Elsevier

Journal of Controlled Release

Volume 326, 10 October 2020, Pages 335-349
Journal of Controlled Release

TRAIL therapy and prospective developments for cancer treatment

https://doi.org/10.1016/j.jconrel.2020.07.013Get rights and content

Abstract

Tumor Necrosis Factor (TNF) Related Apoptosis-Inducing Ligand (TRAIL), an immune cytokine of TNF-family, has received much attention in late 1990s as a potential cancer therapeutics due to its selective ability to induce apoptosis in cancer cells. TRAIL binds to cell surface death receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5) and facilitates formation of death-inducing signaling complex (DISC), eventually activating the p53-independent apoptotic cascade. This unique mechanism makes the TRAIL a potential anticancer therapeutic especially for p53-mutated tumors. However, recombinant human TRAIL protein (rhTRAIL) and TRAIL-R agonist monoclonal antibodies (mAb) failed to exert robust anticancer activities due to inherent and/or acquired resistance, poor pharmacokinetics and weak potencies for apoptosis induction. To get TRAIL back on track as a cancer therapeutic, multiple strategies including protein modification, combinatorial approach and TRAIL gene therapy are being extensively explored. These strategies aim to enhance the half-life and bioavailability of TRAIL and synergize with TRAIL action ultimately sensitizing the resistant and non-responsive cells. We summarize emerging strategies for enhanced TRAIL therapy in this review and cover a wide range of recent technologies that will provide impetus to rejuvenate the TRAIL therapeutics in the clinical realm.

Introduction

Majority of anticancer drugs are expected to inhibit uncontrolled cell proliferation and induce apoptosis. Among the apoptosis pathways, intrinsic or mitochondrial pathway is activated in response to cellular stress and DNA damage by chemotherapy and/or radiotherapy, and involves activation of p53 and release of pro-apoptotic factors from the mitochondria [1,2]. Conventional chemotherapy, however, generally lacks cell selectivity and, moreover, majority of tumors involves mutation(s) of p53 that impede the activation of innate apoptosis mechanism. Alternatively, Tumor Necrosis Factor Related Apoptosis Inducing Ligand (TRAIL, also known as Apo2L), a member of the Tumor Necrosis Factor (TNF) superfamily, induces extrinsic apoptotic pathway, where p53 appears to be dispensable, to induce apoptosis in p53-mutated cancers. TRAIL is a cytokine secreted by majority of normal cells as a part of natural immune reaction and play a significant role in preventing cell proliferation [3]. TRAIL quickly emerged as a promising cancer therapeutic after its discovery because of its ability to induce apoptosis in a wide range of cancer cells while sparing the normal cells [4,5]. Increased interest in TRAIL therapy led to development of recombinant human TRAIL (rhTRAIL) proteins and TRAIL receptor (TRAIL-R1 and TRAIL-R2) agonist monoclonal antibodies (mAb) as potential cancer therapeutics. The promising preclinical results propelled the TRAIL therapy to several clinical trails to test its safety, pharmacokinetics and efficacy in cancer [6,7]. Most studies showed that TRAIL therapy was safe and well-tolerated in patients, but the therapeutic outcomes were insignificant, only a small population of patients showing an effective response [1,[6], [7], [8], [9]].

The recent interest in TRAIL-based intervention led to development of multiple approaches, including TRAIL conjugates, combinatorial approaches, TRAIL gene therapy, and cell-based therapy (Fig. 1). Numerous studies have explored these approaches in both in vitro and in vivo models with many promising outcomes. The objective of this review is to highlight the opportunities and challenges for improving TRAIL-based cancer therapy. We first start with the mechanism of action for TRAIL protein and catalogue various approaches to enhance TRAIL therapy based on this mechanistic insight. We focussed on TRAIL protein based and expression systems for intervention and avoided modified cell-based delivery approaches. We note that engineered MSCs, as well as other types of cells, could show effective migration to sites of action for improved concentration of TRAIL [10,11]. Adipose tissue derived MSCs [12], bone marrow derived MSCs [13], human umbilical cord MSCs (HUMSC) [14] for TRAIL delivery, as well as several non-viral approaches to engineer human stem cells to secrete TRAIL [[15], [16], [17], [18]]. We refer the reader to the above studies on cell-based TRAIL delivery approaches. We also note that others have reviewed TRAIL therapy elsewhere [7,10,19,20], and we refer the reader for a different perspective on the topic.

Section snippets

Mechanism of TRAIL action

TRAIL is a type II transmembrane protein that was initially identified based on the sequence homology of its extracellular domain with CD95L (28% identical) and TNF (23% identical) [4]. Its C-terminal extracellular end can be proteolytically cleaved from cell surface in vesicle-associated or soluble form [21]. TRAIL has a unique capacity to induce apoptosis in a variety of tumor cell lines, but not in most normal cells [[21], [22], [23]]. TRAIL binds with two receptors TRAIL-R1 (also known as

TRAIL therapy in clinical setting

TRAIL based therapy utilizes two types of pharmacological agents, rhTRAIL and mAbs against TRAIL-Rs. The rhTRAIL (Dulanermin), Mapatumumab (HGS-ETR1), Tigatuzumab (CS-1008), Lexatumumab (HGS-ETR2), Drozitumab (PRO95780), Conatumumab (AMG-655), LBY-135 and Apomab have already been tested in clinical trials [6,7,29,30]. Mapatumumab had the most promising outcome and entered a Phase II trial in patients with non-Hodgkin lymphoma and resulted in 3 responses with one complete recovery out of 40

Therapeutic resistance in TRAIL therapy

Insensitivity of ‘normal’ cells to TRAIL is governed by multiple mechanism that are also deployed in cancer cells to some extent, so that a mixed response has been observed in breast, pancreatic and prostate cancers [22,59]. Most of the triple negative/basal-like breast cancer cells (e.g. MDA-MB-231, MDA-MB-436, ZR75–1, SUP-149) are TRAIL sensitive and undergo effective apoptosis [60,61]. However, many cell lines display insignificant response to TRAIL such as breast cancer MDA-MB-453,

Revival of TRAIL therapy

Recombinant TRAIL protein and mAbs against TRAIL-R1 and TRAIL-R2 were the first line of therapeutics developed. But less than desired efficacy in clinical setting have resulted in identifying key limitations of these therapies (discussed in Section 3). Therefore, many critical approaches have been developed to increase the bioavailability of TRAIL, synergize the TRAIL activity for improved efficacy and sensitize TRAIL resistant cells to the administered agents.

Conclusions and future directions

Clinical development of TRAIL, despite promising preclinical studies, faced multiple hurdles and development of innovative strategies will be needed to overcome these hurdles. Chemical derivatization of TRAIL, development of nano-carriers for delivery of TRAIL protein and plasmid, genetic engineering of host cells for TRAIL expression, and recently explored TRAIL combinations are providing important leads. For evaluation of therapeutic efficacy, most studies were performed in xenografts or

Acknowledgments

The studies in the authors' lab are supported by operating grants from Canadian Institutes of Health Research (CIHR), an Innovation Grant from the Canadian Breast Cancer Foundation (CBCF) and equipment support from the NSERC and Edmonton Civic Employees Charitable Assistance Fund (ECE-CAF). Dr. Bindu Thapa was supported by Alberta Innovates Graduate Studentship. We thank Ms. Kylie Parent for help with literature search and Fig. 4. Remant KC and H. Uludag are founders and shareholders in RJH

References (197)

  • H.A. Wakelee et al.

    Phase I and pharmacokinetic study of lexatumumab (HGS-ETR2) given every 2 weeks in patients with advanced solid tumors

    Ann. Oncol.

    (2010)
  • M. Reck et al.

    A randomized, double-blind, placebo-controlled phase 2 study of tigatuzumab (CS-1008) in combination with carboplatin/paclitaxel in patients with chemotherapy-naive metastatic/unresectable non-small cell lung cancer

    Lung Cancer

    (2013)
  • M. Rahman et al.

    The TRAIL to targeted therapy of breast cancer

    Adv. Cancer Res.

    (2009)
  • D.O. Moon et al.

    Verrucarin A sensitizes TRAIL-induced apoptosis via the upregulation of DR5 in an eIF2alpha/CHOP-dependent manner

    Toxicol. in Vitro

    (2013)
  • A. Krueger et al.

    Cellular FLICE-inhibitory protein splice variants inhibit different steps of caspase-8 activation at the CD95 death-inducing signaling complex

    J. Biol. Chem.

    (2001)
  • A. Golks et al.

    c-FLIPR, a new regulator of death receptor-induced apoptosis

    J. Biol. Chem.

    (2005)
  • O. Micheau et al.

    The long form of FLIP is an activator of caspase-8 at the Fas death-inducing signaling complex

    J. Biol. Chem.

    (2002)
  • P. Schneider

    Production of recombinant TRAIL and TRAIL receptor: Fc chimeric proteins

    Methods Enzymol.

    (2000)
  • R.M. Pitti et al.

    Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family

    J. Biol. Chem.

    (1996)
  • N. Muller et al.

    Superior serum half life of albumin tagged TNF ligands

    Biochem. Biophys. Res. Commun.

    (2010)
  • T.H. Kim et al.

    PEGylated TNF-related apoptosis-inducing ligand (TRAIL)-loaded sustained release PLGA microspheres for enhanced stability and antitumor activity

    J. Control. Release

    (2011)
  • R. Li et al.

    Fusion to an albumin-binding domain with a high affinity for albumin extends the circulatory half-life and enhances the in vivo antitumor effects of human TRAIL

    J. Control. Release

    (2016)
  • A. Ashkenazi

    Targeting the extrinsic apoptotic pathway in cancer: lessons learned and future directions

    J. Clin. Invest.

    (2015)
  • J.E. Allen et al.

    Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects

    Sci. Transl. Med.

    (2013)
  • H. Walczak et al.

    Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo

    Nat. Med.

    (1999)
  • R.S. Herbst 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)
  • J. Lemke et al.

    Getting TRAIL back on track for cancer therapy

    Cell Death Differ.

    (2014)
  • A.W. Tolcher 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)
  • A. Forero-Torres et al.

    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.

    (2013)
  • G.E. Naoum et al.

    Journey of TRAIL from bench to bedside and its potential role in immuno-oncology

    Oncol. Rev.

    (2017)
  • X. Song et al.

    Secretory TRAIL-armed natural killer cell-based therapy: in vitro and in vivo colorectal peritoneal carcinomatosis xenograft

    Mol. Cancer Ther.

    (2016)
  • G. Grisendi et al.

    Mesenchymal progenitors expressing TRAIL induce apoptosis in sarcomas

    Stem Cells

    (2015)
  • M.J. Lathrop et al.

    Antitumor effects of TRAIL-expressing mesenchymal stromal cells in a mouse xenograft model of human mesothelioma

    Cancer Gene Ther.

    (2015)
  • S.M. Kim et al.

    Gene therapy using TRAIL-secreting human umbilical cord blood-derived mesenchymal stem cells against intracranial glioma

    Cancer Res.

    (2008)
  • Y.L. Hu et al.

    Mesenchymal stem cells as a novel carrier for targeted delivery of gene in cancer therapy based on nonviral transfection

    Mol. Pharm.

    (2012)
  • X.J. Tang et al.

    TRAIL-engineered bone marrow-derived mesenchymal stem cells: TRAIL expression and cytotoxic effects on C6 glioma cells

    Anticancer Res.

    (2014)
  • M.R. Moniri et al.

    TRAIL-engineered pancreas-derived mesenchymal stem cells: characterization and cytotoxic effects on pancreatic cancer cells

    Cancer Gene Ther.

    (2012)
  • S.A. Choi et al.

    Therapeutic efficacy and safety of TRAIL-producing human adipose tissue-derived mesenchymal stem cells against experimental brainstem glioma

    Neuro-Oncology

    (2011)
  • P.P.G. Guimaraes et al.

    Nanoparticles for immune cytokine TRAIL-based cancer therapy

    ACS Nano

    (2018)
  • A.L. Kretz et al.

    TRAILblazing strategies for cancer treatment

    Cancers (Basel)

    (2019)
  • S. Wang et al.

    TRAIL and apoptosis induction by TNF-family death receptors

    Oncogene

    (2003)
  • A. Ashkenazi et al.

    Safety and antitumor activity of recombinant soluble Apo2 ligand

    J. Clin. Invest.

    (1999)
  • S.W. Lowe et al.

    p53 status and the efficacy of cancer therapy in vivo

    Science

    (1994)
  • G. Pan et al.

    An antagonist decoy receptor and a death domain-containing receptor for TRAIL

    Science

    (1997)
  • D. Merino et al.

    Differential inhibition of TRAIL-mediated DR5-DISC formation by decoy receptors 1 and 2

    Mol. Cell. Biol.

    (2006)
  • A. Morizot et al.

    Chemotherapy overcomes TRAIL-R4-mediated TRAIL resistance at the DISC level

    Cell Death Differ.

    (2011)
  • D.R. Camidge 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)
  • S.J. Hotte 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)
  • A. Younes et al.

    A phase 1b/2 trial of mapatumumab in patients with relapsed/refractory non-Hodgkin’s lymphoma

    Br. J. Cancer

    (2010)
  • J.C. Soria et al.

    Randomized phase II study of dulanermin in combination with paclitaxel, carboplatin, and bevacizumab in advanced non-small-cell lung cancer

    J. Clin. Oncol.

    (2011)
  • Cited by (39)

    • Mesenchymal stem cells: A promising weapon for cancer therapy

      2023, Mesenchymal Stem Cells: Biological Concepts, Current Advances, Opportunities and Challenges
    • Design and synthesis of highly TRAIL expression HDAC inhibitors based on ONC201 to promote apoptosis of colorectal cancer

      2022, European Journal of Medicinal Chemistry
      Citation Excerpt :

      TRAIL (tumor necrosis factor (TNF)-related apoptosis inducing ligand) is the last member of the TNF family, and it is a cytokine that can induce tumor cell apoptosis. TRAIL is widely distributed in tissues and does not show obvious cytotoxicity to normal cells [4–7]. TRAIL-mediated apoptosis mainly acts through two signaling pathways [8]: the exogenous pathway and the endogenous pathway.

    • Inhibitors of ERp44, PDIA1, and AGR2 induce disulfide-mediated oligomerization of Death Receptors 4 and 5 and cancer cell death

      2022, Cancer Letters
      Citation Excerpt :

      However, TRAIL analogs and DR4/5 agonistic antibodies have yet to be clinically approved for anticancer therapy. This is due in part to pharmacokinetic issues related to these protein drugs themselves, and in part to the ability of cancer cells to acquire resistance through a variety of mechanisms, including downregulation of DR4 and DR5 (reviewed in Ref. [9]). Since DDAs upregulate DR5 and activate DR4 and DR5 in a ligand-independent manner [10,4], DDAs may overcome the resistance mechanisms that suppress the efficacy of other activators of the TRAIL/Death Receptor pathway.

    • Fusion of an EGFR-antagonistic affibody enhances the anti-tumor effect of TRAIL to EGFR positive tumors

      2022, International Journal of Pharmaceutics
      Citation Excerpt :

      Recombinant human soluble TRAIL showed a favorable antitumor activity in vitro and in vivo preclinical studies, and therefore has entered clinical trials over a decade ago. Although with slight side effect and well tolerated, TRAIL showed disappointing therapeutic efficacy in clinical trials, as objective response was observed only in a small portion of cancer patients (Stuckey and Shah, 2013; Thapa et al., 2020). The tumor proapoptotic activity of TRAIL relies on binding to two death receptors (DR4 and DR5) which are preferentially expressed on tumor cells.

    View all citing articles on Scopus
    View full text