Biochemistry & Pharmacology: Open Access

Biochemistry & Pharmacology: Open Access
Open Access

ISSN: 2167-0501

+44-77-2385-9429

Mini Review - (2018) Volume 7, Issue 2

SMAC Mimetics: Promising Therapeutic Agents in Cancer and other Diseases

Laurence Dubrez1,2*
1Université de Bourgogne Franche-Comté, LNC UMR1231, 21000 Dijon, France
2Institut National de la Santé et de la Recherche Médicale (Inserm), LNC UMR1231, 21000 Dijon, France
*Corresponding Author: Laurence Dubrez, Université de Bourgogne Franche-Comté, LNC UMR1231, 21000 Dijon, France, Tel: +33-380-393-356 Email:

Abstract

Smac mimetics are a very promising new class of anticancer agents demonstrating an acceptable safety profile and efficacy in some preclinical models of cancers when used as a single agent or in combination with conventional or nonconventional therapies. Future preclinical and clinical trials could enlarge their spectral of action in inflammatory, fibrotic and infectious diseases.

Keywords: Smac mimetics; Nonconventional therapies; Apoptosis; Inflammasome

Abbreviations

cIAPs: Cellular IAPs; IL: Interleukin; MAPK: Mitogen-activated Protein Kinase; NF-κB: Nuclear Factor kappa-light-chain-enhancer of Activated B Cells; NODs: Nucleotide-binding Oligomerization Domain-containing Proteins; PAMPs: Pathogen-associated Molecular Pat-terns; PRRs: Pattern-recognition Receptors; RIG I: Retinoic Acidinducible Gene I; Smac: Second Mitochondria-derived Activator of Caspases; SMs: Smac Mimetics; TLRs: Toll-like Receptors; TNFR: Tumor Necrosis Factor Receptor; XIAP: X-chromosome Linked IAP.

Introduction

The inhibitor of apoptosis proteins (IAPs) were initially discovered in baculovirus in a genetic screen aiming to identify viral proteins able to block the death of infected cells [1]. IAP homologs were then described in yeasts, nematode, insects, fishes and mammals based on structural feature. The IAP family is defined by the presence of at least one specific domain named Baculovirus IAP Repeat (BIR) located at the N-terminal end of the protein. Some IAPs also harbor a C-terminal RING domain that confers an E3-ubiquitine ligase activity (For review, see [2,3]). Mammalian cells contain 8 IAP members including the Xchromosome linked IAP (XIAP), the cellular IAP 1 and 2 (cIAP1 and cIAP2) and the melanoma apoptosis inhibitory protein (ML-IAP) that exert an anti-apoptotic activity. Among them, XIAP can directly block the activity of initiator caspase-9 and executor caspases-3 and -7, thereby inhibiting both intrinsic and extrinsic pathways of apoptosis [4]. Upon activation of intrinsic pathway, XIAP is neutralized by the second mitochondria-derived activator of caspases (Smac) that is released from the mitochondria as a result of the mitochondria outer membrane permeabilization (MOMP). As an additional checkpoint preventing unforeseen caspase activation, cIAP1, cIAP2 and ML-IAP can bind, sequester and target Smac for ubiquitin proteasome system (UPS)-mediated degradation, thus favoring XIAP in caspase inhibition [5]. The structural characterization of the interaction of XIAP with caspases or Smac demonstrated that they bind at the same binding interface e.i. a surface hydrophobic groove located within some BIR domains. Thus, Smac acts as a competitive inhibitor of XIAP-caspase interaction. These results led to the design of synthetic IAP antagonist compounds mimicking the activity of Smac and named Smac mimetics (SMs) [6,7]. This brief review will summarize advances on the use of SMs in cancer therapy and will present recent preclinical studies highlighting the therapeutic potential of SMs in inflammatory, fibrotic and infectious diseases (Figure 1).

biochemistry-pharmacology-inhibiting-inflammasome-activation

Figure 1: A- Cell signaling activity of IAPs. IAPs are important cell signaling regulators of TNFR signaling pathways. They are required for the canonical NF-κB activation, MAPK activation and pro-inflammatory cytokine production and they inhibit cell death and the non-canonical NF-κB activation. IAPs can also control the canonical NF-κB activation in response to PRRs. XIAP blocks apoptosis by inhibiting caspases and cIAPs and ML-IAP prevent Smac from neutralizing XIAP. Moreover, cIAPs have been involved in inflammasome activation. B- Mechanisms of action of Smac mimetics. Smac mimetics can bind XIAP, cIAPs and ML-IAP. They favor apoptosis by blocking XIAP-caspase interaction. They induce the ubiquitin-proteasome system-mediated degradation of cIAP1 and in some situation cIAP2 and XIAP, inhibiting canonical activation of NF-κB in response to TNFR and PRR engagement, inhibiting inflammasome activation, promoting the non-canonical NF-κB activation and priming cells to TNF-induced cell death.

SMs in Cancer Therapy

Because (i) IAPs display potent anti-apoptotic properties, (ii) their expression were showed to be upregulated in number of human tumor samples and correlated with advanced progressive disease, aggressiveness, and poor prognosis and (iii) they constitute a resistance factor to anticancer therapy, SMs have been developed for the purpose of anti cancer treatments. Number of preclinical studies showed their capacity to inhibit tumor growth in multiple solid tumors, acute lymphocytic leukemia (ALL) and multiple myeloma xenograft models and demonstrated a synergic activity of SMs with conventional therapeutic agents and with novel therapy e.g. tumor necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL), proteasome inhibitors, BH3-mimetics or immune checkpoint inhibitors [6,8-11]. Importantly, these compounds were well tolerated by animals and did not display toxicity against normal lymphocytes, bone marrow stromal cells or normal mammary epithelial cells [12]. SMs have entered into active human clinical trials for review, see [6,13]. They are being evaluated as a single agent or in combination with an anticancer drug in both solid tumors and hematological malignancies. The first clinical trials results demonstrated a good tolerance and target inhibition [14,15].

As expected, SMs abrogate XIAP-mediated caspases inhibition and restore apoptotic response in cancer cells. In addition to target XIAP, SMs also bind the BIR domains of ML-IAP, cIAP1 and cIAP2. SMs appeared to stimulate the E3-ubiquitine ligase activity of cIAP1, which results in the auto-ubiquitination and very fast degradation of cIAP1 and in some situation cIAP2 and XIAP. As a consequences, SM cause the production of TNF-α which can trigger cell death by an autocrine pathway [3]. These unexpected results highlighted the critical role of cIAPs as cell signaling regulators. Because of their capacity to bind and ubiquitinylate important signaling intermediates, they control the activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and MAPK (Mitogen-activated protein kinase) signaling pathways downstream of some members of the TNF receptor (TNFR) superfamily including TNFR1, TNFR2, CD30 (Cluster of differentiation 30), CD40 and TWEAK (TNF-like weak inducer of apoptosis). They are required for the canonical activation of NF-κB and MAPK while they block the non-canonical NF-κB activating pathway and cytoplasmic caspase-activation platform assembly. By neutralizing XIAP and degrading cIAPs, SMs simultaneously stimulated NF-κB-mediated pro-inflammatory cytokines production and prime tumor cells for death receptor-mediated cell death [3]. Thus, tumor cells become highly sensitive to TNF-α produced by tumoral cells themselves or by CD8+ lymphocytes and NK (Natural Killer) cells within the tumor (9). Moreover, the boosted production of proinflammatory signals including IFNγ (Interferon gamma) and IL-2 (Interleukin-2) can elicit antitumoral immune response by promoting maturation of antigen-presenting cells and enhancing T-cell response [8,9,16-21].

SMs in Preventing PRR-Dependent Inflammatory Diseases

More recent studies enlarged the signaling function of IAPs in antimicrobial innate immunity by demonstrating their role in signaling pathways initiated by pattern-recognition receptors (PRRs) [22]. These include cytosolic nucleotide-binding oligomerization domaincontaining proteins (NODs) activated in response to intracellular bacterial infection, membrane-bound Toll-like receptors (TLRs) that sensing pathogen-associated molecular patterns (PAMPs) originating from bacteria, viruses, fungi, parasites and cytosolic retinoic acidinducible gene I (RIG I) that recognizes viral RNA. IAPs are recruited on the receptor-associated intracellular signaling complex upon receptor engagement and participate to the signal transduction inducing NF-κB and MAPK activation and the expression of proinflammatory genes [23]. Moreover, cIAPs have been in involved in the maturation of IL-1β and IL-18 by controlling the assembly of inflammasome (for review, see [22]). Thus, inhibiting IAPs with SMs could ultimately decrease the production of inflammatory cytokines, reduce the recruitment of immune cells at the site of inflammation and therefore could potentially prevent tissue damage caused by sepsis and chronic inflammatory conditions. A recent in vivo investigation demonstrated a benefic effect of the SM birinapant on liver injury and survival in endotoxemic mice [24].

SMs in Fibrotic Diseases

In 2016, Ashley et al. highlighted the therapeutic potential of SMs in fibrotic diseases [25]. Pulmonary fibrosis is a progressive disease with a high mortality rate and the therapeutic options available to patients are limited. Fibrosis can be triggered by exposure to various injuries such as infection, allergens, drugs, toxins or it occurs for unknown reasons (case of the idiopathic pulmonary fibrosis: IPF). Fibrosis is characterized by the differentiation of fibroblasts into myofibroblastlike cells producing extracellular matrix. The accumulation of extracellular matrix in the alveolar space causes irreversible damage affecting vital function of lung. An upregulation of XIAP- encoding gene has been reported in patient-derived IPF fibroblasts [26]. The role of a chronic inflammation and specially IL-1β has been demonstrated in a mice model of bleomycin-induced lung fibrosis [27]. Treatment of fibrotic mice with the SM AT-406 significantly decreases the expression of pro-inflammatory cytokines and reduced collagen accumulation. Interestingly, AT-406 has also an antifibrotic activity when administrated latter, after the early inflammatory phase of the disease [25].

SMs in Clearing Virus-infected Cells

IAPs were initially discovery for their properties to block apoptosis of viral infected insect cells, allowing viral propagation [1]. In mammals, cIAPs are a survival factors in HBV (hepatite V virus) infected hepatocytes, allowing viral persistence [28]. Therefore, SMs could be a good option to selectively clear latently infected cells. The efficiency of Smac mimetics in treating HBV infection has been evaluated in an immunocompetent mouse model of chronic HBV. SMs treatment significantly decreases the serum HBV DNA and serum HBV surface antigen level, reduces the amount of HB core antigen (HBcAG)-positives hepatocytes and decrease the content of HBV genome in infected livers. Moreover, SMs improve the efficiency of the antiviral nucleoside analog entecavir in clearing infection [29].

Conclusion

SMs are very promising novel class of therapeutic agents for treating cancer. Ongoing and future clinical trials will determine the efficiency, safety and drugs combinations. The ongoing investigations of mechanistic effects of SMs and the analysis of the genetic and environmental tumors profile should make possible to define hallmarks for predicting the response to SM treatment and for determining the appropriate indications. More work is required to decipher the relationship between IAPs and several cell signaling pathways, to determine the importance of this class of proteins in the physiopathology of inflammatory, fibrotic and infectious diseases and to analysis the potential therapeutic of IAP antagonists in these diseases. SMs are non-specific compounds targeting several IAPs. Ongoing work will determine whether more selective antagonists to one member of the family or to one specific IAP function could be relevant in a particular pathological context.

References

  1. Crook NE, Clem R J, Miller LK (1993) An apoptosis-inhibiting baculovirus gene with a zinc finger-like motif. J Virol 67: 2168-2174.
  2. Estornes Y, Bertrand MJ (2014) IAPs, regulators of innate immunity and inflammation. Seminars in cell & developmental biology.
  3. Silke J, Meier P (2013) Inhibitor of apoptosis (IAP) proteins-modulators of cell death and inflammation. Cold Spring Harbor perspectives in biology 5.
  4. Eckelman BP, Salvesen GS, Scott FL (2006) Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep 7: 988-994.
  5. Berthelet J, Dubrez L (2013) Regulation of Apoptosis by Inhibitors of Apoptosis (IAPs). Cells 2: 163-187.
  6. Dubrez L, Berthelet J, Glorian V (2013) IAP proteins as targets for drug development in oncology. OncoTargets and therapy 9: 1285-1304.
  7. Rathore R, McCallum JE, Varghese E, Florea AM, Büsselberg D (2017) Overcoming chemotherapy drug resistance by targeting inhibitors of apoptosis proteins (IAPs). Apoptosis 22: 898-919.
  8. Kearney CJ, Lalaoui N, Freeman AJ, Ramsbottom KM, Silke J, et al. (2017) PD-L1 and IAPs co-operate to protect tumors from cytotoxic lymphocyte-derived TNF. Cell Death Differ 24: 1705-1716.
  9. Beug ST, Beauregard CE, Healy C, Sanda T, St-Jean M, et al. (2017) Smac mimetics synergize with immune checkpoint inhibitors to promote tumour immunity against glioblastoma. Nat Commun 8.
  10. Chen KF, Lin JP, Shiau CW, Tai WT, Liu CY, et al. (2012) Inhibition of Bcl-2 improves effect of LCL161, a SMAC mimetic, in hepatocellular carcinoma cells. Biochemical pharmacology 84: 268-277.
  11. Derakhshan A, Chen Z, Van Waes C (2017) Therapeutic Small Molecules Target Inhibitor of Apoptosis Proteins in Cancers with Deregulation of Extrinsic and Intrinsic Cell Death Pathways. Clin Cancer Res 23: 1379-1387.
  12. Flygare JA, Beresini M, Budha N, Chan H, Chan IT, et al. (2012) Discovery of a potent small-molecule antagonist of inhibitor of apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152). J Medicinal Chemistry 55: 4101-4113.
  13. Beug ST, Conrad DP, Alain T, Korneluk RG, Lacasse EC (2015) Combinatorial cancer immunotherapy strategies with proapoptotic small-molecule IAP antagonists. Int J Dev Biol 59: 141-147.
  14. Amaravadi RK, Schilder RJ, Dy GK, Ma WW, Jr GJF, et al. (2010) Phase 1 study of the Smac mimetic TL32711 in adult subjects with advanced solid tumors and lymphoma to evaluate safety, pharmacokinetics, pharmacodynamics, and antitumor activity. in 102rd Annual Meeting of the American Association for cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR 71.
  15. Noonan AM, Bunch KP, Chen JQ, Herrmann MA, Lee JM, et al. (2016) Pharmacodynamic markers and clinical results from the phase 2 study of the SMAC mimetic birinapant in women with relapsed platinum-resistant or -refractory epithelial ovarian cancer. Cancer 122: 588-597.
  16. Clancy-Thompson E, Ali L, Bruck PT, Exley MA, Blumberg RS, et al. (2018) IAP Antagonists Enhance Cytokine Production from Mouse and Human iNKT Cells. Cancer Immunol Res 6: 25-35.
  17. Emeagi PU, Van Lint S, Goyvaerts C, Maenhout S, Cauwels A, et al. (2012) Proinflammatory characteristics of SMAC/DIABLO-induced cell death in antitumor therapy. Cancer Research 72: 1342-1352.
  18. Dougan M, Dougan S, Slisz J, Firestone B, Vanneman M, et al. (2010) IAP inhibitors enhance co-stimulation to promote tumor immunity. J Exp Med 207: 2195-2206.
  19. Chesi M, Mirza NN, Garbitt VM, Sharik ME, Dueck AC, et al. (2016) IAP antagonists induce anti-tumor immunity in multiple myeloma. Nat Med 22: 1411-1420.
  20. Beug ST, Tang VA, LaCasse EC, Cheung HH, Beauregard CE, et al. (2014) Smac mimetics and innate immune stimuli synergize to promote tumor death. Nat Biotechnol 32: 182-190.
  21. Beug ST, LaCasse EC, Korneluk RG (2014) Smac mimetics combined with innate immune stimuli create the perfect cytokine storm to kill tumor cells. Oncoimmunology 3: e28541.
  22. Vandenabeele P, Bertrand MJ (2012) The role of the IAP E3 ubiquitin ligases in regulating pattern-recognition receptor signalling. Nat Rev Immunol 12: 833-844.
  23. Tseng PH, Matsuzawa A, Zhang W, Mino T, Vignali DA, et al. (2010) Different modes of ubiquitination of the adaptor TRAF3 selectively activate the expression of type I interferons and proinflammatory cytokines. Nat Immunol 11: 70-75.
  24. Liu H, Liao R, He K, Zhu X, Li P, Gong J (2017) The SMAC mimetic birinapant attenuates lipopolysaccharide-induced liver injury by inhibiting the tumor necrosis factor receptor-associated factor 3 degradation in Kupffer cells. Immunol Lett 185: 79-83.
  25. Ashley SL, Sisson TH, Wheaton AK, Kim KK, Wilke CA, et al. (2016) Targeting Inhibitor of Apoptosis Proteins Protects from Bleomycin-Induced Lung Fibrosis. Am J Respir Cell Mol Biol 54: 482-492.
  26. Ajayi IO, Sisson TH, Higgins PD, Booth AJ, Sagana RL, et al. (2013) X-linked inhibitor of apoptosis regulates lung fibroblast resistance to Fas-mediated apoptosis. Am J Respir Cell Mol Biol 49: 86-95.
  27. Burgy O, Bellaye PS, Causse S, Beltramo G, Wettstein G, et al. (2016) Pleural inhibition of the caspase-1/IL-1β pathway diminishes profibrotic lung toxicity of bleomycin. Respir Res 17: 162.
  28. Ebert G, Preston S, Allison C, Cooney J, Toe JG, et al. (2015) Cellular inhibitor of apoptosis proteins prevent clearance of hepatitis B virus. Proc Natl Acad Sci USA 112: 5797-5802.
  29. Ebert G, Allison C, Preston S, Cooney J, Toe JG, et al. (2015) Eliminating hepatitis B by antagonizing cellular inhibitors of apoptosis. Proc Natl Acad Sci USA 112: 5803-5808.
Citation: Dubrez L (2018) SMAC Mimetics: Promising Therapeutic Agents in Cancer and other Diseases. Biochem Pharmacol 7: 244.

Copyright: © 2018 Dubrez L. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top