Apoptosis can be executed through a mitochondria-dependent (intrinsic) and a death receptor–dependent, mitochondria-independent (extrinsic) pathway. The intrinsic pathway is initiated by, for example, chemotherapy and/or radiotherapy, and the extrinsic pathway is initiated by ligand-induced activation of death receptors on the cell membrane. One of the reasons for chemotherapy resistance is the failure of tumor cells to go into apoptosis due to defects in the intrinsic apoptotic pathway (e.g., changes in p53). Circumvention of chemotherapy resistance via apoptosis induction by death ligands belonging to the tumor necrosis factor family of cytokines, including tumor necrosis factor, Fas ligand, and tumor necrosis factor–related apoptosis-inducing ligand (TRAIL), exhibits cytotoxicity in tumor cell lines. The risk of side effects precludes the systemic use of recombinant forms of tumor necrosis factor and Fas ligand, but this is not the case with recombinant human TRAIL (rhTRAIL). Therefore, exploitation of the TRAIL-mediated apoptosis pathway for translation to the clinic is of interest.

TRAIL signaling pathway. TRAIL, is a type II membrane protein and its COOH terminus can be processed proteolytically to form a soluble ligand. Both membrane-expressed TRAIL and soluble TRAIL can induce apoptosis in a wide variety of tumor cell lines, whereas normal cells are relatively resistant. TRAIL binds to five different receptors. Following binding to its death receptors, DR4 and DR5, TRAIL can induce apoptosis. The membrane-bound decoy receptors DcR1 and DcR2, having no or a truncated intracellular death domain, cannot induce apoptosis. Osteoprotegerin is a soluble receptor inhibiting TRAIL (1, 2). In the mouse, only a single receptor resembling human DR4 and DR5 exists, along with two DcRs that are only distantly related by sequence to human DcR1 and DcR2, and one close homologue of osteoprotegerin. Human and murine TRAIL proteins share only 65% sequence identity. This difference underscores the difficulty to predict side effects of rhTRAIL in humans based on studies in mice. In contrast, human and cynomolgus monkey TRAIL are 98% homologous, and the receptor ectodomain sequence identity is 91% for DR4, 88% for DR5, 84% for DcR2, and 99% for osteoprotegerin, whereas cynomolgus DcR1 seems to be a pseudogene (3). Therefore, testing TRAIL in monkeys potentially better predicts its toxicity.

TRAIL binding to DR4 and DR5 at the cell surface leads to formation of the death-inducing signaling complex (Fig. 1). This complex consists of the trimerized receptor, the adaptor protein Fas-associated death domain, and caspase-8. On death-inducing signaling complex formation, caspase-8 is activated, which in type I cells leads to activation of caspase-3 and subsequently apoptosis. In type II cells, TRAIL also triggers the mitochondrial pathway. Active caspase-8 causes cleavage of BID into tBID, which translocates to the mitochondria leading to release of cytochrome c and Smac/DIABLO. Cytochrome c together with Apaf-1 can activate caspase-9, which then activates caspase-3 (Fig. 1).

Fig. 1.

Schematic representation of the intrinsic and extrinsic apoptotic pathway. Apoptosis induction via the death receptor can result in activation of the extrinsic and intrinsic pathways. Chemotherapy and radiotherapy-induced apoptosis is executed via the intrinsic pathway. , activation; , inhibition.

Fig. 1.

Schematic representation of the intrinsic and extrinsic apoptotic pathway. Apoptosis induction via the death receptor can result in activation of the extrinsic and intrinsic pathways. Chemotherapy and radiotherapy-induced apoptosis is executed via the intrinsic pathway. , activation; , inhibition.

Close modal

Function of TRAIL in physiology and pathophysiology. Studies with a neutralizing rat anti-mouse TRAIL antibody showed a role for TRAIL in immune surveillance against tumor development in mice (4, 5). In mice, this antibody promoted tumor outgrowth of TRAIL-sensitive tumors (5). TRAIL gene knockout mice show an increased susceptibility to tumor initiation and metastases and autoimmune diseases (6).

However, recent evidence indicates that in several tumor cells, including human colon cancer cells, TRAIL can mediate tumor cell proliferation, which may have a dominant effect over the apoptotic signal (7). Moreover, a role for DR5 as a candidate tumor suppressor was established. In human colon cancer cell lines, silencing of DR5 in vivo led to accelerated growth of bioluminescent tumor xenografts and conferred resistance of the tumor cells to the chemotherapeutic agent 5-fluorouracil (8).

Chronic TRAIL blockade in mice exacerbates collagen-induced arthritis and experimental autoimmune encephalomyelitis. TRAIL deficiency also increases the susceptibility of mice to autoimmune arthritis and diabetes (9). The Fas-Fas ligand pathway has been studied extensively in humans in relation to systemic lupus erythematosus. Soluble TRAIL concentrations in the serum, however, are also increased in these patients. This seems to be disease specific, as levels in patients with rheumatoid arthritis and Wegener's granulomatosis were not elevated, suggesting a role for TRAIL in systemic lupus erythematosus pathophysiology (10). TRAIL also plays a role in infections. Dysregulated immune and coagulation systems are held responsible for the septic syndrome. However, additional pathophysiologic mechanisms, such as uncontrolled apoptosis induced by death receptor ligands, might well play a role. Endotoxin administration to volunteers caused not only an increase in tumor necrosis factor but also a 10-fold increase in plasma soluble TRAIL 2.5 hours after administration (11). In addition, virally infected cells are more susceptible for TRAIL-induced apoptosis (12). This illustrates that TRAIL does play a role in normal physiology and diseases and illustrates that the human body can tolerate certain TRAIL levels.

Osteoprotegerin is a key regulator of osteoclastogenesis, but recently also osteoprotegerin involvement in the control of endothelial cell survival through inhibition of TRAIL activity was suggested. Endothelial osteoprotegerin expression was associated with high tumor grade and certain histologic types in 400 breast tumors. This suggests a potential biological role for osteoprotegerin in development and/or maintenance of tumor vasculature (13).

Preclinical in vivo antitumor efficacy and toxicity of rhTRAIL and TRAIL receptor antibodies. In animals, rhTRAIL reduced growth of human tumor xenografts as well as xenotransplants and potentiated chemotherapy and radiotherapy effect. Furthermore, rhTRAIL administration in nonhuman primates showed no serious toxicity towards normal tissue. The rhTRAIL half-life in chimpanzees, however, was only 25 minutes (3). Most chimpanzee and human tissues express TRAIL receptors, whereas a few differences were observed between human and chimpanzee tissues. Lack of liver toxicity in chimpanzees after rhTRAIL administration despite DR4 and DR5 expression was reassuring for rhTRAIL application in humans (14). Moreover, no TRAIL hepatotoxicity was observed in human hepatocytes within the chimeric liver of severe combined immunodeficient mice (15).

Several agonistic antibodies against the human receptors DR4 and DR5 have been developed. These antibodies, just like rhTRAIL, showed antitumor activity alone in several preclinical tumor models and both potentiated the effect of chemotherapy (1619). There were no signs of serious side effects in animals following DR4 and DR5 antibody administration. However, one has to take in account that these animals have been exposed to antibodies specific for human DR4 and DR5.

Translational advances. In a few studies, the prognostic significance of TRAIL and TRAIL receptor expression in tumors has been determined. No differences in TRAIL mRNA expression levels were observed between stage I/II and stage III/IV ovarian cancer, but high TRAIL expression was associated with longer survival (20). DR4 was an independent prognostic factor for disease-free survival in 129 patients with stage II/III colon cancer (21). In non–small cell lung cancer, DR5-positive staining was associated with increased risk of death (22). Similarly, high DR5 expression in breast cancer strongly correlated with decreased survival using tissue microarrays containing specimens from 655 breast cancer patients (23).

Clinical studies. Clinical studies with compounds directed at TRAIL receptors are done as phase I study with rhTRAIL (Genentech, South San Francisco, CA) and with one agonistic TRAIL-DR4 antibody and two agonistic TRAIL-DR5 antibodies (all Human Genome Sciences, Rockville, MD). For the clinical study with rhTRAIL, no data are yet available. The TRAIL-DR4 antibody was well tolerated in phase I studies. The maximum tolerated dose was not yet reached with data up to 10 mg/kg per cycle. The mean terminal elimination half-life was 16 days. Stable diseases occurred in heavily pretreated/refractory patients (24, 25). Phase II studies in 38 non–small cell lung cancer patients showed that 10 mg/kg doses i.v. per 21 days was well tolerated and gave stable disease in 29% (26). In 38 colon cancer patients, stable disease was observed in 31.6% (27). In a phase II study with 40 non-Hodgkin's lymphoma patients, one complete and two partial tumor responses, all in follicular lymphoma patients, occurred (28). This study proves that the TRAIL-DR4 antibody has antitumor activity on its own. Based on preclinical data showing potentiation of chemotherapy effect, studies are ongoing in which TRAIL-DR4 antibody is added to gemcitabine/cisplatin or paclitaxel/carboplatin combination regimens (29, 30). In these studies, no pharmacokinetic interaction is observed and the optimal dose is not yet known.

Several studies have been done with TRAIL-DR5 antibodies (i.e., two phase I studies with ETR2 and a phase I with TR2J), but no data of the TR2J study are available yet. With the ETR2, one ongoing study in 28 patients noticed minimal toxicity at doses of 10 mg/kg every 21 days (31). Eight patients experienced a stable disease after two cycles. In the second study, ETR2 was well tolerated at doses of 10 mg/kg every 2 weeks (32).

Early results from the clinical studies show that, perhaps in contrast to the expectations, these drugs can be administered safely at relevant doses to patients. As rhTRAIL and agonistic DR4 and DR5 antibodies differ in pharmacokinetic behavior and target DR4 and DR5 versus DR4 or DR5, it is too early to decide which of these TRAIL pathway targeted drugs is to be preferred in the clinic.

The effect of recombinant osteoprotegerin was studied in patients with juvenile Paget's disease (33). This disease is characterized by an accelerated bone turnover due to inactivating mutations in the gene encoding osteoprotegerin. Recombinant osteoprotegerin increased radical bone mass. It is unknown whether this might potentiate tumor development.

Interestingly, premalignant lesions of the cervix and colon adenomas also overexpress DR4 and DR5 (34, 35). In addition, in vitro exposure of these premalignant tissues to TRAIL can induce apoptosis. This opens opportunities for secondary prevention when the TRAIL receptor targeted compounds can be administered safely.

There is ongoing research directed at the development of more specific and more potent TRAIL analogues (36, 37).

Initially, TRAIL receptor targeted drugs were especially considered as a relevant adjunct to chemotherapy. However, with the availability of numerous smart drugs, targeting relevant players along the TRAIL signaling pathway combinations of several of these drugs, also based on additive effects in preclinical models, seems of major interest. Interesting combinations (Fig. 2) are envisioned with the following:

Fig. 2.

Illustration of how drugs, such as chemotherapeutic drugs as well as proteasome inhibitors, histone deacetylase (HDAC) inhibitors, inhibitors of antiapoptotic Bcl-2 family members (Bcl-2 and Bcl-XL), and inhibitors of apoptosis protein (IAP), interfere in the TRAIL signaling pathway and can potentiate the effect of rhTRAIL. , activation; , inhibition.

Fig. 2.

Illustration of how drugs, such as chemotherapeutic drugs as well as proteasome inhibitors, histone deacetylase (HDAC) inhibitors, inhibitors of antiapoptotic Bcl-2 family members (Bcl-2 and Bcl-XL), and inhibitors of apoptosis protein (IAP), interfere in the TRAIL signaling pathway and can potentiate the effect of rhTRAIL. , activation; , inhibition.

Close modal
  1. Proteasome inhibitors as the proteasome is responsible for the degradation of the vast majority of intracellular proteins, including proapoptotic proteins, and thus affects multiple signaling pathways within cells (38).

  2. Histone deacetylase inhibitors as they induce differentiation and/or apoptosis in a variety of cell types by activating transcription of target genes. Histone deacetylase inhibitors synergize with TRAIL by, for example, inducing TRAIL-DR4 and TRAIL-DR5 through nuclear factor-κB activation and some proapoptotic members of the Bcl-2 family and engaging the mitochondrial pathway (39).

  3. Inhibitors of antiapoptotic Bcl-2 family members (Bcl-2 and Bcl-XL) or inhibitors of apoptosis protein that thus have the capacity to remove roadblocks on the TRAIL signaling pathway (Fig. 2).

Given the large number of proteins that play a role in TRAIL-induced apoptosis, it can be envisioned that tumor profiling of the relevant genes will be major importance for patient tailored therapy with these drugs.

Grant support: Dutch Cancer Society grants 2000-2286, 2000-2289, 2003-2922, 2005-3361, and 2005-3365.

Note: Presented at the AACR-National Cancer Institute-European Organisation for Research and Treatment of Cancer International Conference on Molecular Targets and Cancer Therapeutics: Discovery, Biology, and Clinical Applications, Philadelphia, PA, November 14-18, 2005.

1
LeBlanc HN, Ashkenazi A. Apo2L/TRAIL and its death and decoy receptors.
Cell Death Differ
2003
;
10
:
66
–75.
2
Ashkenazi A. Targeting death and decoy receptors of the tumor necrosis factor superfamily.
Nat Rev Cancer
2002
;
2
:
420
–30.
3
Kelley SK, Harris LA, Xie D, 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.
4
Takeda K, Smyth MJ, Cretney E, et al. Critical role for tumor necrosis factor-related apoptosis-inducing ligand in immune surveillance against tumor development.
J Exp Med
2002
;
195
:
161
–9.
5
Takeda K, Smyth MJ, Cretney E, et al. Involvement of tumor necrosis factor-related apoptosis-inducing ligand in NK cell-mediated and IFN-γ-dependent suppression of subcutaneous tumor growth.
Cell Immunol
2001
;
214
:
194
–200.
6
Sedger LM, Glaccum MB, Schuh JC, et al. Characterization of the in vivo function of tumor necrosis factor-α-related apoptosis-inducing ligand, TRAIL/Apo2L, using TRAIL/Apo2L gene-deficient mice.
Eur J Immunol
2002
;
32
:
2246
–54.
7
Baader E, Tolocko A, Fuchs U, et al. Tumor necrosis factor-related apoptosis-inducing ligand-mediated proliferation of tumor cells with receptor-proximal apoptosis defects.
Cancer Res
2005
;
65
:
7888
–95.
8
Wang S, El-Deiry WS. Inducible silencing of KILLER/DR5 in vivo promotes bioluminescent colon tumor xenograft growth and confers resistance to chemotherapeutic agent 5-fluorouracil.
Cancer Res
2004
;
64
:
6666
–72.
9
Lamhamedi-Cherradi SE, Zheng SJ, Maguschak KA, Peschon J, Chen YH. Defective thymocyte apoptosis and accelerated autoimmune diseases in TRAIL−/− mice.
Nat Immunol
2003
;
4
:
255
–60.
10
Lub-de Hooge MN, de Vries EG, de Jong S, Bijl M. Soluble TRAIL concentrations are raised in patients with systemic lupus erythematosus.
Ann Rheum Dis
2005
;
64
:
854
–8.
11
Lub-de Hooge MN, de Jong S, Vermot-Desroches C, Tulleken JE, de Vries EG, Zijlstra JG. Endotoxin increases plasma soluble tumor necrosis factor-related apoptosis-inducing ligand level mediated by the p38 mitogen-activated protein kinase signaling pathway.
Shock
2004
;
22
:
186
–8.
12
Wei W, Liu YX, Zheng de X. Current understanding on the immunological functions of tumor necrosis factor-related apoptosis-inducing ligand.
Cell Mol Immunol
2005
;
2
:
265
–9.
13
Cross SS, Yang Z, Brown NJ, et al. Osteoprotegerin (OPG)—a potential new role in the regulation of endothelial cell phenotype and tumour angiogenesis?
Int J Cancer
2006
;
118
:
1901
–8.
14
Spierings DC, de Vries EG, Vellenga E, et al. Tissue distribution of the death ligand TRAIL and its receptors.
J Histochem Cytochem
2004
;
52
:
821
–31.
15
Hao C, Song JH, Hsi B, et al. TRAIL inhibits tumor growth but is nontoxic to human hepatocytes in chimeric mice.
Cancer Res
2004
;
64
:
8502
–6.
16
Buchsbaum DJ, Zhou T, Grizzle WE, et al. Antitumor efficacy of TRA-8 anti-DR5 monoclonal antibody alone or in combination with chemotherapy and/or radiation therapy in a human breast cancer model.
Clin Cancer Res
2003
;
9
:
3731
–41.
17
Zeng Y, Wu XX, Fiscella M, et al. Monoclonal antibody to tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAIL-R2) induces apoptosis in primary renal cell carcinoma cells in vitro and inhibits tumor growth in vivo.
Int J Oncol
2006
;
28
:
421
–30.
18
Georgakis GV, Li Y, Humphreys R, et al. Activity of selective fully human agonistic antibodies to the TRAIL death receptors TRAIL-R1 and TRAIL-R2 in primary and cultured lymphoma cells: induction of apoptosis and enhancement of doxorubicin- and bortezomib-induced cell death.
Br J Haematol
2005
;
130
:
501
–10.
19
Pukac L, Kanakaraj P, Humphreys R, et al. HGS-ETR1, a fully human TRAIL-receptor 1 monoclonal antibody, induces cell death in multiple tumour types in vitro and in vivo.
Br J Cancer
2005
;
92
:
1430
–41.
20
Lancaster JM, Sayer R, Blanchette C, et al. High expression of tumor necrosis factor-related apoptosis-inducing ligand is associated with favorable ovarian cancer survival.
Clin Cancer Res
2003
;
9
:
762
–6.
21
Strater J, Hinz U, Walczak H, et al. Expression of TRAIL and TRAIL receptors in colon carcinoma: TRAIL-R1 is an independent prognostic parameter.
Clin Cancer Res
2002
;
8
:
3734
–40.
22
Spierings DC, de Vries EG, Timens W, Groen HJ, Boezen HM, de Jong S. Expression of TRAIL and TRAIL death receptors in stage III non-small cell lung cancer tumors.
Clin Cancer Res
2003
;
9
:
3397
–405.
23
McCarthy MM, Sznol M, DiVito KA, Camp RL, Rimm DL, Kluger HM. Evaluating the expression and prognostic value of TRAIL-R1 and TRAIL-R2 in breast cancer.
Clin Cancer Res
2005
;
11
:
5188
–94.
24
Hirte HW, Hotte SJ, Chen EX, et al. HGS-ETR1, a fully human monoclonal antibody to the tumor necrosis factor-related apoptosis-inducing ligand receptor 1 (TRAIL-R1) in patients with advanced solid cancer: results of a phase 1 trial [abstract C22].
Clin Cancer Res
2005
;
11
:
9104S
.
25
Mita MT, Tolcher AW, Patnaik A, et al. A phase 1, pharmacokinetic (PK) study of HGS-ETR1, an agonistic monoclonal antibody to TRAIL-R1, in patients with advanced solid tumors [abstract 544].
Proc Am Assoc Cancer Res
2005
;
46
:
128
.
26
Bonomi P, Greco FA, Crawford J, et al. Results of a phase 2 trial of HGS-ETR1 (agonistic human monoclonal antibody to TRAIL receptor 1) in subjects with relapsed/recurrent non-small cell lung cancer [abstract 1851]. 11th World Conference on Lung Cancer.
Lung Cancer
2005
;
49
:
S237
.
27
Kanzler S, Trarbach T, Heinemann V, et al. Results of a phase 2 trial of HGS-ETR1 (agonistic human monoclonal antibody to TRAIL receptor 1) in subjects with relapsed or refractory colorectal cancer (CRC) [abstract 630].
Eur J Cancer Suppl
2005
;
3
:
177
.
28
Younes A, Vose J, Zelenetz AD, et al. Results of a phase 2 trial of HGS-ETR1 (agonistic human monoclonal antibody to TRAIL receptor 1) in subjects with relapsed/refractory non-Hodgkin's lymphoma (NHL) (ETR1-01) [abstract 489].
Blood
2005
;
106
:
146a
–7a.
29
Mom CH, Sleijfer S, Gietema JA, et al. A phase 1 study of HGS-ETR1, a fully human monoclonal antibody to TRAIL-R1, in combination with gemcitabine and cisplatin in subjects with advanced solid malignancies [abstract C74].
Clin Cancer Res
2005
;
11
:
9117S
.
30
Hariharan S, Gore L, Eckhardt SG, et al. A phase 1 and pharmacological study of HGS-ETR1, an antibody targeting TRAIL-R1, in combination with paclitaxel and carboplatin in patients with advanced solid malignancies [abstract B109].
Clin Cancer Res
2005
;
11
:
9058S
–9S.
31
Attard G, Plummer R, de Bono JS, et al. Phase 1 and pharmacokinetic study of HGS-ETR2, a human monoclonal antibody to TRAIL-R2, in patients with advanced solid malignancies [abstract B114].
Clin Cancer Res
2005
;
11
:
9060S
.
32
Sarantopoulos J, Wakelee H, Mita M, et al. A phase 1 clinical trial of HGS-ETR2, a fully human activating monoclonal antibody to TRAIL-R2, in patients with advanced solid tumors [abstract C21].
Clin Cancer Res
2005
;
11
:
9104S
.
33
Cundy T, Davidson J, Rutland MD, Stewart C, DePaoli AM. Recombinant osteoprotegerin for juvenile Paget's disease.
N Engl J Med
2005
;
353
:
918
–23.
34
Reesink-Peters N, Hougardy BM, van den Heuvel FA, et al. Death receptors and ligands in cervical carcinogenesis: an immunohistochemical study.
Gynecol Oncol
2005
;
96
:
705
–13.
35
Jalving M, de Jong S, Koornstra JJ, et al. TRAIL induces apoptosis in human colorectal adenoma cell lines and human colorectal adenomas. Clin Cancer Res. In press.
36
Kelley RF, Totpal K, Lindstrom SH, et al. Receptor-selective mutants of apoptosis-inducing ligand 2/tumor necrosis factor-related apoptosis-inducing ligand reveal a greater contribution of death receptor (DR) 5 than DR4 to apoptosis signaling.
J Biol Chem
2005
;
280
:
2205
–12.
37
van der Sloot AM, Mullally MM, Fernandez-Ballester G, Serrano L, Quax WJ. Stabilization of TRAIL, an all-β-sheet multimeric protein, using computational redesign.
Protein Eng Des Sel
2004
;
17
:
673
–80.
38
Hougardy BM, Maduro JH, van der Zee AG, et al. Proteasome inhibitor MG132 sensitizes HPV-positive human cervical cancer cells to rhTRAIL-induced apoptosis.
Int J Cancer
2006
;
118
:
1892
–900.
39
Earel JK, Jr., VanOosten RL, Griffith TS. Histone deacetylase inhibitors modulate the sensitivity of tumor necrosis factor-related apoptosis-inducing ligand-resistant bladder tumor cells.
Cancer Res
2006
;
66
:
499
–507.