The discovery of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) along with its potent and selective antitumor effects initiated a decades-long search for therapeutic strategies to target the TRAIL pathway. First-generation approaches were focused on the development of TRAIL receptor agonists (TRAs), including recombinant human TRAIL (rhTRAIL) and TRAIL receptor-targeted agonistic antibodies. While such TRAIL pathway-targeted therapies showed promise in preclinical data and clinical trials have been conducted, none have advanced to FDA approval. Subsequent second-generation approaches focused on improving upon the specific limitations of first-generation approaches by ameliorating the pharmacokinetic profiles and agonistic abilities of TRAs as well as through combinatorial approaches to circumvent resistance. In this review, we summarize the successes and shortcomings of first- and second-generation TRAIL pathway-based therapies, concluding with an overview of the discovery and clinical introduction of ONC201, a compound with a unique mechanism of action that represents a new generation of TRAIL pathway-based approaches. We discuss preclinical and clinical findings in different tumor types and provide a unique perspective on translational directions of the field.

The evasion of apoptosis was recognized in the late 1980s as a mechanism of cancer development and progression and was included in 2000 among the original key hallmarks of cancer. Since then, much work has been done to pursue the targeted activation of apoptotic pathways in cancer cells as a viable therapeutic strategy [1,2].

Apoptosis itself was first identified as a controlled cell death pathway in 1972, and years since have seen the emergence of two broad ‘pathways' driving the process: cell ‘intrinsic' and cell ‘extrinsic' [3,4]. Though interplay between the two pathways has been observed, the intrinsic pathway largely relies on the mitochondria and the BCL-2 family of proteins to trigger apoptosis in response to internal stress borne by the cell, while for the extrinsic pathway, ligand binding to cell surface death receptors translates various death signals into a caspase cascade which ultimately leads to cell death [5] (Figure 1).

Cell death signaling activated by TRAIL and TRAIL pathway therapeutics.

Figure 1.
Cell death signaling activated by TRAIL and TRAIL pathway therapeutics.

TRAIL pathway signaling is initiated upon TRAIL binding to DR4 or DR5, driving receptor trimerization and recruitment of the FADD to form the DISC. DR4 and DR5 compete with decoy receptors DcR1, DcR2, and the soluble OPG for TRAIL. Activation of DR4 or DR5 triggers apoptotic signaling initially through initiator caspases, primarily caspase-8. Extrinsic apoptotic signaling (blue arrows) continues through activation of executioner caspases-3, -6, and -7, resulting in apoptotic death. Caspase-8 additionally activates intrinsic apoptotic signaling (green arrows) through facilitating conversion of Bid to tBid, driving proapoptotic factor release from the mitochondrion, apoptosome assembly, and ultimately also converging on executioner caspase activation. Several therapies, as described in the text, have been developed to engage at several points along this path (red labels and red arrows). Some, including first and second generation rTRAIL, TRAs, and antibodies, directly agonize DR4 and DR5 (unless otherwise indicated, these agonists have described activity against both DR4 and DR5). Downstream, inhibitors of c-FLIP prevent its competition with pro-caspase-8 for the FADD to enhance caspase-8 activation. Bcl-2 family inhibitors act to mitigate the function of antiapoptotic members of the Bcl-2 family, especially Bcl-2 and Bcl-xL. Smac mimetics also help to sequester anti-apoptotic IAPs. On a transcriptional level, ONC201 (initially referred to as TIC10 for TRAIL-inducing compound 10) antagonizes DRD2 and DRD3 while agonizing ClpP, ultimately bringing about growth arrest through the integrated stress response (ISR). Moreover, downstream effects of ONC201 (alongside p53) drive increased expression of TRAIL and DR5. Pointed arrows indicate activation; blunted arrows indicate inhibition.

Figure 1.
Cell death signaling activated by TRAIL and TRAIL pathway therapeutics.

TRAIL pathway signaling is initiated upon TRAIL binding to DR4 or DR5, driving receptor trimerization and recruitment of the FADD to form the DISC. DR4 and DR5 compete with decoy receptors DcR1, DcR2, and the soluble OPG for TRAIL. Activation of DR4 or DR5 triggers apoptotic signaling initially through initiator caspases, primarily caspase-8. Extrinsic apoptotic signaling (blue arrows) continues through activation of executioner caspases-3, -6, and -7, resulting in apoptotic death. Caspase-8 additionally activates intrinsic apoptotic signaling (green arrows) through facilitating conversion of Bid to tBid, driving proapoptotic factor release from the mitochondrion, apoptosome assembly, and ultimately also converging on executioner caspase activation. Several therapies, as described in the text, have been developed to engage at several points along this path (red labels and red arrows). Some, including first and second generation rTRAIL, TRAs, and antibodies, directly agonize DR4 and DR5 (unless otherwise indicated, these agonists have described activity against both DR4 and DR5). Downstream, inhibitors of c-FLIP prevent its competition with pro-caspase-8 for the FADD to enhance caspase-8 activation. Bcl-2 family inhibitors act to mitigate the function of antiapoptotic members of the Bcl-2 family, especially Bcl-2 and Bcl-xL. Smac mimetics also help to sequester anti-apoptotic IAPs. On a transcriptional level, ONC201 (initially referred to as TIC10 for TRAIL-inducing compound 10) antagonizes DRD2 and DRD3 while agonizing ClpP, ultimately bringing about growth arrest through the integrated stress response (ISR). Moreover, downstream effects of ONC201 (alongside p53) drive increased expression of TRAIL and DR5. Pointed arrows indicate activation; blunted arrows indicate inhibition.

Close modal

One such death receptor ligand is tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), which binds death receptors DR4 and DR5 and several decoy receptors. While DR4 and DR5 are tumor cell-expressed TRAIL-binding receptors that contain death domains to trigger the induction of apoptosis, decoy receptors lack the death domain and are primarily expressed on normal cells. This ability of TRAIL to preferentially induce apoptosis in tumor cells, but not normal cells, provides a therapeutic window. Thus, following its discovery in 1996, the tumoricidal potential of TRAIL was pursued along with a search for TRAIL-based treatments [6,7] (Figure 1). First explored were TRAIL receptor-based therapies, namely recombinant human TRAIL (rhTRAIL) and death receptor agonistic antibodies. We cloned Death Receptor 5 (DR5; KILLER/DR5) as a p53 target gene which provided a rationale for the combination of TRAIL with DNA damaging therapeutics [8]. These first-generation TRAIL treatments were found to both directly lead to tumor cell death as well as result in the sensitization of tumor cells to other chemotherapies and radiotherapies [9]. However, such strategies have limitations, ranging from weak binding to death receptors, to poor pharmacokinetics, to acquired resistance. As such, novel second-generation treatment strategies have been sought to address these shortcomings, working to either increase or supplement the core efficacy of TRAIL receptor-based therapies, or to prevent the acquisition of resistance and re-sensitize tumors to first-generation agents. In addition, the recent discovery and emerging clinical success of TIC10/ONC201 have led to its recognition as a ‘next-generation' approach.

In this review, we provide a brief overview of key therapies exploiting the TRAIL pathway, along with their limitations and subsequent advances, concluding with a perspective on novel strategies of therapeutic TRAIL pathway-mediated induction of tumor cell death.

TRAIL itself can exist in one of two forms: either as a transmembrane protein form expressed by various cell types, or as a soluble form composed of the shed extracellular domain of membrane-bound TRAIL which self-trimerizes [9]. As previously mentioned, both forms are capable of binding target receptors DR4 and DR5 to trigger an apoptotic signal on the death-receptor expressing cell, but may also bind membrane-bound decoy receptors DcR1 and DcR2 or soluble decoy receptor OPG [10]. Consequently, an initial point of apoptotic signal modulation exists through the expression of such decoy receptors, which ultimately can reduce TRAIL-induced death signaling through competition with DR4 and DR5. Indeed, decoy receptor expression has been implicated, among other mechanisms, in the resistance of normal cells to TRAIL [11,12] as well as in cancer cell resistance to TRAIL [13].

Cell death via the extrinsic apoptotic pathway is initiated by ligands such as TRAIL binding to their target death receptors. Upon ligand binding, the death receptors trimerize, leading to their activation. This is followed by the recruitment of the adaptor protein Fas-associated protein with death domain (FADD) as well as initiator caspases-8 and -10. Together, these molecules assemble into the death-inducing signaling complex (DISC) [14]. While lower-order death receptor clustering (i.e. trimerization) is essential to efficient DISC formation, the higher-order clustering of multiple complexes has emerged as being another important factor in apoptotic signal transduction [15]. Downstream of DISC formation, resultant activation of initiator caspase-8 leads to the activation of executioner caspases-3, -6, and -7, which leads to cell death.

In so-called Type I cells, this pathway alone is sufficient to activate the executioner caspases and induce cell death. Conversely, in Type II cells, additional amplification via the intrinsic mitochondrial apoptotic pathway is required. In these cells, Bid cleavage by caspase-8 is followed by its myristoylation and translocation to the mitochondria, where it, along with Bax and Bak, allows for the release of proapoptotic factor cytochrome C [16,17]. Cytochrome C complexes with apoptotic peptidase activating factor 1 (Apaf-1) and initiator caspase-9 to form an executioner caspase cascade-activating apoptosome. TRAIL signaling at DR5 receptors has additionally been shown to translocate proapoptotic Bim and Bax to lysosomes, resulting in a release of cathepsin B that further enhances mitochondrial outer membrane permeabilization and executioner caspase activation through the recruitment of phosphofurin acidic cluster sorting protein-2 (PACS-2) to DR5-positive endosomes [18]. C-Myc was identified as a major determinant of TRAIL sensitivity [19].

An early study in the exploration of TRAIL pathway-based therapeutics assessed the expression of TRAIL receptors by immunohistochemistry in primary glioblastoma (GBM) specimens. The study showed that higher receptor expression is correlated with increased patient survival, supporting the important role played by TRAIL in commanding patient prognosis [20,21]. Thus, receptor-targeted therapies garnered much attention based on their potential as effective anticancer agents. Many first-generation TRAIL receptor-targeted agents focused on recombinant forms of TRAIL as well as agonistic antibodies against TRAIL death receptors.

Recombinant forms of TRAIL

One strategy to target the TRAIL pathway is to develop bioactive recombinant soluble forms of TRAIL. One such example is dulanermin (AMG-951), which is a soluble recombinant human Apo2Ligand/TRAIL (rhApo2L/TRAIL) protein that induces apoptosis in target cells via its binding and activation of DR4 (i.e. TRAIL-R1) and DR5 (i.e. TRAIL-R2), though it is also known to bind to decoy receptors [22]. Developed by Amgen/Genentech, dulanermin is the only rhTRAIL to have reached clinical trials. In a phase 1 dose-escalation study in patients with advanced cancer, dulanermin was shown to have a tolerable safety profile; however, its observed anticancer activity was limited [22]. Of note, the mean terminal phase half-life (t1/2) of rhTRAIL ranges from 0.56 to 1.02 h, likely restricting its ability to reach tumor cells [22,23]. Moreover, because rhTRAIL binds to decoy receptors, its delivery to DR4/5 is diluted. This weakened agonism is worsened by the fact that although rhTRAIL induces lower-order receptor trimerization, it has a limited capacity to induce higher-order receptor clustering, resulting in a weak apoptotic signal [24,25]. Meanwhile, other first-generation TRAIL pathway-based therapies were developed.

Death receptor agonist antibodies

The development of agonistic antibodies against DR4/5 presented another strategy to target TRAIL receptors. Unlike rhTRAIL, which binds to both death receptors and decoy receptors, diluting its apoptotic effect, agonistic antibodies specifically target DR4 or DR5 to activate apoptosis in target cells. Moreover, receptor-targeted monoclonal antibodies (TRAIL-R mAbs) have a significantly longer half-life than rhTRAIL. Therefore, the development of these therapeutic antibodies was pursued as an anti-cancer strategy.

Preclinical studies showed promising results, with significant antitumor activity without toxicity seen both in vivo and in vitro [26]. Consequently, several first-generation TRAIL-R mAbs have been tested in clinical trials including DR5-targeted agents such as lexatumumab, conatumumab, drozitumab, tigatuzumab, and LBY135. DR5-targeted mAbs showed results in early clinical trials that only modestly improved upon clinical results observed with rhTRAIL. For example, in a phase 1 trial of lexatumumab in patients with advanced cancers, the mAb was well-tolerated and was associated with stable disease in several patients [27]. Safety was also demonstrated in the first-in-human study of conatumumab in adult patients with advanced solid tumors [28]. In this study, a single patient with colorectal cancer (CRC) had stable disease for 24 weeks and showed a 24% reduction in tumor size by RECIST (Response Evaluation Criteria in Solid Tumors) criteria. Drozitumab is another DR5-targeted mAb that was evaluated in a phase 1 study in patients with advanced malignancies [29]. In this study, three minor responses were observed in patients with CRC, granulosa cell ovarian cancer, and chondrosarcoma, although no objective responses were observed. Moreover, in a phase 1 trial of DR5-targeted tigatuzumab, stable disease and a tolerable safety profile were observed [30]. A DR4-targeted agent, mapatumumab, which is a fully human mAb also entered clinical trials. In a phase 1 study in patients with advanced solid malignancies, mapatumumab was observed to be well-tolerated, however, no objective responses were observed [31]. Another approach using DR4 atrimers was explored although this was not pursued in the clinic [32].

Thus, although successful in preclinical studies, TRAIL-R mAbs have performed modestly in clinical trials. Despite improving upon the pharmacokinetic ability and specificity of rhTRAIL, the agonistic ability of TRAIL-R mAbs remains limited. One factor contributing to this is that mAbs have an inherently bivalent structure, only allowing for their cross-linking to two death receptors. As lower-order receptor trimerization is required to ensure efficient DISC-formation, additional cross-linking is needed for sufficient apoptotic signaling [33]. Another limiting factor is that while first-generation TRAIL-R mAbs are DR4- or DR5-specific, there are differences in the ability of each receptor to transmit apoptotic signals that vary between tissue types [34], pointing to the need for antibodies that target both receptors.

First-generation therapies were thus divided into two categories: recombinant forms of TRAIL and receptor-specific agonistic antibodies. Although these early TRAIL receptor agonists (TRAs) were shown to be promising in preclinical data [35,36] and well-tolerated in patients [22,37], they ultimately proved to have limited clinical anticancer activity. This can be attributed to several factors, including poor pharmacokinetics, limited agonistic ability, and inherent or acquired resistance [2,38]. Second-generation TRAs were developed to target these limitations by improving upon the pharmacokinetic profile and agonistic ability of these agents and circumventing resistance through combinatorial approaches.

TRAIL derivatives

N-terminal tags

An early second-generation approach was to modulate the rhTRAIL protein through the addition of N-terminal tags to improve its pharmacokinetic profile and agonistic ability. The small molecular weight of rhTRAIL is what leads to its rapid clearance via renal filtration. Adding a molecular tag to rhTRAIL increases its size and thus improves its half-life [34,39]. Several ‘tagged’ forms of rhTRAIL were developed, including the addition of Flag (FLAG-TRAIL), poly-histidine (His-TRAIL), leucine zipper (LZ-TRAIL), isoleucine zipper (IZ-TRAIL), or tenascin-C (TNC) oligomerization domain (TNC-TRAIL) [40]. Both FLAG-TRAIL and His-TRAIL were created to facilitate the purification process [38]. On the other hand, LZ-TRAIL, IZ-TRAIL, and TNC-TRAIL were all developed to stabilize rhTRAIL homotrimerization through hydrophobic interactions within the LZ, IZ, and TNC sequences [6,41,42]. This increased stability enhances the agonistic activity and apoptotic potential of rhTRAIL. Despite promising preclinical data, FLAG-TRAIL and His-TRAIL were found to induce significant apoptosis in primary human hepatocytes [43]. While LZ-TRAIL, IZ-TRAIL, and TNC-TRAIL were not found to be significantly hepatotoxic [41], concern over on-target or off-target toxicity hindered the translation of tagged rhTRAIL into the clinic.

TLY012

Another approach to improving rhTRAIL is to covalently link it to molecules that enhance its pharmacokinetic profile. One such fusion product is TLY012, an N-terminal PEGylated rhTRAIL. Like the tags, the covalent attachment of polyethylene glycol (PEG) to a therapeutic protein increases its size, thereby reducing its clearance by renal filtration and increasing its half-life [44–46]. Indeed, TLY012 was shown to have a greatly increased half-life compared with rhTRAIL in a molecular weight-dependent manner, with 20K- and 30K-PEG-TRAIL having circulating half-lives of 12 and 18 h, respectively, compared with the 1 h half-life of rhTRAIL in ICR mice [47]. This increased half-life was associated with a greater anti-tumor effect both in vitro and in vivo in CRC models [47]. Later, TLY012 was shown also to have activity against fibrotic cells [48,49]. While TLY012 is entering clinical trials for the treatment of fibrotic diseases and was granted Orphan Drug Designation (ODD) by the FDA in 2019 for the treatment of systemic sclerosis, further exploration of its anticancer potential as a single agent and in combination has been ongoing (further discussed below).

TRAIL-Mu3

A similar approach to enhancing the pharmacokinetic profile of rhTRAIL involves making modifications to the amino acid sequence to alter the chemical properties of the complete peptide. Mutagenesis of amino acid sequence VRERGPQR (114–121) into RRRRRRRR, for instance, has been shown to enhance the membrane-penetrating abilities of wild-type TRAIL, leading to the development of a novel membrane-penetrating TRAIL-Mu3 [50]. TRAIL-Mu3 has been shown to have enhanced efficacy both in in vitro and in vivo preclinical models against pancreatic cancers over both rhTRAIL as well as gemcitabine [50]. TRAIL-Mu3 has a greater tendency to activate caspase cascade in pancreatic cancer cell lines compared with rhTRAIL [51]. Though TRAIL-Mu3 is currently confined to the preclinical setting, its deployment into the clinic may allow for a novel therapeutic strategy against gemcitabine-resistant pancreatic cancers.

Eftozanermin alfa

As previously noted, a major limitation of both first- and second-generation rhTRAIL, receptor-specific mAbs, and TRAIL derivatives is their inability to induce efficient lower- and higher-order receptor clustering, leading to reduced apoptotic signaling. In the case of mAbs, it has been shown that due to the bivalent structure of antibodies, additional cross-linking of the Fc region of antibodies to Fcγ receptors (FcγR) is necessary for lower-order clustering and trimerization to occur [33,52]. However, IgG is known to compete with these antibodies for this interaction. Mouse models have very low levels of IgG as compared with cancer patients, providing a likely explanation for the discrepancy between preclinical studies and clinical trials of receptor-specific mAbs [33]. With this in mind, a new family of TRAIL-R agonists was produced.

APG350 is a first-in-class prototype of Eftozanermin alfa (ABBV-621). It was designed to increase receptor clustering while remaining independent of FcγR cross-linking. The molecule is composed of two single chain TRAIL trimers dimerized via fusion to the Fc region of IgG1 and is therefore able to bind to six TRAIL receptors (both DR4 and DR5) with limited FcγR binding capability [33]. APG350 was shown to have an enhanced lower-order clustering efficiency as compared with TRAIL-R mAbs, and because it can simultaneously bind two death receptor trimers, it has a greater ability to induce higher-order receptor clustering as compared with rhTRAIL and its derivatives [53,54]. Moreover, the later developed Eftozanermin alfa has an estimated human half-life of ∼2 days [54]. Thus, Eftozanermin alfa clearly improves upon the agonistic ability and pharmacokinetic profile of first-generation TRAIL therapeutics and has consequently progressed into clinical trials (Table 1). A first-in-human study showed Eftozanermin alfa was well-tolerated in patients with advanced solid tumors and hematological malignancies alone and in combination with venetoclax or chemotherapeutics [55,56] (NCT03082209). The results of dose-escalation and dose-optimization cohorts of patients with advanced solid tumors receiving Eftozanermin alfa monotherapy showed partial responses according to RECIST in three patients (two with CRC and one with pancreatic cancer) among the 105 patients participating in the clinical trial (PMID 35467243). Pharmacodynamics studies demonstrated saturation of Eftozanermin alfa binding sites on the TRIAL receptors and increased levels of M30 and M65 markers of apoptosis in the peripheral blood. Analysis of paired tumor specimens collected during the clinical trial also showed increase tumor infiltration of immune cells including CD4+ T cells in post-treatment biopsies compared with baseline tumor specimens as well as increased PARP cleavage and down-regulation of MEK/Erk/AKT pathway. Another phase 1B study is currently recruiting patients with relapsed or refractory multiple myeloma (NCT04570631).

Table 1
Ongoing clinical trials involving TRAIL-based therapeutics
Cancer typeCombined withPhaseStatusClinicaltrials.gov ID
Eftozanermin alfa 
Multiple myeloma – Recruiting NCT04570631 
Advanced solid tumors, hematological malignancies – Completed NCT03082209 
SCB-313 
Peritoneal malignancies – Completed NCT03443674 
Malignant pleural effusions – Completed NCT03869697 
Peritoneal carcinomatosis – Recruiting NCT04047771 
Malignant ascites – Completed NCT04051112 
Malignant pleural effusions – Completed NCT04123886 
GEN1029 
Solid tumors – 1/2 Terminated NCT03576131 
INBRX-109 
Metastatic solid tumors, including sarcomas – Recruiting NCT03715933 
Conventional chondrosarcoma – Recruiting NCT04950075 
IGM-8444 
Solid tumors Numerous agents Recruiting NCT04553692 
ONC201 
Neuroendocrine – Active, not recruiting NCT03034200 
Acute myeloid leukemia – Recruiting NCT03932643 
Leukemia ­– 1/2 Recruiting NCT02392572 
Multiple myeloma – 1/2 Active, not recruiting NCT02863991 
Metastatic colorectal cancer – 1/2 Terminated NCT03791398 
Ovarian cancer Paclitaxel Recruiting NCT04055649 
Glioma – Active, not recruiting NCT03295396 
H3 K27M Glioma – Recruiting NCT03416530 
Recurrent GBM, H3 K27M Glioma, midline glioma – Active, not recruiting NCT02525692 
Midline glioma Radiation, Paxalisib, or Panobinostat Recruiting NCT05009992 
Cancer typeCombined withPhaseStatusClinicaltrials.gov ID
Eftozanermin alfa 
Multiple myeloma – Recruiting NCT04570631 
Advanced solid tumors, hematological malignancies – Completed NCT03082209 
SCB-313 
Peritoneal malignancies – Completed NCT03443674 
Malignant pleural effusions – Completed NCT03869697 
Peritoneal carcinomatosis – Recruiting NCT04047771 
Malignant ascites – Completed NCT04051112 
Malignant pleural effusions – Completed NCT04123886 
GEN1029 
Solid tumors – 1/2 Terminated NCT03576131 
INBRX-109 
Metastatic solid tumors, including sarcomas – Recruiting NCT03715933 
Conventional chondrosarcoma – Recruiting NCT04950075 
IGM-8444 
Solid tumors Numerous agents Recruiting NCT04553692 
ONC201 
Neuroendocrine – Active, not recruiting NCT03034200 
Acute myeloid leukemia – Recruiting NCT03932643 
Leukemia ­– 1/2 Recruiting NCT02392572 
Multiple myeloma – 1/2 Active, not recruiting NCT02863991 
Metastatic colorectal cancer – 1/2 Terminated NCT03791398 
Ovarian cancer Paclitaxel Recruiting NCT04055649 
Glioma – Active, not recruiting NCT03295396 
H3 K27M Glioma – Recruiting NCT03416530 
Recurrent GBM, H3 K27M Glioma, midline glioma – Active, not recruiting NCT02525692 
Midline glioma Radiation, Paxalisib, or Panobinostat Recruiting NCT05009992 

Modified TRAIL-R antibodies

Besides TRAIL derivatives, modified TRAIL-R mAbs are another class of second-generation therapeutics. Unlike their bivalent first-generation counterparts which are unable to induce efficient receptor trimerization, these antibodies are multivalent, which allows them to bind more than just two receptors, thereby enhancing receptor clustering.

GEN1029 (HexaBody-DR5/DR5) is novel a mixture of two DR5-specific IgG1 mAbs, each targeting two distinct DR5 epitopes and containing E430G mutations that enhance antibody hexamerization via Fc-Fc interactions [57–59]. This unique structure results in the ability to induce lower- and higher-order receptor clustering in an FcγR-independent manner. GEN1029 has shown potent antitumor activity in preclinical models in a variety of solid and hematological malignancies [59,60]. A two-part safety trial consisting of a first-in-human dose-escalating phase 1 and an expansion phase 2A for a variety of solid tumors was recently terminated by its sponsor, Genmab (Copenhagen, Denmark), for unspecified reasons (NCT03576131) (Table 1).

INBRX-109 (Inhibrx, La Jolla, CA, U.S.A.) is another multivalent DR5 agonistic antibody. Previous attempts to produce multivalent antibodies led to the realization that excessive receptor clustering can cause hepatotoxicity (as shown by the terminated phase 1 trial for TAS266, a DR5-targeting tetravalent nanobody [61]). INBRX-109 was engineered to avoid hepatotoxicity by limiting its valency to four. Preclinical studies indeed showed that INBRX-109 maintained superior induction of apoptosis in cancer cells while sparing human hepatocytes. INBRX-109 entered an ongoing phase 1 clinical study to evaluate its single agent efficacy in subjects with locally advanced or metastatic tumors (NCT04950075). The drug was shown to be well-tolerated, which led to the FDA granting it orphan drug designation (ODD) in 2021 as well as a phase 2 study of INBRX-109 in conventional chondrosarcoma (NCT03715933) (Table 1).

Finally, IGM-8444 is an IgG DR5 antibody. With 10 binding units, it is capable of binding multiple receptors at once, facilitating receptor clustering [62]. IGM-8444 was found to induce apoptosis in a broad panel of solid and hematological malignancies [62]. A phase I study is currently recruiting patients to test IGM-8444 alone and in combination in subjects with cancer (NCT04553692).

Combination therapies

Second-generation approaches have clearly improved upon first-generation therapeutics, with larger, multivalent molecules enhancing the pharmacokinetic profiles and agonistic abilities of rhTRAIL and TRAIL-R mAbs. However, it is established that resistance to monotherapy with these TRAs is inherent or acquired [9,38,63,64]. Combinations of TRAs with chemotherapeutics and TRAIL-sensitizers are being explored to help overcome this resistance.

TRAs have been tested in combination with chemotherapeutics including gemcitabine, cisplatin, carboplatin, irinotecan, 5-FU, oxaliplatin, paclitaxel, capecitabine and doxorubicin. In the case of rhTRAIL, in a phase 1B study of patients with CRC, dulanermin was evaluated in combination with FOLFOX6 regimen plus bevacizumab. Study results showed that the combination was well-tolerated. A partial response (PR) was observed in 57% of patients, stable disease was observed in 30% of patients, and progressive disease (PD) was observed in the remaining 13% [65]. Dulanermin was also evaluated in combination with paclitaxel, carboplatin and bevacizumab in a randomized phase 2 study in patients with advanced non-small-cell lung cancer (NSCLC). The addition of dulanermin to these therapeutic regimes was tolerated but did not significantly improve patient outcomes [66]. In a phase 3 study in patients with advanced NSCLC, the combination of dulanermin plus vinorelbine and cisplatin was evaluated. Here, researchers noted that the addition of dulanermin to the vinorelbine and cisplatin regime significantly improved overall response rate (ORR) and progression-free survival (PFS) [67]. Finally, in a phase 1b open-label study, a patient with BRAF-mutant metastatic colon cancer received dulanermin and FOLFIRI. The patient maintained stable disease through 25 doses administered every two weeks, a period which extended beyond the median overall survival (OS) for patients with this disease [68]. An ongoing phase 1 trial with Eftozanermin alfa is investigating the combination with FOLFIRI and bevacizumab for treatment of patients with KRAS-mutant CRC (NCT03082209).

In the case of TRAIL-R antibodies, in a phase 2 study in chemotherapy-naïve patients with unresectable or metastatic pancreatic cancer, the combination of tigatuzumab with gemcitabine was well-tolerated. Here, an ORR of 13.1% was reported while 45.9% of patients had stable disease and 23% of patients had progressive disease [71]. LBY135 has been evaluated in combination with capecitabine in a phase 1 clinical trial of patients with advanced solid tumors. Here, there were some signs of clinical activity and the mAb was shown to be well-tolerated, once again [72]. Numerous mechanisms have been proposed to explain chemotherapy-induced TRA-sensitization, including receptor up-regulation and suppression of anti-apoptotic proteins [38]. Nonetheless, this sensitization remains limited due to the previously mentioned agonistic and pharmacokinetic issues associated with rhTRAIL and TRAIL-R mAbs.

We discovered a potent synergy between rhTRAIL and sorafenib through effects on NF-kB and Mcl-1 [69]. This strategy was pursued in the clinic through a randomized phase II study using mapatumumab in combination with sorafenib in hepatocellular cancer, but with limited efficacy [70].

TRAs have also been tested in combination with apoptotic pathway agents that act as TRAIL-sensitizers. The rationale is that cancer cells often become resistant to TRAIL therapy through cross-talk between the extrinsic and intrinsic pathways and by up-regulating anti-apoptotic proteins [73]. For instance, c-FLIP is known to inhibit DISC formation by competing with caspase-8/-10 for binding to FADD. FLIP is often overexpressed in cancer [19], leading to the development of FLIP inhibitors and siRNAs that silence the protein [74]. Such approaches were shown to sensitize cancer cells to TRAIL therapy [75]. In addition, XIAP interacts with and inhibits caspases, and its high expression is associated with TRAIL resistance. Smac mimetics have been developed to mimic the binding site of the XIAP antagonist Smac and have shown promising preclinical activity [76]. siRNA silencing of XIAP has been shown to effectively sensitize cells to TRAIL [77]. Finally, overexpression of BCL-2 family anti-apoptotic proteins, including BCL-2, Mcl-1, and Bcl-xl, are associated with TRAIL resistance [78]. BH3-mimetics such as ABT-263, ABT-199 and ABT-737 bind to and inhibit one or more of these proteins and have also been shown to sensitize cells to TRAIL therapy [54,79,80].

Regardless of the improvements made by combination therapies, resistance continues to be a limiting factor for some receptor agonist-based therapies, as cancer cells often up-regulate anti-apoptotic proteins and down-regulate TRAIL and its receptors [9]. Thus, agents that increase the expression of TRAIL and its receptors have some therapeutic potential. One such agent is TIC10/ONC201 [81].

Originally discovered as TRAIL-inducing compound 10 (TIC10), ONC201 is a first-in-class molecule and a founding member of the imipridone small molecule family. ONC201 acts directly to agonize ClpP, which leads to the activation of the integrated stress response (ISR) and downstream ATF4 and CHOP-mediated up-regulation of DR5 [82]. Earlier work has shown that ONC201 also inhibits Akt and ERK, which allows for the nuclear translocation of Foxo3a and subsequent up-regulation of TRAIL [83]. Thus, ONC201 up-regulates TRAIL in all cells and DR5 selectively in tumor cells. This up-regulation has been shown to translate to increased expression of TRAIL and DR5 at the cell surface which leads to downstream induction of apoptosis [84,85]. In addition, ONC201 inhibits cancer stem cells and has documented immunostimulatory effects [86,87]. Thus, ONC201 induces tumor-selective apoptosis and promotes immune function through several mechanisms, many of which are still being studied.

ONC201 has shown preclinical activity as a single agent against a wide variety of solid and hematological [88] tumors, including lung [89], breast [90], pancreatic [91], ovarian [92], colorectal [81], prostate [81], hepatocellular [93], leukemia [94] and lymphoma [95]. Of note, ONC201 has shown efficacy against neuroendocrine tumors and brain tumors [96,97]. Such activity can be attributed to ONC201's ability to cross the blood-brain barrier as well as its ability to antagonize DRD2/3. Indeed, ONC201's activity against neuroendocrine tumors including pheochromocytomas and paragangliomas is correlated with their high expression of dopamine receptor type II [97]. Treatment with ONC201 reduced tumor growth in multiple glioma xenograft models, and a single dose doubled the overall survival of mice with an intracranial xenograft of human GBM [81,98]. ONC201 has also shown preclinical anti-metastatic activity in various tissue types, including breast, CRC, and endometrial cancers [99]. This is explained by the fact that ONC201 inhibits cell adhesion, migration, and invasion in a TRAIL-dependent manner [99,100].

The preclinical outcomes of ONC201 led to its approval by the FDA for testing in phase 1 trials for patients with various cancers. ONC201 has been evaluated in 16 clinical trials as a single agent (Table 1). The first-in-human study found ONC201 was well-tolerated in patients with advanced solid tumors. Although objective responses were not achieved by RECIST criteria, a subset of the patients experienced tumor regression, including one patient with chemotherapy-resistant endometrial cancer [101]. More recently, a phase 2 study of ONC201 monotherapy in patients with recurrent GBM once again found that ONC201 was well-tolerated. Of note, this study was the second report of a patient with a H3K27M mutation achieving a complete regression of enhancing multifocal lesions that was maintained for >1.5 years. It is noteworthy that the more potent ONC201 analog ONC212 is active against H3K27M-mutated DIPG cells in preclinical studies, but this analog has not been reported to antagonize dopamine receptors [102].

Although ONC201 has shown some success as a single agent, its activity is limited by resistance-inducing factors such as PI3K/AKT/mTOR upregulation [103] and insufficient TRAIL induction [104,105]. This, along with its multifaceted mechanism of action makes it a good candidate for combination therapy. There is extensive preclinical work showing synergism between ONC201 and radiation [106], chemotherapeutics, targeted therapies [107], and immune-checkpoint agents [96,99]. Most relevant to this review is the combination between ONC201 and TRAs. ONC201 was shown to be ineffective as a single agent in triple-negative breast cancer (TNBC) due to insufficient TRAIL induction [104,105]. TRAIL itself has activity only in a subset of TNBC patients. Combination therapy between ONC201 and rhTRAIL produced a potent apoptotic effect in breast cancer cells which was replicated in other cancer types [104]. A similar effect was seen in non-TNBC cells with the combination between lexatumumab and ONC201 [108]. Finally, the combination of ONC201 with TLY012 was shown to produce a synergistic apoptotic effect both in vitro and in vivo in pancreatic cancer models [109]. Such combinations have potential but remain to be tested in clinical trials. Meanwhile, clinical trials have been initiated using combinations between ONC201 and radiation (NCT05009992) or chemotherapeutics (NCT04055649) (Table 1). Preclinical data demonstrate synergy between ONC201 or other imipridones and EZH2 or HDAC inhibitors and this has been correlated with H3K27 acetylation [102,110].

Unlike other members of the TNF-family, TRAIL possesses the ability to selectively target cancer cells. This exciting realization led to several decades of intense research focused on the development of TRAIL receptor-based therapies. However, clinical studies have demonstrated that such therapies have failed to meet their expectations, with many possessing an inability to induce lower- or higher-order receptor clustering, poor pharmacokinetic profiles including short half-lives, and rapidly built resistance. While novel TRAs are being developed to specifically target these shortcomings and have shown promise in the clinic, it has become clear that a more imaginative approach must be taken to advance the field. The discovery of ONC201/TIC10 provides such an approach. Unlike TRAs, ONC201 acts independently of receptor binding and instead induces TRAIL and DR5 at the transcriptional level. This unique mechanism of action represents a new generation of TRAIL-pathway based approaches. ONC201 has shown promising preclinical and clinical results. Nevertheless, it must be recognized that the nature of cancer is complex, with tumor heterogeneity, evolution, and resistance to anti-cancer therapy acting as major limiting factors to single-drug therapies. Thus, further exploration of ONC201 and other TRAIL-based approaches is needed to address such obstacles.

  • The TRAIL pathway is a powerful innate immune system whose therapeutic potential is yet to be realized.

  • It is likely that harnessing the TRAIL pathway in cancer therapy alone or in combination with other cancer therapeutic modalities holds promise.

  • Mechanistic studies with first- and second-generation TRAIL receptor agonists will help inform translational directions and combinatorial cancer therapeutics.

W.S.E.-D. is a co-founder of Oncoceutics, Inc., a subsidiary of Chimerix. Dr. El-Deiry has disclosed his relationship with Oncoceutics/Chimerix and potential conflict of interest to his academic institution/employer and is fully compliant with NIH and institutional policy that is managing this potential conflict of interest.

W.S.E-D. is an American Cancer Society Research Professor and is supported by the Mencoff Family University Professorship at Brown University. This work was supported by an NIH grant (CA173453) and by a sponsored research agreement from D&D Pharmatech to W.S.E-D.

CRC

colorectal cancer

DISC

death-inducing signaling complex

FADD

Fas-associated protein with death domain

GBM

glioblastoma

ISR

integrated stress response

IZ

isoleucine zipper

LZ

leucine zipper

NSCLC

non-small-cell lung cancer

ODD

Orphan Drug Designation

ORR

overall response rate

PEG

polyethylene glycol

RECIST

Response Evaluation Criteria in Solid Tumors

TIC10

TRAIL-inducing compound 10

TNBC

triple-negative breast cancer

TNC

tenascin-C

TRAIL

tumor necrosis factor-related apoptosis-inducing ligand

TRAs

TRAIL receptor agonists

rhTRAIL

recombinant human TRAIL

1
Hanahan
,
D.
and
Weinberg
,
R.A.
(
2000
)
The hallmarks of cancer
.
Cell
100
,
57
70
2
Carneiro
,
B.A.
and
El-Deiry
,
W.S.
(
2020
)
Targeting apoptosis in cancer therapy
.
Nat. Rev. Clin. Oncol.
17
,
395
417
3
Kerr
,
J.F.
,
Wyllie
,
A.H.
and
Currie
,
A.R.
(
1972
)
Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics
.
Br. J. Cancer
26
,
239
257
4
Igney
,
F.H.
and
Krammer
,
P.H.
(
2002
)
Death and anti-death: tumour resistance to apoptosis
.
Nat. Rev. Cancer
2
,
277
288
5
Elmore
,
S.
(
2007
)
Apoptosis: a review of programmed cell death
.
Toxicol. Pathol.
35
,
495
516
6
Walczak
,
H.
,
Miller
,
R.E.
,
Ariail
,
K.
,
Gliniak
,
B.
,
Griffith
,
T.S.
,
Kubin
,
M.
et al (
1999
)
Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo
.
Nat. Med.
5
,
157
163
7
Pitti
,
R.M.
,
Marsters
,
S.A.
,
Ruppert
,
S.
,
Donahue
,
C.J.
,
Moore
,
A.
,
and Ashkenazi
,
A.
(
1996
)
Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family
.
J. Biol. Chem.
271
,
12687
12690
8
Wu
,
G.S.
,
Burns
,
T.F.
,
McDonald
,
E.R.
, 3rd
,
Jiang
,
W.
,
Meng
,
R.
,
Krantz
,
I.D.
et al (
1997
)
KILLER/DR5 is a DNA damage–inducible p53–regulated death receptor gene
.
Nat. Genet.
17
,
141
143
9
Yuan
,
X.
,
Gajan
,
A.
,
Chu
,
Q.
,
Xiong
,
H.
,
Wu
,
K.
and
Wu
,
G.S.
(
2018
)
Developing TRAIL/TRAIL death receptor-based cancer therapies
.
Cancer Metastasis Rev.
37
,
733
748
10
Wang
,
S.
and
El-Deiry
,
W.S.
(
2003
)
TRAIL and apoptosis induction by TNF-family death receptors
.
Oncogene
22
,
8628
8633
11
Meng
,
R.D.
,
McDonald
, III,
E.R.
,
Sheikh
,
M.S.
,
Fornace
, Jr,
A.J.
and
El-Deiry
,
W.S.
(
2000
)
The TRAIL decoy receptor TRUNDD (DcR2, TRAIL-R4) is induced by adenovirus-p53 overexpression and can delay TRAIL-, p53-, and KILLER/DR5-dependent colon cancer apoptosis
.
Mol. Ther.
1
,
130
144
12
Pan
,
G.
,
Ni
,
J.
,
Wei
,
Y.F.
,
Yu
,
G.
,
Gentz
,
R.
and
Dixit
,
V.M.
(
1997
)
An antagonist decoy receptor and a death domain-containing receptor for TRAIL
.
Science
277
,
815
818
13
Sanlioglu
,
A.D.
,
Dirice
,
E.
,
Aydin
,
C.
,
Erin
,
N.
,
Koksoy
,
S.
and
Sanlioglu
,
S.
(
2005
)
Surface TRAIL decoy receptor-4 expression is correlated with TRAIL resistance in MCF7 breast cancer cells
.
BMC Cancer
5
,
54
14
Wu
,
G.S.
(
2009
)
TRAIL as a target in anti-cancer therapy
.
Cancer Lett.
285
,
1
5
15
Naval
,
J.
,
de Miguel
,
D.
,
Gallego-Lleyda
,
A.
,
Anel
,
A.
and
Martinez-Lostao
,
L.
(
2019
)
Importance of TRAIL molecular anatomy in receptor oligomerization and signaling. implications for cancer therapy
.
Cancers (Basel)
11
,
444
16
Huang
,
K.
,
Zhang
,
J.
,
O'Neill
,
K.L.
,
Gurumurthy
,
C.B.
,
Quadros
,
R.M.
,
Tu
,
Y.
et al (
2016
)
Cleavage by caspase 8 and mitochondrial membrane association activate the BH3-only protein bid during TRAIL-induced apoptosis
.
J. Biol. Chem.
291
,
11843
11851
17
Ozoren
,
N.
and
El-Deiry
,
W.S.
(
2002
)
Defining characteristics of types I and II apoptotic cells in response to TRAIL
.
Neoplasia
4
,
551
557
18
Werneburg
,
N.W.
,
Bronk
,
S.F.
,
Guicciardi
,
M.E.
,
Thomas
,
L.
,
Dikeakos
,
J.D.
,
Thomas
,
G.
et al (
2012
)
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) protein-induced lysosomal translocation of proapoptotic effectors is mediated by phosphofurin acidic cluster sorting protein-2 (PACS-2)
.
J. Biol. Chem.
287
,
24427
24437
19
Ricci
,
M.S.
,
Jin
,
Z.
,
Dews
,
M.
,
Yu
,
D.
,
Thomas-Tikhonenko
,
A.
,
Dicker
,
D.T.
et al (
2004
)
Direct repression of FLIP expression by c-myc is a major determinant of TRAIL sensitivity
.
Mol. Cell. Biol.
24
,
8541
8555
20
Kuijlen
,
J.M.
,
Mooij
,
J.J.
,
Platteel
,
I.
,
Hoving
,
E.W.
,
van der Graaf
,
W.T.
,
Span
,
M.M.
et al (
2006
)
TRAIL-receptor expression is an independent prognostic factor for survival in patients with a primary glioblastoma multiforme
.
J. Neurooncol.
78
,
161
171
21
Finnberg
,
N.
,
Klein-Szanto
,
A.J.
and
El-Deiry
,
W.S.
(
2008
)
TRAIL-R deficiency in mice promotes susceptibility to chronic inflammation and tumorigenesis
.
J. Clin. Invest.
118
,
111
123
22
Herbst
,
R.S.
,
Eckhardt
,
S.G.
,
Kurzrock
,
R.
,
Ebbinghaus
,
S.
,
O'Dwyer
,
P.J.
,
Gordon
,
M.S.
et al (
2010
)
Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cancer
.
J. Clin. Oncol.
28
,
2839
2846
23
Soria
,
J.C.
,
Smit
,
E.
,
Khayat
,
D.
,
Besse
,
B.
,
Yang
,
X.
,
Hsu
,
C.P.
et al (
2010
)
Phase 1b study of dulanermin (recombinant human Apo2L/TRAIL) in combination with paclitaxel, carboplatin, and bevacizumab in patients with advanced non-squamous non-small-cell lung cancer
.
J. Clin. Oncol.
28
,
1527
1533
24
Graves
,
J.D.
,
Kordich
,
J.J.
,
Huang
,
T.H.
,
Piasecki
,
J.
,
Bush
,
T.L.
,
Sullivan
,
T.
et al (
2014
)
Apo2l/TRAIL and the death receptor 5 agonist antibody AMG 655 cooperate to promote receptor clustering and antitumor activity
.
Cancer Cell
26
,
177
189
25
von Karstedt
,
S.
,
Montinaro
,
A.
and
Walczak
,
H.
(
2017
)
Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy
.
Nat. Rev. Cancer
17
,
352
366
26
Adams
,
C.
,
Totpal
,
K.
,
Lawrence
,
D.
,
Marsters
,
S.
,
Pitti
,
R.
,
Yee
,
S.
et al (
2008
)
Structural and functional analysis of the interaction between the agonistic monoclonal antibody Apomab and the proapoptotic receptor DR5
.
Cell Death Differ.
15
,
751
761
27
Plummer
,
R.
,
Attard
,
G.
,
Pacey
,
S.
,
Li
,
L.
,
Razak
,
A.
,
Perrett
,
R.
et al (
2007
)
Phase 1 and pharmacokinetic study of lexatumumab in patients with advanced cancers
.
Clin. Cancer Res.
13
,
6187
6194
28
Herbst
,
R.S.
,
Kurzrock
,
R.
,
Hong
,
D.S.
,
Valdivieso
,
M.
,
Hsu
,
C.P.
,
Goyal
,
L.
et al (
2010
)
A first-in-human study of conatumumab in adult patients with advanced solid tumors
.
Clin. Cancer Res.
16
,
5883
5891
29
Camidge
,
D.R.
,
Herbst
,
R.S.
,
Gordon
,
M.S.
,
Eckhardt
,
S.G.
,
Kurzrock
,
R.
,
Durbin
,
B.
et al (
2010
)
A phase I safety and pharmacokinetic study of the death receptor 5 agonistic antibody PRO95780 in patients with advanced malignancies
.
Clin. Cancer Res.
16
,
1256
1263
30
Forero-Torres
,
A.
,
Shah
,
J.
,
Wood
,
T.
,
Posey
,
J.
,
Carlisle
,
R.
,
Copigneaux
,
C.
et al (
2010
)
Phase I trial of weekly tigatuzumab, an agonistic humanized monoclonal antibody targeting death receptor 5 (DR5)
.
Cancer Biother. Radiopharm.
25
,
13
19
31
Hotte
,
S.J.
,
Hirte
,
H.W.
,
Chen
,
E.X.
,
Siu
,
L.L.
,
Le
,
L.H.
,
Corey
,
A.
et al (
2008
)
A phase 1 study of mapatumumab (fully human monoclonal antibody to TRAIL-R1) in patients with advanced solid malignancies
.
Clin. Cancer Res.
14
,
3450
3455
32
Allen
,
J.E.
,
Ferrini
,
R.
,
Dicker
,
D.T.
,
Batzer
,
G.
,
Chen
,
E.
,
Oltean
,
D.I.
et al (
2012
)
Targeting TRAIL death receptor 4 with trivalent DR4 Atrimer complexes
.
Mol. Cancer Ther.
11
,
2087
2095
33
Gieffers
,
C.
,
Kluge
,
M.
,
Merz
,
C.
,
Sykora
,
J.
,
Thiemann
,
M.
,
Schaal
,
R.
et al (
2013
)
APG350 induces superior clustering of TRAIL receptors and shows therapeutic antitumor efficacy independent of cross-linking via fcgamma receptors
.
Mol. Cancer Ther.
12
,
2735
2747
34
Thapa
,
B.
,
Kc
,
R.
and
Uludag
,
H.
(
2020
)
TRAIL therapy and prospective developments for cancer treatment
.
J. Control. Release
326
,
335
349
35
Ashkenazi
,
A.
,
Pai
,
R.C.
,
Fong
,
S.
,
Leung
,
S.
,
Lawrence
,
D.A.
,
Marsters
,
S.A.
et al (
1999
)
Safety and antitumor activity of recombinant soluble Apo2 ligand
.
J. Clin. Invest.
104
,
155
162
36
Pollack
,
I.F.
,
Erff
,
M.
and
Ashkenazi
,
A.
(
2001
)
Direct stimulation of apoptotic signaling by soluble Apo2l/tumor necrosis factor-related apoptosis-inducing ligand leads to selective killing of glioma cells
.
Clin. Cancer Res.
7
,
1362
1369
37
Tolcher
,
A.W.
,
Mita
,
M.
,
Meropol
,
N.J.
,
von Mehren
,
M.
,
Patnaik
,
A.
,
Padavic
,
K.
et al (
2007
)
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.
25
,
1390
1395
38
Lemke
,
J.
,
von Karstedt
,
S.
,
Zinngrebe
,
J.
and
Walczak
,
H.
(
2014
)
Getting TRAIL back on track for cancer therapy
.
Cell Death Differ.
21
,
1350
1364
39
Liu
,
H.
,
Su
,
D.
,
Zhang
,
J.
,
Ge
,
S.
,
Li
,
Y.
,
Wang
,
F.
et al (
2017
)
Improvement of pharmacokinetic profile of TRAIL via trimer-tag enhances its antitumor activity in vivo
.
Sci. Rep.
7
,
8953
40
de Miguel
,
D.
,
Lemke
,
J.
,
Anel
,
A.
,
Walczak
,
H.
and
Martinez-Lostao
,
L.
(
2016
)
Onto better TRAILs for cancer treatment
.
Cell Death Differ.
23
,
733
747
41
Ganten
,
T.M.
,
Koschny
,
R.
,
Sykora
,
J.
,
Schulze-Bergkamen
,
H.
,
Büchler
,
P.
,
Haas
,
T.L.
et al (
2006
)
Preclinical differentiation between apparently safe and potentially hepatotoxic applications of TRAIL either alone or in combination with chemotherapeutic drugs
.
Clin. Cancer Res.
12
,
2640
2646
42
Berg
,
D.
,
Lehne
,
M.
,
Müller
,
N.
,
Siegmund
,
D.
,
Münkel
,
S.
,
Sebald
,
W.
et al (
2007
)
Enforced covalent trimerization increases the activity of the TNF ligand family members TRAIL and CD95L
.
Cell Death Differ.
14
,
2021
2034
43
Jo
,
M.
,
Kim
,
T.H.
,
Seol
,
D.W.
,
Esplen
,
J.E.
,
Dorko
,
K.
,
Billiar
,
T.R.
et al (
2000
)
Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis-inducing ligand
.
Nat. Med.
6
,
564
567
44
Harris
,
J.M.
and
Chess
,
R.B.
(
2003
)
Effect of pegylation on pharmaceuticals
.
Nat. Rev. Drug Discov.
2
,
214
221
45
Crawford
,
J.
(
2002
)
Clinical benefits of pegylated proteins in oncology
.
Cancer Treat. Rev.
28
,
1
2
46
Chae
,
S.Y.
,
Kim
,
T.H.
,
Park
,
K.
,
Jin
,
C.H.
,
Son
,
S.
,
Lee
,
S.
et al (
2010
)
Improved antitumor activity and tumor targeting of NH(2)-terminal-specific PEGylated tumor necrosis factor-related apoptosis-inducing ligand
.
Mol. Cancer Ther.
9
,
1719
1729
47
Kim
,
T.H.
,
Youn
,
Y.S.
,
Jiang
,
H.H.
,
Lee
,
S.
,
Chen
,
X.
and
Lee
,
K.C.
(
2011
)
PEGylated TNF-related apoptosis-inducing ligand (TRAIL) analogues: pharmacokinetics and antitumor effects
.
Bioconjug. Chem.
22
,
1631
1637
48
Park
,
J.S.
,
Oh
,
Y.
,
Park
,
Y.J.
,
Park
,
O.
,
Yang
,
H.
,
Slania
,
S.
et al (
2019
)
Targeting of dermal myofibroblasts through death receptor 5 arrests fibrosis in mouse models of scleroderma
.
Nat. Commun.
10
,
1128
49
Oh
,
Y.
,
Park
,
O.
,
Swierczewska
,
M.
,
Hamilton
,
J.P.
,
Park
,
J.S.
,
Kim
,
T.H.
et al (
2016
)
Systemic PEGylated TRAIL treatment ameliorates liver cirrhosis in rats by eliminating activated hepatic stellate cells
.
Hepatology
64
,
209
223
50
Huang
,
M.
,
Zhu
,
H.
,
Yi
,
C.
,
Yan
,
J.
,
Wei
,
L.
,
Yang
,
X.
et al (
2018
)
A novel TRAIL mutant-TRAIL-Mu3 enhances the antitumor effects by the increased affinity and the up-expression of DR5 in pancreatic cancer
.
Cancer Chemother. Pharmacol.
82
,
829
838
51
Huang
,
M.
,
Yi
,
C.
,
Huang
,
X.Z.
,
Yan
,
J.
,
Wei
,
L.J.
,
Tang
,
W.J.
et al (
2021
)
Recombinant protein TRAIL-Mu3 enhances the antitumor effects in pancreatic cancer cells by strengthening the apoptotic signaling pathway
.
Oncol. Lett.
21
,
438
52
Wilson
,
N.S.
,
Yang
,
B.
,
Yang
,
A.
,
Loeser
,
S.
,
Marsters
,
S.
,
Lawrence
,
D.
et al (
2011
)
An fcgamma receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells
.
Cancer Cell
19
,
101
113
53
Legler
,
K.
,
Hauser
,
C.
,
Egberts
,
J.H.
,
Willms
,
A.
,
Heneweer
,
C.
,
Boretius
,
S.
et al (
2018
)
The novel TRAIL-receptor agonist APG350 exerts superior therapeutic activity in pancreatic cancer cells
.
Cell Death Dis.
9
,
445
54
Phillips
,
D.C.
,
Buchanan
,
F.G.
,
Cheng
,
D.
,
Solomon
,
L.R.
,
Xiao
,
Y.
,
Xue
,
J.
et al (
2021
)
Hexavalent TRAIL fusion protein eftozanermin alfa optimally clusters apoptosis-inducing TRAIL receptors to induce on-target antitumor activity in solid tumors
.
Cancer Res.
81
,
3402
3414
55
LoRusso
,
P.
,
Ratain
,
M.J.
,
Doi
,
T.
,
Rasco
,
D.W.
,
de Jonge
,
M.J.A.
,
Moreno
,
V.
et al (
2022
)
Eftozanermin alfa (ABBV-621) monotherapy in patients with previously treated solid tumors: findings of a phase 1, first-in-human study
.
Invest. New Drugs
40
,
762
772
56
de Jonge
,
M.J.A.
,
Carneiro
,
B.A.
,
Devriese
,
L.
,
Doi
,
T.
,
Penugonda
,
S.
,
Petrich
,
A.M.
et al (
2019
)
First-in-Human study of abbv-621, a TRAIL receptor agonist fusion protein, in patients (Pts) with relapsed/refractory (RR) acute myeloid leukemia (AML) and diffuse large B-cell lymphoma (DLBCL)
.
Blood
134
,
3924
3924
57
Cook
,
E.M.
,
Lindorfer
,
M.A.
,
van der Horst
,
H.
,
Oostindie
,
S.
,
Beurskens
,
F.J.
,
Schuurman
,
J.
et al (
2016
)
Antibodies that efficiently form hexamers upon antigen binding Can induce complement-dependent cytotoxicity under complement-Limiting conditions
.
J. Immunol.
197
,
1762
1775
58
de Jong
,
R.N.
,
Beurskens
,
F.J.
,
Verploegen
,
S.
,
Strumane
,
K.
,
van Kampen
,
M.D.
,
Voorhorst
,
M.
et al (
2016
)
A novel platform for the potentiation of therapeutic antibodies based on antigen-dependent formation of IgG hexamers at the cell surface
.
PLoS Biol.
14
,
e1002344
59
Overdijk
,
M.B.
,
Strumane
,
K.
,
Beurskens
,
F.J.
,
Ortiz Buijsse
,
A.
,
Vermot-Desroches
,
C.
,
Vuillermoz
,
B.S.
et al (
2020
)
Dual epitope targeting and enhanced hexamerization by DR5 antibodies as a novel approach to induce potent antitumor activity through DR5 agonism
.
Mol. Cancer Ther.
19
,
2126
2138
60
van der Horst
,
H.J.
,
Gelderloos
,
A.T.
,
Chamuleau
,
M.E.D.
,
Breij
,
E.C.W.
,
Zweegman
,
S.
,
Nijhof
,
I.S.
et al (
2021
)
Potent preclinical activity of HexaBody-DR5/DR5 in relapsed and/or refractory multiple myeloma
.
Blood Adv.
5
,
2165
2172
61
Papadopoulos
,
K.P.
,
Isaacs
,
R.
,
Bilic
,
S.
,
Kentsch
,
K.
,
Huet
,
H.A.
,
Hofmann
,
M.
et al (
2015
)
Unexpected hepatotoxicity in a phase I study of TAS266, a novel tetravalent agonistic Nanobody(R) targeting the DR5 receptor
.
Cancer Chemother. Pharmacol.
75
,
887
895
62
Wang
,
B.T.
,
Kothambawala
,
T.
,
Wang
,
L.
,
Matthew
,
T.J.
,
Calhoun
,
S.E.
,
Saini
,
A.K.
et al (
2021
)
Multimeric anti-DR5 IgM agonist antibody IGM-8444 is a potent inducer of cancer cell apoptosis and synergizes with chemotherapy and BCL-2 inhibitor ABT-199
.
Mol. Cancer Ther.
20
,
2483
2494
63
Dianat-Moghadam
,
H.
,
Heidarifard
,
M.
,
Mahari
,
A.
,
Shahgolzari
,
M.
,
Keshavarz
,
M.
,
Nouri
,
M.
et al (
2020
)
TRAIL in oncology: from recombinant TRAIL to nano- and self-targeted TRAIL-based therapies
.
Pharmacol. Res.
155
,
104716
64
Wong
,
S.H.M.
,
Kong
,
W.Y.
,
Fang
,
C.M.
,
Loh
,
H.S.
,
Chuah
,
L.H.
,
Abdullah
,
S.
et al (
2019
)
The TRAIL to cancer therapy: hindrances and potential solutions
.
Crit. Rev. Oncol. Hematol.
143
,
81
94
65
Wainberg
,
Z.A.
,
Messersmith
,
W.A.
,
Peddi
,
P.F.
,
Kapp
,
A.V.
,
Ashkenazi
,
A.
,
Royer-Joo
,
S.
et al (
2013
)
A phase 1B study of dulanermin in combination with modified FOLFOX6 plus bevacizumab in patients with metastatic colorectal cancer
.
Clin. Colorectal Cancer
12
,
248
254
66
Soria
,
J.C.
,
Márk
,
Z.
,
Zatloukal
,
P.
,
Szima
,
B.
,
Albert
,
I.
,
Juhász
,
E.
et al (
2011
)
Randomized phase II study of dulanermin in combination with paclitaxel, carboplatin, and bevacizumab in advanced non-small-cell lung cancer
.
J. Clin. Oncol.
29
,
4442
4451
67
Ouyang
,
X.
,
Shi
,
M.
,
Jie
,
F.
,
Bai
,
Y.
,
Shen
,
P.
,
Yu
,
Z.
et al (
2018
)
Phase III study of dulanermin (recombinant human tumor necrosis factor-related apoptosis-inducing ligand/Apo2 ligand) combined with vinorelbine and cisplatin in patients with advanced non-small-cell lung cancer
.
Invest. New Drugs
36
,
315
322
68
Lim
,
B.
,
Scicchitano
,
A.
,
Beachler
,
C.
,
Gusani
,
N.
,
Sarwani
,
N.
,
Yang
,
Z.
et al (
2013
)
FOLFIRI plus dulanermin (rhApo2L/TRAIL) in a patient with BRAF-mutant metastatic colon cancer
.
Cancer Biol. Ther.
14
,
711
719
69
Ricci
,
M.S.
,
Kim
,
S.H.
,
Ogi
,
K.
,
Plastaras
,
J.P.
,
Ling
,
J.
,
Wang
,
W.
et al (
2007
)
Reduction of TRAIL-induced Mcl-1 and cIAP2 by c-Myc or sorafenib sensitizes resistant human cancer cells to TRAIL-induced death
.
Cancer Cell
12
,
66
80
70
Ciuleanu
,
T.
,
Bazin
,
I.
,
Lungulescu
,
D.
,
Miron
,
L.
,
Bondarenko
,
I.
,
Deptala
,
A.
et al (
2016
)
A randomized, double-blind, placebo-controlled phase II study to assess the efficacy and safety of mapatumumab with sorafenib in patients with advanced hepatocellular carcinoma
.
Ann. Oncol.
27
,
680
687
71
Forero-Torres
,
A.
,
Infante
,
J.R.
,
Waterhouse
,
D.
,
Wong
,
L.
,
Vickers
,
S.
,
Arrowsmith
,
E.
et al (
2013
)
Phase 2, multicenter, open-label study of tigatuzumab (CS-1008), a humanized monoclonal antibody targeting death receptor 5, in combination with gemcitabine in chemotherapy-naive patients with unresectable or metastatic pancreatic cancer
.
Cancer Med.
2
,
925
932
72
Sharma
,
S.
,
de Vries
,
E.G.
,
Infante
,
J.R.
,
Oldenhuis
,
C.
,
Chiang
,
L.
,
Goldbrunner
,
S.
,
Bilic
,
M.
et al (
2008
)
Phase I trial of LBY135, a monoclonal antibody agonist to DR5, alone and in combination with capecitabine in advanced solid tumors
.
J. Clin. Oncol.
26
,
3538
3538
73
Allen
,
J.E.
,
Prabhu
,
V.V.
,
Talekar
,
M.
,
van den Heuvel
,
A.P.
,
Lim
,
B.
,
Dicker
,
D.T.
et al (
2015
)
Genetic and pharmacological screens converge in identifying FLIP, BCL2, and IAP proteins as key regulators of sensitivity to the TRAIL-inducing anticancer agent ONC201/TIC10
.
Cancer Res.
75
,
1668
1674
74
Zhang
,
S.
and
El-Deiry
,
W.S.
(
2022
)
Abstract 5854: Therapeutic targeting of p73-activated c-FLIP switches cell fate from growth arrest and survival to apoptosis in p53-deficient cancer cells
.
Cancer Res.
82
,
5854
5854
75
Chawla-Sarkar
,
M.
,
Bae
,
S.I.
,
Reu
,
F.J.
,
Jacobs
,
B.S.
,
Lindner
,
D.J.
and
Borden
,
E.C.
(
2004
)
Down-regulation of Bcl-2, FLIP or IAPs (XIAP and survivin) by siRNAs sensitizes resistant melanoma cells to Apo2L/TRAIL-induced apoptosis
.
Cell Death Differ.
11,
,
915
923
76
Fulda
,
S.
,
Wick
,
W.
,
Weller
,
M.
and
Debatin
,
K.M.
(
2002
)
Smac agonists sensitize for Apo2L/TRAIL- or anticancer drug-induced apoptosis and induce regression of malignant glioma in vivo
.
Nat. Med.
8
,
808
815
77
Spee
,
B.
,
Jonkers
,
M.D.
,
Arends
,
B.
,
Rutteman
,
G.R.
,
Rothuizen
,
J.
and
Penning
,
L.C.
(
2006
)
Specific down-regulation of XIAP with RNA interference enhances the sensitivity of canine tumor cell-lines to TRAIL and doxorubicin
.
Mol. Cancer
5
,
34
78
Kim
,
K.
,
Nakagawa
,
H.
,
Fei
,
P.
,
Rustgi
,
A.K.
and
El-Deiry
,
W.S.
(
2004
)
Targeting Bcl-xL in esophageal squamous cancer to sensitize to chemotherapy plus TRAIL-induced apoptosis while normal epithelial cells are protected by blockade of caspase 9
.
Cell Death Differ.
11
,
583
587
79
Huang
,
S.
and
Sinicrope
,
F.A.
(
2008
)
BH3 mimetic ABT-737 potentiates TRAIL-mediated apoptotic signaling by unsequestering Bim and Bak in human pancreatic cancer cells
.
Cancer Res.
68
,
2944
2951
80
Wang
,
G.
,
Zhan
,
Y.
,
Wang
,
H.
and
Li
,
W.
(
2012
)
ABT-263 sensitizes TRAIL-resistant hepatocarcinoma cells by downregulating the Bcl-2 family of anti-apoptotic protein
.
Cancer Chemother. Pharmacol.
69
,
799
805
81
Allen
,
J.E.
,
Krigsfeld
,
G.
,
Mayes
,
P.A.
,
Patel
,
L.
,
Dicker
,
D.T.
,
Patel
,
A.S.
et al (
2013
)
Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects
.
Sci. Transl. Med.
5
,
171ra117
82
Kline
,
C.L.
,
Van den Heuvel
,
A.P.
,
Allen
,
J.E.
,
Prabhu
,
V.V.
,
Dicker
,
D.T.
and
El-Deiry
,
W.S.
(
2016
)
ONC201 kills solid tumor cells by triggering an integrated stress response dependent on ATF4 activation by specific eIF2alpha kinases
.
Sci. Signal.
9
,
ra18
83
Prabhu
,
V.V.
,
Allen
,
J.E.
,
Dicker
,
D.T.
and
El-Deiry
,
W.S.
(
2015
)
Small-molecule ONC201/TIC10 targets chemotherapy-resistant colorectal cancer stem-like cells in an Akt/Foxo3a/TRAIL-dependent manner
.
Cancer Res.
75
,
1423
1432
84
Ray
,
J.E.
,
Ralff
,
M.D.
,
Jhaveri
,
A.
,
Zhou
,
L.
,
Dicker
,
D.T.
,
Ross
,
E.A.
et al (
2021
)
Antitumorigenic effect of combination treatment with ONC201 and TRAIL in endometrial cancer in vitro and in vivo
.
Cancer Biol. Ther.
22
,
554
563
85
Parker
,
C.S.
,
Zhou
,
L.
,
Prabhu
,
V.
,
Allen
,
J.
and
El-Deiry
,
W.
(
2021
)
Abstract 1044: ONC201 as a novel anti-cancer therapeutic via modulation of inhibitors of apoptosis and up-regulation of DR5 in gastric adenocarcinoma
.
Cancer Res.
81
,
1044
1044
86
Prabhu
,
V.V.
,
Lulla
,
A.R.
,
Madhukar
,
N.S.
,
Ralff
,
M.D.
,
Zhao
,
D.
,
Kline
,
C.L.
B. et al (
2017
)
Cancer stem cell-related gene expression as a potential biomarker of response for first-in-class imipridone ONC201 in solid tumors
.
PLoS ONE
12
,
e0180541
87
Stein
,
M.N.
,
Malhotra
,
J.
,
Tarapore
,
R.S.
,
Malhotra
,
U.
,
Silk
,
A.W.
,
Chan
,
N.
et al (
2019
)
Safety and enhanced immunostimulatory activity of the DRD2 antagonist ONC201 in advanced solid tumor patients with weekly oral administration
.
J. Immunother. Cancer
7
,
136
88
Prabhu
,
V.V.
,
Talekar
,
M.K.
,
Lulla
,
A.R.
,
Kline
,
C.L.B.
,
Zhou
,
L.
,
Hall
,
J.
et al (
2018
)
Single agent and synergistic combinatorial efficacy of first-in-class small molecule imipridone ONC201 in hematological malignancies
.
Cell Cycle
17
,
468
478
89
Feng
,
Y.
,
Zhou
,
J.
,
Li
,
Z.
,
Jiang
,
Y.
and
Zhou
,
Y.
(
2016
)
Small molecular TRAIL inducer ONC201 induces death in lung cancer cells: a preclinical study
.
PLoS ONE
11
,
e0162133
90
Yuan
,
X.
,
Kho
,
D.
,
Xu
,
J.
,
Gajan
,
A.
,
Wu
,
K.
and
Wu
,
G.S.
(
2017
)
ONC201 activates ER stress to inhibit the growth of triple-negative breast cancer cells
.
Oncotarget
8
,
21626
21638
91
Zhang
,
Q.
,
Wang
,
H.
,
Ran
,
L.
,
Zhang
,
Z.
and
Jiang
,
R.
(
2016
)
The preclinical evaluation of TIC10/ONC201 as an anti-pancreatic cancer agent
.
Biochem. Biophys. Res. Commun.
476
,
260
266
92
Baumeister
,
M.D.
,
Küçükkase
,
O.C.
,
Prabhu
,
V.V.
,
Dicker
,
D.T.
,
Allen
,
J.E.
and
El-Deiry
,
W.S.
(
2017
)
Abstract 3212: ONC201 shows efficacy in BRCA-deficient cancer cells and synergy with PARP inhibitors in glioblastoma, breast, prostate, and ovarian cancers
.
Cancer Res.
77
,
3212
3212
93
Cheng
,
L.
,
Liu
,
Y.Y.
,
Lu
,
P.H.
,
Peng
,
Y.
,
Yuan
,
Q.
,
Gu
,
X.S.
et al (
2017
)
Identification of DNA-PKcs as a primary resistance factor of TIC10 in hepatocellular carcinoma cells
.
Oncotarget
8
,
28385
28394
94
Ishizawa
,
J.
,
Zarabi
,
S.F.
,
Davis
,
R.E.
,
Halgas
,
O.
,
Nii
,
T.
,
Jitkova
,
Y.
et al (
2019
)
Mitochondrial ClpP-mediated proteolysis induces selective cancer cell lethality
.
Cancer Cell
35
,
721
737.e729
95
Talekar
,
M.K.
,
Allen
,
J.E.
,
Dicker
,
D.T.
and
El-Deiry
,
W.S.
(
2015
)
ONC201 induces cell death in pediatric non-Hodgkin's lymphoma cells
.
Cell Cycle
14
,
2422
2428
96
Prabhu
,
V.V.
,
Morrow
,
S.
,
Rahman Kawakibi
,
A.
,
Zhou
,
L.
,
Ralff
,
M.
,
Ray
,
J.
et al (
2020
)
ONC201 and imipridones: anti-cancer compounds with clinical efficacy
.
Neoplasia
22
,
725
744
97
Anderson
,
P.M.
,
Trucco
,
M.M.
,
Tarapore
,
R.S.
,
Zahler
,
S.
,
Thomas
,
S.
,
Gortz
,
J.
et al (
2022
)
Phase II study of ONC201 in neuroendocrine tumors including pheochromocytoma-paraganglioma and desmoplastic small round cell tumor
.
Clin. Cancer Res.
28
,
1773
1782
98
Ishida
,
C.T.
,
Zhang
,
Y.
,
Bianchetti
,
E.
,
Shu
,
C.
,
Nguyen
,
T.T.T.
,
Kleiner
,
G.
et al (
2018
)
Metabolic reprogramming by dual AKT/ERK inhibition through imipridones elicits unique vulnerabilities in glioblastoma
.
Clin. Cancer Res.
24
,
5392
5406
99
Wagner
,
J.
,
Kline
,
C.L.
,
Zhou
,
L.
,
Campbell
,
K.S.
,
MacFarlane
,
A.W.
,
Olszanski
,
A.J.
et al (
2018
)
Dose intensification of TRAIL-inducing ONC201 inhibits metastasis and promotes intratumoral NK cell recruitment
.
J. Clin. Invest.
128
,
2325
2338
100
Fang
,
Z.
,
Wang
,
J.
,
Clark
,
L.H.
,
Sun
,
W.
,
Yin
,
Y.
,
Kong
,
W.
et al (
2018
)
ONC201 demonstrates anti-tumorigenic and anti-metastatic activity in uterine serous carcinoma in vitro
.
Am. J. Cancer Res.
8
,
1551
1563
101
Stein
,
M.N.
,
Bertino
,
J.R.
,
Kaufman
,
H.L.
,
Mayer
,
T.
,
Moss
,
R.
,
Silk
,
A.
et al (
2017
)
First-in-human clinical trial of oral ONC201 in patients with refractory solid tumors
.
Clin. Cancer Res.
23
,
4163
4169
102
Borsuk
,
R.
,
Zhou
,
L.
,
Chang
,
W.I.
,
Zhang
,
Y.
,
Sharma
,
A.
,
Prabhu
,
V.V.
et al (
2021
)
Potent preclinical sensitivity to imipridone-based combination therapies in oncohistone H3K27M-mutant diffuse intrinsic pontine glioma is associated with induction of the integrated stress response, TRAIL death receptor DR5, reduced ClpX and apoptosis
.
Am. J. Cancer Res.
11
,
4607
4623
103
Jin
,
Z.Z.
,
Wang
,
W.
,
Fang
,
D.L.
and
Jin
,
Y.J.
(
2016
)
mTOR inhibition sensitizes ONC201-induced anti-colorectal cancer cell activity
.
Biochem. Biophys. Res. Commun.
478
,
1515
1520
104
Ralff
,
M.D.
,
Kline
,
C.L.B.
,
Küçükkase
,
O.C.
,
Wagner
,
J.
,
Lim
,
B.
,
Dicker
,
D.T.
et al (
2017
)
ONC201 demonstrates antitumor effects in both triple-negative and non-triple-negative breast cancers through TRAIL-dependent and TRAIL-independent mechanisms
.
Mol. Cancer Ther.
16
,
1290
1298
105
Ralff
,
M.D.
,
Jhaveri
,
A.
,
Ray
,
J.E.
,
Zhou
,
L.
,
Lev
,
A.
,
Campbell
,
K.S.
et al (
2020
)
TRAIL receptor agonists convert the response of breast cancer cells to ONC201 from anti-proliferative to apoptotic
.
Oncotarget
11
,
3753
3769
106
Zhou
,
L.
,
Wu
,
J.L.
,
Safran
,
H.P.
and
El-Deiry
,
W.S.
(
2022
)
Abstract 5448: Synergistic antitumor effect of ONC201, radiotherapy and temozolomide in glioblastoma mouse orthotopic models
.
Cancer Res.
82
,
5448
5448
107
Cristofano
,
F.R.D.
,
Fong
,
M.
,
Zhou
,
L.
and
El-Deiry
,
W.S.
(
2022
)
Abstract 3709: Synergistic activity of ABT-263 and ONC201 against solid tumor cell lines is associated with suppression of BAG3, Mcl-1, pAkt, and up-regulation of Noxa along with Bax cleavage during apoptosis
.
Cancer Res.
82
,
3709
3709
108
Ralff
,
M.D.
,
Ray
,
J.E.
,
Lev
,
A.
,
Zhou
,
L.
,
Dicker
,
D.T.
and
El-Deiry
,
W.S.
(
2019
)
Abstract 258: Recombinant human TRAIL or a DR5 agonistic antibody convert the response of non-triple negative breast cancer cells to ONC201 from anti-proliferative to apoptotic
.
Cancer Res.
79
,
258
258
109
Jhaveri
,
A.V.
,
Zhou
,
L.
,
Ralff
,
M.D.
,
Lee
,
Y.S.
,
Navaraj
,
A.
,
Carneiro
,
B.A.
et al (
2021
)
Combination of ONC201 and TLY012 induces selective, synergistic apoptosis in vitro and significantly delays PDAC xenograft growth in vivo
.
Cancer Biol. Ther.
22
,
607
618
110
Zhang
,
Y.
,
Zhou
,
L.
,
Safran
,
H.
,
Borsuk
,
R.
,
Lulla
,
R.
,
Tapinos
,
N.
et al (
2021
)
EZH2i EPZ-6438 and HDACi vorinostat synergize with ONC201/TIC10 to activate integrated stress response, DR5, reduce H3K27 methylation, ClpX and promote apoptosis of multiple tumor types including DIPG
.
Neoplasia
23
,
792
810
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY-NC-ND).