Proapoptotic DR4 and DR5 signaling in cancer cells: toward clinical translation

https://doi.org/10.1016/j.ceb.2010.08.001Get rights and content

Proapoptotic receptor agonists (PARAs) targeting death receptors (DRs) 4 and 5 hold promise for cancer therapy based on their selective ability to kill malignant versus healthy cells. Emerging clinical results have confirmed that DR4/5 PARAs are relatively well-tolerated and suitable for further investigation. Given that some cancer cell lines and models are not sensitive to PARAs, it is important to develop strategies to identify what specific types of tumor cells may be most responsive to PARA-based therapy and how to overcome apoptosis resistance mechanisms in tumors. Here we review the molecular and biological determinants of responsiveness to PARAs in cancer cells, and discuss the potential for predictive biomarkers and drug combination strategies to maximize the anti-tumor activity of these agents.

Section snippets

Background

Apoptosis plays an essential role in development, homeostasis, and tumor suppression. In mammalian cells, two major pathways, often referred to as the extrinsic and intrinsic pathways, contribute to apoptotic signaling. Both require activation of distinct initiator caspases that target downstream effector caspases, which cleave numerous cellular substrates culminating in the hallmark features of apoptosis: plasma membrane blebbing, nuclear condensation and DNA fragmentation. The intrinsic, or

Apo2L/TRAIL signaling  physiological role in tumor suppression?

Studies in mice suggest that the Apo2L/TRAIL pathway has a role in immune surveillance. Mice deficient in either Apo2L/TRAIL or mDR5 (the sole mouse ortholog of human DR4 and 5) show increased susceptibility to tumorigenesis and pathogen infection [3, 8]. In certain models, however, tumor development does not appear to be affected by mDR5 deficiency; for example, there was no impact on the incidence of lymphomas in p53-null mice or intestinal polyps in the APCmin model [9]. Tumors develop

PARAs  co-opting the Apo2L/TRAIL pathway for cancer therapy

Recombinant Apo2L/TRAIL (dulanermin) and agonistic monoclonal antibodies targeting DR4 or DR5 (Figure 1) may have broad potential for cancer therapy [7, 13]. Proapoptotic anti-tumor activity has been demonstrated in models reflecting diverse tumor types, both in vitro and in tumor xenograft settings. However, despite the wide expression of DR4 and DR5, various cancer cell lines and primary tumor isolates exhibit partial responsiveness or resistance to DR4 and DR5 agonists [14••, 15, 16, 17].

Role of death and decoy receptors in resistance to PARAs

DR4 and DR5 mRNAs are widely detected in healthy tissues [18]; however, protein expression appears restricted to damaged, infected, or malignant cells [19, 20]. Cell-surface levels of DR4 and DR5 do not generally correlate with tumor cell sensitivity to Apo2L/TRAIL signaling [14••], although various agents can upregulate receptor expression and sensitize resistant tumor cells to PARAs [8, 21]. This suggests that low receptor density may be a relevant feature of tumor cell resistance. Indeed,

Defects within the DISC

The formation of a functional DISC is a central feature of DR signaling. Engagement of DR4 or DR5 by Apo2L/TRAIL or other agonists induces the recruitment of FADD and caspase-8 or caspase-10 to the plasma membrane. Additional requirements, including translocation of DISC components into membrane lipid rafts, facilitate receptor clustering and autocatalytic processing of the initiator caspase (Figure 1) [40]. These membrane-proximal events are critical for apoptotic signaling via the extrinsic

The mitochondrial apoptosis pathway in resistance to PARAs

In simplest terms, the mitochondrial apoptosis pathway is regulated by the balance of pro apoptotic versus anti apoptotic members of the Bcl-2 family. In most cancer cell lines, an effective apoptotic response to DR signaling requires signal amplification through the mitochondria. As such, alterations in the expression or function of the Bcl-2 family not only contribute to tumor development and chemoresistance, but also represent likely barriers to the therapeutic potential of PARAs.

The

Alternative signaling pathways–apoptosis friend or foe?

In addition to the primary DISC, engagement of DRs can induce a secondary signaling complex, containing core DISC components, as well as RIP1 and TRAF2 (Figure 1) [63]. This complex may activate additional signaling cascades, including the NF-κB, extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38-mitogen activated protein kinase (MAPK) pathways [8, 64]. Many of these alternative signaling events have been proposed to counter proapoptotic activity, although there

Conclusions/perspective

The selective killing of malignant versus normal cells by PARAs targeting the Apo2L/TRAIL pathway provides a unique opportunity to investigate how best to harness the extrinsic apoptosis pathway for cancer therapy. Given that many cancers are inherently refractory to apoptosis activation, the successful clinical translation of PARAs will likely require better understanding of the most relevant determinants of sensitivity versus resistance. Insights from preclinical studies regarding predictive

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

References (70)

  • B. Gillissen et al.

    Endogenous Bak inhibitors Mcl-1 and Bcl-xL: differential impact on TRAIL resistance in Bax-deficient carcinoma

    J Cell Biol

    (2010)
  • X. Chen et al.

    Differential roles of RelA (p65) and c-Rel subunits of nuclear factor kappa B in tumor necrosis factor-related apoptosis-inducing ligand signaling

    Cancer Res

    (2003)
  • S. Boehrer et al.

    Prostate-apoptosis-response-gene-4 increases sensitivity to TRAIL-induced apoptosis

    Leuk Res

    (2006)
  • R. Burikhanov et al.

    The tumor suppressor Par-4 activates an extrinsic pathway for apoptosis

    Cell

    (2009)
  • D.L. Vaux et al.

    IAPs, RINGs and ubiquitylation

    Nat Rev Mol Cell Biol

    (2005)
  • N.S. Wilson et al.

    Death receptor signal transducers: nodes of coordination in immune signaling networks

    Nat Immunol

    (2009)
  • F. Gonzalvez et al.

    New insights into apoptosis signaling by Apo2L/TRAIL

    Oncogene

    (2010)
  • D. Daniel et al.

    Tumor necrosis factor: renaissance as a cancer therapeutic?

    Curr Cancer Drug Targets

    (2008)
  • A. Ashkenazi

    Targeting death and decoy receptors of the tumour-necrosis factor superfamily

    Nat Rev Cancer

    (2002)
  • 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)
  • H.H. Yue et al.

    Loss of TRAIL-R does not affect thymic or intestinal tumor development in p53 and adenomatous polyposis coli mutant mice

    Cell Death Differ

    (2005)
  • G.P. Dunn et al.

    Interferons, immunity and cancer immunoediting

    Nat Rev Immunol

    (2006)
  • E. Cretney et al.

    Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-inducing ligand-deficient mice

    J Immunol

    (2002)
  • K. Takeda et al.

    Critical role for tumor necrosis factor-related apoptosis-inducing ligand in immune surveillance against tumor development

    J Exp Med

    (2002)
  • J. Wiezorek et al.

    Death receptor agonists as a targeted therapy for cancer

    Clin Cancer Res

    (2010)
  • K.W. Wagner et al.

    Death-receptor O-glycosylation controls tumor-cell sensitivity to the proapoptotic ligand Apo2L/TRAIL

    Nat Med

    (2007)
  • T. Naka et al.

    Effects of tumor necrosis factor-related apoptosis-inducing ligand alone and in combination with chemotherapeutic agents on patients’ colon tumors grown in SCID mice

    Cancer Res

    (2002)
  • J. Cheng et al.

    Multiple mechanisms underlie resistance of leukemia cells to Apo2 Ligand/TRAIL

    Mol Cancer Ther

    (2006)
  • B.L. Hylander et al.

    The anti-tumor effect of Apo2L/TRAIL on patient pancreatic adenocarcinomas grown as xenografts in SCID mice

    J Transl Med

    (2005)
  • J.P. Sheridan et al.

    Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors

    Science

    (1997)
  • K. Ichikawa et al.

    Tumoricidal activity of a novel anti-human DR5 monoclonal antibody without hepatocyte cytotoxicity

    Nat Med

    (2001)
  • E.L. Brincks et al.

    CD8 T cells utilize TRAIL to control influenza virus infection

    J Immunol

    (2008)
  • B. Pennarun et al.

    Playing the DISC: turning on TRAIL death receptor-mediated apoptosis in cancer

    Biochim Biophys Acta

    (2010)
  • P. Horak et al.

    Contribution of epigenetic silencing of tumor necrosis factor-related apoptosis inducing ligand receptor 1 (DR4) to TRAIL resistance and ovarian cancer

    Mol Cancer Res

    (2005)
  • Cited by (115)

    • PARP goes the weasel! Emerging role of PARP inhibitors in acute leukemias

      2021, Blood Reviews
      Citation Excerpt :

      Furthermore, IMGN632 when combined with talazoparib or olaparib was found to have synergistic in vitro effects against clinically annotated primary relapsed/refractory AML patient samples. Meng and colleagues reported that both olaparib and veliparib sensitize AML cell lines to TRAIL, the recombinant human tumor necrosis factor-related apoptosis inducing ligand [44] [45]. The human AML cell line ML-1 was treated for 24 h with veliparib and subsequently exposed to varying doses of recombinant human TRAIL agonist monoclonal antibodies, the natural ligand of the death receptor DR5.

    View all citing articles on Scopus
    1

    Both these authors contributed equally to this paper.

    View full text