Elsevier

Cancer Treatment Reviews

Volume 38, Issue 6, October 2012, Pages 760-766
Cancer Treatment Reviews

New Drugs
Inhibitor of Apoptosis (IAP) proteins as therapeutic targets for radiosensitization of human cancers

https://doi.org/10.1016/j.ctrv.2012.01.005Get rights and content

Summary

Radiotherapy initiates a variety of signaling events in cancer cells that eventually lead to cell death in case the DNA damage cannot be repaired. However, the signal transduction pathways that mediate cell death in response to radiation-inflicted DNA damage are frequently disturbed in human cancers, contributing to radioresistance. For example, aberrant activation of antiapoptotic programs such as high expression of Inhibitor of Apoptosis (IAP) proteins has been shown to interfere with the efficacy of radiotherapy. Since IAP proteins have been linked to radioresistance in several malignancies, therapeutic targeting of IAP proteins may open new perspectives to overcome radioresistance. Therefore, molecular targeting of IAP proteins may provide novel opportunities to reactivate cell death pathways that mediate radiation-induced cytotoxicity. A number of strategies have been developed in recent years to antagonize IAP proteins for the treatment of cancers. Some of these approaches have already been translated into a clinical application. While IAP protein-targeting agents are currently being evaluated in early clinical trials alone or in combination with conventional chemotherapy, they have not yet been tested in combination with radiation therapy. Therefore, it is a timely subject to discuss the opportunities of antagonizing IAP proteins for radiosensitization. Preclinical studies demonstrating the potential of this concept in relevant in vitro and in vivo models underscore that this combination approach warrants further clinical investigation. Thus, combination protocols using IAP antagonists together with radiotherapy may pave the avenue to more effective radiation-based treatment options for cancer patients.

Introduction

The anticancer activity of most cytotoxic therapies which are currently used in the clinical management of cancer patients including radiotherapy is based on their ability to activate cell death programs such as apoptosis in cancer cells. Apoptosis or programmed cell death is an intrinsic program that is in place in every cell of the human body, including cancer cells.1 A characteristic feature of human cancers is the inability to mount a proper apoptotic response during tumor progression or upon treatment with cytotoxic therapies.2 Therefore, evasion of apoptosis constitutes a critical cause of primary or acquired treatment resistance that frequently occurs in various human cancers. This also applies to the resistance of cancers to radiotherapy, one of the main pillars of cancer therapy. Therefore, a better understanding of the molecular events that are responsible for the defects encountered in the apoptosis signaling network may offer novel perspectives to tackle treatment resistance, including radioresistance of human cancers. This review focuses on apoptosis resistance caused by aberrant expression and/or function of Inhibitor of Apoptosis (IAP) proteins, a family of antiapoptotic proteins, and discusses the opportunities of how the targeting of IAP proteins can be translated into the design of new radiation-based treatment protocols.

Section snippets

Signaling pathways in radiation-induced cell death

Radiation-induced signaling events can be initiated in distinct cellular compartments, for example the nucleus, the cytosol or the plasma membrane.3 In response to DNA damage that is sensed in the nucleus, the tumor suppressor and checkpoint protein p53 accumulates and becomes activated, thereby initiating cell cycle arrest, DNA repair and, in case of severe DNA damage, the induction of cell death.4 Furthermore, radiation can stimulate the production of reactive oxygen species (ROS) that can

Targeting IAP proteins for radiosensitization

On theoretical grounds, targeting IAP proteins may present a particularly promising approach to overcome radioresistance, since some IAP proteins such as XIAP and cIAP1 are among the proteins that can be regulated at the level of translation under cellular stress conditions.21 Accordingly, the messenger RNA (mRNA) molecules of XIAP and cIAP1 are translated via an internal ribosome entry site (IRES).22, 23 This IRES site allows continued translation of the protein even under cellular stress

Genetic interventions to target IAP proteins

To test the therapeutic potential of neutralizing IAP proteins to increase radiosensitivity of human cancers, a number of different genetic strategies have been employed (Table 1). For example, overexpression of Smac, the endogenous inhibitor of IAP proteins, has been reported to present a potent mean to increase radiation-induced cell death in a variety of human cancers, including neuroblastoma, glioblastoma, pancreatic and breast carcinoma.30, 31 Both the full-length and the mature forms of

Pharmacological interventions to target IAP proteins

In addition to genetic strategies to interfere with aberrant expression of IAP proteins in order to increase radiosensitivity of cancers, a number of pharmacological approaches have been developed to neutralize IAP proteins (Table 2). These efforts focused on the development of small molecule compounds that mimick the N-terminal stretch of the endogenous Smac protein. Small molecule IAP inhibitors were reported to sensitize pancreatic carcinoma cells for irradiation-induced apoptosis.32

Molecular mechanisms

The following common picture has emerged so far from these preclinical studies into the underlying molecular mechanisms that mediate the cooperative induction of apoptosis by IAP antagonists and radiotherapy (Fig. 1). Neutralizing IAP proteins in conjunction with irradiation results in increased activation of the caspase cascade as well as enhanced mitochondrial outer membrane permeabilization involving increased loss of mitochondrial membrane potential and cytochrome c release from the

Clinical development of IAP protein-targeting agents

The concept to target IAP proteins for therapeutic purposes has been transferred into a clinical context in recent years. Both antisense oligonucleotides as well as small molecule inhibitors of IAP proteins have been evaluated in phase I/II clinical trials either as monotherapy or in combination with conventional chemotherapeutics (Table 3). In general, XIAP antisense oligonucleotides and IAP antagonists were well tolerated in these early clinical trials,56, 57, 58, 59, 60, 61 with the

Conclusions

IAP proteins are aberrantly expressed in various human cancers and represent promising molecular targets for therapeutic intervention in order to restore the ability of cancer cells to undergo cell death in response to radiotherapy. IAP protein-targeting agents have shown synergistic antitumor activity in combination with irradiation in a wide variety of preclinical models. Therefore, the concept to antagonize IAP proteins in order to increase radiosensitivity is considered as a promising

Conflict of interest

None to declare.

Acknowledgements

The expert secretarial assistance of C. Hugenberg is greatly appreciated. Work in the author’s laboratory is supported by grants from the Deutsche Forschungsgemeinschaft, the Deutsche Krebshilfe, the Bundesministerium für Forschung und Technologie, IAP6/18 and the European Community (ApopTrain, APO-SYS).

References (64)

  • R.A. Lockshin et al.

    Cell death in health and disease

    J Cell Mol Med

    (2007)
  • S. Fulda

    Tumor resistance to apoptosis

    Int J Cancer

    (2009)
  • R.K. Schmidt-Ullrich et al.

    Signal transduction and cellular radiation responses

    Radiat Res

    (2000)
  • W.P. Roos et al.

    DNA damage-induced cell death by apoptosis

    Trends Mol Med

    (2006)
  • T. Ozben

    Oxidative stress and apoptosis: impact on cancer therapy

    J Pharm Sci

    (2007)
  • R. Kolesnick et al.

    Radiation and ceramide-induced apoptosis

    Oncogene

    (2003)
  • Q. Chen et al.

    The late increase in intracellular free radical oxygen species during apoptosis is associated with cytochrome c release, caspase activation, and mitochondrial dysfunction

    Cell Death Differ

    (2003)
  • P. Dent et al.

    MAPK pathways in radiation responses

    Oncogene

    (2003)
  • B. Gong et al.

    Ionizing radiation-induced, Bax-mediated cell death is dependent on activation of cysteine and serine proteases

    Cell Growth Differ

    (1999)
  • R.C. Taylor et al.

    Apoptosis: controlled demolition at the cellular level

    Nat Rev Mol Cell Biol

    (2008)
  • Q. Chen et al.

    Distinct stages of cytochrome c release from mitochondria: evidence for a feedback amplification loop linking caspase activation to mitochondrial dysfunction in genotoxic stress induced apoptosis

    Cell Death Differ

    (2000)
  • S. Fulda et al.

    Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy

    Oncogene

    (2006)
  • A. Ashkenazi

    Targeting the extrinsic apoptosis pathway in cancer

    Cytokine Growth Factor Rev

    (2008)
  • S. Fulda et al.

    Targeting mitochondria for cancer therapy

    Nat Rev Drug Discov

    (2010)
  • S. Fulda et al.

    Targeting IAP proteins for therapeutic intervention in cancer

    Nat Rev Drug Disc

    (2012)
  • D. Vucic et al.

    Ubiquitylation in apoptosis: a post-translational modification at the edge of life and death

    Nat Rev Mol Cell Biol

    (2011)
  • B.P. Eckelman et al.

    Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family

    EMBO Rep

    (2006)
  • M. Lu et al.

    XIAP induces NF-kappaB activation via the BIR1/TAB1 interaction and BIR1 dimerization

    Mol Cell

    (2007)
  • R. Hofer-Warbinek et al.

    Activation of NF-kappa B by XIAP, the X chromosome-linked inhibitor of apoptosis, in endothelial cells involves TAK1

    J Biol Chem

    (2000)
  • E. Varfolomeev et al.

    (Un)expected roles of c-IAPs in apoptotic and NFkappaB signaling pathways

    Cell Cycle

    (2008)
  • S.M. Lewis et al.

    IRES in distress: translational regulation of the inhibitor of apoptosis proteins XIAP and HIAP2 during cell stress

    Cell Death Differ

    (2005)
  • M. Holcik et al.

    A new internal-ribosome-entry-site motif potentiates XIAP-mediated cytoprotection

    Nat Cell Biol

    (1999)
  • D. Warnakulasuriyarachchi et al.

    The translation of an antiapoptotic protein HIAP2 is regulated by an upstream open reading frame

    Cell Death Differ

    (2003)
  • M. Holcik et al.

    Translational upregulation of X-linked inhibitor of apoptosis (XIAP) increases resistance to radiation induced cell death

    Oncogene

    (2000)
  • L. Gu et al.

    Regulation of XIAP translation and induction by MDM2 following irradiation

    Cancer Cell

    (2009)
  • I. Tamm

    AEG-35156, an antisense oligonucleotide against X-linked inhibitor of apoptosis for the potential treatment of cancer

    Curr Opin Investig Drugs

    (2008)
  • R. Mannhold et al.

    IAP antagonists: promising candidates for cancer therapy

    Drug Discov Today

    (2010)
  • C.S. Straub

    Targeting IAPs as an approach to anti-cancer therapy

    Curr Top Med Chem

    (2011)
  • J. Chai et al.

    Structural and biochemical basis of apoptotic activation by Smac/DIABLO

    Nature

    (2000)
  • S. Giagkousiklidis et al.

    Sensitization for gamma-irradiation-induced apoptosis by second mitochondria-derived activator of caspase

    Cancer Res

    (2005)
  • T.E. Fandy et al.

    Smac/DIABLO enhances the therapeutic potential of chemotherapeutic drugs and irradiation, and sensitizes TRAIL-resistant breast cancer cells

    Mol Cancer

    (2008)
  • S. Giagkousiklidis et al.

    Sensitization of pancreatic carcinoma cells for gamma-irradiation-induced apoptosis by XIAP inhibition

    Oncogene

    (2007)
  • Cited by (33)

    • The Emerging Role of Deubiquitinases in Radiosensitivity

      2024, International Journal of Radiation Oncology Biology Physics
    • Development of injectable thermosensitive polypeptide hydrogel as facile radioisotope and radiosensitizer hotspot for synergistic brachytherapy

      2020, Acta Biomaterialia
      Citation Excerpt :

      The PETyr and PETyr-I131 polymers were evaluated via proton nuclear magnetic resonance (1H NMR) spectroscopy and gel permeation chromatography (GPC). First, the sol-gel transition was determined via a previously reported vial inversion method [33]. The PETyr copolymer solutions (~0.5 mL) at a given concentration were prepared in 2 mL vials, which were incubated at each scheduled temperature for 15 min.

    • Cancer stem cells in prostate cancer radioresistance

      2019, Cancer Letters
      Citation Excerpt :

      These findings highlight a crucial role, and therapeutic target potential, of DNA repair-associated pathways in PCSCs in CaP radioresistance. Another cellular response to significant DNA damage is to activate apoptotic pathways, the evasion of which also confers radioresistance [17]. The Wnt/β-catenin pathway which regulates livin, a preventer of cell apoptosis, has been reported to be overactivated in PCSCs.

    • Targeting the heat shock response in combination with radiotherapy: Sensitizing cancer cells to irradiation-induced cell death and heating up their immunogenicity

      2015, Cancer Letters
      Citation Excerpt :

      Enhancing irradiation-induced cell death is one strategy of combined modality approaches. Manifold efforts were undertaken to enhance irradiation-induced apoptosis by specific targeting of apoptosis regulators [24]. However, recent reports have questioned whether apoptosis is the desired form of cell death that should be induced by or together with RT, since apoptotic cells appear to stimulate the proliferation of surviving cancer cells, favor angiogenesis, and induce immune tolerance [25,26].

    • Mitochondrial protein targets of radiosensitisation by 1,8-dihydroxy-3- acetyl-6-methyl-9,10 anthraquinone on nasopharyngeal carcinoma cells

      2014, European Journal of Pharmacology
      Citation Excerpt :

      Carcinogenesis, which results in a complex biological process, is mediated by many proteins. Recognition of how target proteins interact with each other and how they affect biological function will be indispensable (Fulda, 2012). The key node protein in the network diagram will be our essential observation target.

    View all citing articles on Scopus
    View full text