You are on page 1of 17

ME65CH20-Wang ARI 11 October 2013 11:20

V I E W
E Review in Advance first posted online
R

on N ovember 4, 2013. (Changes may

S
still occur before final publication
online and in print.)
C E
I N

A
D V A

Targeting Apoptosis Pathways


for New Cancer Therapeutics
Longchuan Bai and Shaomeng Wang
University of Michigan Comprehensive Cancer Center and Departments of Internal
Medicine, Pharmacology, and Medicinal Chemistry, University of Michigan, Ann Arbor,
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org

Michigan 48109; email: shaomeng@umich.edu


by Grand Valley State University on 11/13/13. For personal use only.

Annu. Rev. Med. 2014. 65:20.1–20.17 Keywords


The Annual Review of Medicine is online at
med.annualreviews.org BCL-2, BCL-XL, XIAP, cIAPs, MDM2, p53
This article’s doi: Abstract
10.1146/annurev-med-010713-141310
The past decade has witnessed tremendous advances in the discovery
Copyright ⃝ c 2014 by Annual Reviews.
All rights reserved and development of novel small-molecule inhibitors targeting apoptosis
pathways for cancer treatment, with some compounds now in clinical
development. Early promising clinical data have been reported with
these new classes of anticancer drugs. This review highlights the recent
advancements in the development of small-molecule inhibitors target-
ing three major classes of antiapoptotic proteins: antiapoptotic B cell
lymphoma 2 (BCL-2) proteins, inhibitor of apoptosis proteins (IAPs),
and murine double-minute 2 (MDM2). Special emphasis is given to
those that have been advanced into clinical trials. The challenges and
future directions in the further preclinical and clinical development of
these new anticancer drugs are also discussed.

20.1

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

EXPLOITING APOPTOSIS cytosol. B cell lymphoma 2 (BCL-2) family


PATHWAYS FOR CANCER proteins govern the MOMP (1). The release
TREATMENT of cytochrome c from the mitochondria initi-
ates the formation of a multiprotein complex
Cancer is a multifaceted disease comprised
(apoptosome) consisting of cytochrome c, ap-
of complex genetic and epigenetic aberrations
optotic protease activating factor 1 (APAF1),
that disrupt the normal balance of cellular life
and procaspase-9, resulting in the activation
and death. Apoptosis (programmed cell death)
of caspase-9 and downstream caspase cascade.
represents one of the most important forms
The release of SMAC from mitochondria fur-
of cell death in multicellular organisms and
ther stimulates caspase activation by neutral-
is typically dysregulated in human cancers.
izing IAPs, which regulate apoptosis through
Dysfunction of apoptosis pathways can con-
inhibition of caspases. The extrinsic pathway
fer cancer-treatment resistance, as most con-
is activated following the binding of death li-
ventional chemotherapies as well as radiother-
gands of the tumor necrosis factor (TNF) fam-
apy rely on their ability to provoke apoptotic
ily [e.g., TNFα, TRAIL (TNF-related apopto-
cell death in cancer cells. Therefore, apopto-
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

sis inducing ligand), and Fas ligand] to their cell


sis pathways can be therapeutically exploited
surface receptors of the TNF receptor (TNFR)
for cancer treatment. Apoptosis is tightly regu-
superfamily (e.g., TNFR1 and 2, death recep-
lated by the balance between pro- and antiapop-
tor 4 and 5, and Fas). Upon ligand-receptor in-
totic proteins and becomes dysregulated when
teraction, a multiprotein complex is formed at
the balance between pro- and antiapoptotic
the plasma membrane, which provides a signal-
proteins is altered. In human cancers, overex-
ing platform for activation of initiator caspase-8
pression of antiapoptotic proteins is frequently
and subsequent executioner caspases. Caspase-
observed and has been linked to tumor progres-
8 also cleaves other noncaspase substrates, such
sion, treatment resistance, and poor progno-
as Bid. Truncated Bid (tBid) then translocates
sis. Therefore, targeting antiapoptotic proteins
to mitochondrial membrane to stimulate cy-
with the goal to restore the apoptotic balance
tochrome c release and activation of the intrin-
has been considered as a promising cancer ther-
sic pathway. IAPs thus play an important role in
apeutic strategy in the past decade. Drugs tar-
regulating both intrinsic and extrinsic apoptosis
geting antiapoptotic proteins may directly elicit
pathways.
apoptosis or lower the threshold for cell death
A wide array of apoptosis-related genes is
induction when combined with other cancer
transcriptionally regulated by tumor suppres-
therapeutics.
sor p53 protein. It has been well established
Two main apoptotic pathways have been
that p53 plays a central role in the regulation
extensively investigated: the intrinsic (mito-
of apoptosis and cell cycle transition, among
chondrial) and the extrinsic (death receptor)
many other cellular processes. p53 is mutated
pathways (Figure 1). The intrinsic pathway is
and inactivated in ∼50% of all human can-
activated by a myriad of stress signals, such
cers (http://www-p53.iarc.fr), implying the
as DNA damage caused by chemo- and ra-
importance of disabling p53 function during
diotherapies. Upon cellular stresses, a signal
tumorigenesis. The abundance and activity of
is relayed to the mitochondria, leading to
p53 are tightly controlled under normal physi-
the mitochondrial outer membranes perme-
ologic conditions. Changes in the regulation of
abilization (MOMP) and the release of ap-
p53 can result in the disturbance of p53 func-
optotic proteins, such as cytochrome c and
tion. By directly binding to the N terminus
second mitochondrial-derived activator of cas-
of p53 and promoting p53 ubiquitination and
pases (SMAC) [also known as DIABLO, di-
degradation, murine double-minute 2 (MDM2)
rect inhibitor of apotosis protein (IAP)-binding
[HDM2 (human homolog of MDM2) in hu-
protein with low pI] from mitochondria into
mans] is a crucial negative regulator of p53 (2).

20.2 Bai · Wang

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

Death receptors (extrinsic pathway)

SMAC mimetics

cIAP1/2
GDC-0152
AT-406
RIP1 Cas-8 Cas-3/7 Apoptosis
TL32711
LCL161 cIAP1/2
HGS1029 degradation

XIAP
BCL-2 BH3 mimetics
BCL-XL ABT-263
NF-kB ABT-199
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org

GX15-070
by Grand Valley State University on 11/13/13. For personal use only.

MCL-1 Cas-9
Cytokines AT-101
SMAC mimetics
GDC-0152
AT-406
NOXA TL32711
MDM2 p53 PUMA BAX LCL161
HGS1029
BIM BAK
Cytochrome c
BID SMAC
MDM2 antagonists BAD
RO5045337
RO5503781
MI-77301/SAR405838
CGM097

Cellular stresses (intrinsic pathway)

Figure 1
Schematic representation of the extrinsic and intrinsic apoptotic pathways. The extrinsic (death receptor)
apoptotic pathway is initiated at the cellular membrane by the binding of cell death ligands to their
respective receptors, which triggers the formation of a death-inducing signaling complex to initiate caspase
activation cascade. cIAP1/2 can ubiquitinate RIP1 to prevent it from forming a caspases-8-activating
complex. cIAP1/2 also regulate canonical and noncanonical NF-κB pathways. The intrinsic (mitochondrial)
apoptotic pathway can be activated by various stimuli such as chemotherapeutics, which leads to the
compromise of mitochondrial membrane integrity and release of cytochrome c and SMAC into the cytosol.
Cytochrome c stimulates the formation of the apoptosome complex to activate initiator caspase-9 and
subsequent activation of effector caspases. XIAP directly binds to caspases and inhibits their activity. SMAC
blocks XIAP-mediated caspase inhibition. cIAP1/2 can bind to SMAC and sequester it from XIAP. SMAC
interacts with IAPs via its N-terminal AVPI binding motif. SMAC mimetics mimic this motif to antagonize
the inhibitory function of IAPs. BAX and BAK are the “effectors” of mitochondrial permeabilization.
Antiapoptotic members of the BCL-2 proteins counteract the activity of BAX and BAK, whereas the
proapoptotic BH3-only BCL-2 family proteins stimulate apoptosis either by directly interacting with BAX
and BAK or by binding to the antiapoptotic BCL-2 family proteins to release BAX and BAK. BH3 mimetics
competitively disrupt the inhibitory complexes between pro- and antiapoptotic BCL-2 family proteins. p53
promotes apoptosis by transcriptionally regulating apoptosis-related genes or directly activating BAX and
BAK. The stability and transcriptional activity of p53 is tightly regulated by MDM2 in a negative-feedback
loop. MDM2 antagonists competitively displace p53 from the MDM2 complex. Abbreviations: BCL-2,
B cell lymphoma 2; IAP, inhibitor of apotosis protein; MDM2, murine double-minute 2; SMAC, second
mitochondrial-derived activator of caspase.

www.annualreviews.org • Apoptosis-Based Cancer Therapies 20.3

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

Tremendous efforts have been made by both apies (reviewed in Reference 1). Therefore,
academia and the pharmaceutical industry to targeting antiapoptotic BCL-2 family proteins
design and develop small-molecule inhibitors is viewed as an attractive cancer therapeutic
to restore apoptosis function in cancer cells. strategy.
This review focuses on recent advances in the
development and application of small-molecule
inhibitors targeting three major classes of anti- Small Molecular Inhibitors of
apoptotic proteins: BCL-2/BCL-XL, IAP pro- Antiapoptotic BCL-2 Family Proteins
teins, and MDM2. Special emphasis is given to Several approaches have been utilized to target
those compounds that have been advanced into the antiapoptotic BCL-2 family proteins, and
clinical development. one promising approach is to design nonpep-
tide, small molecules to mimic proapoptotic
BH3-only members of the BCL-2 family (BH3
TARGETING ANTIAPOPTOTIC mimetics). The antiapoptotic members of the
BCL-2 FAMILY PROTEINS FOR BCL-2 family contain a surface hydrophobic
CANCER THERAPY
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

groove that can bind and neutralize the BH3


MOMP, a defining event in the intrinsic apo- death domains of proapoptotic members (5).
ptosis pathway, is controlled by BCL-2 family BH3 mimetics competitively bind into the
proapoptotic effector proteins BAX and BAK, hydrophobic groove of these antiapoptotic
which are activated by proapoptotic BH3-only BCL-2 family proteins to liberate the proapop-
BCL-2 family proteins (i.e., BIM, BID, BAD, totic BCL-2 family proteins. A number of
PUMA, BMF, BIK, HRK, and NOXA) and an- natural product derivatives and rationally
tagonized by antiapoptotic BCL-2 family pro- designed compounds have been developed
teins (i.e., BCL-2, BCL-XL, BCL-w, BCL-B, as BH3 mimetics. These compounds include
MCL-1, and A1/BFL-1) (1). Upon activation, ABT-737 and its oral analog ABT-263 (nav-
BAX and BAK form large oligomeric pores in itoclax), ABT-199, GX15-070 (obatoclax),
the mitochondrial outer membrane, resulting R-(−)-gossypol (AT-101), WEHI-539, and
in the release of proapoptotic factors and even- our BM series compounds, among others
tually cell death. Two different but not mutu- (6–12). Although many of the reported com-
ally exclusive models have been proposed to de- pounds exert cytotoxicity through additional
scribe the regulation of BAX and BAK activity mechanisms, ABT-737/263/199, WEHI-539,
by other BCL-2 family proteins. The indirect and our newly identified BM series compounds
model suggests that the sequestration of BAX exhibit antitumor activities by specifically
and BAK by the antiapoptotic BCL-2 family targeting one or more BCL-2 proteins (10, 13).
proteins inhibits MOMP. Binding of the anti-
apoptotic proteins by some but not all BH3- ABT-737 and ABT-263 (navitoclax). ABT-
only BCL-2 proteins causes the activation of 737 was designed using a fragment-based strat-
BAK or BAX. The direct activation model sug- egy and binds to BCL-XL, BCL-2, and BCL-w
gests that BIM, tBID, and PUMA, which are se- with high affinity (Ki < 1 nM) but with much
questered by antiapoptotic BCL-2 family mem- lower affinity to BFL-1 and MCL-1 (7). ABT-
bers such as BCL-2, BCL-XL, and MCL-1, can 263 is an orally available derivative of ABT-737,
directly activate BAK and BAX (3, 4). and compared with ABT-737, it has a very sim-
Members of the antiapoptotic BCL-2 pro- ilar binding profile to these antiapoptotic BCL-
teins are commonly overexpressed in a wide 2 proteins (7).
range of cancers, including both hematologi- Preclinical studies have shown that ABT-
cal malignancies and solid tumors, where they 737 and ABT-263 have potent cytotoxicity
contribute to tumorigenesis and the develop- against a variety of hematologic malignancies.
ment of resistance to chemo- and radiother- However, most solid tumor cell lines that have

20.4 Bai · Wang

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

been studied are insensitive to the drugs, with first-in-class BCL-2-selective inhibitor with
the exception of a subset of small-cell lung can- subnanomolar affinity for BCL-2 (6). ABT-199
cer cell lines (7, 14). Resistance to ABT-737 and potently disrupts BCL-2 function and induces
ABT-263 is frequently associated with over- apoptosis in cancer cells with high BCL-2
expression of the antiapoptotic BCL-2 family expression, such as leukemia and lymphoma
member MCL-1 (15, 16), which is not targeted (6). Compared with ABT-263, ABT-199 is
by these compounds. Interestingly, despite its substantially less toxic against platelets (6, 29).
similar high binding affinities for the antiapop- In phase I trials, ABT-199 has demonstrated
totic BCL-2, BCL-XL, and BCL-w proteins in substantially greater activity than that seen with
biochemical assays, ABT-737 discriminates be- ABT-263 in patients with either R/RCLL or
tween the complexes formed between these an- R/R non-Hodgkin lymphoma and also with no
tiapoptotic proteins and proapoptotic BCL-2 significant effects on platelets counts (6). Tu-
family proteins in cells (17, 18). For instance, mor lysis syndrome is dose limiting for ABT-
ABT-737 targets BCL-2 with preference over 199 in patients with CLL, and an 85% over-
BCL-XL and BCL-w in leukemia cells (17, 18). all response rate was achieved in CLL (30, 31).
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

In vitro, ABT-737 synergizes with conven- Ongoing clinical study of ABT-199 includes a
tional chemotherapies, radiotherapy, as well as combination trial with bendamustine and rit-
tyrosine kinase inhibitors to overcome drug uximab in patients with R/Rnon-Hodgkin lym-
resistance. Similarly, ABT-263 also signifi- phoma (http://clinicaltrials.gov).
cantly enhances the activity of chemothera-
peutic agents and targeted therapies (19–22).
For example, ABT-263 enhances the efficacy of Obatoclax mesylate (GX15-070) (Gemin
both docetaxel and erlotinib in xenograft mod- X/Teva). Obatoclax is a pan-BCL-2 inhibitor
els of ovarian and lung cancers, concomitant that binds to BCL-2, BCL-XL, BCL-w, BCL-
with the downregulation of MCL-1 and/or up- B, BFL-1, and MCL-1 in vitro, albeit with
regulation of BIM (20). much lower affinities than those of ABT-263 to
Several clinical trials of ABT-263 have been BCL-2, BCL-XL, and BCL-w (32, 33). In light
conducted in patients with chronic lymphocytic of the fact that MCL-1 can confer resistance
leukemia (CLL) (23), lymphoma (24), and lung to ABT-263, the ability of obatoclax to target
cancer (25). Partial response was observed in MCL-1 represents a potential advantage over
one-third of patientswith relapsed or refrac- ABT-263. Obatoclax can efficiently diminish
tory (R/R) CLL (23). However, ABT-263 ex- the interactions between MCL-1 and proapop-
hibits limited single agent activity in small-cell totic proteins to activate the intrinsic apopto-
lung cancer (25). BCL-XL is critical for platelet sis pathway (33). However, obatoclax also in-
survival (26). Because ABT-263 potently tar- duces apoptosis in the absence of BAX/BAK
gets BCL-XL, thrombocytopenia was observed and arrests cancer cells at the G2 phase, sug-
across all clinical trials for ABT-263 and is the gesting that it exerts cytotoxicity through mul-
dose-limiting toxicity (27). Nevertheless, the tiple mechanisms (13, 34).
platelet decrease was transient and reversible, Phase I trials of obatoclax in leukemia and
with no clinically significant bleeding (23–25, lymphoma have identified transient neurotox-
28). Several phase II combination trials are icity as the most common adverse effect (35).
ongoing in lymphoid cancers and solid tumors In these studies, a limited number of ob-
(http://clinicaltrials.gov). jective responses were observed, along with
hematological improvement in a larger propor-
ABT-199 (GDC-0199, AbbVie). Given the tion of treated patients (35). Several phase II
dose-limiting toxicity of ABT-263 in patients, combination trials are ongoing in hematolog-
structure-guided reverse engineering of ABT- ical malignancies and advanced solid tumors
263 led to the discovery of ABT-199, the (http://clinicaltrials.gov).

www.annualreviews.org • Apoptosis-Based Cancer Therapies 20.5

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

AT-101 [R-(−)-gossypol] (Ascenta). AT- tial antitumor activity in BCL-2-driven tumors,


101, a polyphenolic compound derived from while avoiding on-target toxicity by ABT-263
cotton seeds and roots, is an oral pan-BCL-2 (6, 29).
inhibitor. AT-101 exhibits affinity for BCL-2,
BCL-XL, and MCL-1 at submicromolar con- MCL-1-mediated drug resistance. MCL-1
centrations. It induces apoptosis by acting as is recognized as one of the major resistance
a BH3 mimetic as well as a p53-independent factors to ABT-737/263. Therefore, strate-
activator of NOXA and PUMA. The cytotoxi- gies are being developed to tackle MCL-
city of AT-101 may also result from additional 1-mediated resistance. One approach is to
mechanisms, such as DNA cleavage and/or the develop high-affinity pan-BCL-2 inhibitors.
generation of reactive oxygen species (36). Other approaches include combinational treat-
AT-101 was well-tolerated in patients and ment with MCL-1-selective inhibitors or with
demonstrated single-agent cytoreductive activ- conventional chemotherapeutics that reduce
ity in several malignancies, including prostate MCL-1 levels or antagonize MCL-1 by upreg-
cancer (37). However, early phase II trials ulating BIM and/or NOXA (19–21, 22).
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

in prostate and lung cancers, either used as Biomarkers for therapeutic response. Pre-
monotherapy or in combination with conven- clinical studies suggest that the ratio between
tional chemotherapies, failed to deliver signif- BCL-2 and MCL-1 or between antiapoptotic
icant clinical activity (37–40). Several phase II and proapoptotic BCL-2 family proteins can
combination trials are ongoing in squamous cell predict cancer cell response to ABT-737. For
carcinoma of the head and neck and advanced example, BH3 profiling has demonstrated that
laryngeal cancer (http://clinicaltrials.gov). the interactions between and the relative ra-
tios of pro- and antiapoptotic BCL-2 family
Challenges and Future Directions members, but not the BCL-2 protein levels,
Preclinical and clinical studies have provided determine cellular sensitivity to ABT-737 (42,
valuable information regarding the tolerability 43). In primary CLL cells, BCL-2 expression
and “on-target” toxicity of BH3 mimetics in alone does not dictate sensitivity to ABT-737.
patients. These studies have also demonstrated Instead, the amount of BCL-2-BIM complex
promising clinical activity in some patients, as predicts the sensitivity to ABT-737 (44). Other
seen with ABT-263 in CLL and more recently preclinical studies have shown that the ratio of
with ABT-199. However, additional transla- (MCL-1 + BFL-1):BCL-2 or BCL-2:MCL1
tional research is required to realize the full po- predicts the response to ABT-737 in CLL and
tential of this exciting class of targeted drugs. MM cell lines (45, 46). In patients with CLL,
low MCL1 expression and high BIM:MCL-1
On-target toxicities. The dependence of or BIM:BCL-2 ratios in leukemic cells corre-
platelets on BCL-XL for survival presents a sig- lated with response to ABT-263 (23). These
nificant challenge to the clinical application of data suggest the importance of profiling both
ABT-263 and other BCL-XL-targeting BH3 anti- and proapoptotic BCL-2 family mem-
mimetics (27). Preclinical studies have demon- bers to identify biomarkers for BH3 mimetics
strated that the on-target toxicities of ABT-263 sensitivity.
on platelets are greatest at high doses and thus
suggest avoiding a loading dose of ABT-263 to
minimize platelet nadir. Indeed, a regimen with TARGETING IAPS FOR
a low priming dose before dose escalation has CANCER THERAPY
proven safe and effective in early-stage clinical Release of apoptotic proteins, such as cy-
trials of ABT-263 monotherapy (23–25). The tochrome c and SMAC, from the mitochondria
clinical data on ABT-199 demonstrated that se- into the cytosol is a deciding event in the
lective targeting of BCL-2 can achieve substan- intrinsic apoptosis pathway. SMAC promotes

20.6 Bai · Wang

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

caspase activation and apoptosis by binding to (Ala-Val-Pro-Ile) inspired the design of small
cIAP1/2 and XIAP proteins via its N-terminal molecules mimicking this motif as SMAC
Ala-Val-Pro-Ile (AVPI) motif to block their mimetics and antagonists of IAPs. There are
antiapoptotic activities (47). two types of SMAC mimetics: monovalent,
Despite their names, IAPs vary in their which imitates one AVPI motif, and bivalent,
ability to block apoptosis. XIAP directly binds which contains two APVI-like structures con-
to caspase-3, -7, and -9 through its BIR2 or nected by a linker. Resembling the endogenous
BIR3 domains and inhibits their activities. active SMAC, bivalent SMAC mimetics have
cIAPs, which are not potent inhibitors of higher affinity to XIAP because they bind
caspases, bind to SMAC with high affini- simultaneously to the BIR2 and BIR3 domains.
ties and can prevent SMAC from blocking As single agents, SMAC mimetics exert cy-
XIAP-mediated inhibition of caspases (48). totoxicity in a small subset of cancer cell lines
Notably, cIAPs regulate the extrinsic apoptosis (55). SMAC mimetics bind to cIAPs, result-
pathway through their E3 ubiquitin ligase ing in their ubiquitination and subsequent pro-
activity. Particularly, cIAPs are responsible teasomal degradation followed by stabilization
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

for RIP1 ubiquitination, preventing it from of NF-κB-inducing kinase and activation of


forming a complex with caspases-8 and Fas- the noncanonical NF-κB pathway (56). Acti-
associated protein with death domain (FADD), vated NF-κB signaling stimulates the transcrip-
and subsequent caspase activation (49, 50). tion and production of TNFα, which activate
cIAPs are also key regulators of canonical TNFR1 signaling in an autocrine or paracrine
and noncanonical NF-κB pathways. The E3 manner (56). In the absence of cIAPs, un-
ligase activity of cIAPs promotes the activation ubiquitinated RIP1, together with FADD and
of canonical NF-κB signaling by stimulating caspase-8, forms a death-signaling activation
K63 ubiquitin on RIP1 (49, 50). Nevertheless, platform to activate caspase-8 and provoke ap-
cIAP1 constitutively suppresses the abundance optosis (56). Targeting both cIAPs and XIAP
of NF-κB-inducing kinase in the noncanonical appears to be critical for efficient cell death in-
NF-κB pathway (51, 52). NF-kB induces the duction by SMAC mimetics (57, 58).
expression of diverse target genes involved Given the limited cytotoxicity of SMAC
in inflammation, proliferation, survival, cell mimetics in most cancer cell lines, the antitu-
death, angiogenesis, migration, and invasion. mor effects of SMAC mimetics have been ex-
Elevated expression of IAPs has been ob- tensively examined in combination with other
served in hematological malignancies and solid cancer therapeutics, such as chemo- and ra-
tumors, and it correlates with disease progres- diotherapy, death receptor agonists, small-
sion, metastasis, treatment resistance, and poor molecule inhibitors of kinases, BCL-2, and
prognosis. For example, XIAP overexpression proteasome. Synergistic responses have also
correlates with chemoresistance and poor prog- been reported in numerous studies using in
nosis in pancreatic cancer and advanced head vitro and mouse xenograft models (reviewed in
and neck cancer. The chromosome 11q21–q23 Reference 53). In most cases, the reported syn-
region, which harbors cIAP1 and cIAP2 loci, is ergy is independent of TNFα but dependent
amplified in a variety of solid tumors (reviewed on the antagonism of XIAP.
in References 53, 54). Thus, IAPs are attractive
targets for novel cancer therapeutics. GDC-0152 and GDC-0917 (CUDC-427)
(Genentech). GDC-0152 was the first SMAC
mimetic to enter clinical trials. It binds to the
Small-Molecule SMAC Mimetics as BIR3 domain of XIAP, cIAP1, and cIAP2 as
Antagonists of IAPs well as the BIR domain of ML-IAP with Ki
The discovery that the IAP-binding motif values <50 nM (59). In humans, GDC-0152
of SMAC contains only four amino acids exhibited linear pharmacokinetics over a wide

www.annualreviews.org • Apoptosis-Based Cancer Therapies 20.7

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

range of doses with intravenous administration served in patients with diverse tumor types, in-
(59). GDC-0917 is an oral monovalent SMAC cluding breast cancer (66).
mimetic that has shown a favorable safety, PK,
and PD profile in patients with advanced ma- AEG40826/HGS1029 (Aegera/Human Ge-
lignancies (60). nome Sciences). HGS1029 is a bivalent
SMAC mimetic, showing a good tolerability
profile with grade 2 transient lymphopaenia
Birinapant (TL32711) (TetraLogic Phar-
and neutrophilia reported in some patients.
maceuticals). Birinapant is a bivalent SMAC
HGS1029 displays proportional pharmacoki-
mimetic (61). It induces IAP- and caspase-
netics across wide dose ranges. It has also
dependent but TNFa-independent apoptosis.
demonstrated predicted biomarker modu-
It also enhances TRAIL potency in inflam-
lation, such as cIAP1 downregulation and
matory breast cancer models (61). A phase
MCP-1 upregulation (68).
I clinical trial of birinapant has showed that
intravenous birinapant is well tolerated with
SM-406/AT-406/Debio 1143 (University
no dose-limiting toxicities (62). Birinapant also
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

of Michigan/Ascenta/DebioPharm). AT-
demonstrated strong correlation among drug
406 is a monovalent, orally bioavailable SMAC
exposure, predicted biomarker modulation
mimetic that binds to XIAP, cIAP1, and cIAP2
[e.g., cIAP1 degradation and upregulation of
proteins, with a Ki of 66.4, 1.9, and 5.1 nM, re-
interleukin-8 (IL-8) and monocyte chemoat-
spectively (69). Preclinical studies have shown
tractant protein 1(MCP-1)], and apoptosis
that AT-406 exerts mechanism-based antitu-
induction in tumors (62). Furthermore,
mor activity in selected cancer cell lines and
birinapant can be combined with multiple
xenograft tumor models. AT-406 has entered
chemotherapies with excellent tolerability at
clinical trials for studies of its safety, pharma-
standard dosing, and Bell’s palsy was consid-
cological properties, and pharmacodynamics
ered as a dose-limiting toxicity (63). A phase II
biomarkers.
study demonstrated clinical benefit of the
combination of birinapant with irinotecan
over irinotecan alone in R/R colorectal cancer Challenges and Future Directions
patients, with a 57% overall clinical benefit and
In the past few years, considerable progress has
the greatest benefit in KRAS mutant colorectal
been made in the preclinical proof-of-efficacy
cancers (64). An ascending-dose strategy of
studies of SMAC mimetics. Extensive preclini-
birinapant also shows benefits in mitigating
cal studies have also identified several outstand-
Bell’s palsy risk (64).
ing challenges related to developing SMAC
mimetics as a novel therapy for cancer. These
LCL161 (Novartis). LCL161, an orally avail- challenges include the knowledge gaps in our
able IAP antagonist, displays antiproliferative understanding of the biological consequences
activities in a subset of tumors from diverse of IAP antagonism in normal human tissues, as
lineages as a single agent and acts synergisti- well as rational design of SMAC mimetic-based
cally with paclitaxel in preclinical models (65, combination strategies, among others.
66). LCL161 demonstrates target antagonism
by causing cIAP1 protein degradation and up- TNFα-mediated toxicity. All SMAC mimet-
regulation of circulating chemokines. It is also ics, perhaps with the exception of TL32711
well tolerated, with neutropenia, fatigue, and (61), induce TNFα-dependent apoptosis fol-
neuropathy as the principal dose-limiting tox- lowing cIAPs degradation and NF-κB activa-
icities (66, 67). LCL161 was tested in combi- tion in cancer cells. TNFα is a pleiotropic
nation with paclitaxel in patients with advanced cytokine that provokes a variety of cellular
solid tumors, and objective responses were ob- responses, depending on the cellular context.

20.8 Bai · Wang

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

Pertinent concerns include the potential ad- resulting in the sensitization of TMZ-induced
verse effects of SMAC mimetic–stimulated NF- apoptosis (73). Because chemotherapeutics
κB activation and cytokine production on nor- are commonly associated with undesirable
mal tissues. GDC-0152, for example, causes an side effects, their combination with SMAC
acute induction of TNFα in the plasma of dogs mimetics may lower the therapeutic doses for
and rats (70). Accordingly, GDC-0152 demon- chemotherapies without compromising the
strates a toxicity profile consistent with TNFα- efficacy and accordingly may ameliorate the
mediated toxicity. The onset and resolution of side effects associated with chemotherapies.
these acute toxicities generally track with the Therefore, combining SMAC mimetics with
GDC-0152-induced cytokine time course (70). conventional chemotherapies may hold con-
Despite these potential concerns in preclini- siderable clinical promise. Promising evidence
cal studies, severe cytokine release syndrome, from clinical activity in phase I trials provides
caused by an acute increase in plasma TNFα support for this notion: Combining birinapant
and other inflammatory cytokines, has not been with multiple chemotherapeutics at standard
reported in patients (66–68). dosing schedules has yielded excellent tolera-
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

bility in patients with advanced solid cancers


Nonapoptotic functions of IAPs. Accumu- (63).
lating evidence has shown that IAP proteins
also exert nonapoptotic functions, which mostly
stem from IAPs’ characteristics as E3 ubiquitin TARGETING MDM2 FOR
ligases and regulators of NF-kB signaling. For CANCER THERAPY
example, IAPs play a role in cell motility, mi- The p53 tumor suppressor protein plays a cen-
gration, invasion, and metastasis. Therefore, by tral role in the regulation of apoptosis, the
targeting IAPs, SMAC mimetics can have an cell cycle, and senescence, among many other
effect on cell motility, migration, invasion, and cellular processes, through both transcription-
metastasis. dependent and -independent mechanisms (re-
viewed in 74). p53 promotes apoptosis by
Combination with chemotherapeutic transcriptionally regulating genes directly in-
drugs. Chemotherapies, along with radio- volved in apoptosis, such as death receptors
therapy and surgery, are still the mainstays of (e.g., Fas and DR5), proapoptotic BCL-2 fam-
cancer treatments today. Genotoxic agents, ily members (e.g., BAX, NOXA, and PUMA),
such as etoposide, can activate cell death and APAF1, among many others. Moreover,
pathways via proteasome-dependent depletion cytosolic p53 can directly activate proapop-
of cIAPs and XIAP to promote the assembly of totic BAX, leading to mitochondrial mem-
a cytoplasmic cell death–activating platform, brane permeabilization and cytochrome c re-
ripoptosome (71, 72). The core components of lease (75). p53 present at the mitochondria
ripoptosome are RIP1, FADD, and caspase-8; physically associates with proapoptotic BAK,
these components are similar to those in disrupting BAK sequestration by MCL-1 and
complex IIB originating from an interaction resulting in BAK activation (76). In addition,
between the death receptor and death ligand. mitochondrial-localized p53 can bind to BCL-
However, the formation of ripoptosome is 2 or BCL-XL, promoting the release of se-
independent of any death receptors. Notably, questered proapoptotic proteins (77).
SMAC mimetics stimulate the formation p53 is normally expressed at low levels so
of ripoptosome in etoposide-treated SMAC that it would not cause abnormal cell death
mimetic- -resistant cancer cells (72). In or cell cycle arrest. Multiple mechanisms ex-
glioblastoma cells, SMAC mimetics cooperate ist to control the transcriptional activity, stabil-
with temozolomide (TMZ) to stimulate the for- ity, and subcellular localization of p53 protein.
mation of a RIP1/caspase-8/FADD complex, MDM2 is a crucial negative regulator of p53.

www.annualreviews.org • Apoptosis-Based Cancer Therapies 20.9

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

MDM2 contains a p53 binding domain at the N and subsequent cell cycle arrest, apoptosis,
terminus and a RING (really interesting gene) and senescence in cancer cells with WT p53
domain at the C terminus functioning as an E3 (84). Nutlin-3 has been used extensively as a
ligase. MDM2 inhibits p53 by at least two dif- tool compound to investigate the therapeutic
ferent mechanisms: (a) It binds to the transac- potential and mechanisms of action of MDM2
tivation domain of p53 and blocks its transcrip- inhibitors in vitro and in vivo. Nutlin-3
tional activity (78, 79), and (b) it functions as selectively inhibits cell growth in tumor cell
an E3 ubiquitin ligase and promotes the ubiq- lines with WT p53 over those with mutated or
uitination and proteasome-mediated degrada- deleted p53 status. Although Nutlin-3 activates
tion of p53 [1]. p53 also activates the tran- p53 in all tumor cell lines with WT p53 and in-
scription of MDM2. Increased expression of duces p53-dependent cell cycle arrest, it induces
MDM2 leads to the degradation and inactiva- apoptosis in only a subset of cancer cell lines.
tion of p53 in a feedback loop. Gene amplifica- RG7112 (RO5045337), a derivative of
tion and/or overexpression of MDM2 has been Nutlin-3 with improved binding affinity to
reported in a wide range of human malignancies MDM2 and pharmacokinetics, was advanced
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

and is mutually exclusive to p53 mutation in a into clinical trials (85, 86). RG7112 was well
significant subset of tumors, underscoring the tolerated in patients with liposarcoma, some
role of MDM2 in tumorigenesis (81). There- of whom experienced drug-related hemato-
fore, antagonizing MDM2 to release p53 from logical toxicity such as thrombocytopenia,
its negative control is an appealing strategy for neutropenia, and neutropenic fever (87). Of
the treatment of cancers with wildtype (WT) the 17 patients with MDM2 gene ampli-
p53. fication, 1 patient had a confirmed partial
response and 14 had stable disease (87).
RG7112 is currently in Phase I studies for pa-
Small-Molecule Inhibitors of tients with advanced solid tumor or leukemia
MDM2-p53 Interaction and in combination trial in patients with
Targeting MDM2 AML (http://clinicaltrials.gov). RO5503781
Genetic and biochemical studies have mapped (RG7388, Roche) from a different chemical
p53-MDM2 binding sites to the N termi- class is another orally bioavailable MDM2
nus of MDM2 and the transactivation domain inhibitor [2] in phase I clinical trials in patients
of p53 (78). The crystal structure of a p53- with advanced malignancies (monotherapy)
derived peptide bound to the p53 binding do- or with AML (monotherapy or in combination
main of MDM2 revealed that three amino acid with cytarabine) (http://clinicaltrials.gov).
residues from the p53 peptide (Phe19, Trp23,
and Leu26) project residues deep into the hy- MI-219 and MI-77301 (University of
drophobic cavity of the p53 pocket in MDM2, Michigan/Ascenta/Sanofi). MI-219 contains
making them critical to the binding of these a spiro-oxindole core structure and was discov-
two proteins (82). Small molecule inhibitors of ered using a structure-based de novo design
MDM2 have been developed by mimicking the strategy to mimic the key p53 binding residues
p53 binding residues, and several such com- by the Wang laboratory at the University of
pounds are now in clinical development for can- Michigan. It binds to MDM2 with a high affin-
cer treatment. ity and demonstrates a very high specificity over
MDMX and other proteins (88). MI-219 has a
Nutlins (Roche). Nutlins were the first class well-defined mechanism of action as a potent
of potent, specific, and orally bioavailable and specific MDM2 inhibitor in vitro and in
MDM2 inhibitors (83). Nutlins mimic p53 and vivo and demonstrates potent on-target antitu-
bind to the p53-binding pocket in MDM2, mor activity and excellent tolerability in animal
leading to p53 stabilization and activation models (88). Further chemical modifications of

20.10 Bai · Wang

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

MI-219 by the Wang laboratory to improve its DNA-damaging agent topotecan in preclini-
binding affinity to MDM2, on-target cellular cal retinoblastoma models without any obvious
activity, and pharmacokinetics led to the discov- side effects in mice (96), implying that MDM2
ery of MI-77301 (SAR405838), which has been inhibitors may not be toxic to normal tissues
advanced into clinical development by Sanofi. even when combined with genotoxic therapies.
These observations suggest that pharmacolog-
CGM097 (Novartis). CGM097 is another ical inhibition of MDM2 activity does not fully
oral MDM2 inhibitor entering human clinical phenocopy those of MDM2 gene deletion in
trials in early 2013 (http://clinicaltrial.gov), terms of the toxicity to normal tissues.
although no information is disclosed about its One critical difference between activation of
chemical structure and in vitro and in vivo p53 via genetic deletion of MDM2 and pharma-
characterizations. cological activation of p53 by MDM2 inhibitors
is the presence of high levels of MDM2 in tis-
sues where p53 is activated for the pharmaco-
Challenges and Future Directions logical approach due to the feedback loop of
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

Targeting MDM2 to reactivate p53 offers excit- p53-MDM2 regulation (83, 88). The induction
ing prospects in cancer treatment. This notion of MDM2 accumulation by MDM2 inhibitors
is supported by promising preclinical results. may achieve attenuated p53 activation that is
As with other targeted drugs, additional studies sufficient for strong antitumor activity with-
are needed to thoroughly investigate the poten- out causing toxicity in normal tissues. Indeed,
tial side effects of and emergence of drug resis- phase I data for RG7112 from patients with ad-
tance to MDM2 inhibitors in clinically relevant vanced cancer demonstrate a good tolerability
settings. with neutropenia being the major toxicity, sug-
gesting that activation of p53 in the bone mar-
Potential side effects. Several studies have row by MDM2 inhibitors may be responsible
shown that unleashing p53 from MDM2 sup- for its on-target toxicity.
pression could have detrimental effects on
the normal tissues. For example, deletion of MDMX-mediated drug resistance. Al-
MDM2 in mice is embryonic lethal, which can though MDM2 inhibitors such as nutlins and
be rescued by concurrent deletion of p53 (89, MI-219 efficiently induce cell cycle arrest
90). Deletion of MDM2 in specific types of in all tumor cell lines with WT p53 status,
cells, such as neuronal progenitors and car- they induce apoptosis in only a subset of
diomyocytes, also leads to p53-dependent em- WT p53 cancer lines (84, 88). MDMX,
bryonic lethality (91–93). Mice with mutations which is not efficiently antagonized by nutlins
at S21 and T23 of p53 to evade MDM2 bind- or MI-219, has been proposed as a major
ing exhibit constitutive p53 activation and seg- determinant for drug resistance (88, 97).
mental progeria that is correlated with the de- Indeed, knockdown of MDMX by siRNAs
pletion of adult stem cells in multiple tissues, sensitizes WT p53 cancer cells to nutlin- or
including bone marrow, brain and testes (94). MI-219-induced apoptosis (88, 98). Therefore,
Further, MDM2 heterozygous mice are sensi- combinations of selective MDM2 inhibitors
tive to low-dose irradiation treatment (95). All with agents that can directly or indirectly target
these observations raise the question whether MDMX may achieve synergy. It is recently
activation of p53 by MDM2 inhibitors will be reported that Hsp90 inhibitor 17AAG can
toxic to normal tissues, especially when under destabilize MDMX protein and potentiates
stresses. Preclinical studies have demonstrated nutlin-induced p53 transcription of PUMA.
good tolerability of Nutlin-3 and MI-219 in Consequently, 17AAG synergizes with nutlin,
mice with no manifest signs of toxicity (83, in part through inhibiting MDMX and AKT
88). Moreover, Nutlin-3 synergizes with the signaling, to induce apoptosis in WT p53

www.annualreviews.org • Apoptosis-Based Cancer Therapies 20.11

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

cell lines and their xenografts in mice (99). hibitors targeting the IAP proteins (cIAP1/2
Dual inhibitors of MDM2 and MDMX or and XIAP), and inhibitors targeting MDM2
MDMX-selective inhibitors are also under to reactivate p53. In each case, several com-
intensive development, which is beyond the pounds have been advanced into clinical de-
scope of this review. velopment. Evidence of on-target activity has
Because the activity of MDM2 inhibitors de- been observed in patients for each class of com-
pends on activation of WT p53, MDM2 in- pounds with a manageable toxicity profile for
hibitors may induce p53 mutation, which would these apoptosis-inducing agents, and objective
render tumor cells resistant. In fact, treatment clinical responses have been demonstrated. Per-
of p53 WT SJSA-1 cells with Nutlin-3 in vitro haps not surprisingly, these molecularly tar-
leads to the selection for p53 mutations and geted agents are effective only as single agents in
subsequent drug resistance (100), although this a subset of human cancer models in preclinical
observation has yet to be confirmed in vivo. evaluation. Accordingly, development of pre-
Nevertheless, drug-induced p53 mutation by dictive biomarkers will be critical for their suc-
MDM2 inhibitors should be evaluated in clini- cessful development as monotherapies. In ad-
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

cal trials. dition, because it is expected that these new


compounds will be combined with standard of
care (chemotherapy and radiation therapy) in
CONCLUDING REMARKS most cases, development of rational combina-
Over the past decade, major advancements tion strategies will be essential. Moreover, in-
have been made in the development of small depth understanding of de novo or acquired
molecule inhibitors to target the apoptosis resistant mechanisms is needed to develop
pathways. In this review, we focus on three strategies to combat clinical resistance that has
different classes of compounds: inhibitors tar- been observed for all molecularly targeted an-
geting the antiapoptotic BCL-2 proteins, in- ticancer drugs.

DISCLOSURE STATEMENT
S.W. is a consultant and owns stock in Ascenta and Ascentage Pharma, which have licensed IAP,
MDM2, or BCL-2/BCL-XL inhibitors from the University of Michigan for clinical development,
and receives royalty from the University of Michigan on these inventions.

ACKNOWLEDGMENTS
We thank the past and present members of the Wang laboratory for their contributions to the
discovery of small molecule inhibitors targeting BCL-2, IAP, and MDM2 and our development
partners (Ascenta, Sanofi, DebioPharma, and Ascentage) for clinical development of these new
agents. Funding from the National Cancer Institute, National Institutes of Health, the Department
of Defense, the Breast Cancer Research Foundation, the Prostate Cancer Foundation, the Susan
G. Komen Foundation, the Leukemia and Lymphoma Society, Ascenta, Sanofi, and Ascentage
Pharma is greatly appreciated. The authors apologize to the many authors whose works have not
been cited owing to space limitations or the narrow scope of this review.

LITERATURE CITED
1. Cory S, Huang DC, Adams JM. 2003. The BCL-2 family: roles in cell survival and oncogenesis. Oncogene
22(53):8590–607

20.12 Bai · Wang

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

2. Li Q, Lozano G. 2013. Molecular pathways: targeting MDM2 and MDM4 in cancer therapy.
Clin. Cancer Res. 19(1):34–41
3. Letai A, Bassik MC, Walensky LD, et al. 2002. Distinct BH3 domains either sensitize or activate mito-
chondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2(3):183–92
4. Kim H, Rafiuddin-Shah M, Tu HC, et al. 2006. Hierarchical regulation of mitochondrion-dependent
apoptosis by BCL-2 subfamilies. Nat. Cell Biol. 8(12):1348–58
5. Sattler M, Liang H, Nettesheim D, et al. 1997. Structure of BCL-XL-BAK peptide complex: recognition
between regulators of apoptosis. Science 275(5302):983–86
6. Souers AJ, Leverson JD, Boghaert ER, et al. 2013. ABT-199, a potent and selective BCL-2 inhibitor,
achieves antitumor activity while sparing platelets. Nat. Med. 19(2):202–8
7. Oltersdorf T, Elmore SW, Shoemaker AR, et al. 2005. An inhibitor of BCL-2 family proteins induces
regression of solid tumours. Nature 435(7042):677–81
8. Zheng CH, Yang H, Zhang M, et al. 2012. Design, synthesis, and activity evaluation of broad-spectrum
small-molecule inhibitors of anti-apoptotic BCL-2 family proteins: characteristics of broad-spectrum
protein binding and its effects on anti-tumor activity. Bioorg. Med. Chem. Lett. 22(1):39–44
9. Tse C, Shoemaker AR, Adickes J, et al. 2008. ABT-263: a potent and orally bioavailable BCL-2 family
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

inhibitor. Cancer Res. 68(9):3421–28


10. Lessene G, Czabotar PE, Sleebs BE, et al. 2013. Structure-guided design of a selective BCL-XL inhibitor.
Nat. Chem. Biol. 9(6):390–97
11. Zhou H, Chen J, Meagher JL, et al. 2012. Design of BCL-2 and BCL-XL inhibitors with subnanomolar
binding affinities based upon a new scaffold. J. Med. Chem. 55(10):4664–82
12. Chen J, Zhou H, Aguilar A, et al. 2012. Structure-based discovery of BM-957 as a potent small-
molecule inhibitor of BCL-2 and BCL-XL capable of achieving complete tumor regression. J. Med.
Chem. 55(19):8502–14
13. Vogler M, Weber K, Dinsdale D, et al. 2009. Different forms of cell death induced by putative BCL2
inhibitors. Cell Death Differ. 16(7):1030–39
14. Tahir SK, Yang X, Anderson MG, 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):1176–83
15. Konopleva M, Contractor R, Tsao T, et al. 2006. Mechanisms of apoptosis sensitivity and resistance to
the BH3 mimetic ABT-737 in acute myeloid leukemia. Cancer Cell 10(5):375–88
16. Tahir SK, Wass J, Joseph MK, et al. 2010. Identification of expression signatures predictive of sensitivity
to the BCL-2 family member inhibitor ABT-263 in small cell lung carcinoma and leukemia/lymphoma
cell lines. Mol. Cancer Ther. 9(3):545–57
17. Rooswinkel RW, van de Kooij B, Verheij M, et al. 2012. BCL-2 is a better ABT-737 target than BCL-XL
or BCL-w and only Noxa overcomes resistance mediated by MCL-1, BFL-1, or BCL-B. Cell Death Dis.
3:e366
18. Merino D, Khaw SL, Glaser SP, et al. 2012. BCL-2, BCL-x(L), and BCL-w are not equivalent targets
of ABT-737 and navitoclax (ABT-263) in lymphoid and leukemic cells. Blood 119(24):5807–16
19. Ackler S, Mitten MJ, Foster K, 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):869–80
20. Chen J, Jin S, Abraham V, 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):2340–49
21. Shi J, Zhou Y, Huang HC, et al. 2011. Navitoclax (ABT-263) accelerates apoptosis during drug-induced
mitotic arrest by antagonizing BCL-XL. Cancer Res. 71(13):4518–26
22. Tan N, Wong M, Nannini MA, et al. 2013. BCL-2/BCL-XL inhibition increases the efficacy of MEK
inhibition alone and in combination with PI3 kinase inhibition in lung and pancreatic tumor models.
Mol. Cancer Ther. 12:853–64
23. Roberts AW, Seymour JF, Brown JR, 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):488–96

www.annualreviews.org • Apoptosis-Based Cancer Therapies 20.13

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

24. Wilson WH, O’Connor OA, Czuczman MS, et al. 2010. Navitoclax, a targeted high-affinity inhibitor
of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, phar-
macodynamics, and antitumour activity. Lancet Oncol. 11(12):1149–59
25. Gandhi L, Camidge DR, Ribeiro de Oliveira M, 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):909–16
26. Zhang H, Nimmer PM, Tahir SK, et al. 2007. BCL-2 family proteins are essential for platelet survival.
Cell Death Differ. 14(5):943–51
27. Schoenwaelder SM, Jarman KE, Gardiner EE, et al. 2011. BCL-xL-inhibitory BH3 mimetics can induce
a transient thrombocytopathy that undermines the hemostatic function of platelets. Blood 118(6):1663–74
28. Rudin CM, Hann CL, Garon EB, 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):3163–69
29. Vandenberg CJ, Cory S. 2013. ABT-199, a new BCL-2-specific BH3 mimetic, has in vivo efficacy against
aggressive Myc-driven mouse lymphomas without provoking thrombocytopenia. Blood 121(12):2285–88
30. Humerickhouse R. 2013. Remarkable early clinical activity with the BCL-2 selective inhibitor ABT-199:
proving the concept. Presented at AACR Annu. Meet., Apr. 6–10, Washington, DC
31. Seymour JF, Davids MS, Pagel JM, et al. 2013. Updated results of a phase I first-in-human study of the
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

BCL-2 inhibitor ABT-199 (GDC-0199) in patients with relapsed/refractory (R/R) chronic lymphocytic
leukemia (CLL). J. Clin. Oncol. 31(Suppl.): abstr. 7018
32. Zhai D, Jin C, Satterthwait AC, et al. 2006. Comparison of chemical inhibitors of antiapoptotic BCL-
2-family proteins. Cell Death Differ. 13(8):1419–21
33. Nguyen M, Marcellus RC, Roulston A, et al. 2007. Small molecule obatoclax (GX15-070) antago-
nizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis. Proc. Natl. Acad. Sci. USA
104(49):19512–17
34. Konopleva M, Watt J, Contractor R, et al. 2008. Mechanisms of antileukemic activity of the novel BCL-2
homology domain-3 mimetic GX15-070 (obatoclax). Cancer Res. 68(9):3413–20
35. Goard CA, Schimmer AD. 2013. An evidence-based review of obatoclax mesylate in the treatment of
hematological malignancies. Core Evid. 8:15–26
36. Balakrishnan K, Wierda WG, Keating MJ, et al. 2008. Gossypol, a BH3 mimetic, induces apoptosis in
chronic lymphocytic leukemia cells. Blood 112(5):1971–80
37. Liu G, Kelly WK, Wilding G, et al. 2009. An open-label, multicenter, phase I/II study of single-agent
AT-101 in men with castrate-resistant prostate cancer. Clin. Cancer Res. 15(9):3172–76
38. Baggstrom MQ, Qi Y, Koczywas M, et al. 2011. A phase II study of AT-101 (Gossypol) in chemotherapy-
sensitive recurrent extensive-stage small cell lung cancer. J. Thorac. Oncol. 6(10):1757–60
39. Heist RS, Fain J, Chinnasami B, et al. 2010. Phase I/II study of AT-101 with topotecan in relapsed and
refractory small cell lung cancer. J. Thorac. Oncol. 5(10):1637–43
40. Ready N, Karaseva NA, Orlov SV, et al. 2011. Double-blind, placebo-controlled, randomized phase II
study of the proapoptotic agent AT-101 plus docetaxel, in second-line non-small cell lung cancer.
J. Thorac. Oncol. 6(4):781–85
41. Wong M, Tan N, Zha J, et al. 2012. Navitoclax (ABT-263) reduces BCL-X(L)-mediated chemoresistance
in ovarian cancer models. Mol. Cancer Ther. 11(4):1026–35
42. Deng J, Carlson N, Takeyama K, et al. 2007. BH3 profiling identifies three distinct classes of apoptotic
blocks to predict response to ABT-737 and conventional chemotherapeutic agents. Cancer Cell 12(2):171–
85
43. Morales AA, Kurtoglu M, Matulis SM, et al. 2011. Distribution of BIM determines MCL-1 dependence
or codependence with BCL-xL/BCL-2 in MCL-1-expressing myeloma cells. Blood 118(5):1329–39
44. Del Gaizo Moore V, Brown JR, Certo M, et al. 2007. Chronic lymphocytic leukemia requires BCL2 to
sequester prodeath BIM, explaining sensitivity to BCL2 antagonist ABT-737. J. Clin. Invest. 117(1):112–
21
45. Al-Harbi S, Hill BT, Mazumder S, et al. 2011. An antiapoptotic BCL-2 family expression index predicts
the response of chronic lymphocytic leukemia to ABT-737. Blood 118(13):3579–90
46. Bodet L, Gomez-Bougie P, Touzeau C, et al. 2011. ABT-737 is highly effective against molecular
subgroups of multiple myeloma. Blood 118(14):3901–10

20.14 Bai · Wang

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

47. Du C, Fang M, Li Y, et al. 2000. SMAC, a mitochondrial protein that promotes cytochrome c-dependent
caspase activation by eliminating IAP inhibition. Cell 102(1):33–42
48. 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):988–94
49. Varfolomeev E, Goncharov T, Fedorova AV, et al. 2008. c-IAP1 and c-IAP2 are critical mediators of
tumor necrosis factor alpha (TNFα)-induced NF-κB activation. J. Biol. Chem. 283(36):24295–99
50. Bertrand MJ, Milutinovic S, Dickson KM, 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
51. Vallabhapurapu S, Matsuzawa A, Zhang W, et al. 2008. Nonredundant and complementary functions
of TRAF2 and TRAF3 in a ubiquitination cascade that activates NIK-dependent alternative NF-κB
signaling. Nat. Immunol. 9(12):1364–70
52. Zarnegar BJ, Wang Y, Mahoney DJ, et al. 2008. Noncanonical NF-κB activation requires coordinated
assembly of a regulatory complex of the adaptors cIAP1, cIAP2, TRAF2 and TRAF3 and the kinase
NIK. Nat. Immunol. 9(12):1371–78
53. Fulda S, Vucic D. 2012. Targeting IAP proteins for therapeutic intervention in cancer. Nat. Rev. Drug
Discov. 11(2):109–24
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org

54. Fulda S. 2008. Targeting inhibitor of apoptosis proteins (IAPs) for cancer therapy. Anticancer Agents
by Grand Valley State University on 11/13/13. For personal use only.

Med. Chem. 8(5):533–39


55. Cheung HH, Mahoney DJ, Lacasse EC, et al. 2009. Down-regulation of c-FLIP enhances death of
cancer cells by SMAC mimetic compound. Cancer Res. 69(19):7729–38
56. Varfolomeev E, Blankenship JW, Wayson SM, et al. 2007. IAP antagonists induce autoubiquitination
of c-IAPs, NF-κB activation, and TNFα-dependent apoptosis. Cell 131(4):669–81
57. Lu J, Bai L, Sun H, et al. 2008. SM-164: a novel, bivalent SMAC mimetic that induces apoptosis and tumor
regression by concurrent removal of the blockade of cIAP-1/2 and XIAP. Cancer Res. 68(22):9384–93
58. Ndubaku C, Varfolomeev E, Wang L, et al. 2009. Antagonism of c-IAP and XIAP proteins is required
for efficient induction of cell death by small-molecule IAP antagonists. ACS Chem. Biol. 4(7):557–66
59. Flygare JA, Beresini M, Budha N, et al. 2012. 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):4101–13
60. Tolcher AW, Papadopoulos KP, Patnaik A, et al. 2013. Phase I study of safety and pharmacokinetics
(PK) of GDC-0917, an antagonist of inhibitor of apoptosis (IAP) proteins in patients (Pts) with refractory
solid tumors or lymphoma. J. Clin. Oncol. 31(Suppl.): abstr. 2503
61. Allensworth JL, Sauer SJ, Lyerly HK, et al. 2013. SMAC mimetic birinapant induces apoptosis and
enhances TRAIL potency in inflammatory breast cancer cells in an IAP-dependent and TNF-α-
independent mechanism. Breast Cancer Res. Treat. 137(2):359–71
62. Amaravadi RK, Schilder RJ, Dy GK, et al. 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. Proc. Annu. Meet. Am. Assoc. Cancer Res., 102nd, Orlando, FL, Apr. 2–6.
Philadelphia, PA: AACR
63. Amaravadi RK, Senzer NN, Martin LP, et al. 2013. A phase I study of birinapant (TL32711) combined
with multiple chemotherapies evaluating tolerability and clinical activity for solid tumor patients. J. Clin.
Oncol. 31(Suppl.): abstr. 2504
64. Senzer NN, LoRusso P, Martin LP, et al. 2013. Phase II clinical activity and tolerability of the SMAC-
mimetic birinapant (TL32711) plus irinotecan in irinotecan-relapsed/refractory metastatic colorectal
cancer. J. Clin. Oncol. 31(Suppl.): abstr. 3621
65. Sharma SK, Straub C, Zawel L. 2006. Development of peptidomimetics targeting IAPs. Int. J. Pept. Res.
Ther. 12(1):21–32
66. Dienstmann R, Vidal L, Dees EC, et al. 2012. A phase Ib study of LCL161, an oral inhibitor of apoptosis
(IAP) antagonist, in combination with weekly paclitaxel in patients with advanced solid tumors. Cancer
Res. 72(Suppl. 24):P6–11-06
67. Houghton PJ, Kang MH, Reynolds CP, et al. 2012. Initial testing (stage 1) of LCL161, a SMAC mimetic,
by the Pediatric Preclinical Testing Program. Pediatr. Blood Cancer 58(4):636–39

www.annualreviews.org • Apoptosis-Based Cancer Therapies 20.15

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

68. Sikic BI, Eckhardt SG, Gallant G, et al. 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. 29(Suppl.): abstr. 3008
69. Cai Q, Sun H, Peng Y, et al. 2011. A potent and orally active antagonist (SM-406/AT-406) of multi-
ple inhibitor of apoptosis proteins (IAPs) in clinical development for cancer treatment. J. Med. Chem.
54(8):2714–26
70. Erickson RI, Tarrant J, Cain G, et al. 2013. Toxicity profile of small-molecule IAP antagonist GDC-0152
is linked to TNF-α pharmacology. Toxicol. Sci. 131(1):247–58
71. Feoktistova M, Geserick P, Kellert B, et al. 2011. cIAPs block Ripoptosome formation, a RIP1/caspase-
8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol. Cell
43(3):449–63
72. Tenev T, Bianchi K, Darding M, et al. 2011. The ripoptosome, a signaling platform that assembles in
response to genotoxic stress and loss of IAPs. Mol. Cell 43(3):432–48
73. Wagner L, Marschall V, Karl S, et al. 2013. SMAC mimetic sensitizes glioblastoma cells to
temozolomide-induced apoptosis in a RIP1- and NF-κB-dependent manner. Oncogene 32(8):988–97
74. Fridman JS, Lowe SW. 2003. Control of apoptosis by p53. Oncogene 22(56):9030–40
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org

75. Chipuk JE, Kuwana T, Bouchier-Hayes L, et al. 2004. Direct activation of BAX by p53 mediates mito-
by Grand Valley State University on 11/13/13. For personal use only.

chondrial membrane permeabilization and apoptosis. Science 303(5660):1010–14


76. Leu JI, Dumont P, Hafey M, et al. 2004. Mitochondrial p53 activates BAK and causes disruption of a
BAK-MCL1 complex. Nat. Cell Biol. 6(5):443–50
77. Mihara M, Erster S, Zaika A, et al. 2003. p53 has a direct apoptogenic role at the mitochondria.
Mol. Cell 11(3):577–90
78. Momand J, Zambetti GP, Olson DC, et al. 1992. The MDM-2 oncogene product forms a complex with
the p53 protein and inhibits p53-mediated transactivation. Cell 69(7):1237–45
79. Oliner JD, Pietenpol JA, Thiagalingam S, et al. 1993. Oncoprotein MDM2 conceals the activation
domain of tumour suppressor p53. Nature 362(6423):857–60
80. Barak Y, Juven T, Haffner R, et al. 1993. MDM2 expression is induced by wild type p53 activity. EMBO
J. 12(2):461–68
81. Momand J, Jung D, Wilczynski S, et al. 1998. The MDM2 gene amplification database. Nucleic Acids
Res. 26(15):3453–59
82. Kussie PH, Gorina S, Marechal V, et al. 1996. Structure of the MDM2 oncoprotein bound to the p53
tumor suppressor transactivation domain. Science 274(5289):948–53
83. Vassilev LT, Vu BT, Graves B, et al. 2004. In vivo activation of the p53 pathway by small-molecule
antagonists of MDM2. Science 303(5659):844–48
84. Tovar C, Rosinski J, Filipovic Z, et al. 2006. Small-molecule MDM2 antagonists reveal aberrant p53
signaling in cancer: implications for therapy. Proc. Natl. Acad. Sci. USA 103(6):1888–93
85. Tovar C, Graves B, Packman K, et al. 2013. MDM2 small-molecule antagonist RG7112 activates p53
signaling and regresses human tumors in preclinical cancer models. Cancer Res. 73(8):2587–97
86. Vu B, Wovkulich P, Pizzolato G, et al. 2013. Discovery of RG7112: a small-molecule MDM2 inhibitor
in clinical development. ACS Med. Chem. Lett. 4(5):466–69
87. Ray-Coquard I, Blay JY, Italiano A, et al. 2012. Effect of the MDM2 antagonist RG7112 on the p53
pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: an ex-
ploratory proof-of-mechanism study. Lancet Oncol. 13(11):1133–40
88. Shangary S, Qin D, McEachern D, et al. 2008. Temporal activation of p53 by a specific MDM2 inhibitor
is selectively toxic to tumors and leads to complete tumor growth inhibition. Proc. Natl. Acad. Sci. USA
105(10):3933–38
89. Montes de Oca Luna R, Wagner DS, Lozano G. 1995. Rescue of early embryonic lethality in MDM2-
deficient mice by deletion of p53. Nature 378(6553):203–6
90. Jones SN, Roe AE, Donehower LA, et al. 1995. Rescue of embryonic lethality in MDM2-deficient mice
by absence of p53. Nature 378(6553):206–8
91. Grier JD, Xiong S, Elizondo-Fraire AC, et al. 2006. Tissue-specific differences of p53 inhibition by
MSM2 and MDM4. Mol. Cell. Biol. 26(1):192–98

20.16 Bai · Wang

Changes may still occur before final publication online and in print
ME65CH20-Wang ARI 11 October 2013 11:20

92. Francoz S, Froment P, Bogaerts S, et al. 2006. MDM4 and MDM2 cooperate to inhibit p53 activity in
proliferating and quiescent cells in vivo. Proc. Natl. Acad. Sci. USA 103(9):3232–37
93. Xiong S, Van Pelt CS, Elizondo-Fraire AC, et al. 2006. Synergistic roles of MDM2 and MDM4 for p53
inhibition in central nervous system development. Proc. Natl. Acad. Sci. USA 103(9):3226–31
94. Liu D, Ou L, Clemenson GD Jr, et al. 2010. Puma is required for p53-induced depletion of adult stem
cells. Nat. Cell Biol. 12(10):993–98
95. Terzian T, Wang Y, Van Pelt CS, et al. 2007. Haploinsufficiency of MDM2 and MDM4 in tumorigenesis
and development. Mol. Cell. Biol. 27(15):5479–85
96. Laurie NA, Donovan SL, Shih CS, et al. 2006. Inactivation of the p53 pathway in retinoblastoma. Nature
444(7115):61–66
97. Wade M, Wong ET, Tang M, et al. 2006. HDMX modulates the outcome of p53 activation in human
tumor cells. J. Biol. Chem. 281(44):33036–44
98. Patton JT, Mayo LD, Singhi AD, et al. 2006. Levels of HDMX expression dictate the sensitivity of
normal and transformed cells to Nutlin-3. Cancer Res. 66(6):3169–76
99. Vaseva AV, Yallowitz AR, Marchenko ND, et al. 2011. Blockade of Hsp90 by 17AAG antagonizes
MDMX and synergizes with Nutlin to induce p53-mediated apoptosis in solid tumors. Cell Death Dis.
2:e156
Annu. Rev. Med. 2014.65. Downloaded from www.annualreviews.org
by Grand Valley State University on 11/13/13. For personal use only.

100. Aziz MH, Shen H, Maki CG. 2011. Acquisition of p53 mutations in response to the non-genotoxic p53
activator Nutlin-3. Oncogene 30(46):4678–86

www.annualreviews.org • Apoptosis-Based Cancer Therapies 20.17

Changes may still occur before final publication online and in print

You might also like