TRAIL and agonistic antibodies raised against TRAIL death receptors are highly promising new anticancer agents. In this brief review, we describe the recent advances in the molecular understanding of TRAIL signaling and the progress made in using TRAIL or agonistic antibodies clinically in mono- and combination therapies. Synergies have been reported in various scenarios of TRAIL-based multidrug treatments, and these can be used to potentiate the efficacy of therapies targeting TRAIL death receptors. We pay particular attention to structure the current knowledge on the diverse molecular mechanisms that are thought to give rise to these synergies and describe how different signaling features evoking synergies can be associated with distinct classes of drugs used in TRAIL-based combination treatments. Mol Cancer Ther; 11(1); 3–13. ©2012 AACR.

Since the discovery of members of the TNF family and their so-called death receptors, the possibility of a new avenue for apoptosis-inducing cancer therapies has emerged. The death ligand–dependent extrinsic apoptotic pathway can induce apoptosis independent of the transcription factor p53 and, therefore, can circumvent the apoptosis resistance acquired by many tumors through loss-of-function mutations in this tumor suppressor. However, TNFα and agonistic antibodies activating the Fas receptor were found to be highly cytotoxic toward primary hepatocytes and other nontransformed cells when applied at clinically relevant concentrations (1, 2). In contrast, soluble TRAIL as well as leucine zipper–trimerized TRAIL potently induce apoptosis in a range of cancer cell lines, can inhibit tumor xenografts in mice, and were found to be of very low systemic toxicity in mice and nonhuman primates (3, 4). These findings sparked tremendous interest in exploring the potential of TRAIL as an anticancer therapeutic for a broad range of human malignant neoplasms. In this review, we briefly describe the key signaling events during TRAIL-induced apoptosis initiation, give a short overview on the status of TRAIL-based clinical research and trials, and describe the mechanisms of signaling processes that evoke synergistic apoptosis responses in TRAIL-based combination treatments.

Endogenous TRAIL is expressed as a 281–amino acid type II trans-membrane protein, which is anchored to the plasma membrane and presented on the cell surface. TRAIL was independently identified by Wiley and colleagues and Pitti and colleagues in 1995 and 1996, respectively, and sequence alignments indicated its close relation to other death ligands, with highest sequence similarities reported for Fas ligand (FasL; refs. 5, 6). TRAIL is expressed by natural killer cells, which, following the establishment of cell–cell contacts, can induce TRAIL-dependent apoptosis in target cells (7). Physiologically, the TRAIL-signaling system was shown to be essential for immune surveillance, for shaping the immune system through regulating T-helper cell 1 versus T-helper cell 2 as well as “helpless” CD8+ T-cell numbers, and for the suppression of spontaneous tumor formation (8–10).

TRAIL induces apoptosis through ligation with its cognate death receptors TRAIL-R1 (also known as DR4) and TRAIL-R2 (DR5; Fig. 1; refs. 11–13), and the trimerization of TRAIL around a central zinc atom via cysteine230 is essential for its apoptotic potential (14, 15). TRAIL binding is followed by receptor trimerization, and groups of trimerized death receptors can further cluster together to form bigger aggregates. These aggregates predominantly accumulate in lipid raft microdomains in the plasma membrane (16). The molecular composition of the receptor trimers has been controversially discussed, and it has not been fully resolved whether, at physiologic conditions, exclusively homotypic interactions or also heterocomplexes can be observed (17–20). Following trimerization of TRAIL-R1 or TRAIL-R2, the adaptor protein Fas-associated death domain containing protein (FADD) can bind to the intracellular death domains of the receptors and promote the recruitment and activation of initiator caspase-8 within the death-inducing signaling complex (DISC; Fig. 1D). In most cells, caspase-8 then initiates apoptosis through Bid cleavage and the mitochondrial apoptosis pathway (Fig. 1E). Both the formation of the caspase-activating DISC, as well as the subsequent signaling network leading to apoptosis execution, can be modulated through a multitude of complex regulatory processes (21, 22).

Figure 1.

Simplified overview of TRAIL-mediated apoptosis signaling. A, trimers of TRAIL can bind to the extracellular regions of TRAIL-R1, TRAIL-R2, TRAIL-R3, and TRAIL-R4. The proapoptotic TRAIL-R1 and TRAIL-R2 comprise cytosolic regions, including a death domain (DD), which is missing in the antiapoptotic TRAIL-R3 and TRAIL-R4. TRAIL-R3 also lacks the transmembrane region. Osteoprotegerin can serve as a soluble decoy receptor. B, antiapoptotic TRAIL-R3 forms homotrimers following TRAIL binding. TRAIL-R4 can also form heterotrimers with TRAIL-R1 and/or TRAIL-R2. These receptor clusters are unable to transduce the TRAIL signal further and, therefore, act as antiapoptotic TRAIL scavengers. C, TRAIL-R1 or TRAIL-R2 can recruit FADD to form the DISC. Procaspase-8 and cFLIP (a catalytically inactive caspase-8 homolog) compete for FADD binding via their death effector domains (DED). Procaspase-8 consists of a DED-containing prodomain, as well as large and small catalytic subunits. Heterodimers of caspase-8 and the long splice variant cFLIP have a very limited substrate repertoire and, in most cases, cannot transduce the apoptosis signal. The short splice variant of cFLIP (not shown) forms inactive heterodimers with caspase-8. D, induced proximity of 2 procaspase-8 zymogens at the DISC results in autocatalytic cleavage of the linkers between the procaspase-8 subunits and caspase-8 activation. Active caspase-8 either resides at the DISC or, once fully processed, can be released into the cytosol. E, active caspase-8 cleaves Bid, inducing the mitochondrial apoptosis pathway, or directly activates effector caspase-3. XIAP suppresses caspase-9 and caspase-3 activity.

Figure 1.

Simplified overview of TRAIL-mediated apoptosis signaling. A, trimers of TRAIL can bind to the extracellular regions of TRAIL-R1, TRAIL-R2, TRAIL-R3, and TRAIL-R4. The proapoptotic TRAIL-R1 and TRAIL-R2 comprise cytosolic regions, including a death domain (DD), which is missing in the antiapoptotic TRAIL-R3 and TRAIL-R4. TRAIL-R3 also lacks the transmembrane region. Osteoprotegerin can serve as a soluble decoy receptor. B, antiapoptotic TRAIL-R3 forms homotrimers following TRAIL binding. TRAIL-R4 can also form heterotrimers with TRAIL-R1 and/or TRAIL-R2. These receptor clusters are unable to transduce the TRAIL signal further and, therefore, act as antiapoptotic TRAIL scavengers. C, TRAIL-R1 or TRAIL-R2 can recruit FADD to form the DISC. Procaspase-8 and cFLIP (a catalytically inactive caspase-8 homolog) compete for FADD binding via their death effector domains (DED). Procaspase-8 consists of a DED-containing prodomain, as well as large and small catalytic subunits. Heterodimers of caspase-8 and the long splice variant cFLIP have a very limited substrate repertoire and, in most cases, cannot transduce the apoptosis signal. The short splice variant of cFLIP (not shown) forms inactive heterodimers with caspase-8. D, induced proximity of 2 procaspase-8 zymogens at the DISC results in autocatalytic cleavage of the linkers between the procaspase-8 subunits and caspase-8 activation. Active caspase-8 either resides at the DISC or, once fully processed, can be released into the cytosol. E, active caspase-8 cleaves Bid, inducing the mitochondrial apoptosis pathway, or directly activates effector caspase-3. XIAP suppresses caspase-9 and caspase-3 activity.

Close modal

Additional TRAIL receptors, which are incapable of transducing the death signal because they lack a functional intracellular death domain, have been identified. These receptors are TRAIL-R3, TRAIL-R4, and osteoprotegerin (22). TRAIL-R3 and TRAIL-R4 are alternatively also referred to as decoy receptors 1 and 2 (DcR1, DcR2). TRAIL-R4 (DcR2) bears huge similarities to TRAIL-R1 and TRAIL-R2; however, it contains only a truncated cytosolic death domain (Fig. 1A; ref. 23). TRAIL-R3 (DcR1) consists of an extracellular cysteine-rich structure that resembles the proapoptotic TRAIL receptors and is associated with the plasma membrane through a COOH-terminal glycosyl-phosphatidylinositol anchor (Fig. 1A; refs. 11, 24). It is still highly debated whether overexpression of TRAIL-R3 or TRAIL-R4 correlates with cellular apoptosis resistance upon TRAIL exposure, and high expression levels are rarely found naturally in isolated cancer cell lines. The exact molecular details of how decoy receptors inhibit TRAIL-induced apoptosis in addition to their obvious role as TRAIL scavengers are not fully resolved and may differ between TRAIL-R3 and TRAIL-R4. For example, it was reported that TRAIL-R3 exclusively forms homotrimers upon TRAIL binding, whereas TRAIL-R4 may also aggregate into heterotrimers with activated TRAIL-R1 and TRAIL-R2 (Fig. 1B; ref. 25). Such heterotrimers are incapable of forming a functional DISC required for apoptosis initiation. Relatively little is known about osteoprotegerin, the fifth receptor capable of TRAIL binding. Osteoprotegerin negatively regulates osteoclastogenesis and is largely secreted as a soluble protein that might act as a scavenger for soluble TRAIL (26). Uncertainties remain about the molecular mechanisms of death and decoy receptor interaction, and it remains to be conclusively shown whether the relative abundance of death versus decoy receptors could serve to indicate cellular susceptibility to TRAIL-induced apoptosis.

TRAIL treatment strategies largely build on soluble variants of recombinant human TRAIL (rhTRAIL, a 60-kDa homotrimer TRAIL protein without its membrane anchor), as well as on monoclonal agonistic antibodies, which specifically target death receptor TRAIL-R1 or TRAIL-R2, thereby potentially circumventing decoy receptor–mediated resistance, and which in addition have the benefit of a significantly longer plasma half-life (27, 28). Additionally, to increase the local concentration and cancer cell–directed cytotoxicity of TRAIL, fusion proteins have been designed to translocate TRAIL to specific tissues or cells, for example, the epidermal growth factor receptor (EGFR)–selective delivery of soluble TRAIL, combined with an EGFR-targeting antibody fragment (29). Soluble TRAIL fused to a melanoma-associated chondroitin sulfate proteoglycan (MCSP)–specific antibody fragment combines inhibition of MCSP tumorigenic signaling with TRAIL-mediated apoptosis induction in melanoma cells (30). A multitude of studies using models of human tumor xenografts in mice showed a high anticancer activity of TRAIL or receptor-specific ligands in vivo, and TRAIL efficacy can be potentiated in combination treatments with kinase and proteasome inhibitors, genotoxic drugs, histone deacetylase inhibitors, and others drugs (20, 31).

It has frequently been debated whether TRAIL can be cytotoxic to untransformed cells in vivo. In contrast to the high tolerability of TRAIL found in the seminal studies of Ashkenazi and colleagues and Walczak and colleagues (3, 4), other studies reported toxicities of TRAIL in isolated human hepatocytes (32, 33) and human brain tissue (34). However, significant TRAIL hepatotoxicities seem to be restricted to ex vivo cultures of primary liver cells or to highly aggregated TRAIL variants carrying flag or polyhistidine tags (35, 36). In contrast, rhTRAIL, as well as leucine or isoleucine zipper variants of TRAIL, was shown to be well tolerated in animal models. This finding is in stark contrast to the generally high hepatotoxicities that were reported for death ligands FasL and TNFα (3, 4). As a note of caution, however, it was shown that liver steatosis or hepatitis C infection can increase human hepatocyte sensitivity toward TRAIL (37), suggesting that patients from these risk groups should be excluded from TRAIL-based therapies.

Soluble rhTRAIL (also named dulanermin), the TRAIL-R1–targeting monoclonal agonistic antibody mapatumumab, and the TRAIL-R2–targeting monoclonal agonistic antibodies lexatumumab, conatumumab, tigatuzumab, and DAB4 (also named PRO95780) have entered clinical studies. A large number of phase I and phase II clinical trials have been undertaken with these agents by now or are still ongoing, either as monotherapy or in combination with other chemotherapeutic drugs. These trials target a wide range of both solid and nonsolid malignant neoplasms.

Toxicity studies as part of an rhTRAIL phase I trial provided encouraging results. In a dose-escalating monotherapy in patients with advanced cancers, rhTRAIL was well tolerated at serum concentrations that were shown to be effective against cancer cells in preclinical models (38). Similarly, TRAIL-R1–specific mapatumumab seems to be well tolerated, and in a phase II study in patients with relapsed or refractory non-Hodgkin lymphoma, approximately one third of all patients responded to mapatumumab (28, 39, 40). In a phase II clinical trial, mapatumumab was well tolerated in patients with colorectal cancer (41). Lexatumumab, conatumumab, tigatuzumab, PRO95780, and other TRAIL-R2–targeting antibodies, likewise, were investigated in phase I and II clinical trials (42–44). Like mapatumumab, these antibodies only caused low-to-moderate side effects in a small number of patients. Several patients showed partial responses or attenuated disease progression when treated with TRAIL-R2–targeting antibodies (45, 46). In general, TRAIL and TRAIL receptor–binding antibodies, therefore, seem to be well tolerated, and only a few cases of adverse responses have been documented. Furthermore, promising results in patients with advanced or refractory cancers underline the therapeutic potential of TRAIL receptor–targeting molecules. However, as a caveat, signaling through TRAIL death receptors may also induce antiapoptotic responses and proliferation signaling. This alternative response was shown to require the activation of kinases, such as RIP1, IKK, c-jun-NH2-kinase, or p38 in signaling complexes secondary to the DISC (47) and to induce transcription-dependent prosurvival responses that attenuate apoptosis signaling. Indeed, subpopulations of primary cells isolated from children with acute leukemia showed increased proliferation rates following TRAIL treatment (48). These prosurvival responses may, therefore, limit the usability of TRAIL in monotherapies.

Cancers like melanoma, leukemia, multiple myeloma, breast, bladder, prostate, renal, colon, and others would be expected to be largely TRAIL sensitive judging on the basis of preclinical drug response data. However, cases of TRAIL resistance are frequently being observed and often could be associated with the overexpression of antiapoptotic proteins or the low expression of TRAIL receptors. An attractive and preclinically successful strategy, therefore, is to identify combination treatments that sensitize otherwise resistant cancers to TRAIL. Although this strategy holds risks because of the potential sensitization of nontransformed cells to the treatment, initial results are very promising. In a phase Ib clinical study to determine safety, pharmacokinetics, and maximum dose tolerance levels, patients with non–small cell lung cancer were treated with rhTRAIL (dulanermin), combined with paclitaxel, carboplatin, and bevacizumab. No dose-limiting toxicity was observed, and the treatment was well tolerated, with 1 complete response and a high percentage of partial responses observed (49). Other early phase Ib trials assessing combination treatments with mapatumumab and paclitaxel or carboplatin, as well as mapatumumab together with gemcitabine or cisplatin, in patients with advanced solid tumors reported high tolerability of mapatumumab (50, 51). By now, many more additional phase I and II clinical trials have been conducted or are still ongoing in other cancer types and using additional U.S. Food and Drug Administration–approved drugs, such as kinase inhibitors, proteasome inhibitors, histone deacetylase inhibitors, or genotoxic drugs, in combination with TRAIL (Table 1; ref. 45). Even though cases of adverse effects and toxicities were reported, overall, TRAIL combination therapies seem to be remarkably well tolerated. Still, outstanding results from ongoing phase II clinical trials will soon allow assessment of whether patients may benefit from such alternative treatment regimes and whether patient responses are indicative of multidrug synergies that were reported in vitro. The mechanisms bringing about synergistic multidrug responses are very complex. However, significant progress in understanding the underlying signaling cross-talk has been made in recent years, as outlined in the following discussion.

Table 1.

Modulation of TRAIL-mediated apoptosis through combination treatments: clinical trials

Drugs in clinical trials with TRAIL-R ligandTRAIL-R ligandAdditional combinationType of cancerClinical trial phaseReferenceMode of action and/or targetConsequence and/or modulation of the TRAIL pathway
Proteasome inhibitor 
Bortezomib Conatumumab — Lymphoma Ib, susp.a NCT00791011 Inhibition ofproteasomal degradation Stabilization of TRAIL-R, active caspase-8, -3, BH3-only proteins, p53, IkBa. BH3-only protein and p53 expression 
 Mapatumumab — Multiple myeloma II, compl. NCT00315757   
Genotoxic and/or mitosis stress-inducing drugs 
5-FU Conatumumab FOLFOX6, bevacizumab CRC Ib, II NCT00625651 Nucleosidereplacement by analogs p53-mediated transcriptionalupregulation of BH3-only proteins and TRAIL-R2; p53-mediated downregulation of antiapoptotic Bcl-2 family proteins and cFlip 
Capecitabine (5-FU prodrug) Conatumumab Gemcitabine, radiation Pancreatic I, II NCT01017822   
Gemcitabine Conatumumab — Pancreatic Ib, II NCT00630552   
 Mapatumumab Cisplatin Solid tumors I, compl. (60)   
Cisplatin Mapatumumab Radiotherapy Cervical cancer II NCT01088347 DNA alkylation  
Carboplatin Mapatumumab Paclitaxel NSCLC II, compl. NCT00583830   
Doxorubicin Conatumumab — Soft tissue sarcoma II NCT00626704 DNAintercalation  
Paclitaxel Dulanermin Carboplatin, bevacizumab NSCLC Ib, compl. (58) NCT00508625 Tubulindisassembly inhibition Prolonged activation of themitotic checkpoint, p53 signaling 
 Conatumumab Carboplatin NSCLC Ib, II, compl. NCT00534027   
 Mapatumumab Carboplatin Solid tumors I, compl. (59)   
 Mapatumumab Carboplatin NSCLC II, compl. NCT00583830   
 Tigatuzumab — Breast cancer II NCT01307891   
Irinotecan (CPT-11, Camtothecin-11) Dulanermin Cetuximab CRC Ib NCT00671372 Topoisomerase Iinhibition  
 PRO95780 Cetuximab CRC Ib, compl. NCT00497497   
Epigenetic inhibitors 
Vorinostat Conatumumab — Lymphoma Ib, susp.a NCT00791011 Histonedeacetylase inhibition CDK-inhibitor 1 (p21) activation, cell cycle arrest, altered activity of NF-kB, p53 
Irradiation 
Radiotherapy Mapatumumab Cisplatin Cervical cancer II NCT01088347 Alteration of gene transcription, radical generation Induction of BH3-only proteins, ROS production 
3D conformal radiation therapy Conatumumab Gemcitabine, capecitabine Pancreatic I, II NCT01017822   
Growth factor receptor and/or receptor tyrosine kinase inhibitors 
Cetuximab Dulanermin Irinotecan CRC Ib NCT00671372 Inhibition of theepidermal. growth factor receptor Inhibition of PI3K/Aktprosurvival and proliferation signaling 
Panitumumab (mAB) Conatumumab — CRC Ib, II NCT00630786   
Bevacizumab (mAB) Dulanermin Carboplatin, paclitaxel NSCLC II NCT00508625 Inhibition ofvascular endothelial growth factor receptor tyrosine kinases Inhibition of PI3K/Akt signaling: decrease of cFlip and XIAP expression, decrease of NF-kB activation, dephosphorylation of the BH3-only protein Bad and its release from 14-3-3 sequestration 
 Dulanermin FOLFOX CRC NCT00873756   
 Dulanermin FOLFIRI CRC NCT00671372   
 Conatumumab FOLFOX6 CRC Ib, II NCT00625651   
 PRO95780 Carboplatin, paclitaxel NSCLC II, compl. NCT00480831   
 PRO95780 FOLFIRI CRC Ib, compl. NCT00497497   
 PRO95780 FOLFOX CRC Ib, compl. NCT00851136   
Ganitumab (mAB) Conatumumab — NSCLC, CRC, pancreatic, ovarian, sarcoma Ib, II NCT00819169 Inhibition of theinsulin-like growth factor I receptor Downregulation of levels ofactive Akt, ERK1/2, and MAPK p38 
 Conatumumab FOLFIRI KRAS-mutant CRC II NCT00813605   
 Conatumumab FOLFOX6,bevacizumab Solid tumors, CRC, lymphoma, NSCLC II NCT01327612   
Kinase inhibitors 
Sorafenib Mapatumumab — Hepatocellular carcinoma Ib NCT00712855 RAF/MEK/ERKpathway inhibition. Vascular endothelial/platelet-derived growth factor receptor tyrosine kinases inhibition in vasculature. Downregulation of phospho-STAT3, Mcl-1, survivin, and cyclin D1. Downregulation of Mcl-1, cIAP2, cFlip. Inhibition of NF-kB activation and tumor vascularization. 
 Mapatumumab — Hepatocellular carcinoma II NCT01258608   
Cytokines 
IFN-γ Lexatumumab — Pediatric solid tumors NCT00428272 Alteration ofgene transcription Transcriptional downregulation of cFlip 
CD20 antibody 
Rituximab Dulanermin — Non-Hodgkin lymphoma Ib/II, compl. NCT00400764 Inhibition of the B-cell surface protein CD20 Inhibition of NF-kB, downregulation of Bcl-XL, ADCC and CDC induction 
 PRO95780 — Non-Hodgkin lymphoma II, compl. NCT00517049   
Drugs in clinical trials with TRAIL-R ligandTRAIL-R ligandAdditional combinationType of cancerClinical trial phaseReferenceMode of action and/or targetConsequence and/or modulation of the TRAIL pathway
Proteasome inhibitor 
Bortezomib Conatumumab — Lymphoma Ib, susp.a NCT00791011 Inhibition ofproteasomal degradation Stabilization of TRAIL-R, active caspase-8, -3, BH3-only proteins, p53, IkBa. BH3-only protein and p53 expression 
 Mapatumumab — Multiple myeloma II, compl. NCT00315757   
Genotoxic and/or mitosis stress-inducing drugs 
5-FU Conatumumab FOLFOX6, bevacizumab CRC Ib, II NCT00625651 Nucleosidereplacement by analogs p53-mediated transcriptionalupregulation of BH3-only proteins and TRAIL-R2; p53-mediated downregulation of antiapoptotic Bcl-2 family proteins and cFlip 
Capecitabine (5-FU prodrug) Conatumumab Gemcitabine, radiation Pancreatic I, II NCT01017822   
Gemcitabine Conatumumab — Pancreatic Ib, II NCT00630552   
 Mapatumumab Cisplatin Solid tumors I, compl. (60)   
Cisplatin Mapatumumab Radiotherapy Cervical cancer II NCT01088347 DNA alkylation  
Carboplatin Mapatumumab Paclitaxel NSCLC II, compl. NCT00583830   
Doxorubicin Conatumumab — Soft tissue sarcoma II NCT00626704 DNAintercalation  
Paclitaxel Dulanermin Carboplatin, bevacizumab NSCLC Ib, compl. (58) NCT00508625 Tubulindisassembly inhibition Prolonged activation of themitotic checkpoint, p53 signaling 
 Conatumumab Carboplatin NSCLC Ib, II, compl. NCT00534027   
 Mapatumumab Carboplatin Solid tumors I, compl. (59)   
 Mapatumumab Carboplatin NSCLC II, compl. NCT00583830   
 Tigatuzumab — Breast cancer II NCT01307891   
Irinotecan (CPT-11, Camtothecin-11) Dulanermin Cetuximab CRC Ib NCT00671372 Topoisomerase Iinhibition  
 PRO95780 Cetuximab CRC Ib, compl. NCT00497497   
Epigenetic inhibitors 
Vorinostat Conatumumab — Lymphoma Ib, susp.a NCT00791011 Histonedeacetylase inhibition CDK-inhibitor 1 (p21) activation, cell cycle arrest, altered activity of NF-kB, p53 
Irradiation 
Radiotherapy Mapatumumab Cisplatin Cervical cancer II NCT01088347 Alteration of gene transcription, radical generation Induction of BH3-only proteins, ROS production 
3D conformal radiation therapy Conatumumab Gemcitabine, capecitabine Pancreatic I, II NCT01017822   
Growth factor receptor and/or receptor tyrosine kinase inhibitors 
Cetuximab Dulanermin Irinotecan CRC Ib NCT00671372 Inhibition of theepidermal. growth factor receptor Inhibition of PI3K/Aktprosurvival and proliferation signaling 
Panitumumab (mAB) Conatumumab — CRC Ib, II NCT00630786   
Bevacizumab (mAB) Dulanermin Carboplatin, paclitaxel NSCLC II NCT00508625 Inhibition ofvascular endothelial growth factor receptor tyrosine kinases Inhibition of PI3K/Akt signaling: decrease of cFlip and XIAP expression, decrease of NF-kB activation, dephosphorylation of the BH3-only protein Bad and its release from 14-3-3 sequestration 
 Dulanermin FOLFOX CRC NCT00873756   
 Dulanermin FOLFIRI CRC NCT00671372   
 Conatumumab FOLFOX6 CRC Ib, II NCT00625651   
 PRO95780 Carboplatin, paclitaxel NSCLC II, compl. NCT00480831   
 PRO95780 FOLFIRI CRC Ib, compl. NCT00497497   
 PRO95780 FOLFOX CRC Ib, compl. NCT00851136   
Ganitumab (mAB) Conatumumab — NSCLC, CRC, pancreatic, ovarian, sarcoma Ib, II NCT00819169 Inhibition of theinsulin-like growth factor I receptor Downregulation of levels ofactive Akt, ERK1/2, and MAPK p38 
 Conatumumab FOLFIRI KRAS-mutant CRC II NCT00813605   
 Conatumumab FOLFOX6,bevacizumab Solid tumors, CRC, lymphoma, NSCLC II NCT01327612   
Kinase inhibitors 
Sorafenib Mapatumumab — Hepatocellular carcinoma Ib NCT00712855 RAF/MEK/ERKpathway inhibition. Vascular endothelial/platelet-derived growth factor receptor tyrosine kinases inhibition in vasculature. Downregulation of phospho-STAT3, Mcl-1, survivin, and cyclin D1. Downregulation of Mcl-1, cIAP2, cFlip. Inhibition of NF-kB activation and tumor vascularization. 
 Mapatumumab — Hepatocellular carcinoma II NCT01258608   
Cytokines 
IFN-γ Lexatumumab — Pediatric solid tumors NCT00428272 Alteration ofgene transcription Transcriptional downregulation of cFlip 
CD20 antibody 
Rituximab Dulanermin — Non-Hodgkin lymphoma Ib/II, compl. NCT00400764 Inhibition of the B-cell surface protein CD20 Inhibition of NF-kB, downregulation of Bcl-XL, ADCC and CDC induction 
 PRO95780 — Non-Hodgkin lymphoma II, compl. NCT00517049   

NOTE: Drugs listed are currently being investigated in clinical trials in combination with rhTRAIL or TRAIL-receptor ligands. Dulanermin is recombinant human Apo2L/TRAIL; mapatumumab (HGS1012) is a monoclonal antibody directed against TRAIL-R1; lexatumumab (HGS-ETR2), conatumumab, tigatuzumab, and PRO95780 (apomab) are monoclonal antibodies directed against TRAIL-R2. FOLFOX and FOLFIRI are combination chemotherapies including folinic acid and 5-fluorouracil, plus oxaliplatin or irinotecan, respectively.

Abbreviations: ADCC, antibody-dependent cell-mediated cytotoxicity; CDC, complement-dependent cytotoxicity; compl., completed; CRC, colorectal cancer; ERK, extracellular signal-regulated kinase; 5-FU, 5-fluorouracil; mAB, monoclonal antibody; MEK, MAP/ERK kinase; NSCLC, non–small cell lung cancer; ROS, reactive oxygen species; susp., suspended.

aThe clinical trial NCT00791011 was suspended because of a lack of mantle cell lymphoma subjects who were bortezomib naïve.

Trial information was sourced from http://www.clinicaltrials.gov and additional trial-specific information can be accessed online using the respective reference numbers.

Synergistic effects of chemotherapeutics in combination with TRAIL have been reported for various classes of drugs. In general, synergies arise from nonlinear cross-potentiation of drug effects. In the framework of apoptosis signaling, such potentiation may arise from the (often p53-dependent) transcriptional upregulation of proapoptotic proteins, from their accumulation due to impaired protein degradation, from alterations in regulatory posttranslational protein modifications, or from combinations of these processes. In the following paragraphs, we discuss the synergy mechanisms exerted by different drug classes. Related to this discussion, an overview of clinical trials aiming to use drug synergies in TRAIL-based combination treatments is provided in Table 1. Information on additional novel and promising cotreatment strategies that are still at the preclinical stage is provided in Supplementary Table S1.

Different genotoxic drugs have been suggested to enhance TRAIL signaling and its anticancer potential, with 5-fluorouracil and cisplatin having been studied most intensely. A typical stress response to genotoxic monotherapies is the transcriptional induction of BH3-only proteins, such as Puma, which is a potent inducer of apoptosis through the mitochondrial pathway (52). Such responses may synergistically complement the BH3-only protein Bid, which is cleaved and activated by caspase-8 in response to TRAIL. In this respect, similar synergy mechanisms would be relevant in cotreatment scenarios based on synthetic BH3-only protein mimetics, such as AT-101 (a gossypol derivate), ABT-263, ABT-737, and GX-15-070 (obatoclax; ref. 53). These novel drugs are already undergoing evaluation in monotherapy trials and may hold great potential for future combination treatments. However, synergies achieved with genotoxic drugs may also arise from upstream modulation of TRAIL signaling. For example, 5-fluorouracil can prime TRAIL-resistant hepatocellular carcinoma cells to apoptosis, at least partially, through the downregulation of the inactive caspase-8 homolog cFlip (Fig. 1C) and through the parallel upregulation of TRAIL-R2 at physiologically relevant doses (54). Importantly, cytotoxicity to normal cells seems to be limited to acceptable levels in this scenario (54). Similar scenarios of death receptor upregulation or cFlip downregulation were also reported in response to other genotoxic drugs, as well as to ionizing irradiation and epigenetic inhibitors, such as histone deacetylase inhibitors (55, 56). Also IFNs, which are frequently used in hematologic cancer therapy, were shown to potently promote TRAIL-induced apoptosis through evoking transcriptional responses. For example, IFN-α exposure can increase TRAIL and TRAIL-R2 expression in human hepatocellular carcinoma cells (57, 58), whereas IFN-γ can reduce cFlip protein levels (59).

Prolonged proteasome inhibition induces the expression and accumulation of BH3-only proteins, such as Bim, Bik, Puma, and Noxa (60), which leads to synergy mechanisms like those described above for genotoxic drugs. Furthermore, TRAIL-induced NF-κB activation and prosurvival signaling can be limited by proteasome inhibition because the degradation of NF-κB inhibitor IκBα is prevented (23, 61). Maybe more importantly, proteasome inhibition can lead to the direct accumulation of proteins that promote TRAIL-induced apoptosis. An accumulation of both TRAIL-R1 and TRAIL-R2 was described in response to proteasome inhibition in various cancers (62–64), potentially resulting in more efficient DISC formation and caspase activation upon TRAIL addition. The consequences arising from a general inhibition of protein degradation are naturally complex, due to the global disturbance of relative protein abundances in addition to active transcriptional stress responses. Attributing synergies in TRAIL and/or proteasome inhibitor cotreatments to the modulation of single proteins or processes, as proposed in several studies, therefore likely presents an oversimplification of the underlying molecular mechanisms that enhance TRAIL responsiveness. Nevertheless, the fact that proteasome inhibition can promote proapoptotic signaling, at least partially, independent of protein neosynthesis makes proteasome inhibitors particularly attractive for the treatment of cancers that present with deficiencies in transcriptional proapoptotic responses, as can arise from loss-of-function mutations in tumor suppressor p53.

Transcription- or p53-independent synergisms of chemotherapeutics and TRAIL can also be achieved by targeting posttranslational protein modifications such as phosphorylation patterns. For example, inhibiting protein kinase CK2 impairs Bid phosphorylation and enhances Bid cleavage by caspase-8 (65, 66) and may be an attractive cotreatment strategy if outstanding safety tests yield satisfying results. Procaspase-8 itself can be phosphorylated by Src kinase, which impairs its binding to and activation at the DISC (67). In addition, formation of the DISC can be modulated upstream of caspase-8 recruitment through mitogen-activated protein kinases (MAPK; ref. 68). Furthermore, protein kinase C activity limits FADD recruitment into the DISC (69). Kinase inhibition can also directly or indirectly induce transcriptional responses, adding further complexity. For example, inhibiting phosphoinositide 3-kinase (PI3K)/AKT signaling by perifosine can restore TRAIL sensitivity in acute myelogenous leukemia through a p53-independent TRAIL-R2 upregulation and a concomitant cFlip and X-linked inhibitor-of-apoptosis protein (XIAP) downregulation (70). Multitarget kinase inhibitors, such as sorafenib, may therefore hold great potential in enhancing cancer cell responsiveness and are currently being investigated in combination treatments with TRAIL in phase I clinical trials (45). Furthermore, additional posttranslational modifications of the death receptors, such as palmitoylation, nitrosylation, and glycosylation, have been reported, and these modifications can influence cellular sensitivity toward TRAIL (22). Whether these modifications can be therapeutically targeted and used is currently unknown.

Antagonists of inhibitor-of-apoptosis proteins, such as synthetic Smac peptides and Smac mimetics, were shown to sensitize various cancer cell lines to TRAIL-induced apoptosis. In particular, antagonizing XIAP, the most potent inhibitor of executioner caspases, promotes the direct activation of caspase-3 by caspase-8 and results in efficient apoptosis execution (71, 72). Furthermore, Smac mimetics also bind to cellular IAP (cIAP)–1 and cIAP-2. As a consequence, cIAPs are rapidly degraded and cells may respond with secreting TNFα (73, 74). TNFα then may further enhance apoptosis by activating a parallel extrinsic apoptosis pathway that leads to caspase-8 activation. IAP inhibitors have been shown to synergistically kill pancreatic cancer cells in combination with the TRAIL-R1–targeting antibody mapatumumab, whereas the combined treatment with the TRAIL-R2–specific antibody lexatumumab was less efficient, indicating receptor-specific differences in synergy and susceptibility of certain cancers (75). Besides the requirement to better understand and use synergy mechanisms, means to predetermine differential sensitivity to either TRAIL-R1–or TRAIL-R2–transduced apoptosis will be paramount to case specifically identify the most effective treatment strategies in the future.

Experimental preclinical as well as clinical evidence shows that TRAIL has a high potential as a novel anticancer drug, both in monotherapies and in combination treatments (Table 1 and Supplementary Table S1). Given the comprehensive research activities currently focusing on identifying and deciphering the complex intracellular signaling cross-talk that evokes synergies in TRAIL combination treatments, we will soon better understand which conditions need to be fulfilled to more efficiently use the potential of different TRAIL-based treatment strategies. At this stage, TRAIL signaling is already one of the best-characterized apoptotic signaling pathways. In the coming years, this information may greatly assist in identifying potential biomarkers that will allow researchers to rationally predict tumor responsiveness to TRAIL-based therapies. This important, but still outstanding milestone, will need to be achieved before patient benefits can be maximized through appropriately tailored drug administration.

No potential conflicts of interest were disclosed.

This work was supported by grants from Science Foundation Ireland (05/RFP/BIM056), the Health Research Board Ireland (RP/2006/258; RP/2008/7), and the Irish National Biophotonics and Imaging Platform funded under the Irish Higher Education Authority Programme for Third Level Institutions (HEA PRTLI) Cycle 4, cofunded by the Irish Government and the European Union “Investing in your future.”

1.
Costelli
P
,
Aoki
P
,
Zingaro
B
,
Carbó
N
,
Reffo
P
,
Lopez-Soriano
FJ
, et al
Mice lacking TNFalpha receptors 1 and 2 are resistant to death and fulminant liver injury induced by agonistic anti-Fas antibody
.
Cell Death Differ
2003
;
10
:
997
1004
.
2.
Ogasawara
J
,
Watanabe-Fukunaga
R
,
Adachi
M
,
Matsuzawa
A
,
Kasugai
T
,
Kitamura
Y
, et al
Lethal effect of the anti-Fas antibody in mice
.
Nature
1993
;
364
:
806
9
.
3.
Walczak
H
,
Miller
RE
,
Ariail
K
,
Gliniak
B
,
Griffith
TS
,
Kubin
M
, et al
Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo
.
Nat Med
1999
;
5
:
157
63
.
4.
Ashkenazi
A
,
Pai
RC
,
Fong
S
,
Leung
S
,
Lawrence
DA
,
Marsters
SA
, et al
Safety and antitumor activity of recombinant soluble Apo2 ligand
.
J Clin Invest
1999
;
104
:
155
62
.
5.
Wiley
SR
,
Schooley
K
,
Smolak
PJ
,
Din
WS
,
Huang
CP
,
Nicholl
JK
, et al
Identification and characterization of a new member of the TNF family that induces apoptosis
.
Immunity
1995
;
3
:
673
82
.
6.
Pitti
RM
,
Marsters
SA
,
Ruppert
S
,
Donahue
CJ
,
Moore
A
,
Ashkenazi
A
. 
Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family
.
J Biol Chem
1996
;
271
:
12687
90
.
7.
Smyth
MJ
,
Cretney
E
,
Takeda
K
,
Wiltrout
RH
,
Sedger
LM
,
Kayagaki
N
, et al
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) contributes to interferon gamma-dependent natural killer cell protection from tumor metastasis
.
J Exp Med
2001
;
193
:
661
70
.
8.
Cretney
E
,
Takeda
K
,
Yagita
H
,
Glaccum
M
,
Peschon
JJ
,
Smyth
MJ
. 
Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-inducing ligand-deficient mice
.
J Immunol
2002
;
168
:
1356
61
.
9.
Janssen
EM
,
Droin
NM
,
Lemmens
EE
,
Pinkoski
MJ
,
Bensinger
SJ
,
Ehst
BD
, et al
CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death
.
Nature
2005
;
434
:
88
93
.
10.
Zhang
XR
,
Zhang
LY
,
Devadas
S
,
Li
L
,
Keegan
AD
,
Shi
YF
. 
Reciprocal expression of TRAIL and CD95L in Th1 and Th2 cells: role of apoptosis in T helper subset differentiation
.
Cell Death Differ
2003
;
10
:
203
10
.
11.
MacFarlane
M
,
Ahmad
M
,
Srinivasula
SM
,
Fernandes-Alnemri
T
,
Cohen
GM
,
Alnemri
ES
. 
Identification and molecular cloning of two novel receptors for the cytotoxic ligand TRAIL
.
J Biol Chem
1997
;
272
:
25417
20
.
12.
Pan
G
,
O'Rourke
K
,
Chinnaiyan
AM
,
Gentz
R
,
Ebner
R
,
Ni
J
, et al
The receptor for the cytotoxic ligand TRAIL
.
Science
1997
;
276
:
111
3
.
13.
Walczak
H
,
Degli-Esposti
MA
,
Johnson
RS
,
Smolak
PJ
,
Waugh
JY
,
Boiani
N
, et al
TRAIL-R2: a novel apoptosis-mediating receptor for TRAIL
.
EMBO J
1997
;
16
:
5386
97
.
14.
Bodmer
JL
,
Meier
P
,
Tschopp
J
,
Schneider
P
. 
Cysteine 230 is essential for the structure and activity of the cytotoxic ligand TRAIL
.
J Biol Chem
2000
;
275
:
20632
7
.
15.
Hymowitz
SG
,
O'Connell
MP
,
Ultsch
MH
,
Hurst
A
,
Totpal
K
,
Ashkenazi
A
, et al
A unique zinc-binding site revealed by a high-resolution X-ray structure of homotrimeric Apo2L/TRAIL
.
Biochemistry
2000
;
39
:
633
40
.
16.
Song
JH
,
Tse
MC
,
Bellail
A
,
Phuphanich
S
,
Khuri
F
,
Kneteman
NM
, et al
Lipid rafts and nonrafts mediate tumor necrosis factor related apoptosis-inducing ligand induced apoptotic and nonapoptotic signals in non small cell lung carcinoma cells
.
Cancer Res
2007
;
67
:
6946
55
.
17.
Bin
L
,
Thorburn
J
,
Thomas
LR
,
Clark
PE
,
Humphreys
R
,
Thorburn
A
. 
Tumor-derived mutations in the TRAIL receptor DR5 inhibit TRAIL signaling through the DR4 receptor by competing for ligand binding
.
J Biol Chem
2007
;
282
:
28189
94
.
18.
Kischkel
FC
,
Lawrence
DA
,
Chuntharapai
A
,
Schow
P
,
Kim
KJ
,
Ashkenazi
A
. 
Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5
.
Immunity
2000
;
12
:
611
20
.
19.
Tur
V
,
van der Sloot
AM
,
Reis
CR
,
Szegezdi
E
,
Cool
RH
,
Samali
A
, et al
DR4-selective tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) variants obtained by structure-based design
.
J Biol Chem
2008
;
283
:
20560
8
.
20.
van der Sloot
AM
,
Tur
V
,
Szegezdi
E
,
Mullally
MM
,
Cool
RH
,
Samali
A
, et al
Designed tumor necrosis factor-related apoptosis-inducing ligand variants initiating apoptosis exclusively via the DR5 receptor
.
Proc Natl Acad Sci U S A
2006
;
103
:
8634
9
.
21.
Hellwig
CT
,
Passante
E
,
Rehm
M
. 
The molecular machinery regulating apoptosis signal transduction and its implication in human physiology and pathophysiologies
.
Curr Mol Med
2011
;
11
:
31
47
.
22.
Pennarun
B
,
Meijer
A
,
de Vries
EG
,
Kleibeuker
JH
,
Kruyt
F
,
de Jong
S
. 
Playing the DISC: turning on TRAIL death receptor-mediated apoptosis in cancer
.
Biochim Biophys Acta
2010
;
1805
:
123
40
.
23.
Degli-Esposti
MA
,
Dougall
WC
,
Smolak
PJ
,
Waugh
JY
,
Smith
CA
,
Goodwin
RG
. 
The novel receptor TRAIL-R4 induces NF-kappaB and protects against TRAIL-mediated apoptosis, yet retains an incomplete death domain
.
Immunity
1997
;
7
:
813
20
.
24.
Sheridan
JP
,
Marsters
SA
,
Pitti
RM
,
Gurney
A
,
Skubatch
M
,
Baldwin
D
, et al
Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors
.
Science
1997
;
277
:
818
21
.
25.
Mérino
D
,
Lalaoui
N
,
Morizot
A
,
Schneider
P
,
Solary
E
,
Micheau
O
. 
Differential inhibition of TRAIL-mediated DR5-DISC formation by decoy receptors 1 and 2
.
Mol Cell Biol
2006
;
26
:
7046
55
.
26.
Emery
JG
,
McDonnell
P
,
Burke
MB
,
Deen
KC
,
Lyn
S
,
Silverman
C
, et al
Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL
.
J Biol Chem
1998
;
273
:
14363
7
.
27.
Kelley
SK
,
Harris
LA
,
Xie
D
,
Deforge
L
,
Totpal
K
,
Bussiere
J
, et al
Preclinical studies to predict the disposition of Apo2L/tumor necrosis factor-related apoptosis-inducing ligand in humans: characterization of in vivo efficacy, pharmacokinetics, and safety
.
J Pharmacol Exp Ther
2001
;
299
:
31
8
.
28.
Tolcher
AW
,
Mita
M
,
Meropol
NJ
,
von Mehren
M
,
Patnaik
A
,
Padavic
K
, et al
Phase I pharmacokinetic and biologic correlative study of mapatumumab, a fully human monoclonal antibody with agonist activity to tumor necrosis factor-related apoptosis-inducing ligand receptor-1
.
J Clin Oncol
2007
;
25
:
1390
5
.
29.
Bremer
E
,
de Bruyn
M
,
Samplonius
DF
,
Bijma
T
,
ten Cate
B
,
de Leij
LF
, et al
Targeted delivery of a designed sTRAIL mutant results in superior apoptotic activity towards EGFR-positive tumor cells
.
J Mol Med (Berl)
2008
;
86
:
909
24
.
30.
de Bruyn
M
,
Rybczynska
AA
,
Wei
Y
,
Schwenkert
M
,
Fey
GH
,
Dierckx
RA
, et al
Melanoma-associated Chondroitin Sulfate Proteoglycan (MCSP)-targeted delivery of soluble TRAIL potently inhibits melanoma outgrowth in vitro and in vivo
.
Mol Cancer
2010
;
9
:
301
.
31.
Johnstone
RW
,
Frew
AJ
,
Smyth
MJ
. 
The TRAIL apoptotic pathway in cancer onset, progression and therapy
.
Nat Rev Cancer
2008
;
8
:
782
98
.
32.
Jo
M
,
Kim
TH
,
Seol
DW
,
Esplen
JE
,
Dorko
K
,
Billiar
TR
, et al
Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis-inducing ligand
.
Nat Med
2000
;
6
:
564
7
.
33.
Mori
E
,
Thomas
M
,
Motoki
K
,
Nakazawa
K
,
Tahara
T
,
Tomizuka
K
, et al
Human normal hepatocytes are susceptible to apoptosis signal mediated by both TRAIL-R1 and TRAIL-R2
.
Cell Death Differ
2004
;
11
:
203
7
.
34.
Nitsch
R
,
Bechmann
I
,
Deisz
RA
,
Haas
D
,
Lehmann
TN
,
Wendling
U
, et al
Human brain-cell death induced by tumour-necrosis-factor-related apoptosis-inducing ligand (TRAIL)
.
Lancet
2000
;
356
:
827
8
.
35.
Lawrence
D
,
Shahrokh
Z
,
Marsters
S
,
Achilles
K
,
Shih
D
,
Mounho
B
, et al
Differential hepatocyte toxicity of recombinant Apo2L/TRAIL versions
.
Nat Med
2001
;
7
:
383
5
.
36.
Qin
J
,
Chaturvedi
V
,
Bonish
B
,
Nickoloff
BJ
. 
Avoiding premature apoptosis of normal epidermal cells
.
Nat Med
2001
;
7
:
385
6
.
37.
Volkmann
X
,
Fischer
U
,
Bahr
MJ
,
Ott
M
,
Lehner
F
,
Macfarlane
M
, et al
Increased hepatotoxicity of tumor necrosis factor-related apoptosis-inducing ligand in diseased human liver
.
Hepatology
2007
;
46
:
1498
508
.
38.
Herbst
RS
,
Eckhardt
SG
,
Kurzrock
R
,
Ebbinghaus
S
,
O'Dwyer
PJ
,
Gordon
MS
, et al
Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cancer
.
J Clin Oncol
2010
;
28
:
2839
46
.
39.
Hotte
SJ
,
Hirte
HW
,
Chen
EX
,
Siu
LL
,
Le
LH
,
Corey
A
, et al
A phase 1 study of mapatumumab (fully human monoclonal antibody to TRAIL-R1) in patients with advanced solid malignancies
.
Clin Cancer Res
2008
;
14
:
3450
5
.
40.
Younes
A
,
Vose
JM
,
Zelenetz
AD
,
Smith
MR
,
Burris
HA
,
Ansell
SM
, et al
A Phase 1b/2 trial of mapatumumab in patients with relapsed/refractory non-Hodgkin's lymphoma
.
Br J Cancer
2010
;
103
:
1783
7
.
41.
Trarbach
T
,
Moehler
M
,
Heinemann
V
,
Köhne
CH
,
Przyborek
M
,
Schulz
C
, et al
Phase II trial of mapatumumab, a fully human agonistic monoclonal antibody that targets and activates the tumour necrosis factor apoptosis-inducing ligand receptor-1 (TRAIL-R1), in patients with refractory colorectal cancer
.
Br J Cancer
2010
;
102
:
506
12
.
42.
Rosevear
HM
,
Lightfoot
AJ
,
Griffith
TS
. 
Conatumumab, a fully human mAb against death receptor 5 for the treatment of cancer
.
Curr Opin Investig Drugs
2010
;
11
:
688
98
.
43.
Forero-Torres
A
,
Shah
J
,
Wood
T
,
Posey
J
,
Carlisle
R
,
Copigneaux
C
, et al
Phase I trial of weekly tigatuzumab, an agonistic humanized monoclonal antibody targeting death receptor 5 (DR5)
.
Cancer Biother Radiopharm
2010
;
25
:
13
9
.
44.
Camidge
DR
,
Herbst
RS
,
Gordon
MS
,
Eckhardt
SG
,
Kurzrock
R
,
Durbin
B
, et al
A phase I safety and pharmacokinetic study of the death receptor 5 agonistic antibody PRO95780 in patients with advanced malignancies
.
Clin Cancer Res
2010
;
16
:
1256
63
.
45.
Abdulghani
J
,
El-Deiry
WS
. 
TRAIL receptor signaling and therapeutics
.
Expert Opin Ther Targets
2010
;
14
:
1091
108
.
46.
Plummer
R
,
Attard
G
,
Pacey
S
,
Li
L
,
Razak
A
,
Perrett
R
, et al
Phase 1 and pharmacokinetic study of lexatumumab in patients with advanced cancers
.
Clin Cancer Res
2007
;
13
:
6187
94
.
47.
Varfolomeev
E
,
Maecker
H
,
Sharp
D
,
Lawrence
D
,
Renz
M
,
Vucic
D
, et al
Molecular determinants of kinase pathway activation by Apo2 ligand/tumor necrosis factor-related apoptosis-inducing ligand
.
J Biol Chem
2005
;
280
:
40599
608
.
48.
Ehrhardt
H
,
Fulda
S
,
Schmid
I
,
Hiscott
J
,
Debatin
KM
,
Jeremias
I
. 
TRAIL induced survival and proliferation in cancer cells resistant towards TRAIL-induced apoptosis mediated by NF-kappaB
.
Oncogene
2003
;
22
:
3842
52
.
49.
Soria
JC
,
Smit
E
,
Khayat
D
,
Besse
B
,
Yang
X
,
Hsu
CP
, et al
Phase 1b study of dulanermin (recombinant human Apo2L/TRAIL) in combination with paclitaxel, carboplatin, and bevacizumab in patients with advanced non-squamous non-small-cell lung cancer
.
J Clin Oncol
2010
;
28
:
1527
33
.
50.
Leong
S
,
Cohen
RB
,
Gustafson
DL
,
Langer
CJ
,
Camidge
DR
,
Padavic
K
, et al
Mapatumumab, an antibody targeting TRAIL-R1, in combination with paclitaxel and carboplatin in patients with advanced solid malignancies: results of a phase I and pharmacokinetic study
.
J Clin Oncol
2009
;
27
:
4413
21
.
51.
Mom
CH
,
Verweij
J
,
Oldenhuis
CN
,
Gietema
JA
,
Fox
NL
,
Miceli
R
, et al
Mapatumumab, a fully human agonistic monoclonal antibody that targets TRAIL-R1, in combination with gemcitabine and cisplatin: a phase I study
.
Clin Cancer Res
2009
;
15
:
5584
90
.
52.
Yu
J
,
Zhang
L
. 
PUMA, a potent killer with or without p53
.
Oncogene
2008
;
27
[Suppl 1]
:
S71
83
.
53.
Vogler
M
,
Dinsdale
D
,
Dyer
MJ
,
Cohen
GM
. 
Bcl-2 inhibitors: small molecules with a big impact on cancer therapy
.
Cell Death Differ
2009
;
16
:
360
7
.
54.
Ganten
TM
,
Haas
TL
,
Sykora
J
,
Stahl
H
,
Sprick
MR
,
Fas
SC
, et al
Enhanced caspase-8 recruitment to and activation at the DISC is critical for sensitisation of human hepatocellular carcinoma cells to TRAIL-induced apoptosis by chemotherapeutic drugs
.
Cell Death Differ
2004
;
11
[Suppl 1]
:
S86
96
.
55.
Marini
P
,
Schmid
A
,
Jendrossek
V
,
Faltin
H
,
Daniel
PT
,
Budach
W
, et al
Irradiation specifically sensitises solid tumour cell lines to TRAIL mediated apoptosis
.
BMC Cancer
2005
;
5
:
5
.
56.
Guo
F
,
Sigua
C
,
Tao
J
,
Bali
P
,
George
P
,
Li
Y
, et al
Cotreatment with histone deacetylase inhibitor LAQ824 enhances Apo-2L/tumor necrosis factor-related apoptosis inducing ligand-induced death inducing signaling complex activity and apoptosis of human acute leukemia cells
.
Cancer Res
2004
;
64
:
2580
9
.
57.
Shigeno
M
,
Nakao
K
,
Ichikawa
T
,
Suzuki
K
,
Kawakami
A
,
Abiru
S
, et al
Interferon-alpha sensitizes human hepatoma cells to TRAIL-induced apoptosis through DR5 upregulation and NF-kappa B inactivation
.
Oncogene
2003
;
22
:
1653
62
.
58.
Herzer
K
,
Hofmann
TG
,
Teufel
A
,
Schimanski
CC
,
Moehler
M
,
Kanzler
S
, et al
IFN-alpha-induced apoptosis in hepatocellular carcinoma involves promyelocytic leukemia protein and TRAIL independently of p53
.
Cancer Res
2009
;
69
:
855
62
.
59.
Stefanescu
R
,
Bassett
D
,
Modarresi
R
,
Santiago
F
,
Fakruddin
M
,
Laurence
J
. 
Synergistic interactions between interferon-gamma and TRAIL modulate c-FLIP in endothelial cells, mediating their lineage-specific sensitivity to thrombotic thrombocytopenic purpura plasma-associated apoptosis
.
Blood
2008
;
112
:
340
9
.
60.
Brancolini
C
. 
Inhibitors of the Ubiquitin-Proteasome System and the cell death machinery: How many pathways are activated?
Curr Mol Pharmacol
2008
;
1
:
24
37
.
61.
Cusack
JC
 Jr
,
Liu
R
,
Houston
M
,
Abendroth
K
,
Elliott
PJ
,
Adams
J
, et al
Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: implications for systemic nuclear factor-kappaB inhibition
.
Cancer Res
2001
;
61
:
3535
40
.
62.
Koschny
R
,
Ganten
TM
,
Sykora
J
,
Haas
TL
,
Sprick
MR
,
Kolb
A
, et al
TRAIL/bortezomib cotreatment is potentially hepatotoxic but induces cancer-specific apoptosis within a therapeutic window
.
Hepatology
2007
;
45
:
649
58
.
63.
Voortman
J
,
Resende
TP
,
Abou El Hassan
MA
,
Giaccone
G
,
Kruyt
FA
. 
TRAIL therapy in non-small cell lung cancer cells: sensitization to death receptor-mediated apoptosis by proteasome inhibitor bortezomib
.
Mol Cancer Ther
2007
;
6
:
2103
12
.
64.
Liu
X
,
Yue
P
,
Chen
S
,
Hu
L
,
Lonial
S
,
Khuri
FR
, et al
The proteasome inhibitor PS-341 (bortezomib) up-regulates DR5 expression leading to induction of apoptosis and enhancement of TRAIL-induced apoptosis despite up-regulation of c-FLIP and survivin expression in human NSCLC cells
.
Cancer Res
2007
;
67
:
4981
8
.
65.
Desagher
S
,
Osen-Sand
A
,
Montessuit
S
,
Magnenat
E
,
Vilbois
F
,
Hochmann
A
, et al
Phosphorylation of bid by casein kinases I and II regulates its cleavage by caspase 8
.
Mol Cell
2001
;
8
:
601
11
.
66.
Hellwig
CT
,
Ludwig-Galezowska
AH
,
Concannon
CG
,
Litchfield
DW
,
Prehn
JH
,
Rehm
M
. 
Activity of protein kinase CK2 uncouples Bid cleavage from caspase-8 activation
.
J Cell Sci
2010
;
123
:
1401
6
.
67.
Cursi
S
,
Rufini
A
,
Stagni
V
,
Condò
I
,
Matafora
V
,
Bachi
A
, et al
Src kinase phosphorylates Caspase-8 on Tyr380: a novel mechanism of apoptosis suppression
.
EMBO J
2006
;
25
:
1895
905
.
68.
Söderström
TS
,
Poukkula
M
,
Holmström
TH
,
Heiskanen
KM
,
Eriksson
JE
. 
Mitogen-activated protein kinase/extracellular signal-regulated kinase signaling in activated T cells abrogates TRAIL-induced apoptosis upstream of the mitochondrial amplification loop and caspase-8
.
J Immunol
2002
;
169
:
2851
60
.
69.
Harper
N
,
Hughes
MA
,
Farrow
SN
,
Cohen
GM
,
MacFarlane
M
. 
Protein kinase C modulates tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by targeting the apical events of death receptor signaling
.
J Biol Chem
2003
;
278
:
44338
47
.
70.
Tazzari
PL
,
Tabellini
G
,
Ricci
F
,
Papa
V
,
Bortul
R
,
Chiarini
F
, et al
Synergistic proapoptotic activity of recombinant TRAIL plus the Akt inhibitor Perifosine in acute myelogenous leukemia cells
.
Cancer Res
2008
;
68
:
9394
403
.
71.
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
.
72.
Jost
PJ
,
Grabow
S
,
Gray
D
,
McKenzie
MD
,
Nachbur
U
,
Huang
DC
, et al
XIAP discriminates between type I and type II FAS-induced apoptosis
.
Nature
2009
;
460
:
1035
9
.
73.
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
.
74.
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
.
75.
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
.

Supplementary data