Inhibitor of apoptosis (IAP) proteins play a critical role in the control of survival and cell death by regulating key signaling events such as caspase activation and NF-κB signaling. Because aberrantly high expression of IAP proteins represents a frequent oncogenic event in human cancers, therapeutic targeting of IAP proteins is considered as a promising approach. Several small-molecule pharmacologic inhibitors of IAP proteins that mimic the binding domain of the endogenous IAP antagonist second mitochondrial activator of caspases (Smac) to IAP proteins have been developed over the past few years. IAP antagonists have been shown in various preclinical cancer models to either directly initiate cell death or, alternatively, to prime cancer cells for cytotoxic therapies by lowering the threshold for cell death induction. IAP antagonists (i.e., GDC-0917/CUDC-427, LCL161, AT-406, HGS1029, and TL32711) are currently under evaluation in early clinical trials alone or in combination regimens. Thus, the concept to therapeutically target IAP proteins in human cancer has in principle been successfully transferred into a clinical setting and warrants further evaluation as a treatment approach. Clin Cancer Res; 20(2); 289–95. ©2013 AACR.

No potential conflicts of interest were disclosed.

The members of the planning committee have no real or apparent conflict of interest to disclose.

Upon completion of the CME activity, the participant should have a better understanding of the molecular pathways that are regulated by IAP proteins. In addition, the participant should understand the rationale for the design of current therapeutics aimed at antagonizing IAP proteins and the preclinical or clinical development of IAP antagonists in cancer.

This activity does not receive commercial support.

Inhibitor of apoptosis (IAP) proteins are a family of eight human proteins, including neuronal AIP (NAIP), cellular IAP1 (cIAP1), cellular IAP2 (cIAP2), X chromosome-linked IAP (XIAP), survivin, baculovirus IAP repeat (BIR)–containing ubiquitin-conjugating enzyme (BRUCE/Apollon), melanoma IAP (ML-IAP), and IAP-like protein 2 [ILP-2; reviewed in ref. 1)]. As reflected by their name, they were initially characterized as endogenous inhibitors of caspases. However, besides the regulation of apoptosis, IAP proteins have also been implicated in the control of nonapoptotic processes, including differentiation, cell motility, migration, invasion, and metastasis (2).

All IAP proteins contain at least one of the signature BIR domains, a 70–80 amino acid segment that mediates protein–protein interactions between IAP proteins and caspases, thereby inhibiting caspase activation and activity. IAP proteins such as XIAP, cIAP1, cIAP2, and ML-IAP also contain the Really Interesting New Gene (RING) domain that exhibits E3 ubiquitin ligase activity (3). Depending on the chain type (e.g., K5-, K11-, K48-, or K63-linked chains), ubiquitination can lead to proteasomal degradation of substrates or can alter their signaling properties (3).

IAP proteins are key regulators of programmed cell death pathways. Apoptosis represents one of the best-characterized forms of programmed cell death and involves two major signaling pathways (4). In the extrinsic (death receptor) pathway, the binding of death receptor ligands such as TRAIL, to death receptors, such as TRAIL receptors, triggers recruitment of adaptor molecules [e.g., FAS-associated protein with death domain (FADD)] and caspase-8, which in turn drives activation of caspase-8 and, subsequently, of effector caspases such as caspase-3 (4). Furthermore, caspase-8–mediated cleavage of Bid into tBid provides a link between the extrinsic and intrinsic apoptosis pathways (4). Upon the engagement of the intrinsic (mitochondrial) pathway, mitochondrial proteins such as cytochrome c or second mitochondrial activator of caspases (Smac) are released into the cytosol and trigger apoptosis by promoting caspase activation (4). In the case of Smac, caspases are indirectly activated via Smac-mediated neutralization of IAP proteins, which in turn results in caspase activation. IAP proteins, in particular XIAP, block caspase activation by binding to caspases -3, -7, and -9 via the BIR domains (1) and negatively regulate both the intrinsic and extrinsic apoptosis pathways.

Furthermore, IAP proteins play an important role in the regulation of NF-κB signaling, in particular via ubiquitination events (1). NF-κB represents one of the key transcription factors that control various aspects of tumor biology, including cell death and survival signaling (5). Within the canonical NF-κB pathway, binding of TNF-α to TNF receptor 1 (TNFR1) triggers the recruitment of signaling molecules such as TNFR1-associated death domain protein (TRADD), receptor-interacting protein 1 (RIP1), TNF receptor-associated factor 2 (TRAF2), cIAP1, and cIAP2, which results in nondegradative ubiquitination of RIP1 via cIAP proteins and activation of inhibitor of NF-κB kinase (IKK; IκB kinase β) via a multiprotein complex containing TGF-β–activated kinase 1 (TAK1), TAK1-binding protein (TAB), and NF-κB essential modulator (NEMO; ref. 6). This leads to phosphorylation and proteasomal degradation of IκBα followed by nuclear translocation of NF-κB subunits to activate transcription of NF-κB target genes. NF-κB–responsive genes comprise proinflammatory genes, for example TNF-α and interleukin-8, antiapoptotic genes such as XIAP, Bcl-2, or Bcl-XL, as well as proapoptotic genes such as CD95 and TRAIL receptors (5). cIAP proteins promote canonical NF-κB activation by ubiquitinating RIP1 (3).

Within the noncanonical NF-κB pathway, cIAP proteins mediate under resting conditions the constitutive ubiquitination and proteasomal degradation of NF-κB–inducing kinase (NIK) together with TRAF2 and TRAF3, thereby negatively regulating noncanonical NF-κB signaling (7, 8). Activation of noncanonical NF-κB signaling, for example, in response to CD40 stimulation, terminates this cIAP-dependent NIK degradation and allows activation of IKKα via NIK, followed by processing of p100 to p52 and translocation of p52 to the nucleus to activate NF-κB target genes (9).

Moreover, IAP proteins have been implicated in the regulation of additional signaling cascades, for example, the mitogen-activated protein kinase (MAPK) pathway (10), TGF-β signaling (11, 12) and innate and adaptive immunity signaling pathways (13, 14). cIAP proteins have been shown to be required for activation of MAPK signaling by members of the TNFR superfamily (10). XIAP has been described to function as a cofactor in TGF-β signaling (11). The immunomodulatory functions of IAP proteins are mediated via their regulation of the NF-κB and MAPK signaling pathways or via the control of the activity of the inflammasome, a multiprotein complex that regulates an immune response to microbial triggers (13).

IAP proteins are highly expressed in multiple human malignancies and have been implicated in promoting tumor progression, treatment failure, and poor prognosis, indicating that IAP proteins represent relevant targets for therapeutic exploitation (1). Indeed, IAP proteins have been shown to play a critical role in the regulation of sensitivity versus resistance of cancers to current cytotoxic strategies. Therefore, many efforts have been made over the last decade to develop strategies to neutralize IAP proteins, including antisense oligonucleotides and small-molecule inhibitors (15, 16). Due to space limitations, the current review focuses on small-molecule inhibitors for therapeutic inhibition of IAP proteins.

In principle, IAP antagonists mimic the N-terminal part of the endogenous IAP antagonist Smac, which is required for binding to IAP proteins, and are composed of nonpeptidic elements (1). In addition to monovalent compounds that contain one Smac-mimicking motif, bivalent or dimeric IAP antagonists were developed, which consist of two monovalent Smac-mimicking units that are connected via a chemical linker. Bivalent IAP antagonists were reported to exhibit a several-fold higher antitumor activity than monovalent ones, due to their higher binding affinities and their higher potency to promote caspase activation and proteasomal degradation of cIAP proteins (17–20).

Similar to the Smac protein, small-molecule IAP antagonists bind to several IAP proteins, including XIAP, cIAP1, cIAP2, and ML-IAP. In principle, chemically distinct IAP antagonists can differentially neutralize IAP proteins. The binding of IAP antagonists to XIAP results in activation of caspases (21, 22). The interaction of IAP antagonists with cIAP proteins stimulates their dimerization and increases their E3 ligase activity (17–20, 23). This leads to increased autoubiquitination and degradation of cIAP proteins via the proteasome. Because cIAP proteins are responsible for the constitutive proteasomal degradation of NIK, a critical upstream component of the noncanonical NF-κB pathway (7, 8), IAP antagonist–mediated depletion of cIAP proteins leads to NIK accumulation and noncanonical NF-κB activation (Fig. 1). Subsequent upregulation of NF-κB target genes such as TNF-α can then engage TNFR1-mediated caspase-8 activation and apoptosis via a RIP1/FADD/caspase-8 cytosolic complex in an autocrine/paracrine manner (17–20) (Fig. 1). By depleting cIAP proteins, IAP antagonists suppress activation of the canonical NF-κB pathway. Initially, however, they may contribute to canonical NF-κB activation by stimulating the E3 ubiquitin ligase activity of cIAP proteins, thereby promoting RIP1 ubiquitination and NF-κB activation. This initial increase in E3 ligase activity of cIAP proteins is usually only transient because cIAP proteins are themselves autoubiquitinated and degraded. Five distinct IAP antagonists are currently under clinical development (Table 1).

Table 1.

Clinical trials with IAP antagonists

CompoundCombinationCancer typeStatusPhase I/IITrial
LCL-161 None Solid tumors Completed Phase I NCT01098838 
LCL-161 Paclitaxel Solid tumors Recruiting Phase I NCT01240655 
LCL-161 Paclitaxel Breast cancer Recruiting Phase II NCT01617668 
AT-406 None Solid tumors, lymphomas Recruiting Phase I NCT01078649 
AT-406 Daunorubicin, cytarabine AML Terminated Phase I NCT01265199 
HSG1029 None Solid tumors Completed Phase I NCT00708006 
HSG1029 None Lymphoid malignancies Terminated Phase I NCT01013818 
TL32711 None Solid tumors Active, not recruiting Phase I/II NCT01188499 
TL32711 GEM Solid tumors Recruiting Phase I NCT01573780 
TL32711 None Solid tumors, lymphomas Completed Phase I NCT00993239 
TL32711 None AML Recruiting Phase I/II NCT01486784 
TL32711 5-Aza MDS Not yet recruiting Phase I/II NCT01828346 
TL32711 None Ovarian, peritoneal cancer Recruiting Phase II NCT01681368 
GDC-0152 None Solid tumors Completed Phase I NCT00977067 
CUDC-427 None Solid tumors, lymphomas Recruiting Phase I NCT01908413 
CompoundCombinationCancer typeStatusPhase I/IITrial
LCL-161 None Solid tumors Completed Phase I NCT01098838 
LCL-161 Paclitaxel Solid tumors Recruiting Phase I NCT01240655 
LCL-161 Paclitaxel Breast cancer Recruiting Phase II NCT01617668 
AT-406 None Solid tumors, lymphomas Recruiting Phase I NCT01078649 
AT-406 Daunorubicin, cytarabine AML Terminated Phase I NCT01265199 
HSG1029 None Solid tumors Completed Phase I NCT00708006 
HSG1029 None Lymphoid malignancies Terminated Phase I NCT01013818 
TL32711 None Solid tumors Active, not recruiting Phase I/II NCT01188499 
TL32711 GEM Solid tumors Recruiting Phase I NCT01573780 
TL32711 None Solid tumors, lymphomas Completed Phase I NCT00993239 
TL32711 None AML Recruiting Phase I/II NCT01486784 
TL32711 5-Aza MDS Not yet recruiting Phase I/II NCT01828346 
TL32711 None Ovarian, peritoneal cancer Recruiting Phase II NCT01681368 
GDC-0152 None Solid tumors Completed Phase I NCT00977067 
CUDC-427 None Solid tumors, lymphomas Recruiting Phase I NCT01908413 

Abbreviations: AML, acute myeloid leukemia; GEM, gemcitabine; MDS, myelodysplastic syndrome; 5-Aza, 5-azacytidine.

Figure 1.

Regulation of apoptosis and NF-κB signaling by IAP proteins and their antagonists. XIAP negatively regulates both the extrinsic and intrinsic apoptosis pathways by inhibiting caspases-3 and -9. The extrinsic (death receptor) pathway is triggered upon ligation of death receptors such as TRAIL receptors (TRAIL-R) by their ligands such as TRAIL, leading to activation of caspases-8 and -3. The intrinsic (mitochondrial) pathway is engaged by the release of mitochondrial proteins such as cytochrome c (CytC) or Smac into the cytosol, which promotes caspase-9 and -3 activation. Neutralization of XIAP-mediated caspase inhibition by IAP antagonists promotes caspase-dependent apoptosis. cIAP proteins (cIAP) control activation of canonical and noncanonical NF-κB pathways. cIAP proteins promote canonical NF-κB activation via nondegradative ubiquitination of RIP1, whereas they inhibit noncanonical NF-κB signaling via ubiquitination of NIK and its degradation via the proteasome. IAP antagonists stimulate the E3 ubiquitin ligase activity of cIAP proteins, thereby promoting their autoubiquitination and proteasomal degradation. In turn, NIK is stabilized and activates noncanonical NF-κB signaling. Induction of NF-κB target genes such as TNF-α can then, in an autocrine/paracrine manner, trigger TNFR1-mediated caspase-8 activation and apoptosis via a RIP1/FADD/caspase-8 cytosolic complex. IAP antagonists suppress activation of the canonical NF-κB pathway by depleting cIAP proteins. Initially, however, they may stimulate canonical NF-κB signaling by increasing the E3 ubiquitin ligase activity of cIAP proteins, which leads to RIP1 ubiquitination and NF-κB activation. Stars designate IAP antagonists and their targets. Please see the text for more details.

Figure 1.

Regulation of apoptosis and NF-κB signaling by IAP proteins and their antagonists. XIAP negatively regulates both the extrinsic and intrinsic apoptosis pathways by inhibiting caspases-3 and -9. The extrinsic (death receptor) pathway is triggered upon ligation of death receptors such as TRAIL receptors (TRAIL-R) by their ligands such as TRAIL, leading to activation of caspases-8 and -3. The intrinsic (mitochondrial) pathway is engaged by the release of mitochondrial proteins such as cytochrome c (CytC) or Smac into the cytosol, which promotes caspase-9 and -3 activation. Neutralization of XIAP-mediated caspase inhibition by IAP antagonists promotes caspase-dependent apoptosis. cIAP proteins (cIAP) control activation of canonical and noncanonical NF-κB pathways. cIAP proteins promote canonical NF-κB activation via nondegradative ubiquitination of RIP1, whereas they inhibit noncanonical NF-κB signaling via ubiquitination of NIK and its degradation via the proteasome. IAP antagonists stimulate the E3 ubiquitin ligase activity of cIAP proteins, thereby promoting their autoubiquitination and proteasomal degradation. In turn, NIK is stabilized and activates noncanonical NF-κB signaling. Induction of NF-κB target genes such as TNF-α can then, in an autocrine/paracrine manner, trigger TNFR1-mediated caspase-8 activation and apoptosis via a RIP1/FADD/caspase-8 cytosolic complex. IAP antagonists suppress activation of the canonical NF-κB pathway by depleting cIAP proteins. Initially, however, they may stimulate canonical NF-κB signaling by increasing the E3 ubiquitin ligase activity of cIAP proteins, which leads to RIP1 ubiquitination and NF-κB activation. Stars designate IAP antagonists and their targets. Please see the text for more details.

Close modal

Evaluation of IAP antagonists as single agents revealed that they effectively trigger cell death in a small subset of human malignancies (19), indicating that IAP antagonist–based combination therapies might be required in the majority of cancers to achieve sufficient antitumor activity. Therefore, a variety of rational targeted combinations have been developed over the years together with different types of cytotoxic stimuli in a large set of cancer entities in vitro and in vivo to exploit additive or synergistic drug interactions (reviewed in ref. 1). It is interesting to note that there is some evidence from preclinical studies pointing to a therapeutic window in IAP antagonist–based combination regimens to prime malignant cancer cells for cell death induction, while sparing nonmalignant normal cells, e.g., peripheral blood lymphocytes, hematopoietic progenitor cells, and neuronal cells (24–27). However, many more studies need to be performed before any definite conclusion can be drawn. IAP antagonist–based combinations range from conventional chemotherapeutic agents and irradiation, the two pillars of many current treatment protocols, to death receptor agonists and various kinds of small-molecule signal transduction inhibitors.

Chemotherapeutic drugs of different pharmacologic classes, including doxorubicin, etoposide, gemcitabine, paclitaxel, cisplatin, vinorelbine, SN38, 5-fluorouracil (5-FU), and cytarabine, were shown to act in concert with IAP antagonists to exert antitumor activity in preclinical models of cancers (26, 28–32). In clinical trials, paclitaxel, daunorubicin, cytarabine, or gemcitabine have been selected for combination protocols (Table 1). Mechanistically, chemosensitization by IAP antagonists has been linked in some studies to an autocrine/paracrine TNF-α–driven loop that is engaged upon depletion of cIAP proteins (31). However, TNF-α–independent signaling events, i.e., the formation of a cell death complex in the cytosol containing RIP1, FADD, and caspase-8, were also shown to be critical for activation of cell death pathways in response to cotreatment with chemotherapeutics and IAP antagonists (32).

In addition to enhancing the antitumor activity of DNA-damaging drugs, IAP antagonists have been reported to increase radiosensitivity in various cancers (27, 33–36). Interestingly, radiosensitization by IAP antagonists was not restricted to the bulk population of the tumor, but was also found in tumor-initiating cancer stem cells that have been described to be particularly radioresistant (27).

Furthermore, the combination of IAP antagonists together with death receptor agonists turned out to be very potent to induce cell death even in otherwise resistant forms of cancer (22, 24, 25, 37–42). Accordingly, simultaneous neutralization of XIAP and cIAP proteins circumvents not only the requirement for mitochondrial amplification of death receptor–mediated apoptosis in many cancers, but also potentiates death receptor–initiated activation of caspase-8 by promoting the aggregation of caspase-8 together with FADD and RIP1 in a multimeric cytosolic complex (complex II) following the release of these signaling molecules from the receptor-bound death-inducing signaling complex (DISC). Just to give two examples, IAP antagonists were found to be able to prime pancreatic carcinoma cells, which are known to be notoriously refractory to most treatment approaches and to cell death induction, when combined with TRAIL receptor agonists such as soluble TRAIL ligand or monoclonal antibodies directed against one of the agonistic TRAIL receptors, resulting in enhanced apoptosis in vitro and reduced tumor growth in vivo (25, 39). Also, chronic lymphocytic leukemia (CLL) cells derived from patients belonging to the poor prognostic subgroups, e.g., those with TP53 mutation, 17p deletion, chemoresistance, or unmutated V(H) status, turned out to remain susceptible to cell death induction by IAP antagonists and TRAIL receptor agonists (38).

Moreover, IAP antagonists have successfully been combined with a range of signal transduction modulators depending on the cancer entity, including proteasome inhibitors, various kinds of kinase inhibitors, e.g., FMS-like tyrosine kinase (FLT)3 inhibitors, platelet-derived growth factor (PDGF) receptor inhibitors, insulin-like growth factor (IGF) receptor inhibitors, or EGF receptor inhibitors, as well as monoclonal antibodies targeting growth factor receptors (43–46). Also, IAP antagonists have been shown to increase the antitumor effects of immunotherapies both by priming cancer cells to immune cell–mediated cytotoxicity and/or by altering immune cell functions (47).

In addition to promoting apoptosis, there is accumulating evidence indicating that IAP antagonists can also potentiate nonapoptotic forms of cell death such as necroptosis, a recently identified programmed form of necrosis (48). Accordingly, IAP antagonists have been reported to promote the formation of the necrosome, a complex consisting of RIP1 and RIP3, under conditions in which caspase activation is inhibited (49, 50). The IAP antagonist–mediated amplification of necroptosis opens new perspectives to overcome treatment resistance, particularly in apoptosis-refractory forms of cancer.

Five distinct IAP antagonists are currently undergoing evaluation in early clinical trials for the treatment of cancer (www.clinicaltrials.gov). Monovalent agents such as GDC-0917/CUDC-427 (Genentech, Inc./Curis), LCL161 (Novartis), and AT-406 (Ascenta Therapeutics) offer the advantage that they can be administered orally. By comparison, bivalent compounds such as HGS1029 (Aegera Therapeutics/Human Genome Sciences) and TL32711 (TetraLogic Pharmaceuticals) require intravenous administration but may turn out to be more efficacious, as indicated from preclinical studies. Nevertheless, the question remains open about whether mono- or bivalent IAP antagonists are the most promising candidates for further clinical development based on their pharmacodynamic and pharmacokinetic properties as well as their toxicity profiles. In addition, the question about which IAP proteins alone or in combination represent the most critical targets for therapeutic intervention in cancer has not yet been answered. Although neutralization of cIAP proteins has been shown to be instrumental for single-agent activity of IAP antagonists by engaging an NF-κB–dependent autocrine/paracrine TNF-α loop that triggers cell death upon concomitant depletion of cIAP proteins, the release of the XIAP-imposed block on caspases is considered to be critical for full activation of the effector phase of apoptosis.

Phase I trials examining the safety and pharmacologic properties of IAP antagonists trials have been completed for LCL161, HGS1029, and TL32711, demonstrating that IAP antagonists are in principle well tolerated (51–53). Furthermore, several combination protocols with IAP antagonists together with standard-of-care anticancer therapeutics have been initiated, including chemotherapeutic drugs such as paclitaxel, daunorubicin, cytarabine, and gemcitabine (Table 1). In addition, a trial testing TL32711 in combination with the demethylating agent 5-azacytidine has recently been launched (Table 1). Interestingly, there is also very recent evidence from preclinical studies showing synergistic antileukemic effects when an IAP antagonist was combined with 5-azacytidine (54).

Accompanying biomarker studies have demonstrated target inhibition by IAP antagonists by showing depletion of cIAP1 protein levels both in surrogate tissues such as peripheral blood mononuclear cells as well as in tumor tissue (51–53). In addition, increased levels of circulating cytokines and chemokines in plasma specimens were used as pharmacodynamic parameters. However, although the sensitivity of cancer cells to monotherapy with IAP antagonists has been linked to their ability to engage an autocrine/paracrine loop of TNF-α production, corresponding in vivo studies largely failed to detect a substantial increase in TNF-α levels in the circulation (19). This may be explained by local production of TNF-α rather than by its widespread release in the circulation. Also, additional cytokines or chemokines may be relevant for the antitumor activity of IAP antagonists. Although these markers may serve as indicators of target antagonism, they will likely not be suitable to predict treatment response.

Pharmacodynamic assays to determine treatment response include, for example, the detection of apoptotic markers in tumor biopsies such as cleavage products of caspases-3, -8, and PARP. However, the determination of markers of apoptotic cell death may not be sufficient to properly assess treatment response, because under certain conditions IAP antagonists can also trigger alternative forms of cell death besides apoptosis, for example necroptosis (49, 50).

In summary, IAP proteins represent promising targets for the development of small-molecule cancer therapeutics, as they are expressed at aberrantly high levels in multiple human malignancies and as they block cell death pathways while supporting cancer cell survival. The therapeutic potential of IAP antagonists may particularly reside in rational combinations together with other cytotoxic strategies to take advantage of additive or synergistic drug interactions. This has convincingly been demonstrated in various preclinical cancer models. In addition, clinical studies have recently been launched to test this concept. As many of these clinical trials are currently ongoing, it is too early to draw conclusions on the clinical efficacy of IAP antagonists.

The author thanks C. Hugenberg for his expert secretarial assistance.

This work has been partially supported by grants from the Deutsche Forschungsgemeinschaft, the Ministerium für Bildung und Forschung (01GM1104C), European Community, IUAP, Wilhelm Sander-Stiftung, and Jose Carreras-Stiftung.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Fulda
S
,
Vucic
D
. 
Targeting IAP proteins for therapeutic intervention in cancer
.
Nat Rev Drug Discov
2012
;
11
:
109
24
.
2.
Fulda
S
. 
Regulation of cell migration, invasion and metastasis by IAP proteins and their antagonists
.
Oncogene
2013
Mar 11. [Epub ahead of print]
.
3.
Vucic
D
,
Dixit
VM
,
Wertz
IE
. 
Ubiquitylation in apoptosis: a post-translational modification at the edge of life and death
.
Nat Rev Mol Cell Biol
2011
;
12
:
439
52
.
4.
Fulda
S
,
Debatin
KM
. 
Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy
.
Oncogene
2006
;
25
:
4798
811
.
5.
Scheidereit
C
. 
IκB kinase complexes: gateways to NF-κB activation and transcription
.
Oncogene
2006
;
25
:
6685
705
.
6.
Chu
WM
. 
Tumor necrosis factor
.
Cancer Lett
2013
;
328
:
222
5
.
7.
Vallabhapurapu
S
,
Matsuzawa
A
,
Zhang
W
,
Tseng
PH
,
Keats
JJ
,
Wang
H
, et al
Nonredundant and complementary functions of TRAF2 and TRAF3 in a ubiquitination cascade that activates NIK-dependent alternative NF-kappaB signaling
.
Nat Immunol
2008
;
9
:
1364
70
.
8.
Zarnegar
BJ
,
Wang
Y
,
Mahoney
DJ
,
Dempsey
PW
,
Cheung
HH
,
He
J
, et al
Noncanonical NF-kappaB activation requires coordinated assembly of a regulatory complex of the adaptors cIAP1, cIAP2, TRAF2 and TRAF3 and the kinase NIK
.
Nat Immunol
2008
;
9
:
1371
8
.
9.
Sun
SC
. 
Non-canonical NF-kappaB signaling pathway
.
Cell Res
2011
;
21
:
71
85
.
10.
Varfolomeev
E
,
Goncharov
T
,
Maecker
H
,
Zobel
K
,
Komuves
LG
,
Deshayes
K
, et al
Cellular inhibitors of apoptosis are global regulators of NF-kappaB and MAPK activation by members of the TNF family of receptors
.
Sci Signal
2012
;
5
:
ra22
.
11.
Birkey Reffey
S
,
Wurthner
JU
,
Parks
WT
,
Roberts
AB
,
Duckett
CS
. 
X-linked inhibitor of apoptosis protein functions as a cofactor in transforming growth factor-beta signaling
.
J Biol Chem
2001
;
276
:
26542
9
.
12.
Hofer-Warbinek
R
,
Schmid
JA
,
Stehlik
C
,
Binder
BR
,
Lipp
J
,
de Martin
R
. 
Activation of NF-kappa B by XIAP, the X chromosome-linked inhibitor of apoptosis, in endothelial cells involves TAK1
.
J Biol Chem
2000
;
275
:
22064
8
.
13.
Beug
ST
,
Cheung
HH
,
Lacasse
EC
,
Korneluk
RG
. 
Modulation of immune signalling by inhibitors of apoptosis
.
Trends Immunol
2012
;
33
:
535
45
.
14.
Lopez
J
,
Meier
P
. 
To fight or die - inhibitor of apoptosis proteins at the crossroad of innate immunity and death
.
Curr Opin Cell Biol
2010
;
22
:
872
81
.
15.
LaCasse
EC
. 
Pulling the plug on a cancer cell by eliminating XIAP with AEG35156
.
Cancer Lett
2013
;
332
:
215
24
.
16.
Straub
CS
. 
Targeting IAPs as an approach to anti-cancer therapy
.
Curr Top Med Chem
2011
;
11
:
291
316
.
17.
Varfolomeev
E
,
Blankenship
JW
,
Wayson
SM
,
Fedorova
AV
,
Kayagaki
N
,
Garg
P
, et al
IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB activation, and TNFalpha-dependent apoptosis
.
Cell
2007
;
131
:
669
81
.
18.
Vince
JE
,
Wong
WW
,
Khan
N
,
Feltham
R
,
Chau
D
,
Ahmed
AU
, et al
IAP antagonists target cIAP1 to induce TNFalpha-dependent apoptosis
.
Cell
2007
;
131
:
682
93
.
19.
Petersen
SL
,
Wang
L
,
Yalcin-Chin
A
,
Li
L
,
Peyton
M
,
Minna
J
, et al
Autocrine TNFalpha signaling renders human cancer cells susceptible to Smac-mimetic-induced apoptosis
.
Cancer Cell
2007
;
12
:
445
56
.
20.
Bertrand
MJ
,
Milutinovic
S
,
Dickson
KM
,
Ho
WC
,
Boudreault
A
,
Durkin
J
, et al
cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination
.
Mol Cell
2008
;
30
:
689
700
.
21.
Gao
Z
,
Tian
Y
,
Wang
J
,
Yin
Q
,
Wu
H
,
Li
YM
, et al
A dimeric Smac/diablo peptide directly relieves caspase-3 inhibition by XIAP. Dynamic and cooperative regulation of XIAP by Smac/Diablo
.
J Biol Chem
2007
;
282
:
30718
27
.
22.
Varfolomeev
E
,
Alicke
B
,
Elliott
JM
,
Zobel
K
,
West
K
,
Wong
H
, et al
X chromosome-linked inhibitor of apoptosis regulates cell death induction by proapoptotic receptor agonists
.
J Biol Chem
2009
;
284
:
34553
60
.
23.
Dueber
EC
,
Schoeffler
AJ
,
Lingel
A
,
Elliott
JM
,
Fedorova
AV
,
Giannetti
AM
, et al
Antagonists induce a conformational change in cIAP1 that promotes autoubiquitination
.
Science
2011
;
334
:
376
80
.
24.
Fakler
M
,
Loeder
S
,
Vogler
M
,
Schneider
K
,
Jeremias
I
,
Debatin
KM
, et al
Small molecule XIAP inhibitors cooperate with TRAIL to induce apoptosis in childhood acute leukemia cells and overcome Bcl-2-mediated resistance
.
Blood
2009
;
113
:
1710
22
.
25.
Vogler
M
,
Walczak
H
,
Stadel
D
,
Haas
TL
,
Genze
F
,
Jovanovic
M
, et al
Small molecule XIAP inhibitors enhance TRAIL-induced apoptosis and antitumor activity in preclinical models of pancreatic carcinoma
.
Cancer Res
2009
;
69
:
2425
34
.
26.
Loeder
S
,
Fakler
M
,
Schoeneberger
H
,
Cristofanon
S
,
Leibacher
J
,
Vanlangenakker
N
, et al
RIP1 is required for IAP inhibitor-mediated sensitization of childhood acute leukemia cells to chemotherapy-induced apoptosis
.
Leukemia
2012
;
26
:
1020
9
.
27.
Vellanki
SH
,
Grabrucker
A
,
Liebau
S
,
Proepper
C
,
Eramo
A
,
Braun
V
, et al
Small-molecule XIAP inhibitors enhance gamma-irradiation-induced apoptosis in glioblastoma
.
Neoplasia
2009
;
11
:
743
52
.
28.
Stadel
D
,
Cristofanon
S
,
Abhari
BA
,
Deshayes
K
,
Zobel
K
,
Vucic
D
, et al
Requirement of nuclear factor kappaB for Smac mimetic-mediated sensitization of pancreatic carcinoma cells for gemcitabine-induced apoptosis
.
Neoplasia
2011
;
13
:
1162
70
.
29.
Carter
BZ
,
Gronda
M
,
Wang
Z
,
Welsh
K
,
Pinilla
C
,
Andreeff
M
, et al
Small-molecule XIAP inhibitors derepress downstream effector caspases and induce apoptosis of acute myeloid leukemia cells
.
Blood
2005
;
105
:
4043
50
.
30.
Dean
EJ
,
Ward
T
,
Pinilla
C
,
Houghten
R
,
Welsh
K
,
Makin
G
, et al
A small molecule inhibitor of XIAP induces apoptosis and synergises with vinorelbine and cisplatin in NSCLC
.
Br J Cancer
2010
;
102
:
97
103
.
31.
Probst
BL
,
Liu
L
,
Ramesh
V
,
Li
L
,
Sun
H
,
Minna
JD
, et al
Smac mimetics increase cancer cell response to chemotherapeutics in a TNF-alpha-dependent manner
.
Cell Death Differ
2010
;
17
:
1645
54
.
32.
Wagner
L
,
Marschall
V
,
Karl
S
,
Cristofanon
S
,
Zobel
K
,
Deshayes
K
, et al
Smac mimetic sensitizes glioblastoma cells to Temozolomide-induced apoptosis in a RIP1- and NF-kappaB-dependent manner
.
Oncogene
2013
;
32
:
988
97
.
33.
Giagkousiklidis
S
,
Vellanki
SH
,
Debatin
KM
,
Fulda
S
. 
Sensitization of pancreatic carcinoma cells for gamma-irradiation-induced apoptosis by XIAP inhibition
.
Oncogene
2007
;
26
:
7006
16
.
34.
Berger
R
,
Jennewein
C
,
Marschall
V
,
Karl
S
,
Cristofanon
S
,
Wagner
L
, et al
NF-{kappa}B is required for Smac mimetic-mediated sensitization of glioblastoma Cells for {gamma}-irradiation-induced apoptosis
.
Mol Cancer Ther
2011
;
10
:
1867
75
.
35.
Ziegler
DS
,
Keating
J
,
Kesari
S
,
Fast
EM
,
Zawel
L
,
Ramakrishna
N
, et al
A small-molecule IAP inhibitor overcomes resistance to cytotoxic therapies in malignant gliomas in vitro and in vivo
.
Neuro Oncol
2011
;
13
:
820
9
.
36.
Yang
D
,
Zhao
Y
,
Li
AY
,
Wang
S
,
Wang
G
,
Sun
Y
. 
Smac-mimetic compound SM-164 induces radiosensitization in breast cancer cells through activation of caspases and induction of apoptosis
.
Breast Cancer Res Treat
2012
;
133
:
189
99
.
37.
Fulda
S
,
Wick
W
,
Weller
M
,
Debatin
KM
. 
Smac agonists sensitize for Apo2L/TRAIL- or anticancer drug-induced apoptosis and induce regression of malignant glioma in vivo
.
Nat Med
2002
;
8
:
808
15
.
38.
Loeder
S
,
Zenz
T
,
Schnaiter
A
,
Mertens
D
,
Winkler
D
,
Dohner
H
, et al
A novel paradigm to trigger apoptosis in chronic lymphocytic leukemia
.
Cancer Res
2009
;
69
:
8977
86
.
39.
Stadel
D
,
Mohr
A
,
Ref
C
,
MacFarlane
M
,
Zhou
S
,
Humphreys
R
, et al
TRAIL-induced apoptosis is preferentially mediated via TRAIL receptor 1 in pancreatic carcinoma cells and profoundly enhanced by XIAP inhibitors
.
Clin Cancer Res
2010
;
16
:
5734
49
.
40.
Li
L
,
Thomas
RM
,
Suzuki
H
,
De Brabander
JK
,
Wang
X
,
Harran
PG
. 
A small molecule Smac mimic potentiates TRAIL- and TNFalpha-mediated cell death
.
Science
2004
;
305
:
1471
4
.
41.
Abhari
BA
,
Cristofanon
S
,
Kappler
R
,
von Schweinitz
D
,
Humphreys
R
,
Fulda
S
. 
RIP1 is required for IAP inhibitor-mediated sensitization for TRAIL-induced apoptosis via a RIP1/FADD/caspase-8 cell death complex
.
Oncogene
2013
;
32
:
3263
73
.
42.
Basit
F
,
Humphreys
R
,
Fulda
S
. 
RIP1 protein-dependent assembly of a cytosolic cell death complex is required for inhibitor of apoptosis (IAP) inhibitor-mediated sensitization to lexatumumab-induced apoptosis
.
J Biol Chem
2012
;
287
:
38767
77
.
43.
Foster
FM
,
Owens
TW
,
Tanianis-Hughes
J
,
Clarke
RB
,
Brennan
K
,
Bundred
NJ
, et al
Targeting inhibitor of apoptosis proteins in combination with ErbB antagonists in breast cancer
.
Breast Cancer Res
2009
;
11
:
R41
.
44.
Weisberg
E
,
Ray
A
,
Barrett
R
,
Nelson
E
,
Christie
AL
,
Porter
D
, et al
Smac mimetics: implications for enhancement of targeted therapies in leukemia
.
Leukemia
2010
;
24
:
2100
9
.
45.
Ziegler
DS
,
Wright
RD
,
Kesari
S
,
Lemieux
ME
,
Tran
MA
,
Jain
M
, et al
Resistance of human glioblastoma multiforme cells to growth factor inhibitors is overcome by blockade of inhibitor of apoptosis proteins
.
J Clin Invest
2008
;
118
:
3109
22
.
46.
Lecis
D
,
Drago
C
,
Manzoni
L
,
Seneci
P
,
Scolastico
C
,
Mastrangelo
E
, et al
Novel SMAC-mimetics synergistically stimulate melanoma cell death in combination with TRAIL and Bortezomib
.
Br J Cancer
2010
;
102
:
1707
16
.
47.
Dougan
M
,
Dougan
S
,
Slisz
J
,
Firestone
B
,
Vanneman
M
,
Draganov
D
, et al
IAP inhibitors enhance co-stimulation to promote tumor immunity
.
J Exp Med
2010
;
207
:
2195
206
.
48.
Galluzzi
L
,
Vitale
I
,
Abrams
JM
,
Alnemri
ES
,
Baehrecke
EH
,
Blagosklonny
MV
, et al
Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012
.
Cell Death Differ
2012
;
19
:
107
20
.
49.
Laukens
B
,
Jennewein
C
,
Schenk
B
,
Vanlangenakker
N
,
Schier
A
,
Cristofanon
S
, et al
Smac mimetic bypasses apoptosis resistance in FADD- or caspase-8-deficient cells by priming for tumor necrosis factor alpha-induced necroptosis
.
Neoplasia
2011
;
13
:
971
9
.
50.
Vanlangenakker
N
,
Vanden Berghe
T
,
Bogaert
P
,
Laukens
B
,
Zobel
K
,
Deshayes
K
, et al
cIAP1 and TAK1 protect cells from TNF-induced necrosis by preventing RIP1/RIP3-dependent reactive oxygen species production
.
Cell Death Differ
2011
;
18
:
656
65
.
51.
Amaravadi
RK
,
Schilder
RJ
,
Dy
GK
,
Ma
WW
,
Fetterly
GJ
,
Weng
DE
, et al
Phase 1 study of the smac mimetic TL32711 in adult subjects with advanced solid tumors & lymphoma to evaluate safety, pharmacokinetics, pharmacodynamics and anti-tumor activity
. In: 
Proceedings of the 2011 Annual Meeting of the American Association for Cancer Research
;
Orlando, FL
; 
2011
(
abstr LB-406
).
52.
Infante
JR
,
Dees
EC
,
Burris
HA
,
Zawel
L
,
Sager
JA
,
Stevenson
C
, et al
A phase I study of LCL-161, an oral inhibitor, in patients with advanced cancer
. In: 
Proceedings of The 2010 Annual Meeting of the American Association for Cancer Research
;
Washington, DC
; 
2010
(
abstr 2775
).
53.
Sikic
BI
,
Eckhardt
SG
,
Gallant
G
,
Burris
HA
,
Camidge
DR
,
Covelas
AD
, et al
Safety, pharmocokinetics (PK), and pharmacodynamics (PD) of HGS1029, an inhibitor of apoptosis protein (IAP), in patients (Pts.) with advanced solid tumors: Results of a Phase I study
.
J Clin Oncol
29
(
suppl
); 
2012
(
abstr 3008
).
54.
Steinhart
L
,
Belz
K
,
Fulda
S
. 
Smac mimetic and demethylating agents synergistically trigger cell death in acute myeloid leukemia cells and overcome apoptosis resistance by inducing necroptosis
.
Cell Death Dis
2013
;
4
:
e802
.