based on
published IC80 values for enzymatic inhibition. Cell viability was assessed after
24 h by Alamar Blue assay. Log2-transformed results were analyzed according to
SSMD scores (strictly standardized mean difference; [85]) with respect to nega-
tive vehicle controls. The results are graphed in Fig. 2.3 (high SSMD values cor-
respond to selective killing of tamoxifen-treated cells). A secondary assay for cell
number (Hoechst 33342 fluorescence of adherent cells) confirmed these results.
44 D. M. Hockenbery et al.
Fig. 2.3 Plot of SSMD scores for 217 compounds tested against MCF10A-MycER cells +/− tamox-
ifen, in relation to negative (vehicle) controls
The top hits include aldehyde dehydrogenase, malic enzyme, aromatic L-amino
acid decarboxylase, and threonine tRNA ligase. Aldehyde dehydrogenases are
markers for cancer stem cells. Disulfiram, the aldehyde dehydrogenase inhibitor
in the screen, is reported to have anticancer activity [86], although the relevant tar-
get has not been validated. Aromatic L-amino acid decarboxylase (also known as
L-dopa decarboxylase) is expressed in several epithelial cancers in addition to neu-
roendocrine tumors and has co-activator activity for the androgen receptor [87, 88].
2 The Warburg Effect and Beyond: Metabolic Dependencies for Cancer Cells 45
Borrelidin, the threonine tRNA ligase inhibitor, induces apoptosis in acute lympho-
blastic leukemia cells associated with the activation of the GCN2 stress kinase and
proapoptotic CHOP transcription factor [89].
2.6 Summary
It is not surprising, based on the intimate role of catabolic and anabolic metab-
olism in furnishing biochemical energy and building blocks for macromolecu-
lar synthesis and repair, that metabolic pathways are vital to cancer cell growth
and survival in different tumor microenvironments. What we have learned more
recently is that metabolic pathways can also direct chromatin structure and gene
expression, differentiation, and stemness [90–95], typified by the discovery of the
oncometabolite, D-2-hydroxyglutarate (2HG). Specific gain-of-function mutations
in the isocitrate dehydrogenase-1 and dehydrogenase-2 enzymes occur in glio-
blastomas and acute myeloid leukemia and alter enzymatic function to produce
2HG from α-ketoglutarate (α-KG) [96]. 2HG is an inhibitor of α-KG-dependent
Jumonji-C domain histone demethylases [97, 98] and is associated with increased
histone methylation and altered gene expression [99, 100]. Comprehensive map-
ping of cancer cell metabolism is at an early stage, but current knowledge suggests
that the Warburg effect may be the “tip of the iceberg”. The ultimate value of
cancer therapies directed against metabolic dependencies is still to be determined,
as the links between metabolism and cell death, and extent of metabolic flexibil-
ity are largely unknown. Further exploration of metabolic networks in cancer will
require novel strategies, but seems likely to yield novel targets for cancer therapy.
References
8. Cuezva JM, Chen G, Alonso AM, Isidoro A, Misek DE, Hanash SM, Beer DG (2004) The
bioenergetics signature of lung carcinomas is a molecular marker of cancer diagnosis and
prognosis. Carcinog 25(7):1157–1163
9. Sanchez-Cenizo L, Formentini L, Aldea M, Ortega AD, Garcia-Huerta P, Sanchez-Arago
M, Cuezva JM (2010) Up-regulation of the ATPase inhibitory factor (IF1) of the mitochon-
drial H+ -ATP synthase in human tumors mediates the metabolic shift of cancer cells to a
Warburg phenotype. J Biol Chem 285(33):25308–25313
10. Crabtree JG (1929) Observations on the carbohydrate metabolism of tumours. Biochem J
23(3):536–545
11. Formentini L, Sanchez-Arago M, Sanchez-Cenizo L, Cuezva JM (2012) The mitochondrial
ATPase inhibitory factor 1 triggers a ROS-mediated retrograde prosurvival and proliferative
response. Mol Cell 45(6):731–742
12. Sussman I, Erecinska M, Wilson DF (1980) Regulation of cellular energy metabolism: the
crabtree effect. Biochim Biophys Acta 591(2):209–223
13. Wu R, Racker E (1959) Regulatory mechanisms in carbohydrate metabolism. IV. Pasteur
effect and crabtree effect in ascites tumor cells. J Biol Chem 234(5):1036–1041
14. Chesney J, Mitchell R, Benigni F, Bacher M, Spiegel L, Al-Abed Y, Han JH, Metz C,
Bucala R (1999) An inducible gene product for 6-phosphofructo-2-kinase with an AU-rich
instability element: role in tumor cell glycolysis and the warburg effect. Proc Natl Acad Sci
U S A 96(6):3047–3052
15. Guppy M, Greiner E, Brand K (1993) The role of the crabtree effect and an endogenous
fuel in the energy metabolism of resting and proliferating thymocytes. Eur J Biochem
212(1):95–99
16. Reitzer LJ, Wice BM, Kennell D (1979) Evidence that glutamine, not sugar, is the major
energy source for cultured HeLa cells. J Biol Chem 254(8):2669–2676
17. Vazquez A, Liu J, Zhou Y, Oltvai ZN (2010) Catabolic efficiency of aerobic glycolysis: the
warburg effect revisited. BMC Syst Biol 6(5):58
18. Boxer GE, Devlin TM (1961) Pathways of intracellular hydrogen transport. Science
134(3489):1495–1501
19. Sato K, Takaya S, Imai F, Hatayama I, Ito N (1978) Different deviation patterns of carbo-
hydrate-metabolizing enzymes in primary rat hepatomas induced by different chemical car-
cinogens. Cancer Res 38(9):3086–3093
20. Mazurek S (2011) Pyruvate kinase type M2: a key regulator of the metabolic budget system
in tumor cells. Int J Biochem Cell Biol 43(7):969–980
21. Hitosugi T, Kang S, Vander Heiden MG, Chung TW, Elf S, Lythgoe K, Dong S, Lonial
S, Wang X, Chen GZ, Xie J, Gu TL, Polakiewicz RD, Roesel JL, Boggon TJ, Khuri FR,
Gilliland DG, Cantley LC, Kaufman J, Chen J (2009) Tyrosine phosphorylation inhibits
PKM2 to promote the warburg effect and tumor growth. Sci Signal 2(97):ra73
22. Christofk HR, Vander Heiden MG, Wu N, Asara JM, Cantley LC (2008) Pyruvate kinase
M2 is a phosphotyrosine-binding protein. Nature 452(7184):181–186
23. Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, Wei R,
Fleming MD, Schreiber SL, Cantley LC (2008) The M2 splice isoform of pyruvate kinase is
important for cancer metabolism and tumour growth. Nature 452(7184):230–233
24. Presek P, Reinacher M, Eigenbrodt E (1988) Pyruvate kinase type M2 is phosphorylated at
tyrosine residues in cells transformed by rous sarcoma virus. FEBS Lett 242(1):194–198
25. Anastasiou D, Poulogiannis G, Asara JM, Boxer MB, Jiang JK, Shen M, Bellinger G,
Sasaki AT, Locasale JW, Auld DS, Thomas CJ, Vander Heiden MG, Cantley LC (2011)
Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxi-
dant responses. Science 334(6060):1278–1283
26. Vander Heiden MG, Locasale JW, Swanson KD, Sharfi H, Heffron GJ, Amador-Noguez D,
Christofk HR, Wagner G, Rabinowitz JD, Asara JM, Cantley LC (2010) Evidence for an
alternative glycolytic pathway in rapidly proliferating cells. Science 329(5998):1492–1499
27. Gao X, Wang H, Yang JJ, Liu X, Liu ZR (2012) Pyruvate kinase M2 regulates gene tran-
scription by acting as a protein kinase. Mol Cell 45(5):598–609
2 The Warburg Effect and Beyond: Metabolic Dependencies for Cancer Cells 47
28. Wellen KE, Lu C, Mancuso A, Lemons JM, Ryczko M, Dennis JW, Rabinowitz JD, Coller
HA, Thompson CB (2010) The hexosamine biosynthetic pathway couples growth factor-
induced glutamine uptake to glucose metabolism. Genes Dev 24(24):2784–2799
29. Shim H, Dolde C, Lewis BC, Wu CS, Dang G, Jungmann RA, Dalla-Favera R, Dang CV
(1997) c-Myc transactivation of LDH-A: implications for tumor metabolism and growth.
Proc Natl Acad Sci U S A 94(13):6658–6663
30. Morrish F, Neretti N, Sedivy JM, Hockenbery DM (2008) The oncogene c-Myc coor-
dinates regulation of metabolic networks to enable rapid cell cycle entry. Cell Cycle
7(8):1054–1066
31. Morrish F, Isern N, Sadilek M, Jeffrey M, Hockenbery DM (2009) c-Myc activates multiple
metabolic networks to generate substrates for cell-cycle entry. Oncogene 28(27):2485–2491
32. Morrish F, Noonan J, Perez-Olsen C, Gafken PR, Fitzgibbon M, Kelleher J, VanGilst M,
Hockenbery D (2010) Myc-dependent mitochondrial generation of acetyl-CoA contrib-
utes to fatty acid biosynthesis and histone acetylation during cell cycle entry. J Biol Chem
285(47):36267–36274
33. Qing G, Skuli N, Mayes PA, Pawel B, Martinez D, Maris JM, Simon MC (2010)
Combinatorial regulation of neuroblastoma tumor progression by N-Myc and hypoxia
inducible factor HIF-1 alpha. Cancer Res 70(24):10351–10361
34. Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, Buechler P, Isaacs
WB, Semenza GL, Simons JW (1999) Overexpression of hypoxia-inducible factor 1 alpha
in common human cancers and their metastases. Cancer Res 59(22):5830–5835
35. Selak MA, Armour SM, MacKenzie ED, Boulahbel H, Watson DG, Mansfield KD, Pan Y,
Simon MC, Thompson CB, Gottlieb E (2005) Succinate links TCA cycle dysfunction to
oncogenesis by inhibiting HIF-alpha prolyl hydroxylase. Cancer Cell 7(1):77–85
36. Lu H, Forbes RA, Verma A (2002) Hypoxia-inducible factor 1 activation by aerobic glyco-
lysis implicates the Warburg effect in carcinogenesis. J Biol Chem 277(26):23111–23115
37. Treins C, Giorgetti-Peraldi S, Murdaca J, Semenza GL, Van Obberghen E (2002) Insulin
stimulates hypoxia-inducible factor 1 through a phosphatidylinositol 3-kinase/target of rapa-
mycin-dependent signaling pathway. J Biol Chem 277(31):27975–27981
38. Koshiji M, Kageyama Y, Pete EA, Horikawa I, Barrett JC, Huang LE (2004) HIF-1 alpha
induces cell cycle arrest by functionally counteracting Myc. EMBO J 23(9):1949–1956
39. Kim JW, Gao P, Liu YC, Semenza GL, Dang CV (2007) Hypoxia-inducible factor 1
and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and
metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol Cell Biol
27(21):7381–7393
40. Zhou QL, Jiang ZY, Holik J, Chawla A, Hagan GN, Leszyk J, Czech MP (2008) Akt sub-
strate TBC1D1 regulates GLUT1 expression through the mTOR pathway in 3T3-L1 adipo-
cytes. Biochem J 411(3):647–655
41. Sakamoto K, Holman GD (2008) Emerging role for AS160/TBC1D4 and TBC1D1 in the
regulation of GLUT4 traffic. Am J Physiol Endocrinol Metab 295(1):E29–E37
42. Deprez J, Vertommen D, Alessi DR, Hue L, Rider MH (1997) Phosphorylation and activa-
tion of heart 6-phosphofructo-2-kinase by protein kinase B and other protein kinases of the
insulin signaling cascades. J Biol Chem 41(3):533–535
43. Gottlob K, Majewski N, Kennedy S, Kandel E, Robey RB, Hay N (2001) Inhibition of early
apoptotic events by Akt/PKB is dependent on the first committed step of glycolysis and
mitochondrial hexokinase. Genes Dev 15(11):1406–1418
44. Pastorino JG, Hoek JB, Shulga N (2005) Activation of glycogen synthase kinase 3 beta disrupts
the binding of hexokinase II to mitochondria by phosphorylating voltage-dependent anion channel
and potentiates chemotherapy-induced cytotoxicity. Cancer Res 65(22):10545–10554
45. Robey IF, Lien AD, Welsh SJ, Baggett BK, Gillies RJ (2005) Hypoxia-inducible factor-1
alpha and the glycolytic phenotype in tumors. Neoplasia 7(4):324–330
46. Lum JJ, Bui T, Gruber M, Gordan JD, DeBerardinis RJ, Covello KL, Simon MC, Thompson
CB (2007) The transcription factor HIF-1 alpha plays a critical role in the growth factor-
dependent regulation of both aerobic and anaerobic glycolysis. Genes Dev 21(9):1037–1049
48 D. M. Hockenbery et al.
47. Elstrom RL, Bauer DE, Buzzai M, Karnauskas R, Harris MH, Plas DR, Zhuang H, Cinalli
RM, Alavi A, Rudin CM, Thompson CB (2004) Akt stimulates aerobic glycolysis in cancer
cells. Cancer Res 64(11):3892–3899
48. Landau BR, Laszlo J, Stengle J, Burk D (1958) Certain metabolic and pharmaco-
logic effects in cancer patients given infusions of 2-deoxy-D-glucose. J Natl Cancer Inst
21(3):475–483
49. Dwarakanath BS, Singh D, Banerji AK, Sarin R, Venkataramana NK, Jalali R, Vishwanath
PN, Mohanti BK, Tripathi RP, Kalia VK, Jain V (2009) Clinical studies for improving radi-
otherapy with 2-deoxy-D-glucose: present status and future prospects. J Cancer Res Ther
Suppl 1:S21–S26
50. Garber K (2010) Oncology’s energetic pipeline. Nat Biotechnol 28(9):888–891
51. Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, Royer RE, Vander Jagt DL,
Semenza GL, Dang CV (2010) Inhibition of lactate dehydrogenase A induces oxidative
stress and inhibits tumor progression. Proc Natl Acad Sci U S A 107(5):2037–2042
52. Granchi C, Bertini S, Macchia M, Minutolo F (2010) Inhibitors of lactate dehydrogenase
isoforms and their therapeutic potentials. Curr Med Chem 17(7):672–697
53. Courtney KD, Corcoran RB, Engelman JA (2010) The PI3K pathway as drug target in
human cancer. J Clin Oncol 28(6):1075–1083
54. Shutt DC, O’Dorisio MS, Aykin-Burns N, Spitz DR (2010) 2-deoxy-D-glucose induces
oxidative stress and cell killing in human neuroblastoma cells. Cancer Biol Ther
9(11):8953–8961
55. Aykin-Burns N, Ahmad IM, Zhu Y, Oberley LW, Spitz DR (2009) Increased levels of super-
oxide and H2O2 mediate the differential susceptibility of cancer cells versus normal cells to
glucose deprivation. Biochem J 418(1):29–37
56. Le A, Lane AN, Hamaker M, Bose S, Gouw A, Barbi J, Tsukamoto T, Rojas CJ, Slusher BS,
Zhang H, Zimmerman LJ, Liebler DC, Slebos RJ, Lorkiewicz PK, Higashi RM, Fan TW,
Dang CV (2012) Glucose-independent glutamine metabolism via TCA cycling for prolifera-
tion and survival in B cells. Cell Metab 15(1):110–121
57. Seltzer MJ, Bennett BD, Joshi AD, Gao P, Thomas AG, Ferraris DV, Tsukamoto T,
Rojas CJ, Slusher BS, Rabinowitz JD, Dang CV, Riggins GJ (2010) Inhibition of glu-
taminase preferentially slows growth of glioma cells with mutant IDH1. Cancer Res
70(22):8981–8987
58. Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, Yang H, Hild M,
Kung C, Wilson C, Myer VE, MacKeigan JP, Porter JA, Wang YK, Cantley LC, Finan PM,
Murphy LO (2009) Bidirectional transport of amino acids regulates mTOR and autophagy.
Cell 136(3):521–534
59. Deberardinis RJ, Mancuso A, Daikhim E, Nissim I, Yudkoff M, Wehrli S, Thompson CB
(2007) Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism
that exceeds the requirement for protein and nucleotide synthesis. Proc Natl Acad Sci U S A
104(49):19345–19350
60. Yuneva M, Zamboni N, Oefner P, Sachidanandam R, Lazebnik Y (2007) Deficiency in
glutamine but not glucose induces MYC-dependent apoptosis in human cells. J Cell Biol
78(1):93–105
61. Wise DR, De Berardinis RJ, Mancuso A, Sayed N, Zhang XY, Pfeiffer HK, Nissim I,
Daikhin E, Yudkoff M, McMahon SB, Thompson CB (2008) Myc regulates a transcriptional
program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction.
Proc Natl Acad Sci U S A 105(48):18782–18787
62. Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, Zeller KI, De Marzo
AM, Van Eyk JE, Mendell JT, Dang CV (2009) c-Myc suppression of miR-23a/b
enhances mitochondrial glutaminase expression and glutamine metabolism. Nature
458(7239):762–765
63. Cheng T, Sudderth J, Yang C, Mullen AR, Jin ES, Mates JM, DeBerardinis RJ (2011)
Pyruvate carboxylase is required for glutamine-independent growth of tumor cells. Proc
Natl Acad Sci U S A 108(21):8674–8679
2 The Warburg Effect and Beyond: Metabolic Dependencies for Cancer Cells 49
64. Kaadige MR, Looper RE, Kamalanaadhan S, Ayer DE (2009) Glutamine-dependent ana-
pleurosis dictates glucose uptake and cell growth by regulating MondoA transcriptional
activity. Proc Natl Acad Sci U S A 106(35):14878–14883
65. Mitsuishi Y, Taguchi K, Kawatani Y, Shibata T, Nukiwa T, Aburatani H, Yamamoto M,
Motohashi H (2012) Nrf2 redirects glucose and glutamine into anabolic pathways in meta-
bolic reprogramming. Cancer Cell 22(1):66–79
66. Kuhajda FP, Jenner K, Wood FD, Hennigar RA, Jacobs LB, Dick JD, Pasternack GR (1994)
Fatty acid synthesis: a potential selective target for antineoplastic therapy. Proc Natl Acad
Sci U S A 91(14):6379–6383
67. Menendez JA, Lupu R (2007) Fatty acid synthase and the lipogenic phenotype in cancer
pathogenesis. Nat Rev Cancer 7(10):763–777
68. Zha S, Ferdinandusse S, Hicks JL, Denis S, Dunn TA, Wanders RJ, Luo J, De Marzo AM,
Isaacs WB (2005) Peroxisomal branched chain fatty acid beta-oxidation pathway is upregu-
lated in prostate cancer. Prostate 63(4):316–323
69. Liu Y (2006) Fatty acid oxidation is a dominant bioenergetic pathway in prostate cancer.
Prostate Cancer Prostatic Dis 9(3):230–234
70. Pike LS, Smift AL, Croteau NJ, Ferrick DA, Wu M (2011) Inhibition of fatty acid oxida-
tion by etomoxir impairs NADPH production and increases reactive oxygen species result-
ing in ATP depletion and cell death in human glioblastoma cells. Biochim Biophys Acta
1807(6):726–734
71. Harper ME, Antoniou A, Villalobos-Menuey E, Russo A, Trauger R, Vendemelio M, George
A, Bartholomew R, Carlo D, Shaikh A, Kupperman J, Newell EW, Bespalov IA, Wallace
SS, Liu Y, Rogers JR, Gibbs GL, Leahy JL, Camley RE, Melamede R, Newell MK (2002)
Characterization of a novel metabolic strategy used by drug-resistant tumor cells. FASEB J
16(12):1550–1557
72. Samudio I, Harmancey R, Fiegl M, Kantarjian H, Konopleva M, Korchin B, Kaluarachchi
K, Bornmann W, Duvvuri S, Taegtmeyer H, Andreeff M (2010) Pharmacologic inhibition
of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction. J Clin Invest
120(1):142–156
73. Zaugg K, Yao Y, Reilly PT, Kannan K, Kiarash R, Mason J, Huang P, Sawyer SK, Fuerth
B, Faubert B, Kalliomaki T, Elia A, Luo X, Nadeem V, Bungard D, Yalavarthi S, Growney
JD, Wakeham A, Moolani Y, Silvester J, Ten AY, Bakker W, Tsuchihara K, Berger SL, Hill
RP, Jones RG, Tsao M, Robinson MO, Thompson CB, Pan G, Mak TW (2011) Carnitine
palmitoyltransferase 1 C promotes cell survival and tumor growth under conditions of met-
abolic stress. Genes Dev 25(10):1041–1051
74. Niemen KM, Kenny HA, Penicka CV, Ladanyi A, Buell-Gutbrod R, Zilihardt MR, Romero
IL, Carey MS, Mills GB, Hotamisligil GS, Yamada SD, Peter ME, Gwin K, Lengyel E
(2011) Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor
growth. Nat Med 17(11):1498–1503
75. Pizer ES, Wood FD, Pasternack GR, Kuhajda FP (1996) Fatty acid synthase (FAS): a tar-
get for cytotoxic antimetabolites in HL60 promyelocytic leukemia cells. Cancer Res
56(4):745–751
76. Duarte NC, Becker SA, Jamshidi N, Thiele I, Mo ML, Vo TD, Srivas R, Palsson BO (2007)
Global reconstruction of the human metabolic network based on genomic and bibliomic
data. Proc Natl Acad Sci U S A 104(6):1777–1782
77. Jain M, Nilsson R, Sharma S, Madhusudhan N, Kitami T, Souza AL, Kafri R, Kirschner
MW, Clish CB, Mootha VK (2012) Metabolite profiling identifies a key role for glycine in
rapid cancer cell proliferation. Science 336(6064):1040–1044
78. Kriegler MP, Pawlowski AM, Livingston DM (1981) Cysteine auxotrophy of human leuke-
mic lymphoblasts is associated with decreased amounts of intracellular cystathionase mes-
senger ribonucleic acid. Biochemistry 20(5):1312–1318
79. Ensor CM, Holtsberg FW, Bomalaski JS, Clark MA (2002) Pegylated arginine deiminase
(ADi-SS PEG 20,000 mw) inhibits human melanomas and hepatocellular carcinomas in
vitro and in vivo. Cancer Res 62(19):5443–5450
50 D. M. Hockenbery et al.
CB (2010) The common feature of leukemia-associated IDH1 and IDH2 mutations is a neo-
morphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell
17(3):225–234
97. Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Wang P, Xiao MT, Liu LX,
Jiang WQ, Liu J, Zhang JY, Wang B, Frye S, Zhang Y, Xu YH, Lei QY, Guan KL, Zhao
SM, Xiong Y (2011) Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-
ketoglutarate-dependent dioxygenases. Cancer Cell 19(1):17–30
98. Chowdhury R, Yeoh KK, Tian YM, Hillringhaus L, Bagg EA, Rose NR, Leung IK, Li
XS, Woon EC, Yang M, McDonough MA, King ON, Clifton IJ, Klose RJ, Claridge TD,
Ratcliffe PJ, Schofield CJ, Kawamura A (2011) The oncometabolite 2-hydroxyglutarate
inhibits histone lysine demethylases. EMBO Rep 12(5):463–469
99. Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, Edwards CR, Khanin R,
Figueroa ME, Melnick A, Wellen KE, O’Rourke DM, Berger SL, Chan TA, Levine RL,
Mellinghoff IK, Thompson CB (2012) IDH mutation impairs histone demethylation and
results in a block to cell differentiation. Nature 483(7390):474–478
100. Turcan S, Rohle D, Goenka A, Walsh LA, Fang F, Yilmaz E, Campos C, Fabius AW, Lu C,
Ward PS, Thompson CB, Kaufman A, Guryanova O, Levine R, Heguy A, Viale A, Morris
LG, Huse JT, Mellinghoff IK, Chan TA (2012) IDH1 mutation is sufficient to establish the
glioma hypermethylator phenotype. Nature 483(7390):479–483
Chapter 3
Emerging Opportunities for Targeting
the Tumor–Stroma Interactions
for Increasing the Efficacy
of Chemotherapy
Abstract It has become evident that tumor cells utilize survival signals that ema-
nate from the tumor microenvironment to aid in survival and disease progression.
Experimental evidence indicates that these same pathways contribute to de novo
drug resistance. Identification of the mechanisms underlying the recruitment of
accessory cells and survival signals provided by normal cells has provided a novel
area for drug discovery for increasing the efficacy of cancer therapy.
3.1 Introduction
due to increased specificity for the cancer cell as well as reduce the emergence of
multi-drug resistant phenotypes associated with standard therapy.
In support of this potential promise of targeted agents are data generated in
chronic myeloid leukemia (CML) which is driven by the BCR-ABL oncogene
[2, 3]. A myriad of experimental data support the findings that the expression of
the fusion oncogene BCR-ABL is sufficient for transformation and disease pro-
gression using transplant and transgene in vivo model systems [4, 5]. The identi-
fication of BCR-ABL as driving transformation of CML provided an ideal target
for drug discovery, and first- and second-generation BCR-ABL inhibitors have
provided proof-of-principle that this approach may indeed lead to clinical success
[6]. However, it became clear that although BCR-ABL inhibitors are very effec-
tive when treating patients in the early chronic stage of the disease, BCR-ABL
inhibitors were not curative and similar to DNA-damaging agents did not bypass
the emergence of drug resistance. Because CML is initially driven by one driver
oncogene, it represents an ideal disease to delineate de novo drug resistance and
the emergence of acquired resistance. Acquired resistance to BCR-ABL inhibitors
includes point mutations in what is commonly referred to as the gatekeeper region
of the molecule [7]. These mutations result in reduced affinity of drug binding
and thereby render the cells insensitive to the compound. The identification of the
mutations, along with structural information, led to the development of second-
generation compounds predicted to be active in the presence of specific gatekeeper
mutations, and indeed, these molecules do show promising activity in the subset of
patients harboring the T315I gatekeeper mutation [8]. Other mechanisms reported
to contribute toward resistance to BCR-ABL inhibitors include increased expres-
sion of transporters, quiescence, and progression of the disease to blast phase
which typically is the result of expression of additional driver mutations and sub-
sequent resistance to BCR-ABL inhibitors.
Of particular interest for this review is the observation that even in patients
exquisitely sensitive to BCR-ABL inhibitors, minimal residual disease can be
detected at the molecular level. Subsequent to the identification of BCR-ABL came
the identification of activating EGFR mutations in lung cancer and head and neck
cancer and expression of BRAF in melanoma. Again, the identification of these
driver oncogenes led to the rapid translation of specific kinase inhibitors. However,
within the landscape of the complexity associated with lung cancer, colon can-
cer, and melanoma, these inhibitors, albeit effective, demonstrated shorter clinical
response time before the emergence of resistance was a clinical concern [9–12].
Thus, it became clear that targeted therapies which potently inhibit the activity of
a driver oncogene do not circumvent the emergence of drug resistance. The emer-
gence of drug resistance can be caused by mutations and overamplification of tar-
gets which are mechanisms that contribute to acquired resistance. However, more
recently, attention has shifted toward the role of the tumors inherent ability for
co-opting and recruiting normal cells into the niche to provide an adaptive envi-
ronment that favors the survival of tumor cells [13], and hence, as the niche is con-
ditioned by the tumor, the dependency of cancer cells on the activity of a specific
oncogene is diminished. This review will focus on mechanisms associated with
3 Emerging Opportunities for Targeting 55
phosphorylation at the Tyr705 residue in the C-terminal domain [33–35]. The acti-
vated phosphorylated STAT3 then undergoes homodimerization by reciprocal inter-
action between the SH2 domain of one monomer and the phosphorylated Tyr705
residue of its dimerizing partner. STAT3 dimer can translocate to the nucleus and bind
specific DNA sequences and regulate the transcription of the responsive gene [36, 37].
Targeting of STAT3 could potentially occur at multiple points of regulation
including inhibition of the activating receptor kinase or cytokine receptor, via inhi-
bition of JAK kinases or by disrupting the dimerization or subsequent DNA bind-
ing. Specifically, disrupting the dimerization and DNA binding has proven to be a
difficult task using small molecule design. Experimental evidence indicates that
in some tumors, IL-6 plays a dominant role in conferring resistance. For example,
in neuroblastoma, Ara et al. found that blocking IL-6 signaling using a blocking
IL-6Rα antibody reversed resistance to the topoisomerase II inhibitor, etoposide
when neuroblastoma cells were cultured in the presence of MSCs [38]. In multiple
myeloma, IL-6 has been shown to be critical for cellular survival via activation
of the JAK/STAT3 pathway and subsequent increased expression of the anti-
apoptotic family member Bcl-XL [39]. More recently, several groups have pro-
vided evidence that targeting IL-6 or JAKs in the context of co-culturing multiple
myeloma cells with bone stroma cells enhances the efficacy of standard therapy
including melphalan and dexamethasone [40–44]. Thus, in myeloma, it appears
that IL-6 is the predominant cytokine-driving drug resistance in the bone marrow
compartment. In contrast, in CML, experimental evidence indicates that multiple
soluble factors can activate STAT3 and induce resistance to BCR-ABL inhibitors
[20]. Due to the plethora of cytokines and growth factors present in the bone mar-
row milieu capable of activating the JAK/STAT3 pathway, it is likely that target-
ing downstream of the cytokine receptor at the level of JAK or STAT3 will be the
most effective strategy and may circumvent resistance due to the selection of cells
that utilize an alternative receptor for activating the JAK/STAT3 pathway [20] (for
a complete list of drugs targeting JAK/STAT3 pathway see Table 3.2). MSCs are
specifically relevant to tumors which home to the bone such as multiple myeloma
or metastasize to the bone such as lung, breast, and prostate.
3 Emerging Opportunities for Targeting 57
3.2.2 Immune Cells
3.2.3 Endothelial Cells
Once tumors have evaded the immune system and established growth that exceeds
a few millimeters, the tumor must quickly establish a blood supply to sustain sur-
vival and growth. This is achieved by the production of stimulatory factors that
attract and activate endothelial cells to form angiogenic vesicles. As the tumor
grows, localized areas of hypoxia arise and results in induced HIF-1α expression
in the tumor, endothelial, and stroma cells. HIF-1α expression is a major player
for driving the expression and secretion of multiple angiogenic factors including
VEGF, PDGF, FGF, angiopoietins, and SDF-1α [55]. Additionally, integrins are
known to augment growth factor receptor signaling and thus may provide an addi-
tional target for inhibiting angiogenesis [56]. One soluble factor that is critical for
the angiogenic switch is vascular endothelial growth factor (VEGF). VEGF stimu-
lates the sprouting and proliferation of endothelial cells. However, it is becoming
clear that targeting VEGF alone will likely not be sufficient for inhibiting angio-
genesis, as resistance has been observed preclinically and clinically [57]. Overall,
due to the ability of tumor cells to quickly adapt to a reduction in blood flow, it
is currently not clear how effective targeting angiogenesis will be in controlling
cancer. Although it is feasible, as we learn more about the bidirectional communi-
cations between the tumor, myeloid population, stroma, and endothelial cells that
such strategies may be fully realized.
3.3.1 Galectins
Cell surface receptors like cytokine and growth factor receptors are
N-glycosylated transmembrane glycoproteins that can bind glycan-binding pro-
teins called lectins [58, 59]. Galectins are members of the lectin family that has
an affinity for β-galactoside sugars, especially N-acetyllactosamine [60]. Galectins
have been demonstrated to crosslink receptor glycoproteins at the cell surface
and form lattices and thereby dictate the distribution and the retention time of the
receptor on the cell surface [61, 62]. Interestingly, the strength of the interaction
between the galectins and the glycoproteins, and thus the lattice structures, can be
modulated by either regulating the cell surface protein glycosylation or by alter-
ing the expression of galectins [63, 64]. This is important because the distribution
pattern of the cell surface receptors and their residency time on the plasma mem-
brane can modulate the cells response to its microenvironment and is a prevailing
feature of oncogenesis [65]. For example, Mgat5 gene encodes a Golgi-localized
enzyme that initiates N-glycan synthesis on newly formed cell surface receptors
60 R. R. Nair et al.
that is recognized by galectins [66]. Granovsky et al. have shown that Mgat5
knockdown inhibits tumor progression and upregulation of Mgat5 increases tumo-
rigenesis in mouse tumor models [67]. Furthermore, Partridge et al. demonstrated
that in Mgat5−/− tumor cells, cell surface density of EGFR and TGF-β receptor
was reduced and activation of these receptors increased their endocytosis, mak-
ing the cells less sensitive to stimulation by EGF, IGF, PDGF, bFGF, and TGF-β
[68]. Also, these receptor glycoproteins were dependent on galectin-mediated lat-
tice formation for surface retention after receptor activation, thereby prolonging
the sensitivity of the tumor cells to EGF and TGF-β [68]. This observation was
later confirmed by Lajoie et al., who showed that EGFR localization, and thus, the
magnitude of EGFR signaling in tumor cells was very much dependent upon the
galectin lattice structures opposing and prevailing over the oligomerized caveo-
lin-1 microdomains that mediate endocytosis [69]. Finally, regulated glycosylation
of cell surface receptors can create or abolish binding domains for galectins and
thereby control important cellular response such as immune responses like T-cell
activation, homing, and survival [70].
Presently, there are 15 members of the galectin family identified that all contain
a conserved globular domain called carbohydrate recognition domain (CRD) that
recognizes and binds to β-galactosidase [71]. The specificity of the galectins for
their substrates is defined by the ligand-binding groove contained within the CRD
of the protein [72]. The 15 family members are divided into three sub-families on
the basis of their molecular structure (see Fig. 3.1): (1) Galectin-1, Galectin-2,
Galectin-5, Galectin-7, Galectin-10, Galectin-11, Galectin-13, Galectin-14, and
Galectin-15 are categorized under the name “proto-type” galectins as they are
comprised of a single polypeptide chain that is able to dimerize; (2) Galectin-3
is categorized under the name “chimera-type” galectin as it consists of one
C-terminal CRD linked to an N-terminal peptide that is able to pentamerize; and
finally, (3) Galectin-4, Galectin-6, Galectin-8, Galectin-9 and Galectin-12 are cat-
egorized under the name “tandem repeat-type” galectins as they are composed of
two CRDs connected by a linker peptide [72–74]. Irrespective of their type, all
galectins have the ability to recognize and bind galactosyl residues without the
requirement for any other catalyst like cations [71]. Additionally, these lectins are
widely localized ranging from within the cytoplasmic and nuclear compartment to
the extracellular compartment where they modulate a myriad of biologic process
(for review see, [75]). Below, we will briefly discuss the role of galectins in tumo-
rigenesis by highlighting the role of each type of the galectin family.
3.3.2 Galectin-1
CRD
CRD
Tandem repeat-type
galectin
Proto-type
galectin Chimera-type galectin
Fig. 3.1 Model structures of the different types of galectins. Galectins are divided into three
major types according to their structures. The prototype galectin contains one CRD and can exist
as a monomer or a dimer. The chimeric type galectin contains one CRD that is covalently linked
to a non-carbohydrate tail. The chimeric type of galectin, like galectin-3, can exist as a pentamer
and a monomer. Finally, the last type of galectin is the tandem repeat-type galectin that harbors
CRD domains that are covalently linked by a small peptide linker
3.3.3 Galectin-3
This protein is a unique chimera-type galectin that has been associated with sev-
eral models of immune disease and cancer [82–84]. Jeon et al. [85] have shown
that galectin-3 can exert cytokine-like regulatory actions by the activation of the
62 R. R. Nair et al.
3.3.4 Galectin-9
Hypoxia can occur at the site of the primary tumor due to a decreased oxygen
content found in the center of the tumor or at specialized niches found in meta-
static sites. For example, the bone marrow microenvironment is characteristically
hypoxic; direct measurement of oxygen levels has estimated the oxygen content
to be 1 to 2 % in the bone marrow [95]. Thus, hypoxia represents an initial selec-
tion pressure that contributes to necrotic cell death but also provides pressure for
adaptation and tumor progression. Indeed, exposure to hypoxia can induce resist-
ance in multiple tumor types including leukemia and lung cancer cell line models
[96, 97]. Moreover, in CML, hypoxic conditions that mimic the O2 content of the
bone marrow confer resistance to BCR-ABL inhibitors, suggesting that hypoxia
may contribute to failure to eradicate minimal residual disease [98]. Hypoxia
induces a myriad of effects on tumor cells including selection for p53 mutations
[99] and increased genomic instability due to increases in reactive oxygen spe-
cies and attenuation of DNA repair pathways [100]. Experimental findings suggest
that hypoxia can contribute to angiogenesis, invasiveness, metastasis as well as
immune suppression [101–103]. Many of the phenotypes associated with hypoxia
are the result of the induction of the hypoxia-inducible factor (HIF) family of tran-
scription factors. Three members constitute the family (HIF-1, HIF-2, and HIF-3).
HIF-1α is targeted for degradation by ubiquitination, which is regulated by the
E3 ligase von Hippel–Lindau (VHL) tumor suppressor. Under normal oxygen
levels, HIF-1α is hydroxylated on proline 402 and 564 by prolyl-4-hydroxylases
(PHDs). The reaction requires oxygen and 2-oxoglutarate as substrates [104, 105].
Thus, under normal oxygen concentration, HIF family members are hydroxylated
and subsequently targeted for ubquitination and degradation. In contrast, under
hypoxic conditions, HIF-1α is stabilized and can form a heterodimer with HIF-1β
and drive transcription of target genes [106].
A hypoxic environment allows for two independent targeting strategies: (1)
development of bioreductive prodrugs and (2) use of drugs that inhibits targets that
are specifically expressed under hypoxic conditions. Bioreductive prodrugs typi-
cally take advantage of redox cycling such that the levels of the prodrug radical
are kept low in oxic cells at the expense of generation of superoxide. Common
moieties that have the potential to be metabolized under hypoxic conditions
include nitro groups, quinones, aromatic N-oxides, aliphatic N-oxides, and transi-
tion metals. One example of a hypoxia-activated prodrug is TH-302. TH-302 is
nitrogen mustard based prodrug with promising activity in a phase I clinical trial
for advanced solid tumors [107]. Because the bone marrow is hypoxic and nitro-
gen mustards such as melphalan are a mainstay of therapy for myeloma which
homes to the bone, it would be of interest to test prodrugs in the context of tumors
which home or metastasize to the bone. The identification of pathways that are
activated upon hypoxia that contributes to cellular survival represents an additional
strategy for specifically targeting the hypoxic tumor microenvironment. Three
main pathways activated by hypoxia are the HIF family of transcription factors,
64 R. R. Nair et al.
the unfolded protein response (UPR), and autophagy. Severe hypoxia disrupts
the formation of disulfide bond formation leading to misfolded proteins and acti-
vation of the UPR. Thus, targeting the UPR pathway is attractive for selectively
eliminating hypoxic tumors. One promising target in the UPR pathway is the ino-
sitol-requiring enzyme 1 or IRE1 [108]. Inhibitors of IRE1 are currently in devel-
opment and show activity in myeloma models which are tipped toward ER stress
due to the large production of monoclonal antibodies in these cancerous cells
[109, 110]. Another strategy is the use of proteasome inhibitors which also can
trigger additional stress on the UPR response, and correspondingly, bortezomib
shows activity in hypoxic tumor sites [111]. Thus, inhibition of the proteasome
exaggerates the UPR and tips the threshold toward cell death under hypoxic con-
ditions and may represent an attractive strategy for eliminating cells residing in
hypoxic microenvironments.
3.5.1 Integrins
Damiano et al. [112] reported in 1999 that adhesion of myeloma cells via α4β1
integrin was sufficient to cause resistance to structurally and mechanistically
diverse chemotherapeutic agents. Furthermore, they reported that selection for
acquired drug resistance selected for cells with increased expression of β1 and β7
integrin. This report suggested that cell adhesion could contribute to an adaptive
response for cells to evade cytotoxic insult. More recently, α4 integrin expression
was shown to be increased in ex vivo samples collected from relapsed myeloma
patients compared with specimens obtained from newly diagnosed myeloma
patients, albeit only a small sample size was reported [113]. VLA-4 integrin is
known to be important for homing, and more recent evidence indicates that VLA-
4/VCAM-1 interactions between osteoclasts and breast cancer cells drive the
cycle of bone destruction and tumor growth in metastatic models of breast can-
cer [114]. A similar finding was reported for myeloma where adhesion to stroma
induced MIP-1alpha and beta which in turn activated osteoclasts [115] and con-
tributed to further bone destruction and tumor growth. Collectively, these data
indicate that VLA-4 integrin is an attractive target for myeloma and perhaps
solid tumors which metastasize to the bone. Integrins are heterodimeric proteins
comprised of an α and β subunit. Eighteen α and 8 β subunits have been identi-
fied and complex to form 24 known αβ heterodimers. Integrins bind to a diverse
set of extracellular ligands which are found in the extracellular matrix. Integrins
do not contain intrinsic kinase activity; however, integrin ligation allows for the
formation of focal adhesions containing adaptor proteins critical for the activa-
tion of signaling complexes (see Fig. 3.2). Molecules contained within the focal
3 Emerging Opportunities for Targeting 65
adhesions include integrins, FAK/Pyk2, Src, and several scaffold proteins includ-
ing talin, actin, and paxillin. Downstream signaling that is amplified following the
ligation of integrins includes activation of the MAPK-, Src- and AKT-mediated
survival pathways [116, 117]. Finally, integrin signaling is known to augment
growth factor– and cytokine-mediated signaling [27, 118]. Due to the importance
of integrins in sensing and transmitting external cues and contribution to survival,
migration, and growth, integrins remain an attractive target for cancer as well
as autoimmune diseases [32]. Integrins can be effectively targeted by utilizing
blocking antibodies and peptidomimetics which typically compete with consen-
sus ligand sequences such as RGD peptides. The development of small molecules
demonstrating specificity for integrins has been difficult due to inherent difficul-
ties of targeting protein–protein interactions with small molecules.
The structure of integrins has recently been elucidated utilizing crystallogra-
phy to inform structural models [119–121]. The α subunit is comprised of a short
66 R. R. Nair et al.
Fig. 3.3 Steps leading to the activation and clustering of integrins. In resting cells, the integrins
are in a bent, low-affinity state. However, once the cell is exposed to an environment with an
abundance of cytokines or growth factors, an inside-out activation pathway is triggered that cul-
minates in an extended, high-affinity state of integrins. The integrins are now receptive to ligand
binding, either in the form of adhesion to the extracellular matrix or adhesion to other cells
within its microenvironment. Ligand binding causes clustering of integrins, with polymerization
of actin, and induction of an outside-in activation cascade
3 Emerging Opportunities for Targeting 67
integrin and competes with RGD sequences found in components of the matrix
[141]. RGD sequences are contained in multiple ligands, yet integrins demon-
strate specificity which cannot be entirely explained by flanking sequences. Thus,
it was postulated that secondary structure was critical for sequence recognition.
Cyclization represents a strategy to constrain the recognition sequence in a well-
defined secondary structure. Kessler and colleagues performed extensive structure
activity relationship prior to knowledge of crystal structures, and thus, they applied
the following strategies for optimizing RGD sequences for inhibition of αVβ3-
mediated adhesion. They reduced the conformation space using cyclization strat-
egies, spatial screening of peptides, and N-methyl scanning [141]. N-methylation
of peptide bonds often increases their metabolic stability and bioavailability [142].
Merck-Serona has published on the phase II trial and just completed the accrual of
a phase III clinical trial with cilengitide for the treatment for glioblastoma [143].
Preclinically, cilengitide was shown to have activity by inhibiting growth directly,
as well as inhibiting invasion and angiogenesis [144]. These studies will provide
proof-of-principle whether targeting cell adhesion receptors with cyclized peptides
is a viable strategy for drug discovery and development in oncology.
3.5.2 CD44
CD44 is a family of cell surface receptors that similar to integrins mediate cell–
matrix adhesion. One gene encodes the CD44 family, but alternative splicing leads
to multiple variants which are thought to be, in part, responsible for the apparent
diverse functions attributed to this single-pass membrane-spanning cell surface pro-
tein. CD44 facilitates cell adhesion and metastasis, augments growth factor signal-
ing, and expression of CD44 protects against hypoxia-induced lung injury
[145–155]. CD44 can be shed from the cell membrane and soluble CD44 is associ-
ated with advanced disease in chronic lymphocytic leukemia [156–160].
Hyaluronic acid (HA) is the most predominant ligand for CD44. HA is an abundant
polysaccharide found in extracellular matrixes. However, CD44 can also bind
fibronectin and osteopontin [161]. The structure of CD44 consists of a constant
region spanning the first five exons which defines the HA-binding domain and is
found in all splice variants. Exon 7 through 15 (corresponding to v2–v10) is the
variable region of the molecule. This region is localized on the external domain and
adds to the stem region of CD44. The highly conserved cytoplasmic region has part
of exon 18 combined with exon 19 and 20 [162]. CD44 variant expression associ-
ates with markers of cancer stem cells and is a poor prognostic indicator for many
cancer types including multiple myeloma [163–165]. Paradoxically, the activation
of CD44, depending on the cell context, is associated with both cell death and sur-
vival [64, 166–168]. Moreover, CD44v associates with α4β1 integrin in CLL and
positively regulates adhesion of myeloma cells to stroma and extracellular matrices
[148, 165, 169]. Finally, it is known that CD44 associates with VLA-4 integrin in
T cells and CLL [169, 170]. Along with VLA-4, CD44 is considered the major
3 Emerging Opportunities for Targeting 69
homing receptor for hematopoietic cells for trafficking to the bone marrow com-
partment [171] and thus, similar to VLA-4, may play a role in the recruitment of
accessory cells to the tumor site as well as metastasis of the tumor. CD44 has an
N-terminal ligand-binding ectodomain containing the Link module; comprised of
three intradisulfide bonds, a stalk region containing one or multiple variant exons, a
transmembrane domain, and a short C-terminal tail which is important for cytoskel-
etal attachment and signal transduction. CD44 has a highly conserved amino acid
sequence among mammalian and avian species sharing between 47–93 % sequence
homology with humans and an almost identical homology in the Link module,
transmembrane, and cytoplasmic tail regions [172]. The CD44 receptor also con-
tains many posttranslational modifications. It has been estimated that 25–40 % of
CD44 molecules are phosphorylated at Ser325 and that CD44 is constitutively
phosphorylated at this site in cultured cells [172]. While the exact role of Ser325
phosphorylation is unclear, mutations at this site have been shown to inhibit HA
binding and cell migration, as well as modulating the interaction of CD44 with the
ERM (ezrin, radixin, moesin) family of proteins [173]. CD44 is also palmitoylated
at Cys286 and Cys295, and while the exact roles these modifications play are not
yet fully understood, it appears that palmitoylation enhances the association
between CD44 and ankyrin and may also induce clustering of CD44 into mem-
brane subdomains [172]. The CD44 receptor is also highly glycosylated and linked
to chondroitin sulfate; with at least 5 conserved N-glycosylation sites in the ectodo-
main as well as being highly O-glycosylated in the extracellular region, both of
which have been shown to affect ligand-binding affinity [173]. The crystal structure
of HA bound to murine CD44 has been solved, and it has been observed that CD44
adopts two conformational binding states and this binding is the result of hydrogen
bonding [174]. Further studies by nuclear magnetic resonance (NMR) and surface
plasmon resonance (SPR) have further elucidated the two binding configurations of
the CD44 receptor, designated as ordered (O) and partial disordered (PO) [175,
176]. It has been shown that the binding of HA oligomers at an allosteric site on the
ectodomain leads to conformational change in the Link module and switches the
module from the O to PO configuration. Furthermore, while the minimum binding
requires an HA hexamer, recent studies have demonstrated that there is a positive
correlation with the length of HA and binding affinity. It has long been speculated
that this is the result of multiple CD44 molecules binding to one HA polysaccha-
ride. This was recently confirmed by Wolny et al. using an artificial membrane sys-
tem in which they demonstrated that multiple CD44 molecules can bind a single
HA polysaccharide through a multivalent nature [176]. While the crystal structures
of CD44 bound to other ligands have yet to be solved, it is postulated that other
ligands bind in the ectodomain adjacent to the Link module. The CD44 transcript
contains twenty exons that give rise to seventeen known isoforms through alterna-
tive splicing and posttranslational modifications. The most ubiquitously expressed
isoform is the standard form (CD44s), which is comprised of exons 1–5 and 16–20
[172]. CD44 expression is regulated through receptor-mediated endocytosis as well
as the proteolytic cleavage of the extracellular domain by various matrix metallo-
proteinases (MMPs). CD44 activation through ligand binding has been shown to
70 R. R. Nair et al.
have diverse cellular effects. CD44s activation has been shown to activate apoptosis
in Jurkat cell lines, whereas CD44v induction has been suggested to do the con-
verse via sequestering FasR [147]. Similar to integrins, CD44 has no intrinsic
kinase activity; however, it gains access to kinases such as focal adhesion kinase
(FAK), protein kinase C (PKC), and phosphatidylinositol 3-kinase (PI3K) through
adaptor proteins such as the ERM family [177]. In addition to CD44 gaining access
to kinases through adapter proteins, it has been demonstrated that CD44 can also
gain access to other signaling complexes through association with other adhesion
receptors, most notably α4 integrin in lymphocyte extravasation as well as cell
growth and proliferation signaling through the human epidermal growth factor
receptor 2 (HER2), c-Met, and VEGF [178–180]. CD44-deficient mice exhibit nor-
mal development; however, the mice are resistant to T-cell activation-induced cell
death (AICD) [181]. Recently, several in vitro studies targeting CD44 with mono-
clonal antibodies alone and in combination with low molecular weight HA have
shown induction of apoptosis in myeloid cells as well as a novel caspase-independ-
ent pathway in erythroleukemia cells. Together, these data suggest that agonistic
signals may actually activate cell death in tumor cells. More specifically, one study
was able to eradicate human acute myeloid leukemia stem cells in non-diabetic
severe combined immune-deficient (SCID) mice by targeting CD44 receptor with a
CD44 monoclonal antibody that recognized all isoforms and was characterized as
an activating antibody [64]. There has also been an interest in targeting CD44 via
peptidomimetics. In one study, a group using a CD44v6-derived peptide has indi-
cated a role of CD44 in VEGF signaling and angiogenesis [182]. A second peptide
named A6 (acetyl-KPSSPPEE-amino) has been shown to activate CD44 and inhibit
migration and metastasis in CD44-expressing cells [183]. Furthermore, this peptide
has shown promise in phase I and phase II clinical trials in patients with ovarian
cancer. Another molecule that has shown promise in early stage clinical trials is the
immunoconjugate bivatuzumab, a human anti-CD44v6 monoclonal antibody cou-
pled with a benzoansamacrolide moiety, which has been tested in head and neck
squamous cell carcinomas [184]. While these molecules have shown promise in the
clinic, targeting CD44 will remain a difficult task until the diverse expression and
signaling of the many CD44 isoforms is more fully understood in both healthy and
diseased cell lineages.
3.6.1 FAK/Pyk2
Due to the existence of multiple cell adhesion receptors and potential r edundancy, it
is attractive to speculate that downstream targets will be more efficacious for target-
ing cell adhesion mediated drug resistance or CAM-DR. Immediately, downstream
3 Emerging Opportunities for Targeting 71
of multiple cell adhesion receptors, as well as cytokine and growth factor recep-
tors, is FAK and/or Pyk2 which is a FAK homolog. FAK is a non-receptor tyrosine
kinase, is ubiquitously expressed, and localizes to focal adhesions. FAK contains
three major structural domains: (1) an N-terminal FERM domain, (2) a tyrosine
kinase catalytic domain, and (3) a C-terminal focal adhesion–targeting domain
[185]. The activation of FAK requires autophosphorylation at Y397 following the
integrin ligation [186]. Phosphorylation of FAK leads to the recruitment and acti-
vation of Src family members as well as other components of the focal adhesion
including p130Cas and paxillin [187–189]. The activation of FAK leads to the inte-
gration and spatial organization of signaling pathways including activation of AKT,
Src, and the MAPK pathway. Pyk2 is a closely related homolog to FAK, with a
more restricted tissue distribution as expression is limited predominately to hemat-
opoietic cells, vascular smooth muscle, endothelium, spleen, kidney, and the cen-
tral nervous system [190]. Although Pyk2 and FAK are functionally related, they
are not redundant with respect to the phenotype observed in the respective knock-
out mice. FAK knockout mice are embryonic lethal due to defects in the axial
mesodermal tissue and cardiovascular system [191]. In contrast, Pyk2 knockout
mice develop normally, but do exhibit defective macrophage migration and have
increased bone density [192, 193]. PF-562,271 is a promising dual FAK/Pyk2
inhibitor being developed by Pfizer. This compound competes for the ATP-binding
domain and is a reversible inhibitor of FAK and Pyk2 catalytic activity. This com-
pound has shown anti-tumor activity in multiple in vivo xenograft tumor models
[194]. Moreover, in models of lytic bone disease involving implantation of breast
cancer cells into the tibia, investigators have shown that the FAK/Pyk2 inhibitor
reduced the tumor burden and lytic lesions [195]. This is consistent with data show-
ing that Pyk2−/− knockout mice show increased bone mass, indicating that the inhi-
bition of Pyk2 may be helpful for diseases such as myeloma, breast, and lung that
either home or metastasize to the bone and induce lytic lesions [193]. Recently, a
phase 1 clinical trial was reported that showed the compound was well tolerated,
and 12 % of patients showed prolonged stable disease for six or more cycles [196].
3.7 Conclusion
Historically, drug development has focused on developing strategies that will tar-
get the Achilles heel of tumors. Strategies with a track record of success in the
clinic include targeting rapidly dividing cells with DNA-damaging agents or radia-
tion. More recently, focus has centered on targeting oncogene addiction intrinsic to
the tumor cells. However, emerging evidence indicates tumors are very efficient at
using opportunistic signals provided by the microenvironment, and in fact, current
therapies may select for cells more fit to utilize survival signals contained within
the microenvironment allowing for survival during exposure to therapy. The chal-
lenge with targeting the microenvironment will be to a) determine redundancy
72 R. R. Nair et al.
and delineate whether multiple targets will need to be inhibited and b) determine
patient variability contained within the tumor microenvironment and whether a
personalized medicine approach will be required for targeting the tumor micro-
environment. Targeting the microenvironment will present challenges due to the
complexity and the inherent ability of tumors to provide adaptive signals that con-
tinue to inform the surrounding niche to favor tumor progression in a dynamic
rather than static fashion. However, it is clear that target validation and drug dis-
covery will need to consider both the intrinsic oncogenic signals and the tumor
microenvironment for increasing the efficacy of standard therapy in cancer indica-
tions that are currently incurable.
Acknowledgments We are grateful to Deepa G Rathod for her assistance in preparation of the
tables & figures.
References
1. Gottesman MM, Ling V (2006) The molecular basis of multidrug resistance in cancer: the
early years of P-glycoprotein research. FEBS Lett 580:998–1009
2. Buchdunger E, Zimmermann J, Mett H, Meyer T, Muller M, Druker BJ, Lydon NB (1996)
Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimi-
dine derivative. Cancer Res 56:100–104
3. Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM, Fanning S, Zimmermann J, Lydon
NB (1996) Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-
Abl positive cells. Nat Med 2:561–566
4. Carlesso N, Griffin JD, Druker BJ (1994) Use of a temperature-sensitive mutant to define
the biological effects of the p210BCR-ABL tyrosine kinase on proliferation of a factor-
dependent murine myeloid cell line. Oncogene 9:149–156
5. Zhang X, Ren R (1998) Bcr-Abl efficiently induces a myeloproliferative disease and pro-
duction of excess interleukin-3 and granulocyte-macrophage colony-stimulating factor in
mice: a novel model for chronic myelogenous leukemia. Blood 92:3829–3840
6. Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger
MW, Silver RT, Goldman JM, Stone RM et al (2006) Five-year follow-up of patients receiv-
ing imatinib for chronic myeloid leukemia. N Engl J Med 355:2408–2417
7. Shah NP, Nicoll JM, Nagar B, Gorre ME, Paquette RL, Kuriyan J, Sawyers CL (2002)
Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine
kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leuke-
mia. Cancer Cell 2:117–125
8. O’Hare T, Shakespeare WC, Zhu X, Eide CA, Rivera VM, Wang F, Adrian LT, Zhou T,
Huang WS, Xu Q et al (2009) AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leu-
kemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer
Cell 16:401–412
9. Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, Lee MK, Attar N, Sazegar
H et al (2010) Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or
N-RAS upregulation. Nature 468:973–977
10. Kobayashi S, Boggon TJ, Dayaram T, Janne PA, Kocher O, Meyerson M, Johnson BE, Eck
MJ, Tenen DG, Halmos B (2005) EGFR mutation and resistance of non-small-cell lung can-
cer to gefitinib. N Engl J Med 352:786–792
11. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O’Dwyer PJ, Lee
RJ, Grippo JF, Nolop K et al (2010) Inhibition of mutated, activated BRAF in metastatic
melanoma. N Engl J Med 363:809–819
3 Emerging Opportunities for Targeting 73
12. Poulikakos PI, Rosen N (2011) Mutant BRAF melanomas–dependence and resistance.
Cancer Cell 19:11–15
13. Hanahan D, Coussens LM (2012) Accessories to the crime: functions of cells recruited to
the tumor microenvironment. Cancer Cell 21:309–322
14. Meads MB, Gatenby RA, Dalton WS (2009) Environment-mediated drug resistance: a
major contributor to minimal residual disease. Nat Rev Cancer 9:665–674
15. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA,
Simonetti DW, Craig S, Marshak DR (1999) Multilineage potential of adult human mesen-
chymal stem cells. Science 284:143–147
16. Bewry NN, Nair RR, Emmons MF, Boulware D, Pinilla-Ibarz J, Hazlehurst LA (2008) Stat3
contributes to resistance toward BCR-ABL inhibitors in a bone marrow microenvironment
model of drug resistance. Mol Cancer Ther 7:3169–3175
17. Nair RR, Tolentino J, Hazlehurst LA (2010) The bone marrow microenvironment as a sanc-
tuary for minimal residual disease in CML. Biochem Pharmacol 80(5):602–612
18. Nefedova Y, Landowski TH, Dalton WS (2003) Bone marrow stromal-derived soluble fac-
tors and direct cell contact contribute to de novo drug resistance of myeloma cells by dis-
tinct mechanisms. Leukemia 17:1175–1182
19. Lagneaux L, Delforge A, De Bruyn C, Bernier M, Bron D (1999) Adhesion to bone mar-
row stroma inhibits apoptosis of chronic lymphocytic leukemia cells. Leuk Lymphoma
35:445–453
20. Nair RR, Tolentino JH, Argilagos RF, Zhang L, Pinilla-Ibarz J, Hazlehurst LA (2012)
Potentiation of Nilotinib-mediated cell death in the context of the bone marrow microenvi-
ronment requires a promiscuous JAK inhibitor in CML. Leuk Res 36:756–763
21. Hazlehurst LA, Valkov N, Wisner L, Storey JA, Boulware D, Sullivan DM, Dalton WS
(2001) Reduction in drug-induced DNA double-strand breaks associated with beta1 integ-
rin-mediated adhesion correlates with drug resistance in U937 cells. Blood 98:1897–1903
22. Hoyt DG, Rusnak JM, Mannix RJ, Modzelewski RA, Johnson CS, Lazo JS (1996) Integrin
activation suppresses etoposide-induced DNA strand breakage in cultured murine tumor-
derived endothelial cells. Cancer Res 56:4146–4149
23. Ihle JN (2001) The stat family in cytokine signaling. Curr Opin Cell Biol 13:211–217
24. Herrington J, Smit LS, Schwartz J, Carter-Su C (2000) The role of STAT proteins in growth
hormone signaling. Oncogene 19:2585–2597
25. Reddy EP, Korapati A, Chaturvedi P, Rane S (2000) IL-3 signaling and the role of Src
kinases, JAKs and STATs: a covert liaison unveiled. Oncogene 19:2532–2547
26. Marrero MB, Schieffer B, Paxton WG, Heerdt L, Berk BC, Delafontaine P, Bernstein KE
(1995) Direct stimulation of Jak/STAT pathway by the angiotensin II AT1 receptor. Nature
375:247–250
27. Shain KH, Yarde DN, Meads MB, Huang M, Jove R, Hazlehurst LA, Dalton WS (2009)
Beta1 integrin adhesion enhances IL-6-mediated STAT3 signaling in myeloma cells: impli-
cations for microenvironment influence on tumor survival and proliferation. Cancer Res
69:1009–1015
28. Epling-Burnette PK, Liu JH, Catlett-Falcone R, Turkson J, Oshiro M, Kothapalli R, Li Y,
Wang JM, Yang-Yen HF, Karras J et al (2001) Inhibition of STAT3 signaling leads to apop-
tosis of leukemic large granular lymphocytes and decreased Mcl-1 expression. J Clin Invest
107:351–362
29. Gritsko T, Williams A, Turkson J, Kaneko S, Bowman T, Huang M, Nam S, Eweis I, Diaz
N, Sullivan D et al (2006) Persistent activation of stat3 signaling induces survivin gene
expression and confers resistance to apoptosis in human breast cancer cells. Clin Cancer
Res 12:11–19
30. Cuevas P, Diaz-Gonzalez D, Sanchez I, Lozano RM, Gimenez-Gallego G, Dujovny M
(2006) Dobesilate inhibits the activation of signal transducer and activator of transcription 3,
and the expression of cyclin D1 and bcl-XL in glioma cells. Neurol Res 28:127–130
31. Bhattacharya S, Ray RM, Johnson LR (2005) STAT3-mediated transcription of Bcl-2,
Mcl-1 and c-IAP2 prevents apoptosis in polyamine-depleted cells. Biochem J 392:335–344
74 R. R. Nair et al.
32. Cox D, Brennan M, Moran N (2010) Integrins as therapeutic targets: lessons and opportuni-
ties. Nat Rev Drug Discov 9:804–820
33. Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L (1998) Interleukin-6-type
cytokine signalling through the gp130/Jak/STAT pathway. Biochem J 334(Pt 2):297–314
34. Quesnelle KM, Boehm AL, Grandis JR (2007) STAT-mediated EGFR signaling in cancer. J
Cell Biochem 102:311–319
35. Heinrich PC, Behrmann I, Haan S, Hermanns HM, Muller-Newen G, Schaper F (2003)
Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J
374:1–20
36. Shuai K, Stark GR, Kerr IM, Darnell JE Jr (1993) A single phosphotyrosine residue of
Stat91 required for gene activation by interferon-gamma. Science 261:1744–1746
37. Shuai K, Horvath CM, Huang LH, Qureshi SA, Cowburn D, Darnell JE Jr (1994)
Interferon activation of the transcription factor Stat91 involves dimerization through SH2-
phosphotyrosyl peptide interactions. Cell 76:821–828
38. Ara T, Song L, Shimada H, Keshelava N, Russell HV, Metelitsa LS, Groshen SG, Seeger
RC, DeClerck YA (2009) Interleukin-6 in the bone marrow microenvironment promotes the
growth and survival of neuroblastoma cells. Cancer Res 69:329–337
39. Catlett-Falcone R, Landowski TH, Oshiro MM, Turkson J, Levitzki A, Savino R, Ciliberto
G, Moscinski L, Fernandez-Luna JL, Nunez G et al (1999) Constitutive activation of
Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity
10:105–115
40. Monaghan KA, Khong T, Burns CJ, Spencer A (2011) The novel JAK inhibitor CYT387
suppresses multiple signalling pathways, prevents proliferation and induces apoptosis in
phenotypically diverse myeloma cells. Leukemia 25:1891–1899
41. Hunsucker SA, Magarotto V, Kuhn DJ, Kornblau SM, Wang M, Weber DM, Thomas SK,
Shah JJ, Voorhees PM, Xie H et al (2011) Blockade of interleukin-6 signalling with sil-
tuximab enhances melphalan cytotoxicity in preclinical models of multiple myeloma. Br J
Haematol 152:579–592
42. Burger R, Le Gouill S, Tai YT, Shringarpure R, Tassone P, Neri P, Podar K, Catley L,
Hideshima T, Chauhan D et al (2009) Janus kinase inhibitor INCB20 has antiprolifera-
tive and apoptotic effects on human myeloma cells in vitro and in vivo. Mol Cancer Ther
8:26–35
43. Li J, Favata M, Kelley JA, Caulder E, Thomas B, Wen X, Sparks RB, Arvanitis A, Rogers
JD, Combs AP et al (2010) INCB16562, a JAK1/2 selective inhibitor, is efficacious against
multiple myeloma cells and reverses the protective effects of cytokine and stromal cell sup-
port. Neoplasia 12:28–38
44. Honemann D, Chatterjee M, Savino R, Bommert K, Burger R, Gramatzki M, Dorken B,
Bargou RC (2001) The IL-6 receptor antagonist SANT-7 overcomes bone marrow stromal
cell-mediated drug resistance of multiple myeloma cells. Int J Cancer 93:674–680
45. Hu M, Yao J, Cai L, Bachman KE, van den Brule F, Velculescu V, Polyak K (2005) Distinct
epigenetic changes in the stromal cells of breast cancers. Nat Genet 37:899–905
46. Wang W, Li Q, Yamada T, Matsumoto K, Matsumoto I, Oda M, Watanabe G, Kayano Y, Nishioka
Y, Sone S et al (2009) Crosstalk to stromal fibroblasts induces resistance of lung cancer to epi-
dermal growth factor receptor tyrosine kinase inhibitors. Clin Cancer Res 15:6630–6638
47. Martinez-Outschoorn UE, Goldberg A, Lin Z, Ko YH, Flomenberg N, Wang C, Pavlides
S, Pestell RG, Howell A, Sotgia F et al (2011) Anti-estrogen resistance in breast cancer is
induced by the tumor microenvironment and can be overcome by inhibiting mitochondrial
function in epithelial cancer cells. Cancer Biol Ther 12:924–938
48. Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, Banham AH (2006)
Quantification of regulatory T cells enables the identification of high-risk breast cancer
patients and those at risk of late relapse. J Clin Oncol 24:5373–5380
49. Sasada T, Kimura M, Yoshida Y, Kanai M, Takabayashi A (2003) CD4+CD25+ regulatory
T cells in patients with gastrointestinal malignancies: possible involvement of regulatory T
cells in disease progression. Cancer 98:1089–1099
3 Emerging Opportunities for Targeting 75
50. Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the
immune system. Nat Rev Immunol 9:162–174
51. Mazzoni A, Bronte V, Visintin A, Spitzer JH, Apolloni E, Serafini P, Zanovello P, Segal DM
(2002) Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J
Immunol 168:689–695
52. Lu T, Ramakrishnan R, Altiok S, Youn JI, Cheng P, Celis E, Pisarev V, Sherman S, Sporn
MB, Gabrilovich D (2011) Tumor-infiltrating myeloid cells induce tumor cell resistance to
cytotoxic T cells in mice. J Clin Invest 121:4015–4029
53. Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, Fuh G, Gerber HP, Ferrara
N (2007) Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ mye-
loid cells. Nat Biotechnol 25:911–920
54. Schmid MC, Avraamides CJ, Foubert P, Shaked Y, Kang SW, Kerbel RS, Varner JA (2011)
Combined Blockade of Integrin-alpha4beta1 Plus Cytokines SDF-1alpha or IL-1beta
Potently Inhibits Tumor Inflammation and Growth. Cancer Res 71:6965–6975
55. Du R, Lu KV, Petritsch C, Liu P, Ganss R, Passegue E, Song H, Vandenberg S, Johnson RS,
Werb Z et al (2008) HIF1alpha induces the recruitment of bone marrow-derived vascular
modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 13:206–220
56. Alam N, Goel HL, Zarif MJ, Butterfield JE, Perkins HM, Sansoucy BG, Sawyer
TK, Languino LR (2007) The integrin-growth factor receptor duet. J Cell Physiol
213:649–653
57. Bergers G, Hanahan D (2008) Modes of resistance to anti-angiogenic therapy. Nat Rev
Cancer 8:592–603
58. Takahashi M, Tsuda T, Ikeda Y, Honke K, Taniguchi N (2004) Role of N-glycans in growth
factor signaling. Glycoconj J 20:207–212
59. Waetzig GH, Chalaris A, Rosenstiel P, Suthaus J, Holland C, Karl N, Valles Uriarte L, Till
A, Scheller J, Grotzinger J et al (2010) N-linked glycosylation is essential for the stability
but not the signaling function of the interleukin-6 signal transducer glycoprotein 130. J Biol
Chem 285:1781–1789
60. Barondes SH, Cooper DN, Gitt MA, Leffler H (1994) Galectins. Structure and function of a
large family of animal lectins. J Biol Chem 269:20807–20810
61. Brewer CF, Miceli MC, Baum LG (2002) Clusters, bundles, arrays and lattices: novel
mechanisms for lectin-saccharide-mediated cellular interactions. Curr Opin Struct Biol
12:616–623
62. Sacchettini JC, Baum LG, Brewer CF (2001) Multivalent protein-carbohydrate interac-
tions. A new paradigm for supermolecular assembly and signal transduction. Biochemistry
40:3009–3015
63. Collins BE, Paulson JC (2004) Cell surface biology mediated by low affinity multivalent
protein-glycan interactions. Curr Opin Chem Biol 8:617–625
64. Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE (2006) Targeting of CD44 eradicates
human acute myeloid leukemic stem cells. Nat Med 12:1167–1174
65. Varki A, Kannagi R, Toole BP (2009) Glycosylation changes in cancer. In: Varki A,
Cummings RD, Esko JD, Freeze HH, Stanley P, Bertozzi CR, Hart GW, Etzler ME (eds.)
Essentials of glycobiology. Cold Spring Harbor, NY
66. Hirabayashi J, Hashidate T, Arata Y, Nishi N, Nakamura T, Hirashima M, Urashima T, Oka
T, Futai M, Muller WE et al (2002) Oligosaccharide specificity of galectins: a search by
frontal affinity chromatography. Biochim Biophys Acta 1572:232–254
67. Granovsky M, Fata J, Pawling J, Muller WJ, Khokha R, Dennis JW (2000) Suppression of
tumor growth and metastasis in Mgat5-deficient mice. Nat Med 6:306–312
68. Partridge EA, Le Roy C, Di Guglielmo GM, Pawling J, Cheung P, Granovsky M, Nabi IR,
Wrana JL, Dennis JW (2004) Regulation of cytokine receptors by Golgi N-glycan process-
ing and endocytosis. Science 306:120–124
69. Lajoie P, Partridge EA, Guay G, Goetz JG, Pawling J, Lagana A, Joshi B, Dennis JW, Nabi
IR (2007) Plasma membrane domain organization regulates EGFR signaling in tumor cells.
J Cell Biol 179:341–356
76 R. R. Nair et al.
70. Daniels MA, Hogquist KA, Jameson SC (2002) Sweet ‘n’ sour: the impact of differential
glycosylation on T cell responses. Nat Immunol 3:903–910
71. Barondes SH, Castronovo V, Cooper DN, Cummings RD, Drickamer K, Feizi T, Gitt MA,
Hirabayashi J, Hughes C, Kasai K et al (1994) Galectins: a family of animal beta-galacto-
side-binding lectins. Cell 76:597–598
72. Liao DI, Kapadia G, Ahmed H, Vasta GR, Herzberg O (1994) Structure of S-lectin, a devel-
opmentally regulated vertebrate beta-galactoside-binding protein. Proc Nat Acad Sci USA
91:1428–1432
73. Hirabayashi J, Kasai K (1993) The family of metazoan metal-independent beta-galactoside-
binding lectins: structure, function and molecular evolution. Glycobiology 3:297–304
74. Houzelstein D, Goncalves IR, Fadden AJ, Sidhu SS, Cooper DN, Drickamer K, Leffler
H, Poirier F (2004) Phylogenetic analysis of the vertebrate galectin family. Mol Biol Evol
21:1177–1187
75. Di Lella S, Sundblad V, Cerliani JP, Guardia CM, Estrin DA, Vasta GR, Rabinovich GA
(2011) When galectins recognize glycans: from biochemistry to physiology and back again.
Biochemistry 50:7842–7857
76. de Waard A, Hickman S, Kornfeld S (1976) Isolation and properties of beta-galactoside
binding lectins of calf heart and lung. J Biol Chem 251:7581–7587
77. Paz A, Haklai R, Elad-Sfadia G, Ballan E, Kloog Y (2001) Galectin-1 binds oncogenic H-Ras
to mediate Ras membrane anchorage and cell transformation. Oncogene 20:7486–7493
78. Ellerhorst J, Nguyen T, Cooper DN, Lotan D, Lotan R (1999) Differential expression of
endogenous galectin-1 and galectin-3 in human prostate cancer cell lines and effects of
overexpressing galectin-1 on cell phenotype. Int J Oncol 14:217–224
79. van den Brule F, Califice S, Garnier F, Fernandez PL, Berchuck A, Castronovo V (2003)
Galectin-1 accumulation in the ovary carcinoma peritumoral stroma is induced by ovary
carcinoma cells and affects both cancer cell proliferation and adhesion to laminin-1 and
fibronectin. Lab Invest J Tech Methods Pathol 83:377–386
80. Clausse N, van den Brule F, Waltregny D, Garnier F, Castronovo V (1999) Galectin-1
expression in prostate tumor-associated capillary endothelial cells is increased by prostate
carcinoma cells and modulates heterotypic cell-cell adhesion. Angiogenesis 3:317–325
81. Horiguchi N, Arimoto K, Mizutani A, Endo-Ichikawa Y, Nakada H, Taketani S (2003)
Galectin-1 induces cell adhesion to the extracellular matrix and apoptosis of non-adherent
human colon cancer Colo201 cells. J Biochem 134:869–874
82. Lippert E, Gunckel M, Brenmoehl J, Bataille F, Falk W, Scholmerich J, Obermeier F, Rogler
G (2008) Regulation of galectin-3 function in mucosal fibroblasts: potential role in mucosal
inflammation. Clin Exp Immunol 152:285–297
83. Park SH, Min HS, Kim B, Myung J, Paek SH (2008) Galectin-3: a useful biomarker for
differential diagnosis of brain tumors. Neuropathol: Official J Jpn Soc Neuropathol
28:497–506
84. Jiang HR, Al Rasebi Z, Mensah-Brown E, Shahin A, Xu D, Goodyear CS, Fukada SY, Liu
FT, Liew FY, Lukic ML (2009) Galectin-3 deficiency reduces the severity of experimental
autoimmune encephalomyelitis. J Immunol 182:1167–1173
85. Jeon SB, Yoon HJ, Chang CY, Koh HS, Jeon SH, Park EJ (2010) Galectin-3 exerts cytokine-
like regulatory actions through the JAK-STAT pathway. J Immunol 185:7037–7046
86. Wan SY, Zhang TF, Ding Y (2011) Galectin-3 enhances proliferation and angiogenesis of
endothelial cells differentiated from bone marrow mesenchymal stem cells. Transpl Proc
43:3933–3938
87. Chauhan D, Li G, Podar K, Hideshima T, Neri P, He D, Mitsiades N, Richardson P, Chang
Y, Schindler J et al (2005) A novel carbohydrate-based therapeutic GCS-100 overcomes
bortezomib resistance and enhances dexamethasone-induced apoptosis in multiple myeloma
cells. Cancer Res 65:8350–8358
88. Streetly MJ, Maharaj L, Joel S, Schey SA, Gribben JG, Cotter FE (2010) GCS-100, a novel
galectin-3 antagonist, modulates MCL-1, NOXA, and cell cycle to induce myeloma cell
death. Blood 115:3939–3948
3 Emerging Opportunities for Targeting 77
89. Cheng YL, Huang WC, Chen CL, Tsai CC, Wang CY, Chiu WH, Chen YL, Lin YS, Chang
CF, Lin CF (2011) Increased galectin-3 facilitates leukemia cell survival from apoptotic
stimuli. Biochem Biophys Res Commun 412:334–340
90. Yamamoto-Sugitani M, Kuroda J, Ashihara E, Nagoshi H, Kobayashi T, Matsumoto
Y, Sasaki N, Shimura Y, Kiyota M, Nakayama R et al (2011) Galectin-3 (Gal-3) induced
by leukemia microenvironment promotes drug resistance and bone marrow lodgment in
chronic myelogenous leukemia. Proc Nat Acad Sci USA 108:17468–17473
91. Kageshita T, Kashio Y, Yamauchi A, Seki M, Abedin MJ, Nishi N, Shoji H, Nakamura T,
Ono T, Hirashima M (2002) Possible role of galectin-9 in cell aggregation and apoptosis
of human melanoma cell lines and its clinical significance. Int J Cancer J Int du Cancer
99:809–816
92. Lu LH, Nakagawa R, Kashio Y, Ito A, Shoji H, Nishi N, Hirashima M, Yamauchi A,
Nakamura T (2007) Characterization of galectin-9-induced death of Jurkat T cells. J
Biochem 141:157–172
93. Kuroda J, Yamamoto M, Nagoshi H, Kobayashi T, Sasaki N, Shimura Y, Horiike S, Kimura
S, Yamauchi A, Hirashima M et al (2010) Targeting activating transcription factor 3 by
Galectin-9 induces apoptosis and overcomes various types of treatment resistance in chronic
myelogenous leukemia. Mol Cancer Res: MCR 8:994–1001
94. Kobayashi T, Kuroda J, Ashihara E, Oomizu S, Terui Y, Taniyama A, Adachi S, Takagi T,
Yamamoto M, Sasaki N et al. (2010) Galectin-9 exhibits anti-myeloma activity through
JNK and p38 MAP kinase pathways. Leuk: Official J Leuk Soc Am Leuk Res Fund UK
24:843–850
95. Cipolleschi MG, Dello Sbarba P, Olivotto M (1993) The role of hypoxia in the maintenance
of hematopoietic stem cells. Blood 82:2031–2037
96. Wohlkoenig C, Leithner K, Deutsch A, Hrzenjak A, Olschewski A, Olschewski H (2011)
Hypoxia-induced cisplatin resistance is reversible and growth rate independent in lung can-
cer cells. Cancer Lett 308:134–143
97. Tanturli M, Giuntoli S, Barbetti V, Rovida E, Dello Sbarba P (2011) Hypoxia selects borte-
zomib-resistant stem cells of chronic myeloid leukemia. PLoS ONE 6:e17008
98. Giuntoli S, Rovida E, Barbetti V, Cipolleschi MG, Olivotto M, Dello Sbarba P (2006)
Hypoxia suppresses BCR/Abl and selects imatinib-insensitive progenitors within clonal
CML populations. Leukemia 20:1291–1293
99. Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe SW, Giaccia AJ (1996)
Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours.
Nature 379:88–91
100. Bristow RG, Hill RP (2008) Hypoxia and metabolism. Hypoxia, DNA repair and genetic
instability. Nat Rev Cancer 8:180–192
101. Tang N, Wang L, Esko J, Giordano FJ, Huang Y, Gerber HP, Ferrara N, Johnson RS (2004)
Loss of HIF-1alpha in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop nec-
essary for tumorigenesis. Cancer Cell 6:485–495
102. Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM (2003)
Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene.
Cancer Cell 3:347–361
103. Lukashev D, Klebanov B, Kojima H, Grinberg A, Ohta A, Berenfeld L, Wenger RH,
Sitkovsky M (2006) Cutting edge: hypoxia-inducible factor 1alpha and its activation-induc-
ible short isoform I.1 negatively regulate functions of CD4+ and CD8+ T lymphocytes. J
Immunol 177:4962–4965
104. Masson N, Willam C, Maxwell PH, Pugh CW, Ratcliffe PJ (2001) Independent function
of two destruction domains in hypoxia-inducible factor-alpha chains activated by prolyl
hydroxylation. EMBO J 20:5197–5206
105. Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, Kriegsheim A,
Hebestreit HF, Mukherji M, Schofield CJ et al (2001) Targeting of HIF-alpha to the von
Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science
292:468–472
78 R. R. Nair et al.
106. Mole DR, Blancher C, Copley RR, Pollard PJ, Gleadle JM, Ragoussis J, Ratcliffe PJ
(2009) Genome-wide association of hypoxia-inducible factor (HIF)-1alpha and HIF-2alpha
DNA binding with expression profiling of hypoxia-inducible transcripts. J Biol Chem
284:16767–16775
107. Weiss GJ, Infante JR, Chiorean EG, Borad MJ, Bendell JC, Molina JR, Tibes R,
Ramanathan RK, Lewandowski K, Jones SF et al (2011) Phase 1 study of the safety, tol-
erability, and pharmacokinetics of TH-302, a hypoxia-activated prodrug, in patients with
advanced solid malignancies. Clin Cancer Res 17:2997–3004
108. Romero-Ramirez L, Cao H, Nelson D, Hammond E, Lee AH, Yoshida H, Mori K, Glimcher
LH, Denko NC, Giaccia AJ et al (2004) XBP1 is essential for survival under hypoxic condi-
tions and is required for tumor growth. Cancer Res 64:5943–5947
109. Papandreou I, Denko NC, Olson M, Van Melckebeke H, Lust S, Tam A, Solow-Cordero
DE, Bouley DM, Offner F, Niwa M et al (2011) Identification of an Ire1alpha endonu-
clease specific inhibitor with cytotoxic activity against human multiple myeloma. Blood
117:1311–1314
110. Volkmann K, Lucas JL, Vuga D, Wang X, Brumm D, Stiles C, Kriebel D, Der-Sarkissian A,
Krishnan K, Schweitzer C et al (2011) Potent and selective inhibitors of the inositol-requir-
ing enzyme 1 endoribonuclease. J Biol Chem 286:12743–12755
111. Fels DR, Ye J, Segan AT, Kridel SJ, Spiotto M, Olson M, Koong AC, Koumenis C (2008)
Preferential cytotoxicity of bortezomib toward hypoxic tumor cells via overactivation of
endoplasmic reticulum stress pathways. Cancer Res 68:9323–9330
112. Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS (1999) Cell adhesion medi-
ated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human mye-
loma cell lines. Blood 93:1658–1667
113. Emmons MF, Gebhard AW, Nair RR, Baz R, McLaughlin M, Cress AE, Hazlehurst LA
(2011) Acquisition of resistance towards HYD1 correlates with a reduction in cleaved
alpha 4 integrin expression and a compromised CAM-DR phenotype. Mol Cancer Ther
10(12):2257–2266
114. Lu X, Mu E, Wei Y, Riethdorf S, Yang Q, Yuan M, Yan J, Hua Y, Tiede BJ, Haffty BG et al
(2011) VCAM-1 promotes osteolytic expansion of indolent bone micrometastasis of breast
cancer by engaging alpha4beta1-positive osteoclast progenitors. Cancer Cell 20:701–714
115. Abe M, Hiura K, Ozaki S, Kido S, Matsumoto T (2009) Vicious cycle between myeloma
cell binding to bone marrow stromal cells via VLA-4-VCAM-1 adhesion and macrophage
inflammatory protein-1alpha and MIP-1beta production. J Bone Miner Metab 27:16–23
116. Chen Q, Lin TH, Der CJ, Juliano RL (1996) Integrin-mediated activation of MEK
and mitogen-activated protein kinase is independent of Ras (corrected). J Biol Chem
271:18122–18127
117. Lee JW, Juliano RL (2002) The alpha5beta1 integrin selectively enhances epidermal growth
factor signaling to the phosphatidylinositol-3-kinase/Akt pathway in intestinal epithelial
cells. Biochim Biophys Acta 1542:23
118. Bill HM, Knudsen B, Moores SL, Muthuswamy SK, Rao VR, Brugge JS, Miranti CK
(2004) Epidermal growth factor receptor-dependent regulation of integrin-mediated signal-
ing and cell cycle entry in epithelial cells. Mol Cell Biol 24:8586–8599
119. Xiao T, Takagi J, Coller BS, Wang JH, Springer TA (2004) Structural basis for allostery in
integrins and binding to fibrinogen-mimetic therapeutics. Nature 432:59–67
120. Xiong JP, Stehle T, Diefenbach B, Zhang R, Dunker R, Scott DL, Joachimiak A, Goodman
SL, Arnaout MA (2001) Crystal structure of the extracellular segment of integrin alpha
Vbeta3. Science 294:339–345
121. Rosano C, Rocco M (2010) Solution properties of full-length integrin alpha(IIb)beta3
refined models suggest environment-dependent induction of alternative bent/extended rest-
ing states. FEBS J 277:3190–3202
122. Lu C, Takagi J, Springer TA (2001) Association of the membrane proximal regions of the
alpha and beta subunit cytoplasmic domains constrains an integrin in the inactive state. J
Biol Chem 276:14642–14648
3 Emerging Opportunities for Targeting 79
123. Luo BH, Carman CV, Springer TA (2007) Structural basis of integrin regulation and
signaling. Annu Rev Immunol 25:619–647
124. Takagi J, Erickson HP, Springer TA (2001) C-terminal opening mimics ‘inside-out’ activa-
tion of integrin alpha5beta1. Nat Struct Biol 8:412–416
125. Calderwood DA, Zent R, Grant R, Rees DJ, Hynes RO, Ginsberg MH (1999) The Talin
head domain binds to integrin beta subunit cytoplasmic tails and regulates integrin activa-
tion. J Biol Chem 274:28071–28074
126. Anthis NJ, Haling JR, Oxley CL, Memo M, Wegener KL, Lim CJ, Ginsberg MH, Campbell
ID (2009) Beta integrin tyrosine phosphorylation is a conserved mechanism for regulating
talin-induced integrin activation. J Biol Chem 284:36700–36710
127. Critchley DR, Gingras AR (2008) Talin at a glance. J Cell Sci 121:1345–1347
128. Arai A, Nosaka Y, Kanda E, Yamamoto K, Miyasaka N, Miura O (2001) Rap1 is activated
by erythropoietin or interleukin-3 and is involved in regulation of beta1 integrin-mediated
hematopoietic cell adhesion. J Biol Chem 276:10453–10462
129. Hazlehurst LA, Enkemann SA, Beam CA, Argilagos RF, Painter J, Shain KH, Saporta S,
Boulware D, Moscinski L, Alsina M et al (2003) Genotypic and phenotypic comparisons of
de novo and acquired melphalan resistance in an isogenic multiple myeloma cell line model.
Cancer Res 63:7900–7906
130. Rice GP, Hartung HP, Calabresi PA (2005) Anti-alpha4 integrin therapy for multiple
sclerosis: mechanisms and rationale. Neurology 64:1336–1342
131. Targan SR, Feagan BG, Fedorak RN, Lashner BA, Panaccione R, Present DH, Spehlmann
ME, Rutgeerts PJ, Tulassay Z, Volfova M et al (2007) Natalizumab for the treatment of
active Crohn’s disease: results of the ENCORE Trial. Gastroenterology 132:1672–1683
132. Matsunaga T, Takemoto N, Sato T, Takimoto R, Tanaka I, Fujimi A, Akiyama T, Kuroda H,
Kawano Y, Kobune M et al (2003) Interaction between leukemic-cell VLA-4 and stromal
fibronectin is a decisive factor for minimal residual disease of acute myelogenous leukemia.
Nat Med 9:1158–1165
133. Olson DL, Burkly LC, Leone DR, Dolinski BM, Lobb RR (2005) Anti-alpha4 integrin mon-
oclonal antibody inhibits multiple myeloma growth in a murine model. Mol Cancer Ther
4:91–99
134. Pierschbacher MD, Ruoslahti E (1984) Cell attachment activity of fibronectin can be dupli-
cated by small synthetic fragments of the molecule. Nature 309:30–33
135. Gould RJ, Polokoff MA, Friedman PA, Huang TF, Holt JC, Cook JJ, Niewiarowski S (1990)
Disintegrins: a family of integrin inhibitory proteins from viper venoms. Proc Soc Exp Biol
Med 195:168–171
136. Lu X, Lu D, Scully MF, Kakkar VV (2006) Integrins in drug targeting-RGD templates in
toxins. Curr Pharm Des 12:2749–2769
137. DeRoock IB, Pennington ME, Sroka TC, Lam KS, Bowden GT, Bair EL, Cress AE (2001)
Synthetic peptides inhibit adhesion of human tumor cells to extracellular matrix proteins.
Cancer Res 61:3308–3313
138. Pennington ME, Lam KS, Cress AE (1996) The use of a combinatorial library method to
isolate human tumor cell adhesion peptides. Mol Divers 2:19–28
139. Sroka TC, Pennington ME, Cress AE (2006) Synthetic D-amino acid peptide inhibits tumor
cell motility on laminin-5. Carcinogenesis 27(9):1748–1757
140. Nair RR, Emmons MF, Cress AE, Argilagos RF, Lam K, Kerr WT, Wang HG, Dalton
WS, Hazlehurst LA (2009) HYD1-induced increase in reactive oxygen species leads to
autophagy and necrotic cell death in multiple myeloma cells. Mol Cancer Ther 8:2441–2451
141. Mas-Moruno C, Rechenmacher F, Kessler H (2010) Cilengitide: the first anti-angiogenic
small molecule drug candidate design, synthesis and clinical evaluation. Anticancer Agents
Med Chem 10:753–768
142. Doedens L, Opperer F, Cai M, Beck JG, Dedek M, Palmer E, Hruby VJ, Kessler H (2010)
Multiple N-methylation of MT-II backbone amide bonds leads to melanocortin receptor
subtype hMC1R selectivity: pharmacological and conformational studies. J Am Chem Soc
132:8115–8128
80 R. R. Nair et al.
143. Reardon DA, Fink KL, Mikkelsen T, Cloughesy TF, O’Neill A, Plotkin S, Glantz M, Ravin
P, Raizer JJ, Rich KM et al (2008) Randomized phase II study of cilengitide, an integrin-tar-
geting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin
Oncol 26:5610–5617
144. Yamada S, Bu XY, Khankaldyyan V, Gonzales-Gomez I, McComb JG, Laug WE (2006)
Effect of the angiogenesis inhibitor Cilengitide (EMD 121974) on glioblastoma growth in
nude mice. Neurosurgery 59:1304–1312 (discussion 1312)
145. Basoni C, Reuzeau E, Croft D, Genot E, Kramer IM (2006) CD44 and TGFbeta1 syner-
gise to induce expression of a functional NADPH oxidase in promyelocytic cells. Biochem
Biophys Res Commun 343:609–616
146. Khaldoyanidi S, Karakhanova S, Sleeman J, Herrlich P, Ponta H (2002) CD44 variant-
specific antibodies trigger hemopoiesis by selective release of cytokines from bone marrow
macrophages. Blood 99:3955–3961
147. Mielgo A, Brondani V, Landmann L, Glaser-Ruhm A, Erb P, Stupack D, Gunthert U
(2007) The CD44 standard/ezrin complex regulates Fas-mediated apoptosis in Jurkat cells.
Apoptosis 12:2051–2061
148. Okada T, Hawley RG, Kodaka M, Okuno H (1999) Significance of VLA-4-VCAM-1 inter-
action and CD44 for transendothelial invasion in a bone marrow metastatic myeloma model.
Clin Exp Metastasis 17:623–629
149. van der Windt GJ, Schouten M, Zeerleder S, Florquin S, van der Poll T CD44 is Protective
during hyperoxia-induced lung injury. Am J Respir Cell Mol Biol 44(3):377–383
150. Pure E, Assoian RK (2009) Rheostatic signaling by CD44 and hyaluronan. Cell Signal
21:651–655
151. Gunthert U, Hofmann M, Rudy W, Reber S, Zoller M, Haussmann I, Matzku S, Wenzel A,
Ponta H, Herrlich P (1991) A new variant of glycoprotein CD44 confers metastatic potential
to rat carcinoma cells. Cell 65:13–24
152. Weber GF, Bronson RT, Ilagan J, Cantor H, Schmits R, Mak TW (2002) Absence of the
CD44 gene prevents sarcoma metastasis. Cancer Res 62:2281–2286
153. Asosingh K, Gunthert U, Bakkus MH, De Raeve H, Goes E, Van Riet I, Van Camp B,
Vanderkerken K (2000) In vivo induction of insulin-like growth factor-I receptor and
CD44v6 confers homing and adhesion to murine multiple myeloma cells. Cancer Res
60:3096–3104
154. Caers J, Gunthert U, De Raeve H, Van Valckenborgh E, Menu E, Van Riet I, Van Camp B,
Vanderkerken K (2006) The involvement of osteopontin and its receptors in multiple mye-
loma cell survival, migration and invasion in the murine 5T33MM model. Br J Haematol
132:469–477
155. Masellis-Smith A, Belch AR, Mant MJ, Pilarski LM (1997) Adhesion of multiple myeloma
peripheral blood B cells to bone marrow fibroblasts: a requirement for CD44 and alpha-
4beta7. Cancer Res 57:930–936
156. Cichy J, Bals R, Potempa J, Mani A, Pure E (2002) Proteinase-mediated release of epithelial
cell-associated CD44. Extracellular CD44 complexes with components of cellular matrices.
J Biol Chem 277:44440–44447
157. Cichy J, Kulig P, Pure E (2005) Regulation of the release and function of tumor cell-derived
soluble CD44. Biochim Biophys Acta 1745:59–64
158. Astier A, Manie SN, Avraham H, Hirai H, Law SF, Zhang Y, Golemis EA, Fu Y, Druker BJ,
Haghayeghi N et al (1997) The related adhesion focal tyrosine kinase differentially phos-
phorylates p130Cas and the Cas-like protein, p105HEF1. J Biol Chem 272:19719–19724
159. Eisterer W, Bechter O, Soderberg O, Nilsson K, Terol M, Greil R, Thaler J, Herold M, Finke
L, Gunthert U et al (2004) Elevated levels of soluble CD44 are associated with advanced
disease and in vitro proliferation of neoplastic lymphocytes in B-cell chronic lymphocytic
leukaemia. Leuk Res 28:1043–1051
160. Molica S, Vitelli G, Levato D, Giannarelli D, Gandolfo GM (2001) Elevated serum levels
of soluble CD44 can identify a subgroup of patients with early B-cell chronic lymphocytic
leukemia who are at high risk of disease progression. Cancer 92:713–719
3 Emerging Opportunities for Targeting 81
161. Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B (1990) CD44 is the principal
cell surface receptor for hyaluronate. Cell 61:1303–1313
162. Goodison S, Urquidi V, Tarin D (1999) CD44 cell adhesion molecules. Mol Pathol
52:189–196
163. Meyer MJ, Fleming JM, Lin AF, Hussnain SA, Ginsburg E, Vonderhaar BK (2010)
CD44posCD49fhiCD133/2hi defines xenograft-initiating cells in estrogen receptor-negative
breast cancer. Cancer Res 70:4624–4633
164. Stauder R, Van Driel M, Schwarzler C, Thaler J, Lokhorst HM, Kreuser ED, Bloem AC,
Gunthert U, Eisterer W (1996) Different CD44 splicing patterns define prognostic sub-
groups in multiple myeloma. Blood 88:3101–3108
165. Van Driel M, Gunthert U, van Kessel AC, Joling P, Stauder R, Lokhorst HM, Bloem AC
(2002) CD44 variant isoforms are involved in plasma cell adhesion to bone marrow stromal
cells. Leukemia 16:135–143
166. Bourguignon LY, Gilad E, Brightman A, Diedrich F, Singleton P (2006) Hyaluronan-CD44
interaction with leukemia-associated RhoGEF and epidermal growth factor receptor promotes
Rho/Ras co-activation, phospholipase C epsilon-Ca2+ signaling, and cytoskeleton modification in
head and neck squamous cell carcinoma cells. J Biol Chem 281:14026–14040
167. Bourguignon LY, Singleton PA, Zhu H, Zhou B (2002) Hyaluronan promotes signaling
interaction between CD44 and the transforming growth factor beta receptor I in metastatic
breast tumor cells. J Biol Chem 277:39703–39712
168. Lee JL, Wang MJ, Sudhir PR, Chen JY (2008) CD44 engagement promotes matrix-derived
survival through the CD44-SRC-integrin axis in lipid rafts. Mol Cell Biol 28:5710–5723
169. Redondo-Munoz J, Ugarte-Berzal E, Garcia-Marco JA, del Cerro MH, Van den Steen PE,
Opdenakker G, Terol MJ, Garcia-Pardo A (2008) Alpha4beta1 integrin and 190-kDa CD44v
constitute a cell surface docking complex for gelatinase B/MMP-9 in chronic leukemic but
not in normal B cells. Blood 112:169–178
170. Nandi A, Estess P, Siegelman M (2004) Bimolecular complex between rolling and firm
adhesion receptors required for cell arrest; CD44 association with VLA-4 in T cell extrava-
sation. Immunity 20:455–465
171. Krause DS, Lazarides K, von Andrian UH, Van Etten RA (2006) Requirement for CD44 in
homing and engraftment of BCR-ABL-expressing leukemic stem cells. Nat Med 12:1175–1180
172. Thorne RF, Legg JW, Isacke CM (2004) The role of the CD44 transmembrane and cytoplas-
mic domains in co-ordinating adhesive and signalling events. J Cell Sci 117:373–380
173. Lesley J, Hascall VC, Tammi M, Hyman R (2000) Hyaluronan binding by cell surface
CD44. J Biol Chem 275:26967–26975
174. Banerji S, Wright AJ, Noble M, Mahoney DJ, Campbell ID, Day AJ, Jackson DG (2007)
Structures of the Cd44-hyaluronan complex provide insight into a fundamental carbohy-
drate-protein interaction. Nat Struct Mol Biol 14:234–239
175. Ogino S, Nishida N, Umemoto R, Suzuki M, Takeda M, Terasawa H, Kitayama J,
Matsumoto M, Hayasaka H, Miyasaka M et al (2010) Two-state conformations in
the hyaluronan-binding domain regulate CD44 adhesiveness under flow condition.
Structure 18:649–656
176. Wolny PM, Banerji S, Gounou C, Brisson AR, Day AJ, Jackson DG, Richter RP (2010) Analysis
of CD44-hyaluronan interactions in an artificial membrane system: insights into the distinct bind-
ing properties of high and low molecular weight hyaluronan. J Biol Chem 285:30170–30180
177. Ilangumaran S, Borisch B, Hoessli DC (1999) Signal transduction via CD44: role of plasma
membrane microdomains. Leuk Lymphoma 35:455–469
178. Marhaba R, Freyschmidt-Paul P, Zoller M (2006) In vivo CD44-CD49d complex formation
in autoimmune disease has consequences on T cell activation and apoptosis resistance. Eur J
Immunol 36:3017–3032
179. Verfaillie CM, Benis A, Iida J, McGlave PB, McCarthy JB (1994) Adhesion of committed
human hematopoietic progenitors to synthetic peptides from the C-terminal heparin-binding
domain of fibronectin: cooperation between the integrin alpha 4 beta 1 and the CD44 adhe-
sion receptor. Blood 84:1802–1811
82 R. R. Nair et al.
180. van der Voort R, Taher TE, Wielenga VJ, Spaargaren M, Prevo R, Smit L, David G,
Hartmann G, Gherardi E, Pals ST (1999) Heparan sulfate-modified CD44 promotes hepato-
cyte growth factor/scatter factor-induced signal transduction through the receptor tyrosine
kinase c-Met. J Biol Chem 274:6499–6506
181. Ruffell B, Johnson P (2008) Hyaluronan induces cell death in activated T cells through CD44. J
Immunol 181:7044–7054
182. Tremmel M, Matzke A, Albrecht I, Laib AM, Olaku V, Ballmer-Hofer K, Christofori G, Heroult
M, Augustin HG, Ponta H et al (2009) A CD44v6 peptide reveals a role of CD44 in VEGFR-2
signaling and angiogenesis. Blood 114:5236–5244
183. Piotrowicz RS, Damaj BB, Hachicha M, Incardona F, Howell SB, Finlayson M (2011) A6 peptide
activates CD44 adhesive activity, induces FAK and MEK phosphorylation, and inhibits the migra-
tion and metastasis of CD44-expressing cells. Mol Cancer Ther 10:2072–2082
184. Riechelmann H, Sauter A, Golze W, Hanft G, Schroen C, Hoermann K, Erhardt T, Gronau S
(2008) Phase I trial with the CD44v6-targeting immunoconjugate bivatuzumab mertansine in
head and neck squamous cell carcinoma. Oral Oncol 44:823–829
185. Tilghman RW, Parsons JT (2008) Focal adhesion kinase as a regulator of cell tension in the pro-
gression of cancer. Semin Cancer Biol 18:45–52
186. Grigera PR, Jeffery ED, Martin KH, Shabanowitz J, Hunt DF, Parsons JT (2005) FAK phospho-
rylation sites mapped by mass spectrometry. J Cell Sci 118:4931–4935
187. Harte MT, Hildebrand JD, Burnham MR, Bouton AH, Parsons JT (1996) p130Cas, a substrate
associated with v-Src and v-Crk, localizes to focal adhesions and binds to focal adhesion kinase. J
Biol Chem 271:13649–13655
188. Hildebrand JD, Schaller MD, Parsons JT (1995) Paxillin, a tyrosine phosphorylated focal adhe-
sion-associated protein binds to the carboxyl terminal domain of focal adhesion kinase. Mol Biol
Cell 6:637–647
189. Schaller MD, Parsons JT (1995) pp125FAK-dependent tyrosine phosphorylation of paxillin cre-
ates a high-affinity binding site for Crk. Mol Cell Biol 15:2635–2645
190. Avraham H, Park SY, Schinkmann K, Avraham S (2000) RAFTK/Pyk2-mediated cellular signal-
ling. Cell Signal 12:123–133
191. Ilic D, Furuta Y, Kanazawa S, Takeda N, Sobue K, Nakatsuji N, Nomura S, Fujimoto J, Okada M,
Yamamoto T (1995) Reduced cell motility and enhanced focal adhesion contact formation in cells
from FAK-deficient mice. Nature 377:539–544
192. Guinamard R, Okigaki M, Schlessinger J, Ravetch JV (2000) Absence of marginal zone B cells in
Pyk-2-deficient mice defines their role in the humoral response. Nat Immunol 1:31–36
193. Buckbinder L, Crawford DT, Qi H, Ke HZ, Olson LM, Long KR, Bonnette PC, Baumann AP,
Hambor JE, Grasser WA 3rd et al (2007) Proline-rich tyrosine kinase 2 regulates osteoprogenitor
cells and bone formation, and offers an anabolic treatment approach for osteoporosis. Proc Natl
Acad Sci USA 104:10619–10624
194. Roberts WG, Ung E, Whalen P, Cooper B, Hulford C, Autry C, Richter D, Emerson E, Lin J,
Kath J et al (2008) Antitumor activity and pharmacology of a selective focal adhesion kinase
inhibitor, PF-562,271. Cancer Res 68:1935–1944
195. Bagi CM, Roberts GW, Andresen CJ (2008) Dual focal adhesion kinase/Pyk2 inhibitor has posi-
tive effects on bone tumors: implications for bone metastases. Cancer 112:2313–2321
196. Infante JR, Camidge DR, Mileshkin LR, Chen EX, Hicks RJ, Rischin D, Fingert H, Pierce KJ,
Xu H, Roberts WG et al. (2012) Safety, pharmacokinetic, and pharmacodynamic phase I dose-
escalation trial of PF-00562271, an inhibitor of focal adhesion kinase, in advanced solid tumors. J
Clin Oncol 30(13):1527–1533
197. Wei LH, Kuo ML, Chen CA, Chou CH, Lai KB, Lee CN, Hsieh CY (2003) Interleukin-6 pro-
motes cervical tumor growth by VEGF-dependent angiogenesis via a STAT3 pathway. Oncogene
22:1517–1527
198. Hung SC, Pochampally RR, Chen SC, Hsu SC, Prockop DJ (2007) Angiogenic effects of
human multipotent stromal cell conditioned medium activate the PI3 K-Akt pathway in
hypoxic endothelial cells to inhibit apoptosis, increase survival, and stimulate angiogenesis.
Stem Cells 25:2363–2370
3 Emerging Opportunities for Targeting 83
199. Bid HK, Oswald D, Li C, London CA, Lin J, Houghton PJ (2012) Anti-angiogenic activity
of a small molecule STAT3 inhibitor LLL12. PLoS ONE 7:e35513
200. Haynesworth SE, Baber MA, Caplan AI (1996) Cytokine expression by human marrow-
derived mesenchymal progenitor cells in vitro: effects of dexamethasone and IL-1 alpha. J
Cell Physiol 166:585–592
201. Casanova EA, Shakhova O, Patel SS, Asner IN, Pelczar P, Weber FA, Graf U, Sommer L,
Burki K, Cinelli P (2011) Pramel7 mediates LIF/STAT3-dependent self-renewal in embry-
onic stem cells. Stem Cells 29:474–485
202. Nasef A, Mazurier C, Bouchet S, Francois S, Chapel A, Thierry D, Gorin NC, Fouillard L
(2008) Leukemia inhibitory factor: Role in human mesenchymal stem cells mediated immu-
nosuppression. Cell Immunol 253:16–22
203. Majumdar MK, Thiede MA, Mosca JD, Moorman M, Gerson SL (1998) Phenotypic and
functional comparison of cultures of marrow-derived mesenchymal stem cells (MSCs) and
stromal cells. J Cell Physiol 176:57–66
204. Ahr B, Denizot M, Robert-Hebmann V, Brelot A, Biard-Piechaczyk M (2005) Identification
of the cytoplasmic domains of CXCR4 involved in Jak2 and STAT3 phosphorylation. J Biol
Chem 280:6692–6700
205. Hattermann K, Mentlein R, Held-Feindt J (2012) CXCL12 mediates apoptosis resistance in
rat C6 glioma cells. Oncol Rep 27:1348–1352
206. Hartmann TN, Burger JA, Glodek A, Fujii N, Burger M (2005) CXCR4 chemokine receptor
and integrin signaling co-operate in mediating adhesion and chemoresistance in small cell
lung cancer (SCLC) cells. Oncogene 24:4462–4471
207. Lis R, Touboul C, Mirshahi P, Ali F, Mathew S, Nolan DJ, Maleki M, Abdalla SA, Raynaud
CM, Querleu D et al (2011) Tumor associated mesenchymal stem cells protects ovarian can-
cer cells from hyperthermia through CXCL12. Int J Cancer J Int du Cancer 128:715–725
208. Welte G, Alt E, Devarajan E, Krishnappa S, Jotzu C, Song YH (2011) Interleukin-8 derived
from local tissue-resident stromal cells promotes tumor cell invasion. Mol Carcinog
doi:10.1002/mc.20854
209. Neiva KG, Zhang Z, Miyazawa M, Warner KA, Karl E, Nor JE (2009) Cross talk initiated
by endothelial cells enhances migration and inhibits anoikis of squamous cell carcinoma
cells through STAT3/Akt/ERK signaling. Neoplasia 11:583–593
210. Mellado M, Rodriguez-Frade JM, Aragay A, del Real G, Martin AM, Vila-Coro AJ, Serrano A,
Mayor F Jr, Martinez AC (1998) The chemokine monocyte chemotactic protein 1 triggers Janus
kinase 2 activation and tyrosine phosphorylation of the CCR2B receptor. J Immunol 161:805–813
211. Fujimoto H, Sangai T, Ishii G, Ikehara A, Nagashima T, Miyazaki M, Ochiai A (2009)
Stromal MCP-1 in mammary tumors induces tumor-associated macrophage infiltration and
contributes to tumor progression. Int J Cancer J Int du Cancer 125:1276–1284
212. Tsuyada A, Chow A, Wu J, Somlo G, Chu P, Loera S, Luu T, Li X, Wu X, Ye W et al. (2012)
CCL2 mediates crosstalk between cancer cells and stromal fibroblasts that regulates breast
cancer stem cells. Cancer Res 72(11):2768–79
213. Faderl S, Pal A, Bornmann W, Albitar M, Maxwell D, Van Q, Peng Z, Harris D, Liu Z,
Hazan-Halevy I et al (2009) Kit inhibitor APcK110 induces apoptosis and inhibits prolifera-
tion of acute myeloid leukemia cells. Cancer Res 69:3910–3917
214. Chaix A, Lopez S, Voisset E, Gros L, Dubreuil P, De Sepulveda P (2011) Mechanisms of STAT
protein activation by oncogenic KIT mutants in neoplastic mast cells. J Biol Chem 286:5956–5966
215. Hasegawa T, Suzuki K, Sakamoto C, Ohta K, Nishiki S, Hino M, Tatsumi N, Kitagawa S
(2003) Expression of the inhibitor of apoptosis (IAP) family members in human neutro-
phils: up-regulation of cIAP2 by granulocyte colony-stimulating factor and overexpression
of cIAP2 in chronic neutrophilic leukemia. Blood 101:1164–1171
216. Sakamoto C, Suzuki K, Hato F, Akahori M, Hasegawa T, Hino M, Kitagawa S (2003)
Antiapoptotic effect of granulocyte colony-stimulating factor, granulocyte-macrophage col-
ony-stimulating factor, and cyclic AMP on human neutrophils: protein synthesis-dependent
and protein synthesis-independent mechanisms and the role of the Janus kinase-STAT path-
way. Int J Hematol 77:60–70
84 R. R. Nair et al.
217. Kucerova L, Matuskova M, Hlubinova K, Altanerova V, Altaner C (2010) Tumor cell behav-
iour modulation by mesenchymal stromal cells. Mol Cancer 9:129
218. Gu L, Chiang KY, Zhu N, Findley HW, Zhou M (2007) Contribution of STAT3 to the acti-
vation of survivin by GM-CSF in CD34+ cell lines. Exp Hematol 35:957–966
219. Hofer EL, Labovsky V, La Russa V, Vallone VF, Honegger AE, Belloc CG, Wen HC,
Bordenave RH, Bullorsky EO, Feldman L et al (2010) Mesenchymal stromal cells, colony-
forming unit fibroblasts, from bone marrow of untreated advanced breast and lung can-
cer patients suppress fibroblast colony formation from healthy marrow. Stem Cells Dev
19:359–370
220. Syed ZA, Yin W, Hughes K, Gill JN, Shi R, Clifford JL (2011) HGF/c-met/Stat3 signal-
ing during skin tumor cell invasion: indications for a positive feedback loop. BMC Cancer
11:180
221. Patel ZS, Grugan KD, Rustgi AK, Cucinotta FA, Huff JL (2012) Ionizing radiation enhances
esophageal epithelial cell migration and invasion through a paracrine mechanism involving
stromal-derived hepatocyte growth factor. Radiat Res 177:200–208
222. Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S,
Gianni AM (2002) Human bone marrow stromal cells suppress T-lymphocyte proliferation
induced by cellular or nonspecific mitogenic stimuli. Blood 99:3838–3843
223. Carmo CR, Lyons-Lewis J, Seckl MJ, Costa-Pereira AP (2011) A novel requirement for
Janus kinases as mediators of drug resistance induced by fibroblast growth factor-2 in
human cancer cells. PLoS ONE 6:e19861
224. Udayakumar TS, Nagle RB, Bowden GT (2004) Fibroblast growth factor-1 transcriptionally
induces membrane type-1 matrix metalloproteinase expression in prostate carcinoma cell
line. Prostate 58:66–75
225. Nishimori H, Ehata S, Suzuki HI, Katsuno Y, Miyazono K (2012) Prostate cancer cells and
bone stromal cells mutually interact with each other through bone morphogenetic protein-
mediated signals. J Biol Chem 287(24):20037–20046
226. Di Maggio N, Mehrkens A, Papadimitropoulos A, Schaeren S, Heberer M, Banfi A, Martin I
(2012) FGF-2 Maintains a niche-dependent population of self-renewing highly potent non-
adherent mesenchymal progenitors through FGFR2c. Stem Cells 30(7):1455–1464
227. Dorff TB, Goldman B, Pinski JK, Mack PC, Lara PN, Jr Van Veldhuizen PJ, Jr Quinn DI,
Vogelzang NJ, Thompson IM, Jr Hussain MH (2010) Clinical and correlative results of
SWOG S0354: a phase II trial of CNTO328 (siltuximab), a monoclonal antibody against
interleukin-6, in chemotherapy-pretreated patients with castration-resistant prostate cancer.
Clin Cancer Res: Official J Am Assoc Cancer Res 16:3028–3034
228. Puchalski T, Prabhakar U, Jiao Q, Berns B, Davis HM (2010) Pharmacokinetic and phar-
macodynamic modeling of an anti-interleukin-6 chimeric monoclonal antibody (siltuximab)
in patients with metastatic renal cell carcinoma. Clinical Cancer Res: Official J Am Assoc
Cancer Res 16:1652–1661
229. Hedvat M, Huszar D, Herrmann A, Gozgit JM, Schroeder A, Sheehy A, Buettner R, Proia
D, Kowolik CM, Xin H et al (2009) The JAK2 inhibitor AZD1480 potently blocks Stat3
signaling and oncogenesis in solid tumors. Cancer Cell 16:487–497
230. Ioannidis S, Lamb ML, Wang T, Almeida L, Block MH, Davies AM, Peng B, Su M, Zhang
HJ, Hoffmann E et al (2011) Discovery of 5-chloro-N2-[(1S)-1-(5-fluoropyrimidin-2-yl)
ethyl]-N4-(5-methyl-1H-pyrazol-3-yl)pyrimidine-2,4-diamine (AZD1480) as a novel inhibi-
tor of the Jak/Stat pathway. J Med Chem 54:262–276
231. Ramakrishnan V, Kimlinger T, Haug J, Timm M, Wellik L, Halling T, Pardanani A, Tefferi
A, Rajkumar SV, Kumar S (2010) TG101209, a novel JAK2 inhibitor, has significant in
vitro activity in multiple myeloma and displays preferential cytotoxicity for CD45+ mye-
loma cells. Am J Hematol 85:675–686
232. Hart S, Goh KC, Novotny-Diermayr V, Hu CY, Hentze H, Tan YC, Madan B, Amalini C,
Loh YK, Ong LC et al. (2011) SB1518, a novel macrocyclic pyrimidine-based JAK2 inhibi-
tor for the treatment of myeloid and lymphoid malignancies. Leuk: Official J Leuk Soc Am
Leuk Res Fund UK 25:1751–1759
3 Emerging Opportunities for Targeting 85
248. Seavey MM, Lu LD, Stump KL, Wallace NH, Hockeimer W, O’Kane TM, Ruggeri BA,
Dobrzanski P (2012) Therapeutic efficacy of CEP-33779, a novel selective JAK2 inhibitor,
in a mouse model of colitis-induced colorectal cancer. Mol Cancer Ther 11:984–993
249. Michaud-Levesque J, Bousquet-Gagnon N, Beliveau R (2012) Quercetin abrogates IL-6/
STAT3 signaling and inhibits glioblastoma cell line growth and migration. Exp Cell Res
318:925–935
Chapter 4
The Role of Autophagy in Drug Resistance
and Potential for Therapeutic Targeting
4.1 Introduction
Cancer cells are faced with multiple metabolic and therapeutic stresses and rely
on intracellular stress response systems for survival. One such stress response that
can be activated by nutrient deprivation, hypoxia, intracellular pathogens, chemo-
therapy, targeted therapies, and/or radiotherapy is autophagy. Autophagy is a term
derived from the Greek word, “to eat” (-phag, y) “oneself” (auto). The term was
first coined to describe vesicular structures noted on electron microscopy (EM)
that contained cytoplasmic contents. Subsequent studies demonstrated that these
contents consisted of organelles and proteins that were in various stages of catabo-
lism [1, 2]. Autophagy is the only mechanism that eukaryotic cells possess that
allows for the bulk degradation of intracellular organelles, which are often dam-
aged and serve as a liability in stressed cells. Autophagy consists of multiple
related vesicular trafficking programs within the cancer cell and is coordinated by
a complex interplay between dedicated enzymes, cellular membranes, cytoskele-
ton, and motor proteins.
Initially, autophagy was described as type II programmed cell death, and “self-
eating”, and, if persistent, can result in exhaustion of all intracellular resources,
4.2 Defining Autophagy
Three forms of autophagy have been identified, each defined on the basis by which
the lysosome obtains the material targeted for recycling. In “macroautophagy,”
(hereafter referred to as autophagy) a double-membrane structure defined as the
autophagosome, or autophagic vesicle, envelopes the cargo and then fuses with
the lysosome. In “microautophagy,” an invaginated lysosomal membrane engulfs
the cargo [7]. In “chaperone-mediated autophagy,” a chaperone protein deliv-
ers protein cargo directly to the LAMP2 receptor on lysosomal membranes [8].
Unlike the other two forms of autophagy, chaperone-mediated autophagy has been
characterized in higher eukaryotes but not in yeast.
In addition to these described forms of autophagy, there is an increasing aware-
ness of the distinct roles of “basal autophagy” and “stress-induced autophagy”.
Basal autophagy likely plays an essential homeostatic role in removing and recy-
cling damaged parts, cellular metabolism and may also play a role in atypical
protein secretion processes [9]. Basal autophagy may be regulated entirely at the
posttranslational level in order to maintain the integrity of cellular constituents,
while stress-induced autophagy may involve the regulatory input from pathways
described below.
While autophagy has been generally considered a non-selective degradative
process, selective autophagy has been described and includes mitophagy (mito-
chondria), ribophagy (ribosomes), pexophagy (peroxisomes), and reticulophagy
4 The Role of Autophagy in Drug Resistance 89
Vesicle nucleation is the initial process by which proteins and lipids are recruited
for autophagosome formation. Studies indicate that the lipid membrane compo-
nent of the autophagosome may be derived in whole or from parts of the mito-
chondria, the endoplasmic reticulum, the plasma membrane or the nuclear
membrane [27–29]. As further evidence of this, Atg9 has been shown to traffic
between the trans-Golgi network, endosomes, and autophagosome precursors
[30], implying that the membrane may be derived from the above precursors.
Finally, electron tomography has demonstrated connections between the ER and
the autophagosomal membranes indicating that the ER cisternae associate with the
developing autophagosomes [31, 32].
Vesicle nucleation starts with the recruitment of Atg proteins to the phagophore
assembly site (PAS). While the exact mechanism of this step is unclear, activation
of a phosphatidylinositol 3-kinase (PtdIns3K) complex is necessary. Generation
of phosphoinositide signals on the surface of source membranes is accomplished
by various protein complexes that include the class III phosphoinositide 3-kinase
(PI3K), Vps34, and Beclin1 [33], which further complexes with UVRAG and
Bif-1 and thus activates autophagy [34]. Association with Rubicon inhibits traf-
ficking of autophagic vesicles [35]. These findings implicate Vps34/Beclin1 com-
plexes in early and late roles in autophagic flux.
Table 4.1 Yeast autophagy genes and mammalian homologs and their respective functions
Yeast Mammal Function References
Atg1 complex Induction
Atg1 Ulk1 and Ulk 2 Serine/threonine [24, 25]
protein kinase
Atg13 Atg13 Unknown [24, 25]
Atg101 Atg13-binding protein [15]
Atg17 FIP200 (RB1CC1) Modulates response of [24]
autophagy
Atg24 (Snx4) PtdIns(3)P-binding protein [200]
Table 4.1 (Continued)
Yeast Mammal Function References
Atg9 mAtg9A Transmembrane protein, [30]
serves as a lipid carrier
Ypt1 Rab1 Required for correct [208]
localization of Atg8 to
the PAS; in mammals,
Rab1 is also required for
autophagosome formation
Ypt7 Rab7 Small GTP-binding protein, [209]
facilitates transport from
early to late endosomes
and from late endosomes
to lysosomes, facilitates
clearance of autophagic
compartments
Sec18 NSF ATPase responsible for [42]
SNARE disassembly
Atg Autophagy related, ULK Unc51-like kinase, FIP200 focal adhesion kinase (FAK) family–
interacting protein of 200 kDa, RB1CC1 retinoblastoma 1-inducible coiled-coil 1, Beclin1 Bcl-2
interacting myosin/moesin-like coiled-coil protein 1, Vps vacuolar protein sorting, UVRAG UV
irradiation resistance-associated gene, WIPI WD repeat protein interacting with phosphoinositides,
Barkor Beclin1-associated autophagy-related key regulator, GATE-16 Golgi-associated ATPase
enhancer of 16 kDa/GABARAPL2, GABARAP gamma-aminobutyric acid receptor-associated
protein, LC3 microtubule-associated protein 1 light chain 3, TSC1/2 tuberous sclerosis complex ½,
Ypt yeast protein, PE phosphatidylethanolamine
LC3 not only plays a direct role in vesicle nucleation, but also plays a role in the
recruitment of cargo into the developing autophagic vesicle. p62/SQSTM1 has been
shown to recruit aggregated proteins to autophagic vesicles [43], while NBR and
NIX are responsible for the recruitments of organelles [44]. NBR and p62 contain
ubiquitin-binding domains and LC3-binding domains. These domains allow for the
4 The Role of Autophagy in Drug Resistance 93
The significance of membrane composition and the fate of the membrane follow-
ing autolysosome formation are unanswered questions. Furthermore, studies are
needed to identify the mechanisms by which the non-selective versus selective
autophagy pathways are discriminated, the mechanism(s) by which organelles
and/or cytosolic debris are identified and marked for autophagy, and the regulatory
components that limit degradation.
There are multiple in vitro tools to measure autophagy [62]. LC3-I is localized in
the cytosol and LC3-II on the autophagosome surface; assessment of the ratios of
LC3-I to LC3-II using protein-based assays can be employed as a surrogate for
autophagosome induction and flux. Use of inhibitors to cathepsin B, H and L and
pepstatin A can inhibit lysosomal function [63] and further define these processes.
Lysosomal inhibition causes accumulation of autolysosomes and therefore LC3-II.
This finding can be recapitulated by using other lysosomotropic agents like chloro-
quine derivatives or bafilomycin A1, an inhibitor of vacuolar H+-ATPase. Because
V-ATPase contributes to the acidification of other organelles, including endosomes,
bafilomycin A1 may show multiple off-target effects.
In situ imaging of LC3 dynamics can be visualized by fusing LC3 to a green
fluorescent probe (GFP). In the absence of autophagy induction, GFP-LC3 fluo-
rescence is diffuse, while discrete puncta are formed upon induction or blockade
of autophagic flux. It is important to note that in certain circumstances, GFP-LC3
can form puncta in cells independent of autophagy [64, 65] and that GFP fluores-
cence in lysosomes may occur even after degradation of the LC3 moiety. As such,
this method may overestimate the number of autophagosomes. A recent iteration of
the GFP-LC3 assay involves the mRFP-GFP-LC3 color change assay. This assay
capitalizes on differential pH stability between GFP and mRFP, and differential
pH of autophagosomes and lysosomes, the latter which has an acidic pH. In acidic
environments, the fluorescence of mRFP is stable, while GFP is decreased. Merged
mRFP-GFP-LC3 in autophagosomes is yellow, whereas it is red in autolysosomes
[66]. Cells lacking Atg3 continue to have impaired but present autophagosome for-
mation [67], indicating that LC3 lipidation is not the only process required for mem-
brane maturation and should not be the only marker used to assess induction/flux.
Adaptor molecules such as p62/SQSTM1 and NBR1 bind to ubiquitin-labeled
structures and recruit them inside autophagosomal. P62/SQSTM, which facilitates
protein aggregate clearance by autophagy, is degraded by autophagy and can be
found in cellular inclusion bodies that are presumably remnants of autophagic diges-
tion [43, 68]. Inhibition of autophagy leads to the accumulation of p62/SQSTMI.
EM is often viewed as the gold standard for qualitatively assessing autophagy.
Although EM measurements can be subjective, criteria have been defined for
4 The Role of Autophagy in Drug Resistance 95
4.5.1 PI3K/Akt/mTOR
4.5.2 MAPK Signaling
abnormal mitochondria, depleting metabolites for the Krebs cycle, and decreas-
ing oxygen consumption [85]. Data also indicate that hyperactivation of oncogenic
BRAF induces autophagy; subsequent inhibition and/or depletion of its down-
stream effectors, MEK or ERK, inhibits it [86].
4.5.3 Bcl-2
4.5.4 AMPK
Autophagy is required for and/or significantly impacts a wide variety of other cel-
lular processes including but not limited to apoptosis [97, 98], necrosis [99], cell
cycle control [100], immune modulation [101], angiogenesis [102], cell metabo-
lism [103], protein and organelle turnover [104], and cell survival [104]. These
internal and external processes are, in turn, mediated by nutrient, energy, oxygen,
and hormonal demands. Processing these diverse cellular inputs is critical for
maintaining cellular homeostasis. The primary role of autophagy is to protect cells
98 R. Rangwala and R. Amaravadi
under stress conditions, such as starvation and hypoxia. During periods of nutri-
ent deprivation, autophagy degrades cytoplasmic materials to produce amino acids
and fatty acids that can be used to synthesize new proteins or ATP [105]. When
induction of autophagy exceeds homeostatic control, autophagic cell death, also
known as type II programmed cell death, is induced [105–108]. Understanding
how autophagic regulation is effected by glucose, oxygen, and amino acid stores
may provide further insight that will ultimately allow us to leverage autophagy to
our therapeutic advantage in treating disease states such as cancer.
Incubation of murine breast cancer cells in media containing less than 5 mM of
glucose induces autophagy [123]. Glucose deprivation activates the AMP kinase
(AMPK) and TSC2, which, in turn, inhibits the GTPase Rheb and its downstream
4 The Role of Autophagy in Drug Resistance 99
effector mTORC1 [124, 125] by direct activation of the ULK1 complex [126]. Also,
oncogenic Ras and Myc can shunt energy production from oxidative phosphoryla-
tion to glycolysis [127] as a result of PI3K/Akt/mTOR pathway activation. The loss
of the tumor suppressor p53 promotes glycolysis through enhanced glucose trans-
port [128], enhanced phosphoglycerate mutase expression [129], and suppression of
the mitochondrial enzyme synthesis of cytochrome c oxidase 2 (SCO2) [130].
Amino acid deprivation is one of the most potent inducers of autophagy [137].
mTORC1 regulates both protein and cytoplasmic amino acid levels by regulating
the amino acid transporter expression and autophagy [138]. Furthermore, mTORC1
facilitates the transportation of leucine well as its degradation by autophagy and the
ubiquitin-proteasomal system [139, 140]. Additional regulation occurs as a result
of convergence of AMPK onto mTORC1. Ammonia, a glutaminolysis byproduct,
stimulates autophagy by a mechanism dependent on Atg5 [141, 142].
organ explants treated with either agent had an increased number of autophagic vesi-
cles as compared to controls [145]. Autophagy induction has also been implicated as
a resistance mechanism to topoisomerase I and topoisomerase II inhibitors [146].
4.6.8 Proteasomal Inhibitors
Autophagy and the ubiquitin-proteasome system (UPS) are the two intracellu-
lar mechanisms by which proteins are degraded. Autophagy degrades long-lived,
cytosolic proteins and damaged organelles; while shorter-lived proteins are tar-
geted for catabolism by the UPS. Inhibition of the UPS by proteasomal inhibitors,
like bortezomib, induces autophagy by multiple pathways including activation
of HDAC6, activation of IRE1-JNK, stabilization of ATF4, inhibition of mTOR
signaling, and decreased proteasomal degradation of LC3. Not surprisingly, these
pathways show commonality with the regulatory mechanisms of ER stress. The
induction of autophagy has also been shown to mitigate the anti-tumor effects of
proteasome inhibition, including in multiple myeloma [149].
that the HDAC inhibitors induce autophagy dependent and independent of other
intersecting pathways including mTOR, tyrosine kinase induction as a result of
BCR-ABL fusion, and estrogen receptor–activated signaling [151–154] (see
Fig. 4.1).
The above data implicate autophagy as a guardian against genomic instability and
resultant tumorigenesis and therefore support a role for autophagy as a tumor sup-
pressor mechanism. However, the critical role of autophagy as a stress response in
tumor cells, suggests that human tumors growing within the harsh conditions of
the tumor microenvironment may in fact have high levels of autophagy. The ear-
liest data supporting this view come from experiments characterizing autophagy
in mouse models of Akt-driven solid tumors that are deficient in apoptosis [165].
Furthermore, these studies demonstrated that autophagy was induced in tumors
grown in hypoxic environments.
4 The Role of Autophagy in Drug Resistance 103
Additional evidence to support this tumor potentiating role is the high levels of
autophagy, as assessed by EM and LC3 immunohistochemistry, in both metastatic
melanoma and pancreas cancer [85]. LC3B was elevated in 84 % of cases of 20
advanced cancer histologies, and elevated levels of autophagy in breast cancer and
melanoma primary tumors correlated with lymph node metastases and poor sur-
vival [131].
Although monoallelic deletion of beclin has been found in a large subset of
common tumors such as breast and prostate cancer, the retained allele of beclin
in these tumors is always wild type. Beclin1 “deficient” cells usually have simi-
lar levels of autophagy induced by stresses as compared to beclin WT cells.
Beclin1 is bound tightly in a complex involving cytosolic p53. In cells derived
from beclin+/− mice, p53 levels were found to be lower than in cells derived from
beclin WT mice. This finding suggests that p53 deficiency and not autophagy
deficiency may explain why beclin-deficient mice develop malignancies, with
the caveat that Liu and colleagues did not differentiate cytosolic from nuclear
p53 [176]. Furthermore, no other mouse model of genetic autophagy deficiency
has produced true spontaneous malignancies. For example, FIP200−/− mice are
found to have impaired tumorigenesis [177]. In addition, inactivating somatic
mutations have yet to be reported in any other autophagy genes, save for poly-
morphisms in the ATG16L1 gene, which predispose patients to Crohn’s disease
[178] and not cancer. Finally, there is compelling evidence that Ras transforma-
tion produces a massive derangement in cancer cell metabolism that necessitates
high levels of autophagy to avoid oncogene-induced senescence or cell death [84].
Therefore, autophagy may be essential for tumorigenesis and cancer maintenance
and autophagy-deficient tumors may be rare.
How can these dual, opposing roles be reconciled? As detailed above, autophagy
is regulated by multiple, intersecting pathways. Emerging evidence supports the
concept that its tumor suppressor versus tumor potentiator role may be pathway
specific and not necessarily mutually exclusive. Given the multiple mechanisms
by which autophagy is regulated, mutations in any of these signaling nodes will
affect cellular homeostasis and therefore the role of autophagy within that cel-
lular environment. This, in turn, effects whether autophagy modulates pro-sur-
vival versus pro-death pathways. Both the degree and duration of autophagy has
been proposed as an explanation as to the role in which autophagy functions. In
C. elegans, physiological levels of autophagy promote survival during nutri-
ent poor conditions, while either insufficient or excessive autophagy under these
same nutrient poor conditions leads to cell death [179]. In addition, excessive
autophagy likely compromises the integrity of the lysosomal membranes result-
ing in the release of cathepsins into the cytosol [180–182] and may underlie the
104 R. Rangwala and R. Amaravadi
The cytotoxic effects of blocking autophagy with chloroquine in cells that are
reliant on autophagy for survival was first demonstrated in apoptosis-defective
cells exposed to the stress of growth factor withdrawal [186]. Chloroquine also
augmented chemotherapy-induced tumor impairment in a model of Myc-induced
lymphoma [3]. Since these papers, numerous investigators have demonstrated that
combining chloroquine or siRNA against essential autophagy genes can augment
the efficacy of many existing and emerging cancer therapies in many different dis-
ease models (see selected examples in Table 4.2). Because effective autophagy
inhibition can be achieved in vivo with the anti-malarial drug chloroquine (CQ)
[3], and there is extensive experience with CQ derivatives for the treatment for
malaria [187], rheumatoid arthritis [188], and HIV [189], multiple trials have been
launched across a wide variety of tumor types.
A phase III trial in glioblastoma patients treated with radiation and carmustine with
or without daily CQ found a median overall survival of 24 and 11 months in CQ-
and placebo-treated patients, respectively [190]. While it was not adequately pow-
ered to detect a significant difference in survival, it established the safety of adding
low dose CQ to DNA alkylators. Its long half-life and low potency may limit its effi-
cacy as an autophagy inhibitor in patients [191]. To address these concerns, a phase
I/II trial of HCQ with temozolomide and radiation for glioblastoma patients was
launched and included pharmacodynamic (PD) and pharmacokinetic (PK) analyses.
PD evidence of HCQ dose–dependent autophagy inhibition was observed using an
EM assay on serial blood mononuclear cells [192]. The implications of autophagy
as a mechanism by which tumor growth is potentiated have significant clinical
implications: clinical effect observed with standard therapies may be improved
upon by the addition of drugs that inhibit autophagy. To date, only the chloroquine
4 The Role of Autophagy in Drug Resistance 105
derivatives (chloroquine and hydroxychloroquine) have been used in the clinical set-
ting. Since 2007, more than 20 trials have been launched involving HCQ. A com-
plete listing of ongoing and/or completed trials can be found at clinicaltrials.gov.
The knowledge gained from the PD, PK, and predictive biomarkers in these studies
will guide the development of more potent and specific autophagy inhibitors.
Fig. 4.2 Inhibitors of autophagy. Spautin, wortmannin, LY294002, and 3-MA are pan inhibitors
of PtdIns3K. Lucanthone, Lys05, and chloroquine inhibit lysosomal function. Bafilomycin A1
prevents the maturation of autophagic vacuoles
Providing for cellular homeostasis in the face of rapid proliferation can be a challenge
as the cell integrates signals generated from intracellular alterations in glucose, oxygen,
energy, and amino acid stores. An additional layer of complexity lies in the fact that the
cell is also responsible for assimilating extracellular signals arising from the immune
system, pathogens, radio-, targeted-, hormonal-, and/or chemotherapies. These diverse,
sometimes diametrically opposed signals can each stimulate autophagy. In this capac-
ity, autophagy can serve as a stress mechanism that can maintain survival despite limi-
tations in essential nutrients and has been described as such in both physiological and
pathophysiological settings. Interestingly, though, unchecked autophagy can also lead
to cell death. Predicting these diametrically opposed roles is at the heart of leveraging
autophagy as a novel target for cancer therapy. Currently, the therapeutic strategy tar-
geting autophagy as a means to enhance clinical cancer benefit is by inhibiting it. An
abundance of preclinical evidence exists supporting autophagy as a mechanism by
which a cancerous cell protects itself from reactive oxygen and reactive nitrogen spe-
cies accumulation, genetic instability, hypoxia, and diminished glucose stores. Multiple
clinical trials have been launched utilizing chloroquine and the chloroquine derivative,
hydroxychloroquine. Despite their extensive clinical use for the treatment for malaria,
rheumatoid arthritis, and HIV, their oncology use may be limited by their narrow thera-
peutic window, their long half-life, and accumulating toxicities. Development of new
autophagy inhibitors is being pursued and will likely advance this target as a cancer
therapy adjunct.
Hand in hand with the development of new autophagy inhibitors is our ability
to prospectively identify cancer patients who will benefit from autophagy inhibi-
tion. Currently, data from ongoing and completed trials that couple standard thera-
peutics with autophagy inhibition show mixed response. Further work needs to be
pursued concerning pre-identification of sensitive tumor types, recognizing appro-
priate therapeutic regimens that synergistically act with autophagy inhibition to
enhance clinical benefit, and development of more robust pharmacodynamic mark-
ers to assess autophagy inhibition in vivo.
A yet theoretical application for autophagy lies in the use of pharmacologic
autophagic inducers in those cells or tumors in which autophagy stimulates cell
death. The challenge for this application lies in the development of such drugs as
well as our ability to prospectively identify those cells in which autophagy induces
cell death, not cell survival.
108 R. Rangwala and R. Amaravadi
Regardless of the above challenges, research into the metabolic drivers, iden-
tification of the delicate interplay between the immune system and autophagy as
well as development of mouse models is being actively pursued. These insights
will improve our understanding of the role of autophagy in normal, dysplastic,
and cancerous cells and therefore will allow us to better target autophagy and ulti-
mately improve cancer therapy.
References
1. Ashford TP, Porter KR (1962) Cytoplasmic components in hepatic cell lysosomes. J Cell
Biol 12:198–202
2. Watanabe E et al (2009) Sepsis induces extensive autophagic vacuolization in hepatocytes: a
clinical and laboratory-based study. Lab Invest 89(5):549–561
3. Amaravadi RK et al (2007) Autophagy inhibition enhances therapy-induced apoptosis in a
Myc-induced model of lymphoma. J Clin Invest 117(2):326–336
4. Klionsky DJ (2007) Autophagy: from phenomenology to molecular understanding in less
than a decade. Nat Rev Mol Cell Biol 8(11):931–937
5. Kroemer G, Jaattela M (2005) Lysosomes and autophagy in cell death control. Nat Rev
Cancer 5(11):886–897
6. Levine B, Deretic V (2007) Unveiling the roles of autophagy in innate and adaptive
immunity. Nat Rev Immunol 7(10):767–777
7. Muller O et al (2000) Autophagic tubes: vacuolar invaginations involved in lateral mem-
brane sorting and inverse vesicle budding. J Cell Biol 151(3):519–528
8. Bandyopadhyay U et al (2008) The chaperone-mediated autophagy receptor organizes in
dynamic protein complexes at the lysosomal membrane. Mol Cell Biol 28(18):5747–5763
9. Chen HY, White E (2011) Role of autophagy in cancer prevention. Cancer Prev Res (Phila)
4(7):973–983
10. He C, Klionsky DJ (2009) Regulation mechanisms and signaling pathways of autophagy.
Annu Rev Genet 43:67–93
11. Gu Y, Wang C, Cohen A (2004) Effect of IGF-1 on the balance between autophagy of dys-
functional mitochondria and apoptosis. FEBS Lett 577(3):357–360
12. Bjorkoy G et al (2005) p62/SQSTM1 forms protein aggregates degraded by autophagy and
has a protective effect on huntingtin-induced cell death. J Cell Biol 171(4):603–614
13. Wild P, Dikic I (2010) Mitochondria get a Parkin’ ticket. Nat Cell Biol 12(2):104–106
14. Di Bartolomeo S, Nazio F, Cecconi F (2010) The role of autophagy during development in
higher eukaryotes. Traffic 11(10):1280–1289
15. Mizushima N et al (2008) Autophagy fights disease through cellular self-digestion. Nature
451(7182):1069–1075
16. Wang CW, Klionsky DJ (2003) The molecular mechanism of autophagy. Mol Med
9(3–4):65–76
17. Kovacs AL, Zhang H (2010) Role of autophagy in Caenorhabditis elegans. FEBS Lett
584(7):1335–1341
18. Chang YY, Neufeld TP (2010) Autophagy takes flight in drosophila. FEBS Lett
584(7):1342–1349
19. Rong Y et al (2011) Spinster is required for autophagic lysosome reformation and mTOR
reactivation following starvation. Proc Natl Acad Sci USA 108(19):7826–7831
20. Teter SA et al (2001) Degradation of lipid vesicles in the yeast vacuole requires function of
Cvt17, a putative lipase. J Biol Chem 276(3):2083–2087
21. Epple UD et al (2001) Aut5/Cvt17p, a putative lipase essential for disintegration of
autophagic bodies inside the vacuole. J Bacteriol 183(20):5942–5955
4 The Role of Autophagy in Drug Resistance 109
22. Yang Z et al (2006) Atg22 recycles amino acids to link the degradative and recycling func-
tions of autophagy. Mol Biol Cell 17(12):5094–5104
23. Kabeya Y et al (2005) Atg17 functions in cooperation with Atg1 and Atg13 in yeast
autophagy. Mol Biol Cell 16(5):2544–2553
24. Jung CH et al (2009) ULK-Atg13-FIP200 complexes mediate mTOR signaling to the
autophagy machinery. Mol Biol Cell 20(7):1992–2003
25. Mizushima N (2010) The role of the Atg1/ULK1 complex in autophagy regulation. Curr
Opin Cell Biol 22(2):132–9
26. Cheong H et al (2011) Ammonia-induced autophagy is independent of ULK1/ULK2
kinases. Proc Natl Acad Sci USA 108(27):11121–11126
27. Hailey DW et al (2010) Mitochondria supply membranes for autophagosome biogenesis
during starvation. Cell 141(4):656–667
28. Ravikumar B et al (2010) Plasma membrane contributes to the formation of pre-autophago-
somal structures. Nat Cell Biol 12(8):747–757
29. English L et al (2009) Autophagy enhances the presentation of endogenous viral antigens on
MHC class I molecules during HSV-1 infection. Nat Immunol 10(5):480–487
30. Webber JL, Tooze SA (2010) New insights into the function of Atg9. FEBS Lett
584(7):1319–1326
31. Hayashi-Nishino M et al (2009) A subdomain of the endoplasmic reticulum forms a cradle
for autophagosome formation. Nat Cell Biol 11(12):1433–1437
32. Yla-Anttila P et al (2009) 3D tomography reveals connections between the phagophore and
endoplasmic reticulum. Autophagy 5(8):1180–1185
33. Funderburk SF, Wang QJ, Yue Z (2010) The Beclin 1-VPS34 complex–at the crossroads of
autophagy and beyond. Trends Cell Biol 20(6):355–2
34. Itakura E et al (2008) Beclin 1 forms two distinct phosphatidylinositol 3-kinase complexes
with mammalian Atg14 and UVRAG. Mol Biol Cell 19(12):5360–5372
35. Matsunaga K et al (2009) Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally
regulate autophagy at different stages. Nat Cell Biol 11(4):385–396
36. Kabeya Y et al (2000) LC3, a mammalian homologue of yeast Apg8p, is localized in
autophagosome membranes after processing. EMBO J 19(21):5720–5728
37. Ichimura Y et al (2000) A ubiquitin-like system mediates protein lipidation. Nature
408(6811):488–492
38. Yoshimura K et al (2006) Effects of RNA interference of Atg4B on the limited pro-
teolysis of LC3 in PC12 cells and expression of Atg4B in various rat tissues. Autophagy
2(3):200–208
39. Satoo K et al (2009) The structure of Atg4B-LC3 complex reveals the mechanism of LC3
processing and delipidation during autophagy. EMBO J 28(9):1341–1350
40. Tanida I et al (2002) Human Apg3p/Aut1p homologue is an authentic E2 enzyme for mul-
tiple substrates, GATE-16, GABARAP, and MAP-LC3, and facilitates the conjugation of
hApg12p to hApg5p. J Biol Chem 277(16):13739–13744
41. Tanida I et al (2001) The human homolog of Saccharomyces cerevisiae Apg7p is a protein-
activating enzyme for multiple substrates including human Apg12p, GATE-16, GABARAP,
and MAP-LC3. J Biol Chem 276(3):1701–1706
42. Nair U et al (2011) SNARE proteins are required for macroautophagy. Cell 146(2):290–302
43. Pankiv S et al (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of
ubiquitinated protein aggregates by autophagy. J Biol Chem 282(33):24131–24145
44. Schweers RL et al (2007) NIX is required for programmed mitochondrial clearance during
reticulocyte maturation. Proc Natl Acad Sci USA 104(49):19500–19505
45. Kim PK et al (2008) Ubiquitin signals autophagic degradation of cytosolic proteins and per-
oxisomes. Proc Natl Acad Sci USA 105(52):20567–20574
46. Novak I et al (2010) Nix is a selective autophagy receptor for mitochondrial clearance.
EMBO Rep 11(1):45–51
47. Sandoval H et al (2008) Essential role for nix in autophagic maturation of erythroid cells.
Nature 454(7201):232–235
110 R. Rangwala and R. Amaravadi
48. Jager S et al (2004) Role for Rab7 in maturation of late autophagic vacuoles. J Cell Sci
117(Pt 20):4837–4848
49. Djeddi A et al (2012) Induction of autophagy in ESCRT mutants is an adaptive response for
cell survival in C. elegans. J Cell Sci 125(Pt 3):685–694
50. Fader CM, Colombo MI (2009) Autophagy and multivesicular bodies: two closely related
partners. Cell Death Differ 16(1):70–78
51. Young AR et al (2006) Starvation and ULK1-dependent cycling of mammalian Atg9
between the TGN and endosomes. J Cell Sci 119(Pt 18):3888–3900
52. Manjithaya R et al (2010) Unconventional secretion of Pichia pastoris Acb1 is depend-
ent on GRASP protein, peroxisomal functions, and autophagosome formation. J Cell Biol
188(4):537–46
53. Kabeya Y et al (2004) LC3, GABARAP and GATE16 localize to autophagosomal mem-
brane depending on form-II formation. J Cell Sci 117(Pt 13):2805–2812
54. Tanida I et al (2003) GATE-16 and GABARAP are authentic modifiers mediated by Apg7
and Apg3. Biochem Biophys Res Commun 300(3):637–644
55. Tanida I, Ueno T, Kominami E (2004) LC3 conjugation system in mammalian autophagy.
Int J Biochem Cell Biol 36(12):2503–2518
56. Weidberg H et al (2010) LC3 and GATE-16/GABARAP subfamilies are both essential yet
act differently in autophagosome biogenesis. EMBO J 29(11):1792–1802
57. Marino G et al (2003) Human autophagins, a family of cysteine proteinases potentially
implicated in cell degradation by autophagy. J Biol Chem 278(6):3671–3678
58. Behrends C et al (2010) Network organization of the human autophagy system. Nature
466(7302):68–76
59. Lee SJ et al (2011) Beclin 1 deficiency is associated with increased hypoxia-induced angio-
genesis. Autophagy 7(8):829–839
60. Wirawan E et al (2012) Beclin1: a role in membrane dynamics and beyond. Autophagy
8(1):6–17
61. Nishida Y et al (2009) Discovery of Atg5/Atg7-independent alternative macroautophagy.
Nature 461(7264):654–658
62. Klionsky DJ et al (2008) Guidelines for the use and interpretation of assays for monitoring
autophagy in higher eukaryotes. Autophagy 4(2):151–175
63. Tanida I et al (2005) Lysosomal turnover, but not a cellular level, of endogenous LC3 is a
marker for autophagy. Autophagy 1(2):84–91
64. Ciechomska IA, Tolkovsky AM (2007) Non-autophagic GFP-LC3 puncta induced by sapo-
nin and other detergents. Autophagy 3(6):586–590
65. Kuma A, Matsui M, Mizushima N (2007) LC3, an autophagosome marker, can be incorpo-
rated into protein aggregates independent of autophagy: caution in the interpretation of LC3
localization. Autophagy 3(4):323–328
66. Kimura S, Noda T, Yoshimori T (2007) Dissection of the autophagosome maturation process
by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy 3(5):452–460
67. Sou YS et al (2008) The Atg8 conjugation system is indispensable for proper development
of autophagic isolation membranes in mice. Mol Biol Cell 19(11):4762–4775
68. Mathew R et al (2009) Autophagy suppresses tumorigenesis through elimination of p62.
Cell 137(6):1062–1075
69. Ma X et al (2011) Measurements of tumor cell autophagy predict invasiveness, resistance to
chemotherapy, and survival in melanoma. Clin Cancer Res 17(10):3478–3489
70. Mendoza MC, Er EE, Blenis J (2011) The Ras-ERK and PI3 K-mTOR pathways: cross-talk
and compensation. Trends Biochem Sci 36(6):320–328
71. Steelman LS et al (2011) Roles of the Raf/MEK/ERK and PI3 K/PTEN/Akt/mTOR path-
ways in controlling growth and sensitivity to therapy-implications for cancer and aging.
Aging (Albany NY) 3(3):192–222
72. Blommaart EF et al (1995) Phosphorylation of ribosomal protein S6 is inhibitory for
autophagy in isolated rat hepatocytes. J Biol Chem 270(5):2320–2326
73. Hosokawa N et al (2009) Nutrient-dependent mTORC1 association with the ULK1-Atg13-
FIP200 complex required for autophagy. Mol Biol Cell 20(7):1981–1991
4 The Role of Autophagy in Drug Resistance 111
74. Sancak Y et al (2010) Ragulator-Rag complex targets mTORC1 to the lysosomal surface
and is necessary for its activation by amino acids. Cell 141(2):290–303
75. Arico S et al (2001) The tumor suppressor PTEN positively regulates macroautophagy
by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway. J Biol Chem
276(38):35243–35246
76. Ueno T et al (2008) Loss of Pten, a tumor suppressor, causes the strong inhibition of
autophagy without affecting LC3 lipidation. Autophagy 4(5):692–700
77. Laane E et al (2009) Cell death induced by dexamethasone in lymphoid leukemia is medi-
ated through initiation of autophagy. Cell Death Differ 16(7):1018–1029
78. Kuo HP et al (2010) ARD1 stabilization of TSC2 suppresses tumorigenesis through the
mTOR signaling pathway. Sci Signal 3(108):ra9
79. Zeng X, Kinsella TJ (2008) Mammalian target of rapamycin and S6 kinase 1 positively reg-
ulate 6-thioguanine-induced autophagy. Cancer Res 68(7):2384–2390
80. Yaswen P, Campisi J (2007) Oncogene-induced senescence pathways weave an intricate tap-
estry. Cell 128(2):233–234
81. Serrano M et al (1997) Oncogenic ras provokes premature cell senescence associated with
accumulation of p53 and p16INK4a. Cell 88(5):593–602
82. Elgendy M et al (2011) Oncogenic Ras-induced expression of Noxa and Beclin-1 promotes
autophagic cell death and limits clonogenic survival. Mol Cell 42(1):23–35
83. Wu SY et al (2011) Ras-related tumorigenesis is suppressed by BNIP3-mediated autophagy
through inhibition of cell proliferation. Neoplasia 13(12):1171–1182
84. Guo JY et al (2011) Activated Ras requires autophagy to maintain oxidative metabolism and
tumorigenesis. Genes Dev 25(5):460–470
85. Yang S et al (2011) Pancreatic cancers require autophagy for tumor growth. Genes Dev
25(7):717–729
86. Maddodi N et al (2010) Induction of autophagy and inhibition of melanoma growth in vitro
and in vivo by hyperactivation of oncogenic BRAF. J Invest Dermatol 130(6):1657–1667
87. Zhou W et al (2011) Small interfering RNA targeting mcl-1 enhances proteasome inhibitor-
induced apoptosis in various solid malignant tumors. BMC Cancer 11:485
88. Pattingre S et al (2005) Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy.
Cell 122(6):927–939
89. Ku B et al (2008) An insight into the mechanistic role of Beclin 1 and its inhibition by pro-
survival Bcl-2 family proteins. Autophagy 4(4):519–520
90. Hoyer-Hansen M, Jaattela M (2007) AMP-activated protein kinase: a universal regulator of
autophagy? Autophagy 3(4):381–383
91. Liang J et al (2007) The energy sensing LKB1-AMPK pathway regulates p27(kip1)
phosphorylation mediating the decision to enter autophagy or apoptosis. Nat Cell Biol
9(2):218–224
92. Egan D et al (2011) The autophagy initiating kinase ULK1 is regulated via opposing phos-
phorylation by AMPK and mTOR. Autophagy 7(6):643–644
93. Kim J, Guan KL (2011) Regulation of the autophagy initiating kinase ULK1 by nutrients:
Roles of mTORC1 and AMPK. Cell Cycle 10(9):1337–1338
94. Alexander A, Walker CL (2010) Differential localization of ATM is correlated with activa-
tion of distinct downstream signaling pathways. Cell Cycle 9(18):3685–3686
95. Herrero-Martin G et al (2009) TAK1 activates AMPK-dependent cytoprotective autophagy
in TRAIL-treated epithelial cells. EMBO J 28(6):677–685
96. Tzatsos A, Tsichlis PN (2007) Energy depletion inhibits phosphatidylinositol 3-kinase/Akt
signaling and induces apoptosis via AMP-activated protein kinase-dependent phosphoryla-
tion of IRS-1 at Ser-794. J Biol Chem 282(25):18069–18082
97. Xu J et al (2012) MicroRNAs in autophagy and their emerging roles in crosstalk with apop-
tosis. Autophagy 8(6):873–882
98. Baldwin AS (2012) Regulation of cell death and autophagy by IKK and NF-kappaB: critical
mechanisms in immune function and cancer. Immunol Rev 246(1):327–345
99. Jin Y et al (2012) Autophagic proteins: New facets of the oxygen paradox. Autophagy
8(3):426–428
112 R. Rangwala and R. Amaravadi
100.
Wang RC, Levine B (2010) Autophagy in cellular growth control. FEBS Lett
584(7):1417–1426
101. Chaturvedi A, Pierce SK (2009) Autophagy in immune cell regulation and dysregulation.
Curr Allergy Asthma Rep 9(5):341–346
102. Wang L et al (2012) The roles of integrin beta4 in vascular endothelial cells. J Cell Physiol
227(2):474–478
103. Lee J, Giordano S, Zhang J (2012) Autophagy, mitochondria and oxidative stress: cross-talk
and redox signalling. Biochem J 441(2):523–540
104. Yang Z, Klionsky DJ (2010) Eaten alive: a history of macroautophagy. Nat Cell Biol
12(9):814–822
105. Levine B, Yuan J (2005) Autophagy in cell death: an innocent convict? J Clin Invest
115(10):2679–2688
106. Platini F et al (2010) Understanding autophagy in cell death control. Curr Pharm Des
16(1):101–113
107. Maiuri MC et al (2007) Self-eating and self-killing: crosstalk between autophagy and apop-
tosis. Nat Rev Mol Cell Biol 8(9):741–752
108. Chen Y, Azad MB, Gibson SB (2010) Methods for detecting autophagy and determining
autophagy-induced cell death. Can J Physiol Pharmacol 88(3):285–295
109. Malhotra JD, Kaufman RJ (2007) The endoplasmic reticulum and the unfolded protein
response. Semin Cell Dev Biol 18(6):716–731
110. Ron D, Walter P (2007) Signal integration in the endoplasmic reticulum unfolded protein
response. Nat Rev Mol Cell Biol 8(7):519–529
111. Bertolotti A et al (2000) Dynamic interaction of BiP and ER stress transducers in the
unfolded- protein response. Nat Cell Biol 2(6):326–332
112. Bernales S, Schuck S, Walter P (2007) ER-phagy: selective autophagy of the endoplasmic
reticulum. Autophagy 3(3):285–287
113. Yorimitsu T et al (2006) Endoplasmic reticulum stress triggers autophagy. J Biol Chem
281(40):30299–30304
114. Kruse KB, Brodsky JL, McCracken AA (2006) Autophagy: an ER protein quality control
process. Autophagy 2(2):135–137
115. Younce CW, Kolattukudy PE (2010) MCP-1 causes cardiomyoblast death via autophagy
resulting from ER stress caused by oxidative stress generated by inducing a novel zinc-fin-
ger protein, MCPIP. Biochem J 426(1):43–53
116. Rodriguez-Rocha H et al (2011) DNA damage and autophagy. Mutat Res 711(1–2):158–166
117. Rieber M, Rieber MS (2008) Sensitization to radiation-induced DNA damage accelerates loss of
bcl-2 and increases apoptosis and autophagy. Cancer Biol Ther 7(10):1561–1566
118. Feng Z et al (2005) The coordinate regulation of the p53 and mTOR pathways in cells. Proc
Natl Acad Sci USA 102(23):8204–8209
119. Malzer E et al (2010) Impaired tissue growth is mediated by checkpoint kinase 1 (CHK1) in
the integrated stress response. J Cell Sci 123(Pt 17):2892–2900
120. Crighton D et al (2006) DRAM, a p53-induced modulator of autophagy, is critical for apop-
tosis. Cell 126(1):121–134
121. Lorin S et al (2010) Evidence for the interplay between JNK and p53-DRAM signalling
pathways in the regulation of autophagy. Autophagy 6(1):153–154
122. Feng Z, Levine AJ (2010) The regulation of energy metabolism and the IGF-1/mTOR path-
ways by the p53 protein. Trends Cell Biol 20(7):427–434
123. Wu H et al (2011) Central role of lactic acidosis in cancer cell resistance to glucose depriva-
tion-induced cell death. J Pathol 227(2):189–199
124. Takagi H et al (2007) AMPK mediates autophagy during myocardial ischemia in vivo.
Autophagy 3(4):405–407
125. Mihaylova MM, Shaw RJ (2011) The AMPK signalling pathway coordinates cell growth,
autophagy and metabolism. Nat Cell Biol 13(9):1016–1023
126. Egan DF et al (2011) Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase
connects energy sensing to mitophagy. Science 331(6016):456–461
4 The Role of Autophagy in Drug Resistance 113
151. Carew JS et al (2007) Targeting autophagy augments the anticancer activity of the his-
tone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood
110(1):313–322
152. Park JH et al (2011) A new synthetic HDAC inhibitor, MHY218, induces apoptosis or
autophagy-related cell death in tamoxifen-resistant MCF-7 breast cancer cells. Invest New Drugs
30(5):1887–1898
153. Yamamoto S et al (2008) Suberoylanilide hydroxamic acid (SAHA) induces apoptosis or
autophagy-associated cell death in chondrosarcoma cell lines. Anticancer Res 28(3A)1585–91
154. Hrzenjak A et al (2008) SAHA induces caspase-independent, autophagic cell death of endo-
metrial stromal sarcoma cells by influencing the mTOR pathway. J Pathol 216(4):495–504
155. Saitoh T, Akira S (2010) Regulation of innate immune responses by autophagy-related pro-
teins. J Cell Biol 189(6):925–935
156. Zhao Z et al (2008) Autophagosome-independent essential function for the autophagy pro-
tein Atg5 in cellular immunity to intracellular pathogens. Cell Host Microbe 4(5):458–469
157.
Dreux M, Chisari FV (2010) Viruses and the autophagy machinery. Cell Cycle
9(7):1295–1307
158. Amaravadi RK et al (2011) Principles and current strategies for targeting autophagy for can-
cer treatment. Clin Cancer Res 17(4):654–666
159. Michaud M et al (2011) Autophagy-dependent anticancer immune responses induced by
chemotherapeutic agents in mice. Science 334(6062):1573–1577
160. Liang XH et al (1999) Induction of autophagy and inhibition of tumorigenesis by beclin 1.
Nature 402(6762):672–676
161. Aita VM et al (1999) Cloning and genomic organization of beclin 1, a candidate tumor sup-
pressor gene on chromosome 17q21. Genomics 59(1):59–65
162. Qu X et al (2003) Promotion of tumorigenesis by heterozygous disruption of the beclin 1
autophagy gene. J Clin Invest 112(12):1809–1820
163. Yue Z et al (2003) Beclin 1, an autophagy gene essential for early embryonic
development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci USA
100(25):15077–15082
164. Takamura A et al (2011) Autophagy-deficient mice develop multiple liver tumors. Genes
Dev 25(8):795–800
165. Degenhardt K et al (2006) Autophagy promotes tumor cell survival and restricts necrosis,
inflammation, and tumorigenesis. Cancer Cell 10(1):51–64
166. Karantza-Wadsworth V et al (2007) Autophagy mitigates metabolic stress and genome dam-
age in mammary tumorigenesis. Genes Dev 21(13):1621–1635
167. Mathew R, White E (2007) Why sick cells produce tumors: the protective role of autophagy.
Autophagy 3(5):502–505
168. Komatsu M et al (2010) The selective autophagy substrate p62 activates the stress respon-
sive transcription factor Nrf2 through inactivation of Keap1. Nat Cell Biol 12(3):213–223
169. Lau A et al (2010) A noncanonical mechanism of Nrf2 activation by autophagy deficiency:
direct interaction between Keap1 and p62. Mol Cell Biol 30(13):3275–3285
170. Inami Y et al (2011) Persistent activation of Nrf2 through p62 in hepatocellular carcinoma
cells. J Cell Biol 193(2):275–284
171. Young AR et al (2009) Autophagy mediates the mitotic senescence transition. Genes Dev
23(7):798–803
172. Sinha S, Levine B (2008) The autophagy effector Beclin 1: a novel BH3-only protein.
Oncogene 27(Suppl 1):S137–S148
173. Guertin DA, Sabatini DM (2007) Defining the role of mTOR in cancer. Cancer Cell 12(1):9–22
174. Diaz-Troya S et al (2008) The role of TOR in autophagy regulation from yeast to plants and
mammals. Autophagy 4(7):851–865
175. Maiuri MC et al (2007) BH3-only proteins and BH3 mimetics induce autophagy by competi-
tively disrupting the interaction between Beclin 1 and Bcl-2/Bcl-X(L). Autophagy 3(4):374–376
176. Liu J et al (2011) Beclin1 controls the levels of p53 by regulating the deubiquitination
activity of USP10 and USP13. Cell 147(1):223–234
4 The Role of Autophagy in Drug Resistance 115
177. Wei H et al (2011) Suppression of autophagy by FIP200 deletion inhibits mammary tumori-
genesis. Genes Dev 25(14):1510–1527
178. Zhang HF et al (2009) ATG16L1 T300A polymorphism and Crohn’s disease susceptibility:
evidence from 13,022 cases and 17,532 controls. Hum Genet 125(5–6):627–631
179. Kang C, Avery L (2008) To be or not to be, the level of autophagy is the question: dual roles
of autophagy in the survival response to starvation. Autophagy 4(1):82–84
180. Park MA et al (2008) PERK-dependent regulation of HSP70 expression and the regulation
of autophagy. Autophagy 4(3):364–367
181. Hsu KF et al (2009) Cathepsin L mediates resveratrol-induced autophagy and apoptotic cell
death in cervical cancer cells. Autophagy 5(4):451–460
182. Bhoopathi P et al (2010) Cathepsin B facilitates autophagy-mediated apoptosis in
SPARC overexpressed primitive neuroectodermal tumor cells. Cell Death Differ
17(10):1529–1539
183. Mathieu V et al (2007) Galectin-1 knockdown increases sensitivity to temozolomide in a
B16F10 mouse metastatic melanoma model. J Invest Dermatol 127(10):2399–2410
184. Debnath J, Baehrecke EH, Kroemer G (2005) Does autophagy contribute to cell death?
Autophagy 1(2):66–74
185. Lu Z et al (2008) The tumor suppressor gene ARHI regulates autophagy and tumor dor-
mancy in human ovarian cancer cells. J Clin Invest 118(12):3917–3929
186. Lum JJ et al (2005) Growth factor regulation of autophagy and cell survival in the absence
of apoptosis. Cell 120(2):237–248
187. O’Neill PM et al (1998) 4-Aminoquinolines–past, present, and future: a chemical perspec-
tive. Pharmacol Ther 77(1):29–58
188. Kremer JM (2001) Rational use of new and existing disease-modifying agents in rheuma-
toid arthritis. Ann Intern Med 134(8):695–706
189. Romanelli F, Smith KM, Hoven AD (2004) Chloroquine and hydroxychloroquine as inhibi-
tors of human immunodeficiency virus (HIV-1) activity. Curr Pharm Des 10(21):2643–2648
190. Sotelo J, Briceno E, Lopez-Gonzalez MA (2006) Adding chloroquine to conventional treat-
ment for glioblastoma multiforme: a randomized, double-blind, placebo-controlled trial.
Ann Intern Med 144(5):337–343
191. Carmichael SJ, Charles B, Tett SE (2003) Population pharmacokinetics of hydroxychloro-
quine in patients with rheumatoid arthritis. Ther Drug Monit 25(6):671–681
192. Rosenfeld MRGS, Brem S, Mikkelson T, Wang D, Piao S, Davis L, O’Dwyer PJ,
Amaravadi RK (2010) Pharmacokinetic analysis and pharmacodynamic evidence of
autophagy inhibition in patients with newly diagnosed glioblastoma treated on a phase I trial
of hydroxychloroquine in combination with adjuvant temozolomide and radiation (ABTC
0603). J Clin Oncol 28(15s):3086
193. Petiot A et al (2000) Distinct classes of phosphatidylinositol 3’-kinases are involved in sign-
aling pathways that control macroautophagy in HT-29 cells. J Biol Chem 275(2):992–998
194. Wu YT et al (2010) Dual role of 3-methyladenine in modulation of autophagy via different
temporal patterns of inhibition on class I and III phosphoinositide 3-kinase. J Biol Chem
285(14):10850–10861
195. Werner G et al (1984) Metabolic products of microorganisms. 224. Bafilomycins, a new
group of macrolide antibiotics. Production, isolation, chemical structure and biological
activity. J Antibiot (Tokyo) 37(2):110–117
196. Dassonneville L, Bailly C (1999) Stimulation of topoisomerase II-mediated DNA cleavage
by an indazole analogue of lucanthone. Biochem Pharmacol 58(8):1307–1312
197. Luo M, Kelley MR (2004) Inhibition of the human apurinic/apyrimidinic endonuclease
(APE1) repair activity and sensitization of breast cancer cells to DNA alkylating agents with
lucanthone. Anticancer Res 24(4):2127–2134
198. Carew JS et al (2011) Lucanthone is a novel inhibitor of autophagy that induces cathepsin
D-mediated apoptosis. J Biol Chem 286(8):6602–6613
199. Leu JI et al (2011) HSP70 inhibition by the small-molecule 2-phenylethynesulfonamide
impairs protein clearance pathways in tumor cells. Mol Cancer Res 9(7):936–947
116 R. Rangwala and R. Amaravadi
200. Nice DC et al (2002) Cooperative binding of the cytoplasm to vacuole targeting pathway
proteins, Cvt13 and Cvt20, to phosphatidylinositol 3-phosphate at the pre-autophagosomal
structure is required for selective autophagy. J Biol Chem 277(33):30198–30207
201. Guan J et al (2001) Cvt18/Gsa12 is required for cytoplasm-to-vacuole transport, pex-
ophagy, and autophagy in Saccharomyces cerevisiae and Pichia pastoris. Mol Biol Cell
12(12):3821–3838
202. Stromhaug PE et al (2004) Atg21 is a phosphoinositide binding protein required for effi-
cient lipidation and localization of Atg8 during uptake of aminopeptidase I by selective
autophagy. Mol Biol Cell 15(8):3553–3566
203. Tanida I et al (2006) Atg8L/Apg8L is the fourth mammalian modifier of mammalian Atg8
conjugation mediated by human Atg4B, Atg7 and Atg3. FEBS J 273(11):2553–2562
204. Li M et al (2011) Kinetics comparisons of mammalian Atg4 homologues indicate selective
preferences toward diverse Atg8 substrates. J Biol Chem 286(9):7327–7338
205. Mizushima N et al (1998) A protein conjugation system essential for autophagy. Nature
395(6700):395–398
206. Tanida I et al (1999) Apg7p/Cvt2p: A novel protein-activating enzyme essential for
autophagy. Mol Biol Cell 10(5):1367–1379
207. Mizushima N, Noda T, Ohsumi Y (1999) Apg16p is required for the function of the
Apg12p-Apg5p conjugate in the yeast autophagy pathway. EMBO J 18(14):3888–3896
208. Lynch-Day MA et al (2010) Trs85 directs a Ypt1 GEF, TRAPPIII, to the phagophore to pro-
mote autophagy. Proc Natl Acad Sci USA 107(17):7811–7816
209. Balderhaar HJ et al (2010) The Rab GTPase Ypt7 is linked to retromer-mediated receptor
recycling and fusion at the yeast late endosome. J Cell Sci 123(Pt 23):4085–4094
210. Degtyarev M et al (2008) Akt inhibition promotes autophagy and sensitizes PTEN-null
tumors to lysosomotropic agents. J Cell Biol 183(1):101–116
211. Bellodi C et al (2009) Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell
death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin
Invest 119(5):1109–1123
212. Gupta A et al (2010) Autophagy inhibition and antimalarials promote cell death in gastroin-
testinal stromal tumor (GIST). Proc Natl Acad Sci USA 107(32):14333–14338
213. Ding WX et al (2009) Oncogenic transformation confers a selective susceptibility to the
combined suppression of the proteasome and autophagy. Mol Cancer Ther 8(7):2036–2045
214. Rouschop KM et al (2009) Autophagy is required during cycling hypoxia to lower produc-
tion of reactive oxygen species. Radiother Oncol 92(3):411–416
215. Fan QW et al (2010) Akt and autophagy cooperate to promote survival of drug-resistant gli-
oma. Sci Signal 3(147):81
216. Pan Y et al (2011) Targeting autophagy augments in vitro and in vivo antimyeloma activity
of DNA-damaging chemotherapy. Clin Cancer Res 17(10):3248–3258
217. Sheen JH et al (2011) Defective regulation of autophagy upon leucine deprivation
reveals a targetable liability of human melanoma cells in vitro and in vivo. Cancer Cell
19(5):613–628
Chapter 5
MicroRNAs in Cell Death and Cancer
5.1 Introduction
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 117
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_5,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
118 J. K. Park and T. D. Schmittgen
Genome location analysis of miRNAs has suggested that over 70 % of miRNAs are
transcribed from intronic regions of both protein-coding and long non-protein-cod-
ing genes [13, 14]. Expression of these miRNAs shows the remarkable coincidence
with the expression of their host gene transcripts. Otherwise, miRNAs are located in
intergenic regions and apparently possess independent transcription units. Long pri-
mary transcripts containing multiple miRNAs are commonly located as clusters of
polycistronic units. The first step of miRNA biogenesis is the transcription of the nas-
cent primary miRNA precursors (pri-miRNAs), which is mainly mediated by RNA
polymerase II [15] (Fig. 5.1). The pri-miRNAs have a hairpin structure containing
the mature miRNA sequences. Pri-miRNAs also have a 5’ cap structure and poly-
A tails, similar to mRNAs [16]. Binding of DiGeorge syndrome critical region gene
8 (DGCR8) to the pri-miRNAs recruits the class 2 RNase III enzyme, Drosha, fol-
lowing the formation of a multi-protein complex called the microprocessor [17, 18].
Co-factors including the DEAD box RNA helicases p68 (DDX5), p72 (DDX17), and
heterogeneous nuclear ribonucleoproteins (hnRNPs) also compose the microproces-
sor complex [18]. DGCR8 contains two double-stranded RNA-binding domains
(dsRBDs) and is able to bind to a single-stranded portion of the pri-miRNAs for
the appropriate processing [19]. Cleavage of the pri-miRNAs by Drosha produces
~60–70-nt precursor miRNAs (pre-miRNAs) (Fig. 5.1). Following the initial cleav-
age by Drosha, the pre-miRNAs are then recognized by the nuclear export protein,
Exportin-5, and actively transported to the cytoplasm in a Ran-GTP-dependent man-
ner [20]. Recently, it was proposed that other factors such as nuclear export recep-
tor Exportin 1 (XPO1), cap-binding complex (CBC), and arsenite resistance protein 2
(ARS2) act a part of the pri-miRNAs processing [21, 22]. Once inside the cytoplasm,
the endoribonuclease Dicer cleaves the terminal loop of the pre-miRNA. A part-
ner protein, HIV-1 transactivating response (TAR) RNA-binding protein (TRBP) in
humans (R2D2 and loquacious (Loqs), in Drosophila), assists the cleavage resulting in
22–23-nt mature miRNA duplexes (miRNA/miRNA*) (Fig. 5.1) [23–25]. Following
Dicer processing, the guide strand is preferentially incorporated into miRNA-induced
silencing complex (miRISC) and guides Argonaute (AGO) proteins to target genes
(Fig. 5.1). miRNA*, the passenger strand, is commonly degraded [26, 27].
5.3 miRNA Decay
PNAPII PNAPII
PNAPII
PNAPII
pri-miRNA
m7G AAAA
m7G AAAA
DGCR8
Drosha
pre-miRNA
Exportin-5
AGO
AGO
TRBP
Dicer miRNA/miRNA* duplex
miRISC
m7G polyA
3 UTR of a target gene
from several hours to days in cell lines and organs [28]. Since miRNAs can act
as on and off switches and play critical roles in developmental transitions, it is
required that expression of miRNAs is actively regulated in cells. In mouse reti-
nal neurons, turnover of several miRNAs due to rapid decay can be prevented by
blocking the action potential or glutamate receptors. This suggests that turnover-
mediated transition of miRNA expression plays an important role for neuronal
function [29]. miR-29b, which is primarily confined in the nucleus, is an exam-
ple of sequence-specific decomposition of miRNA [30]. miR-29b rapidly decays
due to the uracils at position 9–11. Uracil-rich sequence affects the dynamics
of miRNA decay depending on the sequence context [31]. m169, a transcript of
murine cytomegalovirus (MCMV), inhibits the function of miR-27 through deg-
radation. Its expression kinetics correlate with miR-27 degradation which implies
120 J. K. Park and T. D. Schmittgen
and the cumulation of target genes in P-bodies. For example, cationic amino acid
transporter-1 (CAT-1) mRNA is localized to P-bodies when it is repressed by
miR-122. Upon stress, the interaction of CAT1 with HuR results in the release
of CAT-1 from P-bodies followed by re-initiation of translation activation [38].
Disappearance of P-bodies in developing mouse oocytes shows the coincidence
with global suppression of miRNA functions even though there are a plentiful
amount of miRNA [39, 40].
In addition to the inhibitory function of protein synthesis, miRNA can have
a decoy activity. Posttranscriptional CCAAT/enhancer-binding protein (C/EBP),
alpha (CEBPα) suppression is mediated by interaction with a heterogeneous ribo-
nucleoprotein hnRNP-E2 (also known as poly(rC) binding protein 2). miR-328
interacts with hnRNP-E2 independently of seed sequence, resulting in the release
of CEBPα mRNA from the translation inhibitory effect of hnRNP-E2 [41].
Furthermore, miRNAs can also paradoxically activate the target mRNA’s transla-
tion. Under serum deprivation conditions, both AGO and fragile X mental retarda-
tion–related protein 1 (FXR1) are assembled into an AU-rich element (ARE) in
tumor necrosis factor alpha (TNFα) mRNA, which is mediated by base-pairing of
miR-369-3 with ARE to activate translation [42]. In addition, let-7 and the syn-
thetic miR-cxcr4 repress the translation of their target genes in proliferating cells,
but increase the translation of target genes in serum starved cells, all of which
propose that miRNA’s functions can be shifted depending on cell proliferation
status [42].
5.5 miRNAs in Cancer
It is generally accepted that tumors are initially developed by genetic and epige-
netic alterations of protein-coding oncogenes and tumor suppressors. On account
of structured endeavors to identify additional alterations contributing to human
malignancy, the importance of miRNAs has been realized in the pathogenesis of
cancer over the past several years. Extensive profiling studies have shown that
various miRNAs are differentially expressed in distinct types of human cancer
[43–50]. Furthermore, comprehensive repression of miRNA biogenesis by target-
ing DGCR8, Drosha, or Dicer1 enhances cellular transformation and tumorigen-
esis, which distinctly signifies the role of miRNAs in cancer-related signaling
pathways [51]. Deregulation of miRNAs can result from various causes such as
genomic alterations (i.e., regional gain and loss at chromosomal loci), modification
of transcription factors, epigenetic mechanisms, and abnormal processing of miR-
NAs. Undoubtedly, a number of miRNAs have been identified to have a promising
role in the pathophysiology of many types of tumors. Overexpressed miRNAs in
cancer can serve as oncogenes by targeting protein-coding tumor suppressor genes.
On the contrary, miRNAs have been suggested as tumor suppressors based on their
reduced expression, deletion status, and interaction with protein-coding oncogenes
(Tables 5.1 and 5.2).
122 J. K. Park and T. D. Schmittgen
Table 5.1 Tumor-suppressive miRNAs
microRNA Expression pattern Identified targets Regulation factors References
miR-15a/16-1 Downregulated in BCL2, MCL1, Deletion [52–55, 57–59]
prostate cancer, CCND1,
pituitary WNT3A,
adenomas, multi- FGF2, FGFR1,
ple myeloma, MAP3KIP3
and CLL
miR-26a-1, miR- Reduced in EZH2, CDK6, Transcribed by [60, 63, 65–68]
26a-2 HCC and CCNE1, C/EBP alpha
nasopharyngeal CCNE2,
carcinoma CCND2,
BDNF, GSK3B
miR-34a, miR- Loss in breast, lung, CCND1, CCNE2, Deletion, hyper- [61, 69, 70]
34b/34c colon, pancreatic CDK4, CDK6, methylation,
cancer, HCC, c-MYC, MET, transactivated
and neuroblas- E2F3, SIRT1, by TP53
toma BCL2, CD44
miR-122 Downregulated in ADAM10, SRF, Controlled by [62, 71–73, 77, 78]
HCC IGF1R, HNF1A,
ADAM17, HNF3A,
CUTL1, HNF3B, and
SMARCD1 HNF4A.
Transcription-
ally activated by
C/EBP alpha
miR-143/145 Downregulated in DNMT3A, Repressed by Ras [79, 80, 82–85, 87,
colon, lung, RREB1, MLL– activation. 88]
cervical, and AF4, ERK5, Epigenetically
prostate cancer OCT4, SOX2, repressed in
KLF4, EGFR, ALL
NUDT1,
c-MYC,
MMP11,
ADAM17,
CCNA2
5.5.1 Tumor-Suppressive miRNAs
Association of miRNAs with human cancer was first suggested in 2002. Both miR-
15a and miR-16-1 are located on chromosome 13q14 and are frequently deleted
or downregulated in chronic lymphocytic leukemia (CLL) [52]. Expression of
both miRNAs is also downregulated in pituitary adenomas, prostate carcinoma,
multiple myeloma, and diffuse B-cell lymphomas [53–55]. The tumor-suppres-
sive function of these two miRNAs is corroborated by their specific target genes.
Anti-apoptotic proteins, B-cell lymphoma 2 (BCL2) and myeloid cell leukemia-1
(MCL1) are critical cellular oncogenes which determine proliferation, differentia-
tion, and tumorigenesis [56]. BCL2 expression is inversely proportional to miR-15a
5 MicroRNAs in Cell Death and Cancer 123
Table 5.2 Oncogenic miRNAs
microRNA Expression pattern Identified targets Regulation factors References
hsa-miR-21 Upregulated in PDCD4, TPM1, Induced by [44, 90, 91, 100,
most cancer SPRY2, Ras/ERK, 101]
TIMP3, RECK, AP-1, NFκB,
MASPIN, deltaEF1,
PTEN and STAT3.
Suppressed by
FOXO3A and
NFIB
miR-221/222 Overexpressed in CDKN1B, Induced by NFκB, [102, 103,
lung, breast, CDKN1C, c-Jun, and 105–108, 110,
prostate cancer, PTEN, TIMP3, FOSL1. Nega- 111]
and HCC TRPS1, PUMA tively regulated
by ER in breast
cancer. Modu-
lated by EGFR
and MET
miR-17-92 Upregulated in ZBTB4, E2F1, Transactivated by [113–119]
colon, lung, PTEN, E2F, MYCN,
breast cancer, BCL2L11, and c-Myc.
and B-cell CDKN1A, Negatively
lymphomas DKK3, regulated by
TGFBR2, p53 under
SMAD4, TSP1, hypoxic
CTGF condition
miR-106b-25 Increased in CDKN1A, Activated by E2F1 [120–125]
pancreas, BCL2L11, and E2F3
prostate, colon ITGB8, PTEN,
cancer, and E2F1, E2F3,
HCC BCL2L11
miR-130/301 Upregulated in HOXA5, GAX, Transcribed by [126–131]
breast, colon, TP53INP1, NFκB
and pancreatic RUNX3,
cancer CDKN1A,
FOXF2,
BBC3, PTEN,
COL2A1,
NKRF
miR-155 Upregulated in SHIP1, TP53INP1, Epigenetically [132–142]
thyroid, breast, HGAL, repressed
colon, cervical, SOCS1, by BRCA1.
lung cancer, PPP2CA, Transcription-
and PDAC FOXO3A ally activated
by NFκB and
MYB
and miR-16-1 in CLL, and both miRNAs post transcriptionally repress BCL2
[57]. Transcriptome and proteome analysis in MEG-01 cells showed that ectopic
expression of miR-15a and miR-16-1 results in upregulation or downregulation
124 J. K. Park and T. D. Schmittgen
5.5.2 Oncogenic miRNAs
One of the first miRNAs identified as oncogenic in the human genome is miR-21.
miR-21 was first identified in glioblastoma [90] and is upregulated in most
types of human malignancies [44]. A term “oncomiR addiction” markedly
describes the momentous function of miR-21 on all stages of tumor development.
Overexpression of miR-21 in a genetically engineered mouse model facilitates
a pre-B malignant lymphoid-like phenotype [91]. Switching off miR-21 expres-
sion leads to tumor regression and recovery from symptoms of lymphoma, clearly
illustrating the role of miR-21 in tumor initiation as well as maintenance of estab-
lished tumors. Furthermore, a survival advantage followed by the suppression of
miR-21 also demonstrates the clinical impact of targeting one aberrantly expressed
miRNA [91]. Posttranscriptional regulation of miR-21 is mediated by the receptor-
mediated SMADs (R-SMADs). Interactions of R-SMADs with primary transcripts
of miR-21 (pri-miR-21) promote Drosha processing by stabilizing the binding
of pri-miR-21 with Drosha. TGF-β and BMP4 enhance the expression of mature
miR-21 through the complex formation of R-SMADs, pri-miR-21, and DEAD
(Asp-Glu-Ala-Asp) box polypeptide 5 (DDX5, RNA helicase, p68, a component
of microprocessor complex) [92]. Moreover, miR-21 is located at a TMEM49
126 J. K. Park and T. D. Schmittgen
intronic region, and several pathways modulate the expression of miR-21 at the
transcription level. Akt2 transcriptionally activates miR-21 induction through
binding of NFκB, CREB, and CBP/p300 to the miR-21 promoter regions under
conditions of hypoxia [93]. AP-1 and deltaEF1 increase miR-21 expression by
binding to the miR-21 promoter. Treatment of BMP-6 transcriptionally inhibits
miR-21 expression via suppression of AP-1 and deltaEF1 in breast tumor cells
[94]. In addition, signal transducer and activator of transcription 3 (STAT3) also
directly binds to the miR-21 promoter and regulates miR-21 levels [94]. Interferon
(IFN) increases miR-21 through co-regulation of STAT3 and NFκB. Apoptosis
induced by IFN can be suppressed due to an increased amount of mature miR-21
[95]. Enhanced expression of miR-21 resulted from activation of EGFR signaling
and neurotensin treatment in lung cancer and colon tumors, respectively [96, 97].
In contrast, suppression of miR-21 promoter activity is induced by the binding
of FOXO3A to its promoter region. FOXO3A is a trigger of apoptosis of tumor
cells via upregulation of several pro-apoptotic genes such as Bim and PUMA. Fas
ligand (FasL) is a target of miR-21, and upregulation of FasL by FOXO3A partly
results from miR-21 depletion [98]. Nuclear factor I/B (NFIB) also binds to the
miR-21 promoter and acts as a negative regulator of miR-21 transcription [99].
The potential oncogenic activity of miR-21 has been further supported by numer-
ous validated target genes (reviewed in [100] and summarized in Table 5.2). All of
these are closely related with growth, apoptosis, invasion, and metastasis of tumor
cells. Overexpression of miR-21 therefore leads to facilitation of growth, evasion
of apoptosis, potent metastasis, and resistance toward anticancer agents [100, 101].
Deregulation of other oncogenic miRNAs including miR-221 and miR-222
has also been observed in human malignancies. miR-221 and miR-222 are clus-
tered in an intergenic region on chromosome Xp11.3. They have the same seed
sequence, thus share common predicted target genes. They are upregulated in
lung, breast, prostate cancer, and HCC [102–104]. Contribution of the miR-
221/222 cluster to tumorigenesis of lung cancer and HCC has been suggested.
Expression of miR-221/222 is modulated by EGFR and MET receptors in lung
cancer, and they contribute to gefitinib resistance and tumorigenesis of non-small-
cell lung cancer (NSCLC). Overexpression of miR-221 facilitates the growth of
tumorigenic murine hepatic progenitor cells in vivo, suggesting the important role
of miR-221 in hepatocarcinogenesis [105]. Oncogenic activities of miR-221/222
are also related with their transcription modulators. NFκB and c-Jun contribute to
the oncogenesis of prostate carcinoma and glioblastoma and trigger the expression
of miR-221/222 [106]. A downstream transcription factor of EMT-promoting Ras-
ERK pathway, FOSL1, stimulates the expression of miR-221/222 in breast cancer.
FOSL1 is present in basal-like (BL) sub-type breast cancers; therefore, upregula-
tion of miR-221/222 by FOSL1 in breast cancer contributes to aggressiveness of
BL sub-type partly by targeting tricho-rhino-phalangeal syndrome type 1 (TRPS1)
which represses the GATA family at the transcriptional level and decreases ZEB2
expression [107]. miR-221/222 are negatively modulated by the binding of ER
alpha together with nuclear receptor co-repressor (NCoR) and silencing media-
tor of retinoic acid and thyroid hormone receptor (SMRT) [108]. A number of
5 MicroRNAs in Cell Death and Cancer 127
References
4. Carthew RW, Sontheimer EJ (2009) Origins and mechanisms of miRNAs and siRNAs. Cell
136(4):642–655
5. Malone CD, Hannon GJ (2009) Small RNAs as guardians of the genome. Cell
136(4):656–668
6. Ambros V (2004) The functions of animal microRNAs. Nature 431(7006):350–355
7. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes
small RNAs with antisense complementarity to lin-14. Cell 75(5):843–854
8. Wightman B, Ha I, Ruvkun G (1993) Posttranscriptional regulation of the heterochronic
gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 75(5):855–862
9. Wiemer EA (2007) The role of microRNAs in cancer: no small matter. Eur J Cancer
43(10):1529–1544
10. Zhao Y, Samal E, Srivastava D (2005) Serum response factor regulates a muscle-specific
microRNA that targets Hand2 during cardiogenesis. Nature 436(7048):214–220
11. Schaefer A, O’Carroll D, Tan CL, Hillman D, Sugimori M, Llinas R, Greengard P (2007)
Cerebellar neurodegeneration in the absence of microRNAs. J Exp Med 204(7):1553–1558
12. Taganov KD, Boldin MP, Chang KJ, Baltimore D (2006) NF-kappaB-dependent induc-
tion of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune
responses. Proc Nat Acad Sci USA 103(33):12481–12486
13. Rodriguez A, Griffiths-Jones S, Ashurst JL, Bradley A (2004) Identification of mammalian
microRNA host genes and transcription units. Genome Res 14(10A):1902–1910
14. Berezikov E, Chung WJ, Willis J, Cuppen E, Lai EC (2007) Mammalian mirtron genes. Mol
Cell 28(2):328–336
15. Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, Kim VN (2004) MicroRNA genes are
transcribed by RNA polymerase II. EMBO J 23(20):4051–4060
16. Cai X, Hagedorn CH, Cullen BR (2004) Human microRNAs are processed from capped,
polyadenylated transcripts that can also function as mRNAs. RNA 10(12):1957–1966
17. Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ (2004) Processing of primary
microRNAs by the microprocessor complex. Nature 432(7014):231–235
18. Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, Shiekhattar
R (2004) The microprocessor complex mediates the genesis of microRNAs. Nature
432(7014):235–240
19. Senturia R, Faller M, Yin S, Loo JA, Cascio D, Sawaya MR, Hwang D, Clubb RT, Guo F
(2010) Structure of the dimerization domain of DiGeorge critical region 8. Protein Sci publ
Protein Soc 19(7):1354–1365
20. Yi R, Qin Y, Macara IG, Cullen BR (2003) Exportin-5 mediates the nuclear export of pre-
microRNAs and short hairpin RNAs. Genes Dev 17(24):3011–3016
21. Bussing I, Yang JS, Lai EC, Grosshans H (2010) The nuclear export receptor XPO-1 sup-
ports primary miRNA processing in C. elegans and Drosophila. EMBO J 29(11):1830–1839
22. Gruber JJ, Zatechka DS, Sabin LR, Yong J, Lum JJ, Kong M, Zong WX, Zhang Z, Lau CK,
Rawlings J, Cherry S, Ihle JN, Dreyfuss G, Thompson CB (2009) Ars2 links the nuclear
cap-binding complex to RNA interference and cell proliferation. Cell 138(2):328–339
23. Haase AD, Jaskiewicz L, Zhang H, Laine S, Sack R, Gatignol A, Filipowicz W (2005)
TRBP, a regulator of cellular PKR and HIV-1 virus expression, interacts with Dicer and
functions in RNA silencing. EMBO Rep 6(10):961–967
24. Tsutsumi A, Kawamata T, Izumi N, Seitz H, Tomari Y (2011) Recognition of the pre-
miRNA structure by Drosophila dicer-1. Nat Struct Mol Biol 18(10):1153–1158
25. Saito K, Ishizuka A, Siomi H, Siomi MC (2005) Processing of pre-microRNAs by the dicer-
1-loquacious complex in Drosophila cells. PLoS Biol 3(7):e235
26. Matranga C, Tomari Y, Shin C, Bartel DP, Zamore PD (2005) Passenger-strand cleav-
age facilitates assembly of siRNA into Ago2-containing RNAi enzyme complexes. Cell
123(4):607–620
27. Okamura K, Ishizuka A, Siomi H, Siomi MC (2004) Distinct roles for argonaute proteins in
small RNA-directed RNA cleavage pathways. Genes Dev 18(14):1655–1666
130 J. K. Park and T. D. Schmittgen
28. Gantier MP, McCoy CE, Rusinova I, Saulep D, Wang D, Xu D, Irving AT, Behlke MA,
Hertzog PJ, Mackay F, Williams BR (2011) Analysis of microRNA turnover in mammalian
cells following dicer1 ablation. Nucleic Acids Res 39(13):5692–5703
29. Krol J, Busskamp V, Markiewicz I, Stadler MB, Ribi S, Richter J, Duebel J, Bicker S,
Fehling HJ, Schubeler D, Oertner TG, Schratt G, Bibel M, Roska B, Filipowicz W (2010)
Characterizing light-regulated retinal microRNAs reveals rapid turnover as a common prop-
erty of neuronal microRNAs. Cell 141(4):618–631
30. Hwang HW, Wentzel EA, Mendell JT (2007) A hexanucleotide element directs microRNA
nuclear import. Science 315(5808):97–100
31. Zhang Z, Zou J, Wang GK, Zhang JT, Huang S, Qin YW, Jing Q (2011) Uracils at nucleo-
tide position 9–11 are required for the rapid turnover of miR-29 family. Nucleic Acids Res
39(10):4387–4395
32. Libri V, Helwak A, Miesen P, Santhakumar D, Borger JG, Kudla G, Grey F, Tollervey D,
Buck AH (2012) Murine cytomegalovirus encodes a miR-27 inhibitor disguised as a target.
Proc Nat Acad Sci USA 109(1):279–284
33. Katoh T, Sakaguchi Y, Miyauchi K, Suzuki T, Kashiwabara S, Baba T, Suzuki T (2009)
Selective stabilization of mammalian microRNAs by 3’ adenylation mediated by the cyto-
plasmic poly(A) polymerase GLD-2. Genes Dev 23(4):433–438
34. Chatterjee S, Fasler M, Bussing I, Grosshans H (2011) Target-mediated protection of endog-
enous microRNAs in C. elegans. Dev Cell 20(3):388–396
35. Hammell CM (2008) The microRNA-argonaute complex: a platform for mRNA modula-
tion. RNA Biol 5(3):123–127
36. Wang Y, Li Y, Ma Z, Yang W, Ai C (2010) Mechanism of microRNA-target interac-
tion: molecular dynamics simulations and thermodynamics analysis. PLoS Comput Biol
6(7):e1000866
37. Liu J, Valencia-Sanchez MA, Hannon GJ, Parker R (2005) MicroRNA-dependent localiza-
tion of targeted mRNAs to mammalian P-bodies. Nat Cell Biol 7(7):719–723
38. Bhattacharyya SN, Habermacher R, Martine U, Closs EI, Filipowicz W (2006) Stress-
induced reversal of microRNA repression and mRNA P-body localization in human cells.
Cold Spring Harb Symp Quant Biol 71:513–521
39. Ma J, Flemr M, Stein P, Berninger P, Malik R, Zavolan M, Svoboda P, Schultz RM (2010)
MicroRNA activity is suppressed in mouse oocytes. Current Biol CB 20(3):265–270
40. Suh N, Baehner L, Moltzahn F, Melton C, Shenoy A, Chen J, Blelloch R (2010) MicroRNA
function is globally suppressed in mouse oocytes and early embryos. Current Biol CB
20(3):271–277
41. Eiring AM, Harb JG, Neviani P, Garton C, Oaks JJ, Spizzo R, Liu S, Schwind S, Santhanam
R, Hickey CJ, Becker H, Chandler JC, Andino R, Cortes J, Hokland P, Huettner CS, Bhatia
R, Roy DC, Liebhaber SA, Caligiuri MA, Marcucci G, Garzon R, Croce CM, Calin GA,
Perrotti D (2010) miR-328 functions as an RNA decoy to modulate hnRNP E2 regulation of
mRNA translation in leukemic blasts. Cell 140(5):652–665
42. Vasudevan S, Tong Y, Steitz JA (2007) Switching from repression to activation: microRNAs
can up-regulate translation. Science 318(5858):1931–1934
43. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL,
Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR (2005) MicroRNA
expression profiles classify human cancers. Nature 435(7043):834–838
44. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C,
Ferracin M, Prueitt RL, Yanaihara N, Lanza G, Scarpa A, Vecchione A, Negrini M, Harris
CC, Croce CM (2006) A microRNA expression signature of human solid tumors defines
cancer gene targets. Proc Nat Acad Sci USA 103(7):2257–2261
45. Budhu A, Jia HL, Forgues M, Liu CG, Goldstein D, Lam A, Zanetti KA, Ye QH, Qin LX,
Croce CM, Tang ZY, Wang XW (2008) Identification of metastasis-related microRNAs in
hepatocellular carcinoma. Hepatology 47(3):897–907
5 MicroRNAs in Cell Death and Cancer 131
61. Tryndyak VP, Ross SA, Beland FA, Pogribny IP (2009) Down-regulation of the microR-
NAs miR-34a, miR-127, and miR-200b in rat liver during hepatocarcinogenesis induced by
a methyl-deficient diet. Mol Carcinog 48(6):479–487
62. Tsai WC, Hsu PW, Lai TC, Chau GY, Lin CW, Chen CM, Lin CD, Liao YL, Wang JL, Chau
YP, Hsu MT, Hsiao M, Huang HD, Tsou AP (2009) MicroRNA-122, a tumor suppressor
microRNA that regulates intrahepatic metastasis of hepatocellular carcinoma. Hepatology
49(5):1571–1582
63. Mohamed JS, Lopez MA, Boriek AM (2010) Mechanical stretch up-regulates microRNA-
26a and induces human airway smooth muscle hypertrophy by suppressing glycogen syn-
thase kinase-3beta. J Biol Chem 285(38):29336–29347
64. Harris TE, Albrecht JH, Nakanishi M, Darlington GJ (2001) CCAAT/enhancer-binding
protein-alpha cooperates with p21 to inhibit cyclin-dependent kinase-2 activity and induces
growth arrest independent of DNA binding. J Biol Chem 276(31):29200–29209
65. Kota J, Chivukula RR, O’Donnell KA, Wentzel EA, Montgomery CL, Hwang HW, Chang
TC, Vivekanandan P, Torbenson M, Clark KR, Mendell JR, Mendell JT (2009) Therapeutic
microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell
137(6):1005–1017
66. Zhu Y, Lu Y, Zhang Q, Liu JJ, Li TJ, Yang JR, Zeng C, Zhuang SM (2011) MicroRNA-
26a/b and their host genes cooperate to inhibit the G1/S transition by activating the pRb
protein. Nucleic Acids Res 40:4615–4625
67. Caputo V, Sinibaldi L, Fiorentino A, Parisi C, Catalanotto C, Pasini A, Cogoni C, Pizzuti A
(2011) Brain derived neurotrophic factor (BDNF) expression is regulated by microRNAs
miR-26a and miR-26b allele-specific binding. PLoS ONE 6(12):e28656
68. Lu J, He ML, Wang L, Chen Y, Liu X, Dong Q, Chen YC, Peng Y, Yao KT, Kung HF, Li
XP (2011) MiR-26a inhibits cell growth and tumorigenesis of nasopharyngeal carcinoma
through repression of EZH2. Cancer Res 71(1):225–233
69. Hermeking H (2010) The miR-34 family in cancer and apoptosis. Cell Death Differ
17(2):193–199
70. Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C,
Honorio S, Wiggins JF, Bader AG, Fagin R, Brown D, Tang DG (2011) The microRNA
miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat
Med 17(2):211–215
71. Zeng C, Wang R, Li D, Lin XJ, Wei QK, Yuan Y, Wang Q, Chen W, Zhuang SM (2010)
A novel GSK-3 beta-C/EBP alpha-miR-122-insulin-like growth factor 1 receptor regulatory
circuitry in human hepatocellular carcinoma. Hepatology 52(5):1702–1712
72. Xu H, He JH, Xiao ZD, Zhang QQ, Chen YQ, Zhou H, Qu LH (2010) Liver-enriched tran-
scription factors regulate microRNA-122 that targets CUTL1 during liver development.
Hepatology 52(4):1431–1442
73. Gatfield D, Le Martelot G, Vejnar CE, Gerlach D, Schaad O, Fleury-Olela F, Ruskeepaa AL,
Oresic M, Esau CC, Zdobnov EM, Schibler U (2009) Integration of microRNA miR-122 in
hepatic circadian gene expression. Genes Dev 23(11):1313–1326
74. Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow M, Munk ME, Kauppinen
S, Orum H (2010) Therapeutic silencing of microRNA-122 in primates with chronic hepati-
tis C virus infection. Science 327(5962):198–201
75. Esau C, Davis S, Murray SF, Yu XX, Pandey SK, Pear M, Watts L, Booten SL, Graham M,
McKay R, Subramaniam A, Propp S, Lollo BA, Freier S, Bennett CF, Bhanot S, Monia BP
(2006) miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell
Metab 3(2):87–98
76. Jopling C (2012) Liver-specific microRNA-122: biogenesis and function. RNA Biol
9(2):137–142
77. Bai S, Nasser MW, Wang B, Hsu SH, Datta J, Kutay H, Yadav A, Nuovo G, Kumar P,
Ghoshal K (2009) MicroRNA-122 inhibits tumorigenic properties of hepatocellular carci-
noma cells and sensitizes these cells to sorafenib. J Biol Chem 284(46):32015–32027
5 MicroRNAs in Cell Death and Cancer 133
131. Shi W, Gerster K, Alajez NM, Tsang J, Waldron L, Pintilie M, Hui AB, Sykes J, P’ng C,
Miller N, McCready D, Fyles A, Liu FF (2011) MicroRNA-301 mediates proliferation and
invasion in human breast cancer. Cancer Res 71(8):2926–2937
132. Kluiver J, Poppema S, de Jong D, Blokzijl T, Harms G, Jacobs S, Kroesen BJ, van den Berg
A (2005) BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and dif-
fuse large B cell lymphomas. J Pathol 207(2):243–249
133. Costinean S, Zanesi N, Pekarsky Y, Tili E, Volinia S, Heerema N, Croce CM (2006) Pre-B
cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155
transgenic mice. Proc Nat Acad Sci USA 103(18):7024–7029
134. Faraoni I, Antonetti FR, Cardone J, Bonmassar E (2009) miR-155 gene: a typical multifunc-
tional microRNA. Biochim Biophys Acta 1792(6):497–505
135. Gironella M, Seux M, Xie MJ, Cano C, Tomasini R, Gommeaux J, Garcia S, Nowak J,
Yeung ML, Jeang KT, Chaix A, Fazli L, Motoo Y, Wang Q, Rocchi P, Russo A, Gleave
M, Dagorn JC, Iovanna JL, Carrier A, Pebusque MJ, Dusetti NJ (2007) Tumor protein
53-induced nuclear protein 1 expression is repressed by miR-155, and its restoration inhibits
pancreatic tumor development. Proc Nat Acad Sci USA 104(41):16170–16175
136. Kong W, He L, Coppola M, Guo J, Esposito NN, Coppola D, Cheng JQ (2010)
MicroRNA-155 regulates cell survival, growth, and chemosensitivity by targeting FOXO3a
in breast cancer. J Biol Chem 285(23):17869–17879
137. Lee DW, Futami M, Carroll M, Feng Y, Wang Z, Fernandez M, Whichard Z, Chen Y,
Kornblau S, Shpall EJ, Bueso-Ramos CE, Corey SJ (2012) Loss of SHIP-1 protein expres-
sion in high-risk myelodysplastic syndromes is associated with miR-210 and miR-155.
Oncogene 31(37):4085–4094
138. Dagan LN, Jiang X, Bhatt S, Cubedo E, Rajewsky K, Lossos IS (2012) miR-155 regulates
HGAL expression and increases lymphoma cell motility. Blood 119(2):513–520
139. Jiang S, Zhang HW, Lu MH, He XH, Li Y, Gu H, Liu MF, Wang ED (2010) MicroRNA-155
functions as an OncomiR in breast cancer by targeting the suppressor of cytokine signaling
1 gene. Cancer Res 70(8):3119–3127
140. Vargova K, Curik N, Burda P, Basova P, Kulvait V, Pospisil V, Savvulidi F, Kokavec
J, Necas E, Berkova A, Obrtlikova P, Karban J, Mraz M, Pospisilova S, Mayer J, Trneny
M, Zavadil J, Stopka T (2011) MYB transcriptionally regulates the miR-155 host gene in
chronic lymphocytic leukemia. Blood 117(14):3816–3825
141. Wang B, Majumder S, Nuovo G, Kutay H, Volinia S, Patel T, Schmittgen TD, Croce C,
Ghoshal K, Jacob ST (2009) Role of microRNA-155 at early stages of hepatocarcinogen-
esis induced by choline-deficient and amino acid-defined diet in C57BL/6 mice. Hepatology
50(4):1152–1161
142. Chang S, Wang RH, Akagi K, Kim KA, Martin BK, Cavallone L, Kathleen Cuningham
Foundation Consortium for Research into Familial Breast C, Haines DC, Basik M, Mai P,
Poggi E, Isaacs C, Looi LM, Mun KS, Greene MH, Byers SW, Teo SH, Deng CX, Sharan
SK (2011) Tumor suppressor BRCA1 epigenetically controls oncogenic microRNA-155.
Nat Med 17(10):1275–1282
Chapter 6
Targeting DNA Repair Pathways
for Cancer Therapy
Abstract DNA repair pathways maintain the integrity of the genome, reducing the
onset of cancer, disease, and aging. The majority of anticancer therapeutics (radia-
tion and chemotherapy) function as genotoxins, eliciting genomic DNA damage
in an attempt to induce cell death in the tumor. However, cellular DNA repair pro-
teins counteract the effectiveness of these therapeutic genotoxins by repairing and
removing the cell death-inducing DNA lesions, implicating DNA repair proteins
as prime targets for improving response to currently available anticancer regimens.
To trigger a tumor-specific cell death response (with minimal normal cell toxicity),
the level of genomic DNA damage must therefore surpass the DNA repair capac-
ity of the tumor without overwhelming the DNA repair potential of normal tis-
sue. Interestingly, cancer-specific DNA repair defects offer novel approaches for
tumor-selective therapy. This has become highly relevant as it is suggested that
most cancer cells are likely to be defective in some aspect of DNA repair. Herein,
we describe the molecular pathways that participate in the repair of DNA damage
induced by radiation- and chemotherapeutics and discuss strategies that are being
developed to target DNA repair for cancer treatment and highlight key DNA repair
inhibitors that can enhance response. Further, we present novel therapeutic strate-
gies being considered to exploit inherent weaknesses in tumor cells such as defects
in one or more DNA repair pathways or related processes that may provide the
opportunity to selectively increase tumor-specific cell death.
C. Vens
Division of Biological Stress Response, The Netherlands Cancer Institute, NL,
Amsterdam, The Netherlands
R. W. Sobol (*)
Department of Pharmacology and Chemical Biology, University of Pittsburgh Cancer Institute,
Hillman Cancer Center; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
e-mail: [email protected]
R. W. Sobol
Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh,
PA, USA
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 137
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_6,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
138 C. Vens and R. W. Sobol
Fig. 6.1 Schematic representation of cellular DNA damage repair. This figure depicts cellular
repair processes that deal with chemo- and radiotherapy-induced DNA lesions. Prevalent DNA
repair targets for cancer treatment are highlighted in bold
Human cells must repair tens of thousands of DNA lesions per day [1]. If they are
not repaired, these lesions lead to mutations or genome aberrations that threaten cell
survival and genomic integrity. To combat these threats, cells have evolved multiple
DNA repair and DNA damage response (DDR) mechanisms that signal the presence
of lesions and promote their repair or regulate cellular processes in response to the
DNA damage (Fig. 6.1) [2]. Defects in these repair and response pathways can pro-
mote tumorigenesis and, indeed, are common in human cancers [3, 4]. On the other
hand, current therapy options for cancer patients exploit the DNA-damaging proper-
ties of certain drugs and agents. The success of radiation exposure during radiother-
apy and the success of most chemotherapy agents rely on the destructive nature that
these agents have on cellular DNA, ultimately resulting in death and hopefully eradi-
cation of the tumor cells. Hence, DNA damage and repair mechanisms play a crucial
role in determining treatment outcome. On a cellular level, resistance to treatment is
profoundly determined by the capacity of the cancer cell to respond to and repair the
individual DNA lesions that are induced by the chemotherapeutic agents or radiation.
Our increasing knowledge of these processes has led to the development of new
concepts that target and exploit the cancer cell DDR [5]. Counteracting resistance to
chemotherapy by targeting the appropriate DNA repair pathway is a promising strat-
egy in cancer treatment. Another is to exploit the DNA repair and response defects
that are present in cancer cells thereby specifically targeting tumor cells while spar-
ing healthy cells from a high load of unrepaired DNA damage [6, 7]. Together,
these strategies might provide promising avenues in the conquest against cancer.
6 Targeting DNA Repair Pathways for Cancer Therapy 139
Here, we will briefly describe essential cellular DNA repair and response mecha-
nisms and illustrate novel concepts and promising strategies to exploit and target
DNA repair for cancer treatment.
The initial DDR of a cell involves the recognition of the DNA damage followed by
the propagation of a series of signals ranging from alterations in RNA or protein
expression and modification of protein function or stability through p ost-translation
modification, among other signals. The cell’s defense to genotoxic lesions is trig-
gered and accomplished by a series of events that mediate and regulate prolifera-
tion, cell death, or DNA repair crucial to its survival [2, 4]. The initial steps for an
appropriate response require detection of the lesion, signaling of its presence and
promotion of repair. Cells act upon DNA damage not only by promoting and exe-
cuting repair but also respond by halting the cell cycle or by promoting cell death
mechanisms in order to prevent propagation of the damage. The DDR therefore has
an impact on transcription, cellular metabolism, cell cycle regulators, as well as
cell death, via apoptosis and senescence.
One of the most prominent members of the DNA damage signaling pathway
that links DNA damage with cell cycle checkpoints is the protein ataxia telangi-
ectasia-mutated (ATM) [8], a protein kinase that is recruited to DNA double-
strand breaks (DSB) such as those induced by ionizing radiation. The formation
of DSBs triggers the activation of ATM and activated ATM then phosphorylates a
wide range of downstream substrate proteins thereby signaling the presence of the
damage throughout the cell to facilitate repair [9]. Initial activation of ATM is pro-
moted by its autophosphorylation that initiates a signaling cascade of further phos-
phorylation events that constitute the DDR [10]. With excessive unrepaired DNA
damage present, this cellular response can culminate in an apoptotic response in
which p53 is central but not necessarily always required.
Another key DDR signaling component is the ataxia telangiectasia-RAD3-
related (ATR) kinase that gets activated after replication stress-induced DNA
damage. Replication stress, caused, for example, by exposure to hydroxyurea (HU),
results in the formation of large stretches of single-stranded DNA coated with
replication protein A (RPA) that triggers activation of ATR. Similar to processes
in ATM-mediated signaling, ATR signaling is promoted by regulatory proteins.
ATRIP and TopBP1, together with RAD17-mediated 9-1-1 (Rad9-Rad1-Hus1)
clamp loading, “sense” the damage and trigger the activation of ATR.
Further downstream of these initial events, the cell cycle checkpoint-regulating
protein kinases CHK1 and CHK2 are among the most important targets (substrates)
of ATM and ATR. Supported by the activation of p53 and mediated via multiple
paths, this signaling cascade ultimately results in the reduction of cyclin-dependent
kinase activity that drives cell cycle progression. The halt in cell cycle progression is
thought to allow time for repair and, most importantly, if not successfully repaired,
140 C. Vens and R. W. Sobol
to prevent propagation of the DNA damage. Cell cycle checkpoints are in place at
the border from G1 to S, within S and at the G2/M border. The prevalent blocks
and their extent depend on the damaging agent and the number and type of lesion.
Another important downstream target of ATM and ATR is p53, an essential player
in the induction of apoptosis upon DNA damage. Thus, DDR mechanisms have a
crucial role in the protection against genome instability and chemo/radiotherapy
response.
ATM/ATR signaling also enhances repair by recruiting repair factors to the
site of the lesion and activating DNA repair proteins through phosphoryla-
tion or indirectly, by modulating acetylation, ubiquitylation, SUMOylation, or
DNA repair gene transcription. These kinases also influence chromatin structure
through phosphorylation of the histone H2A variant (γH2AX). Thereby, they
facilitate recruitment of DDR factors and expedite DNA repair while amplifying
DSB signaling that is crucial to cellular survival following exposure to DNA-
damaging agents.
The role of the DDR is broad with respect to the type of cancer therapeutic. DDR
activity is involved upon exposure of a whole range of chemotherapeutic agents and
upon radiation. Indeed, DDR and cell cycle blocks are induced by radiation, topoi-
somerase I and II poisons, anthracyclines, alkylating drugs including platinum ana-
logues and antitumor antibiotics. Interference in DDR by the use of inhibitors will
likely affect the response and cellular survival in most cancer treatment options.
One of the first DNA repair proteins to be considered as a viable target for improving
chemotherapy was O6-alkylguanine-DNA alkyltransferase (MGMT or AGT), a pro-
tein encoded by the O6-methylguanine-DNA methyltransferase gene (MGMT) [11].
In depth, discussion on the function of MGMT and its role in cancer and chemother-
apy can be found in many excellent reviews [12–14]. MGMT falls within the cate-
gory of direct-reversal (DR) DNA repair proteins that also include the AlkB family of
proteins [13, 15]. Unlike most other DNA repair pathways that correct lesions by
removing the base containing the lesion [base excision repair (BER), see below],
removing a short oligonucleotide containing the lesion [nucleotide excision repair
(NER), see below] or removing long tracts of DNA (mismatch repair, MMR) fol-
lowed by a DNA synthesis step (repair-directed DNA synthesis), DR proteins such
as MGMT or the AlkB proteins reverse the damage to the DNA base directly, and
the mechanism of repair does not involve a DNA synthesis step. This section will
focus on the role of MGMT in DNA lesion repair and the subsequent role that the
MMR proteins play in the cellular response when MGMT is unable to repair the
O6-alkylguanine lesion. Further discussion on the mechanism of action of AlkB pro-
teins can be found elsewhere [13].
Like many DNA repair proteins, MGMT repairs lesions from both carcinogenic
compounds and from chemotherapeutic agents. As such, MGMT acts to suppress
6 Targeting DNA Repair Pathways for Cancer Therapy 141
analogue O6-benzylguanine [34] (Sect. 6.3.2). Improved prognosis has also been
reported in tumors with loss of MGMT expression due to promoter methylation
[35] whereas poor prognosis is observed when MGMT expression levels are high
or MMR capacity is compromised. Hence, elevated expression of MGMT and/or a
non-functional MMR pathway contribute much of the observed resistance to TMZ
in many tumor cell lines and in clinical trials.
race [39]. Such a process or hand-off mechanism has the advantage of elimi-
nating or avoiding the accumulation of free BER intermediates that are prone
to induce cell death [13]. Once formed by the glycosylase, the resulting aba-
sic site is then handed off to APE1 to be hydrolyzed on the 5′ end. The resulting
single-nucleotide repair gap contains a 3′OH and a 5′deoxyribose-phosphate
(5′dRP) moiety at the margins. It has been suggested that this BER intermediate
(a single-strand break with a 5′dRP moiety) recruits poly(ADP)ribose polymerase
(PARP)1 to the lesion site. Recruitment then triggers activation of PARP1. Activated
PARP1 polymerizes NAD+ to yield the polymer poly (ADP) ribose (PAR), an
essential posttranslational modification. The first protein to be modified by PAR is
PARP1 itself (auto-modification). Subsequently, it has been observed that XRCC1
and many other proteins are modified [40]. Once modified, activated PARP1 then
facilitates chromatin relaxation (likely to provide access to the lesions for repair)
[41, 42] and recruitment of the remaining BER proteins required to complete repair,
including XRCC1, DNA Ligase III, and DNA polymerase β (Polβ). Whereas
XRCC1 is a scaffold protein, Polβ carries out two essential enzymatic functions in
BER. First, the repair gap is tailored by the 5′dRP lyase activity of Polβ. Next, Polβ
fills the single-nucleotide gap, preparing the strand for ligation by either DNA ligase
I (LigI) or a complex of DNA ligase III (LigIII), and XRCC1 [36].
Although some BER substrates (base lesions) induced by chemotherapeutic
agents are cytotoxic [43], most are found to be mutagenic. However, essentially,
every intermediate throughout the BER pathway (abasic sites, 5′dRP lesions, and
single-strand DNA breaks) is toxic [13] and as such, there has been considerable
interest in developing BER inhibitors to enhance the accumulation of the cytotoxic
repair intermediates following chemotherapy or radiation treatment. This is dis-
cussed further in the sections below.
Another multi-protein, highly complex DNA repair pathway is the NER pathway.
NER plays an important role in the repair of DNA lesions induced by many geno-
toxins and chemotherapeutics including DNA cross-linking agents such as chloro-
ethylating agents (see Sect. 6.1.2), cisplatin, carboplatin, and lesions induced by
photodynamic therapy (PTD). Put simply, NER facilitates the removal of bulky
DNA adducts that grossly distort the DNA double helix and those that cause a
block to transcription. Molecular details on the proteins involved in NER can be
found in several excellent reviews [44–47]. Overall, the pathway consists of two
complementary sub-pathways that have some overlap. The two sub-pathways are
distinct regarding the lesion recognition step but converge and utilize the same
proteins to remove the oligonucleotide containing the lesion and for the steps
involving new DNA synthesis.
The global genomic NER (GG-NER) pathway surveys the entire genome
for DNA helix distorting lesions whereas the transcription-coupled repair NER
144 C. Vens and R. W. Sobol
6.1.5 Non-Homologous-End-Joining
One of the most cytotoxic lesions is a DNA double-strand break (DSB). If not
repaired, DSBs lead to chromosome breaks, loss of genetic material, and gross
genomic rearrangements. Whereas tolerance to the presence of DSBs might vary
in different cell types and cellular states, only a few DSBs will cause cell death or
prevent clonogenicity in most cells including cancer cells [53]. These lesions are
induced by multiple agents such as ionizing radiation, bleomycin, and topoisomer-
ase II inhibitors, but can also be induced indirectly at replication forks when con-
verting DNA single-strand breaks (SSBs) induced by topoisomerase I inhibitors
(camptothecin).
Two major cellular pathways deal with the repair of DNA DSBs. The use of
homologous DNA for repair distinguishes those repair pathways. As indicated
by the name, the non-homologous-end-joining (NHEJ) repair pathway does not
require any homologous sequences. Proteins of the NHEJ pathway can repair the
two ends in a DSB by simple end joining while the homologous recombination
(HR) repair pathway (see Sect. 6.1.6) requires homologous DNA stretches as tem-
plates for DNA synthesis and repair.
After the initial recognition of the DSB that is held in place and stabilized
by the binding of the MRN complex (MRE11, RAD50, NBS) and promoted by
ATM (see above), DSB repair is executed by the DNA-dependent protein kinase
(DNA-PK). DNA-PK is comprised of the catalytic subunit DNA-PKcs and the rel-
atively small Ku proteins (Ku70/80). They promote the simple ligation of the two
broken DNA ends. Damage-induced DSBs, in particular after ionizing radiation,
are rarely re-ligateable, and some end processing might be required that is accom-
plished by other enzymes such as Artemis. The DNA ligase IV-XRCC4 complex
6 Targeting DNA Repair Pathways for Cancer Therapy 145
finally re-ligates the two ends. DNA PK–independent DSB end-joining activity
has been observed in cells that have impaired NHEJ activity, the so-called alterna-
tive or B-NHEJ pathway. PARP and Ligase III activity appears to be implicated in
this cellular DSB repair option [54, 55].
The role of ATM seems to be of particular importance in the repair of a certain pro-
portion of DSBs, namely those in heterochromatic regions of the genome [56, 57].
These, judging from the repair kinetics, require more time to repair but influence sur-
vival substantially as indicated by the hypersensitivity to radiation of cells with impaired
ATM function.
Based on the cytotoxic nature of DSBs, cells impaired in any step of the NHEJ
process are highly sensitive to ionizing radiation. Genetic defects in or inhibition
of NHEJ also profoundly affects survival of cells by other DNA-damaging agents
that cause DSBs (directly or indirectly) such as DNA cross-linkers, bleomycin,
and topoisomerase inhibitors.
that is, crucial to resolve the complex-branched structures that arise in this pro-
cess. Notably, the products of the breast cancer susceptibility genes BRCA1 and
BRCA2 are involved in the FA and HR repair pathways, assisting DSB and cross-
link repair.
To allow the resolution of blocked replication fork structures, in particular fol-
lowing exposure to DNA cross-linking agents, another replication-associated
repair process is required, the FA pathway. Its members were discovered while
analyzing FA patients, victims of a human genetic disease that is characterized,
among other features [61], by extreme cellular sensitivity to drugs that pro-
duce ICLs. Subsequently, their role and actions in cellular cross-link repair was
revealed.
The products of at least 15 genes have been currently implicated in this path-
way [61–63]. This pathway constitutes a major signaling cascade upon replication
fork stalling: the FA “core complex” consists of at least 8 FA elements (FANCA,
FANCB, FANCC, FANCE, FANCF, FANCG, FANCL, and FANCM) and acts by
realizing the mono-ubiquitylation of the FA ID complex (FANCD2 and FANCI).
This activation of the ID complex allows chromatin binding and is thought to
facilitate DNA repair, in particular HR. The FA-mediated recruitment of the
RAD51 recombinase and the BRCA1-FANCJ helicase activity allows re-establish-
ment of the replication fork. Resolution of stalled replication forks appears to be
also supported by the translocase activity of FANCM that can remodel branched
DNA structures. The FA core complex is regulated by ATR and cell cycle check-
point elements (CHK1) allowing the activation of the pathway [64]. Importantly,
any mutation upstream of the FA pathway that will disrupt the mono-ubiquityla-
tion of FANCD2 will result in the cellular ICL hypersensitivity phenotype.
As illustrated above, upon exposure to DNA-damaging agents, it is the multi-
tude of cellular repair capacities that ultimately determines survival. HR and FA
have been shown to determine the cellular sensitivity to a wide range of cancer
therapeutics. HR-defective cells are hypersensitive to cross-linkers, IR, topoi-
somerase inhibitors, and alkylators as they induce DSBs and replication stalling.
The requirement for DNA repair and genome maintenance in response to radiation
and genotoxic chemotherapeutics implicates DNA repair proteins as prime targets
for improving response to currently available anticancer regimens. In addition, fre-
quent cancer-specific DNA repair and DDR defects offer tumor-selective therapy
options. Thus, strategies targeting DNA repair pathways represent promising new
avenues to improve outcome in cancer treatment.
Targeting DNA repair pathways in cancer treatment has been proposed in
several settings (Fig. 6.2). Most evidently, inhibition of cellular DNA repair will
cause increased sensitivity to chemotherapeutic agents or radiotherapy [65].
As illustrated above, cellular death upon exposure to most chemotherapeutic
6 Targeting DNA Repair Pathways for Cancer Therapy 147
Fig. 6.2 Strategies to improve cancer treatment by targeting DNA damage response and repair.
The overall goal is to increase tumor cell kill (y-axis) while sparing normal tissue by increasing
tumor specificity (x-axis) of the cancer treatment. While some strategies will achieve increased
cell death and thereby an increased probability to control the tumor (for example by applying
DNA-PK inhibitors in combination with radiotherapy or by re-sensitizing chemotherapy-resistant
tumors), others (such as those based on the exploitation of tumor-specific defects) will increase
tumor specificity of cancer therapies. However, in the manner in which current chemo- and radio-
therapy regimens are largely applied, each of the targeted approaches will be only beneficial in a
small fraction of patients with tumors that harbor the respective DNA repair defects. Neverthe-
less, considering the wide range of combination possibilities and the high number of targets and
exploitation opportunities, together they represent a promising avenue in cancer treatment
agents and ionizing radiation partly depends on the repair capacity for the
respective lesions. HR, NHEJ, BER, and DR processes are responsible for the
resistance to these agents. Hence, inhibitors to DNA repair elements might be
useful as dose intensifiers, augmenting the cell-killing properties of many if
not all therapeutic agents. This can be particularly useful in a setting in which
cancer cells obtained resistance to certain agents due to an improved repair capac-
ity. Thus, counteracting marked cancer cell resistance is one strategy in which DNA
repair inhibitors are proposed to act as dose intensifiers. By lowering the tolerance
and inhibiting alternative repair routes while augmenting cell kill, the application
of “intelligent” dose intensification by DNA repair inhibition can also prevent the
development of chemotherapy resistance.
Dose intensification will not be, in general, however, well tolerated, since
chemotherapeutic drugs and radiotherapy doses are often administered at
maximum-tolerated levels. Non-cancerous cells, with few exceptions, are as
148 C. Vens and R. W. Sobol
much exposed to the chemotherapeutic agents as are the cancer cells. Indeed, it
is the normal tissue response that defines the dose level and use of dose intensi-
fiers. Some tumor properties could, however, provide a tumor-specific effect when
targeting DNA repair. Further, in line with the rationale of most classical cancer
therapeutics, the proliferative nature of tumors is exploitable. For example, target-
ing replication-associated repair pathways such as HR with novel targeted agents
could be beneficial in radiotherapy regimens in which the healthy cells in the irra-
diated area are nonproliferative. A gain could also be expected if chemotherapy
dose limits are not defined by the proliferative cells, allowing dose intensification
by targeting replication-associated DNA repair. Other tumor-specific properties
such as exposure to hypoxic conditions or the altered metabolic status in cancer
cells can offer opportunities to achieve tumor-specific dose intensification and will
be discussed in the following paragraphs.
Another implication of DNA repair-targeting strategies has, however, become
highly relevant. It is suggested that most cancer cells are likely to be defective in
some aspect of DNA repair. Considering the multitude of repair options of healthy
cells, DNA repair defects in cancer cells can be exploited by targeting the remain-
ing repair processes. The combined lethal effect of two genetic variations that are
otherwise non-lethal is termed “synthetic lethality” [66]. In compliance with the
synthetic lethality concept, cancer cells defective in the primary repair pathway are
viable but rely heavily on secondary backup repair for survival. As this is not only
restricted to repair of endogenously produced lesions, this concept also applies
to cells exposed to exogenous damage by exacerbating the effects of chemo- and
radiotherapy in the defective cancer cells only (also may be called synthetic sick-
ness). Despite mutations and genetic defects, the differential expression of DNA
repair proteins or the altered engagement of DNA repair sub-pathways can be a
base of tumor-specific activities. Hence, these tumor-specific DDR and repair
defects offer promising novel approaches to tumor-selective therapy.
Investigation of the functionality of the individual DNA repair pathways in can-
cer cells and the knowledge on which pathways are implicated upon the inhibition
of DNA repair drug targets are necessary to combine these cancer therapy options
in an intelligent manner while focusing on a differential effect in the cancer versus
normal cells.
The recognition that DNA repair processes are prime targets for chemo-
and radiosensitization has driven the development of specific inhibitors to
elements of DDR, NHEJ, HR, DR, and BER. The more recent discovery of
tumor-specific targeting opportunities by the inhibition of DNA repair processes
fueled such attempts and has yielded a multitude of DNA repair inhibitors [5].
Some of these novel agents are currently being evaluated in the clinic while oth-
ers are being tested preclinically. Novel DNA repair targets have been identified,
Table 6.1 Targets in DNA damage response and repair
DNA Repair DNA Repair Compounds (DNA
protein target pathway involved repair inhibitors) Context Strategy Development stage Reference
PARP1/2 BER Olaparib Monotherapy BRCA1 or BRCA2 Clinical validation [248]
deficiency
PARP1/2 BER Olaparib Chemosensitizer Combined with cisplatin Clinical validation [249]
and gemcitabine
PARP1/2 BER Olaparib Radiation sensitizer Combined with radiation Preclinical [250]
258]
MGMT DR O6-benzylguanine Chemosensitizer Combined with carmustine Clinical validation [259]
(continued)
149
Table 6.1 (continued)
150
None (Abasic BER TRC102 Chemosensitizer Combined with temozolimide Preclinical [133–135]
site in DNA) (methoxyamine)
None (Abasic BER TRC102 Radio- and Combined with Preclinical [136]
site in DNA) (methoxyamine) chemosensitizer iododeoxyuridine +
radiation
None (Abasic site BER TRC102 Chemosensitizer Combined with BCNU Preclinical [137]
in DNA) (methoxyamine)
None (Abasic site BER TRC102 (methoxyamine) Chemosensitizer Combined with manumycin A Preclinical [138]
in DNA)
None (Abasic site BER TRC102 (methoxyamine) Chemosensitizer Combined with fludarabine Preclinical [139]
in DNA)
None (Abasic site BER TRC102 (methoxyamine) Radiation sensitizer Combined with radiation Preclinical [140, 141]
in DNA)
ATR DDR NU6027 Chemosensitizer Combined with PARP inhibitors Preclinical [151]
ATR DDR NVP-BEZ235 Monotherapy Cyclin-E overexpressing cells Compound screen [152]
ATM DDR KU55933 Radiation sensitizer Combined with radiation Compound screen [266]
and preclinical
ATM DDR KU60019 Radiation sensitizer Combined with radiation Preclinical [167]
ATM DDR CP466722 Radiation sensitizer Combined with radiation Preclinical [153]
(continued)
C. Vens and R. W. Sobol
Table 6.1 (continued)
DNA Repair DNA Repair Compounds (DNA
protein target pathway involved repair inhibitors) Context Strategy Development stage Reference
DNA-PK DDR NU7026 Radiation sensitizer Combined with radiation in Preclinical [164]
EGFRvIII-expressing tumors
DNA-PK DDR NU7441 Radio- and Chemosen- Combined with doxorubicin or Compound screen [267]
sitizer radiation and preclinical
DNA-PK DDR DT01 (dbait, DRIIM) Radiation sensitizer Metastatic melanoma with Compound screen [158]
relapsed cutaneous tumors and clinical
validation
DNA-PK DDR DT01 (dbait, DRIIM) Chemosensitizer Combined with 5-fluorouracil or Preclinical [268]
irinotecan
DNA-PK DDR HNI-38 Radiation sensitizer Combined with radiation Preclinical [269]
DNA-PK DDR KU-0060648; A dual Chemosensitizer Combined with etoposide Preclinical [159]
inhibitor of DNA-PK
and PI-3 K
NAMPT NAD+ biosyn- FK866 (Apo866) Chemosensitizer Combined with various Preclinical [190–192]
thesis chemotherapeutics
6 Targeting DNA Repair Pathways for Cancer Therapy
NAMPT NAD+ biosyn- GMX1778 Chemosensitizer Combined with various Preclinical [183]
thesis chemotherapeutics
NAMPT NAD+ biosyn- CB30865 Chemosensitizer Combined with various Preclinical [184]
thesis chemotherapeutics
NAMPT NAD+ biosyn- CHS-828 Chemosensitizer Combined with various Preclinical [185]
thesis chemotherapeutics
151
152 C. Vens and R. W. Sobol
and compounds that specifically inhibit their activity are sought in order to apply
tumor-specific anticancer strategies.
We will list currently explored DNA repair targets and some of the most
advanced compounds according to their developmental stage (Table 6.1).
Rationales and applied strategies will be discussed while pointing to opportunities
on combinations and other DNA repair targets.
6.3.1 Poly(ADP)Ribose Polymerase
One of the most advanced and applied DNA repair target inhibitors to date are the
PARP inhibitors [67–69]. Since the discovery that cells with defects in the BRCA
genes are selectively killed by the inhibition of PARP, PARP inhibitors have rap-
idly made their way into the clinic [70, 71]. As tumors from carriers of mutations
in the breast cancer susceptibility genes BRCA1 and BRCA2 are almost exclu-
sively composed of such BRCA-defected cells while normal cells of these carri-
ers still carry a functional allele, hence are HR proficient, these PARP inhibitors
achieve tumor-specific kill with little normal cell toxicity. The proposed mecha-
nism that causes such selectivity points to the dependence of BER-inhibited cells
on HR due to secondarily induced cytotoxic DSBs (Fig. 6.3a) [60]. Indeed, early
synthetic lethality screens in yeast indicated such an opportunity revealing a cru-
cial link between BER and HR for cellular survival [72]. Other hypotheses assume
a direct role of PARP inhibitors in replication fork stalling [73]. Most compounds
with PARP inhibitory activity target PAR generation by blocking the catalytic
activity of the enzyme. In principle, these compounds compete with NAD+ for the
PARP catalytic site and are therefore not necessarily specific to PARP1 and could
impact the activity of the other PARP isoforms [74]. Several PARP inhibitors are
in clinical development. To date, the leading compounds Olaparib (AZD2281,
AstraZeneca; originally developed by KuDos) and Veliparib (ABT-888, Abbott)
are probably the two most extensively studied in the clinic whereas at least one
compound, namely Iniparib (BSI-201; Sanofi-Aventis), has been reported to lack
effective PARP inhibitory activity [75, 76].
Although registration of these drugs is still awaiting approval, several studies
have shown their beneficial application [77, 78]. One obstacle could be that these
PARP inhibitors were expected to act in a fraction of tumors, those exhibiting HR
defects due to BRCA1 & BRCA2 mutations only. It should be noted that those early
clinical trials revealed that not all BRCA mutation carriers benefit, indicating that a
certain degree and type of HR defect is required to be exploitable with PARP inhi-
bition. The impact of individual BRCA mutations with respect to HR functionality
and/or PARP inhibitor sensitivity could be variable [79]. The status and propensity
to use the remaining DSB repair mechanism NHEJ, for example via 53BP1 chan-
neling, also influences the extent of PARP inhibitor toxicity [60, 80]. In addition,
other general drug resistance mechanisms such as increased compound rejection by
6 Targeting DNA Repair Pathways for Cancer Therapy 153
(a)
(b)
Fig. 6.3 Cellular functions of PARP and opportunities for cancer treatment. a The mode of
action of PARP inhibitors is based primarily on inhibition of the poly(ADP)ribosylation activ-
ity of the enzyme PARP1. As a result, BER or SSBR cannot be executed. In addition, the lack of
poly(ADP)ribosylation is likely to negatively impact chromatin and lesion accessibility. Auto-
poly(ADP)ribosylation of PARP1 is thought to promote its repulsion from DNA. A failure to
recruit downstream BER elements or an increase in lesion shielding by trapping PARP1 on the
DNA further inhibits BER. BER intermediates accumulated upon chemo- and radiotherapy, how-
ever, will then cause replication problems that in turn will induce DSBs. Those will ultimately
lead to cell death via apoptosis and/or mitotic catastrophe in particular if not repaired by HR.
b PARP activity has several cellular roles that can be taken advantage of in cancer treatment.
PARP promotes DNA repair and its inhibition, when combined with chemotherapy or with
tumor-specific defects, enhances tumor cell kill. NAD+ depletion, as a consequence of chemo-
or radiotherapy-induced PARP activity, also induces cell kill. Agents depleting cellular NAD+
levels could indirectly inhibit PARP. Conversely, PARP inhibitors can have the effect of lowering
chemotherapy-induced NAD+ depletion, thereby altering the mode of cell death. Lastly PARP
also has regulatory functions regarding DNA damage–induced gene expression that is often con-
nected to an inflammatory or fibrotic response. PARP inhibitors have been reported to exhibit
anti-inflammatory properties
154 C. Vens and R. W. Sobol
6.3.2 O6-Methylguanine-DNA Methyltransferase
As described in Sect. 6.1 above, MGMT is the sole protein responsible for the
repair of O6-alkylguanine lesions (formed by chemotherapeutic agents such as
TMZ). Tumors with elevated MGMT expression are resistant to TMZ and related
chemotherapeutic agents, and so, an active area of investigation has been the devel-
opment of MGMT inhibitors. If MGMT is inhibited (or MGMT is not expressed),
the tumor becomes highly sensitive to the agent (provided the tumor cell is
proficient in MMR—see Sect. 6.1.2). There have been multiple methodologies
proposed to inhibit or overcome resistance mediated by MGMT expression in the
tumor as well as to prevent sensitivity of normal tissue (primarily hematopoietic
cells) [12]. As we alluded in Sect. 6.1.2, the mechanism of action of MGMT in the
repair or de-alkylation of guanine suggested O6-benzylguanine as an ideal inhib-
itor [104]. This inhibitor, also called BG, is an analogue of the O6-alkylguanine
base and contains a benzyl ring instead of an alkyl group. The BG compound read-
ily reacts with MGMT, and the benzyl moiety is transferred to Cys145 as shown
(Fig. 6.4), releasing free guanine and rendering MGMT inactive. In some cases,
this has been shown to trigger ubiquitylation and proteasome-mediated destruction
156 C. Vens and R. W. Sobol
Fig. 6.4 Schematic
representation depicting
the mechanism of action
of the MGMT inhibitor
O6-benzylguanine (BG).
The benzyl moiety of BG
is transferred to the Cys145
residue in MGMT, releasing
free guanine, rendering
MGMT inactive. In some
cases, this has been shown
to trigger ubiquitylation
and proteasome-mediated
destruction of MGMT
checkpoints to prevent cell division and propagation of the damage. Thus, the
S/G2 checkpoint is an attractive target for cancer-specific sensitization to DNA-
damaging agents [111]. In order to exploit the cancer-specific defects, inhibitors
have been developed that abrogate the G2 checkpoint. CHK1 has been a prime
target for such attempts, as activated CHK1 mediates the arrest by phosphorylat-
ing Cdc25A and Cdc25C leading to their degradation and inactivation that oth-
erwise promote S-phase progression and entry into mitosis. Loss of the G2 cell
cycle checkpoint, despite the presence of unrepaired damage, is thought to pro-
voke mitotic catastrophe and ultimately cell kill. Consistent with this idea, loss of
intra-S or G2/M checkpoints increases the cytotoxicity of DNA-damaging agents
such as ionizing radiation and cisplatin. CHK1 knockdown sensitizes cells to
5-fluorouracil, doxorubicin, and etoposide. Despite the proposed mechanism that
CHK1 inhibition causes sensitization in p53-defective cancer cells due to the G2
block abrogation in a G1 block-deficient background, the data to support this are
contradictory [112–114]. Xeno-transplant studies on human triple-negative breast
cancer demonstrated the benefit of combining irinotecan with CHK1 inhibitors,
inducing checkpoint bypass and apoptosis. A role of p53 was supported by the
gain of CHK1 sensitization after p53 knockdown in the resistant tumors [115].
A more complicated rationale, however, argues that a series of DDR defects in
tumors should be considered and could be exploited by the inhibition of CHK1.
These are based on the secondary effects of CHK1 inactivation on replication and
are discussed below.
A large battery of CHK1 inhibitors is available to support cancer treatment in
combination with radio- and chemotherapy [116]. Older CHK1 inhibitors such as
UCN01 were not very selective but did demonstrate potent chemosensitization to
cisplatin and camptothecin. More potent and specific CHK1 inhibitors were devel-
oped; however, concomitant CHK2 inhibition to some degree is common to most
CHK1 inhibitors. In general, cells appear to depend on a functional G2 checkpoint
when exposed to agents that cause replication stress. Hence, potentiation is greatest
to cross-linkers, topoisomerase I poisons and nucleoside analogues such as gemcit-
abine. Consistent with replication stress hypersensitivity when G2 arrest is abrogated,
PARP inhibitors also cause problems. PARP inhibition can cause a G2 checkpoint
dependence and the combination of PARP inhibitors with CHK1 inhibitors is syn-
thetic lethal [117]. These cellular sensitivity features are the basis for the clini-
cal trials testing the combination of older (UCN01) or later generation compounds
(for example AZD7762, AstraZeneca; PF477736, Pfizer; LY2606368, Eli Lilly and
SCH900776, Schering Plough) with cisplatin, topotecan, and gemcitabine. For a
more detailed review, see [116]. Unfortunately, safety requirements as assessed in
these initial studies were not met in at least one compound (AZD7762) [118]. Similar
to chemotherapy regimens, CHK1 inhibitors prevent ionizing radiation (IR) induced
S and G2 arrest and demonstrated some potential to radiosensitize in a p53-depend-
ent manner. CHK1 is upregulated in Myc-overexpressing lymphomas, and single-
agent activity is expected since CHK1 inhibition is cytotoxic to these cells.
Interestingly, recent data indicate that CHK1 activity might prevent
replication-induced DNA damage or is implicated in DNA repair [119, 120]. A
158 C. Vens and R. W. Sobol
6.3.4 AP-Endonuclease 1
In parallel, there are a number of groups developing direct APE1 active site
inhibitors [143–146], and an overview of the development of APE1 inhibitors has
just been reported [147]. As might be expected, many of these APE1 inhibitors
are themselves cytotoxic and enhance the cytotoxicity of DNA-damaging agents
such as TMZ [144, 145] or other alkylating agents [143]. Although Ape1 KO or
GzmA-mediated Ape1 cleavage in mouse cells induces apoptosis, the cell death
mechanism(s) induced by these recently developed APE1 inhibitors has yet to be
resolved. Interestingly, the cell death mechanism triggered by some of these APE1
inhibitors may involve the accumulation of DNA DSBs since it was observed that
the compounds are more cytotoxic in cells deficient in the HR proteins BRCA1 or
BRCA2 [148].
Similar strategies and rationales that apply to the CHK1 target also apply to
ATR. A direct link connects ATR with CHK1 [121]. After initial Rad17 binding,
Claspin-loaded single-strand DNA mediates the activation of CHK1 by ATR upon
replication stress. ATR is essential in the surveillance of replication stress, espe-
cially when associated with exposed single-stranded DNA mostly connected to
replication problems. Stalled replication forks can collapse which results in the for-
mation of DSBs, a signal that mainly, but not exclusively, triggers ATM whereas
the single-stranded DNA-induced damage response appears to be ATM independ-
ent. Endogenous and exogenous damage induces replication stress that requires
a proficient ATR/CHK1 response for survival. One prevalent rationale to inhibit
ATR for cancer therapy is to exacerbate the levels of replication stress that might
be augmented in cancer cells due to inherited defects in the DDR and DNA repair
pathways. Cancer cells are exposed to a higher load of replication stress com-
pared to normal cells and will suffer most from targeting ATR. In addition, since
targeting replication-associated processes, any selective killing property will be
augmented in highly proliferative cells. Such a strategy is supported by two obser-
vations: (1) the activated DDR found in early stages of tumorigenesis [149, 150]
indicates an increased load of “endogenous” DNA damage and (2) the discovery
that a wide variety of oncogenes generate such damage.
Based on this proposed endogenous damage (replication stress)-induced kill-
ing mechanism, such ATR inhibitors have been proposed to act as single agents.
However, combination strategies similar to those for the CHK1 inhibitor are envi-
sioned. As noted before, the response of normal cells should be carefully taken
into consideration. A few compounds have been pursued by industry. The ATR
inhibitor NU6027 appears to impair HR and enhances the cytotoxicity of PARP
inhibitors [151], a theme that is consistent to other DDR inhibitors. One com-
pound (NVP-BEZ235) has been recently discovered and found to trigger pref-
erential cell kill in cyclin-E overexpressing cells [152] and is awaiting entry into
clinical trials for cancer therapy.
160 C. Vens and R. W. Sobol
6.3.6 Ataxia Telangiectasia-Mutated
ATM inhibitors are less advanced in their clinical development than PARP or
CHK1 inhibitors. Since involved in DSB repair, NHEJ, and HR, ATM inhibition
results in reduced cellular DSB repair activity and cell cycle checkpoint defects.
As a result, ATM inhibitors are highly potent radiosensitizers while exhibiting
little toxicity on their own. Therefore, they have been proposed to be applied in
this context [153]. Several ATM-inhibiting compounds have been identified and
pursued in preclinical studies: KU55933 and KU60019 (KuDos/AstraZeneca) and
CP466722 (Pfizer).
Combination strategies for ATM inhibitors have been proposed to enhance the
cytotoxicity of PARP inhibitors (see above). Interestingly, p53 disruption in normal
cells sensitized those cells to the combination of PARP and ATM inhibitors [83].
Due to its crucial role in DSB repair, the inhibition of ATM will radio- and chemo-
sensitize most cells, including normal cells, with no evident DNA repair defects.
However, synergistic cytotoxicity can be observed under certain conditions that
could be exploited for tumor-targeted strategies. Cells with BER defects for exam-
ple rely on secondary DSB repair pathways such as HR for survival upon dam-
aging agents. Similar to the PARP inhibitor/HR-defect synthetic lethal interaction,
the conversion of unrepaired SSBs to DSBs could be a mechanism that underlies
such dependence. BER defects of different kinds have been reported in tumors, and
it is suggested that inhibition of DSB repair processes will be beneficial in such a
setting [154].
Targeting the cellular signaling pathways via EGFR, PI3K/AKT, or MAPK can
modulate the DNA repair status of cells. The effects are multiple and involve dif-
ferent pathways. Cellular signaling influences DNA repair in multiple ways includ-
ing changes in the expression levels of crucial DNA repair proteins such as RAD51
or the regulation of the activation or translocation of enzymes and kinases such as
DNA-PK. For example, the inhibition of the MAPK pathway can lead to reduced
DSB repair by both homologous and non-homologous pathways [165, 166]. Links
between the AKT, MAPK, and EGFR pathways and DNA repair have been found,
particularly with DSB repair by NHEJ or HR [166–171]. Hence, targeting these
pathways can result in the concomitant inhibition of DNA repair.
Based on knowledge that the PI3K pathway promotes DNA repair and survival,
inhibitors of AKT have been evaluated as chemosensitizers for alkylating agents
[172, 173]. AKT inhibitors such as LY294002 and wortmannin radiosensitized and
caused enhanced sensitivity to alkylating agents such as TMZ, or the cross-linkers
cisplatin in various human tumor cell lines.
The suppression of ATR/CHK1 checkpoints has been observed upon treatment
with wortmannin (a fairly non-specific PI3K inhibitor). For more details, see [174]
and references within. Note that an influence on Rad51 expression or on NHEJ via
DNA-PK has also been observed.
162 C. Vens and R. W. Sobol
Co-targeting PARP and the PI3K pathway has been shown to synergistically
decrease growth and further induce apoptosis [175]. The molecular mechanism of
this combination effect is not completely clear but could originate from secondary
effects on DNA repair pathways and the DDR from the inhibition of cellular sign-
aling in human cancer cells. Further detail on AKT inhibitors is discussed in more
detail in Chap. 13.
6.4.2 Proteasome Inhibitors
Proteasome-mediated destruction and removal of key DNA repair and DDR pro-
teins are an essential aspect of the cellular response to genotoxins [4, 176]. Failure
to remove critical DNA repair and DDR proteins in a timely fashion effectively
halts the process, triggering a cascade of events leading to cell death [177].
As the endpoint in ubiquitin-mediated protein turnover, targeting the proteasome
is a high-profile target to enhance DNA damage–induced cell death [178, 179].
Further detail on the proteasome pathway and proteasome inhibitors is discussed
in more detail in Chap. 12.
The metabolite NAD+ is an essential substrate for all PARPs [74]. Since PARP1,
as well as PARP2 and PARP3, is a critical protein involved in DNA repair
and the DDR [36, 103], we have included NAD+ biosynthesis inhibitors as an
indirect DNA repair modulator. All of the NAD+ biosynthesis inhibitors developed
to date target NAMPT, a pivotal and rate-limiting enzyme in the salvage pathway of
NAD+ biosynthesis [180]. NAMPT catalyzes the synthesis of nicotinamide mononu-
cleotide (NMN) from nicotinamide to 5-phosphoribosyl-1-pyrophosphate [181]. The
resulting NMN is then converted to NAD+ by one of the three isoforms of NMNAT
(1, 2 or 3), located in the nucleus, cytosol, or mitochondria, respectively [182]. The
most studied inhibitor of NAMPT is FK866 (also referred to as Apo866) although
several other NAMPT inhibitors have been reported including GMX1778 [183],
CB30865 [184], and CHS-828 [185], and one group is actively screening for NAMPT
inhibitors [186]. FK866 binds at the interface of the NAMPT dimer [187] and effec-
tively inhibits NAMPT and depletes cellular NAD+ levels within 24 h, inducing apop-
tosis [188] although the overall level of cell death may be offset by FK866-mediated
induction of autophagy [189]. Further detail on the shift of cell death triggered by
NAD+ biosynthesis inhibitors is discussed in Chap. 2.
As an NAMPT inhibitor, FK866 does have a chemo-potentiating effect
[190–192] that is more pronounced when combined with a second DNA repair inhibi-
tor [133] or when combined with TRAIL [193]. Interestingly, it has been suggested
that some tumors are deficient in the NAPRT1-mediated NAD+ biosynthesis pathway
6 Targeting DNA Repair Pathways for Cancer Therapy 163
that is responsible for generating NAD+ from nicotinic acid (NA) [183, 194]. In sum,
these studies suggest that NAMPT is a potentially valuable target to impact both
metabolism and DNA repair and may provide a type of synthetic lethality based on
DNA repair and NAD+ biosynthesis status [133, 195].
6.5.1 ERCC1
6.5.2 DNA Polymerases
There are 15 DNA polymerases in the human cell [204] and several have been
suggested to be viable drug targets either because they facilitate repair of DNA
damage or may be overexpressed in cancer [205]. DNA polymerase β (Polβ) is a
member of the X family of DNA polymerases [204, 206] and is an essential BER
protein, as detailed above. Polβ has two active sites and several critical functional
domains that may be considered as targets to inhibit Polβ and BER. Further, the
central and pivotal role of Polβ in BER implicates this protein as a prime target
to enhance the response to chemotherapeutic agents or radiation. Although many
164 C. Vens and R. W. Sobol
inhibitors of Polβ have been developed and characterized very few, if any, show
specificity and all are cytotoxic even in Polβ KO cells. Three recent reviews on
Polβ inhibitors should be referred to for more detail [195, 207, 208].
Other polymerases have been also identified as potential targets. A siRNA screen
and large-scale tumor sample analysis identified DNA polymerase theta (POLQ) as a
target that determines radiosensitivity and is overexpressed in tumors. POLQ depletion
radiosensitized these cells arguing for the development of POLQ inhibitors [209–211].
6.5.3 DNA Glycosylases
DNA glycosylases are the initiating enzymes in BER (see above). In general,
DNA glycosylases probe the DNA helix for base lesions by a base-flipping
mechanism, interrogating each base as it is flipped out of the major groove as
the enzyme moves along the DNA helix. Identification of the lesion then pro-
motes hydrolysis [212]. As discussed above, radiation and chemotherapeu-
tic agents induce the formation of BER substrates and many of these base
lesions block replication and are therefore cytotoxic. For example, the methyl-
ated base 3-methyladenine, induced by the chemotherapeutic agent TMZ and
repaired by MPG, triggers lesion-induced apoptosis as well as sister chroma-
tid exchange, chromatid and chromosome gaps and breaks, and S-phase arrest
[213]. As such, it has been hypothesized that inhibition of MPG would enhance
response to TMZ and other alkylators. Similarly, UNG, the primary glycosy-
lase responsible for the removal of deoxyuracil, is reported to govern the effi-
cacy of pemetrexed [142], and therefore, UNG might be considered a potential
target to enhance the response to this chemotherapeutic agent. Finally, it was
recently demonstrated that the DNA glycosylase OGG1 is in a complex with
the BER protein PARP1. The OGG1–PARP1 complex promotes PARP1 activ-
ity whereas loss of OGG1 suppressed PARP1 activity [214]. From this study, it
might be suggested that an inhibitor designed to disrupt the OGG1–PARP1 inter-
action may function as a pseudo- or indirect PARP1 inhibitor. However, there are
multiple challenges to developing effective DNA glycosylase inhibitors such as
the large overlap in enzyme substrate specificity [36, 37] and the requirement for
some glycosylases in normal cell survival [215, 216] and immunoglobulin class-
switch recombination [217]. To date, we know of no DNA glycosylase inhibitors
with the exception of Ugi, a virally encoded UNG inhibitor [218].
6.5.4 DUBs
and chemotherapeutic response [221, 222]. Toward that end, several groups have
reported the development of DUB inhibitors that are effective in preclinical studies
[223–226].
6.5.5 HR/FA
6.5.6 Poly(ADP)ribose Glycohydrolase
knockdown PARG in human cells have suggested that blocking PAR degrada-
tion is an effective means to modulate chemotherapy and radiation response
[134, 236, 237]. Pre-clinical evaluations using early phase small-molecule inhibi-
tors such as gallotannin, tannic acid, and related small molecules have implicated
PARG in genotoxin or chemotherapy sensitivity [238–240]. The inhibitor GPI-
16552 (N-bis-(3-phenyl-propyl)9-oxo-fluorene-2,7-diamide) has shown early suc-
cess in cell and mouse models as a chemotherapy sensitizer [97] but has mostly
been effective to reduce inflammation [241–245]. Recent drug discovery efforts
seem promising and have yielded PARG inhibitors with increased specificity and
cell permeability [246, 247].
References
1. Lindahl T (1993) Instability and decay of the primary structure of DNA. Nature
362(6422):709–715
2. Jackson SP, Bartek J (2009) The DNA-damage response in human biology and disease.
Nature 461(7267):1071–1078. Epub 2009/10/23
3. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell
144(5):646–674. Epub 2011/03/08
4. Harper JW, Elledge SJ (2007) The DNA damage response: ten years after. Mol Cell
28(5):739–745. Epub 2007/12/18
6 Targeting DNA Repair Pathways for Cancer Therapy 167
5. Ljungman M (2009) Targeting the DNA damage response in cancer. Chem Rev
109(7):2929–2950. Epub 2009/06/24
6. Alberts B (2009) Redefining cancer research. Science 325(5946):1319. Epub 2009/09/12
7. Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA (2008) DNA repair pathways
as targets for cancer therapy. Nat Rev Cancer 8(3):193–204
8. Lavin MF (2008) Ataxia-telangiectasia: from a rare disorder to a paradigm for cell signal-
ling and cancer. Nat Rev Mol Cell Biol 9(10):759–769. Epub 2008/09/25
9. Matsuoka S, Ballif BA, Smogorzewska A, McDonald ER 3rd, Hurov KE, Luo J et al (2007)
ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA
damage. Science 316(5828):1160–1166
10. Bakkenist CJ, Kastan MB (2003) DNA damage activates ATM through intermolecular
autophosphorylation and dimer dissociation. Nature 421(6922):499–506
11. Wood RD, Mitchell M, Sgouros J, Lindahl T (2001) Human DNA repair genes. Science
291(5507):1284–1289
12. Gerson SL (2004) MGMT: its role in cancer aetiology and cancer therapeutics. Nat Rev
Cancer 4(4):296–307
13. Fu D, Calvo JA, Samson LD (2012) Balancing repair and tolerance of DNA damage caused
by alkylating agents. Nat Rev Cancer 12(2):104–120. Epub 2012/01/13
14. Kaina B, Christmann M, Naumann S, Roos WP (2007) MGMT: key node in the battle
against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA
Repair (Amst) 6(8):1079–1099
15. Friedberg EC, Walker GC, Siede W, Wood RD, Schultz RA, Ellenberger T (2006) DNA
repair and mutagenesis. 2nd Edn. ASM Press, Washington, p 1164
16. Wyatt MD, Pittman DL (2006) Methylating agents and DNA repair responses: methylated
bases and sources of strand breaks. Chem Res Toxicol 19(12):1580–1594
17. Wang JY, Edelmann W (2006) Mismatch repair proteins as sensors of alkylation DNA dam-
age. Cancer Cell 9(6):417–418
18. Friedman HS, Johnson SP, Dong Q, Schold SC, Rasheed BK, Bigner SH et al (1997)
Methylator resistance mediated by mismatch repair deficiency in a glioblastoma multiforme
xenograft. Cancer Res 57(14):2933–2936
19. Roos WP, Batista LF, Naumann SC, Wick W, Weller M, Menck CF et al (2007)
Apoptosis in malignant glioma cells triggered by the temozolomide-induced DNA lesion
O6-methylguanine. Oncogene 26(2):186–197
20. Caporali S, Falcinelli S, Starace G, Russo MT, Bonmassar E, Jiricny J et al (2004) DNA
damage induced by temozolomide signals to both ATM and ATR: role of the mismatch
repair system. Mol Pharmacol 66(3):478–491
21. Jiricny J (2006) The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol
7(5):335–346
22. Pabla N, Ma Z, McIlhatton MA, Fishel R, Dong Z (2011) hMSH2 recruits ATR to
DNA damage sites for activation during DNA damage-induced apoptosis. J Biol Chem
286(12):10411–10418. Epub 2011/02/03
23. Wang Y, Qin J. MSH2 and ATR form a signaling module and regulate two branches of the
damage response to DNA methylation. Proceedings Nat Acad Sci 100(26):15387–15392
24. Hegi ME, Liu L, Herman JG, Stupp R, Wick W, Weller M et al (2008) Correlation of
O6-methylguanine methyltransferase (MGMT) promoter methylation with clinical outcomes in
glioblastoma and clinical strategies to modulate MGMT activity. J Clin Oncol 26(25):4189–4199
25. Sarkaria JN, Kitange GJ, James CD, Plummer R, Calvert H, Weller M et al (2008)
Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clin Cancer Res
14(10):2900–2908
26. Pollack IF, Hamilton RL, Sobol RW, Burnham J, Yates AJ, Holmes EJ et al (2006) MGMT
expression strongly correlates with outcome in childhood malignant gliomas: results from
the CCG-945 cohort. J Clin Oncol 24(21):3431–3437
27. Holland EC (2000) Glioblastoma multiforme: the terminator. Proc Natl Acad Sci USA
97(12):6242–6244. Epub 2000/06/07
168 C. Vens and R. W. Sobol
28. Cohen MH, Johnson JR, Pazdur R (2005) Food and drug administration drug approval sum-
mary: temozolomide plus radiation therapy for the treatment of newly diagnosed glioblas-
toma multiforme. Clin Cancer Res 11(19 Pt 1):6767–6771
29. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC et al (2009)
Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy
alone on survival in glioblastoma in a randomised phase III study: 5 year analysis of the
EORTC-NCIC trial. Lancet Oncol 10(5):459–466
30. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ et al (2005)
Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med
352(10):987–996
31. Cahill DP, Levine KK, Betensky RA, Codd PJ, Romany CA, Reavie LB et al (2007) Loss
of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor pro-
gression during temozolomide treatment. Clin Cancer Res 13(7):2038–2045
32. Yip S, Miao J, Cahill DP, Iafrate AJ, Aldape K, Nutt CL et al (2009) MSH6 mutations arise
in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin
Cancer Res 15(14):4622–4629
33. Friedman HS, Keir S, Pegg AE, Houghton PJ, Colvin OM, Moschel RC et al (2002) O6-
benzylguanine-mediated enhancement of chemotherapy. Mol Cancer Ther 1(11):943–948
34. Tserng KY, Ingalls ST, Boczko EM, Spiro TP, Li X, Majka S et al (2003) Pharmacokinetics
of O6-benzylguanine (NSC637037) and its metabolite, 8-oxo-O6-benzylguanine. J Clin
Pharmacol 43(8):881–893
35. Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, Vanaclocha V et al
(2000) Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to
alkylating agents. N Engl J Med 343(19):1350–1354
36. Almeida KH, Sobol RW (2007) A unified view of base excision repair: lesion-dependent
protein complexes regulated by post-translational modification. DNA Repair 6(6):695–711
37. Svilar D, Goellner EM, Almeida KH, Sobol RW (2011) Base excision repair and lesion-
dependent sub-pathways for repair of oxidative DNA damage. Antioxid Redox Signal
14(12):2491–2507. Epub 2010/07/24
38. Sobol RW (2009) Temozolomide. In: Schwab M (ed) Encyclopedia of cancer, 2nd edn.
Springer, Berlin Heidelberg, pp 2928–2933
39. Wilson SH, Kunkel TA (2000) Passing the baton in base excision repair. Nat Struct Biol
7(3):176–178
40. Gagne JP, Pic E, Isabelle M, Krietsch J, Ethier C, Paquet E et al (2012) Quantitative pro-
teomics profiling of the poly(ADP-ribose)-related response to genotoxic stress. Nucleic
Acids Res 40(16):7788–7805. Epub 2012/06/07
41. Gibson BA, Kraus WL (2012) New insights into the molecular and cellular functions of
poly(ADP-ribose) and PARPs. Nat Rev Mol Cell Biol 13(7):411–424. Epub 2012/06/21
42. Sousa FG, Matuo R, Soares DG, Escargueil AE, Henriques JA, Larsen AK et al PARPs and
the DNA damage response. Carcinogenesis 33(8):1433–1440. Epub 2012/03/21
43. Roos WP, Kaina B (2012) DNA damage-induced apoptosis: from specific DNA lesions to
the DNA damage response and apoptosis. Cancer Lett In Press. Epub 2012/01/21
44. Hoeijmakers JH (2001) Genome maintenance mechanisms for preventing cancer. Nature
411(6835):366–374
45. De Laat WL, Jaspers NG, Hoeijmakers JH (1999) Molecular mechanism of nucleotide exci-
sion repair. Genes Dev 13(7):768–785. Epub 1999/04/10
46. Wood RD (1996) DNA repair in eukaryotes. Annu Rev Biochem 65:135–167. Epub
1996/01/01
47. Shuck SC, Short EA, Turchi JJ (2008) Eukaryotic nucleotide excision repair: from under-
standing mechanisms to influencing biology. Cell Res 18(1):64–72. Epub 2008/01/02
48. Kapetanaki MG, Guerrero-Santoro J, Bisi DC, Hsieh CL, Rapic-Otrin V, Levine AS
(2006) The DDB1-CUL4ADDB2 ubiquitin ligase is deficient in xeroderma pigmentosum
group E and targets histone H2A at UV-damaged DNA sites. Proc Natl Acad Sci USA
103(8):2588–2593
6 Targeting DNA Repair Pathways for Cancer Therapy 169
49. Barakat K, Gajewski M, Tuszynski JA (2012) DNA repair inhibitors: the next major step to
improve cancer therapy. Curr Top Med Chem 12(12):1376–1390. Epub 2012/07/17
50. Postel-Vinay S, Vanhecke E, Olaussen KA, Lord CJ, Ashworth A, Soria JC (2012) The
potential of exploiting DNA-repair defects for optimizing lung cancer treatment. Nat Rev
Clin Oncol 9(3):144–155. Epub 2012/02/15
51. Bhagwat NR, Roginskaya VY, Acquafondata MB, Dhir R, Wood RD, Niedernhofer LJ
(2009) Immunodetection of DNA repair endonuclease ERCC1-XPF in human tissue.
Cancer Res 69(17):6831–6838. Epub 2009/09/03
52. Niedernhofer LJ, Bhagwat N, Wood RD. ERCC1 and non-small-cell lung cancer. N Engl J
Med. 2007;356(24):2538-40; author reply 40-1. Epub 2007/06/15
53. Lobrich M, Shibata A, Beucher A, Fisher A, Ensminger M, Goodarzi AA et al (2010)
GammaH2AX foci analysis for monitoring DNA double-strand break repair: strengths, limi-
tations and optimization. Cell Cycle 9(4):662–669. Epub 2010/02/09
54. Audebert M, Salles B, Calsou P (2004) Involvement of poly(ADP-ribose) polymerase-1 and
XRCC1/DNA ligase III in an alternative route for DNA double-strand breaks rejoining. J
Biol Chem 279(53):55117–55126
55. Mladenov E, Iliakis G (2011) Induction and repair of DNA double strand breaks: the
increasing spectrum of non-homologous end joining pathways. Mutat Res 711(1–2):61–72.
Epub 2011/02/19
56. Goodarzi AA, Noon AT, Deckbar D, Ziv Y, Shiloh Y, Lobrich M et al (2008) ATM signaling
facilitates repair of DNA double-strand breaks associated with heterochromatin. Mol Cell
31(2):167–177. Epub 2008/07/29
57. Murray JM, Stiff T, Jeggo PA (2012) DNA double-strand break repair within heterochro-
matic regions. Biochem Soc Trans 40(1):173–178. Epub 2012/01/21
58. Moynahan ME, Jasin M (2010) Mitotic homologous recombination maintains genomic
stability and suppresses tumorigenesis. Nat Rev Mol Cell Biol 11(3):196–207. Epub
2010/02/24
59. Jeggo PA, Geuting V, Lobrich M (2011) The role of homologous recombination in
radiation-induced double-strand break repair. Radiother Oncol 101(1):7–12. Epub
2011/07/09
60. Allen C, Ashley AK, Hromas R, Nickoloff JA (2011) More forks on the road to replication
stress recovery. J Mol Cell Biol 3(1):4–12. Epub 2011/02/01
61. Kim H, D’Andrea AD (2012) Regulation of DNA cross-link repair by the Fanconi
anemia/BRCA pathway. Genes Dev 26(13):1393–1408. Epub 2012/07/04
62. Crossan GP, Patel KJ (2012) The fanconi anaemia pathway orchestrates incisions at sites of
crosslinked DNA. J Pathol 226(2):326–337. Epub 2011/10/01
63. Yuan F, Song L, Qian L, Hu JJ, Zhang Y (2010) Assembling an orchestra: Fanconi anemia
pathway of DNA repair. Front Biosci 15:1131–1149. Epub 2010/06/03
64. Wang W (2008) A major switch for the Fanconi anemia DNA damage-response pathway.
Nat Struct Mol Biol 15(11):1128–1130. Epub 2008/11/06
65. Begg AC, Stewart FA, Vens C (2011) Strategies to improve radiotherapy with targeted
drugs. Nat Rev Cancer 11(4):239–253. Epub 2011/03/25
66. Kaelin WG Jr (2005) The concept of synthetic lethality in the context of anticancer therapy.
Nat Rev Cancer 5(9):689–698
67. Javle M, Curtin NJ (2011) The role of PARP in DNA repair and its therapeutic exploitation.
Br J Cancer 105(8):1114–1122. Epub 2011/10/13
68. Sandhu SK, Yap TA, De Bono JS (2010) Poly(ADP-ribose) polymerase inhibitors in cancer
treatment: a clinical perspective. Eur J Cancer 46(1):9–20. Epub 2009/11/21
69. Peralta-Leal A, Rodriguez MI, Oliver FJ (2008) Poly(ADP-ribose)polymerase-1 (PARP-1)
in carcinogenesis: potential role of PARP inhibitors in cancer treatment. Clin Transl Oncol
10(6):318–323
70. Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E et al (2005) Specific
killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase.
Nature 434(7035):913–917
170 C. Vens and R. W. Sobol
71. Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB et al (2005)
Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature
434(7035):917–921
72. Hendricks CA, Razlog M, Matsuguchi T, Goyal A, Brock AL, Engelward BP (2002) The
S.cerevisiae Mag1 3-methyladenine DNA glycosylase modulates susceptibility to homolo-
gous recombination. DNA Repair 1:645–659
73. Shaheen M, Allen C, Nickoloff JA, Hromas R (2011) Synthetic lethality: exploiting the
addiction of cancer to DNA repair. Blood 117(23):6074–6082. Epub 2011/03/29
74. Hassa PO, Haenni SS, Elser M, Hottiger MO (2006) Nuclear ADP-ribosylation reactions
in mammalian cells: where are we today and where are we going? Microbiol Mol Biol Rev
70(3):789–829
75. Patel AG, De Lorenzo SB, Flatten KS, Poirier GG, Kaufmann SH (2012) Failure of iniparib
to inhibit poly(ADP-Ribose) polymerase in vitro. Clin Cancer Res 18(6):1655–1662. Epub
2012/02/01
76. Liu X, Shi Y, Maag DX, Palma JP, Patterson MJ, Ellis PA et al (2012) Iniparib nonselec-
tively modifies cysteine-containing proteins in tumor cells and is not a bona fide PARP
inhibitor. Clin Cancer Res 18(2):510–523. Epub 2011/12/01
77. Audeh MW, Carmichael J, Penson RT, Friedlander M, Powell B, Bell-McGuinn KM et
al (2010) Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1
or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet
376(9737):245–251. Epub 2010/07/09
78. Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN et al (2010) Oral
poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 muta-
tions and advanced breast cancer: a proof-of-concept trial. Lancet 376(9737):235–244.
Epub 2010/07/09
79. Vollebergh MA, Jonkers J, Linn SC (2012) Genomic instability in breast and ovarian can-
cers: translation into clinical predictive biomarkers. Cell Mol Life Sci 69(2):223–245. Epub
2011/09/17
80. Bunting SF, Callen E, Kozak ML, Kim JM, Wong N, Lopez-Contreras AJ et al (2012)
BRCA1 functions independently of homologous recombination in DNA interstrand cross-
link repair. Mol Cell 46(2):125–135. Epub 2012/03/27
81. McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S et al (2006) Deficiency
in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-
Ribose) polymerase inhibition. Cancer Res 66(16):8109–8115
82. Yap TA, Sandhu SK, Carden CP, De Bono JS (2011) Poly(ADP-ribose) polymerase (PARP)
inhibitors: exploiting a synthetic lethal strategy in the clinic. CA Cancer J Clin 61(1):31–49.
Epub 2011/01/06
83. Williamson CT, Kubota E, Hamill JD, Klimowicz A, Ye R, Muzik H et al (2012) Enhanced
cytotoxicity of PARP inhibition in mantle cell lymphoma harbouring mutations in both
ATM and p53. EMBO Mol Med 4(6):515–527. Epub 2012/03/15
84. Williamson CT, Muzik H, Turhan AG, Zamo A, O’Connor MJ, Bebb DG et al ATM defi-
ciency sensitizes mantle cell lymphoma cells to poly(ADP-ribose) polymerase-1 inhibitors.
Mol Cancer Ther 9(2):347–57. Epub 2010/02/04
85. McEllin B, Camacho CV, Mukherjee B, Hahm B, Tomimatsu N, Bachoo RM et al (2010)
PTEN loss compromises homologous recombination repair in astrocytes: implications
for glioblastoma therapy with temozolomide or poly(ADP-ribose) polymerase inhibitors.
Cancer Res 70(13):5457–5464. Epub 2010/06/10
86. Mendes-Pereira AM, Martin SA, Brough R, McCarthy A, Taylor JR, Kim JS et al (2009)
Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO molecular
medicine 1(6–7):315–322. Epub 2010/01/06
87. Fraser M, Zhao H, Luoto KR, Lundin C, Coackley C, Chan N et al (2011) PTEN
deletion in prostate cancer cells does not associate with loss of RAD51 function: impli-
cations for radiotherapy and chemotherapy. Clin Cancer Res 18(4):1015–1027. Epub
2011/11/25
6 Targeting DNA Repair Pathways for Cancer Therapy 171
88. Chan N, Pires IM, Bencokova Z, Coackley C, Luoto KR, Bhogal N et al (2010) Contextual
synthetic lethality of cancer cell kill based on the tumor microenvironment. Cancer Res
70(20):8045–8054. Epub 2010/10/07
89. Liu SK, Coackley C, Krause M, Jalali F, Chan N, Bristow RG (2008) A novel poly(ADP-
ribose) polymerase inhibitor, ABT-888, radiosensitizes malignant human cell lines under
hypoxia. Radiother Oncol 88(2):258–268. Epub 2008/05/06
90. Chalmers AJ (2010) Overcoming resistance of glioblastoma to conventional cytotoxic ther-
apies by the addition of PARP inhibitors. Anti-Cancer Agents Med Chem 10(7):520–533.
Epub 2010/10/01
91. Chalmers AJ, Lakshman M, Chan N, Bristow RG (2010) Poly(ADP-ribose) polymerase
inhibition as a model for synthetic lethality in developing radiation oncology targets. Semin
Radiat Oncol 20(4):274–281. Epub 2010/09/14
92. Aguilar-Quesada R, Munoz-Gamez JA, Martin-Oliva D, Peralta-Leal A, Quiles-Perez R,
Rodriguez-Vargas JM et al (2007) Modulation of transcription by PARP-1: consequences in
carcinogenesis and inflammation. Curr Med Chem 14(11):1179–1187
93. Giansanti V, Dona F, Tillhon M, Scovassi AI (2010) PARP inhibitors: new tools to protect
from inflammation. Biochem pharmacol 80(12):1869–1877. Epub 2010/04/27
94. De la Lastra CA, Villegas I, Sanchez-Fidalgo S (2007) Poly(ADP-ribose) polymerase inhib-
itors: new pharmacological functions and potential clinical implications. Curr Pharm Des
13(9):933–962. Epub 2007/04/14
95. Wedge SR, Porteous JK, Newlands ES (1996) 3-Aminobenzamide and/or
O6-benzylguanine evaluated as an adjuvant to temozolomide or BCNU treatment in cell
lines of variable mismatch repair status and O6-alkylguanine-DNA alkyltransferase activity.
Br J Cancer 74(7):1030–1036. Epub 1996/10/01
96. Tentori L, Turriziani M, Franco D, Serafino A, Levati L, Roy R et al (1999) Treatment with
temozolomide and poly(ADP-ribose) polymerase inhibitors induces early apoptosis and
increases base excision repair gene transcripts in leukemic cells resistant to triazene com-
pounds. Leukemia 13(6):901–909
97. Tentori L, Leonetti C, Scarsella M, D’Amati G, Vergati M, Portarena I et al (2003) Systemic
administration of GPI 15427, a novel poly(ADP-ribose) polymerase-1 inhibitor, increases
the antitumor activity of temozolomide against intracranial melanoma, glioma, lymphoma.
Clin Cancer Res 9(14):5370–5379
98. Curtin NJ, Wang LZ, Yiakouvaki A, Kyle S, Arris CA, Canan-Koch S et al (2004) Novel
poly(ADP-ribose) polymerase-1 inhibitor, AG14361, restores sensitivity to temozolomide in
mismatch repair-deficient cells. Clin Cancer Res 10(3):881–889
99. Rottenberg S, Jaspers JE, Kersbergen A, van der Burg E, Nygren AO, Zander SA
et al (2008) High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibi-
tor AZD2281 alone and in combination with platinum drugs. Proc Natl Acad Sci USA
105(44):17079–17084. Epub 2008/10/31
100. Evers B, Drost R, Schut E, de Bruin M, van der Burg E, Derksen PW et al (2008) Selective
inhibition of BRCA2-deficient mammary tumor cell growth by AZD2281 and cisplatin. Clin
Cancer Res 14(12):3916–3925 Epub 2008/06/19
101. Berger NA (1985) Poly(ADP-ribose) in the cellular response to DNA damage. Radiat Res
101(1):4–15
102. Hottiger MO, Hassa PO, Luscher B, Schuler H, Koch-Nolte F (2010) Toward a unified
nomenclature for mammalian ADP-ribosyltransferases. Trends Biochem Sci 35(4):208–219.
Epub 2010/01/29
103. Tang J, Goellner EM, Wang XW, Trivedi RN, St. Croix CM, Jelezcova E et al (2010)
Bioenergetic metabolites regulate base excision repair-dependent cell death in response to
DNA damage. Mol Cancer Res 8(1):67–79. Epub 2010/01/14
104. Dolan ME, Pegg AE (1997) O6-Benzylguanine and its role in chemotherapy. Clin Cancer
Res 3(6):837–847. Epub 1997/06/01
105. Xu-Welliver M, Pegg AE (2002) Degradation of the alkylated form of the DNA repair protein,
O(6)-alkylguanine-DNA alkyltransferase. Carcinogenesis 23(5):823–30. Epub 2002/05/23
172 C. Vens and R. W. Sobol
106. Pegg AE (2011) Multifaceted roles of alkyltransferase and related proteins in DNA
repair, DNA damage, resistance to chemotherapy, and research tools. Chem Res Toxicol
24(5):618–639. Epub 2011/04/07
107. Middleton MR, Kelly J, Thatcher N, Donnelly DJ, McElhinney RS, McMurry TB et al
(2000) O(6)-(4-bromothenyl)guanine improves the therapeutic index of temozolomide
against A375 M melanoma xenografts. Int J Cancer 85(2):248–252. Epub 2000/01/11
108. Zhang J, Stevens MF, Bradshaw TD (2012) Temozolomide: mechanisms of action, repair
and resistance. Curr Mol Pharmacol 5(1):102–114. Epub 2011/11/30
109. Khan O, Middleton MR (2007) The therapeutic potential of O6-alkylguanine DNA alkyl-
transferase inhibitors. Expert Opin Investig Drugs 16(10):1573–1584. Epub 2007/10/10
110. Davis BM, Roth JC, Liu L, Xu-Welliver M, Pegg AE, Gerson SL (1999) Characterization
of the P140 K, PVP(138-140)MLK, and G156A O6-methylguanine-DNA methyltransferase
mutants: implications for drug resistance gene therapy. Hum Gene Ther 10(17):2769–2778.
Epub 1999/12/10
111. Chen T, Stephens PA, Middleton FK, Curtin NJ (2011) Targeting the S and G2 checkpoint
to treat cancer. Drug Discovery Today 17(5–6):194–202. Epub 2011/12/24
112. Zenvirt S, Kravchenko-Balasha N, Levitzki A (2010) Status of p53 in human cancer cells
does not predict efficacy of CHK1 kinase inhibitors combined with chemotherapeutic
agents. Oncogene 29(46):6149–6159. Epub 2010/08/24
113. Cho SH, Toouli CD, Fujii GH, Crain C, Parry D (2004) Chk1 is essential for tumor cell
viability following activation of the replication checkpoint. Cell Cycle 4(1):131–139. Epub
2004/11/13
114. Sorensen CS, Hansen LT, Dziegielewski J, Syljuasen RG, Lundin C, Bartek J et al (2005)
The cell-cycle checkpoint kinase Chk1 is required for mammalian homologous recombina-
tion repair. Nat Cell Biol 7(2):195–201
115. Ma CX, Cai S, Li S, Ryan CE, Guo Z, Schaiff WT et al (2012) Targeting Chk1 in p53-defi-
cient triple-negative breast cancer is therapeutically beneficial in human-in-mouse tumor
models. J Clin Invest 122(4):1541–1552. Epub 2012/03/27
116. Ma CX, Janetka JW, Piwnica-Worms H (2010) Death by releasing the breaks: CHK1 inhibi-
tors as cancer therapeutics. Trends Mol Med 17(2):88–96. Epub 2010/11/23
117. Mitchell C, Park M, Eulitt P, Yang C, Yacoub A, Dent P (2010) Poly(ADP-ribose) poly-
merase 1 modulates the lethality of CHK1 inhibitors in carcinoma cells. Mol Pharmacol
78(5):909–917. Epub 2010/08/11
118. Sausville EA, LoRusso P, Carducci MA, Barker PN, Agbo F, Oakes P et al (2011) Phase I
dose-escalation study of AZD7762 in combination with gemcitabine (gem) in patients (pts)
with advanced solid tumors. J Clin Oncol 29(suppl):abstr 3058
119. McNeely S, Conti C, Sheikh T, Patel H, Zabludoff S, Pommier Y et al (2010) Chk1 inhibi-
tion after replicative stress activates a double strand break response mediated by ATM and
DNA-dependent protein kinase. Cell Cycle 9(5):995–1004. Epub 2010/02/18
120. Sorensen CS, Syljuasen RG (2012) Safeguarding genome integrity: the checkpoint kinases
ATR, CHK1 and WEE1 restrain CDK activity during normal DNA replication. Nucleic
Acids Res 40(2):477–486. Epub 2011/09/23
121. Toledo LI, Murga M, Fernandez-Capetillo O (2011) Targeting ATR and Chk1 kinases for
cancer treatment: a new model for new (and old) drugs. Mol Oncol 5(4):368–373. Epub
2011/08/09
122. Xanthoudakis S, Smeyne RJ, Wallace JD, Curran T (1996) The redox/DNA repair pro-
tein, Ref-1, is essential for early embryonic development in mice. Proc Nat Acad Sci
93(17):8919–8923
123. Izumi T, Brown DB, Naidu CV, Bhakat KK, Macinnes MA, Saito H et al (2005) Two essen-
tial but distinct functions of the mammalian abasic endonuclease. Proc Natl Acad Sci USA
102(16):5739–5743
124. Fan Z, Beresford PJ, Zhang D, Xu Z, Novina CD, Yoshida A et al (2003) Cleaving the
oxidative repair protein Ape1 enhances cell death mediated by granzyme A. Nat Immunol
4(2):145–153
6 Targeting DNA Repair Pathways for Cancer Therapy 173
125. Fishel ML, He Y, Reed AM, Chin-Sinex H, Hutchins GD, Mendonca MS et al (2008)
Knockdown of the DNA repair and redox signaling protein Ape1/Ref-1 blocks ovarian can-
cer cell and tumor growth. DNA Repair (Amst) 7(2):177–186. Epub 2007/11/03
126. Bapat A, Fishel ML, Kelley MR (2009) Going ape as an approach to cancer therapeutics.
Antioxid Redox Signal 11(3):651–668. Epub 2008/08/22
127. Abbotts R, Madhusudan S (2010) Human AP endonuclease 1 (APE1): from mechanistic
insights to druggable target in cancer. Cancer Treat Rev 36(5):425–435. Epub 2010/01/09
128. Phillps JH, Brown DM, Grossman L (1996) The efficiency of induction of mutations by
hydroxylamine. J Mol Biol 21(3):405–419. Epub 1966/11/28
129. Goldszer F, Tindell GL, Walle UK, Walle T (1981) Chemical trapping of labile aldehyde
intermediates in the metabolism of propranolol and oxprenolol. Res Commun Chem Pathol
Pharmacol 34(2):193–205. Epub 1981/11/01
130. Beger RD, Bolton PH (1998) Structures of apurinic and apyrimidinic sites in duplex DNAs.
J Biol Chem 273(25):15565–15573. Epub 1998/06/23
131. Talpaert-Borle M, Liuzzi M (1983) Reaction of apurinic/apyrimidinic sites with [14C]
methoxyamine. A method for the quantitative assay of AP sites in DNA. Biochim Biophys
Acta 740(4):410–416. Epub 1983/09/09
132. Liu L, Taverna P, Whitacre CM, Chatterjee S, Gerson SL (1999) Pharmacologic disruption
of base excision repair sensitizes mismatch repair-deficient and -proficient colon cancer
cells to methylating agents. Clin Cancer Res 5(10):2908–2917
133. Goellner EM, Grimme B, Brown AR, Lin YC, Wang XH, Sugrue KF et al (2011)
Overcoming temozolomide resistance in glioblastoma via dual inhibition of NAD + biosyn-
thesis and base excision repair. Cancer Res 71(6):2308–2317. Epub 2011/03/17
134. Tang JB, Svilar D, Trivedi RN, Wang XH, Goellner EM, Moore B et al (2011)
N-methylpurine DNA glycosylase and DNA polymerase beta modulate BER inhibitor poten-
tiation of glioma cells to temozolomide. Neurooncology 13(5):471–486. Epub 2011/03/08
135. Taverna P, Liu L, Hwang HS, Hanson AJ, Kinsella TJ, Gerson SL (2001) Methoxyamine
potentiates DNA single strand breaks and double strand breaks induced by temozolomide in
colon cancer cells. Mutat Res 485(4):269–281
136. Taverna P, Hwang HS, Schupp JE, Radivoyevitch T, Session NN, Reddy G et al (2003)
Inhibition of base excision repair potentiates iododeoxyuridine-induced cytotoxicity and
radiosensitization. Cancer Res 63(4):838–846. Epub 2003/02/20
137. Liu L, Yan L, Donze JR, Gerson SL (2003) Blockage of abasic site repair enhances antitu-
mor efficacy of 1,3-bis-(2-chloroethyl)-1-nitrosourea in colon tumor xenografts. Mol Cancer
Ther 2(10):1061–1066. Epub 2003/10/28
138. She M, Pan I, Sun L, Yeung SC (2005) Enhancement of manumycin A-induced apoptosis by
methoxyamine in myeloid leukemia cells. Leukemia 19(4):595–602. Epub 2005/03/04
139. Bulgar AD, Snell M, Donze JR, Kirkland EB, Li L, Yang S et al (2010) Targeting base exci-
sion repair suggests a new therapeutic strategy of fludarabine for the treatment of chronic
lymphocytic leukemia. Leukemia 24(10):1795–1799. Epub 2010/09/03
140. Kinsella TJ (2009) Coordination of DNA mismatch repair and base excision repair process-
ing of chemotherapy and radiation damage for targeting resistant cancers. Clin Cancer Res
15(6):1853–1859. Epub 2009/02/26
141. Vermeulen C, Verwijs-Janssen M, Cramers P, Begg AC, Vens C (2007) Role for DNA poly-
merase beta in response to ionizing radiation. DNA Repair (Amst) 6(2):202–212
142. Bulgar AD, Weeks LD, Miao Y, Yang S, Xu Y, Guo C et al (2012) Removal of uracil by
uracil DNA glycosylase limits pemetrexed cytotoxicity: overriding the limit with methoxy-
amine to inhibit base excision repair. Cell Death Dis 3:e252. Epub 2012/01/13
143. Srinivasan A, Wang L, Cline CJ, Xie Z, Sobol RW, Xie XQ et al (2012) Identification and
characterization of human apurinic/apyrimidinic endonuclease-1 inhibitors. Biochemistry
51(31):6246–6259. Epub 2012/07/14
144. Rai G, Vyjayanti VN, Dorjsuren D, Simeonov A, Jadhav A, Wilson DM 3rd et al (2012)
Synthesis, biological evaluation, and structure-activity relationships of a novel class of apurinic/
apyrimidinic endonuclease 1 inhibitors. J Med chem 55(7):3101–3112. Epub 2012/03/30
174 C. Vens and R. W. Sobol
145. Bapat A, Glass LS, Luo M, Fishel ML, Long EC, Georgiadis MM et al (2010) Novel small-
molecule inhibitor of apurinic/apyrimidinic endonuclease 1 blocks proliferation and reduces
viability of glioblastoma cells. J Pharmacol Exp Ther 334(3):988–998. Epub 2010/05/28
146. Zawahir Z, Dayam R, Deng J, Pereira C, Neamati N (2009) Pharmacophore guided discov-
ery of small-molecule human apurinic/apyrimidinic endonuclease 1 inhibitors. J Med Chem
52(1):20–32
147. Al-Safi RI, Odde S, Shabaik Y, Neamati N (2012) Small-molecule inhibitors of APE1 DNA
repair function: an overview. Curr Mol Pharmacol 5(1):14–35. Epub 2011/11/30
148. Sultana R, McNeill DR, Abbotts R, Mohammed MZ, Zdzienicka MZ, Qutob H et al (2012)
Synthetic lethal targeting of DNA double-strand break repair deficient cells by human apu-
rinic/apyrimidinic endonuclease inhibitors. Int J Cancer 131(10):2433–2444. Epub 2012/03/02
149. Bartkova J, Horejsi Z, Koed K, Kramer A, Tort F, Zieger K et al (2005) DNA dam-
age response as a candidate anti-cancer barrier in early human tumorigenesis. Nature
434(7035):864–870. Epub 2005/04/15
150. Gorgoulis VG, Vassiliou LV, Karakaidos P, Zacharatos P, Kotsinas A, Liloglou T et al (2005)
Activation of the DNA damage checkpoint and genomic instability in human precancerous
lesions. Nature 434(7035):907–913. Epub 2005/04/15
151. Peasland A, Wang LZ, Rowling E, Kyle S, Chen T, Hopkins A et al (2011) Identification
and evaluation of a potent novel ATR inhibitor, NU6027, in breast and ovarian cancer cell
lines. Br J Cancer 105(3):372–381. Epub 2011/07/07
152. Toledo LI, Murga M, Zur R, Soria R, Rodriguez A, Martinez S et al (2011) A cell-based
screen identifies ATR inhibitors with synthetic lethal properties for cancer-associated muta-
tions. Nat Struct Mol Biol 18(6):721–727. Epub 2011/05/10
153. Rainey MD, Charlton ME, Stanton RV, Kastan MB (2008) Transient inhibition of ATM
kinase is sufficient to enhance cellular sensitivity to ionizing radiation. Cancer Res
68(18):7466–7474. Epub 2008/09/17
154. Vens C, Begg AC (2010) Targeting base excision repair as a sensitization strategy in radio-
therapy. Semin Radiat Oncol 20(4):241–249. Epub 2010/09/14
155. O’Connor MJ, Martin NM, Smith GC (2007) Targeted cancer therapies based on the inhibi-
tion of DNA strand break repair. Oncogene 26(56):7816–7824. Epub 2007/12/11
156. Nutley BP, Smith NF, Hayes A, Kelland LR, Brunton L, Golding BT et al (2005) Preclinical
pharmacokinetics and metabolism of a novel prototype DNA-PK inhibitor NU7026. Br J
Cancer 93(9):1011–1018. Epub 2005/10/27
157. Zhao Y, Thomas HD, Batey MA, Cowell IG, Richardson CJ, Griffin RJ et al (2006)
Preclinical evaluation of a potent novel DNA-dependent protein kinase inhibitor NU7441.
Cancer Res 66(10):5354–5362. Epub 2006/05/19
158. Quanz M, Berthault N, Roulin C, Roy M, Herbette A, Agrario C et al (2009) Small-
molecule drugs mimicking DNA damage: a new strategy for sensitizing tumors to radio-
therapy. Clin Cancer Res 15(4):1308–1316. Epub 2009/02/05
159. Munck JM, Batey MA, Zhao Y, Jenkins H, Richardson CJ, Cano C et al (2012)
Chemosensitization of cancer cells by KU-0060648, A Dual Inhibitor of DNA-PK and
PI-3 K. Mol Cancer Ther. Epub 2012/05/12
160. Veuger SJ, Curtin NJ, Richardson CJ, Smith GC, Durkacz BW (2003) Radiosensitization and
DNA repair inhibition by the combined use of novel inhibitors of DNA-dependent protein
kinase and poly(ADP-ribose) polymerase-1. Cancer Res 63(18):6008–6015. Epub 2003/10/03
161. Azad A, Jackson S, Cullinane C, Natoli A, Neilsen PM, Callen DF et al (2011) Inhibition of
DNA-dependent protein kinase induces accelerated senescence in irradiated human cancer
cells. Mol Cancer Res 9(12):1696–1707. Epub 2011/10/20
162. Gurley KE, Kemp CJ (2001) Synthetic lethality between mutation in ATM and DNA-PK(cs)
during murine embryogenesis. Curr Biol 11(3):191–194. Epub 2001/03/07
163. Taccioli GE, Amatucci AG, Beamish HJ, Gell D, Xiang XH, Torres Arzayus MI et al
(1998) Targeted disruption of the catalytic subunit of the DNA-PK gene in mice confers
severe combined immunodeficiency and radiosensitivity. Immunity 9(3):355–366. Epub
1998/10/13
6 Targeting DNA Repair Pathways for Cancer Therapy 175
182. Berger F, Lau C, Dahlmann M, Ziegler M (2005) Subcellular compartmentation and differ-
ential catalytic properties of the three human nicotinamide mononucleotide adenylyltrans-
ferase isoforms. J Biol Chem 280(43):36334–36341
183. Watson M, Roulston A, Belec L, Billot X, Marcellus R, Bedard D et al (2009) The small
molecule GMX1778 is a potent inhibitor of NAD + biosynthesis: strategy for enhanced
therapy in nicotinic acid phosphoribosyltransferase 1-deficient tumors. Mol Cell Biol
29(21):5872–5888. Epub 2009/08/26
184. Fleischer TC, Murphy BR, Flick JS, Terry-Lorenzo RT, Gao ZH, Davis T et al Chemical
proteomics identifies Nampt as the target of CB30865, an orphan cytotoxic compound.
Chem Biol 17(6):659–664. Epub 2010/07/09
185. Olesen UH, Christensen MK, Bjorkling F, Jaattela M, Jensen PB, Sehested M et al (2008)
Anticancer agent CHS-828 inhibits cellular synthesis of NAD. Biochem Biophys Res
Commun 367(4):799–804
186. Zhang RY, Qin Y, Lv XQ, Wang P, Xu TY, Zhang L et al (2011) A fluorometric assay for
high-throughput screening targeting nicotinamide phosphoribosyltransferase. Anal Biochem
412(1):18–25. Epub 2011/01/08
187. Khan JA, Tao X, Tong L (2006) Molecular basis for the inhibition of human NMPRTase, a
novel target for anticancer agents. Nat Struct Mol Biol 13(7):582–588
188. Hasmann M, Schemainda I (2003) FK866, a highly specific noncompetitive inhibitor of nic-
otinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor
cell apoptosis. Cancer Res 63(21):7436–7442
189. Billington RA, Genazzani AA, Travelli C, Condorelli F (2008) NAD depletion by FK866
induces autophagy. Autophagy 4(3):385–387. Epub 2008/01/30
190. Pogrebniak A, Schemainda I, Azzam K, Pelka-Fleischer R, Nussler V, Hasmann M (2006)
Chemopotentiating effects of a novel NAD biosynthesis inhibitor, FK866, in combination
with antineoplastic agents. Eur J Med Res 11(8):313–321
191. Bi TQ, Che XM, Liao XH, Zhang DJ, Long HL, Li HJ et al (2011) Overexpression of
nampt in gastric cancer and chemopotentiating effects of the nampt inhibitor FK866 in com-
bination with fluorouracil. Oncol Rep 26(5):1251–1257. Epub 2011/07/12
192. Travelli C, Drago V, Maldi E, Kaludercic N, Galli U, Boldorini R et al (2011) Reciprocal
potentiation of the antitumoral activities of FK866, an inhibitor of nicotinamide phospho-
ribosyltransferase, and etoposide or cisplatin in neuroblastoma cells. J Pharmacol Exp Ther
338(3):829–840. Epub 2011/06/21
193. Zoppoli G, Cea M, Soncini D, Fruscione F, Rudner J, Moran E et al (2010) Potent synergis-
tic interaction between the Nampt inhibitor APO866 and the apoptosis activator TRAIL in
human leukemia cells. Exp Hematol 38(11):979–988. Epub 2010/08/11
194. Olesen UH, Thougaard AV, Jensen PB, Sehested M (2010) A preclinical study on the rescue
of normal tissue by nicotinic acid in high-dose treatment with APO866, a specific nicotina-
mide phosphoribosyltransferase inhibitor. Mol Cancer Ther 9(6):1609–1617
195. Goellner EM, Svilar D, Almeida KH, Sobol RW (2012) Targeting DNA polymerase β for
therapeutic intervention. Curr Mol Pharmacol 5(1):68–87. Epub 2011/11/30
196. Martin LP, Hamilton TC, Schilder RJ (2008) Platinum resistance: the role of DNA repair
pathways. Clin Cancer Res 14(5):1291–1295. Epub 2008/03/05
197. Orelli B, McClendon TB, Tsodikov OV, Ellenberger T, Niedernhofer LJ, Scharer OD (2010)
The XPA-binding domain of ERCC1 is required for nucleotide excision repair but not other
DNA repair pathways. J Biol Chem 285(6):3705–3712. Epub 2009/11/27
198. Su Y, Orelli B, Madireddy A, Niedernhofer LJ, Scharer OD (2012) Multiple DNA binding
domains mediate the function of the ERCC1-XPF protein in nucleotide excision repair. J
Biol Chem 287(26):21846–21855. Epub 2012/05/02
199. Tsodikov OV, Ivanov D, Orelli B, Staresincic L, Shoshani I, Oberman R et al (2007)
Structural basis for the recruitment of ERCC1-XPF to nucleotide excision repair complexes
by XPA. Embo J 26(22):4768–4776. Epub 2007/10/20
200. Sijbers AM, van der Spek PJ, Odijk H, van den Berg J, van Duin M, Westerveld A et al
(1996) Mutational analysis of the human nucleotide excision repair gene ERCC1. Nucleic
Acids Res 24(17):3370–3380. Epub 1996/09/01
6 Targeting DNA Repair Pathways for Cancer Therapy 177
201. Barakat KH, Torin Huzil J, Luchko T, Jordheim L, Dumontet C, Tuszynski J (2009)
Characterization of an inhibitory dynamic pharmacophore for the ERCC1-XPA interaction
using a combined molecular dynamics and virtual screening approach. J Mol Graph Model
28(2):113–130. Epub 2009/05/29
202. Lee-Kwon W, Park D, Bernier M (1998) Involvement of the ras/extracellular signal-reg-
ulated kinase signalling pathway in the regulation of ERCC-1 mRNA levels by insulin.
Biochem J 331 (Pt 2):591–597. Epub 1998/06/11
203. Yadav A, Kumar B, Teknos TN, Kumar P (2011) Sorafenib enhances the antitumor effects
of chemoradiation treatment by downregulating ERCC-1 and XRCC-1 DNA repair proteins.
Mol Cancer Ther 10(7):1241–1251. Epub 2011/05/10
204. Burgers PM, Koonin EV, Bruford E, Blanco L, Burtis KC, Christman MF et al (2001)
Eukaryotic DNA polymerases: proposal for a revised nomenclature. J Biol Chem
276(47):43487–43490
205. Lange SS, Takata K, Wood RD (2011) DNA polymerases and cancer. Nat Rev Cancer
11(2):96–110. Epub 2011/01/25
206. Bebenek K, Kunkel TA (2004) Functions of DNA polymerases. Adv Protein Chem 69:137–
65. Epub 2004/12/14
207. Wilson SH, Beard WA, Shock DD, Batra VK, Cavanaugh NA, Prasad R et al (2010) Base
excision repair and design of small molecule inhibitors of human DNA polymerase beta.
Cell Mol Life Sci 67(21):3633–3647. Epub 2010/09/17
208. Barakat KH, Gajewski MM, Tuszynski JA (2012) DNA polymerase beta (pol beta)
inhibitors: a comprehensive overview. Drug discovery today 17(15–16):913–920. Epub
2012/05/09
209. Higgins GS, Prevo R, Lee YF, Helleday T, Muschel RJ, Taylor S et al (2010) A small inter-
fering RNA screen of genes involved in DNA repair identifies tumor-specific radiosensitiza-
tion by POLQ knockdown. Cancer Res 70(7):2984–2993. Epub 2010/03/18
210. Higgins GS, Harris AL, Prevo R, Helleday T, McKenna WG, Buffa FM (2010)
Overexpression of POLQ confers a poor prognosis in early breast cancer patients.
Oncotarget 1(3):175–184. Epub 2010/08/12
211. Lemee F, Bergoglio V, Fernandez-Vidal A, Machado-Silva A, Pillaire MJ, Bieth A et al
(2010) DNA polymerase theta up-regulation is associated with poor survival in breast can-
cer, perturbs DNA replication, and promotes genetic instability. Proc Natl Acad Sci USA
107(30):13390–13395. Epub 2010/07/14
212. Slupphaug G, Mol CD, Kavli B, Arvai AS, Krokan HE, Tainer JA (1996) A nucleotide-
flipping mechanism from the structure of human uracil-DNA glycosylase bound to DNA.
Nature 384(6604):87–92. Epub 1996/11/07
213. Engelward BP, Allan JM, Dreslin AJ, Kelly JD, Wu MM, Gold B et al (1998) A chemi-
cal and genetic approach together define the biological consequences of 3-methyladenine
lesions in the mammalian genome. J Biol Chem 273(9):5412–5418
214. Noren Hooten N, Kompaniez K, Barnes J, Lohani A, Evans MK (2011) Poly(ADP-ribose)
polymerase 1 (PARP-1) binds to 8-oxoguanine-DNA glycosylase (OGG1). J Biol Chem
286(52):44679–44690. Epub 2011/11/08
215. Endres M, Biniszkiewicz D, Sobol RW, Harms C, Ahmadi M, Lipski A et al (2004)
Increased postischemic brain injury in mice deficient in uracil-DNA glycosylase. J Clin
Invest 113(12):1711–1721
216. Kruman II, Schwartz E, Kruman Y, Cutler RG, Zhu X, Greig NH et al (2004)
Suppression of uracil-DNA glycosylase induces neuronal apoptosis. J Biol Chem
279(42):43952–43960
217. Imai K, Slupphaug G, Lee WI, Revy P, Nonoyama S, Catalan N et al (2003) Human uracil-
DNA glycosylase deficiency associated with profoundly impaired immunoglobulin class-
switch recombination. Nat Immunol 4(10):1023–1028. Epub 2003/09/06
218. Wang Z, Mosbaugh DW. Uracil-DNA glycosylase inhibitor gene of bacteriophage
PBS2 encodes a binding protein specific for uracil-DNA glycosylase. J Biol Chem.
1989;264(2):1163-71. Epub 1989/01/15
178 C. Vens and R. W. Sobol
219. Komander D, Clague MJ, Urbe S (2009) Breaking the chains: structure and function of the
deubiquitinases. Nat Rev Mol Cell Biol 10(8):550–563. Epub 2009/07/25
220. Shabek N, Ciechanover A (2010) Degradation of ubiquitin: the fate of the cellular reaper.
Cell Cycle 9(3):523–530. Epub 2010/01/29
221. Mattern MR, Wu J, Nicholson B (2012) Ubiquitin-based anticancer therapy: carpet bombing
with proteasome inhibitors vs surgical strikes with E1, E2, E3, or DUB inhibitors. Biochim
Biophys Acta 1823(11):2014–2021. Epub 2012/05/23
222. Opferman JT, Green DR (2010) DUB-le trouble for cell survival. Cancer Cell 17(2):117–
119. Epub 2010/02/18
223. Altun M, Kramer HB, Willems LI, McDermott JL, Leach CA, Goldenberg SJ et al (2011)
Activity-based chemical proteomics accelerates inhibitor development for deubiquitylating
enzymes. Chem Biol 18(11):1401–1412. Epub 2011/11/29
224. Kapuria V, Peterson LF, Fang D, Bornmann WG, Talpaz M, Donato NJ (2010)
Deubiquitinase inhibition by small-molecule WP1130 triggers aggresome formation and
tumor cell apoptosis. Cancer Res 70(22):9265–9276. Epub 2010/11/04
225. Kramer HB, Nicholson B, Kessler BM, Altun M (2012) Detection of ubiquitin-proteasome
enzymatic activities in cells: application of activity-based probes to inhibitor development.
Biochim Biophys Acta 1823(11):2029–2037. Epub 2012/05/23
226. Seiberlich V, Goldbaum O, Zhukareva V, Richter-Landsberg C (2012) The small molecule
inhibitor PR-619 of deubiquitinating enzymes affects the microtubule network and causes
protein aggregate formation in neural cells: implications for neurodegenerative diseases.
Biochim Biophys Acta 1823(11):2057–2068. Epub 2012/05/09
227. Chernikova SB, Game JC, Brown JM (2012) Inhibiting homologous recombination for can-
cer therapy. Cancer Biol Ther 13(2):61–68. Epub 2012/02/18
228. Huang F, Motlekar NA, Burgwin CM, Napper AD, Diamond SL, Mazin AV (2011)
Identification of specific inhibitors of human RAD51 recombinase using high-throughput
screening. ACS chemical biology 6(6):628–635. Epub 2011/03/25
229. Quiros S, Roos WP, Kaina B (2011) Rad51 and BRCA2–New molecular targets for sensitiz-
ing glioma cells to alkylating anticancer drugs. PLoS ONE 6(11):e27183. Epub 2011/11/11
230. Short SC, Giampieri S, Worku M, Alcaide-German M, Sioftanos G, Bourne S et al (2011)
Rad51 inhibition is an effective means of targeting DNA repair in glioma models and
CD133 + tumor-derived cells. Neurooncology 13(5):487–499. Epub 2011/03/03
231. Sprong D, Janssen HL, Vens C, Begg AC (2005) Resistance of hypoxic cells to ionizing
radiation is influenced by homologous recombination status. Int J Radiat Oncol Biol Phys
64(2):562–572. Epub 2005/12/14
232. Neijenhuis S, Verwijs-Janssen M, van den Broek LJ, Begg AC, Vens C (2010) Targeted
radiosensitization of cells expressing truncated DNA polymerase {beta}. Cancer Res
70(21):8706–8714. Epub 2010/10/28
233. Slade D, Dunstan MS, Barkauskaite E, Weston R, Lafite P, Dixon N et al (2011) The
structure and catalytic mechanism of a poly(ADP-ribose) glycohydrolase. Nature
477(7366):616–620. Epub 2011/09/06
234. Brochu G, Duchaine C, Thibeault L, Lagueux J, Shah GM, Poirier GG (1994) Mode of
action of poly(ADP-ribose) glycohydrolase. Biochim Biophys Acta 1219(2):342–350
235. Affar EB, Germain M, Winstall E, Vodenicharov M, Shah RG, Salvesen GS et al (2001)
Caspase-3-mediated processing of poly(ADP-ribose) glycohydrolase during apoptosis. J
Biol Chem 276(4):2935–42. Epub 2000/10/29
236. Li Q, Li M, Wang YL, Fauzee NJ, Yang Y, Pan J et al (2012) RNA interference of PARG
could inhibit the metastatic potency of colon carcinoma cells via PI3-kinase/Akt pathway.
Cell Physiol Biochem Int J Exp Cell Physiol Biochem Pharmacol 29(3–4):361–72. Epub
2012/04/18
237. Koh DW, Lawler AM, Poitras MF, Sasaki M, Wattler S, Nehls MC et al (2004) Failure to
degrade poly(ADP-ribose) causes increased sensitivity to cytotoxicity and early embryonic
lethality. Proc Natl Acad Sci USA 101(51):17699–17704
6 Targeting DNA Repair Pathways for Cancer Therapy 179
255. Kummar S, Ji J, Morgan R, Lenz HJ, Puhalla SL, Belani CP et al (2012) A phase I study of
veliparib in combination with metronomic cyclophosphamide in adults with refractory solid
tumors and lymphomas. Clin Cancer Res 18(6):1726–1734. Epub 2012/02/07
256. Barreto-Andrade JC, Efimova EV, Mauceri HJ, Beckett MA, Sutton HG, Darga TE
et al (2011) Response of human prostate cancer cells and tumors to combining PARP inhibi-
tion with ionizing radiation. Mol Cancer Ther 10(7):1185–1193. Epub 2011/05/17
257. Efimova EV, Mauceri HJ, Golden DW, Labay E, Bindokas VP, Darga TE et al (2010)
Poly(ADP-ribose) polymerase inhibitor induces accelerated senescence in irradiated breast
cancer cells and tumors. Cancer Res 70(15):6277–6282. Epub 2010/07/09
258. Nowsheen S, Bonner JA, Yang ES (2011) The poly(ADP-Ribose) polymerase inhibitor
ABT-888 reduces radiation-induced nuclear EGFR and augments head and neck tumor
response to radiotherapy. Radiother Oncol 99(3):331–338. Epub 2011/07/02
259. Apisarnthanarax N, Wood GS, Stevens SR, Carlson S, Chan DV, Liu L et al (2012) Phase
I clinical trial of O6-benzylguanine and topical carmustine in the treatment of cutane-
ous T-cell lymphoma, mycosis fungoides type. Arch Dermatol 148(5):613–620. Epub
2012/01/18
260. Warren KE, Gururangan S, Geyer JR, McLendon RE, Poussaint TY, Wallace D et al (2012)
A phase II study of O6-benzylguanine and temozolomide in pediatric patients with recur-
rent or progressive high-grade gliomas and brainstem gliomas: a pediatric brain tumor con-
sortium study. J Neurooncol 106(3):643–649. Epub 2011/10/05
261. Kefford RF, Thomas NP, Corrie PG, Palmer C, Abdi E, Kotasek D et al (2009) A phase I
study of extended dosing with lomeguatrib with temozolomide in patients with advanced
melanoma. Br J Cancer 100(8):1245–1249. Epub 2009/04/16
262.
Watson AJ, Middleton MR, McGown G, Thorncroft M, Ranson M, Hersey P
et al (2009) O(6)-methylguanine-DNA methyltransferase depletion and DNA damage in
patients with melanoma treated with temozolomide alone or with lomeguatrib. Br J Cancer
100(8):1250–1256. Epub 2009/04/16
263. Ma Z, Yao G, Zhou B, Fan Y, Gao S, Feng X (2012) The Chk1 inhibitor AZD7762 sensitises
p53 mutant breast cancer cells to radiation in vitro and in vivo. Mol Med Rep 6(4):897–903.
Epub 2012/07/25
264. Montano R, Chung I, Garner KM, Parry D, Eastman A (2011) Preclinical development of
the novel Chk1 inhibitor SCH900776 in combination with DNA-damaging agents and anti-
metabolites. Mol Cancer Ther 11(2):427–438. Epub 2011/12/29
265. Ferrao PT, Bukczynska EP, Johnstone RW, McArthur GA (2012) Efficacy of CHK inhibi-
tors as single agents in MYC-driven lymphoma cells. Oncogene 31(13):1661–1672. Epub
2011/08/16
266. Hickson I, Zhao Y, Richardson CJ, Green SJ, Martin NM, Orr AI et al (2004) Identification
and characterization of a novel and specific inhibitor of the ataxia-telangiectasia mutated
kinase ATM. Cancer Res 64(24):9152–9159. Epub 2004/12/18
267. Leahy JJ, Golding BT, Griffin RJ, Hardcastle IR, Richardson C, Rigoreau L et al (2004)
Identification of a highly potent and selective DNA-dependent protein kinase (DNA-
PK) inhibitor (NU7441) by screening of chromenone libraries. Bioorg Med Chem Lett
14(24):6083–6087
268. Devun F, Bousquet G, Biau J, Herbette A, Roulin C, Berger F et al (2012) Preclinical study
of the DNA repair inhibitor Dbait in combination with chemotherapy in colorectal cancer. J
gastroenterol 47(3):266–275. Epub 2011/11/10
269. Kim CH, Park SJ, Lee SH (2002) A targeted inhibition of DNA-dependent protein
kinase sensitizes breast cancer cells following ionizing radiation. J Pharmacol Exp Ther
303(2):753–759. Epub 2002/10/22
Chapter 7
Molecular Chaperones and How Addiction
Matters in Cancer Therapy
To allow for normal functioning, cells use intricate molecular machineries com-
prised of thousands of proteins programmed to execute well-defined functions.
Dysregulation in these pathways may lead to altered functions that confer a patho-
logic phenotype. In cancer, not one but multiple pathways and molecules lose their
inherited state leading to cells characterized by many defects such as aberrant pro-
liferation, cell cycle, invasive potential, and evasion of apoptosis [1]. While at the
M. L. Guzman
Division of Hematology and Oncology, Weill Cornell Medical College, NY, USA
M. A. Lowery
Department of Medicine, Gastrointestinal Service,
Memorial Sloan-Kettering Cancer Center, NY, USA
T. Taldone· J. Koren III· E. D. Gomes· G. Chiosis (*)
Department of Medicine, Breast Cancer Service,
Memorial Sloan-Kettering Cancer Center, NY, USA
e-mail: [email protected]
T. Taldone· J. Koren III· E. D. Gomes· G. Chiosis
Department of Molecular Pharmacology and Chemistry, Sloan-Kettering Institute, NY, USA
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 181
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_7,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
182 M. L. Guzman et al.
The major cancer HSPs are the HSP90 and HSP70 family of chaperones. In
spite of several recent efforts to target HSP70, HSP90 remains to date the only
one with a timely promise of delivering a new anticancer therapy [9, 10]. It is
the most studied and perhaps understood cancer HSP family with four main
known mammalian paralogs, the cytoplasmic heat shock protein 90 alpha and
beta (HSP90α and β) [11], the endoplasmic reticulum (ER) glucose-regulated
protein94 (GRP94) [12], and the mitochondrial tumor necrosis factor receptor–
associated protein-1 (TRAP-1) [13]. Each is known to be highly expressed in
cancer, but while HSP90α/β have been widely investigated, and important roles
assigned to them in maintaining the functional conformation of a large number
of aberrant malignancy-driving proteins, little is known on the roles of GRP94
and TRAP-1 [14, 15]. Because most small molecules used to investigate the
7 Molecular Chaperones and How Addiction Matters in Cancer Therapy 183
role of HSP90 are pan-HSP90 inhibitors and few studies differentiate the role of
HSP90α from that of HSP90β, we will here refer to HSP90 without delineating
the role of individual paralogs.
HSP90 is a member of the GHKL family, proteins characterized by an atypi-
cal fold in their ATP-binding regulatory pocket, referred to as the Bergerat fold
[16]. Its structure consists of three domains: mainly an amino-terminal region
(N-domain) that contains the ATP pocket and co-chaperone-interacting motifs,
a middle domain that provides docking sites for client proteins and co-chaper-
ones and that participates in forming the active ATPase, and finally a carboxy-
terminal domain that contains a dimerization motif, a second ligand-binding
region and interaction sites for other co-chaperones [17, 18]. Dimerization of
two HSP90 molecules through their C-domains is believed to be necessary for
chaperone function.
HSP90 function is regulated by a plethora of co-chaperones, of which
approximately 20 have been identified to date. A full list of these co-chaperones
has recently been reviewed by Johnson [6]. Many of these co-chaperones com-
pete for the same binding site on HSP90 or bind alternate HSP90 conforma-
tions, and thus, distinct HSP90-co-chaperone complexes likely exist at any time.
Several of these co-chaperones have tetratricopeptide (TPR) domains and bind
to the MEEVD acceptor site on HSP90. Others, such as SGT1 and GCUNC-
45, bind to a sequence in the N-terminal domain of HSP90. Most have no effect
on the ATPase activity of HSP90, while others stimulate (i.e., AHA1) or inhibit
(i.e., p23, Cdc37) it. Co-chaperones also regulate the conformation of HSP90,
such as p23 that is believed to stabilize the closed conformation, and control the
chaperoning of client proteins. Cdc37, for example, pre-binds clients that are
kinases and delivers them to HSP90, whereas HSP-organizing protein (HOP)
presents HSP70-bound clients to HSP90.
HSP90 interacts with and regulates its client proteins in a cycle that is driven
by the binding and hydrolysis of ATP. Through this catalytic cycle, HSP90
undergoes considerable structural changes, and this dynamic nature of HSP90
is key to its ability to function as a chaperone [19]. This conformational flux is
constantly altered by the binding of regulatory nucleotides (i.e., ATP/ADP) and
co-chaperones (i.e., HOP, Cdc37, p23, AHA1, and immunophilins) and regu-
lated in part by a series of tyrosine phosphorylation events.
Because HSP90 activity is intrinsically linked to its conformation, which
is in turn dependent on the binding and release of ATP/ADP, co-chaperones,
and client proteins, regulating the HSP90 cycle offers several ways of interfer-
ing with HSP90 chaperoning function. Specifically, small molecules that bind
to the regulatory ATP/ADP pocket, interfere with binding of its co-chaperones
or with the conformational flexibility of HSP90, have all been shown to lead
to HSP90-mediated activities in cancer cells [10]. To date, most advanced in
development are those inhibitors that bind to the N-terminal regulatory pocket,
with several in clinical evaluation in cancers [20, 21]. Section 7.5 will detail
these efforts.
184 M. L. Guzman et al.
Fig. 7.2 a Within normal cells, constitutive expression of HSP90 is required for its evolutionar-
ily conserved housekeeping function of folding and translocating cellular proteins to their proper
cellular compartment (“housekeeping complex”). Upon malignant transformation, cellular pro-
teins are perturbed through mutations, hyperactivity, and retention in incorrect cellular compart-
ments or other means. The presence of these functionally altered proteins is required to initiate
and maintain the malignant phenotype, and it is these oncogenic proteins that are specifically
maintained by a subset of stress-modified HSP90 (“oncogenic complex”). Certain HSP90 inhibi-
tors specifically bind to the “oncogenic complex”. b By acting specifically on the “oncogenic
HSP90”, pharmacologic HSP90 inhibition is more efficient than genetic means at lowering
HSP90 function below the threshold necessary for cancer cell survival
Interest on the target potential of HSP90 grew, however, after the serendipi-
tous discovery of a natural product, geldanamycin (GM) [25]. Found in a screen
searching for compounds able to revert the phenotype of cells transfected with the
v-src oncogene, it was later demonstrated to do so by specifically binding to the
N-terminal regulatory pocket of HSP90 and, by this, to inhibit its function. While
surprising in light of the available genetic data, low concentrations of this natural
product were active on many cancer cells and induced differentiation, reduced cell
186 M. L. Guzman et al.
Now that we understand that HSP90 chaperones a significant portion of the altered
cancer proteome, we will detail below the nature of such proteome. Significant
research over the past 30 years has identified that HSP90 regulates many cancer pro-
teins and protein networks involved in signaling, cell cycle and proliferation, DNA
7 Molecular Chaperones and How Addiction Matters in Cancer Therapy 187
Fig. 7.3 Mechanisms by which HSP90 chaperones the cancer proteome. Cancer cells require
HSP90 to regulate the folding and stability of several cancer-driving proteins (a) and to facilitate
the complex formation of molecules involved in aberrantly activated complexes (b, c). In these
cases, HSP90 acts a scaffolding molecule that maintains protein complexes in an active configu-
ration (b) or as a facilitator of protein complex assembly (c)
damage and repair, transcriptional and translational activity, and epigenetic regulation.
These, while potentially partly dependent on HSP90 in normal cells, may become
addicted to HSP90 for their function in the course of cell progression from a normal
to a cancerous state. In such cases, the cancer cells require HSP90 to regulate the fold-
ing and stability of overexpressed (i.e., HER2), mutated (i.e., mutant B-Raf , mutant
FLT3), activated (i.e., AKT), or chimeric proteins (i.e., BCR-ABL ) [28] (Fig. 7.3a).
Recent studies have started to unveil additional roles for HSP90, such as to facilitate
complex formation of molecules involved in aberrantly activated complexes. In these
cases, HSP90 acts as a scaffolding molecule that maintains protein complexes in an
active configuration (i.e., STAT5) [27] (Fig. 7.3b) or as a facilitator of protein com-
plex assembly (i.e., RNA polymerase II, RISC) [29] (Fig. 7.3c).
the proteins that require HSP90 for this activity are several kinases (Sect. 7.4.2),
transcription factors (Sect. 7.4.3), epigenetic regulators (Sect. 7.4.4), and several
proteins involved in DNA repair (Sect. 7.4.5). In general, inhibition of HSP90
leads to a reduction in the steady-state levels of these proteins. The major path-
way by which the cell degrades these HSP90 client proteins is the proteasome.
Specifically, upon HSP90 inhibition, an E3 ligase is recruited to the complex, and
upon ubiquitination, the protein is directed to the proteasome for degradation,
thus short-circuiting further cycles of HSP90-mediated refolding [22, 28, 32].
HSP90, therefore, sits at the crossroad of folding/stabilization and degradation
pathways, with small molecule inhibitors pushing the equilibrium strongly toward
the degradative fate.
including the steroid receptors, the STAT family, and p53, appear to require
HSP90 to facilitate increased signaling and transcriptional robustness.
The tumor suppressor p53 is another transcription factor that requires HSP90.
Somatic mutations in p53 are found in many types of human tumors and often
result in the inactivation of its tumor suppressor function. These mutations result in
conformation changes and prolonged protein half-life that require HSP90 for their
stabilization [35]. Furthermore, binding of HSP90 to mutant p53 inhibits the ability
of MDM2 to promote p53 ubiquitination and degradation, leading to the stabiliza-
tion of both mutant p53 and MDM2 [42, 43]. Interestingly, while HSP90 associates
with both the wild-type and the mutated p53, inhibition of HSP90 upregulates the
wild-type protein but downregulates the mutant one, suggesting a distinct role for
HSP90 in the regulation of the normal (wild-type) and aberrant (mutant) p53 [44].
7.4.3.3 HIF-1α
HSP90 has also been shown to interact with proteins involved in chromatin regu-
lation by modifying histone methylation, including EZH2, PRMT5, CARM1,
SMYD3, and SMYD2. EZH2 is the catalytic core in the polycomb repressor com-
plex 2 (PRC2) that is involved in the trimethylation of histone-3 lysine-27 (H3K27),
resulting in chromatin condensation and gene silencing [51]. EZH2 is aberrantly
7 Molecular Chaperones and How Addiction Matters in Cancer Therapy 191
regulated in a wide range of cancer types, and it is involved in stem cell mainte-
nance and tumorigenesis [52, 53]. Thus, disruption of EZH2 is of interest as a thera-
peutic strategy for cancer. HSP90 was recently shown to regulate EZH2 in leukemia
cells, with its inhibition leading to decreased EZH2 protein expression, suggesting
that HSP90 is required to maintain the stability of EZH2 [54]. PRMT5 is a protein
arginine methyltransferases (PRMTs) involved in arginine methylation of histones
and other proteins that play a role in the regulation of major signaling pathways
that control survival and malignant transformation [55]. PRMT5 is a client protein
of HSP90 in ovarian cancer cells, and PRMT5 protein levels decreased upon treat-
ment with an HSP90 inhibitor [56]. PRMT4/CARM1 (co-activator-associated argi-
nine methyltransferase 1), an enzyme that catalyzes the transfer of a methyl groups
to arginine residues resulting in the activation of signal transduction cascades and
transcriptional activation [57], has been found to be deregulated in tumor cells [58].
CARM1 was recently reported to be a client protein of HSP90 in leukemia cells,
and this interaction shown to be critical for their survival [27]. The SMYD (SET and
MYND domain) family of lysine methyltransferases (KMTs) plays pivotal roles in
various cellular processes, including gene expression regulation and DNA damage
response. Initially identified as genuine histone methyltransferases, specific mem-
bers of this family have recently been shown to methylate non-histone proteins such
as p53 , VEGFR, and the retinoblastoma tumor suppressor (pRb). Both SMYD3 and
SMYD2 (SET and MYND domain–containing protein 3) are histone methyltrans-
ferases that interact with HSP90 [59, 60]. HSP90 inhibition decreased the expression
of SMYD3 and inhibited migration and proliferation of breast cancer cells. [60]
The DNA damage response refers to a series of tightly regulated, complex cellular
mechanisms that repair DNA breaks occurring through normal cellular metabolism or
environmental exposures. These pathways include base excision repair (BER), nucleo-
tide excision repair (NER), mismatch repair (MMR), non-homologous end-joining
(NHEJ), and homologous recombination (HR). The choice of repair pathway used is
determined by the type of lesion and by the expression of repair proteins at specific
cell cycle phases to ensure that the most accurate repair is performed [61]. DNA dou-
ble-strand breaks (DSBs), which pose the most significant threat to genomic integrity,
are repaired by two major DNA repair pathways, NHEJ and HR. Deficiency in the
HR pathway results in repair of all DSBs by the error-prone NHEJ pathway leading to
the accumulation of chromosomal aberrations. All steps of the DNA damage response
require the recruitment and modification of multiple key proteins; several of which
have been proposed as potential client proteins of the molecular chaperone HSP90
[62]. Several proteins crucial to the HR and NHEJ pathways for repair of DSBs
including BRCA2, FANCA, ATR, CHK1, and DNA-PK are also HSP90-chaperoned
[63–65]. Interestingly, HSP90 can be modified during the DNA damage response.
HSP90 is phosphorylated by DNA-PK, and both phosphorylated HSP90 and DNA-PK
192 M. L. Guzman et al.
have been found to co-localize to the H2AX apoptotic ring that contains activated
ATM, CHK2, and H2AX [65, 66]. HSP90 inhibition may therefore promote the deg-
radation of proteins involved in DNA repair and thereby potentiate DNA damage sus-
tained by ionizing radiation (IR) or chemotherapy. These observations are supported
by the enhancement of radiosensitivity of multiple human cancer cell lines including
glioblastoma, breast, colon, sarcoma, and pancreatic cancer cell lines by treatment
with inhibitors of HSP90 [67, 68].
In addition to regulating protein stability and folding, HSP90 has been recently shown
to act as a scaffolding molecule that increases the competency of protein complexes
involved in signaling and in transcription (Fig. 7.3b). Specifically, in chronic myeloid
leukemia (CML), HSP90 was found to facilitate increased STAT5 signaling by binding
to and influencing the conformation of STAT5 and by maintaining STAT5 in an active
conformation directly within STAT5-containing transcriptional complexes [27]. In the
cytosol, HSP90 binding to STAT5 modulated the conformation of the protein to alter
STAT5 phosphorylation and dephosphorylation kinetics. In addition, HSP90 maintained
STAT5 in an active conformation in STAT5-containing transcriptional complexes.
Unlike the classic HSP90 clients that require HSP90 for stability (Sect. 7.4.1), HSP90
inhibition led to inhibition but not degradation of STAT5.
Recently, HSP90 has also been identified as a critical protein in promoting and
maintaining the assembly of several cancer-related multi-protein complexes
(Fig. 7.3c). The involvement of HSP90 in these complexes varies, and in some, it
relates to stabilizing an unstable protein subunit and facilitating its incorporation into
the mega-complex. In other cases, HSP90 promotes a change in the composition of
a given complex. In any case, HSP90 is no longer present in the final assembled
complex. Such effects of the chaperone on the assembly of the following seven com-
plexes: snoRNP, RNA polymerase II, phosphatidylinositol-3 kinase–related protein
kinase (PIKK), telomere complex, kinetochore, RNA-induced silencing complexes
(RISC), and 26S proteasome was recently detailed in a review article [29].
While the above studies led to the identification of important HSP90 client
proteins, it is clear that due to the complexity of the altered proteome in any given
cancer cell, large proteomic and genomic analyses combined with bioinformatic
7 Molecular Chaperones and How Addiction Matters in Cancer Therapy 193
bound preferentially to the fraction of tumor HSP90 associated with altered client
proteins (i.e., “oncogenic HSP90”; Fig. 7.2a) and moreover, upon binding, locked
HSP90 in an onco-client bound configuration. Together these features greatly facili-
tated the chemical affinity purification of tumor-associated protein clients by mass
spectrometry, providing tumor-by-tumor global insights into the HSP90-sheltered
proteome, including in primary patient specimens. Top scoring networks identified
by this method to be HSP90-sheltered in CML were those used by BCR-ABL to
propagate aberrant signaling: PI3K /AKT/mTOR -, Raf -MAPK-, NFκB-, STAT5-,
and FAK-mediated signaling pathways. Other important transforming pathways in
CML, driven by MYC and TGF-β as well as others involved in disease progres-
sion and aberrant cell cycle and proliferation of CML, were identified. In addition
to signaling proteins, proteins that regulate carbohydrate and lipid metabolism, pro-
tein synthesis, epigenetics, gene expression, and cellular assembly and organization
were identified, in accord with the postulated broad roles of HSP90 as an impor-
tant mediator of cell transformation. The method also identified the histone-arginine
methyltransferase CARM1 as a novel HSP90 interactor, implicating the HSP90-
facilitated CARM1 activity in CML leukemogenesis.
As delineated in Sect. 7.4, HSP90 shelters a complex proteome vital for cancer
progression and maintenance. The HSP90-chaperoned networks and pathways
differ from tumor-to-tumor, depending on the molecular wiring of the alterations
that characterize a specific tumor. These are appealing features for a cancer target,
because by inhibiting one protein, HSP90, one could have beneficial effects in a
variety of tumors. Such effects were not appreciated early on when HSP90 was
discovered, because as indicated above, HSP90 is an abundant protein expressed
in most human cells. However, its persuasive roles in regulating the malignant pro-
teome, the observation that it can be easily pharmacologically modulated by small
molecule ligands, and findings demonstrating the differences between tumor and
normal cell HSP90 have together given it credibility as a cancer target. Indeed, at
the moment, more than 20 inhibitors have been or still are in clinical testing, with
many more coming in the pipeline (Fig. 7.4) [20, 21].
As indicated, the first compounds identified as HSP90 inhibitors were those
that modulate HSP90 chaperone activity by inhibiting the N-terminal domain ATP-
binding site [25, 72]. Targeted approaches such as structure-based drug design,
biochemical and cell-based screening, virtual screening, fragment-based drug
design, and medicinal chemistry have since led to the identification and devel-
opment of several novel inhibitors that act by similar or alternate mechanisms to
inhibit HSP90 function [10]. The molecules currently advanced to clinical trials
are those that bind to the regulatory pocket. These constitute a diverse array of
structures, which at a close inspection classify according to their similarity to GM
(Sect. 7.5.1), to the purine-scaffold (Sect. 7.5.2), or to radicicol (Sect. 7.5.3).
7 Molecular Chaperones and How Addiction Matters in Cancer Therapy 195
Ansamycins Purine-scaffold
benzoquinone NH2 Br O
O Cl- N
N
NH2 I
R H H OH O S O
O N N N N
C17 O N
S O
N
H N N N
O H
OH
OH H3 CO
H3CO OH H3 CO N
H3CO HN O
OCONH2
MPC-3100
OCONH2
PU-H71 HO
Geldanamycin R= -OMe IPI-504
17-AAG R= -NHCH2 CHCH2 NH2 N Cl
17-DMAG R= -NHCH2 CH 2 N(CH 3) 2 N O
N N
IPI-493 R= -NH2 N
S
N O N
H2N N
Resorcinols O
N N
O NH CH3
Cl
HO
H 3C OCH3
O HO
O O
Debio 0932 BIIB021
OH O N
H
OH O N
Radicicol Other
NVP-AUY922
H2N O
N
H
N
O
N
NH2
HO O O
N
N O N
CH3 O
OH O O N
F3C
N NH
HO O O SNX-5422
AT13387 N
O H2N N
O
O H3CO N
N F
OH O
N
KW-2478
HO H2N
N O NVP-HSP990
N
O
NH
OH N NH
XL888
HN
STA-9090 O
7.5.1 Geldanamycin Derivatives
The availability of crystal structures of ADP, GM, and radicicol bound to the
N-terminal nucleotide-binding domain of HSP90 and the realization that HSP90
possesses a unique ATP-binding pocket made the rational design of synthetic
inhibitors possible [10]. As mentioned, HSP90 is part of a small protein family
called the GHKL (Gyrase, HSP90, histidine kinases, and MutL) family, which
contains a unique fold in their regulatory pocket region, called the Bergerat fold
[16], a feature that portends selectivity for its ligands over other ATP-binding
pockets. Taking advantage of the distinct shape of the ATP pocket, Chiosis et al.
[72] designed PU3 as the first reported synthetic inhibitor. PU3, a purine-scaf-
fold compound, maintained the purine core of the endogenous ligands. This was
linked at the 8-position to an aryl moiety via a methylene linker (Purine-CH2-
Aryl). This essential motif was maintained, with some variations, for essentially
all the purine-scaffold HSP90 inhibitors (Fig. 7.4 purine-scaffold). From this
interesting lead, a large effort from multiple groups resulted in four molecules
to enter clinical evaluation in cancers: PU-H71, BIIB021, MPC-3100, and Debio
0932 (formerly CUDC-305) [20, 21].
7 Molecular Chaperones and How Addiction Matters in Cancer Therapy 197
The compounds within this class are distinguished by the presence of a resorcinol
moiety, and for this, they are related to radicicol (Fig. 7.4 resorcinols), a natural
product inhibitor of HSP90 isolated from Monosporium bonorden. Although a
potent HSP90 inhibitor, radicicol is unstable in vivo, and no attempts to directly
modify its structure have resulted in a clinical candidate. However, a number of
agents currently investigated maintain the resorcinol moiety. One of these, NVP-
AUY922, was developed from a lead molecule discovered during a high-through-
put screen designed to measure the ability of a compound to inhibit yeast HSP90
ATPase activity [10, 21, 22]. AT13387, on the other hand, was developed using
a fragment-based approach using NMR screening and X-ray crystallography.
A number of other novel resorcinol compounds are currently being evaluated in
clinical trials, including STA-9090 (ganetespib), KW-2478, and DS2248 [20, 21].
7.7 Conclusion
HSPs and HSP90, in particular, have emerged as important targets in cancer due to
their ability to buffer wide proteome alterations such as are characteristic in malig-
nant transformation. While not easily accepted initially as potential cancer targets,
several studies now spanning almost three decades have revealed important dif-
ferences in the biochemical and functional roles of HSP90 in cancer cells that are
different from those in normal cells. These findings, together with the now almost
explosive interest in the discovery and development of small molecule HSP90
inhibitors, and with the encouraging clinical studies with these agents, have
cemented HSP90 as a cancer target. The journey is, however, yet to be complete.
7 Molecular Chaperones and How Addiction Matters in Cancer Therapy 199
Several issues, including the lack of a biomarker for patient selection and the ina-
bility to measure target inhibition in clinic, remain to be addressed.
Acknowledgments G Chiosis is funded in part by Mr. William H. and Mrs. Alice Goodwin
and the Commonwealth Foundation for Cancer Research and “The Experimental Therapeutics
Center of Memorial Sloan-Kettering Cancer Center”, the Geoffrey Beene Cancer Research
Center of MSKCC, Leukemia and Lymphoma Society, Breast Cancer Research Fund,
the SPORE Pilot Award and Research and Therapeutics Program in Prostate Cancer, the
Hirshberg Foundation for Pancreatic Cancer, the Byrne Fund, National Institutes of Health
(1U01 AG032969, 1R01CA155226, 1R21AI090501, 1R21CA158609, 3P30CA008748,
P50CA086438), MSKCC Society, Department of Defense (R03-BC085588), Susan G Komen
for the Cure and the Institute for the Study of Aging and The Association for Frontotemporal
Dementias (Grant #281207 AFTD). T Taldone discloses a grant support from the Department of
Defense (PDF-BC093421). M Guzman is funded by the US National Institutes of Health (NIH)
through the NIH Director’s New Innovator Award Program, 1 DP2 OD007399-01, National
Cancer Institute (R21 CA158728-01A1), Leukemia and Lymphoma Foundation (LLS 6330-11),
and she is a V Foundation Scholar.
References
1. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–
674 (Epub 2011/03/08)
2. Nahleh Z, Tfayli A, Najm A, El Sayed A, Nahle Z (2012) Heat shock proteins in cancer: tar-
geting the chaperones. Future Med Chem 4(7):927–935 (Epub 2012/05/11)
3. Khalil AA, Kabapy NF, Deraz SF, Smith C (2011) Heat shock proteins in oncology:
diagnostic biomarkers or therapeutic targets? Biochim Biophys 1816(2):89–104 (Epub
2011/05/25)
4. Calderwood SK (2010) Heat shock proteins in breast cancer progression–a suitable case for
treatment? Int J Hyperth: official J Eur Soc Hyperth Oncol, North American Hyperthermia
Group 26(7):681–685 (Epub 2010/07/27)
5. Soo ET, Yip GW, Lwin ZM, Kumar SD, Bay BH (2008) Heat shock proteins as novel thera-
peutic targets in cancer. In vivo 22(3):311–315 (Epub 2008/07/10)
6. Johnson JL (2012) Evolution and function of diverse Hsp90 homologs and cochaperone pro-
teins. Biochim Biophys 1823(3):607–613 (Epub 2011/10/20)
7. Cotto JJ, Morimoto RI (1999) Stress-induced activation of the heat-shock response: cell and
molecular biology of heat-shock factors. Biochem Soc Symp 64:105–118 (Epub 1999/04/20)
8. Solimini NL, Luo J, Elledge SJ (2007) Non-oncogene addiction and the stress phenotype of
cancer cells. Cell 130(6):986–988 (Epub 2007/09/25)
9. Jego G, Hazoume A, Seigneuric R, Garrido C (2010) Targeting heat shock proteins in cancer.
Cancer Lett Epub (2010/11/17)
10. Patel HJ, Modi S, Chiosis G, Taldone T (2011) Advances in the discovery and development
of heat-shock protein 90 inhibitors for cancer treatment. Expert Opin Drug Discov 6(5):559–
587 (Epub 2012/03/09)
11. Prodromou C, Roe SM, O’Brien R, Ladbury JE, Piper PW, Pearl LH (1997) Identification
and structural characterization of the ATP/ADP-binding site in the Hsp90 molecular chaper-
one. Cell 90(1):65–75 (Epub 1997/07/11)
12. Sorger PK, Pelham HRB (1987) The glucose-regulated protein grp94 is related to heat shock
protein hsp90. J Mol Biol 194(2):341–344
200 M. L. Guzman et al.
13. Altieri DC, Stein GS, Lian JB, Languino LR (2012) TRAP-1, the mitochondrial Hsp90.
Biochim Biophys 1823(3):767–773 (Epub 2011/09/01)
14. Sreedhar AS, Kalmar E, Csermely P, Shen YF (2004) Hsp90 isoforms: functions, expression
and clinical importance. FEBS Lett 562(1–3):11–15 (Epub 2004/04/09)
15. da Silva VC, Ramos CH (2012) The network interaction of the human cytosolic 90 kDa heat
shock protein Hsp90: A target for cancer therapeutics. J Proteomics 75(10):2790–2802 (Epub
2012/01/13)
16. Chene P (2002) ATPases as drug targets: learning from their structure. Nat Rev Drug Discov
1(9):665–673 (Epub 2002/09/05)
17. Pearl LH, Prodromou C (2006) Structure and mechanism of the Hsp90 molecular chaperone
machinery. Annu Rev Biochem 75:271–294 (Epub 2006/06/08)
18. Prodromou C (2012) The active life of Hsp90 complexes. Biochim Biophys 1823(3):
614–623 (Epub 2011/08/16)
19. Pearl LH, Prodromou C, Workman P (2008) The Hsp90 molecular chaperone: an open and
shut case for treatment. Biochem J 410(3):439–453 (Epub 2008/02/23)
20. Jhaveri K, Taldone T, Modi S, Chiosis G (2012) Advances in the clinical development of heat
shock protein 90 (Hsp90) inhibitors in cancers. Biochim Biophys 1823(3):742–755 (Epub
2011/11/09)
21. Travers J, Sharp S, Workman P (2012) HSP90 inhibition: two-pronged exploitation of cancer
dependencies. Drug Discovery Today 17(5–6):242–252 (Epub 2012/01/17)
22. Neckers L (2006) Using natural product inhibitors to validate Hsp90 as a molecular target in
cancer. Curr Top Med Chem 6(11):1163–1171 (Epub 2006/07/18)
23. Voss AK, Thomas T, Gruss P (2000) Mice lacking HSP90beta fail to develop a placental lab-
yrinth. Development 127(1):1–11 (Epub 2000/02/02)
24. Ross JS, Schenkein DP, Pietrusko R, Rolfe M, Linette GP, Stec J et al (2004) Targeted thera-
pies for cancer 2004. Am J Clin pathol 122(4):598–609 (Epub 2004/10/19)
25. Neckers L, Schulte TW, Mimnaugh E (1999) Geldanamycin as a potential anti-cancer
agent: its molecular target and biochemical activity. Invest New Drugs 17(4):361–373 (Epub
2000/04/12)
26. Kamal A, Thao L, Sensintaffar J, Zhang L, Boehm MF, Fritz LC et al (2003) A high-
affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature
425(6956):407–410 (Epub 2003/09/26)
27. Moulick K, Ahn JH, Zong H, Rodina A, Cerchietti L, Gomes DaGama EM et al (2011)
Affinity-based proteomics reveal cancer-specific networks coordinated by Hsp90. Nat Chem
Biol 7(11):818–826 (Epub 2011/09/29)
28. Workman P, Burrows F, Neckers L, Rosen N (2007) Drugging the cancer chaperone HSP90:
combinatorial therapeutic exploitation of oncogene addiction and tumor stress. Ann N Y
Acad Sci 1113:202–216 (Epub 2007/05/22)
29. Makhnevych T, Houry WA (2012) The role of Hsp90 in protein complex assembly. Biochim
Biophys 1823(3):674–682 (Epub 2011/09/29)
30. Walter S, Buchner J (2002) Molecular chaperones–cellular machines for protein folding.
Angewandte Chemie 41(7):1098–1113 (Epub 2002/12/20)
31. Young JC, Moarefi I, Hartl FU (2001) Hsp90: a specialized but essential protein-folding tool.
J Cell Biol 154(2):267–273 (Epub 2001/07/27)
32. Whitesell L, Lindquist SL (2005) HSP90 and the chaperoning of cancer. Nat Rev Cancer
5(10):761–772 (Epub 2005/09/22)
33. Perdew GH, Whitelaw ML (1991) Evidence that the 90-kDa heat shock protein (HSP90)
exists in cytosol in heteromeric complexes containing HSP70 and three other proteins with
Mr of 63,000, 56,000, and 50,000. J Biol Chem 266(11):6708–6713 (Epub 1991/04/15)
34. Whitesell L, Mimnaugh EG, De Costa B, Myers CE, Neckers LM (1994) Inhibition of heat
shock protein HSP90-pp60v-src heteroprotein complex formation by benzoquinone ansamy-
cins: essential role for stress proteins in oncogenic transformation. Proc Nat Acad Sci U. S. A.
91(18):8324–8328 (Epub 1994/08/30)
7 Molecular Chaperones and How Addiction Matters in Cancer Therapy 201
35. Blagosklonny MV, Toretsky J, Neckers L (1995) Geldanamycin selectively destabilizes and
conformationally alters mutated p53. Oncogene 11(5):933–939 (Epub 1995/09/07)
36. Grbovic OM, Basso AD, Sawai A, Ye Q, Friedlander P, Solit D et al (2006) V600E B-Raf
requires the Hsp90 chaperone for stability and is degraded in response to Hsp90 inhibitors.
Proc Nat Acad Sci U. S. A. 103(1):57–62 (Epub 2005/12/24)
37. Pratt WB, Galigniana MD, Morishima Y, Murphy PJ (2004) Role of molecular chaperones in
steroid receptor action. Essays Biochem 40:41–58 (Epub 2004/07/10)
38. Pratt WB, Toft DO (1997) Steroid receptor interactions with heat shock protein and immuno-
philin chaperones. Endocrine Rev 18(3):306–360 (Epub 1997/06/01)
39. Echeverria PC, Mazaira G, Erlejman A, Gomez-Sanchez C, Piwien Pilipuk G, Galigniana
MD (2009) Nuclear import of the glucocorticoid receptor-hsp90 complex through the nuclear
pore complex is mediated by its interaction with Nup62 and importin beta. Mol Cell Biol
29(17):4788–4797 (Epub 2009/07/08)
40. Galigniana MD, Erlejman AG, Monte M, Gomez-Sanchez C, Piwien-Pilipuk G (2010) The
hsp90-FKBP52 complex links the mineralocorticoid receptor to motor proteins and per-
sists bound to the receptor in early nuclear events. Mol Cell Biol 30(5):1285–1298 (Epub
2009/12/30)
41. Conway-Campbell BL, George CL, Pooley JR, Knight DM, Norman MR, Hager GL et al
(2011) The HSP90 molecular chaperone cycle regulates cyclical transcriptional dynamics of
the glucocorticoid receptor and its coregulatory molecules CBP/p300 during ultradian ligand
treatment. Mol Endocrinol 25(6):944–954 (Epub 2011/04/23)
42. Nagata Y, Anan T, Yoshida T, Mizukami T, Taya Y, Fujiwara T et al (1999) The stabilization
mechanism of mutant-type p53 by impaired ubiquitination: the loss of wild-type p53 func-
tion and the hsp90 association. Oncogene 18(44):6037–6049 (Epub 1999/11/11)
43. Peng Y, Chen L, Li C, Lu W, Chen J (2001) Inhibition of MDM2 by hsp90 contributes to
mutant p53 stabilization. J Biol Chem 276(44):40583–40590 (Epub 2001/08/17)
44. Lin K, Rockliffe N, Johnson GG, Sherrington PD, Pettitt AR (2008) Hsp90 inhibition has
opposing effects on wild-type and mutant p53 and induces p21 expression and cytotox-
icity irrespective of p53/ATM status in chronic lymphocytic leukaemia cells. Oncogene
27(17):2445–2455 (Epub 2007 Nov 5)
45. Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME et al (1999)
The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent
proteolysis. Nature 399(6733):271–275 (Epub 1999/06/03)
46. Isaacs JS, Jung YJ, Mimnaugh EG, Martinez A, Cuttitta F, Neckers LM (2002) Hsp90 regu-
lates a von Hippel Lindau-independent hypoxia-inducible factor-1 alpha-degradative path-
way. J Biol Chem 277(33):29936–29944 (Epub 2002/06/08)
47. Cerchietti LC, Lopes EC, Yang SN, Hatzi K, Bunting KL, Tsikitas LA et al (2009) A purine
scaffold Hsp90 inhibitor destabilizes BCL-6 and has specific antitumor activity in BCL-6-
dependent B cell lymphomas. Nat Med 15(12):1369–1376 (Epub 2009/12/08)
48. Bird A (2001) Molecular biology: Methylation talk between histones and DNA. Science
294(5549):2113–2115 (Epub 2001/12/12)
49. Zhou Q, Agoston AT, Atadja P, Nelson WG, Davidson NE (2008) Inhibition of histone
deacetylases promotes ubiquitin-dependent proteasomal degradation of DNA methyl-
transferase 1 in human breast cancer cells. Mol Cancer Res: MCR 6(5):873–883 (Epub
2008/05/29)
50. Fiskus W, Buckley K, Rao R, Mandawat A, Yang Y, Joshi R et al (2009) Panobinostat treatment
depletes EZH2 and DNMT1 levels and enhances decitabine mediated de-repression of JunB and
loss of survival of human acute leukemia cells. Cancer Biol Ther 8(10):939–950 (Epub 2009/03/13)
51. Chang CJ, Hung MC (2012) The role of EZH2 in tumour progression. Br J Cancer
106(2):243–247 (Epub 2011/12/22)
52. He Y, Korboukh I, Jin J, Huang J (2012) Targeting protein lysine methylation and demethyla-
tion in cancers. Biochim Biophys Sinica 44(1):70–79 (Epub 2011/12/24)
53. Ho L, Crabtree GR (2008) An EZ mark to miss. Cell Stem Cell 3(6):577–578 (Epub 2008/12/02)
202 M. L. Guzman et al.
54. Fiskus W, Buckley K, Rao R, Mandawat A, Yang Y, Joshi R et al (2009) Panobinostat treat-
ment depletes EZH2 and DNMT1 levels and enhances decitabine mediated de-repression of
JunB and loss of survival of human acute leukemia cells. Cancer Biol Ther 8(10):939–950
(Epub 2009/03/13)
55. Karkhanis V, Hu YJ, Baiocchi RA, Imbalzano AN, Sif S (2011) Versatility of PRMT5-
induced methylation in growth control and development. Trends Biochem Sci 36(12):
633–641 (Epub 2011/10/07)
56. Maloney A, Clarke PA, Naaby-Hansen S, Stein R, Koopman JO, Akpan A et al (2007)
Gene and protein expression profiling of human ovarian cancer cells treated with the heat
shock protein 90 inhibitor 17-allylamino-17-demethoxygeldanamycin. Cancer Res 67(7):
3239–3253 (Epub 2007/04/06)
57. Imhof A (2003) Histone modifications: an assembly line for active chromatin? Curr Biol: CB
13(1):R22–R24 (Epub 2003/01/16)
58. Kim YR, Lee BK, Park RY, Nguyen NT, Bae JA, Kwon DD et al (2010) Differential CARM1
expression in prostate and colorectal cancers. BMC Cancer 10:197 (Epub 2010/05/14)
59. Abu-Farha M, Lanouette S, Elisma F, Tremblay V, Butson J, Figeys D et al (2011) Proteomic
analyses of the SMYD family interactomes identify HSP90 as a novel target for SMYD2. J
Mol cell Biol 3(5):301–308 (Epub 2011/10/27)
60. Luo XG, Zou JN, Wang SZ, Zhang TC, Xi T (2010) Novobiocin decreases SMYD3 expres-
sion and inhibits the migration of MDA-MB-231 human breast cancer cells. IUBMB Life
62(3):194–199 (Epub 2009/12/30)
61. Lord CJ, Ashworth A (2012) The DNA damage response and cancer therapy. Nature
481(7381):287–294 (Epub 2012/01/20)
62. Samant RS, Clarke PA, Workman P (2012) The expanding proteome of the molecular chap-
erone HSP90. Cell Cycle 11(7):1301–1308 (Epub 2012/03/17)
63. Noguchi M, Yu D, Hirayama R, Ninomiya Y, Sekine E, Kubota N et al (2006) Inhibition of
homologous recombination repair in irradiated tumor cells pretreated with Hsp90 inhibitor
17-allylamino-17-demethoxygeldanamycin. Biochem Biophys Res Commun 351(3):658–
663 (Epub 2006/11/07)
64. Ha K, Lee GE, Palii SS, Brown KD, Takeda Y, Liu K et al (2011) Rapid and transient recruit-
ment of DNMT1 to DNA double-strand breaks is mediated by its interaction with multiple
components of the DNA damage response machinery. Human Mol Genet 20(1):126–40 (Epub
2010/10/14)
65. Solier S, Kohn KW, Scroggins B, Xu W, Trepel J, Neckers L et al (2012) Feature Article:
Heat shock protein 90alpha (HSP90alpha), a substrate and chaperone of DNA-PK necessary
for the apoptotic response. Proc Nat Acad Sci U S A (Epub 2012/07/04)
66. Solier S, Pommier Y (2009) The apoptotic ring: a novel entity with phosphorylated histones
H2AX and H2B and activated DNA damage response kinases. Cell Cycle 8(12):1853–1859
(Epub 2009/05/19)
67. Camphausen K, Tofilon PJ (2007) Inhibition of Hsp90: a multitarget approach to radiosen-
sitization. Clin Cancer Res Official J Am Assoc Cancer Res 13(15 Pt 1):4326–4330 (Epub
2007/08/03)
68. Stingl L, Niewidok N, Muller N, Selle M, Djuzenova CS, Flentje M (2012) Radiosensitizing
effect of the novel Hsp90 inhibitor NVP-AUY922 in human tumour cell lines silenced for
Hsp90alpha. Strahlentherapie und Onkologie : Organ Der Deutschen Rontgengesellschaft
188(6):507–515 (Epub 2012/03/24)
69. Hartson SD, Matts RL (2012) Approaches for defining the Hsp90-dependent proteome.
Biochim Biophys 1823(3):656–667 (Epub 2011/09/13)
70. Zhao R, Davey M, Hsu YC, Kaplanek P, Tong A, Parsons AB et al (2005) Navigating the
chaperone network: an integrative map of physical and genetic interactions mediated by the
hsp90 chaperone. Cell 120(5):715–727 (Epub 2005/03/16)
71. Sharma K, Vabulas RM, Macek B, Pinkert S, Cox J, Mann M et al (2012) Quantitative pro-
teomics reveals that Hsp90 inhibition preferentially targets kinases and the DNA damage
response. Mol Cell Proteomics: MCP 11(3):M111 014654 (Epub 2011/12/15)
Molecular Chaperones and How Addiction Matters in Cancer Therapy 203
72. Chiosis G, Timaul MN, Lucas B, Munster PN, Zheng FF, Sepp-Lorenzino L et al (2001) A
small molecule designed to bind to the adenine nucleotide pocket of Hsp90 causes Her2 deg-
radation and the growth arrest and differentiation of breast cancer cells. Chem Biol 8(3):289–
99 (Epub 2001/04/18)
73. Trepel J, Mollapour M, Giaccone G, Neckers L (2010) Targeting the dynamic HSP90 com-
plex in cancer. Nat Rev Cancer 10(8):537–549 (Epub 2010/07/24)
Chapter 8
Sphingolipid Metabolism and Signaling
as a Target for Cancer Treatment
8.1 Sphingolipid Metabolism
Sphingolipids are a class of lipids with a sphingosine back bone that are formed
from non-sphingolipid precursors in the ER and get metabolized further within
different sub-cellular compartments thereby giving rise to a plethora of metabo-
lites. Of all these metabolites, ceramide is one of the most widely studied bioactive
molecules. It is formed through three distinct pathways (Fig 8.1) (1) de novo
synthesis—synthesis from non-sphingolipid precursors; (2) turnover p athways—
break down products from complex sphingolipids; and (3) recycling and sal-
vage pathways—The de novo pathway starts with the condensation of serine
V. Rajagopalan
Department of Biochemistry and Molecular Biology, Medical University of South Carolina,
Charleston, SC, USA
Y. A. Hannun (*)
Stony Brook University, Stony Brook, NY, USA
e-mail: [email protected]
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 205
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_8,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
206 V. Rajagopalan and Y. A. Hannun
KDS S1PPase SK
Dihydrosphingosine Sphingosine
CerS + Fatty acyl coA CDase CDase CerS + Fatty acyl coA
DES C1PP
Dihydroceramide Ceramide
Ceramide-1-phosphate
CK
SMase GCS
CGT GCase
SMS
Sphingomyelin Glucosylceramide
Galactosylceramide
Glycosphingolipids
Fig. 8.1 Ceramide can be formed by de novo pathway from serine and palmitoyl-coA or from
hydrolysis of sphingomyelin or cerebrosides (glucosyl or galactosyl ceramide). Ceramide, thus
formed, can be phosphorylated by CK to yield ceramide-1-phosphate or serves as a substrate for
the synthesis of sphingomyelin or glycosphingolipids. Ceramide can be deacylated by cerami-
dases to form sphingosine which can be phosphorylated by SKs to generate S1P which can be
acted upon by phosphatases to generate sphingosine or by lyase to form ethanolamine-1-phos-
phate and hexadecanal, an aldehyde. Abbreviations: SPT serine palmitoyl transferase, KDS
3-keto-dihydrosphingosine reductase, DES dihydroceramide desaturase, SPPase Sph-1- phosphate
phosphatase, CK cer kinase, C1PP C1P phosphatase, SMS SM synthase, GCS glucosylceramide
synthase, GCase glucosyl CDase, CGT UDP-galactose ceramide-galactosyltransferase
Ceramide regulates many biological processes such as cancer cell growth, dif-
ferentiation, apoptosis, and senescence [17, 18]. Many signals such as cytokines,
anticancer drugs, and stress-inducers upregulate ceramide through the de novo
or salvage pathways [19, 20]. Ceramide triggers signaling cascades by regulat-
ing phosphatases, cathepsin D, or kinase suppressor of RAS (KSR) as described
below.
Phosphatases such as protein phosphatase 1 (PP1) and protein phosphatase 2A
(PP2A) are activated by ceramide in vitro. Inhibition of these phosphatases inhib-
its the ability of ceramide to dephosphorylate (inactivate) several pro-proliferative
proteins such as PKCα, Akt/PKB, c-Jun, Bcl-2, Rb, and SR [21, 22].
Many studies have documented the translocation of cathepsin D from lys-
osomes in response to oxidative stress followed by activation of caspase 3 and
cell death [23]. Interestingly, cathepsin D was found to be a ceramide-binding and
ceramide-activated protein [24]. Besides, acid sphingomyelinase-derived ceramide
has been shown to favor autocatalytic proteolysis of inactive cathepsin D to enzy-
matically active cathepsin D isoform [25].
Similarly, KSR has been found to be ceramide responsive [26–30]. Mammalian
KSR activates Raf, and activation of this pathway results in apoptosis [31, 32].
Also, ceramide has been shown to activate the zeta isoform of protein kinase C
(PKCz) by phosphorylation [33]. Ceramide-activation of PKC-zeta has been
8 Sphingolipid Metabolism and Signaling 209
CDase
SK
Ceramide Sphingosine-1-phosphate
Fig. 8.3 Ceramide regulates many protein signaling molecules such as cathepsin D and
ceramide-activated protein phosphatases (CAPPs), KSR, RAF, MEKK. Proteins that are modulated
by these pathways include RB,SR, AKT, PKC-α, c-JUN, Bcl-2,telomerase, c-MYC, caspases, and
cyclin-dependent kinases (CDKs) which in turn brings about cellular responses such as growth
arrest, apoptosis, and/or senescence. Ceramide that gets metabolised to S1P by ceramidase and SK,
regulates proteins such as ERK, NF-κB, Cox-2 which in turn brings about cellular responses such
as inhibition of apoptosis, malignant transformation, angiogenesis, and inflammation
Ceramide has been implicated in many cell death paradigms. Birbes et al. [39]
showed that selective targeting of bacterial sphingomyelinase (bSMase) to mito-
chondria and not to any other compartments such as plasma membrane, ER, or
Golgi resulted in apoptosis that was associated with generation of ceramide and
release of cytochrome c in MCF-7 cells. Overexpression of Bcl-2 prevented the
mitochondria-targeted bSMase effects on apoptosis. Dai et al. [40] showed that
UV-induced apoptosis was marked by increase in SM in all sub-cellular loca-
tions, particularly mitochondria, in HeLa cells. Ceramide levels were found to
be elevated in mitochondria at 2–6 h, consistent with the cell death time course.
D609, an inhibitor of sphingomyelin synthase to a marked extent and fumonisin
B1 (FB1), a ceramide synthase inhibitor to a lesser extent, rescued the cells from
increases in SM and ceramide, and consequently cell death. On the other hand,
the SPT inhibitor myriocin did not rescue the UV effects on cell death, suggest-
ing the involvement of the turnover pathway-generated ceramide in bringing about
UV-triggered cell death.
In another study in Caenorhabditis elegans, upon inactivation of ceramide syn-
thase, somatic apoptosis was unaffected, but ionizing radiation-induced apoptosis
of germ cells was obliterated, and this phenotype was reversed by microinjec-
tion of long-chain natural ceramide. Radiation-induced ceramide accumulation in
mitochondria consequently activated CED-3 caspase and apoptosis [41].
In Ramos B cells, surface B-cell receptor (BcR)-triggered cell death was marked
by an early increase in C16 ceramide. Pulse labeling with sphinoglipid precursor,
palmitate, in the presence of ceramide synthase inhibitor, FB1, demonstrated that the
de novo ceramide-generating pathway was activated following BcR activation. The
apoptotic cell death induced by cross-linking of BcR was mediated through mito-
chondrial cell death pathways followed by caspase activation [42]. In LNCaP pros-
tate cancer cells, androgen ablation, which is considered as one of the therapeutic
8 Sphingolipid Metabolism and Signaling 211
modalities, was found to increase C16 ceramide level followed by G0/G1 cell cycle
arrest and apoptosis. 5alpha-dihydrotestosterone (DHT) or fumonisin B1 treatment
rescued LNCaP cells from apoptosis [43].
In another study, ceramide acting via PP1, dephosphorylated SR proteins that
regulated the alternate splicing of Bcl-x(L) and caspase 9. In A549 lung adeno-
carcinoma cell lines, cell-permeable D-e-C(6) ceramide downregulated the mRNA
levels of anti-apoptotic Bcl-x(L) and caspase 9b with concomitant increase in the
mRNA of pro-apoptotic Bcl-x(s) and caspase 9. The chemotherapeutic agent,
gemcitabine, induced de novo generation of ceramide and brought about afore-
mentioned alternate splicing of Bcl-x(L) and caspase 9b and consequent loss of
cell viability as measured by MTT assay [44].
In several studies, aSMase was shown to be necessary for radiation-induced
apoptosis in endothelial cells and mice lacking aSMase were protected from gas-
trointestinal and CNS apoptosis [45–47]. In another study, the endolysosomal
aspartate protease cathepsin D (CTSD) was identified as a target of ceramide gen-
erated by acid sphingomyelinase in response to TNFα. CTSD cleaved pro-apop-
totic Bid and activated it in vitro. The lack of Bid activation in cathepsin-deficient
fibroblasts suggested Bid is downstream of cathepsin D in bringing about apopto-
sis as a result of TNFα treatment [48].
In addition to aSMase, neutral sphingomyelinase has been implicated in stress
response pathways initiated by TNFα in MCF-7 cells [49]; amyloid-β peptide in
neuronal cells [50]; ethanol in HepG2 hepatoma cells [51]; and staurosporine in
several neuronal cell lines [52].
In addition to the sphingomyelinases, ceramidases (CDases) were also found to
regulate apoptosis. In one study, nitric oxide induced the degradation of nCDase,
thereby, enabling ceramide accumulation and cell death [53]. In another study, the
degeneration of photoreceptor cells was marked by an increase in ceramide which
was rescued by overexpression of CDase that cleared the ceramide and prevented
its apoptotic effect [54]. These studies clearly implicate ceramide in apoptosis and
in mediating the cellular response to various stress causing stimuli.
8.3.2 Ceramide in Senescence
S1P is considered a pro-survival lipid. For example, S1P stimulated the invasive-
ness of glioblastoma tumor cells [77], promoted estrogen-dependent tumorigen-
esis of breast cancer cells [78] and conferred resistance to the cytotoxic actions of
TNF-α and daunorubicin [10]. A number of studies documented the role of S1P/
S1PR in proliferation, inhibition of apoptosis, vasculogenesis/angiogenesis, and
inflammation. These topics will be discussed in the following sections.
214 V. Rajagopalan and Y. A. Hannun
S1P promotes vasculogenesis and angiogenesis. S1P, the natural ligand for S1P3
receptor or KRX-725, a synthetic peptide that mimics S1P action on this recep-
tor, favored angiogenesis, as demonstrated by assessment of vascular sprouting
using aortic rings as an ex vivo model of angiogenesis. When S1P or KRX-725
were combined with other growth factors such as basic fibroblast growth factor
(b-FGF), stem cell factor, or vascular endothelial growth factor (VEGF), the inves-
tigators observed synergistic induction of angiogenesis [84]. In a cultured mouse
allantois explant model of blood vessel formation, Argraves et al. [85] showed that
S1P, synthesized via the action of SK2, promoted vasculogenesis by promoting
migratory activities of angioblasts and early endothelial cells to expand the vascu-
lar network.
VEGF has been shown to stimulate SK1 activity with an increase in the pro-
duction of S1P and activation of H and N Ras oncogenes in T24 bladder tumor
cell lines [86]. Endothelial cells undergo morphogenesis into capillary networks
in response to S1P involving G protein receptors [87]. S1P has been shown to
induce endothelial cell invasion and morphogenesis in physiologically relevant
collagen and fibrin matrices [88]. Based on studies employing inhibitors and
functional antagonists of S1P receptors, it has been hypothesized that the angi-
ogenic function of S1P is mediated by S1P1 and S1P3 signaling [87, 89, 90].
8 Sphingolipid Metabolism and Signaling 215
8.4.3 S1P in Inflammation
The SK1/S1P pathway has been implicated in inflammation. For instance, TNF-
alpha resulted in activation of SK1/S1P pathway specifically leading to extracel-
lular signal-regulated kinases and NF-kappa B activation [36] and consequently
expression of vascular cell adhesion molecule (VCAM) and intercellular adhe-
sion molecule (ICAM) [93]. S1P also induced cyclooxygenase 2 (COX2) and
prostaglandin E2 (PGE2) production in L929 fibrosarcoma and A549 lung ade-
nocarcinoma cells and genetic knockdown using siRNA blocked their produc-
tion. Additionally, preventing S1P clearance using siRNAs against S1P lyase/
phosphatase resulted in increased production of COX2 and PGE2, implicating a
key role of S1P in this pathway [94]. Microglial activation has been implicated
in neuroinflammation. LPS treatment increased SK1 mRNA and protein levels
and consequently upregulated expression of proinflammatory cytokines such as
TNF-alpha, IL-1beta, and iNOS in microglia. Chronic production of inflamma-
tory cytokines by microglia has been implicated in neuroinflammation [95, 96].
Further, the SK/S1P pathway has been implicated in many other inflammatory
disease paradigms such as asthma, rheumatoid arthritis, and inflammatory bowel
diseases [97].
In summary, these data collectively demonstrate that S1P regulates cancer cell
viability, angiogenesis, and inflammation which favor cancer pathogenesis.
Among the bioactive sphingolipids, ceramide and S1P act as pro-apoptotic and
anti-apoptotic lipids, respectively, and therefore, modulation of these lipids may be
effective as a treatment strategy for cancer (refer to Fig. 8.4 [109]). Such strategies
to increase the accumulation of ceramide and attenuation of S1P are discussed in
detail in the following section.
8 Sphingolipid Metabolism and Signaling 217
FTY 720
Tumor proliferation
S1P Angiogenesis
Inflammation
SK1
Chemoresistance
S1P
Sphingosine
GSL
P-gp
Golgi Apoptosis
CDase
Glucer
B13
SMS
SM
nSMase
D609 Ceramide
analogues
Mitochondria
Chemotherapy
Fig. 8.4 Chemotherapeutic agents increases ceramide levels and induces apoptosis through the
de novo pathway or through the neutral sphingomyelinase (N-SMase) pathway. Induction of SK1
in colon cancer leads to accumulation of S1P, possibly leading to tumor proliferation, angiogene-
sis, and inflammation. Clearance of ceramide to Glucosylceramide by GCS in breast cancer cells
leads to the development of drug resistance. P-glycoprotein (P-gp) expression might potentiate
the chemo-resistant phenotype. Marked in red arrows are modulators of SPL metabolism. B13 is
an inhibitor of acid CDase, OGT2378 is GCS inhibitor, D609 is an inhibitor of SMS, FTY720 is
a sphingosine analogue, and ceramide analogues mimic endogenous ceramides, and Pyridinium
ceramide targets mitochondria and promotes mitochondria mediated apoptosis. Abbreviations:
CDase ceramidase, ER endoplasmic reticulum, PP1 protein phosphatase 1, PP2A protein phos-
phatase 2A, GSL glycosphingolipid, GCS glucosylceramide synthase
that these drugs manifest their cytotoxic effects partly through ceramide production
[110–112]. Besides the de novo pathway, certain drugs, including daunorubicin,
also induce ceramide generation through the activation of nSMase which hydrolyzes
sphingomyelin to generate ceramide. For instance, in leukemia cells, cytosine arabi-
noside (Ara-C) induced activation of nSMase [113]. Mechanistically, this induction of
nSMase was brought about by generation of reactive oxygen species followed by Jun
N-terminal kinase phosphorylation and apoptosis [114]. In another study, actinomycin
D and etoposide induced nSMase activity in a p53- and ROS-dependent manner [115].
Alternatively, studies on reagents that inhibit the enzymes that favor the cera-
mide clearance pathway, leading to accumulation of ceramide and, thereby, poten-
tiating the cytotoxic effects were tested. For instance, compounds such as B13 that
inhibited acid CDase or tricyclodecan-9-yl-xanthogenate (D609) that inhibited
SMS, induced apoptosis in colon cancer and in U937 human monocytic leukemia
cells, respectively [116, 117] (refer to Fig. 8.4 and Table 8.1).
Interestingly, SM was found to potentiate the chemotherapeutic response of gemi-
citabine in prostate cancer cell lines [118]. In a study involving combination of SM
with chemotherapeutic agents, doxorubicin, epirubicin, or topotecan, it was found
that the combination therapy increased the cytotoxic effect of the drugs by increas-
ing their bioavailability, possibly by modulation of plasma membrane lipophilicity,
facilitating entry of these agents into the various cancer cell lines studied [119].
Ceramide analogues (such as the soluble short-chain C2- and C6-ceramides) have
been shown to bring about cell death in many types of cancer cell lines tested [120].
C16-serinol, 4, 6-diene-ceramide, 5R-OH-3E-C8-ceramide, adamantyl-ceramide,
and benzene-C4-ceramide (Table 8.1) are some of the ceramide analogues that
8 Sphingolipid Metabolism and Signaling 219
induced cell death in cell lines such as neuroblastoma and breast cancer [121–124].
A novel, cationic, water soluble, pyridinium ceramide (Table 8.1) accumulated pre-
dominantly in cellular compartments that are negatively charged such as mitochon-
dria and the nucleus and caused changes in mitochondrial structure and function
and inhibited growth in various human head and neck cancer cell lines [125], while
inducing apoptosis in squamous cell carcinoma (HNSCC) cell lines [126–128].
Experiments to uncover the most efficient means of delivery of these ceramide
analogues have been tried extensively. Pegylated liposomes were very effective in
bringing about ceramide-mediated cell death in breast cancer cell lines (Table 8.1).
Liposomal delivery of ceramide decreased phosphorylated AKT and activation of cas-
pase-3/7 more effectively than non-liposomal ceramide [129]. Vincristine incorporated
in SM-liposomes called sphingosomes (Table 8.1) was found to be effective in animal
models for treatment of acute lymphocytic leukemia (ALL) to the extent that it is cur-
rently in Phase II clinical trials [130]. These studies clearly demonstrate that targeting
ceramide generation might be an effective method to bring about cancer cell death.
(DMH), a chemical colonic carcinogen [143]. Merrill and co-workers found that milk
sphingomyelin dietary supplementation reduced the incidence of DMH-induced pre-
malignant lesions of colon tumors in CF1 mice [144]. In addition, mice fed with SM
developed fewer adenocarcinomas. These findings suggest that milk SM might sup-
press advanced malignant tumors in colon [145]. Administration of synthetic SM and
ceramide analogues also suppressed colonic crypt foci formation [146, 147]. In another
study, azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced colon carcinogen-
esis was modulated by dietary SM in the early stages by activation of peroxisome pro-
liferator-activated receptor γ (PPAR-γ), but its anti-carcinogenic effect was independent
of PPAR-γ [148]. Therefore, dietary SM might modulate the proteins expressed during
early stages of colon carcinogenesis and therefore may be a potential therapeutic candi-
date in the context of colon carcinogenesis.
One of the reasons for the failure of chemotherapy in cancer is the development
of tumor cell resistance. Part of the basis for chemoresistance might be attributed
to a re-wiring of sphingolipid metabolism. For instance, in many cases of leuke-
mia, breast cancer, and melanoma, chemotherapeutic agents increase the activity
of GCS which thereby attenuates ceramide levels, resulting in a drug resistance
phenotype [20, 149]. Overexpression of GCS offered increased resistance to doxo-
rubicin whereas siRNA knockdown promoted increased sensitivity to doxorubicin,
paclitaxel, and etoposide in breast cancer cells [150–152]. Mechanistically, GCS
upregulated P-glycoprotein (P-gp) which is an ABC transporter implicated in drug
resistance. Knockdown of GCS inhibited MDR1, a gene that encodes P-gp, revers-
ing drug resistance [153, 154].
Based on the above studies, GCS inhibition has been predicted to improve the
effectiveness of chemotherapeutic drugs. Some studies suggest that this hypothesis
is in fact true. For instance, OGT2378 (Table 8.1), an inhibitor of GCS, inhibited
melanoma growth in a syngeneic orthotopic murine model [155]. In separate stud-
ies, combination of fenretinide, a compound that induces accumulation of dihy-
droceramide [156] through direct inhibition of dihydroceramide desaturase [157],
with GCS inhibitors resulted in synergistic suppression of the growth of various
tumors. Additionally, fenretinide combined with SK inhibitors such as PPMP or
safingol caused growth inhibition [158, 159].
Sphingosine kinase and S1P have also been implicated in drug resistance phe-
notypes. For instance, it has been brought to light that certain drug-resistant mela-
noma cell lines such as Mel-2a and M221 are resistant to Fas-induced cell death
due to a decrease in ceramide and an increase in S1P compared with Fas-sensitive
counterparts such as A-375 and M186. Downregulation of SK1 with siRNA
decreased the resistance of Mel-2a cells to apoptosis [160]. Similar inference was
made in camptothecin resistant prostate cancer cell lines [161]. In a recent study,
SK1 was found to be upregulated in imatinib-resistant chronic myeloid leukemia
8 Sphingolipid Metabolism and Signaling 221
cell line concomitant with increased BCR-ABL mRNA and protein levels. The
PI3K/AKT/mTOR pathway was also found to be upregulated. Knocking down
SK1 expression using siRNA reversed the imatinib resistance to apoptosis and
returned BCR-ABL to normal levels [162], suggesting a role for SK1 in conferring
drug resistance.
8.8 Conclusions
Acknowledgments We thank Benjamin Newcomb for his critical review of this chapter. We
also thank the members of Yusuf Hannun and Lina Obeid laboratory for their helpful discussion.
We apologize to those investigators whose important works were not included in this chapter
because of the space limitations. The Yusuf Hannun laboratory is supported by research grants
from the National Institutes of Health, USA.
References
1. Mandon EC, Ehses I, Rother J, van Echten G, Sandhoff K (1992) Subcellular localization
and membrane topology of serine palmitoyltransferase, 3-dehydrosphinganine reductase,
and sphinganine N-acyltransferase in mouse liver. J Biol Chem 267(16):11144–11148
2. Michel C, van Echten-Deckert G (1997) Conversion of dihydroceramide to ceramide occurs
at the cytosolic face of the endoplasmic reticulum. FEBS Lett 416(2):153–155
3. Tafesse FG, Ternes P, Holthuis JC (2006) The multigenic sphingomyelin synthase family.
J Biol Chem 281(40):29421–29425
4. Ichikawa S, Hirabayashi Y (1998) Glucosylceramide synthase and glycosphingolipid syn-
thesis. Trends Cell Biol 8(5):198–202
5. Shinghal R, Scheller RH, Bajjalieh SM (1993) Ceramide 1-phosphate phosphatase activity
in brain. J Neurochem 61(6):2279–2285
222 V. Rajagopalan and Y. A. Hannun
6. Marchesini N, Hannun YA (2004) Acid and neutral sphingomyelinases: roles and mecha-
nisms of regulation. Biochem Cell Biol Biochimie et Biologie Cellulaire 82(1):27–44
7. Tettamanti G (2004) Ganglioside/glycosphingolipid turnover: new concepts. Glycoconj
J 20(5):301–317
8. Mao C, Obeid LM (2008) Ceramidases: regulators of cellular responses mediated by cera-
mide, sphingosine, and sphingosine-1-phosphate. Biochim Biophys Acta 1781(9):424–434
9. Hait NC, Oskeritzian CA, Paugh SW, Milstien S, Spiegel S (2006) Sphingosine
kinases, sphingosine 1-phosphate, apoptosis and diseases. Biochim Biophys Acta
1758(12):2016–2026
10. Johnson KR, Johnson KY, Becker KP, Bielawski J, Mao C, Obeid LM (2003) Role of
human sphingosine-1-phosphate phosphatase 1 in the regulation of intra- and extracellular
sphingosine-1-phosphate levels and cell viability. J Biol Chem 278(36):34541–34547
11. Bandhuvula P, Saba JD (2007) Sphingosine-1-phosphate lyase in immunity and cancer:
silencing the siren. Trends Mol Med 13(5):210–217
12. Hanada K, Kumagai K, Tomishige N, Kawano M (2007) CERT and intracellular trafficking
of ceramide. Biochim Biophys Acta 1771(6):644–653
13. Yamaji T, Kumagai K, Tomishige N, Hanada K (2008) Two sphingolipid transfer proteins,
CERT and FAPP2: their roles in sphingolipid metabolism. IUBMB Life 60(8):511–518
14. Tani M, Hannun YA (2007) Analysis of membrane topology of neutral sphingomyelinase 2.
FEBS Lett 581(7):1323–1328
15. Hannun YA, Obeid LM (2008) Principles of bioactive lipid signalling: lessons from sphin-
golipids. Nat Rev Mol Cell Biol 9(2):139–150
16. Bartke N, Hannun YA (2009) Bioactive sphingolipids: metabolism and function. J Lipid Res
50:S91–S96 Suppl
17. Hannun YA (1996) Functions of ceramide in coordinating cellular response to stress.
Science 274:1855–1859
18. Hannun YA, Obeid LM (2002) The ceramide-centric universe of lipid-mediated cell regula-
tion: stress encounters of the lipid kind. J Biol Chem 277:25847–25850
19. Andrieu-Abadie N (2001) Ceramide in apoptosis signaling: relationship with oxidative
stress. Free Radic Biol Med 31:717–718
20. Ogretmen B, Hannun YA (2001) Updates on functions of ceramide in chemotherapy-
induced cell death and in multidrug resistance. Drug Resist Updat 4:368–377
21. Hannun YA (1996) Functions of ceramide in coordinating cellular responses to stress.
Science 274(5294):1855–1859
22. Chalfant CE, Ogretmen B, Galadari S, Kroesen BJ, Pettus BJ, Hannun YA (2001) FAS acti-
vation induces dephosphorylation of SR proteins; dependence on the de novo generation of
ceramide and activation of protein phosphatase 1. J Biol Chem 276(48):44848–44855
23. Kagedal K, Johansson U, Ollinger K (2001) The lysosomal protease cathepsin D medi-
ates apoptosis induced by oxidative stress. FASEB J: Official Publ Fed Am Soc Exp Biol
15(9):1592–1594
24. Heinrich M, Wickel M, Winoto-Morbach S, Schneider-Brachert W, Weber T, Brunner J,
Saftig P, Peters C, Kronke M, Schutze S (2000) Ceramide as an activator lipid of cathepsin
D. Adv Exp Med Biol 477:305–315
25. Heinrich M, Wickel M, Schneider-Brachert W, Sandberg C, Gahr J, Schwandner R, Weber
T, Saftig P, Peters C, Brunner J et al (1999) Cathepsin D targeted by acid sphingomyelinase-
derived ceramide. EMBO J 18(19):5252–5263
26. Basu S, Bayoumy S, Zhang Y, Lozano J, Kolesnick R (1998) BAD enables ceramide to sig-
nal apoptosis via Ras and Raf-1. J Biol Chem 273(46):30419–30426
27. Mathias S, Dressler KA, Kolesnick RN (1991) Characterization of a ceramide-activated
protein kinase: stimulation by tumor necrosis factor alpha. Proc Nat Acad Sci USA
88(22):10009–10013
28. Yao B, Zhang Y, Delikat S, Mathias S, Basu S, Kolesnick R (1995) Phosphorylation of Raf
by ceramide-activated protein kinase. Nature 378(6554):307–310
8 Sphingolipid Metabolism and Signaling 223
29. Liu J, Mathias S, Yang Z, Kolesnick RN (1994) Renaturation and tumor necrosis fac-
tor-alpha stimulation of a 97-kDa ceramide-activated protein kinase. J Biol Chem
269(4):3047–3052
30. Joseph CK, Byun HS, Bittman R, Kolesnick RN (1993) Substrate recognition by ceramide-
activated protein kinase. Evidence that kinase activity is proline-directed. J Biol Chem
268(27):20002–20006
31. Xing HR, Kolesnick R (2001) Kinase suppressor of Ras signals through Thr269 of c-Raf-1.
J Biol Chem 276(13):9733–9741
32. Basu S, Kolesnick R (1998) Stress signals for apoptosis: ceramide and c-Jun kinase.
Oncogene 17(25):3277–3285
33. Fox TE, Houck KL, O’Neill SM, Nagarajan M, Stover TC, Pomianowski PT, Unal O, Yun
JK, Naides SJ, Kester M (2007) Ceramide recruits and activates protein kinase C zeta (PKC
zeta) within structured membrane microdomains. J Biol Chem 282(17):12450–12457
34. Bourbon NA, Sandirasegarane L, Kester M (2002) Ceramide-induced inhibition of Akt
is mediated through protein kinase Czeta: implications for growth arrest. J Biol Chem
277(5):3286–3292
35. Payne SG, Milstien S, Spiegel S (2002) Sphingosine-1-phosphate: dual messenger func-
tions. FEBS Lett 531:54–57
36. Xia P, Wang L, Moretti PA, Albanese N, Chai F, Pitson SM, D’Andrea RJ, Gamble JR,
Vadas MA (2002) Sphingosine kinase interacts with TRAF2 and dissects tumor necrosis
factor-alpha signaling. J Biol Chem 277(10):7996–8003
37. French KJ, Schrecengost RS, Lee BD, Zhuang Y, Smith SN, Eberly JL, Yun JK, Smith CD
(2003) Discovery and evaluation of inhibitors of human sphingosine kinase. Cancer Res
63(18):5962–5969
38. Rylova SN, Somova OG, Dyatlovitskaya EV (1998) Comparative investigation of sphin-
goid bases and fatty acids in ceramides and sphingomyelins from human ovarian malignant
tumors and normal ovary. Biochemistry 63:1057–1060
39. Birbes H, El Bawab S, Hannun YA, Obeid LM (2001) Selective hydrolysis of a mitochon-
drial pool of sphingomyelin induces apoptosis. FASEB J: Official Publ Fed Am Soc Exp
Biol 15(14):2669–2679
40. Dai Q, Liu J, Chen J, Durrant D, McIntyre TM, Lee RM (2004) Mitochondrial ceramide
increases in UV-irradiated HeLa cells and is mainly derived from hydrolysis of sphingomy-
elin. Oncogene 23(20):3650–3658
41. Deng X, Yin X, Allan R, Lu DD, Maurer CW, Haimovitz-Friedman A, Fuks Z, Shaham S,
Kolesnick R (2008) Ceramide biogenesis is required for radiation-induced apoptosis in the
germ line of C. elegans. Science 322(5898):110–115
42. Kroesen BJ, Pettus B, Luberto C, Busman M, Sietsma H, de Leij L, Hannun YA (2001)
Induction of apoptosis through B-cell receptor cross-linking occurs via de novo generated
C16-ceramide and involves mitochondria. J Biol Chem 276(17):13606–13614
43. Eto M, Bennouna J, Hunter OC, Hershberger PA, Kanto T, Johnson CS, Lotze MT,
Amoscato AA (2003) C16 ceramide accumulates following androgen ablation in LNCaP
prostate cancer cells. Prostate 57(1):66–79
44. Chalfant CE, Rathman K, Pinkerman RL, Wood RE, Obeid LM, Ogretmen B, Hannun
YA (2002) De novo ceramide regulates the alternative splicing of caspase 9 and Bcl-x in
A549 lung adenocarcinoma cells. Dependence on protein phosphatase-1. J Biol Chem
277(15):12587–12595
45. Santana P, Pena LA, Haimovitz-Friedman A, Martin S, Green D, McLoughlin M,
Cordon-Cardo C, Schuchman EH, Fuks Z, Kolesnick R (1996) Acid sphingomyelinase-
deficient human lymphoblasts and mice are defective in radiation-induced apoptosis. Cell
86(2):189–199
46. Paris F, Fuks Z, Kang A, Capodieci P, Juan G, Ehleiter D, Haimovitz-Friedman A, Cordon-
Cardo C, Kolesnick R (2001) Endothelial apoptosis as the primary lesion initiating intesti-
nal radiation damage in mice. Science 293(5528):293–297
224 V. Rajagopalan and Y. A. Hannun
47. Pena LA, Fuks Z, Kolesnick RN (2000) Radiation-induced apoptosis of endothelial cells in
the murine central nervous system: protection by fibroblast growth factor and sphingomyeli-
nase deficiency. Cancer Res 60(2):321–327
48. Heinrich M, Neumeyer J, Jakob M, Hallas C, Tchikov V, Winoto-Morbach S, Wickel M,
Schneider-Brachert W, Trauzold A, Hethke A et al (2004) Cathepsin D links TNF-induced
acid sphingomyelinase to Bid-mediated caspase-9 and -3 activation. Cell Death Differ
11(5):550–563
49. Luberto C, Hassler DF, Signorelli P, Okamoto Y, Sawai H, Boros E, Hazen-Martin DJ,
Obeid LM, Hannun YA, Smith GK (2002) Inhibition of tumor necrosis factor-induced
cell death in MCF7 by a novel inhibitor of neutral sphingomyelinase. J Biol Chem
277(43):41128–41139
50. Lee JT, Xu J, Lee JM, Ku G, Han X, Yang DI, Chen S, Hsu CY (2004) Amyloid-beta pep-
tide induces oligodendrocyte death by activating the neutral sphingomyelinase-ceramide
pathway. J Cell Biol 164(1):123–131
51. Liu JJ, Wang JY, Hertervig E, Cheng Y, Nilsson A, Duan RD (2000) Activation of neu-
tral sphingomyelinase participates in ethanol-induced apoptosis in Hep G2 cells. Alcohol
Alcohol 35(6):569–573
52. Testai FD, Landek MA, Dawson G (2004) Regulation of sphingomyelinases in cells of the
oligodendrocyte lineage. J Neurosci Res 75(1):66–74
53. Franzen R (2002) Nitric oxide induces degradation of the neutral ceramidase in rat renal
mesangial cells and is counterregulated by protein kinase C. J Biol Chem 277:46184–46190
54. Acharya U (2003) Modulating sphingolipid biosynthetic pathway rescues photoreceptor
degeneration. Science 299:1740–1743
55. Venable ME, Lee JY, Smyth MJ, Bielawska A, Obeid LM (1995) Role of ceramide in cel-
lular senescence. J Biol Chem 270(51):30701–30708
56. Venable ME, Yin X (2009) Ceramide induces endothelial cell senescence. Cell Biochem
Funct 27(8):547–551
57. Modrak DE, Leon E, Goldenberg DM, Gold DV (2009) Ceramide regulates gemcitabine-
induced senescence and apoptosis in human pancreatic cancer cell lines. Mol Cancer Res:
MCR 7(6):890–896
58. Guillas I (2001) C26-CoA-dependent ceramide synthesis of Saccharomyces cerevisiae is
operated by Lag1p and Lac1p. EMBO J 20:2655–2665
59. Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright
WE, Weinrich SL, Shay JW (1994) Specific association of human telomerase activity with
immortal cells and cancer. Science 266(5193):2011–2015
60. Shay JW, Bacchetti S (1997) A survey of telomerase activity in human cancer. Eur J Cancer
33(5):787–791
61. Ogretmen B, Schady D, Usta J, Wood R, Kraveka JM, Luberto C, Birbes H, Hannun YA,
Obeid LM (2001) Role of ceramide in mediating the inhibition of telomerase activity in
A549 human lung adenocarcinoma cells. J Biol Chem 276(27):24901–24910
62. Okazaki T, Bell RM, Hannun YA (1989) Sphingomyelin turnover induced by vitamin D3 in
HL-60 cells. Role in cell differentiation. J Biol Chem 264(32):19076–19080
63. Dobrowsky RT, Werner MH, Castellino AM, Chao MV, Hannun YA (1994) Activation
of the sphingomyelin cycle through the low-affinity neurotrophin receptor. Science
265(5178):1596–1599
64. Fishbein JD, Dobrowsky RT, Bielawska A, Garrett S, Hannun YA (1993) Ceramide-
mediated growth inhibition and CAPP are conserved in Saccharomyces cerevisiae. J Biol
Chem 268(13):9255–9261
65. Dbaibo GS (1995) Retinoblastoma gene product as a downstream target for a ceramide-
dependent pathway of growth arrest. Proc Natl Acad Sci USA 92:1347–1351
66. Daido S, Kanzawa T, Yamamoto A, Takeuchi H, Kondo Y, Kondo S (2004) Pivotal role of
the cell death factor BNIP3 in ceramide-induced autophagic cell death in malignant glioma
cells. Cancer Res 64(12):4286–4293
8 Sphingolipid Metabolism and Signaling 225
87. Lee MJ, Thangada S, Claffey KP, Ancellin N, Liu CH, Kluk M, Volpi M, Sha’afi RI, Hla T
(1999) Vascular endothelial cell adherens junction assembly and morphogenesis induced by
sphingosine-1-phosphate. Cell 99(3):301–312
88. Bayless KJ, Davis GE (2003) Sphingosine-1-phosphate markedly induces matrix metal-
loproteinase and integrin-dependent human endothelial cell invasion and lumen forma-
tion in three-dimensional collagen and fibrin matrices. Biochem Biophys Res Commun
312(4):903–913
89. Yonesu K, Kawase Y, Inoue T, Takagi N, Tsuchida J, Takuwa Y, Kumakura S, Nara F (2009)
Involvement of sphingosine-1-phosphate and S1P1 in angiogenesis: analyses using a new S1P1
antagonist of non-sphingosine-1-phosphate analog. Biochem Pharmacol 77(6):1011–1020
90. LaMontagne K, Littlewood-Evans A, Schnell C, O’Reilly T, Wyder L, Sanchez T, Probst B,
Butler J, Wood A, Liau G et al (2006) Antagonism of sphingosine-1-phosphate receptors by
FTY720 inhibits angiogenesis and tumor vascularization. Cancer Res 66(1):221–231
91. Liu Y, Wada R, Yamashita T, Mi Y, Deng CX, Hobson JP, Rosenfeldt HM, Nava VE, Chae
SS, Lee MJ et al (2000) Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate,
is essential for vascular maturation. J Clin Investig 106(8):951–961
92. Mizugishi K, Yamashita T, Olivera A, Miller GF, Spiegel S, Proia RL (2005) Essential role for
sphingosine kinases in neural and vascular development. Mol Cell Biol 25(24):11113–11121
93. Xia P, Gamble JR, Rye KA, Wang L, Hii CS, Cockerill P, Khew-Goodall Y, Bert AG, Barter
PJ, Vadas MA (1998) Tumor necrosis factor-alpha induces adhesion molecule expression
through the sphingosine kinase pathway. Proc Nat Acad Sci USA 95(24):14196–14201
94. Pettus BJ, Bielawski J, Porcelli AM, Reames DL, Johnson KR, Morrow J, Chalfant CE,
Obeid LM, Hannun YA (2003) The sphingosine kinase 1/sphingosine-1-phosphate path-
way mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB J:
Official Publ Fed Am Soc Exp Biol 17(11):1411–1421
95. Nayak D, Huo Y, Kwang WX, Pushparaj PN, Kumar SD, Ling EA, Dheen ST (2010)
Sphingosine kinase 1 regulates the expression of proinflammatory cytokines and nitric oxide
in activated microglia. Neuroscience 166(1):132–144
96. Hammad SM, Crellin HG, Wu BX, Melton J, Anelli V, Obeid LM (2008) Dual and distinct
roles for sphingosine kinase 1 and sphingosine 1 phosphate in the response to inflammatory
stimuli in RAW macrophages. Prostaglandins Other Lipid Mediat 85(3–4):107–114
97. Snider AJ (2010) Orr Gandy KA, Obeid LM: Sphingosine kinase: Role in regulation of bio-
active sphingolipid mediators in inflammation. Biochimie 92(6):707–715
98. Massiello A, Salas A, Pinkerman RL, Roddy P, Roesser JR, Chalfant CE (2004)
Identification of two RNA cis-elements that function to regulate the 5′ splice site selection
of Bcl-x pre-mRNA in response to ceramide. J Biol Chem 279(16):15799–15804
99. Gomez-Munoz A, Kong JY, Parhar K, Wang SW, Gangoiti P, Gonzalez M, Eivemark S,
Salh B, Duronio V, Steinbrecher UP (2005) Ceramide-1-phosphate promotes cell survival
through activation of the phosphatidylinositol 3-kinase/protein kinase B pathway. FEBS
Lett 579(17):3744–3750
100. Nava VE, Cuvillier O, Edsall LC, Kimura K, Milstien S, Gelmann EP, Spiegel S (2000)
Sphingosine enhances apoptosis of radiation-resistant prostate cancer cells. Cancer Res
60(16):4468–4474
101. Sweeney EA, Sakakura C, Shirahama T, Masamune A, Ohta H, Hakomori S, Igarashi Y (1996)
Sphingosine and its methylated derivative N, N-dimethylsphingosine (DMS) induce apoptosis in a
variety of human cancer cell lines. Int J Cancer J Int du Cancer 66(3):358–366
102. Sawai H, Okazaki T, Domae N (2002) Sphingosine-induced c-jun expression: differences between
sphingosine- and C2-ceramide-mediated signaling pathways. FEBS Lett 524(1–3):103–106
103. Phillips DC, Martin S, Doyle BT, Houghton JA (2007) Sphingosine-induced apoptosis in rhabdo-
myosarcoma cell lines is dependent on pre-mitochondrial Bax activation and post-mitochondrial
caspases. Cancer Res 67(2):756–764
104. Guan F, Handa K, Hakomori SI (2009) Specific glycosphingolipids mediate epithelial-to-
mesenchymal transition of human and mouse epithelial cell lines. Proc Nat Acad Sci USA
106(18):7461–7466
8 Sphingolipid Metabolism and Signaling 227
105. Bremer EG, Schlessinger J, Hakomori S (1986) Ganglioside-mediated modulation of cell growth.
Specific effects of GM3 on tyrosine phosphorylation of the epidermal growth factor receptor. J
Biol Chem 261(5):2434–2440
106. Zhou Q, Hakomori S, Kitamura K, Igarashi Y (1994) GM3 directly inhibits tyrosine phosphoryla-
tion and de-N-acetyl-GM3 directly enhances serine phosphorylation of epidermal growth factor
receptor, independently of receptor–receptor interaction. J Biol Chem 269(3):1959–1965
107. Meuillet EJ, Kroes R, Yamamoto H, Warner TG, Ferrari J, Mania-Farnell B, George D, Rebbaa
A, Moskal JR, Bremer EG (1999) Sialidase gene transfection enhances epidermal growth factor
receptor activity in an epidermoid carcinoma cell line, A431. Cancer Res 59(1):234–240
108. Coskun U, Grzybek M, Drechsel D, Simons K (2011) Regulation of human EGF receptor by
lipids. Proc Nat Acad Sci USA 108(22):9044–9048
109. Ogretmen B, Hannun YA (2004) Biologically active sphingolipids in cancer pathogenesis and
treatment. Nat Rev Cancer 4(8):604–616
110. Bose R (1995) Ceramide synthase mediates daunorubicin-induced apoptosis: an alternative mech-
anism for generating death signals. Cell 82:405–414
111. Perry DK (2000) Serine palmitoyltransferase regulates de novo ceramide generation during etopo-
side-induced apoptosis. J Biol Chem 275:9078–9084
112. Chauvier D, Morjani H, Manfait M (2002) Ceramide involvement in homocamptothecin- and
camptothecin-induced cytotoxicity and apoptosis in colon HT29 cells. Int J Oncol 20:855–863
113. Strum JC (1994) 1-[beta]-D-Arabinofuranosylcytosine stimulates ceramide and digylceride for-
mation in HL-60 cells. J Biol Chem 269:15493–15497
114. Bezombes C (2001) Oxidative stress-induced activation of Lyn recruits sphingomyelinase and is
requisite for its stimulation by Ara-C. FASEB J 15:1583–1585
115. Dbaibo GS (1998) p53-dependent ceramide response to genotoxic stress. J Clin Invest
102:329–339
116. Selzner M, Bielawska A, Morse MA, Rudiger HA, Sindram D, Hannun YA, Clavien PA (2001)
Induction of apoptotic cell death and prevention of tumor growth by ceramide analogues in meta-
static human colon cancer. Cancer Res 61(3):1233–1240
117. Meng A, Luberto C, Meier P, Bai A, Yang X, Hannun YA, Zhou D (2004) Sphingomyelin syn-
thase as a potential target for D609-induced apoptosis in U937 human monocytic leukemia cells.
Exp Cell Res 292(2):385–392
118. Modrak DE, Cardillo TM, Newsome GA, Goldenberg DM, Gold DV (2004) Synergistic interac-
tion between sphingomyelin and gemcitabine potentiates ceramide-mediated apoptosis in pancre-
atic cancer. Cancer Res 64(22):8405–8410
119. Veldman RJ, Zerp S, van Blitterswijk WJ, Verheij M (2004) N-hexanoyl-sphingomyelin potenti-
ates in vitro doxorubicin cytotoxicity by enhancing its cellular influx. Br J Cancer 90(4):917–925
120. Radin NS (2003) Killing tumours by ceramide-induced apoptosis: a critique of available drugs.
Biochem J 371(Pt 2):243–256
121. Bieberich E, Kawaguchi T, Yu RK (2000) N-acylated serinol is a novel ceramide mimic inducing
apoptosis in neuroblastoma cells. J Biol Chem 275(1):177–181
122. Bieberich E (2002) Synthesis and characterization of novel ceramide analogs for induction of
apoptosis in human cancer cells. Cancer Lett 181:55–64
123. Struckhoff AP (2004) Novel ceramide analogs as potential chemotherapeutic agents in breast can-
cer. J Pharmacol Exp Ther 309:523–532
124. Crawford KW (2003) Novel ceramide analogs display selective cytotoxicity in drug-resistant
breast tumor cell lines compared to normal breast epithelial cells. Cell Mol Biol 49:1017–1023
125. Dindo D, Dahm F, Szulc Z, Bielawska A, Obeid LM, Hannun YA, Graf R, Clavien PA (2006)
Cationic long-chain ceramide LCL-30 induces cell death by mitochondrial targeting in SW403
cells. Mol Cancer Ther 5(6):1520–1529
126. Senkal CE, Ponnusamy S, Rossi MJ, Sundararaj K, Szulc Z, Bielawski J, Bielawska A,
Meyer M, Cobanoglu B, Koybasi S et al (2006) Potent antitumor activity of a novel cati-
onic pyridinium-ceramide alone or in combination with gemcitabine against human
head and neck squamous cell carcinomas in vitro and in vivo. J Pharmacol Exp Ther
317(3):1188–1199
228 V. Rajagopalan and Y. A. Hannun
127. Rossi MJ, Sundararaj K, Koybasi S, Phillips MS, Szulc ZM, Bielawska A, Day TA, Obeid LM,
Hannun YA, Ogretmen B (2005) Inhibition of growth and telomerase activity by novel cationic
ceramide analogs with high solubility in human head and neck squamous cell carcinoma cells.
Otolaryngol Head Neck Surg 132(1):55–62
128. Novgorodov SA, Szulc ZM, Luberto C, Jones JA, Bielawski J, Bielawska A, Hannun YA, Obeid
LM (2005) Positively charged ceramide is a potent inducer of mitochondrial permeabilization. J
Biol Chem 280(16):16096–16105
129. Stover T, Kester M (2003) Liposomal delivery enhances short-chain ceramide-induced apoptosis
of breast cancer cells. J Pharmacol Exp Ther 307:468–475
130. Thomas DA, Sarris AH, Cortes J, Faderl S, O’Brien S, Giles FJ, Garcia-Manero G, Rodriguez
MA, Cabanillas F, Kantarjian H (2006) Phase II study of sphingosomal vincristine in patients with
recurrent or refractory adult acute lymphocytic leukemia. Cancer 106(1):120–127
131. Shirahama T, Sweeney EA, Sakakura C, Singhal AK, Nishiyama K, Akiyama S, Hakomori
S, Igarashi Y (1997) In vitro and in vivo induction of apoptosis by sphingosine and N,
N-dimethylsphingosine in human epidermoid carcinoma KB-3-1 and its multidrug-resistant cells.
Clinical cancer research : an official journal of the American Association for Cancer Research
3(2):257–264
132. Sweeney EA (1996) Sphingosine and its methylated derivative N, N-dimethylsphingosine (DMS)
induce apoptosis in a variety of human cancer cell lines. Int J Cancer 66:358–366
133. Cuvillier O, Levade T (2001) Sphingosine 1-phosphate antagonizes apoptosis of human leuke-
mia cells by inhibiting release of cytochrome c and Smac/DIABLO from mitochondria. Blood
98(9):2828–2836
134. Tilly JL, Kolesnick RN (2002) Sphingolipids, apoptosis, cancer treatments and the ovary: investi-
gating a crime against female fertility. Biochim Biophys Acta 1585(2–3):135–138
135. Suomalainen L, Hakala JK, Pentikainen V, Otala M, Erkkila K, Pentikainen MO, Dunkel L
(2003) Sphingosine-1-phosphate in inhibition of male germ cell apoptosis in the human testis. J
Clin Endocrinol Metab 88(11):5572–5579
136. Graler MH, Goetzl EJ (2004) The immunosuppressant FTY720 down-regulates sphingosine
1-phosphate G protein-coupled receptors. FASEB J 10
137. Billich A (2003) Phosphorylation of the immunomodulatory drug FTY720 by sphingosine
kinases. J Biol Chem 278:47408–47415
138. Paugh SW (2003) The immunosuppressant FTY720 is phosphorylated by sphingosine kinase
type 2. FEBS Lett 554:189–193
139. Azuma H (2002) Marked prevention of tumor growth and metastasis by a novel immunosuppres-
sive agent, FTY720, in mouse breast cancer models. Cancer Res 62:1410–1419
140. Wang JD (1999) Early induction of apoptosis in androgen-independent prostate cancer cell line by
FTY720 requires caspase-3 activation. Prostate 40:50–55
141. Sonoda Y (2001) FTY720, a novel immunosuppressive agent, induces apoptosis in human glioma
cells. Biochem Biophys Res Commun 281:282–288
142. Azuma H (2003) Induction of apoptosis in human bladder cancer cells in vitro and in vivo caused
by FTY720 treatment. J Urol 169:2372–2377
143. Dudeja PK, Dahiya R, Brasitus TA (1986) The role of sphingomyelin synthetase and sphingo-
myelinase in 1, 2-dimethylhydrazine-induced lipid alterations of rat colonic plasma membranes.
Biochim Biophys Acta 863(2):309–312
144. Dillehay DL, Webb SK, Schmelz EM, Merrill AH Jr (1994) Dietary sphingomyelin inhibits 1,
2-dimethylhydrazine-induced colon cancer in CF1 mice. J Nutr 124(5):615–620
145. Schmelz EM, Dillehay DL, Webb SK, Reiter A, Adams J, Merrill AH Jr (1996) Sphingomyelin
consumption suppresses aberrant colonic crypt foci and increases the proportion of adenomas ver-
sus adenocarcinomas in CF1 mice treated with 1, 2-dimethylhydrazine: implications for dietary
sphingolipids and colon carcinogenesis. Cancer Res 56(21):4936–4941
146. Schmelz EM, Bushnev AS, Dillehay DL, Sullards MC, Liotta DC, Merrill AH Jr (1999)
Ceramide-beta-D-glucuronide: synthesis, digestion, and suppression of early markers of colon
carcinogenesis. Cancer Res 59(22):5768–5772
8 Sphingolipid Metabolism and Signaling 229
147. Schmelz EM, Bushnev AS, Dillehay DL, Liotta DC, Merrill AH Jr (1997) Suppression of aber-
rant colonic crypt foci by synthetic sphingomyelins with saturated or unsaturated sphingoid base
backbones. Nutr Cancer 28(1):81–85
148. Mazzei JC, Zhou H, Brayfield BP, Hontecillas R, Bassaganya-Riera J, Schmelz EM (2011)
Suppression of intestinal inflammation and inflammation-driven colon cancer in mice by dietary
sphingomyelin: importance of peroxisome proliferator-activated receptor gamma expression. J
Nutr Biochem 22(12):1160–1171
149. Bleicher RJ, Cabot MC (2002) Glucosylceramide synthase and apoptosis. Biochim Biophys Acta
1585:172–178
150. Liu X, Ryland L, Yang J, Liao A, Aliaga C, Watts R, Tan SF, Kaiser J, Shanmugavelandy SS,
Rogers A et al (2010) Targeting of survivin by nanoliposomal ceramide induces complete remis-
sion in a rat model of NK-LGL leukemia. Blood 116(20):4192–4201
151. Liu YY (2001) Ceramide glycosylation potentiates cellular multidrug resistance. FASEB J
15:719–730
152. Liu YY (2004) Oligonucleotides blocking glucosylceramide synthase expression selectively
reverse drug resistance in cancer cells. J Lipid Res 45:933–940
153. Gouaze V, Liu YY, Prickett CS, Yu JY, Giuliano AE, Cabot MC (2005) Glucosylceramide syn-
thase blockade down-regulates P-glycoprotein and resensitizes multidrug-resistant breast cancer
cells to anticancer drugs. Cancer Res 65(9):3861–3867
154. Gouaze-Andersson V, Yu JY, Kreitenberg AJ, Bielawska A, Giuliano AE, Cabot MC (2007)
Ceramide and glucosylceramide upregulate expression of the multidrug resistance gene MDR1 in
cancer cells. Biochim Biophys Acta 1771(12):1407–1417
155. Weiss M (2003) Inhibition of melanoma tumor growth by a novel inhibitor of glucosylceramide
synthase. Cancer Res 63:3654–3658
156. Wang H, Maurer BJ, Liu YY, Wang E, Allegood JC, Kelly S, Symolon H, Liu Y, Merrill AH
Jr, Gouaze-Andersson V et al (2008) N-(4-Hydroxyphenyl)retinamide increases dihydrocera-
mide and synergizes with dimethylsphingosine to enhance cancer cell killing. Mol Cancer Ther
7(9):2967–2976
157. Rahmaniyan M, Curley RW Jr, Obeid LM, Hannun YA, Kraveka JM (2011) Identification
of dihydroceramide desaturase as a direct in vitro target for fenretinide. J Biol Chem
286(28):24754–24764
158. Maurer BJ (1999) Increase of ceramide and induction of mixed apoptosis/necrosis by N-(4-
hydroxyphenyl)-retinamide in neuroblastoma cell lines. J Natl Cancer Inst 91:1138–1146
159. Maurer BJ (2000) Synergistic cytotoxicity in solid tumor cell lines between N-(4-
hydroxyphenyl)retinamide and modulators of ceramide metabolism. J Natl Cancer Inst
92:1897–1909
160. Bektas M, Jolly PS, Muller C, Eberle J, Spiegel S, Geilen CC (2005) Sphingosine kinase activ-
ity counteracts ceramide-mediated cell death in human melanoma cells: role of Bcl-2 expression.
Oncogene 24(1):178–187
161. Akao Y, Banno Y, Nakagawa Y, Hasegawa N, Kim TJ, Murate T, Igarashi Y, Nozawa Y (2006)
High expression of sphingosine kinase 1 and S1P receptors in chemotherapy-resistant prostate
cancer PC3 cells and their camptothecin-induced up-regulation. Biochem Biophys Res Commun
342(4):1284–1290
162. Marfe G, Di Stefano C, Gambacurta A, Ottone T, Martini V, Abruzzese E, Mologni L, Sinibaldi-
Salimei P, de Fabritis P, Gambacorti-Passerini C et al (2011) Sphingosine kinase 1 overexpression
is regulated by signaling through PI3K, AKT2, and mTOR in imatinib-resistant chronic myeloid
leukemia cells. Exp Hematol 39(6):653–665 e656
Chapter 9
Leading Small Molecule Inhibitors
of Anti-Apoptotic Bcl-2 Family Members
9.1 Introduction
V. Y. Yazbeck (*) · D. E. Johnson
Department of Medicine, University of Pittsburgh
and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
e-mail: [email protected]
D. E. Johnson
Department of Pharmacology and Chemical Biology, University of Pittsburgh
and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 231
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_9,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
232 V. Y. Yazbeck and D. E. Johnson
drug design, and fragment-based drug design. To date, several inhibitors have
been isolated from natural sources, while others have been identified in chemi-
cal libraries or developed via medicinal chemistry efforts. Among the inhibitors
that have been reported are peptides derived from BH3 domains of proapoptotic
proteins [1–4], antimycin A3 [5], HA14-1 [6], BH3I compounds [7], several tea
polyphenol compounds [8, 9], chelerythrine [10], sanguinarine [11], (-)-gossypol
[12–14], apogossypolone [15, 16], TW-37 [13], ABT-737 [17], ABT-263 [18], and
GX15-070 [19]. This chapter will focus on three of these inhibitors, ABT-737,
ABT-263, and GX15-070, that have shown particular promise in preclinical stud-
ies and have advanced rapidly to clinical testing. The identification and initial in
vitro evaluation of these compounds will first be described. We will then summa-
rize the current understanding regarding their mechanism of action and the mecha-
nisms which lead to resistance to these inhibitors. Additionally, reported synergies
between these inhibitors and conventional chemotherapeutic agents will be dis-
cussed. Lastly, results obtained from evaluation of the inhibitors in preclinical in
vivo models and human clinical trials will be presented.
Fig. 9.1 Chemical structures
of ABT-737, ABT-263, and
GX15-070
overexpression of the anti-apoptotic Bcl-2 family member Mcl-1 [20, 28, 29]. For
example, Mcl-1 is highly expressed in HeLa cells that are resistant to ABT-737,
but reduction of Mcl-1 levels following CDK2 inhibition, DNA-damaging chemo-
therapy, or shRNA treatment, results in enhanced sensitivity to the compound [29].
In solid tumor cell lines, the induction of proapoptotic NOXA following the treat-
ment with chemotherapy or radiation leads to the inhibition of Mcl-1 and result-
ing synergy with ABT-737 [27, 30]. Efficient cell killing by ABT-737 requires
that binding of the compound to anti-apoptotic Bcl-2 family members results in
the release of bound proapoptotic proteins (e.g., Bim, Bax, Bak). Moreover, the
amount of released proapoptotic proteins must be sufficient to saturate empty
Mcl-1 and A1/Bfl-1 that are present in the cell [21].
Deng et al. have studied the mechanism whereby lymphoma cell lines develop
resistance to ABT-737 and have classified three forms of resistance. Class A resist-
ance is observed in cells that express only low levels of BH3-only activator pro-
teins, whereas Class B resistance is due to significant loss or mutation of Bax
234 V. Y. Yazbeck and D. E. Johnson
9.3.1 Leukemia
9.3.2 Multiple Myeloma
9.3.3 Lymphoma
In several lymphoma cell lines, including those representing MCL and DLBCL,
ABT-737 showed dose-dependent cytotoxicity and synergized with proteasome
inhibitors [55] and the histone deacetylase inhibitor vorinostat [56]. Combination
with bortezomib also demonstrated synergy in primary samples of MCL, DLBCL,
and CLL, but no significant cytotoxic effects were observed in PBMCs from
healthy donors [55]. In Hodgkin’s lymphoma (HL) cell lines, ABT-737 exhibits
dose- and time-dependant cytotoxicity as a single agent and enhanced the activity
of several conventional anti-lymphoma agents [57]. In cutaneous T-cell lymphoma
(CTCL), ABT-737 was found to synergize with the pan-histone deacetylase inhibi-
tor panobinostat [58]. Also, in a murine model of c-myc-driven B-cell lymphoma,
ABT-737 improved overall survival when compared to vehicle alone, even in the
face of Bcl-2 overexpression [29].
236 V. Y. Yazbeck and D. E. Johnson
9.3.4 Lung Cancer
9.3.5 Gastrointestinal Malignancies
In hepatoblastoma cell lines, ABT-737 and GX15-070 (see Sects. 9.7, 9.8, 9.9)
exhibit additive effects when combined with standard chemotherapy [61]. ABT-
737 also synergizes with sorafenib against hepatocellular carcinoma (HCC) cell
lines and HCC xenograft tumors [62, 63]. Inhibition of cholangiocarcinoma cell
line proliferation with ABT-737 is observed with IC50 values in the 4–17 μM
range, and synergy with zoledronic acid has also been reported [64]. In pancreatic
cancer cell lines, ABT-737 synergizes with actinomycin D [38] and TRAIL [65].
Similarly, in human colorectal carcinoma (CRC) cell lines, ABT-737 induced dose-
dependent apoptosis and synergized with irinotecan [66] and oxaliplatin [67]. ABT-
737 also enhanced celecoxib-induced apoptosis and autophagy [68], and overcame
resistance to immunotoxin-mediated apoptosis in CRC cell lines [69]. Single-
agent ABT-737 activity against imatinib-sensitive and imatinib-resistant gastro-
intestinal stromal tumor (GIST) cell lines (IC50s of 1–10 μM) has been reported.
Furthermore, potent synergy with imatinib was observed in these models [70].
ABT-737 synergizes with fenretinide in neuroblastoma cell lines, and the combination
increased event-free survival in mice harboring neuroblastoma xenograft tumors [78].
9.3.8 Genitourinary Malignancies
Renal cell carcinoma (RCC) cell lines exhibit very little sensitivity to ABT-737
as a single agent. However, strong super-additive effects were observed when
ABT-737 was combined with etoposide, vinblastine, or paclitaxel [79]. In pros-
tate cancer, ABT-737 synergizes with docetaxel in vitro [80] and with Pim kinase
inhibitors both in vitro and in vivo [81].
9.3.9 Other Malignancies
Single-agent ABT-737 is largely ineffective against melanoma cell lines, but dem-
onstrates synergy when combined with temodar [82], dacarbazine, fotemustine,
imiquimod [83], pseudomonas exotoxin A [84], and p38 MAPK inhibitor [85].
Similarly, ABT-737 alone is ineffective against head and neck squamous cell carci-
noma (HNSCC) cell lines, but produces marked synergy characterized by NOXA
upregulation when combined with cisplatin or etoposide [27]. In human erythroid
cells expressing mutant JAK2, ABT-737 enhances apoptosis induced by JAK2
inhibitor [86]. ABT-737 also enhances the effect of interferon-α (IFN-α) against
JAK2V617F-positive polycythemia vera hematopoietic progenitor cells [87].
Interestingly, ABT-737 induces apoptosis of mast cells in vitro and in vivo [88]
and is effective in treating animal models of arthritis and lupus [89].
Despite the potency of ABT-737 in targeting Bcl-2 and Bcl-XL, the compound
is not orally bioavailable and exhibits low solubility, hindering intravenous
delivery. Hence, Abbott Laboratories sought to develop second-generation deriv-
atives that would address these issues. Through medicinal chemistry efforts and
further structure-based rational design, an effective ABT-737 derivative named
ABT-263 (see Fig. 9.1b; ABT-263 is also called navitoclax) was developed [18].
ABT-263, like ABT-737, was shown to bind to purified anti-apoptotic Bcl-2 fam-
ily proteins with high affinity (Ki’s < 1 nM for Bcl-2, Bcl-XL, and Bcl-w). The
compound acts by disrupting the interactions of Bcl-2 and Bcl-XL with proap-
optotic Bcl-2 family members, leading to the Bax activation and induction of
238 V. Y. Yazbeck and D. E. Johnson
Initial reports demonstrated that ABT-263 is cytotoxic for cell lines represent-
ing SCLC and various hematologic malignancies [18]. ABT-263 also induced
complete tumor regression in xenograft models of SCLC [18, 90] and ALL [18].
However, in xenograft models of aggressive B-cell lymphoma and MM, single-
agent ABT-263 exhibited only modest or no activity. On the other hand, ABT-263
significantly enhances the activity of other clinically relevant agents in xenograft
tumor models [18], including enhancement of erlotinib activity and the activities
of several conventional chemotherapy drugs [91, 92].
9.5.1 Solid Tumors
9.5.2 Hematologic Malignancies
cell lines and primary patient specimens, ABT-263 synergizes with the histone dea-
cetylase inhibitor, vorinostat [100]. The addition of ABT-263 to rapamycin enhances
apoptosis in FL cell lines and promotes tumor regression in vivo [101]. ABT-263
also enhances the activity of bendamustine against DLBCL, MCL, and Burkitt’s
lymphoma in vivo and improves the response to a bendamustine/rituximab regimen
in a subset of these xenograft tumors [102].
9.6.1 Leukemia
9.6.2 Lymphoma
9.6.3 Solid Tumors
9.8.1 Leukemia
Using AML cell lines, Konopleva et al. [20] determined that GX15-070 induces
time- and dose-dependent cell death through activation of the intrinsic apoptotic
pathway, with IC50 values in the low micromolar range (1.1–5.0 μM). At lower
concentrations, GX15-070 induced cell cycle arrest at S-G2 phase. Treatment of
AML progenitor cells with GX15-070 resulted in potent induction of apoptosis
(IC50 = 3.6 ± 1.2 μM), and clonogenicity was inhibited by concentrations in
the 75–100 nM range. GX15-070 synergized with ABT-737 against AML cell
lines and synergized with Ara-C against leukemia cell lines and primary AML
specimens [20].
In other studies, GX15-070 demonstrated synergy with a histone deacetylase
inhibitor [112], and the multi-kinase inhibitor sorafenib against AML cell lines
and primary AML cells, but not against normal CD34+ cells [113]. Combination
of GX15-070 and sorafenib markedly reduced AML tumor growth in a xenograft
model and significantly enhanced the survival of mice bearing these tumors when
compared with either agent alone [113]. Single-agent GX15-070 was found to
inhibit growth and activate both apoptosis and autophagy in ALL cell lines that
were either sensitive or resistant to dexamethasone [114]. In primary CLL cells
derived from patients who had not received treatment in the past 3 months, GX15-
070 demonstrated killing activity in most specimens, including those characterized
by poor prognostic features such as deletion of 11q or 17p [115]. However, ZAP-
70-positive cases, associated with poor prognosis, were less sensitive to GX15-070
than were ZAP-70 negative cases. Additionally, inhibition of extracellular signal-
regulated kinase (ERK)-1/2 was found to inhibit Bcl-2 phosphorylation in CLL
cells, increasing the sensitivities of these cells to GX15-070 alone or in combina-
tion with proteasome inhibitors [115].
9.8.2 Lymphoma
In B-cell lymphoma cell lines, GX15-070 has been reported to enhance ritux-
imab activity [116]. Moreover, GX15-070 synergized with chemotherapy
to induce apoptosis in both rituximab/chemotherapy-sensitive (RSCL) and
rituximab/chemotherapy-resistant cell lines and primary tumor cells from B-cell
non-Hodgkin’s lymphoma patients [116]. Via NFκB inhibition and inhibition
242 V. Y. Yazbeck and D. E. Johnson
of the DR5 repressor Yin Yang 1 (YY1), GX15-070 induced DR5 expression
and sensitized B-cell lymphoma cells to TRAIL-induced apoptosis [111].
In Hodgkin’s lymphomas, ABT-737 and GX15-070 demonstrated synergy with
the histone deacetylase inhibitor SNDX-275 [117], while in MCL cell lines and
primary cells, GX15-070 demonstrated single-agent activity and synergy with
bortezomib [118].
9.8.3 Multiple Myeloma
9.8.4 Solid Tumors
9.9.1 Leukemia
The first phase I study of single-agent obatoclax mesylate (the clinical formulation of
GX15-070; hereafter referred to as obatoclax) was undertaken in patients with refrac-
tory hematological malignancies [126]. A total of 44 patients with refractory AML,
ALL, CLL, myelodysplasia (MDS), or CML in blast crisis were given 24-h infusions
of the drug, with a median of five infusions per patient. The drug was found to be
well tolerated, aside from grade 1/2 CNS symptoms, and the highest planned dose
was reached without any DLT. One AML patient obtained complete remission for
eight months, but subsequently relapsed, causing the authors to suggest that GX15-
070 induced proliferation arrest, as opposed to differentiation. The final recom-
mended dose for phase II was 28 mg/m2, given as an infusion for 24 h up to four
consecutive days. The authors concluded that obatoclax was well tolerated and sug-
gested further investigation in patients with leukemia and myelodysplasia [126].
A subsequent phase I trial of single-agent obatoclax enrolled 26 CLL patients
[127]. Patients were treated with single-agent obatoclax doses ranging from 3.5
to 14 mg/m2 as a 1-h infusion and from 20 to 40 mg/m2 as a 3-h infusion every
3 weeks. The observed neurological dose-limiting reactions (euphoria, ataxia, som-
nolence) were dose related, but quickly resolved after cessation of drug infusion.
Only one patient achieved partial remission at an obatoclax dose of 3.5 mg/m2. This
patient had been pre-treated with fludarabine, Rituxan, and alemtuzumab but was
the only patient in the trial who was naïve to alkylating agents. In other patients,
there was reduction in transfusion dependence. The MTD was determined to be
28 mg/m2 over 3 h every three weeks and was recommended for phase II studies.
The authors concluded that the biologic activity of obatoclax is not only dependant
on the dose, but also on tumor biology. The activity was modest in this population
and suggested investigation of the drug in a less heavily pre-treated population and
in combination with other agents [127].
9.9.2 Lymphoma
In another series of trials, 35 patients with previously treated lymphoma and solid
tumors, who were not candidates for standard therapies, were enrolled in two
phase I trials (GX001 and GX005) evaluating the safety and tolerability of single-
agent obatoclax given as 1-h (GX001) or 3-h (GX005) weekly infusions [128].
With the 1-h infusion schedule, neurological DLT (somnolence) was observed at
5 and 7 mg/m2. The MTD was 1.25 mg/m2. Patients on the 3-h infusion schedule
showed better tolerability, with an MTD of 20 mg/m2. Stable disease by RECIST
criteria was achieved in 25 % (2/8) of patients with the 1-h infusion and in 18 %
(5/27) of patients with the 3-h infusion. A partial response was observed in one
244 V. Y. Yazbeck and D. E. Johnson
patient with large cell lymphoma stage IV assigned to the 28 mg/m2 dose group,
who received a total of 32 weeks of therapy. The authors concluded that the 3-h
weekly infusion of obatoclax was better tolerated with an MTD of 20 mg/m2 and
evidence of clinical activity [128].
Recently, Oki et al. [129] reported a phase II trial in patients with relapsed or
refractory classical Hodgkin’s lymphoma (cHL) treated with single-agent obato-
clax administered intravenously at 60 mg over 24 h and given every two weeks.
Thirteen patients received at least one dose of obatoclax, with a median of four
cycles per patient (range 1–24). The drug was found to be well tolerated, with
grade 1 toxicities: dizziness (n = 5), euphoria (n = 3), and hypotension (n = 1).
There were no objective responses; 38 % (5/13) of patients had stable disease. The
authors concluded that obatoclax showed limited activity in this heavily pre-treated
population of cHL. They recommended investigation of more potent Bcl-2 family
inhibitors with pharmacodynamic studies to ensure target inhibition and biomarker
analysis for plausible patient selection [129].
9.9.3 Myelofibrosis
Twenty-two patients who had previously been treated were enrolled in a multi-
center, open-label, non-comparative phase II study of single-agent obatoclax
administered as a 24-h infusion every 2 weeks at a fixed dose of 60 mg [130].
Patients received a median of 7 cycles. No objective responses were observed.
Only one patient had clinical improvement (a decrease in transfusion require-
ment). The most common side effects were low-grade ataxia and fatigue, observed
in 50 % of the patients. One patient had a dose reduction secondary to toxicity,
and two patients were taken off the study due to grade 3 toxicity (ataxia and heart
failure). The authors concluded that obatoclax showed no clinical activity in this
patient population at the dose and schedule used in the study [130].
9.9.4 Solid Tumors
A phase I trial studied the safety, MTD, and early anti-tumor activity of the com-
bination of topotecan and obatoclax in patients (n = 14) with solid tumors who
would benefit from treatment with topotecan. Obatoclax was given at a start-
ing dose of 14 mg/m2 over a 3-h intravenous infusion on a 3-week cycle, while
topotecan was given at 1.25 mg/m2 on days 1–5 on a every 3-week cycle [131]. Of
the observed toxicities, 88 % were grade 1 and 2, mainly neurologic, in the form
of ataxia, somnolence, mood alterations, and cognitive dysfunction. Two-fifths
of the patients developed grade 3 neurological DLTs at the 20 mg/m2 dose. Two
patients (SCLC) exhibited partial responses, and four (three with SCLC and one
with pulmonary carcinoid) had stable disease. The recommended dose for phase
9 Leading Small Molecule Inhibitors of Anti-Apoptotic Bcl-2 Family Members 245
Acknowledgments We apologize to those authors whose works have not been cited due to
space limitations, or our oversight. This work was supported by National Institutes of Health
grants R01 CA137260 and P50 CA097190.
References
1. Finnegan NM, Curtin JF, Prevost G, Morgan B, Cotter TG (2001) Induction of apoptosis in
prostate carcinoma cells by BH3 peptides which inhibit Bak/Bcl-2 interactions. Br J Cancer
85(1):115–121 Epub 2001/07/05
2. Certo M, Del Gaizo Moore V, Nishino M, Wei G, Korsmeyer S, Armstrong SA et al (2006)
Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2
family members. Cancer Cell 9(5):351–365 Epub 2006/05/16
3. Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG et al (2005) Differential target-
ing of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apop-
totic function. Mol Cell 17(3):393–403 Epub 2005/02/08
4. Willis SN, Fletcher JI, Kaufmann T, van Delft MF, Chen L, Czabotar PE et al (2007)
Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak.
Science 315(5813):856–859 Epub 2007/02/10
5. Tzung SP, Kim KM, Basanez G, Giedt CD, Simon J, Zimmerberg J et al (2001) Antimycin
A mimics a cell-death-inducing Bcl-2 homology domain 3. Nat Cell Biol 3(2):183–191
Epub 2001/02/15
246 V. Y. Yazbeck and D. E. Johnson
6. Wang JL, Liu D, Zhang ZJ, Shan S, Han X, Srinivasula SM et al (2000) Structure-based
discovery of an organic compound that binds Bcl-2 protein and induces apoptosis of tumor
cells. Proc Natl Acad Sci U S A 97(13):7124–7129 Epub 2000/06/22
7. Degterev A, Lugovskoy A, Cardone M, Mulley B, Wagner G, Mitchison T et al (2001)
Identification of small-molecule inhibitors of interaction between the BH3 domain and
Bcl-xL. Nat Cell Biol 3(2):173–182 Epub 2001/02/15
8. Nishikawa T, Nakajima T, Moriguchi M, Jo M, Sekoguchi S, Ishii M et al (2006) A green
tea polyphenol, epigallocatechin-3-gallate, induces apoptosis of human hepatocellular car-
cinoma, possibly through inhibition of Bcl-2 family proteins. J Hepatol 44(6):1074–1082
Epub 2006/02/17
9. Leone M, Zhai D, Sareth S, Kitada S, Reed JC, Pellecchia M (2003) Cancer prevention by
tea polyphenols is linked to their direct inhibition of antiapoptotic Bcl-2-family proteins.
Cancer Res 63(23):8118–8121 Epub 2003/12/18
10. Chan SL, Lee MC, Tan KO, Yang LK, Lee AS, Flotow H et al (2003) Identification of
chelerythrine as an inhibitor of BclXL function. J Biol Chem 278(23):20453–20456 Epub
2003/04/19
11. Zhang YH, Bhunia A, Wan KF, Lee MC, Chan SL, Yu VC et al (2006) Chelerythrine and
sanguinarine dock at distinct sites on BclXL that are not the classic BH3 binding cleft.
J Mol Biol 364(3):536–549 Epub 2006/10/03
12. Kitada S, Leone M, Sareth S, Zhai D, Reed JC, Pellecchia M (2003) Discovery, charac-
terization, and structure-activity relationships studies of proapoptotic polyphenols targeting
B-cell lymphocyte/leukemia-2 proteins. J Med Chem 46(20):4259–4264 Epub 2003/09/19
13. Wang G, Nikolovska-Coleska Z, Yang CY, Wang R, Tang G, Guo J et al (2006) Structure-
based design of potent small-molecule inhibitors of anti-apoptotic Bcl-2 proteins. J Med
Chem 49(21):6139–6142 Epub 2006/10/13
14. Qiu J, Levin LR, Buck J, Reidenberg MM (2002) Different pathways of cell killing by gos-
sypol enantiomers. Exp Biol Med (Maywood) 227(6):398–401 Epub 2002/05/31
15. Sun Y, Wu J, Aboukameel A, Banerjee S, Arnold AA, Chen J et al (2008) Apogossypolone,
a nonpeptidic small molecule inhibitor targeting Bcl-2 family proteins, effectively inhib-
its growth of diffuse large cell lymphoma cells in vitro and in vivo. Cancer Biol Ther
7(9):1418–1426 Epub 2008/09/05
16. Becattini B, Kitada S, Leone M, Monosov E, Chandler S, Zhai D et al (2004) Rational
design and real time, in-cell detection of the proapoptotic activity of a novel compound tar-
geting Bcl-X(L). Chem Biol 11(3):389–395 Epub 2004/05/05
17. Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA et al
(2005) An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature
435(7042):677–681 Epub 2005/05/20
18. Tse C, Shoemaker AR, Adickes J, Anderson MG, Chen J, Jin S et al (2008) ABT-263: a
potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res 68(9):3421–3428 Epub
2008/05/03
19. Trudel S, Li ZH, Rauw J, Tiedemann RE, Wen XY, Stewart AK (2007) Preclinical studies of
the pan-Bcl inhibitor obatoclax (GX015-070) in multiple myeloma. Blood 109(12):5430–
5438 Epub 2007/03/03
20. Konopleva M, Contractor R, Tsao T, Samudio I, Ruvolo PP, Kitada S et al (2006)
Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute
myeloid leukemia. Cancer Cell 10(5):375–388 Epub 2006/11/14
21. Del Gaizo Moore V, Brown JR, Certo M, Love TM, Novina CD, Letai A (2007) Chronic
lymphocytic leukemia requires BCL2 to sequester prodeath BIM, explaining sensitivity to
BCL2 antagonist ABT-737. J Clin Invest 117(1):112–121. Epub 2007/01/04
22. Howard AN, Bridges KA, Meyn RE, Chandra J (2009) ABT-737, a BH3 mimetic, induces
glutathione depletion and oxidative stress. Cancer Chemother Pharmacol 65(1):41–54 Epub
2009/05/01
23. Maiuri MC, Criollo A, Tasdemir E, Vicencio JM, Tajeddine N, Hickman JA et al (2007)
BH3-only proteins and BH3 mimetics induce autophagy by competitively disrupting the
interaction between Beclin 1 and Bcl-2/Bcl-X(L). Autophagy 3(4):374–376 Epub 2007/04/18
9 Leading Small Molecule Inhibitors of Anti-Apoptotic Bcl-2 Family Members 247
24. Kohl TM, Hellinger C, Ahmed F, Buske C, Hiddemann W, Bohlander SK et al (2007) BH3
mimetic ABT-737 neutralizes resistance to FLT3 inhibitor treatment mediated by FLT3-
independent expression of BCL2 in primary AML blasts. Leukemia 21(8):1763–1772 Epub
2007/06/08
25. Trudel S, Stewart AK, Li Z, Shu Y, Liang SB, Trieu Y et al (2007) The Bcl-2 family protein
inhibitor, ABT-737, has substantial antimyeloma activity and shows synergistic effect with
dexamethasone and melphalan. Clin Cancer Res 13(2 Pt 1):621–629 Epub 2007/01/27
26. Kang MH, Kang YH, Szymanska B, Wilczynska-Kalak U, Sheard MA, Harned TM et al
(2007) Activity of vincristine, L-ASP, and dexamethasone against acute lymphoblastic leuke-
mia is enhanced by the BH3-mimetic ABT-737 in vitro and in vivo. Blood 110(6):2057–2066
Epub 2007/05/31
27. Li R, Zang Y, Li C, Patel NS, Grandis JR, Johnson DE (2009) ABT-737 synergizes with
chemotherapy to kill head and neck squamous cell carcinoma cells via a Noxa-mediated
pathway. Mol Pharmacol 75(5):1231–1239 Epub 2009/02/28
28. Tahir SK, Yang X, Anderson MG, Morgan-Lappe SE, Sarthy AV, Chen J et al (2007)
Influence of Bcl-2 family members on the cellular response of small-cell lung cancer cell
lines to ABT-737. Cancer Res 67(3):1176–1183 Epub 2007/02/07
29. van Delft MF, Wei AH, Mason KD, Vandenberg CJ, Chen L, Czabotar PE et al (2006) The
BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via
Bak/Bax if Mcl-1 is neutralized. Cancer Cell 10(5):389–399 Epub 2006/11/14
30. Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T, Yamashita T et al (2000) Noxa, a BH3-
only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science
288(5468):1053–1058 Epub 2000/05/12
31. Deng J, Carlson N, Takeyama K, Dal Cin P, Shipp M, Letai A (2007) BH3 profiling identi-
fies three distinct classes of apoptotic blocks to predict response to ABT-737 and conven-
tional chemotherapeutic agents. Cancer Cell 12(2):171–185 Epub 2007/08/19
32. Chen S, Dai Y, Harada H, Dent P, Grant S (2007) Mcl-1 down-regulation potentiates ABT-
737 lethality by cooperatively inducing Bak activation and Bax translocation. Cancer Res
67(2):782–791 Epub 2007/01/20
33. Kline MP, Rajkumar SV, Timm MM, Kimlinger TK, Haug JL, Lust JA et al (2007) ABT-
737, an inhibitor of Bcl-2 family proteins, is a potent inducer of apoptosis in multiple mye-
loma cells. Leukemia 21(7):1549–1560 Epub 2007/04/27
34. Chauhan D, Velankar M, Brahmandam M, Hideshima T, Podar K, Richardson P et al
(2007) A novel Bcl-2/Bcl-X(L)/Bcl-w inhibitor ABT-737 as therapy in multiple myeloma.
Oncogene 26(16):2374–2380 Epub 2006/10/04
35. Miller LA, Goldstein NB, Johannes WU, Walton CH, Fujita M, Norris DA et al (2009) BH3
mimetic ABT-737 and a proteasome inhibitor synergistically kill melanomas through Noxa-
dependent apoptosis. J Invest Dermatol 129(4):964–971 Epub 2008/11/07
36. Cragg MS, Jansen ES, Cook M, Harris C, Strasser A, Scott CL (2008) Treatment of B-RAF
mutant human tumor cells with a MEK inhibitor requires Bim and is enhanced by a BH3
mimetic. J Clin Invest 118(11):3651–3659 Epub 2008/10/25
37. Bhat UG, Pandit B, Gartel AL (2010) ARC synergizes with ABT-737 to induce apoptosis in
human cancer cells. Mol Cancer Ther 9(6):1688–1696 Epub 2010/06/03
38. Olberding KE, Wang X, Zhu Y, Pan J, Rai SN, Li C (2010) Actinomycin D synergistically
enhances the efficacy of the BH3 mimetic ABT-737 by downregulating Mcl-1 expression.
Cancer Biol Ther 10(9):918–929 Epub 2010/09/08
39. Zhang C, Cai TY, Zhu H, Yang LQ, Jiang H, Dong XW et al (2011) Synergistic antitumor
activity of gemcitabine and ABT-737 in vitro and in vivo through disrupting the interaction
of USP9X and Mcl-1. Mol Cancer Ther 10(7):1264–1275 Epub 2011/05/14
40. Yin S, Dong Y, Li J, Fan L, Wang L, Lu J et al (2012) Methylseleninic acid potentiates
multiple types of cancer cells to ABT-737-induced apoptosis by targeting Mcl-1 and Bad.
Apoptosis 17(4):388–399 Epub 2011/12/20
41. Song JH, Kandasamy K, Kraft AS (2008) ABT-737 induces expression of the death
receptor 5 and sensitizes human cancer cells to TRAIL-induced apoptosis. J Biol Chem
283(36):25003–25013 Epub 2008/07/05
248 V. Y. Yazbeck and D. E. Johnson
42. Zhang W, Konopleva M, Ruvolo VR, McQueen T, Evans RL, Bornmann WG et al (2008)
Sorafenib induces apoptosis of AML cells via Bim-mediated activation of the intrinsic apop-
totic pathway. Leukemia 22(4):808–818 Epub 2008/01/18
43. Cencic R, Carrier M, Trnkus A, Porco JA Jr, Minden M, Pelletier J (2010) Synergistic effect
of inhibiting translation initiation in combination with cytotoxic agents in acute myeloge-
nous leukemia cells. Leuk Res 34(4):535–541 Epub 2009/09/04
44. Konopleva M, Milella M, Ruvolo P, Watts JC, Ricciardi MR, Korchin B et al (2012) MEK inhibi-
tion enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1
induction and modulation of MCL-1/BIM complex. Leukemia 26(4):778–787 Epub 2011/11/09
45. Kang MH, Wan Z, Kang YH, Sposto R, Reynolds CP (2008) Mechanism of synergy of
N-(4-hydroxyphenyl)retinamide and ABT-737 in acute lymphoblastic leukemia cell lines:
Mcl-1 inactivation. J Natl Cancer Inst 100(8):580–595 Epub 2008/04/10
46. High LM, Szymanska B, Wilczynska-Kalak U, Barber N, O’Brien R, Khaw SL et al (2010)
The Bcl-2 homology domain 3 mimetic ABT-737 targets the apoptotic machinery in acute
lymphoblastic leukemia resulting in synergistic in vitro and in vivo interactions with estab-
lished drugs. Mol Pharmacol 77(3):483–494 Epub 2009/12/30
47. Ishitsuka K, Kunami N, Katsuya H, Nogami R, Ishikawa C, Yotsumoto F et al (2012)
Targeting Bcl-2 family proteins in adult T-cell leukemia/lymphoma: in vitro and in vivo
effects of the novel Bcl-2 family inhibitor ABT-737. Cancer Lett 317(2):218–225 Epub
2011/12/06
48. Mason KD, Khaw SL, Rayeroux KC, Chew E, Lee EF, Fairlie WD et al (2009) The BH3
mimetic compound, ABT-737, synergizes with a range of cytotoxic chemotherapy agents in
chronic lymphocytic leukemia. Leukemia 23(11):2034–2041 Epub 2009/07/31
49. Kojima K, Duvvuri S, Ruvolo V, Samaniego F, Younes A, Andreeff M (2012) Decreased
sensitivity of 17p-deleted chronic lymphocytic leukemia cells to a small molecule BCL-2
antagonist ABT-737. Cancer 118(4):1023–1031 Epub 2011/07/16
50. Kuroda J, Kimura S, Andreeff M, Ashihara E, Kamitsuji Y, Yokota A et al (2008) ABT-737
is a useful component of combinatory chemotherapies for chronic myeloid leukaemias with
diverse drug-resistance mechanisms. Br J Haematol 140(2):181–190 Epub 2007/11/22
51. Kuroda J, Puthalakath H, Cragg MS, Kelly PN, Bouillet P, Huang DC et al (2006) Bim and Bad
mediate imatinib-induced killing of Bcr/Abl+ leukemic cells, and resistance due to their loss is
overcome by a BH3 mimetic. Proc Natl Acad Sci U S A 103(40):14907–14912 Epub 2006/09/26
52. Kuroda J, Kimura S, Strasser A, Andreeff M, O’Reilly LA, Ashihara E et al (2007)
Apoptosis-based dual molecular targeting by INNO-406, a second-generation Bcr-Abl
inhibitor, and ABT-737, an inhibitor of antiapoptotic Bcl-2 proteins, against Bcr-Abl-
positive leukemia. Cell Death Differ 14(9):1667–1677 Epub 2007/05/19
53. Bodet L, Gomez-Bougie P, Touzeau C, Dousset C, Descamps G, Maiga S et al (2011)
ABT-737 is highly effective against molecular subgroups of multiple myeloma. Blood
118(14):3901–3910 Epub 2011/08/13
54. Li M, Chen F, Clifton N, Sullivan DM, Dalton WS, Gabrilovich DI et al (2010) Combined
inhibition of Notch signaling and Bcl-2/Bcl-xL results in synergistic antimyeloma effect.
Mol Cancer Ther 9(12):3200–3209 Epub 2010/12/17
55. Paoluzzi L, Gonen M, Bhagat G, Furman RR, Gardner JR, Scotto L et al (2008) The BH3-
only mimetic ABT-737 synergizes the antineoplastic activity of proteasome inhibitors in
lymphoid malignancies. Blood 112(7):2906–2916 Epub 2008/07/02
56. Wiegmans AP, Alsop AE, Bots M, Cluse LA, Williams SP, Banks KM et al (2011)
Deciphering the molecular events necessary for synergistic tumor cell apoptosis mediated
by the histone deacetylase inhibitor vorinostat and the BH3 mimetic ABT-737. Cancer Res
71(10):3603–3615 Epub 2011/03/15
57. Jayanthan A, Howard SC, Trippett T, Horton T, Whitlock JA, Daisley L et al (2009)
Targeting the Bcl-2 family of proteins in Hodgkin lymphoma: in vitro cytotoxicity, target
modulation and drug combination studies of the Bcl-2 homology 3 mimetic ABT-737. Leuk
Lymphoma 50(7):1174–1182 Epub 2009/06/27
9 Leading Small Molecule Inhibitors of Anti-Apoptotic Bcl-2 Family Members 249
58. Chen J, Fiskus W, Eaton K, Fernandez P, Wang Y, Rao R et al (2009) Cotreatment with
BCL-2 antagonist sensitizes cutaneous T-cell lymphoma to lethal action of HDAC7-Nur77-
based mechanism. Blood 113(17):4038–4048 Epub 2008/12/17
59. Xu H, Krystal GW (2010) Actinomycin D decreases Mcl-1 expression and acts synergisti-
cally with ABT-737 against small cell lung cancer cell lines. Clin Cancer Res 16(17):4392–
4400 Epub 2010/08/25
60. Cragg MS, Kuroda J, Puthalakath H, Huang DC, Strasser A (2007) Gefitinib-induced killing
of NSCLC cell lines expressing mutant EGFR requires BIM and can be enhanced by BH3
mimetics. PLoS Med 4(10):1681–1689. Discussion 90. Epub 2007/11/02
61. Lieber J, Ellerkamp V, Wenz J, Kirchner B, Warmann SW, Fuchs J et al (2012) Apoptosis
sensitizers enhance cytotoxicity in hepatoblastoma cells. Pediatr Surg Int 28(2):149–159
Epub 2011/10/06
62. Galmiche A, Ezzoukhry Z, Francois C, Louandre C, Sabbagh C, Nguyen-Khac E et al
(2010) BAD, a proapoptotic member of the BCL2 family, is a potential therapeutic target in
hepatocellular carcinoma. Mol Cancer Res 8(8):1116–1125 Epub 2010/07/22
63. Hikita H, Takehara T, Shimizu S, Kodama T, Shigekawa M, Iwase K et al (2010) The
Bcl-xL inhibitor, ABT-737, efficiently induces apoptosis and suppresses growth of
hepatoma cells in combination with sorafenib. Hepatology 52(4):1310–1321 Epub
2010/08/28
64. Romani AA, Desenzani S, Morganti MM, Baroni MC, Borghetti AF, Soliani P (2011) The
BH3-mimetic ABT-737 targets the apoptotic machinery in cholangiocarcinoma cell lines
resulting in synergistic interactions with zoledronic acid. Cancer Chemother Pharmacol
67(3):557–567 Epub 2010/05/18
65. Huang S, Sinicrope FA (2008) BH3 mimetic ABT-737 potentiates TRAIL-mediated apop-
totic signaling by unsequestering Bim and Bak in human pancreatic cancer cells. Cancer
Res 68(8):2944–2951 Epub 2008/04/17
66. Okumura K, Huang S, Sinicrope FA (2008) Induction of Noxa sensitizes human colorectal
cancer cells expressing Mcl-1 to the small-molecule Bcl-2/Bcl-xL inhibitor, ABT-737. Clin
Cancer Res 14(24):8132–8142 Epub 2008/12/18
67. Raats DA, de Bruijn MT, Steller EJ, Emmink BL, Borel-Rinkes IH, Kranenburg O (2011)
Synergistic killing of colorectal cancer cells by oxaliplatin and ABT-737. Cell Oncol
(Dordr) 34(4):307–313 Epub 2011/04/07
68. Huang S, Sinicrope FA (2010) Celecoxib-induced apoptosis is enhanced by ABT-737 and
by inhibition of autophagy in human colorectal cancer cells. Autophagy 6(2):256–269 Epub
2010/01/28
69. Traini R, Ben-Josef G, Pastrana DV, Moskatel E, Sharma AK, Antignani A et al (2010)
ABT-737 overcomes resistance to immunotoxin-mediated apoptosis and enhances the
delivery of pseudomonas exotoxin-based proteins to the cell cytosol. Mol Cancer Ther
9(7):2007–2015 Epub 2010/07/01
70. Reynoso D, Nolden LK, Yang D, Dumont SN, Conley AP, Dumont AG et al (2011)
Synergistic induction of apoptosis by the Bcl-2 inhibitor ABT-737 and imatinib
mesylate in gastrointestinal stromal tumor cells. Mol Oncol 5(1):93–104 Epub
2010/12/01
71. Chen ZJ, Zhang B, Pan SH, Zhao HM, Zhang Y, Feng WH et al (2011) Bcl-2 inhibitor ABT-
737 enhances the cisplatin-induced apoptosis in breast cancer T47D cells. Zhonghua Zhong
Liu Za Zhi 33(12):891–895 Epub 2012/02/22
72. Seveno C, Loussouarn D, Brechet S, Campone M, Juin P, Barille-Nion S (2012) Gamma-
secretase inhibition promotes cell death, Noxa upregulation, and sensitization to BH3
mimetic ABT-737 in human breast cancer cells. Breast Cancer Res 14(3):R96 Epub
2012/06/19
73. Zheng L, Yang W, Zhang C, Ding WJ, Zhu H, Lin NM et al (2011) GDC-0941 sensitizes
breast cancer to ABT-737 in vitro and in vivo through promoting the degradation of Mcl-1.
Cancer Lett 309(1):27–36 Epub 2011/06/15
250 V. Y. Yazbeck and D. E. Johnson
74. Oakes SR, Vaillant F, Lim E, Lee L, Breslin K, Feleppa F et al (2012) Sensitization of BCL-
2-expressing breast tumors to chemotherapy by the BH3 mimetic ABT-737. Proc Natl Acad
Sci U S A 109(8):2766–2771 Epub 2011/07/20
75. Witham J, Valenti MR, De-Haven-Brandon AK, Vidot S, Eccles SA, Kaye SB et al (2007)
The Bcl-2/Bcl-XL family inhibitor ABT-737 sensitizes ovarian cancer cells to carboplatin.
Clin Cancer Res 13(23):7191–7198 Epub 2007/12/07
76. Premkumar DR, Jane EP, DiDomenico JD, Vukmer NA, Agostino NR, Pollack IF (2012)
ABT-737 synergizes with bortezomib to induce apoptosis, mediated by Bid cleavage, Bax
activation, and mitochondrial dysfunction in an Akt-dependent context in malignant human
glioma cell lines. J Pharmacol Exp Ther 341(3):859–872 Epub 2012/03/07
77. Tagscherer KE, Fassl A, Campos B, Farhadi M, Kraemer A, Bock BC et al (2008)
Apoptosis-based treatment of glioblastomas with ABT-737, a novel small molecule inhibitor
of Bcl-2 family proteins. Oncogene 27(52):6646–6656 Epub 2008/07/30
78. Fang H, Harned TM, Kalous O, Maldonado V, DeClerck YA, Reynolds CP (2011)
Synergistic activity of fenretinide and the Bcl-2 family protein inhibitor ABT-737 against
human neuroblastoma. Clin Cancer Res 17(22):7093–7104 Epub 2011/09/22
79. Zall H, Weber A, Besch R, Zantl N, Hacker G (2010) Chemotherapeutic drugs sensi-
tize human renal cell carcinoma cells to ABT-737 by a mechanism involving the Noxa-
dependent inactivation of Mcl-1 or A1. Mol Cancer 9:164 Epub 2010/06/26
80. Hao JW, Mao XP, Ding DG, Du GH, Liu ZH (2012) The effect of cell killing by ABT-737
synergized with docetaxel in human prostate cancer PC-3 cells. Zhonghua Wai Ke Za Zhi
50(2):161–165 Epub 2012/04/12
81. Song JH, Kraft AS (2012) Pim kinase inhibitors sensitize prostate cancer cells to apoptosis
triggered by Bcl-2 family inhibitor ABT-737. Cancer Res 72(1):294–303 Epub 2011/11/15
82. Reuland SN, Goldstein NB, Partyka KA, Cooper DA, Fujita M, Norris DA et al (2011) The
combination of BH3-mimetic ABT-737 with the alkylating agent temozolomide induces
strong synergistic killing of melanoma cells independent of p53. PLoS ONE 6(8):e24294
Epub 2011/09/08
83. Weber A, Kirejczyk Z, Potthoff S, Ploner C, Hacker G (2009) Endogenous Noxa determines
the strong proapoptotic synergism of the BH3-mimetic ABT-737 with chemotherapeutic
agents in human melanoma cells. Transl Oncol 2(2):73–83 Epub 2009/05/05
84. Risberg K, Fodstad O, Andersson Y (2011) Synergistic anticancer effects of the 9.2.27PE
immunotoxin and ABT-737 in melanoma. PLoS ONE 6(9):e24012 Epub 2011/09/15
85. Keuling AM, Andrew SE, Tron VA (2010) Inhibition of p38 MAPK enhances ABT-
737-induced cell death in melanoma cell lines: novel regulation of PUMA. Pigment Cell
Melanoma Res 23(3):430–440 Epub 2010/03/27
86. Will B, Siddiqi T, Jorda MA, Shimamura T, Luptakova K, Staber PB et al (2010) Apoptosis
induced by JAK2 inhibition is mediated by Bim and enhanced by the BH3 mimetic ABT-
737 in JAK2 mutant human erythroid cells. Blood 115(14):2901–2909 Epub 2010/02/18
87. Lu M, Wang J, Li Y, Berenzon D, Wang X, Mascarenhas J et al (2010) Treatment with
the Bcl-xL inhibitor ABT-737 in combination with interferon alpha specifically tar-
gets JAK2V617F-positive polycythemia vera hematopoietic progenitor cells. Blood
116(20):4284–4287 Epub 2010/07/14
88. Karlberg M, Ekoff M, Huang DC, Mustonen P, Harvima IT, Nilsson G (2010) The BH3-
mimetic ABT-737 induces mast cell apoptosis in vitro and in vivo: potential for therapeu-
tics. J Immunol 185(4):2555–2562 Epub 2010/07/20
89. Bardwell PD, Gu J, McCarthy D, Wallace C, Bryant S, Goess C et al (2009) The Bcl-2
family antagonist ABT-737 significantly inhibits multiple animal models of autoimmunity.
J Immunol 182(12):7482–7489 Epub 2009/06/06
90. Shoemaker AR, Mitten MJ, Adickes J, Ackler S, Refici M, Ferguson D et al (2008) Activity
of the Bcl-2 family inhibitor ABT-263 in a panel of small cell lung cancer xenograft models.
Clin Cancer Res 14(11):3268–3277 Epub 2008/06/04
91. Chen J, Jin S, Abraham V, Huang X, Liu B, Mitten MJ et al (2011) The Bcl-2/Bcl-
X(L)/Bcl-w inhibitor, navitoclax, enhances the activity of chemotherapeutic agents in vitro
and in vivo. Mol Cancer Ther 10(12):2340–2349 Epub 2011/09/15
9 Leading Small Molecule Inhibitors of Anti-Apoptotic Bcl-2 Family Members 251
92. Ackler S, Mitten MJ, Foster K, Oleksijew A, Refici M, Tahir SK et al (2010) The Bcl-2
inhibitor ABT-263 enhances the response of multiple chemotherapeutic regimens
in hematologic tumors in vivo. Cancer Chemother Pharmacol 66(5):869–880 Epub
2010/01/26
93. Sakuma Y, Tsunezumi J, Nakamura Y, Yoshihara M, Matsukuma S, Koizume S et al (2011)
ABT-263, a Bcl-2 inhibitor, enhances the susceptibility of lung adenocarcinoma cells treated
with Src inhibitors to anoikis. Oncol Rep 25(3):661–667 Epub 2011/01/06
94. Fitzgerald AR (2012) Combination treatments with ABT-263 and an immunotoxin produce
synergistic killing of ABT-263-resistant small cell lung cancer cell lines. Int J Cancer. Epub
2012/07/24
95. Wang G, Zhan Y, Wang H, Li W (2012) ABT-263 sensitizes TRAIL-resistant hepatocarci-
noma cells by downregulating the Bcl-2 family of anti-apoptotic protein. Cancer Chemother
Pharmacol 69(3):799–805 Epub 2011/11/01
96. Zhao X, Ogunwobi OO, Liu C (2011) Survivin inhibition is critical for Bcl-2 inhibi-
tor-induced apoptosis in hepatocellular carcinoma cells. PLoS ONE 6(8):e21980 Epub
2011/08/11
97. Yamaguchi R, Janssen E, Perkins G, Ellisman M, Kitada S, Reed JC (2011) Efficient elimi-
nation of cancer cells by deoxyglucose-ABT-263/737 combination therapy. PLoS ONE
6(9):e24102 Epub 2011/09/29
98. Wong M, Tan N, Zha J, Peale FV, Yue P, Fairbrother WJ et al (2012) Navitoclax (ABT-263)
reduces Bcl-x(L)-mediated chemoresistance in ovarian cancer models. Mol Cancer Ther
11(4):1026–1035 Epub 2012/02/04
99. Sasaki N, Kuroda J, Nagoshi H, Yamamoto M, Kobayashi S, Tsutsumi Y et al (2011) Bcl-2
is a better therapeutic target than c-Myc, but attacking both could be a more effective treat-
ment strategy for B-cell lymphoma with concurrent Bcl-2 and c-Myc overexpression. Exp
Hematol 39(8):817–28 e1. Epub 2011/06/07
100. Xargay-Torrent S, Lopez-Guerra M, Saborit-Villarroya I, Rosich L, Campo E, Roue G et
al (2011) Vorinostat-induced apoptosis in mantle cell lymphoma is mediated by acetyla-
tion of proapoptotic BH3-only gene promoters. Clin Cancer Res 17(12):3956–3968 Epub
2011/06/10
101. Ackler S, Xiao Y, Mitten MJ, Foster K, Oleksijew A, Refici M et al (2008) ABT-263 and
rapamycin act cooperatively to kill lymphoma cells in vitro and in vivo. Mol Cancer Ther
7(10):3265–3274 Epub 2008/10/15
102. Ackler S, Mitten MJ, Chen J, Clarin J, Foster K, Jin S et al (2012) Navitoclax (ABT 263)
and bendamustine ± rituximab induce enhanced killing of non-Hodgkin’s lymphoma tumors
in vivo. Br J Pharmacol 167:881–891 Epub 2012/05/26
103. Roberts AW, Seymour JF, Brown JR, Wierda WG, Kipps TJ, Khaw SL et al (2012)
Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: results of
a phase I study of navitoclax in patients with relapsed or refractory disease. J Clin Oncol
30(5):488–496 Epub 2011/12/21
104. Wilson WH, O’Connor OA, Czuczman MS, LaCasce AS, Gerecitano JF, Leonard JP et al
(2010) Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a
phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitu-
mour activity. Lancet Oncol 11(12):1149–1159 Epub 2010/11/26
105. Gandhi L, Camidge DR, Ribeiro de Oliveira M, Bonomi P, Gandara D, Khaira D et al
(2011) Phase I study of Navitoclax (ABT-263), a novel Bcl-2 family inhibitor, in patients
with small-cell lung cancer and other solid tumors. J Clin Oncol 29(7):909–916 Epub
2011/02/02
106. Rudin CM, Hann CL, Garon EB, Ribeiro de Oliveira M, Bonomi PD, Camidge DR et al
(2012) Phase II study of single-agent navitoclax (ABT-263) and biomarker correlates in
patients with relapsed small cell lung cancer. Clin Cancer Res 18(11):3163–3169 Epub
2012/04/13
107. Furstner A (2003) Chemistry and biology of roseophilin and the prodigiosin alka-
loids: a survey of the last 2500 years. Angew Chem Int Ed Engl 42(31):3582–3603 Epub
2003/08/14
252 V. Y. Yazbeck and D. E. Johnson
108. Shore GC, Viallet J (2005) Modulating the bcl-2 family of apoptosis suppressors for poten-
tial therapeutic benefit in cancer. Hematol Am Soc Hematol Educ Program 2005:226–230
Epub 2005/11/24
109. Zhai D, Jin C, Satterthwait AC, Reed JC (2006) Comparison of chemical inhibitors of antia-
poptotic Bcl-2-family proteins. Cell Death Differ 13(8):1419–1421 Epub 2006/04/29
110. Nguyen M, Marcellus RC, Roulston A, Watson M, Serfass L, Murthy Madiraju SR et al
(2007) Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-
1-mediated resistance to apoptosis. Proc Natl Acad Sci U S A 104(49):19512–19517 Epub
2007/11/28
111. Martinez-Paniagua MA, Baritaki S, Huerta-Yepez S, Ortiz-Navarrete VF, Gonzalez-Bonilla
C, Bonavida B et al (2011) Mcl-1 and YY1 inhibition and induction of DR5 by the BH3-
mimetic Obatoclax (GX15-070) contribute in the sensitization of B-NHL cells to TRAIL
apoptosis. Cell Cycle 10(16):2792–2805 Epub 2011/08/09
112. Wei Y, Kadia T, Tong W, Zhang M, Jia Y, Yang H et al (2010) The combination of a histone
deacetylase inhibitor with the BH3-mimetic GX15-070 has synergistic antileukemia activity
by activating both apoptosis and autophagy. Autophagy 6(7):976–978 Epub 2010/08/24
113. Rahmani M, Aust MM, Attkisson E, Williams DC Jr, Ferreira-Gonzalez A, Grant S (2012)
Inhibition of Bcl-2 antiapoptotic members by obatoclax potently enhances sorafenib-
induced apoptosis in human myeloid leukemia cells through a Bim-dependent process.
Blood 119(25):6089–6098 Epub 2012/03/27
114. Heidari N, Hicks MA, Harada H (2010) GX15-070 (obatoclax) overcomes glucocorticoid
resistance in acute lymphoblastic leukemia through induction of apoptosis and autophagy.
Cell Death Dis 1:e76 Epub 2011/03/03
115. Perez-Galan P, Roue G, Lopez-Guerra M, Nguyen M, Villamor N, Montserrat E et al (2008)
BCL-2 phosphorylation modulates sensitivity to the BH3 mimetic GX15-070 (Obatoclax)
and reduces its synergistic interaction with bortezomib in chronic lymphocytic leukemia
cells. Leukemia 22(9):1712–1720 Epub 2008/07/04
116. Brem EA, Thudium K, Khubchandani S, Tsai PC, Olejniczak SH, Bhat S et al (2011)
Distinct cellular and therapeutic effects of obatoclax in rituximab-sensitive and -resistant
lymphomas. Br J Haematol 153(5):599–611 Epub 2011/04/16
117. Jona A, Khaskhely N, Buglio D, Shafer JA, Derenzini E, Bollard CM et al (2011) The his-
tone deacetylase inhibitor entinostat (SNDX-275) induces apoptosis in Hodgkin lymphoma
cells and synergizes with Bcl-2 family inhibitors. Exp Hematol 39(10):1007–1017 e1. Epub
2011/07/20
118. Perez-Galan P, Roue G, Villamor N, Campo E, Colomer D (2007) The BH3-mimetic GX15-
070 synergizes with bortezomib in mantle cell lymphoma by enhancing Noxa-mediated
activation of Bak. Blood 109(10):4441–4449 Epub 2007/01/18
119. Chen S, Dai Y, Pei XY, Myers J, Wang L, Kramer LB et al (2012) CDK inhibitors up-regu-
late BH3-only proteins to sensitize human myeloma cells to BH3 mimetic therapies. Cancer
Res 72(16):4225–4237 Epub 2012/06/14
120. Jiang CC, Wroblewski D, Yang F, Hersey P, Zhang XD (2009) Human melanoma cells
under endoplasmic reticulum stress are more susceptible to apoptosis induced by the BH3
mimetic obatoclax. Neoplasia 11(9):945–955 Epub 2009/09/03
121. Witters LM, Witkoski A, Planas-Silva MD, Berger M, Viallet J, Lipton A (2007) Synergistic
inhibition of breast cancer cell lines with a dual inhibitor of EGFR-HER-2/neu and a Bcl-2
inhibitor. Oncol Rep 17(2):465–469 Epub 2007/01/05
122. Cruickshanks N, Hamed HA, Bareford MD, Poklepovic A, Fisher PB, Grant S et al (2012)
Lapatinib and obatoclax kill tumor cells through blockade of ERBB1/3/4 and through inhi-
bition of BCL-XL and MCL-1. Mol Pharmacol 81(5):748–758 Epub 2012/02/24
123. Pan J, Cheng C, Verstovsek S, Chen Q, Jin Y, Cao Q (2010) The BH3-mimetic GX15-070
induces autophagy, potentiates the cytotoxicity of carboplatin and 5-fluorouracil in esopha-
geal carcinoma cells. Cancer Lett 293(2):167–174 Epub 2010/02/16
124. Huang S, Okumura K, Sinicrope FA (2009) BH3 mimetic obatoclax enhances TRAIL-mediated
apoptosis in human pancreatic cancer cells. Clin Cancer Res 15(1):150–159 Epub 2009/01/02
9 Leading Small Molecule Inhibitors of Anti-Apoptotic Bcl-2 Family Members 253
125. Smoot RL, Blechacz BR, Werneburg NW, Bronk SF, Sinicrope FA, Sirica AE et al (2010)
A Bax-mediated mechanism for obatoclax-induced apoptosis of cholangiocarcinoma cells.
Cancer Res 70(5):1960–1969 Epub 2010/02/18
126. Schimmer AD, O’Brien S, Kantarjian H, Brandwein J, Cheson BD, Minden MD et al
(2008) A phase I study of the pan bcl-2 family inhibitor obatoclax mesylate in patients with
advanced hematologic malignancies. Clin Cancer Res 14(24):8295–8301 Epub 2008/12/18
127. O’Brien SM, Claxton DF, Crump M, Faderl S, Kipps T, Keating MJ et al (2009) Phase I
study of obatoclax mesylate (GX15-070), a small molecule pan-Bcl-2 family antagonist,
in patients with advanced chronic lymphocytic leukemia. Blood 113(2):299–305 Epub
2008/10/22
128. Hwang JJ, Kuruvilla J, Mendelson D, Pishvaian MJ, Deeken JF, Siu LL et al (2010) Phase I
dose finding studies of obatoclax (GX15-070), a small molecule pan-BCL-2 family antago-
nist, in patients with advanced solid tumors or lymphoma. Clin Cancer Res 16(15):4038–
4045 Epub 2010/06/12
129. Oki Y, Copeland A, Hagemeister F, Fayad LE, Fanale M, Romaguera J et al (2012)
Experience with obatoclax mesylate (GX15-070), a small molecule pan-Bcl-2 fam-
ily antagonist in patients with relapsed or refractory classical Hodgkin lymphoma. Blood
119(9):2171–2172 Epub 2012/03/03
130. Parikh SA, Kantarjian H, Schimmer A, Walsh W, Asatiani E, El-Shami K et al (2010)
Phase II study of obatoclax mesylate (GX15-070), a small-molecule BCL-2 family antago-
nist, for patients with myelofibrosis. Clin Lymphoma Myeloma Leuk 10(4):285–289 Epub
2010/08/17
131. Paik PK, Rudin CM, Brown A, Rizvi NA, Takebe N, Travis W et al (2010) A phase I study
of obatoclax mesylate, a Bcl-2 antagonist, plus topotecan in solid tumor malignancies.
Cancer Chemother Pharmacol 66(6):1079–1085 Epub 2010/02/19
132. Paik PK, Rudin CM, Pietanza MC, Brown A, Rizvi NA, Takebe N et al (2011) A phase II
study of obatoclax mesylate, a Bcl-2 antagonist, plus topotecan in relapsed small cell lung
cancer. Lung Cancer 74(3):481–485 Epub 2011/05/31
133. Chiappori AA, Schreeder MT, Moezi MM, Stephenson JJ, Blakely J, Salgia R et al (2012)
A phase I trial of pan-Bcl-2 antagonist obatoclax administered as a 3-h or a 24-h infusion in
combination with carboplatin and etoposide in patients with extensive-stage small cell lung
cancer. Br J Cancer 106(5):839–845 Epub 2012/02/16
Chapter 10
SMAC IAP Addiction in Cancer
10.1 Introduction
M. F. Brown · K. He · J. Yu (*)
Department of Pathology, University of Pittsburgh School of Medicine,
University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
e-mail: [email protected]
M. F. Brown · J. Yu
Department of Pathology and Molecular and Cellular Pathology Graduate Training Program,
University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute,
Pittsburgh, PA, USA
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 255
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_10,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
256 M. F. Brown et al.
Modern virology has produced some of the most valuable insights into the mecha-
nisms of cell death. Many viruses encode gene products that suppress cell death
by disabling tumor suppressor proteins and producing viral versions of anti-apop-
totic Bcl-2 proteins and/or caspase inhibitors. This is critical for viral replication
and carcinogenesis in certain patients [8–10]. The baculovirus protein, IAP, was
the first inhibitor of apoptosis protein (IAP) discovered and shown to promote
viral replication [11, 12]. Through sequence homology-based screens and other
methods, eight IAP family members have been discovered in human, including
NAIP, XIAP, cIAP1, cIAP2, Survivin, BRUCE (Apollon), ILP2, and ML-IAP.
Seven of them have homologs in mice (Fig. 10.1 and Table 10.1). All IAP fam-
ily members in viruses and metazoans contain at least one signature baculovirus
IAP repeat (BIR) domain (Fig. 10.1 and Table 10.1), which is capable of interact-
ing with caspases and various IAP antagonists. Several of these, including XIAP,
cIAP1, cIAP2, survivin, and ILP2, directly bind to and suppress caspases through
their BIR domains [13]. XIAP, cIAP1, and cIAP2 also contain a RING domain
(Fig. 10.1), a signature of E3 ligases [13], and play critical regulatory roles in
NF-κB signaling and necrosis. Certain IAP family members, including XIAP and
10 SMAC IAP Addiction in Cancer 257
Fig. 10.1 Human IAP family members. Eight human IAP proteins have been identified and
all contain at least one baculoviral IAP repeat (BIR) domain. Several IAPs contain a carboxy-
terminal RING domain with E3 ubiquitin ligase activities, as well as ubiquitin-binding domains
(UBA/C). Other domains such as leucine-rich repeats (LRR), nucleotide oligomerization domain
(NOD) and caspase activation and recruitment domain (CARD) are present in some IAPs, which
mediate protein–protein interactions
10.3 Apoptotic Pathways
Fig. 10.2 IAP-mediated cell survival and signaling. IAP proteins serve as a signaling hub for
cell survival. IAPs are negative regulators of programmed apoptosis and necrosis. XIAP and
cIAPs can inhibit both the initiator and effector caspases to block apoptosis. In addition, cIAPs
positively regulate canonical NF-κB signaling to allow TNFα production and inflammation,
while negatively regulating non-canonical NF-κB signaling, necrosis and caspase-8 activation,
all through complex assembly. SMAC or SMAC mimetics suppress XIAP and cIAP-dependent
cell survival by relieving IAP caspase inhibition through competitive binding, as well as by
depletion of cIAPs via activation of their E3 ligase activity (see text for details). (P)-phosphoryl-
ated and (Ub)-ubiquitinated
IAPs can inhibit caspases through two major mechanisms. Some IAPs bind
directly to active sites of caspases to prevent processing or activity. Among all
IAPs, mammalian XIAP is the only IAP that has high-affinity interactions with
caspase-3, caspase-7, and caspase-9, and functions as a direct caspase inhibi-
tor [28]. XIAP inhibits caspases-3, caspase-7, and caspase-9 via its BIR2 and
BIR3 domains [29–33]. Specifically, insertion of the linker region between
BIR1 and BIR2 into the catalytic pocket of active effector caspases (such as
caspase-3 or caspase-7) prevents substrate entry. In addition, XIAP can prevent
initiator caspases such as caspase-9 from dimerization and subsequent activa-
tion, by hindering a conformational change required for a functional catalytic
pocket [34, 35].
IAPs can also induce ubiquitin-mediated degradation of caspases. The RING
finger domain in DIAP1 regulates Ub conjugation of caspases, and therefore,
apoptosis in Drosophila, though the precise role of mono- versus poly-ubiquityla-
tion in this process, is not clear [36–39]. Consistent with these findings, cIAP1 and
cIAP2, or their homologs in lower eukaryotes do not inhibit caspases efficiently in
vitro [40]. Conversely, in living cells, XIAP promotes the degradation of active-
form caspase-3 [30], but not pro-caspase-3 [38]. Interestingly, the RING domain
in XIAP is not required for its inhibition of caspase-3, caspase-7, or caspase-9 in
vitro or when overexpressed [41, 42]. These findings suggest multiple mechanisms
are involved in IAP-mediated caspase inhibition.
cIAP1 and cIAP2 were initially identified as interacting proteins of the scaf-
fold protein TRAF2 (tumor necrosis factor receptor-associated factor 2). They
are now known to play crucial roles in regulating NF-кB signaling, DNA dam-
age response, and necrosis by regulating the assembly and stability of distinct
signaling complexes through their E3 ligase function [4, 43]. This knowledge
was largely obtained by using small molecules that deplete both cIAPs. In the
canonical NF-кB pathway, cIAP1 and cIAP2 regulate Ub-dependent activation
of NF-кB downstream of TNFR1, which in turn drives transcriptional programs
important for cell survival and inflammation [4, 43]. In addition, cIAPs sup-
press non-canonical NF-кB signaling, necrosis, and the assembly of caspase-8
activation complexes in resting cells. Rapid degradation of cIAP1 and cIAP2
triggered by SMAC mimetics, or DNA damage can lead to activation of the
non-canonical NF-кB pathway, apoptosis, or necrosis if caspase activation is
blocked [44–49].
10 SMAC IAP Addiction in Cancer 261
Several cellular proteins can antagonize the anti-apoptotic activities of IAPs. The
best studied endogenous IAP inhibitor is second mitochondria-derived activator
of caspase (SMAC), also known as direct inhibitor of apoptosis protein (IAP)-
binding protein with low pI (Diablo) or SMAC/Diablo. Upon apoptotic induction,
SMAC is released into the cytosol and binds to BIRs in IAPs through its amino
(N)-terminus to facilitate caspase activation [22, 73]. The Drosophila death pro-
teins Hid, Grim, and Reaper have limited homology with SMAC, in the BIR bind-
ing motif [22, 73, 74] (Fig. 10.3a), suggesting that regulation of caspase activation
is an ancient function of these proteins [41, 75].
SMAC/Diablo functions as a general IAP inhibitor and binds to XIAP, cIAP1,
cIAP2, survivin, livin, and BRUCE, but not NAIP [22, 73, 76–79]. The four
amino-terminal residues of mature SMAC/DIABLO, Ala-Val-Pro-Ile (AVPI) are
both necessary and sufficient for SMAC/DIABLO–IAP interaction [3, 80–83].
10 SMAC IAP Addiction in Cancer 263
Fig. 10.3 IAP-binding motif (IBM) and SMAC mimetics. a The conserved BIR binding motif.
This tetrapeptide motif has the consensus sequence of A-(V/T/I)-(P/A)-(F/Y/I/V) and is found in
SMAC, the drosophila proteins Reaper, Grim, and Hid [41]. b The chemical structure of SMAC
AVPI [151]. c Structures of several small-molecule (IAP) antagonists: monovalent SMAC mimet-
ics GDC-0152 (Genentech) [200] and AT-406 (Aegera Therapeutics) [201], and the bivalent
SMAC mimetic TL32711 [4] and natural product Embelin [151]
This tetrapeptide motif is exposed following SMAC release from the mitochon-
dria after proteolytical removal of a 55-residue N terminal mitochondria-targeting
sequence. Crystallography data revealed that SMAC/DIABLO homodimerizes
through an extensive hydrophobic interface, which is essential for its high-affin-
ity binding to IAPs and its pro-apoptotic functions [81]. Several SMAC isoforms
have been reported to regulate IAPs through ubiquitination-dependent or caspase-
dependent mechanisms to potentiate apoptosis in vitro [3].
Additional pro-apoptotic IAP-binding proteins have been identified and are
capable of binding to several IAP members, including serine protease HtrA2
(high-temperature-requirement protein A2) also known as Omi [23, 84–86], and
XIAP-associated factor 1 (XAF1) [87, 88]. However, a SMAC AVPI-like element
is only found in HtrA2/Omi [86], and the role of these proteins as selective IAP
antagonists in the regulation of apoptosis is much less understood compared with
SMAC.
264 M. F. Brown et al.
The functions of IAPs, including XIAP, cIAPs, and their inhibitors have been
examined extensively in human cells, mice, and other model organisms using both
loss-of-function and overexpression systems (reviewed in [7, 60, 89]). In general,
the findings support that IAP overexpression inhibits caspase activation and apop-
tosis mediated by both intrinsic and extrinsic apoptotic pathways. Overexpression
of SMAC, HtrA2/Omi, or XAF1 increases apoptosis in a variety of cancer cell
lines when combined with other anticancer agents, particularity with death recep-
tor ligands such as TRAIL (TNF-related apoptosis-inducing ligand).
Knockout of IAP in mice or human cancer cell lines results in a range of phe-
notypes associated with altered apoptosis or cell survival, and also supports func-
tion of IAPs in regulating NF-κB activation and cell proliferation [7]. IAPs appear
to regulate apoptosis with a great deal of redundancy. Total knockout of XIAP in
mice resulted in highly elevated apoptosis in response to brain injury, increased
NF-κB activation, and an overall reduced survival [90, 91]. XIAP knockout
HCT116 colon cancer cells are sensitized to TRAIL-induced apoptosis [92]. Mice
with total knockout of cIAP1 have no discernable phenotype due to redundant
pathway activation by cIAP2 [13]. Knockout of cIAP2 in mice leads to increased
macrophage death and resistance to LPS-induced sepsis [93]. Double knockout of
cIAP1/cIAP2 or XIAP/cIAP1 is embryonic lethal due to apoptotic defects, while
XIAP/cIAP2 double-knockout mice showed little change in apoptosis due to com-
pensation by cIAP1 [94]. Conditional cIAP1/2 double-knockout mice show uncon-
trolled B-cell proliferation independent of growth factor [95].
Endogenous IAP inhibitors also exhibit redundant functions in apoptosis regula-
tion [3, 7]. SMAC deficiency results in apoptosis resistance to a selective group of
agents, including TRAIL and non-steroidal anti-inflammatory drugs (NSAID) in
HCT116 colon cancer cells and mice, yet has little to no impact on apoptosis induced
by most DNA-damaging agents [96–99]. HtrA2/Omi knockout mice or cells, or
SMAC/HtrA2 double-knockout mice have minimal changes in apoptosis [100].
Extensive studies have examined genetic and expression alterations in IAPs and
their inhibitors in cancer, and their correlation with clinical outcomes (reviewed
in [3, 4, 43, 101]). Genetic alterations in cIAP1/2 have been reported in multiple
cancers. Overexpression of IAP members, including XIAP, survivin, and cIAP1/2
and reduced expression of SMAC, HtrA2/Omi, or XFA1 are common in cancer
and suggested to correlate with chemoresistance, disease progression, and poor
prognosis. Although these results support targeting IAPs in cancer, they should be
10 SMAC IAP Addiction in Cancer 265
interpreted with caution for several reasons. For example, some studies reported
nuclear but not cytosolic expression as having prognostic value. In certain cases,
either a lack of correlation or a reverse correlation is reported in a different cohort
of the same tumor type. Notably, the sample size was limited in several studies.
Lastly, genetic evidence supports that loss of cIAPs can contribute to tumor initia-
tion or progression in some cases.
While IAP overexpression can result from genetic changes such as gene ampli-
fication or chromosomal aberrations, the reason for their overexpression is not
known in most cases. To date, genetic alterations have been reported for cIAPs,
but not survivin, XIAP or ML-IAP. Amplification and translocation of cIAPs
have been found in both solid tumors and hematological malignancies. The
11q21–q23 amplification, encompassing both cIAP1 and cIAP2 loci, has been
reported in esophageal cancer [102], cervical cancer [103], liver cancer [104],
lung cancer [105], pancreatic cancer [106], medulloblastoma [107], and glioblas-
toma [108]. Interestingly, recurrent amplification of cIAP1 and cIAP2 is found
in Myc-driven liver cancer [104] as well as spontaneous osteosarcomas in mice
[109]. Additionally, the cIAP2–MALT1 fusion protein resulting from t(11;18)
(q21;q21) translocation is found in mucosa-associated lymphoid tissue lymphoma
[110–114].
In rare cases, cIAPs can function as tumor suppressors. For example, 20 %
of patients with multiple myeloma have activated non-canonical NF-κB sign-
aling associated with frequent genetic alterations, including biallelic deletions
of cIAP1, cIAP2, or TRAF2, TRAF3, CYLD, as well as enhanced expression of
CD40, lymphotoxin-β receptor (LTβR), TNFRSF13B, NFKB2, and NIK [5, 115].
Mutational changes leading to increased NIK and non-canonical activation of NF-
κB have also been reported in breast [116] and pancreatic cancers [117].
Elevated IAP expression has been reported to be a predictor of poor progno-
sis in many cancers. For example, elevated XIAP levels are correlated with dis-
ease progression, metastasis, and poor survival in colon cancer [118], liver cancer
[119], gastric cancer [120], breast cancer [121], and melanoma [122]. High lev-
els of cIAP1 are associated with poor overall survival and local recurrence-free
survival in cervical squamous cell carcinomas [103], and nodal metastasis in
squamous cell carcinoma of the tongue [123]. Furthermore, high levels of cIAP2
expression are correlated with reduced survival in colorectal cancer [124] and pan-
creatic ductal adenocarcinomas [125]. Increased ML-IAP expression in mRNA or
protein is found in melanoma and renal cancer [126–129] and is associated with
disease progression or poor prognosis in superficial bladder cancer [130], adult
ALL [131], osteosarcoma [132], and a subset of N-Myc-amplified neuroblastoma
[133]. Lastly, elevated survivin expression is prevalent in cancer and associated
with poor prognosis [101, 134–136].
266 M. F. Brown et al.
10.9.1 SMAC Mimetics
Detailed structural information on the binding between SMAC and IAPs prompted
the development of peptidomimetics and small-molecule SMAC mimetics, pro-
viding an excellent example of rational drug design [151]. SMAC AVPI-derived
peptides were first tested and found to effectively block IAP–caspase interactions
10 SMAC IAP Addiction in Cancer 267
Over 50 patents have been filed on IAP antagonists as potential anticancer agents,
and some of these agents have entered clinical development [4, 43]. Currently, a
large number of clinical trials are underway to assess their safety, pharmacologi-
cal properties and efficacy in patients with advanced solid tumor or hematological
malignancies (Table 10.2).
[4]. Therefore, additional studies are required to determine which classes of agents
have more desirable pharmacodynamic or pharmacokinetic properties for clinical
application.
Two major strategies have been used to target XIAP by preventing it from binding
to caspase-3 [182] or downregulation with antisense. A number of small-molecule
XIAP antagonists have shown efficacy in vitro and are in preclinical development,
including dTWX-024 [183], TPI-1396-34 [184], XAC 1396-11 [185], and the natu-
ral product embelin [186]. The XIAP antisense oligonucleotides AEG35156 (Aegera
270 M. F. Brown et al.
Therapeutics) displays potent antitumor activities in vitro as a single agent and syn-
ergizes with various chemotherapeutic compounds, such as the death receptor ligand,
TRAIL, and radiation to induce apoptosis in cell lines and mouse xenograft models
[187–190]. Results from Phase I and II clinical trials indicated that AEG35156 is
well tolerated and exhibits predictable pharmacokinetic properties, dose-dependent
changes in circulating biomarkers of cell death, as well as some antitumor activities
(Table 10.2) [191, 192], consistent with in vitro data on the suppression of XIAP
mRNA and protein levels [187–190]. Future studies will be needed to determine
whether modalities containing this agent improve efficacy over standard therapies.
10.10 Conclusions
Acknowledgments We are grateful to Dr. Lin Zhang for critical reading and comments, and
Mrs. Laurice Vance-Carr for excellent secretarial assistance. The work in authors’ laboratory is
supported in part by NIH grants CA129829, UO1-DK085570, American Cancer Society grant
RGS-10-124-01-CCE, and Flight Attendant Medical Research Institute (FAMRI).
References
24. Joza N et al (2001) Essential role of the mitochondrial apoptosis-inducing factor in pro-
grammed cell death. Nature 410(6828):549–554
25. Wang X et al (2002) Mechanisms of AIF-mediated apoptotic DNA degradation in
Caenorhabditis elegans. Science 298(5598):1587–1592
26. Luo X et al (1998) Bid, a Bcl2 interacting protein, mediates cytochrome c release from
mitochondria in response to activation of cell surface death receptors. Cell 94(4):481–490
27. Li H et al (1998) Cleavage of BID by caspase 8 mediates the mitochondrial damage in the
Fas pathway of apoptosis. Cell 94(4):491–501
28. Eckelman BP, Salvesen GS, Scott FL (2006) Human inhibitor of apoptosis proteins: why
XIAP is the black sheep of the family. EMBO Rep 7(10):988–994
29. Sun C et al (1999) NMR structure and mutagenesis of the inhibitor-of-apoptosis protein
XIAP. Nature 401(6755):818–822
30. Silke J et al (2001) Direct inhibition of caspase 3 is dispensable for the anti-apoptotic activ-
ity of XIAP. EMBO J 20(12):3114–3123
31. Huang Y et al (2001) Structural basis of caspase inhibition by XIAP: differential roles of the
linker versus the BIR domain. Cell 104(5):781–790
32. Chai J et al (2001) Structural basis of caspase-7 inhibition by XIAP. Cell 104(5):769–780
33. Riedl SJ et al (2001) Structural basis for the inhibition of caspase-3 by XIAP. Cell
104(5):791–800
34. Srinivasula SM et al (2001) A conserved XIAP-interaction motif in caspase-9 and Smac/
DIABLO regulates caspase activity and apoptosis. Nature 410(6824):112–116
35. Shiozaki EN et al (2003) Mechanism of XIAP-mediated inhibition of caspase-9. Mol Cell
11(2):519–527
36. Wang SL et al (1999) The Drosophila caspase inhibitor DIAP1 is essential for cell survival
and is negatively regulated by HID. Cell 98(4):453–463
37. Ditzel M et al (2008) Inactivation of effector caspases through nondegradative polyubiquity-
lation. Mol Cell 32(4):540–553
38. Suzuki Y, Nakabayashi Y, Takahashi R (2001) Ubiquitin-protein ligase activity of
X-linked inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3
and enhances its anti-apoptotic effect in Fas-induced cell death. Proc Natl Acad Sci USA
98(15):8662–8667
39. Choi YE et al (2009) The E3 ubiquitin ligase cIAP1 binds and ubiquitinates cas-
pase-3 and -7 via unique mechanisms at distinct steps in their processing. J Biol Chem
284(19):12772–12782
40. Riedl SJ, Shi Y (2004) Molecular mechanisms of caspase regulation during apoptosis. Nat
Rev Mol Cell Biol 5(11):897–907
41. Shi Y (2002) Mechanisms of caspase activation and inhibition during apoptosis. Mol Cell
9(3):459–470
42. Deveraux QL et al (1999) Cleavage of human inhibitor of apoptosis protein XIAP results in
fragments with distinct specificities for caspases. EMBO J 18(19):5242–5251
43. Gyrd-Hansen M, Meier P (2010) IAPs: from caspase inhibitors to modulators of NF-
kappaB, inflammation and cancer. Nat Rev Cancer 10(8):561–574
44. Varfolomeev E et al (2007) IAP antagonists induce autoubiquitination of c-IAPs, NF-
kappaB activation, and TNFalpha-dependent apoptosis. Cell 131(4):669–681
45. Gaither A et al (2007) A Smac mimetic rescue screen reveals roles for inhibitor of apoptosis
proteins in tumor necrosis factor-alpha signaling. Cancer Res 67(24):11493–11498
46. Wang L, Du F, Wang X (2008) TNF-alpha induces two distinct caspase-8 activation path-
ways. Cell 133(4):693–703
47. Zhang DW et al (2009) RIP3, an energy metabolism regulator that switches TNF-induced
cell death from apoptosis to necrosis. Science 325(5938):332–336
48. He S et al (2009) Receptor interacting protein kinase-3 determines cellular necrotic
response to TNF-alpha. Cell 137(6):1100–1111
49. Feoktistova M et al (2012) Pick your poison: the ripoptosome, a cell death platform regulat-
ing apoptosis and necroptosis. Cell Cycle 11(3):460–467
10 SMAC IAP Addiction in Cancer 273
50. Karin M, Greten FR (2005) NF-kappaB: linking inflammation and immunity to cancer
development and progression. Nat Rev Immunol 5(10):749–759
51. Nathan C, Ding A (2010) Nonresolving inflammation. Cell 140(6):871–882
52. Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and cancer. Cell
140(6):883–899
53. Perkins ND (2007) Integrating cell-signalling pathways with NF-kappaB and IKK function.
Nat Rev Mol Cell Biol 8(1):49–62
54. Bonizzi G, Karin M (2004) The two NF-kappaB activation pathways and their role in innate
and adaptive immunity. Trends Immunol 25(6):280–288
55. Bhoj VG, Chen ZJ (2009) Ubiquitylation in innate and adaptive immunity. Nature
458(7237):430–437
56. Micheau O, Tschopp J (2003) Induction of TNF receptor I-mediated apoptosis via two
sequential signaling complexes. Cell 114(2):181–190
57. Leulier F et al (2006) The Drosophila inhibitor of apoptosis protein DIAP2 functions in
innate immunity and is essential to resist gram-negative bacterial infection. Mol Cell Biol
26(21):7821–7831
58. Gesellchen V et al (2005) An RNA interference screen identifies Inhibitor of apoptosis pro-
tein 2 as a regulator of innate immune signalling in Drosophila. EMBO Rep 6(10):979–984
59. Kleino A et al (2005) Inhibitor of apoptosis 2 and TAK1-binding protein are components of
the Drosophila Imd pathway. EMBO J 24(19):3423–3434
60. Orme M, Meier P (2009) Inhibitor of apoptosis proteins in Drosophila: gatekeepers of
death. Apoptosis 14(8):950–960
61. Winkles JA (2008) The TWEAK-Fn14 cytokine-receptor axis: discovery, biology and thera-
peutic targeting. Nat Rev Drug Discov 7(5):411–425
62. Vallabhapurapu S et al (2008) Nonredundant and complementary functions of TRAF2 and
TRAF3 in a ubiquitination cascade that activates NIK-dependent alternative NF-kappaB
signaling. Nat Immunol 9(12):1364–1370
63. Zarnegar BJ et al (2008) Noncanonical NF-kappaB activation requires coordinated assem-
bly of a regulatory complex of the adaptors cIAP1, cIAP2, TRAF2 and TRAF3 and the
kinase NIK. Nat Immunol 9(12):1371–1378
64. Galluzzi L et al (2011) Programmed necrosis from molecules to health and disease. Int Rev
Cell Mol Biol 289:1–35
65. Petersen SL et al (2007) Autocrine TNFalpha signaling renders human cancer cells suscepti-
ble to Smac-mimetic-induced apoptosis. Cancer Cell 12(5):445–456
66. Vercammen D et al (1998) Inhibition of caspases increases the sensitivity of L929 cells to
necrosis mediated by tumor necrosis factor. J Exp Med 187(9):1477–1485
67. Vanden Berghe T et al (2003) Disruption of HSP90 function reverts tumor necrosis factor-
induced necrosis to apoptosis. J Biol Chem 278(8):5622–5629
68. Zheng L et al (2006) Competitive control of independent programs of tumor necrosis factor
receptor-induced cell death by TRADD and RIP1. Mol Cell Biol 26(9):3505–3513
69. Geserick P et al (2009) Cellular IAPs inhibit a cryptic CD95-induced cell death by limiting
RIP1 kinase recruitment. J Cell Biol 187(7):1037–1054
70. Jackson SP, Bartek J (2009) The DNA-damage response in human biology and disease.
Nature 461(7267):1071–1078
71. Tenev T et al (2011) The Ripoptosome, a signaling platform that assembles in response to
genotoxic stress and loss of IAPs. Mol Cell 43(3):432–448
72. Biton S, Ashkenazi A (2011) NEMO and RIP1 control cell fate in response to extensive
DNA damage via TNF-alpha feedforward signaling. Cell 145(1):92–103
73. Verhagen AM et al (2000) Identification of DIABLO, a mammalian protein that promotes
apoptosis by binding to and antagonizing IAP proteins. Cell 102(1):43–53
74. Steller H (2008) Regulation of apoptosis in Drosophila. Cell Death Differ 15(7):1132–1138
75. Shi Y (2004) Caspase activation: revisiting the induced proximity model. Cell 117(7):855–858
76. Vucic D et al (2002) SMAC negatively regulates the anti-apoptotic activity of melanoma
inhibitor of apoptosis (ML-IAP). J Biol Chem 277(14):12275–12279
274 M. F. Brown et al.
77. Davoodi J et al (2004) Neuronal apoptosis-inhibitory protein does not interact with Smac
and requires ATP to bind caspase-9. J Biol Chem 279(39):40622–40628
78. Hao Y et al (2004) Apollon ubiquitinates SMAC and caspase-9, and has an essential cyto-
protection function. Nat Cell Biol 6(9):849–860
79. Qiu XB, Goldberg AL (2005) The membrane-associated inhibitor of apoptosis protein,
BRUCE/Apollon, antagonizes both the precursor and mature forms of Smac and caspase-9.
J Biol Chem 280(1):174–182
80. Shiozaki EN, Shi Y (2004) Caspases, IAPs and Smac/DIABLO: mechanisms from struc-
tural biology. Trends Biochem Sci 29(9):486–494
81. Chai J et al (2000) Structural and biochemical basis of apoptotic activation by Smac/
DIABLO. Nature 406(6798):855–862
82. Liu Z et al (2000) Structural basis for binding of Smac/DIABLO to the XIAP BIR3 domain.
Nature 408(6815):1004–1008
83. Wu G et al (2000) Structural basis of IAP recognition by Smac/DIABLO. Nature
408(6815):1008–1012
84. Hegde R et al (2002) Identification of Omi/HtrA2 as a mitochondrial apoptotic ser-
ine protease that disrupts inhibitor of apoptosis protein-caspase interaction. J Biol Chem
277(1):432–438
85. Verhagen AM et al (2002) HtrA2 promotes cell death through its serine protease activity
and its ability to antagonize inhibitor of apoptosis proteins. J Biol Chem 277(1):445–454
86. Martins LM et al (2002) The serine protease Omi/HtrA2 regulates apoptosis by binding
XIAP through a reaper-like motif. J Biol Chem 277(1):439–444
87. Liston P et al (2001) Identification of XAF1 as an antagonist of XIAP anti-Caspase activity.
Nat Cell Biol 3(2):128–133
88. Arora V et al (2007) Degradation of survivin by the X-linked inhibitor of apoptosis (XIAP)-
XAF1 complex. J Biol Chem 282(36):26202–26209
89. Kashkar H (2010) X-linked inhibitor of apoptosis: a chemoresistance factor or a hollow
promise. Clin Cancer Res 16(18):4496–4502
90. West T et al (2009) Lack of X-linked inhibitor of apoptosis protein leads to increased apop-
tosis and tissue loss following neonatal brain injury. ASN Neuro 1(1):e00004
91. Bauler LD, Duckett CS, O’Riordan MX (2008) XIAP regulates cytosol-specific innate
immunity to Listeria infection. PLoS Pathog 4(8):e1000142
92. Cummins JM et al (2004) X-linked inhibitor of apoptosis protein (XIAP) is a nonredundant
modulator of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated
apoptosis in human cancer cells. Cancer Res 64(9):3006–3008
93. Conte D et al (2006) Inhibitor of apoptosis protein cIAP2 is essential for lipopolysaccha-
ride-induced macrophage survival. Mol Cell Biol 26(2):699–708
94. Moulin M et al (2012) IAPs limit activation of RIP kinases by TNF receptor 1 during devel-
opment. EMBO J 31(7):1679–1691. doi: http://10.1038/ebomj.2012.18
95. Gardam S et al (2011) Deletion of cIAP1 and cIAP2 in murine B lymphocytes constitu-
tively activates cell survival pathways and inactivates the germinal center response. Blood
117(15):4041–4051
96. Bank A et al (2008) SMAC mimetics sensitize nonsteroidal anti-inflammatory drug-induced apop-
tosis by promoting caspase-3-mediated cytochrome c release. Cancer Res 68(1):276–284
97. Kohli M et al (2004) SMAC/Diablo-dependent apoptosis induced by nonsteroidal antiinflammatory
drugs (NSAIDs) in colon cancer cells. Proc Natl Acad Sci USA 101(48):16897–16902
98. Okada H et al (2002) Generation and characterization of Smac/DIABLO-deficient mice.
Mol Cell Biol 22(10):3509–3517
99. Qiu W et al (2010) Chemoprevention by nonsteroidal anti-inflammatory drugs eliminates
oncogenic intestinal stem cells via SMAC-dependent apoptosis. Proc Natl Acad Sci USA
107(46):20027–20032
100. Martins LM et al (2004) Neuroprotective role of the reaper-related serine protease
HtrA2/Omi revealed by targeted deletion in mice. Mol Cell Biol 24(22):9848–9862
10 SMAC IAP Addiction in Cancer 275
101. Altieri DC (2008) Survivin, cancer networks and pathway-directed drug discovery. Nat Rev
Cancer 8(1):61–70
102. Imoto I et al (2001) Identification of cIAP1 as a candidate target gene within an amplicon at
11q22 in esophageal squamous cell carcinomas. Cancer Res 61(18):6629–6634
103. Imoto I et al (2002) Expression of cIAP1, a target for 11q22 amplification, correlates with
resistance of cervical cancers to radiotherapy. Cancer Res 62(17):4860–4866
104. Zender L et al (2006) Identification and validation of oncogenes in liver cancer using an
integrative oncogenomic approach. Cell 125(7):1253–1267
105. Dai Z et al (2003) A comprehensive search for DNA amplification in lung cancer identi-
fies inhibitors of apoptosis cIAP1 and cIAP2 as candidate oncogenes. Hum Mol Genet
12(7):791–801
106. Bashyam MD et al (2005) Array-based comparative genomic hybridization identifies
localized DNA amplifications and homozygous deletions in pancreatic cancer. Neoplasia
7(6):556–562
107. Reardon DA et al (1997) Extensive genomic abnormalities in childhood medulloblastoma
by comparative genomic hybridization. Cancer Res 57(18):4042–4047
108. Weber RG et al (1996) Clinically distinct subgroups of glioblastoma multiforme studied by
comparative genomic hybridization. Lab Invest 74(1):108–119
109. Ma O et al (2009) MMP13, Birc2 (cIAP1), and Birc3 (cIAP2), amplified on chromo-
some 9, collaborate with p53 deficiency in mouse osteosarcoma progression. Cancer Res
69(6):2559–2567
110. Dierlamm J et al (1999) The apoptosis inhibitor gene API2 and a novel 18q gene, MLT,
are recurrently rearranged in the t(11;18)(q21;q21) associated with mucosa-associated lym-
phoid tissue lymphomas. Blood 93(11):3601–3609
111. Akagi T et al (1999) A novel gene, MALT1 at 18q21, is involved in t(11;18) (q21;q21)
found in low-grade B-cell lymphoma of mucosa-associated lymphoid tissue. Oncogene
18(42):5785–5794
112. Zhou H, Du MQ, Dixit VM (2005) Constitutive NF-kappaB activation by the t(11;18)
(q21;q21) product in MALT lymphoma is linked to deregulated ubiquitin ligase activity.
Cancer Cell 7(5):425–431
113. Morgan JA et al (1999) Breakpoints of the t(11;18)(q21;q21) in mucosa-associated lym-
phoid tissue (MALT) lymphoma lie within or near the previously undescribed gene MALT1
in chromosome 18. Cancer Res 59(24):6205–6213
114. Varfolomeev E et al (2006) The inhibitor of apoptosis protein fusion c-IAP2.MALT1
stimulates NF-kappaB activation independently of TRAF1 AND TRAF2. J Biol Chem
281(39):29022–29029
115. Annunziata CM et al (2007) Frequent engagement of the classical and alternative NF-kappaB
pathways by diverse genetic abnormalities in multiple myeloma. Cancer Cell 12(2):115–130
116. Yamaguchi N et al (2009) Constitutive activation of nuclear factor-kappaB is preferen-
tially involved in the proliferation of basal-like subtype breast cancer cell lines. Cancer Sci
100(9):1668–1674
117. Wharry CE et al (2009) Constitutive non-canonical NF kappaB signaling in pancreatic can-
cer cells. Cancer Biol Ther 8(16):1567–1576
118. Xiang G et al (2009) Expression of X-linked inhibitor of apoptosis protein in human colo-
rectal cancer and its correlation with prognosis. J Surg Oncol 100(8):708–712
119. Augello C et al (2009) Inhibitors of apoptosis proteins (IAPs) expression and their prognos-
tic significance in hepatocellular carcinoma. BMC Cancer 9:125
120. Shibata T et al (2007) Disturbed expression of the apoptosis regulators XIAP, XAF1, and
Smac/DIABLO in gastric adenocarcinomas. Diagn Mol Pathol 16(1):1–8
121. Zhang Y et al (2011) X-linked inhibitor of apoptosis positive nuclear labeling: a new inde-
pendent prognostic biomarker of breast invasive ductal carcinoma. Diagn Pathol 6:49
122. Hiscutt EL et al (2010) Targeting X-linked inhibitor of apoptosis protein to increase the
efficacy of endoplasmic reticulum stress-induced apoptosis for melanoma therapy. J Invest
Dermatol 130(9):2250–2258
276 M. F. Brown et al.
123. Qi S et al (2008) Expression of cIAP-1 correlates with nodal metastasis in squamous cell
carcinoma of the tongue. Int J Oral Maxillofac Surg 37(11):1047–1053
124. Krajewska M et al (2005) Analysis of apoptosis protein expression in early-stage colo-
rectal cancer suggests opportunities for new prognostic biomarkers. Clin Cancer Res
11(15):5451–5461
125. Esposito I et al (2007) Overexpression of cellular inhibitor of apoptosis protein 2 is an early
event in the progression of pancreatic cancer. J Clin Pathol 60(8):885–895
126. Vucic D et al (2000) ML-IAP, a novel inhibitor of apoptosis that is preferentially expressed
in human melanomas. Curr Biol 10(21):1359–1366
127. Wagener N et al (2007) Expression of inhibitor of apoptosis protein Livin in renal cell carci-
noma and non-tumorous adult kidney. Br J Cancer 97(9):1271–1276
128. Gong J et al (2005) Melanoma inhibitor of apoptosis protein is expressed differentially in
melanoma and melanocytic naevus, but similarly in primary and metastatic melanomas. J Clin
Pathol 58(10):1081–1085
129. Kempkensteffen C et al (2007) Expression of the apoptosis inhibitor livin in renal cell carci-
nomas: correlations with pathology and outcome. Tumour Biol 28(3):132–138
130. Gazzaniga P et al (2003) Expression and prognostic significance of LIVIN, SURVIVIN and other
apoptosis-related genes in the progression of superficial bladder cancer. Ann Oncol 14(1):85–90
131. El-Mesallamy HO, Hegab HM, Kamal AM (2011) Expression of inhibitor of apoptosis pro-
tein (IAP) livin/BIRC7 in acute leukemia in adults: correlation with prognostic factors and
outcome. Leuk Res 35(12):1616–1622
132. Nedelcu T et al (2008) Livin and Bcl-2 expression in high-grade osteosarcoma. J Cancer
Res Clin Oncol 134(2):237–244
133. Kim DK et al (2005) Expression of inhibitor-of-apoptosis protein (IAP) livin by neuroblas-
toma cells: correlation with prognostic factors and outcome. Pediatr Dev Pathol 8(6):621–629
134. Duffy MJ et al (2007) Survivin: a promising tumor biomarker. Cancer Lett 249(1):49–60
135. Ambrosini G, Adida C, Altieri DC (1997) A novel anti-apoptosis gene, survivin, expressed
in cancer and lymphoma. Nat Med 3(8):917–921
136. Ambrosini G et al (1998) Induction of apoptosis and inhibition of cell proliferation by sur-
vivin gene targeting. J Biol Chem 273(18):11177–11182
137. Endo K et al (2009) Clinical significance of Smac/DIABLO expression in colorectal cancer.
Oncol Rep 21(2):351–355
138. Xu Y et al (2011) Role of smac in determining the chemotherapeutic response of esophageal
squamous cell carcinoma. Clin Cancer Res 17(16):5412–5422
139. Martinez-Ruiz G et al (2008) Role of Smac/DIABLO in cancer progression. J Exp Clin
Cancer Res 27:48
140. Fong WG et al (2000) Expression and genetic analysis of XIAP-associated factor 1 (XAF1)
in cancer cell lines. Genomics 70(1):113–122
141. Sekimura A et al (2004) Expression of Smac/DIABLO is a novel prognostic marker in lung
cancer. Oncol Rep 11(4):797–802
142. Pluta P et al (2011) Correlation of Smac/DIABLO protein expression with the clinico-path-
ological features of breast cancer patients. Neoplasma 58(5):430–435
143. Mizutani Y, Katsuoka Y, Bonavida B (2010) Prognostic significance of second mitochon-
dria-derived activator of caspase (Smac/DIABLO) expression in bladder cancer and target
for therapy. Int J Oncol 37(2):503–508
144. Mizutani Y et al (2005) Downregulation of Smac/DIABLO expression in renal cell
carcinoma and its prognostic significance. J Clin Oncol 23(3):448–454
145. Kempkensteffen C et al (2008) Expression levels of the mitochondrial IAP antagonists
Smac/DIABLO and Omi/HtrA2 in clear-cell renal cell carcinomas and their prognostic
value. J Cancer Res Clin Oncol 134(5):543–550
146. Dobrzycka B et al (2010) Prognostic significance of smac/DIABLO in endometrioid endo-
metrial cancer. Folia Histochem Cytobiol 48(4):678–681
147. Arellano-Llamas A et al (2006) High Smac/DIABLO expression is associated with early
local recurrence of cervical cancer. BMC Cancer 6:256
10 SMAC IAP Addiction in Cancer 277
148. Pluta A et al (2010) Influence of high expression of Smac/DIABLO protein on the clinical
outcome in acute myeloid leukemia patients. Leuk Res 34(10):1308–1313
149. Ndubaku C et al (2009) Targeting inhibitor of apoptosis proteins for therapeutic interven-
tion. Future Med Chem 1(8):1509–1525
150. Vucic D, Fairbrother WJ (2007) The inhibitor of apoptosis proteins as therapeutic targets in
cancer. Clin Cancer Res 13(20):5995–6000
151. Wang S (2011) Design of small-molecule Smac mimetics as IAP antagonists. Curr Top
Microbiol Immunol 348:89–113
152. Fulda S, Meyer E, Debatin KM (2002) Inhibition of TRAIL-induced apoptosis by Bcl-2
overexpression. Oncogene 21(15):2283–2294
153. Li L et al (2004) A small molecule Smac mimic potentiates TRAIL- and TNFalpha-
mediated cell death. Science 305(5689):1471–1474
154. Arnt CR et al (2002) Synthetic Smac/DIABLO peptides enhance the effects of chemothera-
peutic agents by binding XIAP and cIAP1 in situ. J Biol Chem 277(46):44236–44243
155. Yang L et al (2003) Predominant suppression of apoptosome by inhibitor of apoptosis pro-
tein in non-small cell lung cancer H460 cells: therapeutic effect of a novel polyarginine-
conjugated Smac peptide. Cancer Res 63(4):831–837
156. Sun H et al (2007) Design, synthesis, and characterization of a potent, nonpeptide, cell-per-
meable, bivalent Smac mimetic that concurrently targets both the BIR2 and BIR3 domains
in XIAP. J Am Chem Soc 129(49):15279–15294
157. Bertrand MJ et al (2008) cIAP1 and cIAP2 facilitate cancer cell survival by functioning as
E3 ligases that promote RIP1 ubiquitination. Mol Cell 30(6):689–700
158. Varfolomeev E et al (2008) c-IAP1 and c-IAP2 are critical mediators of tumor necrosis fac-
tor alpha (TNFalpha)-induced NF-kappaB activation. J Biol Chem 283(36):24295–24299
159. Vince JE et al (2007) IAP antagonists target cIAP1 to induce TNFalpha-dependent apopto-
sis. Cell 131(4):682–693
160. Gao Z et al (2007) A dimeric Smac/diablo peptide directly relieves caspase-3 inhibi-
tion by XIAP. dynamic and cooperative regulation of XIAP by Smac/Diablo. J Biol Chem
282(42):30718–30727
161. Varfolomeev E et al (2009) X chromosome-linked inhibitor of apoptosis regulates cell death
induction by proapoptotic receptor agonists. J Biol Chem 284(50):34553–34560
162. Sun Q et al (2011) Smac Modulates chemosensitivity in head and neck cancer cells through
the mitochondrial apoptotic pathway. Clin Cancer Res 17(8):2361–2372 [Epub ahead of print]
163. Feltham R et al (2011) Smac mimetics activate the E3 ligase activity of cIAP1 protein by
promoting RING domain dimerization. J Biol Chem 286(19):17015–17028
164. Mace PD et al (2008) Structures of the cIAP2 RING domain reveal conformational
changes associated with ubiquitin-conjugating enzyme (E2) recruitment. J Biol Chem
283(46):31633–31640
165. Dueber EC et al (2011) Antagonists induce a conformational change in cIAP1 that promotes
autoubiquitination. Science 334(6054):376–380
166. Dineen SP et al (2010) Smac mimetic increases chemotherapy response and improves sur-
vival in mice with pancreatic cancer. Cancer Res 70(7):2852–2861
167. Cheung HH et al (2009) Down-regulation of c-FLIP enhances death of cancer cells by smac
mimetic compound. Cancer Res 69(19):7729–7738
168. Lu J et al (2011) Therapeutic potential and molecular mechanism of a novel, potent, nonpep-
tide, Smac mimetic SM-164 in combination with TRAIL for cancer treatment. Mol Cancer
Ther 10(5):902–914
169. Foster FM et al (2009) Targeting inhibitor of apoptosis proteins in combination with ErbB
antagonists in breast cancer. Breast Cancer Res 11(3):R41
170. Aird KM et al (2010) X-linked inhibitor of apoptosis protein inhibits apoptosis in inflam-
matory breast cancer cells with acquired resistance to an ErbB1/2 tyrosine kinase inhibitor.
Mol Cancer Ther 9(5):1432–1442
171. Fulda S et al (2002) Smac agonists sensitize for Apo2L/TRAIL- or anticancer drug-induced
apoptosis and induce regression of malignant glioma in vivo. Nat Med 8(8):808–815
278 M. F. Brown et al.
193. Crnkovic-Mertens I et al (2006) Isoform-specific silencing of the livin gene by RNA inter-
ference defines Livin beta as key mediator of apoptosis inhibition in HeLa cells. J Mol Med
(Berl) 84(3):232–240
194. Tseng PH et al (2010) Different modes of ubiquitination of the adaptor TRAF3 selectively
activate the expression of type I interferons and proinflammatory cytokines. Nat Immunol
11(1):70–75
195. Kim S et al (2009) Carcinoma-produced factors activate myeloid cells through TLR2 to
stimulate metastasis. Nature 457(7225):102–106
196. Wu Y, Zhou BP (2010) TNF-alpha/NF-kappaB/Snail pathway in cancer cell migration and
invasion. Br J Cancer 102(4):639–644
197. Petersen SL et al (2010) Overcoming cancer cell resistance to Smac mimetic induced apop-
tosis by modulating cIAP-2 expression. Proc Natl Acad Sci USA 107(26):11936–11941
198. Fesik SW (2005) Promoting apoptosis as a strategy for cancer drug discovery. Nat Rev
Cancer 5(11):876–885
199. Zhang L, Ming L, Yu J (2007) BH3 mimetics to improve cancer therapy; mechanisms and
examples. Drug Resist Updat 10(6):207–217
200. Flygare J et al (2012) The discovery of a potent small-molecule antagonist of inhibitor of
apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152). J
Med Chem 55(9):4101–4113
201. Cai Q et al (2011) A potent and orally active antagonist (SM-406/AT-406) of multiple inhib-
itor of apoptosis proteins (IAPs) in clinical development for cancer treatment. J Med Chem
54(8):2714–2726
202. Infante JR (2010) A phase I study of LCL-161, an oral IAP inhibitor, in patients with
advanced cancer. In: Proceedings of the 101st annual meeting of the american association
for cancer research. Washington
203. Sikic B (2011) Safety, pharmacokinetics (PK), and pharmacodynamics (PD) of HGS1029,
an inhibitor of apoptosis protein (IAP) inhibitor, in patients (Pts) with advanced solid
tumors: Results of a phase I study. J Clin Oncol (Meeting abstract)
204. Amaravadi RK (2011) Phase 1 study of the Smac mimetic TL32711 in adult subjects with
advanced solid tumors and lymphoma to evaluate safety, pharmacokinetics, pharmacody-
namics and antitumor activity. In: Proceedings of the 102nd annual meeting of the american
association for cancer research. Orlando, Florida
205. Weisberg E et al (2010) Smac mimetics: implications for enhancement of targeted therapies
in leukemia. Leukemia 24(12):2100–2109
206. Mahadevan D et al (2012) Phase I trial of AEG35156 an antisense oligonucleotide to XIAP
plus gemcitabine in patients with metastatic pancreatic ductal adenocarcinoma. Am J Clin
Oncol
Chapter 11
Harnessing Death Receptor Signaling
for Cancer Treatment
Simone Fulda
11.1 Introduction
S. Fulda (*)
Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt,
Frankfurt, Germany
e-mail: [email protected]
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 281
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_11,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
282 S. Fulda
cell death pathways for therapeutic purposes. The current review focuses on the
opportunities to target death receptor signaling in order to develop new therapeutic
strategies for the treatment for cancer.
11.2 Death Receptors
Death receptors belong to the superfamily of tumor necrosis factor (TNF) recep-
tors, which consist of more than 20 members with a wide spectrum of biologi-
cal functions including regulation of cell death, survival, differentiation, and
immune regulation [7]. All TNF receptor family members share a similar, cyto-
plasmic motif of about 80 amino acids, the so-called death domain, which is
critical for transmitting the death signal from the cell’s surface to intracellular
signaling pathways. In addition, death receptors harbor cysteine-rich extracellu-
lar domains for ligand binding. Among the death receptors CD95 (APO-1/Fas),
TNF receptor 1 (TNFR1) and TNF-related apoptosis-inducing ligand (TRAIL)
receptors have been extensively studied in the last two decades [7]. The corre-
sponding death receptor ligands of the TNF superfamily comprise among others
CD95 ligand, TNFα, lymphotoxin-α (the latter two bind to TNFR1), TRAIL and
TWEAK, a ligand for DR3 [7]. The CD95 receptor/CD95 ligand system repre-
sents a major signaling pathway that mediates apoptosis in several different cell
types, for example in the immune system [8]. TRAIL was identified in 1995 based
on its sequence homology to other members of the TNF superfamily and is consti-
tutively expressed in a wide range of tissues [7]. There are two agonistic TRAIL
receptors, that is, TRAIL-R1 and TRAIL-R2, that contain a death domain, and
therefore a signal to cell death upon ligand binding, whereas TRAIL-R3 to R-5
are antagonistic decoy receptors, which bind TRAIL, but are not able to transmit a
death signal [7].
11.3 Apoptosis Pathways
Two major apoptosis signaling pathways have been identified, that is the death
receptor (extrinsic) and the mitochondrial (intrinsic) pathway [4]. Stimulation
of death receptors such as CD95, TRAIL-R1 or TRAIL-R2 by CD95 ligand,
TRAIL or agonistic antibodies results in receptor oligomerization and recruit-
ment of FADD and caspase-8 to activated death receptors to form the death-
inducing signaling complex (DISC) [7]. This multimeric complex drives the
activation of caspase-8, which in turn transmits the apoptosis signal. To this end,
caspase-8 can directly activate effector pathways of apoptosis by cleaving cas-
pase-3. Alternatively, caspase-8 can initiate a crosstalk to the mitochondrial path-
way of apoptosis by processing Bid into its active form tBid [9]. Bid belongs to the
proapoptotic proteins of the Bcl-2 family that contains a BH3-only domain [10].
11 Harnessing Death Receptor Signaling for Cancer Treatment 283
tBid translocates from the cytosol to the mitochondria and triggers mitochondrial
outer membrane permeabilization. This results in the release of mitochondrial pro-
teins from the intermembrane space of the mitochondria into the cytosol, for exam-
ple of cytochrome c or second mitochondrial activator of caspases (Smac) [10].
Cytochrome c forms a complex in the cytosol together with Apaf-1 and caspase-9
to trigger caspase-9 and subsequently caspase-3 activation [10]. Smac antagonizes
Inhibitor of Apoptosis (IAP) proteins, which function as endogenous inhibitors of
caspases [11]. The release of caspases from the inhibition of IAP proteins by Smac
promotes caspase activation and apoptosis. The engagement of either the extrinsic
or the intrinsic apoptosis pathway results in activation of caspases, a family of pro-
teases that function as executioners in multiple modes of cell death [12].
Cell death pathways are tightly regulated by pro- and antiapoptotic factors.
This should ensure that they are rapidly activated upon stimulation, for example
upon death receptor ligation. Vice versa, this tight control should prevent their
accidental engagement, which could have detrimental effects on cellular survival.
Importantly, cancer cells have adopted many of these antiapoptotic mechanisms to
escape programmed cell death. Accordingly, evasion of apoptosis represents one of
the hallmarks of cancer cells. This also implies that targeting defective cell death
pathways bears the potential to tackle one of the key properties of m alignant cells.
One of the hallmarks of human cancers is their ability to evade apoptosis in order
to survive environmental or oncogenic stress signals. This favors the progressive
growth of a tumor and, as such, cooperates with proliferative signals to foster
tumor development as well as its progression. On a theoretical ground, apoptosis
programs can be disrupted either via a reduction in the apoptosis promoting fac-
tors or via the dominance of processes that block apoptosis. Both genetic as well
as epigenetic events can cause inactivation of apoptosis pathways, implying that at
least a proportion of these events is in principle reversible and amenable for thera-
peutic interventions.
Signaling via the death receptor pathway of apoptosis may be disturbed at multiple
levels in human cancers. Along the signaling cascade, the surface levels of death
receptors have been described to be downregulated in human cancers. CD95 was
reduced in CD95-resistant tumor cells [13, 14] as well as in cells that were refrac-
tory to various anticancer drugs [13, 14], indicating that CD95 expression also
284 S. Fulda
regulates drug responsiveness. Genetic alterations of CD95 have also been impli-
cated in tumorigenesis. Hematological malignancies, as well as solid tumors, were
reported to harbor CD95 gene mutations [15]. In addition to genetic lesions, epige-
netic alterations including hypermethylation of the CD95 promoter have also been
implicated as an underlying cause for reduced CD95 expression in cancers [16, 17].
This mechanism may contribute to tumor immune escape, as cancer with epigeneti-
cally inactivated CD95 displayed low sensitivity to immune cell-mediated killing.
Consequently, restoration of CD95 expression by treatment with epigenetic drugs
such as histone deacetylase inhibitors concomitantly enhanced NK cell-dependent
tumor cell killing as well as the response to chemotherapy [18].
Along the same lines, resistance toward TRAIL has been linked to low or absent
surface expression of one of the two agonistic TRAIL receptors TRAIL-R1 and
TRAIL-R2. Interestingly, the chromosomal localization of these TRAIL receptors
on chromosome 8p falls within a region that is often genetically altered in human
cancers, for example by the loss of heterozygosity (LOH) [19]. Furthermore, the
loss of both copies of TRAIL-R1 or TRAIL-R2 due to deletions or mutations has
been detected in a small percentage of various cancers, for example several car-
cinomas (colorectal, breast, head and neck, lung), non-Hodgkin’s lymphoma and
osteosarcoma [20, 21]. In addition to these genetic events, aberrations in the sub-
cellular distribution of TRAIL receptors may account for the evasion of TRAIL-
induced apoptosis. In this respect, it has been reported in colon carcinoma cells
that the apoptosis-inducing TRAIL receptors, TRAIL-R1 and TRAIL-R2, are
retained in intracellular stores, for example the endoplasmic reticulum, and are not
properly transported to the cell surface, the location where they usually engage
with their ligand TRAIL to initiate proapoptotic signaling [22].
Another mechanism to block death receptor signaling resides in the rela-
tive abundance of decoy receptors. Both the CD95 and TRAIL cascade can be
impaired by such decoy receptors. Decoy receptor 3 (DcR3) has been shown to
competitively bind to CD95 ligand, thereby blocking CD95-induced apoptosis
[23, 24]. Of note, high expression levels of DcR3 or genetic amplification of
CcR3 were detected in lung or colon cancer and in glioblastoma [23, 24], indi-
cating that DcR3 may contribute to CD95 resistance in these cancers. TRAIL-R3
and TRAIL-R4 represent the decoy receptors that bind TRAIL, but are not able to
transmit a death signal, since they are devoid of a functional death domain. The
death domain is the intracellular region of the receptor that is required for the
recruitment of signaling proteins to activated death receptors and the subsequent
activation of initiator caspases. Interestingly, it was described that TRAIL-R3
and TRAIL-R4 inhibit TRAIL-R1 and TRAIL-R2-mediated apoptosis upon
treatment with the soluble ligand TRAIL via distinct mechanisms [25]. While
TRAIL-R3 inhibits the assembly of the DISC complex by sequestrating TRAIL
within lipid rafts, TRAIL-R4 is co-recruited together with TRAIL-R2 into the
11 Harnessing Death Receptor Signaling for Cancer Treatment 285
DISC and interferes with the activation of initiator caspases [25]. Overexpression
of TRAIL-R3 was found in several cancers, for example in gastrointestinal can-
cers or leukemia [26–28]. In colorectal cancer, concomitant high TRAIL-R3 and
low/medium TRAIL-R1 expression was shown to correlate with a poor response
to 5-FU-based first-line chemotherapy and with shorter progression-free survival
[27]. AML blasts were recently reported to express TRAIL-R3 in a substantial
proportion of patients in addition to the proapoptotic TRAIL receptors [26]. Of
note, co-expression of this decoy receptor correlated with a significant shortened
overall survival. [26]. Knockdown of TRAIL-R3 resulted in TRAIL-induced
cell death confirming the decoy function of TRAIL-R3 on AML blasts [26].
Also, treatment with TRAIL-R2-specific antibodies resulted in higher cell death
rates [26]. This underlines that specific targeting of agonistic TRAIL receptors
is required in cancers that express TRAIL decoy receptors in order to exploit the
apoptosis-inducing activities of TRAIL [26].
Furthermore, DNA-damaging events such as anticancer drugs or ionizing
radiation were found to transcriptionally activate expression levels of TRAIL-R3
[28, 29]. This p53-stimulated transactivation occurred via a p53 consensus ele-
ment located within the first intron of the human TRAIL-R3 gene [29]. Similarly,
TRAIL-R4 was found to be induced by p53 upon ectopic expression of p53 [30].
This indicates that genotoxic drugs not only induce proapoptotic TRAIL recep-
tors [31], but may also stimulate antiapoptotic genes such as TRAIL-R3 and
TRAIL-R4.
TRAIL-R4 has recently been demonstrated to stimulate activation of signaling
pathways in an Akt-dependent manner in addition to its ability to inhibit TRAIL-
mediated signaling and cell death at the membrane by forming a heteromeric com-
plex with the agonistic receptor TRAIL-R2 [32]. Overexpression of TRAIL-R4
triggered morphological changes such as cell rounding, loss of adherence,
increased cell proliferation in vitro, and promoted tumor growth in vivo, indicating
that it contributes to carcinogenesis [32].
c-FLIP was not detected in normal pancreatic ducts [37]. Concomitant knockdown
of both c-FLIPL and c-FLIPS isoforms and individual silencing of either c-FLIPL
or c-FLIPS by RNA interference significantly increased TRAIL- and CD95-
induced apoptosis [37]. In addition, downregulation of c-FLIP by pretreatment
with chemotherapeutic drugs, for example 5-fluorouracil (5-FU), sensitized pan-
creatic carcinoma cells to death receptor-mediated apoptosis [37]. Of note, pri-
mary cultured pancreatic cancer cells were similarly primed for TRAIL-triggered
apoptosis by pre-exposure to anticancer drugs [37].
The notion that caspase-8, a key factor of death receptor-triggered apoptosis, may
restrict tumor development is underlined by a study, showing that a deficiency in
caspase-8 can facilitate cellular transformation [38]. Furthermore, expression of
caspase-8 can be downregulated by epigenetic inactivation. Caspase-8 expres-
sion was shown to be silenced by hypermethylation of a regulatory sequence
of the caspase-8 gene mapped to the boundary between exon 3 and intron 3 in
several malignancies, for example neuroectodermal tumors, sarcoma, and lung
carcinoma [39–43]. While this regulatory region of caspase-8 is not a classical
CpG island and is devoid of promoter activity, its methylation status was associ-
ated with caspase-8 expression levels in cell lines and primary tumor specimens
[11, 13, 14, 44–46]. Interestingly, co-methylation for caspase-8 and FLIP was
identified in neuroblastoma in one study, suggesting that caspase-8 is epigeneti-
cally silenced in a non-random fashion [47]. Hypermethylation of caspase-8 was
recently shown to be associated with relapse susceptibility in neuroblastoma [48].
In medulloblastoma, the loss of caspase-8 expression correlated with unfavorable
outcome in childhood medulloblastoma [42]. The loss of caspase-8 protein expres-
sion was identified in the majority of neuroblastoma tumor samples and was not
restricted to advanced disease stages [46]. No correlation was observed between
caspase-8 expression and MYCN amplification or other variables of high-risk dis-
ease (e.g., 1p36 aberrations, disease stage, age at diagnosis, or tumor histology)
[46]. Also, the loss of caspase-8 protein had no effect on event-free or overall sur-
vival in the overall study population or in distinct subgroups of patients [46], indi-
cating that inactivation of caspase-8 is not a characteristic feature of aggressive
neuroblastoma.
Moreover, caspase-8L is a dominant-negative variant of caspase-8 that is pro-
duced by alternative splicing, for example in CD34+ progenitor cells as well as in
leukemia and neuroblastoma [49–52]. Caspase-8L was found to block the bind-
ing of caspase-8 to FADD [49]. In addition, caspase-8L was recruited to the DISC
after CD95 stimulation instead of caspase-8, thereby interfering with CD95 signal-
ing at the receptor level by preventing caspase-8 activation [52].
Further, the tyrosine kinase Src was identified as a kinase that p hosphorylates
caspase-8 on tyrosine 308 in the linker loop of caspase-8, resulting in the sup-
pression of the proapoptotic activity of caspase-8 [53]. Phosphorylation of
11 Harnessing Death Receptor Signaling for Cancer Treatment 287
Since the death receptor pathway of apoptosis represents a signaling cascade that
directly connects to an intrinsic cell death machinery and that is amenable to ther-
apeutic targeting from the outside, it has attracted much attention in the last dec-
ade for the development of molecular cancer therapeutics. Most strategies focused
on targeting the two agonistic TRAIL receptors as discussed in more detail in the
following paragraphs.
one of the characteristics of TRAIL receptor agonists, as they have been shown
to predominantly trigger apoptosis in malignant cells with little effect on normal
cells [7].
There is now a large body of evidence from preclinical studies that recombinant
soluble TRAIL or antibodies against the proapoptotic TRAIL receptors TRAIL-R1
or TRAIL-R2 trigger apoptosis in a wide range of cancer cell lines and human
cancer xenograft models [19, 58, 59]. Of note, TRAIL-R2 antibodies were found
to trigger tumor-specific T-cell memory besides their cytotoxic effects against can-
cer cells, thereby protecting from tumor recurrence [60]. In addition to soluble
recombinant TRAIL ligand or TRAIL receptor specific antibodies, gene therapy
approaches have been launched to deliver TRAIL to the tumor site. An adenoviral
vector-based system based on the hTERT promoter yielded high levels of TRAIL,
leading to tumor-specific induction of apoptosis, suppression of tumor growth in a
xenograft model of breast cancer, and increased tumor-free survival of mice [61].
Furthermore, the apoptosis-inducing activity of TRAIL has been combined with
the ability of mesenchymal stem cells (MSCs) to infiltrate tumors as well as lym-
phatic tissues in order to deliver TRAIL to the primary tumor site as well as to
disseminated cancer cells [62]. In a lung cancer model, MSCs expressing TRAIL
were shown to cause tumor growth inhibition by triggering apoptosis [63].
11.5.3 Upregulation of Caspase-8
TRAIL receptor agonists have been evaluated in a number of early clinical tri-
als over the last years. This includes protocols with recombinant human TRAIL
(e.g., dulanermin) [164–169] that is directed against both agonistic TRAIL recep-
tors TRAIL-R1 and TRAIL‐R2 as well as regimens with agonistic monoclonal
antibodies targeting selectively either TRAIL‐R1, for example mapatumumab
[170–178], or TRAIL‐R2, for example lexatumumab [179–182], conatumumab
[166, 169, 183–189], drozitumab [190–194], tigatuzumab [195], and LBY135
[196]. Initially, these trials were conducted with TRAIL or TRAIL receptor anti-
bodies as single agents. Subsequently, combination studies were launched to
exploit additive or even synergistic interactions by simultaneously targeting both
death receptor and additional signaling pathways. The design of these combina-
tion protocols was based on a large body of data from preclinical studies show-
ing that the incorporation of additional agents that concomitantly trigger, for
example, mitochondrial apoptosis such as chemotherapeutic drugs significantly
11 Harnessing Death Receptor Signaling for Cancer Treatment 291
11.6 Conclusion
References
1. Taylor RC, Cullen SP, Martin SJ (2008) Apoptosis: controlled demolition at the cellular
level. Nat Rev Mol Cell Biol 9:231–241
2. Evan GI, Vousden KH (2001) Proliferation, cell cycle and apoptosis in cancer. Nature
411:342–348
3. Fulda S (2009) Tumor resistance to apoptosis. Int J Cancer 124:511–515
4. Fulda S, Debatin KM (2006) Extrinsic versus intrinsic apoptosis pathways in anticancer
chemotherapy. Oncogene 25:4798–4811
5. Johnstone RW, Ruefli AA, Lowe SW (2002) Apoptosis: a link between cancer genetics and
chemotherapy. Cell 108:153–164
292 S. Fulda
6. Makin G, Dive C (2001) Apoptosis and cancer chemotherapy. Trends Cell Biol 11:22–26
7. Ashkenazi A (2008) Targeting the extrinsic apoptosis pathway in cancer. Cytokine Growth
Factor Rev 19:325–331
8. Lavrik IN, Krammer PH (2012) Regulation of CD95/Fas signaling at the DISC. Cell Death
Differ 19:36–41
9. Kroemer G, Galluzzi L, Brenner C (2007) Mitochondrial membrane permeabilization in cell
death. Physiol Rev 87:99–163
10. Fulda S, Galluzzi L, Kroemer G (2010) Targeting mitochondria for cancer therapy. Nat Rev
Drug Discov 9:447–464
11. Fulda S, Vucic D (2012) Targeting IAP proteins for therapeutic intervention in cancer. Nat
Rev Drug Discov 11:109–124
12. Logue SE, Martin SJ (2008) Caspase activation cascades in apoptosis. Biochem Soc Trans 36:1–9
13. Friesen C, Fulda S, Debatin KM (1997) Deficient activation of the CD95 (APO-1/Fas) sys-
tem in drug-resistant cells. Leukemia 11:1833–1841
14. Fulda S, Scaffidi C, Susin SA et al (1998) Activation of mitochondria and release of mito-
chondrial apoptogenic factors by betulinic acid. J Biol Chem 273:33942–33948
15. Fulda S (2009) Inhibitor of apoptosis proteins in hematological malignancies. Leukemia
23:467–476
16. Petak I, Danam RP, Tillman DM et al (2003) Hypermethylation of the gene promoter and
enhancer region can regulate Fas expression and sensitivity in colon carcinoma. Cell Death
Differ 10:211–217
17. Van Noesel MM, Van Bezouw S, Salomons GS et al (2002) Tumor-specific down-regulation
of the tumor necrosis factor-related apoptosis-inducing ligand decoy receptors DcR1 and
DcR2 is associated with dense promoter hypermethylation. Cancer Res 62:2157–2161
18. Maecker HL, Yun Z, Maecker HT et al (2002) Epigenetic changes in tumor Fas levels deter-
mine immune escape and response to therapy. Cancer Cell 2:139–148
19. Ashkenazi A (2008) Directing cancer cells to self-destruct with pro-apoptotic receptor ago-
nists. Nat Rev Drug Discov 7:1001–1012
20. Dechant MJ, Fellenberg J, Scheuerpflug CG et al (2004) Mutation analysis of the apoptotic
“death-receptors” and the adaptors TRADD and FADD/MORT-1 in osteosarcoma tumor
samples and osteosarcoma cell lines. Int J Cancer 109:661–667
21. Pai SI, Wu GS, Ozoren N et al (1998) Rare loss-of-function mutation of a death receptor
gene in head and neck cancer. Cancer Res 58:3513–3518
22. Jin Z, Mcdonald ER 3rd, Dicker DT et al (2004) Deficient tumor necrosis factor-related
apoptosis-inducing ligand (TRAIL) death receptor transport to the cell surface in human
colon cancer cells selected for resistance to TRAIL-induced apoptosis. J Biol Chem
279:35829–35839
23. Pitti RM, Marsters SA, Lawrence DA et al (1998) Genomic amplification of a decoy recep-
tor for Fas ligand in lung and colon cancer. Nature 396:699–703
24. Roth W, Isenmann S, Nakamura M et al (2001) Soluble decoy receptor 3 is expressed by
malignant gliomas and suppresses CD95 ligand-induced apoptosis and chemotaxis. Cancer
Res 61:2759–2765
25. Merino D, Lalaoui N, Morizot A et al (2006) Differential inhibition of TRAIL-mediated
DR5-DISC formation by decoy receptors 1 and 2. Mol Cell Biol 26:7046–7055
26. Chamuleau ME, Ossenkoppele GJ, Van Rhenen A et al (2011) High TRAIL-R3TRAIL-R3
expression on leukemic blasts is associated with poor outcome and induces apopto-
sis-resistance which can be overcome by targeting TRAIL-R2TRAIL-R2. Leuk Res
35:741–749
27. Granci V, Bibeau F, Kramar A et al (2008) Prognostic significance of TRAIL-R1 and
TRAIL-R3 expression in metastatic colorectal carcinomas. Eur J Cancer 44:2312–2318
28. Sheikh MS, Huang Y, Fernandez-Salas EA et al (1999) The antiapoptotic decoy receptor
TRID/TRAIL-R3 is a p53-regulated DNA damage-inducible gene that is overexpressed in
primary tumors of the gastrointestinal tract. Oncogene 18:4153–4159
11 Harnessing Death Receptor Signaling for Cancer Treatment 293
29. Ruiz De Almodovar C, Ruiz-Ruiz C, Rodriguez A et al (2004) Tumor necrosis factor-related apop-
tosis-inducing ligand (TRAIL) decoy receptor TRAIL-R3 is up-regulated by p53 in breast tumor
cells through a mechanism involving an intronic p53-binding site. J Biol Chem 279:4093–4101
30. Meng RD, Mcdonald ER 3rd, Sheikh MS et al (2000) The TRAIL decoy receptor TRUNDD
(DcR2, TRAIL-R4) is induced by adenovirus-p53 overexpression and can delay TRAIL-,
p53-, and KILLER/DR5-dependent colon cancer apoptosis. Mol Ther 1:130–144
31. Sheikh MS, Burns TF, Huang Y et al (1998) P53-dependent and -independent regulation of
the death receptor KILLER/DR5 gene expression in response to genotoxic stress and tumor
necrosis factor alpha. Cancer Res 58:1593–1598
32. Lalaoui N, Morle A, Merino D et al (2011) TRAIL-R4 promotes tumor growth and resist-
ance to apoptosis in cervical carcinoma HeLa cells through AKT. PLoS One 6:19679
33. Hao C, Beguinot F, Condorelli G et al (2001) Induction and intracellular regulation of tumor
necrosis factor-related apoptosis-inducing ligand (TRAIL) mediated apoptosis in human
malignant glioma cells. Cancer Res 61:1162–1170
34. Krueger A, Baumann S, Krammer PH et al (2001) FLICE-inhibitory proteins: regulators of
death receptor-mediated apoptosis. Mol Cell Biol 21:8247–8254
35. Fulda S, Meyer E, Debatin KM (2000) Metabolic inhibitors sensitize for CD95 (APO-
1/Fas)-induced apoptosis by down-regulating Fas-associated death domain-like interleukin
1-converting enzyme inhibitory protein expression. Cancer Res 60:3947–3956
36. Longley DB, Wilson TR, Mcewan M et al (2006) c-FLIP inhibits chemotherapy-induced
colorectal cancer cell death. Oncogene 25:838–848
37. Haag C, Stadel D, Zhou S et al (2011) Identification of c-FLIP(L) and c-FLIP(S) as critical
regulators of death receptor-induced apoptosis in pancreatic cancer cells. Gut 60:225–237
38. Krelin Y, Zhang L, Kang TB et al (2008) Caspase-8 deficiency facilitates cellular transfor-
mation in vitro. Cell Death Differ 15:1350–1355
39. Fulda S, Kufer MU, Meyer E et al (2001) Sensitization for death receptor- or drug-induced apopto-
sis by re-expression of caspase-8 through demethylation or gene transfer. Oncogene 20:5865–5877
40. Harada K, Toyooka S, Shivapurkar N et al (2002) Deregulation of caspase 8 and 10 expres-
sion in pediatric tumors and cell lines. Cancer Res 62:5897–5901
41. Hopkins-Donaldson S, Ziegler A, Kurtz S et al (2003) Silencing of death receptor and cas-
pase-8 expression in small cell lung carcinoma cell lines and tumors by DNA methylation.
Cell Death Differ 10:356–364
42. Pingoud-Meier C, Lang D, Janss AJ et al (2003) Loss of caspase-8 protein expression correlates
with unfavorable survival outcome in childhood medulloblastoma. Clin Cancer Res 9:6401–6409
43. Teitz T, Wei T, Valentine MB et al (2000) Caspase 8 is deleted or silenced preferentially in
childhood neuroblastomas with amplification of MYCN. Nat Med 6:529–535
44. Finlay D, Howes A, Vuori K (2009) Critical role for caspase-8 in epidermal growth factor
signaling. Cancer Res 69:5023–5029
45. Fulda S, Debatin KM (2006) 5-Aza-2′-deoxycytidine and IFN-gamma cooperate to sensitize
for TRAIL-induced apoptosis by upregulating caspase-8caspase-8. Oncogene 25:5125–5133
46. Fulda S, Poremba C, Berwanger B et al (2006) Loss of caspase-8 expression does not cor-
relate with MYCN amplification, aggressive disease, or prognosis in neuroblastoma. Cancer
Res 66:10016–10023
47. Fulda S, Wick W, Weller M et al (2002) Smac agonists sensitize for Apo2L/TRAIL- or anticancer
drug-induced apoptosis and induce regression of malignant glioma in vivo. Nat Med 8:808–815
48. Grau E, Martinez F, Orellana C et al (2011) Hypermethylation of apoptotic genes as inde-
pendent prognostic factor in neuroblastoma disease. Mol Carcinog 50:153–162
49. Himeji D, Horiuchi T, Tsukamoto H et al (2002) Characterization of caspase-8L: a
novel isoform of caspase-8 that behaves as an inhibitor of the caspase cascade. Blood
99:4070–4078
50. Horiuchi T, Himeji D, Tsukamoto H et al (2000) Dominant expression of a novel splice var-
iant of caspase-8 in human peripheral blood lymphocytes. Biochem Biophys Res Commun
272:877–881
294 S. Fulda
51. Miller MA, Karacay B, Zhu X et al (2006) Caspase 8L, a novel inhibitory isoform of cas-
pase 8, is associated with undifferentiated neuroblastoma. Apoptosis 11:15–24
52. Mohr A, Zwacka RM, Jarmy G et al (2005) Caspase-8L expression protects CD34+ hematopoi-
etic progenitor cells and leukemic cells from CD95-mediated apoptosis. Oncogene 24:2421–2429
53. Cursi S, Rufini A, Stagni V et al (2006) Src kinase phosphorylates Caspase-8 on Tyr380: a
novel mechanism of apoptosis suppression. EMBO J 25:1895–1905
54. Barbero S, Barila D, Mielgo A et al (2008) Identification of a critical tyrosine residue in cas-
pase 8 that promotes cell migration. J Biol Chem 283:13031–13034
55. Senft J, Helfer B, Frisch SM (2007) Caspase-8 interacts with the p85 subunit of phosphati-
dylinositol 3-kinase to regulate cell adhesion and motility. Cancer Res 67:11505–11509
56. Barbero S, Mielgo A, Torres V et al (2009) Caspase-8 association with the focal adhesion
complex promotes tumor cell migration and metastasis. Cancer Res 69:3755–3763
57. Torres VA, Mielgo A, Barbero S et al (2010) Rab5 mediates caspase-8-promoted cell motil-
ity and metastasis. Mol Biol Cell 21:369–376
58. Chuntharapai A, Dodge K, Grimmer K et al (2001) Isotype-dependent inhibition of tumor
growth in vivo by monoclonal antibodies to death receptor 4. J Immunol 166:4891–4898
59. Ichikawa K, Liu W, Zhao L et al (2001) Tumoricidal activity of a novel anti-human DR5
monoclonal antibody without hepatocyte cytotoxicity. Nat Med 7:954–960
60. Takeda K, Yamaguchi N, Akiba H et al (2004) Induction of tumor-specific T cell immunity
by anti-DR5 antibody therapy. J Exp Med 199:437–448
61. Lin T, Huang X, Gu J et al (2002) Long-term tumor-free survival from treatment with
the GFP-TRAIL fusion gene expressed from the hTERT promoter in breast cancer cells.
Oncogene 21:8020–8028
62. Mohr A, Albarenque SM, Deedigan L et al (2010) Targeting of XIAP combined with sys-
temic mesenchymal stem cell-mediated delivery of sTRAIL ligand inhibits metastatic
growth of pancreatic carcinoma cells. Stem Cells 28:2109–2120
63. Mohr A, Lyons M, Deedigan L et al (2008) Mesenchymal stem cells expressing TRAIL
lead to tumour growth inhibition in an experimental lung cancer model. J Cell Mol Med
12:2628–2643
64. Belka C, Schmid B, Marini P et al (2001) Sensitization of resistant lymphoma cells to irra-
diation-induced apoptosis by the death ligand TRAIL. Oncogene 20:2190–2196
65. Chinnaiyan AM, Prasad U, Shankar S et al (2000) Combined effect of tumor necrosis fac-
tor-related apoptosis-inducing ligand and ionizing radiation in breast cancer therapy. Proc
Natl Acad Sci USA 97:1754–1759
66. Gliniak B, Le T (1999) Tumor necrosis factor-related apoptosis-inducing ligand’s anti-
tumor activity in vivo is enhanced by the chemotherapeutic agent CPT-11. Cancer Res
59:6153–6158
67. Keane MM, Rubinstein Y, Cuello M et al (2000) Inhibition of NF-kappaB activity enhances
TRAIL mediated apoptosis in breast cancer cell lines. Breast Cancer Res Treat 64:211–219
68. Nagane M, Pan G, Weddle JJ et al (2000) Increased death receptor 5 expression by chem-
otherapeutic agents in human gliomas causes synergistic cytotoxicity with tumor necrosis
factor-related apoptosis-inducing ligand in vitro and in vivo. Cancer Res 60:847–853
69. Ray S, Almasan A (2003) Apoptosis induction in prostate cancer cells and xenografts by
combined treatment with Apo2 ligand/tumor necrosis factor-related apoptosis-inducing
ligand and CPT-11. Cancer Res 63:4713–4723
70. Rohn TA, Wagenknecht B, Roth W et al (2001) CCNU-dependent potentiation of
TRAIL/Apo2L-induced apoptosis in human glioma cells is p53-independent but may
involve enhanced cytochrome c release. Oncogene 20:4128–4137
71. Singh TR, Shankar S, Chen X et al (2003) Synergistic interactions of chemotherapeutic
drugs and tumor necrosis factor-related apoptosis-inducing ligand/Apo-2 ligand on apopto-
sis and on regression of breast carcinoma in vivo. Cancer Res 63:5390–5400
72. Meng RD, El-Deiry WS (2001) p53-independent upregulation of KILLER/DR5 TRAIL
receptor expression by glucocorticoids and interferon-gamma. Exp Cell Res 262:154–169
11 Harnessing Death Receptor Signaling for Cancer Treatment 295
73. Takimoto R, El-Deiry WS (2000) Wild-type p53 transactivates the KILLER/DR5 gene
through an intronic sequence-specific DNA-binding site. Oncogene 19:1735–1743
74. Wang S, El-Deiry WS (2003) Requirement of p53 targets in chemosensitization of colonic
carcinoma to death ligand therapy. Proc Natl Acad Sci USA 100:15095–15100
75. Lacour S, Micheau O, Hammann A et al (2003) Chemotherapy enhances TNF-related
apoptosis-inducing ligand DISC assembly in HT29 human colon cancer cells. Oncogene
22:1807–1816
76. Morizot A, Merino D, Lalaoui N et al (2011) Chemotherapy overcomes TRAIL-R4-
mediated TRAIL resistance at the DISC level. Cell Death Differ 18:700–711
77. Baritaki S, Suzuki E, Umezawa K et al (2008) Inhibition of Yin Yang 1-dependent repressor
activity of DR5 transcription and expression by the novel proteasome inhibitor NPI-0052
contributes to its TRAIL-enhanced apoptosis in cancer cells. J Immunol 180:6199–6210
78. Brooks AD, Jacobsen KM, Li W et al (2010) Bortezomib sensitizes human renal cell carci-
nomas to TRAIL apoptosis through increased activation of caspase-8caspase-8 in the death-
inducing signaling complex. Mol Cancer Res 8:729–738
79. Concannon CG, Koehler BF, Reimertz C et al (2007) Apoptosis induced by protea-
some inhibition in cancer cells: predominant role of the p53/PUMA pathway. Oncogene
26:1681–1692
80. Conticello C, Adamo L, Giuffrida R et al (2007) Proteasome inhibitors synergize with
tumor necrosis factor-related apoptosis-induced ligand to induce anaplastic thyroid carci-
noma cell death. J Clin Endocrinol Metab 92:1938–1942
81. Ding WX, Ni HM, Chen X et al (2007) A coordinated action of Bax, PUMA, and p53 pro-
motes MG132-induced mitochondria activation and apoptosis in colon cancer cells. Mol
Cancer Ther 6:1062–1069
82. Ganten TM, Koschny R, Haas TL et al (2005) Proteasome inhibition sensitizes hepatocel-
lular carcinoma cells, but not human hepatocytes, to TRAIL. Hepatology 42:588–597
83. He Q, Huang Y, Sheikh MS (2004) Proteasome inhibitor MG132 upregulates death receptor
5 and cooperates with Apo2L/TRAIL to induce apoptosis in Bax-proficient and -deficient
cells. Oncogene 23:2554–2558
84. Hetschko H, Voss V, Seifert V et al (2008) Upregulation of DR5 by proteasome inhibitors
potently sensitizes glioma cells to TRAIL-induced apoptosis. FEBS J 275:1925–1936
85. Inoue T, Shiraki K, Fuke H et al (2006) Proteasome inhibition sensitizes hepatocellular car-
cinoma cells to TRAIL by suppressing caspase inhibitors and AKT pathway. Anticancer
Drugs 17:261–268
86. Johnson TR, Stone K, Nikrad M et al (2003) The proteasome inhibitor PS-341 overcomes TRAIL
resistance in Bax and caspase 9-negative or Bcl-xL overexpressing cells. Oncogene 22:4953–4963
87. Kandasamy K, Kraft AS (2008) Proteasome inhibitor PS-341 (VELCADE) induces stabili-
zation of the TRAIL receptor DR5 mRNA through the 3′-untranslated region. Mol Cancer
Ther 7:1091–1100
88. Kashkar H, Deggerich A, Seeger JM et al (2007) NF-kappaB-independent down-reg-
ulation of XIAP by bortezomib sensitizes HL B cells against cytotoxic drugs. Blood
109:3982–3988
89. Khanbolooki S, Nawrocki ST, Arumugam T et al (2006) Nuclear factor-kappaB maintains
TRAIL resistance in human pancreatic cancer cells. Mol Cancer Ther 5:2251–2260
90. Koschny R, Ganten TM, Sykora J et al (2007) TRAIL/bortezomib cotreatment is potentially
hepatotoxic but induces cancer-specific apoptosis within a therapeutic window. Hepatology
45:649–658
91. Koschny R, Holland H, Sykora J et al (2007) Bortezomib sensitizes primary human astro-
cytoma cells of WHO grades I to IV for tumor necrosis factor-related apoptosis-inducing
ligand-induced apoptosis. Clin Cancer Res 13:3403–3412
92. Lashinger LM, Zhu K, Williams SA et al (2005) Bortezomib abolishes tumor necrosis fac-
tor-related apoptosis-inducing ligand resistance via a p21-dependent mechanism in human
bladder and prostate cancer cells. Cancer Res 65:4902–4908
296 S. Fulda
93. Leverkus M, Sprick MR, Wachter T et al (2003) Proteasome inhibition results in TRAIL
sensitization of primary keratinocytes by removing the resistance-mediating block of effec-
tor caspase maturation. Mol Cell Biol 23:777–790
94. Liu FT, Agrawal SG, Gribben JG et al (2008) Bortezomib blocks Bax degradation in malig-
nant B cells during treatment with TRAIL. Blood 111:2797–2805
95. Liu X, Yue P, Chen S et al (2007) The proteasome inhibitor PS-341 (bortezomib) up-regu-
lates DR5 expression leading to induction of apoptosis and enhancement of TRAIL-induced
apoptosis despite up-regulation of c-FLIP and surviving expression in human NSCLC cells.
Cancer Res 67:4981–4988
96. Nagy K, Szekely-Szuts K, Izeradjene K et al (2006) Proteasome inhibitors sensitize colon
carcinoma cells to TRAIL-induced apoptosis via enhanced release of Smac/DIABLO from
the mitochondria. Pathol Oncol Res 12:133–142
97. Naumann I, Kappler R, Von Schweinitz D et al (2011) Bortezomib primes neuroblastoma
cells for TRAIL-induced apoptosis by linking the death receptor to the mitochondrial path-
way. Clin Cancer Res 17:3204–3218
98. Nikrad M, Johnson T, Puthalalath H et al (2005) The proteasome inhibitor bortezomib sen-
sitizes cells to killing by death receptor ligand TRAIL via BH3-only proteins Bik and Bim.
Mol Cancer Ther 4:443–449
99. Sayers TJ, Brooks AD, Koh CY et al (2003) The proteasome inhibitor PS-341 sensitizes neoplastic
cells to TRAIL-mediated apoptosis by reducing levels of c-FLIP. Blood 102:303–310
100. Shanker A, Brooks AD, Tristan CA et al (2008) Treating metastatic solid tumors with bort-
ezomib and a tumor necrosis factor-related apoptosis-inducing ligand receptor agonist anti-
body. J Natl Cancer Inst 100:649–662
101. Tan TT, Degenhardt K, Nelson DA et al (2005) Key roles of BIM-driven apoptosis in epi-
thelial tumors and rational chemotherapy. Cancer Cell 7:227–238
102. Thorpe JA, Christian PA, Schwarze SR (2008) Proteasome inhibition blocks caspase-8 degrada-
tion and sensitizes prostate cancer cells to death receptor-mediated apoptosis. Prostate 68:200–209
103. Unterkircher T, Cristofanon S, Vellanki SH et al (2011) Bortezomib primes glioblastoma,
including glioblastoma stem cells, for TRAIL by increasing tBid stability and mitochondrial
apoptosis. Clin Cancer Res 17:4019–4030
104. Voortman J, Resende TP, Abou El Hassan MA et al (2007) TRAIL therapy in non-small cell
lung cancer cells: sensitization to death receptor-mediated apoptosis by proteasome inhibi-
tor bortezomib. Mol Cancer Ther 6:2103–2112
105. Yoshida T, Shiraishi T, Nakata S et al (2005) Proteasome inhibitor MG132 induces death recep-
tor 5 through CCAAT/enhancer-binding protein homologous protein. Cancer Res 65:5662–5667
106. Zhao X, Qiu W, Kung J et al (2008) Bortezomib induces caspase-dependent apoptosis in
Hodgkin lymphoma cell lines and is associated with reduced c-FLIP expression: a gene
expression profiling study with implications for potential combination therapies. Leuk Res
32:275–285
107. Zhu H, Guo W, Zhang L et al (2005) Proteasome inhibitors-mediated TRAIL resensitization
and Bik accumulation. Cancer Biol Ther 4:781–786
108. Fulda S (2012) Histone deacetylase (HDAC) inhibitors and regulation of TRAIL-induced
apoptosis. Exp Cell Res 318:1208–1212
109. Butler LM, Liapis V, Bouralexis S et al (2006) The histone deacetylase inhibitor, suber-
oylanilide hydroxamic acid, overcomes resistance of human breast cancer cells to Apo2L/
TRAIL. Int J Cancer 119:944–954
110. Chopin V, Slomianny C, Hondermarck H et al (2004) Synergistic induction of apoptosis in
breast cancer cells by cotreatment with butyrate and TNF-alpha, TRAIL, or anti-FasFas ago-
nist antibody involves enhancement of death receptor’s signaling and requires P21(waf1).
Exp Cell Res 298:560–573
111. Earel JK Jr, Vanoosten RL, Griffith TS (2006) Histone deacetylase inhibitors modulate
the sensitivity of tumor necrosis factor-related apoptosis-inducing ligand-resistant bladder
tumor cells. Cancer Res 66:499–507
11 Harnessing Death Receptor Signaling for Cancer Treatment 297
112. Guo F, Sigua C, Tao J et al (2004) Cotreatment with histone deacetylase inhibitor LAQ824
enhances Apo-2L/tumor necrosis factor-related apoptosis inducing ligand-induced death
inducing signaling complex activity and apoptosis of human acute leukemia cells. Cancer
Res 64:2580–2589
113. Inoue S, Macfarlane M, Harper N et al (2004) Histone deacetylase inhibitors potentiate
TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in lymphoid malignan-
cies. Cell Death Differ 11(Suppl 2):193–206
114. Kim YH, Park JW, Lee JY et al (2004) Sodium butyrate sensitizes TRAIL-mediated apop-
tosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon
cancer cells. Carcinogenesis 25:1813–1820
115. Nakata S, Yoshida T, Horinaka M et al (2004) Histone deacetylase inhibitors upregulate
death receptor 5/TRAIL-R2 and sensitize apoptosis induced by TRAIL/APO2-L in human
malignant tumor cells. Oncogene 23:6261–6271
116. Singh TR, Shankar S, Srivastava RK (2005) HDAC inhibitors enhance the apoptosis-induc-
ing potential of TRAIL in breast carcinoma. Oncogene 24:4609–4623
117. Vanoosten RL, Moore JM, Karacay B et al (2005) Histone deacetylase inhibitors modu-
late renal cell carcinoma sensitivity to TRAIL/Apo-2L-induced apoptosis by enhancing
TRAIL-R2 expression. Cancer Biol Ther 4:1104–1112
118. Vanoosten RL, Moore JM, Ludwig AT et al (2005) Depsipeptide (FR901228) enhances the
cytotoxic activity of TRAIL by redistributing TRAIL receptor to membrane lipid rafts. Mol
Ther 11:542–552
119. Hacker S, Dittrich A, Mohr A et al (2009) Histone deacetylase inhibitors cooperate with
IFN-gamma to restore caspase-8 expression and overcome TRAIL resistance in cancers
with silencing of caspase-8. Oncogene 28:3097–3110
120. Aron JL, Parthun MR, Marcucci G et al (2003) Depsipeptide (FR901228) induces histone
acetylation and inhibition of histone deacetylase in chronic lymphocytic leukemia cells con-
current with activation of caspase 8-mediated apoptosis and down-regulation of c-FLIPc-
FLIP protein. Blood 102:652–658
121. Hernandez A, Thomas R, Smith F et al (2001) Butyrate sensitizes human colon cancer cells
to TRAIL-mediated apoptosis. Surgery 130:265–272
122. Pathil A, Armeanu S, Venturelli S et al (2006) HDAC inhibitor treatment of hepatoma
cells induces both TRAIL-independent apoptosis and restoration of sensitivity to TRAIL.
Hepatology 43:425–434
123. Schuchmann M, Schulze-Bergkamen H, Fleischer B et al (2006) Histone deacetylase
inhibition by valproic acid down-regulates c-FLIP/CASH and sensitizes hepatoma cells
towards CD95- and TRAIL receptor-mediated apoptosis and chemotherapy. Oncol Rep
15:227–230
124. Watanabe K, Okamoto K, Yonehara S (2005) Sensitization of osteosarcoma cells to death
receptor-mediated apoptosis by HDAC inhibitors through downregulation of cellular FLIP.
Cell Death Differ 12:10–18
125. Reddy RM, Yeow WS, Chua A et al (2007) Rapid and profound potentiation of Apo2L/
TRAIL-mediated cytotoxicity and apoptosis in thoracic cancer cells by the histone deacety-
lase inhibitor Trichostatin A: the essential role of the mitochondria-mediated caspase activa-
tion cascade. Apoptosis 12:55–71
126. El-Zawahry A, Lu P, White SJ et al (2006) In vitro efficacy of AdTRAIL gene therapy of
bladder cancer is enhanced by trichostatin A-mediated restoration of CAR expression and
downregulation of cFLIP and Bcl-XLBcl-XL. Cancer Gene Ther 13:281–289
127. Gillespie S, Borrow J, Zhang XD et al (2006) Bim plays a crucial role in synergistic induc-
tion of apoptosis by the histone deacetylase inhibitor SBHA and TRAIL in melanoma cells.
Apoptosis 11:2251–2265
128. Muhlethaler-Mottet A, Flahaut M, Bourloud KB et al (2006) Histone deacetylase inhibitors
strongly sensitise neuroblastoma cells to TRAIL-induced apoptosis by a caspases-dependent
increase of the pro- to anti-apoptotic proteins ratio. BMC Cancer 6:214
298 S. Fulda
148. Das A, Banik NL, Ray SK (2009) Molecular mechanisms of the combination of retinoid and
interferon-gamma for inducing differentiation and increasing apoptosis in human glioblas-
toma T98G and U87MG cells. Neurochem Res 34:87–101
149. De Ambrosis A, Casciano I, Croce M et al (2007) An interferon-sensitive response element
is involved in constitutive caspase-8caspase-8 gene expression in neuroblastoma cells. Int J
Cancer 120:39–47
150. Fulda S, Debatin KM (2002) IFN gamma sensitizes for apoptosis by upregulating caspase-8
expression through the Stat1 pathway. Oncogene 21:2295–2308
151. Johnsen JI, Pettersen I, Ponthan F et al (2004) Synergistic induction of apoptosis in neuro-
blastoma cells using a combination of cytostatic drugs with interferon-gamma and TRAIL.
Int J Oncol 25:1849–1857
152. Kim KB, Choi YH, Kim IK et al (2002) Potentiation of Fas- and TRAIL-mediated apoptosis
by IFN-gamma in A549 lung epithelial cells: enhancement of caspase-8 expression through
IFN-response element. Cytokine 20:283–288
153. Kontny HU, Hammerle K, Klein R et al (2001) Sensitivity of Ewing’s sarcoma to TRAIL-
induced apoptosis. Cell Death Differ 8:506–514
154. Lissat A, Vraetz T, Tsokos M et al (2007) Interferon-gamma sensitizes resistant Ewing’s
sarcoma cells to tumor necrosis factor apoptosis-inducing ligand-induced apoptosis by up-
regulation of caspase-8 without altering chemosensitivity. Am J Pathol 170:1917–1930
155. Meister N, Shalaby T, Von Bueren AO et al (2007) Interferon-gamma mediated up-regula-
tion of caspase-8 sensitizes medulloblastoma cells to radio- and chemotherapy. Eur J Cancer
43:1833–1841
156. Merchant MS, Yang X, Melchionda F et al (2004) Interferon gamma enhances the effective-
ness of tumor necrosis factor-related apoptosis-inducing ligand receptor agonists in a xeno-
graft model of Ewing’s sarcoma. Cancer Res 64:8349–8356
157. Ruiz-Ruiz C, Munoz-Pinedo C, Lopez-Rivas A (2000) Interferon-gamma treatment elevates
caspase-8 expression and sensitizes human breast tumor cells to a death receptor-induced
mitochondria-operated apoptotic program. Cancer Res 60:5673–5680
158. Tekautz TM, Zhu K, Grenet J et al (2006) Evaluation of IFN-gamma effects on apop-
tosis and gene expression in neuroblastoma–preclinical studies. Biochim Biophys Acta
1763:1000–1010
159. Levy DE, Darnell JE Jr (2002) Stats: transcriptional control and biological impact. Nat Rev
Mol Cell Biol 3:651–662
160. Wexler L, Thiele CJ, Mcclure L et al (1992) Adoptive immunotherapy of refractory neuro-
blastoma with tumor-infiltrating lymphocytes, interferon-{gamma}, and interleukin-2. Proc
Ann Meet Am Soc Clin Oncol 11:368
161. Liedtke C, Groger N, Manns MP et al (2006) Interferon-alpha enhances TRAIL-mediated apopto-
sis by up-regulating caspase-8 transcription in human hepatoma cells. J Hepatol 44:342–349
162. Ehrhardt H, Hacker S, Wittmann S et al (2008) Cytotoxic drug-induced, p53-mediated
upregulation of caspase-8 in tumor cells. Oncogene 27:783–793
163. Jiang M, Zhu K, Grenet J et al (2008) Retinoic acid induces caspase-8 transcription via
phospho-CREB and increases apoptotic responses to death stimuli in neuroblastoma cells.
Biochim Biophys Acta 1783:1055–1067
164. Belada D, Smolej L, Stepankova P et al (2010) Diffuse large B-cell lymphoma in a patient
with hyper-IgE syndrome: Successful treatment with risk-adapted rituximab-based immuno-
chemotherapy. Leuk Res 34:232–234
165. Fanale MA, Younes A (2008) Nodular lymphocyte predominant Hodgkin’s lymphoma.
Cancer Treat Res 142:367–381
166. Herbst RS, Eckhardt SG, Kurzrock R et al (2010) Phase I Dose-Escalation study of
recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with
advanced cancer. J Clin Oncol 28:2839–2846
167. Soria JC, Mark Z, Zatloukal P et al (2011) Randomized phase II study of dulanermin in
combination with paclitaxel, carboplatin, and bevacizumab in advanced non-small-cell lung
cancer. J Clin Oncol 29:4442–4451
300 S. Fulda
168. Soria JC, Smit E, Khayat D et al (2010) Phase 1b study of dulanermin (recombinant human
Apo2L/TRAIL) in combination with paclitaxel, carboplatin, and bevacizumab in patients
with advanced non-squamous non-small-cell lung cancer. J Clin Oncol 28:1527–1533
169. Yee YK, Tan VP, Chan P et al (2009) Epidemiology of colorectal cancer in Asia. J
Gastroenterol Hepatol 24:1810–1816
170. Greco FA, Bonomi P, Crawford J et al (2008) Phase 2 study of mapatumumab, a fully
human agonistic monoclonal antibody which targets and activates the TRAIL receptor-1, in
patients with advanced non-small cell lung cancer. Lung Cancer 61:82–90
171. Hotte SJ, Hirte HW, Chen EX et al (2008) A Phase 1 Study of Mapatumumab (Fully Human
Monoclonal Antibody to TRAIL-R1) in Patients with Advanced Solid Malignancies. Clin
Cancer Res 14:3450–3455
172. Leong S, Cohen RB, Gustafson DL et al (2009) Mapatumumab, an antibody targeting
TRAIL-R1, in combination with paclitaxel and carboplatin in patients with advanced solid
malignancies: results of a phase I and pharmacokinetic study. J Clin Oncol 27:4413–4421
173. Mom CH, Verweij J, Oldenhuis CN et al (2009) Mapatumumab, a fully human agonistic
monoclonal antibody that targets TRAIL-R1, in combination with gemcitabine and cispl-
atin: a phase I study. Clin Cancer Res 15:5584–5590
174. Pawel JV, Harvey JH, Spigel DR et al (2010) A randomized phase II trial of mapatumumab,
a TRAIL-R1 agonist monoclonal antibody, in combination with carboplatin and paclitaxel
in patients with advanced NSCLC. J Clin Oncol 28 suppl
175. Sun W, Sohal D, Haller DG et al (2011) Phase 2 trial of bevacizumab, capecitabine, and
oxaliplatin in treatment of advanced hepatocellular carcinoma. Cancer 117:3187–3192
176. Tolcher AW, Mita M, Meropol NJ et al (2007) Phase I pharmacokinetic and biologic cor-
relative study of mapatumumab, a fully human monoclonal antibody with agonist activ-
ity to tumor necrosis factor-related apoptosis-inducing ligand receptor-1. J Clin Oncol
25:1390–1395
177. Trarbach T, Moehler M, Heinemann V et al (2010) Phase II trial of mapatumumab, a fully
human agonistic monoclonal antibody that targets and activates the tumour necrosis factor
apoptosis-inducing ligand receptor-1 (TRAIL-R1), in patients with refractory colorectal
cancer. Br J Cancer 102:506–512
178. Younes A, Vose JM, Zelenetz AD et al (2010) A Phase 1b/2 trial of mapatumumab in
patients with relapsed/refractory non-Hodgkin’s lymphoma. Br J Cancer 103:1783–1787
179. Merchant MS, Chou AJ, Price A et al (2010) Lexatumumab: Results of a phase I trial in
pediatric patients with advanced solid tumors. J Clin Oncol 28:9500 (Meeting Abstracts)
180. Plummer R, Attard G, Pacey S et al (2007) Phase 1 and pharmacokinetic study of lexatu-
mumab in patients with advanced cancers. Clin Cancer Res 13:6187–6194
181. Sikic BI, Wakelee HA, Mehren MV et al (2007) A phase 1b study to assess the safety of lexatu-
mumab, a human monoclonal antibody that activates TRAIL-R2, in 32 combination with gem-
citabine, pemetrexed, doxorubicin or FOLFIRI. J Clin Oncol 25:14006 (Meeting Abstracts)
182. Wakelee HA, Patnaik A, Sikic BI et al (2010) Phase I and pharmacokinetic study of lexa-
tumumab (HGS-ETR2) given every 2 weeks in patients with advanced solid tumors. Ann
Oncol 21:376–381
183. Chawla SP, Tabernero J, Kindler HL et al (2010) Phase I evaluation of the safety
of conatumumab (AMG 655) in combination with AMG 479 in patients (pts) with
advanced, refractory solid tumors. J Clin Oncol 28:3102 (Meeting Abstracts)
184. Doi T, Murakami H, Ohtsu A et al (2011) Phase 1 study of conatumumab, a pro-apoptotic
death receptor 5 agonist antibody, in Japanese patients with advanced solid tumors. Cancer
Chemother Pharmacol 68:733–741
185. Kindler HL, Garbo L, Stephenson J et al (2009) A phase 1b study to evaluate the safety and
efficacy of AMG 655 in combination with gemcitabine (G) in patients (pts) with metastatic
pancreatic cancer (PC). J Clin Oncol 27:4501 (Meeting Abstracts)
186. Kindler HL, Richards DA, Stephenson J et al (2010) A placebo-controlled, randomized phase
34 II study of conatumumab (C) or AMG 479 (A) or placebo (P) plus gemcitabine (G) in
patients (pts) with metastatic pancreatic cancer (mPC). Clin Oncol 28:4035 (Meeting Abstracts)
11 Harnessing Death Receptor Signaling for Cancer Treatment 301
187. Paz-Ares L, Torres JMS, Diaz-Padilla I et al (2009) Safety and efficacy of AMG 655 in 33
combination with paclitaxel and carboplatin (PC) in patients with advanced non-small cell
lung cancer (NSCLC). J Clin Oncol 27:19048 (Meeting Abstracts)
188. Peeters M, Infante P, PLR J et al (2010) Phase Ib/II trial of conatumumab and panitumumab
(pmab) for the treatment (tx) of metastatic colorectal cancer (mCRC): Safety and efficacy,
ASCO Gastrointestinal Cancers Symposium, abstract 443
189. Saltz L, Infante J, Schwartzberg L et al (2009) Safety and efficacy of AMG 655 plus modi-
fied FOLFOX6 (mFOLFOX6) and bevacizumab (B) for the first-line treatment of patients
(pts) with metastatic colorectal cancer (mCRC). J Clin Oncol 27:4079 (Meeting Abstracts)
190. Baron AD, O’bryant CL, Choi Y et al (2011) Phase 1b study of drozitumab combined with
cetuximab (CET) plus irinotecan (IRI) or with FOLFIRI±bevacizumab (BV) in previ-
ously treated patients (pts) with metastatic colorectal cancer (mCRC). Clin Oncol 29:3581
(Meeting Abstracts)
191. Camidge DR, Herbst RS, Gordon MS et al (2010) A phase I safety and pharmacokinetic
study of the death receptor 5 agonistic antibody PRO95780 in patients with advanced malig-
nancies. Clin Cancer Res 16:1256–1263
192. Karapetis CS, Clingan PR, Leighl NB et al (2010) Phase II study of PRO95780 plus pacli-
taxel, carboplatin, and bevacizumab (PCB) in non-small cell lung cancer (NSCLC). J Clin
Oncol 28:7535 (Meeting Abstracts)
193. Rocha CSL, Baranda JC, Wallmark J et al (2011) Phase 1b study of drozitumab combined
with first-line FOLFOX plus bevacizumab (BV) in patients (pts) with metastatic colorectal
cancer (mCRC). J Clin Oncol 29:546 (Meeting Abstracts)
194. Wittebol S, Ferrant A, Wickham NW et al (2010) Phase II study of PRO95780 plus rituxi-
mab in patients with relapsed follicular non-Hodgkin’s lymphoma (NHL). J Clin Oncol
28:18511 (Meeting Abstracts)
195. Forero-Torres A, Shah J, Wood T et al (2010) Phase I trial of weekly tigatuzumab, an ago-
nistic humanized monoclonal antibody targeting death receptor 5 (DR5). Cancer biother
radiopharm 25:13–19
196. Sharma S, Vries EGD, Infante JR et al (2008) Phase I trial of LBY135, a monoclonal anti-
body agonist to DR5, alone and in combination with capecitabine in advanced solid tumors.
J Clin Oncol 26:3538 (Meeting Abstracts)
Chapter 12
Proteasome Inhibition as a Novel Strategy
for Cancer Treatment
12.1 Introduction
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 303
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_12,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
304 M. Shen and Q. P. Dou
role in various cellular events, such as protein quality control, cell cycle progres-
sion, cell differentiation, embryonic development, apoptosis, signal transduction,
gene expression, circadian clocks, as well as immune and inflammatory responses
[1, 2]. In all eukaryotic cells, protein degradation is executed by two major path-
ways, namely the lysosomal pathway, which mainly degrades extracellular and
transmembrane proteins, and the ubiquitin–proteasome pathway (UPP), which
mainly degrades intracellular proteins [2]. Dysregulation of proteasome function
is implicated in several pathological processes, including neoplastic disease and
autoimmune disease which exhibit increased proteasome function. Furthermore,
neurodegenerative disease has been found to be associated with decreased protea-
some function. These properties grant the proteasome potential to be a therapeu-
tic target. Indeed, the first proteasome inhibitor bortezomib was launched in the
United States in 2003 and subsequently in Europe in 2004 for the treatment of
multiple myeloma (MM) and mantle cell lymphoma (MCL) [3].
12.2 Ubiquitin–Proteasome System
The ubiquitin–proteasome system (UPS) was discovered in the late 1970s and
early 1980s. The importance of this discovery was acknowledged through the
award of the 2004 Nobel Prize in Chemistry to its discoverers, Aaron Ciechanover,
Avram Hershko, and Irwin Rose.
12.2.1 Ubiquitin–Proteasome Pathway
12.2.2 Proteasome
AMP+Pi
Ub
Ub
Ub
E1
ATP Repeated Ub
Ub Ub
E2
ubiquitination
Ub Target
Ub
E1 E3
Target
E2
E3
Target ATP
lid base
AMP+Pi
T
A
G
T Ub
R Ub
E Ub
Ub
19S 20S 19S Recycled
26S
Fig. 12.1 A schematic diagram of the UPP and the structure of the 26S proteasome
The 20S core is formed by two identical α rings and two identical β rings stacked
in a symmetric manner with the outside α rings surrounding the inner β rings
(Fig. 12.1). Each α or β ring contains seven different subunits, named α1–α7
or β1–β7, among which only β1, β2, and β5 possess proteolytic activity. The
β1 subunit possesses caspase-like or peptidyl-glutamyl peptide-hydrolyzing-
like (PGPH-like) activity which preferentially cleaves after acidic residues such
as aspartate and glutamate; the β2 subunit possesses trypsin-like activity which
preferentially cleaves after basic residues such as arginine and lysine; and the
β5 subunit possesses chymotrypsin-like activity which preferentially cleaves
after hydrophobic residues such as tyrosine and phenylalanine [3, 6]. The pro-
teasome also exhibits two other enzymatic activities, the “branched chain amino
acid–preferring” (BrAAP) activity and the “small neutral amino acid–prefer-
ring” (SNAAP) activity [7]. The 19S regulatory particle binds to both ends of the
20S core proteasome and is composed of a “lid” and a “base” (Fig. 12.1). The
“lid” contains at least nine non-ATPase subunits, which remove the poly-ubiq-
uitin chain from the substrate by a process called deubiquitylation. The “base”
contains six ATPase and four non-ATPase subunits and opens the gate of the 20S
proteasome, unfolds the substrate proteins, and promotes their entry into the 20S
proteasome [3].
306 M. Shen and Q. P. Dou
In addition to the 19S regulatory particle, the 20S core can alternatively interact
with other regulatory particles at one or both ends, resulting in the formation of
asymmetric or symmetric isoforms of the proteasome. These regulatory particles
include PA28 (also referred to as the 11S regulatory particle) and PA200 [5, 8].
12.2.3 Immunoproteasome
In addition to the constitutive proteasome described thus far, which contains cat-
alytic subunits β1, β2, and β5, there also exists the immunoproteasome, which
harbors different sets of catalytic subunits designated as β1i, β2i, and β5i, corre-
spondingly. Upon cytokine stimulation, especially interferon-γ and tumor necrosis
factor-α, the expression of β1i, β2i, and β5i dramatically increases and coopera-
tively assembles into nascent 20S core particles to form immunoproteasomes.
Interferon-γ stimulation also induces the expression of regulatory subunits PA28α
and PA28β, both of which are non-ATPase subunits. Three PA28α and four PA28β
comprise the heteroheptameric PA28αβ regulatory particle, which facilitates the
assembly of the immunoproteasome and enhances its activity. The immunopro-
teasome is a key component of antigen processing. Compared with the constitu-
tive proteasome, the immunoproteasome possesses enhanced chymotrypsin- and
trypsin-like activities and reduced caspase-like activity. This specialized enzymatic
property endows the immunoproteasome the ability to generate peptides that are
suitable for MHC class I-mediated antigen presentation [9, 10].
12.3 Proteasome Inhibitors
B. Boronates
O O
OH Cl O O OH
O O OH H
H OH H N B
N N B N N B N O
N OH N OH H
H O H O
N O HO O
Cl
Bortezomib CEP-18770 MLN9708
D. -lactones
O O
Cl NH NH
O OH O OH
O O
Marizomib Omuralide
E. Epoxyketones
O O
H H O
N N O O O
N N N H
H H N O
O O O O N N
N H H
S O O
O
Carfilzomib ONX-0912
F. Cyclic peptides
O S
H
HO NH O N O
HO N
NH
O N
O
N O O NH
H O O
HO O NH NH2 H O
O HN N
N
H
O O HN
N
H
O
TMC-95A Argyrin A
Peptide boronates are aldehyde surrogates with improved potency and specific-
ity for the proteasome. For example, MG262 (Z-LLL-Boronate), the boronate surro-
gate of MG132, was shown to be 100-fold more potent than MG132. Besides MG262,
308 M. Shen and Q. P. Dou
Table 12.1 A summary of proteasome inhibitors in clinical use or clinical trials [11, 135]
Binding Route of
Chemical mode and Target selec- administra- Development
Inhibitor Developer nature kinetics tivity tion stage
Bortezomib Millennium Boronate Covalent, β5 > β1 IV/SC FDA
(PS341) slowly approved
reversible
Carfilzomib ONXY Epoxyketone Covalent, β5, β5i IV Phase III
(PR-171) irrevers-
ible
Marizomib Nereus β-lactone Covalent, β5 > β2 > β1 IV Phase I
(NPI- irrevers-
0052) ible
MLN-9708 Millennim Boronate Covalent, β5 > β1 IV/oral Phase I-II
rapidly
reversible
Delanzomib Cephalon Boronate Covalent, β5 > β1 IV/oral Phase I-II
(CEP- slowly
18770) reversible
Oprozomib ONXY Epoxyketone Covalent, β5, β5i Oral Phase I-II
(ONX- irrevers-
0912) ible
IV intravenous; SC subcutaneous; FDA Food and Drug Administration
bortezomib (PS341), delanzomib (CEP-18770), and MLN9708 are the most success-
ful representatives in this class (Fig. 12.2b). Peptide boronates form tetrahedral adducts
with Thr1 in catalytic β subunits, which are further stabilized by two extra hydrogen
bonds. Therefore, although the inhibition is reversible, the dissociation rate is very slow.
Furthermore, unlike peptide aldehydes, peptide boronates have increased bioavailability
and are metabolically stable under physiological conditions. These characteristics make
peptide boronates suitable for therapeutic development. Indeed, bortezomib (Velcade®)
was the first proteasome inhibitor launched in 2003 (Table 12.1). Both delanzomib and
MLN9708 are currently under clinical trials. MLN9708 is sophisticatedly designed as a
prodrug in the form of a boronic ester so that it is orally bioavailable [11, 12].
Peptide vinyl sulfones are irreversible proteasome inhibitors. They were first
described as inhibitors of cysteine proteases. Not surprisingly, they exhibit less
potency and specificity toward the proteasome. Nevertheless, this feature of irre-
versible inhibition offers them certain advantages of being activity-based pro-
teasome probes in basic research. The representatives of this class are ZLVS
(Z-LLL-vs), NLVS (NIP-LLL-vs) (Fig. 12.2c), and AdaAhx3-LLL-vs [3, 11].
A large portion of proteasome inhibitors were initially derived from natural prod-
ucts, and the number of such is still growing. This large family can be further
12 Proteasome Inhibition as a Novel Strategy for Cancer Treatment 309
divided into four groups based on their mechanisms of action: covalent inhibi-
tors, non-covalent inhibitors, non-proteasome-specific inhibitors, and allosteric
inhibitors.
Some natural cyclic peptides have been found to inhibit proteasome func-
tion in a non-covalent manner. The representatives of this class are TMC-95A
(Fig. 12.2f), argyrin A (Fig. 12.2f), and more recently scytonemides [14].
The TMC-95 family is a group of cyclic tripeptides produced by the fungus
Apiospora montagnei. They interact with the active sites of catalytic β subunits
through five hydrogen bonds, thereby reversibly inhibiting proteasome func-
tion [15]. TMC-95A is able to selectively and competitively inhibit all three
310 M. Shen and Q. P. Dou
catalytic β subunits in the low nanomolar range [15]. The shortcoming for the
development of these large inhibitors is that their de novo synthesis is very dif-
ficult and expensive. Argyrin A is a cyclic octapeptide produced by the bacteria
Archangium gephyra. It also reversibly inhibits proteasomal activity in the low
nanomolar range, making itself a promising candidate [16]. However, given its
large size, the exact mechanism by which argyrin A interacts with the protea-
some remains unclear.
Besides the natural proteasome inhibitors discussed above, most of which are
metabolites of bacteria or fungus, a rapidly growing group of natural proteasome
inhibitors has also been isolated from the plant kingdom. These natural products
include some flavonoids, triterpenoids, and others such as curcumin [17] and shi-
konin [18]. Unlike the proteasome inhibitors discussed hitherto that mainly target
the proteasome, plant-derived natural products usually have multiple cellular tar-
gets including the proteasome.
Flavonoids are a group of natural products with a common structure of two
aromatic rings linked by three carbons (C6–C3–C6) [19]. They are abundant in
various vegetables and fruits. Flavonoids that have been found to possess pro-
teasome-inhibitory activity include (-)-epigallocatechin-3-gallate [(-)-EGCG]
(Fig. 12.3a), apigenin, quercetin, and genistein. Among them, (-)-EGCG, the
major catechin in green tea, is most potent in terms of proteasome inhibition.
It inhibits the proteasomal β5 subunit irreversibly through covalent binding.
Notably, (-)-EGCG is able to interact with not only the β5 subunit of the constitu-
tive proteasome but also the β5i subunit of the immunoproteasome with an even
greater affinity [20]. However, (-)-EGCG has been found to have the potential
to decrease the efficacy of bortezomib and other boronate proteasome inhibitors.
This neutralizing effect is mainly due to the direct interaction between the pyro-
catechol moieties on (-)-EGCG and the boronic acid moiety on bortezomib [21].
Whether patients receiving bortezomib should consume green tea needs a clarifi-
cation by further clinical studies.
Triterpenoids are a group of compounds comprised of six isoprene units with
30 carbons. They occur naturally as complex cyclic structures, with the pentacy-
clic structure being most common. Some have been reported to have proteasome-
inhibitory activity, including celastrol [22] (Fig. 12.3b), withaferin A [23], and
pristimerin [24]. These compounds are present in many herbal medicines.
Allosteric inhibitors refer to inhibitors that bind to the allosteric site (a site other than
the enzyme’s active site) of an enzyme and cause its inactivation. The advantage of
12 Proteasome Inhibition as a Novel Strategy for Cancer Treatment 311
A. Flavonoids B. Triterpenoids
OH O
OH OH
HO O
OH
O O
OH OH
O
HO
OH
(-)-EGCG OH
Celastrol
C. Hydroxyquinolines D. Dithiocarbamates
NH2
S
S N
N S
N
S
OH
5AHQ Disulfiram
O O O O
H H
N N N O
O N N N
H H H
O O O
O
O
IPSI-001 ONX-0914
allosteric inhibitors lies in their ability to overcome some particular types of resist-
ance, for example, resistance caused by mutations at the active site. A number of
different metal-binding organic compounds have been shown to be allosteric protea-
some inhibitors in vitro and in cellulo. However, like proteasome inhibitors derived
from plants, most of the organometallic compounds have multiple cellular targets
with the proteasome being one of them.
Two biggest groups of metal-binding compounds that have proteasome-inhibi-
tory activity are hydroxyquinolines [such as 5-amino-8-hydroxyquinoline (5AHQ)
(Fig. 12.3c) and clioquinol] and dithiocarbamates [such as disulfiram (Fig. 12.3d),
diethyldithiocarbamate, and pyrrolidine dithiocarbamate] [25–27]. NMR data indi-
cated that 5AHQ might bind to proteasomal α subunits [27]. Copper is the metal
most commonly used to form complex with these ligands. Other metals that have
been studied include zinc, gold, and gallium.
312 M. Shen and Q. P. Dou
12.3.3 Immunoproteasome-Specific Inhibitors
progression, it is degraded by the proteasome to release CDK2 and CDK4 for G1/S
phase transition, whereas accumulation of p27KIP1 by proteasome inhibition results
in G1 phase arrest [30]. Tambyrajah et al. [31] reported that p27KIP1 is degraded
by the proteasomal β1-mediated activity, which is upregulated as cells enter the
cell cycle without concomitant changes in the protein levels of β1, β2, or β5 subu-
nits. Furthermore, one of the novel proteasome inhibitors discussed above, argyrin
A, was actually discovered in a screen for compounds that prevent degradation of
p27KIP1 [16]. P53 is a bona fide tumor suppressor. As a transcription factor, accu-
mulation of p53 by proteasome inhibition could lead to increased expression of its
target genes such as p21WAF1, another inhibitor of CDK2 [32, 33], and PUMA,
a proapoptotic Bcl-2 family member [34]. However, p53 is not indispensable in
proteasome inhibitor-induced apoptosis, at least in certain types of cancer cells
[35–37].
The transcription factor NF-κB is a critical player against apoptosis in cells and
proteasome function is essential to two proteolytic processes required for the acti-
vation of NF-κB. First, the NF-κB dimer is trapped in the cytoplasm by inhibitor of
NF-κB (IκB) protein. Degradation of IκB by the proteasome is required to release
the NF-κB complex for its nuclear translocation and activation. Inhibition of pro-
teasome function stabilizes IκB and thereby prevents NF-κB activation [38, 39].
Secondly, p105 and p100, two precursors of NF-κB subunits p50 and p52, respec-
tively, need to be processed by the proteasome; otherwise they function like IκB
and confine their dimeric partners to the cytoplasm [39–41]. In addition, it is well
established that conventional cytotoxic drugs as well as inflammatory factors such
as TNFα and IL-1β often activate NF-κB, which is at least partially responsible
for the development of drug resistance. Moreover, constitutive activation of NF-
κB was observed in a large fraction of advanced cancers that are very resistant to
most therapies. Pioneering work done by Delic et al. [42] demonstrated that a low
dose of proteasome inhibitor lactacystin was sufficient to prevent NF-κB activation
and sensitize chemo- and radio-resistant cancer cells to TNFα-induced apoptosis.
Synergistic apoptosis-inducing effects were also observed when combining a pro-
teasome inhibitor with TNF-related apoptosis-inducing ligand (TRAIL) [43, 44],
paclitaxel [45], or etoposide [46]. Therefore, combining a proteasome inhibitor
with other chemotherapeutic agents may represent an effective way to overcome
NF-κB-mediated drug resistance through resensitizing cancer cells to apopto-
sis [47]. However, a study by Hideshima et al. [48] pointed out that under some
experimental conditions, bortezomib actually induced canonical NF-κB activa-
tion in MM cells through phosphorylation of IκB kinase (IKKβ) and its upstream
receptor-interacting protein 2. More intriguingly, Amschler et al. [49] reported that
NF-κB inhibition by either proteasome inhibition or IKKβ blockade sensitized
314 M. Shen and Q. P. Dou
The Bcl-2 family plays a pivotal role in the mitochondrial apoptotic pathway (the
intrinsic pathway). It is comprised of both pro-apoptotic (e.g., Bax, Bak, Bid,
Bim, Bik, NOXA, and PUMA) and anti-apoptotic (e.g., Bcl-2, Bcl-XL, A1, and
Mcl-1) members, the ratio of which is tightly regulated by different factors. The
proteasomal activity influences the level of Bcl-2 family members both directly
and indirectly. Inhibition of proteasome function by different types of inhibitors
was reported to directly induce the accumulation of its substrates Bax [50, 51],
Bik [52], and Bim [53–55]. However, in other studies, conformational change
and mitochondrial translocation, instead of accumulation, of Bax was observed
under proteasome-inhibitory conditions [56, 57]. Proteasome inhibition was
also reported to indirectly induce the transcriptional upregulation of NOXA,
PUMA, and Mcl-1. Although both NOXA and PUMA are target genes of p53,
only upregulation of PUMA, but not NOXA, upon proteasome inhibition was
found to be p53 dependent [34, 36, 51, 58]. A study by Nikiforov et al. [59] sug-
gested that the oncogene c-Myc, another transcription factor, was responsible for
tumor cell-selective upregulation of NOXA in response to proteasome inhibition.
Dysregulation of c-Myc was listed among the most pivotal events by a genome-
wide siRNA screen looking for modulators of cell death induced by bortezomib
[60]. Intriguingly, the anti-apoptotic protein Mcl-1 was also upregulated upon
proteasome inhibition and was found to be mediated by activating transcription
factor-4 (ATF4), an important effector of the unfolded protein response (UPR)
[61]. However, the effect of Mcl-1 was mostly counteracted by the upregulation
of NOXA, which binds to and functionally represses Mcl-1 [62, 63]. Moreover,
regardless of its accumulation, proteasome inhibition caused Mcl-1 cleavage in a
caspase-3-dependent manner [63–65].
(ERAD), both of which help to relieve ER stress. However, prolonged UPR can
eventually lead to apoptosis through the PERK-, ATF4-, and CHOP/GADD153-
mediated pathway or the IRE1- and JNK-mediated pathway [66]. Although the
proteasome does not directly reside in the ER lumen, it participates in the UPR in
multiple levels. First, increased expression of the ER chaperones GRP78/Bip and
GRP94/gp96 was observed upon proteasome inhibition, suggesting activation of
UPR [67]. Secondly, proteasome inhibitor treatment was found to suppress the acti-
vation of IRE1- and XBP1-mediated survival pathways during UPR [68]. Thirdly,
proteasome inhibitors were reported to induce the proapoptotic/terminal UPR medi-
ated by PERK, ATF4, and CHOP/GADD153 [69]. However, the exact mechanism
by which proteasome inhibition induces ER stress remains unclear and debatable.
Generation of reactive oxygen species (ROS) upon proteasome inhibition has been
observed in different cell types including glioma, thyroid cancer, head and neck
squamous cell carcinoma, and non-small cell lung cancer [70–73]. Pre-treating
cells with an antioxidant was able to relieve oxidative stress and suppress apop-
tosis induction by proteasome inhibition [71, 74]. Consistently, Du et al. [72]
reported that high-level intracellular glutathione protected cells from proteasome
inhibition-induced oxidative stress, suggesting that a glutathione-dependent redox
system might play an important role in the sensitivity of certain cells to protea-
some inhibition-induced apoptosis. More intriguingly, Lee et al. [75] reported that
low-dose lactacystin led to increased oxidative/nitrosative protein damage and
lipid peroxidation with little cell death, while high-dose lactacystin further caused
oxidative DNA damage and induced apoptotic cell death. Similar effects were
observed by using another proteasome inhibitor epoxomicin [75]. A comprehen-
sive proteomic and transcriptomic analysis by Bieler et al. [76] further revealed
that low-dose proteasome inhibition induced a transcriptional profile reminiscent
of a physiological stress response that preconditions and protects cells from oxida-
tive stress, while high-dose proteasome inhibition induced massive transcriptional
dysregulation and pronounced oxidative stress, triggering apoptosis.
Bortezomib (Velcade®) is the first and currently the only proteasome inhibitor
approved by the U.S. Food and Drug Administration (FDA). Based on two phase
316 M. Shen and Q. P. Dou
II trials, the CREST trial [77, 78] and the SUMMIT trial [79], bortezomib received
a fast-track approval from the FDA in 2003 for the treatment of patients with
relapsed or refractory MM who had received at least two prior lines of therapy and
progressed on their last therapy. In 2005, bortezomib was promoted by the FDA to
treat patients with MM who had received at least one prior therapy based on the
phase III APEX trial [80, 81]. In 2008, bortezomib successfully became the front-
line therapy for MM patients who are newly diagnosed or previously untreated
based on the phase III VISTA trial [82, 83]. Bortezomib was initially approved for
intravenous injection, but very recently, in 2012, it received FDA approval for sub-
cutaneous administration based on the results from a phase III, non-inferiority trial
conducted in bortezomib-naive patients with relapsed MM [84].
The rational combinations of bortezomib with other chemotherapeutic agents
have been explored. High-dose dexamethasone is a mainstay of therapy for MM.
The combination of bortezomib and dexamethasone was studied in a phase II
trial, where it was used as the first-line treatment in untreated MM patients. The
results suggest that bortezomib with or without dexamethasone is an effective and
well-tolerated induction regimen for the frontline treatment of MM [85, 86]. In
a global phase IIIb trial, the similar regimen was extended to heavily pre-treated
patients with relapsed or refractory MM and proved to be safe and effective in
these patients as well [87]. Adding a third drug to the combination of bortezomib
and dexamethasone has also been studied. For example, a combination of bort-
ezomib, doxorubicin, and dexamethasone used as induction therapy before stem-
cell transplantation induced >90 % of responses in newly diagnosed MM patients
with well-tolerated and manageable toxicities [88, 89]. Likewise, a combination
of bortezomib, thalidomide, and dexamethasone also achieved favorable outcome
when used as induction regimen in newly diagnosed MM patients [90].
For patients ineligible for stem-cell transplantation, melphalan plus prednisone
has long been the standard treatment with response rates around 50 % and median
survival around 3 years [91, 92]. The previously mentioned pivotal VISTA trial
compared the use of melphalan and prednisone with or without bortezomib in
untreated MM patients who were ineligible for high-dose therapy and eventually
supported the approval of bortezomib as frontline therapy in these patients. In
this trial, adding bortezomib to melphalan and prednisone (VMP) achieved much
higher complete response (CR) rate and partial response (PR) rate as well as sig-
nificantly prolonged overall survival [82, 83]. Replacing melphalan with thalido-
mide in this triple-drug regimen resulted in comparable efficacy but more serious
adverse events and discontinuations, suggesting that the VMP regimen should
remain the upfront treatment for untreated MM patients who are ineligible for
stem-cell transplantation [93]. Rather than replacing melphalan with thalidomide,
adding thalidomide to the VMP regimen as a fourth drug was found to be superior
to VMP alone [94, 95].
In 2006, the FDA extended the application of bortezomib to the treatment
of mantle cell lymphoma (MCL) based on the results from the pivotal phase II
PINNACLE trial where 155 patients with relapsed or refractory MCL received
bortezomib monotherapy [96, 97]. The overall response rate (ORR) was 32 %.
12 Proteasome Inhibition as a Novel Strategy for Cancer Treatment 317
The median time to progression and overall survival time in responders were much
longer than those in the entire patient population, suggesting that single-agent
bortezomib therapy is associated with lengthy responses and notable survival in
relapsed or refractory MCL patients. In another phase II trial, the combination of
bortezomib and gemcitabine was investigated and found to be active in patients
with relapsed or refractory MCL with the ORR being 60 %. This regimen also
offers a chemotherapy backbone to which other less myelosuppressive agents may
be added [98].
Dozens of clinical trials are currently undergoing in order to further expand the
application of bortezomib to other hematological malignancies such as different
types of B-cell lymphoma as well as some advanced-stage solid tumors [99].
The application of bortezomib in different B-cell lymphomas has been
explored. Besides MCL, which accounts for 5 % of all B-cell lymphomas, two
major types of B-cell lymphomas are diffuse large B-cell lymphoma and follicu-
lar lymphoma, accounting for 30–40 and 20 %, respectively [100]. In a phase II
trial, a regimen of weekly or twice-weekly bortezomib plus rituximab was evalu-
ated in patients with relapsed or refractory follicular or marginal-zone B-cell lym-
phoma [101]. Based on the encouraging results, the regimen of weekly bortezomib
plus rituximab was further evaluated in patients with relapsed follicular lymphoma
in a phase III trial. However, comparing with rituximab alone, the improvement
elicited by adding bortezomib was not as great as expected, suggesting that this
regimen might be useful for only some subgroups of patients [102]. In the phase
II VERTICAL trial, a combination of bortezomib, bendamustine, and rituximab
was evaluated and proved to be highly active (ORR of 88 % and CR of 53 %)
in patients with relapsed or refractory follicular lymphoma [103]. In a phase I/II
study, bortezomib was introduced to the standard CHOP (cyclophosphamide, dox-
orubicin, vincristine, and prednisone) plus rituximab regimen (R-CHOP). In dif-
fuse large B-cell lymphoma patients, the evaluable ORR was 100 % with 86 %
CR/unconfirmed CR. The numbers were 91 and 72 % in MCL patients, respec-
tively. These results strongly support the bortezomib plus R-CHOP regimen to
enter phase III trial [104].
The application of bortezomib in solid tumors has also been extensively inves-
tigated. Unfortunately, in contrast to its potent efficacy in hematological malignan-
cies, the efficacy of bortezomib in solid tumors is disappointing. In patients with
castration resistant metastatic prostate cancer, neither bortezomib alone nor bort-
ezomib plus prednisone, exhibited significant antitumor effects [105]. Similarly,
docetaxel plus bortezomib used as first-line treatment showed no improved effi-
cacy versus docetaxel alone [106]. In heavily pre-treated metastatic breast cancer
patients, bortezomib plus pegylated liposomal doxorubicin was well tolerated but
318 M. Shen and Q. P. Dou
had minimal activity [107]. A trial testing the bortezomib monotherapy in chemo-
therapy-naïve patients with advanced-stage non-small cell lung cancer was termi-
nated in the first stage due to lack of response in all patients [108]. The bortezomib
monotherapy was inactive in patients with unresectable or metastatic gastric and
gastroesophageal junction adenocarcinoma, either [109]. A first-line regimen of
bortezomib, paclitaxel, and carboplatin also failed in the treatment of patients with
metastatic esophageal, gastric, and gastroesophageal cancer [110].
Therefore, continuous efforts are required both on the benchtop and at the
bedside. A mechanistic explanation regarding the failure of bortezomib in solid
tumors will help in designing a rational regimen and eventually lead to clinical
success.
12.6.1 Advantages of Bortezomib
clinical trials, bortezomib was introduced into the standard therapy and largely
improved the outcome in terms of ORR, time to progression, etc. The chemo- and
radio-sensitizing effect of bortezomib could also be attributed to its unique target
and mechanism of action. It is conceivable that two drugs with different targets or
mechanisms of action may have better efficacy and less toxicity than two drugs
with the same target or similar mechanism of action [115, 116].
12.6.2 Disadvantages of Bortezomib
At least five second generation proteasome inhibitors are currently under clinical
investigation (Table 12.1).
Carfilzomib is an irreversible proteasome inhibitor, with new protein syn-
thesis being required for recovery of proteasome activity, which gives it greater
potency. Whether it has decreased side effects compared with bortezomib needs
to be determined. In addition to targeting the β5 subunit in the constitutive pro-
teasome, carfilmozib also targets the correlated β5i subunit in the immunoprotea-
some, which appears to be preferentially expressed in MM. Moreover, carfilzomib
is shown to be more specific than borzetomib, with little or no off-target activity
outside of the proteasome [123]. However, like bortezomib, the administration of
carfilzomib is intravenous twice weekly, which is inconvenient for patients.
Carfilzomib has been evaluated in two phase II trials, PX-171-003-A1 [124]
and PX-171-004 [125, 126], as monotherapy for the treatment of relapsed and
refractory MM patients. Both trials observed durable response to carfilzomib
with well-tolerated and manageable side effects, regardless of prior exposure to
bortezomib. Analysis of 136 patients in the above two trials indicated that periph-
eral neuropathy occurred in 15 % of patients, among which 9 % was attributed to
carfilzomib. None of the patients required discontinuation or dose adjustments due
to neurotoxicity, allowing long-term treatment and prolonged disease control by
carfilzomib [127]. These data also indicate that peripheral neuropathy is not a class
effect of proteasome inhibitors [127]. A recent study comparing bortezomib and
carfilzomib further pointed out that the neurotoxicity of bortezomib was related
to the off-target inhibition of HtrA2/Omi by bortezomib, a factor known to be
involved in neuronal survival [128].
Carfilzomib has also been evaluated in combination with lenalidomide, and
low-dose dexamethasone (CRd) in both relapsed and/or refractory MM patients
[129] and newly diagnosed MM patients [130] in two separate phase II trials.
CRd regimen is highly active and well tolerated in both trials. In newly diagnosed
patients, the responses are rapid and improve over time reaching 100 % very good
PR or better, which compare favorably with the best frontline regimens in MM
therapy [130]. In relapsed and/or refractory patients, the ORR was 78 % [129].
Based on these encouraging results, the phase III ASPIRE trial of this regimen is
underway for previously bortezomib-treated MM patients.
12 Proteasome Inhibition as a Novel Strategy for Cancer Treatment 321
12.7 Conclusion
The proteasome has been shown to be a validated, novel and valuable target for
cancer treatment. Suppression of proteasome function by synthetic or natural com-
pounds is proven to be an effective way to induce cancer cell death with minimal
effects on normal healthy cells. The first FDA-approved proteasome inhibitor bort-
ezomib has achieved great success in the treatment of MM and MCL. However,
some limitations and side effects have been noted during clinical application, such
as the occurrence of bortezomib resistance, dose-limiting toxicities, and interac-
tion with some natural compounds, all of which compromise the clinical benefits
of bortezomib. Furthermore, the efficaciousness of bortezomib in solid tumors
is disappointing. Therefore, the development of novel, new-generation protea-
some inhibitors is necessary to improve the efficacy, reduce toxicity, and expand
322 M. Shen and Q. P. Dou
Acknowledgments The authors thank Sara Schmitt and Daniela Buac for critical reading of the
manuscript. This work was partially supported by the National Cancer Institute (1R01CA120009,
3R01CA120009-04S1, and 5R01CA127258-05, to QPD).
References
1. Hershko A (2005) The ubiquitin system for protein degradation and some of its roles in the
control of the cell division cycle. Cell Death Differ 12(9):1191–1197
2. Spataro V, Norbury C, Harris AL (1998) The ubiquitin-proteasome pathway in cancer. Br
J Cancer 77(3):448–455
3. de Bettignies G, Coux O (2010) Proteasome inhibitors: Dozens of molecules and still count-
ing. Biochimie 92(11):1530–1545
4. Adams J (2004) The development of proteasome inhibitors as anticancer drugs. Cancer Cell
5(5):417–421
5. Sorokin AV, Kim ER, Ovchinnikov LP (2009) Proteasome system of protein degradation
and processing. Biochemistry (Mosc) 74(13):1411–1442
6. Adams J (2004) The proteasome: a suitable antineoplastic target. Nat Rev Cancer
4(5):349–360
7. Orlowski M, Cardozo C, Michaud C (1993) Evidence for the presence of five distinct pro-
teolytic components in the pituitary multicatalytic proteinase complex. Properties of two
components cleaving bonds on the carboxyl side of branched chain and small neutral amino
acids. Biochemistry 32(6):1563–1572
8. Nandi D, Tahiliani P, Kumar A, Chandu D (2006) The ubiquitin-proteasome system.
J Biosci 31(1):137–155
9. Strehl B, Seifert U, Kruger E, Heink S, Kuckelkorn U, Kloetzel PM (2005) Interferon-
gamma, the functional plasticity of the ubiquitin-proteasome system, and MHC class I anti-
gen processing. Immunol Rev 207:19–30
10. Angeles A, Fung G, Luo H (2012) Immune and non-immune functions of the immunopro-
teasome. Front Biosci 17:1904–1916
11. Kisselev AF, van der Linden WA, Overkleeft HS (2012) Proteasome inhibitors: an expand-
ing army attacking a unique target. Chem Biol 19(1):99–115
12. Borissenko L, Groll M (2007) 20S proteasome and its inhibitors: crystallographic knowl-
edge for drug development. Chem Rev 107(3):687–717
13. Tsukamoto S, Yokosawa H (2010) Inhibition of the ubiquitin-proteasome system by natural
products for cancer therapy. Planta Med 76(11):1064–1074
14. Krunic A, Vallat A, Mo S, Lantvit DD, Swanson SM, Orjala J (2010) Scytonemides A and
B, cyclic peptides with 20S proteasome inhibitory activity from the cultured cyanobacte-
rium Scytonema hofmanii. J Nat Prod 73(11):1927–1932
15. Koguchi Y, Kohno J, Nishio M, Takahashi K, Okuda T, Ohnuki T et al (2000) TMC-95A, B,
C, and D, novel proteasome inhibitors produced by Apiospora montagnei Sacc. TC 1093.
Taxonomy, production, isolation, and biological activities. J Antibiot 53(2):105–109
12 Proteasome Inhibition as a Novel Strategy for Cancer Treatment 323
34. Concannon CG, Koehler BF, Reimertz C, Murphy BM, Bonner C, Thurow N et al (2007)
Apoptosis induced by proteasome inhibition in cancer cells: predominant role of the p53/
PUMA pathway. Oncogene 26(12):1681–1692
35. Strauss SJ, Higginbottom K, Juliger S, Maharaj L, Allen P, Schenkein D et al (2007) The
proteasome inhibitor bortezomib acts independently of p53 and induces cell death via apop-
tosis and mitotic catastrophe in B-cell lymphoma cell lines. Cancer Res 67(6):2783–2790
36. Pandit B, Gartel AL (2011) Proteasome inhibitors induce p53-independent apoptosis in
human cancer cells. Am J Pathol 178(1):355–360
37. Wagenknecht B, Hermisson M, Eitel K, Weller M (1999) Proteasome inhibitors induce p53/
p21-independent apoptosis in human glioma cells. Cell Physiol Biochem 9(3):117–125
38. Traenckner EB, Wilk S, Baeuerle PA (1994) A proteasome inhibitor prevents activation
of NF-kappa B and stabilizes a newly phosphorylated form of I kappa B-alpha that is still
bound to NF-kappa B. EMBO J 13(22):5433–5441
39. Palombella VJ, Rando OJ, Goldberg AL, Maniatis T (1994) The ubiquitin-proteasome path-
way is required for processing the NF-kappa B1 precursor protein and the activation of NF-
kappa B. Cell 78(5):773–785
40. Sears C, Olesen J, Rubin D, Finley D, Maniatis T (1998) NF-kappa B p105 processing via
the ubiquitin-proteasome pathway. J Biol Chem 273(3):1409–1419
41. Heusch M, Lin L, Geleziunas R, Greene WC (1999) The generation of nfkb2 p52: mecha-
nism and efficiency. Oncogene 18(46):6201–6208
42. Delic J, Masdehors P, Omura S, Cosset JM, Dumont J, Binet JL et al (1998) The protea-
some inhibitor lactacystin induces apoptosis and sensitizes chemo- and radioresistant human
chronic lymphocytic leukaemia lymphocytes to TNF-alpha-initiated apoptosis. Br J Cancer
77(7):1103–1107
43. Voortman J, Resende TP (2007) Abou El Hassan MA, Giaccone G, Kruyt FA. TRAIL ther-
apy in non-small cell lung cancer cells: sensitization to death receptor-mediated apoptosis
by proteasome inhibitor bortezomib. Mol Cancer Ther 6(7):2103–2112
44. Nencioni A, Wille L, Dal Bello G, Boy D, Cirmena G, Wesselborg S et al (2005) Cooperative cyto-
toxicity of proteasome inhibitors and tumor necrosis factor-related apoptosis-inducing ligand in
chemoresistant Bcl-2-overexpressing cells. Clin Cancer Res 11(11):4259–4265
45. Oyaizu H, Adachi Y, Okumura T, Okigaki M, Oyaizu N, Taketani S et al (2001) Proteasome
inhibitor 1 enhances paclitaxel-induced apoptosis in human lung adenocarcinoma cell line.
Oncol Rep 8(4):825–829
46. von Metzler I, Heider U, Mieth M, Lamottke B, Kaiser M, Jakob C et al (2009) Synergistic
interaction of proteasome and topoisomerase II inhibition in multiple myeloma. Exp Cell
Res 315(14):2471–2478
47. Mitsiades N, Mitsiades CS, Richardson PG, Poulaki V, Tai YT, Chauhan D et al (2003) The
proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conven-
tional chemotherapeutic agents: therapeutic applications. Blood 101(6):2377–2380
48. Hideshima T, Ikeda H, Chauhan D, Okawa Y, Raje N, Podar K et al (2009) Bortezomib
induces canonical nuclear factor-kappaB activation in multiple myeloma cells. Blood
114(5):1046–1052
49. Amschler K, Schon MP, Pletz N, Wallbrecht K, Erpenbeck L, Schon M (2010) NF-kappaB
inhibition through proteasome inhibition or IKKbeta blockade increases the susceptibil-
ity of melanoma cells to cytostatic treatment through distinct pathways. J Invest Dermatol.
130(4):1073–1086
50. Li B, Dou QP (2000) Bax degradation by the ubiquitin/proteasome-dependent path-
way: involvement in tumor survival and progression. Proc Natl Acad Sci U S A
97(8):3850–3855
51. Ding WX, Ni HM, Chen X, Yu J, Zhang L, Yin XM (2007) A coordinated action of Bax,
PUMA, and p53 promotes MG132-induced mitochondria activation and apoptosis in colon
cancer cells. Mol Cancer Ther 6(3):1062–1069
12 Proteasome Inhibition as a Novel Strategy for Cancer Treatment 325
70. Fan WH, Hou Y, Meng FK, Wang XF, Luo YN, Ge PF (2011) Proteasome inhibitor MG-132
induces C6 glioma cell apoptosis via oxidative stress. Acta Pharmacol Sin 32(5):619–625
71. Ling YH, Liebes L, Zou Y, Perez-Soler R (2003) Reactive oxygen species genera-
tion and mitochondrial dysfunction in the apoptotic response to Bortezomib, a novel
proteasome inhibitor, in human H460 non-small cell lung cancer cells. J Biol Chem
278(36):33714–33723
72. Du ZX, Zhang HY, Meng X, Guan Y, Wang HQ (2009) Role of oxidative stress and intracel-
lular glutathione in the sensitivity to apoptosis induced by proteasome inhibitor in thyroid
cancer cells. BMC Cancer 9:56
73. Fribley A, Zeng Q, Wang CY (2004) Proteasome inhibitor PS-341 induces apoptosis
through induction of endoplasmic reticulum stress-reactive oxygen species in head and neck
squamous cell carcinoma cells. Mol Cell Biol 24(22):9695–9704
74. Papa L, Gomes E, Rockwell P (2007) Reactive oxygen species induced by proteasome inhi-
bition in neuronal cells mediate mitochondrial dysfunction and a caspase-independent cell
death. Apoptosis 12(8):1389–1405
75. Lee MH, Hyun DH, Jenner P, Halliwell B (2001) Effect of proteasome inhibition on cel-
lular oxidative damage, antioxidant defences and nitric oxide production. J Neurochem
78(1):32–41
76. Bieler S, Meiners S, Stangl V, Pohl T, Stangl K (2009) Comprehensive proteomic and tran-
scriptomic analysis reveals early induction of a protective anti-oxidative stress response by
low-dose proteasome inhibition. Proteomics 9(12):3257–3267
77. Jagannath S, Barlogie B, Berenson J, Siegel D, Irwin D, Richardson PG et al (2004) A
phase 2 study of two doses of bortezomib in relapsed or refractory myeloma. Br J Haematol
127(2):165–172
78. Jagannath S, Barlogie B, Berenson JR, Siegel DS, Irwin D, Richardson PG et al
(2008) Updated survival analyses after prolonged follow-up of the phase 2, multicenter
CREST study of bortezomib in relapsed or refractory multiple myeloma. Br J Haematol
143(4):537–540
79. Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D et al (2003)
A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med
348(26):2609–2617
80. Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T et al (2005)
Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med
352(24):2487–2498
81. Richardson PG, Sonneveld P, Schuster M, Irwin D, Stadtmauer E, Facon T et al (2007)
Extended follow-up of a phase 3 trial in relapsed multiple myeloma: final time-to-event
results of the APEX trial. Blood 110(10):3557–3560
82. San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M et al
(2008) Bortezomib plus melphalan and prednisone for initial treatment of multiple mye-
loma. N Engl J Med 359(9):906–917
83. Mateos MV, Richardson PG, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O
et al (2010) Bortezomib plus melphalan and prednisone compared with melphalan and pred-
nisone in previously untreated multiple myeloma: updated follow-up and impact of subse-
quent therapy in the phase III VISTA trial. J Clin Oncol 28(13):2259–2266
84. Moreau P, Pylypenko H, Grosicki S, Karamanesht I, Leleu X, Grishunina M et al (2011)
Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple
myeloma: a randomised, phase 3, non-inferiority study. Lancet Oncol 12(5):431–440
85. Jagannath S, Durie BG, Wolf J, Camacho E, Irwin D, Lutzky J et al (2005) Bortezomib ther-
apy alone and in combination with dexamethasone for previously untreated symptomatic
multiple myeloma. Br J Haematol 129(6):776–783
86. Jagannath S, Durie BG, Wolf JL, Camacho ES, Irwin D, Lutzky J et al (2009) Extended
follow-up of a phase 2 trial of bortezomib alone and in combination with dexamethasone for
the frontline treatment of multiple myeloma. Br J Haematol 146(6):619–626
12 Proteasome Inhibition as a Novel Strategy for Cancer Treatment 327
87. Mikhael JR, Belch AR, Prince HM, Lucio MN, Maiolino A, Corso A et al (2009) High
response rate to bortezomib with or without dexamethasone in patients with relapsed or
refractory multiple myeloma: results of a global phase 3b expanded access program. Br
J Haematol 144(2):169–175
88. Oakervee HE, Popat R, Curry N, Smith P, Morris C, Drake M et al (2005) PAD combina-
tion therapy (PS-341/bortezomib, doxorubicin and dexamethasone) for previously untreated
patients with multiple myeloma. Br J Haematol 129(6):755–762
89. Popat R, Oakervee HE, Hallam S, Curry N, Odeh L, Foot N et al (2008) Bortezomib, doxo-
rubicin and dexamethasone (PAD) front-line treatment of multiple myeloma: updated results
after long-term follow-up. Br J Haematol 141(4):512–516
90. Cavo M, Tacchetti P, Patriarca F, Petrucci MT, Pantani L, Galli M et al (2010) Bortezomib
with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone
as induction therapy before, and consolidation therapy after, double autologous stem-cell
transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet
376(9758):2075–2085
91. Driscoll JJ, Burris J, Annunziata CM (2012) Targeting the proteasome with bortezomib in
multiple myeloma: update on therapeutic benefit as an upfront single agent, induction regi-
men for stem-cell transplantation and as maintenance therapy. Am J Ther 19(2):133–144
92. Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC (2009) Multiple myeloma.
Lancet 374(9686):324–339
93. Mateos MV, Oriol A, Martinez-Lopez J, Gutierrez N, Teruel AI, de Paz R et al (2010)
Bortezomib, melphalan, and prednisone versus bortezomib, thalidomide, and prednisone
as induction therapy followed by maintenance treatment with bortezomib and thalidomide
versus bortezomib and prednisone in elderly patients with untreated multiple myeloma: a
randomised trial. Lancet Oncol 11(10):934–941
94. Palumbo A, Ambrosini MT, Benevolo G, Pregno P, Pescosta N, Callea V et al (2007)
Bortezomib, melphalan, prednisone, and thalidomide for relapsed multiple myeloma. Blood
109(7):2767–2772
95. Palumbo A, Bringhen S, Rossi D, Cavalli M, Larocca A, Ria R et al (2010) Bortezomib-
melphalan-prednisone-thalidomide followed by maintenance with bortezomib-thalidomide
compared with bortezomib-melphalan-prednisone for initial treatment of multiple myeloma:
a randomized controlled trial. J Clin Oncol 28(34):5101–5109
96. Fisher RI, Bernstein SH, Kahl BS, Djulbegovic B, Robertson MJ, de Vos S et al (2006)
Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell
lymphoma. J Clin Oncol 24(30):4867–4874
97. Goy A, Bernstein SH, Kahl BS, Djulbegovic B, Robertson MJ, de Vos S et al (2009)
Bortezomib in patients with relapsed or refractory mantle cell lymphoma: updated time-to-
event analyses of the multicenter phase 2 PINNACLE study. Ann Oncol 20(3):520–525
98. Kouroukis CT, Fernandez LA, Crump M, Gascoyne RD, Chua NS, Buckstein R et al (2011)
A phase II study of bortezomib and gemcitabine in relapsed mantle cell lymphoma from the
National Cancer Institute of Canada Clinical Trials Group (IND 172). Leukemia lymphoma
52(3):394–399
99. Yang H, Zonder JA, Dou QP (2009) Clinical development of novel proteasome inhibitors
for cancer treatment. Expert Opin Investig Drugs 18(7):957–971
100. Kuppers R (2005) Mechanisms of B-cell lymphoma pathogenesis. Nat Rev Cancer 5(4):251–262
101. de Vos S, Goy A, Dakhil SR, Saleh MN, McLaughlin P, Belt R et al (2009) Multicenter
randomized phase II study of weekly or twice-weekly bortezomib plus rituximab in patients
with relapsed or refractory follicular or marginal-zone B-cell lymphoma. J Clin Oncol
27(30):5023–5030
102. Coiffier B, Osmanov EA, Hong X, Scheliga A, Mayer J, Offner F et al (2011) Bortezomib
plus rituximab versus rituximab alone in patients with relapsed, rituximab-naive or
rituximab-sensitive, follicular lymphoma: a randomised phase 3 trial. Lancet Oncol.
12(8):773–784
328 M. Shen and Q. P. Dou
103. Fowler N, Kahl BS, Lee P, Matous JV, Cashen AF, Jacobs SA et al (2011) Bortezomib, ben-
damustine, and rituximab in patients with relapsed or refractory follicular lymphoma: the
phase II VERTICAL study. J Clin Oncol 29(25):3389–3395
104. Ruan J, Martin P, Furman RR, Lee SM, Cheung K, Vose JM et al (2011) Bortezomib plus
CHOP-rituximab for previously untreated diffuse large B-cell lymphoma and mantle cell
lymphoma. J Clin Oncol 29(6):690–697
105. Morris MJ, Kelly WK, Slovin S, Ryan C, Eicher C, Heller G et al (2007) A phase II trial
of bortezomib and prednisone for castration resistant metastatic prostate cancer. J Urology
178(6):2378–2383, discussion 83-4
106. Hainsworth JD, Meluch AA, Spigel DR, Barton J Jr, Simons L, Meng C et al (2007) Weekly
docetaxel and bortezomib as first-line treatment for patients with hormone-refractory pros-
tate cancer: a Minnie Pearl Cancer Research Network phase II trial. Clin Genitourinary
Cancer 5(4):278–283
107. Irvin WJ Jr, Orlowski RZ, Chiu WK, Carey LA, Collichio FA, Bernard PS et al (2010)
Phase II study of bortezomib and pegylated liposomal doxorubicin in the treatment of meta-
static breast cancer. Clin Breast Cancer 10(6):465–470
108. Li T, Ho L, Piperdi B, Elrafei T, Camacho FJ, Rigas JR et al (2010) Phase II study of the
proteasome inhibitor bortezomib (PS-341, Velcade) in chemotherapy-naive patients with
advanced stage non-small cell lung cancer (NSCLC). Lung Cancer 68(1):89–93
109. Shah MA, Power DG, Kindler HL, Holen KD, Kemeny MM, Ilson DH et al (2011) A mul-
ticenter, phase II study of bortezomib (PS-341) in patients with unresectable or metastatic
gastric and gastroesophageal junction adenocarcinoma. Invest New Drugs 29(6):1475–1481
110. Jatoi A, Dakhil SR, Foster NR, Ma C, Rowland KM Jr, Moore DF Jr et al (2008)
Bortezomib, paclitaxel, and carboplatin as a first-line regimen for patients with metastatic
esophageal, gastric, and gastroesophageal cancer: phase II results from the North Central
Cancer Treatment Group (N044B). J Thoracic Oncol 3(5):516–520
111. Pleban E, Bury M, Mlynarczuk I, Wojcik C (2001) Effects of proteasome inhibitor PSI on
neoplastic and non-transformed cell lines. Folia Histochem Cytobiol 39(2):133–134
112. Adams J (2003) Potential for proteasome inhibition in the treatment of cancer. Drug Discov
Today 8(7):307–315
113. Chen D, Frezza M, Schmitt S, Kanwar J, Dou QP (2011) Bortezomib as the first proteasome
inhibitor anticancer drug: current status and future perspectives. Curr Cancer Drug Targets
11(3):239–253
114. Shah JJ, Orlowski RZ (2009) Proteasome inhibitors in the treatment of multiple myeloma.
Leukemia 23(11):1964–1979
115. Orlowski RZ (2004) Bortezomib in combination with other therapies for the treatment of
multiple myeloma. J Natl Compr Canc Netw (Suppl 4):S16–S20
116. Reddy N, Czuczman MS (2010) Enhancing activity and overcoming chemoresistance in
hematologic malignancies with bortezomib: preclinical mechanistic studies. Ann Oncol
21(9):1756–1764
117. Appel A (2011) Drugs: More shots on target. Nature 480(7377):S40–S42
118. Lu S, Chen Z, Yang J, Chen L, Gong S, Zhou H et al (2008) Overexpression of the PSMB5
gene contributes to bortezomib resistance in T-lymphoblastic lymphoma/leukemia cells
derived from Jurkat line. Exp Hematol 36(10):1278–1284
119. Shuqing L, Jianmin Y, Chongmei H, Hui C, Wang J (2011) Upregulated expression of the
PSMB5 gene may contribute to drug resistance in patient with multiple myeloma when
treated with bortezomib-based regimen. Exp Hematol 39(12):1117–1118
120. Oerlemans R, Franke NE, Assaraf YG, Cloos J, van Zantwijk I, Berkers CR et al (2008)
Molecular basis of bortezomib resistance: proteasome subunit beta5 (PSMB5) gene muta-
tion and overexpression of PSMB5 protein. Blood 112(6):2489–2499
121. Franke NE, Niewerth D, Assaraf YG, van Meerloo J, Vojtekova K, van Zantwijk CH et al
(2011) Impaired bortezomib binding to mutant beta5 subunit of the proteasome is the under-
lying basis for bortezomib resistance in leukemia cells. Leukemia 26(4):757–768
12 Proteasome Inhibition as a Novel Strategy for Cancer Treatment 329
122. Markovina S, Callander NS, O’Connor SL, Kim J, Werndli JE, Raschko M et al (2008)
Bortezomib-resistant nuclear factor-kappaB activity in multiple myeloma cells. Mol Cancer
Res 6(8):1356–1364
123. Ruschak AM, Slassi M, Kay LE, Schimmer AD (2011) Novel proteasome inhibitors to over-
come bortezomib resistance. J Natl Cancer Inst 103(13):1007–1017
124. Samuel D, Martin T, Wang M, Vij R, Jakubowiak AJ, Jagannath S et al (2010) Results
of PX-171-003-A1, An Open-Label, Single-Arm, Phase 2 (Ph 2) Study of Carfilzomib
(CFZ) In Patients (pts) with Relapsed and Refractory Multiple Myeloma (MM). Blood
116(21):433
125. Vii R, Kaufman JL, Jakubowiak AJ, Wang M, Jagannath S, Kukreti V et al (2011) Final
results from the bortezomib-naive group of PX-171-004, a phase 2 study of single-agent
carfilzomib in patients with relapsed and/or refractory MM. Blood 118(21):369–370
126. Stewart K, Siegel D, Wang M, Kaufman J, Jakubowiak A, Jagannath S et al (2010) Results
of Px-171-004, an ongoing open-label, phase ii study of carfilzomib in patients with
relapsed and/or refractory multiple myeloma (R/R Mm) with or without prior bortezomib
exposure. Haematologica 95:452
127. Vij R, Wang LH, Orlowski RZ, Stewart AK, Jagannath S, Lonial S et al (2009) Carfilzomib
(CFZ), a novel proteasome inhibitor for relapsed or refractory multiple myeloma, is associ-
ated with minimal peripheral neuropathic effects. Blood 114(22):178–179
128. Arastu-Kapur S, Anderl JL, Kraus M, Parlati F, Shenk KD, Lee SJ et al (2011)
Nonproteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: a link to
clinical adverse events. Clin Cancer Res 17(9):2734–2743
129. Wang M, Bensinger W, Martin T, Alsina M (2011) Interim results from PX-171-006, a
phase (Ph) II multicenter dose-expansion study of carfilzomib (CFZ), lenalidomide (LEN),
and low-dose dexamethasone (loDex) in relapsed and/or refractory multiple myeloma (R/R
MM). J Clin Oncol 2011(suppl):abstr 8025
130. Jakubowiak AJ, Dytfeld D, Jagannath S, Vesole DH, Anderson TB, Nordgren BK et al
(2011) Final results of a frontline phase 1/2 Study of carfilzomib, lenalidomide, and low-
dose dexamethasone (CRd) in multiple myeloma (MM). Blood 118(21):288–289
131. Richardson PG, Spencer A, Cannel P, Harrison SJ, Catley L, Underhill C et al (2011) Phase
1 clinical evaluation of twice-weekly marizomib (NPI-0052), a novel proteasome inhibitor,
in patients with relapsed/refractory multiple myeloma (MM). Blood 118(21):140–141
132. Richardson PG, Baz R, Wang LH, Jakubowiak AJ, Berg D, Liu GH et al (2011)
Investigational agent MLN9708, an oral proteasome inhibitor, in patients (Pts) with relapsed
and/or refractory multiple myeloma (MM): Results from the expansion cohorts of a phase 1
dose-escalation study. Blood 118(21):140
133. Molineaux SM (2012) Molecular pathways: targeting proteasomal protein degradation in
cancer. Clin Cancer Res 18(1):15–20
134. Chauhan D, Singh AV, Aujay M, Kirk CJ, Bandi M, Ciccarelli B et al (2010) A novel orally
active proteasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxicity in multiple
myeloma. Blood 116(23):4906–4915
135. Bedford L, Lowe J, Dick LR, Mayer RJ, Brownell JE (2011) Ubiquitin-like protein conjuga-
tion and the ubiquitin-proteasome system as drug targets. Nat Rev Drug Discov 10(1):29–46
Chapter 13
New Agents and Approaches
for Targeting the RAS/RAF/MEK/ERK
and PI3K/AKT/mTOR Cell Survival
Pathways
James A. McCubrey, Linda S. Steelman, William H. Chappell, Stephen L.
Abrams, Richard A. Franklin, Giuseppe Montalto, Melchiorre Cervello,
Ferdinando Nicoletti, Graziella Malaponte, Clorinda Massarino, Massimo
Libra, Jörg Bäsecke, Agostino Tafuri, Michele Milella, Francesca Chiarini,
Camilla Evangelisti, Lucio Cocco and Alberto M. Martelli
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 331
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_13,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
332 J. A. McCubrey et al.
cells to certain small molecule inhibitors. These pathways have profound effects on
proliferative, apoptotic, and differentiation pathways. Dysregulation of components of
these pathways can contribute to: malignant transformation, resistance to other pathway
inhibitors, and chemotherapeutic drug resistance. This chapter will first briefly describe
these pathways and then evaluate potential uses of Raf, MEK, PI3K, Akt, and mTOR
inhibitors that have been investigated in preclinical and clinical investigations.
13.1 Introduction
Since the discovery of the RAS, RAF, MEK1, PIK3CA, and AKT oncogenes and neu-
rofibromin 1 (NF1), PTEN, TSC1, and TSC2 tumor suppressor genes, the Ras/Raf/
MEK/ERK, and Ras/PI3K/PTEN/Akt/mTOR signaling cascades have been extensively
investigated with the ultimate goal of determining how these genes become activated/
inactivated and whether it is possible to suppress their activity in human cancer and
other diseases [1]. Furthermore, these pathways are also implicated in the resistance and
sometimes sensitivity to therapy [2]. There have been breakthroughs in the discovery of
complex interacting pathway components, and their genetic and epigenetic regulation.
Furthermore, elucidation of the mechanisms by which mutations of components of the
pathways can lead to aberrant signaling, uncontrolled proliferation, and in some cases
confer sensitivity to targeted therapy has greatly advanced the field. This chapter will
review some of the current inhibitors, their targets, and how they are being used to treat
cancer and overcome therapeutic resistance.
Usually signaling commences upon ligation of a growth factor/cytokine/interleu-
kin/mitogen (ligand) to its cognate receptor at the cell surface. This event can result
in the activation of many downstream signaling cascades including the Ras/Raf/
MEK/ERK and Ras/PI3K/PTEN/Akt/mTOR pathways. These pathways can fur-
ther transmit their signals to different subcellular components, namely to the nucleus
to control gene expression, to the translational apparatus to enhance the translation of
“weak” mRNAs, to the apoptotic machinery to regulate apoptosis, or to other events
involved in the regulation of cellular proliferation (e.g., interactions with the p53 path-
way to regulate cell cycle progression). Regulation of the Ras/Raf/MEK/ERK and Ras/
PI3K/PTEN/Akt/mTOR pathways is mediated by a series of kinases, phosphatases,
GTP:GDP exchange, and scaffolding proteins. There are also many tumor suppressor
proteins which interact with these cascades which frequently serve to fine tune or limit
activity (e.g., NF1, PTEN, RKIP, PP2A, TSC1, and TSC2). Mutations occur in many of
the genes in these pathways leading to uncontrolled regulation and aberrant signaling.
An overview of the Ras/Raf/MEK/ERK pathway and the sites where small mol-
ecule inhibitors act is presented in Fig. 13.1. This figure serves to illustrate the flow
of information through this pathway from a growth factor to a specific receptor
13 New Agents and Approaches for Targeting 333
Fig. 13.1 Overview of the Ras/Raf/MEK/ERK cascade and small molecule inhibitors used for
targeting this pathway. Activation of this pathway can occur by mutations in upstream Growth
factor receptors (GFR) or by stimulation by the appropriate growth factors (GF). In addition,
mutations can occur in intrinsic members of the pathway (RAS, RAF, MEK1, or the tumor sup-
pressor Neurofibromin (NF1)). Sites where NF1, protein phosphatase 2A (PP2A), Raf kinase
inhibitory protein (RKIP), kinase suppressor of Ras (KSR) interact with this pathway are on the
right hand side of the Ras/Raf/MEK/ERK pathway. NF1, PP2A, and RKIP are depicted in black
rectangles as they normally serve to dampen the activity of this pathway. Sites where various
small molecule inhibitors function are in black octagons on the left hand side of the pathway.
Representative inhibitors are listed in boxes next to the octagons
growth factor receptor (GFR), for example, epidermal growth factor receptor
(EGFR), insulin-like growth factor-1 receptor (IGF-1R), vascular endothelial
growth factor receptor (VEGFR) and many others [1, 2]. Shc recruits the growth
factor receptor–bound protein 2 (Grb2) protein and the son of sevenless (SOS)
homolog protein [a guanine nucleotide exchange factor (GEF)], resulting in the
loading of the membrane-bound GDP:GTP exchange protein (GTPase). GEFs
promote Ras activation by displacing GDP from Ras which leads to GTP binding.
Ras activation is suppressed by the GTPase-activating proteins (GAPs) that stimu-
late the GTPase activity of Ras. There are two prominent GAP proteins, p120GAP
and NF1. Ras can also be activated by growth factor receptor tyrosine kinases
(GFRTK), such as insulin receptor (IR), via intermediates like insulin receptor sub-
strate (IRS) proteins that bind Grb2 [3]. Ras:GTP then recruits the serine/threonine
(S/T) kinase Raf to the membrane where it becomes activated [1, 2].
Both RAS and RAF are members of multi-gene families, and there are three
RAS members (KRAS, NRAS, and HRAS) and three RAF members (BRAF, RAF1
(a.k.a c-Raf), and ARAF) [1, 2]. Raf-1 and A-Raf are activated, in part, by a Src-
family kinase, while B-Raf does not require the Src-family kinase for activation.
Raf-1 can be regulated by dephosphorylation by the protein serine/threonine phos-
phatase 2A (PP2A). PP2A has been reported to positively and negatively regulate
Raf-1 [4, 5].
Raf is responsible for S/T phosphorylation of mitogen-activated protein kinase
kinase-1 (MEK1) (a dual specificity kinase (T/Y) [1, 2]. Other proteins such as
kinase suppressor of Ras (KSR) have recently been shown to phosphorylate
MEK1 [6]. KSR has scaffolding properties and interacts with Raf, MEK, and
ERK which regulates ERK activation. KSR can form dimers with various Raf pro-
teins which alter the effects of Raf inhibitors. KSR competes with Raf-1 for Raf
inhibitor–induced binding to B-Raf which decreases the normal ERK activation
observed after Raf-inhibitor treatment [7].
MEK1 phosphorylates extracellular signal-regulated kinases 1/2 (ERK1 and 2)
at specific T and Y residues [1, 2]. MEK1 was originally not thought to be mutated
frequently in human cancer. However, recent large-scale mutation screening stud-
ies and studies aimed at determining mechanisms of resistance to small molecule
inhibitors have observed that MEK1 is mutated in certain human cancers and also
is mutated in certain inhibitor-resistant cells [8].
Activated ERK1 and ERK2 serine S/T kinases phosphorylate and activate a
variety of substrates, including p90 ribosomal six kinase-1 (p90Rsk1) [2]. ERK
also phosphorylates MAPK signal-integrating kinases (Mnk1/2) which can in turn
phosphorylate eukaryotic translation initiation factor 4E (eIF4E), a key protein
involved in the translation of difficult mRNAs [9].
p90Rsk1 can activate the cAMP-response element-binding protein (CREB) tran-
scription factor as well as proteins involved in regulation of protein translation
(e.g., Mnk-1, p70 ribosomal S6 kinase (p70S6K), eukaryotic translation initiation
factor 4B, (eIF4B), and ribosomal protein S6 (rpS6) [10].
The number of ERK1/2 substrate/targets is easily in the hundreds. These
substrates/targets include different types of molecules including other kinases,
13 New Agents and Approaches for Targeting 335
Fig. 13.2 Overview of the PI3 K/Akt/mTOR cascade and small molecule inhibitors used for
targeting this pathway. Activation of this pathway can occur by mutations in upstream growth
factor receptors (GFR) or by stimulation by the appropriate GF. In addition, mutations can occur
in intrinsic members of the pathway (RAS, PIK3CA, AKT, or the tumor suppressors (NF1, PTEN,
TSC1, TSC2). Sites where NF1, PTEN, TSC1, TSC2 are depicted in black rectangles as they nor-
mally serve to dampen the activity of this pathway. Sites where various small molecule inhibitors
function are in black octagons. Representative inhibitors are listed in boxes next to the octagons
The mechanism by which Rheb-GTP activates mTORC1 has not been fully
elucidated yet; however, it requires Rheb farnesylation and can be blocked by
Farnesyl transferase (FT) inhibitors. It has been proposed that Rheb-GTP would
relieve the inhibitory function of FKBP38 on mTOR, thus leading to mTORC1
activation [33].
As stated previously, TSC1 and TSC2 have important roles in the regulation of
mTORC1. Two additional molecules important in this regulation are liver kinase B
(LKB1 also known as STK11). LKB1 is an upstream activator of 5′AMP-activated
protein kinase (AMPK) which activates TSC2 that negatively regulates mTORC1
[34]. LKB1 mediates the effects of the diabetes drug metformin [35]. Metformin
has also been shown to be effective in suppressing the developments of certain
cancers [36–38].
Akt also phosphorylates PRAS40, an inhibitor of mTORC1, and by doing
so, it prevents the ability of PRAS40 to suppress mTORC1 signaling (recently
reviewed in [15]). Thus, this could be yet another mechanism by which Akt acti-
vates mTORC1. Moreover, PRAS40 is a substrate of mTORC1 itself, and it has
been demonstrated that mTORC1-mediated phosphorylation of PRAS40 prevents
the inhibition of additional mTORC1 signaling [31].
Ras/Raf/MEK/ERK signaling positively impinges on mTORC1. Both p90Rsk−1
and ERK 1/2 phosphorylate TSC2, thus suppressing its inhibitory function [31].
Moreover, mTORC1 inhibition resulted in ERK 1/2 activation, through p70S6K/
PI3K/Ras/Raf/MEK [39].
The relationship between Akt and mTOR is further complicated by the exist-
ence of the mTOR/Rictor complex (mTORC2), which, in some cell types, displays
rapamycin-insensitive activity. mTORC2 is comprised of rapamycin-insensitive
companion of mTOR (Rictor), mTOR, DEPTOR, mLST8, stress-activated pro-
tein kinase interacting protein 1 (SIN1), and protein observed with Rictor (Protor).
mTORC2 phosphorylates Akt on S473 in vitro which facilitates T308 phospho-
rylation [15]. Thus, mTORC2 can function as the elusive PDK-2 which phos-
phorylates Akt-1 on S473 in response to growth factor stimulation [40]. Akt
and mTOR are linked to each other via positive and negative regulatory circuits,
which restrain their simultaneous hyperactivation through mechanisms involving
p70S6K and PI3K [15, 31]. Assuming that equilibrium exists between these two
complexes, when the mTORC1 complex is formed, it could antagonize the forma-
tion of the mTORC2 complex and reduce Akt activity. Thus, at least in principle,
inhibition of the mTORC1 complex could result in Akt hyperactivation. This is
one problem associated with therapeutic approaches using rapamycin or modified
rapamycins (rapalogs) that block some actions of mTOR but not all.
mTOR is a 289 kDa S/T kinase. mTOR was the first identified member of
the phosphatidylinositol 3-kinase-related kinase (PIKK) family [15]. mTOR has
been referred to as the gatekeeper of autophagy [41]. mTOR regulates transla-
tion in response to nutrients and growth factors by phosphorylating components
of the protein synthesis machinery, including p70S6K and eukaryotic initiation
factor (eIF)-4E binding protein-1 (4EBP-1), the latter resulting in the release of
the eukaryotic initiation factor-4E (eIF-4E) allowing eIF-4E to participate in the
13 New Agents and Approaches for Targeting 339
13.4.1 Raf Inhibitors
Raf inhibitors have been developed, and some are being used for therapy while
others are being evaluated in clinical trials. Raf inhibitors have, in general, exhib-
ited greater response rates in clinical trials than MEK inhibitors which may be
related to the broader therapeutic index of Raf inhibitors that suppress ERK activ-
ity in a mutant-allele-specific fashion as opposed to MEK inhibitors which sup-
press MEK activity in tumor and normal cells [42].
13.4.1.1 Sorafenib
Sorafenib (Bayer) was initially thought to specifically inhibit Raf but has been
subsequently shown to have multiple targets (e.g., VEGF-R, Flt-3, PDGF-R) [43].
340 J. A. McCubrey et al.
However, that does not preclude its usefulness in cancer therapy. Sorafenib is
approved for the treatment for certain cancers (e.g., renal cell Carcinoma (RCC)
and patients with unresectable HCC and was further evaluated in the Sorafenib
Hepatocellular carcinoma Assessment Randomized Protocol (SHARP) trial, which
demonstrated that the drug was effective in prolonging median survival and time
to progression in patients with advanced HCC [44, 45]. Sorafenib is generally well
tolerated in HCC patients with a manageable adverse events profile [44]. While
sorafenib is not considered effective for the treatment for most melanomas with
BRAF V600E mutations, it may be effective in the treatment for a minority of mela-
nomas with G469E and D594G mutations which express constitutive ERK1/2 but
low levels of MEK. These melanomas are sensitive to sorafenib, potentially because
they signal through Raf-1. Raf-1 also exerts anti-apoptotic effects at the mitochon-
drion in association with Bcl-2 family members [46].
13.4.1.2 Vemurafenib
13.4.1.3 Dabrafenib
13.4.1.4 CCT239065
13.4.1.5 GDC-0879
13.4.1.6 AZ628
AZ628 is a selective Raf inhibitor developed by Astra Zeneca. It has been shown
that when BRAF-mutant melanoma cells, which are normally very sensitive to
AZ628, are grown for prolonged periods of time, they become resistant to AZ628
by upregulating the expression of Raf-1 [55].
13.4.1.7 XL281
13.4.1.8 PLX5568
13.4.1.9 Raf-265
13.4.1.10 Regorafenib
13.4.2 MEK Inhibitors
Most MEK inhibitors differ from most other kinase inhibitors as they do not
compete with ATP binding (non-ATP competitive), which confers a high specific-
ity [60–62]. Most MEK inhibitors are specific and do not inhibit many different
protein kinases [62] although as will be discussed below, certain MEK inhibitors
are more specific than others.
Molecular modeling studies indicate that many MEK bind to an allosteric bind-
ing site on MEK1/MEK2. The binding sites on MEK1/MEK2 are relatively unique
to these kinases and may explain the high specificity of MEK inhibitors. This
binding may lock MEK1/2 in an inactivate conformation that enables binding of
ATP and substrate, but prevents the molecular interactions required for catalysis
and access to the ERK activation loop [61].
A distinct advantage of inhibiting MEK is that it can be targeted without
knowledge of the precise genetic mutation that results in its aberrant activation.
This is not true with targeting Raf as certain Raf inhibitors will activate Raf and
also certain B-Raf-specific inhibitors will not be effective in the presence of RAS
mutations.
An advantage of targeting MEK is that the Ras/Raf/MEK/ERK pathway is a
convergence point where a number of upstream signaling pathways can be blocked
with the inhibition of MEK. For example, MEK inhibitors, such as Selumetinib
(AZD6244), are also being investigated for the treatment for pancreatic cancers,
breast cancers, and other cancers such as hematopoietic malignancies, including
multiple myeloma [1, 63].
13.4.2.1 Selumetinib
13.4.2.2 PD-0325901
13.4.2.3 Refametinib
13.4.2.4 Trametinib
13.4.2.5 GDC-0973
13.4.2.6 AS703026
13.4.2.7 RO4987655
13.4.2.8 TAK-733
13.4.2.9 MEK162
13.4.2.10 SL327
SL337 is a MEK inhibitor that has been used in many neurological and drug
addiction studies [78].
Other MEK inhibitors are being developed. RG422 is one such inhibitor.
The possibility of treating certain patients with Raf and MEK inhibitors is a con-
cept which is gaining more acceptance as it may be a therapeutic possibility to
13 New Agents and Approaches for Targeting 345
overcome resistance [42]. Raf inhibitors induce Raf activity in cells with WT RAF
if Ras is active [79]. The addition of a MEK inhibitor would suppress the acti-
vation of MEK and ERK in the normal cells of the cancer patient. Thus, B-Raf
would be suppressed by the B-Raf-selective inhibitor in the cancer patient, while
the consequences of Raf activation in the normal cells would be suppressed by the
MEK inhibitor. These concepts are being examined in clinical trials.
The effects of combining MEK and Bcl-2/Bcl-XL inhibitors have been examined
in preclinical studies with AML cell lines and patient samples [80]. The Bcl-2
inhibitor, ABT-737, was observed to induce ERK activation and Mcl-1 expres-
sion. However, when the ABT-737 inhibitor was combined with the MEK inhibi-
tor PD0325901, a synergistic response was observed in terms of the induction of
cell death both on AML cell lines and on primary tumor cells with the properties
of leukemia stem cells. Furthermore, these studies were also extended into tumor
transplant models with the MOLT-13 cell line, and synergy between ABT-737 and
PD0325901 were also observed in vivo.
13.4.5 ERK Inhibitors
There are at least two ERK molecules regulated by the Raf/MEK/ERK cascade,
ERK1 and ERK2. Little is known about the differential in vivo targets of ERK1
and ERK2. The development of specific ERK1 and ERK2 inhibitors is ongoing
and may be useful in the treatment for certain diseases such as those leukemias
where elevated ERK activation is associated with a poor prognosis (e.g., AML,
ALL) [81]. ERK inhibitors have been described [82].
13.4.5.1 AEZS-131
AEZS-131 has been reported on the Internet to be a highly selective ERK 1/2
inhibitor developed by Aeterna Zentaris and has been examined on human breast
cancer cells.
and CRCs demonstrated that dual inhibition of MEK and ERK by small molecule
inhibitors was synergistic. Furthermore, inhibition of both MEK and ERK acted
to suppress the emergence of resistance and overcome the acquired resistance to
MEK inhibitors in these breast and CRC cell line models.
13.4.5.3 SCH772984
13.4.6 PI3K/Akt/mTOR Inhibitors
Numerous PI3K, Akt, mTOR, and dual PI3K/mTOR inhibitors have been devel-
oped and evaluated. The PI3K and mTOR inhibitors have been used in basic sci-
ence studies for years and have provided much information about the role of the
PI3K/Akt/mTOR pathway in many biologic and diseases processes. We will focus
on the newer inhibitors of this pathway and how they are now being used in clini-
cal trials.
13.4.6.1 PX-866
The modified wortmannin PX-866 has been evaluated as a PI3K inhibitor [84]. It
is being evaluated in phase II clinical trials for patients with advanced metastatic
prostate cancer by Oncothyreon.
13.4.6.2 GDC-0941
13.4.6.3 IC87114
13.4.6.4 CAL-101
The catalytic sites of PI3K and mTOR share a high degree of sequence homol-
ogy. This feature has allowed the synthesis of ATP-competitive compounds that
target the catalytic site of both PI3K and mTOR. Several dual PI3K/mTOR inhibi-
tors have also been developed. In preclinical settings, dual PI3K/mTOR inhibitors
displayed a much stronger cytotoxicity against leukemic cells than either PI3K
inhibitors or allosteric mTOR inhibitors, such as rapamycin and its derivatives
(rapalogs). In contrast to rapamycin/rapalogs, dual PI3K/mTOR inhibitors tar-
geted both mTOR complex 1 and mTOR complex 2 and inhibited the rapamycin-
resistant phosphorylation of eIF4B-1 and inhibited protein translation of many
gene products associated with oncogenesis (enhanced proliferation) in leukemic
cells. The dual inhibitors strongly reduced the proliferation rate and induced an
important apoptotic response [16].
348 J. A. McCubrey et al.
13.4.7.1 PI-103
PI-103 was the first reported ATP-competitive kinase inhibitor of mTOR which
also blocked the enzymatic activity of PI3K p110 isoforms. It was developed at
UCSF in 2006. PI-103 exhibits good selectivity over the rest of the human kinome
in terms of non-selective inhibition of other kinases [100, 101]. PI-103 is a pan-
class I PI3K inhibitor with IC50 values in the 2 nm (p110α PI3K) to 15 nm range
(p110γ PI3K) PI-103 inhibits both mTORC1 (IC50 = 0.02 μm) and mTORC2
(IC50 = 0.083 μm).
in AML cells. This resulted in the reduced levels of the expression of c-Myc,
cyclin D1, and Bcl-xL known to be regulated at the translation initiation level
[102]. NVP-BEZ235 suppressed proliferation and induced an important apoptotic
response in AML cells without affecting healthy CD34+ cell survival. Importantly,
it suppressed the clonogenic activity of leukemic, but not healthy, CD34+ cells
[103]. NVP-BEZ235 targeted the side population (SP) of both T-ALL cell lines
and patient lymphoblasts, which might correspond to Leukemia-Initiating Cells
(LIC), and synergized with several chemotherapeutic agents (cyclophosphamide,
cytarabine, dexamethasone) currently used for treating T-ALL patients [104].
Also, NVP-BEZ235 reduced chemoresistance to vincristine induced in Jurkat cells
by co-culturing with MS-5 stromal cells, which mimic the bone marrow micro-
environment [105]. In this study, NVP-BEZ235 was cytotoxic to T-ALL patient
lymphoblasts displaying pathway activation, where the drug dephosphorylated
4E-BP1, in contrast to the results with obtained rapamycin. Taken together, these
findings indicated that longitudinal inhibition at two nodes of the PI3K/Akt/mTOR
network with NVP-BEZ235, either alone or in combination with chemothera-
peutic drugs, may be an effective therapy for of those T-ALLs that have aberrant
upregulation of this signaling pathway.
NVP-BEZ235 has been evaluated also in a mouse model consisting of
BA/F3 cells overexpressing either WT BCR-ABL or its imatinib-resistant BCR-
ABL mutants (E255K and T315I) [106]. NVP-BEZ235 inhibited proliferation of
both cytokine-independent WT BCR-ABL and mutant BCR-ABL (E255K and
T315I) overexpressing cells, whereas parental cytokine-dependent Ba/F3 cells
were much less sensitive. The drug also induced apoptosis and inhibited both
mTORC1 and mTORC2 signaling. Remarkably, the drug displayed cytotoxic
activity in vivo against leukemic cells expressing the E255K and T315I BCRABL
mutant forms. However, in this experimental model, NVP-BEZ235 induced an
overactivation of MEK/ERK signaling, most likely due to the well-known com-
pensatory feedback mechanism that involves p70S6K [39]. NVP-BEZ235 has
been intensively investigated and is in clinical trials for patients with advanced
cancers [107]. In some trials, NVP-BEZ235 is being evaluated in combination
with either paclitaxel or trastuzumab (herceptin). NVP-BTG226 is a recently
developed PI3K/mTOR inhibitor [15].
13.4.7.4 XL765
13.4.7.7 WJD008
13.4.8 PDK Inhibitors
Some compounds have been reported to be PDK inhibitors, including the osteo-
arthritis drug celecoxib [116], the modified celecoxib, OSU-03012 [76, 117], and
2-O-BN-InsP(5) [118]. Celecoxib (Celebrex, Pfizer) obviously has other targets
than PDK, such as cyclooxygenase-2 (Cox-2). Celecoxib is used to treat CRC
patients to reduce the number of polyps in the colon. OSU-03012 is reported
not to inhibit Cox-2 [117]. 2-O-BN-InsP(5) is based on the structure of inositol
1,3,4,5,6-pentakisphosphate, it may inhibit both PDK and mTOR [118].
13.4.9 Akt Inhibitors
Many attempts to develop Akt inhibitors have been performed over the years. In
many of the earlier attempts, the various Akt inhibitors either lacked specificity or
had deleterious side effects. Part of the deleterious side effects is probably related
to the numerous critical functions that Akt plays in normal physiology. Namely,
some Akt inhibitors will alter the downstream effects of insulin on Glut-4 translo-
cation and glucose transport.
13.4.9.1 Triciribine
Triciribine (API-2) is an Akt inhibitor that has been used in many studies: at least
92 are listed on PubMed. Triciribine suppressed the phosphorylation of all three
Akt isoforms in vitro and the growth of tumor cells overexpressing Akt in mouse
xenograft models [119]. The mechanism(s) by which triciribine inhibits Akt activ-
ity are not clear. The drug has been evaluated in a phase I clinical trial in patients
with advanced hematologic malignancies, including refractory/relapsed AML. In
this trial, triciribine was administered on a weekly schedule. The drug was well
tolerated, with preliminary evidence of pharmacodynamic activity as measured by
decreased levels of activated Akt in primary blast cells [120]. Triciribine has also
been examined in clinical trial with Akt+ metastatic cancers.
13.4.9.2 MK-2206
MK-2206 (Merck) is an allosteric Akt inhibitor which inhibits both T308 and
S473 phosphorylation. It also inhibits the downstream effects of insulin on Glut-4
352 J. A. McCubrey et al.
13.4.9.4 KP372-1
KP372-1 inhibits PDK1, Akt, and Fms-like tyrosine kinase 3 (Flt-3) signaling and
induces mitochondrial dysfunction and apoptosis in AML cells but not normal
hematopoietic progenitor cells [124]. It also suppressed colony formation of pri-
mary AML patient sample cells but not normal hematopoietic progenitor cells. It
has also been investigated in other cancer types, including squamous cell carcino-
mas of the head and neck, thyroid cancers, and glioblastomas.
13.4.9.5 Enzasturin
Enzasturin (LY317615) is a protein kinase C-β (PKC-β) and Akt inhibitor devel-
oped by Lilly. It has been investigated in clinical trials either by itself or in
13 New Agents and Approaches for Targeting 353
combination with other agents in various types of cancer patients including brain
[125] and NSC [126], CRC [127] as well as other cancer types. It is reported to be
in approximately 48 clinical trials on the ClinicalTrials.gov website.
13.4.9.6 Perifosine
13.4.9.8 PBI-05204
13.4.9.9 RX-0201
13.4.9.10 XL-418
13.4.10 mTORC1 Inhibitors
13.4.11 mTOR Inhibitors
Small molecules designed for inhibiting the catalytic site of mTOR have shown
promising effects on the suppression of signaling downstream of mTOR. mTOR
kinase inhibitor has been developed which directly inhibits mTORC1 and
mTORC2. The mTOR kinase inhibitors have advantages over rapamycin and the
13 New Agents and Approaches for Targeting 355
rapalogs as mTOR inhibitors will inhibit both mTORC1 and mTORC2, while
rapamycin and the rapalogs only inhibit mTORC1. Also, the mTOR kinases inhib-
itors do not induce the feedback pathways which result in Akt activation. In vitro
studies with purified mTOR and PI3K proteins have demonstrated that the mTOR
inhibitors selectively bind mTOR more than PI3K.
13.4.11.1 OSI-027
AZD8055 and AZD2014 are pan-mTOR inhibitors with potent anti-tumor activity
[151]. They are being evaluated in clinical trials patients with gliomas who have not
responded to standard glioma therapies as well as patients with other types of cancer.
than either mTORC1 or mTORC2. These inhibitors were modified which resulted
in WYE132 (WYE125132)/WYE132 has 5000-fold greater selectivity for mTOR
over PI3K. It caused tumor regression in breast, glioma, lung, renal tumors [153].
Many other TOR inhibitors have been described which include Ku0063794
(KuDOS Pharmaceuticals) [154] and OXA-01 (OSI Pharmaceuticals) [155]. Torin2
has been developed by optimizing from Torin1 [156]. TORKiCC223 is a pan-TOR
inhibitor developed by Celgene. Other companies are developing mTOR inhibitors;
clearly, this is a very competitive but important research and clinical area.
In the following section, we discuss the potential of combining inhibitors that tar-
get two pathways to more effectively limit cancer growth. Treatment for inducible
murine lung cancers containing KRAS and PIK3CA mutations with PI3K/mTOR
(NVP-BEZ235) and MEK (selumetinib) inhibitors led to an enhanced response
[157]. Synergistic responses between sorafenib and mTOR inhibitors were
observed in xenograft studies with a highly metastatic human HCC tumor [158].
Some recent studies in thyroid cancer have documented the benefit of combining
Raf and PI3K/mTOR inhibitors [159].
Intermittent dosing of MEK and PI3K inhibitors has been observed to suppress
the growth of tumor xenografts in mice [74]. This study demonstrated that con-
tinuous administration of MEK and PI3K inhibitors is not required to suppress
xenograft growth. These important results were obtained by performing washout
studies in vitro and alternate dosing schedules in mice with MEK and PI3K inhibi-
tors with cancer cells having mutations at BRAF and KRAS.
The combined effects of inhibiting MEK with PD-0329501 and
mTOR with rapamycin or its analog, the rapalog AP-23573 (ARIAD
Pharmaceuticals/Merck) were examined in human NSCLC cell lines, as
well as in animal models of human lung cancer. PD-0325901 and rapamy-
cin demonstrated synergistic inhibition of proliferation and protein translation.
Suppression of both MEK and mTOR inhibited ribosomal biogenesis and was
associated with a block in the initiation phase of translation [160]. The pan-
TOR inhibitor AZD-8055 has been examined as a single agent and in combina-
tion with the MEK inhibitor selumetinib in a NSCLC xenograft and increased
cell death and tumor regression [151, 161]. These preclinical results support the
13 New Agents and Approaches for Targeting 357
suppression of both the MEK and mTOR pathways in lung cancer therapy and
indicate that both pathways converge to regulate the initiation of protein transla-
tion. ERK phosphorylates Mnk1/2 and p90Rsk, which regulate the activity of the
eukaryotic translation initiation factor eIF4E. The phosphorylation of 4EBP1 is
altered in cells containing BRAF mutations. It should also be pointed out that
4EBP1 is also regulated by Akt, mTOR, and p70S6K. This may result in the effi-
cient translation of certain mRNAs in BRAF-mutant cells. This could explain
how co-inhibition of MEK and PI3K/Akt/mTOR synergizes to inhibit protein
translation and growth in certain lung cancer cells.
examine the effects of combining MK-2206 with gefitinib (iressa, EGFR inhibitor
AstraZenica). Clinical trials with postmenopausal metastatic breast cancer patients
are in progress to examine the effects of combining anastrozole, letrozole, exemes-
tane (aromatase-inhibitors), or fulvestrant (an estrogen receptor antagonist).
Clinical trials are also underway examining the effects of combining MK-2206
with bendamustin (nitrogen mustard alkylating agent) and Rituximab (chimeric
monoclonal antibody targeting CD20 from IDEC Pharmaceuticals/Genentech)
on CLL cancer patients who have relapsed or on cancer patients with small
lymphocytic lymphoma. Clinical trials combining MK-2206 and various other
drugs including dalotuzumab (a MoAb which targets IGF-1R from Merck) and
MK-0752 (a Y-secretase inhibitor which inhibits the NOTCH pathway from
Merck) are in progress. The effects of MK-8669 (ridaforolimus an mTORC1
inhibitor from Merck) and dalotuzumab are being examined in patients with
advanced cancers. Clinical trials combining MK-2206 and paclitaxel in cancer
patients with locally advanced, metastatic solid tumors, or metastatic breast can-
cers are in progress. The above mentioned clinical trials document the importance
of targeting Akt and other signaling molecules as well as critical targets involved
in cellular division. Furthermore, the clinical trials document how basic research
on these pathways is being translated into clinical therapy for cancer and other
types of patients.
served as a radiosensitizer. The other two cancer cell lines examined in this study
(A549 and MiaPaCa2) had KRAS mutations and both were radiosensitized by the
MEK inhibitor. Although these studies document the ability of a MEK inhibitor to
radiosensitize certain cells, clearly other cancer cell lines without activating muta-
tions in the Ras/Raf/MEK/ERK pathway or autocrine growth stimulation should
be examined for radiosensitization by the MEK inhibitor as the KRAS mutation
may also activate the PI3K pathway which could lead to therapy resistance.
PI3K/Akt/mTOR inhibitors will sensitize the tumor vasculature to radiation
both in vitro in cell lines and in vivo in xenografts [171, 172]. mTOR and radia-
tion play critical roles in the regulation of autophagy [173, 174]. When mTOR is
blocked by rapamycin, there is an increase in autophagy. This is important as apop-
totic cell death is a minor component to cell death in many solid tumors. These
studies document the potential beneficial use of combining mTOR inhibitors and
radiation to improve the induction of autophagy in the treatment for solid tumors.
13.9 Conclusions
not very cytotoxic when it comes to killing tumor cells. This inherent property of
rapamycins may also contribute to their low toxicity in humans.
Mutations at many of the upstream receptor genes or RAS can result in abnor-
mal Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathway activation. Hence,
targeting these cascade components with small molecule inhibitors may inhibit
cell growth. The usefulness of these inhibitors may depend on the mechanism of
transformation of the particular cancer. If the tumor exhibits a dependency on the
Ras/Raf/MEK/ERK pathway, then it may be sensitive to Raf and MEK inhibitors.
In contrast, tumors that do not display enhanced expression of the Ras/Raf/MEK/
ERK pathway may not be sensitive to either Raf or MEK inhibitors, but if the Ras/
PI3K/Akt/mTOR pathway is activated, these cancers may be sensitive to specific
inhibitors that target this pathway. Finally, it is likely that many of the inhibitors
that we have discussed in this review will be more effective in inhibiting tumor
growth in combination with MoAb, cytotoxic chemotherapeutic drugs, or radia-
tion. This is documented by the large number of clinical trials combining signal
transduction inhibitors with these various therapeutic agents.
Some scientists and clinicians have considered that the simultaneous targeting
of Raf and MEK by individual inhibitors may be more effective in cancer therapy
than just targeting Raf or MEK by themselves. This is based in part on the fact
that there are intricate feedback loops from ERK which can inhibit Raf and MEK.
For example, when MEK1 is targeted, ERK1,2 is inhibited, and the negative feed-
back loop on MEK is broken and activated MEK accumulates. However, if Raf
is also inhibited, it may be possible to completely shut down the pathway. This is
a rationale for treatment with both MEK and Raf inhibitors. Likewise, targeting
both PI3K and mTOR may be more effective than targeting either PI3K or mTOR
by themselves. If it is a single inhibitor which targets both molecules, such as the
new PI3K and mTOR dual inhibitors, this becomes a realistic therapeutic option.
Although it should be pointed out that some studies are examining the effects of
combining rapamycins and PI3K/mTOR inhibitors. Finally, an emerging concept
is the dual targeting of two different signal transduction pathways, Raf/MEK/ERK
and PI3K/PTEN/Akt/mTOR, for example. This has been explored in some preclin-
ical models as discussed in the text. The rationale for targeting of both pathways
may be dependent on the presence of mutations in either/or both pathways or in
upstream Ras in the particular cancer which can activate both pathways.
It is not always clear why a particular combination of a signal transduction
inhibitor and chemotherapeutic drug works in one tumor type but not at all in a
different tumor type. This has also been experienced with the development of indi-
vidual chemotherapeutic drugs, some work in some cells but not others. This may
result from many different complex interacting events. Some of these events could
include percentage of cells in different phases of the cell cycle, persistence of can-
cer stem cells, presence of multiple mutated activated oncogenes, or repressed
tumor suppressor genes, epigenetic modifications and many other factors. Finally,
chemotherapeutic drug therapy and other types of therapy (radiotherapy, antibody
therapy) may induce certain signaling pathways (e.g., the reactive oxygen spe-
cies generated by chemotherapy and radiotherapy induce the Ras/Raf/MEK/ERK
362 J. A. McCubrey et al.
pathway). The induction of these signaling pathways may counteract some of the
effects of the signal transduction inhibitors. These effects could indicate that the
timing of each therapy will be important for effective treatments.
In summary, targeted therapy has advanced from basic research studies to the
treatment of cancer patients in less than 25 years. We have learned a lot regard-
ing how specific inhibitors exert their effects and how the Ras/Raf/MEK/ERK and
PI3K/Akt/mTOR pathways function; we still need to discover more about resist-
ance mechanisms and how we can overcome therapeutic resistance and improve
the effectiveness of targeted therapy.
References
1. Chappell WH, Steelman LS, Long JM, Kempf RC, Abrams SL, Franklin RA et al (2011)
Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR inhibitors: rationale and importance to
inhibiting these pathways in human health. Oncotarget 2(3):135–164
2. McCubrey JA, Steelman LS, Kempf CR, Chappell WH, Abrams SL, Stivala F et al
(2011) Therapeutic resistance resulting from mutations in Raf/MEK/ERK and
PI3 K/PTEN/Akt/mTOR signaling pathways. J Cell Physiol 226(11):2762–2781
3. Hayashi K, Shibata K, Morita T, Iwasaki K, Watanabe M, Sobue K (2004) Insulin recep-
tor substrate-1/SHP-2 interaction, a phenotype-dependent switching machinery of
insulin-like growth factor-I signaling in vascular smooth muscle cells. J Biol Chem
279(39):40807–40818
4. Mischak H, Seitz T, Janosch P, Eulitz M, Steen H, Schellerer M et al (1996) Negative regu-
lation of Raf-1 by phosphorylation of serine 621. Mol Cell Biol 16(10):5409–5418
5. Abraham D, Podar K, Pacher M, Kubicek M, Welzel N, Hemmings BA et al (2000) Raf-1-
associated protein phosphatase 2A as a positive regulator of kinase activation. J Biol Chem
275(29):22300–22304
6. Brennan DF, Dar AC, Hertz NT, Chao WC, Burlingame AL, Shokat KM et al (2011) A
Raf-induced allosteric transition of KSR stimulates phosphorylation of MEK. Nature
472(7343):366–369
7. McKay MM, Ritt DA, Morrison DK (2011) RAF inhibitor-induced KSR1/B-RAF binding
and its effects on ERK cascade signaling. Curr Biol 21(7):563–568
13 New Agents and Approaches for Targeting 363
8. Pao W, Girard N (2011) New driver mutations in non-small-cell lung cancer. Lancet Oncol
12(2):175–180
9. Topisirovic I, Sonenberg N (2011) mRNA translation and energy metabolism in cancer: the
role of the MAPK and mTORC1 pathways. Cold Spring Harb Symp Quant Biol 28:28
10. Xing J, Ginty DD, Greenberg ME (1996) Coupling of the RAS-MAPK pathway to gene
activation by RSK2, a growth factor-regulated CREB kinase. Science 273(5277):959–963
11. Balan V, Leicht DT, Zhu J, Balan K, Kaplun A, Singh-Gupta V et al (2006) Identification of
novel in vivo Raf-1 phosphorylation sites mediating positive feedback Raf-1 regulation by
extracellular signal-regulated kinase. Mol Biol Cell 17(3):1141–1153
12. Dougherty MK, Muller J, Ritt DA, Zhou M, Zhou XZ, Copeland TD et al (2005) Regulation
of Raf-1 by direct feedback phosphorylation. Mol Cell 17(2):215–224
13. Catalanotti F, Reyes G, Jesenberger V, Galabova-Kovacs G, de Matos Simoes R, Carugo O
et al (2009) A Mek1-Mek2 heterodimer determines the strength and duration of the Erk sig-
nal. Nat Struct Mol Biol 16(3):294–303
14. Sturm OE, Orton R, Grindlay J, Birtwistle M, Vyshemirsky V, Gilbert D et al (2010) The
mammalian MAPK/ERK pathway exhibits properties of a negative feedback amplifier. Sci
Signal 3(153)
15. Martelli AM, Evangelisti C, Chappell W, Abrams SL, Basecke J, Stivala F et al
(2011) Targeting the translational apparatus to improve leukemia therapy: roles of the
PI3K/PTEN/Akt/mTOR pathway. Leukemia 25(7):1064–1079
16. Martelli AM, Chiarini F, Evangelisti C, Cappellini A, Buontempo F, Bressanin D et al
(2012) Two hits are better than one: targeting both phosphatidylinositol 3-kinase and
mammalian target of rapamycin as a therapeutic strategy for acute leukemia treatment.
Oncotarget 3(4):371–394
17. Steelman LS, Chappell WH, Abrams SL, Kempf RC, Long J, Laidler P et al (2011) Roles of
the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensi-
tivity to therapy-implications for cancer and aging. Aging 3(3):192–222
18. Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese CB et al (1997)
Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates
and activates protein kinase Balpha. Curr Biol 7(4):261–269
19. Coffer PJ, Woodgett JR (1992) Molecular cloning and characterisation of a novel putative
protein-serine kinase related to the cAMP-dependent and protein kinase C families. Eur J
Biochem 205(3):1217
20. Gonzalez E, McGraw TE (2009) The Akt kinases: isoform specificity in metabolism and
cancer. Cell Cycle 8(16):2502–2508
21. Du K, Montminy M (1998) CREB is a regulatory target for the protein kinase Akt/PKB. J
Biol Chem 273(49):32377–32379
22. Brennan P, Babbage JW, Burgering BM, Groner B, Reif K, Cantrell DA (1997)
Phosphatidylinositol 3-kinase couples the interleukin-2 receptor to the cell cycle regulator
E2F. Immunity 7(5):679–689
23. Kane LP, Shapiro VS, Stokoe D, Weiss A (1999) Induction of NF-kappaB by the Akt/PKB
kinase. Curr Biol 9(11):601–604
24. Buitenhuis M, Coffer PJ (2009) The role of the PI3K-PKB signaling module in regulation
of hematopoiesis. Cell Cycle 8(4):560–566
25. del Peso L, Gonzalez-Garcia M, Page C, Herrera R, Nunez G (1997) Interleukin-3-induced
phosphorylation of BAD through the protein kinase Akt. Science 278(5338):687–689
26. Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA (1995) Inhibition of glyco-
gen synthase kinase-3 by insulin mediated by protein kinase B. Nature 378(6559):785–789
27. Chalhoub N, Baker SJ (2009) PTEN and the PI3-kinase pathway in cancer. Annu Rev
Pathol 4:127–150
28. Weng LP, Brown JL, Baker KM, Ostrowski MC, Eng C (2002) PTEN blocks insulin-medi-
ated ETS-2 phosphorylation through MAP kinase, independently of the phosphoinositide
3-kinase pathway. Hum Mol Genet 11(15):1687–1696
364 J. A. McCubrey et al.
49. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J et al (2011)
Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J
Med 364(26):2507–2516
50. Long G, Kefford RF, Carr PJA, Brown MP, Curtis M, Ma B, Lebowitz P, Kim KB, Kurzrock
R, Flachook G(2010) Phase 1/2 study of GSK2118436, a selective inhibitor of V600 mutant
(mut) BRAF kinase: evidence of activity in melanoma brain metastases (mets). Annals of
Oncology. 21(LBA27 (supplement 8)):viii12
51. Kefford R, Arkenau H, Brown MP, Millward M, Infante JR, Long GV, et al (2010) Phase
I/II study of GSK2118436, a selective inhibitor of oncogenic mutant BRAF kinase, in
patients with metastatic melanoma and other solid tumors. ASCO Meeting Abstracts 2010
June 14, 28(15_suppl):8503
52. Greger JG, Eastman SD, Zhang V, Bleam MR, Hughes AM, Smitheman KN et al (2012)
Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to
the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol
Cancer Ther 11(4):909–920
53. Whittaker S, Menard D, Kirk R, Ogilvie L, Hedley D, Zambon A et al (2010) A novel,
selective and efficacious nanomolar pyridopyrazinone inhibitor of V600EBRAF. Cancer Res
70(20):8036–8044
54. Hoeflich KP, Herter S, Tien J, Wong L, Berry L, Chan J et al (2009) Antitumor efficacy of
the novel RAF inhibitor GDC-0879 is predicted by BRAFV600E mutational status and sus-
tained extracellular signal-regulated kinase/mitogen-activated protein kinase pathway sup-
pression. Cancer Res 69(7):3042–3051
55. Montagut C, Sharma SV, Shioda T, McDermott U, Ulman M, Ulkus LE et al (2008)
Elevated CRAF as a potential mechanism of acquired resistance to BRAF inhibition in mel-
anoma. Cancer Res 68(12):4853–4861
56. Schwartz GK, Robertson S, Shen A, Wang E, Pace L, Dials H et al (2009) A phase I study
of XL281, a selective oral RAF kinase inhibitor, in patients (Pts) with advanced solid
tumors. ASCO Meeting Abstracts. 27(15S):3513 (2009 June 8)
57. Buchholz B, Klanke B, Schley G, Bollag G, Tsai J, Kroening S et al (2011) The Raf kinase
inhibitor PLX5568 slows cyst proliferation in rat polycystic kidney disease but promotes
renal and hepatic fibrosis. Nephrol Dial Transplant 26(11):3458–3465
58. Zitzmann K, de Toni E, von Ruden J, Brand S, Goke B, Laubender RP et al (2011) The
novel Raf inhibitor Raf265 decreases Bcl-2 levels and confers TRAIL-sensitivity to neu-
roendocrine tumour cells. Endocr Relat Cancer 18(2):277–285
59. Wilhelm SM, Dumas J, Adnane L, Lynch M, Carter CA, Schutz G et al (2011) Regorafenib
(BAY 73–4506): a new oral multikinase inhibitor of angiogenic, stromal and onco-
genic receptor tyrosine kinases with potent preclinical antitumor activity. Int J Cancer
129(1):245–255
60. Davies SP, Reddy H, Caivano M, Cohen P (2000) Specificity and mechanism of action of
some commonly used protein kinase inhibitors. Biochem J 351(Pt 1):95–105
61. Ohren JF, Chen H, Pavlovsky A, Whitehead C, Zhang E, Kuffa P et al (2004) Structures
of human MAP kinase kinase 1 (MEK1) and MEK2 describe novel noncompetitive kinase
inhibition. Nat Struct Mol Biol 11(12):1192–1197
62. Sebolt-Leopold JS (2008) Advances in the development of cancer therapeutics
directed against the RAS-mitogen-activated protein kinase pathway. Clin Cancer Res
14(12):3651–3656
63. McCubrey JA, Steelman LS, Abrams SL, Chappell WH, Russo S, Ove R et al (2010)
Emerging MEK inhibitors. Expert Opin Emerg Drugs 15(2):203–223
64. Davies BR, Logie A, McKay JS, Martin P, Steele S, Jenkins R, et al (2007) AZD6244
(ARRY-142886), a potent inhibitor of mitogen-activated protein kinase/extracellular signal-
regulated kinase kinase 1/2 kinases: mechanism of action in vivo, pharmacokinetic/phar-
macodynamic relationship, and potential for combination in preclinical models. Molecular
Cancer Therapeutics. 6(8):2209–2219
366 J. A. McCubrey et al.
65. Tai YT, Fulciniti M, Hideshima T, Song W, Leiba M, Li XF et al (2007) Targeting MEK
induces myeloma-cell cytotoxicity and inhibits osteoclastogenesis. Blood 110(5):1656–1663
66. Yeh TC, Marsh V, Bernat BA, Ballard J, Colwell H, Evans RJ et al (2007) Biological char-
acterization of ARRY-142886 (AZD6244), a potent, highly selective mitogen-activated pro-
tein kinase 1/2 inhibitor. Clin Cancer Res 13(5):1576–1583
67. Friday BB, Yu C, Dy GK, Smith PD, Wang L, Thibodeau SN et al (2008) BRAF V600E
disrupts AZD6244-induced abrogation of negative feedback pathways between extracellular
signal-regulated kinase and Raf proteins. Cancer Res 68(15):6145–6153
68. Yoon J, Koo KH, Choi KY (2011) MEK1/2 inhibitors AS703026 and AZD6244 may be
potential therapies for KRAS mutated colorectal cancer that is resistant to EGFR monoclo-
nal antibody therapy. Cancer Res 71(2):445–453
69. Mohammad RM, Goustin AS, Aboukameel A, Chen B, Banerjee S, Wang G et al (2007)
Preclinical Studies of TW-37, a New Nonpeptidic Small-Molecule Inhibitor of Bcl-2, in
Diffuse Large Cell Lymphoma Xenograft Model Reveal Drug Action on Both Bcl-2 and
Mcl-1. Clin Cancer Res 13(7):2226–2235
70. Haura EB, Ricart AD, Larson TG, Stella PJ, Bazhenova L, Miller VA et al (2010) A phase II
study of PD-0325901, an oral MEK inhibitor, in previously treated patients with advanced
non-small cell lung cancer. Clin Cancer Res 16(8):2450–2457
71. LoRusso PM, Krishnamurthi SS, Rinehart JJ, Nabell LM, Malburg L, Chapman PB
et al (2010) Phase I pharmacokinetic and pharmacodynamic study of the oral MAPK/
ERK kinase inhibitor PD-0325901 in patients with advanced cancers. Clin Cancer Res
16(6):1924–1937
72. Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G, Basso A et al (2006) BRAF muta-
tion predicts sensitivity to MEK inhibition. Nature 439(7074):358–362
73. Iverson C, Larson G, Lai C, Yeh LT, Dadson C, Weingarten P et al (2009) RDEA119/BAY
869766: a potent, selective, allosteric inhibitor of MEK1/2 for the treatment of cancer.
Cancer Res 69(17):6839–6847
74. Hoeflich KP, Merchant M, Orr C, Chan J, Den Otter D, Berry L et al (2012) Intermittent
administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust
apoptosis and tumor growth inhibition. Cancer Res 72(1):210–219
75. Kim K, Kong SY, Fulciniti M, Li X, Song W, Nahar S et al (2010) Blockade of the MEK/
ERK signalling cascade by AS703026, a novel selective MEK1/2 inhibitor, induces pleio-
tropic anti-myeloma activity in vitro and in vivo. Br J Haematol 149(4):537–549
76. Lee TX, Packer MD, Huang J, Akhmametyeva EM, Kulp SK, Chen CS et al (2009) Growth
inhibitory and anti-tumour activities of OSU-03012, a novel PDK-1 inhibitor, on vestibular
schwannoma and malignant schwannoma cells. Eur J Cancer 45(9):1709–1720
77. Dong Q, Dougan DR, Gong X, Halkowycz P, Jin B, Kanouni T et al (2011) Discovery of
TAK-733, a potent and selective MEK allosteric site inhibitor for the treatment of cancer.
Bioorg Med Chem Lett 21(5):1315–1319
78. Longoni R, Spina L, Vinci S, Acquas E (2011) The MEK inhibitor SL327 blocks acquisition
but not expression of lithium-induced conditioned place aversion: a behavioral and immuno-
histochemical study. Psychopharmacology 216(1):63–73
79. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N (2010) RAF inhibitors trans-
activate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature
464(7287):427–430
80. Konopleva M, Milella M, Ruvolo P, Watts JC, Ricciardi MR, Korchin B et al (2012)
MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of
ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex. Leukemia
26(4):778–787
81. Ricciardi MR, Scerpa MC, Bergamo P, Ciuffreda L, Petrucci MT, Chiaretti S et al (2012)
Therapeutic potential of MEK inhibition in acute myelogenous leukemia: rationale for “ver-
tical” and “lateral” combination strategies. J Mol Med 8:8
82. Aronov AM, Tang Q, Martinez-Botella G, Bemis GW, Cao J, Chen G et al (2009) Structure-
guided design of potent and selective pyrimidylpyrrole inhibitors of extracellular signal-reg-
ulated kinase (ERK) using conformational control. J Med Chem 52(20):6362–6368
13 New Agents and Approaches for Targeting 367
83. Hatzivassiliou G, Liu B, O’Brien C, Spoerke JM, Hoeflich KP, Haverty PM et al (2012)
ERK inhibition overcomes acquired resistance to MEK inhibitors. Mol Cancer Ther
11(5):1143–1154
84. Ihle NT, Williams R, Chow S, Chew W, Berggren MI, Paine-Murrieta G et al (2004)
Molecular pharmacology and antitumor activity of PX-866, a novel inhibitor of phosphoi-
nositide-3-kinase signaling. Mol Cancer Ther 3(7):763–772
85. Burrows N, Babur M, Resch J, Ridsdale S, Mejin M, Rowling EJ et al (2011) GDC-0941
inhibits metastatic characteristics of thyroid carcinomas by targeting both the phosphoi-
nositide-3 kinase (PI3K) and hypoxia-inducible factor-1alpha (HIF-1alpha) pathways. J Clin
Endocrinol Metab 96(12):12
86. Zou ZQ, Zhang LN, Wang F, Bellenger J, Shen YZ, Zhang XH (2012) The novel dual
PI3K/mTOR inhibitor GDC-0941 synergizes with the MEK inhibitor U0126 in non-small
cell lung cancer cells. Mol Med Report 5(2):503–508
87. Sujobert P, Bardet V, Cornillet-Lefebvre P, Hayflick JS, Prie N, Verdier F et al (2005)
Essential role for the p110delta isoform in phosphoinositide 3-kinase activation and cell
proliferation in acute myeloid leukemia. Blood 106(3):1063–1066
88. Billottet C, Grandage VL, Gale RE, Quattropani A, Rommel C, Vanhaesebroeck B et
al (2006) A selective inhibitor of the p110delta isoform of PI 3-kinase inhibits AML
cell proliferation and survival and increases the cytotoxic effects of VP16. Oncogene
25(50):6648–6659
89. Tamburini J, Chapuis N, Bardet V, Park S, Sujobert P, Willems L et al (2008) Mammalian
target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-
regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: ration-
ale for therapeutic inhibition of both pathways. Blood 111(1):379–382
90. Workman P, van Montfort RL (2010) PI(3) kinases: revealing the delta lady. Nat Chem Biol
6(2):82–83
91. Berndt A, Miller S, Williams O, Le DD, Houseman BT, Pacold JI et al (2010) The p110delta
structure: mechanisms for selectivity and potency of new PI(3)K inhibitors. Nat Chem Biol
6(3):244
92. Lannutti BJ, Meadows SA, Herman SE, Kashishian A, Steiner B, Johnson AJ et al
(2011) CAL-101, a p110delta selective phosphatidylinositol-3-kinase inhibitor for the
treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability. Blood
117(2):591–594
93. Meadows SA, Vega F, Kashishian A, Johnson D, Diehl V, Miller LL et al (2012) PI3Kdelta
inhibitor, GS-1101 (CAL-101), attenuates pathway signaling, induces apoptosis, and over-
comes signals from the microenvironment in cellular models of Hodgkin lymphoma. Blood
119(8):1897–1900
94. Gale S, Croasdell G (2010) 28th Annual JPMorgan healthcare conference–forest laborato-
ries and Icagen. IDrugs 13(3):145–148
95. Maira SM, Pecchi S, Huang A, Burger M, Knapp M, Sterker D et al (2012) Identification
and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor.
Mol Cancer Ther 11(2):317–328
96. Bendell JC, Rodon J, Burris HA, de Jonge M, Verweij J, Birle D et al (2012) Phase I, dose-
escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced
solid tumors. J Clin Oncol 30(3):282–290
97. Brachmann S, Fritsch C, Maira SM, Garcia-Echeverria C (2009) PI3K and mTOR inhibi-
tors: a new generation of targeted anticancer agents. Curr Opin Cell Biol 21(2):194–198
98. Molckovsky A, Siu LL (2008) First-in-class, first-in-human phase I results of targeted
agents: highlights of the 2008 American society of clinical oncology meeting. J Hematol
Oncol 1:20
99. Xu CX, Li Y, Yue P, Owonikoko TK, Ramalingam SS, Khuri FR et al (2011) The combina-
tion of RAD001 and NVP-BEZ235 exerts synergistic anticancer activity against non-small
cell lung cancer in vitro and in vivo. PLoS One 6(6):14
100. Fan QW, Knight ZA, Goldenberg DD, Yu W, Mostov KE, Stokoe D et al (2006) A dual PI3
kinase/mTOR inhibitor reveals emergent efficacy in glioma. Cancer Cell 9(5):341–349
368 J. A. McCubrey et al.
101. Fan QW, Cheng CK, Nicolaides TP, Hackett CS, Knight ZA, Shokat KM et al (2007) A dual
phosphoinositide-3-kinase alpha/mTOR inhibitor cooperates with blockade of epidermal
growth factor receptor in PTEN-mutant glioma. Cancer Res 67(17):7960–7965
102. Maira SM, Stauffer F, Brueggen J, Furet P, Schnell C, Fritsch C et al (2008) Identification
and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol
3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity.
Mol Cancer Ther 7(7):1851–1863
103. Chapuis N, Tamburini J, Green AS, Vignon C, Bardet V, Neyret A et al (2010) Dual inhibi-
tion of PI3K and mTORC1/2 signaling by NVP-BEZ235 as a new therapeutic strategy for
acute myeloid leukemia. Clin Cancer Res 16(22):5424–5435
104. Chiarini F, Fala F, Tazzari PL, Ricci F, Astolfi A, Pession A et al (2009) Dual inhibi-
tion of class IA phosphatidylinositol 3-kinase and mammalian target of rapamy-
cin as a new therapeutic option for T-cell acute lymphoblastic leukemia. Cancer Res
69(8):3520–3528
105. Chiarini F, Grimaldi C, Ricci F, Tazzari PL, Evangelisti C, Ognibene A et al (2010) Activity
of the novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor
NVP-BEZ235 against T-cell acute lymphoblastic leukemia. Cancer Res 70(20):8097–8107
106. Schuster K, Zheng J, Arbini AA, Zhang CC, Scaglioni PP (2011) Selective targeting of the
mTORC1/2 protein kinase complexes leads to antileukemic effects in vitro and in vivo.
Blood Cancer J 1:e34
107. Shuttleworth SJ, Silva FA, Cecil AR, Tomassi CD, Hill TJ, Raynaud FI et al (2011) Progress
in the preclinical discovery and clinical development of class I and dual class I/IV phospho-
inositide 3-kinase (PI3K) inhibitors. Curr Med Chem 18(18):2686–2714
108. Mallon R, Feldberg LR, Lucas J, Chaudhary I, Dehnhardt C, Santos ED et al (2011)
Antitumor efficacy of PKI-587, a highly potent dual PI3K/mTOR kinase inhibitor. Clin
Cancer Res 17(10):3193–3203 (Epub 2011 Feb 15)
109. Yuan J, Mehta PP, Yin MJ, Sun S, Zou A, Chen J et al (2011) PF-04691502, a potent and
selective oral inhibitor of PI3K and mTOR kinases with antitumor activity. Mol Cancer Ther
10(11):2189–2199
110. Mallon R, Hollander I, Feldberg L, Lucas J, Soloveva V, Venkatesan A et al (2010)
Antitumor efficacy profile of PKI-402, a dual phosphatidylinositol 3-kinase/mammalian tar-
get of rapamycin inhibitor. Mol Cancer Ther 9(4):976–984
111. Prasad G, Sottero T, Yang X, Mueller S, James CD, Weiss WA et al (2011) Inhibition of
PI3K/mTOR pathways in glioblastoma and implications for combination therapy with temo-
zolomide. Neuro Oncol 13(4):384–392
112. Mirzoeva OK, Hann B, Hom YK, Debnath J, Aftab D, Shokat K et al (2011) Autophagy
suppression promotes apoptotic cell death in response to inhibition of the PI3K-mTOR
pathway in pancreatic adenocarcinoma. J Mol Med 89(9):877–889
113. Heffron TP, Berry M, Castanedo G, Chang C, Chuckowree I, Dotson J et al (2010)
Identification of GNE-477, a potent and efficacious dual PI3K/mTOR inhibitor. Bioorg Med
Chem Lett 20(8):2408–2411
114. Wallin JJ, Edgar KA, Guan J, Berry M, Prior WW, Lee L et al (2011) GDC-0980 is a novel
class I PI3K/mTOR kinase inhibitor with robust activity in cancer models driven by the
PI3K pathway. Mol Cancer Ther 10(12):2426–2436
115. Li T, Wang J, Wang X, Yang N, Chen SM, Tong LJ et al (2010) WJD008, a dual phos-
phatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin inhibitor, prevents PI3K
signaling and inhibits the proliferation of transformed cells with oncogenic PI3K mutant. J
Pharmacol Exp Ther 334(3):830–838
116. Zhu J, Huang JW, Tseng PH, Yang YT, Fowble J, Shiau CW et al (2004) From the cycloox-
ygenase-2 inhibitor celecoxib to a novel class of 3-phosphoinositide-dependent protein
kinase-1 inhibitors. Cancer Res 64(12):4309–4318
117. Ding H, Han C, Guo D, Wang D, Duan W, Chen CS et al (2008) Sensitivity to the non-COX
inhibiting celecoxib derivative, OSU03012, is p21(WAF1/CIP1) dependent. Int J Cancer
123(12):2931–2938
13 New Agents and Approaches for Targeting 369
118. Falasca M, Chiozzotto D, Godage HY, Mazzoletti M, Riley AM, Previdi S et al (2010) A
novel inhibitor of the PI3K/Akt pathway based on the structure of inositol 1,3,4,5,6-pentaki-
sphosphate. Br J Cancer 102(1):104–114
119. Yang L, Dan HC, Sun M, Liu Q, Sun XM, Feldman RI et al (2004) Akt/protein kinase B
signaling inhibitor-2, a selective small molecule inhibitor of Akt signaling with antitumor
activity in cancer cells overexpressing Akt. Cancer Res 64(13):4394–4399
120. Garrett CR, Coppola D, Wenham RM, Cubitt CL, Neuger AM, Frost TJ et al (2011) Phase
I pharmacokinetic and pharmacodynamic study of triciribine phosphate monohydrate, a
small-molecule inhibitor of AKT phosphorylation, in adult subjects with solid tumors con-
taining activated AKT. Invest New Drugs 29(6):1381–1389
121. Tan S, Ng Y, James DE (2011) Next-generation Akt inhibitors provide greater specificity:
effects on glucose metabolism in adipocytes. Biochem J 435(2):539–544
122. Simioni C, Neri LM, Tabellini G, Ricci F, Bressanin D, Chiarini F et al (2012) Cytotoxic
activity of the novel Akt inhibitor, MK-2206, in T-cell acute lymphoblastic leukemia.
Leukemia 22(10):136
123. Rhodes N, Heerding DA, Duckett DR, Eberwein DJ, Knick VB, Lansing TJ et al (2008)
Characterization of an Akt kinase inhibitor with potent pharmacodynamic and antitumor
activity. Cancer Res 68(7):2366–2374
124. Zeng Z, Samudio IJ, Zhang W, Estrov Z, Pelicano H, Harris D et al (2006) Simultaneous
inhibition of PDK1/AKT and Fms-like tyrosine kinase 3 signaling by a small-molecule
KP372-1 induces mitochondrial dysfunction and apoptosis in acute myelogenous leukemia.
Cancer Res 66(7):3737–3746
125. Rampling R, Sanson M, Gorlia T, Lacombe D, Lai C, Gharib M et al (2012) A phase I
study of LY317615 (enzastaurin) and temozolomide in patients with gliomas (EORTC trial
26054). Neuro Oncol 14(3):344–350
126. Vansteenkiste J, Ramlau R, von Pawel J, San Antonio B, Eschbach C, Szczesna A
(2012) A phase II randomized study of cisplatin-pemetrexed plus either enzastaurin or
placebo in chemonaive patients with advanced non-small cell lung cancer. Oncology
82(1):25–29
127. Wolff RA, Fuchs M, Di Bartolomeo M, Hossain AM, Stoffregen C, Nicol S et al (2011)
A double-blind, randomized, placebo-controlled, phase 2 study of maintenance enzastaurin
with 5-fluorouracil/leucovorin plus bevacizumab after first-line therapy for metastatic colo-
rectal cancer. Cancer 27(10):26692
128. Kondapaka SB, Singh SS, Dasmahapatra GP, Sausville EA, Roy KK (2003) Perifosine,
a novel alkylphospholipid, inhibits protein kinase B activation. Mol Cancer Ther
2(11):1093–1103
129. Pal SK, Reckamp K, Yu H, Figlin RA (2010) Akt inhibitors in clinical development for the
treatment of cancer. Expert Opin Investig Drugs 19(11):1355–1366
130. Handrick R, Rubel A, Faltin H, Eibl H, Belka C, Jendrossek V (2006) Increased cytotoxicity
of ionizing radiation in combination with membrane-targeted apoptosis modulators involves
downregulation of protein kinase B/Akt-mediated survival-signaling. Radiother Oncol
80(2):199–206
131. Martelli AM, Papa V, Tazzari PL, Ricci F, Evangelisti C, Chiarini F et al (2010)
Erucylphosphohomocholine, the first intravenously applicable alkylphosphocholine, is cyto-
toxic to acute myelogenous leukemia cells through JNK- and PP2A-dependent mechanisms.
Leukemia 24(4):687–698
132. Bidyasar S, Kurzrock R, Falchook GS, Naing A, Wheler JJ, Durand J et al (2009) A first-in-
human phase I trial of PBI-05204 (oleandrin), an inhibitor of Akt, FGF-2, NF-Kb, and p70S6K
in advanced solid tumor patients. ASCO Meeting Abstracts. 27(15S):3537 (2009 June 8)
133. Dunn DE, He DN, Yang P, Johansen M, Newman RA, Lo DC (2011) In vitro and in vivo
neuroprotective activity of the cardiac glycoside oleandrin from Nerium oleander in brain
slice-based stroke models. J Neurochem 119(4):805–814
134. Yoon H, Kim DJ, Ahn EH, Gellert GC, Shay JW, Ahn CH et al (2009) Antitumor activity of
a novel antisense oligonucleotide against Akt1. J Cell Biochem 108(4):832–838
370 J. A. McCubrey et al.
154. Garcia-Martinez JM, Moran J, Clarke RG, Gray A, Cosulich SC, Chresta CM et al (2009)
Ku-0063794 is a specific inhibitor of the mammalian target of rapamycin (mTOR).
Biochem J 421(1):29–42
155. Falcon BL, Barr S, Gokhale PC, Chou J, Fogarty J, Depeille P et al (2011) Reduced VEGF
production, angiogenesis, and vascular regrowth contribute to the antitumor properties of
dual mTORC1/mTORC2 inhibitors. Cancer Res 71(5):1573–1583
156. Liu Q, Wang J, Kang SA, Thoreen CC, Hur W, Ahmed T et al (2011) Discovery of
9-(6-aminopyridin-3-yl)-1-(3-(trifluoromethyl)phenyl)benzo[h][1,6]naphthyridin-2(1H)-one
(Torin2) as a potent, selective, and orally available mammalian target of rapamycin (mTOR)
inhibitor for treatment of cancer. J Med Chem 54(5):1473–1480
157. Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R et al (2008) Effective
use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine
lung cancers. Nat Med 14(12):1351–1356
158. Wang Z, Zhou J, Fan J, Qiu SJ, Yu Y, Huang XW et al (2008) Effect of rapamycin alone and
in combination with sorafenib in an orthotopic model of human hepatocellular carcinoma.
Clin Cancer Res 14(16):5124–5130
159. Jin N, Jiang T, Rosen DM, Nelkin BD, Ball DW (2011) Synergistic action of a RAF inhibi-
tor and a dual PI3K/mTOR inhibitor in thyroid cancer. Clin Cancer Res 17(20):6482–6489
160. Legrier ME, Yang CP, Yan HG, Lopez-Barcons L, Keller SM, Perez-Soler R et al (2007)
Targeting protein translation in human non small cell lung cancer via combined MEK and
mammalian target of rapamycin suppression. Cancer Res 67(23):11300–11308
161. Marshall G, Howard Z, Dry J, Fenton S, Heathcote D, Gray N et al (2011) Benefits of
mTOR kinase targeting in oncology: pre-clinical evidence with AZD8055. Biochem Soc
Trans 39(2):456–459
162. Shapiro G, LoRusso P, Kwak EL, Cleary JM, Musib L, Jones C et al (2011) Clinical
combination of the MEK inhibitor GDC-0973 and the PI3 K inhibitor GDC-0941: A
first-in-human phase Ib study testing daily and intermittent dosing schedules in patients
with advanced solid tumors. ASCO Meeting Abstracts. 29(15_suppl):3005 (2011
June 9)
163. Yang H, Higgins B, Kolinsky K, Packman K, Bradley WD, Lee RJ et al (2012) Antitumor
activity of BRAF inhibitor vemurafenib in preclinical models of BRAF-mutant colorectal
cancer. Cancer Res 72(3):779–789
164. Hirai H, Sootome H, Nakatsuru Y, Miyama K, Taguchi S, Tsujioka K et al (2010)
MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chem-
otherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol Cancer Ther
9(7):1956–1967
165. Baumann P, Mandl-Weber S, Oduncu F, Schmidmaier R (2009) The novel orally bioavailable
inhibitor of phosphoinositol-3-kinase and mammalian target of rapamycin, NVP-BEZ235, inhib-
its growth and proliferation in multiple myeloma. Exp Cell Res 315(3):485–497
166. Manara MC, Nicoletti G, Zambelli D, Ventura S, Guerzoni C, Landuzzi L et al (2010) NVP-
BEZ235 as a new therapeutic option for sarcomas. Clin Cancer Res 16(2):530–540
167. Zhang YJ, Duan Y, Zheng XF (2011) Targeting the mTOR kinase domain: the second
generation of mTOR inhibitors. Drug Discov Today 16(7–8):325–331
168. Gravina GL, Marampon F, Petini F, Biordi L, Sherris D, Jannini EA et al (2011) The
TORC1/TORC2 inhibitor, Palomid 529, reduces tumor growth and sensitizes to docetaxel
and cisplatin in aggressive and hormone-refractory prostate cancer cells. Endocr Relat
Cancer 18(4):385–400
169. Diaz R, Nguewa PA, Diaz-Gonzalez JA, Hamel E, Gonzalez-Moreno O, Catena R et al
(2009) The novel Akt inhibitor Palomid 529 (P529) enhances the effect of radiotherapy in
prostate cancer. Br J Cancer 100(6):932–940
170. Chung EJ, Brown AP, Asano H, Mandler M, Burgan WE, Carter D et al (2009) In vitro
and in vivo radiosensitization with AZD6244 (ARRY-142886), an inhibitor of mitogen-acti-
vated protein kinase/extracellular signal-regulated kinase 1/2 kinase. Clin Cancer Res 15(9):
3050–3057 (Epub 2009 Apr 14)
372 J. A. McCubrey et al.
171.
Edwards E, Geng L, Tan J, Onishko H, Donnelly E, Hallahan DE (2002)
Phosphatidylinositol 3-kinase/Akt signaling in the response of vascular endothelium to ion-
izing radiation. Cancer Res 62(16):4671–4677
172. Shinohara ET, Cao C, Niermann K, Mu Y, Zeng F, Hallahan DE et al (2005) Enhanced radi-
ation damage of tumor vasculature by mTOR inhibitors. Oncogene 24(35):5414–5422
173. Paglin S, Lee NY, Nakar C, Fitzgerald M, Plotkin J, Deuel B et al (2005) Rapamycin-
sensitive pathway regulates mitochondrial membrane potential, autophagy, and survival in
irradiated MCF-7 cells. Cancer Res 65(23):11061–11070
174. Moretti L, Attia A, Kim KW, Lu B (2007) Crosstalk between Bak/Bax and mTOR signaling
regulates radiation-induced autophagy. Autophagy 3(2):142–144
175. Kudchadkar R, Paraiso KH, Smalley KS (2012) Targeting mutant BRAF in mela-
noma: current status and future development of combination therapy strategies. Cancer J
18(2):124–131
Chapter 14
Activation of Immune-Mediated Tumor Cell
Death by Chemotherapy
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 373
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0_14,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
374 M. J. McCoy et al.
14.1 Introduction
which migrate to the tumor site and kill tumor cells displaying the relevant anti-
gen via perforin/granzyme-induced apoptosis or ligation of death receptors (cross-
priming), or are tolerized and subsequently deleted (cross-tolerization) [11].
There are several lines of evidence that suggests CD8+ T cells are critical to the
development of anti-tumor immunity. Depletion of CD8+ T cells prevents the
rejection of chemically or UV-induced tumors, or transplanted tumors formed
in immuno-compromised hosts, all of which are highly immunogenic in normal
mice [12–14]. Mice deficient in the cytolytic molecule perforin are not only more
susceptible to chemically induced tumor formation [15], but also develop spon-
taneous lymphoma in later life [13]. While perforin is required for both CTL and
natural killer (NK) cell cytotoxicity, Smyth et al., demonstrated that CTL were
essential for protection from lymphoma, since tumors that developed in perforin
knockout mice were rejected when transplanted into wild-type mice depleted of
NK cells, but not in those depleted of CD8+ T cells. The efficacy of anti-tumor
immunotherapy has also been shown to be dependent on the presence of CD8+
T cells in a variety of murine models [16–19]. In humans, tumor infiltration by
CD8+ T cells has been associated with improved prognosis in many different can-
cers, as will be discussed in more detail later in this chapter.
The role of CD4+ T cells in generating an anti-tumor immune response is not
so well understood. Traditionally, activated CD4+ T cells have been divided into
two major subsets according to their cytokine secretion profile. T helper 1 (Th1)
cells secrete cytokines usually associated with inflammation including interleu-
kin-2 (IL-2), interferon gamma (IFNγ), tumor necrosis factor alpha (TNFα), and
interleukin-12 (IL-12) and classically promote cell-mediated immunity against
intracellular bacteria and viruses. T helper 2 (Th2) cells generally promote
humoral immunity against extracellular pathogens and secrete IL-10, IL-4, and
transforming growth factor beta (TGFβ) [20, 21] More recently, a third subset of
T helper cells, characterized by secretion of the pro-inflammatory cytokines IL-17
and IL-22, has been defined. Termed Th17 cells, this subset protects against a vari-
ety of pathogens including intracellular and extracellular bacteria [22]. However,
with CD4+ T cells often showing non-classical cytokine expression profiles and
increasing evidence that the tissue in which an immune response occurs, as well
as the pathogen invading it, plays a major role in fine-tuning the class of response
[23], the division of activated CD4+ T cells into distinct helper subsets is perhaps
less meaningful than once thought.
As their name suggests, CD4+ helper T cells support CD8+ T-cell-driven
responses, not only through IL-2 production, which is required for T-cell expan-
sion, but also through the licensing of APC via CD40–CD40L interactions
[24–27]. Interaction of the co-stimulatory molecule CD40 (expressed on APC)
with its ligand CD40L (expressed on CD4+ T cells and mast cells) results in the
14 Activation of Immune-Mediated Tumor Cell Death by Chemotherapy 377
γδ T cells, suggests that such a clear division between the two arms of the immune
system may be overly simplistic [41].
Research into immunity against infection has shown that the generation of a
potent adaptive immune response is usually dependent upon the initial stimulation
of a strong innate immune response [42–44]. Consistent with this finding, it is now
becoming clear that the generation of an effective anti-tumor immune response
involves a complex interplay between the innate and adaptive arms of the immune
system. In the early stages of neoplasia, it seems that NK cells play a vital role
in the recognition of cancerous and precancerous cells. NK cells detect cell stress
markers through receptors such as NK group 2 member D (NKG2D), and reduced
MHC class I expression (lack of “self”), both of which are a common feature of
malignant cells. NK cells, like CTL, can exert direct anti-tumor activity through
secretion of perforin and granzymes, and via Fas and tumor necrosis factor-related
apoptosis inducing ligand (TRAIL)-mediated cell death [45–48]. IFNγ secre-
tion by NK cells stimulates DC maturation [49] and the induction of chemokines
including IFN-inducible protein-10 (IP-10), monokine induced by IFNγ (MIG)
and interferon-inducible T-cell α chemoattractant (I-TAC) [50, 51], which pro-
motes lymphocyte recruitment. Mature, activated DC are then able to migrate to
the draining lymph node, where they can cross-present antigen to tumor-specific
CD8+ T cells.
Type I IFN (IFNα/β) are known to play an important role in the initiation of an
innate immune response [52]. Injection of activated plasmacytoid DC (pDC) has
been shown to induce NK and CD8+ T-cell-dependent rejection of transplanted
tumors in a mouse melanoma model through secretion of type I IFN [53], illus-
trating the close collaboration between innate and adaptive immunity in generat-
ing an effective anti-tumor immune response. However, mice deficient in both an
adaptive immune system and type I and type II IFN signaling (RAG2−/− STAT−/−
double knockout mice) are more susceptible to spontaneous tumor development
than either RAG2−/− or STAT−/− single knockouts [54]. This suggests that while
both the innate and adaptive arms of the immune system are required for optimal
immunosurveillance, each plays a partially independent role.
There are therefore three vital sequential steps in the generation of an anti-tumor
immune response. Firstly, tumor cells must be recognized by innate and/or adap-
tive immune effectors. Secondly, recognition of tumor cells must lead to activa-
tion of an immune response rather than tolerance. Thirdly, immune effectors must
effectively kill the tumor cells. Interruption of any one of these three steps can lead
to immuno subversion and uncontrolled tumor growth, that is, transition from the
elimination phase to escape. Tumors employ many mechanisms to subvert anti-
tumor immunity (Table 14.1). One key means by which tumors avoid recognition
by the immune system is through loss or reduced expression of tumor antigens.
14 Activation of Immune-Mediated Tumor Cell Death by Chemotherapy 379
This has been observed in human and mouse cell lines resistant to CTL lysis
in vitro [55, 56], in outgrowing tumors following immunotherapy in mouse mod-
els [57, 58] and in recurrent melanoma in patients treated with antigen-specific
immunotherapy [59]. Aberrant human leukocyte antigen (HLA) expression, which
impedes direct recognition by CD8+ T cells, has also been detected in a variety
of human cancers (reviewed in [60]), and human tumor cell lines lacking HLA
class I expression evade CTL-mediated cell death in vitro [61]. Both loss of tumor
antigen and HLA expression likely represent passive selection rather than active
adaptation, reflecting survival of the least immunogenic cells.
380 M. J. McCoy et al.
Secretion of soluble factors including IL-10 and TGFβ by tumors can skew the
immune system toward tolerance rather than active immunity. IL-10 can inhibit
DC recruitment [62], while TGFβ prevents the cross-priming of CD8+ T cells [63]
and promotes the generation of Treg [64]. Tumor cells can also express inhibitory
molecules on their surface, which negatively regulate T-cell function. For exam-
ple, a variety of human tumors express programed death ligand-1 (PDL-1), which
inhibits T-cell proliferation and cytokine production through interaction with its
receptor, programed death-1 (PD-1), expressed by activated T cells [65, 66].
Finally, tumor cell evolution can also select for cells less susceptible to CTL
and NK cell-mediated cell death. Mutations in death receptors including Fas and
TRAIL receptors, DR4 and DR5, have been observed in B-cell lymphoma [67]
and in metastatic breast cancer [68]. Interestingly, tumor cells have also been
found to express Fas ligand (Fas-L) [69], creating a role reversal situation in which
tumor cells can induce apoptotic cell death in activated T cells.
While original evidence for the existence of anti-tumor immunity came from ani-
mal models, this has now been corroborated by data demonstrating that anti-tumor
immune responses can be detected in cancer patients, even in the context of pro-
gressive disease, and that such responses have clinical significance.
Melanoma is considered one of the most immunogenic cancers with tumor-
associated antigen (TAA)-specific CD8+ T cells detectable in around 50 % of
patients [70]. Vitiligo-like hypopigmentation, thought to result from the destruc-
tion of normal melanocytes by autoantibodies and autoreactive T cells, is also
observed in melanoma patients and is associated with improved outcome [71].
While less frequently, circulating TAA-specific T cells have now been detected
in most cancer types [72–77]. Cancer patients are also commonly found to have
an increased proportion of peripheral Treg, consistent with the idea of an under-
lying anti-tumor immune response blocked by the concurrent expansion of Treg
[78–81].
Perhaps, the most convincing evidence for the clinical relevance of anti-tumor
immunity in patients is the strong prognostic significance of the balance of T-cell
subsets within the local tumor environment. Tumor infiltration by CD8+ T cells
has been associated with an improved prognosis in many cancer types including
ovarian [82], colorectal [83], esophageal [84], and lung [85]. Conversely, tumor-
infiltrating Treg predict poorer survival in most solid cancers [86–89], again
consistent with the suppressive role played by this T-cell subset. An exception,
however, appears to be colorectal cancer, where a higher density of tumor-infiltrat-
ing (Ti) Treg has been identified as a good prognostic factor in several independ-
ent studies [90–93]. One explanation for this seemingly paradoxical finding is that
expansion of Treg within the tumor occurs in response to the activation and expan-
sion of tumor-specific CD8+ T cells, thereby representing an indirect measure of
14 Activation of Immune-Mediated Tumor Cell Death by Chemotherapy 381
an anti-tumor immune response. Alternatively, it has been suggested that Treg may
limit the tumorigenic effects of Th17 cell-mediated inflammation [94]. In support
of this, Th17 cells are more frequently identified in colorectal tumors than other
cancer types, likely due to the abundance of pathogens present in the gut [95], and
a lower density of Ti-Th17 cells has recently been associated with improved dis-
ease-free survival [96]. Ti-Treg have also been associated with improved outcome
in nasopharyngeal and head and neck squamous cell carcinoma [97, 98], both of
which also develop in a non-sterile environment. It may be therefore that it is the
balance of T-cell subsets within a tumor, in relation to the stage of tumor develop-
ment and the environment in which this occurs, that is important. In any case, this
serves to highlight the complexity of the relationship between the immune system
and a developing tumor and the potential value of harnessing anti-tumor immunity
to improve the efficacy of cancer therapy.
The major classes of cytotoxic drugs currently used for the treatment of cancer
include anti-metabolites, anti-folates, alkylating agents, topoisomerase inhibi-
tors, anthracyclines, vinca alkaloids, and taxanes (Table 14.2). While differing
in their precise mode of action, most interfere with DNA synthesis resulting
in apoptotic tumor cell death. The anti-metabolites gemcitabine and 5-fluoro-
uracil (5-FU), for example, are synthetic nucleoside analogues, which upon
intracellular conversion to their active forms, cause apoptosis by two different
mechanisms. They are incorporated into DNA (and RNA in the case of 5-FU),
disrupting DNA synthesis and/or RNA processing and function, and also inhibit
enzymes essential for DNA synthesis and repair [99, 100]. Alkylating agents,
including cyclophosphamide, and platinum-based drugs such as cisplatin and
oxaliplatin, cross-link DNA, inhibiting DNA synthesis and RNA transcription,
disrupting the cell cycle and activating several DNA damage-mediated signal
transduction pathways, leading to apoptosis [101, 102]. Anthracycline antibi-
otics, including doxorubicin, idarubicin and epirubicin also intercalate DNA/
RNA base pairs, but primarily cause apoptotic cell death through inhibition of
topoisomerase II activity, which is vital for DNA structuring and repair [103].
Vinca alkaloids (e.g., vinorelbine and vinblastine) and taxanes (paclitaxel and
docetaxel) differ in their mode of action in that they inhibit mitosis rather than
DNA synthesis through stabilizing or destabilizing microtubules and, thereby
preventing cell cycle progression [104]. However, like the other cytotoxics men-
tioned, this ultimately results in apoptosis. Most commonly used chemotherapy
drugs are therefore not specifically toxic to tumor cells, but target all dividing
cells, including lymphocytes. Consequently, chemotherapy has traditionally
382 M. J. McCoy et al.
Fig. 14.2 Mechanisms by which chemotherapy can alter the level and/or context of antigen
presentation and the immunological milieu to promote anti-tumor immunity: 1 Physical destruc-
tion of the tumor-releasing antigen into the cross-presentation pathway. 2 Induction of “eat me”
and “danger” signals from dying tumor cells including CRT translocation and HMGB1/ATP
release. 3 Upregulation of tumor or cell-surface receptors including MHC class I and NKG2D. 4
Promotion of DC expansion and activation. 5 Homeostatic T-cell proliferation. 6 Selective Treg
depletion. 7 Selective B-cell depletion. ATP adenosine triphosphate, CRT calreticulin, CTL cyto-
toxic T lymphocyte, DC dendritic cell, HMGB1 high-mobility group box 1, MHC major histo-
compatibility complex, NK natural killer, Treg regulatory T cell
Perhaps, the most obvious way by which chemotherapy could increase the level
of antigen presentation, is through physical destruction of the tumor, releas-
ing antigens from the dead and dying cells for entry into the cross-presentation
384 M. J. McCoy et al.
pathway. It is likely that the availability of some tumor antigens, normally pre-
sent at levels too low to induce an immune response, could be increased above
the required threshold in this process. In our murine model of mesothelioma,
using hemagglutinin (HA) antigen-transduced tumor cells (AB1-HA) injected
subcutaneously, gemcitabine significantly increases the level of antigen pres-
entation to tumor-specific CD8+ T cells for a given tumor size [108]. For
example, proliferation of adoptively transferred HA-specific CD8+ T cells in
a mouse bearing a 40 mm2 tumor treated with gemcitabine was almost dou-
ble the level observed in animals with an untreated tumor of equivalent size.
Vaccination with the HA antigen-bearing PR8 virus following gemcitabine
treatment resulted in a marked decrease in tumor growth and increased sur-
vival compared with vaccination without gemcitabine. It is also plausible that
so-called cryptic antigens, not usually visible to the immune system, could be
released following chemotherapy-induced cell death, along with many of the
~90 neoepitopes generated by point mutations that are harbored by the average
tumor [109]. In support of this, we found that animals cured of AB1-HA tumors
following combination chemo-immunotherapy with gemcitabine and the CD40
activating antibody FGK45 were resistant to rechallenge with both AB1-HA
and non-transfected AB1 tumor cells, demonstrating memory to tumor antigens
other than HA [110].
With regard to altering the context of antigen presentation, chemotherapy can
induce the expression or release of molecules associated with dangerous, and
therefore immunogenic, cell death [111, 112]. Using an assay in which tumor
cells are exposed to cytotoxic drugs in vitro, injected into immunocompetent
naive mice, and assessed for their capacity to elicit a protective immune response
against rechallenge with live tumor cells, Zitvogel, Kroemer, and colleagues
have demonstrated that oxaliplatin and the anthracyclines doxorubicin, idaru-
bicin, and mitoxantrone, induce immunogenic cell death in several tumor mod-
els [105, 113–115]. Characteristics of immunogenic versus non-immunogenic
tumor cell death in their system include cell-surface exposure of the calcium
ion-binding protein, calreticulin [115], secretion of the non-histone chromatin-
binding nuclear constituent, high-mobility group box 1 (HMGB1) [113], and
release of adenosine triphosphate (ATP) [105]. Calreticulin is usually seques-
tered within the endoplasmic reticulum. Its translocation to the cell surface
occurs before cells become apoptotic and is required for uptake of dying tumor
cells by DC [115]. Blockade of cell-surface calreticulin with specific antibodies,
or knockdown of its expression with siRNA, inhibited phagocytosis of anthra-
cycline-treated tumor cells, while addition of exogenous recombinant calreticu-
lin, enabled uptake of treated tumor cells transfected with siRNA [115]. Whereas
calreticulin exposure therefore provides an “eat me” signal for DC, binding of
HMGB1 to toll-like receptor 4 (TLR4) on DC, and subsequent signaling through
the MyD88 pathway is thought to represent a danger signal required to induce
antigen processing and presentation. Mice deficient in TLR4 or the MyD88
adapter molecule were unable to mount a tumor-specific immune response fol-
lowing vaccination with oxaliplatin or doxorubicin-treated tumor cells and were
14 Activation of Immune-Mediated Tumor Cell Death by Chemotherapy 385
14.4.1 Combination Chemoimmunotherapy
While cytotoxic chemotherapy is still the mainstay of most cancer therapy, the
emphasis is now shifting from a one size fits all approach to more individualized
treatment strategies. Many factors contribute to chemotherapy efficacy and these
390 M. J. McCoy et al.
pertain as much to the tumor and to the host as to intrinsic properties of the drug.
Not least of these factors is the potential for development of an anti-tumor immune
response, and the capacity for chemotherapy to induce or enhance this. Patients
whose cancers have “druggable” targets may be treated with novel-targeted thera-
pies, and our understanding of the interaction between targeted therapies and the
immune response is even less mature. Nevertheless, many of these patients will
also have chemotherapy before or after treatment with a targeted agent.
Chemoimmunotherapy using carefully selected drug combinations rep-
resents a promising treatment option. However, it is likely that this strategy
will be most effective when used in an individualized setting. Adjunct immu-
notherapies may need to be tailored not only to the type of chemotherapy, but
also to the immunogenicity of the patient’s tumor (expression of tumor anti-
gens) and any defects in the patient’s immune system (e.g., TLR4/P2xr7 muta-
tions). Measurement of immunological parameters prior to initiating therapy
may prove a useful tool in predicting which patients may benefit from adjunct
immunotherapy and in selecting the type of immunotherapy to administer, for
example, tumor-specific CTL stimulating or Treg depleting. More work needs
to be done to determine how the precise balance of T-cell subsets within the
local tumor environment before and after chemotherapy affects response, and
whether assessment of peripheral blood T-cell subsets could potentially repre-
sent a less invasive surrogate measure, which could be more easily translated
into the clinic.
14.5 Conclusions
References
1. Burnet M (1957) Cancer: a biological approach. III. Viruses associated with neoplastic con-
ditions. IV. Practical applications. Br Med J 1:841–847
2. Dunn GP, Bruce AT, Ikeda H et al (2002) Cancer immunoediting: from immunosurveillance
to tumor escape. Nat Immunol 3:991–998
3. Neller MA, Lopez JA, Schmidt CW (2008) Antigens for cancer immunotherapy. Semin
Immunol 20:286–295
4. Huang AY, Golumbek P, Ahmadzadeh M et al (1994) Role of bone marrow-derived cells in
presenting MHC class I-restricted tumor antigens. Science 264:961–965
5. Marzo AL, Lake RA, Lo D et al (1999) Tumor antigens are constitutively presented in the
draining lymph nodes. J Immunol 162:5838–5845
6. van Mierlo GJ, Boonman ZF, Dumortier HM et al (2004) Activation of dendritic cells that
cross-present tumor-derived antigen licenses CD8 + CTL to cause tumor eradication. J
Immunol 173:6753–6759
7. Rock KL, York IA, Goldberg AL (2004) Post-proteasomal antigen processing for major his-
tocompatibility complex class I presentation. Nat Immunol 5:670–677
8. Watts C (2004) The exogenous pathway for antigen presentation on major histocompatibil-
ity complex class II and CD1 molecules. Nat Immunol 5:685–692
9. Bevan MJ (1976) Cross-priming for a secondary cytotoxic response to minor H antigens with H-2
congenic cells which do not cross-react in the cytotoxic assay. J Exp Med 143:1283–1288
10. Lin ML, Zhan Y, Villadangos JA et al (2008) The cell biology of cross-presentation and the
role of dendritic cell subsets. Immunol Cell Biol 86:353–362
11. van der Most RG, Currie A, Robinson BWS et al (2006) Cranking the immunologic engine
with chemotherapy: using context to drive tumor antigen cross-presentation towards useful
antitumor immunity. Cancer Res 66:601–604
12. Boesen M, Svane IM, Engel AM et al (2000) CD8 + T cells are crucial for the ability of
congenic normal mice to reject highly immunogenic sarcomas induced in nude mice with
3-methylcholanthrene. Clin Exp Immunol 121:210–215
13. Smyth MJ, Thia KY, Street SE et al (2000) Perforin-mediated cytotoxicity is critical for sur-
veillance of spontaneous lymphoma. J Exp Med 192:755–760
14. Ward PL, Koeppen HK, Hurteau T et al (1990) Major histocompatibility complex class I
and unique antigen expression by murine tumors that escaped from CD8 + T-cell-dependent
surveillance. Cancer Res 50:3851–3858
15. van den Broek ME, Kagi D, Ossendorp F et al (1996) Decreased tumor surveillance in per-
forin-deficient mice. J Exp Med 184:1781–1790
16. Broomfield SA, van der Most RG, Prosser AC et al (2009) Locally administered TLR7 ago-
nists drive systemic antitumor immune responses that are enhanced by anti-CD40 immuno-
therapy. J Immunol 182:5217–5224
17. Serba S, Schmidt J, Wentzensen N et al (2008) Transfection with CD40L induces tumour
suppression by dendritic cell activation in an orthotopic mouse model of pancreatic adeno-
carcinoma. Gut 57:344–351
18. Slos P, De Meyer M, Leroy P et al (2001) Immunotherapy of established tumors in mice by
intratumoral injection of an adenovirus vector harboring the human IL-2 cDNA: induction
of CD8(+) T-cell immunity and NK activity. Cancer Gene Ther 8:321–332
19. Sutmuller RP, van Duivenvoorde LM, van Elsas A et al (2001) Synergism of cytotoxic
T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells
in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic
T lymphocyte responses. J Exp Med 194:823–832
20. Constant SL, Bottomly K (1997) Induction of Th1 and Th2 CD4 + T cell responses: the
alternative approaches. Annu Rev Immunol 15:297–322
21. Pulendran B, Palucka K, Banchereau J (2001) Sensing pathogens and tuning immune
responses. Science 293:253–256
392 M. J. McCoy et al.
22. Martinez GJ, Nurieva RI, Yang XO et al (2008) Regulation and function of proinflammatory
TH17 cells. Ann N Y Acad Sci 1143:188–211
23. Matzinger P, Kamala T (2011) Tissue-based class control: the other side of tolerance. Nat
Rev Immunol 11:221–230
24. Bennett SR, Carbone FR, Karamalis F et al (1998) Help for cytotoxic-T-cell responses is
mediated by CD40 signalling. Nature 393:478–480
25. Lanzavecchia A (1998) Immunology. Licence to kill. Nature 393:413–414
26. Ridge JP, Di Rosa F, Matzinger P (1998) A conditioned dendritic cell can be a temporal
bridge between a CD4 + T-helper and a T-killer cell. Nature 393:474–478
27. Schoenberger SP, Toes RE, van der Voort EI et al (1998) T-cell help for cytotoxic T lympho-
cytes is mediated by CD40-CD40L interactions. Nature 393:480–483
28. Bennett SR, Carbone FR, Karamalis F et al (1997) Induction of a CD8 + cytotoxic T lymphocyte
response by cross-priming requires cognate CD4 + T cell help. J Exp Med 186:65–70
29. Ossendorp F, Mengede E, Camps M et al (1998) Specific T helper cell requirement for opti-
mal induction of cytotoxic T lymphocytes against major histocompatibility complex class II
negative tumors. J Exp Med 187:693–702
30. Quezada SA, Simpson TR, Peggs KS et al (2010) Tumor-reactive CD4 + T cells develop
cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic
hosts. J Exp Med 207:637–650
31. Hung K, Hayashi R, Lafond-Walker A et al (1998) The central role of CD4(+) T cells in the
antitumor immune response. J Exp Med 188:2357–2368
32. Currie AJ, Prosser A, McDonnell A et al (2009) Dual control of antitumor CD8 T cells
through the programmed death-1/programmed death-ligand 1 pathway and immunosuppres-
sive CD4 T cells: regulation and counterregulation. J Immunol 183:7898–7908
33. Belkaid Y, Rouse BT (2005) Natural regulatory T cells in infectious disease. Nat Immunol
6:353–360
34. Sakaguchi S, Yamaguchi T, Nomura T et al (2008) Regulatory T cells and immune toler-
ance. Cell 133:775–787
35. Zou W (2006) Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol
6:295–307
36. Murugaiyan G, Saha B (2009) Protumor vs antitumor functions of IL-17. J Immunol
183:4169–4175
37. Bennett SR, Carbone FR, Toy T et al (1998) B cells directly tolerize CD8(+) T cells. J Exp
Med 188:1977–1983
38. Qin Z, Richter G, Schuler T et al (1998) B cells inhibit induction of T cell-dependent tumor
immunity. Nat Med 4:627–630
39. Mueller DL, Jenkins MK, Schwartz RH (1989) Clonal expansion versus functional clonal
inactivation: a costimulatory signalling pathway determines the outcome of T cell antigen
receptor occupancy. Annu Rev Immunol 7:445–480
40. Medzhitov R, Janeway C Jr (2000) Innate immunity. New Engl J Med 343:338–344
41. Borghesi L, Milcarek C (2007) Innate versus adaptive immunity: a paradigm past its prime?
Cancer Res 67:3989–3993
42. Dalod M, Hamilton T, Salomon R et al (2003) Dendritic cell responses to early murine cyto-
megalovirus infection: subset functional specialization and differential regulation by inter-
feron alpha/beta. J Exp Med 197:885–898
43. Fearon DT, Locksley RM (1996) The instructive role of innate immunity in the acquired
immune response. Science 272:50–53
44. Megjugorac NJ, Young HA, Amrute SB et al (2004) Virally stimulated plasmacytoid den-
dritic cells produce chemokines and induce migration of T and NK cells. J Leukoc Biol
75:504–514
45. Bradley M, Zeytun A, Rafi-Janajreh A et al (1998) Role of spontaneous and interleukin-
2-induced natural killer cell activity in the cytotoxicity and rejection of Fas + and Fas-
tumor cells. Blood 92:4248–4255
14 Activation of Immune-Mediated Tumor Cell Death by Chemotherapy 393
46. Pardo J, Balkow S, Anel A et al (2002) Granzymes are essential for natural killer cell-mediated
and perf-facilitated tumor control. Eur J Immunol 32:2881–2887
47. Takeda K, Hayakawa Y, Smyth MJ et al (2001) Involvement of tumor necrosis factor-related
apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells.
Nat Med 7:94–100
48. van den Broek MF, Kagi D, Zinkernagel RM et al (1995) Perforin dependence of natural
killer cell-mediated tumor control in vivo. Eur J Immunol 25:3514–3516
49. Mocikat R, Braumuller H, Gumy A et al (2003) Natural killer cells activated by MHC class
I(low) targets prime dendritic cells to induce protective CD8 T cell responses. Immunity
19:561–569
50. Cole KE, Strick CA, Paradis TJ et al (1998) Interferon-inducible T cell alpha chemoattract-
ant (I-TAC): a novel non-ELR CXC chemokine with potent activity on activated T cells
through selective high affinity binding to CXCR3. J Exp Med 187:2009–2021
51. Farber JM (1997) Mig and IP-10: CXC chemokines that target lymphocytes. J Leukoc Biol
61:246–257
52. Barchet W, Cella M, Colonna M (2005) Plasmacytoid dendritic cells–virus experts of innate
immunity. Semin Immunol 17:253–261
53. Liu C, Lou Y, Lizee G et al (2008) Plasmacytoid dendritic cells induce NK cell-depend-
ent, tumor antigen-specific T cell cross-priming and tumor regression in mice. J Clin Invest
118:1165–1175
54. Shankaran V, Ikeda H, Bruce AT et al (2001) IFNgamma and lymphocytes prevent primary
tumour development and shape tumour immunogenicity. Nature 410:1107–1111
55. Sanchez-Perez L, Kottke T, Diaz RM et al (2005) Potent selection of antigen loss vari-
ants of B16 melanoma following inflammatory killing of melanocytes in vivo. Cancer Res
65:2009–2017
56. Slingluff CL Jr, Colella TA, Thompson L et al (2000) Melanomas with concordant loss of
multiple melanocytic differentiation proteins: immune escape that may be overcome by tar-
geting unique or undefined antigens. Cancer Immunol Immunother 48:661–672
57. Singh R, Paterson Y (2007) Immunoediting sculpts tumor epitopes during immunotherapy.
Cancer Res 67:1887–1892
58. Zhou G, Lu Z, McCadden JD et al (2004) Reciprocal changes in tumor antigenicity and
antigen-specific T cell function during tumor progression. J Exp Med 200:1581–1592
59. Yee C, Thompson JA, Byrd D et al (2002) Adoptive T cell therapy using antigen-specific
CD8 + T cell clones for the treatment of patients with metastatic melanoma: in vivo per-
sistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci USA
99:16168–16173
60. So T, Takenoyama M, Mizukami M et al (2005) Haplotype loss of HLA class I antigen as an
escape mechanism from immune attack in lung cancer. Cancer Res 65:5945–5952
61. Baba T, Hanagiri T, Ichiki Y et al (2007) Lack and restoration of sensitivity of lung cancer cells
to cellular attack with special reference to expression of human leukocyte antigen class I and/or
major histocompatibility complex class I chain related molecules A/B. Cancer Sci 98:1795–1802
62. Qin Z, Noffz G, Mohaupt M et al (1997) Interleukin-10 prevents dendritic cell accumula-
tion and vaccination with granulocyte-macrophage colony-stimulating factor gene-modified
tumor cells. J Immunol 159:770–776
63. Gorelik L, Flavell RA (2001) Immune-mediated eradication of tumors through the blockade
of transforming growth factor-beta signaling in T cells. Nat Med 7:1118–1122
64. Chen W, Jin W, Hardegen N et al (2003) Conversion of peripheral CD4 + CD25- naive T cells
to CD4 + CD25 + regulatory T cells by TGF-beta induction of transcription factor Foxp3. J
Exp Med 198:1875–1886
65. Dong H, Strome SE, Salomao DR et al (2002) Tumor-associated B7–H1 promotes T-cell
apoptosis: a potential mechanism of immune evasion. Nat Med 8:793–800
66. Okazaki T, Honjo T (2007) PD-1 and PD-1 ligands: from discovery to clinical application.
Int Immunol 19:813–824
394 M. J. McCoy et al.
89. Zhou J, Ding T, Pan W et al (2009) Increased intratumoral regulatory T cells are related
to intratumoral macrophages and poor prognosis in hepatocellular carcinoma patients. Int J
Cancer 125:1640–1648
90. Correale P, Rotundo MS, Del Vecchio MT et al (2010) Regulatory (FoxP3 +) T-cell tumor
infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing
chemo or chemoimmunotherapy. J Immunother 33:435–441
91. Frey DM, Droeser RA, Viehl CT et al (2010) High frequency of tumor-infiltrating
FOXP3(+) regulatory T cells predicts improved survival in mismatch repair-proficient colo-
rectal cancer patients. Int J Cancer 126:2635–2643
92. Lee WS, Park S, Lee WY et al (2010) Clinical impact of tumor-infiltrating lymphocytes for
survival in stage II colon cancer. Cancer 116:5188–5199
93. Salama P, Phillips M, Grieu F et al (2009) Tumor-infiltrating FOXP3 + T regulatory cells
show strong prognostic significance in colorectal cancer. J Clin Oncol 27:186–192
94. Ladoire S, Martin F, Ghiringhelli F (2011) Prognostic role of FOXP3 + regulatory T cells
infiltrating human carcinomas: the paradox of colorectal cancer. Cancer Immunol Immunother
60:909–918
95. Su X, Ye J, Hsueh EC et al (2010) Tumor microenvironments direct the recruitment and
expansion of human Th17 cells. J Immunol 184:1630–1641
96. Tosolini M, Kirilovsky A, Mlecnik B et al (2011) Clinical impact of different classes of
infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal can-
cer. Cancer Res 71:1263–1271
97. Badoual C, Hans S, Rodriguez J et al (2006) Prognostic value of tumor-infiltrating
CD4 + T-cell subpopulations in head and neck cancers. Clin Cancer Res 12:465–472
98. Zhang YL, Li J, Mo HY et al (2010) Different subsets of tumor infiltrating lymphocytes cor-
relate with NPC progression in different ways. Mol Cancer 9:4
99. Longley DB, Harkin DP, Johnston PG (2003) 5-fluorouracil: mechanisms of action and clin-
ical strategies. Nat Rev Cancer 3:330–338
100. Mini E, Nobili S, Caciagli B et al (2006) Cellular pharmacology of gemcitabine. Ann Oncol
17(Suppl 5):v7–v12
101. Hall AG, Tilby MJ (1992) Mechanisms of action of, and modes of resistance to, alkylating
agents used in the treatment of haematological malignancies. Blood Rev 6:163–173
102. Siddik ZH (2003) Cisplatin: mode of cytotoxic action and molecular basis of resistance.
Oncogene 22:7265–7279
103. Nielsen D, Maare C, Skovsgaard T (1996) Cellular resistance to anthracyclines. Gen
Pharmacol 27:251–255
104. Perez EA (2009) Microtubule inhibitors: differentiating tubulin-inhibiting agents based on
mechanisms of action, clinical activity, and resistance. Mol Cancer Ther 8:2086–2095
105. Ghiringhelli F, Apetoh L, Tesniere A et al (2009) Activation of the NLRP3 inflammasome
in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med
15:1170–1178
106. Nowak AK, Mahendran S, van der Most RG et al (2008) Cisplatin and pemetrexed syn-
ergises with immunotherapy to result in cures in established murine mesothelioma. In:
Proceedings of the American association of cancer research annual meeting 487 (Abstract
2073, 2008)
107. van der Most RG, Currie AJ, Mahendran S et al (2009) Tumor eradication after cyclophos-
phamide depends on concurrent depletion of regulatory T cells: a role for cycling TNFR2-
expressing effector-suppressor T cells in limiting effective chemotherapy. Cancer Immunol
Immunother 58:1219–1228
108. Nowak AK, Lake RA, Marzo AL et al (2003) Induction of tumor cell apoptosis in vivo
increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host
tumor-specific CD8 T cells. J Immunol 170:4905–4913
109. Sjoblom T, Jones S, Wood LD et al (2006) The consensus coding sequences of human
breast and colorectal cancers. Science 314:268–274
396 M. J. McCoy et al.
110. Nowak AK, Robinson BW, Lake RA (2003) Synergy between chemotherapy and immuno-
therapy in the treatment of established murine solid tumors. Cancer Res 63:4490–4496
111. Matzinger P (2002) The danger model: a renewed sense of self. Science 296:301–305
112. Zitvogel L, Kepp O, Senovilla L et al (2010) Immunogenic tumor cell death for optimal
anticancer therapy: the calreticulin exposure pathway. Clin Cancer Res 16:3100–3104
113. Apetoh L, Ghiringhelli F, Tesniere A et al (2007) Toll-like receptor 4-dependent con-
tribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med
13:1050–1059
114. Casares N, Pequignot MO, Tesniere A et al (2005) Caspase-dependent immunogenicity of
doxorubicin-induced tumor cell death. J Exp Med 202:1691–1701
115. Obeid M, Tesniere A, Ghiringhelli F et al (2007) Calreticulin exposure dictates the immuno-
genicity of cancer cell death. Nat Med 13:54–61
116. Liu WM, Fowler DW, Smith P et al (2010) Pre-treatment with chemotherapy can enhance
the antigenicity and immunogenicity of tumours by promoting adaptive immune responses.
Br J Cancer 102:115–123
117. Ramakrishnan R, Assudani D, Nagaraj S et al (2010) Chemotherapy enhances tumor cell
susceptibility to CTL-mediated killing during cancer immunotherapy in mice. J Clin Invest
120:1111–1124
118. van der Most RG, Currie AJ, Cleaver AL et al (2009) Cyclophosphamide chemotherapy sen-
sitizes tumor cells to TRAIL-dependent CD8 T cell-mediated immune attack resulting in
suppression of tumor growth. PLoS One 4:e6982
119. Soriani A, Zingoni A, Cerboni C et al (2009) ATM-ATR-dependent up-regulation of
DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results
in enhanced NK-cell susceptibility and is associated with a senescent phenotype. Blood
113:3503–3511
120. Fine JH, Chen P, Mesci A et al (2010) Chemotherapy-induced genotoxic stress promotes
sensitivity to natural killer cell cytotoxicity by enabling missing-self recognition. Cancer
Res 70:7102–7113
121. Karre K (2008) Natural killer cell recognition of missing self. Nat Immunol 9:477–480
122. Radojcic V, Bezak KB, Skarica M et al (2010) Cyclophosphamide resets dendritic cell
homeostasis and enhances antitumor immunity through effects that extend beyond regula-
tory T cell elimination. Cancer Immunol Immunother 59:137–148
123. Salem ML, Al-Khami AA, El-Naggar SA et al (2010) Cyclophosphamide induces dynamic
alterations in the host microenvironments resulting in a Flt3 ligand-dependent expansion of
dendritic cells. J Immunol 184:1737–1747
124. Salem ML, El-Naggar SA, Cole DJ (2010) Cyclophosphamide induces bone marrow to
yield higher numbers of precursor dendritic cells in vitro capable of functional antigen pres-
entation to T cells in vivo. Cell Immunol 261:134–143
125. Pfannenstiel LW, Lam SS, Emens LA et al (2010) Paclitaxel enhances early dendritic cell
maturation and function through TLR4 signaling in mice. Cell Immunol 263:79–87
126. Shurin GV, Tourkova IL, Kaneno R et al (2009) Chemotherapeutic agents in noncytotoxic
concentrations increase antigen presentation by dendritic cells via an IL-12-dependent
mechanism. J Immunol 183:137–144
127. Goldrath AW, Bevan MJ (1999) Selecting and maintaining a diverse T-cell repertoire.
Nature 402:255–262
128. Mackall CL, Hakim FT, Gress RE (1997) Restoration of T-cell homeostasis after T-cell
depletion. Semin Immunol 9:339–346
129. Schluns KS, Kieper WC, Jameson SC et al (2000) Interleukin-7 mediates the homeostasis of
naive and memory CD8 T cells in vivo. Nat Immunol 1:426–432
130. Tan JT, Ernst B, Kieper WC et al (2002) Interleukin (IL)-15 and IL-7 jointly regulate home-
ostatic proliferation of memory phenotype CD8 + cells but are not required for memory
phenotype CD4 + cells. J Exp Med 195:1523–1532
131. Marleau AM, Sarvetnick N (2005) T cell homeostasis in tolerance and immunity. J Leukoc
Biol 78:575–584
14 Activation of Immune-Mediated Tumor Cell Death by Chemotherapy 397
132. Theofilopoulos AN, Dummer W, Kono DH (2001) T cell homeostasis and systemic
autoimmunity. J Clin Invest 108:335–340
133. Dummer W, Niethammer AG, Baccala R et al (2002) T cell homeostatic proliferation elicits
effective antitumor autoimmunity. J Clin Invest 110:185–192
134. Maguire HC Jr, Ettore VL (1967) Enhancement of dinitrochlorobenzene (DNCB) contact
sensitization by cyclophosphamide in the guinea pig. J Invest Dermatol 48:39–43
135. Berd D, Mastrangelo MJ, Engstrom PF et al (1982) Augmentation of the human immune
response by cyclophosphamide. Cancer Res 42:4862–4866
136. Ghiringhelli F, Larmonier N, Schmitt E et al (2004) CD4 + CD25 + regulatory T cells sup-
press tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy
of established tumors to be curative. Eur J Immunol 34:336–344
137. Ghiringhelli F, Menard C, Puig PE et al (2007) Metronomic cyclophosphamide regimen
selectively depletes CD4 + CD25 + regulatory T cells and restores T and NK effector func-
tions in end stage cancer patients. Cancer Immunol Immunother 56:641–648
138. Rettig L, Seidenberg S, Parvanova I et al (2011) Gemcitabine depletes regulatory T-cells in
human and mice and enhances triggering of vaccine-specific cytotoxic T-cells. Int J Cancer
129:832–838
139. Vicari AP, Luu R, Zhang N et al (2009) Paclitaxel reduces regulatory T cell numbers and
inhibitory function and enhances the anti-tumor effects of the TLR9 agonist PF-3512676 in
the mouse. Cancer Immunol Immunother 58:615–628
140. Fisson S, Darrasse-Jeze G, Litvinova E et al (2003) Continuous activation of autoreactive
CD4 + CD25 + regulatory T cells in the steady state. J Exp Med 198:737–746
141. Vukmanovic-Stejic M, Zhang Y, Cook JE et al (2006) Human CD4 + CD25hi Foxp3 + reg-
ulatory T cells are derived by rapid turnover of memory populations in vivo. J Clin Invest
116:2423–2433
142. Chen X, Subleski JJ, Kopf H et al (2008) Cutting edge: expression of TNFR2 defines a
maximally suppressive subset of mouse CD4 + CD25 + FoxP3 + T regulatory cells: appli-
cability to tumor-infiltrating T regulatory cells. J Immunol 180:6467–6471
143. Chen Y, Shen S, Gorentla BK et al (2012) Murine regulatory T cells contain hyperprolifera-
tive and death-prone subsets with differential ICOS expression. J Immunol 188:1698–1707
144. Strauss L, Bergmann C, Szczepanski MJ et al (2008) Expression of ICOS on human mela-
noma-infiltrating CD4 + CD25highFoxp3 + T regulatory cells: implications and impact on
tumor-mediated immune suppression. J Immunol 180:2967–2980
145. Nowak AK, Robinson BW, Lake RA (2002) Gemcitabine exerts a selective effect on the
humoral immune response: implications for combination chemo-immunotherapy. Cancer
Res 62:2353–2358
146. Wijayahadi N, Haron MR, Stanslas J et al (2007) Changes in cellular immunity dur-
ing chemotherapy for primary breast cancer with anthracycline regimens. J Chemother
19:176–723
147. Demaria S, Volm MD, Shapiro RL et al (2001) Development of tumor-infiltrating lym-
phocytes in breast cancer after neoadjuvant paclitaxel chemotherapy. Clin Cancer Res
7:3025–3030
148. Ladoire S, Arnould L, Apetoh L et al (2008) Pathologic complete response to neoadjuvant
chemotherapy of breast carcinoma is associated with the disappearance of tumor-infiltrating
foxp3 + regulatory T cells. Clin Cancer Res 14:2413–2420
149. Yasuda K, Nirei T, Sunami E et al (2011) Density of CD4(+) and CD8(+) T lymphocytes in
biopsy samples can be a predictor of pathological response to chemoradiotherapy (CRT) for
rectal cancer. Radiat Oncol 6:49
150. Peng RQ, Chen YB, Ding Y et al (2010) Expression of calreticulin is associated with infil-
tration of T-cells in stage IIIB colon cancer. World J Gastroenterol: WJG 16:2428–2434
151. Hirota S, Isozaki K, Moriyama Y et al (1998) Gain-of-function mutations of c-kit in human
gastrointestinal stromal tumors. Science 279:577–580
152. Blanke CD, Rankin C, Demetri GD et al (2008) Phase III randomized, intergroup trial
assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic
398 M. J. McCoy et al.
gastrointestinal stromal tumors expressing the kit receptor tyrosine kinase: S0033. J Clin
Oncol 26:626–632
153. Borg C, Terme M, Taieb J et al (2004) Novel mode of action of c-kit tyrosine kinase inhibi-
tors leading to NK cell-dependent antitumor effects. J Clin Invest 114:379–388
154. Balachandran VP, Cavnar MJ, Zeng S et al (2011) Imatinib potentiates antitumor T
cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat Med
17:1094–1100
155. Anraku M, Tagawa T, Wu L et al (2010) Synergistic antitumor effects of regulatory T
cell blockade combined with pemetrexed in murine malignant mesothelioma. J Immunol
185:956–966
156. Correale P, Del Vecchio MT, La Placa M et al (2008) Chemotherapeutic drugs may be
used to enhance the killing efficacy of human tumor antigen peptide-specific CTLs. J
Immunother 31:132–147
157. Fridlender ZG, Sun J, Singhal S et al (2010) Chemotherapy delivered after viral immuno-
gene therapy augments antitumor efficacy via multiple immune-mediated mechanisms.
Mol Ther 18:1947–1959
158. Tseng CW, Hung CF, Alvarez RD et al (2008) Pretreatment with cisplatin enhances
E7-specific CD8 + T-cell-mediated antitumor immunity induced by DNA vaccination. Clin
Cancer Res 14:3185–3192
159. Wada S, Yoshimura K, Hipkiss EL et al (2009) Cyclophosphamide augments antitumor
immunity: studies in an autochthonous prostate cancer model. Cancer Res 69:4309–4318
160. Zhong H, Han B, Tourkova IL et al (2007) Low-dose paclitaxel prior to intratumoral den-
dritic cell vaccine modulates intratumoral cytokine network and lung cancer growth. Clin
Cancer Res 13:5455–5462
161. Robert C, Thomas L, Bondarenko I et al (2011) Ipilimumab plus dacarbazine for previously
untreated metastatic melanoma. New Engl J Med 364:2517–2526
162. Wolchok JD, Neyns B, Linette G et al (2010) Ipilimumab monotherapy in patients with pre-
treated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging
study. Lancet Oncol 11:155–164
163. Correale P, Cusi MG, Tsang KY et al (2005) Chemo-immunotherapy of metastatic colo-
rectal carcinoma with gemcitabine plus FOLFOX 4 followed by subcutaneous granulocyte
macrophage colony-stimulating factor and interleukin-2 induces strong immunologic and
antitumor activity in metastatic colon cancer patients. J Clin Oncol 23:8950–8958
164. Manegold C, Gravenor D, Woytowitz D et al (2008) Randomized phase II trial of a toll-like
receptor 9 agonist oligodeoxynucleotide, PF-3512676, in combination with first-line taxane
plus platinum chemotherapy for advanced-stage non-small-cell lung cancer. J Clin Oncol
26:3979–3986
165. Emens LA, Asquith JM, Leatherman JM et al (2009) Timed sequential treatment with cyclo-
phosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating
factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety
and immune activation. J Clin Oncol 27:5911–5918
166. Hegmans JP, Veltman JD, Lambers ME et al (2010) Consolidative dendritic cell-based
immunotherapy elicits cytotoxicity against malignant mesothelioma. Am J Respir Crit Care
Med 181:1383–1390
167. Ramlau R, Quoix E, Rolski J et al (2008) A phase II study of Tg4010 (Mva-Muc1-Il2) in asso-
ciation with chemotherapy in patients with stage III/IV non-small cell lung cancer. J Thorac
Oncol 3:735–744
168. Walker DG, Laherty R, Tomlinson FH et al (2008) Results of a phase I dendritic cell vaccine
trial for malignant astrocytoma: potential interaction with adjuvant chemotherapy. J Clin
Neurosci 15:114–121
169. Ciampricotti M, Hau CS, Doornebal CW et al (2012) Chemotherapy response of spontane-
ous mammary tumors is independent of the adaptive immune system. Nat Med 18:344–346.
Author reply 346
14 Activation of Immune-Mediated Tumor Cell Death by Chemotherapy 399
170. O’Gorman WE, Dooms H, Thorne SH et al (2009) The initial phase of an immune response
functions to activate regulatory T cells. J Immunol 183:332–339
171. Darrasse-Jeze G, Bergot AS, Durgeau A et al (2009) Tumor emergence is sensed by self-
specific CD44hi memory Tregs that create a dominant tolerogenic environment for tumors
in mice. J Clin Invest 119:2648–2662
172. Turtle CJ, Swanson HM, Fujii N et al (2009) A distinct subset of self-renewing human
memory CD8 + T cells survives cytotoxic chemotherapy. Immunity 31:834–844
173. Dunker K, Schlaf G, Bukur J et al (2008) Expression and regulation of non-classical
HLA-G in renal cell carcinoma. Tissue Antigens 72:137–148
174. Lin A, Zhang X, Zhou WJ et al (2011) HLA-G expression is associated with a poor progno-
sis in patients with esophageal squamous cell carcinoma. Int J Cancer 129:1382–1390
175. Paul P, Cabestre FA, Le Gal FA et al (1999) Heterogeneity of HLA-G gene transcription and
protein expression in malignant melanoma biopsies. Cancer Res 59:1954–1960
176. Chang CC, Ferrone S (2006) NK cell activating ligands on human malignant cells: molecu-
lar and functional defects and potential clinical relevance. Semin Cancer Biol 16:383–392
177. Clayton A, Mitchell JP, Court J et al (2008) Human tumor-derived exosomes down-modu-
late NKG2D expression. J Immunol 180:7249–7258
178. Uyttenhove C, Pilotte L, Theate I et al (2003) Evidence for a tumoral immune resistance
mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med
9:1269–1274
179. Villablanca EJ, Raccosta L, Zhou D et al (2010) Tumor-mediated liver X receptor-alpha
activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens anti-
tumor responses. Nat Med 16:98–105
180. Adjei AA (2004) Pharmacology and mechanism of action of pemetrexed. Clin Lung Cancer
5(Suppl 2):S51–S55
181. Longo-Sorbello GS, Bertino JR (2001) Current understanding of methotrexate phar-
macology and efficacy in acute leukemias. Use of newer antifolates in clinical trials.
Haematologica 86:121–127
182. Hande KR (1998) Etoposide: four decades of development of a topoisomerase II inhibitor.
Eur J Cancer 34:1514–1521
About the Editor
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 401
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
Index
D. E. Johnson (ed.), Cell Death Signaling in Cancer Biology and Treatment, 403
Cell Death in Biology and Diseases, DOI: 10.1007/978-1-4614-5847-0,
© Springer Science+Business Media New York 2013
X.-M. Yin and Z. Dong (Series eds.), Cell Death in Biology and Diseases
404 Index
C (cont.)
Caspase-8, 3, 5–7, 15–18, 62, 258–260, 262, E
267, 268, 282, 285–290 Enzastaurin, 352
Caspase-9, 3, 5–10, 62, 216, 259, 260, ERCC1, 144, 163
283, 289 ERK, 62, 65, 96–98, 126, 163, 193, 209, 214,
Caspase-10, 5–7, 16, 18, 258, 285 241, 332, 334, 335, 337–340, 344–346,
CCT239065, 340 359, 360
CD44, 68–70 ERK inhibitors, 97, 345, 346
CD95, 262, 268, 282–286 ER stress response, 88, 98
Ced-3, 2, 210 Erucylphosphocholine (ErPC), 353
Ced-4, 2, 3 Erucylphosphohomocholine (ErPC3), 353
Ced-9, 2 Extrinsic apoptosis pathway, 4, 5, 15, 17, 18,
Caenorhabditis elegans, 2, 3, 18, 103, 128 257, 264
Ceramide, 205–219
c-FLIP, 16, 18, 268, 285, 286, 289
Chaperones, 88, 182, 186, 188, 192, 314, 315 F
Chemoimmunotherapy, 373, 388–390 Fanconi pathway, 145
CHK1 (checkpoint kinase 1), 139, 141, 146, Fas, 3–5, 16, 17, 126, 220, 262, 282, 377–380
157, 159, 191 Fas-associated death domain protein (FADD),
CHK2 (checkpoint kinase 2), 139, 141, 156, 4, 5, 16–18, 258, 262, 267, 286
157, 192
Chloroquine (CQ), 94, 104–107, 350
Chronic myeloid leukemia (CML), 54, 55, G
127, 192, 220, 235 Galectin-1, 60, 61
cIAP1, 8–10, 256–258, 260–262, 264, 267, Galectin-3, 60–62
268, 270 Galectin-9, 60, 62
cIAP2, 8–10, 256, 258, 260–262, 264, 265, GDC-0879, 340, 360
267, 270 GDC-0941, 236, 343, 346
Co-chaperones, 183, 186, 193 GDC-0973, 343
Crabtree effect, 36, 37 Geldanamycin, 185, 195, 196
Cytochrome c, 3–6, 11, 13, 15, 18, 99, 210, Glutamine metabolism, 40, 41
216, 259, 283 Glycolysis, 35–39, 41, 99, 238
Cytotoxic T cells (CTL), 101, 375, 379, 383 GX15-070 (also called Obatoclax), 231–233,
236, 240–243
D
Dabrafenib, 340, 343, 357, 360 H
Death domain (DD), 4, 17, 258, 261, 262, HDAC inhibitors, 100, 101, 190, 289
282, 285 Heat Shock Proteins (HSPs), 106, 182
Death-effector domain (DED), 5, 18 Helper T cells, 376
Death-inducing signaling complex (DISC), 4, Histone methyl transferase, 45, 190, 191
5, 16–18, 258, 259, 282, 284–286, 288 Homologous recombination, 144, 145, 191
Delanzomib, 308 HSP90 (heat shock protein 90), 376
DNA damage response, 88, 98, 138, 139, Hypoxia, 63, 64, 99, 105, 189
144, 147 Hypoxia-inducible factor (HIF), 43, 63, 99,
DNA-dependent protein kinase (DNA-PK), 189, 346
144, 160
DNA glycosylases, 142, 164
DNA methyl transferase (DNMT), 190 I
DNA polymerases, 143, 158, 163, 164, 382 IC87114, 346
DNA repair, 14, 53, 63, 138–140, 143, Immune system, 2, 59, 88, 101, 102, 107,
146–148, 152, 155, 159, 161–163, 108, 282, 373, 374, 377, 378, 380–382,
166, 192 384, 387
Index 405
K N
KP372-1, 252 NAD+ biosynthesis inhibitors, 162
Natural killer cells (NK cells), 376–379
Nuclear factor-κB (NF-κB), 102, 209, 215,
L 256, 259, 261, 262, 264, 265, 268, 270,
LC3-I, 92, 94 289, 313, 319, 337, 353
LC3-II, 92, 94 Neutral Sphingomyelinase (nSMase), 208,
Lin-4, 117, 128 211, 212, 217, 218
Non-homologous-end-joining (NHEJ), 144
NOXA, 11, 13–15, 62, 233, 237, 314
M Nucleotide excision repair (NER), 140, 191
3-methyladenine (3-MA), 105, 164
Marizomib, 307–309, 321
Mcl-1, 11–13, 15, 55, 62, 96, 231–234, 238, O
240, 314, 345 Oprozomib, 308, 309, 321
MDM2, 14, 189 OSI-027, 355
MEK, 65, 96, 97, 234, 332, 334, 335,
338–340, 342–346, 356–360
MEK inhibitors, 339, 342–344, 346, 358, P
360, 361 p53, 14, 15, 17, 62, 63, 98, 103, 105, 106,
MEK162, 344, 347, 357, 358 122–124, 127, 128, 139, 140, 156, 161,
Mesenchymal stem cells (MSCs), 55, 288 189, 190, 262, 285, 289, 314
MGMT (06-methylguanine DNA methyl trans- PARP [poly(ADP)ribose polymerase], 143,
ferase), 140–142, 149, 154–156, 165 152
microRNA (miRNA), 12 , 13, 41, 117, PBI-05204, 353
122, 123 PD-0325901, 343, 356, 357
miR-15a, 12, 122–124 Perifosine, 344, 353
miR-16-1, 12, 122–124 PI-103, 348
miR-21, 123, 125, 126, 128 PI3K, 39, 70, 89, 90, 95–97, 99, 102, 161,
miR-26a, 124 162, 193, 194, 213, 221, 236, 332,
miR-34, 124 335–339, 341, 343, 346–350, 355–362
miR-122, 120–122, 124 PLX5568, 341
miR-130/131, 123, 127 Proteasome, 162, 304, 306, 308, 310,
miR-143, 122, 125 312–315, 318, 320
miR-145, 125 PTEN, 95, 123, 127, 128, 154, 242, 332,
miR-155, 123, 128 335–337, 339, 350, 356, 357, 360, 361
miR-221, 126 PUMA, 11, 13–15, 98, 126, 127, 314
miR-222, 126 Purine and purine-like inhibitors, 196
406 Index
P (cont.)
PX-866, 346 Tumor necrosis factor (TNF), 3, 216, 282
Pyk2, 65, 70, 71 Tumor necrosis factor receptor--associated
death domain protein (TRADD), 17,
18, 261, 270
R Tumor necrosis factor receptor--associated
Raf-265, 341 factor -1/-2 (TRAF-1/-2), 17
Raf, 96, 163, 187, 188, 194, 208, 209, Tumor necrosis factor--related apoptosis-
332–335, 337–345, 356, 357, 359, 362 inducing ligand (TRAIL), 3, 16, 17, 97,
Raf inhibitors, 334, 339, 342, 345, 360, 361 162, 234, 236, 238, 242, 264, 267, 268,
Ras, 60, 65, 67, 95, 96, 99, 102, 125, 163, 208, 270, 282, 284–291
214, 332–339, 342, 345, 347, 359–362 TRAIL-R1, 16, 17, 282, 284, 285, 288–290
Receptor-interacting protein (RIP), 17, 313 TRAIL-R2, 16, 17, 282, 284, 285, 288, 289
Refametinib, 343 TRAIL-R3, 282, 284, 285
Regorafenib, 341 TRAIL-R4, 284, 285, 288
Regulatory T cells (Treg), 377 Trametinib, 343
Resorcinol class inhibitors, 197 Triciribine, 351
RO4987655, 344
RX-201, 353–354
U
Ubiquitin-proteasome pathway (UPP), 304
S Ubiquitin-proteasome system (UPS), 100, 304
SCH772984, 346 Unfolded protein response (UPR), 64, 314
Second mitochondria-derived activator of
caspase (SMAC), 9, 10, 18, 255, 256,
259, 260, 262–264, 266–269, 270 V
Selumetinib, 342, 344, 356, 357, 359 Vemurafenib, 340, 357, 358, 360
SL327, 344
SMAC mimetics, 256, 259, 266–268, 270
Sorafenib, 57, 124, 163, 234, 236, 241, 339, W
340, 356, 360 Warburg effect, 36, 42, 45
Sphingosine, 42, 205–208, 214, 216, 217, 220 WJD008, 350
Sphingosine kinase 1 (SK1), 207, 208, 214,
215, 217, 219, 221
Sphingosine kinase 2 (SK2), 207, 208 X
Sphingosine-1 phosphate (S1P), 205, 207 XIAP, 8–10, 256, 258–260, 262–267, 269, 270
STAT3, 55–57, 126 XIAP antagonists, 269
Steroid hormone receptors, 189 XL147, 347
XL281, 341
XL418, 354
T XL765, 350
TAK-733, 334
Tumor cell metabolism, 35