Cancer Letters

Cancer Letters

Volume 383, Issue 2, 28 December 2016, Pages 250-260
Cancer Letters

Original Article
High-order TRAIL oligomer formation in TRAIL-coated lipid nanoparticles enhances DR5 cross-linking and increases antitumour effect against colon cancer

https://doi.org/10.1016/j.canlet.2016.10.005Get rights and content

Highlights

  • Liposomes with TRAIL show greater cytotoxicity than soluble TRAIL in colon cancer cells.

  • Liposomes with TRAIL overcome resistance to soluble TRAIL in colon cancer cells.

  • TRAIL anchored to liposome surface form high-order oligomers which in turn, induce a more potent clusterization of DR5.

  • Liposomes with TRAIL show greater in vivo anti-tumour activity than soluble TRAIL.

Abstract

During the last years, a great effort has been invested into developing new TRAIL formulations with increased bioactivity, trying to overcome the resistance to conventional soluble TRAIL (sTRAIL) exhibited by many primary tumours. In our group, we have generated artificial lipid nanoparticles decorated with sTRAIL (LUV-TRAIL), emulating the physiological TRAIL-containing exosomes by which T-cells release TRAIL upon activation. We already demonstrated that LUV-TRAIL has greater cytotoxicity against both chemoresistant haematologic tumour cells and epithelial carcinoma cells compared to a form of sTRAIL similar to that used in clinical trials. In this study we have tested LUV-TRAIL in several human colon cancer cell lines with different sensitivity to sTRAIL. LUV-TRAIL significantly improved sTRAIL cytotoxicity in all colon cancer cell lines tested. Trying to ascertain the molecular mechanism by which LUV-TRAIL exhibited improved cytotoxicity, we demonstrated that TRAIL-coated lipid nanoparticles were able to activate DR5 more efficiently than sTRAIL, and this relied on LUV-TRAIL ability to promote DR5 clustering on the cell surface. Moreover, we show that TRAIL molecules are arranged in higher order oligomers only in LUV-TRAIL, which may explain their enhanced DR5 clustering ability. Finally, LUV-TRAIL showed significantly better antitumour activity than sTRAIL in an in vivo model using HCT-116 xenograft tumours in nude mice, validating its potential clinical application.

Introduction

Colorectal cancer (CRC) is one of the most frequent solid tumours in western countries. In fact, recent estimations indicate that CRC is the third most common cancer both in males and females [1]. The survival rate throughout all stages has improved mainly due to the implementation of screening programs, leading to the detection of CRC in earlier stages, together with the development of more effective treatment options. However, CRC still accounts for about 9% of the estimated cancer-related deaths [1].

Resistance of cancer cells to apoptosis is one of the so-called hallmarks of cancer [2]. Since impairments in the intrinsic apoptotic pathway are often involved in this resistance, targeting the extrinsic apoptotic pathway is especially attractive for cancer therapy [3], [4]. Among the death ligands capable of triggering apoptosis through the extrinsic pathway, CD95L showed a remarkable strong cytotoxic potential on cancer cells. However, disappointingly it also displayed a severe systemic toxicity, therefore making unrealistic its use as anti-cancer agent [5]. On the other hand, another death ligand, namely tumour necrosis factor (TNF)-related apoptosis–inducing ligand (TRAIL), was found to be able to induce apoptosis in tumour cells without affecting most normal cells [6], [7]. Hence, it was soon considered as potentially useful for anti-cancer therapy [8], [9]. However, despite the encouraging initial results, TRAIL-based therapies showed very limited therapeutic activity in phase II/III clinical trials carried out on a wide variety of human cancers [3], [10]. In order to overcome TRAIL resistance, better sensitization strategies [11], [12], [13], as well as novel TRAIL formulations with improved bioactivity can be of great usefulness for its future clinical use [14], [15], [16], [17].

TRAIL has four membrane-bound receptors in humans: TRAIL-R1/DR4, TRAIL-R2/DR5, TRAIL-R3/DcR1 and TRAIL-R4/DcR2 [18], [19], [20], [21], [22], [23], [24]. In addition, TRAIL has been described to be able to bind to the soluble receptor osteoprotegerin (OPG) [25]. Among these five receptors, only DR4 and DR5 are able to transduce the apoptotic signal upon TRAIL binding. Although they share high similarity, both receptors present certain functional differences. For example, DR5 has been described to present higher affinity for TRAIL [26]. However, apparently DR5 requires further clustering, so it can only be properly activated by membrane-bound TRAIL or by artificially cross-linked versions of the molecule [27], [28]. On the contrary, DR4 can readily be activated by soluble versions of TRAIL, although cross-linked versions of TRAIL proved again to be more active than soluble TRAIL [29]. Regarding the relative contribution of both receptors to TRAIL-induced apoptosis, DR4 has been described to be the pre-eminent pro-apoptotic receptor in haematological cells, while in epithelial cells expressing both receptors DR5 appears to be the main pro-apoptotic receptor [30], [31], [32], [33], [34]. However, the differential contribution of both receptors is still controversial.

Our group demonstrated that, in physiological conditions, TRAIL was indeed released by activated human T cells in its transmembrane form, inserted in the membrane of lipid microvesicles called exosomes [35], [36]. On this basis, we have generated artificial lipid nanoparticles containing membrane-bound TRAIL (LUV-TRAIL) resembling those natural exosomes. We already demonstrated that LUV-TRAIL was more effective inducing apoptosis than soluble TRAIL against leukemia cells that presented resistance to soluble TRAIL and chemotherapeutic drugs [37], [38] and also in epithelial-derived cancer cells [39], [40]. This increased cytotoxicity induced by LUV-TRAIL was due to a superior DR5 activation, which led to an enhanced DISC recruitment [40], [41].

In the present work, we have tested LUV-TRAIL in several human colon tumour cell lines with different sensitivity to soluble TRAIL, finding that LUV-TRAIL was more efficient than soluble TRAIL inducing apoptosis in all cancer cells tested. Finally, the in vivo antitumour potential of LUV-TRAIL against colon carcinoma cells was tested in a model of nude mice xenografted with the human colon carcinoma cell line HCT-116. This novel TRAIL formulation showed an enhanced antitumour ability, and could be potentially useful to improve therapy against colon cancer.

Section snippets

Generation of liposomes covered with soluble recombinant TRAIL

Large Unilamellar Vesicles (LUV) with soluble recombinant TRAIL (hereafter sTRAIL) anchored on their surface were generated as previously described [37], [42]. Briefly, after generating the lipid nanoparticles, a version of sTRAIL (rTRAIL-His6, corresponding to amino acids 95–281, kindly provided by Dr. M. MacFarlane [22]) was attached to their surface by incubation at 37 °C for 30 min.

Cell culture

HCT-116, HT29 and CACO-2 cell lines were obtained from ATCC. Bax-deficient HCT-116 cells deficient (HCT-116-Bax

LUV-TRAIL show a significant higher cytotoxic ability compared with sTRAIL in human CRC cell lines

Firstly, we checked the surface expression of the TRAIL receptors DR4 and DR5 in all CRC cell lines studied (Fig. S2). finding that all cell lines expressed both pro-apoptotic TRAIL receptors. Noteworthy, all cell lines showed a higher expression of DR5 than DR4. Then, we characterized the overall sensitivity of the four CRC cell lines to both forms of TRAIL. For that, cell viability was analysed after carrying out dose-response assays using both sTRAIL and LUV-TRAIL (Fig. 1A). CRC cell lines

Discussion

Despite the initial optimism that TRAIL incited as a possible new and highly selective antitumour agent, the clinical effectiveness of TRAIL, both in monotherapy and in combined regimes, has been disappointing [3], [4], [10]. It is currently accepted that in order to overcome the poor responsiveness of some cancers to recombinant TRAIL formulations currently available [17] it appears as absolutely indispensable to improve its pro-apoptotic potential by the development of novel TRAIL

Acknowledgments

We gratefully acknowledge Dr. Gorka Basáñez for supporting in LUV-TRAIL generation. We are also indebted to Dr. Christoph Borner for providing HCT-Bax−/− cells. Finally, we gratefully acknowledge Dr. Henning Walczak for support throughout the years.

This work was supported by Grants PI13/00416 (LML) from Instituto de Salud Carlos III, SAF2013-48626-C2-1-R (AA) from Ministerio de Economía y Competitividad. Additionally, this work has been also supported in part by Grant DPI2011-28262-C04-01 (LJF)

References (69)

  • M. MacFarlane et al.

    Identification and molecular cloning of two novel receptors for the cytotoxic ligand TRAIL

    J. Biol. Chem.

    (1997)
  • P. Schneider et al.

    Characterization of two receptors for TRAIL

    FEBS Lett.

    (1997)
  • J.G. Emery et al.

    Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL

    J. Biol. Chem.

    (1998)
  • A. Truneh et al.

    Temperature-sensitive differential affinity of TRAIL for its receptors. DR5 is the highest affinity receptor

    J. Biol. Chem.

    (2000)
  • F. Muhlenbeck et al.

    The tumor necrosis factor-related apoptosis-inducing ligand receptors TRAIL-R1 and TRAIL-R2 have distinct cross-linking requirements for initiation of apoptosis and are non-redundant in JNK activation

    J. Biol. Chem.

    (2000)
  • S.K. Kelley et al.

    Targeting death receptors in cancer with Apo2L/TRAIL

    Curr. Opin. Pharmacol.

    (2004)
  • R.F. Kelley 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)
  • D. De Miguel et al.

    Liposome-bound TRAIL induces superior DR5 clustering and enhanced DISC recruitment in histiocytic lymphoma U937 cells

    Leuk. Res.

    (2015)
  • T. Mosmann

    Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays

    J. Immunol. Methods

    (1983)
  • A. Astashkina et al.

    A critical evaluation of in vitro cell culture models for high-throughput drug screening and toxicity

    Pharmacol. Ther.

    (2012)
  • S. Navran

    The application of low shear modeled microgravity to 3-D cell biology and tissue engineering

    Biotechnol. Annu. Rev.

    (2008)
  • P.E. Carrington et al.

    The structure of FADD and its mode of interaction with procaspase-8

    Mol. Cell

    (2006)
  • L.S. Dickens et al.

    The 'complexities' of life and death: death receptor signalling platforms

    Exp. Cell Res.

    (2012)
  • J.D. Graves et al.

    Apo2L/TRAIL and the death receptor 5 agonist antibody AMG 655 cooperate to promote receptor clustering and antitumor activity

    Cancer Cell

    (2014)
  • M. Gomez-Benito et al.

    Membrane expression of DR4, DR5 and caspase-8 levels, but not Mcl-1, determine sensitivity of human myeloma cells to Apo2L/TRAIL

    Exp. Cell Res.

    (2007)
  • R. Siegel et al.

    Cancer statistics, 2014

    CA Cancer J. Clin.

    (2014)
  • J. Lemke et al.

    Getting TRAIL back on track for cancer therapy

    Cell Death Differ.

    (2014)
  • J. Ogasawara et al.

    Lethal effect of the anti-Fas antibody in mice

    Nature

    (1993)
  • A. Ashkenazi

    Directing cancer cells to self-destruct with pro-apoptotic receptor agonists

    Nat. Rev. Drug Discov.

    (2008)
  • R.W. Johnstone et al.

    The TRAIL apoptotic pathway in cancer onset, progression and therapy

    Nat. Rev. Cancer

    (2008)
  • O. Micheau et al.

    Death receptors as targets in cancer

    Br. J. Pharmacol.

    (2013)
  • S. Bernardi et al.

    State of art and recent developments of anti-cancer strategies based on TRAIL

    Recent Pat. Anticancer Drug Discov.

    (2012)
  • L.Y. Dimberg et al.

    On the TRAIL to successful cancer therapy? Predicting and counteracting resistance against TRAIL-based therapeutics

    Oncogene

    (2013)
  • D. De Miguel et al.

    Onto better TRAILs for cancer treatment

    Cell Death Differ.

    (2016)
  • Cited by (42)

    • A self-sustained nanoplatform reverses TRAIL-resistance of pancreatic cancer through coactivating of exogenous and endogenous apoptotic pathway

      2021, Biomaterials
      Citation Excerpt :

      ART could improve the antitumor efficacy of TRAIL and reverse TRAIL-resistance of many tumor cell lines by upregulating the expression of DR4 and/or DR5 [24−28]. Hence, a self-sustained nanoplatform (SSN) was prepared to reverse TRAIL-resistance of pancreatic cancer through simultaneously promoting exogenous and endogenous apoptotic pathway, and as well to elongate circulation time [29–37]. Accordingly, once administrated, the enhanced circulation time provided enough time for TRAIL fragment to target to death receptors on tumor cells, which could be further improved the by interaction of positively charged R6 fragment with negatively charged tumor cell membrane [38].

    • DR5-targeted, chemotherapeutic drug-loaded nanoparticles induce apoptosis and tumor regression in pancreatic cancer in vivo models

      2020, Journal of Controlled Release
      Citation Excerpt :

      An alternative approach to enhance multivalency of antibodies is their attachment to the surface of NPs to promote clustering of antibody paratopes, and we have previously shown that efficient DR5 receptor clustering can be achieved through conjugation of AMG 655 to PEGylated poly(lactic-co-glycolide) (PLGA) nanoparticles, leading to activation of apoptosis in colorectal models [14,15]. Other groups have also shown that conjugation of monovalent recombinant TRAIL to the surface of multiple nanoparticle types causes an increase in its potency [16–19]. Upregulation of FLIP (Fas-associated death domain (FADD)-like IL-1β- converting enzyme-inhibitory protein) is a potential contributor to the lack of efficacy of DR5 targeting therapies through inhibiting caspase 8 activation [20].

    View all citing articles on Scopus
    1

    Current address: Cell Death, Cancer and Inflammation, University College of London, Grower Street, London, WC1E 6BT, United Kingdom.

    2

    Current address: Servicio de Inmunología, Hospital Clínico Universitario Lozano Blesa, Avda/San Juan Bosco 15, Zaragoza, 50009, Spain.

    View full text